1
|
Ye M, Ji Q, Liu Q, Kang X, Zhan Y. Longitudinal associations of lipid profiles with sleep disorders in patients with Parkinson's disease. Lipids 2024. [PMID: 39702743 DOI: 10.1002/lipd.12428] [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: 08/29/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024]
Abstract
To examine the associations of apolipoprotein E (APOE) carrier status and lipid profiles with sleep disorders, including excessive daytime sleepiness (EDS) and probable rapid eye movement sleep behavior disorder (pRBD), among patients with early Parkinson's disease (PD) over 5-year follow-up periods. The Parkinson's Progression Markers Initiative is a multicenter cohort study based on an ongoing and open-ended registry. Data from baseline and 5-year follow-up visits from participants of de novo PD were analyzed. Longitudinal associations of APOE carrier status and lipid profiles with sleep disorders were estimated via linear mixed-effects models. A total of 657 participants with complete APOE genotypes were enrolled at baseline. Among them, 153 (25.3%) had available lipid profiles at baseline. In the linear mixed-effects models, baseline APOE ε2/ε3/ε4 carrier status did not exhibit significant associations with EDS and pRBD (all p > 0.05) in all models. However, reduced high-density lipoprotein (HDL) and elevated triglycerides (TG) were associated with developing EDS (β = -0.04, 95% CI: -0.07, -0.00) and pRBD (β = 0.01, 95% CI: 0.00, 0.02) in PD patients, respectively. In the APOE ε4+ subgroup, decreased HDL and increased TG displayed substantial associations with developing EDS and sleep disorders (all p < 0.05) in all models, respectively, whereas no significant differences were noted in the APOE ε4- subgroup (all p > 0.05). Our study did not demonstrate a clear association between APOE ε2/ε3/ε4 and sleep disorders in PD patients. However, the presence of APOE ε4 was associated with changes in lipid profiles, notably affecting TG and HDL levels.
Collapse
Affiliation(s)
- Meijie Ye
- Department of Epidemiology, School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Qianqian Ji
- Department of Epidemiology, School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Qi Liu
- Department of Epidemiology, School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Xiaoying Kang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Yiqiang Zhan
- Department of Epidemiology, School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
2
|
Chen L, Lu H, Mao L, Lin J, Liu P. Unraveling the interplay of β-amyloid pathology and Parkinson's disease progression: Insights from autopsy-confirmed patients. Heliyon 2024; 10:e39194. [PMID: 39524781 PMCID: PMC11543873 DOI: 10.1016/j.heliyon.2024.e39194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/05/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Background Parkinson's disease (PD) is a prevalent neurodegenerative disorder that manifests with both motor and non-motor symptoms, with α-synuclein misfolding recognized as a key contributor. Cognitive decline in advanced PD stages prompts interest in amyloid deposition, a hallmark of Alzheimer's disease (AD), as a potential factor. This study explores the impact of β-amyloid (Aβ) pathology in PD patients on disease progression, aiming to elucidate the role of Aβ in PD development and progression. Methods This study included autopsy-confirmed PD patients with post-mortem analyses from the Parkinson's Progression Markers Initiative. Comprehensive clinical assessments, including demographic data, clinical features, CSF markers, and neuroimaging, were conducted. Statistical analyses assessed differences between groups based on the severity of AD neuropathological changes. Results All 16 PD participants exhibited severe Lewy body pathology, with 75 % displaying AD neuropathological changes. At baseline, PD patients with severe or moderate AD neuropathological changes had a lower Aβ42 levels (p = 0.022) and Aβ42/tau ratio (p = 0.001). Longitudinal follow-up data indicated that individuals with severe or moderate AD neuropathological changes exhibited a more rapid decline in MOCA score and BJLOT score, along with a quicker increase in MDS-UPDRS Ⅲ score. Conclusions The study underscores the presence of severe Aβ pathology in PD, suggesting a role in accelerated disease progression. Cross-seeding between Aβ and α-synuclein may contribute to rapid clinical symptom progression. Further research is needed for a comprehensive understanding of neurodegenerative disease complexities and exploring potential therapeutic interventions targeting protein aggregation.
