α-Synuclein accumulation and GBA deficiency due to L444P GBA mutation contributes to MPTP-induced parkinsonism

Abstract Background Mutations in glucocerebrosidase (GBA) cause Gaucher disease (GD) and increase the risk of developing Parkinson’s disease (PD) and Dementia with Lewy Bodies (DLB). Since both genetic and environmental factors contribute to the pathogenesis of sporadic PD, we investigated the susce...

Full description

Bibliographic Details
Main Authors: Seung Pil Yun, Donghoon Kim, Sangjune Kim, SangMin Kim, Senthilkumar S. Karuppagounder, Seung-Hwan Kwon, Saebom Lee, Tae-In Kam, Suhyun Lee, Sangwoo Ham, Jae Hong Park, Valina L. Dawson, Ted M. Dawson, Yunjong Lee, Han Seok Ko
Format: Article
Language:English
Published: BMC 2018-01-01
Series:Molecular Neurodegeneration
Subjects:
GBA
Online Access:http://link.springer.com/article/10.1186/s13024-017-0233-5
id doaj-213271272ea14b04817b478ead2a71ae
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Seung Pil Yun
Donghoon Kim
Sangjune Kim
SangMin Kim
Senthilkumar S. Karuppagounder
Seung-Hwan Kwon
Saebom Lee
Tae-In Kam
Suhyun Lee
Sangwoo Ham
Jae Hong Park
Valina L. Dawson
Ted M. Dawson
Yunjong Lee
Han Seok Ko
spellingShingle Seung Pil Yun
Donghoon Kim
Sangjune Kim
SangMin Kim
Senthilkumar S. Karuppagounder
Seung-Hwan Kwon
Saebom Lee
Tae-In Kam
Suhyun Lee
Sangwoo Ham
Jae Hong Park
Valina L. Dawson
Ted M. Dawson
Yunjong Lee
Han Seok Ko
α-Synuclein accumulation and GBA deficiency due to L444P GBA mutation contributes to MPTP-induced parkinsonism
Molecular Neurodegeneration
Parkinson’s disease
GBA
MPTP
Mitochondrial dysfunction
α-synuclein
author_facet Seung Pil Yun
Donghoon Kim
Sangjune Kim
SangMin Kim
Senthilkumar S. Karuppagounder
Seung-Hwan Kwon
Saebom Lee
Tae-In Kam
Suhyun Lee
Sangwoo Ham
Jae Hong Park
Valina L. Dawson
Ted M. Dawson
Yunjong Lee
Han Seok Ko
author_sort Seung Pil Yun
title α-Synuclein accumulation and GBA deficiency due to L444P GBA mutation contributes to MPTP-induced parkinsonism
title_short α-Synuclein accumulation and GBA deficiency due to L444P GBA mutation contributes to MPTP-induced parkinsonism
title_full α-Synuclein accumulation and GBA deficiency due to L444P GBA mutation contributes to MPTP-induced parkinsonism
title_fullStr α-Synuclein accumulation and GBA deficiency due to L444P GBA mutation contributes to MPTP-induced parkinsonism
title_full_unstemmed α-Synuclein accumulation and GBA deficiency due to L444P GBA mutation contributes to MPTP-induced parkinsonism
title_sort α-synuclein accumulation and gba deficiency due to l444p gba mutation contributes to mptp-induced parkinsonism
publisher BMC
series Molecular Neurodegeneration
issn 1750-1326
publishDate 2018-01-01
description Abstract Background Mutations in glucocerebrosidase (GBA) cause Gaucher disease (GD) and increase the risk of developing Parkinson’s disease (PD) and Dementia with Lewy Bodies (DLB). Since both genetic and environmental factors contribute to the pathogenesis of sporadic PD, we investigated the susceptibility of nigrostriatal dopamine (DA) neurons in L444P GBA heterozygous knock-in (GBA +/L444P ) mice to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a selective dopaminergic mitochondrial neurotoxin. Method We used GBA +/L444P mice, α-synuclein knockout (SNCA −/− ) mice at 8 months of age, and adeno-associated virus (AAV)-human GBA overexpression to investigate the rescue effect of DA neuronal loss and susceptibility by MPTP. Mitochondrial morphology and functional assay were used to identify mitochondrial defects in GBA +/L444P mice. Motor behavioral test, immunohistochemistry, and HPLC were performed to measure dopaminergic degeneration by MPTP and investigate the relationship between GBA mutation and α-synuclein. Mitochondrial immunostaining, qPCR, and Western blot were also used