1
|
Hui H, Yu Y, Yiwei L, Li Y, Liling X, Dongguang Z. Genetic etiology and clinical features of non-syndromic pediatric obesity in the Chinese population: a large cohort study. BMC Pediatr 2025; 25:358. [PMID: 40329189 DOI: 10.1186/s12887-025-05702-9] [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: 01/03/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND This study aimed to investigate the genetic etiology and clinical features of non-syndromic pediatric obesity in a large Chinese cohort, providing insights into the genetic profile and its correlation with clinical phenotypes. METHODS We enrolled 391 children, aged 7-14 years, diagnosed with non-syndromic pediatric obesity at Jiangxi Provincial Children's Hospital from January 2020 to June 2022. Whole-exome sequencing was employed to identify potential genetic causes, focusing on 79 candidate genes associated with obesity. Multivariate logistic regression analysis was performed on the clinical data of the non-syndromic obesity gene-positive group and the gene-negative group. RESULTS Among the 391 patients, 32 (8.2%) carried 18 non-syndromic obesity genes, with UCP3 and MC4R being the most common. Seven cases (1.8%) were rated as likely pathogenic by the American College of Medical Genetics and Genomics (ACMG). Clinical phenotype and genetic correlation analysis revealed that urinary microalbumin, fT4, GGT, uric acid, serum phosphorus, paternal weight, family history, impaired glucose tolerance (IGT), non-HDL cholesterol (non-HDL-C), and metabolic syndrome (MetS) showed significant statistical differences (P < 0.05). Serum phosphorus is an independent risk factor associated with genetic predispositions to obesity in children and adolescents (P < 0.05). CONCLUSION Our findings highlight the genetic heterogeneity of non-syndromic pediatric obesity and identify UCP3 and MC4R as potential hotspot genes in the Chinese population. The study underscores the potential of genetic testing for early diagnosis and personalized management of pediatric obesity.
Collapse
Affiliation(s)
- Huang Hui
- Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Child Development and Genetics, Jiangxi Provincial Children's Hospital, Nanchang, China
| | - Yang Yu
- Jiangxi Provincial Key Laboratory of Child Development and Genetics, Jiangxi Provincial Children's Hospital, Nanchang, China.
- Department of Endocrinology, Genetics and Metabolism, Jiangxi Provincial Children's Hospital Clinical Medical Research Center of Genetic Metabolic Diseases in Children, Nanchang, China.
| | - Liang Yiwei
- Department of Child Health, Jiangxi Provincial Children's Hospital, Nanchang, China
| | - Yang Li
- Department of Endocrinology, Genetics and Metabolism, Jiangxi Provincial Children's Hospital Clinical Medical Research Center of Genetic Metabolic Diseases in Children, Nanchang, China
| | - Xie Liling
- Department of Endocrinology, Genetics and Metabolism, Jiangxi Provincial Children's Hospital Clinical Medical Research Center of Genetic Metabolic Diseases in Children, Nanchang, China
| | - Zhang Dongguang
- Department of Endocrinology, Genetics and Metabolism, Jiangxi Provincial Children's Hospital Clinical Medical Research Center of Genetic Metabolic Diseases in Children, Nanchang, China
| |
Collapse
|
2
|
Grabowski GA, Kishnani PS, Alcalay RN, Prakalapakorn SG, Rosenbloom BE, Tuason DA, Weinreb NJ. Challenges in Gaucher disease: Perspectives from an expert panel. Mol Genet Metab 2025; 145:109074. [PMID: 40112481 DOI: 10.1016/j.ymgme.2025.109074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/25/2024] [Accepted: 02/25/2025] [Indexed: 03/22/2025]
Abstract
This focused review concentrates on eight topics of high importance for Gaucher disease (GD) clinicians and researchers: 1) The consideration of GD as distinct types rather than a spectrum. A review of the literature clearly supports the view that there are distinct types of GD. Type 1 is characterized by the absence of primary neuronopathic involvement, while types 2 and 3 are characterized by progressive primary neuronopathic disease. 2) Neurologic and neuronopathic manifestations. A growing body of evidence indicates that the peripheral nervous system may be involved in GD type 1 and that there may also be signs and symptoms of central nervous system (CNS) disease in this group. However, GD type 1 is characterized by the absence of primary neuronopathic disease, whereas GD types 2 and 3 are characterized by progressive, albeit variable, primary neuronopathic disease. Abnormalities in saccadic eye movements have been suggested as being diagnostic for neuronopathic GD, but they may also occur in GD type 1 and in other inflammatory diseases. 3) The importance of whole GBA1 sequencing. This approach is superior to exome sequencing because of potential effects of deep intronic variants on gene expression. It also has the capacity to detect variant alleles that might be missed with gene panels. 4) Monoclonal gammopathy of undetermined significance (MGUS). The risks of MGUS, multiple myeloma, and non-Hodgkin's lymphoma are elevated in patients with GD compared to the general population and strong evidence indicates that lyso-Gb1 stimulates the formation of monoclonal immunoglobulins (M-protein) in patients with GD and MGUS. 5) Pulmonary involvement in GD. Pulmonary complications can be identified through spirometry in up to 45 % of patients with GD type 1 and 55 % of those with GD type 3. Limited evidence exists that enzyme replacement therapy (ERT) reduces the severity of these complications in patients with GD type 1. 6) Gaucheromas. These may occur in patients with GD types 1 or 3, but there is little detailed information about their inception, mechanisms underlying growth, cellular organization, and biochemical activities, and no definitive guidance for their management. Gaucheromas behave like benign (i.e. non-metastasizing) neoplasms, and it may be reasonable to classify them as such. 7) Bone and joint involvement. Dual-energy X-ray absorptiometry scans alone are insufficient for monitoring all changes in bone that may occur in patients with GD. Quantitative magnetic resonance imaging (MRI) techniques using Dixon quantitative chemical shift imaging have provided results that correlate with GD severity scores, bone complications, and biomarkers for GD bone involvement. Thoracic kyphosis is a common complication of GD types 1 and 3, and there is very limited information regarding the effects of ERT or substrate synthesis inhibition therapy (SSIT) on this condition. 8) Treatment initiation, selection, combination, and switching. Prompt initiation of treatment in pediatric patients is important as GD can lead to impaired growth, lower peak bone mass, and delayed puberty. These adverse outcomes can often be ameliorated or prevented with timely treatment. Either ERT or eliglustat, a SSIT agent, is suitable as first-line treatment of adults with GD. Studies of switching from ERT to eliglustat, or between different ERT products, have indicated that changing treatment is safe, although efficacy outcomes vary. A critical remaining issue is the lack of treatments capable of reaching the CNS to slow or halt the progression of neuronopathic disease in patients with GD type 2 or 3 and potentially reduce the risk of Parkinson's disease in GD type 1 patients and heterozygotes for GBA1 variants.
Collapse
Affiliation(s)
- Gregory A Grabowski
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH 45267, USA; Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA.
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, 905 Lasalle Street, GSRB1, 4th Floor, Room 4010, Durham, NC 27710, USA.
| | - Roy N Alcalay
- Neurological Institute of New York, Columbia University, 710 West 168th Street, New York, NY 10032, USA.
| | - S Grace Prakalapakorn
- Department of Ophthalmology and Pediatrics, Duke University Medical Center, 2351 Erwin Rd, Box 3802, DUMC, Durham, NC 27705-4699, USA.
| | - Barry E Rosenbloom
- Cedars-Sinai Tower Hematology Oncology Medical Group, 9090 Wilshire Blvd #300, Beverly Hills, CA 90211, USA.
| | - Dominick A Tuason
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, 800 Howard Ave, New Haven, CT 06510, USA.
| | - Neal J Weinreb
- University of Miami UHealth Sylvester Cancer Center Coral Springs, 8170 Royal Palm Blvd, Coral Springs, FL 33065, USA
| |
Collapse
|
3
|
Somerville EN, James A, Beetz C, Schwieger R, Barrel G, Kandaswamy KK, Iurascu MI, Bauer P, Ta M, Iwaki H, Senkevich K, Yu E, Alcalay RN, Gan‐Or Z. Plasma Glucosylceramide Levels Are Regulated by ATP10D and Are Not Involved in Parkinson's Disease Pathogenesis. Ann Neurol 2025; 97:873-878. [PMID: 40022584 PMCID: PMC12010051 DOI: 10.1002/ana.27219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/24/2025] [Accepted: 02/04/2025] [Indexed: 03/03/2025]
Abstract
GBA1 variants and decreased glucocerebrosidase activity are implicated in Parkinson's disease (PD). We investigated the hypothesis that increased levels of glucosylceramide (GlcCer), a main substrate of glucocerebrosidase, are involved in PD pathogenesis. Using multiple genetic methods, we show that ATPase phospholipid transporting 10D (ATP10D), not GBA1, is the main regulator of plasma GlcCer levels, yet it is not involved in PD pathogenesis. Plasma GlcCer levels were associated with PD, but not in a causative manner, and are not predictive of disease status. These results argue against targeting GlcCer in GBA1-PD, and underscore the need to explore alternative mechanisms and biomarkers for PD. ANN NEUROL 2025;97:873-878.
Collapse
Affiliation(s)
- Emma N. Somerville
- The Neuro (Montréal Neurological Institute‐Hospital)McGill UniversityMontréalQCCanada
- Department of Human GeneticsMcGill UniversityMontréalQCCanada
| | | | | | | | | | | | | | | | - Michael Ta
- Laboratory of NeurogeneticsNational Institute on Aging, National Institutes of HealthBethesdaMDUSA
- DataTecnica InternationalWashingtonDCUSA
| | - Hirotaka Iwaki
- Laboratory of NeurogeneticsNational Institute on Aging, National Institutes of HealthBethesdaMDUSA
- DataTecnica InternationalWashingtonDCUSA
| | - Konstantin Senkevich
- The Neuro (Montréal Neurological Institute‐Hospital)McGill UniversityMontréalQCCanada
- Department of Neurology and NeurosurgeryMcGill UniversityMontréalQCCanada
| | - Eric Yu
- The Neuro (Montréal Neurological Institute‐Hospital)McGill UniversityMontréalQCCanada
- Department of Human GeneticsMcGill UniversityMontréalQCCanada
| | - Roy N. Alcalay
- Department of NeurologyColumbia University Irving Medical CenterNew YorkNYUSA
- Tel Aviv Sourasky Medical CenterTel AvivIsrael
- Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Ziv Gan‐Or
- The Neuro (Montréal Neurological Institute‐Hospital)McGill UniversityMontréalQCCanada
- Department of Human GeneticsMcGill UniversityMontréalQCCanada
- Department of Neurology and NeurosurgeryMcGill UniversityMontréalQCCanada
| |
Collapse
|
4
|
Boura I, Poplawska-Domaszewicz K, Limbachiya N, Trivedi D, Batzu L, Chaudhuri KR. Prodromal Parkinson's Disease: A Snapshot of the Landscape. Neurol Clin 2025; 43:209-228. [PMID: 40185519 DOI: 10.1016/j.ncl.2024.12.004] [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: 04/07/2025]
Abstract
Early observations of specific nonmotor and subtle motor symptoms preceding clinical diagnosis of Parkinson's disease (PD) have paved the way for prodromal PD research, significantly propelling our understanding of early, subclinical stages of neurodegeneration. Prodromal PD has emerged as a complex concept with some researchers suggesting that the period before PD onset is divided into the "at-risk," "preclinical," and "prodromal" phases. Advances in genetic, imaging, laboratory, and digital technologies have enabled the identification of pathophysiological patterns and the potential development of diagnostic, progressive, and therapeutic biomarkers, which could lead to early PD detection and intervention.
Collapse
Affiliation(s)
- Iro Boura
- School of Medicine, University of Crete, Heraklion, Greece; Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK.
| | - Karolina Poplawska-Domaszewicz
- Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK; Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland.
| | - Naomi Limbachiya
- Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| | - Dhaval Trivedi
- Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| | - Lucia Batzu
- Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| | - Kallol Ray Chaudhuri
- Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| |
Collapse
|
5
|
Sabir MS, Makarious MB, Huizing M, Gahl WA, Platt FM, Malicdan MCV. Comprehensive analysis of SLC17A5 variants in large European cohorts reveals no association with Parkinson's disease risk. Parkinsonism Relat Disord 2025; 134:107790. [PMID: 40088783 DOI: 10.1016/j.parkreldis.2025.107790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/13/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative disorder characterized by dopaminergic neuron loss and α-synuclein aggregation. Aging is the primary risk factor, with both rare and common genetic variants playing a role. Previous studies have implicated lysosomal storage disorder (LSD)-related genes, including SLC17A5, in PD susceptibility. OBJECTIVE This study aimed to investigate the association of SLC17A5 variants, including rare and common variants and the FSASD-associated p.Arg39Cys missense variant, with PD risk in large European ancestry cohorts. METHODS Rare variant burden analyses were performed at minor allele frequency (MAF) thresholds of ≤1 % and ≤0.1 % in 7,184 PD cases and 51,650 controls using whole-genome and whole-exome sequencing data. Association testing of the p.Arg39Cys variant was conducted across five cohorts, encompassing both Finnish and non-Finnish Europeans. Common variant associations were examined using summary statistics from the largest European GWAS of PD. RESULTS No significant association was observed between rare SLC17A5 variants and PD at either MAF threshold. The p.Arg39Cys variant, though enriched in Finnish Europeans, showed no significant association with PD across several cohorts. Similarly, common SLC17A5 variants (MAF ≥1%) were not associated with PD risk. CONCLUSION Our findings do not support a role for SLC17A5 variants in PD susceptibility. While lysosomal dysfunction is central to PD pathogenesis, its contribution appears pathway-specific, with SLC17A5 unlikely to influence risk. Larger, multiethnic studies and functional analyses are needed to further investigate sialic acid metabolism in PD and related disorders.
Collapse
Affiliation(s)
- Marya S Sabir
- UDP Translational Laboratory, NIH Undiagnosed Diseases Program, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA; NIH Oxford-Cambridge Scholars Program, University of Oxford, Oxford, UK
| | - Mary B Makarious
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA; DataTecnica LLC, Washington, DC, USA
| | - Marjan Huizing
- Human Biochemical Genetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - William A Gahl
- UDP Translational Laboratory, NIH Undiagnosed Diseases Program, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA; Human Biochemical Genetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - May Christine V Malicdan
- UDP Translational Laboratory, NIH Undiagnosed Diseases Program, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA; Human Biochemical Genetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
6
|
Bouhadoun S, Delva A, Schwarzschild MA, Postuma RB. Preparing for Parkinson's disease prevention trials: Current progress and future directions. JOURNAL OF PARKINSON'S DISEASE 2025:1877718X251334050. [PMID: 40289581 DOI: 10.1177/1877718x251334050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
In recent decades, numerous clinical trials have aimed to delay or prevent Parkinson's disease (PD) progression. Despite the theoretical promise and encouraging preclinical data, none have shown clear efficacy in slowing or preventing PD progression, related to several key limitations. Conventional motor and non-motor scales often fall short in detecting early disease changes, while the heterogeneity of PD phenotypes complicates treatment efficacy. The timing of interventions is also critical, as most trials target patients already in advanced stages of neurodegeneration. A deeper understanding of the preclinical phase and the emergence of new pathological frameworks have shifted the focus toward preventing the onset of clinical PD. Recent advances in biomarker research, including tissue, fluid, and imaging markers, are poised to transform PD research by improving patient selection, stratification, and disease progression monitoring. New biologically grounded frameworks for classifying synucleinopathies aim to distinguish biological subtypes from clinical phenotypes, enabling more targeted prevention trials. Successful PD prevention trials will require early enrollment of individuals at the highest risk, employing low-risk personalized interventions, with biomarkers or sensitive clinical markers as endpoints. Early involvement of key stakeholders will be essential to ensure that trials are timely, ethically sound, and aligned with the needs of the PD community.
Collapse
Affiliation(s)
- Sarah Bouhadoun
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montreal, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Aline Delva
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montreal, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Michael A Schwarzschild
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ronald B Postuma
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montreal, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Centre for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Montreal, Canada
- Research Institute of McGill University Health Centre, Montreal, Canada
| |
Collapse
|
7
|
Lange LM, Cerquera-Cleves C, Schipper M, Panagiotaropoulou G, Braun A, Kraft J, Awasthi S, Bell N, Posthuma D, Ripke S, Blauwendraat C, Heilbron K. Prioritizing Parkinson's disease risk genes in genome-wide association loci. NPJ Parkinsons Dis 2025; 11:77. [PMID: 40240380 PMCID: PMC12003903 DOI: 10.1038/s41531-025-00933-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Many drug targets in ongoing Parkinson's disease (PD) clinical trials have strong genetic links. While genome-wide association studies (GWAS) nominate regions associated with disease, pinpointing causal genes is challenging. Our aim was to prioritize additional druggable genes underlying PD GWAS signals. The polygenic priority score (PoPS) integrates genome-wide information from MAGMA gene-level associations and over 57,000 gene-level features. We applied PoPS to East Asian and European PD GWAS data and prioritized genes based on PoPS, distance to the GWAS signal, and non-synonymous credible set variants. We prioritized 46 genes, including well-established PD genes (SNCA, LRRK2, GBA1, TMEM175, VPS13C), genes with strong literature evidence supporting a mechanistic link to PD (RIT2, BAG3, SCARB2, FYN, DYRK1A, NOD2, CTSB, SV2C, ITPKB), and genes relatively unexplored in PD. Many hold potential for drug repurposing or development. We prioritized high-confidence genes with strong links to PD pathogenesis that may represent our next-best candidates for developing disease-modifying therapeutics.
