151
|
Volta M, Milnerwood AJ, Farrer MJ. Insights from late-onset familial parkinsonism on the pathogenesis of idiopathic Parkinson's disease. Lancet Neurol 2015; 14:1054-64. [PMID: 26376970 DOI: 10.1016/s1474-4422(15)00186-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 07/17/2015] [Accepted: 07/20/2015] [Indexed: 01/24/2023]
Abstract
Disease-modifying therapies that slow or halt the progression of Parkinson's disease are an unmet clinical need. Many hypotheses have been put forward to explain the pathogenesis of the disease, but none has led to the development of disease-modifying drugs. Here we focus on familial forms of late-onset parkinsonism that most closely resemble idiopathic Parkinson's disease and present a synthesis of emerging molecular advances. Genetic discoveries and mechanistic investigations have highlighted early alterations to synaptic function, endosomal maturation, and protein sorting that might lead to an intracellular proteinopathy. We propose that these cellular processes constitute one pathway to pathogenesis and suggest that neuroprotection, as an adjunct to current symptomatic treatments, need not remain an elusive goal.
Collapse
Affiliation(s)
- Mattia Volta
- Department of Medical Genetics, Centre for Applied Neurogenetics, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Austen J Milnerwood
- Division of Neurology, Centre for Applied Neurogenetics, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Matthew J Farrer
- Department of Medical Genetics, Centre for Applied Neurogenetics, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
152
|
Xing P, Xu Y, Li H, Liu S, Lu A, Sun S. Ratiometric and colorimetric near-infrared sensors for multi-channel detection of cyanide ion and their application to measure β-glucosidase. Sci Rep 2015; 5:16528. [PMID: 26549546 PMCID: PMC4637889 DOI: 10.1038/srep16528] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 10/15/2015] [Indexed: 11/09/2022] Open
Abstract
A near-infrared sensor for cyanide ion (CN(-)) was developed via internal charge transfer (ICT). This sensor can selectively detect CN(-) either through dual-ratiometric fluorescence (logarithm of I414/I564 and I803/I564) or under various absorption (356 and 440 nm) and emission (414, 564 and 803 nm) channels. Especially, the proposed method can be employed to measure β-glucosidase by detecting CN(-) traces in commercial amygdalin samples.
Collapse
Affiliation(s)
- Panfei Xing
- College of Science, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yongqian Xu
- College of Science, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Hongjuan Li
- College of Science, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Shuhui Liu
- College of Science, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, 999077, China
| | - Shiguo Sun
- College of Science, Northwest A&F University, Yangling, Shaanxi, 712100, China
| |
Collapse
|
153
|
Kumaran R, Cookson MR. Pathways to Parkinsonism Redux: convergent pathobiological mechanisms in genetics of Parkinson's disease. Hum Mol Genet 2015; 24:R32-44. [PMID: 26101198 PMCID: PMC4571999 DOI: 10.1093/hmg/ddv236] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 06/18/2015] [Indexed: 12/19/2022] Open
Abstract
In the past few years, there have been a large number of genes identified that contribute to the lifetime risk of Parkinson's disease (PD). Some genes follow a Mendelian inheritance pattern, but others are risk factors for apparently sporadic PD. Here, we will focus on those genes nominated by genome-wide association studies (GWAS) in sporadic PD, with a particular emphasis on genes that overlap between familial and sporadic disease such as those encoding a-synuclein (SNCA), tau (MAPT), and leucine-rich repeat kinase 2 (LRRK2). We will advance the view that there are likely relationships between these genes that map not only to neuronal processes, but also to neuroinflammation. We will particularly discuss evidence for a role of PD proteins in microglial activation and regulation of the autophagy-lysosome system that is dependent on microtubule transport in neurons. Thus, there are at least two non-mutually exclusive pathways that include both non-cell-autonomous and cell-autonomous mechanisms in the PD brain. Collectively, these data have highlighted the amount of progress made in understanding PD and suggest ways forward to further dissect this disorder.
Collapse
Affiliation(s)
- Ravindran Kumaran
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, 35 Convent Drive, Bethesda, MD 20892-3707, USA
| | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, 35 Convent Drive, Bethesda, MD 20892-3707, USA
| |
Collapse
|
154
|
Gegg ME, Schapira AHV. Mitochondrial dysfunction associated with glucocerebrosidase deficiency. Neurobiol Dis 2015; 90:43-50. [PMID: 26388395 PMCID: PMC4838669 DOI: 10.1016/j.nbd.2015.09.006] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 09/08/2015] [Accepted: 09/16/2015] [Indexed: 02/06/2023] Open
Abstract
The lysosomal hydrolase glucocerebrosidase (GCase) is encoded for by the GBA gene. Homozygous GBA mutations cause Gaucher disease (GD), a lysosomal storage disorder. Furthermore, homozygous and heterozygous GBA mutations are numerically the greatest genetic risk factor for developing Parkinson's disease (PD), the second most common neurodegenerative disorder. The loss of GCase activity results in impairment of the autophagy‐lysosome pathway (ALP), which is required for the degradation of macromolecules and damaged organelles. Aberrant protein handling of α-synuclein by the ALP occurs in both GD and PD. α-synuclein is the principle component of Lewy bodies, a defining hallmark of PD. Mitochondrial dysfunction is also observed in both GD and PD. In this review we will describe how mitochondria are affected following loss of GCase activity. The pathogenic mechanisms leading to mitochondria dysfunction will also be discussed, focusing on the likely inhibition of the degradation of mitochondria by the ALP, also termed mitophagy. Other pathogenic cellular processes associated with GBA mutations that might contribute, such as the unfolding of GCase in the endoplasmic reticulum, calcium dysregulation and neuroinflammation will also be described. Impairment of the ALP and mitochondria dysfunction are common pathogenic themes between GD and PD and probably explain why GBA mutations increase the risk of developing PD that is very similar to sporadic forms of the disease.
Collapse
Affiliation(s)
- Matthew E Gegg
- Department of Clinical Neuroscience, UCL Institute of Neurology, London NW3 2PF, UK
| | - Anthony H V Schapira
- Department of Clinical Neuroscience, UCL Institute of Neurology, London NW3 2PF, UK.
| |
Collapse
|
155
|
Keatinge M, Bui H, Menke A, Chen YC, Sokol AM, Bai Q, Ellett F, Da Costa M, Burke D, Gegg M, Trollope L, Payne T, McTighe A, Mortiboys H, de Jager S, Nuthall H, Kuo MS, Fleming A, Schapira AHV, Renshaw SA, Highley JR, Chacinska A, Panula P, Burton EA, O'Neill MJ, Bandmann O. Glucocerebrosidase 1 deficient Danio rerio mirror key pathological aspects of human Gaucher disease and provide evidence of early microglial activation preceding alpha-synuclein-independent neuronal cell death. Hum Mol Genet 2015; 24:6640-52. [PMID: 26376862 PMCID: PMC4634372 DOI: 10.1093/hmg/ddv369] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 09/04/2015] [Indexed: 01/12/2023] Open
Abstract
Autosomal recessively inherited glucocerebrosidase 1 (GBA1) mutations cause the lysosomal storage disorder Gaucher's disease (GD). Heterozygous GBA1 mutations (GBA1+/−) are the most common risk factor for Parkinson's disease (PD). Previous studies typically focused on the interaction between the reduction of glucocerebrosidase (enzymatic) activity in GBA1+/− carriers and alpha-synuclein-mediated neurotoxicity. However, it is unclear whether other mechanisms also contribute to the increased risk of PD in GBA1+/− carriers. The zebrafish genome does not contain alpha-synuclein (SNCA), thus providing a unique opportunity to study pathogenic mechanisms unrelated to alpha-synuclein toxicity. Here we describe a mutant zebrafish line created by TALEN genome editing carrying a 23 bp deletion in gba1 (gba1c.1276_1298del), the zebrafish orthologue of human GBA1. Marked sphingolipid accumulation was already detected at 5 days post-fertilization with accompanying microglial activation and early, sustained up-regulation of miR-155, a master regulator of inflammation. gba1c.1276_1298del mutant zebrafish developed a rapidly worsening phenotype from 8 weeks onwards with striking reduction in motor activity by 12 weeks. Histopathologically, we observed marked Gaucher cell invasion of the brain and other organs. Dopaminergic neuronal cell count was normal through development but reduced by >30% at 12 weeks in the presence of ubiquitin-positive, intra-neuronal inclusions. This gba1c.1276_1298del zebrafish line is the first viable vertebrate model sharing key pathological features of GD in both neuronal and non-neuronal tissue. Our study also provides evidence for early microglial activation prior to alpha-synuclein-independent neuronal cell death in GBA1 deficiency and suggests upregulation of miR-155 as a common denominator across different neurodegenerative disorders.
Collapse
Affiliation(s)
- Marcus Keatinge
- The Bateson Centre, Sheffield Institute for Translational Neuroscience (SITraN)
| | - Hai Bui
- Lilly Research Laboratories, Eli Lilly & Company, Indianapolis, USA
| | | | - Yu-Chia Chen
- Neuroscience Center and Department of Anatomy, University of Helsinki, Finland
| | - Anna M Sokol
- Laboratory of Mitochondrial Biogenesis, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Qing Bai
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | | | - Marc Da Costa
- The Bateson Centre, Sheffield Institute for Translational Neuroscience (SITraN)
| | - Derek Burke
- Molecular and Genetics Unit, University College London Institute of Child Health, Enzyme Unit and Metabolic Unit, Chemical Pathology, Great Ormond Street Hospital, London, UK
| | - Matthew Gegg
- Department of Clinical Neurosciences, University College London Institute of Neurology, London, UK
| | - Lisa Trollope
- The Bateson Centre, Sheffield Institute for Translational Neuroscience (SITraN)
| | - Thomas Payne
- The Bateson Centre, Sheffield Institute for Translational Neuroscience (SITraN)
| | - Aimee McTighe
- The Bateson Centre, Sheffield Institute for Translational Neuroscience (SITraN)
| | | | - Sarah de Jager
- Department of Medical Genetics, Cambridge Institute for Medical Research University of Cambridge, Cambridge, UK and
| | - Hugh Nuthall
- Lilly Research Laboratories, Eli Lilly & Company, Indianapolis, USA
| | - Ming-Shang Kuo
- Lilly Research Laboratories, Eli Lilly & Company, Indianapolis, USA
| | - Angeleen Fleming
- Department of Medical Genetics, Cambridge Institute for Medical Research University of Cambridge, Cambridge, UK and
| | - Anthony H V Schapira
- Department of Clinical Neurosciences, University College London Institute of Neurology, London, UK
| | - Stephen A Renshaw
- The Bateson Centre, Department of Infection and Immunity, University of Sheffield, Sheffield, UK
| | | | - Agnieszka Chacinska
- Laboratory of Mitochondrial Biogenesis, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Pertti Panula
- Neuroscience Center and Department of Anatomy, University of Helsinki, Finland
| | - Edward A Burton
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | | | - Oliver Bandmann
- The Bateson Centre, Sheffield Institute for Translational Neuroscience (SITraN),
| |
Collapse
|
156
|
Suzuki M, Fujikake N, Takeuchi T, Kohyama-Koganeya A, Nakajima K, Hirabayashi Y, Wada K, Nagai Y. Glucocerebrosidase deficiency accelerates the accumulation of proteinase K-resistant α-synuclein and aggravates neurodegeneration in aDrosophilamodel of Parkinson's disease. Hum Mol Genet 2015; 24:6675-86. [DOI: 10.1093/hmg/ddv372] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/07/2015] [Indexed: 11/14/2022] Open
|
157
|
Beilina A, Cookson MR. Genes associated with Parkinson's disease: regulation of autophagy and beyond. J Neurochem 2015. [PMID: 26223426 DOI: 10.1111/jnc.13266] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Substantial progress has been made in the genetic basis of Parkinson's disease (PD). In particular, by identifying genes that segregate with inherited PD or show robust association with sporadic disease, and by showing the same genes are found on both lists, we have generated an outline of the cause of this condition. Here, we will discuss what those genes tell us about the underlying biology of PD. We specifically discuss the relationships between protein products of PD genes and show that common links include regulation of the autophagy-lysosome system, an important way by which cells recycle proteins and organelles. We also discuss whether all PD genes should be considered to be in the same pathway and propose that in some cases the relationships are closer, whereas in other cases the interactions are more distant and might be considered separate. Beilina and Cookson review the links between genes for Parkinson's disease (red) and the autophagy-lysosomal system. They propose the hypothesis that many of the known PD genes can be assigned to pathways that affect (I) turnover of mitochondria via mitophagy (II) turnover of several vesicular structures via macroautophagy or chaperone-mediated autophagy or (III) general lysosome function. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Alexandra Beilina
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
| | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA.
| |
Collapse
|
158
|
Dansithong W, Paul S, Scoles DR, Pulst SM, Huynh DP. Generation of SNCA Cell Models Using Zinc Finger Nuclease (ZFN) Technology for Efficient High-Throughput Drug Screening. PLoS One 2015; 10:e0136930. [PMID: 26317803 PMCID: PMC4552753 DOI: 10.1371/journal.pone.0136930] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 08/10/2015] [Indexed: 12/17/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder caused by loss of dopaminergic neurons of the substantia nigra. The hallmark of PD is the appearance of neuronal protein aggregations known as Lewy bodies and Lewy neurites, of which α-synuclein forms a major component. Familial PD is rare and is associated with missense mutations of the SNCA gene or increases in gene copy number resulting in SNCA overexpression. This suggests that lowering SNCA expression could be therapeutic for PD. Supporting this hypothesis, SNCA reduction was neuroprotective in cell line and rodent PD models. We developed novel cell lines expressing SNCA fused to the reporter genes luciferase (luc) or GFP with the objective to enable high-throughput compound screening (HTS) for small molecules that can lower SNCA expression. Because SNCA expression is likely regulated by far-upstream elements (including the NACP-REP1 located at 8852 bp upstream of the transcription site), we employed zinc finger nuclease (ZFN) genome editing to insert reporter genes in-frame downstream of the SNCA gene in order to retain native SNCA expression control. This ensured full retention of known and unknown up- and downstream genetic elements controlling SNCA expression. Treatment of cells with the histone deacetylase inhibitor valproic acid (VPA) resulted in significantly increased SNCA-luc and SNCA-GFP expression supporting the use of our cell lines for identifying small molecules altering complex modes of expression control. Cells expressing SNCA-luc treated with a luciferase inhibitor or SNCA siRNA resulted in Z'-scores ≥ 0.75, suggesting the suitability of these cell lines for use in HTS. This study presents a novel use of genome editing for the creation of cell lines expressing α-synuclein fusion constructs entirely under native expression control. These cell lines are well suited for HTS for compounds that lower SNCA expression directly or by acting at long-range sites to the SNCA promoter and 5'-UTR.
