Original Article
POPDC2 a novel susceptibility gene for conduction disorders

https://doi.org/10.1016/j.yjmcc.2020.06.005Get rights and content

Abstract

Despite recent progress in the understanding of cardiac ion channel function and its role in inherited forms of ventricular arrhythmias, the molecular basis of cardiac conduction disorders often remains unresolved. We aimed to elucidate the genetic background of familial atrioventricular block (AVB) using a whole exome sequencing (WES) approach. In monozygotic twins with a third-degree AVB and in another, unrelated family with first-degree AVB, we identified a heterozygous nonsense mutation in the POPDC2 gene causing a premature stop at position 188 (POPDC2W188⁎), deleting parts of its cAMP binding-domain. Popeye-domain containing (POPDC) proteins are predominantly expressed in the skeletal muscle and the heart, with particularly high expression of POPDC2 in the sinoatrial node of the mouse. We now show by quantitative PCR experiments that in the human heart the POPDC-modulated two-pore domain potassium (K2P) channel TREK-1 is preferentially expressed in the atrioventricular node. Co-expression studies in Xenopus oocytes revealed that POPDC2W188⁎ causes a loss-of-function with impaired TREK-1 modulation. Consistent with the high expression level of POPDC2 in the murine sinoatrial node, POPDC2W188⁎ knock-in mice displayed stress-induced sinus bradycardia and pauses, a phenotype that was previously also reported for POPDC2 and TREK-1 knock-out mice. We propose that the POPDC2W188⁎ loss-of-function mutation contributes to AVB pathogenesis by an aberrant modulation of TREK-1, highlighting that POPDC2 represents a novel arrhythmia gene for cardiac conduction disorders.

Introduction

Disorders affecting the cardiac conduction system can induce serious and potentially life-threatening bradyarrhythmias. These dysfunctions can become evident by impaired cardiac conduction through the atrioventricular (AV) node, the His-bundle or the Purkinje system with right or left bundle branch block (RBBB or LBBB) and/or a broadening of the QRS complex. Hereditary forms of arrhythmias are predominantly caused by mutations in genes encoding ion channels or their modulating subunits. Mutations in the SCN5A gene encoding the cardiac sodium channel or its β-subunits are associated with progressive cardiac conduction disorder (PCCD) [[1], [2], [3]] and with congenital sick sinus syndrome (SSS) [1,4]. In addition, mutations in the TRPM4 channel gene cause autosomal-dominant hereditary forms of PCCD including AV conduction abnormalities [5]. Moreover, mutations in the LMNA gene may first present as AV conduction disorder and later as progressive heart failure and dilated cardiomyopathy [6]. Despite recent progress in the understanding of cardiac ion channel function and their role in inherited forms of ventricular arrhythmias, the genetic background of cardiac conduction disorders often remains unresolved.

The Popeye domain containing (POPDC) gene family encodes proteins with three transmembrane domains which are predominantly expressed in the heart and skeletal muscle [7,8]. The C-terminal parts of the proteins harboring the highly conserved Popeye-domain which contains a cAMP binding site are localized to the cytoplasm [9]. POPDC proteins are known to interact with the two-pore domain potassium (K2P) channel TREK-1 thereby enhancing its current amplitude [9]. We have previously reported that cAMP binds with high affinity to the Popeye domain, abolishing the current modulation of TREK-1 channels [9].

TREK-1 plays a major role in murine sinoatrial pacemaking as cardiac-specific ablation of TREK-1 in mice causes a bradycardia with episodes of sinus pauses following stress [10]. In the mouse heart, POPDC2 is expressed in atrial and ventricular myocytes [8], however a dominant expression in the sinoatrial and AV node and other parts of the cardiac conduction system was described [9]. Interestingly, in mice, POPDC1 and POPDC2 null mutants presented a comparable phenotype as the TREK-1 knock-out mice, with a strong stress-induced sinoatrial node dysfunction [9,10]. On the other hand, in POPDC1/POPDC2 double knock-out mice also an AVB was prevalent [11], compatible with observations from zebrafish in which morpholino oligonucleotide-mediated knock-down of POPDC2 led to AVB [12]. In humans we recently identified a POPDC1S201F mutation which causes AV block (AVB) and autosomal recessive limb-girdle muscular dystrophy (LGMDR25) [13]. Another three recessive mutations in POPDC1 are associated with AVB and LGMD of variable severity and age at onset [14], while mutations in POPDC3 are causing LGMD (LGMDR26) with no cardiac involvement [15].

To elucidate the genetic background of familial AVB we used a whole exome sequencing (WES) approach in a family with AVB and identified the heterozygous POPDC2 nonsense variant POPDC2W188⁎. The same POPDC2W188⁎ mutation was subsequently identified in another family with dominant inheritance. Mechanistically, our data suggest that the POPDC2W188⁎ mutation causes impaired AV conduction that is mediated by a dysregulation of TREK-1 potassium channels.

