51
|
Pang SYY, Lo RCN, Ho PWL, Liu HF, Chang EES, Leung CT, Malki Y, Choi ZYK, Wong WY, Kung MHW, Ramsden DB, Ho SL. LRRK2, GBA and their interaction in the regulation of autophagy: implications on therapeutics in Parkinson's disease. Transl Neurodegener 2022; 11:5. [PMID: 35101134 PMCID: PMC8805403 DOI: 10.1186/s40035-022-00281-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/12/2022] [Indexed: 02/06/2023] Open
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) and glucocerebrosidase (GBA) represent two most common genetic causes of Parkinson’s disease (PD). Both genes are important in the autophagic-lysosomal pathway (ALP), defects of which are associated with α-synuclein (α-syn) accumulation. LRRK2 regulates macroautophagy via activation of the mitogen activated protein kinase/extracellular signal regulated protein kinase (MAPK/ERK) kinase (MEK) and the calcium-dependent adenosine monophosphate (AMP)-activated protein kinase (AMPK) pathways. Phosphorylation of Rab GTPases by LRRK2 regulates lysosomal homeostasis and endosomal trafficking. Mutant LRRK2 impairs chaperone-mediated autophagy, resulting in α-syn binding and oligomerization on lysosomal membranes. Mutations in GBA reduce glucocerebrosidase (GCase) activity, leading to glucosylceramide accumulation, α-syn aggregation and broad autophagic abnormalities. LRRK2 and GBA influence each other: GCase activity is reduced in LRRK2 mutant cells, and LRRK2 kinase inhibition can alter GCase activity in GBA mutant cells. Clinically, LRRK2 G2019S mutation seems to modify the effects of GBA mutation, resulting in milder symptoms than those resulting from GBA mutation alone. However, dual mutation carriers have an increased risk of PD and earlier age of onset compared with single mutation carriers, suggesting an additive deleterious effect on the initiation of PD pathogenic processes. Crosstalk between LRRK2 and GBA in PD exists, but its exact mechanism is unclear. Drugs that inhibit LRRK2 kinase or activate GCase are showing efficacy in pre-clinical models. Since LRRK2 kinase and GCase activities are also altered in idiopathic PD (iPD), it remains to be seen if these drugs will be useful in disease modification of iPD.
Collapse
|
52
|
Abeliovich A, Hefti F, Sevigny J. Gene Therapy for Parkinson's Disease Associated with GBA1 Mutations. JOURNAL OF PARKINSON'S DISEASE 2022; 11:S183-S188. [PMID: 34151863 PMCID: PMC8543272 DOI: 10.3233/jpd-212739] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 11/24/2022]
Abstract
Human genetic studies as well as studies in animal models indicate that lysosomal dysfunction plays a key role in the pathogenesis of Parkinson's disease. Among the lysosomal genes involved, GBA1 has the largest impact on Parkinson's disease risk. Deficiency in the GBA1 encoded enzyme glucocerebrosidase (GCase) leads to the accumulation of the GCase glycolipid substrates glucosylceramide and glucosylsphingosine and ultimately results in toxicity and inflammation and negatively affect many clinical aspects of Parkinson's disease, including disease risk, the severity of presentation, age of onset, and likelihood of progression to dementia. These findings support the view that re-establishing normal levels of GCase enzyme activity may reduce the progression of Parkinson's disease in patients carrying GBA1 mutations. Studies in mouse models indicate that PR001, a AAV9 vector-based gene therapy designed to deliver a functional GBA1 gene to the brain, suggest that this therapeutic approach may slow or stop disease progression. PR001 is currently being evaluated in clinical trials with Parkinson's disease patients carrying GBA1 mutations.
Collapse
Affiliation(s)
- Asa Abeliovich
- Prevail Therapeutics, A Wholly-Owned Subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Franz Hefti
- Prevail Therapeutics, A Wholly-Owned Subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Jeffrey Sevigny
- Prevail Therapeutics, A Wholly-Owned Subsidiary of Eli Lilly and Company, New York, NY, USA
| |
Collapse
|
53
|
Grosso Jasutkar H, Oh SE, Mouradian MM. Therapeutics in the Pipeline Targeting α-Synuclein for Parkinson's Disease. Pharmacol Rev 2022; 74:207-237. [PMID: 35017177 PMCID: PMC11034868 DOI: 10.1124/pharmrev.120.000133] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder and the fastest growing neurologic disease in the world, yet no disease-modifying therapy is available for this disabling condition. Multiple lines of evidence implicate the protein α-synuclein (α-Syn) in the pathogenesis of PD, and as such, there is intense interest in targeting α-Syn for potential disease modification. α-Syn is also a key pathogenic protein in other synucleionpathies, most commonly dementia with Lewy bodies. Thus, therapeutics targeting this protein will have utility in these disorders as well. Here we discuss the various approaches that are being investigated to prevent and mitigate α-Syn toxicity in PD, including clearing its pathologic aggregates from the brain using immunization strategies, inhibiting its misfolding and aggregation, reducing its expression level, enhancing cellular clearance mechanisms, preventing its cell-to-cell transmission within the brain and perhaps from the periphery, and targeting other proteins associated with or implicated in PD that contribute to α-Syn toxicity. We also discuss the therapeutics in the pipeline that harness these strategies. Finally, we discuss the challenges and opportunities for the field in the discovery and development of therapeutics for disease modification in PD. SIGNIFICANCE STATEMENT: PD is the second most common neurodegenerative disorder, for which disease-modifying therapies remain a major unmet need. A large body of evidence points to α-synuclein as a key pathogenic protein in this disease as well as in dementia with Lewy bodies, making it of leading therapeutic interest. This review discusses the various approaches being investigated and progress made to date toward discovering and developing therapeutics that would slow and stop progression of these disabling diseases.
Collapse
Affiliation(s)
- Hilary Grosso Jasutkar
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| | - Stephanie E Oh
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| | - M Maral Mouradian
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| |
Collapse
|
54
|
Bonam SR, Tranchant C, Muller S. Autophagy-Lysosomal Pathway as Potential Therapeutic Target in Parkinson's Disease. Cells 2021; 10:3547. [PMID: 34944054 PMCID: PMC8700067 DOI: 10.3390/cells10123547] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 01/18/2023] Open
Abstract
Cellular quality control systems have gained much attention in recent decades. Among these, autophagy is a natural self-preservation mechanism that continuously eliminates toxic cellular components and acts as an anti-ageing process. It is vital for cell survival and to preserve homeostasis. Several cell-type-dependent canonical or non-canonical autophagy pathways have been reported showing varying degrees of selectivity with regard to the substrates targeted. Here, we provide an updated review of the autophagy machinery and discuss the role of various forms of autophagy in neurodegenerative diseases, with a particular focus on Parkinson's disease. We describe recent findings that have led to the proposal of therapeutic strategies targeting autophagy to alter the course of Parkinson's disease progression.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe-Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Christine Tranchant
- Service de Neurologie, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France;
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, 67400 Illkirch, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67000 Strasbourg, France
| | - Sylviane Muller
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67000 Strasbourg, France
- CNRS and Strasbourg University, Unit Biotechnology and Cell Signaling/Strasbourg Drug Discovery and Development Institute (IMS), 67000 Strasbourg, France
- University of Strasbourg Institute for Advanced Study (USIAS), 67000 Strasbourg, France
| |
Collapse
|
55
|
Vieira SRL, Schapira AHV. Glucocerebrosidase mutations: A paradigm for neurodegeneration pathways. Free Radic Biol Med 2021; 175:42-55. [PMID: 34450264 DOI: 10.1016/j.freeradbiomed.2021.08.230] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/06/2021] [Accepted: 08/12/2021] [Indexed: 02/07/2023]
Abstract
Biallelic (homozygous or compound heterozygous) glucocerebrosidase gene (GBA) mutations cause Gaucher disease, whereas heterozygous mutations are numerically the most important genetic risk factor for Parkinson disease (PD) and are associated with the development of other synucleinopathies, notably Dementia with Lewy Bodies. This phenomenon is not limited to GBA, with converging evidence highlighting further examples of autosomal recessive disease genes increasing neurodegeneration risk in heterozygous mutation carriers. Nevertheless, despite extensive research, the cellular mechanisms by which mutations in GBA, encoding lysosomal enzyme β-glucocerebrosidase (GCase), predispose to neurodegeneration remain incompletely understood. Alpha-synuclein (A-SYN) accumulation, autophagic lysosomal dysfunction, mitochondrial abnormalities, ER stress and neuroinflammation have been proposed as candidate pathogenic pathways in GBA-linked PD. The observation of GCase and A-SYN interactions in PD initiated the development and evaluation of GCase-targeted therapeutics in PD clinical trials.
Collapse
Affiliation(s)
- Sophia R L Vieira
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, United Kingdom
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, United Kingdom.
| |
Collapse
|
56
|
Levodopa-Induced Ocular Dyskinesia in an Early-Onset Parkinson Disease Patient With GBA Mutation. Clin Neuropharmacol 2021; 44:201-204. [PMID: 34654015 PMCID: PMC8594500 DOI: 10.1097/wnf.0000000000000484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Supplemental digital content is available in the text. Objectives The aim of this study was to report a case of levodopa-induced ocular dyskinesia in an early-onset Parkinson disease patient and to investigate the pathogenic gene. Methods We report the case of a 49-year-old male patient with a 13-year history of Parkinson disease. Involuntary eye movements were noticed after treatment with amantadine for limb dyskinesias. Levodopa-induced ocular dyskinesias involving repetitive, transient, and stereotyped rightward deviations of gaze appeared after intake of an antiparkinsonian drug. Limb dyskinesias also occurred simultaneously. We used a next-generation sequencing targeted gene panel and found a heterozygous missense mutation (p.R535H) in GBA. Direct Sanger sequencing verified the missense mutation. Conclusions We report the case of an uncommon early-onset PD patient carrying a GBA mutation presenting ocular dyskinesia. Genetic screening may provide a better mechanistic insight into dyskinesias.
Collapse
|
57
|
Lee CY, Menozzi E, Chau KY, Schapira AHV. Glucocerebrosidase 1 and leucine-rich repeat kinase 2 in Parkinson disease and interplay between the two genes. J Neurochem 2021; 159:826-839. [PMID: 34618942 DOI: 10.1111/jnc.15524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/18/2021] [Accepted: 09/22/2021] [Indexed: 01/24/2023]
Abstract
The glucocerebrosidase 1 gene (GBA1), bi-allelic variants of which cause Gaucher disease (GD), encodes the lysosomal enzyme glucocerebrosidase (GCase) and is a risk factor for Parkinson Disease (PD). GBA1 variants are linked to a reduction in GCase activity in the brain. Variants in Leucine-Rich Repeat Kinase 2 (LRRK2), such as the gain-of-kinase-function variant G2019S, cause the most common familial form of PD. In patients without GBA1 and LRRK2 mutations, GCase and LRRK2 activity are also altered, suggesting that these two genes are implicated in all forms of PD and that they may play a broader role in PD pathogenesis. In this review, we review the proposed roles of GBA1 and LRRK2 in PD, focussing on the endolysosomal pathway. In particular, we highlight the discovery of Ras-related in brain (Rab) guanosine triphosphatases (GTPases) as LRRK2 kinase substrates and explore the links between increased LRRK2 activity and Rab protein function, lysosomal dysfunction, alpha-synuclein accumulation and GCase activity. We also discuss the discovery of RAB10 as a potential mediator of LRRK2 and GBA1 interaction in PD. Finally, we discuss the therapeutic implications of these findings, including current approaches and future perspectives related to novel drugs targeting LRRK2 and GBA1.