Collapse
Affiliation(s)
- Warunee Dansithong
- Department of Neurology, University of Utah, 175 North Medical Center Drive East, 5th Floor, Salt Lake City, Utah, 84132, United States of America
| | - Sharan Paul
- Department of Neurology, University of Utah, 175 North Medical Center Drive East, 5th Floor, Salt Lake City, Utah, 84132, United States of America
| | - Daniel R. Scoles
- Department of Neurology, University of Utah, 175 North Medical Center Drive East, 5th Floor, Salt Lake City, Utah, 84132, United States of America
| | - Stefan M. Pulst
- Department of Neurology, University of Utah, 175 North Medical Center Drive East, 5th Floor, Salt Lake City, Utah, 84132, United States of America
| | - Duong P. Huynh
- Department of Neurology, University of Utah, 175 North Medical Center Drive East, 5th Floor, Salt Lake City, Utah, 84132, United States of America
- * E-mail:
| |
Collapse
|
159
|
Han F, Grimes DA, Li F, Wang T, Yu Z, Song N, Wu S, Racacho L, Bulman DE. Mutations in the glucocerebrosidase gene are common in patients with Parkinson's disease from Eastern Canada. Int J Neurosci 2015; 126:415-21. [PMID: 26000814 DOI: 10.3109/00207454.2015.1023436] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Mutations in the β-glucocerebrosidase gene (GBA) have been implicated as a risk factor for Parkinson's disease (PD). However, GBA mutations in PD patients of different ethnic origins were reported to be inconsistent. METHODS We sequenced all exons of the GBA gene in 225 PD patients and 110 control individuals from Eastern Canada. RESULT Two novel GBA variants of c.-119 A/G and S(-35)N, five known GBA mutations of R120W, N370S, L444P, RecNciI and RecTL mutation (del55/D409H/RecNciI) as well as two non-pathological variants of E326K and T369M were identified from PD patients while only one mutation of S13L and two non-pathological variants of E326K and T369M were found in the control individuals. The frequency of GBA mutations within PD patients (4.4%) is 4.8 times higher than the 0.91% observed in control individuals (X(2) = 2.91, p = 0.088; odds ratio = 4.835; 95% confidence interval = 2.524-9.123). The most common mutations of N370S and L444P accounted for 36.0% (9/25) of all the GBA mutations in this Eastern Canadian PD cohort. The frequency (6.67%) of E326K and T369M in PD patients is comparable to 7.27% in control individuals (X(2) = 0.042, p = 0.8376), further supporting that these two variants have no pathological effects on PD. Phenotype analysis showed that no significant difference in family history, age at onset and cognitive impairment was identified between the GBA mutation carriers and non-GBA mutation carriers. CONCLUSION GBA mutations were found to be a common genetic risk factor for PD in Eastern Canadian patients.
Collapse
Affiliation(s)
- Fabin Han
- a 1 Centre for Stem cells and Regenerative Medicine, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital , Taishan Medical University , Liaocheng , China.,b 2 Department of Neurology, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital , Taishan Medical University , Liaocheng , China
| | - David A Grimes
- c 3 Department of Medicine, The Ottawa Hospital , University of Ottawa , Ottawa , Canada
| | - Fang Li
- c 3 Department of Medicine, The Ottawa Hospital , University of Ottawa , Ottawa , Canada
| | - Ting Wang
- a 1 Centre for Stem cells and Regenerative Medicine, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital , Taishan Medical University , Liaocheng , China
| | - Zhe Yu
- a 1 Centre for Stem cells and Regenerative Medicine, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital , Taishan Medical University , Liaocheng , China
| | - Na Song
- a 1 Centre for Stem cells and Regenerative Medicine, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital , Taishan Medical University , Liaocheng , China
| | - Shichao Wu
- a 1 Centre for Stem cells and Regenerative Medicine, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital , Taishan Medical University , Liaocheng , China
| | - Lemuel Racacho
- d 4 Department of Pediatrics, Children's Hospital of Eastern Ontario Research Institute , University of Ottawa , Ottawa , Canada
| | - Dennis E Bulman
- d 4 Department of Pediatrics, Children's Hospital of Eastern Ontario Research Institute , University of Ottawa , Ottawa , Canada
| |
Collapse
|
160
|
Kenney DL, Benarroch EE. The autophagy-lysosomal pathway: General concepts and clinical implications. Neurology 2015. [PMID: 26203091 DOI: 10.1212/wnl.0000000000001860] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Daniel L Kenney
- From the Departments of Child and Adolescent Neurology (D.L.K.) and Neurology (E.E.B.), Mayo Clinic, Rochester, MN
| | - Eduardo E Benarroch
- From the Departments of Child and Adolescent Neurology (D.L.K.) and Neurology (E.E.B.), Mayo Clinic, Rochester, MN.
| |
Collapse
|
161
|
Abstract
The last 2 decades represent a period of unparalleled advancement in the understanding of the pathogenesis of Parkinson disease (PD). The discovery of several forms of familial parkinsonism with mendelian inheritance has elucidated insights into the mechanisms underlying the degeneration of dopaminergic neurons of the substantia nigra that histologically characterize PD. α-Synuclein, the principal component of Lewy bodies, remains the presumed pathogen at the heart of the current model; however, concurrently, a diverse range of other mechanisms have been implicated. The creation of a coherent disease model will be crucial to the development of effective disease modifying therapies for sporadic PD.
Collapse
Affiliation(s)
- Stephen Mullin
- Department of Clinical Neurosciences, UCL Institute of Neurology, Rowland Hill Street, Hampstead, London NW3 2PF, UK
| | - Anthony H V Schapira
- Department of Clinical Neurosciences, UCL Institute of Neurology, Rowland Hill Street, Hampstead, London NW3 2PF, UK.
| |
Collapse
|
162
|
Alcalay RN, Levy OA, Waters CC, Fahn S, Ford B, Kuo SH, Mazzoni P, Pauciulo MW, Nichols WC, Gan-Or Z, Rouleau GA, Chung WK, Wolf P, Oliva P, Keutzer J, Marder K, Zhang X. Glucocerebrosidase activity in Parkinson's disease with and without GBA mutations. Brain 2015; 138:2648-58. [PMID: 26117366 DOI: 10.1093/brain/awv179] [Citation(s) in RCA: 299] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/27/2015] [Indexed: 11/14/2022] Open
Abstract
Glucocerebrosidase (GBA) mutations have been associated with Parkinson's disease in numerous studies. However, it is unknown whether the increased risk of Parkinson's disease in GBA carriers is due to a loss of glucocerebrosidase enzymatic activity. We measured glucocerebrosidase enzymatic activity in dried blood spots in patients with Parkinson's disease (n = 517) and controls (n = 252) with and without GBA mutations. Participants were recruited from Columbia University, New York, and fully sequenced for GBA mutations and genotyped for the LRRK2 G2019S mutation, the most common autosomal dominant mutation in the Ashkenazi Jewish population. Glucocerebrosidase enzymatic activity in dried blood spots was measured by a mass spectrometry-based assay and compared among participants categorized by GBA mutation status and Parkinson's disease diagnosis. Parkinson's disease patients were more likely than controls to carry the LRRK2 G2019S mutation (n = 39, 7.5% versus n = 2, 0.8%, P < 0.001) and GBA mutations or variants (seven homozygotes and compound heterozygotes and 81 heterozygotes, 17.0% versus 17 heterozygotes, 6.7%, P < 0.001). GBA homozygotes/compound heterozygotes had lower enzymatic activity than GBA heterozygotes (0.85 µmol/l/h versus 7.88 µmol/l/h, P < 0.001), and GBA heterozygotes had lower enzymatic activity than GBA and LRRK2 non-carriers (7.88 µmol/l/h versus 11.93 µmol/l/h, P < 0.001). Glucocerebrosidase activity was reduced in heterozygotes compared to non-carriers when each mutation was compared independently (N370S, P < 0.001; L444P, P < 0.001; 84GG, P = 0.003; R496H, P = 0.018) and also reduced in GBA variants associated with Parkinson's risk but not with Gaucher disease (E326K, P = 0.009; T369M, P < 0.001). When all patients with Parkinson's disease were considered, they had lower mean glucocerebrosidase enzymatic activity than controls (11.14 µmol/l/h versus 11.85 µmol/l/h, P = 0.011). Difference compared to controls persisted in patients with idiopathic Parkinson's disease (after exclusion of all GBA and LRRK2 carriers; 11.53 µmol/l/h, versus 12.11 µmol/l/h, P = 0.036) and after adjustment for age and gender (P = 0.012). Interestingly, LRRK2 G2019S carriers (n = 36), most of whom had Parkinson's disease, had higher enzymatic activity than non-carriers (13.69 µmol/l/h versus 11.93 µmol/l/h, P = 0.002). In patients with idiopathic Parkinson's, higher glucocerebrosidase enzymatic activity was associated with longer disease duration (P = 0.002) in adjusted models, suggesting a milder disease course. We conclude that lower glucocerebrosidase enzymatic activity is strongly associated with GBA mutations, and modestly with idiopathic Parkinson's disease. The association of lower glucocerebrosidase activity in both GBA mutation carriers and Parkinson's patients without GBA mutations suggests that loss of glucocerebrosidase function contributes to the pathogenesis of Parkinson's disease. High glucocerebrosidase enzymatic activity in LRRK2 G2019S carriers may reflect a distinct pathogenic mechanism. Taken together, these data suggest that glucocerebrosidase enzymatic activity could be a modifiable therapeutic target.
Collapse
Affiliation(s)
- Roy N Alcalay
- 1 Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA 2 Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Oren A Levy
- 1 Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA 2 Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Cheryl C Waters
- 1 Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Stanley Fahn
- 1 Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Blair Ford
- 1 Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Sheng-Han Kuo
- 1 Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Pietro Mazzoni
- 1 Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Michael W Pauciulo
- 3 Division of Human Genetics, Cincinnati Children's Hospital Medical Center and the Department of Pediatrics; University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - William C Nichols
- 3 Division of Human Genetics, Cincinnati Children's Hospital Medical Center and the Department of Pediatrics; University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ziv Gan-Or
- 4 Montréal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Guy A Rouleau
- 4 Montréal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Wendy K Chung
- 5 Department of Pediatrics and Medicine, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Pavlina Wolf
- 6 Global BioTherapeutics, Genzyme, a Sanofi company, Framingham, MA, USA
| | - Petra Oliva
- 6 Global BioTherapeutics, Genzyme, a Sanofi company, Framingham, MA, USA
| | - Joan Keutzer
- 6 Global BioTherapeutics, Genzyme, a Sanofi company, Framingham, MA, USA
| | - Karen Marder
- 1 Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA 2 Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA 7 Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Xiaokui Zhang
- 6 Global BioTherapeutics, Genzyme, a Sanofi company, Framingham, MA, USA
| |
Collapse
|
163
|
Gegg ME, Sweet L, Wang BH, Shihabuddin LS, Sardi SP, Schapira AHV. No evidence for substrate accumulation in Parkinson brains with GBA mutations. Mov Disord 2015; 30:1085-9. [PMID: 26096906 PMCID: PMC4529481 DOI: 10.1002/mds.26278] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/20/2015] [Accepted: 05/03/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND To establish whether Parkinson's disease (PD) brains previously described to have decreased glucocerebrosidase activity exhibit accumulation of the lysosomal enzyme's substrate, glucosylceramide, or other changes in lipid composition. METHODS Lipidomic analyses and cholesterol measurements were performed on the putamen (n = 5-7) and cerebellum (n = 7-14) of controls, Parkinson's disease brains with heterozygote GBA1 mutations (PD+GBA), or sporadic PD. RESULTS Total glucosylceramide levels were unchanged in both PD+GBA and sporadic PD brains when compared with controls. No changes in glucosylsphingosine (deacetylated glucosylceramide), sphingomyelin, gangliosides (GM2, GM3), or total cholesterol were observed in either putamen or cerebellum. CONCLUSIONS This study did not demonstrate glucocerebrosidase substrate accumulation in PD brains with heterozygote GBA1 mutations in areas of the brain with low α-synuclein pathology.
