201
|
Chen Y, Sam R, Sharma P, Chen L, Do J, Sidransky E. Glucocerebrosidase as a therapeutic target for Parkinson's disease. Expert Opin Ther Targets 2020; 24:287-294. [PMID: 32106725 PMCID: PMC7113099 DOI: 10.1080/14728222.2020.1733970] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 02/20/2020] [Indexed: 12/21/2022]
Abstract
Introduction: The association between Gaucher disease, caused by the inherited deficiency of glucocerebrosidase, and Parkinson's disease was first recognized in the clinic, noting that patients with Gaucher disease and their carrier relatives had an increased incidence of Parkinson's disease. Currently, mutations in glucocerebrosidase (GBA1) are the most common genetic risk factor for Parkinson's disease and dementia with Lewy bodies, with an inverse relationship between glucocerebrosidase and α-synuclein, a key factor in Parkinson pathogenesis. The hypothesis that therapeutic enhancement of brain glucocerebrosidase levels might reduce the aggregation, accumulation or spread of α-synuclein has spurred great interest in glucocerebrosidase as a novel therapeutic target.Area covered: This article explores the potential molecular mechanisms underlying the association between GBA1 mutations and Parkinson's disease and outlines therapeutic strategies to increase brain glucocerebrosidase, including gene therapy, targeted delivery of recombinant glucocerebrosidase to the brain, small-molecule chaperones to rescue mutant glucocerebrosidase, and small-molecule modulators to activate wild-type glucocerebrosidase.Expert opinion: Although an improved understanding of the mechanistic basis for GBA1-associated parkinsonism is essential, enhancing levels of brain glucocerebrosidase may have wide therapeutic implications. While gene therapy may ultimately be effective, less expensive and invasive small-molecule non-inhibitory chaperones or activators could significantly impact the disease course.
Collapse
Affiliation(s)
- Yu Chen
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Richard Sam
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Pankaj Sharma
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Lu Chen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Jenny Do
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ellen Sidransky
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
202
|
Valdez C, Ysselstein D, Young TJ, Zheng J, Krainc D. Progranulin mutations result in impaired processing of prosaposin and reduced glucocerebrosidase activity. Hum Mol Genet 2020; 29:716-726. [PMID: 31600775 PMCID: PMC7104673 DOI: 10.1093/hmg/ddz229] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/22/2019] [Accepted: 09/23/2019] [Indexed: 02/05/2023] Open
Abstract
Frontotemporal dementia (FTD) is a common neurogenerative disorder characterized by progressive degeneration in the frontal and temporal lobes. Heterozygous mutations in the gene encoding progranulin (PGRN) are a common genetic cause of FTD. Recently, PGRN has emerged as an important regulator of lysosomal function. Here, we examine the impact of PGRN mutations on the processing of full-length prosaposin to individual saposins, which are critical regulators of lysosomal sphingolipid metabolism. Using FTD-PGRN patient-derived cortical neurons differentiated from induced pluripotent stem cells, as well as post-mortem tissue from patients with FTLD-PGRN, we show that PGRN haploinsufficiency results in impaired processing of prosaposin to saposin C, a critical activator of the lysosomal enzyme glucocerebrosidase (GCase). Additionally, we found that PGRN mutant neurons had reduced lysosomal GCase activity, lipid accumulation and increased insoluble α-synuclein relative to isogenic controls. Importantly, reduced GCase activity in PGRN mutant neurons is rescued by treatment with saposin C. Together, these findings suggest that reduced GCase activity due to impaired processing of prosaposin may contribute to pathogenesis of FTD resulting from PGRN mutations.
Collapse
Affiliation(s)
- Clarissa Valdez
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Daniel Ysselstein
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tiffany J Young
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jianbin Zheng
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dimitri Krainc
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
203
|
Miliukhina IV, Usenko TS, Senkevich KA, Nikolaev MA, Timofeeva AA, Agapova EA, Semenov AV, Lubimova NE, Totolyan AA, Pchelina SN. Plasma Cytokines Profile in Patients with Parkinson's Disease Associated with Mutations in GBA Gene. Bull Exp Biol Med 2020; 168:423-426. [PMID: 32146630 DOI: 10.1007/s10517-020-04723-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Indexed: 10/24/2022]
Abstract
Plasma cytokine concentration in patients with Parkinson's disease and mutation in GBA gene, in patients with sporadic Parkinson's disease, and in healthy volunteers were measured by ELISA and multiplex analysis. In patients with Parkinson's disease and mutation in GBA gene, elevated plasma concentrations of IL-1β and TNFα were revealed by ELISA in comparison with both controls and patients with sporadic form of Parkinson's disease. Multiplex analysis revealed enhanced secretion of IL-1β, IL-2, IFNγ and reduced plasma levels of monocyte chemoattractant protein-1 (MCP-1) in patients with Parkinson's disease and mutation in GBA gene (in comparison with other groups) and increased plasma levels of IL-13 (only in comparison with the healthy volunteers). Our results support the hypothesis that the concentrations of inflammatory mediators are increased in patients with Parkinson's disease and mutation in GBA gene.
Collapse
Affiliation(s)
- I V Miliukhina
- Institute of Experimental Medicine, St. Petersburg, Russia.
- Ministry of Health of Russian Federation, I. P. Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia.
- B. P. Konstantinov St. Petersburg Nuclear Physics Institute, National Research Center Kurchatov Institute, Gatchina, Leningrad region, Russia.
| | - T S Usenko
- Ministry of Health of Russian Federation, I. P. Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia
- B. P. Konstantinov St. Petersburg Nuclear Physics Institute, National Research Center Kurchatov Institute, Gatchina, Leningrad region, Russia
| | - K A Senkevich
- Institute of Experimental Medicine, St. Petersburg, Russia
- Ministry of Health of Russian Federation, I. P. Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia
- B. P. Konstantinov St. Petersburg Nuclear Physics Institute, National Research Center Kurchatov Institute, Gatchina, Leningrad region, Russia
| | - M A Nikolaev
- Ministry of Health of Russian Federation, I. P. Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia
- B. P. Konstantinov St. Petersburg Nuclear Physics Institute, National Research Center Kurchatov Institute, Gatchina, Leningrad region, Russia
| | - A A Timofeeva
- Ministry of Health of Russian Federation, I. P. Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia
| | - E A Agapova
- B. P. Konstantinov St. Petersburg Nuclear Physics Institute, National Research Center Kurchatov Institute, Gatchina, Leningrad region, Russia
| | - A V Semenov
- Ministry of Health of Russian Federation, I. P. Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia
- Pasteur St. Petersburg Research Institute of Epidemiology and Microbiology, St. Petersburg, Russia
| | - N E Lubimova
- Pasteur St. Petersburg Research Institute of Epidemiology and Microbiology, St. Petersburg, Russia
| | - A A Totolyan
- Ministry of Health of Russian Federation, I. P. Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia
- Pasteur St. Petersburg Research Institute of Epidemiology and Microbiology, St. Petersburg, Russia
| | - S N Pchelina
- Institute of Experimental Medicine, St. Petersburg, Russia
- Ministry of Health of Russian Federation, I. P. Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia
- B. P. Konstantinov St. Petersburg Nuclear Physics Institute, National Research Center Kurchatov Institute, Gatchina, Leningrad region, Russia
| |
Collapse
|
204
|
Gurda BL, Vite CH. Large animal models contribute to the development of therapies for central and peripheral nervous system dysfunction in patients with lysosomal storage diseases. Hum Mol Genet 2020; 28:R119-R131. [PMID: 31384936 DOI: 10.1093/hmg/ddz127] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 04/16/2019] [Accepted: 06/07/2019] [Indexed: 12/12/2022] Open
Abstract
Lysosomal storage diseases (LSDs) are a group of 70 monogenic disorders characterized by the lysosomal accumulation of a substrate. As a group, LSDs affect ~1 in 5000 live births; however, each individual storage disease is rare, limiting the ability to perform natural history studies or to perform clinical trials. Perhaps in no other biomedical field have naturally occurring large animal (canine, feline, ovine, caprine, and bovine) models been so essential for understanding the fundamentals of disease pathogenesis and for developing safe and effective therapies. These models were critical for the development of hematopoietic stem cell transplantation in α- and β- mannosidosis, fucosidosis, and the mucopolysaccharidoses; enzyme replacement therapy for fucosidosis, the mucopolysaccharidoses, and neuronal ceroid lipofuscinosis; and small molecule therapy in Niemann-Pick type C disease. However, their most notable contributions to the biomedical field are in the development of gene therapy for LSDs. Adeno-associated viral vectors to treat nervous system disease have been evaluated in the large animal models of α-mannosidosis, globoid cell leukodystrophy, GM1 and GM2 gangliosidosis, the mucopolysaccharidoses, and neuronal ceroid lipofuscinosis. This review article will summarize the large animal models available for study as well as their contributions to the development of central and peripheral nervous system dysfunction in LSDs.
Collapse
Affiliation(s)
- Brittney L Gurda
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles H Vite
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
205
|
Challis C, Hori A, Sampson TR, Yoo BB, Challis RC, Hamilton AM, Mazmanian SK, Volpicelli-Daley LA, Gradinaru V. Gut-seeded α-synuclein fibrils promote gut dysfunction and brain pathology specifically in aged mice. Nat Neurosci 2020; 23:327-336. [PMID: 32066981 PMCID: PMC7065967 DOI: 10.1038/s41593-020-0589-7] [Citation(s) in RCA: 263] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 01/09/2020] [Indexed: 01/02/2023]
Abstract
Parkinson's disease is a synucleinopathy that is characterized by motor dysfunction, death of midbrain dopaminergic neurons and accumulation of α-synuclein (α-Syn) aggregates. Evidence suggests that α-Syn aggregation can originate in peripheral tissues and progress to the brain via autonomic fibers. We tested this by inoculating the duodenal wall of mice with α-Syn preformed fibrils. Following inoculation, we observed gastrointestinal deficits and physiological changes to the enteric nervous system. Using the AAV-PHP.S capsid to target the lysosomal enzyme glucocerebrosidase for peripheral gene transfer, we found that α-Syn pathology is reduced due to the increased expression of this protein. Lastly, inoculation of α-Syn fibrils in aged mice, but not younger mice, resulted in progression of α-Syn histopathology to the midbrain and subsequent motor defects. Our results characterize peripheral synucleinopathy in prodromal Parkinson's disease and explore cellular mechanisms for the gut-to-brain progression of α-Syn pathology.
Collapse
Affiliation(s)
- Collin Challis
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Acacia Hori
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Timothy R Sampson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Bryan B Yoo
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Rosemary C Challis
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Adam M Hamilton
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sarkis K Mazmanian
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Laura A Volpicelli-Daley
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
206
|
Systematic review of genetic variants associated with cognitive impairment and depressive symptoms in Parkinson's disease. Acta Neuropsychiatr 2020; 32:10-22. [PMID: 31292011 DOI: 10.1017/neu.2019.28] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Cognitive impairment and depression are among the most prevalent and most disabling non-motor symptoms in Parkinson's disease (PD). The genetic factors that are associated with these symptoms remain uncertain. This systematic review aims to summarise the prevailing evidence from all genetic association studies investigating the genetic variants associated with cognitive impairment and depressive symptoms in people with PD. METHOD A systematic review using five online databases: PubMed, PsycINFO, CINAHL, EMBASE and OpenGrey (PROSPERO protocol: CRD42017067431). We completed the quality assessment using the Q-Genie tool. RESULTS 2353 articles were screened, and 43 articles were found to be eligible to be included. A meta-analysis of studies investigating LRRK2 rs34637584 confirmed that the minor allele carriers had significantly less cognitive impairment (p = 0.015). Further meta-analyses showed that GBA variants rs76763715 (p < 0.001) and rs421016 (p = 0.001) were significantly associated with more cognitive impairment in people with PD. Minor alleles of GBA variants rs76763715, rs421016, rs387906315 and rs80356773 were associated with more depressive symptoms in PD. Moreover, APOE ε4 allele has been associated with more cognitive impairment in PD. BDNF (rs6265) and CRY1 (rs2287161) variants have been associated with more depressive symptoms in people with PD. CONCLUSIONS PD carriers of GBA variants are at high risk for cognitive decline and depression. Screening for these variants may facilitate early identification and effective management of these non-motor symptoms. The molecular mechanisms underlying favourable cognitive functioning in LRRK2 rs34637584 variant carriers warrant further investigation.