to study the effects of α-synuclein knockout or GBA overexpression on MPTP-induced mitochondrial defects and susceptibility. Results L444P GBA heterozygous mutation reduced GBA protein levels, enzymatic activity and a concomitant accumulation of α-synuclein in the midbrain of GBA +/L444P mice. Furthermore, the deficiency resulted in defects in mitochondria of cortical neurons cultured from GBA +/L444P mice. Notably, treatment with MPTP resulted in a significant loss of dopaminergic neurons and striatal dopaminergic fibers in GBA +/L444P mice compared to wild type (WT) mice. Levels of striatal DA and its metabolites were more depleted in the striatum of GBA +/L444P mice. Behavioral deficits, neuroinflammation, and mitochondrial defects were more exacerbated in GBA +/L444P mice after MPTP treatment. Importantly, MPTP induced PD-like symptoms were significantly improved by knockout of α-synuclein or augmentation of GBA via AAV5-hGBA injection in both WT and GBA +/L444P mice. Intriguingly, the degree of reduction in MPTP induced PD-like symptoms in GBA +/L444P α-synuclein (SNCA) −/− mice was nearly equal to that in SNCA −/− mice after MPTP treatment. Conclusion Our results suggest that GBA deficiency due to L444P GBA heterozygous mutation and the accompanying accumulation of α-synuclein render DA neurons more susceptible to MPTP intoxication. Thus, GBA and α-synuclein play dual physiological roles in the survival of DA neurons in response to the mitochondrial dopaminergic neurotoxin, MPTP.
topic Parkinson’s disease
GBA
MPTP
Mitochondrial dysfunction
α-synuclein
url http://link.springer.com/article/10.1186/s13024-017-0233-5
work_keys_str_mv AT seungpilyun asynucleinaccumulationandgbadeficiencyduetol444pgbamutationcontributestomptpinducedparkinsonism
AT donghoonkim asynucleinaccumulationandgbadeficiencyduetol444pgbamutationcontributestomptpinducedparkinsonism
AT sangjunekim asynucleinaccumulationandgbadeficiencyduetol444pgbamutationcontributestomptpinducedparkinsonism
AT sangminkim asynucleinaccumulationandgbadeficiencyduetol444pgbamutationcontributestomptpinducedparkinsonism
AT senthilkumarskaruppagounder asynucleinaccumulationandgbadeficiencyduetol444pgbamutationcontributestomptpinducedparkinsonism
AT seunghwankwon asynucleinaccumulationandgbadeficiencyduetol444pgbamutationcontributestomptpinducedparkinsonism
AT saebomlee asynucleinaccumulationandgbadeficiencyduetol444pgbamutationcontributestomptpinducedparkinsonism
AT taeinkam asynucleinaccumulationandgbadeficiencyduetol444pgbamutationcontributestomptpinducedparkinsonism
AT suhyunlee asynucleinaccumulationandgbadeficiencyduetol444pgbamutationcontributestomptpinducedparkinsonism
AT sangwooham asynucleinaccumulationandgbadeficiencyduetol444pgbamutationcontributestomptpinducedparkinsonism
AT jaehongpark asynucleinaccumulationandgbadeficiencyduetol444pgbamutationcontributestomptpinducedparkinsonism
AT valinaldawson asynucleinaccumulationandgbadeficiencyduetol444pgbamutationcontributestomptpinducedparkinsonism
AT tedmdawson asynucleinaccumulationandgbadeficiencyduetol444pgbamutationcontributestomptpinducedparkinsonism
AT yunjonglee asynucleinaccumulationandgbadeficiencyduetol444pgbamutationcontributestomptpinducedparkinsonism
AT hanseokko asynucleinaccumulationandgbadeficiencyduetol444pgbamutationcontributestomptpinducedparkinsonism
_version_ 1725884826771259392