Collapse
Affiliation(s)
- Matthew E Gegg
- Department of Clinical Neuroscience, UCL Institute of Neurology, London, UK
| | - Lindsay Sweet
- Genzyme, a Sanofi Company, Framingham, Massachusetts, USA
| | - Bing H Wang
- Genzyme, a Sanofi Company, Framingham, Massachusetts, USA
| | | | | | | |
Collapse
|
164
|
Abstract
BACKGROUND Parkinson's disease (PD) was previously described as the prototypical sporadic disease; however, rapid advances in population and molecular genetics have revealed the existence of a significant number genetic risk factors, prompting its redefinition as a primarily genetic disorder. SOURCES OF DATA Data for this review have been gathered from the published literature. AREAS OF AGREEMENT Multiple haplotypes conveying variable but quantifiable genetic risk, acting concurrently and possibly interacting with one another, provide the basis for a new model of PD. The beginning of this revolution in our understanding came from the clinical observation of parkinsonism with a Mendelian pattern of inheritance in a number of families. The functional work that followed elucidated multiple disease pathways leading to the degeneration of the substantia nigra that characterizes PD. It is however only in recent years, with the emergence of large cohort genome-wide association studies (GWAS), that the relevance of these pathways to so-called sporadic PD has become apparent. AREAS OF CONTROVERSY A substantial portion of the presumed genetic inheritance of PD remains at present undefined. Although it is likely that so-called intermediate risk genetic risk factors are the principal component of this 'missing heritability', this is yet to be proved. GROWING POINTS Although the picture is by now means complete, the beginnings of rational basis for genetic screening of PD risk have begun to emerge. Equally, this enhanced understanding of the various genetic and in turn biochemical pathways shows promising signs of producing fruitful therapeutic strategies. Technological advances promise to reduce the costs associated with and further increase our capability to understand the complex influence of genetics on the pathogenesis of PD. AREAS TIMELY FOR DEVELOPING RESEARCH The coming years will require the enhancement of current techniques and the development of new ones to define PD's missing heritability. It will also require functional work to define better and in turn potentially reverse the mechanisms that contribute with large effect sizes to the risk of sporadic PD.
Collapse
Affiliation(s)
- Stephen Mullin
- Leonard Wolfson Clinical Research Fellow, UCL, Institute of Neurology, Rowland Hill Street, Hampstead, London NW3 2PF, UK
| | - Anthony Schapira
- Department of Clinical Neurosciences, UCL, Institute of Neurology, Hampstead, London, UK
| |
Collapse
|
165
|
Schapira AHV. Glucocerebrosidase and Parkinson disease: Recent advances. Mol Cell Neurosci 2015; 66:37-42. [PMID: 25802027 PMCID: PMC4471139 DOI: 10.1016/j.mcn.2015.03.013] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/10/2015] [Accepted: 03/12/2015] [Indexed: 12/13/2022] Open
Abstract
Mutations of the glucocerebrosidase (GBA) gene are the most important risk factor yet discovered for Parkinson disease (PD). Homozygous GBA mutations result in Gaucher disease (GD), a lysosomal storage disorder. Heterozygous mutations have not until recently been thought to be associated with any pathological process. However, it is clear that the presence of a GBA mutation in homozygous or heterozygous form is associated with an approximately 20-fold increase in the risk for PD, with little if any difference in risk burden related to gene dose. Most studies suggest that 5-10% of PD patients have GBA mutations, although this figure is greater in the Ashkenazi population and may be an underestimate overall if the entire exome is not sequenced. GBA-associated PD is clinically indistinguishable from idiopathic PD, except for slightly earlier age of onset and a greater frequency of cognitive impairment. Pathological and imaging features, and response to pharmacotherapy are identical to idiopathic PD. GBA mutations result in reduced enzyme activity and mutant protein may become trapped in the endoplasmic reticulum (ER) leading to unfolded protein response and ER associated degradation and stress. Both mechanisms may be relevant in GD and PD pathogenesis and lead to impaired lysosomal function. Of particular relevance to PD is the interaction of glucocerebrosidase enzyme (GCase) with alpha-synuclein (SNCA). There appears to be a bi-directional reciprocal relationship between GCase levels and those of SNCA. Thus reduced GCase in GBA mutation PD brain is associated with increased SNCA, and increased SNCA deposition is associated with reduced GCase even in GBA wild-type PD brains. It is noteworthy that GBA mutations are also associated with an increase in risk for dementia with Lewy bodies, another synucleinopathy. It has been suggested that the relationship between GCase and SNCA may be leveraged to reduce SNCA levels in PD by enhancing GCase levels and activity. This hypothesis has been confirmed in GBA mutant mice, PD patient fibroblasts and cells with SNCA overexpression, and offers an important target pathway for future neuroprotection therapy in PD. This article is part of a Special Issue entitled 'Neuronal Protein'.
Collapse
Affiliation(s)
- Anthony H V Schapira
- Department of Clinical Neurosciences, UCL Institute of Neurology, UCL Royal Free Campus, Rowland Hill Street, London NW3 2PF, United Kingdom.
| |
Collapse
|
166
|
Autophagy in neurodegenerative diseases: From pathogenic dysfunction to therapeutic modulation. Semin Cell Dev Biol 2015; 40:115-26. [PMID: 25843774 DOI: 10.1016/j.semcdb.2015.03.005] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/05/2015] [Accepted: 03/06/2015] [Indexed: 12/12/2022]
Abstract
Neuronal homeostasis depends on the proper functioning of quality control systems like autophagy. This mechanism is responsible of the clearance of misfolded proteins, aggregates and the turnover of organelles within the neuron. Autophagic dysfunction has been described in many neurodegenerative diseases. It can occur at several steps of the autophagic machinery and can contribute to the formation of intracellular aggregates and ultimately to neuronal death. Accordingly restoring autophagy activity in affected neurons can be an attractive therapeutic approach to fight neurodegeneration. In this review we summarize the present encouraging strategies that have been achieved with pharmacological and genetic treatments aimed to induce neuronal autophagy in experimental models of neurodegenerative diseases.
Collapse
|
167
|
Sun Y, Florer J, Mayhew CN, Jia Z, Zhao Z, Xu K, Ran H, Liou B, Zhang W, Setchell KDR, Gu J, Grabowski GA. Properties of neurons derived from induced pluripotent stem cells of Gaucher disease type 2 patient fibroblasts: potential role in neuropathology. PLoS One 2015; 10:e0118771. [PMID: 25822147 PMCID: PMC4378893 DOI: 10.1371/journal.pone.0118771] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 01/06/2015] [Indexed: 11/30/2022] Open
Abstract
Gaucher disease (GD) is caused by insufficient activity of acid β-glucosidase (GCase) resulting from mutations in GBA1. To understand the pathogenesis of the neuronopathic GD, induced pluripotent stem cells (iPSCs) were generated from fibroblasts isolated from three GD type 2 (GD2) and 2 unaffected (normal and GD carrier) individuals. The iPSCs were converted to neural precursor cells (NPCs) which were further differentiated into neurons. Parental GD2 fibroblasts as well as iPSCs, NPCs, and neurons had similar degrees of GCase deficiency. Lipid analyses showed increases of glucosylsphingosine and glucosylceramide in the GD2 cells. In addition, GD2 neurons showed increased α-synuclein protein compared to control neurons. Whole cell patch-clamping of the GD2 and control iPSCs-derived neurons demonstrated excitation characteristics of neurons, but intriguingly, those from GD2 exhibited consistently less negative resting membrane potentials with various degree of reduction in action potential amplitudes, sodium and potassium currents. Culture of control neurons in the presence of the GCase inhibitor (conduritol B epoxide) recapitulated these findings, providing a functional link between decreased GCase activity in GD and abnormal neuronal electrophysiological properties. To our knowledge, this study is first to report abnormal electrophysiological properties in GD2 iPSC-derived neurons that may underlie the neuropathic phenotype in Gaucher disease.
Collapse
Affiliation(s)
- Ying Sun
- Division of Human Genetics, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Jane Florer
- Division of Human Genetics, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Christopher N. Mayhew
- Division of Developmental Biology, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Zhanfeng Jia
- Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Zhiying Zhao
- Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Kui Xu
- Division of Human Genetics, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Huimin Ran
- Division of Human Genetics, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Benjamin Liou
- Division of Human Genetics, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Wujuan Zhang
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Kenneth D. R. Setchell
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Jianguo Gu
- Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Gregory A. Grabowski
- Division of Human Genetics, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Synageva BioPharma Corp., Lexington, Massachusetts, United States of America
| |
Collapse
|
168
|
Elstein D, Alcalay R, Zimran A. The emergence of Parkinson disease among patients with Gaucher disease. Best Pract Res Clin Endocrinol Metab 2015; 29:249-59. [PMID: 25987177 DOI: 10.1016/j.beem.2014.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
In the last decade, several lines of evidence have been presented that document the clinical manifestations, genetic associations, and sub-cellular mechanisms of the inter-relatedness of β-glucocerebrosidase mutations and the emergence of Parkinson disease among carriers and patients with Gaucher disease. This review is an attempt to apprise the reader of the recent literature with the caveat that this is an area of intensive exploration that is constantly being updated because of the immediate clinical ramifications but also because of the impact on our understanding of Parkinson disease, and finally because of the unexpected inter-reactions between these entities on the molecular level. It has been an unexpected happenstance that it has been discovered that a rare monogenetic disease has an interface at many points with a neurological disorder of the elderly that has both familial and sporadic forms: to date there is no cure for either of these disorders.
Collapse
Affiliation(s)
- Deborah Elstein
- Gaucher Clinic, Shaare Zedek Medical Center Affiliated with the Hebrew University School of Medicine, Jerusalem Israel.
| | - Roy Alcalay
- Department of Neurology and the Taub Institute, Columbia University Medical Center, New York, NY, USA.
| | - Ari Zimran
- Gaucher Clinic, Shaare Zedek Medical Center Affiliated with the Hebrew University School of Medicine, Jerusalem Israel.
| |
Collapse
|
169
|
Autophagy in neuronal cells: general principles and physiological and pathological functions. Acta Neuropathol 2015; 129:337-62. [PMID: 25367385 DOI: 10.1007/s00401-014-1361-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 10/21/2014] [Accepted: 10/25/2014] [Indexed: 12/12/2022]
Abstract
Autophagy delivers cytoplasmic components and organelles to lysosomes for degradation. This pathway serves to degrade nonfunctional or unnecessary organelles and aggregate-prone and oxidized proteins to produce substrates for energy production and biosynthesis. Macroautophagy delivers large aggregates and whole organelles to lysosomes by first enveloping them into autophagosomes that then fuse with lysosomes. Chaperone-mediated autophagy (CMA) degrades proteins containing the KFERQ-like motif in their amino acid sequence, by transporting them from the cytosol across the lysosomal membrane into the lysosomal lumen. Autophagy is especially important for the survival and homeostasis of postmitotic cells like neurons, because these cells are not able to dilute accumulating detrimental substances and damaged organelles by cell division. Our current knowledge on the autophagic pathways and molecular mechanisms and regulation of autophagy will be summarized in this review. We will describe the physiological functions of macroautophagy and CMA in neuronal cells. Finally, we will summarize the current evidence showing that dysfunction of macroautophagy and/or CMA contributes to neuronal diseases. We will give an overview of our current knowledge on the role of autophagy in aging neurons, and focus on the role of autophagy in four types of neurodegenerative diseases, i.e., amyotrophic lateral sclerosis and frontotemporal dementia, prion diseases, lysosomal storage diseases, and Parkinson's disease.