Collapse
|
207
|
Ji S, Wang C, Qiao H, Gu Z, Gan-Or Z, Fon EA, Chan P. Decreased Penetrance of Parkinson's Disease in Elderly Carriers of Glucocerebrosidase Gene L444P/R Mutations: A Community-Based 10-Year Longitudinal Study. Mov Disord 2020; 35:672-678. [PMID: 31912918 DOI: 10.1002/mds.27971] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/04/2019] [Accepted: 12/13/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Heterozygous mutations in the glucocerebrosidase gene (GBA) have been shown to be an important genetic risk factor for Parkinson's disease (PD) worldwide. However, the penetrance of GBA heterozygote for L444P, the common mutation for Asian population, is not known in older Chinese people. OBJECTIVES To assess the conversion rate to PD in identified carriers of GBA L444P/R mutations in Chinese community-dwelling older adults. METHODS The GBA gene was sequenced for mutations at position 444 in 8405 people older than 55 years who participated in the Beijing Longitudinal Study on Aging II cohort. Nine subjects were identified as heterozygous carriers of GBA L444P or L444R mutations at baseline and clinically followed up from 2009 to 2019 to investigate their PD conversion, motor and nonmotor symptoms, and change of vesicular monoamine transporter type 2 using tracer of [18 F]9-fluoropropyl-(+)-dihydrotetrabenazine (18 F-DTBZ, also known as 18 F-AV-133). RESULTS Eight heterozygous GBA L444P and 1 L444R mutation carriers were identified without PD at baseline, and none of them developed clinical parkinsonism after a 10-year follow-up. CONCLUSIONS Although GBA mutations may lead to an earlier onset PD, the majority of GBA L444P heterozygotes in older adults may not convert to PD. Further studies are warranted to identify factors that modify the risk of conversion. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Shaozhen Ji
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Chaodong Wang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Clinical and Research Center for Parkinson's Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory for Parkinson's Disease, Parkinson Disease Center of Beijing Institute for Brain Disorders, Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Hongwen Qiao
- Department of Nuclear Medicine, Xuanwu Hospital of Capital Medical University, Beijing, China.,Department of Geriatrics, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Zhuqin Gu
- Clinical and Research Center for Parkinson's Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory for Parkinson's Disease, Parkinson Disease Center of Beijing Institute for Brain Disorders, Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ziv Gan-Or
- Montreal Neurological Institute and Hospital, Department of Neurology & Neurosurgery, McGill University, Montreal, Canada
| | - Edward A Fon
- Montreal Neurological Institute and Hospital, Department of Neurology & Neurosurgery, McGill University, Montreal, Canada
| | - Piu Chan
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Clinical and Research Center for Parkinson's Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory for Parkinson's Disease, Parkinson Disease Center of Beijing Institute for Brain Disorders, Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China.,Department of Geriatrics, Xuanwu Hospital of Capital Medical University, Beijing, China
| |
Collapse
|
208
|
Nielsen JJJ, Lillethorup TP, Glud AN, Sørensen JCH, Orlowski D. The application of iPSCs in Parkinson’s disease. Acta Neurobiol Exp (Wars) 2020. [DOI: 10.21307/ane-2020-024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
209
|
Caiazza MC, Lang C, Wade-Martins R. What we can learn from iPSC-derived cellular models of Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2019; 252:3-25. [PMID: 32247368 DOI: 10.1016/bs.pbr.2019.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Parkinson's disease (PD) is an age-related neurodegenerative disorder with no known cure. In order to better understand the pathological mechanisms which lead to neuronal cell death and to accelerate the process of drug discovery, a reliable in vitro model is required. Unfortunately, research into PD and neurodegeneration in general has long suffered from a lack of adequate in vitro models, mainly due to the inaccessibility of live neurons from vulnerable areas of the human brain. Recent reprogramming technologies have recently made it possible to reliably derive human induced pluripotent stem cells (iPSCs) from patients and healthy subjects to generate specific, difficult to obtain, cellular sub-types. These iPSC-derived cells can be employed to model disease to better understand pathological mechanisms and underlying cellular vulnerability. Therefore, in this chapter, we will discuss the techniques involved in the reprogramming of somatic cells into iPSCs, the evolution of iPSC differentiation methods and their application in neurodegenerative disease modeling.
Collapse
Affiliation(s)
- Maria Claudia Caiazza
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Charmaine Lang
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
210
|
Schindlbeck KA, Vo A, Nguyen N, Tang CC, Niethammer M, Dhawan V, Brandt V, Saunders-Pullman R, Bressman SB, Eidelberg D. LRRK2 and GBA Variants Exert Distinct Influences on Parkinson's Disease-Specific Metabolic Networks. Cereb Cortex 2019; 30:2867-2878. [PMID: 31813991 DOI: 10.1093/cercor/bhz280] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/23/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022] Open
Abstract
The natural history of idiopathic Parkinson's disease (PD) varies considerably across patients. While PD is generally sporadic, there are known genetic influences: the two most common, mutations in the LRRK2 or GBA1 gene, are associated with slower and more aggressive progression, respectively. Here, we applied graph theory to metabolic brain imaging to understand the effects of genotype on the organization of previously established PD-specific networks. We found that closely matched PD patient groups with the LRRK2-G2019S mutation (PD-LRRK2) or GBA1 variants (PD-GBA) expressed the same disease networks as sporadic disease (sPD), but PD-LRRK2 and PD-GBA patients exhibited abnormal increases in network connectivity that were not present in sPD. Using a community detection strategy, we found that the location and modular distribution of these connections differed strikingly across genotypes. In PD-LRRK2, connections were gained within the network core, with the formation of distinct functional pathways linking the cerebellum and putamen. In PD-GBA, by contrast, the majority of functional connections were formed outside the core, involving corticocortical pathways at the network periphery. Strategically localized connections within the core in PD-LRRK2 may maintain PD network activity at lower levels than in PD-GBA, resulting in a less aggressive clinical course.
Collapse
Affiliation(s)
- Katharina A Schindlbeck
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY 10030, USA
| | - An Vo
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY 10030, USA
| | - Nha Nguyen
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Chris C Tang
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY 10030, USA
| | - Martin Niethammer
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY 10030, USA
| | - Vijay Dhawan
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY 10030, USA
| | - Vicky Brandt
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY 10030, USA
| | - Rachel Saunders-Pullman
- Department of Neurology, Mount Sinai Beth Israel, Mount Sinai Hospital, New York, NY 10003, USA
| | - Susan B Bressman
- Department of Neurology, Mount Sinai Beth Israel, Mount Sinai Hospital, New York, NY 10003, USA
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY 10030, USA
| |
Collapse
|
211
|
Ysselstein D, Nguyen M, Young TJ, Severino A, Schwake M, Merchant K, Krainc D. LRRK2 kinase activity regulates lysosomal glucocerebrosidase in neurons derived from Parkinson's disease patients. Nat Commun 2019; 10:5570. [PMID: 31804465 PMCID: PMC6895201 DOI: 10.1038/s41467-019-13413-w] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 11/07/2019] [Indexed: 12/22/2022] Open
Abstract
Mutations in LRRK2 and GBA1 are common genetic risk factors for Parkinson's disease (PD) and major efforts are underway to develop new therapeutics that target LRRK2 or glucocerebrosidase (GCase). Here we describe a mechanistic and therapeutic convergence of LRRK2 and GCase in neurons derived from patients with PD. We find that GCase activity was reduced in dopaminergic (DA) neurons derived from PD patients with LRRK2 mutations. Inhibition of LRRK2 kinase activity results in increased GCase activity in DA neurons with either LRRK2 or GBA1 mutations. This increase is sufficient to partially rescue accumulation of oxidized dopamine and alpha-synuclein in PD patient neurons. We have identified the LRRK2 substrate Rab10 as a key mediator of LRRK2 regulation of GCase activity. Together, these results suggest an important role of mutant LRRK2 as a negative regulator of lysosomal GCase activity.
Collapse
Affiliation(s)
- Daniel Ysselstein
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, IL, 60611, USA
| | - Maria Nguyen
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, IL, 60611, USA
| | - Tiffany J Young
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, IL, 60611, USA
| | - Alex Severino
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, IL, 60611, USA
| | - Michael Schwake
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, IL, 60611, USA
| | - Kalpana Merchant
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, IL, 60611, USA
| | - Dimitri Krainc
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, IL, 60611, USA.
| |
Collapse
|
212
|
Graham OEE, Pitcher TL, Liau Y, Miller AL, Dalrymple-Alford JC, Anderson TJ, Kennedy MA. Nanopore sequencing of the glucocerebrosidase (GBA) gene in a New Zealand Parkinson's disease cohort. Parkinsonism Relat Disord 2019; 70:36-41. [PMID: 31809948 DOI: 10.1016/j.parkreldis.2019.11.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/22/2019] [Accepted: 11/25/2019] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Bi-allelic mutations in the gene for glucocerebrosidase (GBA) cause Gaucher disease, an autosomal recessive lysosomal storage disorder. Gaucher disease causing GBA mutations in the heterozygous state are also high risk factors for Parkinson's disease (PD). GBA analysis is challenging due to a related pseudogene and structural variations (SVs) that can occur at this locus. We have applied and refined a recently developed nanopore DNA sequencing method to analyze GBA variants in a clinically assessed New Zealand longitudinal cohort of PD. METHOD We examined amplicons encompassing the coding region of GBA (8.9 kb) from 229 PD cases and 50 healthy controls using the GridION nanopore sequencing platform, and Sanger validation. RESULTS We detected 23 variants in 21 PD cases (9.2% of patients). We detected modest PD risk variant p.N409S (rs76763715) in one case, p.E365K (rs2230288) in 12 cases, and p.T408 M (rs75548401) in seven cases, one of whom also had p.E365K. We additionally detected the possible risk variants p.R78C (rs146774384) in one case, p.D179H (rs147138516) in one case which occurred on the same haplotype as p.E365K, and one novel variant c.335C > T or p.(L335 = ), that potentially impacts splicing of GBA transcripts. Additionally, we found a higher prevalence of dementia among patients with GBA variants. CONCLUSION This work confirmed the utility of nanopore sequencing as a high-throughput method to identify known and novel GBA variants, and to assign precise haplotypes. Our observations may contribute to improved understanding of the effects of variants on disease pathogenesis, and to the development of more targeted treatments.
Collapse
Affiliation(s)
- O E E Graham
- Gene Structure and Function Laboratory, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand.
| | - T L Pitcher
- Department of Medicine, University of Otago, Christchurch, New Zealand; New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Y Liau
- Gene Structure and Function Laboratory, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - A L Miller
- Gene Structure and Function Laboratory, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - J C Dalrymple-Alford
- New Zealand Brain Research Institute, Christchurch, New Zealand; School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
| | - T J Anderson
- Department of Medicine, University of Otago, Christchurch, New Zealand; New Zealand Brain Research Institute, Christchurch, New Zealand; Neurology Department, Christchurch Hospital, Christchurch, New Zealand
| | - M A Kennedy
- Gene Structure and Function Laboratory, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| |
Collapse
|
213
|
Zheng YQ, Zhang Y, Yau Y, Zeighami Y, Larcher K, Misic B, Dagher A. Local vulnerability and global connectivity jointly shape neurodegenerative disease propagation. PLoS Biol 2019; 17:e3000495. [PMID: 31751329 PMCID: PMC6894889 DOI: 10.1371/journal.pbio.3000495] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 12/05/2019] [Accepted: 10/31/2019] [Indexed: 01/06/2023] Open
Abstract
It is becoming increasingly clear that brain network organization shapes the course and expression of neurodegenerative diseases. Parkinson disease (PD) is marked by progressive spread of atrophy from the midbrain to subcortical structures and, eventually, to the cerebral cortex. Recent discoveries suggest that the neurodegenerative process involves the misfolding and prion-like propagation of endogenous α-synuclein via axonal projections. However, the mechanisms that translate local "synucleinopathy" to large-scale network dysfunction and atrophy remain unknown. Here, we use an agent-based epidemic spreading model to integrate structural connectivity, functional connectivity, and gene expression and to predict sequential volume loss due to neurodegeneration. The dynamic model replicates the spatial and temporal patterning of empirical atrophy in PD and implicates the substantia nigra as the disease epicenter. We reveal a significant role for both connectome topology and geometry in shaping the distribution of atrophy. The model also demonstrates that SNCA and GBA transcription influence α-synuclein concentration and local regional vulnerability. Functional coactivation further amplifies the course set by connectome architecture and gene expression. Altogether, these results support the theory that the progression of PD is a multifactorial process that depends on both cell-to-cell spreading of misfolded proteins and regional vulnerability.
