Piezo1 and BKCa channels in human atrial fibroblasts: Interplay and remodelling in atrial fibrillation

https://doi.org/10.1016/j.yjmcc.2021.05.002Get rights and content

Highlights

  • Piezo1 and BKCa are present and mechano-modulated in human atrial fibroblasts.

  • Fibroblast Piezo1 activity is larger in atrial fibrillation compared to sinus rhythm.

  • Fibroblast BKCa activity is smaller in atrial fibrillation compared to sinus rhythm.

  • BKCa mechanosensitivity depends in part on stretch-activated Ca2+ influx via Piezo1.

  • Piezo1 and BKCa channels are linked functionally, but not structurally.

Abstract

Aims

Atrial Fibrillation (AF) is an arrhythmia of increasing prevalence in the aging populations of developed countries. One of the important indicators of AF is sustained atrial dilatation, highlighting the importance of mechanical overload in the pathophysiology of AF. The mechanisms by which atrial cells, including fibroblasts, sense and react to changing mechanical forces, are not fully elucidated. Here, we characterise stretch-activated ion channels (SAC) in human atrial fibroblasts and changes in SAC- presence and activity associated with AF.

Methods and results

Using primary cultures of human atrial fibroblasts, isolated from patients in sinus rhythm or sustained AF, we combine electrophysiological, molecular and pharmacological tools to identify SAC. Two electrophysiological SAC- signatures were detected, indicative of cation-nonselective and potassium-selective channels. Using siRNA-mediated knockdown, we identified the cation-nonselective SAC as Piezo1. Biophysical properties of the potassium-selective channel, its sensitivity to calcium, paxilline or iberiotoxin (blockers), and NS11021 (activator), indicated presence of calcium-dependent ‘big potassium channels’ (BKCa). In cells from AF patients, Piezo1 activity and mRNA expression levels were higher than in cells from sinus rhythm patients, while BKCa activity (but not expression) was downregulated. Both Piezo1-knockdown and removal of extracellular calcium from the patch pipette resulted in a significant reduction of BKCa current during stretch. No co-immunoprecipitation of Piezo1 and BKCa was detected.

Conclusions

Human atrial fibroblasts contain at least two types of ion channels that are activated during stretch: Piezo1 and BKCa. While Piezo1 is directly stretch-activated, the increase in BKCa activity during mechanical stimulation appears to be mainly secondary to calcium influx via SAC such as Piezo1. During sustained AF, Piezo1 is increased, while BKCa activity is reduced, highlighting differential regulation of both channels. Our data support the presence and interplay of Piezo1 and BKCa in human atrial fibroblasts in the absence of physical links between the two channel proteins.

Introduction

Atrial Fibrillation (AF) is a supraventricular arrhythmia with increasing prevalence in countries with an aging population. Although AF is one of the most common cardiovascular causes of hospitalization [[1], [2], [3]], its pathophysiology is not fully elucidated, and it represents an unmet need for effective prevention and treatment. One hallmark of AF is its progressive nature. As AF becomes increasingly resistant over time to pharmacological or electrical attempts at conversion back to sinus rhythm (SR) [2], atrial tissue undergoes pronounced remodelling [2,4]. Structural and functional changes involve cell electrophysiological and tissue morphological alterations. Whilst electrical remodelling of atrial cardiomyocytes is characterised by a shortening of action potential duration and of effective refractory period, as well as by impaired adaptation of these parameters to changes in heart rate [5], fibrosis – a prominent feature of AF-related structural remodelling – may in parallel contribute to slowing of conduction. The combination of short effective refractory period and slow conduction favours maintenance of AF via re-entry mechanisms [6].

Many of the risk factors for AF, e.g. heart failure, hypertension, or valvulopathies, are accompanied by mechanical overload of the atria [7]. Since stretch enhances the susceptibility to AF induction [8,9], it has been suggested, that mechanical overload may contribute to initiation and perpetuation of AF in vivo [[10], [11], [12], [13]]. In addition, acute stretch of control atrial tissue induces complex and regionally varying changes in action potential shape [10] and diastolic depolarizations that may trigger extrasystoles [9,14,15]. This ‘mechano-electric feedback’ [16,17] requires cells to be able to sense their mechanical environment, and to translate this into an electrophysiologically relevant signal.