Collapse
|
170
|
Burrow TA, Sun Y, Prada CE, Bailey L, Zhang W, Brewer A, Wu SW, Setchell KDR, Witte D, Cohen MB, Grabowski GA. CNS, lung, and lymph node involvement in Gaucher disease type 3 after 11 years of therapy: clinical, histopathologic, and biochemical findings. Mol Genet Metab 2015; 114:233-241. [PMID: 25219293 PMCID: PMC4312736 DOI: 10.1016/j.ymgme.2014.08.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 08/27/2014] [Accepted: 08/27/2014] [Indexed: 11/25/2022]
Abstract
A Caucasian male with Gaucher disease type 3, treated with continuous enzyme therapy (ET) for 11 years, experienced progressive mesenteric and retroperitoneal lymphadenopathy, lung disease, and neurological involvement leading to death at an age of 12.5 years. Autopsy showed significant pathology of the brain, lymph nodes, and lungs. Liver and spleen glucosylceramide (GluCer) and glucosylsphingosine (GluS) levels were nearly normal and storage cells were cleared. Clusters of macrophages and very elevated GluCer and GluS levels were in the lungs, and brain parenchymal and perivascular regions. Compared to normal brain GluCer (GC 18:0), GluCer species with long fatty acid acyl chains were increased in the patient's brain. This profile was similar to that in the patient's lungs, suggesting that these lipids were present in brain perivascular macrophages. In the patient's brain, generalized astrogliosis, and enhanced LC3, ubiquitin, and Tau signals were identified in the regions surrounding macrophage clusters, indicating proinflammation, altered autophagy, and neurodegeneration. These findings highlight the altered phenotypes resulting from increased longevity due to ET, as well as those in poorly accessible compartments of brain and lung, which manifested progressive disease involvement despite ET.
Collapse
Affiliation(s)
- Thomas A Burrow
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Ohio
| | - Ying Sun
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Ohio
| | - Carlos E Prada
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati Ohio
- Centro de Medicina Genómica y Metabolismo, Fundación Cardiovascular de Colombia, Colombia
| | - Laurie Bailey
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati Ohio
| | - Wujuan Zhang
- Department of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Amanda Brewer
- Department of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Steve W Wu
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Ohio
- Division of Pediatric Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kenneth D R Setchell
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Ohio
- Department of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David Witte
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Ohio
- Department of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Mitchell B Cohen
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Ohio
- Division of Pediatric Gastroenterology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Gregory A Grabowski
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Ohio
| |
Collapse
|
171
|
Gruschus JM, Jiang Z, Yap TL, Hill SA, Grishaev A, Piszczek G, Sidransky E, Lee JC. Dissociation of glucocerebrosidase dimer in solution by its co-factor, saposin C. Biochem Biophys Res Commun 2015; 457:561-6. [PMID: 25600808 DOI: 10.1016/j.bbrc.2015.01.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 01/08/2015] [Indexed: 12/23/2022]
Abstract
Mutations in the gene for the lysosomal enzyme glucocerebrosidase (GCase) cause Gaucher disease and are the most common risk factor for Parkinson disease (PD). Analytical ultracentrifugation of 8 μM GCase shows equilibrium between monomer and dimer forms. However, in the presence of its co-factor saposin C (Sap C), only monomer GCase is seen. Isothermal calorimetry confirms that Sap C associates with GCase in solution in a 1:1 complex (Kd = 2.1 ± 1.1 μM). Saturation cross-transfer NMR determined that the region of Sap C contacting GCase includes residues 63-66 and 74-76, which is distinct from the region known to enhance GCase activity. Because α-synuclein (α-syn), a protein closely associated with PD etiology, competes with Sap C for GCase binding, its interaction with GCase was also measured by ultracentrifugation and saturation cross-transfer. Unlike Sap C, binding of α-syn to GCase does not affect multimerization. However, adding α-syn reduces saturation cross-transfer from Sap C to GCase, confirming displacement. To explore where Sap C might disrupt multimeric GCase, GCase x-ray structures were analyzed using the program PISA, which predicted stable dimer and tetramer forms. For the most frequently predicted multimer interface, the GCase active sites are partially buried, suggesting that Sap C might disrupt the multimer by binding near the active site.
Collapse
Affiliation(s)
- James M Gruschus
- Laboratory of Molecular Biophysics, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda MD 20892, United States.
| | - Zhiping Jiang
- Laboratory of Molecular Biophysics, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda MD 20892, United States
| | - Thai Leong Yap
- Laboratory of Molecular Biophysics, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda MD 20892, United States
| | - Stephanie A Hill
- Laboratory of Molecular Biophysics, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda MD 20892, United States
| | - Alexander Grishaev
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Grzegorz Piszczek
- Biophysics Core, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Ellen Sidransky
- Section on Molecular Neurogenetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Jennifer C Lee
- Laboratory of Molecular Biophysics, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda MD 20892, United States.
| |
Collapse
|
172
|
Sardi SP, Cheng SH, Shihabuddin LS. Gaucher-related synucleinopathies: the examination of sporadic neurodegeneration from a rare (disease) angle. Prog Neurobiol 2015; 125:47-62. [PMID: 25573151 DOI: 10.1016/j.pneurobio.2014.12.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 12/01/2014] [Accepted: 12/27/2014] [Indexed: 10/24/2022]
Abstract
Gaucher disease, the most common lysosomal storage disease, is caused by a recessively inherited deficiency in glucocerebrosidase and subsequent accumulation of toxic lipid substrates. Heterozygous mutations in the lysosomal glucocerebrosidase gene (GBA1) have recently been recognized as the highest genetic risk factor for the development of α-synuclein aggregation disorders ("synucleinopathies"), including Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Despite the wealth of experimental, clinical and genetic evidence that supports the association between mutant genotypes and synucleinopathy risk, the precise mechanisms by which GBA1 mutations lead to PD and DLB remain unclear. Decreased glucocerebrosidase activity has been demonstrated to promote α-synuclein misprocessing. Furthermore, aberrant α-synuclein species have been reported to downregulate glucocerebrosidase activity, which further contributes to disease progression. In this review, we summarize the recent findings that highlight the complexity of this pathogenetic link and how several pathways that connect glucocerebrosidase insufficiency with α-synuclein misprocessing have emerged as potential therapeutic targets. From a translational perspective, we discuss how various therapeutic approaches to lysosomal dysfunction have been explored for the treatment of GBA1-related synucleinopathies, and potentially, for non-GBA1-associated neurodegenerative diseases. In summary, the link between GBA1 and synucleinopathies has become the paradigm of how the study of a rare lysosomal disease can transform the understanding of the etiopathology, and hopefully the treatment, of a more prevalent and multifactorial disorder.
Collapse
Affiliation(s)
- S Pablo Sardi
- Genzyme, a Sanofi Company, 49 New York Avenue, Framingham, MA 01701, USA.
| | - Seng H Cheng
- Genzyme, a Sanofi Company, 49 New York Avenue, Framingham, MA 01701, USA
| | | |
Collapse
|
173
|
Gan-Or Z, Dion PA, Rouleau GA. Genetic perspective on the role of the autophagy-lysosome pathway in Parkinson disease. Autophagy 2015; 11:1443-57. [PMID: 26207393 PMCID: PMC4590678 DOI: 10.1080/15548627.2015.1067364] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/10/2015] [Accepted: 06/24/2015] [Indexed: 02/09/2023] Open
Abstract
Parkinson disease (PD), once considered as a prototype of a sporadic disease, is now known to be considerably affected by various genetic factors, which interact with environmental factors and the normal process of aging, leading to PD. Large studies determined that the hereditary component of PD is at least 27%, and in some populations, single genetic factors are responsible for more than 33% of PD patients. Interestingly, many of these genetic factors, such as LRRK2, GBA, SMPD1, SNCA, PARK2, PINK1, PARK7, SCARB2, and others, are involved in the autophagy-lysosome pathway (ALP). Some of these genes encode lysosomal enzymes, whereas others correspond to proteins that are involved in transport to the lysosome, mitophagy, or other autophagic-related functions. Is it possible that all these factors converge into a single pathway that causes PD? In this review, we will discuss these genetic findings and the role of the ALP in the pathogenesis of PD and will try to answer this question. We will suggest a novel hypothesis for the pathogenic mechanism of PD that involves the lysosome and the different autophagy pathways.
Collapse
Affiliation(s)
- Ziv Gan-Or
- The Department of Human Genetics; McGill University; Montreal, QC Canada
- Montreal Neurological Institute; McGill University; Montreal, QC Canada
| | - Patrick A Dion
- The Department of Human Genetics; McGill University; Montreal, QC Canada
- Montreal Neurological Institute; McGill University; Montreal, QC Canada
- The Department of Neurology & Neurosurgery; McGill University; Montreal, QC Canada
| | - Guy A Rouleau
- The Department of Human Genetics; McGill University; Montreal, QC Canada
- Montreal Neurological Institute; McGill University; Montreal, QC Canada
- The Department of Neurology & Neurosurgery; McGill University; Montreal, QC Canada
| |
Collapse
|
174
|
|
175
|
Yu Z, Wang T, Xu J, Wang W, Wang G, Chen C, Zheng L, Pan L, Gong D, Li X, Qu H, Li F, Zhang B, Le W, Han F. Mutations in the glucocerebrosidase gene are responsible for Chinese patients with Parkinson's disease. J Hum Genet 2014; 60:85-90. [PMID: 25518742 DOI: 10.1038/jhg.2014.110] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 11/10/2014] [Accepted: 11/18/2014] [Indexed: 01/16/2023]
Abstract
Pathological mutations in the glucocerebrosidase gene (GBA) have been suggested to be associated with Parkinson's disease (PD) in various ethnic populations. Most studies on Chinese PD patients have only screened the N370S and L444P mutations in the GBA gene. To investigate the GBA mutations in Chinese population, we performed complete sequencing of the GBA gene in 184 Chinese PD patients and 130 Chinese control individuals. As a result, we identified three novel and nine reported GBA mutations. The novel mutations include 5-bp deletion (c.334_338delCAGAA), L264I and L314V and the nine reported GBA mutations are R163Q, F213I, E326K, S364S, F347L, V375L, L444P, RecNciI and Q497R. The novel 5-bp deletion (CAGAA) produces a short truncated GBA protein of 142 amino acids, which loses major function domains of the 536 amino acids. Our data also reveals that the frequency of GBA mutations within this Chinese PD cohort was 8.7%, which is significantly higher than 1.54% observed in the Chinese control cohort (χ(2) = 7.22, P = 0.0072; odds ratio (OR) = 6.095, 95% confidence interval of OR = 1.546-24.030). The most common L444P mutation accounts 2.74%, which confer more genetic risk for PD in this Chinese population. In conclusion, novel and known GBA mutations were identified and were found to be associated to PD in this Chinese population.
Collapse
Affiliation(s)
- Zhe Yu
- Centre for Stem cells and Regenerative Medicine, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital, Taishan Medical University, Shandong, China
| | - Ting Wang
- Centre for Stem cells and Regenerative Medicine, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital, Taishan Medical University, Shandong, China
| | - Jun Xu
- 1] Centre for Stem cells and Regenerative Medicine, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital, Taishan Medical University, Shandong, China [2] Department of Neurology, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital, Taishan Medical University, Shandong, China
| | - Wei Wang
- Centre for Stem cells and Regenerative Medicine, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital, Taishan Medical University, Shandong, China
| | - Guifang Wang
- Department of Neurology, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital, Taishan Medical University, Shandong, China
| | - Chao Chen
- Centre for Stem cells and Regenerative Medicine, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital, Taishan Medical University, Shandong, China
| | - Lili Zheng
- Centre for Stem cells and Regenerative Medicine, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital, Taishan Medical University, Shandong, China
| | - Li Pan
- Centre for Stem cells and Regenerative Medicine, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital, Taishan Medical University, Shandong, China
| | - Dianrong Gong
- Department of Neurology, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital, Taishan Medical University, Shandong, China
| | - Xueli Li
- Department of Neurology, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital, Taishan Medical University, Shandong, China
| | - Huaiqian Qu
- Department of Neurology, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital, Taishan Medical University, Shandong, China
| | - Fang Li
- Department of Medicine, The Ottawa Hospital, University of Ottawa, Ontario, Canada
| | - Bin Zhang
- Centre for Stem cells and Regenerative Medicine, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital, Taishan Medical University, Shandong, China
| | - Weidong Le
- Institute of Neurology, Department of Neurology, The Affiliated Reijing Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fabin Han
- 1] Centre for Stem cells and Regenerative Medicine, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital, Taishan Medical University, Shandong, China [2] Department of Neurology, The Affiliated Liaocheng Hospital/Liaocheng People's Hospital, Taishan Medical University, Shandong, China
| |
Collapse
|
176
|
Woodard CM, Campos BA, Kuo SH, Nirenberg MJ, Nestor MW, Zimmer M, Mosharov EV, Sulzer D, Zhou H, Paull D, Clark L, Schadt EE, Sardi SP, Rubin L, Eggan K, Brock M, Lipnick S, Rao M, Chang S, Li A, Noggle SA. iPSC-derived dopamine neurons reveal differences between monozygotic twins discordant for Parkinson's disease. Cell Rep 2014; 9:1173-82. [PMID: 25456120 DOI: 10.1016/j.celrep.2014.10.023] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 09/03/2014] [Accepted: 10/10/2014] [Indexed: 01/01/2023] Open
Abstract
Parkinson's disease (PD) has been attributed to a combination of genetic and nongenetic factors. We studied a set of monozygotic twins harboring the heterozygous glucocerebrosidase mutation (GBA N370S) but clinically discordant for PD. We applied induced pluripotent stem cell (iPSC) technology for PD disease modeling using the twins' fibroblasts to evaluate and dissect the genetic and nongenetic contributions. Utilizing fluorescence-activated cell sorting, we obtained a homogenous population of "footprint-free" iPSC-derived midbrain dopaminergic (mDA) neurons. The mDA neurons from both twins had ∼50% GBA enzymatic activity, ∼3-fold elevated α-synuclein protein levels, and a reduced capacity to synthesize and release dopamine. Interestingly, the affected twin's neurons showed an even lower dopamine level, increased monoamine oxidase B (MAO-B) expression, and impaired intrinsic network activity. Overexpression of wild-type GBA and treatment with MAO-B inhibitors normalized α-synuclein and dopamine levels, suggesting a combination therapy for the affected twin.