Collapse
Affiliation(s)
- Ying-Qiu Zheng
- McConnell Brain Imaging Centre, Montréal Neurological Institute, McGill University, Montréal, Quebec, Canada
| | - Yu Zhang
- McConnell Brain Imaging Centre, Montréal Neurological Institute, McGill University, Montréal, Quebec, Canada
- Centre de Recherche de I'Institut Universitaire de Gériatrie de Montréal, Montréal, Canada
| | - Yvonne Yau
- McConnell Brain Imaging Centre, Montréal Neurological Institute, McGill University, Montréal, Quebec, Canada
| | - Yashar Zeighami
- McConnell Brain Imaging Centre, Montréal Neurological Institute, McGill University, Montréal, Quebec, Canada
| | - Kevin Larcher
- McConnell Brain Imaging Centre, Montréal Neurological Institute, McGill University, Montréal, Quebec, Canada
| | - Bratislav Misic
- McConnell Brain Imaging Centre, Montréal Neurological Institute, McGill University, Montréal, Quebec, Canada
- * E-mail: (BM); (AD)
| | - Alain Dagher
- McConnell Brain Imaging Centre, Montréal Neurological Institute, McGill University, Montréal, Quebec, Canada
- * E-mail: (BM); (AD)
| |
Collapse
|
214
|
Pozzilli V, Giona F, Ceccanti M, Cambieri C, Frasca V, Onesti E, Libonati L, Di Bari S, Fiorini I, Cardarelli L, Santopietro M, Inghilleri M. A case of motor neuron involvement in Gaucher disease. Mol Genet Metab Rep 2019; 21:100540. [PMID: 31844629 PMCID: PMC6895677 DOI: 10.1016/j.ymgmr.2019.100540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/01/2019] [Accepted: 11/03/2019] [Indexed: 12/13/2022] Open
Abstract
Gaucher disease (GD) is a genetic disorder characterized by an accumulation of glucosylceramide in cells in the monocyte-macrophage system. We describe a case of a 33-year-old man with a previous diagnosis of type 3 GD who displayed a progressive weakening of the limbs followed by upper motor neuron involvement. A diagnosis of definite Amyotrophic Lateral Sclerosis was made. This is the first reported case of concurrent Gaucher disease and the ALS phenotype in the same patient.
Collapse
Affiliation(s)
- V Pozzilli
- Rare Neuromuscular Diseases Centre, Department of Human Neurosciences, Sapienza University, Rome, Italy
| | - F Giona
- Haematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - M Ceccanti
- Rare Neuromuscular Diseases Centre, Department of Human Neurosciences, Sapienza University, Rome, Italy
| | - C Cambieri
- Rare Neuromuscular Diseases Centre, Department of Human Neurosciences, Sapienza University, Rome, Italy
| | - V Frasca
- Rare Neuromuscular Diseases Centre, Department of Human Neurosciences, Sapienza University, Rome, Italy
| | - E Onesti
- Rare Neuromuscular Diseases Centre, Department of Human Neurosciences, Sapienza University, Rome, Italy
| | - L Libonati
- Rare Neuromuscular Diseases Centre, Department of Human Neurosciences, Sapienza University, Rome, Italy
| | - S Di Bari
- Rare Neuromuscular Diseases Centre, Department of Human Neurosciences, Sapienza University, Rome, Italy
| | - I Fiorini
- Rare Neuromuscular Diseases Centre, Department of Human Neurosciences, Sapienza University, Rome, Italy
| | - L Cardarelli
- Haematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - M Santopietro
- Haematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - M Inghilleri
- Rare Neuromuscular Diseases Centre, Department of Human Neurosciences, Sapienza University, Rome, Italy
| |
Collapse
|
215
|
Huebecker M, Moloney EB, van der Spoel AC, Priestman DA, Isacson O, Hallett PJ, Platt FM. Reduced sphingolipid hydrolase activities, substrate accumulation and ganglioside decline in Parkinson's disease. Mol Neurodegener 2019; 14:40. [PMID: 31703585 PMCID: PMC6842240 DOI: 10.1186/s13024-019-0339-z] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/20/2019] [Indexed: 12/20/2022] Open
Abstract
Background Haploinsufficiency in the Gaucher disease GBA gene, which encodes the lysosomal glucocerebrosidase GBA, and ageing represent major risk factors for developing Parkinson’s disease (PD). Recently, more than fifty other lysosomal storage disorder gene variants have been identified in PD, implicating lysosomal dysfunction more broadly as a key risk factor for PD. Despite the evidence of multiple lysosomal genetic risks, it remains unclear how sphingolipid hydrolase activities, other than GBA, are altered with ageing or in PD. Moreover, it is not fully known if levels of glycosphingolipid substrates for these enzymes change in vulnerable brain regions of PD. Finally, little is known about the levels of complex gangliosides in substantia nigra which may play a significant role in ageing and PD. Methods To study sphingolipid hydrolase activities and glycosphingolipid expression in ageing and in PD, two independent cohorts of human substantia nigra tissues were obtained. Fluorescent 4-methylumbelliferone assays were used to determine multiple enzyme activities. The lysosomal GBA and non-lysosomal GBA2 activities were distinguished using the inhibitor NB-DGJ. Sensitive and quantitative normal-phase HPLC was performed to study glycosphingolipid levels. In addition, glycosphingolipid levels in cerebrospinal fluid and serum were analysed as possible biomarkers for PD. Results The present study demonstrates, in two independent cohorts of human post-mortem substantia nigra, that sporadic PD is associated with deficiencies in multiple lysosomal hydrolases (e.g. α-galactosidase and β-hexosaminidase), in addition to reduced GBA and GBA2 activities and concomitant glycosphingolipid substrate accumulation. Furthermore, the data show significant reductions in levels of complex gangliosides (e.g. GM1a) in substantia nigra, CSF and serum in ageing, PD, and REM sleep behaviour disorder, which is a strong predictor of PD. Conclusions These findings conclusively demonstrate reductions in GBA activity in the parkinsonian midbrain, and for the first time, reductions in the activity of several other sphingolipid hydrolases. Furthermore, significant reductions were seen in complex gangliosides in PD and ageing. The diminished activities of these lysosomal hydrolases, the glycosphingolipid substrate accumulation, and the reduced levels of complex gangliosides are likely major contributors to the primary development of the pathology seen in PD and related disorders with age.
Collapse
Affiliation(s)
- Mylene Huebecker
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Elizabeth B Moloney
- Neuroregeneration Institute, McLean Hospital / Harvard Medical School, Belmont, MA, 02478, USA
| | - Aarnoud C van der Spoel
- Departments of Pediatrics and Biochemistry & Molecular Biology, Atlantic Research Centre, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - David A Priestman
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Ole Isacson
- Neuroregeneration Institute, McLean Hospital / Harvard Medical School, Belmont, MA, 02478, USA.
| | - Penelope J Hallett
- Neuroregeneration Institute, McLean Hospital / Harvard Medical School, Belmont, MA, 02478, USA.
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK.
| |
Collapse
|
216
|
Schneider SA, Tahirovic S, Hardy J, Strupp M, Bremova-Ertl T. Do heterozygous mutations of Niemann-Pick type C predispose to late-onset neurodegeneration: a review of the literature. J Neurol 2019; 268:2055-2064. [PMID: 31701332 DOI: 10.1007/s00415-019-09621-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/30/2019] [Accepted: 11/02/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND/METHODS Monogenic diseases are important models for the study of neurodegenerative diseases, such as Parkinson's disease (PD) and dementia. Notably, for some disorders, homozygosity is associated with a complex metabolic disease, while heterozygosity predisposes to late-onset neurodegeneration. For instance, biallelic glucocerebrosidase gene mutations cause Gaucher's disease, while heterozygous mutations are a common genetic risk factor for late-onset PD. Little is known about similar risks of related diseases, such as Niemann-Pick type C (NPC). Given that both conditions map into related, i.e., lysosomal, pathways, we hypothesize a similar risk of single-NPC gene mutations. Indeed, there is increasing evidence based on clinical observations in humans and animal studies. Here we review the current knowledge of NPC heterozygosity. RESULTS Family history studies suggest a high proportion of late-onset neurodegenerative diseases in NPC families. We identified 19 cases with heterozygous NPC mutations in the literature who presented with a neurodegenerative disease, including levodopa-responsive PD, atypical parkinsonism (PSP, CBD), dystonia or dementia with a mean age at onset of about 57 years (range 8-87). Consistent splenomegaly and mildly abnormal filipin staining results have also been reported in heterozygous gene mutation carriers. Imaging and pathological data support this notion. DISCUSSION/CONCLUSION This finding has wider implications in so far as NPC-related forms of Parkinsonian syndromes, dementia, motor neuron disease and other neurodegenerative disorders may benefit from NPC-mechanistic therapies, in particular related to lysosomal dysfunction. Further research is warranted to generate systematic data of heterozygous mutation carriers, including longitudinal data.
Collapse
Affiliation(s)
- Susanne A Schneider
- Department of Neurology, Ludwig-Maximilians-University, Marchioninistr 15, 81377, Munich, Germany.
| | - Sabina Tahirovic
- German Center for Neurodegenerative Diseases (DZNE) Within the Helmholtz Association, Feodor-Lynen-Strasse 17, Munich, Germany
| | - John Hardy
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Michael Strupp
- Department of Neurology, Ludwig-Maximilians-University, Marchioninistr 15, 81377, Munich, Germany
| | - Tatiana Bremova-Ertl
- Department of Neurology, Ludwig-Maximilians-University, Marchioninistr 15, 81377, Munich, Germany.,Department of Neurology, Inselspital, University Hospital Bern, Bern, Switzerland
| |
Collapse
|
217
|
The Overcrowded Crossroads: Mitochondria, Alpha-Synuclein, and the Endo-Lysosomal System Interaction in Parkinson's Disease. Int J Mol Sci 2019; 20:ijms20215312. [PMID: 31731450 PMCID: PMC6862467 DOI: 10.3390/ijms20215312] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/21/2019] [Accepted: 10/24/2019] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder worldwide, mainly affecting the elderly. The disease progresses gradually, with core motor presentations and a multitude of non-motor manifestations. There are two neuropathological hallmarks of PD, the dopaminergic neuronal loss and the alpha-synuclein-containing Lewy body inclusions in the substantia nigra. While the exact pathomechanisms of PD remain unclear, genetic investigations have revealed evidence of the involvement of mitochondrial function, alpha-synuclein (α-syn) aggregation, and the endo-lysosomal system, in disease pathogenesis. Due to the high energy demand of dopaminergic neurons, mitochondria are of special importance acting as the cellular powerhouse. Mitochondrial dynamic fusion and fission, and autophagy quality control keep the mitochondrial network in a healthy state. Should defects of the organelle occur, a variety of reactions would ensue at the cellular level, including disrupted mitochondrial respiratory network and perturbed calcium homeostasis, possibly resulting in cellular death. Meanwhile, α-syn is a presynaptic protein that helps regulate synaptic vesicle transportation and endocytosis. Its misfolding into oligomeric sheets and fibrillation is toxic to the mitochondria and neurons. Increased cellular oxidative stress leads to α-syn accumulation, causing mitochondrial dysfunction. The proteasome and endo-lysosomal systems function to regulate damage and unwanted waste management within the cell while facilitating the quality control of mitochondria and α-syn. This review will analyze the biological functions and interactions between mitochondria, α-syn, and the endo-lysosomal system in the pathogenesis of PD.
