351
|
Rockenstein E, Clarke J, Viel C, Panarello N, Treleaven CM, Kim C, Spencer B, Adame A, Park H, Dodge JC, Cheng SH, Shihabuddin LS, Masliah E, Sardi SP. Glucocerebrosidase modulates cognitive and motor activities in murine models of Parkinson's disease. Hum Mol Genet 2016; 25:2645-2660. [PMID: 27126635 PMCID: PMC5181635 DOI: 10.1093/hmg/ddw124] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 04/08/2016] [Accepted: 04/18/2016] [Indexed: 12/11/2022] Open
Abstract
Mutations in GBA1, the gene encoding glucocerebrosidase, are associated with an enhanced risk of developing synucleinopathies such as Parkinson’s disease (PD) and dementia with Lewy bodies. A higher prevalence and increased severity of motor and non-motor symptoms is observed in PD patients harboring mutant GBA1 alleles, suggesting a link between the gene or gene product and disease development. Interestingly, PD patients without mutations in GBA1 also exhibit lower levels of glucocerebrosidase activity in the central nervous system (CNS), implicating this lysosomal enzyme in disease pathogenesis. Here, we investigated whether modulation of glucocerebrosidase activity in murine models of synucleinopathy (expressing wild type Gba1) affected α-synuclein accumulation and behavioral phenotypes. Partial inhibition of glucocerebrosidase activity in PrP-A53T-SNCA mice using the covalent inhibitor conduritol-B-epoxide induced a profound increase in soluble α-synuclein in the CNS and exacerbated cognitive and motor deficits. Conversely, augmenting glucocerebrosidase activity in the Thy1-SNCA mouse model of PD delayed the progression of synucleinopathy. Adeno-associated virus-mediated expression of glucocerebrosidase in the Thy1-SNCA mouse striatum led to decrease in the levels of the proteinase K-resistant fraction of α-synuclein, amelioration of behavioral aberrations and protection from loss of striatal dopaminergic markers. These data indicate that increasing glucocerebrosidase activity can influence α-synuclein homeostasis, thereby reducing the progression of synucleinopathies. This study provides robust in vivo evidence that augmentation of CNS glucocerebrosidase activity is a potential therapeutic strategy for PD, regardless of the mutation status of GBA1.
Collapse
Affiliation(s)
- Edward Rockenstein
- Neuroscience Department, University of California San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | - Changyoun Kim
- Neuroscience Department, University of California San Diego, La Jolla, CA 92093, USA
| | - Brian Spencer
- Neuroscience Department, University of California San Diego, La Jolla, CA 92093, USA
| | - Anthony Adame
- Neuroscience Department, University of California San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | - E Masliah
- Neuroscience Department, University of California San Diego, La Jolla, CA 92093, USA.,Pathology Department, University of California San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
352
|
Jung O, Patnaik S, Marugan J, Sidransky E, Westbroek W. Progress and potential of non-inhibitory small molecule chaperones for the treatment of Gaucher disease and its implications for Parkinson disease. Expert Rev Proteomics 2016; 13:471-9. [PMID: 27098312 DOI: 10.1080/14789450.2016.1174583] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Gaucher disease, caused by pathological mutations GBA1, encodes the lysosome-resident enzyme glucocerebrosidase, which cleaves glucosylceramide into glucose and ceramide. In Gaucher disease, glucocerebrosidase deficiency leads to lysosomal accumulation of substrate, primarily in cells of the reticulo-endothelial system. Gaucher disease has broad clinical heterogeneity, and mutations in GBA1 are a risk factor for the development of different synucleinopathies. Insights into the cell biology and biochemistry of glucocerebrosidase have led to new therapeutic approaches for Gaucher disease including small chemical chaperones. Such chaperones facilitate proper enzyme folding and translocation to lysosomes, thereby preventing premature breakdown of the enzyme in the proteasome. This review discusses recent progress in developing chemical chaperones as a therapy for Gaucher disease, with implications for the treatment of synucleinopathies. It focuses on the development of non-inhibitory glucocerebrosidase chaperones and their therapeutic advantages over inhibitory chaperones, as well as the challenges involved in identifying and validating chemical chaperones.
Collapse
Affiliation(s)
- Olive Jung
- a Section on Molecular Neurogenetics, Medical Genetics Branch , National Human Genome Research Institute, NIH , Bethesda , MD , USA
| | - Samarjit Patnaik
- b National Center for Advancing Translational Sciences , National Institutes of Health , Bethesda , MD , USA
| | - Juan Marugan
- b National Center for Advancing Translational Sciences , National Institutes of Health , Bethesda , MD , USA
| | - Ellen Sidransky
- a Section on Molecular Neurogenetics, Medical Genetics Branch , National Human Genome Research Institute, NIH , Bethesda , MD , USA
| | - Wendy Westbroek
- a Section on Molecular Neurogenetics, Medical Genetics Branch , National Human Genome Research Institute, NIH , Bethesda , MD , USA
| |
Collapse
|
353
|
Glucocerebrosidase Deficiency in Drosophila Results in α-Synuclein-Independent Protein Aggregation and Neurodegeneration. PLoS Genet 2016; 12:e1005944. [PMID: 27019408 PMCID: PMC4809718 DOI: 10.1371/journal.pgen.1005944] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 02/25/2016] [Indexed: 02/07/2023] Open
Abstract
Mutations in the glucosidase, beta, acid (GBA1) gene cause Gaucher’s disease, and are the most common genetic risk factor for Parkinson’s disease (PD) and dementia with Lewy bodies (DLB) excluding variants of low penetrance. Because α-synuclein-containing neuronal aggregates are a defining feature of PD and DLB, it is widely believed that mutations in GBA1 act by enhancing α-synuclein toxicity. To explore this hypothesis, we deleted the Drosophila GBA1 homolog, dGBA1b, and compared the phenotypes of dGBA1b mutants in the presence and absence of α-synuclein expression. Homozygous dGBA1b mutants exhibit shortened lifespan, locomotor and memory deficits, neurodegeneration, and dramatically increased accumulation of ubiquitinated protein aggregates that are normally degraded through an autophagic mechanism. Ectopic expression of human α-synuclein in dGBA1b mutants resulted in a mild enhancement of dopaminergic neuron loss and increased α-synuclein aggregation relative to controls. However, α-synuclein expression did not substantially enhance other dGBA1b mutant phenotypes. Our findings indicate that dGBA1b plays an important role in the metabolism of protein aggregates, but that the deleterious consequences of mutations in dGBA1b are largely independent of α-synuclein. Future work with dGBA1b mutants should reveal the mechanism by which mutations in dGBA1b lead to accumulation of protein aggregates, and the potential influence of this protein aggregation on neuronal integrity. Mutations in the glucosidase, beta, acid (GBA1) gene cause Gaucher’s disease (GD), a lysosomal storage disease that includes neurodegenerative phenotypes. Recently, mutations in GBA1 were identified as the strongest genetic risk factor for Parkinson’s disease (PD) and dementia with Lewy bodies (DLB), which are neurodegenerative conditions characterized by intraneuronal protein aggregates containing α-synuclein. To explore how GBA1 mutations lead to neurodegeneration in GD, PD and DLB, we developed a novel invertebrate model of GBA1 insufficiency by deleting the Drosophila GBA1 homolog, dGBA1b. We found that dGBA1b mutants have multiple phenotypes consistent with neuronal dysfunction as seen in PD, DLB and GD, and dramatically increased protein aggregation that is normally cleared through an autophagic mechanism. dGBA1b mutants expressing human α-synuclein in dopaminergic neurons led to dopaminergic neuron loss and α-synuclein aggregation. However, α-synuclein expression had minimal effect on dGBA1b mutant phenotypes, suggesting that the deleterious consequences of glucocerebrosidase deficiency are independent of α-synuclein. These findings significantly contribute to our understanding of the role of GBA1 mutations in the pathogenesis of PD, DLB, and GD, and further studies using this model should elucidate mechanisms underlying these diseases.
Collapse
|
354
|
Glucocerebrosidase and parkinsonism: lessons to learn. J Neurol 2016; 263:1033-1044. [PMID: 26995357 DOI: 10.1007/s00415-016-8085-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/26/2016] [Accepted: 02/27/2016] [Indexed: 12/23/2022]
Abstract
Both homo- (causing autosomal-recessive Gaucher's disease; GD) and heterozygous mutations in the glucocerebrosidase gene (GBA) are associated with Parkinson's disease (PD), and represent the most robust known genetic susceptibility factors identified in PD. Since the accumulation of α-synuclein has been considered critical to the pathogenesis of PD among several possible pathways through which glucocerebrosidase (GCase) deficiency may promote the pathogenesis of PD, particular attention was given to the reciprocity with α-synuclein levels, lysosomal dysfunction, endoplasmatic reticulum-Golgi trafficking of GCase, dysregulation of calcium homeostasis and mitochondrial abnormalities. The proportion of PD patients that carry GBA mutations is estimated to be approximately between 5 and 10 %. Individual PD patients with or without GBA mutations cannot be discriminated on clinical or pathological grounds. However, GBA mutation carriers may have slightly earlier age at PD onset, more likely have a positive family history for PD, and more prevalent non-motor symptoms when compared to those patients who are not carriers. Establishing the concept of GBA-related PD promoted a search for the pathogenic mechanisms through which GCase deficiency may influence pathogenesis of PD, suggesting that targeting the GCase-lysosomal pathway might be a rational approach for the development of neuroprotective drugs in PD.
Collapse
|
355
|
Methyl-Arginine Profile of Brain from Aged PINK1-KO+A53T-SNCA Mice Suggests Altered Mitochondrial Biogenesis. PARKINSONS DISEASE 2016; 2016:4686185. [PMID: 27034888 PMCID: PMC4791501 DOI: 10.1155/2016/4686185] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 11/29/2022]
Abstract
Hereditary Parkinson's disease can be triggered by an autosomal dominant overdose of alpha-Synuclein (SNCA) or the autosomal recessive deficiency of PINK1. We recently showed that the combination of PINK1-knockout with overexpression of A53T-SNCA in double mutant (DM) mice potentiates phenotypes and reduces survival. Now we studied brain hemispheres of DM mice at age of 18 months in a hypothesis-free approach, employing a quantitative label-free global proteomic mass spectrometry scan of posttranslational modifications focusing on methyl-arginine. The strongest effects were documented for the adhesion modulator CMAS, the mRNA decapping/deadenylation factor PATL1, and the synaptic plasticity mediator CRTC1/TORC1. In addition, an intriguing effect was observed for the splicing factor PSF/SFPQ, known to interact with the dopaminergic differentiation factor NURR1 as well as with DJ-1, the protein responsible for the autosomal recessive PARK7 variant of PD. CRTC1, PSF, and DJ-1 are modulators of PGC1alpha and of mitochondrial biogenesis. This pathway was further stressed by dysregulations of oxygen sensor EGLN3 and of nuclear TMPO. PSF and TMPO cooperate with dopaminergic differentiation factors LMX1B and NURR1. Further dysregulations concerned PRR18, TRIO, HNRNPA1, DMWD, WAVE1, ILDR2, DBNDD1, and NFM. Thus, we report selective novel endogenous stress responses in brain, which highlight early dysregulations of mitochondrial homeostasis and midbrain vulnerability.
