1
|
Cardoso F, Schmidt P. Proposed biological definitions of Parkinson's disease confuse understanding without delivering meaningful advances. JOURNAL OF PARKINSON'S DISEASE 2025:1877718X241308482. [PMID: 39973501 DOI: 10.1177/1877718x241308482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
This article reflects on two proposals to classify and stage Parkinson's disease (PD) and related conditions based on the presence of aggregated alpha-synuclein (ASN) detected by alpha-synuclein aggregation assay (SAA). The authors highlight the significant shortcomings of the proposals: detection of ASN by SAA is not specific of PD or other well-established clinical conditions; they do not allow prediction of clinical course and prognosis; and they are not suitable as an endpoint for clinical trials. To move forward with proposals reflecting so many uncertainties and unknowns will just sow confusion and misunderstanding within the PD community.
Collapse
Affiliation(s)
- Francisco Cardoso
- Movement Disorders Unit, Neurology Service, Internal Medicine Department, The Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Peter Schmidt
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
- Rho, Inc., Durham, NC, USA
| |
Collapse
|
2
|
Menozzi E, Schapira AHV. Prospects for Disease Slowing in Parkinson Disease. Annu Rev Pharmacol Toxicol 2025; 65:237-258. [PMID: 39088860 DOI: 10.1146/annurev-pharmtox-022124-033653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
The increasing prevalence of Parkinson disease (PD) highlights the need to develop interventions aimed at slowing or halting its progression. As a result of sophisticated disease modeling in preclinical studies, and refinement of specific clinical/genetic/pathological profiles, our understanding of PD pathogenesis has grown over the years, leading to the identification of several targets for disease modification. This has translated to the development of targeted therapies, many of which have entered clinical trials. Nonetheless, up until now, none of these treatments have satisfactorily shown disease-modifying effects in PD. In this review, we present the most up-to-date disease-modifying pharmacological interventions in the clinical trial pipeline for PD. We focus on agents that have reached more advanced stages of clinical trials testing, highlighting both positive and negative results, and critically reflect on strengths, weaknesses, and challenges of current disease-modifying therapeutic avenues in PD.
Collapse
Affiliation(s)
- Elisa Menozzi
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom;
| | - Anthony H V Schapira
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom;
| |
Collapse
|
3
|
Orimo K, Mitsui J, Matsukawa T, Tanaka M, Nomoto J, Ishiura H, Omae Y, Kawai Y, Tokunaga K, Toda T, Tsuji S. Association study of GBA1 variants with MSA based on comprehensive sequence analysis -Pitfalls in short-read sequence analysis depending on the human reference genome. J Hum Genet 2024; 69:613-621. [PMID: 39020124 PMCID: PMC11599039 DOI: 10.1038/s10038-024-01266-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/22/2024] [Accepted: 06/07/2024] [Indexed: 07/19/2024]
Abstract
Multiple system atrophy (MSA) is a neurodegenerative disorder characterized by various combinations of autonomic failure, parkinsonism, and cerebellar ataxia. To elucidate variants associated with MSA, we have been conducting short-read-based whole-genome sequence analysis. In the process of the association studies, we initially focused on GBA1, a previously proposed susceptibility gene for MSA, to evaluate whether GBA1 variants can be efficiently identified despite its extraordinarily high homology with its pseudogene, GBA1LP. To accomplish this, we conducted a short-read whole-genome sequence analysis with alignment to GRCh38 as well as Sanger sequence analysis and compared the results. We identified five variants with inconsistencies between the two pipelines, of which three variants (p.L483P, p.A495P-p.V499V, p.L483_M489delinsW) were the results of misalignment due to minor alleles in GBA1P1 registered in GRCh38. The miscalling events in these variants were resolved by alignment to GRCh37 as the reference genome, where the major alleles are registered. In addition, a structural variant was not properly identified either by short-read or by Sanger sequence analyses. Having accomplished correct variant calling, we identified three variants pathogenic for Gaucher disease (p.S310G, p.L483P, and p.L483_M489delinsW). Of these variants, the allele frequency of p.L483P (0.003) in the MSA cases was higher than that (0.0011) in controls. The meta-analysis incorporating a previous report demonstrated a significant association of p.L483P with MSA with an odds ratio of 2.85 (95% CI; 1.05 - 7.76, p = 0.0400).
Collapse
Affiliation(s)
- Kenta Orimo
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Jun Mitsui
- Department of Precision Medicine Neurology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takashi Matsukawa
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Masaki Tanaka
- Institute of Medical Genomics, International University of Health and Welfare, 4-3, Kozunomori, Narita-shi, Chiba, 286-8686, Japan
| | - Junko Nomoto
- Institute of Medical Genomics, International University of Health and Welfare, 4-3, Kozunomori, Narita-shi, Chiba, 286-8686, Japan
| | - Hiroyuki Ishiura
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Yosuke Omae
- Genome Medical Science Project, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Yosuke Kawai
- Genome Medical Science Project, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Katsushi Tokunaga
- Genome Medical Science Project, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Tatsushi Toda
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Shoji Tsuji
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
- Institute of Medical Genomics, International University of Health and Welfare, 4-3, Kozunomori, Narita-shi, Chiba, 286-8686, Japan.
| |
Collapse
|
4
|
Vieira SR, Mezabrovschi R, Toffoli M, Del Pozo SL, Menozzi E, Mullin S, Yalkic S, Limbachiya N, Koletsi S, Loefflad N, Lopez GJ, Gan‐Or Z, Alcalay RN, Sidransky E, Schapira AH. Consensus Guidance for Genetic Counseling in GBA1 Variants: A Focus on Parkinson's Disease. Mov Disord 2024; 39:2144-2154. [PMID: 39258449 PMCID: PMC11657020 DOI: 10.1002/mds.30006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/16/2024] [Indexed: 09/12/2024] Open
Abstract
Glucocerebrosidase (GBA1) variants constitute numerically the most common known genetic risk factor for Parkinson's disease (PD) and are distributed worldwide. Access to GBA1 genotyping varies across the world and even regionally within countries. Guidelines for GBA1 variant counseling are evolving. We review the current knowledge of the link between GBA1 and PD, and discuss the practicalities of GBA1 testing. Lastly, we provide a consensus for an approach to counseling people with GBA1 variants, notably the communication of PD risk. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sophia R.L. Vieira
- Department of Clinical and Movement NeurosciencesUniversity College London Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Roxana Mezabrovschi
- Department of Clinical and Movement NeurosciencesUniversity College London Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Marco Toffoli
- Department of Clinical and Movement NeurosciencesUniversity College London Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Sara Lucas Del Pozo
- Department of Clinical and Movement NeurosciencesUniversity College London Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Elisa Menozzi
- Department of Clinical and Movement NeurosciencesUniversity College London Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Stephen Mullin
- Department of Clinical and Movement NeurosciencesUniversity College London Queen Square Institute of NeurologyLondonUnited Kingdom
- Faculty of HealthUniversity of PlymouthPlymouthUnited Kingdom
| | - Selen Yalkic
- Department of Clinical and Movement NeurosciencesUniversity College London Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Naomi Limbachiya
- Department of Clinical and Movement NeurosciencesUniversity College London Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Sofia Koletsi
- Department of Clinical and Movement NeurosciencesUniversity College London Queen Square Institute of NeurologyLondonUnited Kingdom
- Aligning Science Across Parkinson's Collaborative Research NetworkChevy ChaseMarylandUSA
| | - Nadine Loefflad
- Department of Clinical and Movement NeurosciencesUniversity College London Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Grisel J. Lopez
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Ziv Gan‐Or
- Department of Neurology and Neurosurgery, The Neuro (Montreal Neurological Institute‐Hospital), and Department of Human GeneticsMcGill UniversityMontrealQuebecCanada
| | - Roy N. Alcalay
- Columbia University Irving Medical CenterNew YorkNew YorkUSA
- Tel Aviv Sourasky Medical Center, Tel Aviv School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Ellen Sidransky
- Aligning Science Across Parkinson's Collaborative Research NetworkChevy ChaseMarylandUSA
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Anthony H.V. Schapira
- Department of Clinical and Movement NeurosciencesUniversity College London Queen Square Institute of NeurologyLondonUnited Kingdom
- Aligning Science Across Parkinson's Collaborative Research NetworkChevy ChaseMarylandUSA
| |
Collapse
|
5
|
Zhang X, Wu H, Tang B, Guo J. Clinical, mechanistic, biomarker, and therapeutic advances in GBA1-associated Parkinson's disease. Transl Neurodegener 2024; 13:48. [PMID: 39267121 PMCID: PMC11391654 DOI: 10.1186/s40035-024-00437-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 08/17/2024] [Indexed: 09/14/2024] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease. The development of PD is closely linked to genetic and environmental factors, with GBA1 variants being the most common genetic risk. Mutations in the GBA1 gene lead to reduced activity of the coded enzyme, glucocerebrosidase, which mediates the development of PD by affecting lipid metabolism (especially sphingolipids), lysosomal autophagy, endoplasmic reticulum, as well as mitochondrial and other cellular functions. Clinically, PD with GBA1 mutations (GBA1-PD) is characterized by particular features regarding the progression of symptom severity. On the therapeutic side, the discovery of the relationship between GBA1 variants and PD offers an opportunity for targeted therapeutic interventions. In this review, we explore the genotypic and phenotypic correlations, etiologic mechanisms, biomarkers, and therapeutic approaches of GBA1-PD and summarize the current state of research and its challenges.
Collapse
Affiliation(s)
- Xuxiang Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Heng Wu
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, University of South China, Hengyang, 421001, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province, Hengyang, 421001, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, University of South China, Hengyang, 421001, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China.
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China.
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410008, China.
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
6
|
Zhong W, Li D, Fei Y, Hong P. A review of type 3 Gaucher disease: unique neurological manifestations and advances in treatment. Acta Neurol Belg 2024; 124:1213-1223. [PMID: 38413480 DOI: 10.1007/s13760-024-02493-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 02/01/2024] [Indexed: 02/29/2024]
Abstract
Gaucher disease (GD) is a rare lysosomal storage disease that is caused by mutations in the GBA gene. It is classified into three main phenotypes according to the patient's clinical presentation. Of these, chronic neuronopathic GD (GD3) is characterized by progressive neurological damage. Understanding the unique neurological manifestations of GD3 has important diagnostic and therapeutic implications. Our article summarizes the neurological symptoms specific to GD3 and related therapeutic advances, and it highlights the relevance of the gene to clinical symptoms, so as to provide a reference for the diagnosis and treatment of GD3.
Collapse
Affiliation(s)
- Wei Zhong
- Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Dan Li
- Department of Hematology, Shaoxing People's Hospital, 568 Zhongxing North Road, Shaoxing, 312000, Zhejiang, China
| | - Yue Fei
- Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Pan Hong
- Department of Hematology, Shaoxing People's Hospital, 568 Zhongxing North Road, Shaoxing, 312000, Zhejiang, China.
| |
Collapse
|
7
|
Mulroy E, Erro R, Bhatia KP, Hallett M. Refining the clinical diagnosis of Parkinson's disease. Parkinsonism Relat Disord 2024; 122:106041. [PMID: 38360507 PMCID: PMC11069446 DOI: 10.1016/j.parkreldis.2024.106041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/17/2024]
Abstract
Our ability to define, understand, and classify Parkinson's disease (PD) has undergone significant changes since the disorder was first described in 1817. Clinical features and neuropathologic signatures can now be supplemented by in-vivo interrogation of genetic and biological substrates of disease, offering great opportunity for further refining the diagnosis of PD. In this mini-review, we discuss the historical perspectives which shaped our thinking surrounding the definition and diagnosis of PD. We highlight the clinical, genetic, pathologic and biologic diversity which underpins the condition, and proceed to discuss how recent developments in our ability to define biologic and pathologic substrates of disease might impact PD definition, diagnosis, individualised prognostication, and personalised clinical care. We argue that Parkinson's 'disease', as currently diagnosed in the clinic, is actually a syndrome. It is the outward manifestation of any array of potential dysfunctional biologic processes, neuropathological changes, and disease aetiologies, which culminate in common outward clinical features which we term PD; each person has their own unique disease, which we can now define with increasing precision. This is an exciting time in PD research and clinical care. Our ability to refine the clinical diagnosis of PD, incorporating in-vivo assessments of disease biology, neuropathology, and neurogenetics may well herald the era of biologically-based, precision medicine approaches PD management. With this however comes a number of challenges, including how to integrate these technologies into clinical practice in a way which is acceptable to patients, promotes meaningful changes to care, and minimises health economic impact.
