1
|
Towns C, Fang ZH, Tan MMX, Jasaityte S, Schmaderer TM, Stafford EJ, Pollard M, Tilney R, Hodgson M, Wu L, Labrum R, Hehir J, Polke J, Lange LM, Schapira AHV, Bhatia KP, Singleton AB, Blauwendraat C, Klein C, Houlden H, Wood NW, Jarman PR, Morris HR, Real R. Parkinson's families project: a UK-wide study of early onset and familial Parkinson's disease. NPJ Parkinsons Dis 2024; 10:188. [PMID: 39420034 PMCID: PMC11487259 DOI: 10.1038/s41531-024-00778-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 08/12/2024] [Indexed: 10/19/2024] Open
Abstract
The Parkinson's Families Project is a UK-wide study aimed at identifying genetic variation associated with familial and early-onset Parkinson's disease (PD). We recruited individuals with a clinical diagnosis of PD and age at motor symptom onset ≤45 years and/or a family history of PD in up to third-degree relatives. Where possible, we also recruited affected and unaffected relatives. We analysed DNA samples with a combination of single nucleotide polymorphism (SNP) array genotyping, multiplex ligation-dependent probe amplification (MLPA), and whole-genome sequencing (WGS). We investigated the association between identified pathogenic mutations and demographic and clinical factors such as age at motor symptom onset, family history, motor symptoms (MDS-UPDRS) and cognitive performance (MoCA). We performed baseline genetic analysis in 718 families, of which 205 had sporadic early-onset PD (sEOPD), 113 had familial early-onset PD (fEOPD), and 400 had late-onset familial PD (fLOPD). 69 (9.6%) of these families carried pathogenic variants in known monogenic PD-related genes. The rate of a molecular diagnosis increased to 28.1% in PD with motor onset ≤35 years. We identified pathogenic variants in LRRK2 in 4.2% of families, and biallelic pathogenic variants in PRKN in 3.6% of families. We also identified two families with SNCA duplications and three families with a pathogenic repeat expansion in ATXN2, as well as single families with pathogenic variants in VCP, PINK1, PNPLA6, PLA2G6, SPG7, GCH1, and RAB32. An additional 73 (10.2%) families were carriers of at least one pathogenic or risk GBA1 variant. Most early-onset and familial PD cases do not have a known genetic cause, indicating that there are likely to be further monogenic causes for PD.
Collapse
Affiliation(s)
- Clodagh Towns
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Zih-Hua Fang
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Manuela M X Tan
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Simona Jasaityte
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Theresa M Schmaderer
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Eleanor J Stafford
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Miriam Pollard
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Russel Tilney
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Megan Hodgson
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
| | - Lesley Wu
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Robyn Labrum
- Neurogenetics Laboratory, National Hospital for Neurology & Neurosurgery, Queen Square, London, UK
| | - Jason Hehir
- Neurogenetics Laboratory, National Hospital for Neurology & Neurosurgery, Queen Square, London, UK
| | - James Polke
- Neurogenetics Laboratory, National Hospital for Neurology & Neurosurgery, Queen Square, London, UK
| | - Lara M Lange
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Neurology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Kailash P Bhatia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
| | - 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
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 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, USA
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Nicholas W Wood
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Paul R Jarman
- National Hospital for Neurology & Neurosurgery, Queen Square, London, UK
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.
- UCL Movement Disorders Centre, University College London, London, UK.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Raquel Real
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.
- UCL Movement Disorders Centre, University College London, London, UK.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
2
|
Williams D, Glasstetter LM, Jong TT, Chen T, Kapoor A, Zhu S, Zhu Y, Calvo R, Gehrlein A, Wong K, Hogan AN, Vocadlo DJ, Jagasia R, Marugan JJ, Sidransky E, Henderson MJ, Chen Y. High-throughput screening for small-molecule stabilizers of misfolded glucocerebrosidase in Gaucher disease and Parkinson's disease. Proc Natl Acad Sci U S A 2024; 121:e2406009121. [PMID: 39388267 PMCID: PMC11494340 DOI: 10.1073/pnas.2406009121] [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: 09/10/2024] [Indexed: 10/12/2024] Open
Abstract
Glucocerebrosidase (GCase) is implicated in both a rare, monogenic disorder (Gaucher disease, GD) and a common, multifactorial condition (Parkinson's disease, PD); hence, it is an urgent therapeutic target. To identify correctors of severe protein misfolding and trafficking obstruction manifested by the pathogenic L444P-variant of GCase, we developed a suite of quantitative, high-throughput, cell-based assays. First, we labeled GCase with a small proluminescent HiBiT peptide reporter tag, enabling quantitation of protein stabilization in cells while faithfully maintaining target biology. TALEN-based gene editing allowed for stable integration of a single HiBiT-GBA1 transgene into an intragenic safe-harbor locus in GBA1-knockout H4 (neuroglioma) cells. This GD cell model was amenable to lead discovery via titration-based quantitative high-throughput screening and lead optimization via structure-activity relationships. A primary screen of 10,779 compounds from the NCATS bioactive collections identified 140 stabilizers of HiBiT-GCase-L444P, including both pharmacological chaperones (ambroxol and noninhibitory chaperone NCGC326) and proteostasis regulators (panobinostat, trans-ISRIB, and pladienolide B). Two complementary high-content imaging-based assays were deployed to triage hits: The fluorescence-quenched substrate LysoFix-GBA captured functional lysosomal GCase activity, while an immunofluorescence assay featuring antibody hGCase-1/23 directly visualized GCase lysosomal translocation. NCGC326 was active in both secondary assays and completely reversed pathological glucosylsphingosine accumulation. Finally, we tested the concept of combination therapy by demonstrating synergistic actions of NCGC326 with proteostasis regulators in enhancing GCase-L444P levels. Looking forward, these physiologically relevant assays can facilitate the identification, pharmacological validation, and medicinal chemistry optimization of small molecules targeting GCase, ultimately leading to a viable therapeutic for GD and PD.
Collapse
Affiliation(s)
- Darian Williams
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD20850
| | - Logan M. Glasstetter
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Tiffany T. Jong
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Tiffany Chen
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Abhijeet Kapoor
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD20850
| | - Sha Zhu
- Department of Chemistry, Simon Fraser University, Burnaby, BCV5A 1S6, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BCV5A 1S6, Canada
| | - Yanping Zhu
- Department of Chemistry, Simon Fraser University, Burnaby, BCV5A 1S6, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BCV5A 1S6, Canada
| | - Raul Calvo
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD20850
| | - Alexandra Gehrlein
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, 4070Basel, Switzerland
| | - Kimberly Wong
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Andrew N. Hogan
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - David J. Vocadlo
- Department of Chemistry, Simon Fraser University, Burnaby, BCV5A 1S6, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BCV5A 1S6, Canada
| | - Ravi Jagasia
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, 4070Basel, Switzerland
| | - Juan J. Marugan
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD20850
| | - Ellen Sidransky
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Mark J. Henderson
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD20850
| | - Yu Chen
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD20892
| |
Collapse
|
3
|
Kikuya A, Tsukita K, Sawamura M, Yoshimura K, Takahashi R. Distinct Clinical Implications of Patient- Versus Clinician-Rated Motor Symptoms in Parkinson's Disease. Mov Disord 2024; 39:1799-1808. [PMID: 39092513 DOI: 10.1002/mds.29962] [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: 05/11/2024] [Revised: 07/10/2024] [Accepted: 07/17/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Patient-rated motor symptoms (PRMS) and clinician-rated motor symptoms (CRMS) often differ in Parkinson's disease (PD). OBJECTIVE Our goal was to investigate the determinants and clinical implications of PRMS compared with CRMS in PD. METHODS This retrospective, observational cohort study analyzed the cross-sectional associations and longitudinal impacts of PRMS as assessed by the Movement Disorders Society-sponsored Unified PD Rating Scale (MDS-UPDRS) part 2, while controlling for CRMS measured by MDS-UPDRS part 3. Longitudinal analyses used Cox proportional hazards models and multiple linear mixed-effects random intercepts/slope models, adjusting for many clinical predictors. We conducted propensity score matching (PSM) to reinforce our analyses' robustness and surface-based morphometry to investigate neural correlates. RESULTS We enrolled 442 patients with early-stage PD. At baseline, regardless of CRMS, PRMS were associated with the severity of postural instability and gait disturbance (PIGD). Notably, PRMS independently and more accurately predicted faster long-term deterioration in motor function than CRMS (Hoehn and Yahr 4, adjusted hazard ratio per +1 point = 1.19 [95% confidence intervals, 1.08-1.32]), particularly in PIGD (PIGD subscore, β-interaction = 0.052 [95% confidence intervals, 0.018-0.086]). PSM confirmed these findings' robustness. Surface-based morphometry suggested that enhanced sensory processing was distinctively associated with PRMS. CONCLUSIONS In early-stage PD, PRMS weighed different aspects of symptoms and more effectively predicted motor deterioration compared to CRMS, with distinctive brain structural characteristics. The superior sensitivity of PRMS to subtle declines in drug-refractory symptoms like PIGD likely underlie our results, highlighting the importance of understanding the differential clinical implications of PRMS to prevent long-term motor deterioration. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Akihiro Kikuya
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuto Tsukita
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan
- Division of Sleep Medicine, Kansai Electric Power Medical Research Institute, Osaka, Japan
| | - Masanori Sawamura
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenji Yoshimura
- Department of Neurology, Osaka City General Hospital, Osaka, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
4
|
Gibson LL, Weintraub D, Lemmen R, Perera G, Chaudhuri KR, Svenningsson P, Aarsland D. Risk of Dementia in Parkinson's Disease: A Systematic Review and Meta-Analysis. Mov Disord 2024; 39:1697-1709. [PMID: 39036849 DOI: 10.1002/mds.29918] [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: 04/15/2024] [Revised: 06/17/2024] [Accepted: 06/24/2024] [Indexed: 07/23/2024] Open
Abstract
Estimates of the risk of dementia in Parkinson's disease (PDD) vary widely. We aimed to review the incidence of PDD and in a meta-analysis estimate the pooled annual incidence and relative risk of PDD while also exploring factors that may contribute to heterogeneity between studies. Preferred Reporting Items for Systematic reviews and Meta-Analyses guidelines were followed and MEDLINE and EMBASE were searched for articles reporting the number of cases of dementia in a population, followed longitudinally, with a minimum of 100 dementia-free Parkinson's disease (PD) patients at baseline. Meta-analyses and meta-regressions were used to estimate the pooled incidence rate of PDD and the relative risk of PDD versus healthy controls (HC). A total of 32 studies were identified, 25 reporting the incidence of PDD and 10 reporting the relative risk of PDD versus HC. The pooled incidence rate of PDD was 4.45 (95% confidence interval [CI], 3.91-4.99) per 100 person-years at risk, equating to a 4.5% annual risk of dementia in a PD prevalent population. The relative risk of PDD was estimated to be 3.25 (95% CI, 2.62-4.03) times greater than HC. Factors contributing to study heterogeneity and disparities in the estimated risk of PDD include the age of patients, year of recruitment, and study location. Significant gaps remain with no studies identified in several geographical regions. Future studies should stratify by age and standardize reporting to reduce overall heterogeneity. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Lucy L Gibson
- Centre for Healthy Brain Ageing, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
| | - Daniel Weintraub
- Department of Psychiatry and Neurology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
- Parkinson's Disease Research, Education and Clinical Center (PADRECC), Philadelphia Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Roos Lemmen
- Centre for Healthy Brain Ageing, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
| | - Gayan Perera
- Centre for Healthy Brain Ageing, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
| | - Kallol Ray Chaudhuri
- Department of Basic and Clinical Neuroscience, Parkinson Foundation International Centre of Excellence, Kings College Hospital and Kings College London, London, UK
| | - Per Svenningsson
- Basic and Clinical Neuroscience, King's College London, London, UK
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Dag Aarsland
- Centre for Healthy Brain Ageing, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
- Centre for Age-Related Disease, Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
5
|
Ribeiro J, Camacho M, Scott KM, Greenland JC, Evans JR, Breen DP, Wijeyekoon RS, Barker RA, Williams-Gray CH. Validation of a 5-Year Prognostic Model for Parkinson's Disease. Mov Disord Clin Pract 2024. [PMID: 39344276 DOI: 10.1002/mdc3.14215] [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: 07/08/2024] [Revised: 08/16/2024] [Accepted: 09/06/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND A simple prognostic model was previously developed to predict the probability of recently-diagnosed patients reaching negative outcomes (postural instability, dementia or death) in a 5-year period. OBJECTIVES To validate this model in an independent cohort and establish utility at later time points. METHODS Validation was performed using data collected in an incident cohort at baseline, 2 and 4 years. Predicted negative outcome probabilities were compared to actual 5-year outcomes. RESULTS The model, based on age, MDS-UPDRS axial score and 60-second animal fluency, predicted poor 5-year outcome when applied at baseline, (area under the curve (AUC) 0.80), 2 years (AUC 0.82) and 4 years (AUC 0.71). Power calculations showed that selecting a subgroup with prognostic score >0.5 reduced the sample size required for a disease-modifying trial. CONCLUSIONS This 5-year prognostic model has good accuracy when employed up to 4 years from diagnosis and may help stratification for disease-modifying trials.
Collapse
Affiliation(s)
- Joana Ribeiro
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Marta Camacho
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Kirsten M Scott
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Julia C Greenland
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - David P Breen
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, UK
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Ruwani S Wijeyekoon
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Roger A Barker
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
6
|
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
|
7
|
Shiner T, Kavé G, Mirelman A, Regev K, Piura Y, Goldstein O, Gana Weisz M, Bar-Shira A, Gurevich T, Orr-Urtreger A, Alcalay RN, Giladi N, Bregman N. Effect of GBA1 Mutations and APOE Polymorphisms on Survival and Progression Among Ashkenazi Jews with Dementia with Lewy Bodies. Mov Disord 2024. [PMID: 39212252 DOI: 10.1002/mds.30003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/28/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Glucocerebrosidase 1 (GBA1) mutations are associated with reduced survival in Parkinson's disease but their effect on survival in dementia with Lewy bodies (DLB) is unclear. OBJECTIVE To assess the impact of GBA1 mutations on survival among Ashkenazi Jews with DLB, while controlling for APOE status. METHODS One hundred and forty participants from Tel Aviv Medical Center, Israel were genotyped for GBA1 mutations and APOE polymorphisms. Survival rates and follow-up cognitive screening scores were analyzed. RESULTS GBA1 mutation carriers had a two-fold increased risk of death (HR = 1.999), while APOE status did not independently affect survival. In a subset of patients with available clinical data (N = 63), carriers of the APOE ε4 allele showed faster cognitive deterioration, while GBA1 mutation carriers also declined more rapidly albeit not significantly. CONCLUSION Understanding the genetic effects on survival and progression is crucial for patient counseling and inclusion in clinical trials. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Tamara Shiner
- Cognitive Neurology Unit, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Movement Disorders Unit, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Gitit Kavé
- Department of Education and Psychology, The Open University, Raanana, Israel
| | - Anat Mirelman
- Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Laboratory for Early Markers of Neurodegeneration (LEMON), Neurological Institute, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Keren Regev
- Neuroimmunology Unit, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Yoav Piura
- Cognitive Neurology Unit, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Orly Goldstein
- Laboratory of Biomarkers and Genomic of Neurodegeneration, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Mali Gana Weisz
- Laboratory of Biomarkers and Genomic of Neurodegeneration, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Anat Bar-Shira
- Genetic Laboratory, Genetic Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Tanya Gurevich
- Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Movement Disorders Unit, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Avi Orr-Urtreger
- Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Laboratory of Biomarkers and Genomic of Neurodegeneration, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Roy N Alcalay
- Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Movement Disorders Unit, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Laboratory of Biomarkers and Genomic of Neurodegeneration, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Nir Giladi
- Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Movement Disorders Unit, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Noa Bregman
- Cognitive Neurology Unit, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
8
|
Pisani S, Gosse L, Aarsland D, Ray Chaudhuri K, Ballard C, Ffytche D, Velayudhan L, Bhattacharyya S. Parkinson's disease psychosis associated with accelerated multidomain cognitive decline. BMJ MENTAL HEALTH 2024; 27:1-10. [PMID: 39043465 PMCID: PMC11268075 DOI: 10.1136/bmjment-2024-301062] [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: 03/05/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Cognitive deficits are associated with poor quality of life and increased risk of development of dementia in patients with Parkinson's disease (PD) psychosis. The trajectory of cognitive decline in PD psychosis remains however unclear. OBJECTIVE We examined this using data from the Parkinson's Progression Markers Initiative study. METHODS We analysed data from patients with drug-naïve PD (n=676) and healthy controls (HC, n=187) over 5 years, and examined all cognitive measures assessed at each time point. We classified patients with PD into those who developed psychosis over the course of the study (PDP) and those without psychosis throughout (PDnP) using the Movement Disorders Society Unified Parkinson's Disease Rating Scale part I hallucinations/psychosis item. We used linear mixed-effect models with restricted maximum likelihood. Age, sex, ethnicity, education and neuropsychiatric and PD-specific symptoms were entered as covariates of interest. FINDINGS There were no baseline cognitive differences between PD patient groups. There were differences in cognitive performance between PD and HC across the majority of the assessments.Patients with PDP exhibited greater cognitive decline over 5 years compared with PDnP across most domains even after controlling for sociodemographics, depression, sleepiness, rapid eye movement sleep behaviour disorder and motor symptom severity (immediate recall, b=-0.288, p=0.003; delayed recall, b=-0.146, p=0.003; global cognition, Montreal Cognitive Assessment, b=-0.206, p<0.001; visuospatial, b=-0.178, p=0.012; semantic fluency, b=-0.704, p=0.002; processing speed, b=-0.337, p=0.029). CONCLUSIONS Patients with PD psychosis exhibited decline in semantic aspects of language, processing speed, global cognition, visuospatial abilities and memory, regardless of sociodemographic characteristics, neuropsychiatric and motor symptoms. These cognitive domains, particularly semantic aspects of language may therefore play an important role in PD psychosis and warrant further investigation. TRIAL REGISTRATION NUMBER NCT01141023.