Collapse
|
218
|
Li H, Jiang H, Zhang B, Feng J. Modeling Parkinson's Disease Using Patient-specific Induced Pluripotent Stem Cells. JOURNAL OF PARKINSONS DISEASE 2019; 8:479-493. [PMID: 30149462 PMCID: PMC6218140 DOI: 10.3233/jpd-181353] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disorder. It is characterized by the degeneration of nigral dopaminergic (DA) neurons. While over 90% of cases are idiopathic, without a clear etiology, mutations in many genes have been linked to rare, familial forms of PD. It has been quite challenging to develop effective animal models of PD that capture salient features of PD. The discovery of induced pluripotent stem cells (iPSCs) makes it possible to generate patient-specific DA neurons to study PD. Here, we review the methods for the generation of iPSCs and discuss previous studies using iPSC-derived neurons from monogenic forms of PD. These investigations have revealed several converging pathways that intersect with the unique vulnerabilities of human nigral DA neurons. With the rapid development in stem cell biology, it is possible to generate patient-specific neurons that will be increasingly similar to those in the brain of the patient. Combined with the ability to edit the genome to generate isogenic iPSCs, the generation and analysis of patient-specific midbrain DA neurons will transform PD research by providing a valuable tool for mechanistic study and drug discovery.
Collapse
Affiliation(s)
- Hong Li
- Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA.,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Houbo Jiang
- Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA.,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Boyang Zhang
- Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA.,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Jian Feng
- Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA.,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
219
|
Isacson O, Brekk OR, Hallett PJ. Novel Results and Concepts Emerging From Lipid Cell Biology Relevant to Degenerative Brain Aging and Disease. Front Neurol 2019; 10:1053. [PMID: 31649605 PMCID: PMC6794469 DOI: 10.3389/fneur.2019.01053] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/17/2019] [Indexed: 01/26/2023] Open
Abstract
While very rare familial forms of proteinopathy can cause Parkinson's disease (PD), Lewy body dementia (LBD) and age-related dementias, recent in-depth studies of lipid disturbances in the majority of the common forms of these diseases instead suggest a primary pathogenesis in lipid pathways. This review synthesizes a perspective from new data that point to an interdependence of lipids and proteinopathy. This article describes disturbed relationships in lipid homeostasis that causes neuropathology to develop over time and with age, which includes altered mechanisms of glia-neuron exchange of lipids and inflammatory signals.
Collapse
Affiliation(s)
- Ole Isacson
- McLean Hospital and Harvard Medical School, Neuroregeneration Research Institute, Belmont, MA, United States
| | | | | |
Collapse
|
220
|
Ryan E, Seehra GK, Sidransky E. Mutations, modifiers and epigenetics in Gaucher disease: Blurred boundaries between simple and complex disorders. Mol Genet Metab 2019; 128:10-13. [PMID: 31474515 DOI: 10.1016/j.ymgme.2019.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/19/2019] [Accepted: 08/19/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Emory Ryan
- Section on Molecular Neurogenetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Gurpreet Kaur Seehra
- Section on Molecular Neurogenetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ellen Sidransky
- Section on Molecular Neurogenetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
221
|
Chen B, Wen X, Jiang H, Wang J, Song N, Xie J. Interactions between iron and α-synuclein pathology in Parkinson's disease. Free Radic Biol Med 2019; 141:253-260. [PMID: 31233777 DOI: 10.1016/j.freeradbiomed.2019.06.024] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/06/2019] [Accepted: 06/19/2019] [Indexed: 02/06/2023]
Abstract
Both iron deposition and α-synuclein aggregation are neuropathological hallmarks of Parkinson's disease (PD). We aimed to summarize the extensive interactions between these two factors. The direct structural links between iron and α-synuclein suggest that structural reorganization provokes α-synuclein conformational change. Iron post-transcriptionally regulates α-synuclein synthesis in the presence of iron-responsive element. Increased oxidative/nitrative stress induced by iron is believed to be involved in the post-translational modulation of α-synuclein. Iron modulates proteolytic pathways and therefore participates in the regulation of α-synuclein levels. Meanwhile, the recycling of iron through ferritin degradation suggests a link from the aspects of the degradation signaling pathway. Finally, α-synuclein might regulate iron metabolism through its ferrireductase activity. A prominent role of α-synuclein in iron homeostasis is involved in the uptake of transferrin-Fe. These findings suggest that intracellular iron and α-synuclein are closely related to each other, contributing to the vulnerability of dopaminergic neurons or even to a vicious cycle of toxicity in the pathology of PD.
Collapse
Affiliation(s)
- Bingbing Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Medical College of Qingdao University, Qingdao, 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China
| | - Xiaoming Wen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Medical College of Qingdao University, Qingdao, 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Medical College of Qingdao University, Qingdao, 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China
| | - Jun Wang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Medical College of Qingdao University, Qingdao, 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China
| | - Ning Song
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Medical College of Qingdao University, Qingdao, 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China.
| | - Junxia Xie
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Medical College of Qingdao University, Qingdao, 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
222
|
Vidyadhara DJ, Lee JE, Chandra SS. Role of the endolysosomal system in Parkinson's disease. J Neurochem 2019; 150:487-506. [PMID: 31287913 PMCID: PMC6707858 DOI: 10.1111/jnc.14820] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/01/2019] [Accepted: 07/03/2019] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders, affecting 1-1.5% of the total population. While progress has been made in understanding the neurodegenerative mechanisms that lead to cell death in late stages of PD, mechanisms for early, causal pathogenic events are still elusive. Recent developments in PD genetics increasingly point at endolysosomal (E-L) system dysfunction as the early pathomechanism and key pathway affected in PD. Clathrin-mediated synaptic endocytosis, an integral part of the neuronal E-L system, is probably the main early target as evident in auxilin, RME-8, and synaptojanin-1 mutations that cause PD. Autophagy, another important pathway in the E-L system, is crucial in maintaining proteostasis and a healthy mitochondrial pool, especially in neurons considering their inability to divide and requirement to function an entire life-time. PINK1 and Parkin mutations severely perturb autophagy of dysfunctional mitochondria (mitophagy), both in the cell body and synaptic terminals of dopaminergic neurons, leading to PD. Endolysosomal sorting and trafficking is also crucial, which is complex in multi-compartmentalized neurons. VPS35 and VPS13C mutations noted in PD target these mechanisms. Mutations in GBA comprise the most common risk factor for PD and initiate pathology by compromising lysosomal function. This is also the case for ATP13A2 mutations. Interestingly, α-synuclein and LRRK2, key proteins involved in PD, function in different steps of the E-L pathway and target their components to induce disease pathogenesis. In this review, we discuss these E-L system genes that are linked to PD and how their dysfunction results in PD pathogenesis. This article is part of the Special Issue "Synuclein".
Collapse
Affiliation(s)
- D J Vidyadhara
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA
| | - John E Lee
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sreeganga S Chandra
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
223
|
Foffani G, Trigo‐Damas I, Pineda‐Pardo JA, Blesa J, Rodríguez‐Rojas R, Martínez‐Fernández R, Obeso JA. Focused ultrasound in Parkinson's disease: A twofold path toward disease modification. Mov Disord 2019; 34:1262-1273. [DOI: 10.1002/mds.27805] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/14/2019] [Accepted: 06/27/2019] [Indexed: 12/18/2022] Open
Affiliation(s)
- Guglielmo Foffani
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- Hospital Nacional de Parapléjicos Toledo Spain
| | - Inés Trigo‐Damas
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| | - José A. Pineda‐Pardo
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| | - Javier Blesa
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| | - Rafael Rodríguez‐Rojas
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| | - Raul Martínez‐Fernández
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| | - José A. Obeso
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| |
Collapse
|
224
|
Laurence M, Benito-León J, Calon F. Malassezia and Parkinson's Disease. Front Neurol 2019; 10:758. [PMID: 31396143 PMCID: PMC6667642 DOI: 10.3389/fneur.2019.00758] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 07/01/2019] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a common debilitating neurodegenerative disease caused by a loss of dopamine neurons in the substantia nigra within the central nervous system (CNS). The process leading to this neuronal loss is poorly understood. Seborrheic dermatitis (SD) is a common benign inflammatory condition of the skin which mainly affects lipid-rich regions of the head and trunk. SD is caused by over proliferation of the lipophilic fungus Malassezia. PD and SD are strongly associated. The increased PD risk following an SD diagnosis (OR = 1.69, 95% CI 1.36, 2.1; p < 0.001) reported by Tanner and colleagues remains unexplained. Malassezia were historically considered commensals confined to the skin. However, many recent studies report finding Malassezia in internal organs, including the CNS. This raises the possibility that Malassezia might be directly contributing to PD. Several lines of evidence support this hypothesis. AIDS is causally associated with both parkinsonism and SD, suggesting that weak T cell-mediated control of commensal microbes such as Malassezia might contribute to both. Genetic polymorphisms associated with PD (LRRK2, GBA, PINK1, SPG11, SNCA) increase availability of lipids within human cells, providing a suitable environment for Malassezia. Four LRRK2 polymorphisms which increase PD risk also increase Crohn's disease risk; Crohn's disease is strongly associated with an immune response against fungi, particularly Malassezia. Finally, Malassezia hypha formation and melanin synthesis are stimulated by L-DOPA, which could promote Malassezia invasiveness of dopamine neurons, and contribute to the accumulation of melanin in these neurons. Although Malassezia's presence in the substantia nigra remains to be confirmed, if Malassezia play a role in PD etiology, antifungal drugs should be tested as a possible therapeutic intervention.
Collapse
Affiliation(s)
| | - Julián Benito-León
- Department of Neurology, University Hospital "12 de Octubre", Madrid, Spain.,Department of Medicine, Faculty of Medicine, Complutense University, Madrid, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Frédéric Calon
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada.,Neurosciences Unit, CHU de Québec-Université Laval Research Center, Quebec City, QC, Canada
| |
Collapse
|
225
|
Savitt D, Jankovic J. Targeting α-Synuclein in Parkinson's Disease: Progress Towards the Development of Disease-Modifying Therapeutics. Drugs 2019; 79:797-810. [PMID: 30982161 DOI: 10.1007/s40265-019-01104-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD), the second most common neurodegenerative movement disorder, is characterized by progressive motor and non-motor symptoms [1]. Despite treatment with pharmacologic and surgical therapies, the disease will continue to relentlessly advance. Hence, there is a great deal of interest in potential disease-modifying therapies with the hope that the neurodegenerative process can be slowed or halted. The purpose of this review is to highlight the role toxic α-synuclein (α-syn) plays in PD pathogenesis and critically review the relevant literature about therapeutic modalities targeting α-syn. Toxic α-syn plays a key role in PD pathogenesis, disrupting important cellular functions, and, thus, targeting α-syn is a reasonable disease-modifying strategy. Current approaches under investigation include decreasing α-syn production with RNA interference (RNAi), inhibiting α-syn aggregation, promoting intracellular degradation of α-syn aggregates (via enhancing autophagy and enhancing lysosomal degradation), and promoting extracellular degradation of α-syn via active and passive immunization.
Collapse
Affiliation(s)
- Daniel Savitt
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, 7200 Cambridge, Suite 9A, Houston, TX, 77030-4202, USA
| | - Joseph Jankovic
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, 7200 Cambridge, Suite 9A, Houston, TX, 77030-4202, USA.
| |
Collapse
|
226
|
Ferreira-Junior NC, Campos AC, Guimarães FS, Del-Bel E, Zimmermann PMDR, Brum Junior L, Hallak JE, Crippa JA, Zuardi AW. Biological bases for a possible effect of cannabidiol in Parkinson's disease. ACTA ACUST UNITED AC 2019; 42:218-224. [PMID: 31314869 PMCID: PMC7115443 DOI: 10.1590/1516-4446-2019-0460] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/08/2019] [Indexed: 01/10/2023]
Abstract
Current pharmacotherapy of Parkinson’s disease (PD) is palliative and unable to modify the progression of neurodegeneration. Treatments that can improve patients’ quality of life with fewer side effects are needed, but not yet available. Cannabidiol (CBD), the major non-psychotomimetic constituent of cannabis, has received considerable research attention in the last decade. In this context, we aimed to critically review the literature on potential therapeutic effects of CBD in PD and discuss clinical and preclinical evidence supporting the putative neuroprotective mechanisms of CBD. We searched MEDLINE (via PubMed) for indexed articles published in English from inception to 2019. The following keywords were used: cannabis; cannabidiol and neuroprotection; endocannabinoids and basal ganglia; Parkinson’s animal models; Parkinson’s history; Parkinson’s and cannabidiol. Few studies addressed the biological bases for the purported effects of CBD on PD. Six preclinical studies showed neuroprotective effects, while three targeted the antidyskinetic effects of CBD. Three human studies have tested CBD in patients with PD: an open-label study, a case series, and a randomized controlled trial. These studies reported therapeutic effects of CBD on non-motor symptoms. Additional research is needed to elucidate the potential effectiveness of CBD in PD and the underlying mechanisms involved.
