Download PDF Download PDF

Molecular Psychiatry

Publication date: 2022-06-01
Volume: 27 Pages: 2821 - 2832
Publisher: Springer Nature [academic journals on nature.com]

Author:

Petit, Dieter
Fernandez, Sara Gutierrez ; Zoltowska, Katarzyna Marta ; Enzlein, Thomas ; Ryan, Natalie S ; O'Connor, Antoinette ; Szaruga, Maria ; Hill, Elizabeth ; Vandenberghe, Rik ; Fox, Nick C ; Chavez-Gutierrez, Lucia

Keywords:

Science & Technology, Life Sciences & Biomedicine, Biochemistry & Molecular Biology, Neurosciences, Psychiatry, Neurosciences & Neurology, GAMMA-SECRETASE CLEAVAGE, OF-ONSET, PRESENILIN-1 MUTATIONS, APP MUTATIONS, IN-VITRO, PROTEIN, A-BETA-42, MECHANISM, DEFICIENCY, HYPOTHESIS, Humans, Alzheimer Disease, Amyloid beta-Peptides, Amyloid beta-Protein Precursor, Amyloid Precursor Protein Secretases, Mutation, Presenilin-1, G0B2519N#54970002, 1S59621N|1S59623N#55997033, 06 Biological Sciences, 11 Medical and Health Sciences, 17 Psychology and Cognitive Sciences, 3202 Clinical sciences, 5202 Biological psychology, 5203 Clinical and health psychology

Abstract:

Familial Alzheimer's disease (FAD), caused by mutations in Presenilin (PSEN1/2) and Amyloid Precursor Protein (APP) genes, is associated with an early age at onset (AAO) of symptoms. AAO is relatively consistent within families and between carriers of the same mutations, but differs markedly between individuals carrying different mutations. Gaining a mechanistic understanding of why certain mutations manifest several decades earlier than others is extremely important in elucidating the foundations of pathogenesis and AAO. Pathogenic mutations affect the protease (PSEN/γ-secretase) and the substrate (APP) that generate amyloid β (Aβ) peptides. Altered Aβ metabolism has long been associated with AD pathogenesis, with absolute or relative increases in Aβ42 levels most commonly implicated in the disease development. However, analyses addressing the relationships between these Aβ42 increments and AAO are inconsistent. Here, we investigated this central aspect of AD pathophysiology via comprehensive analysis of 25 FAD-linked Aβ profiles. Hypothesis- and data-driven approaches demonstrate linear correlations between mutation-driven alterations in Aβ profiles and AAO. In addition, our studies show that the Aβ (37 + 38 + 40) / (42 + 43) ratio offers predictive value in the assessment of 'unclear' PSEN1 variants. Of note, the analysis of PSEN1 variants presenting additionally with spastic paraparesis, indicates that a different mechanism underlies the aetiology of this distinct clinical phenotype. This study thus delivers valuable assays for fundamental, clinical and genetic research as well as supports therapeutic interventions aimed at shifting Aβ profiles towards shorter Aβ peptides.