Collapse
Affiliation(s)
- Chris M Woodard
- The New York Stem Cell Foundation Research Institute, New York, NY 10032, USA
| | - Brian A Campos
- The New York Stem Cell Foundation Research Institute, New York, NY 10032, USA
| | - Sheng-Han Kuo
- Department of Neurology, Columbia University, New York, NY 10032, USA
| | | | - Michael W Nestor
- The New York Stem Cell Foundation Research Institute, New York, NY 10032, USA
| | - Matthew Zimmer
- The New York Stem Cell Foundation Research Institute, New York, NY 10032, USA
| | - Eugene V Mosharov
- Department of Neurology, Columbia University, New York, NY 10032, USA
| | - David Sulzer
- Department of Neurology, Columbia University, New York, NY 10032, USA; Departments of Psychiatry, Pharmacology, Columbia University, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA
| | - Hongyan Zhou
- The New York Stem Cell Foundation Research Institute, New York, NY 10032, USA
| | - Daniel Paull
- The New York Stem Cell Foundation Research Institute, New York, NY 10032, USA
| | - Lorraine Clark
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Lee Rubin
- The New York Stem Cell Foundation Research Institute, New York, NY 10032, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Kevin Eggan
- The New York Stem Cell Foundation Research Institute, New York, NY 10032, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; The Howard Hughes Medical Institute, Harvard Stem Cell Institute, Stanley Center for Psychiatric Research, Harvard University, Cambridge, MA 02138, USA
| | - Mathew Brock
- Axion Biosystems, 1819 Peachtree Road, Suite 350, Atlanta, GA 30309, USA
| | - Scott Lipnick
- The New York Stem Cell Foundation Research Institute, New York, NY 10032, USA
| | - Mahendra Rao
- The New York Stem Cell Foundation Research Institute, New York, NY 10032, USA
| | - Stephen Chang
- The New York Stem Cell Foundation Research Institute, New York, NY 10032, USA
| | - Aiqun Li
- The New York Stem Cell Foundation Research Institute, New York, NY 10032, USA.
| | - Scott A Noggle
- The New York Stem Cell Foundation Research Institute, New York, NY 10032, USA.
| |
Collapse
|
177
|
Fishbein I, Kuo YM, Giasson BI, Nussbaum RL. Augmentation of phenotype in a transgenic Parkinson mouse heterozygous for a Gaucher mutation. ACTA ACUST UNITED AC 2014; 137:3235-47. [PMID: 25351739 DOI: 10.1093/brain/awu291] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The involvement of the protein α-synuclein (SNCA) in the pathogenesis of Parkinson's disease is strongly supported by the facts that (i) missense and copy number mutations in the SNCA gene can cause inherited Parkinson's disease; and (ii) Lewy bodies in sporadic Parkinson's disease are largely composed of aggregated SNCA. Unaffected heterozygous carriers of Gaucher disease mutations have an increased risk for Parkinson's disease. As mutations in the GBA gene encoding glucocerebrosidase (GBA) are known to interfere with lysosomal protein degradation, GBA heterozygotes may demonstrate reduced lysosomal SNCA degradation, leading to increased steady-state SNCA levels and promoting its aggregation. We have created mouse models to investigate the interaction between GBA mutations and synucleinopathies. We investigated the rate of SNCA degradation in cultured primary cortical neurons from mice expressing wild-type mouse SNCA, wild-type human SNCA, or mutant A53T SNCA, in a background of either wild-type Gba or heterozygosity for the L444P GBA mutation associated with Gaucher disease. We also tested the effect of this Gaucher mutation on motor and enteric nervous system function in these transgenic animals. We found that human SNCA is stable, with a half-life of 61 h, and that the A53T mutation did not significantly affect its half-life. Heterozygosity for a naturally occurring Gaucher mutation, L444P, reduced GBA activity by 40%, reduced SNCA degradation and triggered accumulation of the protein in culture. This mutation also resulted in the exacerbation of motor and gastrointestinal deficits found in the A53T mouse model of Parkinson's disease. This study demonstrates that heterozygosity for a Gaucher disease-associated mutation in Gba interferes with SNCA degradation and contributes to its accumulation, and exacerbates the phenotype in a mouse model of Parkinson's disease.
Collapse
Affiliation(s)
- Ianai Fishbein
- 1 Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Yien-Ming Kuo
- 1 Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Benoit I Giasson
- 2 Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA
| | - Robert L Nussbaum
- 1 Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
178
|
Liou B, Haffey WD, Greis KD, Grabowski GA. The LIMP-2/SCARB2 binding motif on acid β-glucosidase: basic and applied implications for Gaucher disease and associated neurodegenerative diseases. J Biol Chem 2014; 289:30063-74. [PMID: 25202012 PMCID: PMC4208013 DOI: 10.1074/jbc.m114.593616] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 09/04/2014] [Indexed: 12/31/2022] Open
Abstract
The acid β-glucosidase (glucocerbrosidase (GCase)) binding sequence to LIMP-2 (lysosomal integral membrane protein 2), the receptor for intracellular GCase trafficking to the lysosome, has been identified. Heterologous expression of deletion constructs, the available GCase crystal structures, and binding and co-localization of identified peptides or mutant GCases were used to identify and characterize a highly conserved 11-amino acid sequence, DSPIIVDITKD, within human GCase. The binding to LIMP-2 is not dependent upon a single amino acid, but the interactions of GCase with LIMP-2 are heavily influenced by Asp(399) and the di-isoleucines, Ile(402) and Ile(403). A single alanine substitution at any of these decreases GCase binding to LIMP-2 and alters its pH-dependent binding as well as diminishing the trafficking of GCase to the lysosome and significantly increasing GCase secretion. Enterovirus 71 also binds to LIMP-2 (also known as SCARB2) on the external surface of the plasma membrane. However, the LIMP-2/SCARB2 binding sequences for enterovirus 71 and GCase are not similar, indicating that LIMP-2/SCARB2 may have multiple or overlapping binding sites with differing specificities. These findings have therapeutic implications for the production of GCase and the distribution of this enzyme that is delivered to various organs.
Collapse
Affiliation(s)
- Benjamin Liou
- From the Division of Human Genetics, Cincinnati Children's Hospital Medical Center, and the Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229 and
| | - Wendy D Haffey
- the Department of Cancer Biology, Vontz Center for Molecular Studies, University of Cincinnati Medical Center, Cincinnati, Ohio 45229
| | - Kenneth D Greis
- the Department of Cancer Biology, Vontz Center for Molecular Studies, University of Cincinnati Medical Center, Cincinnati, Ohio 45229
| | - Gregory A Grabowski
- From the Division of Human Genetics, Cincinnati Children's Hospital Medical Center, and the Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229 and
| |
Collapse
|
179
|
Richter F, Fleming SM, Watson M, Lemesre V, Pellegrino L, Ranes B, Zhu C, Mortazavi F, Mulligan CK, Sioshansi PC, Hean S, De La Rosa K, Khanna R, Flanagan J, Lockhart DJ, Wustman BA, Clark SW, Chesselet MF. A GCase chaperone improves motor function in a mouse model of synucleinopathy. Neurotherapeutics 2014; 11:840-56. [PMID: 25037721 PMCID: PMC4391384 DOI: 10.1007/s13311-014-0294-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Mutation of the lysosomal hydrolase acid-β-glucosidase (GCase), which leads to reduced GCase activity, is one of the most frequent genetic risk factors for Parkinson's disease (PD) and promotes α-synuclein accumulation in the brain, a hallmark of PD and other synucleinopathies. Whether targeting GCase pharmacologically is a valid therapeutic strategy for sporadic PD in the absence of GCase mutation is unknown. We have investigated whether increasing the stability, trafficking, and activity of wild-type GCase could be beneficial in synucleinopathies by administering the pharmacological chaperone AT2101 (afegostat-tartrate, isofagomine) to mice that overexpress human wild-type α-synuclein (Thy1-aSyn mice). AT2101 administered orally for 4 months to Thy1-aSyn mice improved motor and nonmotor function, abolished microglial inflammatory response in the substantia nigra, reduced α-synuclein immunoreactivity in nigral dopaminergic neurons, and reduced the number of small α-synuclein aggregates, while increasing the number of large α-synuclein aggregates. These data support the further investigation of pharmacological chaperones that target GCase as a therapeutic approach for sporadic PD and other synucleinopathies, even in the absence of glucocerebrosidase mutations.
Collapse
Affiliation(s)
- Franziska Richter
- />The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769 USA
| | - Sheila M. Fleming
- />The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769 USA
| | - Melanie Watson
- />The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769 USA
| | - Vincent Lemesre
- />The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769 USA
| | - Lee Pellegrino
- />Amicus Therapeutics, 1 Cedar Brook Drive, Cranbury, NJ 08512 USA
| | - Brian Ranes
- />Amicus Therapeutics, 1 Cedar Brook Drive, Cranbury, NJ 08512 USA
| | - Chunni Zhu
- />The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769 USA
| | - Farzad Mortazavi
- />The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769 USA
| | - Caitlin K. Mulligan
- />The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769 USA
| | - Pedrom C. Sioshansi
- />The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769 USA
| | - Sindalana Hean
- />The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769 USA
| | - Krystal De La Rosa
- />The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769 USA
| | - Richie Khanna
- />Amicus Therapeutics, 1 Cedar Brook Drive, Cranbury, NJ 08512 USA
| | - John Flanagan
- />Amicus Therapeutics, 1 Cedar Brook Drive, Cranbury, NJ 08512 USA
| | | | - Brandon A. Wustman
- />Amicus Therapeutics, 11099 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Sean W. Clark
- />Amicus Therapeutics, 1 Cedar Brook Drive, Cranbury, NJ 08512 USA
| | | |
Collapse
|
180
|
Swan M, Saunders-Pullman R. The association between ß-glucocerebrosidase mutations and parkinsonism. Curr Neurol Neurosci Rep 2014; 13:368. [PMID: 23812893 DOI: 10.1007/s11910-013-0368-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mutations in the ß-glucocerebrosidase gene (GBA), which encodes the lysosomal enzyme ß-glucocerebrosidase, have traditionally been implicated in Gaucher disease, an autosomal recessive lysosomal storage disorder. Yet the past two decades have yielded an explosion of epidemiological and basic-science evidence linking mutations in GBA with the development of Parkinson disease (PD) as well. Although the specific contribution of mutant GBA to the pathogenesis of parkinsonism remains unknown, evidence suggests that both loss of function and toxic gain of function by abnormal ß-glucocerebrosidase may be important, and implicates a close relationship between ß-glucocerebrosidase and α-synuclein. Furthermore, multiple lines of evidence suggest that although GBA-associated PD closely mimics idiopathic PD (IPD), it may present at a younger age, and is more frequently complicated by cognitive dysfunction. Understanding the clinical association between GBA and PD, and the relationship between ß-glucocerebrosidase and α-synuclein, may enhance understanding of the pathogenesis of IPD, improve prognostication and treatment of GBA carriers with parkinsonism, and furthermore inform therapies for IPD not due to GBA mutations.
Collapse
Affiliation(s)
- Matthew Swan
- Department of Neurology, Beth Israel Medical Center, 10 Union Square East, Suite 5K, New York, NY 10003, USA.
| | | |
Collapse
|
181
|
Emerging insights into the mechanistic link between α-synuclein and glucocerebrosidase in Parkinson's disease. Biochem Soc Trans 2014; 41:1509-12. [PMID: 24256245 DOI: 10.1042/bst20130158] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Mutations in the GBA1 gene, encoding the enzyme glucocerebrosidase, cause the lysosomal storage disorder GD (Gaucher's disease), and are associated with the development of PD (Parkinson's disease) and other Lewy body disorders. Interestingly, GBA1 variants are the most common genetic risk factor associated with PD. Although clinical studies argue a strong case towards a link between GBA1 mutations and the development of PD, mechanistic insights have been lacking. In the present article, we review recent findings that have provided some biochemical evidence to bridge this relationship, focusing on the molecular link between two proteins, α-synuclein and glucocerebrosidase, involved in PD and GD respectively.