Collapse
Affiliation(s)
- Eoin Mulroy
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Roberto Erro
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, (SA), Italy
| | - Kailash P Bhatia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Mark Hallett
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
8
|
Cardoso F, Goetz CG, Mestre TA, Sampaio C, Adler CH, Berg D, Bloem BR, Burn DJ, Fitts MS, Gasser T, Klein C, de Tijssen MAJ, Lang AE, Lim SY, Litvan I, Meissner WG, Mollenhauer B, Okubadejo N, Okun MS, Postuma RB, Svenningsson P, Tan LCS, Tsunemi T, Wahlstrom-Helgren S, Gershanik OS, Fung VSC, Trenkwalder C. A Statement of the MDS on Biological Definition, Staging, and Classification of Parkinson's Disease. Mov Disord 2024; 39:259-266. [PMID: 38093469 DOI: 10.1002/mds.29683] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 12/27/2023] Open
Affiliation(s)
- Francisco Cardoso
- Movement Disorders Unit, Neurology Service, Internal Medicine Department, The Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Christopher G Goetz
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Tiago A Mestre
- Ottawa Hospital Research Institute; University of Ottawa Brain and Mind Research Institute; Division of Neurology, Department of Medicine, University of Ottawa, The Ottawa Hospital Ottawa, Ottawa, Ontario, Canada
| | - Cristina Sampaio
- CHDI Management/CHDI Foundation, Princeton, New Jersey, USA
- Laboratory of Clinical Pharmacology and Therapeutics, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Phoenix, Arizona, USA
| | - Daniela Berg
- Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel and Christian Albrechts-University of Kiel, Kiel, Germany
| | - Bastiaan R Bloem
- Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Department of Neurology, Center of Expertise for Parkinson and Movement Disorders, Nijmegen, The Netherlands
| | - David J Burn
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Michael S Fitts
- UAB Libraries, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Thomas Gasser
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany. German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Marina A J de Tijssen
- Department of Neurology, Expertise Centre Movement Disorders, University Medical Centre Groningen, Groningen, The Netherlands
| | - Anthony E Lang
- Edmond J. Safra Program in Parkinson's Disease, University Health Network and the University of Toronto, Toronto, Ontario, Canada
| | - Shen-Yang Lim
- Division of Neurology, Department of Medicine, and the Mah Pooi Soo and Tan Chin Nam Centre for Parkinson's and Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Irene Litvan
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Wassilios G Meissner
- CHU Bordeaux, Service de Neurologie des Maladies Neurodégénératives, Bordeaux, France
- Univ. Bordeaux, CNRS, IMN, Bordeaux, France
- Department of Medicine, University of Otago, Christchurch, and New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center, Kassel, Germany
| | - Njideka Okubadejo
- Neurology Unit, Department of Medicine, Faculty of Clinical Sciences, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Michael S Okun
- Adelaide Lackner Professor of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainsville, Florida, USA
| | - Ronald B Postuma
- Department of Neurology, McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| | | | | | - Taiji Tsunemi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | | | - Oscar S Gershanik
- Movement Disorders Unit, Institute of Neuroscience, Favaloro Foundation University Hospital, Buenos Aires, Argentina
- Cognitive Neuroscience Laboratory, Institute of Cognitive Neurology (INECO), Buenos Aires, Argentina
| | - Victor S C Fung
- Movement Disorders Unit, Department of Neurology, Westmead Hospital and Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Claudia Trenkwalder
- Paracelsus-Elena Klinik, Kassel, Germany
- Department of Neurosurgery, University Medical Center, Goettingen, Germany
| |
Collapse
|
9
|
Saffie Awad P, Teixeira-Dos-Santos D, Santos-Lobato BL, Camargos S, Cornejo-Olivas M, de Mello Rieder CR, Mata IF, Chaná-Cuevas P, Klein C, Schumacher Schuh AF. Frequency of Hereditary and GBA1-Related Parkinsonism in Latin America: A Systematic Review and Meta-Analysis. Mov Disord 2024; 39:6-16. [PMID: 37921246 DOI: 10.1002/mds.29614] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/28/2023] [Accepted: 09/05/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Identifying hereditary parkinsonism is valuable for diagnosis, genetic counseling, patient prioritization in trials, and studying the disease for personalized therapies. However, most studies were conducted in Europeans, and limited data exist on admixed populations like those from Latin America. OBJECTIVES This study aims to assess the frequency and distribution of genetic parkinsonism in Latin America. METHODS We conducted a systematic review and meta-analysis of the frequency of parkinsonian syndromes associated with genetic pathogenic variants in Latin America. We defined hereditary parkinsonism as those caused by the genes outlined by the MDS Nomenclature of Genetic Movement Disorders and heterozygous carriers of GBA1 pathogenic variants. A systematic search was conducted in PubMed, Web of Science, Embase, and LILACS in August 2022. Researchers reviewed titles and abstracts, and disagreements were resolved by a third researcher. After this screening, five researchers reanalyzed the selection criteria and extracted information based on the full paper. The frequency for each parkinsonism-related gene was determined by the presence of pathogenic/likely pathogenic variants among screened patients. Cochran's Q and I2 tests were used to quantify heterogeneity. Meta-regression, publication bias tests, and sensitivity analysis regarding study quality were also used for LRRK2-, PRKN-, and GBA1-related papers. RESULTS We included 73 studies involving 3014 screened studies from 16 countries. Among 7668 Latin American patients, pathogenic variants were found in 19 different genes. The frequency of the pathogenic variants in LRRK2 was 1.38% (95% confidence interval [CI]: 0.52-2.57), PRKN was 1.16% (95% CI: 0.08-3.05), and GBA1 was 4.17% (95% CI: 2.57-6.08). For all meta-analysis, heterogeneity was high and publication bias tests were negative, except for PRKN, which was contradictory. Information on the number of pathogenic variants in the other genes is further presented in the text. CONCLUSIONS This study provides insights into hereditary and GBA1-related parkinsonism in Latin America. Lower GBA1 frequencies compared to European/North American cohorts may result from limited access to gene sequencing. Further research is vital for regional prevalence understanding, enabling personalized care and therapies. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Paula Saffie Awad
- Programa de Pós-Graduação em Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Centro de Trastornos del Movimiento (CETRAM), Santiago, Chile
- Clínica Santa María, Santiago, Chile
| | | | - Bruno Lopes Santos-Lobato
- Hospital Ophir Loyola, Belém, Brazil
- Laboratório de Neuropatologia Experimental, Universidade Federal do Pará, Belém, Brazil
| | - Sarah Camargos
- Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mario Cornejo-Olivas
- Neurogenetics Working Group, Universidad Científica del Sur, Lima, Peru
- Neurogenetics Research Center, Instituto Nacional de Ciencias Neurologicas, Lima, Peru
| | | | - Ignacio F Mata
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Pedro Chaná-Cuevas
- Centro de Trastornos del Movimiento (CETRAM), Santiago, Chile
- Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Santiago, Chile
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Artur F Schumacher Schuh
- Programa de Pós-Graduação em Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Serviço de Neurologia, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Departamento de Farmacologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
10
|
Armstrong MJ, Barnes LL. Under-Diagnosis of Dementia with Lewy Bodies in Individuals Racialized as Black: Hypotheses Regarding Potential Contributors. J Alzheimers Dis 2024; 97:1571-1580. [PMID: 38277299 PMCID: PMC10894581 DOI: 10.3233/jad-231177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2023] [Indexed: 01/28/2024]
Abstract
Dementia with Lewy bodies (DLB) is one of the most common degenerative dementias after Alzheimer's disease (AD) dementia. DLB is under-diagnosed across populations but may be particularly missed in older Black adults. The object of this review was to examine key features of DLB and potential associations with race in order to hypothesize why DLB may be under-diagnosed in Black adults in the U.S. In terms of dementia, symptoms associated with high rates of co-pathology (e.g., AD, vascular disease) in older Black adults may obscure the clinical picture that might suggest Lewy body pathology. Research also suggests that clinicians may be predisposed to give AD dementia diagnoses to Black adults, potentially missing contributions of Lewy body pathology. Hallucinations in Black adults may be misattributed to AD or primary psychiatric disease rather than Lewy body pathology. Research on the prevalence of REM sleep behavior in diverse populations is lacking, but REM sleep behavior disorder could be under-diagnosed in Black adults due to sleep patterns or reporting by caregivers who are not bed partners. Recognition of parkinsonism could be reduced in Black adults due to clinician biases, cultural effects on self-report, and potentially underlying differences in the frequency of parkinsonism. These considerations are superimposed on structural and systemic contributions to health (e.g., socioeconomic status, education, structural racism) and individual-level social exposures (e.g., social interactions, discrimination). Improving DLB recognition in Black adults will require research to investigate reasons for diagnostic disparities and education to increase identification of core symptoms in this population.
Collapse
Affiliation(s)
- Melissa J. Armstrong
- Department of Neurology, University of Florida College of Medicine, Gainesville, FL, USA
- 1Florida Alzheimer Disease Research Center, Gainesville, FL, USA
| | - Lisa L. Barnes
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
11
|
Schaeffer E, Yilmaz R, St. Louis EK, Noyce AJ. Ethical Considerations for Identifying Individuals in the Prodromal/Early Phase of Parkinson's Disease: A Narrative Review. JOURNAL OF PARKINSON'S DISEASE 2024; 14:S307-S319. [PMID: 38995800 PMCID: PMC11492008 DOI: 10.3233/jpd-230428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/11/2024] [Indexed: 07/14/2024]
Abstract
The ability to identify individuals in the prodromal phase of Parkinson's disease has improved in recent years, raising the question of whether and how those affected should be informed about the risk of future disease. Several studies investigated prognostic counselling for individuals with isolated REM sleep behavior disorder and have shown that most patients want to receive information about prognosis, but autonomy and individual preferences must be respected. However, there are still many unanswered questions about risk disclosure or early diagnosis of PD, including the impact on personal circumstances, cultural preferences and specific challenges associated with different profiles of prodromal symptoms, genetic testing or biomarker assessments. This narrative review aims to summarize the current literature on prognostic counselling and risk disclosure in PD, as well as highlight future perspectives that may emerge with the development of new biomarkers and their anticipated impact on the definition of PD.
Collapse
Affiliation(s)
- Eva Schaeffer
- Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel and Kiel University, Kiel, Germany
| | - Rezzak Yilmaz
- Department of Neurology, Ankara University School of Medicine, Ankara, Turkey
- Ankara University Brain Research Center, Ankara, Turkey
| | - Erik K. St. Louis
- Mayo Center for Sleep Medicine, Mayo Clinic, Rochester, MN, USA
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Health System Southwest Wisconsin, La Crosse, WI, USA
| | - Alastair J. Noyce
- Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
12
|
Alizadeh P, Terroba-Chambi C, Achen B, Bruno V. Pain in monogenic Parkinson's disease: a comprehensive review. Front Neurol 2023; 14:1248828. [PMID: 38020640 PMCID: PMC10643218 DOI: 10.3389/fneur.2023.1248828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
Pain, a challenging symptom experienced by individuals diagnosed with Parkinson's disease (PD), still lacks a comprehensive understanding of its underlying pathophysiological mechanisms. A systematic investigation of its prevalence and impact on the quality of life in patients affected by monogenic forms of PD has yet to be undertaken. This comprehensive review aims to provide an overview of the association between pain and monogenic forms of PD, specifically focusing on pathogenic variants in SNCA, PRKN, PINK1, PARK7, LRRK2, GBA1, VPS35, ATP13A2, DNAJC6, FBXO7, and SYNJ1. Sixty-three articles discussing pain associated with monogenic PD were identified and analyzed. The included studies exhibited significant heterogeneity in design, sample size, and pain outcome measures. Nonetheless, the findings of this review suggest that patients with monogenic PD may experience specific types of pain depending on the pathogenic variant present, distinguishing them from non-carriers. For instance, individuals with SNCA pathogenic variants have reported painful dystonia, lower extremity pain, dorsal pain, and upper back pain. However, these observations are primarily based on case reports with unclear prevalence. Painful lower limb dystonia and lower back pain are prominent symptoms in PRKN carriers. A continual correlation has been noted between LRRK2 mutations and the emergence of pain, though the conflicting research outcomes pose challenges in reaching definitive conclusions. Individuals with PINK1 mutation carriers also frequently report experiencing pain. Pain has been frequently reported as an initial symptom and the most troublesome one in GBA1-PD patients compared to those with idiopathic PD. The evidence regarding pain in ATP13A2, PARK7, VPS35, DNAJC6, FBXO7, and SYNJ1pathogenic variants is limited and insufficient. The potential linkage between genetic profiles and pain outcomes holds promising clinical implications, allowing for the potential stratification of patients in clinical trials and the development of personalized treatments for pain in monogenic PD. In conclusion, this review underscores the need for further research to unravel the intricate relationship between pain and monogenic forms of PD. Standardized methodologies, larger sample sizes, and longitudinal studies are essential to elucidate the underlying mechanisms and develop targeted therapeutic interventions for pain management in individuals with monogenic PD.
Collapse
Affiliation(s)
- Parisa Alizadeh
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Calgary, AB, Canada
| | | | - Beatrice Achen
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Veronica Bruno
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Calgary, AB, Canada
| |
Collapse
|
13
|
Menozzi E, Toffoli M, Schapira AHV. Targeting the GBA1 pathway to slow Parkinson disease: Insights into clinical aspects, pathogenic mechanisms and new therapeutic avenues. Pharmacol Ther 2023; 246:108419. [PMID: 37080432 DOI: 10.1016/j.pharmthera.2023.108419] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/31/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
The GBA1 gene encodes the lysosomal enzyme glucocerebrosidase (GCase), which is involved in sphingolipid metabolism. Biallelic variants in GBA1 cause Gaucher disease (GD), a lysosomal storage disorder characterised by loss of GCase activity and aberrant intracellular accumulation of GCase substrates. Carriers of GBA1 variants have an increased risk of developing Parkinson disease (PD), with odds ratio ranging from 2.2 to 30 according to variant severity. GBA1 variants which do not cause GD in homozygosis can also increase PD risk. Patients with PD carrying GBA1 variants show a more rapidly progressive phenotype compared to non-carriers, emphasising the need for disease modifying treatments targeting the GBA1 pathway. Several mechanisms secondary to GCase dysfunction are potentially responsible for the pathological changes leading to PD. Misfolded GCase proteins induce endoplasmic reticulum stress and subsequent unfolded protein response and impair the autophagy-lysosomal pathway. This results in α-synuclein accumulation and spread, and promotes neurodegenerative changes. Preclinical evidence also shows that products of GCase activity can promote accumulation of α-synuclein, however there is no convincing evidence of substrate accumulation in GBA1-PD brains. Altered lipid homeostasis secondary to loss of GCase activity could also contribute to PD pathology. Treatments that target the GBA1 pathway could reverse these pathological processes and halt/slow the progression of PD. These range from augmentation of GCase activity via GBA1 gene therapy, restoration of normal intracellular GCase trafficking via molecular chaperones, and substrate reduction therapy. This review discusses the pathways associated with GBA1-PD and related novel GBA1-targeted interventions for PD treatment.
