HDAC (Histone Deacetylase) Inhibitor Valproic Acid Attenuates Atrial Remodeling and Delays the Onset of Atrial Fibrillation in Mice

Circ Arrhythm Electrophysiol. 2019 Mar;12(3):e007071. doi: 10.1161/CIRCEP.118.007071.

Abstract

Background: A structural, electrical and metabolic atrial remodeling is central in the development of atrial fibrillation (AF) contributing to its initiation and perpetuation. In the heart, HDACs (histone deacetylases) control remodeling associated processes like hypertrophy, fibrosis, and energy metabolism. Here, we analyzed, whether the HDAC class I/IIa inhibitor valproic acid (VPA) is able to attenuate atrial remodeling in CREM-IbΔC-X (cAMP responsive element modulator isoform IbΔC-X) transgenic mice, a mouse model of extensive atrial remodeling with age-dependent progression from spontaneous atrial ectopy to paroxysmal and finally long-lasting AF.

Methods: VPA was administered for 7 or 25 weeks to transgenic and control mice. Atria were analyzed macroscopically and using widefield and electron microscopy. Action potentials were recorded from atrial cardiomyocytes using patch-clamp technique. ECG recordings documented the onset of AF. A proteome analysis with consecutive pathway mapping identified VPA-mediated proteomic changes and related pathways.

Results: VPA attenuated many components of atrial remodeling that are present in transgenic mice, animal AF models, and human AF. VPA significantly ( P<0.05) reduced atrial dilatation, cardiomyocyte enlargement, atrial fibrosis, and the disorganization of myocyte's ultrastructure. It significantly reduced the occurrence of atrial thrombi, reversed action potential alterations, and finally delayed the onset of AF by 4 to 8 weeks. Increased histone H4-acetylation in atria from VPA-treated transgenic mice verified effective in vivo HDAC inhibition. Cardiomyocyte-specific genetic inactivation of HDAC2 in transgenic mice attenuated the ultrastructural disorganization of myocytes comparable to VPA. Finally, VPA restrained dysregulation of proteins in transgenic mice that are involved in a multitude of AF relevant pathways like oxidative phosphorylation or RhoA (Ras homolog gene family, member A) signaling and disease functions like cardiac fibrosis and apoptosis of muscle cells.

Conclusions: Our results suggest that VPA, clinically available, well-tolerated, and prescribed to many patients for years, has the therapeutic potential to delay the development of atrial remodeling and the onset of AF in patients at risk.

Keywords: atrial fibrillation; atrial remodeling; histone deacetylase inhibitor; proteomics; valproic acid.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Action Potentials
  • Animals
  • Anti-Arrhythmia Agents / pharmacology*
  • Atrial Fibrillation / enzymology
  • Atrial Fibrillation / pathology
  • Atrial Fibrillation / physiopathology
  • Atrial Fibrillation / prevention & control*
  • Atrial Remodeling / drug effects*
  • Cyclic AMP Response Element Modulator / genetics
  • Disease Models, Animal
  • Heart Atria / drug effects*
  • Heart Atria / enzymology
  • Heart Atria / physiopathology
  • Heart Atria / ultrastructure
  • Heart Rate
  • Histone Deacetylase Inhibitors / pharmacology*
  • Male
  • Mice, Transgenic
  • Myocytes, Cardiac / drug effects*
  • Myocytes, Cardiac / enzymology
  • Myocytes, Cardiac / ultrastructure
  • Time Factors
  • Valproic Acid / pharmacology*

Substances

  • Anti-Arrhythmia Agents
  • Histone Deacetylase Inhibitors
  • Cyclic AMP Response Element Modulator
  • Valproic Acid