Skip to main content

EDITORIAL article

Front. Physiol., 28 February 2023
Sec. Cardiac Electrophysiology
This article is part of the Research Topic Transcription Factors and Arrhythmogenesis View all 5 articles

Editorial: Transcription factors and arrhythmogenesis

  • 1Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
  • 2Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
  • 3Department of Internal Medicine, Division of Cardiovascular Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
  • 4Cardiac Electrophysiology and Pacing Laboratory, Division of Cardiovascular Medicine, Makiminato Central Hospital, Urasoe, Japan
  • 5Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
  • 6Department of Medicine, Wilf Family Cardiovascular Research Institute, Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY, United States
  • 7Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Department of Molecular Pharmacology, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY, United States

Editorial on the Research Topic
Transcription factors and arrhythmogenesis

The present Research Topic, entitled “Transcription Factors and Arrhythmogenesis” aims at highlighting the functional role of gene regulatory networks of cardiac enriched transcription factors and their target genes in the pathophysiology of cardiac arrhythmias. Generally speaking, transcription factors bind to the promoter regions of target genes and regulate their expression (Ciccarelli et al., 2011; Larsen et al., 2020; Henley and Koehler, 2021; Tang et al., 2021; de Mattos et al., 2022; Xiao et al., 2022; Jiang and Qian, 2023; Yu et al., 2023; Zhao et al., 2023). Transcription factors can regulate arrhythmogenesis by modulating the expression of genes involved in the cardiac conduction system and/or myocardial structures, as well as through indirect effects on ionic currents, calcium homeostasis, inflammation, oxidative stress, and cardiac remodeling (Liang et al., 2015; Lu et al., 2015; Crespo-Garcia et al., 2022).

Tom McDonald and collaborators elegantly describe the phenotypic variability in iPSC-induced cardiomyocytes and cardiac fibroblasts carrying diverse mutations in the LMNA gene, encoding lamin A/C (Yang et al.). These mutations are known to lead to familial arrhythmogenic cardiomyopathy with a high penetrance; however, there is a phenotypic variability in terms of disease onset, severity, and rate of progression. Recent evidence has shown that induced pluripotent stem cell (iPSC) represent a reliable strategy to create disease-in-a-dish models (Varzideh et al., 2022). Thus, the authors generated seven patient-specific iPSC lines with different LMNA mutations and successfully differentiated them in cardiomyocytes and cardiac fibroblasts. They observed electrophysiological aberrations, sarcomere disarray, and increased apoptosis in cardiomyocytes, and detected several irregularities of the nuclear membrane morphology in cardiac fibroblasts. Intriguingly, co-culture assays of cardiomyocytes and fibroblasts carrying LMNA mutations show exaggerated electrical disturbances (Yang et al.), suggesting that conduction properties of cardiomyocytes might be adversely affected after coculture with fibroblasts in LMNA mutation-associated dilated cardiomyopathy. Additionally, patient- or mutation-specific iPSC may serve as an ideal platform for predicting and testing new effective therapeutics.

The ZFHX3 gene is one of the most studied genes associated with atrial fibrillation (AFib) (Benjamin et al., 2009; Tomomori et al., 2018; Cheng et al., 2019). In a single-center, retrospective, observational cohort study conducted in 1,782 patients who underwent AFib catheter ablation, Hwang et al. found an association between genetic variants of ZFHX3 and extra-pulmonary vein (PV) triggers. Extra-PV triggers are main causes in AFib recurrence post catheter ablation. ZFHX3 knockdown in HL-1 atrial myocytes was found to induce electrical remodeling and increase metabolic stress (Cheng et al., 2019; Lkhagva et al., 2021). These pieces of evidence might explain how extra-PV triggers occur in ZFHX3 genetic variants after ablation.

Qin et al. examined the effects of Gluconolactone (D-glucono-1,5-lactone, GDL) in cardiac ischemia/reperfusion (I/R) injury both in vivo (in mice) and in vitro (in neonatal cardiomyocytes). GDL is a food additive (E-number: E575) present in several dietary products including bread, cheese, wine, yogurt, and tofu (Romo-Rodriguez et al., 2015). The authors observed that GDL attenuated I/R injury and reduced reperfusion-induced arrhythmias and oxidative stress. They also provide a mechanism explaining these findings, showing that GDL acts as a potent activator of PKCε-mediated ERK signaling (Qin et al.). PKCε may provide cardioprotection in I/R injury via activating mitochondrial ALDH2 to scavenge toxic aldehyde-lipid peroxidation products, opening of mitochondrial KATP channels to decrease ROS production and calcium overload, and inhibiting the activation of L-type calcium channels (Inagaki et al., 2006; Ferreira et al., 2012). Moreover, although not directly investigated in the Qin’s paper, PKCε is a known activator of the transcription factors c-Jun and STAT3 (signal transducer and activator of transcription 3) (Batarseh et al., 2010; Wang et al., 2018). Accordingly, GDL was suggested to have cardioprotective potential against I/R injury.