Collapse
Affiliation(s)
- Lara M Lange
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Catalina Cerquera-Cleves
- Neurology Unit, Department of Neurosciences, Hospital Universitario San Ignacio, Bogotá, Colombia
- Centre de recherche du Centre Hospitalier Universitaire de Québec, Axe Neurosciences, Département de Psychiatrie et Neurosciences, Laval University, Québec, QC, Canada
| | | | - Georgia Panagiotaropoulou
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Alice Braun
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Julia Kraft
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Swapnil Awasthi
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Nathaniel Bell
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Danielle Posthuma
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Child and Adolescent Psychiatry and Pediatric Psychology, Section Complex Trait Genetics, Amsterdam Neuroscience, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Stephan Ripke
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Karl Heilbron
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany.
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany.
| |
Collapse
|
8
|
Martín-Bórnez M, Shar N, Nour MA, Murphy D, Elsayed I, Megha SN, Nwaokorie F, Olusanya A, Kuznetsov N, Bandres-Ciga S, Noyce AJ, Iwaki H, Jones L, Gómez-Garre P, Mir P, Periñan MT. Does COMT Play a Role in Parkinson's Disease Susceptibility Across Diverse Ancestral Populations? MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.11.25325572. [PMID: 40297458 PMCID: PMC12036390 DOI: 10.1101/2025.04.11.25325572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Background The catechol-O-methyltransferase (COMT) gene is involved in brain catecholamine metabolism, but its association with Parkinson's disease (PD) risk remains unclear. Objective To investigate the relationship between COMT genetic variants and PD risk across diverse ancestries. Methods We analyzed COMT variants in 2,251 PD patients and 2,835 controls of European descent using whole-genome sequencing from the Accelerating Medicines Partnership-Parkinson Disease (AMP-PD), along with 20,427 PD patients and 11,837 controls from 10 ancestries using genotyping data from the Global Parkinson's Genetics Program (GP2). Results Utilizing the largest case-control datasets to date, no significant enrichment of COMT risk alleles in PD patients was observed across any ancestry group after correcting for multiple testing. Among Europeans, no correlations with cognitive decline, motor function, motor complications, or time to LID onset were observed. Conclusions These findings emphasize the need for larger, diverse cohorts to confirm the role of COMT in PD development and progression.
Collapse
Affiliation(s)
- Miguel Martín-Bórnez
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Nisar Shar
- Department of clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- NED University of Engineering & Technology, Karachi, Pakistan
| | | | - David Murphy
- Department of clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Inas Elsayed
- Faculty of Pharmacy, University of Gezira, Wadmedani, Sudan
- Systems Biology Ireland, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Shri N Megha
- Department of Neurology, National Institute of Mental Health and Neurosciences, India
| | - Francisca Nwaokorie
- Department of Medical Laboratory Science, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, Nigeria
| | - Adedunni Olusanya
- Department of Pharmacology, Therapeutics and Toxicology, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, Nigeria
| | - Nicole Kuznetsov
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica, Washington DC, USA
| | - Sara Bandres-Ciga
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Alastair J Noyce
- Center for Preventive Neurology, Wolfson Institute of Population Health, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Hirotaka Iwaki
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica, Washington DC, USA
| | - Lietsel Jones
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica, Washington DC, USA
| | - Pilar Gómez-Garre
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Maria Teresa Periñan
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
- Center for Preventive Neurology, Wolfson Institute of Population Health, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
9
|
Hall DA, Shulman JM, Singleton A, Bandres Ciga S, S Tosin MH, Ouyang B, Shulman L. Racial Disparities in Parkinson Disease Clinical Phenotype, Management, and Genetics: Protocol for a Prospective Observational Study. JMIR Res Protoc 2025; 14:e60587. [PMID: 40193190 PMCID: PMC12012400 DOI: 10.2196/60587] [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: 05/15/2024] [Revised: 08/16/2024] [Accepted: 09/30/2024] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Parkinson disease (PD) has been described and studied extensively in White populations, with little known about how the disease manifests and progresses in patients from the Black community. Studies investigating disease features in Black populations are uncommon, with some suggesting that the Black population with PD is more disabled and has greater disease severity and different clinical features compared with the White population with PD. These health disparities are likely to influence the quality of care for Black patients with PD. OBJECTIVE This study aimed to investigate the motor and nonmotor symptoms and quality of life in Black and White participants with PD in a case-case design. METHODS This is an observational, prospective, multicenter, case-case design study. Other aims will investigate the management of PD in Black individuals and the presence of shared or unique genetic risk factors among the Black PD population. A total of 400 Black and 200 White participants with PD will be recruited. Data will be collected at 7 US sites and entered into a Research Electronic Data Capture database. Linear multivariate regression analysis will be used, except for comparing PD management, which will be analyzed using the chi-square test or Fisher exact test. Bonferroni correction will be applied. This protocol also describes plans for educational programming for clinicians and patients at the end of the study in partnership with national PD organizations. RESULTS The Rush Institutional Review Board approved the project as the single-site institutional review board in February 2022, and it was funded by the National Institute of Neurological Disorders and Stroke in April 2022. Recruitment began in July 2022. At the time of submission of this manuscript, 131 participants had been recruited. CONCLUSIONS To our knowledge, this is the largest study of PD phenotype and management in Black patients in the United States. The planned collaboration with the Global Parkinson's Genetics Program and PD GENEration will enhance our understanding of genetic risk factors for PD in this understudied population. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/60587.
Collapse
Affiliation(s)
- Deborah A Hall
- Department of Neurological Sciences, Rush University, Chicago, IL, United States
| | - Josh M Shulman
- Departments of Neurology and Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institutes of Aging, National Institutes of Health, Bethesda, MD, United States
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethseda, MD, United States
| | - Sara Bandres Ciga
- Laboratory of Neurogenetics, National Institutes of Aging, National Institutes of Health, Bethesda, MD, United States
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethseda, MD, United States
| | - Michelle Hyczy S Tosin
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States
| | - Bichun Ouyang
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States
| | - Lisa Shulman
- Department of Neurology, University of Maryland School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
10
|
Cummings JL, Teunissen CE, Fiske BK, Le Ber I, Wildsmith KR, Schöll M, Dunn B, Scheltens P. Biomarker-guided decision making in clinical drug development for neurodegenerative disorders. Nat Rev Drug Discov 2025:10.1038/s41573-025-01165-w. [PMID: 40185982 DOI: 10.1038/s41573-025-01165-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 04/07/2025]
Abstract
Neurodegenerative disorders are characterized by complex neurobiological changes that are reflected in biomarker alterations detectable in blood, cerebrospinal fluid (CSF) and with brain imaging. As accessible proxies for processes that are difficult to measure, biomarkers are tools that hold increasingly important roles in drug development and clinical trial decision making. In the past few years, biomarkers have been the basis for accelerated approval of new therapies for Alzheimer disease and amyotrophic lateral sclerosis as surrogate end points reasonably likely to predict clinical benefit.Blood-based biomarkers are emerging for Alzheimer disease and other neurodegenerative disorders (for example, Parkinson disease, frontotemporal dementia), and some biomarkers may be informative across multiple disease states. Collection of CSF provides access to biomarkers not available in plasma, including markers of synaptic dysfunction and neuroinflammation. Molecular imaging is identifying an increasing array of targets, including amyloid plaques, neurofibrillary tangles, inflammation, mitochondrial dysfunction and synaptic density. In this Review, we consider how biomarkers can be implemented in clinical trials depending on their context of use, including providing information on disease risk and/or susceptibility, diagnosis, prognosis, pharmacodynamic outcomes, monitoring, prediction of response to therapy and safety. Informed choice of increasingly available biomarkers and rational deployment in clinical trials support drug development decision making and de-risk the drug development process for neurodegenerative disorders.
Collapse
Affiliation(s)
- Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA.
| | - Charlotte E Teunissen
- Neurochemistry Laboratory and Biobank, Department of Neuroscience, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Brian K Fiske
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | | | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Göteborg, Sweden
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK
| | - Billy Dunn
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Philip Scheltens
- Alzheimer's Center Amsterdam, Amsterdam University Medical Center, Amsterdam, the Netherlands
- EQT Group, Dementia Fund, Stockholm, Sweden
| |
Collapse
|
11
|
Pan L, Yang L, Ding W, Hu Y, Yang W, Wang J, Zhang Z, Fan K, Sun Z, Liang Y, Lin X, Chen J, Zhang Y. Integrated genetic analysis and single cell-RNA sequencing for brain image-derived phenotypes and Parkinson's disease. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111317. [PMID: 40081564 DOI: 10.1016/j.pnpbp.2025.111317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Previous studies have reported Parkinson's disease (PD) patients usually have changes in brain image-derived phenotypes (IDPs). However, the role of genetic factors in their association and biological mechanism remains unclear. We aimed to unveil genetic and biological links between brain IDPs and PD. METHODS Using genome-wide association study (GWAS) summary statistics and single-cell RNA sequencing (scRNA-seq) data, we performed a comprehensive analysis between 624 brain IDPs and PD. The genetic correlations and causality were examined by linkage disequilibrium score regression (LDSC), two-sample bidirectional Mendelian randomization (MR) and meta-analysis. Potential shared genes were identified using MAGMA and PLACO. Finally, pathway enrichment using FUMA and Metascape, and scRNA-seq analysis were performed to determine biological mechanisms and gene expression atlas across various cell types in brain tissue. RESULTS LDSC revealed that 50 brain IDPs were genetically correlated with PD (P < 0.05), in which 5 IDPs, exhibited putative causality on PD through MR (P < 0.05). For instance, we identified that the increased volume of the right thalamus (IVW: OR = 2.08, 95 % CI: 1.33 to 3.25, PFDR = 0.03) was positively correlated with the risk of PD, which was also supported by replicated MR (IVW: OR = 1.63, 95 % CI: 1.17-2.26, PFDR = 0.02) in FinnGen and meta-analysis (OR = 1.78, 95 % CI: 1.36-2.31, PFDR = 5.00 × 10-4). Additionally, we identified 56 unique pleiotropic genes, such as FAM13A, with notable enrichment in neuronal cells. Biological mechanism analysis revealed these genes were enriched in brain tissues and a variety of pathways such as negative regulation of neuron apoptotic processes. CONCLUSION We indicated the shared genetic architecture and biological mechanisms between brain IDPs and PD. These findings might provide insights on the therapeutic intervention and early prediction of PD at the brain imaging level.
Collapse
Affiliation(s)
- Lin Pan
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China
| | - Laiyu Yang
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Weijie Ding
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China
| | - Yongfei Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Dongfeng Road East 651, Guangzhou 510060, China
| | - Wenzhuo Yang
- Department of Neurosurgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jingning Wang
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China
| | - Zhiyun Zhang
- Department of Plastic Surgery, The First Hospital of Jilin University, Changchun 130000, China
| | - Kangli Fan
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China
| | - Zhihui Sun
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China
| | - Yue Liang
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China
| | - Xiaoyue Lin
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China
| | - Jun Chen
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China.
| | - Ying Zhang
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
12
|
Rossi M, Schaake S, Usnich T, Boehm J, Steffen N, Schell N, Krüger C, Gül‐Demirkale T, Bahr N, Kleinz T, Madoev H, Laabs B, Gan‐Or Z, Alcalay RN, Lohmann K, Klein C. Classification and Genotype-Phenotype Relationships of GBA1 Variants: MDSGene Systematic Review. Mov Disord 2025; 40:605-618. [PMID: 39927608 PMCID: PMC12006889 DOI: 10.1002/mds.30141] [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] [Received: 11/13/2024] [Revised: 01/07/2025] [Accepted: 01/23/2025] [Indexed: 02/11/2025] Open
Abstract
Depending on zygosity and the specific change, different variants in the GBA1 gene can cause Parkinson's disease (PD, PARK-GBA1) with reduced penetrance, act as genetic risk factors for PD or parkinsonism, and/or lead to Gaucher's disease (GD). This MDSGene systematic literature review covers 27,963 patients carrying GBA1 variants from 1082 publications with 794 variants, including 13,342 patients with PD or other forms of parkinsonism. It provides a comprehensive overview of demographic, clinical, and genetic findings from an ethnically diverse sample originating from 82 countries across five continents. The most frequent pathogenic or likely pathogenic variants were "N409S" (aka "N370S"; dominating among Jewish and Whites), and "L483P" (aka "L444P"; dominating among Asians and Hispanics), whereas the most common coding risk variants were "E365K" (E326K), and "T408M" (T369M) (both common among Whites). A novel finding is that early-onset PD patients were predominantly of Asian ethnicity, whereas late-onset PD patients were mainly of White ethnicity. Motor cardinal features were similar between PD patients and other forms of parkinsonism, whereas motor complications and non-motor symptoms were more frequently reported in PD patients carrying "severe" variants than in those with "risk" or "mild" variants. Cognitive decline was reported in most patients after surgical treatment, despite achieving a beneficial motor function response. Most GD patients developing PD harbored the "N409S" variant, were of Ashkenazi Jewish ethnicity, and showed a positive response to chronic levodopa treatment. With this review, we start to fill the gaps regarding genotype-phenotype correlations in GBA1 variant carriers, especially concerning PD. © 2025 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Malco Rossi
- Servicio de Movimientos Anormales, Departamento de NeurologíaFleniBuenos AiresArgentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)Buenos AiresArgentina
| | - Susen Schaake
- Institute of NeurogeneticsUniversity of LübeckLübeckGermany
| | - Tatiana Usnich
- Institute of NeurogeneticsUniversity of LübeckLübeckGermany
| | | | - Nina Steffen
- Institute of NeurogeneticsUniversity of LübeckLübeckGermany
| | | | - Clara Krüger
- Institute of NeurogeneticsUniversity of LübeckLübeckGermany
| | - Tuğçe Gül‐Demirkale
- Neurodegeneration Research Laboratory (NDAL), Research Center for Translational Medicine (KUTTAM), School of MedicineKoç UniversityIstanbulTurkey
| | - Natascha Bahr
- Institute of NeurogeneticsUniversity of LübeckLübeckGermany
| | - Teresa Kleinz
- Institute of NeurogeneticsUniversity of LübeckLübeckGermany
| | - Harutyun Madoev
- Institute of Medical Biometry and StatisticsUniversity of LübeckLübeckGermany
| | - Björn‐Hergen Laabs
- Institute of Medical Biometry and StatisticsUniversity of LübeckLübeckGermany
| | - Ziv Gan‐Or
- Department of Neurology and NeurosurgeryMcGill UniversityMontrealQuebecCanada
- Clinical Research Unit, The Neuro (Montreal Neurological Institute‐Hospital)McGill UniversityMontrealQuebecCanada
- Department of Human GeneticsMcGill UniversityMontrealQuebecCanada
| | - Roy N. Alcalay
- Division of Movement DisordersTel Aviv Sourasky Medical CenterTel AvivIsrael
- Columbia University Irving Medical CenterNew York CityNew YorkUSA
| | - Katja Lohmann
- Institute of NeurogeneticsUniversity of LübeckLübeckGermany
| | | |
Collapse
|
13
|
Spargo TP, Sands CF, Juan IR, Mitchell J, Ravanmehr V, Butts JC, De-Paula RB, Kim Y, Hu F, Wang Q, Vitsios D, Garg M, Middleton L, Tyrlik M, Messa M, Del Angel G, Calame DG, Saade H, Robak L, Hollis B, Cuddapah VA, Zoghbi HY, Shulman JM, Petrovski S, Al-Ramahi I, Tachmazidou I, Dhindsa RS. Haploinsufficiency of ITSN1 is associated with a substantial increased risk of Parkinson's disease. Cell Rep 2025; 44:115355. [PMID: 40056900 DOI: 10.1016/j.celrep.2025.115355] [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] [Received: 08/15/2024] [Revised: 12/18/2024] [Accepted: 02/06/2025] [Indexed: 03/10/2025] Open
Abstract
Despite its significant heritability, the genetic basis of Parkinson's disease (PD) remains incompletely understood. Here, in analyzing whole-genome sequence data from 3,809 PD cases and 247,101 controls in the UK Biobank, we discover that protein-truncating variants in ITSN1 confer a substantially increased risk of PD (p = 6.1 × 10-7; odds ratio [95% confidence interval] = 10.5 [5.2, 21.3]). We replicate this association in three independent datasets totaling 8,407 cases and 413,432 controls (combined p = 4.5 × 10-12). Notably, ITSN1 haploinsufficiency has also been associated with autism spectrum disorder, suggesting variable penetrance/expressivity. In Drosophila, we find that loss of the ITSN1 ortholog Dap160 exacerbates α-synuclein-induced neuronal toxicity and motor deficits, and in vitro assays further suggest a physical interaction between ITSN1 and α-synuclein. These results firmly establish ITSN1 as a PD risk gene with an effect size exceeding previously established loci, implicate vesicular trafficking dysfunction in PD pathogenesis, and potentially open new avenues for therapeutic development.