Collapse
Affiliation(s)
- Sara Pisani
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Luca Gosse
- Faculty of Medicine, Dentistry and Health, The University of Sheffield, Sheffield, UK
| | - Dag Aarsland
- Department of Psychological Medicine, Centre for Healthy Brain Ageing, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Stavanger, Norway
| | - K Ray Chaudhuri
- National Parkinson’s Foundation Centre of Excellence, King's College Hospital NHS Foundation Trust, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Clive Ballard
- Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Dominic Ffytche
- Department of Psychological Medicine, Centre for Healthy Brain Ageing, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Latha Velayudhan
- Department of Psychological Medicine, Centre for Healthy Brain Ageing, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Sagnik Bhattacharyya
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| |
Collapse
|
9
|
Young CB, Cholerton B, Smith AM, Shahid-Besanti M, Abdelnour C, Mormino EC, Hu SC, Chung KA, Peterson A, Rosenthal L, Pantelyat A, Dawson TM, Quinn J, Zabetian CP, Montine TJ, Poston KL. The Parkinson's Disease Composite of Executive Functioning: A Measure for Detecting Cognitive Decline in Clinical Trials. Neurology 2024; 103:e209609. [PMID: 38870440 PMCID: PMC11244747 DOI: 10.1212/wnl.0000000000209609] [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: 08/01/2023] [Accepted: 05/07/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Executive functioning is one of the first domains to be impaired in Parkinson disease (PD), and the majority of patients with PD eventually develop dementia. Thus, developing a cognitive endpoint measure specifically assessing executive functioning is critical for PD clinical trials. The objective of this study was to develop a cognitive composite measure that is sensitive to decline in executive functioning for use in PD clinical trials. METHODS We used cross-sectional and longitudinal follow-up data from PD participants enrolled in the PD Cognitive Genetics Consortium, a multicenter setting focused on PD. All PD participants with Trail Making Test, Digit Symbol, Letter-Number Sequencing, Semantic Fluency, and Phonemic Fluency neuropsychological data collected from March 2010 to February 2020 were included. Baseline executive functioning data were used to create the Parkinson's Disease Composite of Executive Functioning (PaCEF) through confirmatory factor analysis. We examined the changes in the PaCEF over time, how well baseline PaCEF predicts time to cognitive progression, and the required sample size estimates for PD clinical trials. PaCEF results were compared with the Montreal Cognitive Assessment (MoCA), individual tests forming the PaCEF, and tests of visuospatial, language, and memory functioning. RESULTS A total of 841 participants (251 no cognitive impairment [NCI], 480 mild cognitive impairment [MCI], and 110 dementia) with baseline data were included, of which the mean (SD) age was 67.1 (8.9) years and 270 were women (32%). Five hundred forty five PD participants had longitudinal neuropsychological data spanning 9 years (mean [SD] 4.5 [2.2] years) and were included in analyses examining cognitive decline. A 1-factor model of executive functioning with excellent fit (comparative fit index = 0.993, Tucker-Lewis index = 0.989, and root mean square error of approximation = 0.044) was used to calculate the PaCEF. The average annual change in PaCEF ranged from 0.246 points per year for PD-NCI participants who remained cognitively unimpaired to -0.821 points per year for PD-MCI participants who progressed to dementia. For PD-MCI, baseline PaCEF, but not baseline MoCA, significantly predicted time to dementia. Sample size estimates were 69%-73% smaller for PD-NCI trials and 16%-19% smaller for PD-MCI trials when using the PaCEF rather than MoCA as the endpoint. DISCUSSION The PaCEF is a sensitive measure of executive functioning decline in PD and will be especially beneficial for PD clinical trials.
Collapse
Affiliation(s)
- Christina B Young
- From the Departments of Neurology and Neurological Sciences (C.B.Y., A.M.S., M.S.-B., C.A., E.C.M., K.L.P.) and (B.C., T.J.M.), Stanford University School of Medicine, CA; Veterans Affairs Puget Sound Health Care System (B.C., S.-C.H., C.P.Z.), Seattle; Department of Neurology (S.-C.H., C.P.Z.), University of Washington School of Medicine, Seattle; Department of Neurology (K.A.C., A. Peterson, J.Q.), Oregon Health and Science University, Portland; Portland Veterans Affairs Health Care System (K.A.C., A. Peterson, J.Q.), Oregon; Department of Neurology (L.R., A. Pantelyat, T.M.D.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Brenna Cholerton
- From the Departments of Neurology and Neurological Sciences (C.B.Y., A.M.S., M.S.-B., C.A., E.C.M., K.L.P.) and (B.C., T.J.M.), Stanford University School of Medicine, CA; Veterans Affairs Puget Sound Health Care System (B.C., S.-C.H., C.P.Z.), Seattle; Department of Neurology (S.-C.H., C.P.Z.), University of Washington School of Medicine, Seattle; Department of Neurology (K.A.C., A. Peterson, J.Q.), Oregon Health and Science University, Portland; Portland Veterans Affairs Health Care System (K.A.C., A. Peterson, J.Q.), Oregon; Department of Neurology (L.R., A. Pantelyat, T.M.D.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alena M Smith
- From the Departments of Neurology and Neurological Sciences (C.B.Y., A.M.S., M.S.-B., C.A., E.C.M., K.L.P.) and (B.C., T.J.M.), Stanford University School of Medicine, CA; Veterans Affairs Puget Sound Health Care System (B.C., S.-C.H., C.P.Z.), Seattle; Department of Neurology (S.-C.H., C.P.Z.), University of Washington School of Medicine, Seattle; Department of Neurology (K.A.C., A. Peterson, J.Q.), Oregon Health and Science University, Portland; Portland Veterans Affairs Health Care System (K.A.C., A. Peterson, J.Q.), Oregon; Department of Neurology (L.R., A. Pantelyat, T.M.D.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Marian Shahid-Besanti
- From the Departments of Neurology and Neurological Sciences (C.B.Y., A.M.S., M.S.-B., C.A., E.C.M., K.L.P.) and (B.C., T.J.M.), Stanford University School of Medicine, CA; Veterans Affairs Puget Sound Health Care System (B.C., S.-C.H., C.P.Z.), Seattle; Department of Neurology (S.-C.H., C.P.Z.), University of Washington School of Medicine, Seattle; Department of Neurology (K.A.C., A. Peterson, J.Q.), Oregon Health and Science University, Portland; Portland Veterans Affairs Health Care System (K.A.C., A. Peterson, J.Q.), Oregon; Department of Neurology (L.R., A. Pantelyat, T.M.D.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Carla Abdelnour
- From the Departments of Neurology and Neurological Sciences (C.B.Y., A.M.S., M.S.-B., C.A., E.C.M., K.L.P.) and (B.C., T.J.M.), Stanford University School of Medicine, CA; Veterans Affairs Puget Sound Health Care System (B.C., S.-C.H., C.P.Z.), Seattle; Department of Neurology (S.-C.H., C.P.Z.), University of Washington School of Medicine, Seattle; Department of Neurology (K.A.C., A. Peterson, J.Q.), Oregon Health and Science University, Portland; Portland Veterans Affairs Health Care System (K.A.C., A. Peterson, J.Q.), Oregon; Department of Neurology (L.R., A. Pantelyat, T.M.D.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Elizabeth C Mormino
- From the Departments of Neurology and Neurological Sciences (C.B.Y., A.M.S., M.S.-B., C.A., E.C.M., K.L.P.) and (B.C., T.J.M.), Stanford University School of Medicine, CA; Veterans Affairs Puget Sound Health Care System (B.C., S.-C.H., C.P.Z.), Seattle; Department of Neurology (S.-C.H., C.P.Z.), University of Washington School of Medicine, Seattle; Department of Neurology (K.A.C., A. Peterson, J.Q.), Oregon Health and Science University, Portland; Portland Veterans Affairs Health Care System (K.A.C., A. Peterson, J.Q.), Oregon; Department of Neurology (L.R., A. Pantelyat, T.M.D.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Shu-Ching Hu
- From the Departments of Neurology and Neurological Sciences (C.B.Y., A.M.S., M.S.-B., C.A., E.C.M., K.L.P.) and (B.C., T.J.M.), Stanford University School of Medicine, CA; Veterans Affairs Puget Sound Health Care System (B.C., S.-C.H., C.P.Z.), Seattle; Department of Neurology (S.-C.H., C.P.Z.), University of Washington School of Medicine, Seattle; Department of Neurology (K.A.C., A. Peterson, J.Q.), Oregon Health and Science University, Portland; Portland Veterans Affairs Health Care System (K.A.C., A. Peterson, J.Q.), Oregon; Department of Neurology (L.R., A. Pantelyat, T.M.D.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kathryn A Chung
- From the Departments of Neurology and Neurological Sciences (C.B.Y., A.M.S., M.S.-B., C.A., E.C.M., K.L.P.) and (B.C., T.J.M.), Stanford University School of Medicine, CA; Veterans Affairs Puget Sound Health Care System (B.C., S.-C.H., C.P.Z.), Seattle; Department of Neurology (S.-C.H., C.P.Z.), University of Washington School of Medicine, Seattle; Department of Neurology (K.A.C., A. Peterson, J.Q.), Oregon Health and Science University, Portland; Portland Veterans Affairs Health Care System (K.A.C., A. Peterson, J.Q.), Oregon; Department of Neurology (L.R., A. Pantelyat, T.M.D.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Amie Peterson
- From the Departments of Neurology and Neurological Sciences (C.B.Y., A.M.S., M.S.-B., C.A., E.C.M., K.L.P.) and (B.C., T.J.M.), Stanford University School of Medicine, CA; Veterans Affairs Puget Sound Health Care System (B.C., S.-C.H., C.P.Z.), Seattle; Department of Neurology (S.-C.H., C.P.Z.), University of Washington School of Medicine, Seattle; Department of Neurology (K.A.C., A. Peterson, J.Q.), Oregon Health and Science University, Portland; Portland Veterans Affairs Health Care System (K.A.C., A. Peterson, J.Q.), Oregon; Department of Neurology (L.R., A. Pantelyat, T.M.D.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Liana Rosenthal
- From the Departments of Neurology and Neurological Sciences (C.B.Y., A.M.S., M.S.-B., C.A., E.C.M., K.L.P.) and (B.C., T.J.M.), Stanford University School of Medicine, CA; Veterans Affairs Puget Sound Health Care System (B.C., S.-C.H., C.P.Z.), Seattle; Department of Neurology (S.-C.H., C.P.Z.), University of Washington School of Medicine, Seattle; Department of Neurology (K.A.C., A. Peterson, J.Q.), Oregon Health and Science University, Portland; Portland Veterans Affairs Health Care System (K.A.C., A. Peterson, J.Q.), Oregon; Department of Neurology (L.R., A. Pantelyat, T.M.D.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alexander Pantelyat
- From the Departments of Neurology and Neurological Sciences (C.B.Y., A.M.S., M.S.-B., C.A., E.C.M., K.L.P.) and (B.C., T.J.M.), Stanford University School of Medicine, CA; Veterans Affairs Puget Sound Health Care System (B.C., S.-C.H., C.P.Z.), Seattle; Department of Neurology (S.-C.H., C.P.Z.), University of Washington School of Medicine, Seattle; Department of Neurology (K.A.C., A. Peterson, J.Q.), Oregon Health and Science University, Portland; Portland Veterans Affairs Health Care System (K.A.C., A. Peterson, J.Q.), Oregon; Department of Neurology (L.R., A. Pantelyat, T.M.D.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ted M Dawson
- From the Departments of Neurology and Neurological Sciences (C.B.Y., A.M.S., M.S.-B., C.A., E.C.M., K.L.P.) and (B.C., T.J.M.), Stanford University School of Medicine, CA; Veterans Affairs Puget Sound Health Care System (B.C., S.-C.H., C.P.Z.), Seattle; Department of Neurology (S.-C.H., C.P.Z.), University of Washington School of Medicine, Seattle; Department of Neurology (K.A.C., A. Peterson, J.Q.), Oregon Health and Science University, Portland; Portland Veterans Affairs Health Care System (K.A.C., A. Peterson, J.Q.), Oregon; Department of Neurology (L.R., A. Pantelyat, T.M.D.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Joseph Quinn
- From the Departments of Neurology and Neurological Sciences (C.B.Y., A.M.S., M.S.-B., C.A., E.C.M., K.L.P.) and (B.C., T.J.M.), Stanford University School of Medicine, CA; Veterans Affairs Puget Sound Health Care System (B.C., S.-C.H., C.P.Z.), Seattle; Department of Neurology (S.-C.H., C.P.Z.), University of Washington School of Medicine, Seattle; Department of Neurology (K.A.C., A. Peterson, J.Q.), Oregon Health and Science University, Portland; Portland Veterans Affairs Health Care System (K.A.C., A. Peterson, J.Q.), Oregon; Department of Neurology (L.R., A. Pantelyat, T.M.D.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Cyrus P Zabetian
- From the Departments of Neurology and Neurological Sciences (C.B.Y., A.M.S., M.S.-B., C.A., E.C.M., K.L.P.) and (B.C., T.J.M.), Stanford University School of Medicine, CA; Veterans Affairs Puget Sound Health Care System (B.C., S.-C.H., C.P.Z.), Seattle; Department of Neurology (S.-C.H., C.P.Z.), University of Washington School of Medicine, Seattle; Department of Neurology (K.A.C., A. Peterson, J.Q.), Oregon Health and Science University, Portland; Portland Veterans Affairs Health Care System (K.A.C., A. Peterson, J.Q.), Oregon; Department of Neurology (L.R., A. Pantelyat, T.M.D.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Thomas J Montine
- From the Departments of Neurology and Neurological Sciences (C.B.Y., A.M.S., M.S.-B., C.A., E.C.M., K.L.P.) and (B.C., T.J.M.), Stanford University School of Medicine, CA; Veterans Affairs Puget Sound Health Care System (B.C., S.-C.H., C.P.Z.), Seattle; Department of Neurology (S.-C.H., C.P.Z.), University of Washington School of Medicine, Seattle; Department of Neurology (K.A.C., A. Peterson, J.Q.), Oregon Health and Science University, Portland; Portland Veterans Affairs Health Care System (K.A.C., A. Peterson, J.Q.), Oregon; Department of Neurology (L.R., A. Pantelyat, T.M.D.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kathleen L Poston
- From the Departments of Neurology and Neurological Sciences (C.B.Y., A.M.S., M.S.-B., C.A., E.C.M., K.L.P.) and (B.C., T.J.M.), Stanford University School of Medicine, CA; Veterans Affairs Puget Sound Health Care System (B.C., S.-C.H., C.P.Z.), Seattle; Department of Neurology (S.-C.H., C.P.Z.), University of Washington School of Medicine, Seattle; Department of Neurology (K.A.C., A. Peterson, J.Q.), Oregon Health and Science University, Portland; Portland Veterans Affairs Health Care System (K.A.C., A. Peterson, J.Q.), Oregon; Department of Neurology (L.R., A. Pantelyat, T.M.D.), Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
10
|
Gorji A, Fathi Jouzdani A. Machine learning for predicting cognitive decline within five years in Parkinson's disease: Comparing cognitive assessment scales with DAT SPECT and clinical biomarkers. PLoS One 2024; 19:e0304355. [PMID: 39018311 PMCID: PMC11253925 DOI: 10.1371/journal.pone.0304355] [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: 10/12/2023] [Accepted: 05/08/2024] [Indexed: 07/19/2024] Open
Abstract
OBJECTIVE Parkinson's disease (PD) is an age-related neurodegenerative condition characterized mostly by motor symptoms. Although a wide range of non-motor symptoms (NMS) are frequently experienced by PD patients. One of the important and common NMS is cognitive impairment, which is measured using different cognitive scales. Monitoring cognitive impairment and its decline in PD is essential for patient care and management. In this study, our goal is to identify the most effective cognitive scale in predicting cognitive decline over a 5-year timeframe initializing clinical biomarkers and DAT SPECT. METHODS Machine Learning has previously shown superior performance in image and clinical data classification and detection. In this study, we propose to use machine learning with different types of data, such as DAT SPECT and clinical biomarkers, to predict PD-CD based on various cognitive scales. We collected 330 DAT SPECT images and their clinical data in baseline, years 2,3,4, and 5 from Parkinson's Progression Markers Initiative (PPMI). We then designed a 3D Autoencoder to extract deep radiomic features (DF) from DAT SPECT images, and we then concatenated it with 17 clinical features (CF) to predict cognitive decline based on Montreal Cognitive Assessment (MoCA) and The Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS-I). RESULTS The utilization of MoCA as a cognitive decline scale yielded better performance in various years compared to MDS-UPDRS-I. In year 4, the application of the deep radiomic feature resulted in the highest achievement, with a cross-validation AUC of 89.28, utilizing the gradient boosting classifier. For the MDS-UPDRS-I scale, the highest achievement was obtained by utilizing the deep radiomic feature, resulting in a cross-validation AUC of 81.34 with the random forest classifier. CONCLUSIONS The study findings indicate that the MoCA scale may be a more effective predictor of cognitive decline within 5 years compared to MDS-UPDRS-I. Furthermore, deep radiomic features had better performance compared to sole clinical biomarkers or clinical and deep radiomic combined. These results suggest that using the MoCA score and deep radiomic features extracted from DAT SPECT could be a promising approach for identifying individuals at risk for cognitive decline in four years. Future research is needed to validate these findings and explore their utility in clinical practice.