Collapse
|
182
|
Altarescu G, Ioscovich D, Alcalay RN, Zimran A, Elstein D. α-Synuclein rs356219 polymorphisms in patients with Gaucher disease and Parkinson disease. Neurosci Lett 2014; 580:104-7. [PMID: 25111979 DOI: 10.1016/j.neulet.2014.07.051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 07/27/2014] [Accepted: 07/29/2014] [Indexed: 11/17/2022]
Abstract
Mutations in β-glucocerebrosidase, the genetic defect in Gaucher disease (GD), are an important susceptibility factor for Parkinson disease (PD). A PD effector is α-synuclein (SNCA) hypothesized to selectively interact with β-glucocerebrosidase under lysosomal conditions. SNCA polymorphism rs356219 may be associated with early-age-onset PD, common among patients with GD+PD. The objective of this study was to ascertain rs356219 genotypes of GD+PD patients. All GD+PD patients at our Gaucher referral clinic were asked to participate. A GD-only sex-, age-, GD genotype-, and enzyme therapy (ERT)-matched control was found for each GD+PD participant. Student's t-test was used (p-value <0.05 as significant). There were 14 GD+PD patients: all Ashkenazi Jewish; 11 males (78.6%); mean (range) age diagnosed GD 34.2 (5-62) years; 50% N370S homozygous; mild to moderate GD; 3 asplenic and only these have osteonecrosis; 5 received ERT; mean age (range) diagnosed PD was 57.8 (43-70) years; first PD sign was tremor in 9 (64.3%); cognitive dysfunction in all. In GD+PD, frequency for AG+GG (9) was greater than AA (5); in GD only, there was equality (7). Odds Ratio risk for PD increases with number minor alleles: but not significantly greater among GD+PD than GD only; in aggregate, there was no difference between cohorts for frequency of minor alleles. The limitation of this study is few GD+PD, albeit virtually all the GD+PD cohort >500 adult GD patients in our clinic. Nonetheless, as a foray into potential genetic GD susceptibility for a synucleinopathy, this study suggests the need for collaboration to achieve larger sample size.
Collapse
Affiliation(s)
- Gheona Altarescu
- Genetics Unit, Shaare Zedek Medical Center, Affliated with the Hadassah-Hebrew University Medical School, Jerusalem, Israel
| | - Daniel Ioscovich
- Gaucher Clinic, Shaare Zedek Medical Center, Affliated with the Hadassah-Hebrew University Medical School, Jerusalem, Israel
| | - Roy N Alcalay
- Department of Neurology and the Taub Institute, Columbia University Medical Center, New York, NY, USA
| | - Ari Zimran
- Gaucher Clinic, Shaare Zedek Medical Center, Affliated with the Hadassah-Hebrew University Medical School, Jerusalem, Israel
| | - Deborah Elstein
- Gaucher Clinic, Shaare Zedek Medical Center, Affliated with the Hadassah-Hebrew University Medical School, Jerusalem, Israel.
| |
Collapse
|
183
|
Deng H, Xiu X, Jankovic J. Genetic convergence of Parkinson's disease and lysosomal storage disorders. Mol Neurobiol 2014; 51:1554-68. [PMID: 25099932 DOI: 10.1007/s12035-014-8832-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 07/23/2014] [Indexed: 01/07/2023]
Abstract
Parkinson's disease is a common progressive neurodegenerative disorder characterized by predominant degeneration of the dopaminergic neurons in the substantia nigra pars compacta and the presence of intracellular inclusions enriched in α-synuclein, resulting in a variety motor and nonmotor symptoms. Lysosomal storage disorders are a group of disorders including Gaucher disease, Niemann-Pick disease, and neuronal ceroid lipofuscinoses caused by the defective activity of lysosomal and nonlysosomal proteins. In addition to an overlap in some clinical features between lysosomal storage disorders and Parkinson's disease, the two disorders may be also linked pathogenically. There is growing support for the notion that mutations in genes causing lysosomal storage disorders including the glucocerebrosidase gene, the sphingomyelin phosphodiesterase 1 gene, and the NPC1 gene may increase risk for developing Parkinson's disease. In this review, we discuss the recent advances in the genetic convergence of Parkinson's disease and lysosomal storage disorders, shedding new light on the understanding of shared pathogenic pathways.
Collapse
Affiliation(s)
- Hao Deng
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan, 410013, China,
| | | | | |
Collapse
|
184
|
Xu YH, Xu K, Sun Y, Liou B, Quinn B, Li RH, Xue L, Zhang W, Setchell KDR, Witte D, Grabowski GA. Multiple pathogenic proteins implicated in neuronopathic Gaucher disease mice. Hum Mol Genet 2014; 23:3943-57. [PMID: 24599400 PMCID: PMC4082362 DOI: 10.1093/hmg/ddu105] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 02/28/2014] [Accepted: 03/03/2014] [Indexed: 11/14/2022] Open
Abstract
Gaucher disease, a prevalent lysosomal storage disease (LSD), is caused by insufficient activity of acid β-glucosidase (GCase) and the resultant glucosylceramide (GC)/glucosylsphingosine (GS) accumulation in visceral organs (Type 1) and the central nervous system (Types 2 and 3). Recent clinical and genetic studies implicate a pathogenic link between Gaucher and neurodegenerative diseases. The aggregation and inclusion bodies of α-synuclein with ubiquitin are present in the brains of Gaucher disease patients and mouse models. Indirect evidence of β-amyloid pathology promoting α-synuclein fibrillation supports these pathogenic proteins as a common feature in neurodegenerative diseases. Here, multiple proteins are implicated in the pathogenesis of chronic neuronopathic Gaucher disease (nGD). Immunohistochemical and biochemical analyses showed significant amounts of β-amyloid and amyloid precursor protein (APP) aggregates in the cortex, hippocampus, stratum and substantia nigra of the nGD mice. APP aggregates were in neuronal cells and colocalized with α-synuclein signals. A majority of APP co-localized with the mitochondrial markers TOM40 and Cox IV; a small portion co-localized with the autophagy proteins, P62/LC3, and the lysosomal marker, LAMP1. In cultured wild-type brain cortical neural cells, the GCase-irreversible inhibitor, conduritol B epoxide (CBE), reproduced the APP/α-synuclein aggregation and the accumulation of GC/GS. Ultrastructural studies showed numerous larger-sized and electron-dense mitochondria in nGD cerebral cortical neural cells. Significant reductions of mitochondrial adenosine triphosphate production and oxygen consumption (28-40%) were detected in nGD brains and in CBE-treated neural cells. These studies implicate defective GCase function and GC/GS accumulation as risk factors for mitochondrial dysfunction and the multi-proteinopathies (α-synuclein-, APP- and Aβ-aggregates) in nGD.
Collapse
Affiliation(s)
- You-hai Xu
- The Division of Human Genetics and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Kui Xu
- The Division of Human Genetics and
| | - Ying Sun
- The Division of Human Genetics and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | | | | | - Rong-hua Li
- The Division of Human Genetics and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Ling Xue
- Attardi Institute of Mitochondrial Biomedicine, Wenzhou Medical College, Wenzhou, Zhejiang, PR China
| | - Wujuan Zhang
- The Division of Pathology and Laboratory, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229-3039, USA
| | - Kenneth D R Setchell
- The Division of Pathology and Laboratory, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229-3039, USA Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - David Witte
- The Division of Pathology and Laboratory, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229-3039, USA Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Gregory A Grabowski
- The Division of Human Genetics and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA Synageva BioPharma, Lexington, MA 02421, USA
| |
Collapse
|
185
|
Zigdon H, Meshcheriakova A, Futerman AH. From sheep to mice to cells: Tools for the study of the sphingolipidoses. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1841:1189-99. [DOI: 10.1016/j.bbalip.2014.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Revised: 02/24/2014] [Accepted: 02/25/2014] [Indexed: 12/12/2022]
|
186
|
Luth ES, Stavrovskaya IG, Bartels T, Kristal BS, Selkoe DJ. Soluble, prefibrillar α-synuclein oligomers promote complex I-dependent, Ca2+-induced mitochondrial dysfunction. J Biol Chem 2014; 289:21490-507. [PMID: 24942732 DOI: 10.1074/jbc.m113.545749] [Citation(s) in RCA: 215] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
α-Synuclein (αSyn) aggregation and mitochondrial dysfunction both contribute to the pathogenesis of Parkinson disease (PD). Although recent studies have suggested that mitochondrial association of αSyn may disrupt mitochondrial function, it is unclear what aggregation state of αSyn is most damaging to mitochondria and what conditions promote or inhibit the effect of toxic αSyn species. Because the neuronal populations most vulnerable in PD are characterized by large cytosolic Ca(2+) oscillations that burden mitochondria, we examined mitochondrial Ca(2+) stress in an in vitro system comprising isolated mitochondria and purified recombinant human αSyn in various aggregation states. Using fluorimetry to simultaneously measure four mitochondrial parameters, we observed that soluble, prefibrillar αSyn oligomers, but not monomeric or fibrillar αSyn, decreased the retention time of exogenously added Ca(2+), promoted Ca(2+)-induced mitochondrial swelling and depolarization, and accelerated cytochrome c release. Inhibition of the permeability transition pore rescued these αSyn-induced changes in mitochondrial parameters. Interestingly, the mitotoxic effects of αSyn were specifically dependent upon both electron flow through complex I and mitochondrial uptake of exogenous Ca(2+). Our results suggest that soluble prefibrillar αSyn oligomers recapitulate several mitochondrial phenotypes previously observed in animal and cell models of PD: complex I dysfunction, altered membrane potential, disrupted Ca(2+) homeostasis, and enhanced cytochrome c release. These data reveal how the association of oligomeric αSyn with mitochondria can be detrimental to the function of cells with high Ca(2+)-handling requirements.
Collapse
Affiliation(s)
- Eric S Luth
- From the Center for Neurologic Diseases, Department of Neurology, and
| | - Irina G Stavrovskaya
- Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Tim Bartels
- From the Center for Neurologic Diseases, Department of Neurology, and
| | - Bruce S Kristal
- Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Dennis J Selkoe
- From the Center for Neurologic Diseases, Department of Neurology, and
| |
Collapse
|
187
|
Alcalay RN, Dinur T, Quinn T, Sakanaka K, Levy O, Waters C, Fahn S, Dorovski T, Chung WK, Pauciulo M, Nichols W, Rana HQ, Balwani M, Bier L, Elstein D, Zimran A. Comparison of Parkinson risk in Ashkenazi Jewish patients with Gaucher disease and GBA heterozygotes. JAMA Neurol 2014; 71:752-7. [PMID: 24756352 PMCID: PMC4082726 DOI: 10.1001/jamaneurol.2014.313] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
IMPORTANCE Information on age-specific risk for Parkinson disease (PD) in patients with Gaucher disease (GD) and glucocerebrosidase (GBA) heterozygotes is important for understanding the pathophysiology of the genetic association and for counseling these populations. OBJECTIVE To estimate the age-specific risk for PD in Ashkenazi Jewish patients with type 1 GD and in GBA heterozygotes. DESIGN, SETTING, AND PARTICIPANTS The study included patients with GD from 2 tertiary centers, Shaare Zedek Medical Center, Jerusalem, Israel (n = 332) and Mount Sinai School of Medicine, New York, New York (n = 95). GBA noncarrier non-PD spouse control participants were recruited at the Center for Parkinson's Disease at Columbia University, New York (n = 77). All participants were Ashekanzi Jewish and most patients (98.1%) with GD carried at least 1 N370S mutation. MAIN OUTCOMES AND MEASURES The main outcome measure was a diagnosis of PD. Diagnosis was established in patients with GD on examination. We used a validated family history interview that identifies PD with a sensitivity of 95.5% and specificity of 96.2% to identify PD in family members. Kaplan-Meier survival curves were used to estimate age-specific PD risk among patients with GD (n = 427), among their parents who are obligate GBA mutation carriers (heterozygotes, n = 694), and among noncarriers (parents of non-PD, non-GD control participants, n = 154). The age-specific risk was compared among groups using the log-rank test. RESULTS Among those who developed PD, patients with GD had a younger age at onset than GBA heterozygotes (mean, 54.2 vs 65.2 years, respectively; P = .003). Estimated age-specific risk for PD at 60 and 80 years of age was 4.7% and 9.1% among patients with GD, 1.5% and 7.7% among heterozygotes, and 0.7% and 2.1% among noncarriers, respectively. The risk for PD was higher in patients with GD than noncarriers (P = .008, log-rank test) and in heterozygotes than noncarriers (P = .03, log-rank test), but it did not reach statistical significance between patients with GD and GBA heterozygotes (P = .07, log-rank test). CONCLUSIONS AND RELEVANCE Patients with GD and GBA heterozygotes have an increased age-specific risk for PD compared with control individuals, with a similar magnitude of PD risk by 80 years of age; however, the number of mutant alleles may play an important role in age at PD onset.
