Please use this identifier to cite or link to this item: http://hdl.handle.net/2445/123830
Title: Rare variants in calcium homeostasis modulator 1 (CALHM1) found in early onset Alzheimer's disease patients alter calcium homeostasis.
Author: Rubio Moscardo, Fanny
Setó Salvia, Núria
Pera, Marta
Bosch Morató, Mònica
Plata, Cristina
Belbin, Olivia
Gené, Gemma
Dols Icardo, Oriol
Ingelsson, Martin
Helisalmi, Seppo
Soininen, Hilkka
Hiltunen, Mikko
Giedraitis, Vilmantas
Lannfelt, Lars
Frank García, Ana
Bullido, María Jesús
Combarros, Onofre
Sánchez Juan, Pascual
Boada, Mercè
Tárraga, Lluís
Pastor, Pau
Pérez Tur, Jordi
Baquero, Miquel
Molinuevo, José Luis
Sánchez del Valle Díaz, Raquel
Fuentes Prior, Pablo
Fortea Ormaechea, Juan
Blesa González, Rafael
Muñoz, Francisco J.
Lleó Bisa, Alberto
Valverde, Miguel Ángel
Clarimón, Jordi
Keywords: Malaltia d'Alzheimer
Homeòstasi
Calci
Alzheimer's disease
Homeostasis
Calcium
Issue Date: 17-Sep-2013
Publisher: Public Library of Science (PLoS)
Abstract: Calcium signaling in the brain is fundamental to the learning and memory process and there is evidence to suggest that its dysfunction is involved in the pathological pathways underlying Alzheimer's disease (AD). Recently, the calcium hypothesis of AD has received support with the identification of the non-selective Ca2+-permeable channel CALHM1. A genetic polymorphism (p. P86L) in CALHM1 reduces plasma membrane Ca2+ permeability and is associated with an earlier age-at-onset of AD. To investigate the role of CALHM1 variants in early-onset AD (EOAD), we sequenced all CALHM1 coding regions in three independent series comprising 284 EOAD patients and 326 controls. Two missense mutations in patients (p.G330D and p.R154H) and one (p.A213T) in a control individual were identified. Calcium imaging analyses revealed that while the mutation found in a control (p.A213T) behaved as wild-type CALHM1 (CALHM1-WT), a complete abolishment of the Ca2+ influx was associated with the mutations found in EOAD patients (p.G330D and p.R154H). Notably, the previously reported p. P86L mutation was associated with an intermediate Ca2+ influx between the CALHM1-WT and the p.G330D and p.R154H mutations. Since neither expression of wild-type nor mutant CALHM1 affected amyloid ß-peptide (Aß) production or Aß-mediated cellular toxicity, we conclude that rare genetic variants in CALHM1 lead to Ca2+ dysregulation and may contribute to the risk of EOAD through a mechanism independent from the classical Aß cascade
Note: Reproducció del document publicat a: https://doi.org/10.1371/journal.pone.0074203
It is part of: PLoS One, 2013, vol. 8, num. 9, p. 1-8
URI: http://hdl.handle.net/2445/123830
Related resource: https://doi.org/10.1371/journal.pone.0074203
ISSN: 1932-6203
Appears in Collections:Articles publicats en revistes (Medicina)
Articles publicats en revistes (IDIBAPS: Institut d'investigacions Biomèdiques August Pi i Sunyer)

Files in This Item:
File Description SizeFormat 
677582.pdf846.25 kBAdobe PDFView/Open


This item is licensed under a Creative Commons License Creative Commons