Collapse
Affiliation(s)
- Chiao-Yin Lee
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Elisa Menozzi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Kai-Yin Chau
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| |
Collapse
|
58
|
Gegg ME, Verona G, Schapira AHV. Glucocerebrosidase deficiency promotes release of α-synuclein fibrils from cultured neurons. Hum Mol Genet 2021; 29:1716-1728. [PMID: 32391886 PMCID: PMC7322566 DOI: 10.1093/hmg/ddaa085] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 01/17/2023] Open
Abstract
Mutations in the GBA gene, which encodes the lysosomal enzyme glucocerebrosidase (GCase), are the most important genetic risk factor for Parkinson disease (PD). GCase activity is also decreased in sporadic PD brains and with normal ageing. Loss of GCase activity impairs the autophagy lysosomal pathway resulting in increased α-synuclein (α-syn) levels. Furthermore, elevated α-syn results in decreased GCase activity. Although the role of α-syn in PD remains unclear, evidence indicates that aggregated α-syn fibrils are a pathogenic species in PD, passing between neurons and inducing endogenous native α-syn to aggregate; spreading pathology through the brain. We have investigated if preformed α-syn fibrils (PFFs) impair GCase activity in mouse cortical neurons and differentiated dopaminergic cells, and whether GCase deficiency in these models increased the transfer of α-syn pathology to naïve cells. Neurons treated with PFFs induced endogenous α-syn to become insoluble and phosphorylated at Ser129 to a greater extent than monomeric α-syn-treatment. PFFs, but not monomeric α-syn, inhibited lysosomal GCase activity in these cells and induced the unfolded protein response. Neurons in which GCase was inhibited by conduritol β-epoxide did not increase the amount of insoluble monomeric α-syn or its phosphorylation status. Instead the release of α-syn fibrils from GCase deficient cells was significantly increased. Co-culture studies showed that the transfer of α-syn pathology to naïve cells was greater from GCase deficient cells. This study suggests that GCase deficiency increases the spread of α-syn pathology and likely contributes to the earlier age of onset and increased cognitive decline associated with GBA-PD.
Collapse
Affiliation(s)
- Matthew E Gegg
- Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, Rowland Hill Street, London NW3 2PF, UK
| | - Guglielmo Verona
- Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, London NW3 2PF, UK
| | - Anthony H V Schapira
- Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, Rowland Hill Street, London NW3 2PF, UK
| |
Collapse
|
59
|
Fletcher EJR, Kaminski T, Williams G, Duty S. Drug repurposing strategies of relevance for Parkinson's disease. Pharmacol Res Perspect 2021; 9:e00841. [PMID: 34309236 PMCID: PMC8311732 DOI: 10.1002/prp2.841] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/29/2021] [Indexed: 01/01/2023] Open
Abstract
Parkinson's disease is a highly disabling, progressive neurodegenerative disease that manifests as a mix of motor and non-motor signs. Although we are equipped with some symptomatic treatments, especially for the motor signs of the disease, there are still no established disease-modifying drugs so the disease progresses unchecked. Standard drug discovery programs for disease-modifying therapies have provided key insights into the pathogenesis of Parkinson's disease but, of the many positive candidates identified in pre-clinical studies, none has yet translated into a successful clinically efficacious drug. Given the huge cost of drug discovery programs, it is not surprising that much attention has turned toward repurposing strategies. The trialing of an established therapeutic has the advantage of bypassing the need for preclinical safety testing and formulation optimization, thereby cutting both time and costs involved in getting a treatment to the clinic. Additional reduced failure rates for repurposed drugs are also a potential bonus. Many different strategies for drug repurposing are open to researchers in the Parkinson's disease field. Some of these have already proven effective in identifying suitable drugs for clinical trials, lending support to such approaches. In this review, we present a summary of the different strategies for drug repurposing, from large-scale epidemiological correlation analysis through to single-gene transcriptional approaches. We provide examples of past or ongoing studies adopting each strategy, where these exist. For strategies that have yet to be applied to Parkinson's disease, their utility is illustrated using examples taken from other disorders.
Collapse
Affiliation(s)
- Edward J. R. Fletcher
- King’s College LondonInstitute of Psychiatry, Psychology & NeuroscienceWolfson Centre for Age‐Related DiseasesLondonUK
| | - Thomas Kaminski
- King’s College LondonInstitute of Psychiatry, Psychology & NeuroscienceWolfson Centre for Age‐Related DiseasesLondonUK
| | - Gareth Williams
- King’s College LondonInstitute of Psychiatry, Psychology & NeuroscienceWolfson Centre for Age‐Related DiseasesLondonUK
| | - Susan Duty
- King’s College LondonInstitute of Psychiatry, Psychology & NeuroscienceWolfson Centre for Age‐Related DiseasesLondonUK
| |
Collapse
|
60
|
Wild-type GBA1 increases the α-synuclein tetramer-monomer ratio, reduces lipid-rich aggregates, and attenuates motor and cognitive deficits in mice. Proc Natl Acad Sci U S A 2021; 118:2103425118. [PMID: 34326260 PMCID: PMC8346893 DOI: 10.1073/pnas.2103425118] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The mechanisms responsible for brain α-synuclein (αS) dyshomeostasis, caused by Gaucher’s GBA1 mutations that increase Parkinson’s disease (PD) risk, are largely unknown. We previously showed that abrogating physiological αS tetramers by a familial PD-E46K–amplified 3K mutation produces PD-like syndrome in mice and that treatment with stearoyl-CoA desaturase inhibitors increased a portion of the αS tetramers, benefitting the motor phenotypes. Here, we show that—similar to previous findings in GBA1-mutant PD culture—GCase elevation prolonged the stabilization of wild-type and 3K mutant αS tetramers in wtGBA1–transduced mouse brains, improving lysosomal integrity and motor and cognitive phenotypes. These data help elucidating lipid modulators that impact the αS physiological state in vivo and the development of PD therapeutic approaches. Loss-of-function mutations in acid beta-glucosidase 1 (GBA1) are among the strongest genetic risk factors for Lewy body disorders such as Parkinson’s disease (PD) and Lewy body dementia (DLB). Altered lipid metabolism in PD patient–derived neurons, carrying either GBA1 or PD αS mutations, can shift the physiological α-synuclein (αS) tetramer–monomer (T:M) equilibrium toward aggregation-prone monomers. A resultant increase in pSer129+ αS monomers provides a likely building block for αS aggregates. 3K αS mice, representing a neuropathological amplification of the E46K PD–causing mutation, have decreased αS T:M ratios and vesicle-rich αS+ aggregates in neurons, accompanied by a striking PD-like motor syndrome. We asked whether enhancing glucocerebrosidase (GCase) expression could benefit αS dyshomeostasis by delivering an adeno-associated virus (AAV)–human wild-type (wt) GBA1 vector into the brains of 3K neonates. Intracerebroventricular AAV-wtGBA1 at postnatal day 1 resulted in prominent forebrain neuronal GCase expression, sustained through 6 mo. GBA1 attenuated behavioral deficits both in working memory and fine motor performance tasks. Furthermore, wtGBA1 increased αS solubility and the T:M ratio in both 3K-GBA mice and control littermates and reduced pS129+ and lipid-rich aggregates in 3K-GBA. We observed GCase distribution in more finely dispersed lysosomes, in which there was increased GCase activity, lysosomal cathepsin D and B maturation, decreased perilipin-stabilized lipid droplets, and a normalized TFEB translocation to the nucleus, all indicative of improved lysosomal function and lipid turnover. Therefore, a prolonged increase of the αS T:M ratio by elevating GCase activity reduced the lipid- and vesicle-rich aggregates and ameliorated PD-like phenotypes in mice, further supporting lipid modulating therapies in PD.
Collapse
|
61
|
Abe T, Kuwahara T. Targeting of Lysosomal Pathway Genes for Parkinson's Disease Modification: Insights From Cellular and Animal Models. Front Neurol 2021; 12:681369. [PMID: 34194386 PMCID: PMC8236816 DOI: 10.3389/fneur.2021.681369] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/20/2021] [Indexed: 01/01/2023] Open
Abstract
Previous genetic studies on hereditary Parkinson's disease (PD) have identified a set of pathogenic gene mutations that have strong impacts on the pathogenicity of PD. In addition, genome-wide association studies (GWAS) targeted to sporadic PD have nominated an increasing number of genetic variants that influence PD susceptibility. Although the clinical and pathological characteristics in hereditary PD are not identical to those in sporadic PD, α-synuclein, and LRRK2 are definitely associated with both types of PD, with LRRK2 mutations being the most frequent cause of autosomal-dominant PD. On the other hand, a significant portion of risk genes identified from GWAS have been associated with lysosomal functions, pointing to a critical role of lysosomes in PD pathogenesis. Experimental studies have suggested that the maintenance or upregulation of lysosomal activity may protect against neuronal dysfunction or degeneration. Here we focus on the roles of representative PD gene products that are implicated in lysosomal pathway, namely LRRK2, VPS35, ATP13A2, and glucocerebrosidase, and provide an overview of their disease-associated functions as well as their cooperative actions in the pathogenesis of PD, based on the evidence from cellular and animal models. We also discuss future perspectives of targeting lysosomal activation as a possible strategy to treat neurodegeneration.
Collapse
Affiliation(s)
- Tetsuro Abe
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomoki Kuwahara
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
62
|
Nakanishi E, Uemura N, Akiyama H, Kinoshita M, Masanori S, Taruno Y, Yamakado H, Matsuzawa SI, Takeda S, Hirabayashi Y, Takahashi R. Impact of Gba2 on neuronopathic Gaucher's disease and α-synuclein accumulation in medaka (Oryzias latipes). Mol Brain 2021; 14:80. [PMID: 33971917 PMCID: PMC8111776 DOI: 10.1186/s13041-021-00790-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 05/05/2021] [Indexed: 12/21/2022] Open
Abstract
Homozygous mutations in the lysosomal glucocerebrosidase gene, GBA1, cause Gaucher’s disease (GD), while heterozygous mutations in GBA1 are a strong risk factor for Parkinson’s disease (PD), whose pathological hallmark is intraneuronal α-synuclein (asyn) aggregates. We previously reported that gba1 knockout (KO) medaka exhibited glucosylceramide accumulation and neuronopathic GD phenotypes, including short lifespan, the dopaminergic and noradrenergic neuronal cell loss, microglial activation, and swimming abnormality, with asyn accumulation in the brains. A recent study reported that deletion of GBA2, non-lysosomal glucocerebrosidase, in a non-neuronopathic GD mouse model rescued its phenotypes. In the present study, we generated gba2 KO medaka and examined the effect of Gba2 deletion on the phenotypes of gba1 KO medaka. The Gba2 deletion in gba1 KO medaka resulted in the exacerbation of glucosylceramide accumulation and no improvement in neuronopathic GD pathological changes, asyn accumulation, or swimming abnormalities. Meanwhile, though gba2 KO medaka did not show any apparent phenotypes, biochemical analysis revealed asyn accumulation in the brains. gba2 KO medaka showed a trend towards an increase in sphingolipids in the brains, which is one of the possible causes of asyn accumulation. In conclusion, this study demonstrated that the deletion of Gba2 does not rescue the pathological changes or behavioral abnormalities of gba1 KO medaka, and GBA2 represents a novel factor affecting asyn accumulation in the brains.
Collapse
Affiliation(s)
- Etsuro Nakanishi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Norihito Uemura
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan. .,Department of Pathology and Laboratory Medicine, Institute On Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104-2676, USA.
| | - Hisako Akiyama
- Laboratory for Neural Cell Dynamics, RIKEN Center for Brain Science, Saitama, 351-0198, Japan
| | - Masato Kinoshita
- Division of Applied Bioscience, Kyoto University Graduate School of Agriculture, Kyoto, 606-8502, Japan
| | - Sawamura Masanori
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Yosuke Taruno
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Hodaka Yamakado
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Shu-Ichi Matsuzawa
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Shunichi Takeda
- Department of Radiation Genetics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | | | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan.
| |
Collapse
|
63
|
Prasuhn J, Brüggemann N. Genotype-driven therapeutic developments in Parkinson's disease. Mol Med 2021; 27:42. [PMID: 33874883 PMCID: PMC8056568 DOI: 10.1186/s10020-021-00281-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/12/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Remarkable advances have been reached in the understanding of the genetic basis of Parkinson's disease (PD), with the identification of monogenic causes (mPD) and a plethora of gene loci leading to an increased risk for idiopathic PD. The expanding knowledge and subsequent identification of genetic contributions fosters the understanding of molecular mechanisms leading to disease development and progression. Distinct pathways involved in mitochondrial dysfunction, oxidative stress, and lysosomal function have been identified and open a unique window of opportunity for individualized treatment approaches. These genetic findings have led to an imminent progress towards pathophysiology-targeted clinical trials and potentially disease-modifying treatments in the future. MAIN BODY OF THE MANUSCRIPT In this review article we will summarize known genetic contributors to the pathophysiology of Parkinson's disease, the molecular mechanisms leading to disease development, and discuss challenges and opportunities in clinical trial designs. CONCLUSIONS The future success of clinical trials in PD is mainly dependent on reliable biomarker development and extensive genetic testing to identify genetic cases. Whether genotype-dependent stratification of study participants will extend the potential application of new drugs will be one major challenge in conceptualizing clinical trials. However, the latest developments in genotype-driven treatments will pave the road to individualized pathophysiology-based therapies in the future.