Collapse
Affiliation(s)
- Thomas P Spargo
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Chloe F Sands
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA; Genetics & Genomics Graduate Program, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA
| | - Isabella R Juan
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jonathan Mitchell
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Vida Ravanmehr
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA
| | - Jessica C Butts
- Department of Bioengineering, George R. Brown School of Engineering, Rice University, Houston, TX, USA
| | - Ruth B De-Paula
- Quantitative and Computational Biology Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Youngdoo Kim
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Fengyuan Hu
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Quanli Wang
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Waltham, MA, USA
| | - Dimitrios Vitsios
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Manik Garg
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Lawrence Middleton
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Michal Tyrlik
- Genetics & Genomics Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Mirko Messa
- Translational Genomics, Centre for Genomics Research, Discovery Sciences BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Guillermo Del Angel
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Waltham, MA, USA
| | - Daniel G Calame
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Hiba Saade
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Laurie Robak
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Ben Hollis
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Vishnu A Cuddapah
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Huda Y Zoghbi
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Center for Alzheimer's and Neurodegenerative Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Joshua M Shulman
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Department of Neurology, Baylor College of Medicine, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Center for Alzheimer's and Neurodegenerative Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Slavé Petrovski
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK; Department of Medicine, Austin Health, University of Melbourne, Melbourne, VIC, Australia.
| | - Ismael Al-Ramahi
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Center for Alzheimer's and Neurodegenerative Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Ioanna Tachmazidou
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Ryan S Dhindsa
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
14
|
Lewis SJG, Sue CM, Cooper A, Halliday GM. Future is now: an Australasian perspective on disease-modifying trials in Parkinson's and prodromal disease. BMJ Neurol Open 2025; 7:e001070. [PMID: 40129903 PMCID: PMC11931937 DOI: 10.1136/bmjno-2025-001070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/05/2025] [Indexed: 03/26/2025] Open
Abstract
There is increasing interest in the role of platform trials, where several investigational products targeting disease modification in Parkinson's Disease can be assessed in parallel. Indeed, several initiatives are currently gearing up across North America and Europe to conduct such studies. However, to date, little attention has been paid to ongoing efforts that already exist in Australia and look soon to expand across to New Zealand as part of greater collaboration. This viewpoint will highlight some of these ongoing efforts addressing the challenges and potential solutions for delivering successful studies.
Collapse
Affiliation(s)
- Simon JG Lewis
- Macquarie Medical School, Macquarie University, Sydney, New South Wales, Australia
| | - Carolyn M Sue
- Kinghorn Chair, Neurodegeneration, Neuroscience Research Australia, Randwick, New South Wales, Australia
- Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Antony Cooper
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Glenda M Halliday
- Brain and Mind Centre, Faculty of Medicine and Health, Central Clinical School, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
15
|
Sun Q, Cao Q, Gu Z, He P, Zhu M, Liang X. Causal relationship between hand grip strength and cognition/dementia risk: a Mendelian randomization study. Am J Transl Res 2025; 17:1910-1924. [PMID: 40225993 PMCID: PMC11982891 DOI: 10.62347/lbnh3951] [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: 02/18/2024] [Accepted: 02/09/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Muscle strength positively correlates with cognitive function, with the bidirectional causal link between hand grip strength and cognition posing a significant but incompletely understood public health challenge. This study aimed to explore the causal relationship between hand grip strength and cognition and its effect on dementia. METHODS A two-sample Mendelian randomization analysis used genome-wide significant single nucleotide polymorphisms (SNPs) (P < 5×10-8, at least P < 5×10-6) linked to hand grip strength (right or left), cognition/dementia risk from the IEU Open GWAS project with 42,484 GWAS summary data sets. The primary analysis employed the inverse variance weighted method, while sensitivity analyses were conducted using the weighted mode and MR-Egger. These analyses aimed to assess the causal relationships between hand grip strength and cognition/dementia risk. RESULTS The inverse variance weighted (IVW) analysis indicated a directional positive causal effects of hand grip strength on cognition (Left-hand grip strength on cognitive function (OR (95% Cl): 1.23 (1.02-1.48), P = 0.026)/performance (OR (95% Cl): 1.16 (1.04-1.30), P = 0.009); Right-hand grip strength on cognitive function (OR (95% Cl): 1.23 (1.02-1.48), P = 0.031)/performance (OR (95% Cl): 1.10 (1.02-1.19), P = 0.018), with almost no reverse causality between cognitive function/performance and hand grip strength. Based on the results above, we then researched the directional causal effects of hand grip strength on neurodegenerative diseases (like dementia) with cognitive decline as the main clinical manifestation. However, the IVW methods yielded no evidence to support a causal effect of left-hand grip strength on dementia (P > 0.05). CONCLUSIONS This MR study indicates a positive directional causal relationship between hand grip strength and cognition, with no observed causal link to dementia. These results hold implications for the development of public health measures and strategies for preventing cognitive decline.
Collapse
Affiliation(s)
- Qian Sun
- Department of Anesthesiology, Jiangnan University Medical Center (Wuxi No. 2 People’s Hospital, Wuxi Clinical College of Nantong University)Wuxi 214000, Jiangsu, China
| | - Qian Cao
- Department of Anesthesiology, Jiangnan University Medical Center (Wuxi No. 2 People’s Hospital, Wuxi Clinical College of Nantong University)Wuxi 214000, Jiangsu, China
| | - Zhen Gu
- Department of Anesthesiology, Jiangnan University Medical Center (Wuxi No. 2 People’s Hospital, Wuxi Clinical College of Nantong University)Wuxi 214000, Jiangsu, China
| | - Peicheng He
- Department of Neurology, Jiangnan University Medical Center (Wuxi No. 2 People’s Hospital, Wuxi Clinical College of Nantong University)Wuxi 214000, Jiangsu, China
| | - Minmin Zhu
- Department of Anesthesiology, Jiangnan University Medical Center (Wuxi No. 2 People’s Hospital, Wuxi Clinical College of Nantong University)Wuxi 214000, Jiangsu, China
| | - Xiao Liang
- Department of Anesthesiology, Jiangnan University Medical Center (Wuxi No. 2 People’s Hospital, Wuxi Clinical College of Nantong University)Wuxi 214000, Jiangsu, China
| |
Collapse
|
16
|
Siddiqi S, Ortiz Z, Simard S, Li J, Lawrence K, Redmond M, Tomlinson JJ, Schlossmacher MG, Salmaso N. Race and ethnicity matter! Moving Parkinson's risk research towards diversity and inclusiveness. NPJ Parkinsons Dis 2025; 11:45. [PMID: 40050644 PMCID: PMC11885646 DOI: 10.1038/s41531-025-00891-7] [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: 02/29/2024] [Accepted: 02/21/2025] [Indexed: 03/09/2025] Open
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disorder that shows considerable heterogeneity of risk factors however, the degree to which race/ethnicity has been actively pursued in PD risk research is unknown. We examined PD literature from 2000-24 and found that less than half accounted for race/ethnicity and only 4.8% of n = 1142 articles included ethno-racial factors as an integral part of the analysis. This demonstrates that race/ethnicity has been critically understudied in PD and further studies that examine ethno-racial contributions to risk for PD are warranted.
Collapse
Affiliation(s)
- Sara Siddiqi
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Zoe Ortiz
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Stephanie Simard
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Juan Li
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Neuroscience Program, Ottawa Hospital Research Institute, University of Ottawa Brain & Mind Research Institute, Ottawa, ON, Canada
| | - Kamaya Lawrence
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Melissa Redmond
- School of Social Work, Carleton University, Ottawa, ON, Canada
| | - Julianna J Tomlinson
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | - Michael G Schlossmacher
- School of Social Work, Carleton University, Ottawa, ON, Canada
- Division of Neurology, The Ottawa Hospital, Ottawa, ON, Canada
| | - Natalina Salmaso
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada.
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada.
| |
Collapse
|
17
|
Akçimen F, Paquette K, Crea PW, Saffie-Awad P, Achoru C, Taiwo F, Ozomma S, Onwuegbuzie G, Khani M, Grant S, Owolabi L, Okereke C, Oshinaike O, Iwuozo E, Lee PS, Oyakhire S, Osemwegie N, Daida K, Abubakar S, Olusanya A, Isayan M, Traurig R, Ogunmodede A, Samuel S, Makarious MB, Sa’ad F, Olanigan R, Levine K, Ogbimi EM, Vitale D, Odiase F, Koretsky MJ, Ojini F, Odeniyi O, Fang ZH, Obianozie N, Hall DA, Nwazor E, Xie T, Nwaokorie F, Padmanaban M, Nwani P, Shamim EA, Nnama A, Standaert D, Komolafe M, Dean M, Osaigbovo G, Disbrow E, Ishola I, Rawls A, Imarhiagbe F, Chandra S, Erameh C, Hinson V, Louie N, Idowu A, Solle J, Norris SA, Ibrahim A, Kilbane C, Sukumar G, Shulman LM, Ezuduemoih D, Staisch J, Breaux S, Dalgard C, Foster ER, Bello A, Ameri A, Real R, Ikwenu E, Morris HR, Anyanwu R, Stimming EF, Billingsley K, Alaofin W, Jerez PA, Agabi O, Hernandez DG, Akinyemi R, Arepalli S, Malik L, Owolabi R, Nyandaiti Y, Leonard HL, Wahab K, Step K, Abiodun O, Hernandez CF, Abdulai F, Iwaki H, Bardien S, Klein C, Hardy J, Houlden H, Galvelis KG, Nalls MA, Dahodwala N, Aamodt W, et alAkçimen F, Paquette K, Crea PW, Saffie-Awad P, Achoru C, Taiwo F, Ozomma S, Onwuegbuzie G, Khani M, Grant S, Owolabi L, Okereke C, Oshinaike O, Iwuozo E, Lee PS, Oyakhire S, Osemwegie N, Daida K, Abubakar S, Olusanya A, Isayan M, Traurig R, Ogunmodede A, Samuel S, Makarious MB, Sa’ad F, Olanigan R, Levine K, Ogbimi EM, Vitale D, Odiase F, Koretsky MJ, Ojini F, Odeniyi O, Fang ZH, Obianozie N, Hall DA, Nwazor E, Xie T, Nwaokorie F, Padmanaban M, Nwani P, Shamim EA, Nnama A, Standaert D, Komolafe M, Dean M, Osaigbovo G, Disbrow E, Ishola I, Rawls A, Imarhiagbe F, Chandra S, Erameh C, Hinson V, Louie N, Idowu A, Solle J, Norris SA, Ibrahim A, Kilbane C, Sukumar G, Shulman LM, Ezuduemoih D, Staisch J, Breaux S, Dalgard C, Foster ER, Bello A, Ameri A, Real R, Ikwenu E, Morris HR, Anyanwu R, Stimming EF, Billingsley K, Alaofin W, Jerez PA, Agabi O, Hernandez DG, Akinyemi R, Arepalli S, Malik L, Owolabi R, Nyandaiti Y, Leonard HL, Wahab K, Step K, Abiodun O, Hernandez CF, Abdulai F, Iwaki H, Bardien S, Klein C, Hardy J, Houlden H, Galvelis KG, Nalls MA, Dahodwala N, Aamodt W, Hill E, Espay A, Factor S, Branson C, Blauwendraat C, Singleton AB, Ojo O, Chahine LM, Okubadejo N, Bandres-Ciga S. Large-scale genetic characterization of Parkinson's disease in the African and African admixed populations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.14.25320205. [PMID: 39867380 PMCID: PMC11759243 DOI: 10.1101/2025.01.14.25320205] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Elucidating the genetic contributions to Parkinson's disease (PD) etiology across diverse ancestries is a critical priority for the development of targeted therapies in a global context. We conducted the largest sequencing characterization of potentially disease-causing, protein-altering and splicing mutations in 710 cases and 11,827 controls from genetically predicted African or African admixed ancestries. We explored copy number variants (CNVs) and runs of homozygosity (ROHs) in prioritized early onset and familial cases. Our study identified rare GBA1 coding variants to be the most frequent mutations among PD patients, with a frequency of 4% in our case cohort. Out of the 18 GBA1 variants identified, ten were previously classified as pathogenic or likely pathogenic, four were novel, and four were reported as of uncertain clinical significance. The most common known disease-associated GBA1 variants in the Ashkenazi Jewish and European populations, p.Asn409Ser, p.Leu483Pro, p.Thr408Met, and p.Glu365Lys, were not identified among the screened PD cases of African and African admixed ancestry. Similarly, the European and Asian LRRK2 disease-causing mutational spectrum, including LRRK2 p.Gly2019Ser and p.Gly2385Arg genetic risk factors, did not appear to play a major role in PD etiology among West African-ancestry populations. However, we found three heterozygous novel missense LRRK2 variants of uncertain significance overrepresented in cases, two of which - p.Glu268Ala and p.Arg1538Cys - had a higher prevalence in the African ancestry population reference datasets. Structural variant analyses revealed the presence of PRKN CNVs with a frequency of 0.7% in African and African admixed cases, with 66% of CNVs detected being compound heterozygous or homozygous in early-onset cases, providing further insights into the genetic underpinnings in early-onset juvenile PD in these populations. Novel genetic variation overrepresented in cases versus controls among screened genes warrants further replication and functional prioritization to unravel their pathogenic potential. Here, we created the most comprehensive genetic catalog of both known and novel coding and splicing variants potentially linked to PD etiology in an underserved population. Our study has the potential to guide the development of targeted therapies in the emerging era of precision medicine. By expanding genetics research to involve underrepresented populations, we hope that future PD treatments are not only effective but also inclusive, addressing the needs of diverse ancestral groups.
