4-Phenylbutyrate promoted wild-type γ-aminobutyric acid type A receptor trafficking, reduced endoplasmic reticulum stress, and mitigated seizures in Gabrg2+/Q390X mice associated with Dravet syndrome

Epilepsia. 2024 Jan;65(1):204-217. doi: 10.1111/epi.17779. Epub 2023 Nov 29.

Abstract

Objective: γ-Aminobutyric acid type A (GABAA ) receptor subunit gene mutations are major causes of various epilepsy syndromes, including severe kinds such as Dravet syndrome. Although the GABAA receptor is a major target for antiseizure medications, treating GABAA receptor mutations with receptor channel modulators is ineffective. Here, we determined the effect of a novel treatment with 4-phenylbutyrate (PBA) in Gabrg2+/Q390X knockin mice associated with Dravet syndrome.

Methods: We used biochemistry in conjunction with differential tagging of the wild-type and the mutant alleles, live brain slice surface biotinylation, microsome isolation, patch-clamp whole-cell recordings, and video-monitoring synchronized electroencephalographic (EEG) recordings in Gabrg2+/Q390X mice to determine the effect of PBA in vitro with recombinant GABAA receptors and in vivo with knockin mice.

Results: We found that PBA reduced the mutant γ2(Q390X) subunit protein aggregates, enhanced the wild-type GABAA receptor subunits' trafficking, and increased the membrane expression of the wild-type receptors. PBA increased the current amplitude of GABA-evoked current in human embryonic kidney 293T cells and the neurons bearing the γ2(Q390X) subunit protein. PBA also proved to reduce endoplasmic reticulum (ER) stress caused by the mutant γ2(Q390X) subunit protein, as well as mitigating seizures and EEG abnormalities in the Gabrg2+/Q390X mice.

Significance: This research has unveiled a promising and innovative approach for treating epilepsy linked to GABAA receptor mutations through an unconventional antiseizure mechanism. Rather than directly modulating the affected mutant channel, PBA facilitates the folding and transportation of wild-type receptor subunits to the cell membrane and synapse. Combining these findings with our previous study, which demonstrated PBA's efficacy in restoring GABA transporter 1 (encoded by SLC6A1) function, we propose that PBA holds significant potential for a wide range of genetic epilepsies. Its ability to target shared molecular pathways involving mutant protein ER retention and impaired protein membrane trafficking suggests broad application in treating such conditions.

Keywords: 4-phenylbutyrate; ER stress; GABAA receptors; Gabrg2+/Q390X knockin mice; epilepsy.

MeSH terms

  • Animals
  • Endoplasmic Reticulum Stress / genetics
  • Epilepsies, Myoclonic* / complications
  • Epilepsies, Myoclonic* / drug therapy
  • Epilepsies, Myoclonic* / genetics
  • Epilepsy* / genetics
  • Humans
  • Mice
  • Phenylbutyrates*
  • Receptors, GABA / metabolism
  • Receptors, GABA-A / genetics
  • Receptors, GABA-A / metabolism
  • Seizures / complications
  • gamma-Aminobutyric Acid

Substances

  • Receptors, GABA-A
  • Receptors, GABA
  • 4-phenylbutyric acid
  • gamma-Aminobutyric Acid
  • Gabrg2 protein, mouse
  • Phenylbutyrates