Collapse
Affiliation(s)
- Nilson C Ferreira-Junior
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto (FMRP), Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Alline C Campos
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto (FMRP), Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Francisco S Guimarães
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto (FMRP), Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Elaine Del-Bel
- Departamento de Morfologia, Fisiologia e Patologia Básica, Faculdade de Odontologia de Ribeirão Preto (FORP), USP, Ribeirão Preto, SP, Brazil
| | | | | | - Jaime E Hallak
- Departamento de Neurociências e Ciências do Comportamento, FMRP, USP, Ribeirão Preto, SP, Brazil
| | - José A Crippa
- Departamento de Neurociências e Ciências do Comportamento, FMRP, USP, Ribeirão Preto, SP, Brazil
| | - Antonio W Zuardi
- Departamento de Neurociências e Ciências do Comportamento, FMRP, USP, Ribeirão Preto, SP, Brazil
| |
Collapse
|
227
|
Farfel-Becker T, Do J, Tayebi N, Sidransky E. Can GBA1-Associated Parkinson Disease Be Modeled in the Mouse? Trends Neurosci 2019; 42:631-643. [PMID: 31288942 DOI: 10.1016/j.tins.2019.05.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/21/2019] [Accepted: 05/31/2019] [Indexed: 02/06/2023]
Abstract
Homozygous and heterozygous mutations in GBA1, the gene implicated in Gaucher disease, increase the risk and severity of Parkinson disease (PD). We evaluated the design, phenotype, strengths, and limitations of current GBA1-associated PD mouse models. Although faithful modeling of a genetic risk factor poses many challenges, the different approaches taken were successful in revealing predisposing abnormalities in heterozygotes for GBA1 mutations and demonstrating the deleterious effects of GBA1 impairment on the PD course in PD models. GBA1-PD models differ in key parameters, with no single model recapitulating all aspects of the GBA1-PD puzzle, emphasizing the importance of selecting the proper in vivo model depending on the specific molecular mechanism or potential therapy being studied.
Collapse
Affiliation(s)
- Tamar Farfel-Becker
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-3706, USA.
| | - Jenny Do
- Section of Molecular Neurogenetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-3708, USA
| | - Nahid Tayebi
- Section of Molecular Neurogenetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-3708, USA
| | - Ellen Sidransky
- Section of Molecular Neurogenetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-3708, USA.
| |
Collapse
|
228
|
Tulisiak CT, Mercado G, Peelaerts W, Brundin L, Brundin P. Can infections trigger alpha-synucleinopathies? PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 168:299-322. [PMID: 31699323 PMCID: PMC6857718 DOI: 10.1016/bs.pmbts.2019.06.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
As synucleinopathies, Parkinson's disease (PD) and multiple system atrophy (MSA) are neurodegenerative diseases that involve the spread of pathogenic alpha-synuclein (αSyn) throughout the brain. Recent studies have suggested a role for αSyn as an antimicrobial peptide in response to PD- and MSA-related infections of peripheral tissues, including those in the respiratory, gastrointestinal, and urogenital systems. In this chapter, we examine epidemiological and experimental evidence for a role of peripheral microbial infections in triggering alpha-synucleinopathies. We propose a model of how infectious triggers, in conjunction with inflammatory, environmental, and genetic facilitators, may result in transfer of pathogenic αSyn strains from the periphery to the brain, where they propagate and spread. Finally, we discuss future research challenges and programs necessary to clarify the role of infections as triggers of PD and MSA and, ultimately, to prevent the onset of these diseases by infectious triggers.
Collapse
Affiliation(s)
- Christopher T Tulisiak
- Center for Neurodegenerative Sciences, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Gabriela Mercado
- Center for Neurodegenerative Sciences, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Wouter Peelaerts
- Center for Neurodegenerative Sciences, Van Andel Research Institute, Grand Rapids, MI, United States; Laboratory for Neurobiology and Gene Therapy, KU Leuven, Leuven, Belgium
| | - Lena Brundin
- Center for Neurodegenerative Sciences, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Patrik Brundin
- Center for Neurodegenerative Sciences, Van Andel Research Institute, Grand Rapids, MI, United States.
| |
Collapse
|
229
|
Ikenaka K, Suzuki M, Mochizuki H, Nagai Y. Lipids as Trans-Acting Effectors for α-Synuclein in the Pathogenesis of Parkinson's Disease. Front Neurosci 2019; 13:693. [PMID: 31333408 PMCID: PMC6619337 DOI: 10.3389/fnins.2019.00693] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/18/2019] [Indexed: 12/15/2022] Open
Abstract
Aggregation of α-synuclein (αSyn) plays a central role in the pathogenesis of Parkinson’s disease (PD) and dementia with Lewy bodies (DLB). Lewy bodies (LBs) and Lewy neurites, which consist mainly of aggregated αSyn, are widely observed in the affected regions of patient brains. Except for some familial forms of PD/DLB, most sporadic PD/DLB patients express the wild-type (WT) αSyn protein without any mutations, and the mechanisms as to how WT αSyn gains the propensity to pathologically aggregate still remains unclear. Furthermore, the mechanisms by which the same αSyn protein can cause different synucleinopathies with distinct phenotypes and pathologies, such as PD, DLB, and multiple system atrophy (MSA), still remain largely unknown. Recently, mutations in the GBA1 gene (encoding glucocerebrosidase), which are responsible for the lysosomal storage disorder Gaucher disease (GD), have been reported to be the strongest risk factor for developing sporadic PD/DLB. We previously demonstrated that glucosylceramide accumulated by GBA1 deficiency promotes the conversion of αSyn into a proteinase K-resistant conformation. Furthermore, decreased glucocerebrosidase activity has also been reported in the brains of patients with sporadic PD/DLB. Moreover, αSyn pathology has also been shown in the brains of lysosomal storage disorder patients, which show glycosphingolipid accumulation. These observations suggest the possibility that altered lipid metabolism and lipid accumulation play roles in αSyn aggregation and PD/DLB pathogenesis. Indeed, several previous studies have demonstrated that lipid interactions affect the conformation of αSyn and induces its oligomerization and aggregation. In this review, we will give an overview of the association between αSyn aggregation and lipid interactions from the viewpoints of the etiology, pathology, and genetics of PD/DLB. We also discuss the distinct species of αSyn aggregates and their association with specific types of synucleinopathies, and introduce our hypothesis that lipid interactions play a role as trans-acting effectors in producing distinct strains of αSyn fibrils.
Collapse
Affiliation(s)
- Kensuke Ikenaka
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mari Suzuki
- Department of Neurotherapeutics, Graduate School of Medicine, Osaka University, Osaka, Japan.,Diabetic Neuropathy Project, Department of Sensory and Motor Systems, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshitaka Nagai
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Neurotherapeutics, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
230
|
Zurbruegg M, Chan MY, Svenningsson P. GBA RNAi but not catalytic inhibition of glucocerebrosidase with Conduritol-β-epoxide increases levels of total α-synuclein in SH-SY5Y cells. Neurosci Lett 2019; 706:217-222. [DOI: 10.1016/j.neulet.2019.05.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/07/2019] [Accepted: 05/14/2019] [Indexed: 01/24/2023]
|
231
|
Jellinger KA. Neuropathology and pathogenesis of extrapyramidal movement disorders: a critical update-I. Hypokinetic-rigid movement disorders. J Neural Transm (Vienna) 2019; 126:933-995. [PMID: 31214855 DOI: 10.1007/s00702-019-02028-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/05/2019] [Indexed: 02/06/2023]
Abstract
Extrapyramidal movement disorders include hypokinetic rigid and hyperkinetic or mixed forms, most of them originating from dysfunction of the basal ganglia (BG) and their information circuits. The functional anatomy of the BG, the cortico-BG-thalamocortical, and BG-cerebellar circuit connections are briefly reviewed. Pathophysiologic classification of extrapyramidal movement disorder mechanisms distinguish (1) parkinsonian syndromes, (2) chorea and related syndromes, (3) dystonias, (4) myoclonic syndromes, (5) ballism, (6) tics, and (7) tremor syndromes. Recent genetic and molecular-biologic classifications distinguish (1) synucleinopathies (Parkinson's disease, dementia with Lewy bodies, Parkinson's disease-dementia, and multiple system atrophy); (2) tauopathies (progressive supranuclear palsy, corticobasal degeneration, FTLD-17; Guamian Parkinson-dementia; Pick's disease, and others); (3) polyglutamine disorders (Huntington's disease and related disorders); (4) pantothenate kinase-associated neurodegeneration; (5) Wilson's disease; and (6) other hereditary neurodegenerations without hitherto detected genetic or specific markers. The diversity of phenotypes is related to the deposition of pathologic proteins in distinct cell populations, causing neurodegeneration due to genetic and environmental factors, but there is frequent overlap between various disorders. Their etiopathogenesis is still poorly understood, but is suggested to result from an interaction between genetic and environmental factors. Multiple etiologies and noxious factors (protein mishandling, mitochondrial dysfunction, oxidative stress, excitotoxicity, energy failure, and chronic neuroinflammation) are more likely than a single factor. Current clinical consensus criteria have increased the diagnostic accuracy of most neurodegenerative movement disorders, but for their definite diagnosis, histopathological confirmation is required. We present a timely overview of the neuropathology and pathogenesis of the major extrapyramidal movement disorders in two parts, the first one dedicated to hypokinetic-rigid forms and the second to hyperkinetic disorders.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|
232
|
Mitochondrial antigen presentation: a mechanism linking Parkinson’s disease to autoimmunity. Curr Opin Immunol 2019; 58:31-37. [DOI: 10.1016/j.coi.2019.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 12/12/2022]
|
233
|
Understanding the schizophrenia phenotype in the first patient with the full SCN2A phenotypic spectrum. Psychiatr Genet 2019; 29:91-94. [PMID: 30741786 DOI: 10.1097/ypg.0000000000000219] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The sodium voltage-gated channel α subunit 2 (SCN2A) gene encodes a subunit of sodium voltage-gated channels expressed primarily in the central nervous system that are responsible for action potential initiation and propagation in excitable cells. SCN2A mutations underlie a spectrum of distinct phenotypes, including seizure disorders, neurodevelopmental disorders, and rarer instances of episodic ataxia and schizophrenia. We report on a 38-year-old patient with adult-onset psychotic symptoms on a background of infantile-onset seizures, autistic features and episodic ataxia. Whole-exome sequencing revealed a de-novo novel SCN2A mutation (c.4966T > C, p.Ser1656Pro). This and other SCN2A mutations associated with the schizophrenia phenotype overlap those seen in neurodevelopmental disorders, suggesting a common underlying mechanism. This is the first report of a patient with the entire known SCN2A phenotypic spectrum. We highlight the importance of recognizing the psychiatric phenotypes associated with SCN2A mutations and that the phenotypic spectrum is more fluid, and less categorical, than previously thought.