Collapse
|
356
|
Moors T, Paciotti S, Chiasserini D, Calabresi P, Parnetti L, Beccari T, van de Berg WDJ. Lysosomal Dysfunction and α-Synuclein Aggregation in Parkinson's Disease: Diagnostic Links. Mov Disord 2016; 31:791-801. [DOI: 10.1002/mds.26562] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 12/31/2015] [Accepted: 01/06/2016] [Indexed: 12/15/2022] Open
Affiliation(s)
- Tim Moors
- Department of Anatomy and Neurosciences; Section Quantitative Morphology, Neuroscience Campus Amsterdam, VU University Medical Center; Amsterdam the Netherlands
| | - Silvia Paciotti
- Department of Pharmaceutical Sciences; Section of Nutrition and Food Science, University of Perugia; Perugia Italy
| | - Davide Chiasserini
- Department of Medicine; Section of Neurology, University of Perugia; Perugia Italy
| | - Paolo Calabresi
- Department of Medicine; Section of Neurology, University of Perugia; Perugia Italy
- Fondazione Santa Lucia-Istituto di Ricovero e Cura a Carattere Scientifico; Roma Italy
| | - Lucilla Parnetti
- Department of Medicine; Section of Neurology, University of Perugia; Perugia Italy
| | - Tommaso Beccari
- Department of Pharmaceutical Sciences; Section of Nutrition and Food Science, University of Perugia; Perugia Italy
| | - Wilma D. J. van de Berg
- Department of Anatomy and Neurosciences; Section Quantitative Morphology, Neuroscience Campus Amsterdam, VU University Medical Center; Amsterdam the Netherlands
| |
Collapse
|
357
|
Barker RA, Williams-Gray CH. Review: The spectrum of clinical features seen with alpha synuclein pathology. Neuropathol Appl Neurobiol 2016; 42:6-19. [DOI: 10.1111/nan.12303] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/06/2016] [Accepted: 01/10/2016] [Indexed: 12/15/2022]
Affiliation(s)
- R. A. Barker
- Department of Clinical Neurosciences; John van Geest Centre for Brain Repair; University of Cambridge; Cambridge UK
| | - C. H. Williams-Gray
- Department of Clinical Neurosciences; John van Geest Centre for Brain Repair; University of Cambridge; Cambridge UK
| |
Collapse
|
358
|
Migdalska-Richards A, Schapira AHV. The relationship between glucocerebrosidase mutations and Parkinson disease. J Neurochem 2016; 139 Suppl 1:77-90. [PMID: 26860875 PMCID: PMC5111601 DOI: 10.1111/jnc.13385] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 09/08/2015] [Accepted: 10/02/2015] [Indexed: 01/12/2023]
Abstract
Parkinson disease (PD) is the second most common neurodegenerative disorder after Alzheimer disease, whereas Gaucher disease (GD) is the most frequent lysosomal storage disorder caused by homozygous mutations in the glucocerebrosidase (GBA1) gene. Increased risk of developing PD has been observed in both GD patients and carriers. It has been estimated that GBA1 mutations confer a 20‐ to 30‐fold increased risk for the development of PD, and that at least 7–10% of PD patients have a GBA1 mutation. To date, mutations in the GBA1 gene constitute numerically the most important risk factor for PD. The type of PD associated with GBA1 mutations (PD‐GBA1) is almost identical to idiopathic PD, except for a slightly younger age of onset and a tendency to more cognitive impairment. Importantly, the pathology of PD‐GBA1 is identical to idiopathic PD, with nigral dopamine cell loss, Lewy bodies, and neurites containing alpha‐synuclein. The mechanism by which GBA1 mutations increase the risk for PD is still unknown. However, given that clinical manifestation and pathological findings in PD‐GBA1 patients are almost identical to those in idiopathic PD individuals, it is likely that, as in idiopathic PD, alpha‐synuclein accumulation, mitochondrial dysfunction, autophagic impairment, oxidative and endoplasmic reticulum stress may contribute to the development and progression of PD‐GBA1. Here, we review the GBA1 gene, its role in GD, and its link with PD.
The impact of glucocerebrosidase 1 (GBA1) mutations on functioning of endoplasmic reticulum (ER), lysosomes, and mitochondria. GBA1 mutations resulting in production of misfolded glucocerebrosidase (GCase) significantly affect the ER functioning. Misfolded GCase trapped in the ER leads to both an increase in the ubiquitin–proteasome system (UPS) and the ER stress. The presence of ER stress triggers the unfolded protein response (UPR) and/or endoplasmic reticulum‐associated degradation (ERAD). The prolonged activation of UPR and ERAD subsequently leads to increased apoptosis. The presence of misfolded GCase in the lysosomes together with a reduction in wild‐type GCase levels lead to a retardation of alpha‐synuclein degradation via chaperone‐mediated autophagy (CMA), which subsequently results in alpha‐synuclein accumulation and aggregation. Impaired lysosomal functioning also causes a decrease in the clearance of autophagosomes, and so their accumulation. GBA1 mutations perturb normal mitochondria functioning by increasing generation of free radical species (ROS) and decreasing adenosine triphosphate (ATP) production, oxygen consumption, and membrane potential. GBA1 mutations also lead to accumulation of dysfunctional and fragmented mitochondria.
This article is part of a special issue on Parkinson disease.
Collapse
|
359
|
Rivero-Ríos P, Madero-Pérez J, Fernández B, Hilfiker S. Targeting the Autophagy/Lysosomal Degradation Pathway in Parkinson's Disease. Curr Neuropharmacol 2016; 14:238-49. [PMID: 26517050 PMCID: PMC4857622 DOI: 10.2174/1570159x13666151030103027] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 06/17/2015] [Accepted: 06/17/2015] [Indexed: 01/08/2023] Open
Abstract
Autophagy is a cellular quality control mechanism crucial for neuronal homeostasis. Defects in autophagy are critically associated with mechanisms underlying Parkinson's disease (PD), a common and debilitating neurodegenerative disorder. Autophagic dysfunction in PD can occur at several stages of the autophagy/lysosomal degradative machinery, contributing to the formation of intracellular protein aggregates and eventual neuronal cell death. Therefore, autophagy inducers may comprise a promising new therapeutic approach to combat neurodegeneration in PD. Several currently available FDA-approved drugs have been shown to enhance autophagy, which may allow for their repurposing for use in novel clinical conditions including PD. This review summarizes our current knowledge of deficits in the autophagy/lysosomal degradation pathways associated with PD, and highlight current approaches which target this pathway as possible means towards novel therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Sabine Hilfiker
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Avda del Conocimiento s/n, 18016 Granada, Spain.
| |
Collapse
|
360
|
McMahon B, Aflaki E, Sidransky E. Chaperoning glucocerebrosidase: a therapeutic strategy for both Gaucher disease and Parkinsonism. Neural Regen Res 2016; 11:1760-1761. [PMID: 28123414 PMCID: PMC5204226 DOI: 10.4103/1673-5374.194717] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Affiliation(s)
- Benjamin McMahon
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elma Aflaki
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ellen Sidransky
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
361
|
Barkhuizen M, Anderson DG, Grobler AF. Advances in GBA-associated Parkinson's disease--Pathology, presentation and therapies. Neurochem Int 2015; 93:6-25. [PMID: 26743617 DOI: 10.1016/j.neuint.2015.12.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/29/2015] [Accepted: 12/04/2015] [Indexed: 12/27/2022]
Abstract
GBA mutations are to date the most common genetic risk factor for Parkinson's disease. The GBA gene encodes the lysomal hydrolase glucocerebrosidase. Whilst bi-allelic GBA mutations cause Gaucher disease, both mono- and bi-allelic mutations confer risk for Parkinson's disease. Clinically, Parkinson's disease patients with GBA mutations resemble idiopathic Parkinson's disease patients. However, these patients have a modest reduction in age-of-onset of disease and a greater incidence of cognitive decline. In some cases, GBA mutations are also responsible for familial Parkinson's disease. The accumulation of α-synuclein into Lewy bodies is the central neuropathological hallmark of Parkinson's disease. Pathologic GBA mutations reduce enzymatic function. A reduction in glucocerebrosidase function increases α-synuclein levels and propagation, which in turn inhibits glucocerebrosidase in a feed-forward cascade. This cascade is central to the neuropathology of GBA-associated Parkinson's disease. The lysosomal integral membrane protein type-2 is necessary for normal glucocerebrosidase function. Glucocerebrosidase dysfunction also increases in the accumulation of β-amyloid and amyloid-precursor protein, oxidative stress, neuronal susceptibility to metal ions, microglial and immune activation. These factors contribute to neuronal death. The Mendelian Parkinson's disease genes, Parkin and ATP13A2, intersect with glucocerebrosidase. These factors sketch a complex circuit of GBA-associated neuropathology. To clinically interfere with this circuit, central glucocerebrosidase function must be improved. Strategies based on reducing breakdown of mutant glucocerebrosidase and increasing the fraction that reaches the lysosome has shown promise. Breakdown can be reduced by interfering with the ability of heat-shock proteins to recognize mutant glucocerebrosidase. This underlies the therapeutic efficacy of certain pharmacological chaperones and histone deacetylase inhibitors. These therapies are promising for Parkinson's disease, regardless of mutation status. Recently, there has been a boom in studies investigating the role of glucocerebrosidase in the pathology of Parkinson's disease. This merits a comprehensive review of the current cell biological processes and pathological pictures involving Parkinson's disease associated with GBA mutations.
