Skip to main content

EDITORIAL article

Front. Physiol., 10 August 2022
Sec. Cardiac Electrophysiology
This article is part of the Research Topic Interplay between the heart and the immune system: Focus on heart rhythm regulation View all 5 articles

Editorial: Interplay between the heart and the immune system: Focus on heart rhythm regulation

  • 1Almazov National Medical Research Centre, Saint Petersburg, Russia
  • 2Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia

Activation or dysregulation of the immune system plays an important role in the development and progression of many cardiovascular diseases and cardiac arrhythmias. Atrial fibrillation (AF) is the most common cardiac arrhythmia. Inflammation contributes to both electrical and structural atrial remodeling and thrombosis in patients with AF, and therapy targeting specific inflammatory cascades may be a potential strategy to prevent and treat AF (Hu et al., 2015). However, AF has a complex and multifactorial developmental mechanism that involves more than just inflammation.

Clinical studies have shown that AF can be triggered by autonomic stimulation, bradycardia, atrial premature beats, tachycardia, accessory pathways, and acute atrial stretch (Weil and Ozcan, 2015). The electrical or focal mechanisms of AF include abnormal automatism, trigger activity and multiple variable reciprocal patterns (Wakili et al., 2011; January et al., 2014). Reduced L-type Ca2+ (ICaL) current, Ca2+ overload, changes in K+ current (IKACh, IK1), Na+ current (INa), and transient outward current (Ito) have each been reported in AF (Nattel, 2002). In turn, ionic currents can directly affect the function of cardiomyocyte mitochondria, since it was previously shown that a high content of Ca2+ inhibits mitochondrial respiration, dissipates the membrane potential, and suppresses ATP production (Holmuhamedov et al., 2001; Jahangir et al., 2001). Remodeling of the calcium cycle can contribute to the progression of AF to persistent and activate profibrotic pathways (Denham et al., 2018).

In addition to the electrophysiological aspects, the complex anatomical structure of the atria is of great importance (Platonov et al., 2008). It is known that the conduction of an electrical impulse goes through wide muscle bundles with a parallel arrangement of muscle fibers (Anderson et al., 1983). The anatomy of the pulmonary veins promotes re-entry due to the combination of abrupt changes in fiber orientation and reduced electrical connectivity between muscle bundles creating areas of heterogeneous conduction velocity and localized block (Hocini et al., 2002; Arora et al., 2003; Hsueh et al., 2013).

Mounting evidence has shown that cardiomyocytes are not the only cells of the heart with an electrical activity. Recent studies have shown that fibroblasts express voltage-gated sodium channels that provide inward current flow (INa) (Chatelier et al., 2012; Koivumäki et al., 2014; Sánchez et al., 2019). Although they are not electrically excitable, they can affect the electrophysiological properties of myocytes (Gaudesius et al., 2003). Fibroblasts have been found to electrically react with myocytes through gap junctions (Jousset et al., 2016). Fibroblasts become active under inflammatory response and then differentiate into myofibroblasts (Chacar et al., 2017) by different pathways (Jalife and Kaur, 2015). Myofibroblasts differ from fibroblasts in that they develop contractile proteins and exhibit a more depolarized resting membrane potential (Salvarani et al., 2017) and greater membrane capacitance (Sridhar et al., 2017).

Recent investigations have shown that the myocardium contains a 3D network of telocytes that tightly cover cardiomyocytes and are involved in contacts with all types of cells and structures (Hinescu and Popescu, 2005; Popescu et al., 2005; Cretoiu et al., 2014). In addition to gap junctions, telocytes have point, nanocontacts, and planar contacts with cells. They can penetrate the basement membrane that envelopes two P cells of the cardiac conduction system together (Mitrofanova et al., 2018). Sheng et al. (2014) demonstrated that atrial and ventricular telocytes express Ca2⁺ activated high conductive K⁺ current (BK(Ca)) and internal rectifying K⁺ current (IK(ir)), but not transient external K⁺ current (I (to)) and an ATP-sensitive potassium current (K (ATP)). The results of these authors showed that functionally competent K+ channels are present in human heart telocytes, and their modulation may be of significant importance in myocardial physiopathology.

Cardiac macrophages have been recently demonstrated to also play an important role in both electrophysiology and arrhythmogenesis. Cardiac macrophages are a heterogeneous group of immune cells including resident macrophages derived from embryonic and fetal progenitors and recruited macrophages derived from circulating bone marrow monocytes (Xia R. et al.).