Collapse
Affiliation(s)
- Fulya Akçimen
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Kimberly Paquette
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Peter Wild Crea
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Paula Saffie-Awad
- Programa de Pós-Graduação em Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Clínica Santa María, Santiago, Chile
| | - Charles Achoru
- Jos University Teaching Hospital, Jos, Plateau State, Nigeria
| | | | - Simon Ozomma
- University of Calabar Teaching Hospital, Calabar, Cross River State, Nigeria
| | | | - Marzieh Khani
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Spencer Grant
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | | | - Chiamaka Okereke
- University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu State, Nigeria
| | | | | | - Paul Suhwan Lee
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Kensuke Daida
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Sani Abubakar
- Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Adedunni Olusanya
- College of Medicine, University of Lagos, Idi-araba, Lagos State, Nigeria
- R-Jolad Hospital, Gbagada, Lagos, Nigeria
| | - Mariam Isayan
- Department of Neurology and Neurosurgery, National Institute of Health, Yerevan, Armenia
| | - Rami Traurig
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | | | - Sarah Samuel
- University of Maiduguri Teaching Hospital, Maiduguri, Borno State
| | - Mary B. Makarious
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- DataTecnica LLC, Washington, DC, USA
| | | | - Rashidat Olanigan
- Lagos State University Teaching Hospital, Ikeja, Lagos State, Nigeria
| | - Kristin Levine
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- DataTecnica LLC, Washington, DC, USA
| | | | - Dan Vitale
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- DataTecnica LLC, Washington, DC, USA
| | | | - Mathew J. Koretsky
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- DataTecnica LLC, Washington, DC, USA
| | - Francis Ojini
- College of Medicine, University of Lagos, Idi-araba, Lagos State, Nigeria
- Lagos University Teaching Hospital, Idi-araba, Lagos State, Nigeria
| | | | - Zih-Hua Fang
- German Center for Neurodegenerative Diseases, DZNE, Tübingen, Germany
| | - Nkechi Obianozie
- University of Abuja Teaching Hospital, Gwagwalada, Federal Capital Territory, Nigeria
| | - Deborah A. Hall
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Ernest Nwazor
- Rivers State University Teaching Hospital, Port Harcourt, Rivers State, Nigeria
| | - Tao Xie
- University of Chicago Medicine, Department of Neurology, Chicago, USA
| | | | - Mahesh Padmanaban
- University of Chicago Medicine, Department of Neurology, Chicago, USA
| | - Paul Nwani
- Nnamdi Azikiwe University Teaching Hospital, Nnewi, Anambra State, Nigeria
| | - Ejaz A. Shamim
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Mid-Atlantic Permanente Medical Group, Department of Neurology, Largo, MD, USA
- Kaiser Permanente, MidAtlantic Permanente Research Institute, Washington, DC, USA
| | - Alero Nnama
- University of Port Harcourt Teaching Hospital, Rivers State, Nigeria
| | - David Standaert
- University of Alabama at Birmingham, Department of Neurology, Birmingham, USA
| | | | - Marissa Dean
- University of Alabama at Birmingham, Department of Neurology, Birmingham, USA
| | | | - Elizabeth Disbrow
- Department of Neurology, LSU Health Shreveport, LSU Health Shreveport Center for Brain Health, Shreveport, USA
| | - Ismaila Ishola
- College of Medicine, University of Lagos, Idi-araba, Lagos State, Nigeria
| | - Ashley Rawls
- University of Florida Norman Fixel Institute for Neurological Diseases, Neurology Movement Disorders, Gainesville, USA
| | | | - Shivika Chandra
- The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Cyril Erameh
- Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Vanessa Hinson
- Medical University of South Carolina, Charleston, SC, USA
| | - Naomi Louie
- Michael J. Fox Foundation for Parkinson’s Research, Department of Clinical Research, New York, USA
| | - Ahmed Idowu
- Obafemi Awolowo University Teaching Hospitals Complex, Ile-Ife, Osun State, Nigeria
| | - J Solle
- Michael J. Fox Foundation for Parkinson’s Research, Department of Clinical Research, New York, USA
| | | | - Abdullahi Ibrahim
- Federal University of Health Sciences Teaching Hospital, Azare, Bauchi State, Nigeria
| | - Camilla Kilbane
- University Hospital in Cleveland Medical Center/Case Western Reserve University (UH)
| | - Gauthaman Sukumar
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services
- University of the Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA
| | | | | | | | | | - Clifton Dalgard
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The American Genome Center, Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | | | - Abiodun Bello
- University of Ilorin Teaching Hospital, Ilorin, Kwara State, Nigeria
| | - Andrew Ameri
- Medical University of South Carolina, Charleston, SC, USA
| | - Raquel Real
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- UCL Movement Disorders Centre, University College London, London WC1N 3BG, UK
| | - Erica Ikwenu
- Lagos University Teaching Hospital, Idi-araba, Lagos State, Nigeria
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- National Hospital for Neurology and Neurosurgery, London, UK
- Department of Neurology, Royal Free Hospital, London, UK
| | - Roosevelt Anyanwu
- College of Medicine, University of Lagos, Idi-araba, Lagos State, Nigeria
| | - Erin Furr Stimming
- The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kimberley Billingsley
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | | | - Pilar Alvarez Jerez
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Osigwe Agabi
- College of Medicine, University of Lagos, Idi-araba, Lagos State, Nigeria
- Lagos University Teaching Hospital, Idi-araba, Lagos State, Nigeria
| | - Dena G. Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Rufus Akinyemi
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Sampath Arepalli
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Laksh Malik
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | | | - Yakub Nyandaiti
- University of Maiduguri Teaching Hospital, Maiduguri, Borno State
| | - Hampton L. Leonard
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- DataTecnica LLC, Washington, DC, USA
| | | | - Kathryn Step
- Department of Biomedical Sciences, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | | | - Carlos F. Hernandez
- Universidad del Desarrollo, Centro de Genética y Genómica, Facultad de Medicina Clínica Alemana, Santiago 7610658, Chile
| | - Fatima Abdulai
- University of Abuja Teaching Hospital, Gwagwalada, Federal Capital Territory, Nigeria
| | - Hirotaka Iwaki
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- DataTecnica LLC, Washington, DC, USA
| | - Soraya Bardien
- Department of Biomedical Sciences, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Christine Klein
- Institute of Neurogenetics and Department of Neurology, University of Lübeck and University Hospital Schleswig-Holstein, Lübeck, Germany
| | - John Hardy
- Reta Lila Weston Institute, University College London Institute of Neurology, Queen Square, London, UK
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | | | - Mike A. Nalls
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- DataTecnica LLC, Washington, DC, USA
| | | | | | - Emily Hill
- University of Cincinnati, Cincinnati, Ohio
| | | | | | | | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Andrew B. Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Oluwadamilola Ojo
- College of Medicine, University of Lagos, Idi-araba, Lagos State, Nigeria
- Lagos University Teaching Hospital, Idi-araba, Lagos State, Nigeria
| | - Lana M. Chahine
- University of Pittsburgh, 3471 Fifth Avenue, Pittsburgh, PA, 15213, USA
| | | | | | | | | | - Njideka Okubadejo
- College of Medicine, University of Lagos, Idi-araba, Lagos State, Nigeria
- Lagos University Teaching Hospital, Idi-araba, Lagos State, Nigeria
| | - Sara Bandres-Ciga
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
18
|
Vitale D, Koretsky MJ, Kuznetsov N, Hong S, Martin J, James M, Makarious MB, Leonard H, Iwaki H, Faghri F, Blauwendraat C, Singleton AB, Song Y, Levine K, Kumar-Sreelatha AA, Fang ZH, Nalls M. GenoTools: an open-source Python package for efficient genotype data quality control and analysis. G3 (BETHESDA, MD.) 2025; 15:jkae268. [PMID: 39566101 PMCID: PMC11708233 DOI: 10.1093/g3journal/jkae268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/20/2024] [Accepted: 10/14/2024] [Indexed: 11/22/2024]
Abstract
GenoTools, a Python package, streamlines population genetics research by integrating ancestry estimation, quality control, and genome-wide association studies capabilities into efficient pipelines. By tracking samples, variants, and quality-specific measures throughout fully customizable pipelines, users can easily manage genetics data for large and small studies. GenoTools' "Ancestry" module renders highly accurate predictions, allowing for high-quality ancestry-specific studies, and enables custom ancestry model training and serialization specified to the user's genotyping or sequencing platform. As the genotype processing engine that powers several large initiatives, including the NIH's Center for Alzheimer's and Related Dementias and the Global Parkinson's Genetics Program, GenoTools was used to process and analyze the UK Biobank and major Alzheimer's disease and Parkinson's disease datasets with over 400,000 genotypes from arrays and 5,000 whole genome sequencing samples and has led to novel discoveries in diverse populations. It has provided replicable ancestry predictions, implemented rigorous quality control, and conducted genetic ancestry-specific genome-wide association studies to identify systematic errors or biases through a single command. GenoTools is a customizable tool that enables users to efficiently analyze and scale genotyping and sequencing (whole genome sequencing and exome) data with reproducible and scalable ancestry, quality control, and genome-wide association studies pipelines.
Collapse
Affiliation(s)
- Dan Vitale
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- DataTecnica LLC., Washington, DC 20037, USA
| | - Mathew J Koretsky
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- DataTecnica LLC., Washington, DC 20037, USA
| | - Nicole Kuznetsov
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Samantha Hong
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jessica Martin
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mikayla James
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mary B Makarious
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- DataTecnica LLC., Washington, DC 20037, USA
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Hampton Leonard
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- DataTecnica LLC., Washington, DC 20037, USA
| | - Hirotaka Iwaki
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- DataTecnica LLC., Washington, DC 20037, USA
| | - Faraz Faghri
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- DataTecnica LLC., Washington, DC 20037, USA
| | - Cornelis Blauwendraat
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew B Singleton
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yeajin Song
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- DataTecnica LLC., Washington, DC 20037, USA
| | - Kristin Levine
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- DataTecnica LLC., Washington, DC 20037, USA
| | - Ashwin Ashok Kumar-Sreelatha
- Centre for Genetic Epidemiology, Institute for Clinical Epidemiology and Applied Biometry, University of Tübingen, Tübingen 72074, Germany
| | - Zih-Hua Fang
- German Center for Neurodegenerative Diseases (DZNE), Tübingen 72076, Germany
| | - Mike Nalls
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- DataTecnica LLC., Washington, DC 20037, USA
| |
Collapse
|
19
|
Step K, Eltaraifee E, Elsayed I, Rasaholiarison N, Okubadejo N, Walker R, Mohamed W, Rizig M, Bandres‐Ciga S, Noyce AJ, Dey S, Bardien S, Periñan MT. Advancing Parkinson's Disease Research in Africa: A Strategic Training Framework of the Global Parkinson's Genetics Program. Mov Disord 2025; 40:51-56. [PMID: 39482233 PMCID: PMC11752974 DOI: 10.1002/mds.30051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 11/03/2024] Open
Affiliation(s)
- Kathryn Step
- Department of Biomedical Sciences, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Esraa Eltaraifee
- Department of Molecular BiologyInstitute of Endemic DiseasesKhartoumSudan
| | - Inas Elsayed
- Faculty of Pharmacy University of GeziraWad MedaniSudan
- Systems Biology Ireland, School of Medicine and Medical ScienceUniversity College DublinDublinIreland
| | | | - Njideka Okubadejo
- Neurology Unit, Department of Medicine, College of MedicineUniversity of LagosLagosNigeria
| | - Richard Walker
- Population Health Sciences InstituteNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Wael Mohamed
- Basic Medical Science Department, Kulliyyah of MedicineInternational Islamic University MalaysiaKuantanMalaysia
- Clinical Pharmacology Department, Menoufia Medical SchoolMenoufia UniversityShebin El‐KomEgypt
| | - Mie Rizig
- Department of Neuromuscular DiseasesUCL Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Sara Bandres‐Ciga
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
| | - Alastair J. Noyce
- Centre for Preventive Neurology, Wolfson Institute of Population HealthQueen Mary University of LondonLondonUnited Kingdom
| | - Sumit Dey
- Centre for Preventive Neurology, Wolfson Institute of Population HealthQueen Mary University of LondonLondonUnited Kingdom
| | | | - Soraya Bardien
- Department of Biomedical Sciences, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences and South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research UnitStellenbosch UniversityCape TownSouth Africa
| | - Maria Teresa Periñan
- Centre for Preventive Neurology, Wolfson Institute of Population HealthQueen Mary University of LondonLondonUnited Kingdom
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de SevillaHospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de SevillaSevilleSpain
| |
Collapse
|
20
|
Guedi AB, Ikram S, Youssef A, Alya G, Amira S, Saloua M, Amina N, Mouna BD, Imen K, Amina GB, Riadh G. Glutathione S-transferase polymorphisms (GSTM1/GSTT1) outcomes in clinical profile and treatment responsiveness among Tunisian cohort of Parkinson's disease. J Neural Transm (Vienna) 2025; 132:117-127. [PMID: 39123072 DOI: 10.1007/s00702-024-02815-w] [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] [Received: 05/15/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024]
Abstract
Glutathione S-transferases are involved in the oxidative stress which contributes to the pathogenesis of Parkinson's disease (PD). our aim was to investigate the influence of GSTM1 and GSTT1 polymorphisms on the clinical features and treatments outcomes among PD Tunisian patients. We included 300-PD patients followed in neurology department at Razi-University-hospital. GSTM1 and GSTT1 were screened using PCR methods. Correlation between the clinical phenotype and the genotypes was then assessed after adequate parameters adjustment. Individuals carrying inactive GSTT1/GSTM1 were estimated to have 2.5-fold higher risk of developing PD, p = 0.035. The demographic and clinical baseline analysis of GSTM1 polymorphism revealed significant association between the inactive gene and development of tremor as first symptoms (p = 0.046), further, it was correlated to asymmetric start (p = 0.044). The evaluation of the impact of GSTM1/GSTT1 activity among PD at last follow-up revealed the significant variability of motor impairment among cases carrier of the active genes (p = 0.048). As patients with inactive GSTM1/GSTT1 had higher UPDRS-III score. Additionally, higher frequency of cases with good treatment responsiveness was reported among PD with active GSTM1/GSTT1 (p = 0.038).No motor complications were observed among PD by considering the GSTs genotypes (p > 0.05). Finally, we noted significant impairment of memory among cases with inactivate GSTs (p = 0.04), attention deficit (p = 0.013) and impaired judgement (p = 0.0031). This study represents one of the most comprehensive and extensive investigation to date regarding the influence of GSTT1/GSTM1 genotype among PD patients.We speculate that the impact of GSTT1/GSTM1 on PD progression may occur through a cumulative effect, potentially not manifesting during the initial PD stages. Further studies are necessary to validate our conclusions.
Collapse
Affiliation(s)
- Ali Barreh Guedi
- Neurology Department LR18SP03, Razi University Hospital, 1 rue des Orangers Manouba, Tunis, PC: 2010, Tunisia
- Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des Orangers Manouba, Tunis, 2010, Tunisia
| | - Sghaier Ikram
- Neurology Department LR18SP03, Razi University Hospital, 1 rue des Orangers Manouba, Tunis, PC: 2010, Tunisia
- Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des Orangers Manouba, Tunis, 2010, Tunisia
| | - Abida Youssef
- Neurology Department LR18SP03, Razi University Hospital, 1 rue des Orangers Manouba, Tunis, PC: 2010, Tunisia
- Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Akhdhar, La Rabta, Tunis, 1007, Tunisia
- Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des Orangers Manouba, Tunis, 2010, Tunisia
| | - Gharbi Alya
- Neurology Department LR18SP03, Razi University Hospital, 1 rue des Orangers Manouba, Tunis, PC: 2010, Tunisia
- Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Akhdhar, La Rabta, Tunis, 1007, Tunisia
- Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des Orangers Manouba, Tunis, 2010, Tunisia
| | - Souissi Amira
- Neurology Department LR18SP03, Razi University Hospital, 1 rue des Orangers Manouba, Tunis, PC: 2010, Tunisia
- Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Akhdhar, La Rabta, Tunis, 1007, Tunisia
- Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des Orangers Manouba, Tunis, 2010, Tunisia
| | - Mrabet Saloua
- Neurology Department LR18SP03, Razi University Hospital, 1 rue des Orangers Manouba, Tunis, PC: 2010, Tunisia
- Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Akhdhar, La Rabta, Tunis, 1007, Tunisia
- Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des Orangers Manouba, Tunis, 2010, Tunisia
| | - Nasri Amina
- Neurology Department LR18SP03, Razi University Hospital, 1 rue des Orangers Manouba, Tunis, PC: 2010, Tunisia
- Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Akhdhar, La Rabta, Tunis, 1007, Tunisia
- Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des Orangers Manouba, Tunis, 2010, Tunisia
| | - Ben Djebara Mouna
- Neurology Department LR18SP03, Razi University Hospital, 1 rue des Orangers Manouba, Tunis, PC: 2010, Tunisia
- Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Akhdhar, La Rabta, Tunis, 1007, Tunisia
- Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des Orangers Manouba, Tunis, 2010, Tunisia
| | - Kacem Imen
- Neurology Department LR18SP03, Razi University Hospital, 1 rue des Orangers Manouba, Tunis, PC: 2010, Tunisia
- Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Akhdhar, La Rabta, Tunis, 1007, Tunisia
- Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des Orangers Manouba, Tunis, 2010, Tunisia
| | - Gargouri-Berrechid Amina
- Neurology Department LR18SP03, Razi University Hospital, 1 rue des Orangers Manouba, Tunis, PC: 2010, Tunisia
- Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Akhdhar, La Rabta, Tunis, 1007, Tunisia
- Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des Orangers Manouba, Tunis, 2010, Tunisia
| | - Gouider Riadh
- Neurology Department LR18SP03, Razi University Hospital, 1 rue des Orangers Manouba, Tunis, PC: 2010, Tunisia.
- Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Akhdhar, La Rabta, Tunis, 1007, Tunisia.
- Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des Orangers Manouba, Tunis, 2010, Tunisia.
- Research Laboratory "Neurodegenerative Diseases and Psychiatric Disorders", LR18SP03, Tunis, poste 522, Tunisia.
| |
Collapse
|
21
|
Intronic variant increases Parkinson disease risk by disrupting branchpoint sequence. Nat Struct Mol Biol 2025; 32:12-13. [PMID: 39747488 DOI: 10.1038/s41594-024-01424-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
22
|
Cui C, Li H, Bao Y, Han Y, Yu H, Song H, Zhang B. Association between GRIN2B polymorphism and Parkinson's disease risk, age at onset, and progression in Southern China. Front Neurol 2024; 15:1459576. [PMID: 39758784 PMCID: PMC11697588 DOI: 10.3389/fneur.2024.1459576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 11/19/2024] [Indexed: 01/07/2025] Open
Abstract
Background and objectives The role of N-methyl-D-aspartate receptor 2B (GRIN2B) single nucleotide polymorphisms (SNPs) in influencing the risk and progression of Parkinson's disease (PD) is still unclear. This study aimed to assess the impact of GRIN2B genotype status on PD susceptibility and symptom progression. Methods We enrolled 165 individuals with sporadic PD and 154 healthy controls, all of whom had comprehensive clinical data available at the start and during follow-up. We used chi-squared (χ2) analysis to compare the allele and genotype frequency distributions between the patient and control groups. Linear mixed-effect models were employed to investigate the link between the GRIN2B genotype and the progression of motor and cognitive symptoms. Results The prevalence of the GG + GT genotype and G allele was higher in patients compared to controls (p = 0.032 and p = 0.001, respectively). Subgroup analysis revealed that the GG + GT genotype and G allele were significantly more frequently observed in late-onset PD (LOPD) patients compared to early-onset PD (EOPD) patients (p = 0.014 and p = 0.035, respectively). Notably, individuals with the GG + GT genotype exhibited an estimated annual progression rate of 6.10 points on the Unified Parkinson's Disease Rating Scale (UPDRS), which is significantly higher than that of the TT genotype carriers. Furthermore, the GG + GT carriers showed a markedly rapid progression in rigidity. In addition, the GG + GT carriers demonstrated significantly faster progression rates in rigidity (1.83 points/year) and axial impairment (1.2 points/year) compared to the TT carriers. Notably, the GG genotype carriers exhibited a more rapid decline in recall function. Conclusion The GRIN2B rs219882 G allele is associated with increased PD susceptibility, particularly in LOPD. The carriers of the GG + GT genotype exhibited more rapid motor symptom progression, with a pronounced impact on rigidity and axial impairment.