Ample evidence points to an essential role of stretch-activated ion channels (SAC) as mechano-sensors in cardiomyocytes (for reviews see [18,19]). SAC are also present and functional in human atrial fibroblasts [20,21], but it is currently not known whether SAC function is altered in AF in the various human heart cells, especially in fibroblasts, which are key players in fibrosis. Therefore, the aim of this study was to compare SAC function in atrial fibroblasts from patients in SR or in sustained AF. The cation-nonselective SAC Piezo1 [[22], [23], [24]] forms a plausible candidate, in line with recently reported Piezo1 effects on remodelling of non-cardiac tissues [25]. A second candidate, the potassium-selective Ca2+-activated channel of large conductance (BKCa), has been reported to respond to stretch, local Ca2+ concentration changes, transforming growth factor beta (TGF-β), and angiotensin II in several cardiac cell types [[26], [27], [28], [29], [30]]. BKCa is further known to modulate fibroblast proliferation [31], a critical event during pathological tissue remodelling in AF. Both Piezo1 and BKCa have previously been detected in human atrial fibroblasts [[31], [32], [33]].

In this study we report AF-related changes in Piezo1 and BKCa channel activity in human atrial fibroblasts, and establish functional interactions between the two channel types.

Section snippets

Tissue collection

Tissue samples were obtained from the right atrial appendage of patients undergoing open-heart surgery. Patients were either in SR, or in sustained AF (which includes patients with persistent, long-standing persistent and permanent AF, defined according to ESC Guidelines [34]). Tissue samples were processed by the Cardiovascular Biobank of the University Heart Center Freiburg Bad Krozingen (approved by the ethics committee of Freiburg University, No 393/16; 214/18) or the Clinical Center of the

Fibroblasts and myofibroblasts are the main constituents of right atrial outgrowth cell cultures

Cells obtained by the outgrowth technique from human right atrial appendage did not contain any cardiomyocytes. The majority (98%) of cells stained positive for vimentin, and 1% of the cells were positive for the endothelial cell marker CD31. Antibody functionality was verified in positive controls using human umbilical vein endothelial cell (HUVEC; Fig. S1A and B). Overall, results confirmed that the outgrowth technique yields predominantly fibroblasts-like cells with negligible contamination

Discussion

We confirm the presence, in human right atrial fibroblasts from patients in SR and AF, of at least two different types of ion currents that are activated during stretch: the cation-nonselective Piezo1 and the potassium-selective BKCa. Our main findings are: (i) activity and expression of Piezo1 are larger in non-passaged right atrial fibroblasts from AF patients than in cells from SR patients; (ii) activity, but not expression, of BKCa channels is lower in AF than in SR cells; (iii)

Author contributions

DJ, AK, SNH, PK, UR and RP contributed to conception, design and interpretation of the study. DJ, AK and ED performed and analysed electrophysiological experiments. DA, EARZ, SP and CS performed and analysed quantitative RT-PCR experiments. DJ and TG performed and analysed immunocytochemical experiments. DA and ASC isolated cells. BA and HG performed and analysed the co-immunoprecipitation and localisation experiments. RE performed the PCR to assess the presence of STREX. SRK provided HAF

Funding

This work was supported by the ERC Advanced Grant CardioNECT (project ID: #323099, PK), a research grant from the Ministry of Science, Research and Arts Baden-Württemberg (MWK-BW Sonderlinie Medizin, #3091311631, PK), and a DFG Emmy Noether Fellowship (to EARZ, 285 #396913060). This research was funded by the German Research Foundation (DFG) under the Excellence Strategy (CIBSS-EXC-2189-Project ID 390939984). ED, PK, UR and RP acknowledge support by Amgen Inc. ED, RE, ASC, EARZ, FB, FAK, CS,

Declaration of Competing Interest

None.

Acknowledgments

The authors thank all colleagues at the Department for Cardiovascular Surgery of the University Heart Centre Freiburg - Bad Krozingen, and at the CardioVascular BioBank Freiburg, for providing access to human atrial tissue. Special thanks for technical support go to Cinthia Walz, Anne Hetkamp, Kristina Kollmar and Gabriele Lechner. We would like to also thank Dr. Simone Nübling and Dr. Hannah Fürniss for their help concerning patient demographics. We thank Dr. Bo Bentzen (University of

References (68)

  • S. Candia et al.

    Mode of action of iberiotoxin, a potent blocker of the large conductance Ca 2+-activated K+ channel

    Biophys. J.

    (1992)
  • K. Takahashi et al.

    Stretch-activated BK channel and heart function

    Prog. Biophys. Mol. Biol.

    (2012)
  • A.H. Lewis et al.

    Transduction of repetitive mechanical stimuli by Piezo1 and Piezo2 ion channels

    Cell Rep.

    (2017)
  • S. Jha et al.

    The β1 subunit of Na+/K+-ATPase interacts with BKCa channels and affects their steady-state expression on the cell surface

    FEBS Lett.

    (2009)
  • L. Han et al.

    Canonical transient receptor potential 3 channels in atrial fibrillation

    Eur. J. Pharmacol.

    (2018)
  • J. Davis et al.