Collapse
Affiliation(s)
- Arman Gorji
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Neuroscience and Artificial Intelligence Research Group (NAIRG), Hamadan University of Medical Sciences, Hamadan, Iran
- USERN Office, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Fathi Jouzdani
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Neuroscience and Artificial Intelligence Research Group (NAIRG), Hamadan University of Medical Sciences, Hamadan, Iran
- USERN Office, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
11
|
Chang CW, Tan CH, Hong WP, Yu RL. GBA moderates cognitive reserve's effect on cognitive function in patients with Parkinson's disease. J Neurol 2024; 271:4392-4405. [PMID: 38656622 DOI: 10.1007/s00415-024-12374-5] [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: 12/04/2023] [Revised: 04/02/2024] [Accepted: 04/05/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Cognitive reserve (CR) involves an individual's ability to maintain cognitive vitality over their lifespan. Glucocerebrosidase (GBA) gene mutations contribute to additional effects on cognitive function in Parkinson's disease (PD) patients, but the interplay between GBA mutations and CR remains unclear. We investigated the interactions among CR, GBA, and diseases, aiming to examine whether the CR established at different stages interacts with specific genotypes to affect cognitive function. METHODS Three hundred and eighteen participants' CR indicators (i.e., education, occupation, and social function) and comprehensive neuropsychological function (i.e., tests for executive function, attention/working memory, visuospatial function, memory, and language) were evaluated. RESULTS We found that CR established in a specific life stage influences the individual's cognitive function, particularly in PD, based on their distinct GBA rs9628662 genotypes. Attention/working memory and memory performance are affected by occupational complexity in midlife in PD patients with the GG genotype (q < 0.0001; q < 0.0001) and healthy adults with the T genotype (q = 0.0440; q < 0.0001). Language is influenced by early education and occupation, and the effects of occupation are also observed in PD patients with the GG genotype (q = 0.0040) and in healthy adults carrying the T genotype (q = 0.0040). CONCLUSIONS CR, established at different life stages, can be influenced by the GBA rs9628662 genotype, impacting later-life cognition. Validating genotypes and incorporating genotype information when assessing cognitive reserve effects is crucial and can enhance targeted cognitive training.
Collapse
Affiliation(s)
- Chia-Wen Chang
- Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chun-Hsiang Tan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wei-Pin Hong
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Rwei-Ling Yu
- Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Office of Strategic Planning, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
12
|
Cao LX, Kong WL, Chan P, Zhang W, Morris MJ, Huang Y. Assessment tools for cognitive performance in Parkinson's disease and its genetic contributors. Front Neurol 2024; 15:1413187. [PMID: 38988604 PMCID: PMC11233456 DOI: 10.3389/fneur.2024.1413187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/14/2024] [Indexed: 07/12/2024] Open
Abstract
Background We have shown that genetic factors associating with motor progression of Parkinson's disease (PD), but their roles in cognitive function is poorly understood. One reason is that while cognitive performance in PD can be evaluated by various cognitive scales, there is no definitive guide indicating which tool performs better. Methods Data were obtained from the Parkinson's Progression Markers Initiative, where cognitive performance was assessed using five cognitive screening tools, including Symbol Digit Modalities Test (SDMT), Montreal Cognitive Assessment, Benton Judgment of Line Orientation, Modified Semantic Fluency Test, and Letter Number Sequencing Test, at baseline and subsequent annual follow-up visit for 5 years. Genetic data including ApoE and other PD risk genetic information were also obtained. We used SPSS-receiver operating characteristic and ANOVA repeated measures to evaluate which cognitive assessment is the best reflecting cognitive performance in PD at early stage and over time. Logistic regression analyses were used to determine the genetic associations with the rapidity of cognitive decline in PD. Results SDMT performed better in detecting mild cognitive impairment at baseline (AUC = 0.763), and SDMT was the only tool showing a steady cognitive decline during longitudinal observation. Multigenetic factors significantly associated with cognitive impairment at early stage of the disease (AUC = 0.950) with IP6K2 rs12497850 more evident, and a significantly faster decline (AUC = 0.831) within 5 years after motor onset, particularly in those carrying FGF20 rs591323. Conclusion SDMT is a preferable cognitive assessment tool for PD and genetic factors synergistically contribute to the cognitive dysfunction in PD.
Collapse
Affiliation(s)
- Ling-Xiao Cao
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wee Lee Kong
- Pharmacology Department, School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Piu Chan
- Department of Neurobiology, Neurology and Geriatrics, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wei Zhang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Margaret J. Morris
- Pharmacology Department, School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Yue Huang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Pharmacology Department, School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
13
|
Zhu SG, Chen ZL, Xiao K, Wang ZW, Lu WB, Liu RP, Huang SS, Zhu JH, Zhang X, Wang JY. Association analyses of apolipoprotein E genotypes and cognitive performance in patients with Parkinson's disease. Eur J Med Res 2024; 29:334. [PMID: 38880878 PMCID: PMC11181540 DOI: 10.1186/s40001-024-01924-2] [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: 03/30/2024] [Accepted: 06/06/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Cognitive impairment is a common non-motor symptom of Parkinson's disease (PD). The apolipoprotein E (APOE) ε4 genotype increases the risk of Alzheimer's disease (AD). However, the effect of APOEε4 on cognitive function of PD patients remains unclear. In this study, we aimed to understand whether and how carrying APOEε4 affects cognitive performance in patients with early-stage and advanced PD. METHODS A total of 119 Chinese early-stage PD patients were recruited. Movement Disorder Society Unified Parkinson's Disease Rating Scale, Hamilton anxiety scale, Hamilton depression scale, non-motor symptoms scale, Mini-mental State Examination, Montreal Cognitive Assessment, and Fazekas scale were evaluated. APOE genotypes were determined by polymerase chain reactions and direct sequencing. Demographic and clinical information of 521 early-stage and 262 advanced PD patients were obtained from Parkinson's Progression Marker Initiative (PPMI). RESULTS No significant difference in cognitive performance was found between ApoEε4 carriers and non-carriers in early-stage PD patients from our cohort and PPMI. The cerebrospinal fluid (CSF) Amyloid Beta 42 (Aβ42) level was significantly lower in ApoEε4 carrier than non-carriers in early-stage PD patients from PPMI. In advanced PD patients from PPMI, the BJLOT, HVLT retention and SDMT scores seem to be lower in ApoEε4 carriers without reach the statistical significance. CONCLUSIONS APOEε4 carriage does not affect the cognitive performance of early-stage PD patients. However, it may promote the decline of CSF Aβ42 level and the associated amyloidopathy, which is likely to further contribute to the cognitive dysfunction of PD patients in the advanced stage.
Collapse
Affiliation(s)
- Shi-Guo Zhu
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Zhu-Ling Chen
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Ke Xiao
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Zi-Wei Wang
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Wen-Bin Lu
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Rong-Pei Liu
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Shi-Shi Huang
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Jian-Hong Zhu
- Department of Preventive Medicine, Institute of Nutrition and Diseases, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Xiong Zhang
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Jian-Yong Wang
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| |
Collapse
|
14
|
Szlepák T, Kossev AP, Csabán D, Illés A, Udvari S, Balicza P, Borsos B, Takáts A, Klivényi P, Molnár MJ. GBA-associated Parkinson's disease in Hungary: clinical features and genetic insights. Neurol Sci 2024; 45:2671-2679. [PMID: 38153678 PMCID: PMC11082009 DOI: 10.1007/s10072-023-07213-w] [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: 07/23/2023] [Accepted: 11/16/2023] [Indexed: 12/29/2023]
Abstract
INTRODUCTION Parkinson's disease (PD) has a complex genetic background involving both rare and common genetic variants. Although a small percentage of cases show a clear Mendelian inheritance pattern, it is much more relevant to identify patients who present with a complex genetic profile of risk variants with different severity. The ß-glucocerebrosidase coding gene (GBA1) is recognized as the most frequent genetic risk factor for PD and Lewy body dementia, irrespective of reduction of the enzyme activity due to genetic variants. METHODS In a selected cohort of 190 Hungarian patients with clinical signs of PD and suspected genetic risk, we performed the genetic testing of the GBA1 gene. As other genetic hits can modify clinical features, we also screened for additional rare variants in other neurodegenerative genes and assessed the APOE-ε genotype of the patients. RESULTS In our cohort, we identified 29 GBA1 rare variant (RV) carriers. Out of the six different detected RVs, the highly debated E365K and T408M variants are composed of the majority of them (22 out of 32). Three patients carried two GBA1 variants, and an additional three patients carried rare variants in other neurodegenerative genes (SMPD1, SPG11, and SNCA). We did not observe differences in age at onset or other clinical features of the patients carrying two GBA1 variants or patients carrying heterozygous APOE-ε4 allele. CONCLUSION We need further studies to better understand the drivers of clinical differences in these patients, as this could have important therapeutic implications.
Collapse
Affiliation(s)
- Tamás Szlepák
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
- HUN-REN, Multiomic Neurodegeneration Research Group, Budapest, Hungary
| | - Annabel P Kossev
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
| | - Dóra Csabán
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
| | - Anett Illés
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Szabolcs Udvari
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
| | - Péter Balicza
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
- HUN-REN, Multiomic Neurodegeneration Research Group, Budapest, Hungary
| | - Beáta Borsos
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
| | - Annamária Takáts
- Department of Neurology, Semmelweis University, Budapest, Hungary
| | - Péter Klivényi
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Mária J Molnár
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary.
- HUN-REN, Multiomic Neurodegeneration Research Group, Budapest, Hungary.
| |
Collapse
|
15
|
Mulroy E, Erro R, Bhatia KP, Hallett M. Refining the clinical diagnosis of Parkinson's disease. Parkinsonism Relat Disord 2024; 122:106041. [PMID: 38360507 PMCID: PMC11069446 DOI: 10.1016/j.parkreldis.2024.106041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/17/2024]
Abstract
Our ability to define, understand, and classify Parkinson's disease (PD) has undergone significant changes since the disorder was first described in 1817. Clinical features and neuropathologic signatures can now be supplemented by in-vivo interrogation of genetic and biological substrates of disease, offering great opportunity for further refining the diagnosis of PD. In this mini-review, we discuss the historical perspectives which shaped our thinking surrounding the definition and diagnosis of PD. We highlight the clinical, genetic, pathologic and biologic diversity which underpins the condition, and proceed to discuss how recent developments in our ability to define biologic and pathologic substrates of disease might impact PD definition, diagnosis, individualised prognostication, and personalised clinical care. We argue that Parkinson's 'disease', as currently diagnosed in the clinic, is actually a syndrome. It is the outward manifestation of any array of potential dysfunctional biologic processes, neuropathological changes, and disease aetiologies, which culminate in common outward clinical features which we term PD; each person has their own unique disease, which we can now define with increasing precision. This is an exciting time in PD research and clinical care. Our ability to refine the clinical diagnosis of PD, incorporating in-vivo assessments of disease biology, neuropathology, and neurogenetics may well herald the era of biologically-based, precision medicine approaches PD management. With this however comes a number of challenges, including how to integrate these technologies into clinical practice in a way which is acceptable to patients, promotes meaningful changes to care, and minimises health economic impact.
Collapse
Affiliation(s)
- Eoin Mulroy
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Roberto Erro
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, (SA), Italy
| | - Kailash P Bhatia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Mark Hallett
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
16
|
Williams D, Glasstetter LM, Jong TT, Kapoor A, Zhu S, Zhu Y, Gehrlein A, Vocadlo DJ, Jagasia R, Marugan JJ, Sidransky E, Henderson MJ, Chen Y. Development of quantitative high-throughput screening assays to identify, validate, and optimize small-molecule stabilizers of misfolded β-glucocerebrosidase with therapeutic potential for Gaucher disease and Parkinson's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.22.586364. [PMID: 38712038 PMCID: PMC11071283 DOI: 10.1101/2024.03.22.586364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Glucocerebrosidase (GCase) is implicated in both a rare, monogenic disorder (Gaucher disease, GD) and a common, multifactorial condition (Parkinson's disease); hence, it is an urgent therapeutic target. To identify correctors of severe protein misfolding and trafficking obstruction manifested by the pathogenic L444P-variant of GCase, we developed a suite of quantitative, high-throughput, cell-based assays. First, we labeled GCase with a small pro-luminescent HiBiT peptide reporter tag, enabling quantitation of protein stabilization in cells while faithfully maintaining target biology. TALEN-based gene editing allowed for stable integration of a single HiBiT-GBA1 transgene into an intragenic safe-harbor locus in GBA1-knockout H4 (neuroglioma) cells. This GD cell model was amenable to lead discovery via titration-based quantitative high-throughput screening and lead optimization via structure-activity relationships. A primary screen of 10,779 compounds from the NCATS bioactive collections identified 140 stabilizers of HiBiT-GCase-L444P, including both pharmacological chaperones (ambroxol and non-inhibitory chaperone NCGC326) and proteostasis regulators (panobinostat, trans-ISRIB, and pladienolide B). Two complementary high-content imaging-based assays were deployed to triage hits: the fluorescence-quenched substrate LysoFix-GBA captured functional lysosomal GCase activity, while an immunofluorescence assay featuring antibody hGCase-1/23 provided direct visualization of GCase lysosomal translocation. NCGC326 was active in both secondary assays and completely reversed pathological glucosylsphingosine accumulation. Finally, we tested the concept of combination therapy, by demonstrating synergistic actions of NCGC326 with proteostasis regulators in enhancing GCase-L444P levels. Looking forward, these physiologically-relevant assays can facilitate the identification, pharmacological validation, and medicinal chemistry optimization of new chemical matter targeting GCase, ultimately leading to a viable therapeutic for two protein-misfolding diseases.
Collapse
Affiliation(s)
- Darian Williams
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Logan M. Glasstetter
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Tiffany T. Jong
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Abhijeet Kapoor
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Sha Zhu
- Department of Chemistry and Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Yanping Zhu
- Department of Chemistry and Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Alexandra Gehrlein
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - David J. Vocadlo
- Department of Chemistry and Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Ravi Jagasia
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - Juan J. Marugan
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Ellen Sidransky
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Mark J. Henderson
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Yu Chen
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
17
|
Kouli A, Spindler LRB, Fryer TD, Hong YT, Malpetti M, Aigbirhio FI, White SR, Camacho M, O’Brien JT, Williams-Gray CH. Neuroinflammation is linked to dementia risk in Parkinson's disease. Brain 2024; 147:923-935. [PMID: 37757857 PMCID: PMC10907093 DOI: 10.1093/brain/awad322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/09/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The development of dementia is a devastating aspect of Parkinson's disease (PD), affecting nearly half of patients within 10 years post-diagnosis. For effective therapies to prevent and slow progression to PD dementia (PDD), the key mechanisms that determine why some people with PD develop early dementia, while others remain cognitively unaffected, need to be understood. Neuroinflammation and tau protein accumulation have been demonstrated in post-mortem PD brains, and in many other neurodegenerative disorders leading to dementia. However, whether these processes mediate dementia risk early on in the PD disease course is not established. To this end, we used PET neuroimaging with 11C-PK11195 to index neuroinflammation and 18F-AV-1451 for misfolded tau in early PD patients, stratified according to dementia risk in our 'Neuroinflammation and Tau Accumulation in Parkinson's Disease Dementia' (NET-PDD) study. The NET-PDD study longitudinally assesses newly-diagnosed PD patients in two subgroups at low and high dementia risk (stratified based on pentagon copying, semantic fluency, MAPT genotype), with comparison to age- and sex-matched controls. Non-displaceable binding potential (BPND) in 43 brain regions (Hammers' parcellation) was compared between groups (pairwise t-tests), and associations between BPND of the tracers tested (linear-mixed-effect models). We hypothesized that people with higher dementia risk have greater inflammation and/or tau accumulation in advance of significant cognitive decline. We found significantly elevated neuroinflammation (11C-PK11195 BPND) in multiple subcortical and restricted cortical regions in the high dementia risk group compared with controls, while in the low-risk group this was limited to two cortical areas. The high dementia risk group also showed significantly greater neuroinflammation than the low-risk group concentrated on subcortical and basal ganglia regions. Neuroinflammation in most of these regions was associated with worse cognitive performance (Addenbrooke's Cognitive Examination-III score). Overall neuroinflammation burden also correlated with serum levels of pro-inflammatory cytokines. In contrast, increases in 18F-AV-1451 (tau) BPND in PD versus controls were restricted to subcortical regions where off-target binding is typically seen, with no relationship to cognition found. Whole-brain 18F-AV-1451 burden correlated with serum phosphorylated tau181 levels. Although there was minimal regional tau accumulation in PD, regional neuroinflammation and tau burden correlated in PD participants, with the strongest association in the high dementia risk group, suggesting possible co-localization of these pathologies. In conclusion, our findings suggest that significant regional neuroinflammation in early PD might underpin higher risk for PDD development, indicating neuroinflammation as a putative early modifiable aetiopathological disease factor to prevent or slow dementia development using immunomodulatory strategies.