Collapse
Affiliation(s)
- Can Cui
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hongxia Li
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yiwen Bao
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yingying Han
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hongxiang Yu
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huan Song
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Bei Zhang
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
23
|
Hong S, Koretsky MJ, Lichtenberg J, Leonard H, Pitz V. Parkinson's Disease Pathogenic Variants: Cross-Ancestry Analysis and Microarray Data Validation. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.16.24319097. [PMID: 39763553 PMCID: PMC11702716 DOI: 10.1101/2024.12.16.24319097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Background Known pathogenic variants in Parkinson's disease (PD) contribute to disease development but have yet to be fully explored by arrays at scale. Objectives This study evaluated genotyping success of the NeuroBooster array (NBA) and determined the frequencies of pathogenic variants across ancestries. Method We analyzed the presence and allele frequency of 34 pathogenic variants in 28,710 PD cases, 9,614 other neurodegenerative disorder cases, and 15,821 controls across 11 ancestries within the Global Parkinson's Genetics Program dataset. Of these, 25 were genotyped on NBA and cluster plots were used to assess their quality. Results Genes previously predicted to have high or very high confidence of causing PD tend to have more pathogenic variants and are present across ancestry groups. Twenty-five of the 34 pathogenic variants were typed by the NBA array and classified "good" (n=12), "medium" (n=4), and "bad" (n=9) variants. Conclusion Our results confirm the likelihood that established PD genes are pathogenic and highlight the importance of ancestrally diverse research in PD. We also show the usefulness of the NBA as a reliable tool for genotyping of rare variants for PD.
Collapse
Affiliation(s)
- Samantha Hong
- Center for Alzheimer’s and Related Dementias, National Institutes of Health, Bethesda, USA
| | - Mathew J. Koretsky
- Center for Alzheimer’s and Related Dementias, National Institutes of Health, Bethesda, USA
- DataTecnica LLC, Washington DC, USA
| | - Jens Lichtenberg
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, USA
| | - Hampton Leonard
- Center for Alzheimer’s and Related Dementias, National Institutes of Health, Bethesda, USA
- DataTecnica LLC, Washington DC, USA
| | - Vanessa Pitz
- Integrative Neurogenomics Unit (INU), Laboratory of Neurogenetics, National Institutes of Health, Bethesda, USA
| | | |
Collapse
|
24
|
Koros C, Bougea A, Alefanti I, Simitsi AM, Papagiannakis N, Pachi I, Sfikas E, Antonelou R, Stefanis L. A Global Perspective of GBA1-Related Parkinson's Disease: A Narrative Review. Genes (Basel) 2024; 15:1605. [PMID: 39766872 PMCID: PMC11675599 DOI: 10.3390/genes15121605] [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] [Received: 11/05/2024] [Revised: 12/02/2024] [Accepted: 12/14/2024] [Indexed: 01/03/2025] Open
Abstract
Parkinson's disease (PD) is considered to be the second most prominent neurodegenerative disease and has a global prevalence. Glucocerebrosidase (GBA1) gene mutations represent a significant hereditary risk factor for the development of PD and have a profound impact on the motor and cognitive progression of the disease. The aim of this review is to summarize the literature data on the prevalence, type, and peculiarities of GBA1 mutations in populations of different ethnic backgrounds. We reviewed articles spanning the 2000-2024 period. GBA1-related PD has a worldwide distribution. It has long been recognized that pathogenic GBA1 mutations are particularly common in certain ethnic populations, including PD patients of Ashkenazi Jewish ancestry. Moreover, a considerable number of studies focused on European ancestry patients from Europe and North America have revealed a high proportion (up to 15%) of carriers among the PD population. GBA1 mutations also appear to play an important role in patient groups with an East Asian background, although the frequency of specific variants may differ as compared to those of European ancestry. Notably, the assessment of underrepresented populations in other parts of Asia (including India) and Latin America is in the spotlight of current research, while a variant with a newly described pathogenic mechanism has been reported in Sub-Saharan Africans. Given the importance of GBA1 mutations for PD genetics and clinical phenotype, a focused assessment of the prevalence and type of GBA1 variants in distinct ethnic populations will possibly inform ongoing PD-related clinical studies and facilitate upcoming therapeutic trials.
Collapse
Affiliation(s)
| | - Anastasia Bougea
- 1st Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (C.K.); (I.A.); (A.M.S.); (N.P.); (I.P.); (E.S.); (R.A.); (L.S.)
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Lange LM, Cerquera-Cleves C, Schipper M, Panagiotaropoulou G, Braun A, Kraft J, Awasthi S, Bell N, Posthuma D, Ripke S, Blauwendraat C, Heilbron K. Prioritizing Parkinson's disease risk genes in genome-wide association loci. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.13.24318996. [PMID: 39711693 PMCID: PMC11661345 DOI: 10.1101/2024.12.13.24318996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Recent advancements in Parkinson's disease (PD) drug development have been significantly driven by genetic research. Importantly, drugs supported by genetic evidence are more likely to be approved. While genome-wide association studies (GWAS) are a powerful tool to nominate genomic regions associated with certain traits or diseases, pinpointing the causal biologically relevant gene is often challenging. Our aim was to prioritize genes underlying PD GWAS signals. The polygenic priority score (PoPS) is a similarity-based gene prioritization method that integrates genome-wide information from MAGMA gene-level association tests and more than 57,000 gene-level features, including gene expression, biological pathways, and protein-protein interactions. We applied PoPS to data from the largest published PD GWAS in East Asian- and European-ancestries. We identified 120 independent associations with P < 5×10-8 and prioritized 46 PD genes across these loci based on their PoPS scores, distance to the GWAS signal, and presence of non-synonymous variants in the credible set. Alongside well-established PD genes (e.g., TMEM175 and VPS13C), some of which are targeted in ongoing clinical trials (i.e., SNCA, LRRK2, and GBA1), we prioritized genes with a plausible mechanistic link to PD pathogenesis (e.g., RIT2, BAG3, and SCARB2). Many of these genes hold potential for drug repurposing or novel therapeutic developments for PD (i.e., FYN, DYRK1A, NOD2, CTSB, SV2C, and ITPKB). Additionally, we prioritized potentially druggable genes that are relatively unexplored in PD (XPO1, PIK3CA, EP300, MAP4K4, CAMK2D, NCOR1, and WDR43). We prioritized a high-confidence list of genes with strong links to PD pathogenesis that may represent our next-best candidates for disease-modifying therapeutics. We hope our findings stimulate further investigations and preclinical work to facilitate PD drug development programs.
Collapse
Affiliation(s)
- Lara M. Lange
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
| | - Catalina Cerquera-Cleves
- Neurology Unit, Department of Neurosciences, Hospital Universitario San Ignacio, Bogotá, Colombia
- CHU de Québec Research Center, Axe Neurosciences, Laval University, Quebec City, Quebec, Canada
| | | | - Georgia Panagiotaropoulou
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Alice Braun
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Julia Kraft
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Swapnil Awasthi
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Nathaniel Bell
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Danielle Posthuma
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Child and Adolescent Psychiatry and Pediatric Psychology, Section Complex Trait Genetics, Amsterdam Neuroscience, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Stephan Ripke
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Karl Heilbron
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
- Current address: Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany
| |
Collapse
|
26
|
Palushaj B, Lewis SJG, Abdelnour C. What is the future for dementia with Lewy bodies? J Neurol 2024; 272:43. [PMID: 39666092 DOI: 10.1007/s00415-024-12734-1] [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] [Received: 10/07/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 12/13/2024]
Abstract
Dementia with Lewy bodies (DLB) is the second most common neurodegenerative dementia after Alzheimer's disease (AD), yet it remains under-recognized and frequently misdiagnosed due to heterogenous clinical presentations, the presence of co-pathology, and the lack of specific diagnostic tools. Pathologically, DLB is characterized by the accumulation of misfolded alpha-synuclein (aSyn) aggregates, known as Lewy bodies. Recent advancements have improved in vivo detection of aSyn pathology through techniques such as seed amplification assays, monoclonal antibodies, and positron emission tomography using novel small-molecule ligands. The ability to detect aSyn in vivo has sparked dialogue about using biomarkers to identify individuals with aSyn, similar to the approach influencing the field of AD. Proponents argue that biological staging could facilitate the detection of preclinical disease stages, allowing for earlier intervention and targets for disease modification, and could improve diagnostic sensitivity and accuracy in selecting patients for clinical trials. However, critics caution that this method may oversimplify the complexity of DLB and overlook its clinical heterogeneity, also highlighting practical challenges related to implementation, cost, and global access to advanced diagnostic technologies. Importantly, although significant progress has been made in detecting aSyn for diagnostic purposes, disease-modifying therapies targeting aSyn have yet to demonstrate clear efficacy in slowing disease progression. Elucidating the physiological and pathophysiological roles of aSyn remains an urgent priority in neurodegenerative research. Other experimental research priorities for DLB include developing improved cellular and animal models that reflect epigenetic and environmental factors, mapping post-translational modifications, and systematically characterizing neurons that are vulnerable and resistant to lewy pathology using a multi-omic approach. Clinically, there is an urgent need for international, prospective, longitudinal studies and for validated, disease-specific outcome measures. Addressing these priorities is essential for advancing our understanding of DLB and developing effective therapies.
Collapse
Affiliation(s)
- Bianca Palushaj
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| | | | - Carla Abdelnour
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
27
|
Lim SY, Tan AH, Ahmad-Annuar A, Okubadejo NU, Lohmann K, Morris HR, Toh TS, Tay YW, Lange LM, Bandres-Ciga S, Mata I, Foo JN, Sammler E, Ooi JCE, Noyce AJ, Bahr N, Luo W, Ojha R, Singleton AB, Blauwendraat C, Klein C. Uncovering the genetic basis of Parkinson's disease globally: from discoveries to the clinic. Lancet Neurol 2024; 23:1267-1280. [PMID: 39447588 DOI: 10.1016/s1474-4422(24)00378-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/29/2024] [Accepted: 09/04/2024] [Indexed: 10/26/2024]
Abstract
Knowledge on the genetic basis of Parkinson's disease has grown tremendously since the discovery of the first monogenic form, caused by a mutation in α-synuclein, and with the subsequent identification of multiple other causative genes and associated loci. Genetic studies provide insights into the phenotypic heterogeneity and global distribution of Parkinson's disease. By shedding light on the underlying biological mechanisms, genetics facilitates the identification of new biomarkers and therapeutic targets. Several clinical trials of genetics-informed therapies are ongoing or imminent. International programmes in populations who have been under-represented in Parkinson's disease genetics research are fostering collaboration and capacity-building, and have already generated novel findings. Many challenges remain for genetics research in these populations, but addressing them provides opportunities to obtain a more complete and equitable understanding of Parkinson's disease globally. These advances facilitate the integration of genetics into the clinic, to improve patient management and personalised medicine.
Collapse
Affiliation(s)
- Shen-Yang Lim
- Division of Neurology, Department of Medicine, and The Mah Pooi Soo & Tan Chin Nam Centre for Parkinson's & Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| | - Ai Huey Tan
- Division of Neurology, Department of Medicine, and The Mah Pooi Soo & Tan Chin Nam Centre for Parkinson's & Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Azlina Ahmad-Annuar
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Njideka Ulunma Okubadejo
- College of Medicine, University of Lagos and Lagos University Teaching Hospital, Idi Araba, Lagos State, Nigeria
| | - Katja Lohmann
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, University College London, Institute of Neurology, London, UK
| | - Tzi Shin Toh
- Division of Neurology, Department of Medicine, and The Mah Pooi Soo & Tan Chin Nam Centre for Parkinson's & Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yi Wen Tay
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Lara M Lange
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany; Department of Neurology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Sara Bandres-Ciga
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Ignacio Mata
- Lerner Research Institute, Genomic Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Jia Nee Foo
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore; Laboratory of Neurogenetics, Genome Institute of Singapore, A*STAR, Singapore
| | - Esther Sammler
- Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK; Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Joshua Chin Ern Ooi
- Department of Neurology, Queen Elizabeth Hospital, Kota Kinabalu, Sabah, Malaysia
| | - Alastair J Noyce
- Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Natascha Bahr
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany; Department of Neurology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Wei Luo
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, People's Republic of China
| | - Rajeev Ojha
- Department of Neurology, Tribhuvan University Teaching Hospital, Kathmandu, Nepal
| | - Andrew B Singleton
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA; Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
| | - Cornelis Blauwendraat
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA; Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
| | - Christine Klein
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany; Department of Neurology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| |
Collapse
|
28
|
Álvarez Jerez P, Wild Crea P, Ramos DM, Gustavsson EK, Radefeldt M, Damianov A, Makarious MB, Ojo OO, Billingsley KJ, Malik L, Daida K, Bromberek S, Hu F, Schneider Z, Surapaneni AL, Stadler J, Rizig M, Morris HR, Pantazis CB, Leonard HL, Screven L, Qi YA, Nalls MA, Bandres-Ciga S, Hardy J, Houlden H, Eng C, Burchard EG, Kachuri L, Lin CH, Black DL, Singleton AB, Fischer S, Bauer P, Reed X, Ryten M, Beetz C, Ward M, Okubadejo NU, Blauwendraat C. African ancestry neurodegeneration risk variant disrupts an intronic branchpoint in GBA1. Nat Struct Mol Biol 2024; 31:1955-1963. [PMID: 39668204 PMCID: PMC11638064 DOI: 10.1038/s41594-024-01423-2] [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: 03/25/2024] [Accepted: 10/10/2024] [Indexed: 12/14/2024]
Abstract
Recently, an African ancestry-specific Parkinson disease (PD) risk signal was identified at the gene encoding glucocerebrosidase (GBA1). This variant ( rs3115534 -G) is carried by ~50% of West African PD cases and imparts a dose-dependent increase in risk for disease. The risk variant has varied frequencies across African ancestry groups but is almost absent in European and Asian ancestry populations. GBA1 is a gene of high clinical and therapeutic interest. Damaging biallelic protein-coding variants cause Gaucher disease and monoallelic variants confer risk for PD and dementia with Lewy bodies, likely by reducing the function of glucocerebrosidase. Interestingly, the African ancestry-specific GBA1 risk variant is a noncoding variant, suggesting a different mechanism of action. Using full-length RNA transcript sequencing, we identified partial intron 8 expression in risk variant carriers (G) but not in nonvariant carriers (T). Antibodies targeting the N terminus of glucocerebrosidase showed that this intron-retained isoform is likely not protein coding and subsequent proteomics did not identify a shorter protein isoform, suggesting that the disease mechanism is RNA based. Clustered regularly interspaced short palindromic repeats editing of the reported index variant ( rs3115534 ) revealed that this is the sequence alteration responsible for driving the production of these transcripts containing intron 8. Follow-up analysis of this variant showed that it is in a key intronic branchpoint sequence and, therefore, has important implications in splicing and disease. In addition, when measuring glucocerebrosidase activity, we identified a dose-dependent reduction in risk variant carriers. Overall, we report the functional effect of a GBA1 noncoding risk variant, which acts by interfering with the splicing of functional GBA1 transcripts, resulting in reduced protein levels and reduced glucocerebrosidase activity. This understanding reveals a potential therapeutic target in an underserved and underrepresented population.