Collapse
|
234
|
Mitochondrial Dysfunction in Parkinson's Disease-Cause or Consequence? BIOLOGY 2019; 8:biology8020038. [PMID: 31083583 PMCID: PMC6627981 DOI: 10.3390/biology8020038] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/25/2019] [Accepted: 02/05/2019] [Indexed: 12/18/2022]
Abstract
James Parkinson first described the motor symptoms of the disease that took his name over 200 years ago. While our knowledge of many of the changes that occur in this condition has increased, it is still unknown what causes this neurodegeneration and why it only affects some individuals with advancing age. Here we review current literature to discuss whether the mitochondrial dysfunction we have detected in Parkinson’s disease is a pathogenic cause of neuronal loss or whether it is itself a consequence of dysfunction in other pathways. We examine research data from cases of idiopathic Parkinson’s with that from model systems and individuals with familial forms of the disease. Furthermore, we include data from healthy aged individuals to highlight that many of the changes described are also present with advancing age, though not normally in the presence of severe neurodegeneration. While a definitive answer to this question may still be just out of reach, it is clear that mitochondrial dysfunction sits prominently at the centre of the disease pathway that leads to catastrophic neuronal loss in those affected by this disease.
Collapse
|
235
|
Alecu I, Bennett SAL. Dysregulated Lipid Metabolism and Its Role in α-Synucleinopathy in Parkinson's Disease. Front Neurosci 2019; 13:328. [PMID: 31031582 PMCID: PMC6470291 DOI: 10.3389/fnins.2019.00328] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/21/2019] [Indexed: 12/23/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, the main pathological hallmark of which is the accumulation of α-synuclein (α-syn) and the formation of filamentous aggregates called Lewy bodies in the brainstem, limbic system, and cortical areas. Lipidomics is a newly emerging field which can provide fresh insights and new answers that will enhance our capacity for early diagnosis, tracking disease progression, predicting critical endpoints, and identifying risk in pre-symptomatic persons. In recent years, lipids have been implicated in many aspects of PD pathology. Biophysical and lipidomic studies have demonstrated that α-syn binds preferentially not only to specific lipid families but also to specific molecular species and that these lipid-protein complexes enhance its interaction with synaptic membranes, influence its oligomerization and aggregation, and interfere with the catalytic activity of cytoplasmic lipid enzymes and lysosomal lipases, thereby affecting lipid metabolism. The genetic link between aberrant lipid metabolism and PD is even more direct, with mutations in GBA and SMPD1 enhancing PD risk in humans and loss of GALC function increasing α-syn aggregation and accumulation in experimental murine models. Moreover, a number of lipidomic studies have reported PD-specific lipid alterations in both patient brains and plasma, including alterations in the lipid composition of lipid rafts in the frontal cortex. A further aspect of lipid dysregulation promoting PD pathogenesis is oxidative stress and inflammation, with proinflammatory lipid mediators such as platelet activating factors (PAFs) playing key roles in arbitrating the progressive neurodegeneration seen in PD linked to α-syn intracellular trafficking. Lastly, there are a number of genetic risk factors of PD which are involved in normal lipid metabolism and function. Genes such as PLA2G6 and SCARB2, which are involved in glycerophospholipid and sphingolipid metabolism either directly or indirectly are associated with risk of PD. This review seeks to describe these facets of metabolic lipid dysregulation as they relate to PD pathology and potential pathomechanisms involved in disease progression, while highlighting incongruous findings and gaps in knowledge that necessitate further research.
Collapse
Affiliation(s)
- Irina Alecu
- Neural Regeneration Laboratory, Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
- Department of Chemistry and Biomolecular Sciences, Centre for Catalysis and Research Innovation, University of Ottawa, Ottawa, ON, Canada
| | - Steffany A. L. Bennett
- Neural Regeneration Laboratory, Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
- Department of Chemistry and Biomolecular Sciences, Centre for Catalysis and Research Innovation, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
236
|
Koster KP, Francesconi W, Berton F, Alahmadi S, Srinivas R, Yoshii A. Developmental NMDA receptor dysregulation in the infantile neuronal ceroid lipofuscinosis mouse model. eLife 2019; 8:40316. [PMID: 30946007 PMCID: PMC6464704 DOI: 10.7554/elife.40316] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 03/31/2019] [Indexed: 12/20/2022] Open
Abstract
Protein palmitoylation and depalmitoylation alter protein function. This post-translational modification is critical for synaptic transmission and plasticity. Mutation of the depalmitoylating enzyme palmitoyl-protein thioesterase 1 (PPT1) causes infantile neuronal ceroid lipofuscinosis (CLN1), a pediatric neurodegenerative disease. However, the role of protein depalmitoylation in synaptic maturation is unknown. Therefore, we studied synapse development in Ppt1-/- mouse visual cortex. We demonstrate that the developmental N-methyl-D-aspartate receptor (NMDAR) subunit switch from GluN2B to GluN2A is stagnated in Ppt1-/- mice. Correspondingly, Ppt1-/- neurons exhibit immature evoked NMDAR currents and dendritic spine morphology in vivo. Further, dissociated Ppt1-/- cultured neurons show extrasynaptic, diffuse calcium influxes and enhanced vulnerability to NMDA-induced excitotoxicity, reflecting the predominance of GluN2B-containing receptors. Remarkably, Ppt1-/- neurons demonstrate hyperpalmitoylation of GluN2B as well as Fyn kinase, which regulates surface retention of GluN2B. Thus, PPT1 plays a critical role in postsynapse maturation by facilitating the GluN2 subunit switch and proteostasis of palmitoylated proteins.
Collapse
Affiliation(s)
- Kevin P Koster
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, United States
| | - Walter Francesconi
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, United States
| | - Fulvia Berton
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, United States
| | - Sami Alahmadi
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, United States
| | - Roshan Srinivas
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, United States
| | - Akira Yoshii
- Department of Pediatrics, University of Illinois at Chicago, Chicago, United States.,Department of Neurology, University of Illinois at Chicago, Chicago, United States
| |
Collapse
|
237
|
Ganoderma lucidum extract ameliorates MPTP-induced parkinsonism and protects dopaminergic neurons from oxidative stress via regulating mitochondrial function, autophagy, and apoptosis. Acta Pharmacol Sin 2019; 40:441-450. [PMID: 29991712 DOI: 10.1038/s41401-018-0077-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 06/14/2018] [Indexed: 12/25/2022]
Abstract
Neuroprotection targeting mitochondrial dysfunction has been proposed as an important therapeutic strategy for Parkinson's disease. Ganoderma lucidum (GL) has emerged as a novel agent that protects neurons from oxidative stress. However, the detailed mechanisms underlying GL-induced neuroprotection have not been documented. In this study, we investigated the neuroprotective effects of GL extract (GLE) and the underlying mechanisms in the classic MPTP(1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)-induced mouse model of PD. Mice were injected with MPTP to induce parkinsonism. Then the mice were administered GLE (400 mg kg-1 d-1, ig) for 4 weeks. We observed that GLE administration significantly improved locomotor performance and increased tyrosine hydroxylase expression in the substantia nigra pars compact (SNpc) of MPTP-treated mice. In in vitro study, treatment of neuroblastoma neuro-2a cells with 1-methyl-4-phenylpyridinium (MPP+, 1 mmol/L) caused mitochondrial membrane potential collapse, radical oxygen species accumulation, and ATP depletion. Application of GLE (800 μg/mL) protected neuroblastoma neuro-2a cells against MPP+ insult. Application of GLE also improved mitochondrial movement dysfunction in cultured primary mesencephalic neurons. In addition, GLE counteracted the decline in NIX (also called BNIP3L) expression and increase in the LC3-II/LC3-I ratio evoked by MPP+. Moreover, GLE reactivated MPP+-inhibited AMPK, mTOR, and ULK1. Similarly, GLE was sufficient to counteract MPP+-induced inhibition of PINK1 and Parkin expression. GLE suppressed MPP+-induced cytochrome C release and activation of caspase-3 and caspase-9. In summary, our results provide evidence that GLE ameliorates parkinsonism pathology via regulating mitochondrial function, autophagy, and apoptosis, which may involve the activation of both the AMPK/mTOR and PINK1/Parkin signaling pathway.
Collapse
|
238
|
Senkevich KA, Miliukhina IV, Pchelina SN. [The genetic predictors of cognitive impairment in Parkinson's disease]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 118:109-117. [PMID: 30251988 DOI: 10.17116/jnevro2018118081109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder that can be both sporadic and familial. A number of studies are devoted to the study of non-motor symptoms in PD today. Cognitive deficits, and especially dementia, are one of the most severe and disabling non-motor symptoms of PD. More than a quarter of patients in the early stages of PD have a moderate cognitive impairment, more than half of patients with PD develop dementia within 10 years from the date of diagnosis. Using genome-wide association studies (GWAS), a number of genes associated with cognitive impairment have been identified based on a comparison of genetic and clinical phenotypes. These genes can be divided into three groups: genes that lead to the development of PD and are inherited according to the laws of Mendel (SNCA), genes that are risk factors for PD development (GBA, MAPT) and genes associated with the development of cognitive impairment, but not with PD (COMT, APOE, BDNF). This review examines the effect of genetic variants in the above-mentioned genes on cognitive functions in patients with PD. The elucidation of the genetic basis of cognitive deficits in PD could help in choice of treatment tactics and in development of new therapeutic strategies.
Collapse
Affiliation(s)
- K A Senkevich
- Institute of Experimental Medicine, St. Petersburg, Russia; Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia; St. Petersburg Nuclear Physics Institute named by Konstantinov of NRC 'Kurchatov Institute', Gatchina, Russia
| | - I V Miliukhina
- Institute of Experimental Medicine, St. Petersburg, Russia; Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia
| | - S N Pchelina
- Institute of Experimental Medicine, St. Petersburg, Russia; Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia; St. Petersburg Nuclear Physics Institute named by Konstantinov of NRC 'Kurchatov Institute', Gatchina, Russia
| |
Collapse
|
239
|
Boudewyn LC, Walkley SU. Current concepts in the neuropathogenesis of mucolipidosis type IV. J Neurochem 2019; 148:669-689. [PMID: 29770442 PMCID: PMC6239999 DOI: 10.1111/jnc.14462] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/29/2018] [Accepted: 05/02/2018] [Indexed: 12/11/2022]
Abstract
Mucolipidosis type IV (MLIV) is an autosomal recessive, lysosomal storage disorder causing progressively severe intellectual disability, motor and speech deficits, retinal degeneration often culminating in blindness, and systemic disease causing a shortened lifespan. MLIV results from mutations in the gene MCOLN1 encoding the transient receptor potential channel mucolipin-1. It is an ultra-rare disease and is currently known to affect just over 100 diagnosed individuals. The last decade has provided a wealth of research focused on understanding the role of the enigmatic mucolipin-1 protein in cell and brain function and how its absence causes disease. This review explores our current understanding of the mucolipin-1 protein in relation to neuropathogenesis in MLIV and describes recent findings implicating mucolipin-1's important role in mechanistic target of rapamycin and TFEB (transcription factor EB) signaling feedback loops as well as in the function of the greater endosomal/lysosomal system. In addition to addressing the vital role of mucolipin-1 in the brain, we also report new data on the question of whether haploinsufficiency as would be anticipated in MCOLN1 heterozygotes is associated with any evidence of neuron dysfunction or disease. Greater insights into the role of mucolipin-1 in the nervous system can be expected to shed light not only on MLIV disease but also on numerous processes governing normal brain function. This article is part of the Special Issue "Lysosomal Storage Disorders".