Collapse
Affiliation(s)
- Jannik Prasuhn
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Norbert Brüggemann
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany.
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany.
- Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
64
|
Yang CN, Peng WY, Lin LC, Tsai TH. Protein unbound pharmacokinetics of ambroxol in the blood and brains of rats and the interaction of ambroxol with Polygala tenuifolia by multiple microdialysis. JOURNAL OF ETHNOPHARMACOLOGY 2021; 269:113764. [PMID: 33383115 DOI: 10.1016/j.jep.2020.113764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/29/2020] [Accepted: 12/24/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ambroxol elevates glucocerebrosidase (GCase) activity and reduces nigrostriatal alpha-synuclein burden to better ameliorate motor function in Parkinson's disease (PD). Polygala tenuifolia is a potential alternative botanical medicine for the treatment of many nonmotor symptoms of PD commonly used in Taiwanese patients. Co-administration of these two medicines pose potential herb-drug interaction. AIM OF THE STUDY Our hypothesis is that ambroxol and P. tenuifolia may potentially possess herbal drug synergetic effects in the blood and brain. MATERIALS AND METHODS To investigate this hypothesis, a multiple microdialysis system coupled with validated ultra-high performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method was developed for rat blood and brain samples. Experimental rats were divided into three groups: low-dose and high-dose ambroxol alone (10 mg/kg, i.v. and 30 mg/kg, i.v., respectively) and ambroxol (10 mg/kg, i.v.) pretreated with P. tenuifolia extract (1 g/kg, p.o. for 5 consecutive days). RESULTS Ambroxol easily penetrated into the brain and reached a maximum concentration in the striatum at approximately 60 min after low- and high-dose treatment. The area under the concentration curve (AUC) ratio increased proportionally at the doses of 10 and 30 mg/kg, which suggested a linear pharmacokinetic manner of ambroxol. The brain penetration of ambroxol was approximately 30-34%, which was defined as the ambroxol AUC blood-to-brain distribution ratio (AUCbrain/AUCblood). The P. tenuifolia extract did not significantly alter the pharmacokinetics of ambroxol in the blood and brain of rats. CONCLUSION The present study suggests that it is safety without pharmacokinetic interactions for this dosing regimen to use P. tenuifolia extract and ambroxol together.
Collapse
Affiliation(s)
- Chao-Nan Yang
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan; Department of Neurology, China Medical University Hospital-Taipei Branch, China Medical University, Taichung, 40402, Taiwan
| | - Wen-Ya Peng
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan
| | - Lie-Chwen Lin
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, 112, Taiwan
| | - Tung-Hu Tsai
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan; Graduate Institute of Acupuncture Science, China Medical University, Taichung, 40402, Taiwan; School of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| |
Collapse
|
65
|
Senkevich KA, Kopytova AE, Usenko TS, Emelyanov AK, Pchelina SN. Parkinson's Disease Associated with GBA Gene Mutations: Molecular Aspects and Potential Treatment Approaches. Acta Naturae 2021; 13:70-78. [PMID: 34377557 PMCID: PMC8327146 DOI: 10.32607/actanaturae.11031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/03/2020] [Indexed: 01/01/2023] Open
Abstract
Parkinson's disease (PD) is a multifactorial neurodegenerative disease. To date, genome-wide association studies have identified more than 70 loci associated with the risk of PD. Variants in the GBA gene encoding glucocerebrosidase are quite often found in PD patients in all populations across the world, which justifies intensive investigation of this gene. A number of biochemical features have been identified in patients with GBA-associated Parkinson's disease (GBA-PD). In particular, these include decreased activity of glucocerebrosidase and accumulation of the glucosylceramide substrate. These features were the basis for putting forward a hypothesis about treatment of GBA-PD using new strategies aimed at restoring glucocerebrosidase activity and reducing the substrate concentration. This paper discusses the molecular and genetic mechanisms of GBA-PD pathogenesis and potential approaches to the treatment of this form of the disease.
Collapse
Affiliation(s)
- K. A. Senkevich
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Saint-Petersburg, 188300 Russia
- First Pavlov State Medical University of St. Petersburg, Saint-Petersburg, 197022 Russia
- Montreal Neurological Institute, McGill University, Montréal, QC, H3A 1A1, Canada
| | - A. E. Kopytova
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Saint-Petersburg, 188300 Russia
- First Pavlov State Medical University of St. Petersburg, Saint-Petersburg, 197022 Russia
| | - T. S. Usenko
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Saint-Petersburg, 188300 Russia
- First Pavlov State Medical University of St. Petersburg, Saint-Petersburg, 197022 Russia
| | - A. K. Emelyanov
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Saint-Petersburg, 188300 Russia
- First Pavlov State Medical University of St. Petersburg, Saint-Petersburg, 197022 Russia
| | - S. N. Pchelina
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre «Kurchatov Institute», Saint-Petersburg, 188300 Russia
- First Pavlov State Medical University of St. Petersburg, Saint-Petersburg, 197022 Russia
- Institute of Experimental Medicine, St. Petersburg, 197376 Russia
| |
Collapse
|
66
|
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder resulting from the death of dopamine neurons in the substantia nigra pars compacta. Our understanding of PD biology has been enriched by the identification of genes involved in its rare, inheritable forms, termed PARK genes. These genes encode proteins including α-syn, LRRK2, VPS35, parkin, PINK1, and DJ1, which can cause monogenetic PD when mutated. Investigating the cellular functions of these proteins has been instrumental in identifying signaling pathways that mediate pathology in PD and neuroprotective mechanisms active during homeostatic and pathological conditions. It is now evident that many PD-associated proteins perform multiple functions in PD-associated signaling pathways in neurons. Furthermore, several PARK proteins contribute to non-cell-autonomous mechanisms of neuron death, such as neuroinflammation. A comprehensive understanding of cell-autonomous and non-cell-autonomous pathways involved in PD is essential for developing therapeutics that may slow or halt its progression.
Collapse
Affiliation(s)
- Nikhil Panicker
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Preston Ge
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA.,Diana Helis Henry Medical Research Foundation, New Orleans, LA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA.,Diana Helis Henry Medical Research Foundation, New Orleans, LA
| |
Collapse
|
67
|
Stott SRW, Wyse RK, Brundin P. Drug Repurposing for Parkinson's Disease: The International Linked Clinical Trials experience. Front Neurosci 2021; 15:653377. [PMID: 33815053 PMCID: PMC8017145 DOI: 10.3389/fnins.2021.653377] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/19/2021] [Indexed: 11/27/2022] Open
Abstract
The international Linked Clinical Trials (iLCT) program for Parkinson's to date represents one of the most comprehensive drug repurposing programs focused on one disease. Since initial planning in 2010, it has rapidly grown - giving rise to seven completed, and 15 ongoing, clinical trials of 16 agents each aimed at delivering disease modification in Parkinson's disease (PD). In this review, we will provide an overview of the history, structure, process, and progress of the program. We will also present some examples of agents that have been selected and prioritized by the program and subsequently evaluated in clinical trials. Our goal with this review is to provide a template that can be considered across other therapeutic areas.
Collapse
Affiliation(s)
| | | | - Patrik Brundin
- Parkinson’s Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| |
Collapse
|
68
|
Ge H, Zhang C, Yang Y, Chen W, Zhong J, Fang X, Jiang X, Tan L, Zou Y, Hu R, Chen Y, Feng H. Ambroxol Upregulates Glucocerebrosidase Expression to Promote Neural Stem Cells Differentiation Into Neurons Through Wnt/β-Catenin Pathway After Ischemic Stroke. Front Mol Neurosci 2021; 13:596039. [PMID: 33551744 PMCID: PMC7855720 DOI: 10.3389/fnmol.2020.596039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/22/2020] [Indexed: 12/29/2022] Open
Abstract
Ischemic stroke has been becoming one of the leading causes resulting in mortality and adult long-term disability worldwide. Post-stroke pneumonia is a common complication in patients with ischemic stroke and always associated with 1-year mortality. Though ambroxol therapy often serves as a supplementary treatment for post-stroke pneumonia in ischemic stroke patients, its effect on functional recovery and potential mechanism after ischemic stroke remain elusive. In the present study, the results indicated that administration of 70 mg/kg and 100 mg/kg enhanced functional recovery by virtue of decreasing infarct volume. The potential mechanism, to some extent, was due to promoting NSCs differentiation into neurons and interfering NSCs differentiation into astrocytes through increasing GCase expression to activate Wnt/β-catenin signaling pathway in penumbra after ischemic stroke, which advanced basic knowledge of ambroxol in regulating NSCs differentiation and provided a feasible therapy for ischemic stroke treatment, even in other brain disorders in clinic.
Collapse
Affiliation(s)
- Hongfei Ge
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chao Zhang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yang Yang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Weixiang Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jun Zhong
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xuanyu Fang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xuheng Jiang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Liang Tan
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yongjie Zou
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Department of Neurosurgery, Hospital of People's Liberation Army, Nanchang, China
| | - Rong Hu
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yujie Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hua Feng
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
69
|
Behl T, Kaur G, Fratila O, Buhas C, Judea-Pusta CT, Negrut N, Bustea C, Bungau S. Cross-talks among GBA mutations, glucocerebrosidase, and α-synuclein in GBA-associated Parkinson's disease and their targeted therapeutic approaches: a comprehensive review. Transl Neurodegener 2021; 10:4. [PMID: 33446243 PMCID: PMC7809876 DOI: 10.1186/s40035-020-00226-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/01/2020] [Indexed: 02/08/2023] Open
Abstract
Current therapies for Parkinson's disease (PD) are palliative, of which the levodopa/carbidopa therapy remains the primary choice but is unable to modulate the progression of neurodegeneration. Due to the complication of such a multifactorial disorder and significant limitations of the therapy, numerous genetic approaches have been proved effective in finding out genes and mechanisms implicated in this disease. Following the observation of a higher frequency of PD in Gaucher's disease (GD), a lysosomal storage condition, mutations of glycosylceramidase beta (GBA) encoding glucocerebrosidase (GCase) have been shown to be involved and have been explored in the context of PD. GBA mutations are the most common genetic risk factor of PD. Various studies have revealed the relationships between PD and GBA gene mutations, facilitating a better understanding of this disorder. Various hypotheses delineate that the pathological mutations of GBA minimize the enzymatic activity of GCase, which affects the proliferation and clearance of α-synuclein; this affects the lysosomal homeostasis, exacerbating the endoplasmic reticulum stress or encouraging the mitochondrial dysfunction. Identification of the pathological mechanisms underlying the GBA-associated parkinsonism (GBA + PD) advances our understanding of PD. This review based on current literature aims to elucidate various genetic and clinical characteristics correlated with GBA mutations and to identify the numerous pathological processes underlying GBA + PD. We also delineate the therapeutic strategies to interfere with the mutant GCase function for further improvement of the related α-synuclein-GCase crosstalks. Moreover, the various therapeutic approaches such as gene therapy, chaperone proteins, and histone deacetylase inhibitors for the treatment of GBA + PD are discussed.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Gagandeep Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ovidiu Fratila
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Camelia Buhas
- Department of Morphological Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Bihor County, Romania
| | - Claudia Teodora Judea-Pusta
- Department of Morphological Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Bihor County, Romania
| | - Nicoleta Negrut
- Department of Psycho-Neuroscience and Recovery, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Cristiana Bustea
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
70
|
Behl T, Kaur G, Fratila O, Buhas C, Judea-Pusta CT, Negrut N, Bustea C, Bungau S. Cross-talks among GBA mutations, glucocerebrosidase, and α-synuclein in GBA-associated Parkinson’s disease and their targeted therapeutic approaches: a comprehensive review. Transl Neurodegener 2021. [DOI: https://doi.org/10.1186/s40035-020-00226-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
AbstractCurrent therapies for Parkinson’s disease (PD) are palliative, of which the levodopa/carbidopa therapy remains the primary choice but is unable to modulate the progression of neurodegeneration. Due to the complication of such a multifactorial disorder and significant limitations of the therapy, numerous genetic approaches have been proved effective in finding out genes and mechanisms implicated in this disease. Following the observation of a higher frequency of PD in Gaucher’s disease (GD), a lysosomal storage condition, mutations of glycosylceramidase beta (GBA) encoding glucocerebrosidase (GCase) have been shown to be involved and have been explored in the context of PD. GBA mutations are the most common genetic risk factor of PD. Various studies have revealed the relationships between PD and GBA gene mutations, facilitating a better understanding of this disorder. Various hypotheses delineate that the pathological mutations of GBA minimize the enzymatic activity of GCase, which affects the proliferation and clearance of α-synuclein; this affects the lysosomal homeostasis, exacerbating the endoplasmic reticulum stress or encouraging the mitochondrial dysfunction. Identification of the pathological mechanisms underlying the GBA-associated parkinsonism (GBA + PD) advances our understanding of PD. This review based on current literature aims to elucidate various genetic and clinical characteristics correlated with GBA mutations and to identify the numerous pathological processes underlying GBA + PD. We also delineate the therapeutic strategies to interfere with the mutant GCase function for further improvement of the related α-synuclein–GCase crosstalks. Moreover, the various therapeutic approaches such as gene therapy, chaperone proteins, and histone deacetylase inhibitors for the treatment of GBA + PD are discussed.