Collapse
Affiliation(s)
- Pilar Álvarez Jerez
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Peter Wild Crea
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Daniel M Ramos
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Emil K Gustavsson
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK
| | | | - Andrey Damianov
- Department of Microbiology, Immunology and Molecular Genetics, The David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Mary B Makarious
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- DataTecnica, Washington, DC, USA
| | - Oluwadamilola O Ojo
- College of Medicine, University of Lagos, Lagos, Nigeria
- Lagos University Teaching Hospital, Lagos, Nigeria
| | - Kimberley J Billingsley
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Laksh Malik
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Kensuke Daida
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Sarah Bromberek
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Fangle Hu
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Zachary Schneider
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Aditya L Surapaneni
- Department of Medicine, New York University Langone School of Medicine, New York, NY, USA
| | - Julia Stadler
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mie Rizig
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Huw R Morris
- UCL Movement Disorders Centre, University College London, London, UK
| | - Caroline B Pantazis
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Hampton L Leonard
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- DataTecnica, Washington, DC, USA
| | - Laurel Screven
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Yue A Qi
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mike A Nalls
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- DataTecnica, Washington, DC, USA
| | - Sara Bandres-Ciga
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - John Hardy
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Celeste Eng
- Department of Biotherapeutic Sciences and Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Esteban González Burchard
- Department of Biotherapeutic Sciences and Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Linda Kachuri
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Chia-Ho Lin
- Department of Microbiology, Immunology and Molecular Genetics, The David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Douglas L Black
- Department of Microbiology, Immunology and Molecular Genetics, The David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Andrew B Singleton
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | | | | | - Xylena Reed
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mina Ryten
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- UK Dementia Research Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - Michael Ward
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Njideka U Okubadejo
- College of Medicine, University of Lagos, Lagos, Nigeria
- Lagos University Teaching Hospital, Lagos, Nigeria
| | - Cornelis Blauwendraat
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA.
| |
Collapse
|
29
|
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
|
30
|
Tan AH, Saffie-Awad P, Schumacher Schuh AF, Lim SY, Madoev H, Ahmad-Annuar A, Solle J, Wegel CE, Doquenia ML, Dey S, Perinan MT, Makarious MB, Fiske B, Morris HR, Noyce AJ, Alcalay RN, Kumar KR, Klein C. Global Perspectives on Returning Genetic Research Results in Parkinson Disease. Neurol Genet 2024; 10:e200213. [PMID: 39807215 PMCID: PMC11727605 DOI: 10.1212/nxg.0000000000200213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/14/2024] [Indexed: 01/16/2025]
Abstract
Background and Objectives In the era of precision medicine, genetic test results have become increasingly relevant in the care of patients with Parkinson disease (PD). While large research consortia are performing widespread research genetic testing to accelerate discoveries, debate continues about whether, and to what extent, the results should be returned to patients. Ethically, it is imperative to keep participants informed, especially when findings are potentially actionable. However, research testing may not hold the same standards required from clinical diagnostic laboratories and hold significant psychosocial implications. The absence of universally recognized protocols complicates the establishment of appropriate guidelines. Methods Aiming to develop recommendations on return of research results (RoR) practice within the Global Parkinson's Genetics Program (GP2), we conducted a global survey to gain insight on GP2 members' perceptions, practice, readiness, and needs surrounding RoR. Results GP2 members (n = 191), representing 147 institutions and 60 countries across 6 continents, completed the survey. Access to clinical genetic testing services was significantly higher in high-income countries compared with low- and middle-income countries (96.6% vs 58.4%), where funding was predominantly covered by patients themselves. While 92.7% of the respondents agreed that genetic research results should be returned, levels of agreement were higher for clinically relevant results relating to pathogenic or likely pathogenic variants in genes known to cause PD or other neurodegenerative diseases. Less than 10% offered separate clinically accredited genetic testing before returning genetic research results. A total of 48.7% reported having a specific statement on RoR policy in their ethics consent form, while 53.9% collected data on participants' preferences on RoR prospectively. 24.1% had formal genetic counselling training. Notably, the comfort level in returning incidental genetic findings or returning results to unaffected individuals remains low. Discussion Given the differences in resources and training for RoR, as well as ethical and regulatory considerations, tailored approaches are required to ensure equitable access to RoR. Several identified strategies to enhance RoR practices include improving informed consent processes, increasing capacity for genetic counselling including providing counselling toolkits for common genetic variants, broadening access to sustainable clinically accredited testing, building logistical infrastructure for RoR processes, and continuing public and health care education efforts on the important role of genetics in PD.
Collapse
Affiliation(s)
- Ai Huey Tan
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Paula Saffie-Awad
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Artur F Schumacher Schuh
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Shen-Yang Lim
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Harutyun Madoev
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Azlina Ahmad-Annuar
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - J Solle
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Claire E Wegel
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Maria Leila Doquenia
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Sumit Dey
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Maria Teresa Perinan
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Mary B Makarious
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Brian Fiske
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Huw R Morris
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Alastair J Noyce
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Roy N Alcalay
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Kishore Raj Kumar
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Christine Klein
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| |
Collapse
|
31
|
Somerville EN, Gan-Or Z. Genetic-based diagnostics of Parkinson's disease and other Parkinsonian syndromes. Expert Rev Mol Diagn 2024:1-13. [PMID: 39545628 DOI: 10.1080/14737159.2024.2427625] [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: 08/20/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024]
Abstract
INTRODUCTION Parkinson's disease (PD) is a complex disorder with vast clinical heterogeneity. Recent genetic, imaging and clinical evidence suggest that there are multiple subtypes of PD, and perhaps even distinct clinical entities, which are being diagnosed under the umbrella of PD. These might have similar clinical presentation, but potentially different underlying mechanisms, which, in future, will require different treatments. Despite extensive genetic research progress, genetic testing is still not a common practice in clinical patient care. AREAS COVERED This review examines the numerous genes that have been discovered to affect the risk of, or cause, PD. We also outline genetic variants that affect PD age at onset, its progression, and the presence or severity of motor and non-motor symptoms. We differentiate between PD, other synucleinopathies, and atypical parkinsonism syndromes, and describe genes responsible for familial forms of typical PD and atypical parkinsonism. Lastly, we present current clinical trails that are underway for targeted therapies, particularly for GBA1-PD and LRRK2-PD which are the most significant subtypes. EXPERT OPINION While genetic studies alone cannot be diagnostic for PD, proper utilization of genetic screening for PD could improve diagnostic accuracy and predictions for prognosis, guide treatment, and identify individuals that qualify for clinical trials.
Collapse
Affiliation(s)
- Emma N Somerville
- The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, Canada
- Department of Human Genetics, McGill University, Montréal, Canada
| | - Ziv Gan-Or
- The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, Canada
- Department of Human Genetics, McGill University, Montréal, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Canada
| |
Collapse
|
32
|
Miao S, Ni Q, Fang J. Relationship Between Mitochondrial Biological Function and Breast Cancer: An Approach Based on Mendelian Randomization Analysis. Breast J 2024; 2024:4434466. [PMID: 39742384 PMCID: PMC11561182 DOI: 10.1155/2024/4434466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/24/2024] [Accepted: 09/21/2024] [Indexed: 01/03/2025]
Abstract
Objective: This study aims to investigate the potential causal link between mitochondrial function and breast cancer using the Mendelian randomization (MR) analysis. Methods: The data used for this study were obtained from genomewide association studies (GWAS) databases on mitochondrial biological function and breast cancer. Mitochondrial function was considered the exposure variable, breast cancer the outcome variable, and specific single nucleotide polymorphisms (SNPs) were selected as instrumental variables (IVs). Two MR methods, inverse variance weighting (IVW) and MR-Egger regression, were used to assess the causal association between mitochondrial function and breast cancer. Data analysis and visualization were performed using R software. Results: The results of the analysis revealed that several genes, including 39S ribosomal protein L34, pyruvate carboxylase, rRNA methyltransferase 3, and cytochrome c oxidase assembly factor 3 homolog, are causally linked to an increased risk of breast cancer in European populations. In addition, cytochrome c oxidase subunit 8A and ADP-ribose pyrophosphatase were found to be protective factors against breast cancer in European populations. In East Asian populations, 39S ribosomal protein L52, ATP synthase subunit beta, and pyruvate dehydrogenase (acetyl-transferring) were identified as causal risk factors for breast cancer. Conversely, 39S ribosomal protein L32, ADP-ribose pyrophosphatase, and cytochrome c oxidase subunit 8A were identified as protective factors against breast cancer in this population. Conclusion: In conclusion, this study provides evidence of a causal relationship between mitochondrial function and breast cancer in both European and East Asian populations. Additional research is warranted to further elucidate the mechanisms underlying this association.
Collapse
Affiliation(s)
- Shichen Miao
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Qichao Ni
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Jun Fang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| |
Collapse
|
33
|
Bandres-Ciga S, Faghri F, Majounie E, Koretsky MJ, Kim J, Levine KS, Leonard H, Makarious MB, Iwaki H, Crea PW, Hernandez DG, Arepalli S, Billingsley K, Lohmann K, Klein C, Lubbe SJ, Jabbari E, Saffie-Awad P, Narendra D, Reyes-Palomares A, Quinn JP, Schulte C, Morris HR, Traynor BJ, Scholz SW, Houlden H, Hardy J, Dumanis S, Riley E, Blauwendraat C, Singleton A, Nalls M, Jeff J, Vitale D. NeuroBooster Array: A Genome-Wide Genotyping Platform to Study Neurological Disorders Across Diverse Populations. Mov Disord 2024; 39:2039-2048. [PMID: 39283294 PMCID: PMC11568947 DOI: 10.1002/mds.29902] [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: 03/28/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND Commercial genome-wide genotyping arrays have historically neglected coverage of genetic variation across populations. OBJECTIVE We aimed to create a multi-ancestry genome-wide array that would include a wide range of neuro-specific genetic content to facilitate genetic research in neurological disorders across multiple ancestral groups, fostering diversity and inclusivity in research studies. METHODS We developed the Illumina NeuroBooster Array (NBA), a custom high-throughput and cost-effective platform on a backbone of 1,914,934 variants from the Infinium Global Diversity Array and added custom content comprising 95,273 variants associated with more than 70 neurological conditions or traits, and we further tested its performance on more than 2000 patient samples. This novel platform includes approximately 10,000 tagging variants to facilitate imputation and analyses of neurodegenerative disease-related genome-wide association study loci across diverse populations. RESULTS In this article, we describe NBA's potential as an efficient means for researchers to assess known and novel disease genetic associations in a multi-ancestry framework. The NBA can identify rare genetic variants and accurately impute more than 15 million common variants across populations. Apart from enabling sample prioritization for further whole-genome sequencing studies, we envisage that NBA will play a pivotal role in recruitment for interventional studies in the precision medicine space. CONCLUSIONS From a broader perspective, the NBA serves as a promising means to foster collaborative research endeavors in the field of neurological disorders worldwide. Ultimately, this carefully designed tool is poised to make a substantial contribution to uncovering the genetic etiology underlying these debilitating conditions. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.
Collapse
Affiliation(s)
- Sara Bandres-Ciga
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Faraz Faghri
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC 20037, USA
| | | | - Mathew J Koretsky
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey Kim
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kristin S Levine
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC 20037, USA
| | - Hampton Leonard
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC 20037, USA
| | - Mary B Makarious
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK
| | - Hirotaka Iwaki
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC 20037, USA
| | - Peter Wild Crea
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dena G Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sampath Arepalli
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kimberley Billingsley
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Steven J Lubbe
- Ken and Ruth Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Simpson Querrey Center for Neurogenetics, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Edwin Jabbari
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK
| | - Paula Saffie-Awad
- Programa de Pós-Graduação em Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brasil
- Centro de Trastornos del Movimiento (CETRAM), Santiago, Chile
- Clínica Santa María, Santiago, Chile
| | - Derek Narendra
- Inherited Movement Disorders Unit, Neurogenetics Branch, Division of Intramural Research, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Armando Reyes-Palomares
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, Málaga, Spain
| | - John P Quinn
- Department of Pharmacology & Therapeutics, University of Liverpool, Liverpool, UK
| | - Claudia Schulte
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tuebingen and German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK
- Aligning Science Against Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Bryan J. Traynor
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Sonja W. Scholz
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Henry Houlden
- Aligning Science Against Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - John Hardy
- Aligning Science Against Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Sonya Dumanis
- Aligning Science Against Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Ekemini Riley
- Aligning Science Against Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Cornelis Blauwendraat
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew Singleton
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mike Nalls
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC 20037, USA
| | | | - Dan Vitale
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC 20037, USA
| |
Collapse
|
34
|
Brolin KA, Bäckström D, Wallenius J, Gan-Or Z, Puschmann A, Hansson O, Swanberg M. GBA1 T369M and Parkinson's disease - Further evidence of a lack of association in the Swedish population. Parkinsonism Relat Disord 2024; 130:107191. [PMID: 39514913 DOI: 10.1016/j.parkreldis.2024.107191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/21/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Variants in GBA1 are important genetic risk factors in Parkinson's disease (PD). GBA1 T369M has been linked to an ∼80 % increased PD risk but the reports are conflicting and the relevance of GBA1 variants in different populations varies. A lack of association between T369M and PD in the Swedish population was recently reported but needs further validation. We therefore investigated T369M in 1,808 PD patients and 2,183 controls and our results support that T369M is not a risk factor for PD in the Swedish population.
Collapse
Affiliation(s)
- Kajsa Atterling Brolin
- Translational Neurogenetics Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden; Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University of London, EC1M 6BQ, London, UK.
| | - David Bäckström
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Joel Wallenius
- Division of Neurology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden; Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Ziv Gan-Or
- Department of Neurology & Neurosurgery, McGill University, Montreal, Quebec, Canada; Clinical Research Unit, The Neuro (Montreal Neurological Institute-Hospital), Montreal, Quebec, Canada; Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Andreas Puschmann
- Division of Neurology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden; Department of Neurology, Skåne University Hospital, Lund, Sweden; SciLifeLab National Research Infrastructure, Lund University, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Maria Swanberg
- Translational Neurogenetics Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
35
|
Chahine LM, Louie N, Solle J, Akçimen F, Ameri A, Augenbraun S, Avripas S, Breaux S, Causey C, Chandra S, Dean M, Disbrow EA, Fanty L, Fernandez J, Foster ER, Furr Stimming E, Hall D, Hinson V, Johnson-Turbes A, Jonas C, Kilbane C, Norris SA, Nguyen BT, Padmanaban M, Paquette K, Parry C, Pessoa Rocha N, Rawls A, Shamim EA, Shulman LM, Sipma R, Staisch J, Traurig R, von Coelln R, Wild Crea P, Xie T, Fang ZH, O'Grady A, Kopil CM, McGuire Kuhl M, Singleton A, Blauwendraat C, Bandres-Ciga S. The Black and African American Connections to Parkinson's Disease (BLAAC PD) study protocol. BMC Neurol 2024; 24:403. [PMID: 39434044 PMCID: PMC11492614 DOI: 10.1186/s12883-024-03914-7] [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: 09/20/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024] Open
Abstract
Determining the genetic contributions to Parkinson's disease (PD) across diverse ancestries is a high priority as this work can guide therapeutic development in a global setting. The genetics of PD spans the etiological risk spectrum, from rare, highly deleterious variants linked to monogenic forms with Mendelian patterns of inheritance, to common variation involved in sporadic disease. A major limitation in PD genomics research is lack of racial and ethnic diversity. Enrollment disparities have detrimental consequences on the generalizability of results and exacerbate existing inequities in care. The Black and African American Connections to Parkinson's Disease (BLAAC PD) study is part of the Global Parkinson's Genetics Program, supported by the Aligning Science Across Parkinson's initiative. The goal of the study is to investigate the genetic architecture underlying PD risk and progression in the Black and/or African American populations. This cross-sectional multicenter study in the United States has a recruitment target of up to 2,000 individuals with PD and up to 2,000 controls, all of Black and/or African American ancestry. The study design incorporates several strategies to reduce barriers to research participation. The multifaceted recruitment strategy aims to involve individuals with and without PD in various settings, emphasizing community outreach and engagement. The BLAAC PD study is an important first step toward informing understanding of the genetics of PD in a more diverse population.