Collapse
Affiliation(s)
- 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, MD 20815, United States of America
| | - Marco Toffoli
- 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, MD 20815, United States of America
| | - 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, MD 20815, United States of America.
| |
Collapse
|
14
|
Baden P, Perez MJ, Raji H, Bertoli F, Kalb S, Illescas M, Spanos F, Giuliano C, Calogero AM, Oldrati M, Hebestreit H, Cappelletti G, Brockmann K, Gasser T, Schapira AHV, Ugalde C, Deleidi M. Glucocerebrosidase is imported into mitochondria and preserves complex I integrity and energy metabolism. Nat Commun 2023; 14:1930. [PMID: 37024507 PMCID: PMC10079970 DOI: 10.1038/s41467-023-37454-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 03/17/2023] [Indexed: 04/08/2023] Open
Abstract
Mutations in GBA1, the gene encoding the lysosomal enzyme β-glucocerebrosidase (GCase), which cause Gaucher's disease, are the most frequent genetic risk factor for Parkinson's disease (PD). Here, we employ global proteomic and single-cell genomic approaches in stable cell lines as well as induced pluripotent stem cell (iPSC)-derived neurons and midbrain organoids to dissect the mechanisms underlying GCase-related neurodegeneration. We demonstrate that GCase can be imported from the cytosol into the mitochondria via recognition of internal mitochondrial targeting sequence-like signals. In mitochondria, GCase promotes the maintenance of mitochondrial complex I (CI) integrity and function. Furthermore, GCase interacts with the mitochondrial quality control proteins HSP60 and LONP1. Disease-associated mutations impair CI stability and function and enhance the interaction with the mitochondrial quality control machinery. These findings reveal a mitochondrial role of GCase and suggest that defective CI activity and energy metabolism may drive the pathogenesis of GCase-linked neurodegeneration.
Collapse
Affiliation(s)
- Pascale Baden
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Maria Jose Perez
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Hariam Raji
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Federico Bertoli
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Stefanie Kalb
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - María Illescas
- Instituto de Investigación Hospital 12 de Octubre (i + 12), Madrid, 28041, Spain
| | - Fokion Spanos
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Claudio Giuliano
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Unit of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, 27100, Pavia, Italy
| | - Alessandra Maria Calogero
- Department of Biosciences, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Marvin Oldrati
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Hannah Hebestreit
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Graziella Cappelletti
- Department of Biosciences, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Kathrin Brockmann
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Thomas Gasser
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Anthony H V Schapira
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, Royal Free Campus, London, NW3 2PF, UK
| | - Cristina Ugalde
- Instituto de Investigación Hospital 12 de Octubre (i + 12), Madrid, 28041, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), U723, Madrid, Spain
| | - Michela Deleidi
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
- Institut Imagine, INSERM UMR1163 Paris Cite' University, 24 boulevard du Montparnasse, 75015, Paris, France.
| |
Collapse
|
15
|
Who is at Risk of Parkinson Disease? Refining the Preclinical Phase of GBA1 and LRRK2 Variant Carriers: a Clinical, Biochemical, and Imaging Approach. Curr Neurol Neurosci Rep 2023; 23:121-130. [PMID: 36881256 PMCID: PMC10119235 DOI: 10.1007/s11910-023-01259-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2023] [Indexed: 03/08/2023]
Abstract
PURPOSE OF REVIEW Genetic variants in GBA1 and LRRK2 genes are the commonest genetic risk factor for Parkinson disease (PD); however, the preclinical profile of GBA1 and LRRK2 variant carriers who will develop PD is unclear. This review aims to highlight the more sensitive markers that can stratify PD risk in non-manifesting GBA1 and LRRK2 variant carriers. RECENT FINDINGS Several case-control and a few longitudinal studies evaluated clinical, biochemical, and neuroimaging markers within cohorts of non-manifesting carriers of GBA1 and LRRK2 variants. Despite similar levels of penetrance of PD in GBA1 and LRRK2 variant carriers (10-30%), these individuals have distinct preclinical profiles. GBA1 variant carriers at higher risk of PD can present with prodromal symptoms suggestive of PD (hyposmia), display increased α-synuclein levels in peripheral blood mononuclear cells, and show dopamine transporter abnormalities. LRRK2 variant carriers at higher risk of PD might show subtle motor abnormalities, but no prodromal symptoms, higher exposure to some environmental factors (non-steroid anti-inflammatory drugs), and peripheral inflammatory profile. This information will help clinicians tailor appropriate screening tests and counseling and facilitate researchers in the development of predictive markers, disease-modifying treatments, and selection of healthy individuals who might benefit from preventive interventions.
Collapse
|
16
|
Usnich T, Olmedillas M, Schell N, Paul JJ, Curado F, Skobalj S, Csoti I, Ertan S, Gruber D, Zittel S, Sammler E, Isaacson SH, Kühn AA, Pedrosa DJ, Reetz K, Kasten M, Rolfs A, Bauer P, Skrahina V, Klein C, Brüggemann N. Frequency of non-motor symptoms in Parkinson's disease patients carrying the E326K and T369M GBA risk variants. Parkinsonism Relat Disord 2023; 107:105248. [PMID: 36565535 DOI: 10.1016/j.parkreldis.2022.105248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Affiliation(s)
- Tatiana Usnich
- Institute of Neurogenetics, University of Lübeck, Germany
| | | | - Nathalie Schell
- Institute of Neurogenetics, University of Lübeck, Germany; Department of Pediatrics, Universitätsmedizin Essen, Germany
| | | | | | | | - Ilona Csoti
- Gertrudis Clinic Biskirchen, Parkinson-Center, Leun, Germany
| | - Sibel Ertan
- Department of Neurology, Koç University School of Medicine, Istanbul, Turkey
| | - Doreen Gruber
- Movement Disorders Clinic, Beelitz-Heilstätten, Germany
| | - Simone Zittel
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Stuart H Isaacson
- Parkinson's Disease and Movement Disorder Center of Boca Raton, Boca Raton, USA
| | - Andrea A Kühn
- Department of Neurology, Movement Disorders and Neuromodulation Unit, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - David J Pedrosa
- Department of Neurology, University Hospital Marburg, Marburg, Germany
| | - Kathrin Reetz
- Department of Neurology, University Hospital Aachen, Aachen, Germany
| | - Meike Kasten
- Institute of Neurogenetics, University of Lübeck, Germany; Department of Psychiatry and Psychotherapy, University of Lübeck, Germany
| | - Arndt Rolfs
- CENTOGENE GmbH, Rostock, Germany; University of Rostock, Albrecht Kossel Institute, Rostock, Germany; Arcensus GmbH, Rostock, Germany
| | | | - Volha Skrahina
- CENTOGENE GmbH, Rostock, Germany; Arcensus GmbH, Rostock, Germany
| | | | - Norbert Brüggemann
- Institute of Neurogenetics, University of Lübeck, Germany; Department of Neurology, University of Luebeck, Luebeck, Germany.
| |
Collapse
|
17
|
GBA1 Gene Mutations in α-Synucleinopathies-Molecular Mechanisms Underlying Pathology and Their Clinical Significance. Int J Mol Sci 2023; 24:ijms24032044. [PMID: 36768367 PMCID: PMC9917178 DOI: 10.3390/ijms24032044] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
α-Synucleinopathies comprise a group of neurodegenerative diseases characterized by altered accumulation of a protein called α-synuclein inside neurons and glial cells. This aggregation leads to the formation of intraneuronal inclusions, Lewy bodies, that constitute the hallmark of α-synuclein pathology. The most prevalent α-synucleinopathies are Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). To date, only symptomatic treatment is available for these disorders, hence new approaches to their therapy are needed. It has been observed that GBA1 mutations are one of the most impactful risk factors for developing α-synucleinopathies such as PD and DLB. Mutations in the GBA1 gene, which encodes a lysosomal hydrolase β-glucocerebrosidase (GCase), cause a reduction in GCase activity and impaired α-synuclein metabolism. The most abundant GBA1 gene mutations are N370S or N409S, L444P/L483P and E326K/E365K. The mechanisms by which GCase impacts α-synuclein aggregation are poorly understood and need to be further investigated. Here, we discuss some of the potential interactions between α-synuclein and GCase and show how GBA1 mutations may impact the course of the most prevalent α-synucleinopathies.
Collapse
|
18
|
Volta M. Lysosomal Pathogenesis of Parkinson's Disease: Insights From LRRK2 and GBA1 Rodent Models. Neurotherapeutics 2023; 20:127-139. [PMID: 36085537 PMCID: PMC10119359 DOI: 10.1007/s13311-022-01290-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2022] [Indexed: 01/18/2023] Open
Abstract
The discovery of mutations in LRRK2 and GBA1 that are linked to Parkinson's disease provided further evidence that autophagy and lysosome pathways are likely implicated in the pathogenic process. Their protein products are important regulators of lysosome function. LRRK2 has kinase-dependent effects on lysosome activity, autophagic efficacy and lysosomal Ca2+ signaling. Glucocerebrosidase (encoded by GBA1) is a hydrolytic enzyme contained in the lysosomes and contributes to the degradation of alpha-synuclein. PD-related mutations in LRRK2 and GBA1 slow the degradation of alpha-synuclein, thus directly implicating the dysfunction of the process in the neuropathology of Parkinson's disease. The development of genetic rodent models of LRRK2 and GBA1 provided hopes of obtaining reliable preclinical models in which to study pathogenic processes and perform drug validation studies. Here, I will review the extensive characterization of these models, their impact on understanding lysosome alterations in the course of Parkinson's disease and what novel insights have been obtained. In addition, I will discuss how these models fare with respect to the features of a "gold standard" animal models and what could be attempted in future studies to exploit LRRK2 and GBA1 rodent models in the fight against Parkinson's disease.
Collapse
Affiliation(s)
- Mattia Volta
- Institute for Biomedicine, Eurac Research - Affiliated Institute of the University of Lübeck, via Volta 21, Bolzano, 39100, Italy.
| |
Collapse
|
19
|
Dursun H, Metli K, Bayram F. Case Report: Novel Pathogenic Variant Detected in Two Siblings with Type 1 Gaucher Disease. Endocr Metab Immune Disord Drug Targets 2023; 23:1780-1783. [PMID: 37157213 DOI: 10.2174/1871530323666230508151529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/17/2023] [Accepted: 03/10/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND Gaucher disease (GD) is an autosomal recessive lysosomal storage disease. The disease develops due to glucocerebrosidase enzyme deficiency caused by biallelic pathogenic variants in the glucosylceramidase beta 1 (GBA1) gene, which encodes the glucocerebrosidase enzyme. The GBA1 gene is located at chromosomal location 1q22 and consists of 11 exons. In this article, we report a novel pathogenic variant in the GBA1 gene. CASE PRESENTATIONS A 32-year-old female patient with no known chronic disease was admitted with complaints of weakness, bone pain, and abdominal pain. Her evaluation included hepatosplenomegaly, thrombocytopenia, osteoporosis, and anemia. The clinical suspicion of Gaucher disease was confirmed by glucocerebrosidase enzyme level and genetic testing. In her family screening, her sister also had hepato-splenomegaly, osteoporosis, thrombocytopenia, and anemia. Both sisters had no neurological symptoms. As a result of GBA1 gene sequence analysis in two of our patients, a missense variant was detected in the c.593C > A homozygous genotype. This variant has not been reported in any previously published case. CONCLUSION In this case report, we aimed to contribute to the literature by reporting a new novel pathogenic variant in the GBA1 gene leading to type 1 Gaucher disease that has not been described before.
Collapse
Affiliation(s)
- Huseyin Dursun
- Department of Endocrinology, Bingol State Hospital, Bingol, Turkey
| | - Kubra Metli
- Department of Medical Genetics, Derince Education And Research Hospital, Kocaeli, Turkey
| | - Fahri Bayram
- Department of Endocrinology, Erciyes University Medical School, Kayseri, Turkey
| |
Collapse
|
20
|
Subbotina TN, Abramov VG, Shaleva AA, Vereschagina SV, Pokhabov DV. [Analysis of GBA mutations in patients with Parkinson's disease in the Krasnoyarsk region]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:103-108. [PMID: 37084373 DOI: 10.17116/jnevro2023123041103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2023]
Abstract
OBJECTIVE To analyze mutations and polymorphisms in exons 2, 7, 8, 9, 10 and 11 of the glucocerebrosidase (GBA) gene in patients of the Krasnoyarsk region diagnosed with Parkinson's disease (PD). MATERIAL AND METHODS Seventy-five patients with sporadic and familial forms of PD were examined. Genomic DNA was isolated from the whole blood of patients. The above mentioned exons of GBA were analyzed using Sanger sequencing. RESULTS Various changes in the DNA structure of GBA were detected in 11 patients, thus, the overall frequency of variants was 14.7%, and the frequency of pathologically significant mutations (p.L444P, p.D409H, p.H255Q) was 5.3%. CONCLUSION The frequencies of variants in GBA, one of the most common high-risk factors for PD, in patients of the Krasnoyarsk region turned out to be quite high and comparable to that in patients in other populations of the world. Thus, screening for GBA mutations is relevant for PD patients living in the Krasnoyarsk region as part of genetic counseling at present, and in the future it may be necessary for personalized treatment.