Collapse
Affiliation(s)
- Roy N. Alcalay
- Department of Neurology and the Taub Institute, Columbia University Medical Center, New York, NY
| | - Tama Dinur
- Gaucher Clinic, Shaare Zedek Medical Center, Jerusalem, affiliated with the Hebrew University – Hadassah Medical School, Ein Karem, Israel
| | - Timothy Quinn
- Department of Neurology and the Taub Institute, Columbia University Medical Center, New York, NY
| | - Karina Sakanaka
- Department of Neurology and the Taub Institute, Columbia University Medical Center, New York, NY
| | - Oren Levy
- Department of Neurology and the Taub Institute, Columbia University Medical Center, New York, NY
| | - Cheryl Waters
- Department of Neurology and the Taub Institute, Columbia University Medical Center, New York, NY
| | - Stanley Fahn
- Department of Neurology and the Taub Institute, Columbia University Medical Center, New York, NY
| | - Tsvyatko Dorovski
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Wendy K Chung
- Departments of Pediatrics and Medicine Columbia University Medical Center, New York, NY, USA
| | - Michael Pauciulo
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - William Nichols
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Huma Q. Rana
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY
| | - Manisha Balwani
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY
| | - Louise Bier
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY
| | - Deborah Elstein
- Gaucher Clinic, Shaare Zedek Medical Center, Jerusalem, affiliated with the Hebrew University – Hadassah Medical School, Ein Karem, Israel
| | - Ari Zimran
- Gaucher Clinic, Shaare Zedek Medical Center, Jerusalem, affiliated with the Hebrew University – Hadassah Medical School, Ein Karem, Israel
| |
Collapse
|
188
|
Brooks P, Tagle DA, Groft S. Expanding rare disease drug trials based on shared molecular etiology. Nat Biotechnol 2014; 32:515-8. [PMID: 24911489 PMCID: PMC4548299 DOI: 10.1038/nbt.2924] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- P.J. Brooks
- Office of Rare Diseases Research, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 6701 Democracy Blvd., Bethesda, MD 20892-7518
| | - Danilo A. Tagle
- Office of Special Initiatives, Office of the Director, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 6701 Democracy Blvd., Bethesda, MD 20892-7518
| | - Steve Groft
- Office of Rare Diseases Research, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 6701 Democracy Blvd., Bethesda, MD 20892-7518
| |
Collapse
|
189
|
Boyd RE, Valenzano KJ. Correction of lysosomal dysfunction as a therapeutic strategy for neurodegenerative diseases. Bioorg Med Chem Lett 2014; 24:3001-5. [PMID: 24894562 DOI: 10.1016/j.bmcl.2014.04.108] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 04/24/2014] [Accepted: 04/25/2014] [Indexed: 11/25/2022]
Abstract
Mutations in the gene that encodes the lysosomal enzyme acid β-glucosidase lead to reduced cellular activity and accumulation of glycosphingolipid substrates, biochemical hallmarks of the lysosomal storage disorder Gaucher disease (GD). Recently such mutations have been identified as risk factors for Parkinson's disease (PD) and related disorders. Both gain-of-function (due to toxic cellular accumulation of mutant enzyme) and loss-of-function (due to accumulation of lipid substrates) hypotheses have been put forth to address the biochemical link between GD and PD. Similarly, links between Alzheimer's disease and other lysosomal enzyme deficiencies have begun to emerge. The use of pharmacological chaperones to restore the cellular trafficking and activity of mutant lysosomal enzymes may offer a novel approach to treat these debilitating neurodegenerative diseases.
Collapse
Affiliation(s)
- Robert E Boyd
- Amicus Therapeutics, 1 Cedar Brook Dr., Cranbury, NJ 08512, United States.
| | | |
Collapse
|
190
|
Sybertz E, Krainc D. Development of targeted therapies for Parkinson's disease and related synucleinopathies. J Lipid Res 2014; 55:1996-2003. [PMID: 24668939 DOI: 10.1194/jlr.r047381] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Therapeutic efforts in neurodegenerative diseases have been very challenging, particularly due to a lack of validated and mechanism-based therapeutic targets and biomarkers. The basic idea underlying the novel therapeutic approaches reviewed here is that by exploring the molecular basis of neurodegeneration in a rare lysosomal disease such as Gaucher's disease (GD), new molecular targets will be identified for therapeutic development in common synucleinopathies. Accumulation of α-synuclein plays a key role in the pathogenesis of Parkinson's disease (PD) and other synucleinopathies, suggesting that improved clearance of α-synuclein may be of therapeutic benefit. To achieve this goal, it is important to identify specific mechanisms and targets involved in the clearance of α-synuclein. Recent discovery of clinical, genetic, and pathological linkage between GD and PD offers a unique opportunity to examine lysosomal glucocerebrosidase, an enzyme mutated in GD, for development of targeted therapies in synucleinopathies. While modulation of glucocerebrosidase and glycolipid metabolism offers a viable approach to treating disorders associated with synuclein accumulation, the compounds described to date either lack the ability to penetrate the CNS or have off-target effects that may counteract or limit their capabilities to mediate the desired pharmacological action. However, recent emergence of selective inhibitors of glycosphingolipid biosynthesis and noninhibitory pharmacological chaperones of glycosphingolipid processing enzymes that gain access to the CNS provide a novel approach that may overcome some of the limitations of compounds reported to date. These new strategies may allow for development of targeted treatments for synucleinopathies that affect both children and adults.
Collapse
Affiliation(s)
| | - Dimitri Krainc
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
191
|
Nelson MP, Tse TE, O’Quinn DB, Percival SM, Jaimes EA, Warnock DG, Shacka JJ. Autophagy-lysosome pathway associated neuropathology and axonal degeneration in the brains of alpha-galactosidase A-deficient mice. Acta Neuropathol Commun 2014; 2:20. [PMID: 24529306 PMCID: PMC3933238 DOI: 10.1186/2051-5960-2-20] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 02/01/2014] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Mutations in the gene for alpha-galactosidase A result in Fabry disease, a rare, X-linked lysosomal storage disorder characterized by a loss of alpha-galactosidase A enzymatic activity. The resultant accumulation of glycosphingolipids throughout the body leads to widespread vasculopathy with particular detriment to the kidneys, heart and nervous system. Disruption in the autophagy-lysosome pathway has been documented previously in Fabry disease but its relative contribution to nervous system pathology in Fabry disease is unknown. Using an experimental mouse model of Fabry disease, alpha-galactosidase A deficiency, we examined brain pathology in 20-24 month old mice with particular emphasis on the autophagy-lysosome pathway. RESULTS Alpha-galactosidase A-deficient mouse brains exhibited enhanced punctate perinuclear immunoreactivity for the autophagy marker microtubule-associated protein light-chain 3 (LC3) in the parenchyma of several brain regions, as well as enhanced parenchymal and vascular immunoreactivity for lysosome-associated membrane protein-1 (LAMP-1). Ultrastructural analysis revealed endothelial cell inclusions with electron densities and a pronounced accumulation of electron-dense lipopigment. The pons of alpha-galactosidase A-deficient mice in particular exhibited a striking neuropathological phenotype, including the presence of large, swollen axonal spheroids indicating axonal degeneration, in addition to large interstitial aggregates positive for phosphorylated alpha-synuclein that co-localized with the axonal spheroids. Double-label immunofluorescence revealed co-localization of phosphorylated alpha-synuclein aggregates with ubiquitin and LC3. CONCLUSION Together these findings indicate widespread neuropathology and focused axonal neurodegeneration in alpha-galactosidase A-deficient mouse brain in association with disruption of the autophagy-lysosome pathway, and provide the basis for future mechanistic assessment of the contribution of the autophagy-lysosome pathway to this histologic phenotype.
Collapse
Affiliation(s)
- Michael P Nelson
- />Department Pathology, Neuropathology Division, University of Alabama at Birmingham, Birmingham, AL USA
| | - Tonia E Tse
- />Department Pathology, Neuropathology Division, University of Alabama at Birmingham, Birmingham, AL USA
- />Birmingham VA Medical Center, Birmingham, AL USA
| | - Darrel B O’Quinn
- />Department Pathology, Anatomic Pathology Division, University of Alabama at Birmingham, Birmingham, AL USA
| | - Stefanie M Percival
- />Department Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Edgar A Jaimes
- />Birmingham VA Medical Center, Birmingham, AL USA
- />Department Medicine, Nephrology Division, University of Alabama at Birmingham, Birmingham, AL USA
| | - David G Warnock
- />Department Medicine, Nephrology Division, University of Alabama at Birmingham, Birmingham, AL USA
| | - John J Shacka
- />Department Pathology, Neuropathology Division, University of Alabama at Birmingham, Birmingham, AL USA
- />Birmingham VA Medical Center, Birmingham, AL USA
| |
Collapse
|
192
|
Siebert M, Sidransky E, Westbroek W. Glucocerebrosidase is shaking up the synucleinopathies. ACTA ACUST UNITED AC 2014; 137:1304-22. [PMID: 24531622 DOI: 10.1093/brain/awu002] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The lysosomal enzyme glucocerebrosidase, encoded by the glucocerebrosidase gene, is involved in the breakdown of glucocerebroside into glucose and ceramide. Lysosomal build-up of the substrate glucocerebroside occurs in cells of the reticulo-endothelial system in patients with Gaucher disease, a rare lysosomal storage disorder caused by the recessively inherited deficiency of glucocerebrosidase. Gaucher disease has a broad clinical phenotypic spectrum, divided into non-neuronopathic and neuronopathic forms. Like many monogenic diseases, the correlation between clinical manifestations and molecular genotype is not straightforward. There is now a well-established clinical association between mutations in the glucocerebrosidase gene and the development of more prevalent multifactorial disorders including Parkinson's disease and other synucleinopathies. In this review we discuss recent studies advancing our understanding of the cellular relationship between glucocerebrosidase and α-synuclein, the potential impact of established and emerging therapeutics for Gaucher disease for the treatment of the synucleinopathies, and the role of lysosomal pathways in the pathogenesis of these neurodegenerative disorders.
Collapse
Affiliation(s)
- Marina Siebert
- 1 Section on Molecular Neurogenetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Building 35 Room 1A213, 35 Convent Drive, MSC 3708, Bethesda, MD 20892-3708, USA
| | | | | |
Collapse
|
193
|
Ginns EI, Mak SKK, Ko N, Karlgren J, Akbarian S, Chou VP, Guo Y, Lim A, Samuelsson S, LaMarca ML, Vazquez-DeRose J, Manning-Boğ AB. Neuroinflammation and α-synuclein accumulation in response to glucocerebrosidase deficiency are accompanied by synaptic dysfunction. Mol Genet Metab 2014; 111:152-62. [PMID: 24388731 DOI: 10.1016/j.ymgme.2013.12.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/04/2013] [Accepted: 12/05/2013] [Indexed: 12/28/2022]
Abstract
Clinical, epidemiological and experimental studies confirm a connection between the common degenerative movement disorder Parkinson's disease (PD) that affects over 1 million individuals, and Gaucher disease, the most prevalent lysosomal storage disorder. Recently, human imaging studies have implicated impaired striatal dopaminergic neurotransmission in early PD pathogenesis in the context of Gaucher disease mutations, but the underlying mechanisms have yet to be characterized. In this report we describe and characterize two novel long-lived transgenic mouse models of Gba deficiency, along with a subchronic conduritol-ß-epoxide (CBE) exposure paradigm. All three murine models revealed striking glial activation within nigrostriatal pathways, accompanied by abnormal α-synuclein accumulation. Importantly, the CBE-induced, pharmacological Gaucher mouse model replicated this change in dopamine neurotransmission, revealing a markedly reduced evoked striatal dopamine release (approximately 2-fold) that indicates synaptic dysfunction. Other changes in synaptic plasticity markers, including microRNA profile and a 24.9% reduction in post-synaptic density size, were concomitant with diminished evoked dopamine release following CBE exposure. These studies afford new insights into the mechanisms underlying the Parkinson's-Gaucher disease connection, and into the physiological impact of related abnormal α-synuclein accumulation and neuroinflammation on nigrostriatal dopaminergic neurotransmission.