In conclusion, the research area investigating how transcription factors are linked to the pathophysiology of cardiac arrhythmias is quite active, and further studies are warranted to identify novel molecular mechanisms and eventually therapeutic targets to tackle this major issue in cardiovascular medicine.

Author contributions

GS wrote the first draft; All the authors edited the manuscript and approved its final version.

Funding

The GS’s Lab is supported in part by the National Institutes of Health (NIH): National Heart, Lung, and Blood Institute (NHLBI: R01-HL164772, R01-HL159062, R01-HL146691, T32-HL144456), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK: R01-DK123259, R01-DK033823) to GS, National Center for Advancing Translational Sciences (NCATS: UL1TR002556-06) to GS, by the Diabetes Action Research and Education Foundation (to GS), and by the Monique Weill-Caulier and Irma T. Hirschl Trusts (to GS).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Batarseh, A., Li, J., and Papadopoulos, V. (2010). Protein kinase C epsilon regulation of translocator protein (18 kDa) Tspo gene expression is mediated through a MAPK pathway targeting STAT3 and c-Jun transcription factors. Biochemistry 49 (23), 4766–4778. doi:10.1021/bi100020e

PubMed Abstract | CrossRef Full Text | Google Scholar

Benjamin, E. J., Rice, K. M., Arking, D. E., Pfeufer, A., van Noord, C., Smith, A. V., et al. (2009). Variants in ZFHX3 are associated with atrial fibrillation in individuals of European ancestry. Nat. Genet. 41 (8), 879–881. doi:10.1038/ng.416

PubMed Abstract | CrossRef Full Text | Google Scholar

Cheng, W. L., Kao, Y. H., Chao, T. F., Lin, Y. K., Chen, S. A., and Chen, Y. J. (2019). MicroRNA-133 suppresses ZFHX3-dependent atrial remodelling and arrhythmia. Acta Physiol. (Oxf). 227 (3), e13322. doi:10.1111/apha.13322

PubMed Abstract | CrossRef Full Text | Google Scholar

Ciccarelli, M., Sorriento, D., Cipolletta, E., Santulli, G., Fusco, A., Zhou, R. H., et al. (2011). Impaired neoangiogenesis in β₂-adrenoceptor gene-deficient mice: Restoration by intravascular human β₂-adrenoceptor gene transfer and role of NFκB and CREB transcription factors. Br. J. Pharmacol. 162 (3), 712–721. doi:10.1111/j.1476-5381.2010.01078.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Crespo-Garcia, T., Camara-Checa, A., Dago, M., Rubio-Alarcon, M., Rapun, J., Tamargo, J., et al. (2022). Regulation of cardiac ion channels by transcription factors: Looking for new opportunities of druggable targets for the treatment of arrhythmias. Biochem. Pharmacol. 204, 115206. doi:10.1016/j.bcp.2022.115206

PubMed Abstract | CrossRef Full Text | Google Scholar

de Mattos, K., Viger, R. S., and Tremblay, J. J. (2022). Transcription factors in the regulation of leydig cell gene expression and function. Front. Endocrinol. (Lausanne). 13, 881309. doi:10.3389/fendo.2022.881309

PubMed Abstract | CrossRef Full Text | Google Scholar

Ferreira, J. C., Mochly-Rosen, D., and Boutjdir, M. (2012). Regulation of cardiac excitability by protein kinase C isozymes. Front. Biosci. Sch. Ed. 4 (2), 532–546. doi:10.2741/283

PubMed Abstract | CrossRef Full Text | Google Scholar

Henley, M. J., and Koehler, A. N. (2021). Advances in targeting 'undruggable' transcription factors with small molecules. Nat. Rev. Drug Discov. 20 (9), 669–688. doi:10.1038/s41573-021-00199-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Inagaki, K., Churchill, E., and Mochly-Rosen, D. (2006). Epsilon protein kinase C as a potential therapeutic target for the ischemic heart. Cardiovasc. Res. 70 (2), 222–230. doi:10.1016/j.cardiores.2006.02.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiang, Y., and Qian, H. Y. (2023). Transcription factors: Key regulatory targets of vascular smooth muscle cell in atherosclerosis. Mol. Med. 29 (1), 2. doi:10.1186/s10020-022-00586-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Larsen, A. I., Valborgland, T., Ogne, C., Lindal, S., Halvorsen, B., Munk, P. S., et al. (2020). Plasma tumour necrosis factor correlates with mRNA expression of tumour necrosis factor and mitochondrial transcription factors in skeletal muscle in patients with chronic heart failure treated with cardiac resynchronization therapy: Potential role in myopathy. Eur. J. Prev. Cardiol. 27 (19), 2362–2366. doi:10.1177/2047487319855796