Collapse
Affiliation(s)
- Melinda Barkhuizen
- DST/NWU Preclinical Drug Development Platform, North-West University, Potchefstroom, 2520, South Africa; Department of Paediatrics, School for Mental Health and Neuroscience, Maastricht University, Maastricht, 6229, The Netherlands.
| | - David G Anderson
- Department of Neurology, Witwatersrand University Donald Gordon Medical Centre, Parktown, Johannesburg, 2193, South Africa
| | - Anne F Grobler
- DST/NWU Preclinical Drug Development Platform, North-West University, Potchefstroom, 2520, South Africa
| |
Collapse
|
362
|
Choi S, Kim D, Kam TI, Yun S, Kim S, Park H, Hwang H, Pletnikova O, Troncoso JC, Dawson VL, Dawson TM, Ko HS. Lysosomal Enzyme Glucocerebrosidase Protects against Aβ1-42 Oligomer-Induced Neurotoxicity. PLoS One 2015; 10:e0143854. [PMID: 26629917 PMCID: PMC4668030 DOI: 10.1371/journal.pone.0143854] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 11/10/2015] [Indexed: 01/31/2023] Open
Abstract
Glucocerebrosidase (GCase) functions as a lysosomal enzyme and its mutations are known to be related to many neurodegenerative diseases, including Gaucher’s disease (GD), Parkinson’s disease (PD), and Dementia with Lewy Bodies (DLB). However, there is little information about the role of GCase in the pathogenesis of Alzheimer’s disease (AD). Here we demonstrate that GCase protein levels and enzyme activity are significantly decreased in sporadic AD. Moreover, Aβ1–42 oligomer treatment results in neuronal cell death that is concomitant with decreased GCase protein levels and enzyme activity, as well as impairment in lysosomal biogenesis and acidification. Importantly, overexpression of GCase promotes the lysosomal degradation of Aβ1–42 oligomers, restores the lysosomal impairment, and protects against the toxicity in neurons treated with Aβ1–42 oligomers. Our findings indicate that a deficiency of GCase could be involved in progression of AD pathology and suggest that augmentation of GCase activity may be a potential therapeutic option for the treatment of AD.
Collapse
Affiliation(s)
- Seulah Choi
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana, United States of America
| | - Donghoon Kim
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana, United States of America
| | - Tae-In Kam
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Seungpil Yun
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana, United States of America
| | - Sangjune Kim
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Hyejin Park
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Heehong Hwang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Olga Pletnikova
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Juan C. Troncoso
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Valina L. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana, United States of America
| | - Ted M. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana, United States of America
| | - Han Seok Ko
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
363
|
Kilpatrick BS, Magalhaes J, Beavan MS, McNeill A, Gegg ME, Cleeter MWJ, Bloor-Young D, Churchill GC, Duchen MR, Schapira AH, Patel S. Endoplasmic reticulum and lysosomal Ca²⁺ stores are remodelled in GBA1-linked Parkinson disease patient fibroblasts. Cell Calcium 2015; 59:12-20. [PMID: 26691915 PMCID: PMC4751977 DOI: 10.1016/j.ceca.2015.11.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/06/2015] [Accepted: 11/25/2015] [Indexed: 11/19/2022]
Abstract
Mutations in β-glucocerebrosidase (encoded by GBA1) cause Gaucher disease (GD), a lysosomal storage disorder, and increase the risk of developing Parkinson disease (PD). The pathogenetic relationship between the two disorders is unclear. Here, we characterised Ca(2+) release in fibroblasts from type I GD and PD patients together with age-matched, asymptomatic carriers, all with the common N370S mutation in β-glucocerebrosidase. We show that endoplasmic reticulum (ER) Ca(2+) release was potentiated in GD and PD patient fibroblasts but not in cells from asymptomatic carriers. ER Ca(2+) signalling was also potentiated in fibroblasts from aged healthy subjects relative to younger individuals but not further increased in aged PD patient cells. Chemical or molecular inhibition of β-glucocerebrosidase in fibroblasts and a neuronal cell line did not affect ER Ca(2+) signalling suggesting defects are independent of enzymatic activity loss. Conversely, lysosomal Ca(2+) store content was reduced in PD fibroblasts and associated with age-dependent alterations in lysosomal morphology. Accelerated remodelling of Ca(2+) stores by pathogenic GBA1 mutations may therefore feature in PD.
Collapse
Affiliation(s)
- Bethan S Kilpatrick
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| | - Joana Magalhaes
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London NW3 2PF, UK
| | - Michelle S Beavan
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London NW3 2PF, UK
| | - Alisdair McNeill
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London NW3 2PF, UK
| | - Matthew E Gegg
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London NW3 2PF, UK
| | - Michael W J Cleeter
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London NW3 2PF, UK
| | | | - Grant C Churchill
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Michael R Duchen
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Anthony H Schapira
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London NW3 2PF, UK
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
364
|
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
|
365
|
Haploinsufficiency of cathepsin D leads to lysosomal dysfunction and promotes cell-to-cell transmission of α-synuclein aggregates. Cell Death Dis 2015; 6:e1901. [PMID: 26448324 PMCID: PMC4632307 DOI: 10.1038/cddis.2015.283] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 07/06/2015] [Accepted: 07/22/2015] [Indexed: 12/17/2022]
Abstract
Lysosomal dysfunction has been implicated both pathologically and genetically in neurodegenerative disorders, such as Alzheimer's disease and Parkinson's disease (PD). Lysosomal gene deficiencies cause lysosomal storage disorders, many of which involve neurodegeneration. Heterozygous mutations of some of these genes, such as GBA1, are associated with PD. CTSD is the gene encoding Cathepsin D (CTSD), a lysosomal protein hydrolase, and homozygous CTSD deficiency results in neuronal ceroid-lipofuscinosis, which is characterized by the early onset, progressive neurodegeneration. CTSD deficiency was also associated with deposition of α-synuclein aggregates, the hallmark of PD. However, whether partial deficiency of CTSD has a role in the late onset progressive neurodegenerative disorders, including PD, remains unknown. Here, we generated cell lines harboring heterozygous nonsense mutations in CTSD with genomic editing using the zinc finger nucleases. Heterozygous mutation in CTSD resulted in partial loss of CTSD activity, leading to reduced lysosomal activity. The CTSD mutation also resulted in increased accumulation of intracellular α-synuclein aggregates and the secretion of the aggregates. When α-synuclein was introduced in the media, internalized α-synuclein aggregates accumulated at higher levels in CTSD+/− cells than in the wild-type cells. Consistent with these results, transcellular transmission of α-synuclein aggregates was increased in CTSD+/− cells. The increased transmission of α-synuclein aggregates sustained during the successive passages of CTSD+/− cells. These results suggest that partial loss of CTSD activity is sufficient to cause a reduction in lysosomal function, which in turn leads to α-synuclein aggregation and propagation of the aggregates.
Collapse
|
366
|
Ambrosi G, Ghezzi C, Zangaglia R, Levandis G, Pacchetti C, Blandini F. Ambroxol-induced rescue of defective glucocerebrosidase is associated with increased LIMP-2 and saposin C levels in GBA1 mutant Parkinson's disease cells. Neurobiol Dis 2015; 82:235-242. [PMID: 26094596 DOI: 10.1016/j.nbd.2015.06.008] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 06/03/2015] [Accepted: 06/15/2015] [Indexed: 11/26/2022] Open
Abstract
Heterozygous mutations in GBA1 gene, encoding for lysosomal enzyme glucocerebrosidase (GCase), are a major risk factor for sporadic Parkinson's disease (PD). Defective GCase has been reported in fibroblasts of GBA1-mutant PD patients and pharmacological chaperone ambroxol has been shown to correct such defect. To further explore this issue, we investigated GCase and elements supporting GCase function and trafficking in fibroblasts from sporadic PD patients--with or without heterozygous GBA1 mutations--and healthy subjects, in basal conditions and following in vitro exposure to ambroxol. We assessed protein levels of GCase, lysosomal integral membrane protein-2 (LIMP-2), which mediates GCase trafficking to lysosomes, GCase endogenous activator saposin (Sap) C and parkin, which is involved in degradation of defective GCase. We also measured activities of GCase and cathepsin D, which cleaves Sap C from precursor prosaposin. GCase activity was reduced in fibroblasts from GBA1-mutant patients and ambroxol corrected this defect. Ambroxol increased cathepsin D activity, GCase and Sap C protein levels in all groups, while LIMP-2 levels were increased only in GBA1-mutant PD fibroblasts. Parkin levels were slightly increased only in the PD group without GBA1 mutations and were not significantly modified by ambroxol. Our study confirms that GCase activity is deficient in fibroblasts of GBA1-mutant PD patients and that ambroxol corrects this defect. The drug increased Sap C and LIMP-2 protein levels, without interfering with parkin. These results confirm that chemical chaperone ambroxol modulates lysosomal markers, further highlighting targets that may be exploited for innovative PD therapeutic strategies.
Collapse
Affiliation(s)
- Giulia Ambrosi
- Center for Research in Neurodegenerative Diseases, C. Mondino National Neurological Institute, Pavia, Italy
| | - Cristina Ghezzi
- Center for Research in Neurodegenerative Diseases, C. Mondino National Neurological Institute, Pavia, Italy
| | - Roberta Zangaglia
- Center for Research in Neurodegenerative Diseases, C. Mondino National Neurological Institute, Pavia, Italy
| | - Giovanna Levandis
- Center for Research in Neurodegenerative Diseases, C. Mondino National Neurological Institute, Pavia, Italy
| | - Claudio Pacchetti
- Center for Research in Neurodegenerative Diseases, C. Mondino National Neurological Institute, Pavia, Italy
| | - Fabio Blandini
- Center for Research in Neurodegenerative Diseases, C. Mondino National Neurological Institute, Pavia, Italy.
| |
Collapse
|
367
|
van der Brug MP, Singleton A, Gasser T, Lewis PA. Parkinson's disease: From human genetics to clinical trials. Sci Transl Med 2015; 7:205ps20. [PMID: 26378242 PMCID: PMC5995146 DOI: 10.1126/scitranslmed.aaa8280] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Combining genetic insights into the pathogenesis of Parkinson's disease (PD) with findings from animal and cellular models of this disorder has advanced our understanding of the pathways that lead to the characteristic degeneration of dopaminergic neurons in the brain's nigrostriatal pathway. This has fueled an increase in candidate compounds designed to modulate these pathways and to alter the processes underlying neuronal death in this disorder. Using mitochondrial quality control and the macroautophagy/lysosomal pathways as examples, we discuss the pipeline from a comprehensive genetic architecture for PD through to clinical trials for drugs targeting pathways linked to neurodegeneration in PD. We also identify opportunities and pitfalls on the road to a clinically effective disease-modifying treatment for this disease.