Several studies have demonstrated specific macrophage-dependent mechanisms regulating electrical, structural, or autonomic remodeling leading to arrhythmias (Monnerat et al., 2016; Liu et al., 2019; Miyosawa et al., 2020; Hiram et al., 2021). Hulsmans et al. (2017) have shown that cardiac macrophages facilitate electrical conduction through the distal atrioventricular node, where conducting cells densely intersperse with elongated macrophages expressing connexin 43. Disruption of the macrophage-cardiomyocyte interaction results in impaired electrical conduction in the AV node. Could it be telocytes? Simon-Chica et al. (2022) revealed that resident murine cardiac macrophages express potassium channels including Kv1.3, Kv1.5, and Kir2.1 which establish several inward and outward rectifying currents. The interaction between macrophages and cardiac fibroblasts regulates the balance of cardiac fibrosis (Van Linthout et al., 2014).

Currently, many researchers believe that inflammatory processes are key factors in the pathophysiology of AF [Ihara and Sasano]. The incidence of AF increases in the presence of systemic inflammation (Christian et al., 2008). Cases of occurrence of AF in COVID-19 are described (Gaine et al., 2021). AF may be associated with chronic inflammation (Choi et al., 2019; Lazzerini et al., 2019). Inflammation can be systemic or focal (eg, myocarditis) (Subahi et al., 2019). It is quite expected, that increased levels of inflammatory markers (e.g., CRP, IL-6, IL-8, TNF-α etc.) are described in patients with AF (Patel et al., 2010).

Traditional clinical practice has failed in several areas such as identifying patients at risk for AF or patients with concomitant undiagnosed paroxysmal AF. New approaches using artificial intelligence may provide new tools to solve some of these old problems (Isaksen et al., 2022). In this regard, the study of Jiang et al. is extremely relevant. The authors showed that aging, overweight, hypertension, diabetes and smoking seemed to be associated with high CRP levels. This study proved the existence of inflammation-related changes in cardiac electrophysiological signals.

The autonomic nervous system plays an important role in the onset and maintenance of AF. Catheter ablation of ganglionated plexi (GP) in the left and right atrium has been proposed in varied clinical conditions. The benefit of adding autonomic ganglion ablation to the standard pulmonary vein exposure procedure for patients with paroxysmal AF is supported by both experimental and clinical data (Stavrakis et al., 2015; Rebecchi et al., 2021). The original experimental study of Ma et al. convincingly demonstrates that local administration of TRAM-34, an inhibitor of intermediate-conductance KCa channels (SK4), into the atrial GP can suppress GP activity and AF vulnerability during rapid atrial pacing. The effects of TRAM-34 may be related to the macrophage polarization and inflammatory response of GP. This study once again underlines the multilevel and complexity of the pathogenesis of AF and opens up new prospects for treatment.

In sum, the articles in this Research Topic demonstrate the complexity of the pathogenesis of AF, which has many levels: electrophysiological, anatomical, histological, cellular, ultrastructural and molecular. Inflammation is the key link. Further study of the pathogenesis of cardiac arrhythmias with the involvement of genetics, the study of the role of transcription factors, LncRNAs and others will allow moving from surgery and pharmacology to gene therapy.

Author contributions

LM and SP prepared the manuscript.

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

Anderson R. H., Ho S. Y., Becker A. E. (1983). The surgical anatomy of the conduction tissues. Thorax 38 (6), 408–420. doi:10.1136/thx.38.6.408

PubMed Abstract | CrossRef Full Text | Google Scholar

Arora R., Verheule S., Scott L., Navarrete A., Katari V., Wilson E., et al. (2003). Arrhythmogenic substrate of the pulmonary veins assessed by high-resolution optical mapping. Circulation 107 (13), 1816–1821. doi:10.1161/01.CIR.0000058461.86339.7E

PubMed Abstract | CrossRef Full Text | Google Scholar

Chacar S., Farès N., Bois P., Faivre J. F. (2017). Basic signaling in cardiac fibroblasts. J. Cell. Physiol. 232 (4), 725–730. doi:10.1002/jcp.25624

PubMed Abstract | CrossRef Full Text | Google Scholar

Chatelier A., Mercier A., Tremblier B., Theriault O., Moubarak M., Benamer N., et al. (2012). A distinct de novo expression of Nav1.5 sodium channels in human atrial fibroblasts differentiated into myofibroblasts. J. Physiol. 590 (17), 4307–4319. doi:10.1113/jphysiol.2012.233593