Collapse
Affiliation(s)
- Lauren C. Boudewyn
- Dominick P. Purpura Department of Neuroscience, Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, New York
| | - Steven U. Walkley
- Dominick P. Purpura Department of Neuroscience, Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
240
|
Arkadir D, Dinur T, Becker Cohen M, Revel-Vilk S, Tiomkin M, Brüggemann N, Cozma C, Rolfs A, Zimran A. Prodromal substantia nigra sonography undermines suggested association between substrate accumulation and the risk for GBA-related Parkinson's disease. Eur J Neurol 2019; 26:1013-1018. [PMID: 30714262 DOI: 10.1111/ene.13927] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 01/23/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND PURPOSE Individuals with GBA (glucocerebrosidase) mutations are at increased risk of Parkinson's disease (PD). It is still debated, however, whether this increased risk results from impaired glucocerebrosidase activity leading to substrate accumulation. Comparing the presence of prodromal PD marker in GBA mutation carriers and patients with Gaucher disease (GD) (in which substrate accumulation is extensive) can assist in clarifying this issue. METHODS In this cross-sectional study, we compared the hyperechogenic area of the substantia nigra, a prodromal PD marker, in large cohorts of GBA mutation carriers (n = 71) and patients with GD (n = 145). Our control populations were healthy, non-carriers (n = 49) and patients with GBA -related PD (n = 11). Substrate accumulation was assessed from dry blood spot levels of glucosylsphingosine. RESULTS Our findings indicate no contribution of substrate accumulation, as the area of hyperechogenicity is similarly enlarged relative to healthy controls in both GBA mutation carriers and patients with GD. Moreover, this similarity between GBA carriers and patients with GD persists when comparing only carriers of the N370S (c.1226A>G) mutation (n = 38) with untreated patients with GD who were homozygotes for the same mutation (n = 47). In addition, measurements of hyperechogenic area did not correlate with levels of glucosylsphingosine in the untreated patients with GD. CONCLUSION The presence of a marker of prodromal PD (substantia nigra hyperechogenicity) is independent of substrate accumulation in a population with mutated GBA . Although further longitudinal studies are needed to determine the precise predictive value of this marker for GBA -related PD, our findings raise doubts regarding the contribution of substance reduction strategies to PD prevention.
Collapse
Affiliation(s)
- D Arkadir
- Department of Neurology, Hadassah Medical Center, The Hebrew University, Jerusalem
| | - T Dinur
- Gaucher Center, Shaare Zedek Medical Center, The Hebrew University, Jerusalem, Israel
| | - M Becker Cohen
- Gaucher Center, Shaare Zedek Medical Center, The Hebrew University, Jerusalem, Israel
| | - S Revel-Vilk
- Gaucher Center, Shaare Zedek Medical Center, The Hebrew University, Jerusalem, Israel
| | - M Tiomkin
- Gaucher Center, Shaare Zedek Medical Center, The Hebrew University, Jerusalem, Israel
| | - N Brüggemann
- Department of Neurology, University of Lübeck, Lübeck.,Institute of Neurogenetics, University of Lübeck, Lübeck
| | | | - A Rolfs
- Centogene AG, Rostock.,Albrecht-Kossel-Institute for Neurodegeneration, University of Rostock, Rostock, Germany
| | - A Zimran
- Gaucher Center, Shaare Zedek Medical Center, The Hebrew University, Jerusalem, Israel
| |
Collapse
|
241
|
Nasrolahi A, Safari F, Farhoudi M, Khosravi A, Farajdokht F, Bastaminejad S, Sandoghchian Shotorbani S, Mahmoudi J. Immune system and new avenues in Parkinson’s disease research and treatment. Rev Neurosci 2019; 30:709-727. [DOI: 10.1515/revneuro-2018-0105] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/28/2018] [Indexed: 12/13/2022]
Abstract
Abstract
Parkinson’s disease (PD) is a progressive neurological disorder characterized by degeneration of dopaminergic neurons in the substantia nigra. However, although 200 years have now passed since the primary clinical description of PD by James Parkinson, the etiology and mechanisms of neuronal loss in this disease are still not fully understood. In addition to genetic and environmental factors, activation of immunologic responses seems to have a crucial role in PD pathology. Intraneuronal accumulation of α-synuclein (α-Syn), as the main pathological hallmark of PD, potentially mediates initiation of the autoimmune and inflammatory events through, possibly, auto-reactive T cells. While current therapeutic regimens are mainly used to symptomatically suppress PD signs, application of the disease-modifying therapies including immunomodulatory strategies may slow down the progressive neurodegeneration process of PD. The aim of this review is to summarize knowledge regarding previous studies on the relationships between autoimmune reactions and PD pathology as well as to discuss current opportunities for immunomodulatory therapy.
Collapse
Affiliation(s)
- Ava Nasrolahi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences , Tabriz 51666-14756 , Iran
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Fatemeh Safari
- Departmant of Medical Biotechnology, School of Advanced Medical Sciences and Technologies , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Mehdi Farhoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences , Tabriz 51666-14756 , Iran
| | - Afra Khosravi
- Department of Immunology, Faculty of Medicine , Ilam University of Medical Sciences , Ilam , Iran
| | - Fereshteh Farajdokht
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences , Tabriz 51666-14756 , Iran
| | - Saiyad Bastaminejad
- Department of Biochemistry and Molecular Medicine, School of Medicine , Ilam University of Medical Sciences , Ilam , Iran
| | | | - Javad Mahmoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences , P.O. 51666-14756, Tabriz , Iran , e-mail:
| |
Collapse
|
242
|
Papadopoulos VE, Nikolopoulou G, Antoniadou I, Karachaliou A, Arianoglou G, Emmanouilidou E, Sardi SP, Stefanis L, Vekrellis K. Modulation of β-glucocerebrosidase increases α-synuclein secretion and exosome release in mouse models of Parkinson's disease. Hum Mol Genet 2019; 27:1696-1710. [PMID: 29547959 DOI: 10.1093/hmg/ddy075] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/23/2018] [Indexed: 11/14/2022] Open
Abstract
Glucocerebrosidase gene (GBA) mutations are the most common genetic contributor to Parkinson's disease (PD) and are associated with decreased glucocerebrosidase (GCase) enzymatic activity in PD. PD patients without GBA mutations also exhibit lower levels of GCase activity in the central nervous system suggesting a potential contribution of the enzyme activity in disease pathogenesis, possibly by alteration of lysosomal function. α-synuclein (ASYN), a protein with a central role in PD pathogenesis, has been shown to be secreted partly in association with exosomes. It is possible that a dysfunction of the endocytic pathway through GCase may result in altered exosome release of ASYN. The aim of this study was to examine whether manipulating GCase activity in vivo and in vitro could affect ASYN accumulation and secretion. GCase overexpression in vitro resulted in a significant decrease of exosome secretion. Chronic inhibition of GCase activity in vivo, by administration of the covalent inhibitor conduritol-B epoxide in A53T-synuclein alpha gene Tg mice significantly elevated intracellular oligomeric ASYN species. Importantly, GCase inhibition, induced a profound increase in the number of brain exosomes released, as well as exosome-associated ASYN oligomers. Finally, virus-mediated expression of mutant GBA in the mouse striatum increased ASYN secretion in the same region. Together, these results provide for the first time evidence that a decrease of GCase or overexpression of mutant GCase in a chronic in vivo setting can affect ASYN secretion. Such effects may mediate enhanced propagation of ASYN, driving pathology in GBA-associated PD.
Collapse
Affiliation(s)
- Vassilis E Papadopoulos
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece.,Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Georgia Nikolopoulou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece.,Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ivi Antoniadou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece.,Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 1516, Cyprus
| | - Antonia Karachaliou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece.,School of Science, Faculty of Biology, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Giovanna Arianoglou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece.,Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Evangelia Emmanouilidou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | | | - Leonidas Stefanis
- Center of Clinical Research and Experimental Surgery, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece.,Second Department of Neurology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Kostas Vekrellis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
243
|
Silveira CRA, MacKinley J, Coleman K, Li Z, Finger E, Bartha R, Morrow SA, Wells J, Borrie M, Tirona RG, Rupar CA, Zou G, Hegele RA, Mahuran D, MacDonald P, Jenkins ME, Jog M, Pasternak SH. Ambroxol as a novel disease-modifying treatment for Parkinson's disease dementia: protocol for a single-centre, randomized, double-blind, placebo-controlled trial. BMC Neurol 2019; 19:20. [PMID: 30738426 PMCID: PMC6368728 DOI: 10.1186/s12883-019-1252-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 02/01/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Currently there are no disease-modifying treatments for Parkinson's disease dementia (PDD), a condition linked to aggregation of the protein α-synuclein in subcortical and cortical brain areas. One of the leading genetic risk factors for Parkinson's disease is being a carrier in the gene for β-Glucocerebrosidase (GCase; gene name GBA1). Studies in cell culture and animal models have shown that raising the levels of GCase can decrease levels of α-synuclein. Ambroxol is a pharmacological chaperone for GCase and is able to raise the levels of GCase and could therefore be a disease-modifying treatment for PDD. The aims of this trial are to determine if Ambroxol is safe and well-tolerated by individuals with PDD and if Ambroxol affects cognitive, biochemical, and neuroimaging measures. METHODS This is a phase II, single-centre, double-blind, randomized placebo-controlled trial involving 75 individuals with mild to moderate PDD. Participants will be randomized into Ambroxol high-dose (1050 mg/day), low-dose (525 mg/day), or placebo treatment arms. Assessments will be undertaken at baseline, 6-months, and 12-months follow up times. Primary outcome measures will be the Alzheimer's disease Assessment Scale-cognitive subscale (ADAS-Cog) and the ADCS Clinician's Global Impression of Change (CGIC). Secondary measures will include the Parkinson's disease Cognitive Rating Scale, Clinical Dementia Rating, Trail Making Test, Stroop Test, Unified Parkinson's disease Rating Scale, Purdue Pegboard, Timed Up and Go, and gait kinematics. Markers of neurodegeneration will include MRI and CSF measures. Pharmacokinetics and pharmacodynamics of Ambroxol will be examined through plasma levels during dose titration phase and evaluation of GCase activity in lymphocytes. DISCUSSION If found effective and safe, Ambroxol will be one of the first disease-modifying treatments for PDD. TRIAL REGISTRATION ClinicalTrials.gov NCT02914366, 26 Sep 2016/retrospectively registered.
Collapse
Affiliation(s)
- C R A Silveira
- Cognitive Neurology and Alzheimer's Disease Research Centre, Parkwood Institute - Main Building, Room A230, 550, Wellington Road, London, Ontario, N6G 0A7, Canada.,Lawson Health Research Institute, London, Ontario, Canada
| | - J MacKinley
- Cognitive Neurology and Alzheimer's Disease Research Centre, Parkwood Institute - Main Building, Room A230, 550, Wellington Road, London, Ontario, N6G 0A7, Canada.,Lawson Health Research Institute, London, Ontario, Canada
| | - K Coleman
- Cognitive Neurology and Alzheimer's Disease Research Centre, Parkwood Institute - Main Building, Room A230, 550, Wellington Road, London, Ontario, N6G 0A7, Canada.,Lawson Health Research Institute, London, Ontario, Canada
| | - Z Li
- Cognitive Neurology and Alzheimer's Disease Research Centre, Parkwood Institute - Main Building, Room A230, 550, Wellington Road, London, Ontario, N6G 0A7, Canada.,Lawson Health Research Institute, London, Ontario, Canada
| | - E Finger
- Cognitive Neurology and Alzheimer's Disease Research Centre, Parkwood Institute - Main Building, Room A230, 550, Wellington Road, London, Ontario, N6G 0A7, Canada.,Lawson Health Research Institute, London, Ontario, Canada.,Deparment of Clinical Neurological Science, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - R Bartha
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,Robarts Research Institute, Western University, London, Ontario, Canada
| | - S A Morrow
- Cognitive Neurology and Alzheimer's Disease Research Centre, Parkwood Institute - Main Building, Room A230, 550, Wellington Road, London, Ontario, N6G 0A7, Canada.,Lawson Health Research Institute, London, Ontario, Canada.,Deparment of Clinical Neurological Science, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - J Wells
- Lawson Health Research Institute, London, Ontario, Canada.,Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - M Borrie
- Lawson Health Research Institute, London, Ontario, Canada.,Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - R G Tirona
- Lawson Health Research Institute, London, Ontario, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - C A Rupar
- Lawson Health Research Institute, London, Ontario, Canada.,Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - G Zou
- Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,Robarts Research Institute, Western University, London, Ontario, Canada
| | - R A Hegele
- Lawson Health Research Institute, London, Ontario, Canada.,Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,Robarts Research Institute, Western University, London, Ontario, Canada
| | - D Mahuran
- Laboratory of Medicine and Pathobiology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - P MacDonald
- Deparment of Clinical Neurological Science, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - M E Jenkins
- Lawson Health Research Institute, London, Ontario, Canada.,Deparment of Clinical Neurological Science, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - M Jog
- Lawson Health Research Institute, London, Ontario, Canada.,Deparment of Clinical Neurological Science, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - S H Pasternak
- Cognitive Neurology and Alzheimer's Disease Research Centre, Parkwood Institute - Main Building, Room A230, 550, Wellington Road, London, Ontario, N6G 0A7, Canada. .,Lawson Health Research Institute, London, Ontario, Canada. .,Deparment of Clinical Neurological Science, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada. .,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada. .,Robarts Research Institute, Western University, London, Ontario, Canada.
