ALDH2 deficiency induces atrial fibrillation through dysregulated cardiac sodium channel and mitochondrial bioenergetics: A multi-omics analysis

Point mutation in alcohol dehydrogenase 2 (ALDH2), ALDH2*2 results in decreased catalytic enzyme activity and has been found to be associated with different human pathologies. Whether ALDH2*2 would induce cardiac remodeling and increase the attack of atrial fibrillation (AF) remains poorly understoo...

Full description

Saved in:
Bibliographic Details
Published inBiochimica et biophysica acta. Molecular basis of disease Vol. 1867; no. 5; p. 166088
Main Authors Hu, Yu-Feng, Wu, Chih-Hsun, Lai, Tsung-Ching, Chang, Yu-Chan, Hwang, Ming-Jing, Chang, Ting-Yung, Weng, Ching-Hui, Chang, Peter Mu-Hsin, Chen, Che-Hong, Mochly-Rosen, Daria, Huang, Chi-Ying F., Chen, Shih-Ann
Format Journal Article
LanguageEnglish
Published Netherlands Elsevier B.V 01.05.2021
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Point mutation in alcohol dehydrogenase 2 (ALDH2), ALDH2*2 results in decreased catalytic enzyme activity and has been found to be associated with different human pathologies. Whether ALDH2*2 would induce cardiac remodeling and increase the attack of atrial fibrillation (AF) remains poorly understood. The present study evaluated the effect of ALDH2*2 mutation on AF susceptibility and unravelled the underlying mechanisms using a multi-omics approach including whole-genome gene expression and proteomics analysis. The in-vivo electrophysiological study showed an increase in the incidence and reduction in the threshold of AF for the mutant mice heterozygous for ALDH2*2 as compared to the wild type littermates. The microarray analysis revealed a reduction in the retinoic acid signals which was accompanied by a downstream reduction in the expression of voltage-gated Na+ channels (SCN5A). The treatment of an antagonist for retinoic acid receptor resulted in a decrease in SCN5A transcript levels. The integrated analysis of the transcriptome and proteome data showed a dysregulation of fatty acid β-oxidation, adenosine triphosphate synthesis via electron transport chain, and activated oxidative responses in the mitochondria. Oral administration of Coenzyme Q10, an essential co-factor known to meliorate mitochondrial oxidative stress and preserve bioenergetics, conferred a protection against AF attack in the mutant ALDH2*2 mice. The multi-omics approach showed the unique pathophysiology mechanisms of concurrent dysregulated SCN5A channel and mitochondrial bioenergetics in AF. This inspired the development of a personalized therapeutic agent, Coenzyme Q10, to protect against AF attack in humans characterized by ALDH2*2 genotype. [Display omitted] •The ALDH2*2 increased atrial fibrillation (AF) susceptibility in the mice model.•Concurrent dysregulated SCN5A channel and mitochondrial bioenergetics led to AF.•Coenzyme Q10 protected against AF attack in humans characterized by ALDH2*2 genotype.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
Y.F.H. and C.H.W. contributed equally to this work.
The present study was conceived and designed by Y. F. Hu and C. H. Wu. The multi-omics and network analysis were performed by C. H. Wu. The microarray experiments were conducted by T. C. Lai and Y. C. Chang. P. M. H. Chang performed the proteomics experiments. In vivo mice studies and molecular biology assays were conducted by T. Y. Chang and C. H. Weng. C. H. Chen and D. Mochly-Rosen provided the mouse model for the study and contributed in the discussion and preparation of the manuscript. The interpretation of the data and manuscript writing was carried out by Y. F. Hu and C. H. Wu. The manuscript was revised by C. H. Chen and C. Y. F. Huang. The study was performed under the supervision of Y. F. Hu, M. J. Hwang, and S. A. Chen.
Author contributions
ISSN:0925-4439
1879-260X
DOI:10.1016/j.bbadis.2021.166088