Your browser doesn't support javascript.
loading
Molecular insights into sex-specific metabolic alterations in Alzheimer's mouse brain using multi-omics approach.
Strefeler, Abigail; Jan, Maxime; Quadroni, Manfredo; Teav, Tony; Rosenberg, Nadia; Chatton, Jean-Yves; Guex, Nicolas; Gallart-Ayala, Hector; Ivanisevic, Julijana.
Afiliação
  • Strefeler A; Metabolomics Unit, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland.
  • Jan M; Bioinformatics Competence Center, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland.
  • Quadroni M; Protein Analysis Facility, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland.
  • Teav T; Metabolomics Unit, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland.
  • Rosenberg N; Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.
  • Chatton JY; Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.
  • Guex N; Bioinformatics Competence Center, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland.
  • Gallart-Ayala H; Metabolomics Unit, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland. hector.gallartayala@unil.ch.
  • Ivanisevic J; Metabolomics Unit, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland. julijana.ivanisevic@unil.ch.
Alzheimers Res Ther ; 15(1): 8, 2023 01 09.
Article em En | MEDLINE | ID: mdl-36624525
ABSTRACT

BACKGROUND:

Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is characterized by altered cellular metabolism in the brain. Several of these alterations have been found to be exacerbated in females, known to be disproportionately affected by AD. We aimed to unravel metabolic alterations in AD at the metabolic pathway level and evaluate whether they are sex-specific through integrative metabolomic, lipidomic, and proteomic analysis of mouse brain tissue.

METHODS:

We analyzed male and female triple-transgenic mouse whole brain tissue by untargeted mass spectrometry-based methods to obtain a molecular signature consisting of polar metabolite, complex lipid, and protein data. These data were analyzed using multi-omics factor analysis. Pathway-level alterations were identified through joint pathway enrichment analysis or by separately evaluating lipid ontology and known proteins related to lipid metabolism.

RESULTS:

Our analysis revealed significant AD-associated and in part sex-specific alterations across the molecular signature. Sex-dependent alterations were identified in GABA synthesis, arginine biosynthesis, and in alanine, aspartate, and glutamate metabolism. AD-associated alterations involving lipids were also found in the fatty acid elongation pathway and lysophospholipid metabolism, with a significant sex-specific effect for the latter.

CONCLUSIONS:

Through multi-omics analysis, we report AD-associated and sex-specific metabolic alterations in the AD brain involving lysophospholipid and amino acid metabolism. These findings contribute to the characterization of the AD phenotype at the molecular level while considering the effect of sex, an overlooked yet determinant metabolic variable.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Doença de Alzheimer Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Doença de Alzheimer Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2023 Tipo de documento: Article