PubMed Abstract | CrossRef Full Text | Google Scholar

Choi Y.-J., Choi E.-K., Han K.-D., Park J., Moon I., Lee E., et al. (2019). Increased risk of atrial fibrillation in patients with inflammatory bowel disease: a nationwide population-based study. World J. Gastroenterol. 25, 2788–2798. doi:10.3748/wjg.v25.i22.2788

PubMed Abstract | CrossRef Full Text | Google Scholar

Christian S. A., Schorr C., Ferchau L., Jarbrink M. E., Parrillo J. E., Gerber D. R. (2008). Clinical characteristics and outcomes of septic patients with new-onset atrial fibrillation. J. Crit. Care 23 (4), 532–536. doi:10.1016/j.jcrc.2007.09.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Cretoiu D., Hummel E., Zimmermann H., Gherghiceanu M., Popescu L. M. (2014). Human cardiac telocytes: 3D imaging by FIB-SEM tomography. J. Cell. Mol. Med. 18 (11), 2157–2164. doi:10.1111/jcmm.12468

PubMed Abstract | CrossRef Full Text | Google Scholar

Denham N. C., Pearman C. M., Caldwell J. L., Madders G. W. P., Eisner D. A., Trafford A. W., et al. (2018). Calcium in the pathophysiology of atrial fibrillation and heart failure. Front. Physiol. 9, 1380. doi:10.3389/fphys.2018.01380

PubMed Abstract | CrossRef Full Text | Google Scholar

Gaine S., Devitt P., Coughlan J. J., Pearson I. (2021). COVID-19-associated myocarditis presenting as new-onset heart failure and atrial fibrillation. BMJ Case Rep. 14, e244027. doi:10.1136/bcr-2021-244027

PubMed Abstract | CrossRef Full Text | Google Scholar

Gaudesius G., Miragoli M., Thomas S. P., Rohr S. (2003). Coupling of cardiac electrical activity over extended distances by fibroblasts of cardiac origin. Circ. Res. 93 (5), 421–428. doi:10.1161/01.RES.0000089258.40661.0C

PubMed Abstract | CrossRef Full Text | Google Scholar

Hinescu M. E., Popescu L. M. (2005). Interstitial Cajal-like cells (ICLC) in human atrial myocardium. J. Cell. Mol. Med. 9 (4), 972–975. doi:10.1111/j.1582-4934.2005.tb00394.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Hiram R., Xiong F., Naud P., Xiao J., Sirois M., Tanguay J. F., et al. (2021). The inflammation-resolution promoting molecule resolvin-D1 prevents atrial proarrhythmic remodelling in experimental right heart disease. Cardiovasc. Res. 117 (7), 1776–1789. doi:10.1093/cvr/cvaa186

PubMed Abstract | CrossRef Full Text | Google Scholar

Hocini M., Ho S. Y., Kawara T., Linnenbank A. C., Potse M., Shah D., et al. (2002). Electrical conduction in canine pulmonary veins: electrophysiological and anatomic correlation. Circulation 105 (20), 2442–2448. doi:10.1161/01.cir.0000016062.80020.11

PubMed Abstract | CrossRef Full Text | Google Scholar

Holmuhamedov E. L., Ozcan C., Jahangir A., Terzic A. (2001). Restoration of Ca2+-inhibited oxidative phosphorylation in cardiac mitochondria by mitochondrial Ca2+ unloading. Mol. Cell. Biochem. 220 (1-2), 135–140. doi:10.1023/a:1010894427373

PubMed Abstract | CrossRef Full Text | Google Scholar

Hsueh C. H., Chang P. C., Hsieh Y. C., Reher T., Chen P. S., Lin S. F. (2013). Proarrhythmic effect of blocking the small conductance calcium activated potassium channel in isolated canine left atrium. Heart Rhythm 10 (6), 891–898. doi:10.1016/j.hrthm.2013.01.033

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu Y. F., Chen Y. J., Lin Y. J., Chen S. A. (2015). Inflammation and the pathogenesis of atrial fibrillation. Nat. Rev. Cardiol. 12, 230–243. doi:10.1038/nrcardio.2015.2

PubMed Abstract | CrossRef Full Text | Google Scholar

Hulsmans M., Clauss S., Xiao L., Aguirre A. D., King K. R., Hanley A., et al. (2017). Macrophages facilitate electrical conduction in the heart. Cell 169 (3), 510–522. doi:10.1016/j.cell.2017.03.050