    A TRPC6-dependent pathway for myofibroblast transdifferentiation and wound healing in vivo

    Dev. Cell

    (2012)
  • S.N. Psychari et al.

    Relation of elevated C-reactive protein and interleukin-6 levels to left atrial size and duration of episodes in patients with atrial fibrillation

    Am. J. Cardiol.

    (2005)
  • G.M. Marcus et al.

    Interleukin-6 and atrial fibrillation in patients with coronary artery disease: data from the heart and soul study

    Am. Heart J.

    (2008)
  • Y. Gao et al.

    ANO1 inhibits cardiac fibrosis after myocardial infraction via TGF-β/smad3 pathway

    Sci. Rep.

    (2017)
  • C.X. Wong et al.

    The increasing burden of atrial fibrillation compared with heart failure and myocardial infarction: a 15-year study of all hospitalizations in Australia

    Arch. Intern. Med.

    (2012)
  • H. Zulkifly et al.

    Epidemiology of atrial fibrillation

    Int. J. Clin. Pract.

    (2018)
  • L. Staerk et al.

    Atrial fibrillation: epidemiology, pathophysiology, and clinical outcomes

    Circ. Res.

    (2017)
  • M.C. Wijffels et al.

    Atrial fibrillation begets atrial fibrillation. A study in awake chronically instrumented goats

    Circulation

    (1995)
  • D. Dobrev et al.

    Remodeling of cardiomyocyte ion channels in human atrial fibrillation

    Basic Res. Cardiol.

    (2003)
  • K. Kumagai et al.

    Electrophysiological properties in chronic lone atrial fibrillation

    Circulation

    (1991)
  • B.M. Psaty et al.

    Incidence of and risk factors for atrial fibrillation in older adults

    Circulation

    (1997)
  • F. Bode et al.

    Tarantula peptide inhibits atrial fibrillation

    Nature

    (2001)
  • F. Ravelli et al.

    Effects of atrial dilatation on refractory period and vulnerability to atrial fibrillation in the isolated Langendorff-perfused rabbit heart

    Circulation

    (1997)
  • F. Ravelli et al.

    Acute atrial dilatation slows conduction and increases AF vulnerability in the human atrium

    J. Cardiovas. Electrophysiol.

    (2011)
  • P. Tavi et al.

    Mechanisms of stretch-induced changes in [Ca2+]i in rat atrial myocytes: role of increased troponin C affinity and stretch-activated ion channels

    Circ. Res.

    (1998)
  • S.A. Nazir et al.

    Mechanoelectric feedback and atrial arrhythmias

    Cardiovasc. Res.

    (1996)
  • R. Kaufmann et al.

    Autonomously promoted extension effect in Purkinje fibers, papillary muscles and trabeculae carneae of rhesus monkeys

    Pflugers Archiv fur die gesamte Physiologie des Menschen und der Tiere.

    (1967)
  • M.J. Lab

    Depolarization produced by mechanical changes in normal and abnormal myocardium [proceedings]

    J. Physiol.

    (1978)
  • R. Peyronnet et al.

    Cardiac mechano-gated ion channels and arrhythmias

    Circ. Res.

    (2016)
  • Cited by (24)

    • Dexmedetomidine and Perioperative Arrhythmias

      2024, Journal of Cardiothoracic and Vascular Anesthesia
    • The role of mechanosensitive Piezo1 channel in diseases

      2022, Progress in Biophysics and Molecular Biology
      Citation Excerpt :

      Although Piezo1 is directly activated via stretch, the increased activity of BKCa during mechanical stress stimulation activated after Ca2+ influx. And during the process of Atrial Fibrillation (AF), Piezo1 is increased, while BKCa activity is opposed, indicating the different regulation of both channels (Jakob et al., 2021). Besides the expression in cardiac fibroblasts, Piezo1 upregulated in both human and mouse diseased hearts due to an autonomic response of cardiomyocytes.

    • Mechanosensitive Piezo1 channel in rat epididymal epithelial cells promotes transepithelial K<sup>+</sup> secretion

      2022, Cell Calcium
      Citation Excerpt :

      However, it is still unknown whether the Ca2+influx via Piezo1 channel occurs upstream of the activation of KCa channels. Recently, the presence and interplay of Piezo1 and BK channels in human atrial fibroblasts have been reported, although physical links between these two channel proteins were absent [37]. However, there has been no report of a structural or functional coupling between Piezo1 channel and IK channels or SK channels.

    • Research Progress on the Novel Mechanosensitive Ion Channel Piezo1 in Cardiac Fibrosis

      2024, Yiyong Shengwu Lixue/Journal of Medical Biomechanics
    View all citing articles on Scopus
    1

    These authors contributed equally to the work.

    View full text