Collapse
Affiliation(s)
- Antonina Kouli
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK
| | - Lennart R B Spindler
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK
| | - Tim D Fryer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Young T Hong
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Maura Malpetti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK
| | - Franklin I Aigbirhio
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Simon R White
- Medical Research Council Biostatistics Unit, Cambridge Institute of Public Health, Cambridge, CB2 0SL, UK
| | - Marta Camacho
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK
| | - John T O’Brien
- Department of Psychiatry, University of Cambridge, Cambridge, CB2 0SZ, UK
| | | |
Collapse
|
18
|
Tunold JA, Tan MMX, Toft M, Ross O, van de Berg WDJ, Pihlstrøm L. Lysosomal Polygenic Burden Drives Cognitive Decline in Parkinson's Disease with Low Alzheimer Risk. Mov Disord 2024; 39:596-601. [PMID: 38124396 DOI: 10.1002/mds.29698] [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: 09/07/2023] [Revised: 11/22/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Genetics influence cognitive progression in Parkinson's disease, possibly through mechanisms related to Lewy and Alzheimer's disease pathology. Lysosomal polygenic burden has recently been linked to more severe Lewy pathology post mortem. OBJECTIVES To assess the influence of lysosomal polygenic burden on cognitive progression in Parkinson's disease patients with low Alzheimer's disease risk. METHODS Using Cox regression we assessed association between lysosomal polygenic scores and time to Montreal Cognitive Assessment score ≤ 21 in the Parkinson's Progression Markers Initiative cohort (n = 374), with replication in data from the Parkinson's Disease Biomarker Program (n = 777). Patients were stratified by Alzheimer's disease polygenic risk. RESULTS The lysosomal polygenic score was associated with faster progression of cognitive decline in patients with low Alzheimer's disease risk in both datasets (P = 0.0032 and P = 0.0054, respectively). CONCLUSION Our study supports complex interplay between genetics and neuropathology in Parkinson's disease-related cognitive impairment, emphasizing the role of lysosomal polygenic burden. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jon-Anders Tunold
- Department of Neurology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Manuela M X Tan
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Mathias Toft
- Department of Neurology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Owen Ross
- Mayo Clinic, Department of Neuroscience, Jacksonville, Florida, USA
| | - Wilma D J van de Berg
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
19
|
Slingerland S, van der Zee S, Carli G, Slomp AC, Boertien JM, d’Angremont E, Bohnen NI, Albin RL, van Laar T. Cholinergic innervation topography in GBA-associated de novo Parkinson's disease patients. Brain 2024; 147:900-910. [PMID: 37748026 PMCID: PMC10907081 DOI: 10.1093/brain/awad323] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/23/2023] [Accepted: 09/12/2023] [Indexed: 09/27/2023] Open
Abstract
The most common genetic risk factors for Parkinson's disease are GBA1 mutations, encoding the lysosomal enzyme glucocerebrosidase. Patients with GBA1 mutations (GBA-PD) exhibit earlier age of onset and faster disease progression with more severe cognitive impairments, postural instability and gait problems. These GBA-PD features suggest more severe cholinergic system pathologies. PET imaging with the vesicular acetylcholine transporter ligand 18F-F-fluoroethoxybenzovesamicol (18F-FEOBV PET) provides the opportunity to investigate cholinergic changes and their relationship to clinical features in GBA-PD. The study investigated 123 newly diagnosed, treatment-naïve Parkinson's disease subjects-with confirmed presynaptic dopaminergic deficits on PET imaging. Whole-gene GBA1 sequencing of saliva samples was performed to evaluate GBA1 variants. Patients underwent extensive neuropsychological assessment of all cognitive domains, motor evaluation with the Unified Parkinson's Disease Rating Scale, brain MRI, dopaminergic PET to measure striatal-to-occipital ratios of the putamen and 18F-FEOBV PET. We investigated differences in regional cholinergic innervation between GBA-PD carriers and non-GBA1 mutation carriers (non-GBA-PD), using voxel-wise and volume of interest-based approaches. The degree of overlap between t-maps from two-sample t-test models was quantified using the Dice similarity coefficient. Seventeen (13.8%) subjects had a GBA1 mutation. No significant differences were found in clinical features and dopaminergic ratios between GBA-PD and non-GBA-PD at diagnosis. Lower 18F-FEOBV binding was found in both the GBA-PD and non-GBA-PD groups compared to controls. Dice (P < 0.05, cluster size 100) showed good overlap (0.7326) between the GBA-PD and non-GBA-PD maps. GBA-PD patients showed more widespread reduction in 18F-FEOBV binding than non-GBA-PD when compared to controls in occipital, parietal, temporal and frontal cortices (P < 0.05, FDR-corrected). In volume of interest analyses (Bonferroni corrected), the left parahippocampal gyrus was more affected in GBA-PD. De novo GBA-PD show a distinct topography of regional cholinergic terminal ligand binding. Although the Parkinson's disease groups were not distinguishable clinically, in comparison to healthy controls, GBA-PD showed more extensive cholinergic denervation compared to non-GBA-PD. A larger group is needed to validate these findings. Our results suggest that de novo GBA-PD and non-GBA-PD show differential patterns of cholinergic system changes before clinical phenotypic differences between carriers versus non-carrier groups are observable.
Collapse
Affiliation(s)
- Sofie Slingerland
- Department of Neurology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Sygrid van der Zee
- Department of Neurology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- Department of Neurology, Division of Clinical Neuropsychology, University of Groningen, University Medical Center, 9713 GZ Groningen, The Netherlands
| | - Giulia Carli
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Anne C Slomp
- Department of Neurology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- Department of Neurology, Division of Clinical Neuropsychology, University of Groningen, University Medical Center, 9713 GZ Groningen, The Netherlands
| | - Jeffrey M Boertien
- Department of Neurology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Emile d’Angremont
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Nicolaas I Bohnen
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
- Neurology Service and GRECC, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
- Morris K. Udall Center of Excellence for Parkinson’s Disease Research, University of Michigan, Ann Arbor, MI 48109, USA
- Parkinson’s Foundation Research Center of Excellence, University of Michigan, Ann Arbor, MI 48109, USA
| | - Roger L Albin
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Neurology Service and GRECC, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
- Morris K. Udall Center of Excellence for Parkinson’s Disease Research, University of Michigan, Ann Arbor, MI 48109, USA
- Parkinson’s Foundation Research Center of Excellence, University of Michigan, Ann Arbor, MI 48109, USA
| | - Teus van Laar
- Department of Neurology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
20
|
Cooper O, Hallett P, Isacson O. Upstream lipid and metabolic systems are potential causes of Alzheimer's disease, Parkinson's disease and dementias. FEBS J 2024; 291:632-645. [PMID: 36165619 PMCID: PMC10040476 DOI: 10.1111/febs.16638] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/02/2022] [Accepted: 09/26/2022] [Indexed: 11/28/2022]
Abstract
Brain health requires circuits, cells and molecular pathways to adapt when challenged and to promptly reset once the challenge has resolved. Neurodegeneration occurs when adaptability becomes confined, causing challenges to overwhelm neural circuitry. Studies of rare and common neurodegenerative diseases suggest that the accumulation of lipids can compromise circuit adaptability. Using microglia as an example, we review data that suggest increased lipid concentrations cause dysfunctional inflammatory responses to immune challenges, leading to Alzheimer's disease, Parkinson's disease and dementia. We highlight current approaches to treat lipid metabolic and clearance pathways and identify knowledge gaps towards restoring adaptive homeostasis in individuals who are at-risk of losing cognition.
Collapse
Affiliation(s)
- Oliver Cooper
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478
| | - Penny Hallett
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478
| | - Ole Isacson
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478
| |
Collapse
|
21
|
Liampas I, Kyriakoulopoulou P, Siokas V, Tsiamaki E, Stamati P, Kefalopoulou Z, Chroni E, Dardiotis E. Apolipoprotein E Gene in α-Synucleinopathies: A Narrative Review. Int J Mol Sci 2024; 25:1795. [PMID: 38339074 PMCID: PMC10855384 DOI: 10.3390/ijms25031795] [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: 12/21/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
In this narrative review, we delved into the intricate interplay between Apolipoprotein E (APOE) alleles (typically associated with Alzheimer's disease-AD) and alpha-synucleinopathies (aS-pathies), involving Parkinson's disease (PD), Parkinson's disease dementia (PDD), dementia with Lewy bodies (DLB), and multiple-system atrophy (MSA). First, in-vitro, animal, and human-based data on the exacerbating effect of APOE4 on LB pathology were summarized. We found robust evidence that APOE4 carriage constitutes a risk factor for PDD-APOE2, and APOE3 may not alter the risk of developing PDD. We confirmed that APOE4 copies confer an increased hazard towards DLB, as well. Again APOE2 and APOE3 appear unrelated to the risk of conversion. Of note, in individuals with DLB APOE4, carriage appears to be intermediately prevalent between AD and PDD-PD (AD > DLB > PDD > PD). Less consistency existed when it came to PD; APOE-PD associations tended to be markedly modified by ethnicity. Finally, we failed to establish an association between the APOE gene and MSA. Phenotypic associations (age of disease onset, survival, cognitive-neuropsychiatric- motor-, and sleep-related manifestations) between APOE alleles, and each of the aforementioned conditions were also outlined. Finally, a synopsis of literature gaps was provided followed by suggestions for future research.
Collapse
Affiliation(s)
- Ioannis Liampas
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, 41100 Larissa, Greece; (V.S.); (P.S.); (E.D.)
| | - Panagiota Kyriakoulopoulou
- Department of Neurology, University Hospital of Patras, School of Medicine, University of Patras, 26504 Rio Patras, Greece; (P.K.); (E.T.); (Z.K.); (E.C.)
| | - Vasileios Siokas
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, 41100 Larissa, Greece; (V.S.); (P.S.); (E.D.)
| | - Eirini Tsiamaki
- Department of Neurology, University Hospital of Patras, School of Medicine, University of Patras, 26504 Rio Patras, Greece; (P.K.); (E.T.); (Z.K.); (E.C.)
| | - Polyxeni Stamati
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, 41100 Larissa, Greece; (V.S.); (P.S.); (E.D.)
| | - Zinovia Kefalopoulou
- Department of Neurology, University Hospital of Patras, School of Medicine, University of Patras, 26504 Rio Patras, Greece; (P.K.); (E.T.); (Z.K.); (E.C.)
| | - Elisabeth Chroni
- Department of Neurology, University Hospital of Patras, School of Medicine, University of Patras, 26504 Rio Patras, Greece; (P.K.); (E.T.); (Z.K.); (E.C.)
| | - Efthimios Dardiotis
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, 41100 Larissa, Greece; (V.S.); (P.S.); (E.D.)
| |
Collapse
|
22
|
Meng T, Zhang Y, Huang J, Pandey V, Fu S, Ma S. Rubusoside mitigates neuroinflammation and cellular apoptosis in Parkinson's disease, and alters gut microbiota and metabolite composition. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 124:155309. [PMID: 38237261 DOI: 10.1016/j.phymed.2023.155309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024]
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative condition characterized by the progressive loss of dopaminergic neurons within the substantia nigra. Neuroinflammation plays a pivotal role in the pathogenesis of PD, involving the activation of microglia cells, heightened production of proinflammatory cytokines, and perturbations in the composition of the gut microbiota. Rubusoside (Ru), the principal steviol bisglucoside present in Rubus chingii var. suavissimus (S.K.Lee) L.T.Lu (Rosaceae), has been documented for its anti-inflammatory properties in diverse disease models. Nonetheless, there is an imperative need to comprehensively assess and elucidate the protective and anti-inflammatory attributes of Ru concerning PD, as well as to uncover the underlying mechanism involved. OBJECTIVE The aim of this study is to evaluate the neuroprotective and anti-inflammatory effects of Ru on PD and investigate its potential mechanisms associated with microbes. RESEARCH DESIGN AND METHODS We pre-treated mice and cell lines with Ru in order to simulate the progression of PD and the neuroinflammatory state. The mouse model was induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), SN4741 cells were induced by 1-methyl-4-phenylpyridine (mpp+), and BV-2 cells were induced by lipopolysaccharide (LPS). We assessed the impact of Ru on motor function, neuroinflammation, neuron apoptosis, the composition of gut microbes, and their metabolites. RESULTS Ru treatment reduces the release of pro-inflammatory mediators by inhibiting microglia activation. It also prevents neuronal apoptosis, thereby safeguarding dopaminergic neurons and ameliorating motor dysfunction. Furthermore, it induces alterations in the fecal microbiota composition and metabolites profile in PD mice. In vitro experiments have demonstrated that Ru inhibits neuronal apoptosis in SN4741 cells induced by mpp+, suppresses the production of pro-inflammatory mediators, and activates the c-Jun N-terminal kinase (JNK), mitogen-activated protein kinase (p38 MAPK), and nuclear factor kappa-B (NF-κB) signaling pathways. CONCLUSION Ru exhibits inhibitory effects on the MPTP-induced PD model by mitigating neuroinflammation and neuronal apoptosis while also inducing changes in the gut microbiota and metabolite composition.
Collapse
Affiliation(s)
- Tianyu Meng
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, PR China; Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, PR China
| | - Yufei Zhang
- College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Jing Huang
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, PR China; Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, PR China
| | - Vijay Pandey
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, PR China; Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, PR China
| | - Shoupeng Fu
- College of Veterinary Medicine, Jilin University, Changchun 130062, PR China.
| | - Shaohua Ma
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, PR China; Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, PR China.
| |
Collapse
|
23
|
Jellinger KA. Pathobiology of Cognitive Impairment in Parkinson Disease: Challenges and Outlooks. Int J Mol Sci 2023; 25:498. [PMID: 38203667 PMCID: PMC10778722 DOI: 10.3390/ijms25010498] [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: 11/23/2023] [Revised: 12/11/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Cognitive impairment (CI) is a characteristic non-motor feature of Parkinson disease (PD) that poses a severe burden on the patients and caregivers, yet relatively little is known about its pathobiology. Cognitive deficits are evident throughout the course of PD, with around 25% of subtle cognitive decline and mild CI (MCI) at the time of diagnosis and up to 83% of patients developing dementia after 20 years. The heterogeneity of cognitive phenotypes suggests that a common neuropathological process, characterized by progressive degeneration of the dopaminergic striatonigral system and of many other neuronal systems, results not only in structural deficits but also extensive changes of functional neuronal network activities and neurotransmitter dysfunctions. Modern neuroimaging studies revealed multilocular cortical and subcortical atrophies and alterations in intrinsic neuronal connectivities. The decreased functional connectivity (FC) of the default mode network (DMN) in the bilateral prefrontal cortex is affected already before the development of clinical CI and in the absence of structural changes. Longitudinal cognitive decline is associated with frontostriatal and limbic affections, white matter microlesions and changes between multiple functional neuronal networks, including thalamo-insular, frontoparietal and attention networks, the cholinergic forebrain and the noradrenergic system. Superimposed Alzheimer-related (and other concomitant) pathologies due to interactions between α-synuclein, tau-protein and β-amyloid contribute to dementia pathogenesis in both PD and dementia with Lewy bodies (DLB). To further elucidate the interaction of the pathomechanisms responsible for CI in PD, well-designed longitudinal clinico-pathological studies are warranted that are supported by fluid and sophisticated imaging biomarkers as a basis for better early diagnosis and future disease-modifying therapies.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, A-1150 Vienna, Austria
| |
Collapse
|
24
|
Pal G, Corcos DM, Metman LV, Israel Z, Bergman H, Arkadir D. Cognitive Effects of Subthalamic Nucleus Deep Brain Stimulation in Parkinson's Disease with GBA1 Pathogenic Variants. Mov Disord 2023; 38:2155-2162. [PMID: 37916476 PMCID: PMC10990226 DOI: 10.1002/mds.29647] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 11/03/2023] Open
Abstract
Genetic subtyping of patients with Parkinson's disease (PD) may assist in predicting the cognitive and motor outcomes of subthalamic deep brain stimulation (STN-DBS). Practical questions were recently raised with the emergence of new data regarding suboptimal cognitive outcomes after STN-DBS in individuals with PD associated with pathogenic variants in glucocerebrosidase gene (GBA1-PD). However, a variety of gaps and controversies remain. (1) Does STN-DBS truly accelerate cognitive deterioration in GBA1-PD? If so, what is the clinical significance of this acceleration? (2) How should the overall risk-to-benefit ratio of STN-DBS in GBA1-PD be established? (3) If STN-DBS has a negative effect on cognition in GBA1-PD, how can this effect be minimized? (4) Should PD patients be genetically tested before STN-DBS? (5) How should GBA1-PD patients considering STN-DBS be counseled? We aim to summarize the currently available relevant data and detail the gaps and controversies that exist pertaining to these questions. In the absence of evidence-based data, all authors strongly agree that clinicians should not categorically deny DBS to PD patients based solely on genotype (GBA1 status). We suggest that PD patients considering DBS may be offered genetic testing for GBA1, where available and feasible, so the potential risks and benefits of STN-DBS can be properly weighed by both the patient and clinician. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Gian Pal
- Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, United States
| | - Daniel M. Corcos
- Department of Physical Therapy and Human Movement Sciences, Northwestern University, Chicago, Illinois, United States
| | - Leo Verhagen Metman
- Parkinson’s Disease and Movement Disorders Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zvi Israel
- Faculty of Medicine, The Hebrew University and Hadassah, Jerusalem, Jerusalem, Israel
- Department of Neurosurgery, Hadassah Medical Center, Jerusalem, Israel
| | - Hagai Bergman
- Faculty of Medicine, The Hebrew University and Hadassah, Jerusalem, Jerusalem, Israel
- Department of Medical Neurobiology, Institute of Medical Research Israel–Canada (IMRIC), The Hebrew University–Hadassah Medical School, Jerusalem, Israel
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem, Israel
| | - David Arkadir
- Faculty of Medicine, The Hebrew University and Hadassah, Jerusalem, Jerusalem, Israel
- Department of Neurology, Hadassah Medical Center, Jerusalem, Israel
| |
Collapse
|
25
|
Santos-Rebouças CB, Cordovil Cotrin J, Dos Santos Junior GC. Exploring the interplay between metabolomics and genetics in Parkinson's disease: Insights from ongoing research and future avenues. Mech Ageing Dev 2023; 216:111875. [PMID: 37748695 DOI: 10.1016/j.mad.2023.111875] [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: 08/22/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 09/27/2023]
Abstract
Parkinson's disease (PD) is a widespread neurodegenerative disorder, whose complex aetiology remains under construction. While rare variants have been associated with the monogenic PD form, most PD cases are influenced by multiple genetic and environmental aspects. Nonetheless, the pathophysiological pathways and molecular networks involved in monogenic/idiopathic PD overlap, and genetic variants are decisive in elucidating the convergent underlying mechanisms of PD. In this scenario, metabolomics has furnished a dynamic and systematic picture of the synergy between the genetic background and environmental influences that impact PD, making it a valuable tool for investigating PD-related metabolic dysfunctions. In this review, we performed a brief overview of metabolomics current research in PD, focusing on significant metabolic alterations observed in idiopathic PD from different biofluids and strata and exploring how they relate to genetic factors associated with monogenic PD. Dysregulated amino acid metabolism, lipid metabolism, and oxidative stress are the critical metabolic pathways implicated in both genetic and idiopathic PD. By merging metabolomics and genetics data, it is possible to distinguish metabolic signatures of specific genetic backgrounds and to pinpoint subgroups of PD patients who could derive personalized therapeutic benefits. This approach holds great promise for advancing PD research and developing innovative, cost-effective treatments.