Collapse
Affiliation(s)
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thomas Gasser
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen and German Centre for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Patrick A Lewis
- School of Pharmacy, University of Reading, Reading RG6 6AP, UK. Centre for Integrated Neuroscience and Neurodynamics, University of Reading, Reading RG6 6AP, UK. Department of Molecular Neuroscience, UCL Institute of Neurology, Queen's Square, London WC1N 3BG, UK.
| |
Collapse
|
368
|
Pal G, Robertson E, O'Keefe J, Hall D. The Neuropsychiatric and Motor Profile of GBA-Associated Parkinson's Disease: A Review. Mov Disord Clin Pract 2015; 3:4-8. [PMID: 30363594 DOI: 10.1002/mdc3.12229] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 05/21/2015] [Accepted: 06/25/2015] [Indexed: 01/15/2023] Open
Abstract
Background Cognitive and motor decline, along with psychiatric symptoms, have a major impact on independence, nursing home admission, caregiver burden, and mortality in Parkinson's disease (PD). The single most common genetic risk factor for developing PD is a mutation in the glucocerebrosidase (GBA) gene. Methods This work is a literature review regarding "GBA" and "Parkinson's disease" as conducted by PubMed search. Results There is a higher prevalence of cognitive decline and more rapid trajectory of disease progression in PD-GBA carriers, compared to noncarriers. PD-GBA carriers also have domain-specific cognitive impairment, particularly in visual memory tasks. PD-GBA carriers may also have a more aggressive motor phenotype than noncarriers. Conclusions Early identification of PD-GBA carriers may lead to targeted therapies and development of new treatments.
Collapse
Affiliation(s)
- Gian Pal
- Department of Neurological Sciences Rush University Chicago Illinois USA
| | - Erin Robertson
- Department of Anatomy & Cell Biology Rush University Chicago Illinois USA
| | - Joan O'Keefe
- Department of Anatomy & Cell Biology Rush University Chicago Illinois USA
| | - Deborah Hall
- Department of Neurological Sciences Rush University Chicago Illinois USA
| |
Collapse
|
369
|
Kubo SI. Membrane lipids as therapeutic targets for Parkinson’s disease: a possible link between Lewy pathology and membrane lipids. Expert Opin Ther Targets 2015; 20:1301-1310. [DOI: 10.1517/14728222.2016.1086340] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
370
|
Soto-Ortolaza AI, Ross OA. Genetic susceptibility variants in parkinsonism. Parkinsonism Relat Disord 2015; 22 Suppl 1:S7-11. [PMID: 26414118 DOI: 10.1016/j.parkreldis.2015.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 09/02/2015] [Indexed: 12/22/2022]
Abstract
Parkinsonism is an umbrella term for a group of disorders characterized by the clinical signs of tremor, bradykinesia, rigidity, and postural instability. On neuropathologic examination parkinsonism can display alternate protein pathologies (e.g. α-synucleinopathy or tauopathy) but the degeneration of nigral neurons is consistent. The main forms of parkinsonism are, Parkinson's disease (PD), Dementia with Lewy Bodies (DLB), Multiple System Atrophy (MSA), Progressive Supranuclear Palsy (PSP) and Corticobasal Degeneration (CBD). Genetic studies from candidate gene, to unbiased genome-wide approaches including association and next-generation sequencing have nominated a number of disease determinants. Within this review we will highlight the genetic loci that are associated with disease and discuss the implications and importance for a better understanding of the genes involved and thus the underlying pathophysiology of these disorders.
Collapse
Affiliation(s)
- Alexandra I Soto-Ortolaza
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Department of Biology, University of North Florida, Jacksonville, FL 32224, USA; School of Medicine and Medical Science, University College Dublin, Dublin, Ireland; Mayo Graduate School, Neurobiology of Disease, Jacksonville, FL, USA.
| |
Collapse
|
371
|
Abstract
Parkinson's disease is a neurological disorder with evolving layers of complexity. It has long been characterised by the classical motor features of parkinsonism associated with Lewy bodies and loss of dopaminergic neurons in the substantia nigra. However, the symptomatology of Parkinson's disease is now recognised as heterogeneous, with clinically significant non-motor features. Similarly, its pathology involves extensive regions of the nervous system, various neurotransmitters, and protein aggregates other than just Lewy bodies. The cause of Parkinson's disease remains unknown, but risk of developing Parkinson's disease is no longer viewed as primarily due to environmental factors. Instead, Parkinson's disease seems to result from a complicated interplay of genetic and environmental factors affecting numerous fundamental cellular processes. The complexity of Parkinson's disease is accompanied by clinical challenges, including an inability to make a definitive diagnosis at the earliest stages of the disease and difficulties in the management of symptoms at later stages. Furthermore, there are no treatments that slow the neurodegenerative process. In this Seminar, we review these complexities and challenges of Parkinson's disease.
Collapse
Affiliation(s)
- Lorraine V Kalia
- Morton and Gloria Shulman Movement Disorders Clinic and the Edmond J Safra Program in Parkinson's Disease, Toronto Western Hospital, Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Anthony E Lang
- Morton and Gloria Shulman Movement Disorders Clinic and the Edmond J Safra Program in Parkinson's Disease, Toronto Western Hospital, Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
372
|
Rocha EM, Smith GA, Park E, Cao H, Graham AR, Brown E, McLean JR, Hayes MA, Beagan J, Izen SC, Perez-Torres E, Hallett PJ, Isacson O. Sustained Systemic Glucocerebrosidase Inhibition Induces Brain α-Synuclein Aggregation, Microglia and Complement C1q Activation in Mice. Antioxid Redox Signal 2015; 23:550-64. [PMID: 26094487 PMCID: PMC4544823 DOI: 10.1089/ars.2015.6307] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AIMS Loss-of-function mutations in GBA1, which cause the autosomal recessive lysosomal storage disease, Gaucher disease (GD), are also a key genetic risk factor for the α-synucleinopathies, including Parkinson's disease (PD) and dementia with Lewy bodies. GBA1 encodes for the lysosomal hydrolase glucocerebrosidase and reductions in this enzyme result in the accumulation of the glycolipid substrates glucosylceramide and glucosylsphingosine. Deficits in autophagy and lysosomal degradation pathways likely contribute to the pathological accumulation of α-synuclein in PD. In this report we used conduritol-β-epoxide (CBE), a potent selective irreversible competitive inhibitor of glucocerebrosidase, to model reduced glucocerebrosidase activity in vivo, and tested whether sustained glucocerebrosidase inhibition in mice could induce neuropathological abnormalities including α-synucleinopathy, and neurodegeneration. RESULTS Our data demonstrate that daily systemic CBE treatment over 28 days caused accumulation of insoluble α-synuclein aggregates in the substantia nigra, and altered levels of proteins involved in the autophagy lysosomal system. These neuropathological changes were paralleled by widespread neuroinflammation, upregulation of complement C1q, abnormalities in synaptic, axonal transport and cytoskeletal proteins, and neurodegeneration. INNOVATION A reduction in brain GCase activity has been linked to sporadic PD and normal aging, and may contribute to the susceptibility of vulnerable neurons to degeneration. This report demonstrates that systemic reduction of GCase activity using chemical inhibition, leads to neuropathological changes in the brain reminiscent of α-synucleinopathy. CONCLUSIONS These data reveal a link between reduced glucocerebrosidase and the development of α-synucleinopathy and pathophysiological abnormalities in mice, and support the development of GCase therapeutics to reduce α-synucleinopathy in PD and related disorders.
Collapse
Affiliation(s)
- Emily M Rocha
- 1 Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital , Belmont, Massachusetts
| | - Gaynor A Smith
- 1 Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital , Belmont, Massachusetts
| | | | | | | | | | - Jesse R McLean
- 1 Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital , Belmont, Massachusetts
| | - Melissa A Hayes
- 1 Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital , Belmont, Massachusetts
| | - Jonathan Beagan
- 1 Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital , Belmont, Massachusetts
| | - Sarah C Izen
- 1 Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital , Belmont, Massachusetts
| | - Eduardo Perez-Torres
- 1 Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital , Belmont, Massachusetts
| | - Penelope J Hallett
- 1 Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital , Belmont, Massachusetts
| | - Ole Isacson
- 1 Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital , Belmont, Massachusetts
| |
Collapse
|
373
|
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
|
374
|
Cox TM, Rosenbloom BE, Barker RA. Gaucher disease and comorbidities: B-cell malignancy and parkinsonism. Am J Hematol 2015; 90 Suppl 1:S25-8. [PMID: 26096744 DOI: 10.1002/ajh.24057] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Data emerging from the International Collaborative Gaucher Group (ICGG) Gaucher Registry together with other contemporary clinical surveys have revealed a close association between Gaucher disease and non-Hodgkin's B-cell lymphoma and myeloma and Gaucher disease and Parkinson's disease. Several possible explanations for increased B-cell proliferation and neoplasia in Gaucher disease have been proposed, including the possible influence of sphingosine (derived from the extra lysosomal metabolism of glucosylceramide), gene modifiers, splenectomy and immune system deregulation induced by cytokines, chemokines, and hydrolases released from Gaucher cells. Parkinson's disease is frequently seen in the otherwise-healthy relatives of Gaucher disease patients leading to the finding that GBA mutations represent a genetic risk factor for Parkinson's disease. The mechanism of the association between GBA mutations and Parkinson's disease has yet to be elucidated but the pathogenesis appears distinct from that of Gaucher disease. Several pathogenic pathways have been proposed including lysosomal and/or mitochondrial dysfunction. The effect of Gaucher disease specific therapies on the incidence of cancer or Parkinson's disease are not clear and will likely be evaluated in future ICGG Gaucher Registry studies.