Collapse
Affiliation(s)
- Lana M Chahine
- University of Pittsburgh, 3471 Fifth Avenue, Pittsburgh, PA, 15213, USA.
| | - Naomi Louie
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - J Solle
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Fulya Akçimen
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Andrew Ameri
- Medical University of South Carolina, Charleston, SC, USA
| | | | | | | | - Christopher Causey
- Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, USA
| | - Shivika Chandra
- The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Marissa Dean
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Elizabeth A Disbrow
- Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, USA
| | | | | | - Erin R Foster
- University of Pittsburgh, 3471 Fifth Avenue, Pittsburgh, PA, 15213, USA
| | - Erin Furr Stimming
- The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Deborah Hall
- Rush University Medical Center, Chicago, IL, USA
| | - Vanessa Hinson
- Medical University of South Carolina, Charleston, SC, USA
| | | | - Cabell Jonas
- Kaiser Permanente Mid-Atlantic States, Largo, MD, USA
| | - Camilla Kilbane
- University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | | | | | | | - Kimberly Paquette
- Center for Alzheimer's Disease and Related Dementias, National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Carly Parry
- NORC at the University of Chicago, Chicago, IL, USA
| | | | | | - Ejaz A Shamim
- Kaiser Permanente Mid-Atlantic States, Largo, MD, USA
| | | | - Rebeka Sipma
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Rami Traurig
- Center for Alzheimer's Disease and Related Dementias, National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | | | - Peter Wild Crea
- Center for Alzheimer's Disease and Related Dementias, National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Tao Xie
- University of Chicago, Chicago, IL, USA
| | - Zih-Hua Fang
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Alyssa O'Grady
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Catherine M Kopil
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | | | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer's Disease and Related Dementias, National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | | | - Sara Bandres-Ciga
- Center for Alzheimer's Disease and Related Dementias, National Institute on Aging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
36
|
Ng ASL, Tan AH, Tan YJ, Lim JL, Lian MM, Dy Closas AM, Ahmad-Annuar A, Viswanathan S, Chia YK, Foo JN, Lim WK, Tan EK, Lim SY. Identification of Genetic Variants in Progressive Supranuclear Palsy in Southeast Asia. Mov Disord 2024; 39:1829-1842. [PMID: 39149795 DOI: 10.1002/mds.29932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/21/2024] [Accepted: 07/01/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Progressive supranuclear palsy (PSP) is largely a sporadic disease with few reported familial cases. Genome-wide association studies (GWAS) in sporadic PSP in Caucasian populations have identified MAPT as the most commonly associated genetic risk locus with the strongest effect size. At present there are limited data on genetic factors associated with PSP in Asian populations. OBJECTIVES Our goal was to investigate the genetic factors associated with PSP in Southeast Asian PSP patients. METHODS Next-generation sequencing (whole-exome, whole-genome and targeted sequencing) was performed in two Asian cohorts, comprising 177 PSP patients. RESULTS We identified 17 pathogenic or likely pathogenic variants in 16 PSP patients (9%), eight of which were novel. The most common relevant genetic variants identified were in MAPT, GBA1, OPTN, SYNJ1, and SQSTM1. Other variants detected were in TBK1, PRNP, and ABCA7-genes that have been implicated in other neurodegenerative diseases. Eighteen patients had a positive family history, of whom two carried pathogenic MAPT variants, and one carried a likely pathogenic GBA1 variant. None of the patients had expanded repeats in C9orf72. Furthermore, we found 16 different variants of uncertain significance in 21 PSP patients in PSEN2, ABCA7, SMPD1, MAPT, ATP13A2, OPTN, SQSTM1, CYLD, and BSN. CONCLUSIONS The genetic findings in our PSP cohorts appear to be somewhat distinct from those in Western populations, and also suggest an overlap of the genetic architecture between PSP and other neurodegenerative diseases. Further functional studies and validation in independent Asian cohorts will be useful for improving our understanding of PSP genetics and guiding genetic screening strategies in these populations. © 2024 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Adeline Su Lyn Ng
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
- Neuroscience and Behavioural Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Ai Huey Tan
- Division of Neurology, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Mah Pooi Soo and Tan Chin Nam Centre for Parkinson's and Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yi Jayne Tan
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
| | - Jia Lun Lim
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Michelle Mulan Lian
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Alfand Marl Dy Closas
- Division of Neurology, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Mah Pooi Soo and Tan Chin Nam Centre for Parkinson's and Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Metro Davao Medical and Research Center, Davao Doctors Hospital, Davao City, Philippines
| | - Azlina Ahmad-Annuar
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Yuen Kang Chia
- Department of Neurology, Queen Elizabeth Hospital, Kota Kinabalu, Sabah, Malaysia
| | - Jia Nee Foo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
| | - Weng Khong Lim
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
- Singhealth Duke-NUS Institute of Precision Medicine, Singapore
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
- SingHealth Duke-NUS Genomic Medicine Centre, Singapore
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
- Neuroscience and Behavioural Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Shen-Yang Lim
- Division of Neurology, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Mah Pooi Soo and Tan Chin Nam Centre for Parkinson's and Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
37
|
McGlinchey E, Duran-Aniotz C, Akinyemi R, Arshad F, Zimmer ER, Cho H, Adewale BA, Ibanez A. Biomarkers of neurodegeneration across the Global South. THE LANCET. HEALTHY LONGEVITY 2024; 5:100616. [PMID: 39369726 PMCID: PMC11540104 DOI: 10.1016/s2666-7568(24)00132-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 10/08/2024] Open
Abstract
Research on neurodegenerative diseases has predominantly focused on high-income countries in the Global North. This Series paper describes the state of biomarker evidence for neurodegeneration in the Global South, including Latin America, Africa, and countries in south, east, and southeast Asia. Latin America shows growth in fluid biomarker and neuroimaging research, with notable advancements in genetics. Research in Africa focuses on genetics and cognition but there is a paucity of data on fluid and neuroimaging biomarkers. South and east Asia, particularly India and China, has achieved substantial progress in plasma, neuroimaging, and genetic studies. However, all three regions face several challenges in the form of a lack of harmonisation, insufficient funding, and few comparative studies both within the Global South, and between the Global North and Global South. Other barriers include scarce infrastructure, lack of knowledge centralisation, genetic and cultural diversity, sociocultural stigmas, and restricted access to tools such as PET scans. However, the diverse ethnic, genetic, economic, and cultural backgrounds in the Global South present unique opportunities for bidirectional learning, underscoring the need for global collaboration to enhance the understanding of dementia and brain health.
Collapse
Affiliation(s)
- Eimear McGlinchey
- Trinity College Dublin, Dublin, Ireland; Global Brain Health Institute, University of California San Francisco (UCSF), San Francisco, CA, USA; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.
| | - Claudia Duran-Aniotz
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibanez, Santiago de Chile, Chile
| | - Rufus Akinyemi
- Global Brain Health Institute, University of California San Francisco (UCSF), San Francisco, CA, USA; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland; Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria; Centre for Genomic and Precision Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Faheem Arshad
- Global Brain Health Institute, University of California San Francisco (UCSF), San Francisco, CA, USA; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland; National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Eduardo R Zimmer
- Department of Pharmacology, Graduate Program in Biological Sciences: Pharmacology and Therapeutics (PPGFT) and Biochemistry (PPGBioq), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Brain Institute of Rio Grande do Sul, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil; McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Hanna Cho
- Global Brain Health Institute, University of California San Francisco (UCSF), San Francisco, CA, USA; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland; Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Boluwatife Adeleye Adewale
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Agustin Ibanez
- Trinity College Dublin, Dublin, Ireland; Global Brain Health Institute, University of California San Francisco (UCSF), San Francisco, CA, USA; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland; Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibanez, Santiago de Chile, Chile.
| |
Collapse
|
38
|
Jones L, Cerquera-Cleves C, Schuh AFS, Makarious MB, Iwaki H, Nalls MA, Noyce AJ, Blauwendraat C, Singleton A, Mata I, Bandres-Ciga S. Multi-ancestry population attributable risk assessment of common genetic variation in Alzheimer's and Parkinson's diseases. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.23.24314240. [PMID: 39371162 PMCID: PMC11451675 DOI: 10.1101/2024.09.23.24314240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Multiple scientific studies, mostly performed within European populations, have unraveled many of the genetic factors associated with Alzheimer's disease (AD) and Parkinson's disease (PD) etiologies, improving our understanding of the molecular pathways implicated in the pathogenesis of these conditions. However, there is increasing evidence that the genetic architecture of these diseases differs across ancestral populations. This raises concerns about the efficacy of therapeutic interventions crafted around genetic targets prevalent only in European ancestry populations. Such interventions neglect potentially distinctive etiological profiles, including Latino, Black/African American, and East Asian populations. In the current study, we explore Population Attributable Risk (PAR) in AD and PD etiologies and assess the proportion of disease attributed to specific genetic factors across diverse populations. Leveraging data from genome-wide association studies across four ancestries, we explore distinct and universal therapeutic targets across diverse populations. Multi-ancestral genetics research is critical to the development of successful therapeutics and treatments for neurodegenerative diseases. By offering insights into genetic disparities, we aim to inform more inclusive and effective therapeutic strategies, advancing personalized healthcare.
Collapse
Affiliation(s)
- Lietsel Jones
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- DataTecnica LLC, Washington DC, USA
| | - Catalina Cerquera-Cleves
- Department of Neurosciences, Neurology Unit, Hospital Universitario San Ignacio, Bogotá, Colombia
- CHU de Québec Research Center, Axe Neurosciences, Université Laval, Quebec City, Canada
| | - Artur FS Schuh
- Departamento de Farmacologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Serviço de Neurologia, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Mary B Makarious
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- DataTecnica LLC, Washington DC, USA
| | - Hirotaka Iwaki
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- DataTecnica LLC, Washington DC, USA
| | - Mike A. Nalls
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- DataTecnica LLC, Washington DC, USA
| | - Alastair J Noyce
- Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | | | - Cornelis Blauwendraat
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
| | - Andrew Singleton
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
| | - Ignacio Mata
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Sara Bandres-Ciga
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
39
|
Somerville EN, James A, Beetz C, Schwieger R, Barrel G, Kandaswamy KK, Iurascu MI, Bauer P, Ta M, Iwaki H, Senkevich K, Yu E, Alcalay RN, Gan-Or Z. Plasma glucosylceramide levels are regulated by ATP10D and are not involved in Parkinson's disease pathogenesis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.13.24313644. [PMID: 39371176 PMCID: PMC11451666 DOI: 10.1101/2024.09.13.24313644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
GBA1 variants and decreased glucocerebrosidase (GCase) activity are implicated in Parkinson's disease (PD). We investigated the hypothesis that increased levels of glucosylceramide (GlcCer), one of GCase main substrates, are involved in PD pathogenesis. Using multiple genetic methods, we show that ATP10D, not GBA1, is the main regulator of plasma GlcCer levels, yet it is not involved in PD pathogenesis. Plasma GlcCer levels were associated with PD, but not in a causative manner, and are not predictive of disease status. These results argue against targeting GlcCer in GBA1-PD and underscore the need to explore alternative mechanisms and biomarkers for PD.
Collapse
Affiliation(s)
- Emma N. Somerville
- The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | | | | | | | | | - Krishna K. Kandaswamy
- CENTOGENE GmbH, Rostock, Germany
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Marius I. Iurascu
- CENTOGENE GmbH, Rostock, Germany
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | | | - Michael Ta
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC, USA
| | - Hirotaka Iwaki
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC, USA
| | - Konstantin Senkevich
- The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Eric Yu
- The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Roy N. Alcalay
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ziv Gan-Or
- The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| |
Collapse
|
40
|
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
|
41
|
Hertz E, Chen Y, Sidransky E. Gaucher disease provides a unique window into Parkinson disease pathogenesis. Nat Rev Neurol 2024; 20:526-540. [PMID: 39107435 DOI: 10.1038/s41582-024-00999-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2024] [Indexed: 09/04/2024]
Abstract
An exciting development in the field of neurodegeneration is the association between the rare monogenic disorder Gaucher disease and the common complex disorder Parkinson disease (PD). Gaucher disease is a lysosomal storage disorder resulting from an inherited deficiency of the enzyme glucocerebrosidase, encoded by GBA1, which hydrolyses the glycosphingolipids glucosylceramide and glucosylsphingosine. The observation of parkinsonism in a rare subgroup of individuals with Gaucher disease first directed attention to the role of glucocerebrosidase deficiency in the pathogenesis of PD. PD occurs more frequently in people heterozygous for Gaucher GBA1 mutations, and 3-25% of people with Parkinson disease carry a GBA1 variant. However, only a small percentage of individuals with GBA1 variants develop parkinsonism, suggesting that the penetrance is low. Despite over a decade of intense research in this field, including clinical and radiological evaluations, genetic studies and investigations using model systems, the mechanism underlying GBA1-PD is still being pursued. Insights from this association have emphasized the role of lysosomal pathways in parkinsonism. Furthermore, different therapeutic strategies considered or developed for Gaucher disease can now inform drug development for PD.
Collapse
Affiliation(s)
- Ellen Hertz
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yu Chen
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ellen Sidransky
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
42
|
Gan-Or Z. Clinical genetic testing in Parkinson's disease should become part of routine patient care. Brain 2024; 147:2595-2597. [PMID: 39087915 PMCID: PMC11292891 DOI: 10.1093/brain/awae181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 08/02/2024] Open
Abstract
This scientific commentary refers to ‘Relevance of genetic testing in the gene-targeted trial era: the Rostock Parkinson’s Disease Study’ by Westenberger et al. (https://doi.org/10.1093/brain/awae188) and ‘Parkinson’s disease variant detection and disclosure: PD GENEration, a North American study’ by Cook et al. (https://doi.org/10.1093/brain/awae142).
Collapse
Affiliation(s)
- Ziv Gan-Or
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
- Clinical Research Unit, The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montreal, QC H3A 2B4, Canada
- Department of Human Genetics, McGill University, Montreal, QC H3A 2B4, Canada
| |
Collapse
|
43
|
Tanner CM, Ostrem JL. Parkinson's Disease. N Engl J Med 2024; 391:442-452. [PMID: 39083773 DOI: 10.1056/nejmra2401857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Affiliation(s)
- Caroline M Tanner
- From the Movement Disorders and Neuromodulation Center, Department of Neurology, Weill Institute for Neuroscience, University of California, San Francisco, San Francisco
| | - Jill L Ostrem
- From the Movement Disorders and Neuromodulation Center, Department of Neurology, Weill Institute for Neuroscience, University of California, San Francisco, San Francisco
| |
Collapse
|
44
|
Jonson C, Levine KS, Lake J, Hertslet L, Jones L, Patel D, Kim J, Bandres‐Ciga S, Terry N, Mata IF, Blauwendraat C, Singleton AB, Nalls MA, Yokoyama JS, Leonard HL. Assessing the lack of diversity in genetics research across neurodegenerative diseases: A systematic review of the GWAS Catalog and literature. Alzheimers Dement 2024; 20:5740-5756. [PMID: 39030740 PMCID: PMC11350004 DOI: 10.1002/alz.13873] [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: 01/19/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 07/22/2024]
Abstract
The under-representation of non-European cohorts in neurodegenerative disease genome-wide association studies (GWAS) hampers precision medicine efforts. Despite the inherent genetic and phenotypic diversity in these diseases, GWAS research consistently exhibits a disproportionate emphasis on participants of European ancestry. This study reviews GWAS up to 2022, focusing on non-European or multi-ancestry neurodegeneration studies. We conducted a systematic review of GWAS results and publications up to 2022, focusing on non-European or multi-ancestry neurodegeneration studies. Rigorous article inclusion and quality assessment methods were employed. Of 123 neurodegenerative disease (NDD) GWAS reviewed, 82% predominantly featured European ancestry participants. A single European study identified over 90 risk loci, compared to a total of 50 novel loci in identified in all non-European or multi-ancestry studies. Notably, only six of the loci have been replicated. The significant under-representation of non-European ancestries in NDD GWAS hinders comprehensive genetic understanding. Prioritizing genomic diversity in future research is crucial for advancing NDD therapies and understanding. HIGHLIGHTS: Eighty-two percent of neurodegenerative genome-wide association studies (GWAS) focus on Europeans. Only 6 of 50 novel neurodegenerative disease (NDD) genetic loci have been replicated. Lack of diversity significantly hampers understanding of NDDs. Increasing diversity in NDD genetic research is urgently required. New initiatives are aiming to enhance diversity in NDD research.
