Abstract

ABSTRACTBi-allelic GBA1 mutations cause Gaucher's disease (GD), the most common lysosomal storage disorder. Neuronopathic manifestations in GD include neurodegeneration, which can be severe and rapidly progressive. GBA1 mutations are also the most frequent genetic risk factors for Parkinson's disease. Dysfunction of the autophagy-lysosomal pathway represents a key pathogenic event in GBA1-associated neurodegeneration. Using an induced pluripotent stem cell (iPSC) model of GD, we previously demonstrated that lysosomal alterations in GD neurons are linked to dysfunction of the transcription factor EB (TFEB). TFEB controls the coordinated expression of autophagy and lysosomal genes and is negatively regulated by the mammalian target of rapamycin complex 1 (mTORC1). To further investigate the mechanism of autophagy-lysosomal pathway dysfunction in neuronopathic GD, we examined mTORC1 kinase activity in GD iPSC neuronal progenitors and differentiated neurons. We found that mTORC1 is hyperactive in GD cells as evidenced by increased phosphorylation of its downstream protein substrates. We also found that pharmacological inhibition of glucosylceramide synthase enzyme reversed mTORC1 hyperactivation, suggesting that increased mTORC1 activity is mediated by the abnormal accumulation of glycosphingolipids in the mutant cells. Treatment with the mTOR inhibitor Torin1 upregulated lysosomal biogenesis and enhanced autophagic clearance in GD neurons, confirming that lysosomal dysfunction is mediated by mTOR hyperactivation. Further analysis demonstrated that increased TFEB phosphorylation by mTORC1 results in decreased TFEB stability in GD cells. Our study uncovers a new mechanism contributing to autophagy-lysosomal pathway dysfunction in GD, and identifies the mTOR complex as a potential therapeutic target for treatment of GBA1-associated neurodegeneration.

Highlights

  • Gaucher’s disease (GD) is the most common lysosomal storage disorder (LSD)

  • Increased mammalian target of rapamycin complex 1 (mTORC1) activity in neuronopathic GD induced pluripotent stem cell (iPSC) neuronal progenitor cells (NPCs) We have previously demonstrated lysosomal alterations in both NPCs and differentiated neurons derived from neuronopathic GD iPSC lines (Awad et al, 2015, 2017)

  • The neuronopathic GD iPSC lines were derived from two neuronopathic type 2 GD patients harboring the bi-allelic mutations L444P/RecNciI and W184R/ D409H, and from one neuronopathic type 3 GD patient with L444P/ L444P mutations

Read more

Summary

Introduction

Gaucher’s disease (GD) is the most common lysosomal storage disorder (LSD). It is caused by bi-allelic mutations in the GBA1 ( known as GBA) gene, which encodes the lysosomal enzyme β-glucocerebrosidase (GCase) (Jmoudiak and Futerman, 2005; Cox, 2010; Bendikov-Bar and Horowitz, 2012). Type 2 (acute neuronopathic) GD patients develop severe rapidly progressive neurodegeneration leading to death during early childhood. GBA1 mutations are the most frequent genetic risk factors for Parkinson’s disease (Brockmann and Berg, 2014; Klein and Westenberger, 2012), and decreased GCase enzyme activity is linked to α-synuclein accumulation and Parkinson’s disease pathogenesis (Almeida, 2012; Mazzulli et al, 2016; Schapira, 2015)

Methods
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.