Collapse
Affiliation(s)
- Timothy M. Cox
- Department of Medicine; University of Cambridge; United Kingdom
| | | | - Roger A. Barker
- Department of Clinical Neurosciences; University of Cambridge; United Kingdom
| |
Collapse
|
375
|
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
|
376
|
Liu G, Chen M, Mi N, Yang W, Li X, Wang P, Yin N, Li Y, Yue F, Chan P, Yu S. Increased oligomerization and phosphorylation of α-synuclein are associated with decreased activity of glucocerebrosidase and protein phosphatase 2A in aging monkey brains. Neurobiol Aging 2015; 36:2649-59. [PMID: 26149921 DOI: 10.1016/j.neurobiolaging.2015.06.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 06/02/2015] [Accepted: 06/03/2015] [Indexed: 12/15/2022]
Abstract
Aging is associated with an increased risk for Parkinson's disease and dementia with Lewy bodies, in which α-synuclein (α-syn) oligomerization plays key pathogenic roles. Here, we show that oligomeric α-syn levels increase with age in the brain of cynomolgus monkeys and are accompanied by a decrease in the expression and activity of glucocerebrosidase (GCase), a lysosomal enzyme whose dysfunction is linked to accumulation of oligomeric α-syn. Besides, levels of α-syn phosphorylated at serine 129 (pS129 α-syn), a modification that promotes α-syn oligomerization also increase with age in the brain and is associated with a reduction in the activity of protein phosphatase 2A (PP2A), an enzyme that facilitates α-syn dephosphorylation. The inverse relationship between levels of oligomeric α-syn and pS129 α-syn and activity of GCase and PP2A was more evident in brain regions susceptible to neurodegeneration (i.e., the striatum and hippocampus) than those that are less vulnerable (i.e., cerebellum and occipital cortex). In vitro experiments showed that GCase activity was more potently inhibited by oligomeric than by monomeric α-syn in the lysosome-enriched fractions isolated from brain tissues and cultured neuronal cells. Inhibition of GCase activity induced an elevation of oligomeric α-syn levels, which was shown to increase pS129 α-syn levels and reduce PP2A activity in cultured neuronal cells. The alterations in oligomeric and pS129 α-syns and their association with GCase and PP2A in aging brains may explain the vulnerability of certain brain regions to neurodegeneration in Parkinson's disease and dementia with Lewy bodies.
Collapse
Affiliation(s)
- Guangwei Liu
- Department of Neurobiology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, China; Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Min Chen
- Department of Neurobiology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory for Parkinson's Disease, Beijing, China
| | - Na Mi
- Department of Neurobiology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, China; Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Weiwei Yang
- Department of Neurobiology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, China; Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Xin Li
- Department of Neurobiology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, China; Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Peng Wang
- Department of Neurobiology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, China; Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Na Yin
- Department of Neurobiology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, China; Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Yaohua Li
- Department of Neurobiology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, China; Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Feng Yue
- Department of Neurobiology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, China; Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Piu Chan
- Department of Neurobiology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, China; Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China; Beijing Key Laboratory for Parkinson's Disease, Beijing, China
| | - Shun Yu
- Department of Neurobiology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, China; Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China; Beijing Key Laboratory for Parkinson's Disease, Beijing, China.
| |
Collapse
|
377
|
Wheeler S, Sillence DJ. Mechanisms of Gaucher disease pathogenesis. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:S1. [PMID: 26046054 DOI: 10.3978/j.issn.2305-5839.2015.03.42] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 02/06/2015] [Indexed: 01/18/2023]
Affiliation(s)
- Simon Wheeler
- School of Pharmacy, Hawthorn Building, De Montfort University, Leicester, UK
| | - Dan John Sillence
- School of Pharmacy, Hawthorn Building, De Montfort University, Leicester, UK
| |
Collapse
|
378
|
Ishibashi Y, Hirabayashi Y. AMP-activated Protein Kinase Suppresses Biosynthesis of Glucosylceramide by Reducing Intracellular Sugar Nucleotides. J Biol Chem 2015; 290:18245-18260. [PMID: 26048992 DOI: 10.1074/jbc.m115.658948] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Indexed: 12/25/2022] Open
Abstract
The membrane glycolipid glucosylceramide (GlcCer) plays a critical role in cellular homeostasis. Its intracellular levels are thought to be tightly regulated. How cells regulate GlcCer levels remains to be clarified. AMP-activated protein kinase (AMPK), which is a crucial cellular energy sensor, regulates glucose and lipid metabolism to maintain energy homeostasis. Here, we investigated whether AMPK affects GlcCer metabolism. AMPK activators (5-aminoimidazole-4-carboxamide 1-β-d-ribofuranoside and metformin) decreased intracellular GlcCer levels and synthase activity in mouse fibroblasts. AMPK inhibitors or AMPK siRNA reversed these effects, suggesting that GlcCer synthesis is negatively regulated by an AMPK-dependent mechanism. Although AMPK did not affect the phosphorylation or expression of GlcCer synthase, the amount of UDP-glucose, an activated form of glucose required for GlcCer synthesis, decreased under AMPK-activating conditions. Importantly, the UDP-glucose pyrophosphatase Nudt14, which degrades UDP-glucose, generating UMP and glucose 1-phosphate, was phosphorylated and activated by AMPK. On the other hand, suppression of Nudt14 by siRNA had little effect on UDP-glucose levels, indicating that mammalian cells have an alternative UDP-glucose pyrophosphatase that mainly contributes to the reduction of UDP-glucose under AMPK-activating conditions. Because AMPK activators are capable of reducing GlcCer levels in cells from Gaucher disease patients, our findings suggest that reducing GlcCer through AMPK activation may lead to a new strategy for treating diseases caused by abnormal accumulation of GlcCer.
Collapse
Affiliation(s)
- Yohei Ishibashi
- Laboratory for Molecular Membrane Neuroscience, RIKEN Brain Science Institute, Wako-shi, Saitama 351-0198, Japan
| | - Yoshio Hirabayashi
- Laboratory for Molecular Membrane Neuroscience, RIKEN Brain Science Institute, Wako-shi, Saitama 351-0198, Japan.
| |
Collapse
|
379
|
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
|
380
|
Kresojević N, Janković M, Petrović I, Kumar KR, Dragašević N, Dobričić V, Novaković I, Svetel M, Klein C, Pekmezović T, Kostić VS. Presenting symptoms of GBA-related Parkinson's disease. Parkinsonism Relat Disord 2015; 21:804-7. [PMID: 25957717 DOI: 10.1016/j.parkreldis.2015.04.028] [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] [Received: 11/20/2014] [Revised: 04/11/2015] [Accepted: 04/14/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND Mutations in the Glucocerebrosidase gene (GBA) are associated with Parkinson's disease (PD). It has been shown that GBA-related PD (PD-GBA) patients had an earlier age at PD onset and more prevalent non-motor symptoms when compared to "sporadic" PD patients without such mutations (sPD). AIM To explore whether presenting symptoms differ between PD-GBA and sPD patients. METHODS Demographic and clinical features (including presenting symptoms) were collected for 578 PD patients. Sequence analysis was performed for exons 8-11 of the GBA gene for all participants. RESULTS 39 PD patients (6.7%) with GBA mutations were compared to 539 PD patients without them. Although no statistically significant differences were found regarding the presenting symptoms, we observed that pain was more frequently reported as an initial problem in the PD-GBA (10.3%) than in the sPD group (3.0%) (chi square p = 0.039; logistic regression analysis OR = 3.74; p = 0.024). CONCLUSIONS Overall, the presenting symptoms were similar in PD-GBA and sPD patients, with the exception that pain might be more frequent in PD-GBA.
Collapse
Affiliation(s)
- Nikola Kresojević
- Institute of Neurology CCS, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milena Janković
- Institute of Neurology CCS, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Igor Petrović
- Institute of Neurology CCS, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Kishore R Kumar
- Department of Neurogenetics, Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, St Leonards, NSW, Australia
| | - Nataša Dragašević
- Institute of Neurology CCS, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Valerija Dobričić
- Institute of Neurology CCS, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ivana Novaković
- Institute for Human Genetics, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Marina Svetel
- Institute of Neurology CCS, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Christine Klein
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Tatjana Pekmezović
- Institute of Epidemiology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vladimir S Kostić
- Institute of Neurology CCS, School of Medicine, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
381
|
Abstract
INTRODUCTION Sialidosis is a neurosomatic, lysosomal storage disease (LSD) caused by mutations in the NEU1 gene, encoding the lysosomal sialidase NEU1. Deficient enzyme activity results in impaired processing/degradation of sialo-glycoproteins, and accumulation of oversialylated metabolites. Sialidosis is considered an orphan disorder for which no therapy is currently available. AREAS COVERED The review describes the clinical forms of sialidosis and the NEU1 mutations so far identified; NEU1 requirement to complex with the protective protein/cathepsin A for stability and activation; and the pathogenic effects of NEU1 deficiency. Studies of the molecular mechanisms of pathogenesis in animal models uncovered basic cellular pathways downstream of NEU1 and its substrates, which may be implicated in more common adult (neurodegenerative) diseases. The development of a Phase I/II clinical trial for patients with galactosialidosis may prove suitable for sialidosis patients with the attenuated form of the disease. EXPERT OPINION Recently, there has been a renewed interest in the development of therapies for orphan LSDs, like sialidosis. Given the small number of potentially eligible patients, the way to treat sialidosis would be through the coordinated effort of clinical centers, which provide diagnosis and care for these patients, and the basic research labs that work towards understanding the disease pathogenesis.
Collapse
Affiliation(s)
- Alessandra d'Azzo
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Eda Machado
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ida Annunziata
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
382
|
Parks MM, Lawrence CE, Raphael BJ. Detecting non-allelic homologous recombination from high-throughput sequencing data. Genome Biol 2015; 16:72. [PMID: 25886137 PMCID: PMC4425883 DOI: 10.1186/s13059-015-0633-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/16/2015] [Indexed: 12/27/2022] Open
Abstract
Non-allelic homologous recombination (NAHR) is a common mechanism for generating genome rearrangements and is implicated in numerous genetic disorders, but its detection in high-throughput sequencing data poses a serious challenge. We present a probabilistic model of NAHR and demonstrate its ability to find NAHR in low-coverage sequencing data from 44 individuals. We identify NAHR-mediated deletions or duplications in 109 of 324 potential NAHR loci in at least one of the individuals. These calls segregate by ancestry, are more common in closely spaced repeats, often result in duplicated genes or pseudogenes, and affect highly studied genes such as GBA and CYP2E1.
Collapse
Affiliation(s)
- Matthew M Parks
- Division of Applied Mathematics, Brown University, Providence, USA.
| | - Charles E Lawrence
- Division of Applied Mathematics, Brown University, Providence, USA. .,Center for Computational Molecular Biology, Brown University, Providence, USA.
| | - Benjamin J Raphael
- Center for Computational Molecular Biology, Brown University, Providence, USA. .,Department of Computer Science, Brown University, Providence, USA.
| |
Collapse
|
383
|
Wexselblatt E, Esko JD, Tor Y. GNeosomes: Highly Lysosomotropic Nanoassemblies for Lysosomal Delivery. ACS NANO 2015; 9:3961-3968. [PMID: 25831231 DOI: 10.1021/nn507382n] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
GNeosomes, lysosomotropic lipid vesicles decorated with guanidinoneomycin, can encapsulate and facilitate the cellular internalization and lysosomal delivery of cargo ranging from small molecules to high molecular weight proteins, in a process that is exclusively dependent on cell surface glycosaminoglycans. Their cellular uptake mechanism and co-localization with lysosomes, as well as the delivery, release, and activity of internalized cargo, are quantified. GNeosomes are proposed as a universal platform for lysosomal delivery with potential as a basic research tool and a therapeutic vehicle.
