Abstract

Human genetic variants predicted to cause loss-of-function of protein-coding genes (pLoF variants) provide natural in vivo models of human gene inactivation and can be valuable indicators of gene function and the potential toxicity of therapeutic inhibitors targeting these genes1,2. Gain-of-kinase-function variants in LRRK2 are known to significantly increase the risk of Parkinson’s disease3,4, suggesting that inhibition of LRRK2 kinase activity is a promising therapeutic strategy. While preclinical studies in model organisms have raised some on-target toxicity concerns5–8, the biological consequences of LRRK2 inhibition have not been well characterized in humans. Here, we systematically analyze pLoF variants in LRRK2 observed across 141,456 individuals sequenced in the Genome Aggregation Database (gnomAD)9, 49,960 exome-sequenced individuals from the UK Biobank and over 4 million participants in the 23andMe genotyped dataset. After stringent variant curation, we identify 1,455 individuals with high-confidence pLoF variants in LRRK2. Experimental validation of three variants, combined with previous work10, confirmed reduced protein levels in 82.5% of our cohort. We show that heterozygous pLoF variants in LRRK2 reduce LRRK2 protein levels but that these are not strongly associated with any specific phenotype or disease state. Our results demonstrate the value of large-scale genomic databases and phenotyping of human loss-of-function carriers for target validation in drug discovery.

Highlights

  • New therapeutic strategies are desperately needed in Parkinson’s disease (PD), one of the most common age-related neurological diseases, which affects about 1% of people over the age of 60 years[11,12]

  • While model organisms are invaluable for understanding the function of leucine-rich repeat kinase 2 (LRRK2), they have important limitations, as exemplified by inconsistencies in phenotypic consequences of reduced LRRK2 activity seen among yeast, fruit flies, worms, mice, rats and nonhuman primates[25]

  • We found no significant difference between the age distribution of LRRK2 pLoF variant carriers and noncarriers in either the Genome Aggregation Database (gnomAD) or 23andMe datasets, suggesting no major impact on longevity, though we note that this analysis is based on age at sample collection, which is not equivalent to longevity and at current sample sizes we are only powered to detect a strong effect (Supplementary Table 6)

Read more

Summary

Introduction

New therapeutic strategies are desperately needed in Parkinson’s disease (PD), one of the most common age-related neurological diseases, which affects about 1% of people over the age of 60 years[11,12].

Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call