Your browser doesn't support javascript.
loading
Peroxisome proliferator-activated receptor α as a novel therapeutic target for schizophrenia.
Wada, Yuina; Maekawa, Motoko; Ohnishi, Tetsuo; Balan, Shabeesh; Matsuoka, Shigeru; Iwamoto, Kazuya; Iwayama, Yoshimi; Ohba, Hisako; Watanabe, Akiko; Hisano, Yasuko; Nozaki, Yayoi; Toyota, Tomoko; Shimogori, Tomomi; Itokawa, Masanari; Kobayashi, Tetsuyuki; Yoshikawa, Takeo.
Affiliation
  • Wada Y; Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan; Department of Biological Science, Graduate School of Humanities and Science, Ochanomizu University, Tokyo 112-8610, Japan; Laboratory for Molecular Psychiatry, RIKEN Center for Brai
  • Maekawa M; Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan; Department of Biological Science, Graduate School of Humanities and Science, Ochanomizu University, Tokyo 112-8610, Japan. Electronic address: motoko.maekawa@riken.jp.
  • Ohnishi T; Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan; Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan.
  • Balan S; Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan; Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan.
  • Matsuoka S; Faculty of Medicine, Oita University, Oita 879-5593, Japan.
  • Iwamoto K; Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan.
  • Iwayama Y; Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan.
  • Ohba H; Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan.
  • Watanabe A; Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan.
  • Hisano Y; Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan.
  • Nozaki Y; Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan.
  • Toyota T; Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan.
  • Shimogori T; Laboratory for Molecular Mechanisms of Brain Development, RIKEN Center for Brain Science, Saitama 351-0198, Japan.
  • Itokawa M; Center for Medical Cooperation, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan.
  • Kobayashi T; Department of Biological Science, Graduate School of Humanities and Science, Ochanomizu University, Tokyo 112-8610, Japan.
  • Yoshikawa T; Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan; Department of Biological Science, Graduate School of Humanities and Science, Ochanomizu University, Tokyo 112-8610, Japan. Electronic address: takeo.yoshikawa@riken.jp.
EBioMedicine ; 62: 103130, 2020 Dec.
Article de En | MEDLINE | ID: mdl-33279456
ABSTRACT

BACKGROUND:

The pathophysiology of schizophrenia, a major psychiatric disorder, remains elusive. In this study, the role of peroxisome proliferator-activated receptor (PPAR)/retinoid X receptor (RXR) families, belonging to the ligand-activated nuclear receptor superfamily, in schizophrenia, was analyzed.

METHODS:

The PPAR/RXR family genes were screened by exploiting molecular inversion probe (MIP)-based targeted next-generation sequencing (NGS) using the samples of 1,200 Japanese patients with schizophrenia. The results were compared with the whole-genome sequencing databases of the Japanese cohort (ToMMo) and the gnomAD. To reveal the relationship between PPAR/RXR dysfunction and schizophrenia, Ppara KO mice and fenofibrate (a clinically used PPARα agonist)-administered mice were assessed by performing behavioral, histological, and RNA-seq analyses.

FINDINGS:

Our findings indicate that c.209-2delA, His117Gln, Arg141Cys, and Arg226Trp of the PPARA gene are risk variants for schizophrenia. The c.209-2delA variant generated a premature termination codon. The three missense variants significantly decreased the activity of PPARα as a transcription factor in vitro. The Ppara KO mice exhibited schizophrenia-relevant phenotypes, including behavioral deficits and impaired synaptogenesis in the cerebral cortex. Oral administration of fenofibrate alleviated spine pathology induced by phencyclidine, an N-methyl-d-aspartate (NMDA) receptor antagonist. Furthermore, pre-treatment with fenofibrate suppressed the sensitivity of mice to another NMDA receptor antagonist, MK-801. RNA-seq analysis revealed that PPARα regulates the expression of synaptogenesis signaling pathway-related genes.

INTERPRETATION:

The findings of this study indicate that the mechanisms underlying schizophrenia pathogenesis involve PPARα-regulated transcriptional machinery and modulation of synapse physiology. Hence, PPARα can serve as a novel therapeutic target for schizophrenia.
Sujet(s)
Mots clés

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Schizophrénie / Marqueurs biologiques / Récepteur PPAR alpha Type d'étude: Etiology_studies / Prognostic_studies Limites: Adult / Aged / Animals / Female / Humans / Male / Middle aged Langue: En Journal: EBioMedicine Année: 2020 Type de document: Article

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Schizophrénie / Marqueurs biologiques / Récepteur PPAR alpha Type d'étude: Etiology_studies / Prognostic_studies Limites: Adult / Aged / Animals / Female / Humans / Male / Middle aged Langue: En Journal: EBioMedicine Année: 2020 Type de document: Article