Collapse
Affiliation(s)
- T N Subbotina
- Siberian federal university, Krasnoyarsk, Russia
- Federal Siberian Research and Clinical Center, Krasnoyarsk, Russia
| | - V G Abramov
- Federal Siberian Research and Clinical Center, Krasnoyarsk, Russia
| | - A A Shaleva
- Siberian federal university, Krasnoyarsk, Russia
- Federal Siberian Research and Clinical Center, Krasnoyarsk, Russia
| | - S V Vereschagina
- Federal Siberian Research and Clinical Center, Krasnoyarsk, Russia
| | - D V Pokhabov
- Federal Siberian Research and Clinical Center, Krasnoyarsk, Russia
- Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| |
Collapse
|
21
|
Abdul Murad NA, Sulaiman SA, Ahmad-Annuar A, Mohamed Ibrahim N, Mohamed W, Md Rani SA, Mok KY. Editorial: Genetic and molecular diversity in Parkinson's disease. Front Aging Neurosci 2022; 14:1094914. [PMID: 36589546 PMCID: PMC9800990 DOI: 10.3389/fnagi.2022.1094914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Affiliation(s)
- Nor Azian Abdul Murad
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| | - Siti Aishah Sulaiman
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| | - Azlina Ahmad-Annuar
- Department of Biomedical Science, Faculty of Medicine, University of Malaya (UM), Kuala Lumpur, Malaysia
| | - Norlinah Mohamed Ibrahim
- Neurology Unit, Department of Medicine, Faculty of Medicine, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia,*Correspondence: Norlinah Mohamed Ibrahim ✉
| | - Wael Mohamed
- Kulliyah of Medicine, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| | - Shahrul Azmin Md Rani
- Neurology Unit, Department of Medicine, Faculty of Medicine, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| | - Kin Ying Mok
- Department of Neurodegenerative Disease, University College London (UCL) Institute of Neurology, University College London, London, United Kingdom,State Key Laboratory of Molecular Neuroscience, Division of Life Science, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong SAR, China,Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, Hong Kong SAR, China,Kin Ying Mok ✉
| |
Collapse
|
22
|
Kim J, Daadi EW, Oh T, Daadi ES, Daadi MM. Human Induced Pluripotent Stem Cell Phenotyping and Preclinical Modeling of Familial Parkinson's Disease. Genes (Basel) 2022; 13:1937. [PMID: 36360174 PMCID: PMC9689743 DOI: 10.3390/genes13111937] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 12/05/2022] Open
Abstract
Parkinson's disease (PD) is primarily idiopathic and a highly heterogenous neurodegenerative disease with patients experiencing a wide array of motor and non-motor symptoms. A major challenge for understanding susceptibility to PD is to determine the genetic and environmental factors that influence the mechanisms underlying the variations in disease-associated traits. The pathological hallmark of PD is the degeneration of dopaminergic neurons in the substantia nigra pars compacta region of the brain and post-mortem Lewy pathology, which leads to the loss of projecting axons innervating the striatum and to impaired motor and cognitive functions. While the cause of PD is still largely unknown, genome-wide association studies provide evidence that numerous polymorphic variants in various genes contribute to sporadic PD, and 10 to 15% of all cases are linked to some form of hereditary mutations, either autosomal dominant or recessive. Among the most common mutations observed in PD patients are in the genes LRRK2, SNCA, GBA1, PINK1, PRKN, and PARK7/DJ-1. In this review, we cover these PD-related mutations, the use of induced pluripotent stem cells as a disease in a dish model, and genetic animal models to better understand the diversity in the pathogenesis and long-term outcomes seen in PD patients.
Collapse
Affiliation(s)
- Jeffrey Kim
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
- Cell Systems and Anatomy, San Antonio, TX 78229, USA
| | - Etienne W. Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Thomas Oh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Elyas S. Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Marcel M. Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
- Cell Systems and Anatomy, San Antonio, TX 78229, USA
- Department of Radiology, Long School of Medicine, University of Texas Health at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
23
|
Sahyadri M, Nadiga APR, Mehdi S, Mruthunjaya K, Nayak PG, Parihar VK, Manjula SN. Mitochondria-lysosome crosstalk in GBA1-associated Parkinson's disease. 3 Biotech 2022; 12:230. [PMID: 35992895 PMCID: PMC9388709 DOI: 10.1007/s13205-022-03261-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 07/17/2022] [Indexed: 11/26/2022] Open
Abstract
Organelle crosstalk is significant in regulating their respective functions and subsequent cell fate. Mitochondria and lysosomes are amongst the essential organelles in maintaining cellular homeostasis. Mitochondria-lysosome connections, which may develop dynamically in the human neurons, have been identified as sites of bidirectional communication. Aberrancies are often associated with neurodegenerative disorders like Parkinson's disease (PD), suggesting the physical and functional link between these two organelles. PD is often linked with genetic mutations of several mutations discovered in the familial forms of the disease; some are considered risk factors. Many of these genes are either associated with mitochondrial function or belong to endo-lysosomal pathways. The recent investigations have indicated that neurons with mutant glucosylceramidase beta (GBA1) exhibit extended mitochondria-lysosome connections in individuals with PD. This may be due to impaired control of the untethering protein, which aids in the hydrolysis of Rab7 GTP required for contact untethering. A GCase modulator may be used to augment the reduced GBA1 lysosomal enzyme activity in the neurons of PD patients. This review focuses on how GBA1 mutation in PD is interlinked with mitochondria-lysosome (ML) crosstalk, exploring the pathways governing these interactions and mechanistically comprehending the mitochondrial and lysosomal miscommunication in the pathophysiology of PD. This review is based on the limited literature available on the topic and hence may be subject to bias in its views. Our estimates may be conservative and limited due to the lack of studies under the said discipline due to its inherent complex nature. The current association of GBA1 to PD pathogenesis is based on the limited scope of study and further research is necessary to explore the risk factors further and identify the relationship with more detail.
Collapse
Affiliation(s)
- M. Sahyadri
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015 Karnataka India
| | - Abhishek P. R. Nadiga
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015 Karnataka India
| | - Seema Mehdi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015 Karnataka India
| | - K. Mruthunjaya
- Department of Pharmacognosy, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015 Karnataka India
| | - Pawan G. Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, 576104 Karnataka India
| | - Vipan K. Parihar
- Department of Pharmacology and Toxicology, NIPER-Hajipur, Bihar, 844102 India
| | - S. N. Manjula
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015 Karnataka India
| |
Collapse
|
24
|
Vieira SRL, Schapira AHV. Glucocerebrosidase mutations and Parkinson disease. J Neural Transm (Vienna) 2022; 129:1105-1117. [PMID: 35932311 PMCID: PMC9463283 DOI: 10.1007/s00702-022-02531-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/07/2022] [Indexed: 11/06/2022]
Abstract
The discovery of glucocerebrosidase (GBA1) mutations as the greatest numerical genetic risk factor for the development of Parkinson disease (PD) resulted in a paradigm shift within the research landscape. Efforts to elucidate the mechanisms behind GBA1-associated PD have highlighted shared pathways in idiopathic PD including the loss and gain-of-function hypotheses, endoplasmic reticulum stress, lipid metabolism, neuroinflammation, mitochondrial dysfunction and altered autophagy-lysosomal pathway responsible for degradation of aggregated and misfolded a-synuclein. GBA1-associated PD exhibits subtle differences in phenotype and disease progression compared to idiopathic counterparts notably an earlier age of onset, faster motor decline and greater frequency of non-motor symptoms (which also constitute a significant aspect of the prodromal phase of the disease). GBA1-targeted therapies have been developed and are being investigated in clinical trials. The most notable are Ambroxol, a small molecule chaperone, and Venglustat, a blood-brain-barrier-penetrant substrate reduction therapy agent. It is imperative that further studies clarify the aetiology of GBA1-associated PD, enabling the development of a greater abundance of targeted therapies in this new era of precision medicine.
Collapse
Affiliation(s)
- Sophia R L Vieira
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, Rowland Hill St., London, NW3 2PF, UK
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, Rowland Hill St., London, NW3 2PF, UK.
| |
Collapse
|
25
|
Smith LJ, Lee CY, Menozzi E, Schapira AHV. Genetic variations in GBA1 and LRRK2 genes: Biochemical and clinical consequences in Parkinson disease. Front Neurol 2022; 13:971252. [PMID: 36034282 PMCID: PMC9416236 DOI: 10.3389/fneur.2022.971252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/25/2022] [Indexed: 11/24/2022] Open
Abstract
Variants in the GBA1 and LRRK2 genes are the most common genetic risk factors associated with Parkinson disease (PD). Both genes are associated with lysosomal and autophagic pathways, with the GBA1 gene encoding for the lysosomal enzyme, glucocerebrosidase (GCase) and the LRRK2 gene encoding for the leucine-rich repeat kinase 2 enzyme. GBA1-associated PD is characterized by earlier age at onset and more severe non-motor symptoms compared to sporadic PD. Mutations in the GBA1 gene can be stratified into severe, mild and risk variants depending on the clinical presentation of disease. Both a loss- and gain- of function hypothesis has been proposed for GBA1 variants and the functional consequences associated with each variant is often linked to mutation severity. On the other hand, LRRK2-associated PD is similar to sporadic PD, but with a more benign disease course. Mutations in the LRRK2 gene occur in several structural domains and affect phosphorylation of GTPases. Biochemical studies suggest a possible convergence of GBA1 and LRRK2 pathways, with double mutant carriers showing a milder phenotype compared to GBA1-associated PD. This review compares GBA1 and LRRK2-associated PD, and highlights possible genotype-phenotype associations for GBA1 and LRRK2 separately, based on biochemical consequences of single variants.
Collapse
Affiliation(s)
- Laura J. Smith
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London (UCL), London, United Kingdom
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| | - Chiao-Yin Lee
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London (UCL), London, United Kingdom
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| | - Elisa Menozzi
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London (UCL), London, United Kingdom
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| | - Anthony H. V. Schapira
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London (UCL), London, United Kingdom
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| |
Collapse
|
26
|
Glucocerebrosidase variant T369M is not a risk factor for Parkinson's disease in Sweden. Neurosci Lett 2022; 784:136767. [PMID: 35779693 DOI: 10.1016/j.neulet.2022.136767] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Genetic variants in the Beta-glucocerebrosidase gene (GBA1) is a known risk factor for Parkinson's disease. The GBA1 mutations L444P, N370S and many other have been shown to associate with the disease in populations with diverse background. Some GBA1 polymorphisms have a less pronounced effect, and their pathogenicity has been debated. We have previously found associations with L444P, N370S and E326K and Parkinson's disease in Sweden. METHOD In this study we used pyrosequencing to genotype the T369M variant in a large Swedish cohort consisting of 1,131 patients with idiopathic Parkinson's disease, and 1,594 control subjects to evaluate the possibility of this variant conferring an increased risk for Parkinson's disease. RESULTS The minor allele frequency was 2.15% in patients and 1.76% in controls. Statistical analysis showed that there was no significant difference in allele frequency between patients and control subjects, p-value 0.37, Odds Ratio 1.23 with a 95% confidence interval of 0.82-1.83. CONCLUSION Our results suggest that T369M is not a risk factor for Parkinson's disease in the Swedish population.
Collapse
|
27
|
Straniero L, Rimoldi V, Monfrini E, Bonvegna S, Melistaccio G, Lake J, Soldà G, Aureli M, Shankaracharya, Keagle P, Foroud T, Landers JE, Blauwendraat C, Zecchinelli A, Cilia R, Di Fonzo A, Pezzoli G, Duga S, Asselta R. Role of Lysosomal Gene Variants in Modulating GBA-Associated Parkinson's Disease Risk. Mov Disord 2022; 37:1202-1210. [PMID: 35262230 PMCID: PMC9310717 DOI: 10.1002/mds.28987] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/08/2022] [Accepted: 02/13/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND To date, variants in the GBA gene represent the most frequent large-effect genetic factor associated with Parkinson's disease (PD). However, the reason why individuals with the same GBA variant may or may not develop neurodegeneration and PD is still unclear. OBJECTIVES Therefore, we evaluated the contribution of rare variants in genes responsible for lysosomal storage disorders (LSDs) to GBA-PD risk, comparing the burden of deleterious variants in LSD genes in PD patients versus asymptomatic subjects, all carriers of deleterious variants in GBA. METHODS We used a custom next-generation sequencing panel, including 50 LSD genes, to screen 305 patients and 207 controls (discovery cohort). Replication and meta-analysis were performed in two replication cohorts of GBA-variant carriers, of 250 patients and 287 controls, for whom exome or genome data were available. RESULTS Statistical analysis in the discovery cohort revealed a significantly increased burden of deleterious variants in LSD genes in patients (P = 0.0029). Moreover, our analyses evidenced that the two strongest modifiers of GBA penetrance are a second variation in GBA (5.6% vs. 1.4%, P = 0.023) and variants in genes causing mucopolysaccharidoses (6.9% vs. 1%, P = 0.0020). These results were confirmed in the meta-analysis, where we observed pooled odds ratios of 1.42 (95% confidence interval [CI] = 1.10-1.83, P = 0.0063), 4.36 (95% CI = 2.02-9.45, P = 0.00019), and 1.83 (95% CI = 1.04-3.22, P = 0.038) for variants in LSD genes, GBA, and mucopolysaccharidosis genes, respectively. CONCLUSION The identification of genetic lesions in lysosomal genes increasing PD risk may have important implications in terms of patient stratification for future therapeutic trials. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson Movement Disorder Society.
