Biallelic variants in POPDC2 cause a novel autosomal recessive syndrome presenting with cardiac conduction defects and variable hypertrophic cardiomyopathy.

Michele Nicastro,Alexa M C Vermeer,Pieter G Postema, Rafik Tadros,Forrest Z Bowling,Hildur M Aegisdottir,Vinicius Tragante,Lukas Mach,Alex V Postma,Elisabeth M Lodder,Karel van Duijvenboden,Rob Zwart,Leander Beekman, Lingshuang Wu, Paul A van der Zwaag,Mariëlle Alders, Mona Allouba,Yasmine Aguib, J Luis Santomel, David de Una,Lorenzo Monserrat,Antonio M A Miranda,Kazumasa Kanemaru, James Cranley, Ingeborg E van Zeggeren, Eleonora M A Aronica,Michela Ripolone,Simona Zanotti,Gardar Sveinbjornsson,Erna V Ivarsdottir,Hilma Hólm, Daníel F Guðbjartsson,Ástrós Th Skúladóttir,Kári Stefánsson,Lincoln Nadauld, Kirk U Knowlton,Sisse Rye Ostrowski, Erik Sørensen,Ole Birger Vesterager Pedersen, Jonas Ghouse, Søren Rand,Henning Bundgaard,Henrik Ullum, Christian Erikstrup, Bitten Aagaard,Mie Topholm Bruun,Mette Christiansen, Henrik K Jensen, Deanna Alexis Carere, Christopher T Cummings, Kristen Fishler,Pernille Mathiesen Tøring, Klaus Brusgaard, Trine Maxel Juul, Lotte Saaby,Bo Gregers Winkel, Jens Mogensen,Francesco Fortunato,Giacomo Pietro Comi, Dario Ronchi,J Peter van Tintelen,Michela Noseda,Michael V Airola,Imke Christiaans,Arthur A M Wilde,Ronald Wilders, Sally-Ann Clur, Arie O Verkerk,Connie R Bezzina,Najim Lahrouchi

medRxiv : the preprint server for health sciences(2024)

Cited 0|Views1
No score
Abstract
POPDC2 encodes for the Popeye domain-containing protein 2 which has an important role in cardiac pacemaking and conduction, due in part to its cAMP-dependent binding and regulation of TREK-1 potassium channels. Loss of Popdc2 in mice results in sinus pauses and bradycardia and morpholino knockdown of popdc2 in zebrafish results in atrioventricular (AV) block. We identified bi-allelic variants in POPDC2 in 4 families that presented with a phenotypic spectrum consisting of sinus node dysfunction, AV conduction defects and hypertrophic cardiomyopathy. Using homology modelling we show that the identified POPDC2 variants are predicted to diminish the ability of POPDC2 to bind cAMP. In in vitro electrophysiological studies we demonstrated that, while co-expression of wild-type POPDC2 with TREK-1 increased TREK-1 current density, POPDC2 variants found in the patients failed to increase TREK-1 current density. While patient muscle biopsy did not show clear myopathic disease, it showed significant reduction of the expression of both POPDC1 and POPDC2, suggesting that stability and/or membrane trafficking of the POPDC1-POPDC2 complex is impaired by pathogenic variants in any of the two proteins. Single-cell RNA sequencing from human hearts demonstrated that co-expression of POPDC1 and 2 was most prevalent in AV node, AV node pacemaker and AV bundle cells. Sinoatrial node cells expressed POPDC2 abundantly, but expression of POPDC1 was sparse. Together, these results concur with predisposition to AV node disease in humans with loss-of-function variants in POPDC1 and POPDC2 and presence of sinus node disease in POPDC2, but not in POPDC1 related disease in human. Using population-level genetic data of more than 1 million individuals we showed that none of the familial variants were associated with clinical outcomes in heterozygous state, suggesting that heterozygous family members are unlikely to develop clinical manifestations and therefore might not necessitate clinical follow-up. Our findings provide evidence for POPDC2 as the cause of a novel Mendelian autosomal recessive cardiac syndrome, consistent with previous work showing that mice and zebrafish deficient in functional POPDC2 display sinus and AV node dysfunction. GRAPHICAL ABSTRACT:
More
Translated text
AI Read Science
Must-Reading Tree
Example
Generate MRT to find the research sequence of this paper
Chat Paper
Summary is being generated by the instructions you defined