Collapse
Affiliation(s)
- Ezequiel Wexselblatt
- †Chemistry and Biochemistry and ‡Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Jeffrey D Esko
- †Chemistry and Biochemistry and ‡Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Yitzhak Tor
- †Chemistry and Biochemistry and ‡Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
384
|
Li Y, Li P, Liang H, Zhao Z, Hashimoto M, Wei J. Gaucher-Associated Parkinsonism. Cell Mol Neurobiol 2015; 35:755-61. [PMID: 25820783 PMCID: PMC4502293 DOI: 10.1007/s10571-015-0176-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 03/10/2015] [Indexed: 12/11/2022]
Abstract
Gaucher disease is associated with Parkinson’s disease (PD) by mutations in glucocerebrosidase (GCase). The gene encoding GCase, glucosidase beta acid (GBA), is an important risk factor for PD. Findings from large studies have shown that patients with PD have an increased frequency of mutations in GBA and that GBA mutation carriers exhibit diverse parkinsonian phenotypes and Lewy body pathology. Although the mechanism for this association remains elusive, some hypotheses have been proposed to explain it, including gain of function caused by GBA mutations, which increases α-synuclein (α-syn) aggregation, loss of function due to lysosomal enzyme deficiency, which affects α-syn clearance, and even a bidirectional feedback loop, but each of these hypotheses has its limitations. It is also worth noting that many findings have implicated the interaction between α-syn and GCase, indicating the essential role of the interaction in the pathogenesis of GBA-associated parkinsonism. Therefore, the current review focuses on α-syn and GCase, and it provides some new thoughts that may be helpful for understanding the α-syn-GCase interaction and unraveling the exact mechanism underlying GBA-associated parkinsonism.
Collapse
Affiliation(s)
- Yaqiong Li
- Laboratory of Brain Function and Disease, Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | | | | | | | | | | |
Collapse
|
385
|
Rocha EM, Smith GA, Park E, Cao H, Brown E, Hallett P, Isacson O. Progressive decline of glucocerebrosidase in aging and Parkinson's disease. Ann Clin Transl Neurol 2015; 2:433-8. [PMID: 25909088 PMCID: PMC4402088 DOI: 10.1002/acn3.177] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 12/29/2014] [Indexed: 12/22/2022] Open
Abstract
The principal risk factor for developing most adult onset neurodegenerative diseases is aging, with incidence rising significantly after age 50. Despite research efforts, the causes of Parkinson's disease (PD) remain unknown. As neurons age, they show signs of diminished lysosomal and mitochondrial function, including increased oxidative stress and accumulation of misfolded proteins, and these changes become exacerbated PD. We show that activity of the lysosomal hydrolase glucocerebrosidase gradually diminishes with age in the substantia nigra and putamen of healthy controls. This reduction is comparable to glucocerebrosidase activity in GBA1-mutation carrier PD patients. These data, demonstrate for the first time that an age-dependent reduction in glucocerebrosidase activity may lower the threshold for developing PD.
Collapse
Affiliation(s)
- Emily M Rocha
- Neuroregeneration Laboratories, Harvard Medical School, McLean Hospital Belmont, Massachusetts, 02478
| | - Gaynor A Smith
- Neuroregeneration Laboratories, Harvard Medical School, McLean Hospital Belmont, Massachusetts, 02478
| | - Eric Park
- Shire 300 Shire Way, Lexington, Massachusetts, 02421
| | - Hongmei Cao
- Shire 300 Shire Way, Lexington, Massachusetts, 02421
| | - Eilish Brown
- Shire 300 Shire Way, Lexington, Massachusetts, 02421
| | - Penelope Hallett
- Neuroregeneration Laboratories, Harvard Medical School, McLean Hospital Belmont, Massachusetts, 02478
| | - Ole Isacson
- Neuroregeneration Laboratories, Harvard Medical School, McLean Hospital Belmont, Massachusetts, 02478
| |
Collapse
|
386
|
Gan-Or Z, Amshalom I, Kilarski LL, Bar-Shira A, Gana-Weisz M, Mirelman A, Marder K, Bressman S, Giladi N, Orr-Urtreger A. Differential effects of severe vs mild GBA mutations on Parkinson disease. Neurology 2015; 84:880-7. [PMID: 25653295 DOI: 10.1212/wnl.0000000000001315] [Citation(s) in RCA: 267] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE To better define the genotype-phenotype correlations between the type of GBA (glucosidase, beta, acid) mutation, severe or mild, and the risk and age at onset (AAO), and potential mechanism of Parkinson disease (PD). METHODS We analyzed 1,000 patients of Ashkenazi-Jewish descent with PD for 7 founder GBA mutations, and conducted a meta-analysis of risk and AAO according to GBA genotype (severe or mild mutation). The meta-analysis included 11,453 patients with PD and 14,565 controls from worldwide populations. The statistical analysis was done with and without continuity correction (constant or empirical), considering biases that could potentially affect the results. RESULTS Among Ashkenazi-Jewish patients with PD, the odds ratios for PD were 2.2 and 10.3 for mild and severe GBA mutation carriers, respectively. The observed frequency of severe GBA mutation carriers among patients with PD was more than 4-fold than expected (4.4% vs 0.9%, respectively, p < 0.0001, Fisher exact test). In the different models of the meta-analysis, the odds ratios for PD ranged between 2.84 and 4.94 for mild GBA mutation carriers and 9.92 and 21.29 for severe GBA mutation carriers (p < 1 × 10(-6) for all analyses). Pooled analysis demonstrated AAO of 53.1 (±11.2) and 58.1 (±10.6) years for severe and mild GBA mutation carriers, respectively (p = 4.3 × 10(-5)). CONCLUSIONS These data demonstrate that mild and severe heterozygous GBA mutations differentially affect the risk and the AAO of PD. Our results have important implications for genetic counseling and clinical follow-up.
Collapse
Affiliation(s)
- Ziv Gan-Or
- From The Genetic Institute (Z.G.-O., I.A., A.B.-S., M.G.-W., A.O.-R.) and Movement Disorders Unit, Parkinson Center, and Department of Neurology (A.M., N.G.), Tel Aviv Sourasky Medical Center; The Sackler Faculty of Medicine (N.G., A.O.-U.), Tel Aviv University, Israel; Stroke and Dementia Research Centre (L.L.K.), St. George's, University of London, UK; Columbia University (K.M.), Columbia Presbyterian Medical Center, New York; and Beth Israel Medical Center (S.B.), New York, NY. Z.G.-O. is currently affiliated with the Department of Human Genetics and Montreal Neurological Institute, McGill University, Montreal, Canada.
| | - Idan Amshalom
- From The Genetic Institute (Z.G.-O., I.A., A.B.-S., M.G.-W., A.O.-R.) and Movement Disorders Unit, Parkinson Center, and Department of Neurology (A.M., N.G.), Tel Aviv Sourasky Medical Center; The Sackler Faculty of Medicine (N.G., A.O.-U.), Tel Aviv University, Israel; Stroke and Dementia Research Centre (L.L.K.), St. George's, University of London, UK; Columbia University (K.M.), Columbia Presbyterian Medical Center, New York; and Beth Israel Medical Center (S.B.), New York, NY. Z.G.-O. is currently affiliated with the Department of Human Genetics and Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Laura L Kilarski
- From The Genetic Institute (Z.G.-O., I.A., A.B.-S., M.G.-W., A.O.-R.) and Movement Disorders Unit, Parkinson Center, and Department of Neurology (A.M., N.G.), Tel Aviv Sourasky Medical Center; The Sackler Faculty of Medicine (N.G., A.O.-U.), Tel Aviv University, Israel; Stroke and Dementia Research Centre (L.L.K.), St. George's, University of London, UK; Columbia University (K.M.), Columbia Presbyterian Medical Center, New York; and Beth Israel Medical Center (S.B.), New York, NY. Z.G.-O. is currently affiliated with the Department of Human Genetics and Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Anat Bar-Shira
- From The Genetic Institute (Z.G.-O., I.A., A.B.-S., M.G.-W., A.O.-R.) and Movement Disorders Unit, Parkinson Center, and Department of Neurology (A.M., N.G.), Tel Aviv Sourasky Medical Center; The Sackler Faculty of Medicine (N.G., A.O.-U.), Tel Aviv University, Israel; Stroke and Dementia Research Centre (L.L.K.), St. George's, University of London, UK; Columbia University (K.M.), Columbia Presbyterian Medical Center, New York; and Beth Israel Medical Center (S.B.), New York, NY. Z.G.-O. is currently affiliated with the Department of Human Genetics and Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Mali Gana-Weisz
- From The Genetic Institute (Z.G.-O., I.A., A.B.-S., M.G.-W., A.O.-R.) and Movement Disorders Unit, Parkinson Center, and Department of Neurology (A.M., N.G.), Tel Aviv Sourasky Medical Center; The Sackler Faculty of Medicine (N.G., A.O.-U.), Tel Aviv University, Israel; Stroke and Dementia Research Centre (L.L.K.), St. George's, University of London, UK; Columbia University (K.M.), Columbia Presbyterian Medical Center, New York; and Beth Israel Medical Center (S.B.), New York, NY. Z.G.-O. is currently affiliated with the Department of Human Genetics and Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Anat Mirelman
- From The Genetic Institute (Z.G.-O., I.A., A.B.-S., M.G.-W., A.O.-R.) and Movement Disorders Unit, Parkinson Center, and Department of Neurology (A.M., N.G.), Tel Aviv Sourasky Medical Center; The Sackler Faculty of Medicine (N.G., A.O.-U.), Tel Aviv University, Israel; Stroke and Dementia Research Centre (L.L.K.), St. George's, University of London, UK; Columbia University (K.M.), Columbia Presbyterian Medical Center, New York; and Beth Israel Medical Center (S.B.), New York, NY. Z.G.-O. is currently affiliated with the Department of Human Genetics and Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Karen Marder
- From The Genetic Institute (Z.G.-O., I.A., A.B.-S., M.G.-W., A.O.-R.) and Movement Disorders Unit, Parkinson Center, and Department of Neurology (A.M., N.G.), Tel Aviv Sourasky Medical Center; The Sackler Faculty of Medicine (N.G., A.O.-U.), Tel Aviv University, Israel; Stroke and Dementia Research Centre (L.L.K.), St. George's, University of London, UK; Columbia University (K.M.), Columbia Presbyterian Medical Center, New York; and Beth Israel Medical Center (S.B.), New York, NY. Z.G.-O. is currently affiliated with the Department of Human Genetics and Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Susan Bressman
- From The Genetic Institute (Z.G.-O., I.A., A.B.-S., M.G.-W., A.O.-R.) and Movement Disorders Unit, Parkinson Center, and Department of Neurology (A.M., N.G.), Tel Aviv Sourasky Medical Center; The Sackler Faculty of Medicine (N.G., A.O.-U.), Tel Aviv University, Israel; Stroke and Dementia Research Centre (L.L.K.), St. George's, University of London, UK; Columbia University (K.M.), Columbia Presbyterian Medical Center, New York; and Beth Israel Medical Center (S.B.), New York, NY. Z.G.-O. is currently affiliated with the Department of Human Genetics and Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Nir Giladi
- From The Genetic Institute (Z.G.-O., I.A., A.B.-S., M.G.-W., A.O.-R.) and Movement Disorders Unit, Parkinson Center, and Department of Neurology (A.M., N.G.), Tel Aviv Sourasky Medical Center; The Sackler Faculty of Medicine (N.G., A.O.-U.), Tel Aviv University, Israel; Stroke and Dementia Research Centre (L.L.K.), St. George's, University of London, UK; Columbia University (K.M.), Columbia Presbyterian Medical Center, New York; and Beth Israel Medical Center (S.B.), New York, NY. Z.G.-O. is currently affiliated with the Department of Human Genetics and Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Avi Orr-Urtreger
- From The Genetic Institute (Z.G.-O., I.A., A.B.-S., M.G.-W., A.O.-R.) and Movement Disorders Unit, Parkinson Center, and Department of Neurology (A.M., N.G.), Tel Aviv Sourasky Medical Center; The Sackler Faculty of Medicine (N.G., A.O.-U.), Tel Aviv University, Israel; Stroke and Dementia Research Centre (L.L.K.), St. George's, University of London, UK; Columbia University (K.M.), Columbia Presbyterian Medical Center, New York; and Beth Israel Medical Center (S.B.), New York, NY. Z.G.-O. is currently affiliated with the Department of Human Genetics and Montreal Neurological Institute, McGill University, Montreal, Canada.