Collapse
Affiliation(s)
- Letizia Straniero
- Department of Biomedical SciencesHumanitas UniversityMilanItaly
- Humanitas Clinical and Research CenterIRCCSMilanItaly
| | - Valeria Rimoldi
- Department of Biomedical SciencesHumanitas UniversityMilanItaly
- Humanitas Clinical and Research CenterIRCCSMilanItaly
| | - Edoardo Monfrini
- IRCCS Foundation Ca' Granda Ospedale Maggiore PoliclinicoNeurology UnitMilanItaly
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | | | | | - Julie Lake
- Laboratory of NeurogeneticsNational Institute on Aging, National Institutes of HealthBethesdaMarylandUSA
| | - Giulia Soldà
- Department of Biomedical SciencesHumanitas UniversityMilanItaly
- Humanitas Clinical and Research CenterIRCCSMilanItaly
| | - Massimo Aureli
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
| | - Shankaracharya
- Department of NeurologyUniversity of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Pamela Keagle
- Department of NeurologyUniversity of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Tatiana Foroud
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - John E. Landers
- Department of NeurologyUniversity of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Cornelis Blauwendraat
- Laboratory of NeurogeneticsNational Institute on Aging, National Institutes of HealthBethesdaMarylandUSA
| | | | - Roberto Cilia
- Fondazione IRCCS Istituto Neurologico Carlo BestaParkinson and Movement Disorders UnitMilanItaly
| | - Alessio Di Fonzo
- IRCCS Foundation Ca' Granda Ospedale Maggiore PoliclinicoNeurology UnitMilanItaly
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Gianni Pezzoli
- Parkinson InstituteASST Gaetano Pini‐CTOMilanItaly
- Fondazione Grigioni per il Morbo di ParkinsonMilanItaly
| | - Stefano Duga
- Department of Biomedical SciencesHumanitas UniversityMilanItaly
- Humanitas Clinical and Research CenterIRCCSMilanItaly
| | - Rosanna Asselta
- Department of Biomedical SciencesHumanitas UniversityMilanItaly
- Humanitas Clinical and Research CenterIRCCSMilanItaly
| |
Collapse
|
28
|
Höglinger G, Schulte C, Jost WH, Storch A, Woitalla D, Krüger R, Falkenburger B, Brockmann K. GBA-associated PD: chances and obstacles for targeted treatment strategies. J Neural Transm (Vienna) 2022; 129:1219-1233. [PMID: 35639160 PMCID: PMC9463270 DOI: 10.1007/s00702-022-02511-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/01/2022] [Indexed: 11/08/2022]
Abstract
Given the clear role of GBA in the pathogenesis of Parkinson’s disease (PD) and its impact on phenotypical characteristics, this review provides an overview of the current knowledge of GBA-associated PD with a special focus on clinical trajectories and the underlying pathological mechanisms. Importantly, differences and characteristics based on mutation severity are recognized, and current as well as potential future treatment options are discussed. These findings will inform future strategies for patient stratification and cohort enrichment as well as suitable outcome measures when designing clinical trials.
Collapse
Affiliation(s)
- Günter Höglinger
- Department of Neurology, Hannover Medical School, 30625, Hannover, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Claudia Schulte
- Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, Center of Neurology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany.,German Center for Neurodegenerative Disease (DZNE), Tuebingen, Germany
| | | | - Alexander Storch
- Department of Neurology, Rostock University, Gehlsheimer Str. 20, 18147, Rostock, Germany.,German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Gehlsheimer Str. 20, 18147, Rostock, Germany
| | - Dirk Woitalla
- Department of Neurology, St. Josef-Hospital, Katholische Kliniken Ruhrhalbinsel, Contilia Gruppe, Essen, Germany
| | - Rejko Krüger
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg.,Translational Neuroscience, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg.,Parkinson Research Clinic, Centre Hospitalier de Luxembourg (CHL), Luxembourg, Luxembourg
| | - Björn Falkenburger
- Department of Neurology, Faculty of Medicine, University Hospital Carl Gustav Carus and Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Kathrin Brockmann
- Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, Center of Neurology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany. .,German Center for Neurodegenerative Disease (DZNE), Tuebingen, Germany.
| |
Collapse
|
29
|
Fernández-Santiago R, Sharma M. What have we learned from genome-wide association studies (GWAS) in Parkinson's disease? Ageing Res Rev 2022; 79:101648. [PMID: 35595184 DOI: 10.1016/j.arr.2022.101648] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/11/2022] [Accepted: 05/11/2022] [Indexed: 11/01/2022]
Abstract
After fifteen years of genome-wide association studies (GWAS) in Parkinson's disease (PD), what have we learned? Addressing this question will help catalogue the progress made towards elucidating disease mechanisms, improving the clinical utility of the identified loci, and envisioning how we can harness the strides to develop translational GWAS strategies. Here we review the advances of PD GWAS made to date while critically addressing the challenges and opportunities for next-generation GWAS. Thus, deciphering the missing heritability in underrepresented populations is currently at the reach of hand for a truly comprehensive understanding of the genetics of PD across the different ethnicities. Moreover, state-of-the-art GWAS designs hold a true potential for enhancing the clinical applicability of genetic findings, for instance, by improving disease prediction (PD risk and progression). Lastly, advanced PD GWAS findings, alone or in combination with clinical and environmental parameters, are expected to have the capacity for defining patient enriched cohorts stratified by genetic risk profiles and readily available for neuroprotective clinical trials. Overall, envisioning future strategies for advanced GWAS is currently timely and can be instrumental in providing novel genetic readouts essential for a true clinical translatability of PD genetic findings.
Collapse
|
30
|
Galvagnion C, Marlet FR, Cerri S, Schapira AHV, Blandini F, Di Monte DA. Sphingolipid changes in Parkinson L444P GBA mutation fibroblasts promote α-synuclein aggregation. Brain 2022; 145:1038-1051. [PMID: 35362022 PMCID: PMC9050548 DOI: 10.1093/brain/awab371] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 08/19/2021] [Accepted: 09/06/2021] [Indexed: 12/30/2022] Open
Abstract
Intraneuronal accumulation of aggregated α-synuclein is a pathological hallmark of Parkinson’s disease. Therefore, mechanisms capable of promoting α-synuclein deposition bear important pathogenetic implications. Mutations of the glucocerebrosidase 1 (GBA) gene represent a prevalent Parkinson’s disease risk factor. They are associated with loss of activity of a key enzyme involved in lipid metabolism, glucocerebrosidase, supporting a mechanistic relationship between abnormal α-synuclein–lipid interactions and the development of Parkinson pathology. In this study, the lipid membrane composition of fibroblasts isolated from control subjects, patients with idiopathic Parkinson’s disease and Parkinson's disease patients carrying the L444P GBA mutation (PD-GBA) was assayed using shotgun lipidomics. The lipid profile of PD-GBA fibroblasts differed significantly from that of control and idiopathic Parkinson’s disease cells. It was characterized by an overall increase in sphingolipid levels. It also featured a significant increase in the proportion of ceramide, sphingomyelin and hexosylceramide molecules with shorter chain length and a decrease in the percentage of longer-chain sphingolipids. The extent of this shift was correlated to the degree of reduction of fibroblast glucocerebrosidase activity. Lipid extracts from control and PD-GBA fibroblasts were added to recombinant α-synuclein solutions. The kinetics of α-synuclein aggregation were significantly accelerated after addition of PD-GBA extracts as compared to control samples. Amyloid fibrils collected at the end of these incubations contained lipids, indicating α-synuclein–lipid co-assembly. Lipids extracted from α-synuclein fibrils were also analysed by shotgun lipidomics. Data revealed that the lipid content of these fibrils was significantly enriched by shorter-chain sphingolipids. In a final set of experiments, control and PD-GBA fibroblasts were incubated in the presence of the small molecule chaperone ambroxol. This treatment restored glucocerebrosidase activity and sphingolipid levels and composition of PD-GBA cells. It also reversed the pro-aggregation effect that lipid extracts from PD-GBA fibroblasts had on α-synuclein. Taken together, the findings of this study indicate that the L444P GBA mutation and consequent enzymatic loss are associated with a distinctly altered membrane lipid profile that provides a biological fingerprint of this mutation in Parkinson fibroblasts. This altered lipid profile could also be an indicator of increased risk for α-synuclein aggregate pathology.
Collapse
Affiliation(s)
- Céline Galvagnion
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany.,Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen Ø, Denmark
| | - Frederik Ravnkilde Marlet
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen Ø, Denmark
| | - Silvia Cerri
- Cellular and Molecular Neurobiology Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Fabio Blandini
- Cellular and Molecular Neurobiology Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy.,Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Donato A Di Monte
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| |
Collapse
|
31
|
Napolioni V, Fredericks CA, Kim Y, Channappa D, Khan RR, Kim LH, Zafar F, Couthouis J, Davidzon GA, Mormino EC, Gitler AD, Montine TJ, Schüle B, Greicius MD. Phenotypic Heterogeneity among GBA p.R202X Carriers in Lewy Body Spectrum Disorders. Biomedicines 2022; 10:biomedicines10010160. [PMID: 35052839 PMCID: PMC8774039 DOI: 10.3390/biomedicines10010160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/03/2022] [Accepted: 01/11/2022] [Indexed: 02/02/2023] Open
Abstract
We describe the clinical and neuropathologic features of patients with Lewy body spectrum disorder (LBSD) carrying a nonsense variant, c.604C>T; p.R202X, in the glucocerebrosidase 1 (GBA) gene. While this GBA variant is causative for Gaucher's disease, the pathogenic role of this mutation in LBSD is unclear. Detailed neuropathologic evaluation was performed for one index case and a structured literature review of other GBA p.R202X carriers was conducted. Through the systematic literature search, we identified three additional reported subjects carrying the same GBA mutation, including one Parkinson's disease (PD) patient with early disease onset, one case with neuropathologically-verified LBSD, and one unaffected relative of a Gaucher's disease patient. Among the affected subjects carrying the GBA p.R202X, all males were diagnosed with Lewy body dementia, while the two females presented as PD. The clinical penetrance of GBA p.R202X in LBSD patients and families argues strongly for a pathogenic role for this variant, although presenting with a striking phenotypic heterogeneity of clinical and pathological features.
Collapse
Affiliation(s)
- Valerio Napolioni
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; (C.A.F.); (Y.K.); (R.R.K.); (L.H.K.); (E.C.M.); (M.D.G.)
- Correspondence: ; Tel.: +1-(669)-287-2586
| | - Carolyn A. Fredericks
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; (C.A.F.); (Y.K.); (R.R.K.); (L.H.K.); (E.C.M.); (M.D.G.)
| | - Yongha Kim
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; (C.A.F.); (Y.K.); (R.R.K.); (L.H.K.); (E.C.M.); (M.D.G.)
| | - Divya Channappa
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; (D.C.); (F.Z.); (T.J.M.); (B.S.)
| | - Raiyan R. Khan
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; (C.A.F.); (Y.K.); (R.R.K.); (L.H.K.); (E.C.M.); (M.D.G.)
| | - Lily H. Kim
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; (C.A.F.); (Y.K.); (R.R.K.); (L.H.K.); (E.C.M.); (M.D.G.)
| | - Faria Zafar
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; (D.C.); (F.Z.); (T.J.M.); (B.S.)
| | - Julien Couthouis
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; (J.C.); (A.D.G.)
| | - Guido A. Davidzon
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Elizabeth C. Mormino
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; (C.A.F.); (Y.K.); (R.R.K.); (L.H.K.); (E.C.M.); (M.D.G.)
| | - Aaron D. Gitler
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; (J.C.); (A.D.G.)
| | - Thomas J. Montine
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; (D.C.); (F.Z.); (T.J.M.); (B.S.)
| | - Birgitt Schüle
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; (D.C.); (F.Z.); (T.J.M.); (B.S.)
| | - Michael D. Greicius
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; (C.A.F.); (Y.K.); (R.R.K.); (L.H.K.); (E.C.M.); (M.D.G.)
| |
Collapse
|
32
|
Usnich T, Hanssen H, Lohmann K, Lohse C, Klein C, Kasten M, Brüggemann N. Pronounced Orthostatic Hypotension in GBA-Related Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:1539-1544. [PMID: 35491800 DOI: 10.3233/jpd-223197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Patients with Parkinson's disease (PD) carrying variants in the Glucocerebrosidase (GBA) gene (GBA-PD) suffer from orthostatic symptoms more frequently than idiopathic PD patients (IPD). Systematic measurements of the blood pressure have not yet been performed. In the present study, a prospective analysis of 33 GBA-PD and 313 IPD patients was carried out. Systolic blood pressure upon changing from the supine to the upright position dropped more strongly in GBA-PD compared to IPD patients. Diastolic blood pressure and heart rate did not differ between groups. This study provides further evidence for a pronounced involvement of the autonomic nervous system in GBA-PD.