PubMed Abstract | CrossRef Full Text | Google Scholar

Isaksen J. L., Baumert M., Hermans A. N. L., Maleckar M., Linz D. (2022). Artificial intelligence for the detection, prediction, and management of atrial fibrillation. Herzschrittmacherther Elektrophysiol 33, 34–41. doi:10.1007/s00399-022-00839-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Jahangir A., Ozcan C., Holmuhamedov E. L., Terzic A. (2001). Increased calcium vulnerability of senescent cardiac mitochondria: protective role for a mitochondrial potassium channel opener. Mech. Ageing Dev. 122 (10), 1073–1086. doi:10.1016/s0047-6374(01)00242-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Jalife J., Kaur K. (2015). Atrial remodeling, fibrosis, and atrial fibrillation. Trends Cardiovasc. Med. 25 (6), 475–484. doi:10.1016/j.tcm.2014.12.015

PubMed Abstract | CrossRef Full Text | Google Scholar

January C. T., Wann L. S., Alpert J. S., Calkins H., Cigarroa J. E., Cleveland J. C., et al. (2014). 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American college of cardiology/American heart association task force on practice guidelines and the heart rhythm society. Circulation 130 (23), e199–267. doi:10.1161/CIR.0000000000000041

PubMed Abstract | CrossRef Full Text | Google Scholar

Jousset F., Maguy A., Rohr S., Kucera J. P. (2016). Myofibroblasts electrotonically coupled to cardiomyocytes alter conduction: insights at the cellular level from a detailed in silico tissue structure model. Front. Physiol. 7, 496. doi:10.3389/fphys.2016.00496

PubMed Abstract | CrossRef Full Text | Google Scholar

Koivumäki J. T., Clark R. B., Belke D., Kondo C., Fedak P. W. M., Maleckar M. M. C., et al. (2014). Na(+) current expression in human atrial myofibroblasts: identity and functional roles. Front. Physiol. 5, 275. doi:10.3389/fphys.2014.00275

PubMed Abstract | CrossRef Full Text | Google Scholar

Lazzerini P. E., Laghi-Pasini F., Acampa M., Srivastava U., Bertolozzi I., Giabbani B., et al. (2019). Systemic inflammation rapidly induces reversible atrial electrical remodeling: the role of interleukin-6-mediated changes in connexin expression. J. Am. Heart Assoc. 8, e011006. doi:10.1161/JAHA.118.011006

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu L., Gan S., Li B., Ge X., Yu H., Zhou H. (2019). Fisetin alleviates atrial inflammation, remodeling, and vulnerability to atrial fibrillation after myocardial infarction. Int. Heart J. 60 (6), 1398–1406. doi:10.1536/ihj.19-131

PubMed Abstract | CrossRef Full Text | Google Scholar

Mitrofanova L. B., Gorshkov A. N., Konovalov P. V., Krylova J. S. (2018). Telocytes in the human sinoatrial node. J. Cell. Mol. Med. 22 (1), 521–532. doi:10.1111/jcmm.13340

PubMed Abstract | CrossRef Full Text | Google Scholar

Miyosawa K., Iwata H., Minami-Takano A., Hayashi H., Tabuchi H., Sekita G., et al. (2020). Enhanced monocyte migratory activity in the pathogenesis of structural remodeling in atrial fibrillation. PLoS One 15 (10), e0240540. doi:10.1371/journal.pone.0240540

PubMed Abstract | CrossRef Full Text | Google Scholar

Monnerat G., Alarcón M. L., Vasconcellos L. R., Hochman-Mendez C., Brasil G., Bassani R. A., et al. (2016). Macrophage-dependent IL-1β production induces cardiac arrhythmias in diabetic mice. Nat. Commun. 7, 13344. doi:10.1038/ncomms13344

PubMed Abstract | CrossRef Full Text | Google Scholar

Nattel S. (2002). New ideas about atrial fibrillation 50 years on. Nature 415 (6868), 219–226. doi:10.1038/415219a

PubMed Abstract | CrossRef Full Text | Google Scholar

Patel P., Dokainish H., Tsai P., Lakkis N. (2010). Update on the association of inflammation and atrial fibrillation. J. Cardiovasc. Electrophysiol. 21 (9), 1064–1070. doi:10.1111/j.1540-8167.2010.01774.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Platonov P. G., Mitrofanova L., Ivanov V., Ho S. Y. (2008). Substrates for intra-atrial and interatrial conduction in the atrial septum: anatomical study on 84 human hearts. Heart Rhythm 5 (8), 1189–1195. doi:10.1016/j.hrthm.2008.04.025