| |
Collapse
|
387
|
Yadav AK, Shen DL, Shan X, He X, Kermode AR, Vocadlo DJ. Fluorescence-quenched substrates for live cell imaging of human glucocerebrosidase activity. J Am Chem Soc 2015; 137:1181-9. [PMID: 25562638 DOI: 10.1021/ja5106738] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Deficiency of the lysosomal glycoside hydrolase glucocerebrosidase (GCase) leads to abnormal accumulation of glucosyl ceramide in lysosomes and the development of the lysosomal storage disease known as Gaucher's disease. More recently, mutations in the GBA1 gene that encodes GCase have been uncovered as a major genetic risk factor for Parkinson's disease (PD). Current therapeutic strategies to increase GCase activity in lysosomes involve enzyme replacement therapy (ERT) and molecular chaperone therapy. One challenge associated with developing and optimizing these therapies is the difficulty in determining levels of GCase activity present within the lysosomes of live cells. Indeed, visualizing the activity of endogenous levels of any glycoside hydrolases, including GCase, has proven problematic within live mammalian cells. Here we describe the successful modular design and synthesis of fluorescence-quenched substrates for GCase. The selection of a suitable fluorophore and quencher pair permits the generation of substrates that allow convenient time-dependent monitoring of endogenous GCase activity within cells as well as localization of activity within lysosomes. These efficiently quenched (∼99.9%) fluorescent substrates also permit assessment of GCase inhibition in live cells by either confocal microscopy or high content imaging. Such substrates should enable improved understanding of GCase in situ as well the optimization of small-molecule chaperones for this enzyme. These findings also suggest routes to generate fluorescence-quenched substrates for other mammalian glycoside hydrolases for use in live cell imaging.
Collapse
Affiliation(s)
- Anuj K Yadav
- Department of Chemistry, ‡Department of Biological Sciences, and §Department of Molecular Biology and Biochemistry, Simon Fraser University , Burnaby, British Columbia V5A 1S6, Canada
| | | | | | | | | | | |
Collapse
|
388
|
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
|
389
|
Complexity of Genotype-Phenotype Correlations in Mendelian Disorders: Lessons from Gaucher Disease. Rare Dis 2015. [DOI: 10.1007/978-94-017-9214-1_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
390
|
Osellame LD, Duchen MR. Quality control gone wrong: mitochondria, lysosomal storage disorders and neurodegeneration. Br J Pharmacol 2014; 171:1958-72. [PMID: 24116849 PMCID: PMC3976615 DOI: 10.1111/bph.12453] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 09/04/2013] [Accepted: 09/23/2013] [Indexed: 12/18/2022] Open
Abstract
The eukaryotic cell possesses specialized pathways to turn over and degrade redundant proteins and organelles. Each pathway is unique and responsible for degradation of distinctive cytosolic material. The ubiquitin-proteasome system and autophagy (chaperone-mediated, macro, micro and organelle specific) act synergistically to maintain proteostasis. Defects in this equilibrium can be deleterious at cellular and organism level, giving rise to various disease states. Dysfunction of quality control pathways are implicated in neurodegenerative diseases and appear particularly important in Parkinson's disease and the lysosomal storage disorders. Neurodegeneration resulting from impaired degradation of ubiquitinated proteins and α-synuclein is often accompanied by mitochondrial dysfunction. Mitochondria have evolved to control a diverse number of processes, including cellular energy production, calcium signalling and apoptosis, and like every other organelle within the cell, they must be ‘recycled.’ Failure to do so is potentially lethal as these once indispensible organelles become destructive, leaking reactive oxygen species and activating the intrinsic cell death pathway. This process is paramount in neurons which have an absolute dependence on mitochondrial oxidative phosphorylation as they cannot up-regulate glycolysis. As such, mitochondrial bioenergetic failure can underpin neural death and neurodegenerative disease. In this review, we discuss the links between cellular quality control and neurodegenerative diseases associated with mitochondrial dysfunction, with particular attention to the emerging links between Parkinson's and Gaucher diseases in which defective quality control is a defining factor. LINKED ARTICLES This article is part of a themed issue on Mitochondrial Pharmacology: Energy, Injury & Beyond. To view the other articles in this issue visit http://dx.doi.org/10.1111/bph.2014.171.issue-8
Collapse
Affiliation(s)
- L D Osellame
- Department of Cell and Developmental Biology and UCL Consortium for Mitochondrial Research, University College London, London, UK
| | | |
Collapse
|
391
|
Yap TL, Jiang Z, Heinrich F, Gruschus JM, Pfefferkorn CM, Barros M, Curtis JE, Sidransky E, Lee JC. Structural features of membrane-bound glucocerebrosidase and α-synuclein probed by neutron reflectometry and fluorescence spectroscopy. J Biol Chem 2014; 290:744-54. [PMID: 25429104 DOI: 10.1074/jbc.m114.610584] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mutations in glucocerebrosidase (GCase), the enzyme deficient in Gaucher disease, are a common genetic risk factor for the development of Parkinson disease and related disorders, implicating the role of this lysosomal hydrolase in the disease etiology. A specific physical interaction exists between the Parkinson disease-related protein α-synuclein (α-syn) and GCase both in solution and on the lipid membrane, resulting in efficient enzyme inhibition. Here, neutron reflectometry was employed as a first direct structural characterization of GCase and α-syn·GCase complex on a sparsely-tethered lipid bilayer, revealing the orientation of the membrane-bound GCase. GCase binds to and partially inserts into the bilayer with its active site most likely lying just above the membrane-water interface. The interaction was further characterized by intrinsic Trp fluorescence, circular dichroism, and surface plasmon resonance spectroscopy. Both Trp fluorescence and neutron reflectometry results suggest a rearrangement of loops surrounding the catalytic site, where they extend into the hydrocarbon chain region of the outer leaflet. Taking advantage of contrasting neutron scattering length densities, the use of deuterated α-syn versus protiated GCase showed a large change in the membrane-bound structure of α-syn in the complex. We propose a model of α-syn·GCase on the membrane, providing structural insights into inhibition of GCase by α-syn. The interaction displaces GCase away from the membrane, possibly impeding substrate access and perturbing the active site. GCase greatly alters membrane-bound α-syn, moving helical residues away from the bilayer, which could impact the degradation of α-syn in the lysosome where these two proteins interact.
Collapse
Affiliation(s)
| | - Zhiping Jiang
- From the Laboratory of Molecular Biophysics, NHLBI, and
| | - Frank Heinrich
- the Department of Physics, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, and the Center for Neutron Research, National Institute of Standards and Technology (NIST), Gaithersburg, Maryland 20899
| | | | | | - Marilia Barros
- the Department of Physics, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, and
| | - Joseph E Curtis
- the Center for Neutron Research, National Institute of Standards and Technology (NIST), Gaithersburg, Maryland 20899
| | - Ellen Sidransky
- the Medical Genetics Branch, NHGRI, National Institutes of Health, Bethesda, Maryland 20892
| | | |
Collapse
|
392
|
Quadri M, Yang X, Cossu G, Olgiati S, Saddi VM, Breedveld GJ, Ouyang L, Hu J, Xu N, Graafland J, Ricchi V, Murgia D, Guedes LC, Mariani C, Marti MJ, Tarantino P, Asselta R, Valldeoriola F, Gagliardi M, Pezzoli G, Ezquerra M, Quattrone A, Ferreira J, Annesi G, Goldwurm S, Tolosa E, Oostra BA, Melis M, Wang J, Bonifati V. An exome study of Parkinson's disease in Sardinia, a Mediterranean genetic isolate. Neurogenetics 2014; 16:55-64. [PMID: 25294124 DOI: 10.1007/s10048-014-0425-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/15/2014] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder of complex aetiology. Rare, highly penetrant PD-causing mutations and common risk factors of small effect size have been identified in several genes/loci. However, these mutations and risk factors only explain a fraction of the disease burden, suggesting that additional, substantial genetic determinants remain to be found. Genetically isolated populations offer advantages for dissecting the genetic architecture of complex disorders, such as PD. We performed exome sequencing in 100 unrelated PD patients from Sardinia, a genetic isolate. SNPs absent from dbSNP129 and 1000 Genomes, shared by at least five patients, and of functional effects were genotyped in an independent Sardinian case-control sample (n = 500). Variants associated with PD with nominal p value <0.05 and those with odds ratio (OR) ≥3 were validated by Sanger sequencing and typed in a replication sample of 2965 patients and 2678 controls from Italy, Spain, and Portugal. We identified novel moderately rare variants in several genes, including SCAPER, HYDIN, UBE2H, EZR, MMRN2 and OGFOD1 that were specifically present in PD patients or enriched among them, nominating these as novel candidate risk genes for PD, although no variants achieved genome-wide significance after Bonferroni correction. Our results suggest that the genetic bases of PD are highly heterogeneous, with implications for the design of future large-scale exome or whole-genome analyses of this disease.