Collapse
Affiliation(s)
- Tatiana Usnich
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Henrike Hanssen
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
- Department of Neurology, University of Luebeck, Luebeck, Germany
| | - Katja Lohmann
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Christina Lohse
- Department of Neurology, University of Luebeck, Luebeck, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Meike Kasten
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
- Department of Psychiatry and Psychotherapy, University of Luebeck, Luebeck, Germany
| | - Norbert Brüggemann
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
- Department of Neurology, University of Luebeck, Luebeck, Germany
| |
Collapse
|
33
|
Zhao YW, Pan HX, Liu Z, Wang Y, Zeng Q, Fang ZH, Luo TF, Xu K, Wang Z, Zhou X, He R, Li B, Zhao G, Xu Q, Sun QY, Yan XX, Tan JQ, Li JC, Guo JF, Tang BS. The Association Between Lysosomal Storage Disorder Genes and Parkinson's Disease: A Large Cohort Study in Chinese Mainland Population. Front Aging Neurosci 2021; 13:749109. [PMID: 34867278 PMCID: PMC8634711 DOI: 10.3389/fnagi.2021.749109] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/29/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Recent years have witnessed an increasing number of studies indicating an essential role of the lysosomal dysfunction in Parkinson’s disease (PD) at the genetic, biochemical, and cellular pathway levels. In this study, we investigated the association between rare variants in lysosomal storage disorder (LSD) genes and Chinese mainland PD. Methods: We explored the association between rare variants of 69 LSD genes and PD in 3,879 patients and 2,931 controls from Parkinson’s Disease & Movement Disorders Multicenter Database and Collaborative Network in China (PD-MDCNC) using next-generation sequencing, which were analyzed by using the optimized sequence kernel association test. Results: We identified the significant burden of rare putative LSD gene variants in Chinese mainland patients with PD. This association was robust in familial or sporadic early-onset patients after excluding the GBA variants but not in sporadic late-onset patients. The burden analysis of variant sets in genes of LSD subgroups revealed a suggestive significant association between variant sets in genes of sphingolipidosis deficiency disorders and familial or sporadic early-onset patients. In contrast, variant sets in genes of sphingolipidoses, mucopolysaccharidoses, and post-translational modification defect disorders were suggestively associated with sporadic late-onset patients. Then, SMPD1 and other four novel genes (i.e., GUSB, CLN6, PPT1, and SCARB2) were suggestively associated with sporadic early-onset or familial patients, whereas GALNS and NAGA were suggestively associated with late-onset patients. Conclusion: Our findings supported the association between LSD genes and PD and revealed several novel risk genes in Chinese mainland patients with PD, which confirmed the importance of lysosomal mechanisms in PD pathogenesis. Moreover, we identified the genetic heterogeneity in early-onset and late-onset of patients with PD, which may provide valuable suggestions for the treatment.
Collapse
Affiliation(s)
- Yu-Wen Zhao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hong-Xu Pan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenhua Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yige Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qian Zeng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Zheng-Huan Fang
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Teng-Fei Luo
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Kun Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Zheng Wang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xun Zhou
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Runcheng He
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Bin Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Guihu Zhao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qian Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qi-Ying Sun
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Xin-Xiang Yan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jie-Qiong Tan
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Jin-Chen Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Ji-Feng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Bei-Sha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| |
Collapse
|
34
|
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: 1.3] [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
|
35
|
Day JO, Mullin S. The Genetics of Parkinson's Disease and Implications for Clinical Practice. Genes (Basel) 2021; 12:genes12071006. [PMID: 34208795 PMCID: PMC8304082 DOI: 10.3390/genes12071006] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/21/2021] [Accepted: 06/28/2021] [Indexed: 12/17/2022] Open
Abstract
The genetic landscape of Parkinson’s disease (PD) is characterised by rare high penetrance pathogenic variants causing familial disease, genetic risk factor variants driving PD risk in a significant minority in PD cases and high frequency, low penetrance variants, which contribute a small increase of the risk of developing sporadic PD. This knowledge has the potential to have a major impact in the clinical care of people with PD. We summarise these genetic influences and discuss the implications for therapeutics and clinical trial design.
Collapse
Affiliation(s)
- Jacob Oliver Day
- Faculty of Health, University of Plymouth, Plymouth PL4 8AA, UK;
| | - Stephen Mullin
- Faculty of Health, University of Plymouth, Plymouth PL4 8AA, UK;
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, London WC1N 3BG, UK
- Correspondence:
| |
Collapse
|
36
|
Menozzi E, Schapira AHV. Exploring the Genotype-Phenotype Correlation in GBA-Parkinson Disease: Clinical Aspects, Biomarkers, and Potential Modifiers. Front Neurol 2021; 12:694764. [PMID: 34248830 PMCID: PMC8264189 DOI: 10.3389/fneur.2021.694764] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/18/2021] [Indexed: 01/01/2023] Open
Abstract
Variants in the glucocerebrosidase (GBA) gene are the most common genetic risk factor for Parkinson disease (PD). These include pathogenic variants causing Gaucher disease (GD) (divided into “severe,” “mild,” or “complex”—resulting from recombinant alleles—based on the phenotypic effects in GD) and “risk” variants, which are not associated with GD but nevertheless confer increased risk of PD. As a group, GBA-PD patients have more severe motor and nonmotor symptoms, faster disease progression, and reduced survival compared with noncarriers. However, different GBA variants impact variably on clinical phenotype. In the heterozygous state, “complex” and “severe” variants are associated with a more aggressive and rapidly progressive disease. Conversely, “mild” and “risk” variants portend a more benign course. Homozygous or compound heterozygous carriers usually display severe phenotypes, akin to heterozygous “complex” or “severe” variants carriers. This article reviews genotype–phenotype correlations in GBA-PD, focusing on clinical and nonclinical aspects (neuroimaging and biochemical markers), and explores other disease modifiers that deserve consideration in the characterization of these patients.
Collapse
Affiliation(s)
- Elisa Menozzi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom
| |
Collapse
|
37
|
Ben-Joseph A, Marshall CR, Lees AJ, Noyce AJ. Ethnic Variation in the Manifestation of Parkinson's Disease: A Narrative Review. JOURNAL OF PARKINSONS DISEASE 2021; 10:31-45. [PMID: 31868680 PMCID: PMC7029316 DOI: 10.3233/jpd-191763] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The global prevalence of Parkinson's disease is increasing, yet the characteristics, risk factors and genetics of PD in Black, Asian and Hispanic populations is little understood. In this paper we review the published literature on clinical variation in the symptoms and signs of Parkinson's disease in different ethnic groups and responses to treatment. We included any study that sampled patients with Parkinson's disease from distinct ethnic backgrounds. We conclude that whilst there is little published evidence for ethnic variation in the clinical features of Parkinson's disease, there are substantial limitations and gaps in the current literature, which mean that the evidence does necessarily not fit with clinical observation. Possible explanations for expected differences in manifestation include genetic determinants, the co-existence of cerebrovascular disease and/or Alzheimer's disease pathology, healthcare inequalities and socio-cultural factors.
Collapse
Affiliation(s)
- Aaron Ben-Joseph
- Preventive Neurology Unit, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | - Charles R Marshall
- Preventive Neurology Unit, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | - Andrew J Lees
- Reta Lila Weston Institute of Neurological Studies and Department of Clinical and Movement Neurosciences, University College London, London, UK
| | - Alastair J Noyce
- Preventive Neurology Unit, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK.,Reta Lila Weston Institute of Neurological Studies and Department of Clinical and Movement Neurosciences, University College London, London, UK
| |
Collapse
|
38
|
Biswas A, Sadhukhan D, Biswas A, Das SK, Banerjee TK, Bal PS, Pal S, Ghosh A, Ray K, Ray J. Identification of GBA mutations among neurodegenerative disease patients from eastern India. Neurosci Lett 2021; 751:135816. [PMID: 33711404 DOI: 10.1016/j.neulet.2021.135816] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 03/04/2021] [Accepted: 03/07/2021] [Indexed: 11/18/2022]
Abstract
INTRODUCTION GBA mutations have been reported in PD, PDD and DLB - but not associated with cognitive impairment for example in PSP, AD or MSA. However, frequencies of GBA mutations are ethnicity dependent. The present study aims to identify commonly reported GBA mutations (mostly from Asia), among eastern Indian patients with neurodegenerative disorders. METHODS The patient cohort consisting of 198 classical PD cases, 136 PD cases with cognitive impairment, 184 cases with Parkinson Plus syndrome, 46 AD and 241 unrelated controls, from eastern India. Subjects were analyzed for IVS2 + 1A > G, p.Arg120Trp, p.His255Gln, p.Arg257Gln, p.Glu326Lys, p.Asn370Ser, p.Asp409His, p.Leu444Pro, & RecNciI by PCR-RFLP techniques and confirmed by Sanger sequencing method. RESULTS We have identified only p.Leu444Pro variant among nine cases; three PDD, one DLB, two PD, two PSP and one AD patients in heterozygous condition. The highest frequency for p.Leu444Pro variant was found among PDD subgroup (3.95 %, P = 0.0134). An overall significant overrepresentation of positive family history (P = 0.000049), impaired recent memory (P = 0.0123) was observed among p.Leu444Pro carriers. Further, subgroup analysis for PD, PD-MCI and PDD, revealed statistically significant higher frequency of early age at onset (P = 0.0455), positive family history (P = 0.0025), higher UPDRS III score (off state) (P = 0.006), advanced H&Y stage (P = 0.045) and anxious behaviour (P = 0.0124) among p.Leu444Pro positive patients. CONCLUSION The p.Leu444Pro mutation of GBA was found in patients with PD, PDD, DLB, PSP and AD. An Overall higher frequency of positive family history and impaired recent memory are significantly associated with for p.Leu444Pro carriers from eastern India. Our study also ascertains contribution of p.Leu444Pro to an earlier onset of PD, PD-MCI and PDD, higher UPDRS III score (off state) against positive family history background. Furthermore, taking into consideration other Indian studies, we can conclude that p.Leu444Pro mutation plays a limited role in PD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Arindam Biswas
- Molecular Biology & Clinical Neuroscience Division, National Neurosciences Centre, Calcutta, India; S. N. Pradhan Centre for Neurosciences, University of Calcutta, Kolkata, India.
| | - Dipanwita Sadhukhan
- S. N. Pradhan Centre for Neurosciences, University of Calcutta, Kolkata, India
| | - Atanu Biswas
- Institute of Post graduate of Medical Education & Research and Bangur Institute of Neurosciences, Kolkata, India
| | - Shyamal K Das
- Institute of Post graduate of Medical Education & Research and Bangur Institute of Neurosciences, Kolkata, India
| | - Tapas K Banerjee
- Molecular Biology & Clinical Neuroscience Division, National Neurosciences Centre, Calcutta, India
| | - Partha Sarathi Bal
- Molecular Biology & Clinical Neuroscience Division, National Neurosciences Centre, Calcutta, India
| | - Sandip Pal
- Medical College & Hospitals, Kolkata, India
| | | | - Kunal Ray
- ATGC Diagnostics Private Limited, Kolkata, India
| | - Jharna Ray
- S. N. Pradhan Centre for Neurosciences, University of Calcutta, Kolkata, India.
| |
Collapse
|
39
|
Wen Y, Zhou Y, Jiao B, Shen L. Genetics of Progressive Supranuclear Palsy: A Review. JOURNAL OF PARKINSON'S DISEASE 2021; 11:93-105. [PMID: 33104043 PMCID: PMC7990399 DOI: 10.3233/jpd-202302] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 09/26/2020] [Indexed: 02/06/2023]
Abstract
Progressive supranuclear palsy (PSP) is an atypical parkinsonism with prominent 4R-tau neuropathology, and the classical clinical phenotype is characterized by vertical supranuclear gaze palsy, unprovoked falls, akinetic-rigid syndrome and cognitive decline. Though PSP is generally regarded as sporadic, there is increasing evidence suggesting that a series of common and rare genetic variants impact on sporadic and familial forms of PSP. To date, more than 10 genes have been reported to show a potential association with PSP. Among these genes, the microtubule-associated protein tau (MAPT) is the risk locus with the strongest effect size on sporadic PSP in the case-control genome-wide association studies (GWAS). Additionally, MAPT mutations are the most common cause of familial PSP while the leucine-rich repeat kinase 2 (LRRK2) is a rare monogenic cause of PSP, and several other gene mutations may mimic the PSP phenotype, like the dynactin subunit 1 (DCTN1). In total, 15 MAPT mutations have been identified in cases with PSP, and the mean age at onset is much earlier than in cases carrying LRRK2 or DCTN1 mutations. GWAS have further identified several risk loci of PSP, proposing molecular pathways related to PSP. The present review focused on genetic studies on PSP and summarized genetic factors of PSP, which may help to elucidate the underlying pathogenesis and provide new perspectives for therapeutic strategies.
Collapse
Affiliation(s)
- Yafei Wen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Yafang Zhou
- Department of Geriatrics Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, PR China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, PR China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, PR China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, PR China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan, PR China
| |
Collapse
|
40
|
Straniero L, Asselta R, Bonvegna S, Rimoldi V, Melistaccio G, Soldà G, Aureli M, Della Porta M, Lucca U, Di Fonzo A, Zecchinelli A, Pezzoli G, Cilia R, Duga S. The SPID-GBA study: Sex distribution, Penetrance, Incidence, and Dementia in GBA-PD. NEUROLOGY-GENETICS 2020; 6:e523. [PMID: 33209983 PMCID: PMC7670574 DOI: 10.1212/nxg.0000000000000523] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/31/2020] [Indexed: 11/15/2022]
Abstract
Objective To provide a variant-specific estimate of incidence, penetrance, sex distribution, and association with dementia of the 4 most common Parkinson disease (PD)-associated GBA variants, we analyzed a large cohort of 4,923 Italian unrelated patients with primary degenerative parkinsonism (including 3,832 PD) enrolled in a single tertiary care center and 7,757 ethnically matched controls. Methods The p.E326K, p.T369M, p.N370S, and p.L444P variants were screened using an allele-specific multiplexed PCR approach. All statistical procedures were performed using R or Plink v1.07. Results Among the 4 analyzed variants, the p.L444P confirmed to be the most strongly associated with disease risk for PD, PD dementia (PDD), and dementia with Lewy bodies (DLB) (odds ratio [OR] for PD 15.63, 95% confidence interval [CI] = 8.04-30.37, p = 4.97*10-16; OR for PDD 29.57, 95% CI = 14.07-62.13, p = 3.86*10-19; OR for DLB 102.7, 95% CI = 31.38-336.1, p = 1.91*10-14). However, an unexpectedly high risk for dementia was conferred by p.E326K (OR for PDD 4.80, 95% CI = 2.87-8.02, p = 2.12*10-9; OR for DLB 12.24, 95% CI = 4.95-30.24, p = 5.71*10-8), which, on the basis of the impact on glucocerebrosidase activity, would be expected to be mild. The 1.5-2:1 male sex bias described in sporadic PD was lost in p.T369M carriers. We also showed that PD penetrance for p.L444P could reach the 15% at age 75 years. Conclusions We report a large monocentric study on GBA-PD assessing mutation-specific data on the sex distribution, penetrance, incidence, and association with dementia of the 4 most frequent deleterious variants in GBA.