Collapse
Affiliation(s)
- Cíntia Barros Santos-Rebouças
- Human Genetics Service, Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| | - Juliana Cordovil Cotrin
- Human Genetics Service, Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Gilson Costa Dos Santos Junior
- LabMet, Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil
| |
Collapse
|
26
|
Lin J, Ou R, Li C, Hou Y, Zhang L, Wei Q, Liu K, Jiang Q, Yang T, Xiao Y, Pang D, Zhao B, Chen X, Yang J, Shang H. Evolution and Predictive Role of Plasma Alzheimer's Disease-related Pathological Biomarkers in Parkinson's Disease. J Gerontol A Biol Sci Med Sci 2023; 78:2203-2213. [PMID: 37560912 DOI: 10.1093/gerona/glad189] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Indexed: 08/11/2023] Open
Abstract
Plasma Alzheimer's disease-related pathological biomarkers' role in Parkinson's disease (PD) remains unknown. We aimed to determine whether plasma Alzheimer's disease-related biomarkers can predict PD progression. A total of 184 PD patients and 86 healthy controls were included and followed up for 5 years. Plasma phosphorylated tau181 (p-tau181), Aβ40, and Aβ42 were measured at baseline and the 1- and 2-year follow-ups using the Quanterix-single-molecule array. Global cognitive function and motor symptoms were assessed using the Montreal Cognitive Assessment and Unified Parkinson's Disease Rating Scale part III. Genetic analyses were conducted to identify APOE and MAPT genotypes. Plasma p-tau181 levels were higher in PD than healthy controls. APOE-ε4 carriers had lower plasma Aβ42 levels and Aβ42/Aβ40 ratio. The linear mixed-effects models showed that Montreal Cognitive Assessment scores were associated with plasma p-tau181/Aβ42 ratio (β -1.719 [-3.398 to -0.040], p = .045). Higher baseline plasma p-tau181 correlated with faster cognitive decline and motor symptoms deterioration in total patients (β -0.170 [-0.322 to -0.018], p = .029; β 0.329 [0.032 to 0.626], p = .030) and APOE-ε4 carriers (β -0.318 [-0.602 to -0.034], p = .030; β 0.632 [0.017 to 1.246], p = .046), but not in the noncarriers. Higher baseline plasma Aβ40 correlated with faster cognitive decline in total patients (β -0.007 [-0.015 to -0.0001], p = .047) and faster motor symptoms deterioration in total patients (β 0.026 [0.010 to 0.041], p = .001) and APOE-ε4 carriers (β 0.044 [-0.026 to 0.049], p = .020), but not in the noncarriers. The plasma p-tau181/Aβ2 ratio monitors the cognitive status of PD. Higher baseline plasma p-tau181 and Aβ40 predict faster cognitive decline and motor symptoms deterioration in PD, especially in APOE-ε4 carriers.
Collapse
Affiliation(s)
- Junyu Lin
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruwei Ou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chunyu Li
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanbing Hou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lingyu Zhang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qianqian Wei
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kuncheng Liu
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qirui Jiang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tianmi Yang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Xiao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dejiang Pang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bi Zhao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xueping Chen
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jing Yang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huifang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
27
|
Connolly KJ, Margaria J, Di Biase E, Cooper O, Hallett PJ, Isacson O. Loss of Lipid Carrier ApoE Exacerbates Brain Glial and Inflammatory Responses after Lysosomal GBA1 Inhibition. Cells 2023; 12:2564. [PMID: 37947642 PMCID: PMC10647680 DOI: 10.3390/cells12212564] [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: 08/04/2023] [Revised: 09/26/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Tightly regulated and highly adaptive lipid metabolic and transport pathways are critical to maintaining brain cellular lipid homeostasis and responding to lipid and inflammatory stress to preserve brain function and health. Deficits in the lipid handling genes APOE and GBA1 are the most significant genetic risk factors for Lewy body dementia and related dementia syndromes. Parkinson's disease patients who carry both APOE4 and GBA1 variants have accelerated cognitive decline compared to single variant carriers. To investigate functional interactions between brain ApoE and GBA1, in vivo GBA1 inhibition was tested in WT versus ApoE-deficient mice. The experiments demonstrated glycolipid stress caused by GBA1 inhibition in WT mice induced ApoE expression in several brain regions associated with movement and dementia disorders. The absence of ApoE in ApoE-KO mice amplified complement C1q elevations, reactive microgliosis and astrocytosis after glycolipid stress. Mechanistically, GBA1 inhibition triggered increases in cell surface and intracellular lipid transporters ABCA1 and NPC1, respectively. Interestingly, the absence of NPC1 in mice also triggered elevations of brain ApoE levels. These new data show that brain ApoE, GBA1 and NPC1 functions are interconnected in vivo, and that the removal or reduction of ApoE would likely be detrimental to brain function. These results provide important insights into brain ApoE adaptive responses to increased lipid loads.
Collapse
Affiliation(s)
| | | | | | | | - Penelope J. Hallett
- Departments of Psychiatry and Neurology Harvard Medical School, Neuroregeneration Institute, McLean Hospital, Belmont, MA 02478, USA
| | - Ole Isacson
- Departments of Psychiatry and Neurology Harvard Medical School, Neuroregeneration Institute, McLean Hospital, Belmont, MA 02478, USA
| |
Collapse
|
28
|
Gasca-Salas C, Trompeta C, López-Aguirre M, Rodríguez Rojas R, Clarimon J, Dols-Icardo O, El Bounasri S, Guida P, Mata-Marín D, Hernández-Fernández F, Marras C, García-Cañamaque L, Plaza de Las Heras I, Obeso I, Vela L, Fernández-Rodríguez B. Brain hypometabolism in non-demented microtubule-associated protein tau H1 carriers with Parkinson's disease. J Neuroimaging 2023; 33:953-959. [PMID: 37726927 DOI: 10.1111/jon.13156] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/06/2023] [Accepted: 09/10/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND AND PURPOSE The microtubule-associated protein tau (MAPT) H1 homozygosity (H1/H1 haplotype) is a genetic risk factor for neurodegenerative diseases, such as Parkinson's disease (PD). MAPT H1 homozygosity has been associated with conversion to PD; however, results are conflicting since some studies did not find a strong influence. Cortical hypometabolism is associated with cognitive impairment in PD. In this study, we aimed to evaluate the metabolic pattern in nondemented PD patients MAPT H1/H1 carriers in comparison with MAPT H1/H2 haplotype. In addition, we evaluated domain-specific cognitive differences according to MAPT haplotype. METHODS We compared a group of 26 H1/H1 and 20 H1/H2 carriers with late-onset PD. Participants underwent a comprehensive neuropsychological cognitive evaluation and a [18F]-Fluorodeoxyglucose PET-MR scan. RESULTS MAPT H1/H1 carriers showed worse performance in the digit span forward test of attention compared to MAPT H1/H2 carriers. In the [18F]-Fluorodeoxyglucose PET comparisons, MAPT H1/H1 displayed hypometabolism in the frontal cortex, parahippocampal, and cingulate gyrus, as well as in the caudate and globus pallidus. CONCLUSION PD patients MAPT H1/H1 carriers without dementia exhibit relative hypometabolism in several cortical areas as well as in the basal ganglia, and worse performance in attention than MAPT H1/H2 carriers. Longitudinal studies should assess if lower scores in attention and dysfunction in these areas are predictors of dementia in MAPT H1/H1 homozygotes.
Collapse
Affiliation(s)
- Carmen Gasca-Salas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- University CEU-San Pablo, Madrid, Spain
| | - Clara Trompeta
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- PhD Program in Health Sciences, University of Alcala de Henares Alcalá de Henares, Madrid, Spain
| | - Miguel López-Aguirre
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- PhD Program in Physics, Complutense University of Madrid, Madrid, Spain
| | - Rafael Rodríguez Rojas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Jordi Clarimon
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- Memory Unit, Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Oriol Dols-Icardo
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- Memory Unit, Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Shaimaa El Bounasri
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- Memory Unit, Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Pasqualina Guida
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- PhD Program in Neuroscience, Autónoma de Madrid University-Cajal Institute, Madrid, Spain
| | - David Mata-Marín
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- PhD Program in Neuroscience, Autónoma de Madrid University-Cajal Institute, Madrid, Spain
| | - Frida Hernández-Fernández
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Department of Nursing and Nutrition, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
| | - Connie Marras
- The Edmond J Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Lina García-Cañamaque
- Nuclear Medicine Department, PET-MRI Centre, HM Puerta del Sur University Hospital, HM Hospitales, Madrid, Spain
| | - Isabel Plaza de Las Heras
- Nuclear Medicine Department, PET-MRI Centre, HM Puerta del Sur University Hospital, HM Hospitales, Madrid, Spain
| | - Ignacio Obeso
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Lydia Vela
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Department of Neurology, Hospital U Fundación Alcorcón, Calle Budapest, Alcorcón, Spain
| | - Beatriz Fernández-Rodríguez
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- PhD Program in Neuroscience, Autónoma de Madrid University-Cajal Institute, Madrid, Spain
| |
Collapse
|
29
|
Kapan A, Haider S, Wakolbinger M, Spatt J. Associations of Apolipoprotein ε4 Genotypes with Motor and Nonmotor Symptoms in Parkinson's Disease: A Cross-Sectional Study. Mov Disord Clin Pract 2023; 10:1611-1619. [PMID: 38026513 PMCID: PMC10654815 DOI: 10.1002/mdc3.13862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/20/2023] [Accepted: 08/06/2023] [Indexed: 12/01/2023] Open
Abstract
Background The apolipoprotein E (APOE) ε4 allele has been associated with cognitive decline in Parkinson's disease (PD), but little is known about its relationship with motor and other nonmotor symptoms and whether APOE ε4 retains an influence on cognition when other factors are considered. Objective To investigate the impact of APOE ε4 on motor/nonmotor symptoms and its relationship with other factors affecting cognition in individuals with PD. Methods We analyzed data from 7616 individuals, comparing motor/nonmotor symptoms in different APOE genotypes using binary logistic regression. Multivariate logistic regression examined factors associated with cognitive impairments, including APOE ε4, Geriatric Depression Scale (GDS) score, Non-motor Symptom Questionnaire (NMS) score, Movement Disorder Society-Sponsored Revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS) Part II score, and physical activity level. Results APOE ε4 heterozygosity was modestly associated with lower cognitive scores (odds ratio [OR], 0.92; 95% confidence interval [CI], 0.87-0.99), whereas no significant association was found for any other nonmotor and motor symptoms. However, in multivariate analysis, cognitive impairment was associated with higher GDS (OR, 1.28; 95% CI, 1.23-1.34), NMS (OR, 1.22; 95% CI, 1.19-1.25), and MDS-UPDRS Part II (OR, 1.07; 95% CI, 1.06-1.09) scores, whereas physical activity was negatively associated (OR, 0.99; 95% CI, 0.98-0.99). APOE ε4 was no longer significant after adjusting for these factors. Conclusions There is a link between cognition and APOE ε4 in patients with PD; however, when considering multiple factors, APOE ε4 plays a subordinate role. Other factors, such as depression, physical activity, and other nonmotor symptoms, demonstrate a stronger influence on cognitive impairment.
Collapse
Affiliation(s)
- Ali Kapan
- Department of Social and Preventive Medicine, Center for Public HealthMedical University of ViennaViennaAustria
| | - Sandra Haider
- Department of Social and Preventive Medicine, Center for Public HealthMedical University of ViennaViennaAustria
| | - Maria Wakolbinger
- Department of Social and Preventive Medicine, Center for Public HealthMedical University of ViennaViennaAustria
| | - Josef Spatt
- Faculty for MedicineSigmund Freud University ViennaViennaAustria
- Neurological DepartmentEvangelical Hospital ViennaViennaAustria
| |
Collapse
|
30
|
Zenuni H, Bovenzi R, Bissacco J, Grillo P, Simonetta C, Mascioli D, Pieri M, Bernardini S, Sancesario GM, Stefani A, Mercuri NB, Schirinzi T. Clinical and neurochemical correlates of the APOE genotype in early-stage Parkinson's disease. Neurobiol Aging 2023; 131:24-28. [PMID: 37572524 DOI: 10.1016/j.neurobiolaging.2023.07.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/14/2023] [Accepted: 07/15/2023] [Indexed: 08/14/2023]
Abstract
Emerging evidence indicates that apolipoprotein E (APOE) genotype may influence Parkinson's disease (PD) course, although clinical and neurochemical correlates have not been completely established. This study aimed to determine the associations of APOE genotypes (ε4 vs. non-ε4) with cerebrospinal fluid (CSF) neurodegeneration biomarkers and clinical parameters in early-stage PD patients. One hundred and seventy-five PD patients and 89 non-neurodegenerative controls grouped in APOE-ε4 carriers (28 PD; 12 controls) and non-APOE-ε4 carriers (147 PD; 78 controls) were enrolled. CSF levels of amyloid-β-42, amyloid-β-40, total and 181-phosphorylated tau, and clinical scores were compared among groups adjusting for main covariates. APOE genotypes prevalence was similar in PD and controls. PD APOE-ε4 carriers had lower amyloid-β-42 CSF levels than PD non-APOE-ε4 carriers and controls, independently from age. PD APOE-ε4 carriers also had higher total and "item 5" (attention and memory) non-motor symptoms scale scores than PD non-APOE-ε4 carriers, independently from confounding factors. APOE-ε4 genotype might thus account for a more vulnerable PD subtype characterized by prominent amyloidopathy and a greater burden of non-motor symptoms in the early disease stages. DATA AVAILABILITY: Data are available upon reasonable request.
Collapse
Affiliation(s)
- Henri Zenuni
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Roberta Bovenzi
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Jacopo Bissacco
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Piergiorgio Grillo
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Clara Simonetta
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Davide Mascioli
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Massimo Pieri
- Clinical Biochemistry Unit, Department of Experimental Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Segio Bernardini
- Clinical Biochemistry Unit, Department of Experimental Medicine, University of Roma Tor Vergata, Rome, Italy
| | | | - Alessandro Stefani
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Tommaso Schirinzi
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy.
| |
Collapse
|
31
|
Chase BA, Krueger R, Pavelka L, Chung SJ, Aasly J, Dardiotis E, Premkumar AP, Schoneburg B, Kartha N, Aunaetitrakul N, Frigerio R, Maraganore D, Markopoulou K. Multifactorial assessment of Parkinson's disease course and outcomes using trajectory modeling in a multiethnic, multisite cohort - extension of the LONG-PD study. Front Aging Neurosci 2023; 15:1240971. [PMID: 37842125 PMCID: PMC10569724 DOI: 10.3389/fnagi.2023.1240971] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/28/2023] [Indexed: 10/17/2023] Open
Abstract
Background The severity, progression, and outcomes of motor and non-motor symptoms in Parkinson's disease (PD) are quite variable. Following PD cohorts holds promise for identifying predictors of disease severity and progression. Methods PD patients (N = 871) were enrolled at five sites. Enrollment occurred within 5 years of initial motor symptom onset. Disease progression was assessed annually for 2-to-10 years after onset. Group-based trajectory modeling was used to identify groups differing in disease progression. Models were developed for UPDRS-III scores, UPDRS-III tremor and bradykinesia-rigidity subscores, Hoehn & Yahr (H&Y) stage, Mini-Mental Status Exam (MMSE) scores, and UPDRS-III, H&Y and MMSE scores considered together. Predictors of trajectory-group membership were modeled simultaneously with the trajectories. Kaplan-Meier survival analysis evaluated survival free of PD outcomes. Results The best fitting models identified three groups. One showed a relatively benign, slowly progressing trajectory (Group 1), a second showed a moderate, intermediately progressing trajectory (Group 2), and a third showed a more severe, rapidly progressing trajectory (Group 3). Stable trajectory-group membership occurred relatively early in the disease course, 5 years after initial motor symptom. Predictors of intermediate and more severe trajectory-group membership varied across the single variable models and the multivariable model jointly considering UPDRS-III, H&Y and MMSE scores. In the multivariable model, membership in Group 2 (28.4% of patients), relative to Group 1 (50.5%), was associated with male sex, younger age-at-onset, fewer education-years, pesticide exposure, absence of reported head injury, and akinetic/rigid subtype at initial presentation. Membership in Group 3 (21.3%), relative to Group 1, was associated with older age-at-onset, fewer education-years, pesticide exposure, and the absence of a tremor-predominant subtype at initial presentation. Persistent freezing, persistent falls, and cognitive impairment occurred earliest and more frequently in Group 3, later and less frequently in Group 2, and latest and least frequently in Group 1. Furthermore, autonomic complications, dysphagia, and psychosis occurred more frequently in Groups 2 and 3 than in Group 1. Conclusion Modeling disease course using multiple objective assessments over an extended follow-up duration identified groups that more accurately reflect differences in PD course, prognosis, and outcomes than assessing single parameters over shorter intervals.
Collapse
Affiliation(s)
- Bruce A. Chase
- Health Information Technology, NorthShore University HealthSystem, Evanston, IL, United States
| | - Rejko Krueger
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Centre Hospitalier de Luxembourg (CLG), Luxembourg, Luxembourg
- Parkinson’s Research Clinic, Centre Hospitalier de Luxembourg (CHL), Luxembourg, Luxembourg
| | - Lukas Pavelka
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Centre Hospitalier de Luxembourg (CLG), Luxembourg, Luxembourg
- Parkinson’s Research Clinic, Centre Hospitalier de Luxembourg (CHL), Luxembourg, Luxembourg
| | - Sun Ju Chung
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jan Aasly
- Department of Neurology, St. Olav’s Hospital, Trondheim, Norway
- Department of Neuroscience, Norwegian University of Science and Technology, Trondheim, Norway
| | - Efthimios Dardiotis
- Department of Neurology, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Ashvini P. Premkumar
- Department of Neurology, NorthShore University HealthSystem, Evanston, IL, United States
| | - Bernadette Schoneburg
- Department of Neurology, NorthShore University HealthSystem, Evanston, IL, United States
| | - Ninith Kartha
- Department of Neurology, NorthShore University HealthSystem, Evanston, IL, United States
| | - Navamon Aunaetitrakul
- Department of Neurology, NorthShore University HealthSystem, Evanston, IL, United States
| | - Roberta Frigerio
- Department of Neurology, NorthShore University HealthSystem, Evanston, IL, United States
| | | | - Katerina Markopoulou
- Department of Neurology, NorthShore University HealthSystem, Evanston, IL, United States
- Department of Neurology, University of Chicago Pritzker School of Medicine, Chicago, IL, United States
| |
Collapse
|
32
|
Ta M, Blauwendraat C, Antar T, Leonard HL, Singleton AB, Nalls MA, Iwaki H. Genome-Wide Meta-Analysis of Cerebrospinal Fluid Biomarkers in Alzheimer's Disease and Parkinson's Disease Cohorts. Mov Disord 2023; 38:1697-1705. [PMID: 37539664 PMCID: PMC11459375 DOI: 10.1002/mds.29511] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/14/2023] [Accepted: 05/30/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Amyloid-β, phosphorylated tau (p-tau), and total tau (t-tau) in cerebrospinal fluid are established biomarkers for Alzheimer's disease (AD). In other neurodegenerative diseases, such as Parkinson's disease (PD), these biomarkers have also been found to be altered, and the molecular mechanisms responsible for these alterations are still under investigation. Moreover, the interplay between these mechanisms and the diverse underlying disease states remains to be elucidated. OBJECTIVE To investigate genetic contributions to the AD biomarkers and assess the commonality and heterogeneity of the associations per underlying disease status. METHODS We conducted genome-wide association studies (GWASs) for the AD biomarkers on subjects from the Parkinson's Progression Markers Initiative, the Fox Investigation for New Discovery of Biomarkers, and the Alzheimer's Disease Neuroimaging Initiative, and meta-analyzed with the largest AD GWAS. We tested heterogeneity of associations of interest between different disease statuses (AD, PD, and control). RESULTS We observed three GWAS signals: the APOE locus for amyloid-β, the 3q28 locus between GEMC1 and OSTN for p-tau and t-tau, and the 7p22 locus (top hit: rs60871478, an intronic variant for DNAAF5, also known as HEATR2) for p-tau. The 7p22 locus is novel and colocalized with the brain DNAAF5 expression. Although no heterogeneity from underlying disease status was observed for the earlier GWAS signals, some disease risk loci suggested disease-specific associations with these biomarkers. CONCLUSIONS Our study identified a novel association at the intronic region of DNAAF5 associated with increased levels of p-tau across all diseases. We also observed some disease-specific genetic associations with these biomarkers. Published 2023. This article is a U.S. Government work and is in the public domain in the USA.
