Back to Search Start Over

Functional characterisation of the amyotrophic lateral sclerosis risk locus GPX3/TNIP1.

Authors :
Restuadi R
Steyn FJ
Kabashi E
Ngo ST
Cheng FF
Nabais MF
Thompson MJ
Qi T
Wu Y
Henders AK
Wallace L
Bye CR
Turner BJ
Ziser L
Mathers S
McCombe PA
Needham M
Schultz D
Kiernan MC
van Rheenen W
van den Berg LH
Veldink JH
Ophoff R
Gusev A
Zaitlen N
McRae AF
Henderson RD
Wray NR
Giacomotto J
Garton FC
Source :
Genome medicine [Genome Med] 2022 Jan 19; Vol. 14 (1), pp. 7. Date of Electronic Publication: 2022 Jan 19.
Publication Year :
2022

Abstract

Background: Amyotrophic lateral sclerosis (ALS) is a complex, late-onset, neurodegenerative disease with a genetic contribution to disease liability. Genome-wide association studies (GWAS) have identified ten risk loci to date, including the TNIP1/GPX3 locus on chromosome five. Given association analysis data alone cannot determine the most plausible risk gene for this locus, we undertook a comprehensive suite of in silico, in vivo and in vitro studies to address this.<br />Methods: The Functional Mapping and Annotation (FUMA) pipeline and five tools (conditional and joint analysis (GCTA-COJO), Stratified Linkage Disequilibrium Score Regression (S-LDSC), Polygenic Priority Scoring (PoPS), Summary-based Mendelian Randomisation (SMR-HEIDI) and transcriptome-wide association study (TWAS) analyses) were used to perform bioinformatic integration of GWAS data (N <subscript>cases</subscript> = 20,806, N <subscript>controls</subscript> = 59,804) with 'omics reference datasets including the blood (eQTLgen consortium N = 31,684) and brain (N = 2581). This was followed up by specific expression studies in ALS case-control cohorts (microarray N <subscript>total</subscript> = 942, protein N <subscript>total</subscript> = 300) and gene knockdown (KD) studies of human neuronal iPSC cells and zebrafish-morpholinos (MO).<br />Results: SMR analyses implicated both TNIP1 and GPX3 (p < 1.15 × 10 <superscript>-6</superscript> ), but there was no simple SNP/expression relationship. Integrating multiple datasets using PoPS supported GPX3 but not TNIP1. In vivo expression analyses from blood in ALS cases identified that lower GPX3 expression correlated with a more progressed disease (ALS functional rating score, p = 5.5 × 10 <superscript>-3</superscript> , adjusted R <superscript>2</superscript> = 0.042, B <subscript>effect</subscript> = 27.4 ± 13.3 ng/ml/ALSFRS unit) with microarray and protein data suggesting lower expression with risk allele (recessive model p = 0.06, p = 0.02 respectively). Validation in vivo indicated gpx3 KD caused significant motor deficits in zebrafish-MO (mean difference vs. control ± 95% CI, vs. control, swim distance = 112 ± 28 mm, time = 1.29 ± 0.59 s, speed = 32.0 ± 2.53 mm/s, respectively, p for all < 0.0001), which were rescued with gpx3 expression, with no phenotype identified with tnip1 KD or gpx3 overexpression.<br />Conclusions: These results support GPX3 as a lead ALS risk gene in this locus, with more data needed to confirm/reject a role for TNIP1. This has implications for understanding disease mechanisms (GPX3 acts in the same pathway as SOD1, a well-established ALS-associated gene) and identifying new therapeutic approaches. Few previous examples of in-depth investigations of risk loci in ALS exist and a similar approach could be applied to investigate future expected GWAS findings.<br /> (© 2021. The Author(s).)

Details

Language :
English
ISSN :
1756-994X
Volume :
14
Issue :
1
Database :
MEDLINE
Journal :
Genome medicine
Publication Type :
Academic Journal
Accession number :
35042540
Full Text :
https://doi.org/10.1186/s13073-021-01006-6