Collapse
Affiliation(s)
- Letizia Straniero
- Department of Biomedical Sciences (L.S., R.A., V.R., G.M., G.S., M.D.P., S.D.), Humanitas University; Humanitas Clinical and Research Center (R.A., V.R., G.S., M.D.P., S.D.), IRCCS, Rozzano; Fondazione Grigioni per il Morbo di Parkinson (S.B., A.Z., G.P.); Parkinson Institute (S.B., A.Z., G.P., R.C.), ASST "Gaetano Pini-CTO"; Department of Medical Biotechnology and Translational Medicine (M.A.), University of Milan; Laboratory of Geriatric Neuropsychiatry (U.L.), Istituto di Ricerche Farmacologiche Mario Negri IRCCS; IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico (A.D.F.), Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan; and Fondazione IRCCS Istituto Neurologico "Carlo Besta" (R.C.), Milan, Italy
| | - Rosanna Asselta
- Department of Biomedical Sciences (L.S., R.A., V.R., G.M., G.S., M.D.P., S.D.), Humanitas University; Humanitas Clinical and Research Center (R.A., V.R., G.S., M.D.P., S.D.), IRCCS, Rozzano; Fondazione Grigioni per il Morbo di Parkinson (S.B., A.Z., G.P.); Parkinson Institute (S.B., A.Z., G.P., R.C.), ASST "Gaetano Pini-CTO"; Department of Medical Biotechnology and Translational Medicine (M.A.), University of Milan; Laboratory of Geriatric Neuropsychiatry (U.L.), Istituto di Ricerche Farmacologiche Mario Negri IRCCS; IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico (A.D.F.), Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan; and Fondazione IRCCS Istituto Neurologico "Carlo Besta" (R.C.), Milan, Italy
| | - Salvatore Bonvegna
- Department of Biomedical Sciences (L.S., R.A., V.R., G.M., G.S., M.D.P., S.D.), Humanitas University; Humanitas Clinical and Research Center (R.A., V.R., G.S., M.D.P., S.D.), IRCCS, Rozzano; Fondazione Grigioni per il Morbo di Parkinson (S.B., A.Z., G.P.); Parkinson Institute (S.B., A.Z., G.P., R.C.), ASST "Gaetano Pini-CTO"; Department of Medical Biotechnology and Translational Medicine (M.A.), University of Milan; Laboratory of Geriatric Neuropsychiatry (U.L.), Istituto di Ricerche Farmacologiche Mario Negri IRCCS; IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico (A.D.F.), Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan; and Fondazione IRCCS Istituto Neurologico "Carlo Besta" (R.C.), Milan, Italy
| | - Valeria Rimoldi
- Department of Biomedical Sciences (L.S., R.A., V.R., G.M., G.S., M.D.P., S.D.), Humanitas University; Humanitas Clinical and Research Center (R.A., V.R., G.S., M.D.P., S.D.), IRCCS, Rozzano; Fondazione Grigioni per il Morbo di Parkinson (S.B., A.Z., G.P.); Parkinson Institute (S.B., A.Z., G.P., R.C.), ASST "Gaetano Pini-CTO"; Department of Medical Biotechnology and Translational Medicine (M.A.), University of Milan; Laboratory of Geriatric Neuropsychiatry (U.L.), Istituto di Ricerche Farmacologiche Mario Negri IRCCS; IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico (A.D.F.), Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan; and Fondazione IRCCS Istituto Neurologico "Carlo Besta" (R.C.), Milan, Italy
| | - Giada Melistaccio
- Department of Biomedical Sciences (L.S., R.A., V.R., G.M., G.S., M.D.P., S.D.), Humanitas University; Humanitas Clinical and Research Center (R.A., V.R., G.S., M.D.P., S.D.), IRCCS, Rozzano; Fondazione Grigioni per il Morbo di Parkinson (S.B., A.Z., G.P.); Parkinson Institute (S.B., A.Z., G.P., R.C.), ASST "Gaetano Pini-CTO"; Department of Medical Biotechnology and Translational Medicine (M.A.), University of Milan; Laboratory of Geriatric Neuropsychiatry (U.L.), Istituto di Ricerche Farmacologiche Mario Negri IRCCS; IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico (A.D.F.), Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan; and Fondazione IRCCS Istituto Neurologico "Carlo Besta" (R.C.), Milan, Italy
| | - Giulia Soldà
- Department of Biomedical Sciences (L.S., R.A., V.R., G.M., G.S., M.D.P., S.D.), Humanitas University; Humanitas Clinical and Research Center (R.A., V.R., G.S., M.D.P., S.D.), IRCCS, Rozzano; Fondazione Grigioni per il Morbo di Parkinson (S.B., A.Z., G.P.); Parkinson Institute (S.B., A.Z., G.P., R.C.), ASST "Gaetano Pini-CTO"; Department of Medical Biotechnology and Translational Medicine (M.A.), University of Milan; Laboratory of Geriatric Neuropsychiatry (U.L.), Istituto di Ricerche Farmacologiche Mario Negri IRCCS; IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico (A.D.F.), Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan; and Fondazione IRCCS Istituto Neurologico "Carlo Besta" (R.C.), Milan, Italy
| | - Massimo Aureli
- Department of Biomedical Sciences (L.S., R.A., V.R., G.M., G.S., M.D.P., S.D.), Humanitas University; Humanitas Clinical and Research Center (R.A., V.R., G.S., M.D.P., S.D.), IRCCS, Rozzano; Fondazione Grigioni per il Morbo di Parkinson (S.B., A.Z., G.P.); Parkinson Institute (S.B., A.Z., G.P., R.C.), ASST "Gaetano Pini-CTO"; Department of Medical Biotechnology and Translational Medicine (M.A.), University of Milan; Laboratory of Geriatric Neuropsychiatry (U.L.), Istituto di Ricerche Farmacologiche Mario Negri IRCCS; IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico (A.D.F.), Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan; and Fondazione IRCCS Istituto Neurologico "Carlo Besta" (R.C.), Milan, Italy
| | - Matteo Della Porta
- Department of Biomedical Sciences (L.S., R.A., V.R., G.M., G.S., M.D.P., S.D.), Humanitas University; Humanitas Clinical and Research Center (R.A., V.R., G.S., M.D.P., S.D.), IRCCS, Rozzano; Fondazione Grigioni per il Morbo di Parkinson (S.B., A.Z., G.P.); Parkinson Institute (S.B., A.Z., G.P., R.C.), ASST "Gaetano Pini-CTO"; Department of Medical Biotechnology and Translational Medicine (M.A.), University of Milan; Laboratory of Geriatric Neuropsychiatry (U.L.), Istituto di Ricerche Farmacologiche Mario Negri IRCCS; IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico (A.D.F.), Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan; and Fondazione IRCCS Istituto Neurologico "Carlo Besta" (R.C.), Milan, Italy
| | - Ugo Lucca
- Department of Biomedical Sciences (L.S., R.A., V.R., G.M., G.S., M.D.P., S.D.), Humanitas University; Humanitas Clinical and Research Center (R.A., V.R., G.S., M.D.P., S.D.), IRCCS, Rozzano; Fondazione Grigioni per il Morbo di Parkinson (S.B., A.Z., G.P.); Parkinson Institute (S.B., A.Z., G.P., R.C.), ASST "Gaetano Pini-CTO"; Department of Medical Biotechnology and Translational Medicine (M.A.), University of Milan; Laboratory of Geriatric Neuropsychiatry (U.L.), Istituto di Ricerche Farmacologiche Mario Negri IRCCS; IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico (A.D.F.), Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan; and Fondazione IRCCS Istituto Neurologico "Carlo Besta" (R.C.), Milan, Italy
| | - Alessio Di Fonzo
- Department of Biomedical Sciences (L.S., R.A., V.R., G.M., G.S., M.D.P., S.D.), Humanitas University; Humanitas Clinical and Research Center (R.A., V.R., G.S., M.D.P., S.D.), IRCCS, Rozzano; Fondazione Grigioni per il Morbo di Parkinson (S.B., A.Z., G.P.); Parkinson Institute (S.B., A.Z., G.P., R.C.), ASST "Gaetano Pini-CTO"; Department of Medical Biotechnology and Translational Medicine (M.A.), University of Milan; Laboratory of Geriatric Neuropsychiatry (U.L.), Istituto di Ricerche Farmacologiche Mario Negri IRCCS; IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico (A.D.F.), Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan; and Fondazione IRCCS Istituto Neurologico "Carlo Besta" (R.C.), Milan, Italy
| | - Anna Zecchinelli
- Department of Biomedical Sciences (L.S., R.A., V.R., G.M., G.S., M.D.P., S.D.), Humanitas University; Humanitas Clinical and Research Center (R.A., V.R., G.S., M.D.P., S.D.), IRCCS, Rozzano; Fondazione Grigioni per il Morbo di Parkinson (S.B., A.Z., G.P.); Parkinson Institute (S.B., A.Z., G.P., R.C.), ASST "Gaetano Pini-CTO"; Department of Medical Biotechnology and Translational Medicine (M.A.), University of Milan; Laboratory of Geriatric Neuropsychiatry (U.L.), Istituto di Ricerche Farmacologiche Mario Negri IRCCS; IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico (A.D.F.), Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan; and Fondazione IRCCS Istituto Neurologico "Carlo Besta" (R.C.), Milan, Italy
| | - Gianni Pezzoli
- Department of Biomedical Sciences (L.S., R.A., V.R., G.M., G.S., M.D.P., S.D.), Humanitas University; Humanitas Clinical and Research Center (R.A., V.R., G.S., M.D.P., S.D.), IRCCS, Rozzano; Fondazione Grigioni per il Morbo di Parkinson (S.B., A.Z., G.P.); Parkinson Institute (S.B., A.Z., G.P., R.C.), ASST "Gaetano Pini-CTO"; Department of Medical Biotechnology and Translational Medicine (M.A.), University of Milan; Laboratory of Geriatric Neuropsychiatry (U.L.), Istituto di Ricerche Farmacologiche Mario Negri IRCCS; IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico (A.D.F.), Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan; and Fondazione IRCCS Istituto Neurologico "Carlo Besta" (R.C.), Milan, Italy
| | - Roberto Cilia
- Department of Biomedical Sciences (L.S., R.A., V.R., G.M., G.S., M.D.P., S.D.), Humanitas University; Humanitas Clinical and Research Center (R.A., V.R., G.S., M.D.P., S.D.), IRCCS, Rozzano; Fondazione Grigioni per il Morbo di Parkinson (S.B., A.Z., G.P.); Parkinson Institute (S.B., A.Z., G.P., R.C.), ASST "Gaetano Pini-CTO"; Department of Medical Biotechnology and Translational Medicine (M.A.), University of Milan; Laboratory of Geriatric Neuropsychiatry (U.L.), Istituto di Ricerche Farmacologiche Mario Negri IRCCS; IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico (A.D.F.), Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan; and Fondazione IRCCS Istituto Neurologico "Carlo Besta" (R.C.), Milan, Italy
| | - Stefano Duga
- Department of Biomedical Sciences (L.S., R.A., V.R., G.M., G.S., M.D.P., S.D.), Humanitas University; Humanitas Clinical and Research Center (R.A., V.R., G.S., M.D.P., S.D.), IRCCS, Rozzano; Fondazione Grigioni per il Morbo di Parkinson (S.B., A.Z., G.P.); Parkinson Institute (S.B., A.Z., G.P., R.C.), ASST "Gaetano Pini-CTO"; Department of Medical Biotechnology and Translational Medicine (M.A.), University of Milan; Laboratory of Geriatric Neuropsychiatry (U.L.), Istituto di Ricerche Farmacologiche Mario Negri IRCCS; IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico (A.D.F.), Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan; and Fondazione IRCCS Istituto Neurologico "Carlo Besta" (R.C.), Milan, Italy
| |
Collapse
|
41
|
Brockmann K. GBA-Associated Synucleinopathies: Prime Candidates for Alpha-Synuclein Targeting Compounds. Front Cell Dev Biol 2020; 8:562522. [PMID: 33102473 PMCID: PMC7545538 DOI: 10.3389/fcell.2020.562522] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/09/2020] [Indexed: 12/13/2022] Open
Abstract
With disease-modifying compounds targeting alpha-synuclein available in clinical trials, patient stratification according to alpha-synuclein-specific enrichment strategies is a much-needed prerequisite. Such a scenario will be exemplified for GBA, one major genetic risk factor that is specifically associated with the alpha-synucleinopathies: Parkinson's disease and dementia with Lewy bodies.