Collapse
|
71
|
Schneider R, Leven P, Glowka T, Kuzmanov I, Lysson M, Schneiker B, Miesen A, Baqi Y, Spanier C, Grants I, Mazzotta E, Villalobos‐Hernandez E, Kalff JC, Müller CE, Christofi FL, Wehner S. A novel P2X2-dependent purinergic mechanism of enteric gliosis in intestinal inflammation. EMBO Mol Med 2021; 13:e12724. [PMID: 33332729 PMCID: PMC7799361 DOI: 10.15252/emmm.202012724] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 12/19/2022] Open
Abstract
Enteric glial cells (EGC) modulate motility, maintain gut homeostasis, and contribute to neuroinflammation in intestinal diseases and motility disorders. Damage induces a reactive glial phenotype known as "gliosis", but the molecular identity of the inducing mechanism and triggers of "enteric gliosis" are poorly understood. We tested the hypothesis that surgical trauma during intestinal surgery triggers ATP release that drives enteric gliosis and inflammation leading to impaired motility in postoperative ileus (POI). ATP activation of a p38-dependent MAPK pathway triggers cytokine release and a gliosis phenotype in murine (and human) EGCs. Receptor antagonism and genetic depletion studies revealed P2X2 as the relevant ATP receptor and pharmacological screenings identified ambroxol as a novel P2X2 antagonist. Ambroxol prevented ATP-induced enteric gliosis, inflammation, and protected against dysmotility, while abrogating enteric gliosis in human intestine exposed to surgical trauma. We identified a novel pathogenic P2X2-dependent pathway of ATP-induced enteric gliosis, inflammation and dysmotility in humans and mice. Interventions that block enteric glial P2X2 receptors during trauma may represent a novel therapy in treating POI and immune-driven intestinal motility disorders.
Collapse
Affiliation(s)
| | | | - Tim Glowka
- Department of SurgeryUniversity of BonnBonnGermany
| | | | | | | | - Anna Miesen
- Department of SurgeryUniversity of BonnBonnGermany
| | - Younis Baqi
- Faculty of ScienceDepartment of ChemistrySultan Qaboos UniversityMuscatOman
- Pharmaceutical InstitutePharmaceutical & Medical ChemistryUniversity of BonnBonnGermany
| | - Claudia Spanier
- Pharmaceutical InstitutePharmaceutical & Medical ChemistryUniversity of BonnBonnGermany
| | - Iveta Grants
- Department of AnesthesiologyWexner Medical CenterThe Ohio State UniversityColumbusOHUSA
| | - Elvio Mazzotta
- Department of AnesthesiologyWexner Medical CenterThe Ohio State UniversityColumbusOHUSA
| | | | - Jörg C Kalff
- Department of SurgeryUniversity of BonnBonnGermany
| | - Christa E Müller
- Pharmaceutical InstitutePharmaceutical & Medical ChemistryUniversity of BonnBonnGermany
| | - Fedias L Christofi
- Department of AnesthesiologyWexner Medical CenterThe Ohio State UniversityColumbusOHUSA
| | - Sven Wehner
- Department of SurgeryUniversity of BonnBonnGermany
| |
Collapse
|
72
|
Aslam M, Kandasamy N, Ullah A, Paramasivam N, Öztürk MA, Naureen S, Arshad A, Badshah M, Khan K, Wajid M, Abbasi R, Ilyas M, Eils R, Schlesner M, Wade RC, Ahmad N, von Engelhardt J. Putative second hit rare genetic variants in families with seemingly GBA-associated Parkinson's disease. NPJ Genom Med 2021; 6:2. [PMID: 33402667 PMCID: PMC7785741 DOI: 10.1038/s41525-020-00163-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 11/12/2020] [Indexed: 11/08/2022] Open
Abstract
Rare variants in the beta-glucocerebrosidase gene (GBA1) are common genetic risk factors for alpha synucleinopathy, which often manifests clinically as GBA-associated Parkinson's disease (GBA-PD). Clinically, GBA-PD closely mimics idiopathic PD, but it may present at a younger age and often aggregates in families. Most carriers of GBA variants are, however, asymptomatic. Moreover, symptomatic PD patients without GBA variant have been reported in families with seemingly GBA-PD. These observations obscure the link between GBA variants and PD pathogenesis and point towards a role for unidentified additional genetic and/or environmental risk factors or second hits in GBA-PD. In this study, we explored whether rare genetic variants may be additional risk factors for PD in two families segregating the PD-associated GBA1 variants c.115+1G>A (ClinVar ID: 93445) and p.L444P (ClinVar ID: 4288). Our analysis identified rare genetic variants of the HSP70 co-chaperone DnaJ homolog subfamily B member 6 (DNAJB6) and lysosomal protein prosaposin (PSAP) as additional factors possibly influencing PD risk in the two families. In comparison to the wild-type proteins, variant DNAJB6 and PSAP proteins show altered functions in the context of cellular alpha-synuclein homeostasis when expressed in reporter cells. Furthermore, the segregation pattern of the rare variants in the genes encoding DNAJB6 and PSAP indicated a possible association with PD in the respective families. The occurrence of second hits or additional PD cosegregating rare variants has important implications for genetic counseling in PD families with GBA1 variant carriers and for the selection of PD patients for GBA targeted treatments.
Collapse
Affiliation(s)
- Muhammad Aslam
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| | - Nirosiya Kandasamy
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Anwar Ullah
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan
- Department of Biochemistry, Quaid-i-Azam University, Islamabad, Pakistan
| | - Nagarajan Paramasivam
- Heidelberg Center for Personalized Oncology (DKFZ-HIPO), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mehmet Ali Öztürk
- Molecular and Cellular Modeling Group, Heidelberg Institute of Theoretical Studies (HITS), Heidelberg, Germany
- The Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104, Freiburg, Germany
| | - Saima Naureen
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Department of Zoology, PMAS-Arid Agriculture University, Rawalpindi, Pakistan
| | - Abida Arshad
- Department of Zoology, PMAS-Arid Agriculture University, Rawalpindi, Pakistan
| | - Mazhar Badshah
- Department of Neurology, Shaheed Zulfiqar Ali Bhutto Medical University, Islamabad, Pakistan
| | - Kafaitullah Khan
- Department of Microbiology, University of Balochistan, Quetta, Pakistan
| | - Muhammad Wajid
- Department of Biological Sciences, University of Okara, Okara, Pakistan
| | - Rashda Abbasi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan
| | - Muhammad Ilyas
- Faculty of Mechanical Engineering, GIK Institute of Engineering Sciences and Technology, Topi, 23460, Pakistan
| | - Roland Eils
- Center for Digital Health, Berlin Institute of Health and Charité Universitätsmedizin Berlin, Berlin, Germany
- Health Data Science Unit, Bioquant, Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Matthias Schlesner
- Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rebecca C Wade
- Molecular and Cellular Modeling Group, Heidelberg Institute of Theoretical Studies (HITS), Heidelberg, Germany
- Center for Molecular Biology of the University of Heidelberg (ZMBH), DKFZ-ZMBH Alliance, and Interdisciplinary Center for Scientific Computing (IWR), Heidelberg, Germany
| | - Nafees Ahmad
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan
| | - Jakob von Engelhardt
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
73
|
Zhang CL, Han QW, Chen NH, Yuan YH. Research on developing drugs for Parkinson's disease. Brain Res Bull 2020; 168:100-109. [PMID: 33387636 DOI: 10.1016/j.brainresbull.2020.12.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 12/22/2020] [Accepted: 12/26/2020] [Indexed: 12/28/2022]
Abstract
Current treatments for Parkinson's disease (PD) are mainly dopaminergic drugs. However, dopaminergic drugs are only symptomatic treatments and limited by several side effects. Recent studies into drug development focused on emerging new molecular mechanisms, including nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, nuclear receptor-related 1 (Nurr1), adenosine receptor A2, nicotine receptor, metabotropic glutamate receptors (mGluRs), and glucocerebrosidase (GCase). Also, immunotherapy and common pathological mechanisms shared with Alzheimer's Disease (AD) and diabetes have attracted much attention. In this review, we summarized the development of preclinical and clinical studies of novel drugs and the improvement of dopaminergic drugs to provide a prospect for PD treatment.
Collapse
Affiliation(s)
- Cheng-Lu Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qi-Wen Han
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yu-He Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
74
|
Minakaki G, Krainc D, Burbulla LF. The Convergence of Alpha-Synuclein, Mitochondrial, and Lysosomal Pathways in Vulnerability of Midbrain Dopaminergic Neurons in Parkinson's Disease. Front Cell Dev Biol 2020; 8:580634. [PMID: 33381501 PMCID: PMC7767856 DOI: 10.3389/fcell.2020.580634] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, characterized by progressive bradykinesia, rigidity, resting tremor, and gait impairment, as well as a spectrum of non-motor symptoms including autonomic and cognitive dysfunction. The cardinal motor symptoms of PD stem from the loss of substantia nigra (SN) dopaminergic (DAergic) neurons, and it remains unclear why SN DAergic neurons are preferentially lost in PD. However, recent identification of several genetic PD forms suggests that mitochondrial and lysosomal dysfunctions play important roles in the degeneration of midbrain dopamine (DA) neurons. In this review, we discuss the interplay of cell-autonomous mechanisms linked to DAergic neuron vulnerability and alpha-synuclein homeostasis. Emerging studies highlight a deleterious feedback cycle, with oxidative stress, altered DA metabolism, dysfunctional lysosomes, and pathological alpha-synuclein species representing key events in the pathogenesis of PD. We also discuss the interactions of alpha-synuclein with toxic DA metabolites, as well as the biochemical links between intracellular iron, calcium, and alpha-synuclein accumulation. We suggest that targeting multiple pathways, rather than individual processes, will be important for developing disease-modifying therapies. In this context, we focus on current translational efforts specifically targeting lysosomal function, as well as oxidative stress via calcium and iron modulation. These efforts could have therapeutic benefits for the broader population of sporadic PD and related synucleinopathies.
Collapse
Affiliation(s)
- Georgia Minakaki
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Dimitri Krainc
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Lena F Burbulla
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
75
|
Wildburger NC, Hartke AS, Schidlitzki A, Richter F. Current Evidence for a Bidirectional Loop Between the Lysosome and Alpha-Synuclein Proteoforms. Front Cell Dev Biol 2020; 8:598446. [PMID: 33282874 PMCID: PMC7705175 DOI: 10.3389/fcell.2020.598446] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/28/2020] [Indexed: 12/28/2022] Open
Abstract
Cumulative evidence collected in recent decades suggests that lysosomal dysfunction contributes to neurodegenerative diseases, especially if amyloid proteins are involved. Among these, alpha-synuclein (aSyn) that progressively accumulates and aggregates in Lewy bodies is undisputedly a main culprit in Parkinson disease (PD) pathogenesis. Lysosomal dysfunction is evident in brains of PD patients, and mutations in lysosomal enzymes are a major risk factor of PD. At first glance, the role of protein-degrading lysosomes in a disease with pathological protein accumulation seems obvious and should guide the development of straightforward and rational therapeutic targets. However, our review demonstrates that the story is more complicated for aSyn. The protein can possess diverse posttranslational modifications, aggregate formations, and truncations, all of which contribute to a growing known set of proteoforms. These interfere directly or indirectly with lysosome function, reducing their own degradation, and thereby accelerating the protein aggregation and disease process. Conversely, unbalanced lysosomal enzymatic processes can produce truncated aSyn proteoforms that may be more toxic and prone to aggregation. This highlights the possibility of enhancing lysosomal function as a treatment for PD, if it can be confirmed that this approach effectively reduces harmful aSyn proteoforms and does not produce novel, toxic proteoforms.