Collapse
Affiliation(s)
- Michael Ta
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC, USA
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Tarek Antar
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Hampton L Leonard
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC, USA
| | - Andrew B. Singleton
- Center for Alzheimer’s and Related Dementias, National Institutes of Health, Bethesda, MD, USA 20892
| | - Mike A. Nalls
- 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
| | | |
Collapse
|
33
|
Zhou Y, Wang Y, Wan J, Zhao Y, Pan H, Zeng Q, Zhou X, He R, Zhou X, Xiang Y, Zhou Z, Chen B, Sun Q, Xu Q, Tan J, Shen L, Jiang H, Yan X, Li J, Guo J, Tang B, Wu H, Liu Z. Mutational spectrum and clinical features of GBA1 variants in a Chinese cohort with Parkinson's disease. NPJ Parkinsons Dis 2023; 9:129. [PMID: 37658046 PMCID: PMC10474275 DOI: 10.1038/s41531-023-00571-4] [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: 04/20/2023] [Accepted: 08/16/2023] [Indexed: 09/03/2023] Open
Abstract
GBA1 variants are important risk factors for Parkinson's disease (PD). Most studies assessing GBA1-related PD risk have been performed in European-derived populations. Although the coding region of the GBA1 gene in the Chinese population has been analyzed, the sample sizes were not adequate. In this study, we aimed to investigate GBA1 variants in a large Chinese cohort of patients with PD and healthy control and explore the associated clinical characteristics. GBA1 variants in 4034 patients and 2931 control participants were investigated using whole-exome and whole-genome sequencing. The clinical features of patients were evaluated using several scales. Regression analysis, chi-square, and Fisher exact tests were used to analyze GBA1 variants and the clinical symptoms of different groups. We identified 104 variants, including 8 novel variants, expanding the spectrum of GBA1 variants. The frequency of GBA1 variants in patients with PD was 7.46%, higher than that in the control (1.81%) (P < 0.001, odds ratio [OR] = 4.38, 95% confidence interval [CI]: 3.26-5.89). Among patients, 176 (4.36%) had severe variants, 34 (0.84%) carried mild variants, three (0.07%) had risk variants, and 88 (2.18%) carried unknown variants. Our study, for the first time, found that p.G241R (P = 0.007, OR = 15.3, 95% CI: 1.25-261.1) and p.S310G (P = 0.005, OR = 4.86, 95% CI: 1.52-28.04) variants increased the risk of PD. Patients with GBA1 variants exhibited an earlier onset age and higher risk of probable rapid-eye-movement sleep behavior disorder, olfactory dysfunction, depression, and autonomic dysfunction than patients without GBA1 variants.
Collapse
Affiliation(s)
- Yangjie Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yige Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan Wan
- Department of Neurology, & Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Yuwen Zhao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongxu Pan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Zeng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xun Zhou
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Runcheng He
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoxia Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yaqin Xiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhou Zhou
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bin Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qiying Sun
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jieqiong Tan
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Changsha, Hunan, China
- Bioinformatics Center & National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Changsha, Hunan, China
| | - Xinxiang Yan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jinchen Li
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Bioinformatics Center & National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Changsha, Hunan, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Neurology, & Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Changsha, Hunan, China
- Bioinformatics Center & National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Heng Wu
- Department of Neurology, & Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China.
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province, Hengyang, Hunan, China.
| | - Zhenhua Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Changsha, Hunan, China.
- Bioinformatics Center & National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
34
|
Martinez-Carrasco A, Real R, Lawton M, Iwaki H, Tan MMX, Wu L, Williams NM, Carroll C, Hu MTM, Grosset DG, Hardy J, Ryten M, Foltynie T, Ben-Shlomo Y, Shoai M, Morris HR. Genetic meta-analysis of levodopa induced dyskinesia in Parkinson's disease. NPJ Parkinsons Dis 2023; 9:128. [PMID: 37652906 PMCID: PMC10471743 DOI: 10.1038/s41531-023-00573-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023] Open
Abstract
The genetic basis of levodopa-induced-dyskinesia (LiD) is poorly understood, and there have been few well-powered genome-wide studies. We performed a genome-wide survival meta-analyses to study the effect of genetic variation on the development of LiD in five separate longitudinal cohorts, and meta-analysed the results. We included 2784 PD patients, of whom 14.6% developed LiD. We found female sex (HR = 1.35, SE = 0.11, P = 0.007) and younger age at onset (HR = 1.8, SE = 0.14, P = 2 × 10-5) increased the probability of developing LiD. We identified three genetic loci significantly associated with time-to-LiD onset. rs72673189 on chromosome 1 (HR = 2.77, SE = 0.18, P = 1.53 × 10-8) located at the LRP8 locus, rs189093213 on chromosome 4 (HR = 3.06, SE = 0.19, P = 2.81 × 10-9) in the non-coding RNA LINC02353 locus, and rs180924818 on chromosome 16 (HR = 3.13, SE = 0.20, P = 6.27 × 10-9) in the XYLT1 locus. Based on a functional annotation analysis on chromosome 1, we determined that changes in DNAJB4 gene expression, close to LRP8, are an additional potential cause of increased susceptibility to LiD. Baseline anxiety status was significantly associated with LiD (OR = 1.14, SE = 0.03, P = 7.4 × 10-5). Finally, we performed a candidate variant analysis of previously reported loci, and found that genetic variability in ANKK1 (rs1800497, HR = 1.27, SE = 0.09, P = 8.89 × 10-3) and BDNF (rs6265, HR = 1.21, SE = 0.10, P = 4.95 × 10-2) loci were significantly associated with time to LiD in our large meta-analysis.
Collapse
Affiliation(s)
- Alejandro Martinez-Carrasco
- Department of Clinical and Movement Neurosciences, UCL 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.
| | - Raquel Real
- Department of Clinical and Movement Neurosciences, UCL 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
| | - Michael Lawton
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, 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, Glen Echo, MD, USA
| | - Manuela M X Tan
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Lesley Wu
- Department of Clinical and Movement Neurosciences, UCL 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
| | - Nigel M Williams
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Camille Carroll
- Faculty of Health, University of Plymouth, Plymouth, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - 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
| | - Donald G Grosset
- School of Neuroscience and Psychology, University of Glasgow, Glasgow, UK
| | - John Hardy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, 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
| | - Mina Ryten
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| | - Tom Foltynie
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Yoav Ben-Shlomo
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Maryam Shoai
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, UCL 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
|
35
|
Siciliano M, De Micco R, Russo AG, Esposito F, Sant'Elia V, Ricciardi L, Morgante F, Russo A, Goldman JG, Chiorri C, Tedeschi G, Trojano L, Tessitore A. Memory Phenotypes In Early, De Novo Parkinson's Disease Patients with Mild Cognitive Impairment. Mov Disord 2023; 38:1461-1472. [PMID: 37319041 DOI: 10.1002/mds.29502] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/17/2023] [Accepted: 05/24/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Memory deficits in mild cognitive impairment related to Parkinson's disease (PD-MCI) are quite heterogeneous, and there is no general agreement on their genesis. OBJECTIVES To define memory phenotypes in de novo PD-MCI and their associations with motor and non-motor features and patients' quality of life. METHODS From a sample of 183 early de novo patients with PD, cluster analysis was applied to neuropsychological measures of memory function of 82 patients with PD-MCI (44.8%). The remaining patients free of cognitive impairment were considered as a comparison group (n = 101). Cognitive measures and structural magnetic resonance imaging-based neural correlates of memory function were used to substantiate the results. RESULTS A three-cluster model produced the best solution. Cluster A (65.85%) included memory unimpaired patients; Cluster B (23.17%) included patients with mild episodic memory disorder related to a "prefrontal executive-dependent phenotype"; Cluster C (10.97%) included patients with severe episodic memory disorder related to a "hybrid phenotype," where hippocampal-dependent deficits co-occurred with prefrontal executive-dependent memory dysfunctions. Cognitive and brain structural imaging correlates substantiated the findings. The three phenotypes did not differ in terms of motor and non-motor features, but the attention/executive deficits progressively increased from Cluster A, through Cluster B, to Cluster C. This last cluster had worse quality of life compared to others. CONCLUSIONS Our results demonstrated the memory heterogeneity of de novo PD-MCI, suggesting existence of three distinct memory-related phenotypes. Identification of such phenotypes can be fruitful in understanding the pathophysiological mechanisms underlying PD-MCI and its subtypes and in guiding appropriate treatments. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Mattia Siciliano
- Department of Advanced Medical and Surgical Sciences-MRI Research Center Vanvitelli-FISM, University of Campania "Luigi Vanvitelli", Naples, Italy
- Department of Psychology, University of Campania "Luigi Vanvitelli", Caserta, Italy
- Neurosciences Research Centre, Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Rosa De Micco
- Department of Advanced Medical and Surgical Sciences-MRI Research Center Vanvitelli-FISM, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Andrea Gerardo Russo
- Department of Advanced Medical and Surgical Sciences-MRI Research Center Vanvitelli-FISM, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Fabrizio Esposito
- Department of Advanced Medical and Surgical Sciences-MRI Research Center Vanvitelli-FISM, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Valeria Sant'Elia
- Department of Advanced Medical and Surgical Sciences-MRI Research Center Vanvitelli-FISM, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Lucia Ricciardi
- Neurosciences Research Centre, Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Francesca Morgante
- Neurosciences Research Centre, Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Antonio Russo
- Department of Advanced Medical and Surgical Sciences-MRI Research Center Vanvitelli-FISM, University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | - Carlo Chiorri
- Department of Educational Sciences, University of Genova, Genoa, Italy
| | - Gioacchino Tedeschi
- Department of Advanced Medical and Surgical Sciences-MRI Research Center Vanvitelli-FISM, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Luigi Trojano
- Department of Psychology, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Alessandro Tessitore
- Department of Advanced Medical and Surgical Sciences-MRI Research Center Vanvitelli-FISM, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
36
|
Lloyd GM, Long B, Quintin S, Sorrentino ZA, Gorion KMM, Bell BM, Carrillo D, Sullivan P, Borchelt D, Giasson BI. Carboxyl truncation of α-synuclein occurs early and is influenced by human APOE genotype in transgenic mouse models of α-synuclein pathogenesis. Acta Neuropathol Commun 2023; 11:119. [PMID: 37482615 PMCID: PMC10363304 DOI: 10.1186/s40478-023-01623-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/15/2023] [Indexed: 07/25/2023] Open
Abstract
Post-translational modifications to the carboxyl (C) terminus domain of α-synuclein can play an important role in promoting the pathologic aggregation of α-synuclein. Various cleavages that diminish this highly charged, proline-rich region can result in exposure of hydrophobic, aggregation-prone regions, thereby accelerating the aggregation kinetics of α-synuclein into misfolded, pathologic forms. C-terminally truncated forms of α-synuclein are abundant in human diseased brains compared to controls, suggesting a role in disease pathogenesis. Factors that alter the homeostatic proteolytic processing of α-synuclein may ultimately tip the balance towards a progressive disease state. Apolipoprotein E (APOE) has been implicated in the acceleration of cognitive impairment in patients with Lewy body diseases. The APOE4 isoform has been found to cause dysregulation in the endosomal-lysosomal pathway, which could result in altered α-synuclein degradation as a potential mechanism for promoting its pathologic misfolding. Herein, we investigate the spatiotemporal accumulation of C-terminally truncated α-synuclein in a seeded and progressive mouse model of synucleinopathy. Furthermore, we study how this process is influenced in the context of mice that are altered to express either the human APOE3 or APOE4 isoforms. We found that specific C-terminal truncation of α-synuclein occurs at early stages of pathogenesis. We also found that proteolytic processing of this domain differs across various brain regions and is influenced by the presence of different human APOE isoforms. Our data demonstrate an early pathogenic role for C-terminally truncated α-synuclein, and highlight the influence of APOE isoforms in modulating its impact.
Collapse
Affiliation(s)
- Grace M Lloyd
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Brooke Long
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
| | - Stephan Quintin
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Zachary A Sorrentino
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Kimberly-Marie M Gorion
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Brach M Bell
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Denise Carrillo
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
| | - Patrick Sullivan
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - David Borchelt
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Benoit I Giasson
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA.
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
37
|
Liu T, Zuo H, Ma D, Song D, Zhao Y, Cheng O. Cerebrospinal fluid GFAP is a predictive biomarker for conversion to dementia and Alzheimer's disease-associated biomarkers alterations among de novo Parkinson's disease patients: a prospective cohort study. J Neuroinflammation 2023; 20:167. [PMID: 37475029 PMCID: PMC10357612 DOI: 10.1186/s12974-023-02843-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/27/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Dementia is a prevalent non-motor manifestation among individuals with advanced Parkinson's disease (PD). Glial fibrillary acidic protein (GFAP) is an inflammatory marker derived from astrocytes. Research has demonstrated the potential of plasma GFAP to forecast the progression to dementia in PD patients with mild cognitive impairment (PD-MCI). However, the predictive role of cerebrospinal fluid (CSF) GFAP on future cognitive transformation and alterations in Alzheimer's disease (AD)-associated CSF biomarkers in newly diagnosed PD patients has not been investigated. METHODS 210 de novo PD patients from the Parkinson's Progression Markers Initiative were recruited. Cognitive progression in PD participants was evaluated using Cox regression. Cross-sectional and longitudinal associations between baseline CSF GFAP and cognitive function and AD-related CSF biomarkers were evaluated using multiple linear regression and generalized linear mixed model. RESULTS At baseline, the mean age of PD participants was 60.85 ± 9.78 years, including 142 patients with normal cognition (PD-NC) and 68 PD-MCI patients. The average follow-up time was 6.42 ± 1.69 years. A positive correlation was observed between baseline CSF GFAP and age (β = 0.918, p < 0.001). There was no statistically significant difference in baseline CSF GFAP levels between PD-NC and PD-MCI groups. Higher baseline CSF GFAP predicted greater global cognitive decline over time in early PD patients (Montreal Cognitive Assessment, β = - 0.013, p = 0.014). Furthermore, Cox regression showed that high baseline CSF GFAP levels were associated with a high risk of developing dementia over an 8-year period in the PD-NC group (adjusted HR = 3.070, 95% CI 1.119-8.418, p = 0.029). In addition, the baseline CSF GFAP was positively correlated with the longitudinal changes of not only CSF α-synuclein (β = 0.313, p < 0.001), but also CSF biomarkers associated with AD, namely, amyloid-β 42 (β = 0.147, p = 0.034), total tau (β = 0.337, p < 0.001) and phosphorylated tau (β = 0.408, p < 0.001). CONCLUSIONS CSF GFAP may be a valuable prognostic tool that can predict the severity and progression of cognitive deterioration, accompanied with longitudinal changes in AD-associated pathological markers in early PD.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Hongzhou Zuo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Di Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Dan Song
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Yuying Zhao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Oumei Cheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| |
Collapse
|
38
|
Ta M, Blauwendraat C, Antar T, Leonard HL, Singleton AB, Nalls MA, Iwaki H. Genome-wide meta-analysis of CSF biomarkers in Alzheimer's disease and Parkinson's disease cohorts. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.13.23291354. [PMID: 37398091 PMCID: PMC10312859 DOI: 10.1101/2023.06.13.23291354] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Background Amyloid beta (Aβ), phosphorylated tau (p-tau), and total tau (t-tau) in cerebrospinal fluid are established biomarkers for Alzheimer's disease (AD). In other neurodegenerative diseases, such as Parkinson's disease (PD), these biomarkers have also been found to be altered, and the molecular mechanisms responsible for these alterations are still under investigation. Moreover, the interplay between these mechanisms and the diverse underlying disease states remains to be elucidated. Objectives To investigate genetic contributions to the AD biomarkers and assess the commonality and heterogeneity of the associations per underlying disease status. Methods We conducted GWAS for the AD biomarkers on subjects from the Parkinson's Progression Markers Initiative (PPMI), the Fox Investigation for New Discovery of Biomarkers (BioFIND), and the Alzheimer's Disease Neuroimaging Initiative (ADNI) and meta-analyzed with the largest AD GWAS.[7] We tested heterogeneity of associations of interest between different disease statuses (AD, PD, and control). Results We observed three GWAS signals: the APOE locus for Aβ, the 3q28 locus between GEMC1 and OSTN for p-tau and t-tau, and the 7p22 locus (top hit: rs60871478, an intronic variant for DNAAF5 , also known as HEATR2 ) for p-tau. The 7p22 locus is novel and co-localized with the brain DNAAF5 expression. While no heterogeneity from underlying disease status was observed for the above GWAS signals, some disease risk loci suggested disease specific associations with these biomarkers. Conclusions Our study identified a novel association at the intronic region of DNAAF5 associated with increased levels of p-tau across all diseases. We also observed some disease specific genetic associations with these biomarkers.