Collapse
Affiliation(s)
- Kathrin Brockmann
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Disease (DZNE), Bonn, Germany
| |
Collapse
|
42
|
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: 13] [Impact Index Per Article: 2.6] [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
|
43
|
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: 218] [Impact Index Per Article: 43.6] [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
|
44
|
Schierding W, Farrow S, Fadason T, Graham OEE, Pitcher TL, Qubisi S, Davidson AJ, Perry JK, Anderson TJ, Kennedy MA, Cooper A, O'Sullivan JM. Common Variants Coregulate Expression of GBA and Modifier Genes to Delay Parkinson's Disease Onset. Mov Disord 2020; 35:1346-1356. [PMID: 32557794 PMCID: PMC7496525 DOI: 10.1002/mds.28144] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/02/2020] [Accepted: 05/20/2020] [Indexed: 12/29/2022] Open
Abstract
Background GBA mutations are numerically the most significant genetic risk factor for Parkinson's disease (PD), yet these mutations have low penetrance, suggesting additional mechanisms. Objectives The objective of this study was to determine if the penetrance of GBA in PD can be explained by regulatory effects on GBA and modifier genes. Methods Genetic variants associated with the regulation of GBA were identified by screening 128 common single nucleotide polymorphisms (SNPs) in the GBA locus for spatial cis‐expression quantitative trail locus (supported by chromatin interactions). Results We identified common noncoding SNPs within GBA that (1) regulate GBA expression in peripheral tissues, some of which display α‐synuclein pathology and (2) coregulate potential modifier genes in the central nervous system and/or peripheral tissues. Haplotypes based on 3 of these SNPs delay disease onset by 5 years. In addition, SNPs on 6 separate chromosomes coregulate GBA expression specifically in either the substantia nigra or cortex, and their combined effect potentially modulates motor and cognitive symptoms, respectively. Conclusions This work provides a new perspective on the haplotype‐specific effects of GBA and the genetic etiology of PD, expanding the role of GBA from the gene encoding the β‐glucocerebrosidase (GCase) to that of a central regulator and modifier of PD onset, with GBA expression itself subject to distant regulation. Some idiopathic patients might possess insufficient GBA‐encoded GCase activity in the substantia nigra as the result of distant regulatory variants and therefore might benefit from GBA‐targeting therapeutics. The SNPs’ regulatory impacts provide a plausible explanation for the variable phenotypes also observed in GBA‐centric Gaucher's disease and dementia with Lewy bodies. © 2020 The Authors. Movement Disorders published by Wiley Periodicals, LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Sophie Farrow
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Tayaza Fadason
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Oscar E E Graham
- Gene Structure and Function Laboratory, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Toni L Pitcher
- Department of Medicine, University of Otago, Christchurch, New Zealand.,Brain Research New Zealand, The University of Auckland, Auckland, New Zealand.,New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Sara Qubisi
- Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Alan J Davidson
- Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Jo K Perry
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Tim J Anderson
- Department of Medicine, University of Otago, Christchurch, New Zealand.,Brain Research New Zealand, The University of Auckland, Auckland, New Zealand.,New Zealand Brain Research Institute, Christchurch, New Zealand.,Neurology Department, Christchurch Hospital, Canterbury District Health Board, Christchurch, New Zealand
| | - Martin A Kennedy
- Gene Structure and Function Laboratory, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand.,New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Antony Cooper
- Australian Parkinsons Mission, Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Justin M O'Sullivan
- Liggins Institute, The University of Auckland, Auckland, New Zealand.,Brain Research New Zealand, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
45
|
Avenali M, Blandini F, Cerri S. Glucocerebrosidase Defects as a Major Risk Factor for Parkinson's Disease. Front Aging Neurosci 2020; 12:97. [PMID: 32372943 PMCID: PMC7186450 DOI: 10.3389/fnagi.2020.00097] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 03/23/2020] [Indexed: 01/05/2023] Open
Abstract
Heterozygous mutations of the GBA1 gene, encoding for lysosomal enzyme glucocerebrosidase (GCase), occur in a considerable percentage of all patients with sporadic Parkinson's disease (PD), varying between 8% and 12% across the world. Genome wide association studies have confirmed the strong correlation between PD and GBA1 mutations, pointing to this element as a major risk factor for PD, possibly the most important one after age. The pathobiological mechanisms underlying the link between a defective function of GCase and the development of PD are still unknown and are currently the focus of intense investigation in the community of pre-clinical and clinical researchers in the PD field. A major controversy regards the fact that, despite the unequivocal correlation between the presence of GBA1 mutations and the risk of developing PD, only a minority of asymptomatic carriers with GBA1 mutations convert to PD in their lifetime. GBA1 mutations reduce the enzymatic function of GCase, impairing lysosomal efficiency and the cellular ability to dispose of pathological alpha-synuclein. Changes in the cellular lipidic content resulting from the accumulation of glycosphingolipids, triggered by lysosomal dysfunction, may contribute to the pathological modification of alpha-synuclein, due to its ability to interact with cell membrane lipids. Mutant GCase can impair mitochondrial function and cause endoplasmic reticulum stress, thereby impacting on cellular energy production and proteostasis. Importantly, reduced GCase activity is associated with clear activation of microglia, a major mediator of neuroinflammatory response within the brain parenchyma, which points to neuroinflammation as a major consequence of GCase dysfunction. In this present review article, we summarize the current knowledge on the role of GBA1 mutations in PD development and their phenotypic correlations. We also discuss the potential role of the GCase pathway in the search for PD biomarkers that may enable the development of disease modifying therapies. Answering these questions will aid clinicians in offering more appropriate counseling to the patients and their caregivers and provide future directions for PD preclinical research.
Collapse
Affiliation(s)
- Micol Avenali
- Neurorehabilitation Unit, IRCCS Mondino Foundation, Pavia, Italy.,Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Fabio Blandini
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy.,Laboratory of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, Pavia, Italy
| | - Silvia Cerri
- Laboratory of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
46
|
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: 4.8] [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
|
47
|
Genetic analysis of N6-methyladenosine modification genes in Parkinson's disease. Neurobiol Aging 2020; 93:143.e9-143.e13. [PMID: 32371107 DOI: 10.1016/j.neurobiolaging.2020.03.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 03/23/2020] [Accepted: 03/29/2020] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease with a relatively unclear etiology. Previous studies have shown that N6-methyladenosine (m6A) is a vital RNA modification enriched in brain tissue, and that the genes involved in m6A modification are implicated in various neurologic diseases. Here, we conducted a comprehensive genetic analysis using targeted sequencing with molecular inversion probes (MIPs) to identify m6A-modification genes (including METTL3, METTL14, WTAP, FTO, ALKBH5, YTHDF1, YTHDF2, YTHDF3, HNRNPC, and ELAVL1) in a total of 1647 sporadic PD patients and 1372 controls of Han Chinese origin. PD patients were divided into early-onset PD (EOPD) and late-onset PD (LOPD) based on whether the onset of motor symptoms occurred before or after 50 years of age. Rare variants were subjected to gene-based burden tests and common variants were subjected to single-variant association analyses. As a result, we identified 214 rare variants in all 10 m6A-modification genes and 16 common variants in 7 genes. Gene-wise association analyses of rare variants in each m6A-modification gene did not achieved a p value of less than 0.05 in either total cohorts or 2 age groups. In fact, p values greater than 0.05 were found when conducting single-variant association analyses on common variants of these genes between PD and control patients. Our comprehensive analyses of m6A-modification genes suggest that there is no significant association between these 10 m6A-modification genes and the risk of sporadic PD.
Collapse
|
48
|
Johnson PH, Weinreb NJ, Cloyd JC, Tuite PJ, Kartha RV. GBA1 mutations: Prospects for exosomal biomarkers in α-synuclein pathologies. Mol Genet Metab 2020; 129:35-46. [PMID: 31761523 PMCID: PMC7002237 DOI: 10.1016/j.ymgme.2019.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/03/2019] [Accepted: 10/12/2019] [Indexed: 12/13/2022]
Abstract
The discovery that patients with Gaucher Disease (GD), a rare lysosomal storage disorder, were developing symptoms similar to Parkinson's disease (PD) led to investigation of the relationship between the two seemingly unrelated pathologies. GD, an autosomal recessive disorder, is the result of a biallelic mutation in the gene GBA1, which encodes for the enzyme glucocerebrosidase (GCase). Since the observation of its relation to PD, GBA1 mutations have become recognized as the most common genetic risk factor for development of synucleinopathies such as PD and dementia with Lewy bodies. Although the exact mechanism by which GBA1 mutations promote PD is unknown, current understanding suggests that impaired GCase inhibits lysosomal activity and decreases the overall ability of the cell to degrade proteins, specifically the neuronal protein α-synuclein. Decreased elimination of α-synuclein can lead to its abnormal accumulation and aggregation, an important component of PD development. Further understanding of how decreased GCase activity increases risk for α-synuclein pathology can assist with the development of clinical biomarkers for early detection of synucleinopathies, as well as promote novel treatments tailored for people with a GBA1 mutation. Historically, α-synuclein has not been a reliable biomarker for PD. However, recent research on α-synuclein content within exosomes, which are small vesicles released by cells that carry specific cellular cargo, has yielded encouraging results. Moreover, decreased GCase activity has been shown to influence exosomal contents. Exosomes have emerged as a promising new avenue for the identification of novel biomarkers and therapeutic targets aimed at improving neuronal GCase function and limiting the development of synucleinopathies.
Collapse
Affiliation(s)
- Parker H Johnson
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Neal J Weinreb
- Department of Human Genetics and Medicine (Hematology), Leonard Miller School of Medicine of University of Miami, Miami, FL, United States of America
| | - James C Cloyd
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States of America; Department of Neurology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Paul J Tuite
- Department of Neurology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Reena V Kartha
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States of America.
| |
Collapse
|
49
|
Blauwendraat C, Nalls MA, Singleton AB. The genetic architecture of Parkinson's disease. Lancet Neurol 2020; 19:170-178. [PMID: 31521533 PMCID: PMC8972299 DOI: 10.1016/s1474-4422(19)30287-x] [Citation(s) in RCA: 661] [Impact Index Per Article: 132.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/14/2019] [Accepted: 06/19/2019] [Indexed: 12/17/2022]
Abstract
Parkinson's disease is a complex neurodegenerative disorder for which both rare and common genetic variants contribute to disease risk, onset, and progression. Mutations in more than 20 genes have been associated with the disease, most of which are highly penetrant and often cause early onset or atypical symptoms. Although our understanding of the genetic basis of Parkinson's disease has advanced considerably, much remains to be done. Further disease-related common genetic variability remains to be identified and the work in identifying rare risk alleles has only just begun. To date, genome-wide association studies have identified 90 independent risk-associated variants. However, most of them have been identified in patients of European ancestry and we know relatively little of the genetics of Parkinson's disease in other populations. We have a limited understanding of the biological functions of the risk alleles that have been identified, although Parkinson's disease risk variants appear to be in close proximity to known Parkinson's disease genes and lysosomal-related genes. In the past decade, multiple efforts have been made to investigate the genetic architecture of Parkinson's disease, and emerging technologies, such as machine learning, single-cell RNA sequencing, and high-throughput screens, will improve our understanding of genetic risk.
Collapse
Affiliation(s)
- Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA; Data Tecnica International, Glen Echo, MD, USA
| | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
50
|
Ji S, Wang C, Qiao H, Gu Z, Gan-Or Z, Fon EA, Chan P. Decreased Penetrance of Parkinson's Disease in Elderly Carriers of Glucocerebrosidase Gene L444P/R Mutations: A Community-Based 10-Year Longitudinal Study. Mov Disord 2020; 35:672-678. [PMID: 31912918 DOI: 10.1002/mds.27971] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/04/2019] [Accepted: 12/13/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Heterozygous mutations in the glucocerebrosidase gene (GBA) have been shown to be an important genetic risk factor for Parkinson's disease (PD) worldwide. However, the penetrance of GBA heterozygote for L444P, the common mutation for Asian population, is not known in older Chinese people. OBJECTIVES To assess the conversion rate to PD in identified carriers of GBA L444P/R mutations in Chinese community-dwelling older adults. METHODS The GBA gene was sequenced for mutations at position 444 in 8405 people older than 55 years who participated in the Beijing Longitudinal Study on Aging II cohort. Nine subjects were identified as heterozygous carriers of GBA L444P or L444R mutations at baseline and clinically followed up from 2009 to 2019 to investigate their PD conversion, motor and nonmotor symptoms, and change of vesicular monoamine transporter type 2 using tracer of [18 F]9-fluoropropyl-(+)-dihydrotetrabenazine (18 F-DTBZ, also known as 18 F-AV-133). RESULTS Eight heterozygous GBA L444P and 1 L444R mutation carriers were identified without PD at baseline, and none of them developed clinical parkinsonism after a 10-year follow-up. CONCLUSIONS Although GBA mutations may lead to an earlier onset PD, the majority of GBA L444P heterozygotes in older adults may not convert to PD. Further studies are warranted to identify factors that modify the risk of conversion. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Shaozhen Ji
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Chaodong Wang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Clinical and Research Center for Parkinson's Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory for Parkinson's Disease, Parkinson Disease Center of Beijing Institute for Brain Disorders, Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Hongwen Qiao
- Department of Nuclear Medicine, Xuanwu Hospital of Capital Medical University, Beijing, China.,Department of Geriatrics, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Zhuqin Gu
- Clinical and Research Center for Parkinson's Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory for Parkinson's Disease, Parkinson Disease Center of Beijing Institute for Brain Disorders, Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ziv Gan-Or
- Montreal Neurological Institute and Hospital, Department of Neurology & Neurosurgery, McGill University, Montreal, Canada
| | - Edward A Fon
- Montreal Neurological Institute and Hospital, Department of Neurology & Neurosurgery, McGill University, Montreal, Canada
| | - Piu Chan
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Clinical and Research Center for Parkinson's Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory for Parkinson's Disease, Parkinson Disease Center of Beijing Institute for Brain Disorders, Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China.,Department of Geriatrics, Xuanwu Hospital of Capital Medical University, Beijing, China
| |
Collapse
|