Collapse
Affiliation(s)
- Caroline Jonson
- Center for Alzheimer's and Related DementiasNational Institutes of HealthBethesdaMarylandUSA
- DataTecnica LLCWashingtonDistrict of ColumbiaUSA
- Pharmaceutical Sciences and Pharmacogenomics Graduate ProgramUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Kristin S. Levine
- Center for Alzheimer's and Related DementiasNational Institutes of HealthBethesdaMarylandUSA
- DataTecnica LLCWashingtonDistrict of ColumbiaUSA
| | - Julie Lake
- Center for Alzheimer's and Related DementiasNational Institutes of HealthBethesdaMarylandUSA
- Laboratory of NeurogeneticsNational Institutes on AgingNational Institutes of HealthBethesdaMarylandUSA
| | - Linnea Hertslet
- Center for Alzheimer's and Related DementiasNational Institutes of HealthBethesdaMarylandUSA
| | - Lietsel Jones
- Center for Alzheimer's and Related DementiasNational Institutes of HealthBethesdaMarylandUSA
- DataTecnica LLCWashingtonDistrict of ColumbiaUSA
| | - Dhairya Patel
- Integrative Neurogenomics UnitLaboratory of NeurogeneticsNational Institute on AgingNational Institutes of HealthBethesdaMarylandUSA
| | - Jeff Kim
- Center for Alzheimer's and Related DementiasNational Institutes of HealthBethesdaMarylandUSA
- Laboratory of NeurogeneticsNational Institutes on AgingNational Institutes of HealthBethesdaMarylandUSA
| | - Sara Bandres‐Ciga
- Center for Alzheimer's and Related DementiasNational Institutes of HealthBethesdaMarylandUSA
| | - Nancy Terry
- Division of Library ServicesOffice of Research ServicesNational Institutes of HealthBethesdaMarylandUSA
| | - Ignacio F. Mata
- Genomic Medicine Institute, Lerner Research Institute, Genomic MedicineCleveland Clinic FoundationClevelandOhioUSA
| | - Cornelis Blauwendraat
- Center for Alzheimer's and Related DementiasNational Institutes of HealthBethesdaMarylandUSA
- Integrative Neurogenomics UnitLaboratory of NeurogeneticsNational Institute on AgingNational Institutes of HealthBethesdaMarylandUSA
| | - Andrew B. Singleton
- Center for Alzheimer's and Related DementiasNational Institutes of HealthBethesdaMarylandUSA
- Laboratory of NeurogeneticsNational Institutes on AgingNational Institutes of HealthBethesdaMarylandUSA
| | - Mike A. Nalls
- Center for Alzheimer's and Related DementiasNational Institutes of HealthBethesdaMarylandUSA
- DataTecnica LLCWashingtonDistrict of ColumbiaUSA
- Laboratory of NeurogeneticsNational Institutes on AgingNational Institutes of HealthBethesdaMarylandUSA
| | - Jennifer S. Yokoyama
- Pharmaceutical Sciences and Pharmacogenomics Graduate ProgramUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Hampton L. Leonard
- Center for Alzheimer's and Related DementiasNational Institutes of HealthBethesdaMarylandUSA
- DataTecnica LLCWashingtonDistrict of ColumbiaUSA
- Laboratory of NeurogeneticsNational Institutes on AgingNational Institutes of HealthBethesdaMarylandUSA
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
| |
Collapse
|
45
|
Skrahin A, Horowitz M, Istaiti M, Skrahina V, Lukas J, Yahalom G, Cohen ME, Revel-Vilk S, Goker-Alpan O, Becker-Cohen M, Hassin-Baer S, Svenningsson P, Rolfs A, Zimran A. GBA1-Associated Parkinson's Disease Is a Distinct Entity. Int J Mol Sci 2024; 25:7102. [PMID: 39000225 PMCID: PMC11241486 DOI: 10.3390/ijms25137102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
GBA1-associated Parkinson's disease (GBA1-PD) is increasingly recognized as a distinct entity within the spectrum of parkinsonian disorders. This review explores the unique pathophysiological features, clinical progression, and genetic underpinnings that differentiate GBA1-PD from idiopathic Parkinson's disease (iPD). GBA1-PD typically presents with earlier onset and more rapid progression, with a poor response to standard PD medications. It is marked by pronounced cognitive impairment and a higher burden of non-motor symptoms compared to iPD. Additionally, patients with GBA1-PD often exhibit a broader distribution of Lewy bodies within the brain, accentuating neurodegenerative processes. The pathogenesis of GBA1-PD is closely associated with mutations in the GBA1 gene, which encodes the lysosomal enzyme beta-glucocerebrosidase (GCase). In this review, we discuss two mechanisms by which GBA1 mutations contribute to disease development: 'haploinsufficiency,' where a single functional gene copy fails to produce a sufficient amount of GCase, and 'gain of function,' where the mutated GCase acquires harmful properties that directly impact cellular mechanisms for alpha-synuclein degradation, leading to alpha-synuclein aggregation and neuronal cell damage. Continued research is advancing our understanding of how these mechanisms contribute to the development and progression of GBA1-PD, with the 'gain of function' mechanism appearing to be the most plausible. This review also explores the implications of GBA1 mutations for therapeutic strategies, highlighting the need for early diagnosis and targeted interventions. Currently, small molecular chaperones have shown the most promising clinical results compared to other agents. This synthesis of clinical, pathological, and molecular aspects underscores the assertion that GBA1-PD is a distinct clinical and pathobiological PD phenotype, necessitating specific management and research approaches to better understand and treat this debilitating condition.
Collapse
Affiliation(s)
- Aliaksandr Skrahin
- Rare Disease Consulting RCV GmbH, Leibnizstrasse 58, 10629 Berlin, Germany
| | - Mia Horowitz
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, 6997801 Ramat Aviv, Israel
| | - Majdolen Istaiti
- Gaucher Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Agyany Pharma Ltd., 9695614 Jerusalem, Israel
| | | | - Jan Lukas
- Translational Neurodegeneration Section Albrecht Kossel, Department of Neurology, University Medical Center Rostock, 18147 Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany
| | - Gilad Yahalom
- Department of Neurology and Movement Disorders Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, 9112102 Jerusalem, Israel
| | - Mikhal E. Cohen
- Department of Neurology and Movement Disorders Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, 9112102 Jerusalem, Israel
| | - Shoshana Revel-Vilk
- Gaucher Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, 9112102 Jerusalem, Israel
| | - Ozlem Goker-Alpan
- Lysosomal and Rare Disorders Research and Treatment Center, Fairfax, VA 22030, USA
| | | | - Sharon Hassin-Baer
- Movement Disorders Institute, Department of Neurology, Chaim Sheba Medical Center, 5262101 Tel-Hashomer, Israel
- Department of Neurology and Neurosurgery, Faculty of Medical and Health Sciences, Tel Aviv University, 6997801 Tel-Aviv, Israel
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
- Department of Basal and Clinical Neuroscience, King’s College London, London SE5 9RT, UK
| | - Arndt Rolfs
- Rare Disease Consulting RCV GmbH, Leibnizstrasse 58, 10629 Berlin, Germany
- Agyany Pharma Ltd., 9695614 Jerusalem, Israel
- Medical Faculty, University of Rostock, 18055 Rostock, Germany
| | - Ari Zimran
- Gaucher Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Agyany Pharma Ltd., 9695614 Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, 9112102 Jerusalem, Israel
| |
Collapse
|
46
|
Tan MMX, Lawton MA, Pollard MI, Brown E, Real R, Carrasco AM, Bekadar S, Jabbari E, Reynolds RH, Iwaki H, Blauwendraat C, Kanavou S, Hubbard L, Malek N, Grosset KA, Bajaj N, Barker RA, Burn DJ, Bresner C, Foltynie T, Wood NW, Williams-Gray CH, Andreassen OA, Toft M, Elbaz A, Artaud F, Brice A, Corvol JC, Aasly J, Farrer MJ, Nalls MA, Singleton AB, Williams NM, Ben-Shlomo Y, Hardy J, Hu MTM, Grosset DG, Shoai M, Pihlstrøm L, Morris HR. Genome-wide determinants of mortality and motor progression in Parkinson's disease. NPJ Parkinsons Dis 2024; 10:113. [PMID: 38849413 PMCID: PMC11161485 DOI: 10.1038/s41531-024-00729-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/24/2024] [Indexed: 06/09/2024] Open
Abstract
There are 90 independent genome-wide significant genetic risk variants for Parkinson's disease (PD) but currently only five nominated loci for PD progression. The biology of PD progression is likely to be of central importance in defining mechanisms that can be used to develop new treatments. We studied 6766 PD patients, over 15,340 visits with a mean follow-up of between 4.2 and 15.7 years and carried out genome-wide survival studies for time to a motor progression endpoint, defined by reaching Hoehn and Yahr stage 3 or greater, and death (mortality). There was a robust effect of the APOE ε4 allele on mortality in PD. We also identified a locus within the TBXAS1 gene encoding thromboxane A synthase 1 associated with mortality in PD. We also report 4 independent loci associated with motor progression in or near MORN1, ASNS, PDE5A, and XPO1. Only the non-Gaucher disease causing GBA1 PD risk variant E326K, of the known PD risk variants, was associated with mortality in PD. Further work is needed to understand the links between these genomic variants and the underlying disease biology. However, these may represent new candidates for disease modification in PD.
Collapse
Affiliation(s)
- Manuela M X Tan
- Department of Neurology, Oslo University Hospital, Oslo, Norway.
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK.
- UCL Movement Disorders Centre, University College London, London, UK.
| | - Michael A Lawton
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Miriam I Pollard
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK
| | - Emmeline Brown
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK
| | - Raquel Real
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Alejandro Martinez Carrasco
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Samir Bekadar
- Sorbonne University, Paris Brain Institute - ICM, Inserm, CNRS, Assistance Publique Hôpitaux de Paris, Departement of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Edwin Jabbari
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
| | - Regina H Reynolds
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Hirotaka Iwaki
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica, Washington DC, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Sofia Kanavou
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Leon Hubbard
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Naveed Malek
- Department of Neurology, Institute of Neurological Sciences, Queen Elizabeth University Hospital, Glasgow, UK
| | - Katherine A Grosset
- Department of Neurology, Institute of Neurological Sciences, Queen Elizabeth University Hospital, Glasgow, UK
| | - Nin Bajaj
- Clinical Neurosciences, University of Nottingham, Nottingham, UK
| | - Roger A Barker
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - David J Burn
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Catherine Bresner
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Thomas Foltynie
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
| | - Nicholas W Wood
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Caroline H Williams-Gray
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Ole A Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Mathias Toft
- Department of Neurology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Alexis Elbaz
- Paris-Saclay University, UVSQ, Inserm, Gustave Roussy, "Exposome and Heredity" team, CESP, 94807, Villejuif, France
| | - Fanny Artaud
- Paris-Saclay University, UVSQ, Inserm, Gustave Roussy, "Exposome and Heredity" team, CESP, 94807, Villejuif, France
| | - Alexis Brice
- Sorbonne University, Paris Brain Institute - ICM, Inserm, CNRS, Assistance Publique Hôpitaux de Paris, Departement of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Jean-Christophe Corvol
- Sorbonne University, Paris Brain Institute - ICM, Inserm, CNRS, Assistance Publique Hôpitaux de Paris, Departement of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Jan Aasly
- Department of Neurology, St. Olavs Hospital, Trondheim, Norway
- Department of Neuromedicine and Movement Science (INB), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Matthew J Farrer
- Department of Neurology, University of Florida, Gainesville, FL, USA
| | - Michael A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica, Washington DC, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Nigel M Williams
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Yoav Ben-Shlomo
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - John Hardy
- UCL Movement Disorders Centre, University College London, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Neurodegenerative Diseases, Queen Square Institute of Neurology, University College London, London, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute, University College London, London, UK
- National Institute for Health Research (NIHR) University College London Hospitals Biomedical Research Centre, London, UK
- Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Michele T M Hu
- Nuffield Department of Clinical Neurosciences, Division of Clinical Neurology, University of Oxford, Oxford, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
- Department of Clinical Neurology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Donald G Grosset
- School of Neuroscience and Psychology, University of Glasgow, Glasgow, UK
| | - Maryam Shoai
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Neurodegenerative Diseases, Queen Square Institute of Neurology, University College London, London, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London, UK
| | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK.
- UCL Movement Disorders Centre, University College London, London, UK.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
47
|
Deliz JR, Tanner CM, Gonzalez-Latapi P. Epidemiology of Parkinson's Disease: An Update. Curr Neurol Neurosci Rep 2024; 24:163-179. [PMID: 38642225 DOI: 10.1007/s11910-024-01339-w] [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] [Accepted: 04/12/2024] [Indexed: 04/22/2024]
Abstract
PURPOSE OF REVIEW In recent decades, epidemiological understanding of Parkinson disease (PD) has evolved significantly. Major discoveries in genetics and large epidemiological investigations have provided a better understanding of the genetic, behavioral, and environmental factors that play a role in the pathogenesis and progression of PD. In this review, we provide an epidemiological update of PD with a particular focus on advances in the last five years of published literature. RECENT FINDINGS We include an overview of PD pathophysiology, followed by a detailed discussion of the known distribution of disease and varied determinants of disease. We describe investigations of risk factors for PD, and provide a critical summary of current knowledge, knowledge gaps, and both clinical and research implications. We emphasize the need to characterize the epidemiology of the disease in diverse populations. Despite increasing understanding of PD epidemiology, recent paradigm shifts in the conceptualization of PD as a biological entity will also impact epidemiological research moving forward and guide further work in this field.
Collapse
Affiliation(s)
- Juan R Deliz
- Ken and Ruth Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Caroline M Tanner
- Weill Institute for Neurosciences, Department of Neurology, University of California -San Francisco, San Francisco, CA, USA
| | - Paulina Gonzalez-Latapi
- Ken and Ruth Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
48
|
Coukos R, Krainc D. Key genes and convergent pathogenic mechanisms in Parkinson disease. Nat Rev Neurosci 2024; 25:393-413. [PMID: 38600347 DOI: 10.1038/s41583-024-00812-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 04/12/2024]
Abstract
Parkinson disease (PD) is a neurodegenerative disorder marked by the preferential dysfunction and death of dopaminergic neurons in the substantia nigra. The onset and progression of PD is influenced by a diversity of genetic variants, many of which lack functional characterization. To identify the most high-yield targets for therapeutic intervention, it is important to consider the core cellular compartments and functional pathways upon which the varied forms of pathogenic dysfunction may converge. Here, we review several key PD-linked proteins and pathways, focusing on the mechanisms of their potential convergence in disease pathogenesis. These dysfunctions primarily localize to a subset of subcellular compartments, including mitochondria, lysosomes and synapses. We discuss how these pathogenic mechanisms that originate in different cellular compartments may coordinately lead to cellular dysfunction and neurodegeneration in PD.
Collapse
Affiliation(s)
- Robert Coukos
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dimitri Krainc
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
49
|
Saffie Awad P, Makarious MB, Elsayed I, Sanyaolu A, Wild Crea P, Schumacher Schuh AF, Levine KS, Vitale D, Korestky MJ, Kim J, Peixoto Leal T, Perinan MT, Dey S, Noyce AJ, Reyes-Palomares A, Rodriguez-Losada N, Foo JN, Mohamed W, Heilbron K, Norcliffe-Kaufmann L, Rizig M, Okubadejo N, Nalls M, Blauwendraat C, Singleton A, Leonard H, Mata IF, Bandres Ciga S. Insights into Ancestral Diversity in Parkinsons Disease Risk: A Comparative Assessment of Polygenic Risk Scores. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.11.28.23299090. [PMID: 38076954 PMCID: PMC10705647 DOI: 10.1101/2023.11.28.23299090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Objectives To evaluate and compare different polygenic risk score (PRS) models in predicting Parkinsons disease (PD) across diverse ancestries, focusing on identifying the most suitable approach for each population and potentially contributing to equitable advancements in precision medicine. Methods We constructed a total of 105 PRS across individual level data from seven diverse ancestries. First, a cross-ancestry conventional PRS comparison was implemented by utilizing the 90 known European risk loci with weighted effects from four independent summary statistics including European, East Asian, Latino/Admixed American, and African/Admixed. These models were adjusted by sex, age, and principal components (28 PRS) and by sex, age, and percentage of admixture (28 PRS) for comparison. Secondly, a novel and refined multi-ancestry best-fit PRS approach was then applied across the seven ancestries by leveraging multi-ancestry meta-analyzed summary statistics and using a p-value thresholding approach (49 PRS) to enhance prediction applicability in a global setting. Results European-based PRS models predicted disease status across all ancestries to differing degrees of accuracy. Ashkenazi Jewish had the highest Odds Ratio (OR): 1.96 (95% CI: 1.69-2.25, p < 0.0001) with an AUC (Area Under the Curve) of 68%. Conversely, the East Asian population, despite having fewer predictive variants (84 out of 90), had an OR of 1.37 (95% CI: 1.32-1.42) and an AUC of 62%, illustrating the cross-ancestry transferability of this model. Lower OR alongside broader confidence intervals were observed in other populations, including Africans (OR =1.38, 95% CI: 1.12-1.63, p=0.001). Adjustment by percentage of admixture did not outperform principal components. Multi-ancestry best-fit PRS models improved risk prediction in European, Ashkenazi Jewish, and African ancestries, yet didn't surpass conventional PRS in admixed populations such as Latino/American admixed and African admixed populations. Interpretation The present study represents a novel and comprehensive assessment of PRS performance across seven ancestries in PD, highlighting the inadequacy of a 'one size fits all' approach in genetic risk prediction. We demonstrated that European based PD PRS models are partially transferable to other ancestries and could be improved by a novel best-fit multi-ancestry PRS, especially in non-admixed populations.
Collapse
|
50
|
Khani M, Cerquera-Cleves C, Kekenadze M, Crea PAW, Singleton AB, Bandres-Ciga S. Towards a Global View of Parkinson's Disease Genetics. Ann Neurol 2024; 95:831-842. [PMID: 38557965 PMCID: PMC11060911 DOI: 10.1002/ana.26905] [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: 12/06/2023] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 04/04/2024]
Abstract
Parkinson's disease (PD) is a global health challenge, yet historically studies of PD have taken place predominantly in European populations. Recent genetics research conducted in non-European populations has revealed novel population-specific genetic loci linked to PD risk, highlighting the importance of studying PD globally. These insights have broadened our understanding of PD etiology, which is crucial for developing disease-modifying interventions. This review comprehensively explores the global genetic landscape of PD, emphasizing the scientific rationale for studying underrepresented populations. It underscores challenges, such as genotype-phenotype heterogeneity and inclusion difficulties for non-European participants, emphasizing the ongoing need for diverse and inclusive research in PD. ANN NEUROL 2024;95:831-842.
Collapse
Affiliation(s)
- Marzieh Khani
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Catalina Cerquera-Cleves
- Pontificia Universidad Javeriana, San Ignacio Hospital, Neurology Unit, Bogotá, Colombia
- CHU de Québec Research Center, Axe Neurosciences, Laval University. Quebec City, Canada
| | - Mariam Kekenadze
- Tbilisi State Medical University, Tbilisi, 0141, Georgia
- University College London, Queen Square Institute of Neurology , WC1N 3BG, London, UK
| | - Peter A. Wild Crea
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Andrew B. Singleton
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Sara Bandres-Ciga
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|