Abstract

The G2019S mutation in LRRK2 is one of the most common known genetic causes of neurodegeneration and Parkinson disease (PD). LRRK2 mutations are thought to enhance LRRK2 kinase activity. Efficacious small molecule LRRK2 kinase inhibitors with favorable drug properties have recently been developed for pre-clinical studies in rodent models, and inhibitors have advanced to safety trials in humans. Rats that express human G2019S-LRRK2 protein and G2019S-LRRK2 knock-in mice provide newly characterized models to better understand the ostensible target for inhibitors. Herein, we explore the relationships between LRRK2 kinase inhibition in the brain and the periphery to establish the link between LRRK2 kinase activity and protein stability, induction of lysosomal defects in kidney and lung, and how G2019S-LRRK2 expression impacts these phenotypes. Using a novel ultra-sensitive scalable assay based on protein capillary electrophoresis with LRRK2 kinase inhibitors included in-diet, G2019S-LRRK2 protein was resilient to inhibition compared to wild-type (WT)-LRRK2 protein, particularly in the brain. Whereas WT-LRRK2 kinase activity could be completed blocked without lowering LRRK2 protein levels, higher inhibitor concentrations were necessary to fully reduce G2019S-LRRK2 activity. G2019S-LRRK2 expression afforded robust protection from inhibitor-induced kidney lysosomal defects, suggesting a gain-of-function for the mutation in this phenotype. In rodents treated with inhibitors, parallel measurements of phospho-Rab10 revealed a poor correlation to phospho-LRRK2, likely due to cells that express Rab10 but poorly express LRRK2 in heterogenous tissues and cell isolates. In summary, our results highlight several challenges associated with the inhibition of the G2019S-LRRK2 kinase that might be considered in initial clinical efforts.

Highlights

  • The leucine-rich repeat kinase 2 gene encodes LRRK2 protein that is expressed primarily in circulating leukocytes, kidney, lung, and the brain in humans (West, 2017)

  • To study the characteristics of G2019S-LRRK2 versus WT-LRRK2 inhibition, two structurally distinct LRRK2 kinase inhibitors would produce more reliable conclusions than those surmised from a single small molecule

  • Whereas MLi2 blocks WT and G2019S-LRRK2 kinase activity with similar potencies, PF-360 is considerably weaker in blocking G2019S-LRRK2 autophosphorylation (~6-fold less potent) as well as G2019S-LRRK2 mediated LRRKtide phosphorylation (~4-fold less potent)

Read more

Summary

Introduction

The leucine-rich repeat kinase 2 gene encodes LRRK2 protein that is expressed primarily in circulating leukocytes, kidney, lung, and the brain in humans (West, 2017). Initial in vitro studies in transfected cell lines revealed that G2019S-LRRK2 increased autophosphorylation activities as well as LRRK2 kinase activity towards generic peptide substrates, usually ~2–5 fold over endogenous wild-type (WT)-LRRK2. Analyses of LRRK2 protein harbored in extracellular exosomes purified from urine from LRRK2 mutation carriers with Parkinson’s disease (PD) suggests a similar effect on LRRK2 autophosphorylation (Fraser et al, 2016a; Wang et al, 2017). Intensive efforts are devoted towards the development of LRRK2 kinase inhibitors for the treatment of LRRK2-linked PD (West, 2017). Safety trials are underway with several LRRK2 kinase inhibitors of as-yet unknown identity (Hyland and Warners, 2017)

Methods
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