Collapse
Affiliation(s)
- Norelle C Wildburger
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hanover, Germany.,Center for Systems Neuroscience, Hanover, Germany
| | - Anna-Sophia Hartke
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hanover, Germany
| | - Alina Schidlitzki
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hanover, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hanover, Germany.,Center for Systems Neuroscience, Hanover, Germany
| |
Collapse
|
76
|
Bouscary A, Quessada C, René F, Spedding M, Turner BJ, Henriques A, Ngo ST, Loeffler JP. Sphingolipids metabolism alteration in the central nervous system: Amyotrophic lateral sclerosis (ALS) and other neurodegenerative diseases. Semin Cell Dev Biol 2020; 112:82-91. [PMID: 33160824 DOI: 10.1016/j.semcdb.2020.10.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/14/2020] [Accepted: 10/19/2020] [Indexed: 12/11/2022]
Abstract
Sphingolipids are complex lipids. They play a structural role in neurons, but are also involved in regulating cellular communication, and neuronal differentiation and maturation. There is increasing evidence to suggest that dysregulated metabolism of sphingolipids is linked to neurodegenerative processes in amyotrophic lateral sclerosis (ALS), Parkinson's disease and Gaucher's disease. In this review, we provide an overview of the role of sphingolipids in the development and maintenance of the nervous system. We describe the implications of altered metabolism of sphingolipids in the pathophysiology of certain neurodegenerative diseases, with a primary focus on ALS. Finally, we provide an update of potential treatments that could be used to target the metabolism of sphingolipids in neurodegenerative diseases.
Collapse
Affiliation(s)
- Alexandra Bouscary
- Université de Strasbourg, UMR_S 1118, Fédération de Médecine Translationnelle, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France; INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France
| | - Cyril Quessada
- Université de Strasbourg, UMR_S 1118, Fédération de Médecine Translationnelle, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France; INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France
| | - Frédérique René
- Université de Strasbourg, UMR_S 1118, Fédération de Médecine Translationnelle, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France; INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France
| | - Michael Spedding
- Spedding Research Solutions SAS, 6 rue Ampere, 78650 Le Vesinet, France
| | - Bradley J Turner
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia
| | | | - Shyuan T Ngo
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Building 75, Cnr College Rd & Cooper Rd, Brisbane city, QLD 4072, Australia; Centre for Clinical Research, The University of Queensland, Building 71/918, Royal Brisbane & Women's Hospital Campus, Herston, QLD 4029, Australia; Queensland Brain Institute Building 79, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Jean-Philippe Loeffler
- Université de Strasbourg, UMR_S 1118, Fédération de Médecine Translationnelle, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France; INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France.
| |
Collapse
|
77
|
Straniero L, Rimoldi V, Melistaccio G, Di Fonzo A, Pezzoli G, Duga S, Asselta R. A rapid and low-cost test for screening the most common Parkinson's disease-related GBA variants. Parkinsonism Relat Disord 2020; 80:138-141. [PMID: 32987361 DOI: 10.1016/j.parkreldis.2020.09.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/10/2020] [Accepted: 09/21/2020] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Deleterious variants in the GBA gene confer a 2- to 20-fold increased risk of Parkinson's disease (PD) and are associated with a more severe disease course. The presence of a highly-similar pseudogene complicates genetic screening, both by Sanger and next-generation sequencing (NGS). Among PD-associated GBA variants, four missense substitutions (p.L444P, p.N370S, p.T369M, p.E326K) account for the majority of cases. Here, we aimed at developing an allele-specific PCR (AS-PCR) able to concomitantly detect the most common PD-related GBA variants. METHODS A multiplex PCR assay was designed using allele-specific oligonucleotides that distinguish the gene from the pseudogene and can exclusively amplify the variant alleles. Primer sequences and molarity, and thermal profiles were empirically optimized. The assay was validated on 4016 DNAs extracted by standard salting-out and previously analyzed by whole-exome sequencing (WES) followed by Sanger validation. RESULTS AS-PCRs performed on 4016 DNAs detected 103 variants; among them, 97 were true positives and 6 false positives. When comparing the results with the original WES data, for the "difficult" p.L444P, the number of false positives was 2/9 and 18/24 for multiplex-AS-PCR and WES, respectively. As we could have missed some p.L444P alleles by NGS, we verified the test performance on 50 DNAs from Sanger-validated p.L444P heterozygotes. All samples tested correctly. CONCLUSION We set up and validated a rapid and inexpensive test for screening large cohorts of individuals for variants conferring a significant PD risk. This screening method is particularly interesting to identify patients who could benefit most from early access to personalized therapies.
Collapse
Affiliation(s)
- Letizia Straniero
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy
| | - Valeria Rimoldi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy
| | - Giada Melistaccio
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy
| | - Alessio Di Fonzo
- IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Gianni Pezzoli
- Fondazione Grigioni per Il Morbo di Parkinson, Milan, Italy; Parkinson Institute, ASST 'Gaetano Pini-CTO', Milan, Italy
| | - Stefano Duga
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy; Humanitas Clinical and Research Center, IRCCS, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy; Humanitas Clinical and Research Center, IRCCS, Via Manzoni 56, 20089, Rozzano, Milan, Italy.
| |
Collapse
|
78
|
Fouka M, Mavroeidi P, Tsaka G, Xilouri M. In Search of Effective Treatments Targeting α-Synuclein Toxicity in Synucleinopathies: Pros and Cons. Front Cell Dev Biol 2020; 8:559791. [PMID: 33015057 PMCID: PMC7500083 DOI: 10.3389/fcell.2020.559791] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/14/2020] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease (PD), multiple system atrophy (MSA) and Dementia with Lewy bodies (DLB) represent pathologically similar, progressive neurodegenerative disorders characterized by the pathological aggregation of the neuronal protein α-synuclein. PD and DLB are characterized by the abnormal accumulation and aggregation of α-synuclein in proteinaceous inclusions within neurons named Lewy bodies (LBs) and Lewy neurites (LNs), whereas in MSA α-synuclein inclusions are mainly detected within oligodendrocytes named glial cytoplasmic inclusions (GCIs). The presence of pathologically aggregated α-synuclein along with components of the protein degradation machinery, such as ubiquitin and p62, in LBs and GCIs is considered to underlie the pathogenic cascade that eventually leads to the severe neurodegeneration and neuroinflammation that characterizes these diseases. Importantly, α-synuclein is proposed to undergo pathogenic misfolding and oligomerization into higher-order structures, revealing self-templating conformations, and to exert the ability of "prion-like" spreading between cells. Therefore, the manner in which the protein is produced, is modified within neural cells and is degraded, represents a major focus of current research efforts in the field. Given that α-synuclein protein load is critical to disease pathogenesis, the identification of means to limit intracellular protein burden and halt α-synuclein propagation represents an obvious therapeutic approach in synucleinopathies. However, up to date the development of effective therapeutic strategies to prevent degeneration in synucleinopathies is limited, due to the lack of knowledge regarding the precise mechanisms underlying the observed pathology. This review critically summarizes the recent developed strategies to counteract α-synuclein toxicity, including those aimed to increase protein degradation, to prevent protein aggregation and cell-to-cell propagation, or to engage antibodies against α-synuclein and discuss open questions and unknowns for future therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | - Maria Xilouri
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
79
|
Menozzi E, Schapira AHV. Enhancing the Activity of Glucocerebrosidase as a Treatment for Parkinson Disease. CNS Drugs 2020; 34:915-923. [PMID: 32607746 DOI: 10.1007/s40263-020-00746-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mutations in the glucocerebrosidase (GBA1) gene are the most common genetic risk factor for Parkinson disease (PD). Homozygous or compound heterozygous GBA1 mutations cause the lysosomal storage disorder Gaucher disease (GD), characterized by deficient activity of the glucocerebrosidase enzyme (GCase). Both individuals with GD type I and heterozygous carriers of pathogenic variants of GBA1 have an increased risk of developing PD, by approximately ten- to 20-fold compared to non-carriers. GCase activity is also reduced in PD patients without GBA1 mutations, suggesting that the GCase lysosomal pathway might be involved in PD pathogenesis. Available evidence indicates that GCase can affect α-synuclein pathology in different ways. Misfolded GCase proteins are retained in the endoplasmic reticulum, altering the lysosomal trafficking of the enzyme and disrupting protein trafficking. Also, deficient GCase leads to accumulation of substrates that in turn may bind α-synuclein and promote pathological formation of aggregates. Furthermore, α-synuclein itself can lower the enzymatic activity of GCase, indicating that a bidirectional interaction exists between GCase and α-synuclein. Targeted therapies aimed at enhancing GCase activity, augmenting the trafficking of misfolded GCase proteins by small molecule chaperones, or reducing substrate accumulation, have been tested in preclinical and clinical trials. This article reviews the molecular mechanisms linking GCase to α-synuclein and discusses the therapeutic drugs that by targeting the GCase pathway can influence PD progression.
Collapse
Affiliation(s)
- Elisa Menozzi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.
| |
Collapse
|
80
|
Migdalska‐Richards A, Wegrzynowicz M, Harrison IF, Verona G, Bellotti V, Spillantini MG, Schapira AHV. L444P Gba1 mutation increases formation and spread of α-synuclein deposits in mice injected with mouse α-synuclein pre-formed fibrils. PLoS One 2020; 15:e0238075. [PMID: 32833982 PMCID: PMC7444808 DOI: 10.1371/journal.pone.0238075] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/07/2020] [Indexed: 11/28/2022] Open
Abstract
Parkinson disease is the most common neurodegenerative movement disorder, estimated to affect one in twenty-five individuals over the age of 80. Mutations in glucocerebrosidase 1 (GBA1) represent the most common genetic risk factor for Parkinson disease. The link between GBA1 mutations and α-synuclein accumulation, a hallmark of Parkinson disease, is not fully understood. Following our recent finding that Gba1 mutations lead to increased α-synuclein accumulation in mice, we have studied the effects of a single injection of mouse α-synuclein pre-formed fibrils into the striatum of Gba1 mice that carry a L444P knock-in mutation. We found significantly greater formation and spread of α-synuclein inclusions in Gba1-transgenic mice compared to wild-type controls. This indicates that the Gba1 L444P mutation accelerates α-synuclein pathology and spread.
Collapse
Affiliation(s)
- Anna Migdalska‐Richards
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London, United Kingdom
- Complex Disease Epigenetics Group, University of Exeter, Royal Devon & Exeter Hospital, Exeter, United Kingdom
- * E-mail: (AHVS); (AMR)
| | - Michal Wegrzynowicz
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
- Laboratory of Molecular Basis of Neurodegeneration, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - Ian F. Harrison
- Centre for Advanced Biomedical Imaging, University College London, London, United Kingdom
| | - Guglielmo Verona
- Centre for Amyloidosis and Acute Phase Proteins, Faculty of Medical Sciences, University College London, London, United Kingdom
| | - Vittorio Bellotti
- Centre for Amyloidosis and Acute Phase Proteins, Faculty of Medical Sciences, University College London, London, United Kingdom
| | | | - Anthony H. V. Schapira
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London, United Kingdom
- * E-mail: (AHVS); (AMR)
| |
Collapse
|
81
|
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease typified by a movement disorder consisting of bradykinesia, rest tremor, rigidity, and postural instability. Treatment options for PD are limited, with most of the current approaches based on restoration of dopaminergic tone in the striatum. However, these do not alter disease course and do not treat the non-dopamine-dependent features of PD such as freezing of gait, cognitive impairment, and other non-motor features of the disorder, which often have the greatest impact on quality of life. As understanding of PD pathogenesis grows, novel therapeutic avenues are emerging. These include treatments that aim to control the symptoms of PD without the problematic side effects seen with currently available treatments and those that are aimed towards slowing pathology, reducing neuronal loss, and attenuating disease course. In this latter regard, there has been much interest in drug repurposing (the use of established drugs for a new indication), with many drugs being reported to affect PD-relevant intracellular processes. This approach offers an expedited route to the clinic, given that pharmacokinetic and safety data are potentially already available. In terms of better symptomatic therapies that are also regenerative, gene therapies and cell-based treatments are beginning to enter clinical trials, and developments in other neurosurgical strategies such as more nuanced deep brain stimulation approaches mean that the landscape of PD treatment is likely to evolve considerably over the coming years. In this review, we provide an overview of the novel therapeutic approaches that are close to, or are already in, clinical trials.