Collapse
Affiliation(s)
- Marialuisa Quadri
- Department of Clinical Genetics, Erasmus MC, PO Box 2040, 3000, CA, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
393
|
Pchelina SN, Nuzhnyi EP, Emelyanov AK, Boukina TM, Usenko TS, Nikolaev MA, Salogub GN, Yakimovskii AF, Zakharova EY. Increased plasma oligomeric alpha-synuclein in patients with lysosomal storage diseases. Neurosci Lett 2014; 583:188-93. [PMID: 25265039 DOI: 10.1016/j.neulet.2014.09.041] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 09/08/2014] [Accepted: 09/18/2014] [Indexed: 01/14/2023]
Abstract
A link between lysosomal storage diseases (LSDs) and neurodegenerative disorders associated with accumulation of presynaptic protein alpha-synuclein has been shown. Particularly, Gaucher disease (GD) patients with a deficiency of the lysosomal enzyme glucocerebrosidase (GBA) and carriers of GBA mutations are at increased risk of Parkinson's disease (PD). It remains unclear whether this link is due to increased alpha-synuclein oligomerization. Here we show that level of oligomeric alpha-synuclein form, associated with PD development, is increased in plasma of GD patients (n=41, median=22.9pg/mL, range1.57-444.58pg/mL; controls (n=40, median=6.02pg/mL, range 1.05-103.14pg/mL, p<0.0001). This difference is absent in GD patients receiving enzyme replacement therapy (ERT) for more than 5 years. Moreover, the levels of alpha-synuclein oligomers in plasma are also higher in patients with other LSDs (Niemann-Pick type C, Krabbe disease, Wolman disease) compared to the median value in controls. Therefore, we suggest that mutations in the GBA gene and at least in several other LSDs genes may be associated with an increase in oligomeric alpha-synuclein in plasma. ERT applied for recovering of GBA functions in GD treatment might decrease formation of plasma oligomeric alpha-synuclein.
Collapse
Affiliation(s)
- S N Pchelina
- Petersburg Nuclear Physics Institute, St. Petersburg, Russia; First Pavlov's State Medical University of Saint-Petersburg, St. Petersburg, Russia; St. Petersburg Academic University - Nanothecnology Research and Education Centre, RAS, St. Petersburg, Russia.
| | - E P Nuzhnyi
- First Pavlov's State Medical University of Saint-Petersburg, St. Petersburg, Russia
| | - A K Emelyanov
- Petersburg Nuclear Physics Institute, St. Petersburg, Russia; First Pavlov's State Medical University of Saint-Petersburg, St. Petersburg, Russia; St. Petersburg Academic University - Nanothecnology Research and Education Centre, RAS, St. Petersburg, Russia
| | - T M Boukina
- Medical-genetics Scientific Center, Moscow, Russia
| | - T S Usenko
- Petersburg Nuclear Physics Institute, St. Petersburg, Russia; First Pavlov's State Medical University of Saint-Petersburg, St. Petersburg, Russia
| | - M A Nikolaev
- Petersburg Nuclear Physics Institute, St. Petersburg, Russia; St. Petersburg Academic University - Nanothecnology Research and Education Centre, RAS, St. Petersburg, Russia
| | - G N Salogub
- First Pavlov's State Medical University of Saint-Petersburg, St. Petersburg, Russia
| | - A F Yakimovskii
- First Pavlov's State Medical University of Saint-Petersburg, St. Petersburg, Russia
| | | |
Collapse
|
394
|
Affiliation(s)
- Glenda M Halliday
- 1 Neuroscience Research Australia, Sydney, NSW 2031, Australia 2 School of Medical Sciences, Faculty of Medicine, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Karen E Murphy
- 1 Neuroscience Research Australia, Sydney, NSW 2031, Australia 2 School of Medical Sciences, Faculty of Medicine, The University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
395
|
Kresojević N, Dobričić V, Svetel M, Kostić V. Mutations in Niemann Pick type C gene are risk factor for Alzheimer's disease. Med Hypotheses 2014; 83:559-62. [PMID: 25220527 DOI: 10.1016/j.mehy.2014.08.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/13/2014] [Accepted: 08/24/2014] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia characterized by deterioration of memory and other cognitive domains which leads to death in 3-9years after diagnosis. In addition to mutations in APP, PSEN1 and PSEN2 genes, that cause early onset autosomal dominant AD, several genetic risk factors for late onset AD are now known. There is another distinctive neurodegenerative lysosomal storage disorder - Niemann-Pick type C (NPC) that is sometimes referred to as "Childhood Alzheimer's". NPC is autosomal recessive disease caused by mutations in the NPC1 or NPC2 genes. NPC and AD share some biochemical and pathological similarities which are discussed in this paper. On the other hand, there is a well documented connection between other autosomal recessive lysosomal storage disorder - Gaucher's disease (GD) and neurodegenerative disorder - Parkinson's disease (PD). It has been shown that GD patients have 20-fold increased life-time risk of developing PD. Surprisingly, even heterozygous carriers of mutations in glucocerebrosidase gene (GBA) have increased risk for developing PD. Having in mind above mentioned correlations, we hypothesized that heterozygous mutations in the NPC gene may act as an independent risk factor for Alzheimer's disease. If true, this would expand link between lysosomal disorders and neurodegenerative diseases. Also, if heterozygous NPC1/2 mutation carriers develop AD we assume it would be worth trying with miglustat-specific therapy recommended for NPC disease.
Collapse
Affiliation(s)
- Nikola Kresojević
- Neurology Clinic, CCS, School of Medicine, University of Belgrade, Dr Subotića starijeg 6, 11000 Belgrade, Serbia
| | - Valerija Dobričić
- Neurology Clinic, CCS, School of Medicine, University of Belgrade, Dr Subotića starijeg 6, 11000 Belgrade, Serbia
| | - Marina Svetel
- Neurology Clinic, CCS, School of Medicine, University of Belgrade, Dr Subotića starijeg 6, 11000 Belgrade, Serbia
| | - Vladimir Kostić
- Neurology Clinic, CCS, School of Medicine, University of Belgrade, Dr Subotića starijeg 6, 11000 Belgrade, Serbia.
| |
Collapse
|
396
|
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
|
397
|
Abstract
The basal ganglia were originally thought to be associated purely with motor control. However, dysfunction and pathology of different regions and circuits are now known to give rise to many clinical manifestations beyond the association of basal ganglia dysfunction with movement disorders. Moreover, disorders that were thought to be caused by dysfunction of the basal ganglia only, such as Parkinson's disease and Huntington's disease, have diverse abnormalities distributed not only in the brain but also in the peripheral and autonomic nervous systems; this knowledge poses new questions and challenges. We discuss advances and the unanswered questions, and ways in which progress might be made.
Collapse
Affiliation(s)
- Jose A Obeso
- Movement Disorders Laboratory, Department of Neurology and Neuroscience Area, Clínica Universitaria and Medical School, and CIMA, University of Navarra, Pamplona, Spain; Centro de Investigación en Redes sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Maria C Rodriguez-Oroz
- Centro de Investigación en Redes sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Department of Neurology, University Hospital Donostia and Neuroscience Unit BioDonostia Research Institute, San Sebastian, Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Maria Stamelou
- Movement Disorders Clinic, Second Department of Neurology, Attiko Hospital, University of Athens, Greece; Sobell Department of Motor Neurosciences and Movement Disorders, UCL Institute of Neurology, London, UK
| | - Kailash P Bhatia
- Sobell Department of Motor Neurosciences and Movement Disorders, UCL Institute of Neurology, London, UK
| | - David J Burn
- Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
398
|
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
|
399
|
Lysosomal NEU1 deficiency affects amyloid precursor protein levels and amyloid-β secretion via deregulated lysosomal exocytosis. Nat Commun 2014; 4:2734. [PMID: 24225533 PMCID: PMC4015463 DOI: 10.1038/ncomms3734] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 10/09/2013] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD) belongs to a category of adult neurodegenerative conditions which are associated with intracellular and extracellular accumulation of neurotoxic protein aggregates. Understanding how these aggregates are formed, secreted and propagated by neurons has been the subject of intensive research, but so far no preventive or curative therapy for AD is available and clinical trials have been largely unsuccessful. Here we show that deficiency of the lysosomal sialidase NEU1 leads to the spontaneous occurrence of an AD-like amyloidogenic process in mice. This involves two consecutive events linked to NEU1 loss-of-function – accumulation and amyloidogenic processing of an oversialylated amyloid precursor protein in lysosomes, and extracellular release of Aβ-peptides by excessive lysosomal exocytosis. Furthermore, cerebral injection of NEU1 in an established AD mouse model substantially reduces β-amyloid plaques. Our findings identify an additional pathway for the secretion of Aβ and define NEU1 as a potential therapeutic molecule for AD.
Collapse
|
400
|
Moore SF, Barker RA. Predictors of Parkinson's disease dementia: towards targeted therapies for a heterogeneous disease. Parkinsonism Relat Disord 2014; 20 Suppl 1:S104-7. [PMID: 24262158 DOI: 10.1016/s1353-8020(13)70026-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Parkinson's disease dementia (PDD) has become an increasing area of research as treatments for the motor features of Parkinson's disease (PD) have improved and the population of patients with PD grows and ages. If predictors could be used to identify a sub-population of patients at risk of developing an early PDD then research into its neuropathological basis and treatment could be more effectively targeted to specific individuals. At present the predictors with the most evidence have arisen from longitudinal studies tracking the development of dementia in populations of incident, newly diagnosed patients with PD. Evidence exists for predictors across multiple domains: clinical, biological, neuroimaging and genetic. Some of the most robust of these suggest that PDD may develop as the result of an age and tau dependent, posterior cortically based process driven in some cases by mutations in the gene for glucocerebrosidase (GBA). It is clear, though, that more research needs to be undertaken into finding reliable predictors of PDD. At present the best approach may be to combine a set of predictors already identified in order to provide a basis for understanding why and how it occurs. Through this, new therapeutic strategies may emerge.
Collapse
Affiliation(s)
- Sarah F Moore
- John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, CB2 0PY, UK.
| | | |
Collapse
|