| |
Collapse
|
244
|
Benson DL, Huntley GW. Are we listening to everything the PARK genes are telling us? J Comp Neurol 2019; 527:1527-1540. [PMID: 30680728 DOI: 10.1002/cne.24642] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/30/2018] [Accepted: 01/02/2019] [Indexed: 12/17/2022]
Abstract
The cardinal motor symptoms that define Parkinson's disease (PD) clinically have been recognized for over 200 years. That these symptoms arise following the loss of dopamine neurons in the substantia nigra has been known for the last 50. These long-established facts have fueled a broadly held expectation that degenerating dopaminergic neurons alone hold the key to understanding and curing PD. This prevalent expectation is at odds with the observation that many nonmotor symptoms, including depression and cognitive inflexibility among others, can appear years earlier than the overt dopaminergic neuron degeneration that drives motor abnormalities and are not improved by levodopa treatment. Thus, preserving or rescuing dopamine neuron health and function is of paramount importance, but this alone fails to capture the underlying neurobiology of earlier-appearing nonmotor symptoms. Insight into the complete landscape of disease-related abnormalities and the context in which they arise can be gleaned from a more comprehensive consideration of the PARK genes that are known to cause PD. Here, we make the case that a full incorporation of research showing when and where PARK genes are expressed as well as the impact of gene mutation on function throughout life, in tandem with research studying how dopaminergic neuron degeneration begins, is essential for a full understanding of the multi-dimensional etiology of PD. A broad view may also reveal something about long-term adjustments cells and systems make in response to gene mutation and help to identify mechanisms conferring the resilience or susceptibility of some cells and systems over others.
Collapse
Affiliation(s)
- Deanna L Benson
- Department of Neuroscience, Friedman Brain Institute and the Graduate School of Biomedical Sciences Icahn School of Medicine at Mount Sinai, New York, New York
| | - George W Huntley
- Department of Neuroscience, Friedman Brain Institute and the Graduate School of Biomedical Sciences Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
245
|
Dunn TM, Tifft CJ, Proia RL. A perilous path: the inborn errors of sphingolipid metabolism. J Lipid Res 2019; 60:475-483. [PMID: 30683667 PMCID: PMC6399501 DOI: 10.1194/jlr.s091827] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/13/2019] [Indexed: 01/19/2023] Open
Abstract
The sphingolipid (SL) metabolic pathway generates structurally diverse lipids that have roles as membrane constituents and as bioactive signaling molecules. The influence of the SL metabolic pathway in biology is pervasive; it exists in all mammalian cells and has roles in many cellular and physiological pathways. Human genetic diseases have long been recognized to be caused by mutations in the pathway, but until recently these mutational defects were only known to affect lysosomal SL degradation. Now, with a nearly complete delineation of the genes constituting the SL metabolic pathway, a growing number of additional genetic disorders caused by mutations in genes within other sectors of the pathway (de novo ceramide synthesis, glycosphingolipid synthesis, and nonlysosomal SL degradation) have been recognized. Although these inborn disorders of SL metabolism are clinically heterogeneous, some common pathogenic mechanisms, derived from the unique properties and functions of the SLs, underlie several of the diseases. These mechanisms include overaccumulation of toxic or bioactive lipids and the disruption of specific critical cellular and physiological processes. Many of these diseases also have commonalities in physiological systems affected, such as the nervous system and skin. While inborn disorders of SL metabolism are rare, gene variants in the pathway have been linked to increased susceptibility to Parkinson’s disease and childhood asthma, implying that the SL metabolic pathway may have a role in these disorders. A more complete understanding of the inborn errors of SL metabolism promises new insights into the convergence of their pathogenesis with those of common human diseases.
Collapse
Affiliation(s)
- Teresa M Dunn
- Department of Biochemistry, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814
| | - Cynthia J Tifft
- Office of the Clinical Director and Medical Genetics Branch National Human Genome Research Institute, Bethesda, MD 20892
| | - Richard L Proia
- Genetics of Development and Disease Branch National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
246
|
Marques ARA, Saftig P. Lysosomal storage disorders - challenges, concepts and avenues for therapy: beyond rare diseases. J Cell Sci 2019; 132:jcs221739. [PMID: 30651381 DOI: 10.1242/jcs.221739] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The pivotal role of lysosomes in cellular processes is increasingly appreciated. An understanding of the balanced interplay between the activity of acidic hydrolases, lysosomal membrane proteins and cytosolic proteins is required. Lysosomal storage diseases (LSDs) are characterized by disturbances in this network and by intralysosomal accumulation of substrates, often only in certain cell types. Even though our knowledge of these diseases has increased and therapies have been established, many aspects of the molecular pathology of LSDs remain obscure. This Review aims to discuss how lysosomal storage affects functions linked to lysosomes, such as membrane repair, autophagy, exocytosis, lipid homeostasis, signalling cascades and cell viability. Therapies must aim to correct lysosomal storage not only morphologically, but reverse its (patho)biochemical consequences. As different LSDs have different molecular causes, this requires custom tailoring of therapies. We will discuss the major advantages and drawbacks of current and possible future therapies for LSDs. Study of the pathological molecular mechanisms underlying these 'experiments of nature' often yields information that is relevant for other conditions found in the general population. Therefore, more common diseases may profit from a correction of impaired lysosomal function.
Collapse
Affiliation(s)
- André R A Marques
- Biochemisches Institut, Christian Albrechts-Universität Kiel, Olshausenstr. 40, D-24098 Kiel, Germany
| | - Paul Saftig
- Biochemisches Institut, Christian Albrechts-Universität Kiel, Olshausenstr. 40, D-24098 Kiel, Germany
| |
Collapse
|
247
|
Zheng J, Jeon S, Jiang W, Burbulla LF, Ysselstein D, Oevel K, Krainc D, Silverman RB. Conversion of Quinazoline Modulators from Inhibitors to Activators of β-Glucocerebrosidase. J Med Chem 2019; 62:1218-1230. [PMID: 30645117 DOI: 10.1021/acs.jmedchem.8b01294] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Gaucher's disease is a lysosomal disease caused by mutations in the β-glucocerebrosidase gene ( GBA1 and GCase) that have been also linked to increased risk of Parkinson's disease (PD) and Diffuse Lewy body dementia. Prior studies have suggested that mutant GCase protein undergoes misfolding and degradation, and therefore, stabilization of the mutant protein represents an important therapeutic strategy in synucleinopathies. In this work, we present a structure-activity relationship (SAR) study of quinazoline compounds that serve as inhibitors of GCase. Unexpectedly, we found that N-methylation of these inhibitors transformed them into GCase activators. A systematic SAR study further revealed that replacement of the key oxygen atom in the linker of the quinazoline derivative also contributed to the activity switch. PD patient-derived fibroblasts and dopaminergic midbrain neurons were treated with a selected compound (9q) that partially stabilized GCase and improved its activity. These results highlight a novel strategy for therapeutic development of noninhibitory GCase modulators in PD and related synucleinopathies.
Collapse
Affiliation(s)
- Jianbin Zheng
- Department of Chemistry, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, and Center for Developmental Therapeutics , Northwestern University , Evanston , Illinois 60208 , United States.,Department of Neurology , Northwestern University Feinberg School of Medicine , Chicago , Illinois 60611 , United States
| | - Sohee Jeon
- Department of Neurology , Northwestern University Feinberg School of Medicine , Chicago , Illinois 60611 , United States
| | - Weilan Jiang
- Department of Neurology , Northwestern University Feinberg School of Medicine , Chicago , Illinois 60611 , United States
| | - Lena F Burbulla
- Department of Neurology , Northwestern University Feinberg School of Medicine , Chicago , Illinois 60611 , United States
| | - Daniel Ysselstein
- Department of Neurology , Northwestern University Feinberg School of Medicine , Chicago , Illinois 60611 , United States
| | - Kristine Oevel
- Department of Neurology , Northwestern University Feinberg School of Medicine , Chicago , Illinois 60611 , United States
| | - Dimitri Krainc
- Department of Neurology , Northwestern University Feinberg School of Medicine , Chicago , Illinois 60611 , United States
| | - Richard B Silverman
- Department of Chemistry, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, and Center for Developmental Therapeutics , Northwestern University , Evanston , Illinois 60208 , United States
| |
Collapse
|
248
|
Tarakad A, Jankovic J. Essential Tremor and Parkinson's Disease: Exploring the Relationship. Tremor Other Hyperkinet Mov (N Y) 2019; 8:589. [PMID: 30643667 PMCID: PMC6329774 DOI: 10.7916/d8md0gvr] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/04/2018] [Indexed: 12/31/2022] Open
Abstract
Background There is longstanding controversy surrounding the possible link between essential tremor (ET) and Parkinson's disease (PD). Inconsistent and unreliable diagnostic criteria may in part account for some of the difficulties in defining the relationship between these two common movement disorders. Methods References for this systematic review were identified using PubMed with the search terms "essential tremor" AND "Parkinson's disease" with articles published in English between 1960 and September 2018 included. Results In this review we provide evidence that some patients diagnosed with ET have an increased risk of developing PD years or decades after onset of action tremor. There are several still unresolved questions about the link between the two disorders including lack of verifiable diagnostic criteria for the two disorders and marked overlap in phenomenology. Here we review clinical, epidemiologic, imaging, pathologic, and genetic studies that address the ET-PD relationship. Several lines of evidence support the association between ET and PD, including overlapping motor and non-motor features, relatively high prevalence of rapid eye movement sleep behavior disorder (26-43%) in ET patients, increased prevalence of PD in patients with longstanding antecedent ET, increased prevalence of ET in family members of patients with PD, and the presence of Lewy bodies in the brains of some ET patients (15-24%). Discussion There is a substantial body of evidence supporting the association between ET and PD within at least a subset of patients, although the nature and possible pathogenic mechanisms of the relationship are not well understood.
Collapse
Affiliation(s)
- Arjun Tarakad
- Parkinson’s Disease Center and Movement Disorders Clinic, Baylor College of Medicine Houston, TX, USA
| | - Joseph Jankovic
- Parkinson’s Disease Center and Movement Disorders Clinic, Baylor College of Medicine Houston, TX, USA
| |
Collapse
|
249
|
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease characterized by a progressive loss of dopaminergic neurons from the nigrostriatal pathway, formation of Lewy bodies, and microgliosis. During the past decades multiple cellular pathways have been associated with PD pathology (i.e., oxidative stress, endosomal-lysosomal dysfunction, endoplasmic reticulum stress, and immune response), yet disease-modifying treatments are not available. We have recently used genetic data from familial and sporadic cases in an unbiased approach to build a molecular landscape for PD, revealing lipids as central players in this disease. Here we extensively review the current knowledge concerning the involvement of various subclasses of fatty acyls, glycerolipids, glycerophospholipids, sphingolipids, sterols, and lipoproteins in PD pathogenesis. Our review corroborates a central role for most lipid classes, but the available information is fragmented, not always reproducible, and sometimes differs by sex, age or PD etiology of the patients. This hinders drawing firm conclusions about causal or associative effects of dietary lipids or defects in specific steps of lipid metabolism in PD. Future technological advances in lipidomics and additional systematic studies on lipid species from PD patient material may improve this situation and lead to a better appreciation of the significance of lipids for this devastating disease.
Collapse
|
250
|
Gustavsson N, Marote A, Pomeshchik Y, Russ K, Azevedo C, Chumarina M, Goldwurm S, Collin A, Pinto L, Salgado AJ, Klementieva O, Roybon L, Savchenko E. Generation of an induced pluripotent stem cell line (CSC-46) from a patient with Parkinson's disease carrying a novel p.R301C mutation in the GBA gene. Stem Cell Res 2019; 34:101373. [DOI: 10.1016/j.scr.2018.101373] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 12/17/2018] [Indexed: 11/26/2022] Open
|