Collapse
Affiliation(s)
- Thomas B Stoker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK
- Department of Neurology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Roger A Barker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK
- Wellcome Trust – Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
82
|
Bouscary A, Quessada C, René F, Spedding M, Henriques A, Ngo S, Loeffler JP. Drug repositioning in neurodegeneration: An overview of the use of ambroxol in neurodegenerative diseases. Eur J Pharmacol 2020; 884:173446. [PMID: 32739173 DOI: 10.1016/j.ejphar.2020.173446] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/30/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common motor neuron disease in adults. While it is primarily characterized by the death of upper and lower motor neurons, there is a significant metabolic component involved in the progression of the disease. Two-thirds of ALS patients have metabolic alterations that are associated with the severity of symptoms. In ALS, as in other neurodegenerative diseases, the metabolism of glycosphingolipids, a class of complex lipids, is strongly dysregulated. We therefore assume that this pathway constitutes an interesting avenue for therapeutic approaches. We have shown that the glucosylceramide degrading enzyme, glucocerebrosidase (GBA) 2 is abnormally increased in the spinal cord of the SOD1G86R mouse model of ALS. Ambroxol, a chaperone molecule that inhibits GBA2, has been shown to have beneficial effects by slowing the development of the disease in SOD1G86R mice. Currently used in clinical trials for Parkinson's and Gaucher disease, ambroxol could be considered as a promising therapeutic treatment for ALS.
Collapse
Affiliation(s)
- Alexandra Bouscary
- INSERM U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, Université de Strasbourg, France; Université de Strasbourg, UMR-S 1118, Fédération de Médecine Translationnelle, Strasbourg, France
| | - Cyril Quessada
- INSERM U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, Université de Strasbourg, France; Université de Strasbourg, UMR-S 1118, Fédération de Médecine Translationnelle, Strasbourg, France
| | - Frédérique René
- INSERM U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, Université de Strasbourg, France; Université de Strasbourg, UMR-S 1118, Fédération de Médecine Translationnelle, Strasbourg, France
| | | | | | - Shyuan Ngo
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia; Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Jean-Philippe Loeffler
- INSERM U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, Université de Strasbourg, France; Université de Strasbourg, UMR-S 1118, Fédération de Médecine Translationnelle, Strasbourg, France.
| |
Collapse
|
83
|
Mullin S, Smith L, Lee K, D'Souza G, Woodgate P, Elflein J, Hällqvist J, Toffoli M, Streeter A, Hosking J, Heywood WE, Khengar R, Campbell P, Hehir J, Cable S, Mills K, Zetterberg H, Limousin P, Libri V, Foltynie T, Schapira AHV. Ambroxol for the Treatment of Patients With Parkinson Disease With and Without Glucocerebrosidase Gene Mutations: A Nonrandomized, Noncontrolled Trial. JAMA Neurol 2020; 77:427-434. [PMID: 31930374 PMCID: PMC6990847 DOI: 10.1001/jamaneurol.2019.4611] [Citation(s) in RCA: 208] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Question Does ambroxol cross the blood-brain barrier, and what are the biochemical changes associated with ambroxol therapy in patients with Parkinson disease with and without glucocerebrosidase gene mutations? Findings In this open-label clinical trial of 17 patients with Parkinson disease, ambroxol crossed the blood-brain barrier and bound to the β-glucocerebrosidase enzyme, and it increased β-glucocerebrosidase enzyme protein levels and cerebrospinal fluid α-synuclein levels in patients both with and without glucocerebrosidase gene mutations. Meaning Ambroxol therapy has potential for study as a neuroprotective compound for the treatment of patients with Parkinson disease both with and without glucocerebrosidase gene mutations. Importance Mutations of the glucocerebrosidase gene, GBA1 (OMIM 606463), are the most important risk factor for Parkinson disease (PD). In vitro and in vivo studies have reported that ambroxol increases β-glucocerebrosidase (GCase) enzyme activity and reduces α-synuclein levels. These observations support a potential role for ambroxol therapy in modifying a relevant pathogenetic pathway in PD. Objective To assess safety, tolerability, cerebrospinal fluid (CSF) penetration, and target engagement of ambroxol therapy with GCase in patients with PD with and without GBA1 mutations. Interventions An escalating dose of oral ambroxol to 1.26 g per day. Design, Setting, and Participants This single-center open-label noncontrolled clinical trial was conducted between January 11, 2017, and April 25, 2018, at the Leonard Wolfson Experimental Neuroscience Centre, a dedicated clinical research facility and part of the University College London Queen Square Institute of Neurology in London, United Kingdom. Participants were recruited from established databases at the Royal Free London Hospital and National Hospital for Neurology and Neurosurgery in London. Twenty-four patients with moderate PD were evaluated for eligibility, and 23 entered the study. Of those, 18 patients completed the study; 1 patient was excluded (failed lumbar puncture), and 4 patients withdrew (predominantly lumbar puncture–related complications). All data analyses were performed from November 1 to December 14, 2018. Main Outcomes and Measures Primary outcomes at 186 days were the detection of ambroxol in the CSF and a change in CSF GCase activity. Results Of the 18 participants (15 men [83.3%]; mean [SD] age, 60.2 [9.7] years) who completed the study, 17 (8 with GBA1 mutations and 9 without GBA1 mutations) were included in the primary analysis. Between days 0 and 186, a 156-ng/mL increase in the level of ambroxol in CSF (lower 95% confidence limit, 129 ng/mL; P < .001) was observed. The CSF GCase activity decreased by 19% (0.059 nmol/mL per hour; 95% CI, –0.115 to –0.002; P = .04). The ambroxol therapy was well tolerated, with no serious adverse events. An increase of 50 pg/mL (13%) in the CSF α-synuclein concentration (95% CI, 14-87; P = .01) and an increase of 88 ng/mol (35%) in the CSF GCase protein levels (95% CI, 40-137; P = .002) were observed. Mean (SD) scores on part 3 of the Movement Disorders Society Unified Parkinson Disease Rating Scale decreased (ie, improved) by 6.8 (7.1) points (95% CI, –10.4 to –3.1; P = .001). These changes were observed in patients with and without GBA1 mutations. Conclusions and Relevance The study results suggest that ambroxol therapy was safe and well tolerated; CSF penetration and target engagement of ambroxol were achieved, and CSF α-synuclein levels were increased. Placebo-controlled clinical trials are needed to examine whether ambroxol therapy is associated with changes in the natural progression of PD. Trial Registration ClinicalTrials.gov identifier: NCT02941822; EudraCT identifier: 2015-002571-24
Collapse
Affiliation(s)
- Stephen Mullin
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, London, United Kingdom.,Institute of Translational and Stratified Medicine, University of Plymouth School of Medicine, Plymouth, United Kingdom
| | - Laura Smith
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, London, United Kingdom
| | - Katherine Lee
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, London, United Kingdom
| | - Gayle D'Souza
- NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Philip Woodgate
- NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Josh Elflein
- NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Jenny Hällqvist
- Translational Mass Spectrometry Research Group, University College London Institute of Child Health, London, United Kingdom
| | - Marco Toffoli
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, London, United Kingdom
| | - Adam Streeter
- Department of Medical Statistics, University of Plymouth School of Medicine, Plymouth, United Kingdom
| | - Joanne Hosking
- Department of Medical Statistics, University of Plymouth School of Medicine, Plymouth, United Kingdom
| | - Wendy E Heywood
- Translational Mass Spectrometry Research Group, University College London Institute of Child Health, London, United Kingdom
| | - Rajeshree Khengar
- NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Philip Campbell
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, London, United Kingdom
| | - Jason Hehir
- Neurogenetics Unit, National Hospital for Neurology and Neurosurgery, UCLH NHS Foundation Trust, London, United Kingdom
| | - Sarah Cable
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, London, United Kingdom
| | - Kevin Mills
- Translational Mass Spectrometry Research Group, University College London Institute of Child Health, London, United Kingdom
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, University College London Institute of Neurology, London, United Kingdom.,UK Dementia Research Institute at University College London, London, United Kingdom.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Molndal, Sweden.,Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Molndal, Sweden
| | - Patricia Limousin
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, London, United Kingdom
| | - Vincenzo Libri
- NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Tom Foltynie
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, London, United Kingdom
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, London, United Kingdom
| |
Collapse
|
84
|
Polissidis A, Petropoulou-Vathi L, Nakos-Bimpos M, Rideout HJ. The Future of Targeted Gene-Based Treatment Strategies and Biomarkers in Parkinson's Disease. Biomolecules 2020; 10:E912. [PMID: 32560161 PMCID: PMC7355671 DOI: 10.3390/biom10060912] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 12/13/2022] Open
Abstract
Biomarkers and disease-modifying therapies are both urgent unmet medical needs in the treatment of Parkinson's disease (PD) and must be developed concurrently because of their interdependent relationship: biomarkers for the early detection of disease (i.e., prior to overt neurodegeneration) are necessary in order for patients to receive maximal therapeutic benefit and vice versa; disease-modifying therapies must become available for patients whose potential for disease diagnosis and prognosis can be predicted with biomarkers. This review provides an overview of the milestones achieved to date in the therapeutic strategy development of disease-modifying therapies and biomarkers for PD, with a focus on the most common and advanced genetically linked targets alpha-synuclein (SNCA), leucine-rich repeat kinase-2 (LRRK2) and glucocerebrosidase (GBA1). Furthermore, we discuss the convergence of the different pathways and the importance of patient stratification and how these advances may apply more broadly to idiopathic PD. The heterogeneity of PD poses a challenge for therapeutic and biomarker development, however, the one gene- one target approach has brought us closer than ever before to an unprecedented number of clinical trials and biomarker advancements.
Collapse
Affiliation(s)
| | | | | | - Hardy J. Rideout
- Laboratory of Neurodegenerative Diseases, Centre for Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (A.P.); (L.P.-V.); (M.N.-B.)
| |
Collapse
|
85
|
Yañez MJ, Marín T, Balboa E, Klein AD, Alvarez AR, Zanlungo S. Finding pathogenic commonalities between Niemann-Pick type C and other lysosomal storage disorders: Opportunities for shared therapeutic interventions. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165875. [PMID: 32522631 DOI: 10.1016/j.bbadis.2020.165875] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/06/2020] [Accepted: 06/03/2020] [Indexed: 12/12/2022]
Abstract
Lysosomal storage disorders (LSDs) are diseases characterized by the accumulation of macromolecules in the late endocytic system and are caused by inherited defects in genes that encode mainly lysosomal enzymes or transmembrane lysosomal proteins. Niemann-Pick type C disease (NPCD), a LSD characterized by liver damage and progressive neurodegeneration that leads to early death, is caused by mutations in the genes encoding the NPC1 or NPC2 proteins. Both proteins are involved in the transport of cholesterol from the late endosomal compartment to the rest of the cell. Loss of function of these proteins causes primary cholesterol accumulation, and secondary accumulation of other lipids, such as sphingolipids, in lysosomes. Despite years of studying the genetic and molecular bases of NPCD and related-lysosomal disorders, the pathogenic mechanisms involved in these diseases are not fully understood. In this review we will summarize the pathogenic mechanisms described for NPCD and we will discuss their relevance for other LSDs with neurological components such as Niemann- Pick type A and Gaucher diseases. We will particularly focus on the activation of signaling pathways that may be common to these three pathologies with emphasis on how the intra-lysosomal accumulation of lipids leads to pathology, specifically to neurological impairments. We will show that although the primary lipid storage defect is different in these three LSDs, there is a similar secondary accumulation of metabolites and activation of signaling pathways that can lead to common pathogenic mechanisms. This analysis might help to delineate common pathological mechanisms and therapeutic targets for lysosomal storage diseases.