Section snippets

Study population: clinical and targeted DNA sequence analysis

In the present study we included patients with isolated atrioventricular block (AVB; n = 22), idiopathic sinus node dysfunction (SND; n = 32), right or left bundle branch block (RBBB, LBBB; n = 13), progressive cardiac conduction disease (PCCD; n = 5) and atrial fibrillation (AF; n = 10). We previously excluded mutations in SCN5A and TRPM4 in patients with PCCD, RBBB, LBBB and AVB and mutations in HCN4 in patients with SND. Stress electrocardiogram (ECG), Holter ECG and transthoracic

Whole exome sequencing (WES) identified a POPDC2 gene mutation in a twin pair with high degree atrioventricular block (AVB)

A 7-years old boy (proband 10031-1) presented with severe AVB (varying between 2nd to 3rd degree) (Fig. 1A and B) and due to symptoms (fainting and dizziness) he subsequently received a pacemaker. An identical clinical and symptomatic phenotype was seen for his monozygotic twin brother (10031–4) (Fig. 1A and B and Supplemental Fig. 1) who also received a pacemaker implant at the age of 10 years. Both twins had no evidence for a structural heart disease (as assessed by transthoracic

Discussion

The genetic basis for congenital disorders affecting pacemaking or AV conduction often remains elusive. To identify novel genes responsible for inherited forms of AVB we performed WES in a family with severe AVB and identified the POPDC2W188⁎ mutation. POPDC proteins were recently identified as novel cAMP binding proteins involved in cardiac pacemaking and POPDC1 or POPDC2 null mutations in the mouse [9], as well as POPDC2 morphants in zebrafish [12] suffered from severe cardiac arrhythmias.

Authors' contributions

B.O.-B. and S.R. performed TEVC measurements and data analyses; A.K.K. and B.O.-B. performed patch clamp experiments and data analyses with HL-1 cells; S.R., B.O.-B. and A.K.K. prepared or edited all figures; B.S. analyzed WES data; C.F. and S.D. performed QPCR experiments; R.F.R.S performed site-directed mutagenesis and cell transfections for Western blot experiments; U.H.-B. helped generating and breeding the transgenic knock-in mouse line; E.S.-B., S.Z., B.S., C.R. and E.A. enrolled subjects

Declaration of Competing Interest

L.Fa has received institutional research grants from European Union, British Heart Foundation, Medical Research Council (UK), DFG and Gilead. L.Fa is listed as inventor on two patents held by University of Birmingham (Atrial Fibrillation Therapy WO 2015140571, Markers for Atrial Fibrillation WO 2016012783).

Acknowledgements

This work was supported by grants of the Universitätsklinikum Giessen/Marburg (UKGM) and the Anneliese Pohl Habilitationsförderung to S.R.. The RARER-Regione Emilia Romagna and EU SOLVE-RD [N. 779257] projects to A.F. are gratefully acknowledged. The work was supported by the Medical Research Council [MR/J010383/1], the British Heart Foundation [PG/14/46/30911 and PG/19/13/34247] and the Magdi Yacoub Institute to T.B. and Grant Agreement No. 633196 [CATCH ME], British Heart Foundation [

References (33)

  • J.J. Schott et al.

    Cardiac conduction defects associate with mutations in SCN5A

    Nat. Genet.

    (1999)
  • H. Watanabe et al.

    Sodium channel beta1 subunit mutations associated with Brugada syndrome and cardiac conduction disease in humans

    J. Clin. Invest.

    (2008)
  • D.W. Benson et al.

    Congenital sick sinus syndrome caused by recessive mutations in the cardiac sodium channel gene (SCN5A)

    J. Clin. Invest.

    (2003)
  • M. Kruse et al.

    Impaired endocytosis of the ion channel TRPM4 is associated with human progressive familial heart block type I

    J. Clin. Invest.

    (2009)
  • A. Froese et al.

    Expression pattern of Popdc2 during mouse embryogenesis and in the adult

    Dev. Dyn.

    (2008)
  • A. Froese et al.

    Popeye domain containing proteins are essential for stress-mediated modulation of cardiac pacemaking in mice

    J. Clin. Invest.

    (2012)
  • Cited by (18)

    • Systemic AAV9.BVES delivery ameliorates muscular dystrophy in a mouse model of LGMDR25

      2023, Molecular Therapy
      Citation Excerpt :

      All POPDC proteins are highly expressed in striated muscles.9 Interestingly, patients carrying mutations in BVES often display a loss of membrane localization of POPDC2,2 a risk protein of cardiac arrhythmia.10 BVES has been implicated in various cellular processes, including protein trafficking,3 myoblast differentiation,11 cell migration,12 cell proliferation,13 and signaling cascades.14

    • Phosphodiesterase type 4 anchoring regulates cAMP signaling to Popeye domain-containing proteins

      2022, Journal of Molecular and Cellular Cardiology
      Citation Excerpt :

      Several protein-protein interaction partners of POPDC proteins have been identified including the tandem pore-domain background potassium channel TWIK-related K+ channel 1 (TREK-1). When TREK-1 is co-expressed with POPDC proteins in Xenopus oocytes, the channel displays enhanced membrane trafficking and conductivity, which is sensitive to changes in cAMP levels [13,17,24]. Effector proteins of the cAMP pathway form complexes with PDEs, which limits their activation in response to elevations in cAMP levels 1.

    View all citing articles on Scopus
    1

    Currently at the Institute for Human Genetics, University Hospital Münster, Münster, Germany.

    2

    Shared senior authors.

    View full text