PubMed Abstract | CrossRef Full Text | Google Scholar

Popescu L. M., Gherghiceanu M., Cretoiu D., Radu E. (2005). The connective connection: interstitial cells of cajal (ICC) and ICC-like cells establish synapses with immunoreactive cells. Electron microscope study in situ. J. Cell. Mol. Med. 9 (3), 714–730. doi:10.1111/j.1582-4934.2005.tb00502.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Rebecchi M., Panattoni G., Edoardo B., de Ruvo E., Sciarra L., Politano A., et al. (2021). Atrial fibrillation and autonomic nervous system: a translational approach to guide therapeutic goals. J. Arrhythm. 37 (2), 320–330. doi:10.1002/joa3.12512

PubMed Abstract | CrossRef Full Text | Google Scholar

Salvarani N., Maguy A., De Simone S. A., Miragoli M., Jousset F., Rohr S. (2017). TGF-β1 (transforming growth factor-β1) plays a pivotal role in cardiac myofibroblast arrhythmogenicity. Circ. Arrhythm. Electrophysiol. 10 (5), e004567. doi:10.1161/CIRCEP.116.004567

PubMed Abstract | CrossRef Full Text | Google Scholar

Sánchez J., Gomez J. F., Martinez-Mateu L., Romero L., Saiz J., Trenor B. (2019). Heterogeneous effects of fibroblast-myocyte coupling in different regions of the human atria under conditions of atrial fibrillation. Front. Physiol. 10, 847. doi:10.3389/fphys.2019.00847

PubMed Abstract | CrossRef Full Text | Google Scholar

Sheng J., Shim W., Lu J., Lim S. Y., Ong B. H., Lim T. S., et al. (2014). Electrophysiology of human cardiac atrial and ventricular telocytes. J. Cell. Mol. Med. 18 (2), 355–362. doi:10.1111/jcmm.12240

PubMed Abstract | CrossRef Full Text | Google Scholar

Simon-Chica A., Fernández M. C., Wülfers E. M., Lother A., Hilgendorf I., Seemann G., et al. (2022). Novel insights into the electrophysiology of murine cardiac macrophages: relevance of voltage-gated potassium channels. Cardiovasc. Res. 118 (3), 798–813. doi:10.1093/cvr/cvab126

PubMed Abstract | CrossRef Full Text | Google Scholar

Sridhar S., Vandersickel N., Panfilov A. V. (2017). Effect of myocyte-fibroblast coupling on the onset of pathological dynamics in a model of ventricular tissue. Sci. Rep. 7, 40985. doi:10.1038/srep40985

PubMed Abstract | CrossRef Full Text | Google Scholar

Stavrakis S., Nakagawa H., Po S. S., Scherlag B. J., Lazzara R., Jackman W. M. (2015). The role of the autonomic ganglia in atrial fibrillation. JACC Clin. Electrophysiol. 1 (1-2), 1–13. doi:10.1016/j.jacep.2015.01.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Subahi A., Akintoye E., Yassin A. S., Abubakar H., Adegbala O., Mishra T., et al. (2019). Impact of atrial fibrillation on patients hospitalized for acute myocarditis: insights from a nationally-representative United States cohort. Clin. Cardiol. 42 (1), 26–31. doi:10.1002/clc.23088

PubMed Abstract | CrossRef Full Text | Google Scholar

Van Linthout S., Miteva K., Tschöpe C. (2014). Crosstalk between fibroblasts and inflammatory cells. Cardiovasc. Res. 102 (2), 258–269. doi:10.1093/cvr/cvu062

PubMed Abstract | CrossRef Full Text | Google Scholar

Wakili R., Voigt N., Kääb S., Dobrev D., Nattel S. (2011). Recent advances in the molecular pathophysiology of atrial fibrillation. J. Clin. Invest. 121 (8), 2955–2968. doi:10.1172/JCI46315

PubMed Abstract | CrossRef Full Text | Google Scholar

Weil B. R., Ozcan C. (2015). Cardiomyocyte remodeling in atrial fibrillation and hibernating myocardium: shared pathophysiologic traits identify novel treatment strategies? Biomed. Res. Int. 2015, 587361. doi:10.1155/2015/587361

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: atrial fibrillation, inflammation, pathogenesis, macrophages, artificial intelligence

Citation: Mitrofanova L and Popov S (2022) Editorial: Interplay between the heart and the immune system: Focus on heart rhythm regulation. Front. Physiol. 13:981499. doi: 10.3389/fphys.2022.981499

Received: 29 June 2022; Accepted: 15 July 2022;
Published: 10 August 2022.

Edited and reviewed by:

Gaetano Santulli, Albert Einstein College of Medicine, United States

Copyright © 2022 Mitrofanova and Popov. 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: Lubov Mitrofanova, lubamitr@yandex.ru

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.