Collapse
Affiliation(s)
- Edward I Ginns
- Lysosomal Disorders Treatment and Research Program, Clinical Labs, University of Massachusetts Medical School, Worcester, MA 01545, USA; Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA 01545, USA; Clinical Neuroscience Branch, IRP, NIMH, Bethesda, MD 20892, USA
| | - Sally K-K Mak
- Center for Health Sciences, Biosciences Division, SRI International, Menlo Park, CA 94025, USA
| | - Novie Ko
- Center for Health Sciences, Biosciences Division, SRI International, Menlo Park, CA 94025, USA
| | - Juliane Karlgren
- Lysosomal Disorders Treatment and Research Program, Clinical Labs, University of Massachusetts Medical School, Worcester, MA 01545, USA
| | - Schahram Akbarian
- Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA 01545, USA
| | - Vivian P Chou
- Center for Health Sciences, Biosciences Division, SRI International, Menlo Park, CA 94025, USA
| | - Yin Guo
- Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA 01545, USA
| | - Arlene Lim
- Lysosomal Disorders Treatment and Research Program, Clinical Labs, University of Massachusetts Medical School, Worcester, MA 01545, USA; Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA 01545, USA
| | - Steven Samuelsson
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA 94025, USA
| | - Mary L LaMarca
- Clinical Neuroscience Branch, IRP, NIMH, Bethesda, MD 20892, USA
| | | | - Amy B Manning-Boğ
- Center for Health Sciences, Biosciences Division, SRI International, Menlo Park, CA 94025, USA.
| |
Collapse
|
194
|
Murphy KE, Gysbers AM, Abbott SK, Tayebi N, Kim WS, Sidransky E, Cooper A, Garner B, Halliday GM. Reduced glucocerebrosidase is associated with increased α-synuclein in sporadic Parkinson's disease. ACTA ACUST UNITED AC 2014; 137:834-48. [PMID: 24477431 DOI: 10.1093/brain/awt367] [Citation(s) in RCA: 382] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Heterozygous mutations in GBA1, the gene encoding lysosomal glucocerebrosidase, are the most frequent known genetic risk factor for Parkinson's disease. Reduced glucocerebrosidase and α-synuclein accumulation are directly related in cell models of Parkinson's disease. We investigated relationships between Parkinson's disease-specific glucocerebrosidase deficits, glucocerebrosidase-related pathways, and α-synuclein levels in brain tissue from subjects with sporadic Parkinson's disease without GBA1 mutations. Brain regions with and without a Parkinson's disease-related increase in α-synuclein levels were assessed in autopsy samples from subjects with sporadic Parkinson's disease (n = 19) and age- and post-mortem delay-matched controls (n = 10). Levels of glucocerebrosidase, α-synuclein and related lysosomal and autophagic proteins were assessed by western blotting. Glucocerebrosidase enzyme activity was measured using a fluorimetric assay, and glucocerebrosidase and α-synuclein messenger RNA expression determined by quantitative polymerase chain reaction. Related sphingolipids were analysed by mass spectrometry. Multivariate statistical analyses were performed to identify differences between disease groups and regions, with non-parametric correlations used to identify relationships between variables. Glucocerebrosidase protein levels and enzyme activity were selectively reduced in the early stages of Parkinson's disease in regions with increased α-synuclein levels although limited inclusion formation, whereas GBA1 messenger RNA expression was non-selectively reduced in Parkinson's disease. The selective loss of lysosomal glucocerebrosidase was directly related to reduced lysosomal chaperone-mediated autophagy, increased α-synuclein and decreased ceramide. Glucocerebrosidase deficits in sporadic Parkinson's disease are related to the abnormal accumulation of α-synuclein and are associated with substantial alterations in lysosomal chaperone-mediated autophagy pathways and lipid metabolism. Our data suggest that the early selective Parkinson's disease changes are likely a result of the redistribution of cellular membrane proteins leading to a chronic reduction in lysosome function in brain regions vulnerable to Parkinson's disease pathology.
Collapse
Affiliation(s)
- Karen E Murphy
- 1 Neuroscience Research Australia, Sydney, NSW 2031, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
195
|
Parnetti L, Chiasserini D, Persichetti E, Eusebi P, Varghese S, Qureshi MM, Dardis A, Deganuto M, De Carlo C, Castrioto A, Balducci C, Paciotti S, Tambasco N, Bembi B, Bonanni L, Onofrj M, Rossi A, Beccari T, El-Agnaf O, Calabresi P. Cerebrospinal fluid lysosomal enzymes and alpha-synuclein in Parkinson's disease. Mov Disord 2014; 29:1019-27. [PMID: 24436092 PMCID: PMC4282452 DOI: 10.1002/mds.25772] [Citation(s) in RCA: 198] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 11/04/2013] [Accepted: 11/12/2013] [Indexed: 11/11/2022] Open
Abstract
To assess the discriminating power of multiple cerebrospinal fluid (CSF) biomarkers for Parkinson's disease (PD), we measured several proteins playing an important role in the disease pathogenesis. The activities of β-glucocerebrosidase and other lysosomal enzymes, together with total and oligomeric α-synuclein, and total and phosphorylated tau, were thus assessed in CSF of 71 PD patients and compared to 45 neurological controls. Activities of β-glucocerebrosidase, β-mannosidase, β-hexosaminidase, and β-galactosidase were measured with established enzymatic assays, while α-synuclein and tau biomarkers were evaluated with immunoassays. A subset of PD patients (n = 44) was also screened for mutations in the β-glucocerebrosidase-encoding gene (GBA1). In the PD group, β-glucocerebrosidase activity was reduced (P < 0.05) and patients at earlier stages showed lower enzymatic activity (P < 0.05); conversely, β-hexosaminidase activity was significantly increased (P < 0.05). Eight PD patients (18%) presented GBA1 sequence variations; 3 of them were heterozygous for the N370S mutation. Levels of total α-synuclein were significantly reduced (P < 0.05) in PD, in contrast to increased levels of α-synuclein oligomers, with a higher oligomeric/total α-synuclein ratio in PD patients when compared with controls (P < 0.001). A combination of β-glucocerebrosidase activity, oligomeric/total α-synuclein ratio, and age gave the best performance in discriminating PD from neurological controls (sensitivity 82%; specificity 71%, area under the receiver operating characteristic curve = 0.87). These results demonstrate the possibility of detecting lysosomal dysfunction in CSF and further support the need to combine different biomarkers for improving the diagnostic accuracy of PD.
Collapse
Affiliation(s)
- Lucilla Parnetti
- Clinica Neurologica, Università degli Studi di Perugia, Perugia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
196
|
Rempel BP, Withers SG. Phosphodiesters serve as potentially tunable aglycones for fluoro sugar inactivators of retaining β-glycosidases. Org Biomol Chem 2014; 12:2592-5. [DOI: 10.1039/c4ob00235k] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
2-Deoxy-2-fluoroglycosides were synthesised and tested as covalent glycosidase inactivators. β-d-Gluco-, -manno- and -galacto-configured benzyl-benzylphosphonate derivatives efficiently inactivate β-gluco-, β-manno- and β-galactosidases, while α-gluco- and α-manno-configured phosphate and phosphonate derivatives instead served as slow substrates for their cognate α-glycosidases.
Collapse
Affiliation(s)
- B. P. Rempel
- Department of Chemistry
- University of British Columbia
- Vancouver, Canada
| | - S. G. Withers
- Department of Chemistry
- University of British Columbia
- Vancouver, Canada
| |
Collapse
|
197
|
Abstract
Parkinson disease (PD) is the second most common neurodegenerative disease after Alzheimer disease with a lifetime risk in the UK population of almost 5%. An association between PD and Gaucher disease (GD) derived from the observation that GD patients and their heterozygous carrier relatives were at increased risk of PD. GD is an autosomal recessive lysosomal storage disorder caused by homozygous mutations in the gene encoding glucocerebrosidase (GBA). Approximately 5%-10% of PD patients have GBA mutations, making these mutations numerically the most important genetic predisposing risk factor for the development of PD identified to date. GBA mutations result in a phenotype that is virtually indistinguishable clinically, pharmacologically, and pathologically from sporadic PD, except GBA mutations result in a slightly earlier age of onset and more frequent cognitive impairment among PD patients. The mechanisms by which GBA mutations result in PD are not yet understood. Both reduced glucocerebrosidase enzyme (GCase) activity with lysosomal dysfunction, and unfolded protein response (UPR) with endoplasmic reticulum-associated degradation (ERAD) and stress are considered contributory.
Collapse
Affiliation(s)
- Michelle S Beavan
- Department of Clinical Neurosciences, University College London Institute of Neurology , London NW3 2PF , United Kingdom
| | | |
Collapse
|
198
|
Yap TL, Gruschus JM, Velayati A, Sidransky E, Lee JC. Saposin C protects glucocerebrosidase against α-synuclein inhibition. Biochemistry 2013; 52:7161-3. [PMID: 24070323 PMCID: PMC3833811 DOI: 10.1021/bi401191v] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Mutations in GBA1, the gene for glucocerebrosidase (GCase), are genetic risk factors for Parkinson disease (PD). α-Synuclein (α-Syn), a protein implicated in PD, interacts with GCase and efficiently inhibits enzyme activity. GCase deficiency causes the lysosomal storage disorder Gaucher disease (GD). We show that saposin C (Sap C), a protein vital for GCase activity in vivo, protects GCase against α-syn inhibition. Using nuclear magnetic resonance spectroscopy, site-specific fluorescence, and Förster energy transfer probes, Sap C was observed to displace α-syn from GCase in solution and on lipid vesicles. Our results suggest that Sap C might play a crucial role in GD-related PD.
Collapse
Affiliation(s)
- Thai Leong Yap
- Laboratory of Molecular Biophysics, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland 20892, United States
| | - James M. Gruschus
- Laboratory of Molecular Biophysics, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland 20892, United States
| | - Arash Velayati
- Section on Molecular Neurogenetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Ellen Sidransky
- Section on Molecular Neurogenetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Jennifer C. Lee
- Laboratory of Molecular Biophysics, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland 20892, United States
| |
Collapse
|
199
|
Li Y, Sekine T, Funayama M, Li L, Yoshino H, Nishioka K, Tomiyama H, Hattori N. Clinicogenetic study of GBA mutations in patients with familial Parkinson's disease. Neurobiol Aging 2013; 35:935.e3-8. [PMID: 24126159 DOI: 10.1016/j.neurobiolaging.2013.09.019] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 09/11/2013] [Accepted: 09/12/2013] [Indexed: 12/13/2022]
Abstract
The glucocerebrosidase gene (GBA) is a known risk factor of Parkinson's disease (PD). We sequenced entire coding exons and exon/intron boundaries of GBA in 147 Japanese familial PD (FPD) patients from 144 families and 100 unrelated control subjects. Twenty-seven of 144 (18.8%) of index patients were heterozygous for known Gaucher disease mutations, suggesting that GBA heterozygous mutations are strongly associated with FPD (odds ratio = 22.9, 95% confidence interval = 3.1-171.2). The frequency was significantly higher in autosomal dominant PD (ADPD) compared with autosomal recessive PD. According to clinical assessments, PD patients with GBA mutations exhibited typical manifestations of PD or dementia with Lewy bodies (DLB), such as L-dopa responsive parkinsonism with psychiatric problems and/or cognitive decline. Interestingly, they also presented with reduced myocardial (123)I-metaiodobenzylguanidine uptake. Our findings suggest that heterozygous GBA mutations are strong risk factors in FPD, especially for autosomal dominant PD. Some patients with GBA heterozygous mutations develop clinical features of DLB. We speculate that GBA dysfunction may promote Lewy body formation, resulting in more severe PD or DLB phenotypes that are inherited in families.
Collapse
Affiliation(s)
- Yuanzhe Li
- Research Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo, Japan; Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Takeshi Sekine
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Manabu Funayama
- Research Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo, Japan; Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Lin Li
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Hiroyo Yoshino
- Research Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo, Japan
| | - Kenya Nishioka
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Hiroyuki Tomiyama
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan; Department of Neuroscience for Neurodegenerative Disorders, Juntendo University School of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Research Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo, Japan; Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan; Department of Neuroscience for Neurodegenerative Disorders, Juntendo University School of Medicine, Tokyo, Japan.
| |
Collapse
|
200
|
Ma L, Wei L, Wu F, Hu Z, Liu Z, Yuan W. Advances with microRNAs in Parkinson's disease research. DRUG DESIGN DEVELOPMENT AND THERAPY 2013; 7:1103-13. [PMID: 24109179 PMCID: PMC3792848 DOI: 10.2147/dddt.s48500] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is the second-most common age-dependent neurodegenerative disorder and is caused by severe degeneration of dopaminergic neurons in the substantia nigra pars compacta. Unfortunately, current treatment only targets symptoms and involves dopamine replacement therapy, which does not counteract progressive degeneration. MicroRNAs (miRNAs) are a class of small RNA molecules implicated in post-transcriptional regulation of gene expression during development. Recent studies show that miRNAs are playing an important role in the pathophysiology of PD. miRNA-based therapy is a powerful tool with which to study gene function, investigate the mechanism of the disease, and validate drug targets. In this review, we focus on the recent advances of the use of miRNAs in the pathogenesis of PD.
Collapse
Affiliation(s)
- Liuqing Ma
- Department of Neurology, Xinhua Hospital Affiliated with Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China ; School of Pharmacy, Shanghai JiaoTong University, Shanghai, People's Republic of China
| | | | | | | | | | | |
Collapse
|