Collapse
Affiliation(s)
- M J Yañez
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - T Marín
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - E Balboa
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - A D Klein
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - A R Alvarez
- Laboratory of Cell Signaling, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago, Chile; CARE UC, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - S Zanlungo
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
86
|
Han TU, Sam R, Sidransky E. Small Molecule Chaperones for the Treatment of Gaucher Disease and GBA1-Associated Parkinson Disease. Front Cell Dev Biol 2020; 8:271. [PMID: 32509770 PMCID: PMC7248408 DOI: 10.3389/fcell.2020.00271] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 03/30/2020] [Indexed: 12/15/2022] Open
Abstract
Parkinson disease, the second most common movement disorder, is a complex neurodegenerative disorder hallmarked by the accumulation of alpha-synuclein, a neural-specific small protein associated with neuronal synapses. Mutations in the glucocerebrosidase gene (GBA1), implicated in the rare, autosomal recessive lysosomal disorder Gaucher disease, are the most common known genetic risk factor for Parkinson disease. Insights into the inverse relationship between glucocerebrosidase and alpha-synuclein have led to new therapeutic approaches for the treatment of Gaucher disease and GBA1-associated Parkinson disease. Unlike the current drugs used to treat Gaucher disease, which are highly expensive and do not cross the blood-brain-barrier, new small molecules therapies, including competitive and non-competitive chaperones that enhance glucocerebrosidase levels are being developed to overcome these limitations. Some of these include iminosugars, ambroxol, other competitive glucocerebrosidase inhibitors, and non-inhibitory chaperones or activators that do not compete for the active site. These drugs, which have been shown in different disease models to increase glucocerebrosidase activity, could have potential as a therapy for Gaucher disease and GBA1- associated Parkinson disease. Some have been demonstrated to reduce α-synuclein levels in pre-clinical studies using cell-based or animal models of GBA1-associated Parkinson disease, and may also have utility for idiopathic Parkinson disease.
Collapse
Affiliation(s)
- Tae-Un Han
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Richard Sam
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ellen Sidransky
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
87
|
Mohamed NV, Larroquette F, Beitel LK, Fon EA, Durcan TM. One Step Into the Future: New iPSC Tools to Advance Research in Parkinson's Disease and Neurological Disorders. JOURNAL OF PARKINSONS DISEASE 2020; 9:265-281. [PMID: 30741685 PMCID: PMC6597965 DOI: 10.3233/jpd-181515] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Studying Parkinson’s disease (PD) in the laboratory presents many challenges, the main one being the limited availability of human cells and tissue from affected individuals. As PD is characterized by a loss of dopaminergic (DA) neurons in the brain, it is nearly impossible for researchers to access and extract these cells from living patients. Thus, in the past PD research has focused on the use of patients’ post-mortem tissues, animal models, or immortalized cell lines to dissect cellular pathways of interest. While these strategies deepened our knowledge of pathological mechanisms in PD, they failed to faithfully capture key mechanisms at play in the human brain. The emergence of induced pluripotent stem cell (iPSC) technology is revolutionizing PD research, as it allows for the differentiation and growth of human DA neurons in vitro, holding immense potential not only for modelling PD, but also for identifying novel therapies. However, to reproduce the complexity of the brain’s environment, researchers are recognizing the need to further develop and refine iPSC-based tools. In this review, we provide an overview of different systems now available for the study of PD, with a particular emphasis on the potential and limitations of iPSC as research tools to generate more relevant models of PD pathophysiology and advance the drug discovery process.
Collapse
Affiliation(s)
- Nguyen-Vi Mohamed
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Frédérique Larroquette
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Lenore K Beitel
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Edward A Fon
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Thomas M Durcan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
88
|
Poewe W, Seppi K, Marini K, Mahlknecht P. New hopes for disease modification in Parkinson's Disease. Neuropharmacology 2020; 171:108085. [PMID: 32298705 DOI: 10.1016/j.neuropharm.2020.108085] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 03/31/2020] [Indexed: 12/11/2022]
Abstract
To date, despite numerous clinical trials, no intervention has been demonstrated to modify the progression of Parkinson's disease (PD). However, over the past decades encouraging progress has been made towards a better understanding of molecular pathways relevant for the neurodegenerative process in PD. This is also based on new insights into the genetic architecture of the disease, revealing multiple novel targets for potentially disease-modifying interventions. Important achievements have also been made in the field of risk markers and combinations thereof, in the form of risk algorithms, will hopefully soon provide the possibility to identify affected individuals at yet prediagnostic or prodromal stages of the illness. Such phases of the disease would provide an ideal window for neuroprotection trials. Taken together, these developments offer hope that a breakthrough towards modifying the course of PD might be reached. In this article we summarize various approaches currently pursued in this quest. This article is part of the special issue entitled 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'.
Collapse
Affiliation(s)
- Werner Poewe
- Department of Neurology, Medical University Innsbruck, Austria.
| | - Klaus Seppi
- Department of Neurology, Medical University Innsbruck, Austria
| | - Kathrin Marini
- Department of Neurology, Medical University Innsbruck, Austria
| | | |
Collapse
|
89
|
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: 4.0] [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
|
90
|
Elmatboly AM, Sherif AM, Deeb DA, Benmelouka A, Bin-Jumah MN, Aleya L, Abdel-Daim MM. The impact of proteostasis dysfunction secondary to environmental and genetic causes on neurodegenerative diseases progression and potential therapeutic intervention. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:11461-11483. [PMID: 32072427 DOI: 10.1007/s11356-020-07914-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 01/28/2020] [Indexed: 06/10/2023]
Abstract
Aggregation of particular proteins in the form of inclusion bodies or plaques followed by neuronal death is a hallmark of neurodegenerative proteopathies such as primary Parkinsonism, Alzheimer's disease, Lou Gehrig's disease, and Huntington's chorea. Complex polygenic and environmental factors implicated in these proteopathies. Accumulation of proteins in these disorders indicates a substantial disruption in protein homeostasis (proteostasis). Proteostasis or cellular proteome homeostasis is attained by the synchronization of a group of cellular mechanisms called the proteostasis network (PN), which is responsible for the stability of the proteome and achieves the equilibrium between synthesis, folding, and degradation of proteins. In this review, we will discuss the different types of PN and the impact of PN component dysfunction on the four major neurodegenerative diseases mentioned earlier. Graphical abstract.
Collapse
Affiliation(s)
| | - Ahmed M Sherif
- Faculty of Medicine, Zagazig University, El-Sharkia, Egypt
| | - Dalia A Deeb
- Faculty of Medicine, Zagazig University, El-Sharkia, Egypt
| | - Amira Benmelouka
- Faculty of Medicine, University of Algiers, Sidi M'Hamed, Algeria
| | - May N Bin-Jumah
- Biology Department, College Of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Besançon Cedex, France
| | - Mohamed M Abdel-Daim
- Department of Zoology, Science College, King Saud University, Riyadh, 11451, Saudi Arabia.
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt.
| |
Collapse
|
91
|
Ambroxol Improves Neuronal Survival and Reduces White Matter Damage through Suppressing Endoplasmic Reticulum Stress in Microglia after Intracerebral Hemorrhage. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8131286. [PMID: 32309438 PMCID: PMC7142346 DOI: 10.1155/2020/8131286] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/23/2020] [Accepted: 02/28/2020] [Indexed: 12/31/2022]
Abstract
Intracerebral hemorrhage (ICH) has been becoming a serious public health problem. Pneumonia, occurring in 43% of all ICH patients, is a common complication heavily influencing outcome and accounting for more than 1/3 of the overall mortality in patients with ICH. Ambroxol may be an effective additional treatment for ICH patients with pneumonia. But its effect and potential mechanism on functional recovery post-ICH still remain elusive. In the present study, the results indicated that 35 mg/kg and 70 mg/kg ambroxol facilitated neuronal survival and reduced white matter fiber bundle damage due to mitigating microglial activation and reducing proinflammatory cytokine accumulation in mice with ICH. The possible mechanism might be due to suppressing endoplasmic reticulum stress involving the IRE1α/TRAF2 signaling pathway, which paves a new path for the treatment of ICH and opens a new window for the use of ambroxol in clinical practice.
Collapse
|
92
|
Teil M, Arotcarena ML, Faggiani E, Laferriere F, Bezard E, Dehay B. Targeting α-synuclein for PD Therapeutics: A Pursuit on All Fronts. Biomolecules 2020; 10:biom10030391. [PMID: 32138193 PMCID: PMC7175302 DOI: 10.3390/biom10030391] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/26/2020] [Accepted: 02/29/2020] [Indexed: 12/15/2022] Open
Abstract
Parkinson's Disease (PD) is characterized both by the loss of dopaminergic neurons in the substantia nigra and the presence of cytoplasmic inclusions called Lewy Bodies. These Lewy Bodies contain the aggregated α-synuclein (α-syn) protein, which has been shown to be able to propagate from cell to cell and throughout different regions in the brain. Due to its central role in the pathology and the lack of a curative treatment for PD, an increasing number of studies have aimed at targeting this protein for therapeutics. Here, we reviewed and discussed the many different approaches that have been studied to inhibit α-syn accumulation via direct and indirect targeting. These analyses have led to the generation of multiple clinical trials that are either completed or currently active. These clinical trials and the current preclinical studies must still face obstacles ahead, but give hope of finding a therapy for PD with time.
Collapse
Affiliation(s)
- Margaux Teil
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Marie-Laure Arotcarena
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Emilie Faggiani
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Florent Laferriere
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Erwan Bezard
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Benjamin Dehay
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- Correspondence:
| |
Collapse
|
93
|
Toffoli M, Smith L, Schapira AHV. The biochemical basis of interactions between Glucocerebrosidase and alpha-synuclein in GBA1 mutation carriers. J Neurochem 2020; 154:11-24. [PMID: 31965564 DOI: 10.1111/jnc.14968] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 12/11/2022]
Abstract
The discovery of genes involved in familial as well as sporadic forms of Parkinson disease (PD) constitutes an important milestone in understanding this disorder's pathophysiology and potential treatment. Among these genes, GBA1 is one of the most common and well-studied, but it is still unclear how mutations in GBA1 translate into an increased risk for developing PD. In this review, we provide an overview of the biochemical and structural relationship between GBA1 and PD to help understand the recent advances in the development of PD therapies intended to target this pathway.
Collapse
Affiliation(s)
- Marco Toffoli
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, UK
| | - Laura Smith
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, UK
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, UK
| |
Collapse
|
94
|
Stott SRW, Wyse RK, Brundin P. Novel approaches to counter protein aggregation pathology in Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2020; 252:451-492. [PMID: 32247372 PMCID: PMC10019778 DOI: 10.1016/bs.pbr.2019.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The primary neuropathological characteristics of the Parkinsonian brain are the loss of nigral dopamine neurons and the aggregation of alpha synuclein protein. Efforts to development potentially disease-modifying treatments have largely focused on correcting these aspects of the condition. In the last decade treatments targeting protein aggregation have entered the clinical pipeline. In this chapter we provide an overview of ongoing clinical trial programs for different therapies attempting to reduce protein aggregation pathology in Parkinson's disease. We will also briefly consider various novel approaches being proposed-and being developed preclinically-to inhibit/reduce aggregated protein pathology in Parkinson's.