PubMed Abstract | CrossRef Full Text | Google Scholar

Liang, X., Zhang, Q., Cattaneo, P., Zhuang, S., Gong, X., Spann, N. J., et al. (2015). Transcription factor ISL1 is essential for pacemaker development and function. J. Clin. Invest. 125 (8), 3256–3268. doi:10.1172/JCI68257

PubMed Abstract | CrossRef Full Text | Google Scholar

Lkhagva, B., Lin, Y. K., Chen, Y. C., Cheng, W. L., Higa, S., Kao, Y. H., et al. (2021). ZFHX3 knockdown dysregulates mitochondrial adaptations to tachypacing in atrial myocytes through enhanced oxidative stress and calcium overload. Acta Physiol. (Oxf). 231 (4), e13604. doi:10.1111/apha.13604

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, L., Sirish, P., Zhang, Z., Woltz, R. L., Li, N., Timofeyev, V., et al. (2015). Regulation of gene transcription by voltage-gated L-type calcium channel, Cav1.3. J. Biol. Chem. 290 (8), 4663–4676. doi:10.1074/jbc.M114.586883

PubMed Abstract | CrossRef Full Text | Google Scholar

Romo-Rodriguez, P., Acevedo-Aguilar, F. J., Lopez-Torres, A., Wrobel, K., Wrobel, K., and Gutierrez-Corona, J. F. (2015). Cr(VI) reduction by gluconolactone and hydrogen peroxide, the reaction products of fungal glucose oxidase: Cooperative interaction with organic acids in the biotransformation of Cr(VI). Chemosphere 134, 563–570. doi:10.1016/j.chemosphere.2014.12.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Tang, X. H., Gambardella, J., Jankauskas, S., Wang, X., Santulli, G., Gudas, L. J., et al. (2021). A retinoic acid receptor beta (2) agonist improves cardiac function in a heart failure model. J. Pharmacol. Exp. Ther. 379 (2), 182–190. doi:10.1124/jpet.121.000806

PubMed Abstract | CrossRef Full Text | Google Scholar

Tomomori, S., Nakano, Y., Ochi, H., Onohara, Y., Sairaku, A., Tokuyama, T., et al. (2018). Maintenance of low inflammation level by the ZFHX3 SNP rs2106261 minor allele contributes to reduced atrial fibrillation recurrence after pulmonary vein isolation. PLoS One 13 (9), e0203281. doi:10.1371/journal.pone.0203281

PubMed Abstract | CrossRef Full Text | Google Scholar

Varzideh, F., Mone, P., and Santulli, G. (2022). Bioengineering strategies to create 3D cardiac constructs from human induced pluripotent stem cells. Bioengineering 9 (4), 168. doi:10.3390/bioengineering9040168

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, C., Li, H., Wang, S., Mao, X., Yan, D., Wong, S. S., et al. (2018). Repeated non-invasive limb ischemic preconditioning confers cardioprotection through PKC-ε/STAT3 signaling in diabetic rats. Cell Physiol. Biochem. 45 (5), 2107–2121. doi:10.1159/000488047

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiao, D., Caldow, M., Kim, H. J., Blazev, R., Koopman, R., Manandi, D., et al. (2022). Time-resolved phosphoproteome and proteome analysis reveals kinase signaling on master transcription factors during myogenesis. iScience 25 (6), 104489. doi:10.1016/j.isci.2022.104489

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, C., Li, X., Zhao, Y., and Hu, Y. (2023). The role of FOXA family transcription factors in glucolipid metabolism and NAFLD. Front. Endocrinol. (Lausanne) 14, 1081500. doi:10.3389/fendo.2023.1081500

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, J., Yu, L., Xue, X., Xu, Y., Huang, T., Xu, D., et al. (2023). Diminished α7 nicotinic acetylcholine receptor (α7nAChR) rescues amyloid-β induced atrial remodeling by oxi-CaMKII/MAPK/AP-1 axis-mediated mitochondrial oxidative stress. Redox Biol. 59, 102594. doi:10.1016/j.redox.2022.102594

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: cardiovascular medicine, induced pluripotent stem cell (iPSC), LMNA, ZFHX3, bioengineering, cardiac arrhythmia, PKCε

Citation: Kao Y-H, Chen Y-J, Higa S, Chattipakorn N and Santulli G (2023) Editorial: Transcription factors and arrhythmogenesis. Front. Physiol. 14:1169747. doi: 10.3389/fphys.2023.1169747

Received: 20 February 2023; Accepted: 21 February 2023;
Published: 28 February 2023.

Edited and reviewed by:

Ruben Coronel, University of Amsterdam, Netherlands

Copyright © 2023 Kao, Chen, Higa, Chattipakorn and Santulli. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Yu-Hsun Kao, yuhsunkao@gmail.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.