Collapse
|
39
|
Béreau M, Van Waes V, Servant M, Magnin E, Tatu L, Anheim M. Apathy in Parkinson's Disease: Clinical Patterns and Neurobiological Basis. Cells 2023; 12:1599. [PMID: 37371068 DOI: 10.3390/cells12121599] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Apathy is commonly defined as a loss of motivation leading to a reduction in goal-directed behaviors. This multidimensional syndrome, which includes cognitive, emotional and behavioral components, is one of the most prevalent neuropsychiatric features of Parkinson's disease (PD). It has been established that the prevalence of apathy increases as PD progresses. However, the pathophysiology and anatomic substrate of this syndrome remain unclear. Apathy seems to be underpinned by impaired anatomical structures that link the prefrontal cortex with the limbic system. It can be encountered in the prodromal stage of the disease and in fluctuating PD patients receiving bilateral chronic subthalamic nucleus stimulation. In these stages, apathy may be considered as a disorder of motivation that embodies amotivational behavioral syndrome, is underpinned by combined dopaminergic and serotonergic denervation and is dopa-responsive. In contrast, in advanced PD patients, apathy may be considered as cognitive apathy that announces cognitive decline and PD dementia, is underpinned by diffuse neurotransmitter system dysfunction and Lewy pathology spreading and is no longer dopa-responsive. In this review, we discuss the clinical patterns of apathy and their treatment, the neurobiological basis of apathy, the potential role of the anatomical structures involved and the pathways in motivational and cognitive apathy.
Collapse
Affiliation(s)
- Matthieu Béreau
- Département de Neurologie, CHU de Besançon, 25000 Besançon, France
- Université de Franche-Comté, LINC Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive, 25000 Besançon, France
| | - Vincent Van Waes
- Université de Franche-Comté, LINC Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive, 25000 Besançon, France
| | - Mathieu Servant
- Université de Franche-Comté, LINC Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive, 25000 Besançon, France
| | - Eloi Magnin
- Département de Neurologie, CHU de Besançon, 25000 Besançon, France
- Université de Franche-Comté, LINC Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive, 25000 Besançon, France
| | - Laurent Tatu
- Département de Neurologie, CHU de Besançon, 25000 Besançon, France
- Université de Franche-Comté, LINC Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive, 25000 Besançon, France
- Laboratoire d'Anatomie, Université de Franche-Comté, 25000 Besançon, France
| | - Mathieu Anheim
- Département de Neurologie, CHU de Strasbourg, 67200 Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67000 Strasbourg, France
- Institut de génétique Et de Biologie Moléculaire Et Cellulaire (IGBMC), INSERM-U964, CNRS-UMR7104, Université de Strasbourg, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
40
|
Weintraub D, Picillo M, Cho HR, Caspell‐Garcia C, Blauwendraat C, Brown EG, Chahine LM, Coffey CS, Dobkin RD, Foroud T, Galasko D, Kieburtz K, Marek K, Merchant K, Mollenhauer B, Poston KL, Simuni T, Siderowf A, Singleton A, Seibyl J, Tanner CM. Impact of the Dopamine System on Long-Term Cognitive Impairment in Parkinson Disease: An Exploratory Study. Mov Disord Clin Pract 2023; 10:943-955. [PMID: 37332638 PMCID: PMC10272925 DOI: 10.1002/mdc3.13751] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/09/2023] [Accepted: 04/02/2023] [Indexed: 06/20/2023] Open
Abstract
Background Little is known about the impact of the dopamine system on development of cognitive impairment (CI) in Parkinson disease (PD). Objectives We used data from a multi-site, international, prospective cohort study to explore the impact of dopamine system-related biomarkers on CI in PD. Methods PD participants were assessed annually from disease onset out to 7 years, and CI determined by applying cut-offs to four measures: (1) Montreal Cognitive Assessment; (2) detailed neuropsychological test battery; (3) Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS) cognition score; and (4) site investigator diagnosis of CI (mild cognitive impairment or dementia). The dopamine system was assessed by serial Iodine-123 Ioflupane dopamine transporter (DAT) imaging, genotyping, and levodopa equivalent daily dose (LEDD) recorded at each assessment. Multivariate longitudinal analyses, with adjustment for multiple comparisons, determined the association between dopamine system-related biomarkers and CI, including persistent impairment. Results Demographic and clinical variables associated with CI were higher age, male sex, lower education, non-White race, higher depression and anxiety scores and higher MDS-UPDRS motor score. For the dopamine system, lower baseline mean striatum dopamine transporter values (P range 0.003-0.005) and higher LEDD over time (P range <0.001-0.01) were significantly associated with increased risk for CI. Conclusions Our results provide preliminary evidence that alterations in the dopamine system predict development of clinically-relevant, cognitive impairment in Parkinson's disease. If replicated and determined to be causative, they demonstrate that the dopamine system is instrumental to cognitive health status throughout the disease course. TRIAL REGISTRATION Parkinson's Progression Markers Initiative is registered with ClinicalTrials.gov (NCT01141023).
Collapse
Affiliation(s)
- Daniel Weintraub
- Department of PsychiatryPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Marina Picillo
- Assistant Professor in Neurology at the Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”University of SalernoItaly
| | - Hyunkeun Ryan Cho
- Department of Biostatistics, College of Public HealthUniversity of IowaIowa CityIowaUSA
| | | | - Cornelis Blauwendraat
- Center for Alzheimer's and Related Dementias, and the Integrative Neurogenomics Unit, Laboratory of NeurogeneticsNational Institute on Aging, National Institutes of HealthBethesdaMarylandUSA
| | - Ethan G. Brown
- Department of NeurologyWeill Institute for Neurosciences, University of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Lana M. Chahine
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Christopher S. Coffey
- Department of Biostatistics, College of Public HealthUniversity of IowaIowa CityIowaUSA
| | - Roseanne D. Dobkin
- Department of PsychiatryRutgers University, Robert Wood Johnson Medical SchoolPiscatawayNew JerseyUSA
| | - Tatiana Foroud
- Department of Medical and Molecular GeneticsIndiana UniversityIndianapolisIndianaUSA
| | - Doug Galasko
- Department of NeurologyUniversity of CaliforniaSan DiegoCaliforniaUSA
| | - Karl Kieburtz
- Department of NeurologyUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - Kenneth Marek
- Institute for Neurodegenerative DisordersNew HavenConnecticutUSA
| | - Kalpana Merchant
- Department of NeurologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Brit Mollenhauer
- Department of NeurologyUniversity Medical Center GoettingenGoettingenGermany
| | - Kathleen L. Poston
- Department of Neurology and Neurological SciencesStanford UniversityStanfordCaliforniaUSA
| | - Tanya Simuni
- Department of NeurologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Andrew Siderowf
- Department of NeurologyPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Andrew Singleton
- Center for Alzheimer's and Related Dementias, and the Molecular Genetics SectionLaboratory of Neurogenetics, National Institute on Aging, National Institutes of HealthBethesdaMarylandUSA
| | - John Seibyl
- Institute for Neurodegenerative DisordersNew HavenConnecticutUSA
| | - Caroline M. Tanner
- Department of NeurologyWeill Institute for Neurosciences, University of California, San FranciscoSan FranciscoCaliforniaUSA
| | | |
Collapse
|
41
|
Gonzalez MC, Tovar‐Rios DA, Alves G, Dalen I, Williams‐Gray CH, Camacho M, Forsgren L, Bäckström D, Lawson RA, Macleod AD, Counsell CE, Paquet C, DeLena C, D'Antonio F, Pilotto A, Padovani A, Blanc F, Falup‐Pecurariu C, Lewis SJ, Rejdak K, Papuc E, Hort J, Nedelska Z, O'Brien J, Bonanni L, Marquié M, Boada M, Pytel V, Abdelnour C, Alcolea D, Beyer K, Tysnes O, Aarsland D, Maple‐Grødem J. Cognitive and Motor Decline in Dementia with Lewy Bodies and Parkinson's Disease Dementia. Mov Disord Clin Pract 2023; 10:980-986. [PMID: 37332651 PMCID: PMC10272890 DOI: 10.1002/mdc3.13752] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/09/2023] [Accepted: 03/29/2023] [Indexed: 06/20/2023] Open
Abstract
Background There is a need to better understand the rate of cognitive and motor decline of Dementia with Lewy bodies (DLB) and Parkinson's disease Dementia (PDD). Objectives To compare the rate of cognitive and motor decline in patients with DLB and PDD from the E-DLB Consortium and the Parkinson's Incidence Cohorts Collaboration (PICC) Cohorts. Methods The annual change in MMSE and MDS-UPDRS part III was estimated using linear mixed regression models in patients with at least one follow-up (DLB n = 837 and PDD n = 157). Results When adjusting for confounders, we found no difference in the annual change in MMSE between DLB and PDD (-1.8 [95% CI -2.3, -1.3] vs. -1.9 [95% CI -2.6, -1.2] [P = 0.74]). MDS-UPDRS part III showed nearly identical annual changes (DLB 4.8 [95% CI 2.1, 7.5]) (PDD 4.8 [95% CI 2.7, 6.9], [P = 0.98]). Conclusions DLB and PDD showed similar rates of cognitive and motor decline. This is relevant for future clinical trial designs.
Collapse
Affiliation(s)
- Maria Camila Gonzalez
- Department of Quality and Health Technology, Faculty of Health SciencesUniversity of StavangerStavangerNorway
- The Norwegian Centre for Movement DisordersStavanger University HospitalStavangerNorway
- Centre for Age‐Related MedicineStavanger University HospitalStavangerNorway
| | - Diego Alejandro Tovar‐Rios
- Centre for Age‐Related MedicineStavanger University HospitalStavangerNorway
- Grupos de investigación INFERIR and PRECEC, Section of BiostatisticsUniversidad del ValleSantiago de CaliColombia
| | - Guido Alves
- The Norwegian Centre for Movement DisordersStavanger University HospitalStavangerNorway
- Department of Chemistry, Bioscience and Environmental EngineeringUniversity of StavangerStavangerNorway
- Department of NeurologyStavanger University HospitalStavangerNorway
| | - Ingvild Dalen
- Department of NeurologyStavanger University HospitalStavangerNorway
| | | | - Marta Camacho
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeEngland
| | - Lars Forsgren
- Department of Clinical Science, NeurosciencesUmeå UniversityUmeåSweden
| | - David Bäckström
- Department of Clinical Science, NeurosciencesUmeå UniversityUmeåSweden
| | - Rachael A. Lawson
- Translational and Clinical Research InstituteNewcastle UniversityTyneUK
| | - Angus D. Macleod
- Institute of Applied Health SciencesUniversity of AberdeenAberdeenUK
| | - Carl E. Counsell
- Institute of Applied Health SciencesUniversity of Aberdeen, Polwarth BuildingAberdeenUK
| | - Claire Paquet
- Université de Paris, Cognitive Neurology Center, APHP, Lariboisière Fernand‐Widal HospitalParisFrance
| | - Carlo DeLena
- Department of Human NeurosciencesSapienza University of RomeRomeItaly
| | | | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
| | - Frédéric Blanc
- Memory Resource and Research Centre (CM2R), Geriatrics Day Hospital, Geriatrics DepartmentUniversity Hospital of StrasbourgStrasbourg CedexFrance
| | - Cristian Falup‐Pecurariu
- Department of Neurology, County Clinic Hospital, Faculty of MedicineTransilvania UniversityBrasovRomania
| | | | - Konrad Rejdak
- Department of NeurologyMedical University of LublinLublinPoland
| | - Ewa Papuc
- Department of NeurologyMedical University of LublinLublinPoland
| | - Jakub Hort
- Memory Clinic, Department of NeurologyCharles University, 2nd Faculty of Medicine and Motol University HospitalPragueCzech Republic
| | - Zuzana Nedelska
- Memory Clinic, Department of NeurologyCharles University, 2nd Faculty of Medicine and Motol University HospitalPragueCzech Republic
| | - John O'Brien
- Department of PsychiatryUniversity of Cambridge School of Clinical MedicineCambridgeUK
| | - Laura Bonanni
- Department of Medicine and Aging SciencesUniversity Gd'Annunzio of Chieti‐PescaraChietiItaly
| | - Marta Marquié
- Ace Alzheimer Center Barcelona—Universitat Internacional de CatalunyaBarcelonaSpain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona—Universitat Internacional de CatalunyaBarcelonaSpain
| | - Vanesa Pytel
- Ace Alzheimer Center Barcelona—Universitat Internacional de CatalunyaBarcelonaSpain
| | - Carla Abdelnour
- Department of Neurology and Neurological SciencesStanford University School of MedicineStanfordCaliforniaUSA
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, IIB Sant Pau—Hospital de Sant PauUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Katrin Beyer
- Department NeuroscienceResearch Institute Germans Trias i PujolBadalonaSpain
| | - Ole‐Bjørn Tysnes
- Department of NeurologyHaukeland University HospitalBergenNorway
| | - Dag Aarsland
- Centre for Age‐Related MedicineStavanger University HospitalStavangerNorway
- Department of Old Age PsychiatryInstitute of Psychiatry, Psychology, and Neuroscience, King's College LondonLondonUK
| | - Jodi Maple‐Grødem
- The Norwegian Centre for Movement DisordersStavanger University HospitalStavangerNorway
- Department of Chemistry, Bioscience and Environmental EngineeringUniversity of StavangerStavangerNorway
| |
Collapse
|
42
|
Martinez-Carrasco A, Real R, Lawton M, Iwaki H, Tan MMX, Wu L, Williams NM, Carroll C, Hu MT, Grosset DG, Hardy J, Ryten M, Foltynie T, Ben-Shlomo Y, Shoai M, Morris HR. Genetic meta-analysis of levodopa induced dyskinesia in Parkinson's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.24.23290362. [PMID: 37425912 PMCID: PMC10327264 DOI: 10.1101/2023.05.24.23290362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Importance Forty percent of Parkinson's disease patients develop levodopa-induced-dyskinesia (LiD) within 4 years of starting levodopa. The genetic basis of LiD remains poorly understood, and there have been few well powered studies. Objective To discover common genetic variants in the PD population that increase the probability of developing LiD. Design setting and Participants We performed survival analyses to study the development of LiD in 5 separate longitudinal cohorts. We performed a meta-analysis to combine the results of genetic association from each study based on a fixed effects model weighting the effect sizes by the inverse of their standard error. The selection criteria was specific to each cohort. We studied individuals that were genotyped from each cohort and that passed our analysis specific inclusion criteria. Main Outcomes and Measures We measured the time for PD patients on levodopa treatment to develop LiD as defined by reaching a score higher or equal than 2 from the MDS-UPDRS part IV, item 1, which is equivalent to a range of 26%-50% of the waking time with dyskinesia. We carried out a genome-wide analysis of the hazard ratio and the association of genome-wide SNPs with the probability of developing LiD using cox proportional hazard models (CPH). Results This study included 2,784 PD patients of European ancestry, of whom 14.6% developed LiD. Consistent with previous studies, we found female gender (HR = 1.35, SE = 0.11, P = 0.007) and younger age at onset (HR = 1.8, SE = 0.14, P = 2 × 10 -5 ) to increase the probability of developing LiD. We identified three loci significantly associated with time-to-LiD onset. rs72673189 on chromosome 1 (HR = 2.77, SE = 0.18, P = 1.53 × 10 -8 ) located in the LRP8 locus, rs189093213 on chromosome 4 (HR = 3.06,, SE = 0.19, P = 2.81 × 10 -9 ) in the non-coding RNA LINC02353 locus, and rs180924818 on chromosome 16 (HR = 3.13, SE = 0.20, P = 6.27 × 10 -9 ) in the XYLT1 locus. Subsequent colocalization analyses on chromosome 1 identified DNAJB4 as a candidate gene associated with LiD through a change in gene expression. We computed a PRS based on our GWAS meta-analysis and found high accuracy to stratify between PD-LID and PD (AUC 83.9). We also performed a stepwise regression analysis for baseline features selection associated with LiD status. We found baseline anxiety status to be significantly associated with LiD (OR = 1.14, SE = 0.03, P = 7.4 × 10 -5 ). Finally, we performed a candidate variant analysis and found that genetic variability in ANKK1 ( rs1800497 , Beta = 0.24, SE = 0.09, P = 8.89 × 10 -3 ) and BDNF ( rs6265 , Beta = 0.19, SE = 0.10, P = 4.95 × 10 -2 ) loci were significantly associated with time to LiD in our large meta-analysis. Conclusion In this association study, we have found three novel genetic variants associated with LiD, as well as confirming reports that variability in ANKK1 and BDNF loci were significantly associated with LiD probability. A PRS nominated from our time-to-LiD meta-analysis significantly differentiated between PD-LiD and PD. In addition, we have found female gender, young PD onset and anxiety to be significantly associated with LiD.