spelling doaj-213271272ea14b04817b478ead2a71ae2020-11-24T21:50:10ZengBMCMolecular Neurodegeneration1750-13262018-01-0113111910.1186/s13024-017-0233-5α-Synuclein accumulation and GBA deficiency due to L444P GBA mutation contributes to MPTP-induced parkinsonismSeung Pil Yun0Donghoon Kim1Sangjune Kim2SangMin Kim3Senthilkumar S. Karuppagounder4Seung-Hwan Kwon5Saebom Lee6Tae-In Kam7Suhyun Lee8Sangwoo Ham9Jae Hong Park10Valina L. Dawson11Ted M. Dawson12Yunjong Lee13Han Seok Ko14Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of MedicineNeuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of MedicineNeuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of MedicineNeuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of MedicineNeuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of MedicineNeuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of MedicineNeuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of MedicineNeuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of MedicineNeuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of MedicineDivision of Pharmacology, Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research InstituteNeuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of MedicineNeuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of MedicineNeuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of MedicineDivision of Pharmacology, Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research InstituteNeuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of MedicineAbstract Background Mutations in glucocerebrosidase (GBA) cause Gaucher disease (GD) and increase the risk of developing Parkinson’s disease (PD) and Dementia with Lewy Bodies (DLB). Since both genetic and environmental factors contribute to the pathogenesis of sporadic PD, we investigated the susceptibility of nigrostriatal dopamine (DA) neurons in L444P GBA heterozygous knock-in (GBA +/L444P ) mice to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a selective dopaminergic mitochondrial neurotoxin. Method We used GBA +/L444P mice, α-synuclein knockout (SNCA −/− ) mice at 8 months of age, and adeno-associated virus (AAV)-human GBA overexpression to investigate the rescue effect of DA neuronal loss and susceptibility by MPTP. Mitochondrial morphology and functional assay were used to identify mitochondrial defects in GBA +/L444P mice. Motor behavioral test, immunohistochemistry, and HPLC were performed to measure dopaminergic degeneration by MPTP and investigate the relationship between GBA mutation and α-synuclein. Mitochondrial immunostaining, qPCR, and Western blot were also used to study the effects of α-synuclein knockout or GBA overexpression on MPTP-induced mitochondrial defects and susceptibility. Results L444P GBA heterozygous mutation reduced GBA protein levels, enzymatic activity and a concomitant accumulation of α-synuclein in the midbrain of GBA +/L444P mice. Furthermore, the deficiency resulted in defects in mitochondria of cortical neurons cultured from GBA +/L444P mice. Notably, treatment with MPTP resulted in a significant loss of dopaminergic neurons and striatal dopaminergic fibers in GBA +/L444P mice compared to wild type (WT) mice. Levels of striatal DA and its metabolites were more depleted in the striatum of GBA +/L444P mice. Behavioral deficits, neuroinflammation, and mitochondrial defects were more exacerbated in GBA +/L444P mice after MPTP treatment. Importantly, MPTP induced PD-like symptoms were significantly improved by knockout of α-synuclein or augmentation of GBA via AAV5-hGBA injection in both WT and GBA +/L444P mice. Intriguingly, the degree of reduction in MPTP induced PD-like symptoms in GBA +/L444P α-synuclein (SNCA) −/− mice was nearly equal to that in SNCA −/− mice after MPTP treatment. Conclusion Our results suggest that GBA deficiency due to L444P GBA heterozygous mutation and the accompanying accumulation of α-synuclein render DA neurons more susceptible to MPTP intoxication. Thus, GBA and α-synuclein play dual physiological roles in the survival of DA neurons in response to the mitochondrial dopaminergic neurotoxin, MPTP.http://link.springer.com/article/10.1186/s13024-017-0233-5Parkinson’s diseaseGBAMPTPMitochondrial dysfunctionα-synuclein