Please use this identifier to cite or link to this item: http://hdl.handle.net/2445/140358
Title: The human brainome: network analysis identifies HSPA2 as a novel Alzheimer's disease target
Author: Petyuk, Vladislav A.
Chang, Rui
Ramirez-Restrepo, Manuel
Beckmann, Noam D.
Henrion, Marc Y.R.
Piehowski, Paul D.
Zhu, Kuixi
Wang, Sven
Clarke, Jennifer
Huentelman, Matthew J.
Xie, Fang
Andreev, Victor
Engel, Anzhelika
Guettoche, Toumy
Navarro, Loida
De Jager, Philip
Schneider, Julie A.
Morris, Christopher M.
McKeith, Ian G.
Perry, Robert H.
Lovestone, Simon
Woltjer, Randall L.
Beach, Thomas G.
Sue, Lucia I.
Serrano, Geidy E.
Lieberman, Andrew P.
Albin, Roger L.
Ferrer, Isidro (Ferrer Abizanda)
Mash, Deborah C.
Hulette, Christine M.
Ervin, John F.
Reiman, Eric M.
Hardy, John A.
Bennett, David A.
Schadt, Eric
Smith, Richard D.
Myers, Amanda J.
Keywords: Malaltia d'Alzheimer
Genètica
Proteïnes
Fisiologia
Alzheimer's disease
Genetics
Proteins
Physiology
Issue Date: 1-Sep-2018
Publisher: Oxford University Press
Abstract: Our hypothesis is that changes in gene and protein expression are crucial to the development of late-onset Alzheimer's disease. Previously we examined how DNA alleles control downstream expression of RNA transcripts and how those relationships are changed in late-onset Alzheimer's disease. We have now examined how proteins are incorporated into networks in two separate series and evaluated our outputs in two different cell lines. Our pipeline included the following steps: (i) predicting expression quantitative trait loci; (ii) determining differential expression; (iii) analysing networks of transcript and peptide relationships; and (iv) validating effects in two separate cell lines. We performed all our analysis in two separate brain series to validate effects. Our two series included 345 samples in the first set (177 controls, 168 cases; age range 65-105; 58% female; KRONOSII cohort) and 409 samples in the replicate set (153 controls, 141 cases, 115 mild cognitive impairment; age range 66-107; 63% female; RUSH cohort). Our top target is heat shock protein family A member 2 (HSPA2), which was identified as a key driver in our two datasets. HSPA2 was validated in two cell lines, with overexpression driving further elevation of amyloid-b40 and amyloid-b42 levels in APP mutant cells, as well as significant elevation of microtubule associated protein tau and phosphorylated-tau in a modified neuroglioma line. This work further demonstrates that studying changes in gene and protein expression is crucial to understanding late onset disease and further nominates HSPA2 as a specific key regulator of late-onset Alzheimer's disease processes.
Note: Versió postprint del document publicat a: https://doi.org/10.1093/brain/awy215
It is part of: Brain, 2018, vol. 141, num. 9, p. 2721-2739
URI: http://hdl.handle.net/2445/140358
Related resource: https://doi.org/10.1093/brain/awy215
ISSN: 0006-8950
Appears in Collections:Articles publicats en revistes (Patologia i Terapèutica Experimental)

Files in This Item:
File Description SizeFormat 
689458.pdf1.55 MBAdobe PDFView/Open


Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.