Collapse
Affiliation(s)
| | | | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States.
| |
Collapse
|
95
|
Bellomo G, Paciotti S, Gatticchi L, Parnetti L. The Vicious Cycle Between
α
‐Synuclein Aggregation and Autophagic‐Lysosomal Dysfunction. Mov Disord 2019; 35:34-44. [DOI: 10.1002/mds.27895] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/31/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022] Open
Affiliation(s)
- Giovanni Bellomo
- Magnetic Resonance Center (CERM) University of Florence Sesto Fiorentino (FI) Italy
| | - Silvia Paciotti
- Laboratory of Clinical Neurochemistry, Section of Neurology University of Perugia Perugia (PG) Italy
- Department of Experimental Medicine University of Perugia Perugia (PG) Italy
| | - Leonardo Gatticchi
- Department of Experimental Medicine University of Perugia Perugia (PG) Italy
| | - Lucilla Parnetti
- Laboratory of Clinical Neurochemistry, Section of Neurology University of Perugia Perugia (PG) Italy
| |
Collapse
|
96
|
Schöndorf DC, Ivanyuk D, Baden P, Sanchez-Martinez A, De Cicco S, Yu C, Giunta I, Schwarz LK, Di Napoli G, Panagiotakopoulou V, Nestel S, Keatinge M, Pruszak J, Bandmann O, Heimrich B, Gasser T, Whitworth AJ, Deleidi M. The NAD+ Precursor Nicotinamide Riboside Rescues Mitochondrial Defects and Neuronal Loss in iPSC and Fly Models of Parkinson's Disease. Cell Rep 2019; 23:2976-2988. [PMID: 29874584 DOI: 10.1016/j.celrep.2018.05.009] [Citation(s) in RCA: 225] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 03/05/2018] [Accepted: 05/02/2018] [Indexed: 11/30/2022] Open
Abstract
While mitochondrial dysfunction is emerging as key in Parkinson's disease (PD), a central question remains whether mitochondria are actual disease drivers and whether boosting mitochondrial biogenesis and function ameliorates pathology. We address these questions using patient-derived induced pluripotent stem cells and Drosophila models of GBA-related PD (GBA-PD), the most common PD genetic risk. Patient neurons display stress responses, mitochondrial demise, and changes in NAD+ metabolism. NAD+ precursors have been proposed to ameliorate age-related metabolic decline and disease. We report that increasing NAD+ via the NAD+ precursor nicotinamide riboside (NR) significantly ameliorates mitochondrial function in patient neurons. Human neurons require nicotinamide phosphoribosyltransferase (NAMPT) to maintain the NAD+ pool and utilize NRK1 to synthesize NAD+ from NAD+ precursors. Remarkably, NR prevents the age-related dopaminergic neuronal loss and motor decline in fly models of GBA-PD. Our findings suggest NR as a viable clinical avenue for neuroprotection in PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- David C Schöndorf
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Tübingen 72076, Germany; Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
| | - Dina Ivanyuk
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Tübingen 72076, Germany; Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
| | - Pascale Baden
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Tübingen 72076, Germany; Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
| | - Alvaro Sanchez-Martinez
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Silvia De Cicco
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Tübingen 72076, Germany; Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
| | - Cong Yu
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Tübingen 72076, Germany; Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
| | - Ivana Giunta
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Lukas K Schwarz
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Tübingen 72076, Germany; Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
| | - Gabriele Di Napoli
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Tübingen 72076, Germany; Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
| | - Vasiliki Panagiotakopoulou
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Tübingen 72076, Germany; Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
| | - Sigrun Nestel
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg 79104, Germany
| | - Marcus Keatinge
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Jan Pruszak
- Emmy Noether-Group for Stem Cell Biology, Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Biological Signaling Studies (BIOSS), University of Freiburg, Freiburg 79104, Germany
| | - Oliver Bandmann
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Bernd Heimrich
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg 79104, Germany
| | - Thomas Gasser
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Tübingen 72076, Germany; Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
| | - Alexander J Whitworth
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Michela Deleidi
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Tübingen 72076, Germany; Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany.
| |
Collapse
|
97
|
Cerri S, Blandini F. Role of Autophagy in Parkinson's Disease. Curr Med Chem 2019; 26:3702-3718. [PMID: 29484979 DOI: 10.2174/0929867325666180226094351] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 01/30/2018] [Accepted: 02/13/2018] [Indexed: 12/11/2022]
Abstract
Autophagy is an essential catabolic mechanism that delivers misfolded proteins and damaged organelles to the lysosome for degradation. Autophagy pathways include macroautophagy, chaperone-mediated autophagy and microautophagy, each involving different mechanisms of substrate delivery to lysosome. Defects of these pathways and the resulting accumulation of protein aggregates represent a common pathobiological feature of neurodegenerative disorders such as Alzheimer, Parkinson and Huntington disease. This review provides an overview of the role of autophagy in Parkinson's disease (PD) by summarizing the most relevant genetic and experimental evidence showing how this process can contribute to disease pathogenesis. Given lysosomes take part in the final step of the autophagic process, the role of lysosomal defects in the impairment of autophagy and their impact on disease will also be discussed. A glance on the role of non-neuronal autophagy in the pathogenesis of PD will be included. Moreover, we will examine novel pharmacological targets and therapeutic strategies that, by boosting autophagy, may be theoretically beneficial for PD. Special attention will be focused on natural products, such as phenolic compounds, that are receiving increasing consideration due to their potential efficacy associated with low toxicity. Although many efforts have been made to elucidate autophagic process, the development of new therapeutic interventions requires a deeper understanding of the mechanisms that may lead to autophagy defects in PD and should take into account the multifactorial nature of the disease as well as the phenotypic heterogeneity of PD patients.
Collapse
Affiliation(s)
- Silvia Cerri
- Laboratory of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, Pavia, Italy
| | - Fabio Blandini
- Laboratory of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
98
|
Neurorestorative effects of sub-chronic administration of ambroxol in rodent model of Parkinson’s disease. Naunyn Schmiedebergs Arch Pharmacol 2019; 393:429-444. [DOI: 10.1007/s00210-019-01737-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023]
|
99
|
Kim YM, Yum MS, Heo SH, Kim T, Jin HK, Bae JS, Seo GH, Oh A, Yoon HM, Lim HT, Kim HW, Ko TS, Lim HS, Osborn MJ, Tolar J, Cozma C, Rolfs A, Zimran A, Lee BH, Yoo HW. Pharmacologic properties of high-dose ambroxol in four patients with Gaucher disease and myoclonic epilepsy. J Med Genet 2019; 57:124-131. [PMID: 31649052 PMCID: PMC7029246 DOI: 10.1136/jmedgenet-2019-106132] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 08/27/2019] [Accepted: 09/14/2019] [Indexed: 11/05/2022]
Abstract
Background Ambroxol (ABX) has been suggested as an augmentative pharmacological agent for neuronopathic Gaucher disease (nGD). This study assessed the long-term safety and efficacy of combined therapy with high-dose ABX and enzyme replacement therapy (ERT) in nGD. Methods ABX+ERT therapy was administered for 4.5 years in four patients with nGD. ABX was initiated at a dose of 1.5 mg/kg/day, and the dose was escalated up to 27 mg/kg/day. The target plasma level was 10 µmol/L or less. The changes in glucocerebrosidase activity, biochemical, safety and neurocognitive findings were assessed. Results Enhanced residual GCcase activity was observed in all patients, as evidenced in both in vitro and in vivo studies. During the first 2 years of study with ABX (up to 21 mg/kg/day), mean seizure frequencies and neurocognitive function worsened. After ABX dosage was increased up to 27 mg/kg/day of ABX, its trough plasma concentration was 3.2–8.8 µmol/L. Drug-to-drug interaction, especially with antiepileptic drug significantly affected the pharmacokinetic parameters of ABX. Importantly, at 27 mg/kg/day of ABX, the seizure frequencies markedly decreased from the baseline, and the neurocognitive function was improved. In addition, Lyso-Gb1, a biomarker for the severity and progression of GD, was normalised in all patients. High-dose ABX was well-tolerated with no severe adverse events. Conclusions Long-term treatment with high-dose ABX+ERT was safe and might help to arrest the progression of the neurological manifestations in GD.
Collapse
Affiliation(s)
- Yoon-Myung Kim
- Department of Pediatrics, Gangneung Asan Hospital, Gangneung, The Republic of Korea
| | - Mi-Sun Yum
- Department of Pediatrics, Asan Medical Center Children's Hospital, College of Medicine University of Ulsan, Seoul, The Republic of Korea
| | - Sun Hee Heo
- Asan Institute for Life Sciences, Asan Medical Center, College of Medicine University of Ulsan, Seoul, The Republic of Korea
| | - Taeho Kim
- Asan Institute for Life Sciences, Asan Medical Center, College of Medicine University of Ulsan, Seoul, The Republic of Korea
| | - Hee Kyung Jin
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, The Republic of Korea
| | - Jae-Sung Bae
- Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, The Republic of Korea
| | - Go Hun Seo
- Department of Pediatrics, Asan Medical Center Children's Hospital, College of Medicine University of Ulsan, Seoul, The Republic of Korea
| | - Arum Oh
- Department of Pediatrics, Asan Medical Center Children's Hospital, College of Medicine University of Ulsan, Seoul, The Republic of Korea
| | - Hee Mang Yoon
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, College of Medicine University of Ulsan, Seoul, The Republic of Korea
| | - Hyun Taek Lim
- Department of Ophthalmology, Asan Medical Center, College of Medicine University of Ulsan, Seoul, The Republic of Korea
| | - Hyo-Won Kim
- Department of Psychiatry, Asan Medical Center, College of Medicine University of Ulsan, Seoul, The Republic of Korea
| | - Tae-Sung Ko
- Department of Pediatrics, Asan Medical Center Children's Hospital, College of Medicine University of Ulsan, Seoul, The Republic of Korea
| | - Hyeong-Seok Lim
- Department of Clinical Pharmacology and Therapeutics, Asan Medical Center, College of Medicine University of Ulsan, Seoul, The Republic of Korea
| | - Mark J Osborn
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Medical Center, Minneapolis, Minnesota, USA
| | - Jakub Tolar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Medical Center, Minneapolis, Minnesota, USA
| | | | - Arndt Rolfs
- Centogene AG, Rostock, Germany.,Albrecht-Kossel-Institute for Neuroregeneration, Medical University of Rostock, Rostock, Germany
| | - Ari Zimran
- The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Beom Hee Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, College of Medicine University of Ulsan, Seoul, The Republic of Korea .,Medical Genetics Center, Asan Medical Center, College of Medicine University of Ulsan, Seoul, The Republic of Korea
| | - Han-Wook Yoo
- Department of Pediatrics, Asan Medical Center Children's Hospital, College of Medicine University of Ulsan, Seoul, The Republic of Korea .,Medical Genetics Center, Asan Medical Center, College of Medicine University of Ulsan, Seoul, The Republic of Korea
| |
Collapse
|
100
|
Yang SY, Gegg M, Chau D, Schapira A. Glucocerebrosidase activity, cathepsin D and monomeric α-synuclein interactions in a stem cell derived neuronal model of a PD associated GBA1 mutation. Neurobiol Dis 2019; 134:104620. [PMID: 31634558 PMCID: PMC6983928 DOI: 10.1016/j.nbd.2019.104620] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/13/2019] [Accepted: 09/23/2019] [Indexed: 11/24/2022] Open
Abstract
The presence of GBA1 gene mutations increases risk for Parkinson's disease (PD), but the pathogenic mechanisms of GBA1 associated PD remain unknown. Given that impaired α-synuclein turnover is a hallmark of PD pathogenesis and cathepsin D is a key enzyme involved in α-synuclein degradation in neuronal cells, we have examined the relationship of glucocerebrosidase (GCase), cathepsin D and monomeric α-synuclein in human neural crest stem cell derived dopaminergic neurons. We found that normal activity of GCase is necessary for cathepsin D to perform its function of monomeric α-synuclein removal from neurons. GBA1 mutations lead to a lower level of cathepsin D protein and activity, and higher level of monomeric α-synuclein in neurons. When GBA1 mutant neurons were treated with GCase replacement or chaperone therapy; cathepsin D protein levels and activity were restored, and monomeric α-synuclein decreased. When cathepsin D was inhibited, GCase replacement failed to reduce monomeric α-synuclein levels in GBA1 mutant neurons. These data indicate that GBA1 gene mutations increase monomeric α-synuclein levels via an effect on lysosomal cathepsin D in neurons. Cathepsin D protein and activity decreased in GBA mutation-associated PD neurons. α-synuclein protein level increased in GBA mutation-associated PD neurons. GBA enzyme replacement treatment increased cathepsin D, decreased α-synuclein levels. GBA enzyme chaperone treatment increased cathepsin D, decreased α-synuclein levels. The influence of GBA enzyme replacement on α-synuclein mediated through cathepsin D.
Collapse
Affiliation(s)
- Shi-Yu Yang
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Matthew Gegg
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - David Chau
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Anthony Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.
| |
Collapse
|