Collapse
Affiliation(s)
- Alejandro Martinez-Carrasco
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, UK
- UCL Movement Disorders Centre, University College London, London, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| | - Raquel Real
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, UK
- UCL Movement Disorders Centre, University College London, London, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| | - Michael Lawton
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, 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, Glen Echo, Maryland, USA
| | | | - Lesley Wu
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, UK
- UCL Movement Disorders Centre, University College London, London, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| | - Nigel M. Williams
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Camille Carroll
- Faculty of Health, University of Plymouth, Plymouth, UK; Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - 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
| | - Donald G. Grosset
- School of Neuroscience and Psychology, University of Glasgow, Glasgow, UK
| | - John Hardy
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, 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
| | - Mina Ryten
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| | - Tom Foltynie
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, UK
| | - Yoav Ben-Shlomo
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Maryam Shoai
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
| | - Huw R. Morris
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, UK
- UCL Movement Disorders Centre, University College London, London, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| |
Collapse
|
43
|
Yang N, Sang S, Peng T, Hu W, Wang J, Bai R, Lu H. Impact of GBA variants on longitudinal freezing of gait progression in early Parkinson's disease. J Neurol 2023; 270:2756-2764. [PMID: 36790548 DOI: 10.1007/s00415-023-11612-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND Freezing of gait (FOG) is a common disabling gait disturbance among patients with Parkinson's disease (PD), but the influence of genetic variants on the incidence of FOG has been poorly studied to date. OBJECTIVES We aimed to evaluate the association of GBA variants with the risk of FOG development in a large early PD cohort. METHODS This study included 371 early PD patients from the Parkinson's Progression Markers Initiative (PPMI) who were divided into a GBA variant carrier group (GBA-PD group, n = 44) and an idiopathic PD group without GBA variants (iPD group, n = 327). They were followed up for up to 5 years to examine the progression of FOG. The cumulative incidence of FOG and risk factors for FOG were assessed using Kaplan‒Meier and Cox regression analyses. RESULTS At baseline, the GBA-PD group had lower CSF β-amyloid 1-42 (Aβ42) levels and more severe motor and nonmotor symptoms than the iPD group. During the 5-year follow-up, the GBA-PD group had a higher incidence of FOG than the iPD group, and the FOG progression rate was related to GBA variant severity. In the multivariable Cox model without CSF Aβ42, GBA variants were significant predictors of future FOG, and the association remained significant after adding CSF Aβ42 to the model. In the subgroup analyses, the effect of GBA variants was not observed in the "low-level" group. However, in the "high-level" group, GBA variants independently increased the risk of FOG, and this association was stronger than the association with CSF Aβ42. CONCLUSION GBA variants are novel genetic risk factors for future FOG development in early PD patients. This association seemed to be mediated by both Aβ-dependent pathways and Aβ-independent pathways.
Collapse
Affiliation(s)
- Nannan Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Shushan Sang
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Tao Peng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Wentao Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jingtao Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Rong Bai
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hong Lu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
44
|
Ren J, Zhou G, Wang Y, Zhang R, Guo Z, Zhou H, Zheng H, Sun Y, Ma C, Lu M, Liu W. Association of GBA genotype with motor and cognitive decline in Chinese Parkinson's disease patients. Front Aging Neurosci 2023; 15:1091919. [PMID: 36845659 PMCID: PMC9950580 DOI: 10.3389/fnagi.2023.1091919] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/23/2023] [Indexed: 02/12/2023] Open
Abstract
Objective Variants in the glucocerebrosidase (GBA) gene are the most common and significant risk factor for Parkinson's disease (PD). However, the impact of GBA variants on PD disease progression in the Chinese population remains unclear. This study aimed to explore the significance of GBA status on motor and cognitive impairment in a longitudinal cohort of Chinese patients with PD. Methods The entire GBA gene was screened by long-range polymerase chain reaction (LR-PCR) and next generation sequencing (NGS). A total of 43 GBA-related PD (GBA-PD) and 246 non-GBA-mutated PD (NM-PD) patients with complete clinical data at baseline and at least one follow-up were recruited for this study. The associations of GBA genotype with rate of motor and cognitive decline, as measured by Unified PD Rating Scale (UPDRS) motor and Montreal Cognitive Assessment (MoCA), were assessed by linear mixed-effect models. Results The estimated (standard error, SE) UPDRS motor [2.25 (0.38) points/year] and MoCA [-0.53 (0.11) points/year] progression rates in the GBA-PD group were significantly faster than those in the NM-PD group [1.35 (0.19); -0.29 (0.04) points/year; respectively]. In addition, the GBA-PD group showed significantly faster estimated (SE) bradykinesia [1.04 (0.18) points/year], axial impairment [0.38 (0.07) points/year], and visuospatial/executive [-0.15 (0.03) points/year] progression rates than the NM-PD group [0.62 (0.10); 0.17 (0.04); -0.07 (0.01) points/year; respectively]. Conclusion GBA-PD is associated with faster motor and cognitive decline, specifically greater disability in terms of bradykinesia, axial impairment, and visuospatial/executive function. Better understanding of GBA-PD progression may help predict prognosis and improve clinical trial design.
Collapse
Affiliation(s)
- Jingru Ren
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Gaiyan Zhou
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yajie Wang
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Ronggui Zhang
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Zhiying Guo
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Zhou
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Huifen Zheng
- Department of Neurology, Geriatric Hospital of Nanjing Medical University, Nanjing, China
| | - Yu Sun
- International Laboratory for Children’s Medical Imaging Research, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, China
| | - Changyan Ma
- Department of Medical Genetics, Nanjing Medical University, Nanjing, China
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Weiguo Liu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China,*Correspondence: Weiguo Liu,
| |
Collapse
|
45
|
Liu JY, Ma LZ, Wang J, Cui XJ, Sheng ZH, Fu Y, Li M, Ou YN, Yu JT, Tan L, Lian Y. Age-Related Association Between APOE ɛ4 and Cognitive Progression in de novo Parkinson's Disease. J Alzheimers Dis 2023; 91:1121-1132. [PMID: 36565124 DOI: 10.3233/jad-220976] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND APOE ɛ4 genotype was correlated with exacerbation of pathology and higher risk of dementia in Parkinson's disease (PD). Meanwhile, the differential influence of APOE ɛ4 on cognition in young and old individuals interpreted as antagonistic pleiotropy. OBJECTIVE To examine whether the effect of APOE ɛ4 on cognitive progression in de novo PD is age dependent. METHODS In this study, 613 de novo PD patients were recruited from Parkinson's Progression Markers Initiative (PPMI). To examine the age-dependent relationship between APOE ɛ4 and cognitive changes, we added 3-way interaction of APOE ɛ4*baseline age*time to the linear mixed-effect (LME) models and evaluated the specific roles of APOE ɛ4 in the middle age group and elderly group separately. Cox regression was utilized to examine the progression of cognition in age-stratified PD participants. RESULTS Age significantly modified relationship between APOE ɛ4 and cognitive changes in most cognitive domains (pinteraction <0.05). In the elderly group, APOE ɛ4 carriers showed steeper decline in global cognition (p = 0.001) as well as in most cognitive domains, and they had a greater risk of cognitive progression (adjusted HR 1.625, 95% CI 1.143-2.310, p = 0.007), compared with non-carriers. However, in the middle age group, no significant relationships between APOE ɛ4 and cognitive decline can be detected. CONCLUSION Our results indicated that the APOE ɛ4 allele has an age-dependent effect on cognitive decline in PD patients. The underlying mechanisms need to be investigated in the future.
Collapse
Affiliation(s)
- Jia-Yao Liu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ling-Zhi Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jun Wang
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.,Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xin-Jing Cui
- Department of Outpatient, Qingdao Municipal Hospital, Qingdao, China
| | - Ze-Hu Sheng
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Meng Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan Lian
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.,Department of Prevention and Health Care, Daping Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
46
|
Miao G, Zhuo D, Han X, Yao W, Liu C, Liu H, Cao H, Sun Y, Chen Z, Feng T. From degenerative disease to malignant tumors: Insight to the function of ApoE. Biomed Pharmacother 2023; 158:114127. [PMID: 36516696 DOI: 10.1016/j.biopha.2022.114127] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/03/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Apolipoprotein E (ApoE) is a multifunctional protein involved in lipid transport and lipoprotein metabolism, mediating lipid distribution/redistribution in tissues and cells. It can also regulate inflammation and immune function, maintain cytoskeleton stability, and improve neural tissue Function. Due to genetic polymorphisms of ApoE (ε2, ε3, and ε4), its three common structural isoforms (ApoE2, ApoE3, ApoE4) are also associated with the risk of many diseases, especially degenerative diseases, such as vascular degenerative diseases including atherosclerosis (AS), coronary heart disease (CHD), and neurodegenerative disease like Alzheimer's disease (AD). The frequency of the ε4 allele and APOE variants were significantly higher than that of the ε2 and ε3 alleles in the patients with CHD or AD. In recent years, ApoE has frequently appeared in tumor research and become a tumor biomarker gradually. It has been found that ApoE is highly expressed in most solid tumor tissues, such as glioblastoma, gastric cancer, pancreatic ductal cell carcinoma, etc. Studies illustrated that ApoE could regulate the polarization changes of macrophages, participate in the construction of tumor immune microenvironment, regulate tumor inflammation and immune response and play a role in tumor progression, invasion, and metastasis. Of course, many functions of ApoE and its relationship with diseases are still under research. By reviewing the structure and function of ApoE from degeneration diseases to tumor neoplasms, we hope to better understand such a biomarker and further explore the value of ApoE in later studies.
Collapse
Affiliation(s)
- Ganggang Miao
- Department of General Surgery, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang, Jiangsu, China; Department of General Surgery, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Danping Zhuo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xue Han
- Department of Clinical Laboratory, the Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Wentao Yao
- Department of Urology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu, China
| | - Chuan Liu
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Transplantation, Chinese Academy of Medical Sciences, Nanjing, China
| | - Hanyuan Liu
- Department of General Surgery, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongyong Cao
- Department of General Surgery, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Yangbai Sun
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Zhiqiang Chen
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Tingting Feng
- Jiangsu Key Laboratory of Infection and Immunity, Institute of Biology and Medical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
47
|
Muñoz-Delgado L, Macías-García D, Periñán MT, Jesús S, Adarmes-Gómez AD, Bonilla Toribio M, Buiza Rueda D, Jiménez-Jaraba MDV, Benítez Zamora B, Díaz Belloso R, García-Díaz S, Martín-Bórnez M, Pineda Sánchez R, Carrillo F, Gómez-Garre P, Mir P. Peripheral inflammatory immune response differs among sporadic and familial Parkinson's disease. NPJ Parkinsons Dis 2023; 9:12. [PMID: 36720879 PMCID: PMC9889312 DOI: 10.1038/s41531-023-00457-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 01/13/2023] [Indexed: 02/01/2023] Open
Abstract
Peripheral inflammatory immune responses are thought to play a major role in the pathogenesis of Parkinson's disease (PD). The neutrophil-to-lymphocyte ratio (NLR), a biomarker of systemic inflammation, has been reported to be higher in patients with PD than in healthy controls (HCs). The present study was aimed at determining if the peripheral inflammatory immune response could be influenced by the genetic background of patients with PD. We included a discovery cohort with 222 patients with PD (132 sporadic PD, 44 LRRK2-associated PD (with p.G2019S and p.R1441G variants), and 46 GBA-associated PD), as well as 299 HCs. Demographic and clinical data were recorded. Leukocytes and their subpopulations, and the NLR were measured in peripheral blood. Multivariate lineal regression and post-hoc tests were applied to determine the differences among the groups. Subsequently, a replication study using the Parkinson's Progression Markers Initiative cohort was performed which included 401 patients with PD (281 sPD patients, 66 LRRK2-PD patients, 54 GBA-PD patients) and a group of 174 HCs. Patients with sporadic PD and GBA-associated PD showed a significantly lower lymphocyte count, a non-significantly higher neutrophil count and a significantly higher NLR than HCs. The peripheral inflammatory immune response of patients with LRRK2-associated PD did not differ from HCs. Our study supports the involvement of a peripheral inflammatory immune response in the pathophysiology of sPD and GBA-associated PD. However, this inflammatory response was not found in LRRK2-associated PD, probably reflecting different pathogenic inflammatory mechanisms.
Collapse
Affiliation(s)
- Laura Muñoz-Delgado
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, 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 (CIBERNED), Madrid, Spain
| | - Daniel Macías-García
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, 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 (CIBERNED), Madrid, Spain
| | - María Teresa Periñán
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, 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 (CIBERNED), Madrid, Spain
| | - Silvia Jesús
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, 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 (CIBERNED), Madrid, Spain
| | - Astrid D Adarmes-Gómez
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, 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 (CIBERNED), Madrid, Spain
| | - Marta Bonilla Toribio
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, 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 (CIBERNED), Madrid, Spain
| | - Dolores Buiza Rueda
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, 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 (CIBERNED), Madrid, Spain
| | - María Del Valle Jiménez-Jaraba
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Belén Benítez Zamora
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Rafael Díaz Belloso
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, 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 (CIBERNED), Madrid, Spain
| | - Sergio García-Díaz
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, 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 (CIBERNED), Madrid, Spain
| | - Miguel Martín-Bórnez
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Rocío Pineda Sánchez
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, 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 (CIBERNED), Madrid, Spain
| | - Fátima Carrillo
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, 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 (CIBERNED), Madrid, Spain
| | - Pilar Gómez-Garre
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, 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 (CIBERNED), Madrid, Spain.
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, 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 (CIBERNED), Madrid, Spain.
- Departamento de Medicina, Universidad de Sevilla, Seville, Spain.
| |
Collapse
|
48
|
Abstract
Parkinson's disease (PD) is clinically, pathologically, and genetically heterogeneous, resisting distillation to a single, cohesive disorder. Instead, each affected individual develops a virtually unique form of Parkinson's syndrome. Clinical manifestations consist of variable motor and nonmotor features, and myriad overlaps are recognized with other neurodegenerative conditions. Although most commonly characterized by alpha-synuclein protein pathology throughout the central and peripheral nervous systems, the distribution varies and other pathologies commonly modify PD or trigger similar manifestations. Nearly all PD is genetically influenced. More than 100 genes or genetic loci have been identified, and most cases likely arise from interactions among many common and rare genetic variants. Despite its complex architecture, insights from experimental genetic dissection coalesce to reveal unifying biological themes, including synaptic, lysosomal, mitochondrial, andimmune-mediated mechanisms of pathogenesis. This emerging understanding of Parkinson's syndrome, coupled with advances in biomarkers and targeted therapies, presages successful precision medicine strategies.
Collapse
Affiliation(s)
- Hui Ye
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA; ,
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Laurie A Robak
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA;
| | - Meigen Yu
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA;
| | - Matthew Cykowski
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA;
- Department of Neurology, Houston Methodist Hospital, Houston, Texas, USA
| | - Joshua M Shulman
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA; ,
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA;
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA;
- Center for Alzheimer's and Neurodegenerative Diseases, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
49
|
GBA1 Gene Mutations in α-Synucleinopathies-Molecular Mechanisms Underlying Pathology and Their Clinical Significance. Int J Mol Sci 2023; 24:ijms24032044. [PMID: 36768367 PMCID: PMC9917178 DOI: 10.3390/ijms24032044] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
α-Synucleinopathies comprise a group of neurodegenerative diseases characterized by altered accumulation of a protein called α-synuclein inside neurons and glial cells. This aggregation leads to the formation of intraneuronal inclusions, Lewy bodies, that constitute the hallmark of α-synuclein pathology. The most prevalent α-synucleinopathies are Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). To date, only symptomatic treatment is available for these disorders, hence new approaches to their therapy are needed. It has been observed that GBA1 mutations are one of the most impactful risk factors for developing α-synucleinopathies such as PD and DLB. Mutations in the GBA1 gene, which encodes a lysosomal hydrolase β-glucocerebrosidase (GCase), cause a reduction in GCase activity and impaired α-synuclein metabolism. The most abundant GBA1 gene mutations are N370S or N409S, L444P/L483P and E326K/E365K. The mechanisms by which GCase impacts α-synuclein aggregation are poorly understood and need to be further investigated. Here, we discuss some of the potential interactions between α-synuclein and GCase and show how GBA1 mutations may impact the course of the most prevalent α-synucleinopathies.
Collapse
|
50
|
Neuronopathic GBA1L444P Mutation Accelerates Glucosylsphingosine Levels and Formation of Hippocampal Alpha-Synuclein Inclusions. J Neurosci 2023; 43:501-521. [PMID: 36639889 PMCID: PMC9864632 DOI: 10.1523/jneurosci.0680-22.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 09/09/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022] Open
Abstract
The most common genetic risk factor for Parkinson's disease (PD) is heterozygous mutations GBA1, which encodes for the lysosomal enzyme, glucocerebrosidase. Reduced glucocerebrosidase activity associates with an accumulation of abnormal α-synuclein (α-syn) called Lewy pathology, which characterizes PD. PD patients heterozygous for the neuronotypic GBA1L444P mutation (GBA1+/L444P) have a 5.6-fold increased risk of cognitive impairments. In this study, we used GBA1+/L444P mice of either sex to determine its effects on lipid metabolism, expression of synaptic proteins, behavior, and α-syn inclusion formation. At 3 months of age, GBA1+/L444P mice demonstrated impaired contextual fear conditioning, and increased motor activity. Hippocampal levels of vGLUT1 were selectively reduced in GBA1+/L444P mice. We show, using mass spectrometry, that GBA1L444P expression increased levels of glucosylsphingosine, but not glucosylceramide, in the brains and serum of GBA1+/L444P mice. Templated induction of α-syn pathology in mice showed an increase in α-syn inclusion formation in the hippocampus of GBA1+/L444P mice compared with GBA1+/+ mice, but not in the cortex, or substantia nigra pars compacta. Pathologic α-syn reduced SNc dopamine neurons by 50% in both GBA1+/+ and GBA1+/L444P mice. Treatment with a GlcCer synthase inhibitor did not affect abundance of α-syn inclusions in the hippocampus or rescue dopamine neuron loss. Overall, these data suggest the importance of evaluating the contribution of elevated glucosylsphingosine to PD phenotypes. Further, our data suggest that expression of neuronotypic GBA1L444P may cause defects in the hippocampus, which may be a mechanism by which cognitive decline is more prevalent in individuals with GBA1-PD.SIGNIFICANCE STATEMENT Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are both pathologically characterized by abnormal α-synuclein (α-syn). Mutant GBA1 is a risk factor for both PD and DLB. Our data show the expression of neuronotypic GBA1L444P impairs behaviors related to hippocampal function, reduces expression of a hippocampal excitatory synaptic protein, and that the hippocampus is more susceptible to α-syn inclusion formation. Further, our data strengthen support for the importance of evaluating the contribution of glucosylsphingosine to PD phenotypes. These outcomes suggest potential mechanisms by which GBA1L444P contributes to the cognitive symptoms clinically observed in PD and DLB. Our findings also highlight the importance of glucosylsphingosine as a relevant biomarker for future therapeutics.
Collapse
|