Collapse
Affiliation(s)
- Linxi Chen
- Department of Pathology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Hongsheng Lu
- Department of Pathology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Lingqun Mao
- Department of Neurology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Junxin Lin
- School of Medicine, Taizhou University, Taizhou, Zhejiang, China
| | - Peng Liu
- Department of Neurology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
- School of Medicine, Taizhou University, Taizhou, Zhejiang, China
| |
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
|
Chen L, He X, Mao L, Liu P. APOE contributes to longitudinal impulse control disorders progression in Parkinson's disease. BMC Psychiatry 2024; 24:632. [PMID: 39334114 PMCID: PMC11438395 DOI: 10.1186/s12888-024-06084-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Impulse control disorders (ICDs) are an increasingly recognized complication in Parkinson disease (PD). The pathogenesis of ICDs is currently unclear. Few genetic studies have been conducted in this area. OBJECTIVE We aimed to ascertain the correlation between APOE and ICDs, and identify clinical predictors of ICDs in PD. METHODS This study included 287 PD patients from the Parkinson's Progression Markers Initiative. They were followed up to investigate the progression of ICDs over a period of 5 years. The cumulative incidence of ICDs and potential risk factors were evaluated using Kaplan-Meier and Cox regression analyses. RESULTS 44.3% (31/70) patients with APOE ɛ4 and 32.3% (70/217) patients without APOE ɛ4 developed ICDs during the five-year follow up period. There were significant differences between the PD with and without ICDs development group in age, MSEADLG score, ESS score, GDS score, and STAI score at baseline. In multivariable Cox regression analysis, APOE ε4 (HR = 1.450, p = 0.048) and STAI score (HR = 1.017, p = 0.001) were predictors of the development of ICDs. Patients with APOE ɛ4 group showed significantly lower CSF Aβ42 and CSF α-syn level than patients without APOE ɛ4 group at baseline. In patients with APOE ɛ4 group, the "low α-syn level" group and the "low ptau/tau ratio" group had a significantly higher incidence of ICDs, respectively. CONCLUSIONS This study provides important insights into the potential role of the APOE gene in the development of ICDs in PD. Further studies are needed to confirm our findings and to investigate the underlying mechanisms in more detail.
Collapse
Affiliation(s)
- Linxi Chen
- Taizhou Central Hospital (Taizhou University Hospital), No. 999, Donghai Avenue, economic development zone, Taizhou, 318000, Zhejiang, China
| | - Xinwei He
- Taizhou Central Hospital (Taizhou University Hospital), No. 999, Donghai Avenue, economic development zone, Taizhou, 318000, Zhejiang, China
| | - Lingqun Mao
- Taizhou Central Hospital (Taizhou University Hospital), No. 999, Donghai Avenue, economic development zone, Taizhou, 318000, Zhejiang, China
| | - Peng Liu
- Taizhou Central Hospital (Taizhou University Hospital), No. 999, Donghai Avenue, economic development zone, Taizhou, 318000, Zhejiang, China.
| |
Collapse
|
5
|
Liu P, Chen L, He X, Mao L. Predictors of the Rapid Progression in Prodromal Parkinson's Disease: A Longitudinal Follow-Up Study. Gerontology 2024; 70:595-602. [PMID: 38565088 DOI: 10.1159/000538515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 03/21/2024] [Indexed: 04/04/2024] Open
Abstract
INTRODUCTION Parkinson's disease (PD) is characterized by a prodromal phase preceding the onset of classic motor symptoms. The duration and clinical manifestations of prodromal PD vary widely, indicating underlying heterogeneity within this stage. This discrepancy prompts the question of whether specific factors contribute to the divergent rates of progression in prodromal PD. METHODS This study included prodromal PD patients from the Parkinson's progression marker initiative. They were followed up to assess the disease progression. The data collected during the follow-up period were analyzed to identify potential predictors of rapid disease progression in prodromal PD. RESULTS In this study, 61 individuals with prodromal PD were enrolled. Among them, 43 patients presented with both RBD and hyposmia, 17 had hyposmia alone, and 1 had RBD alone at baseline. 13 (21.3%) prodromal PD participants exhibited rapid disease progression, with two of these cases advancing to non-neurological diseases. Significant differences were observed between the rapid progression group and no rapid progression group in terms of MDS-UPDRS II score and UPSIT score. Longitudinal analysis showed a significant increase in the MDS-UPDRS III score and MDS-UPDRS total score in the rapid progression group. Regression analyses identified the MDS-UPDRS II score and UPSIT score as predictors of rapid disease progression in prodromal PD. CONCLUSION Our study findings suggest that the MDS-UPDRS II score and UPSIT score may serve as clinical markers associated with rapid disease progression. Further research and development of precise biomarkers and advanced assessment methods are needed to enhance our understanding of prodromal PD and its progression patterns.
Collapse
Affiliation(s)
- Peng Liu
- Department of Neurology, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, China
| | - Linxi Chen
- Department of Neurology, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, China
- Department of Pathology, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, China
| | - Xinwei He
- Department of Neurology, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, China
| | - Lingqun Mao
- Department of Neurology, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, China
| |
Collapse
|
6
|
Park M, Lee YG. Association of Family History and Polygenic Risk Score With Longitudinal Prognosis in Parkinson Disease. Neurol Genet 2024; 10:e200115. [PMID: 38169864 PMCID: PMC10759146 DOI: 10.1212/nxg.0000000000200115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/02/2023] [Indexed: 01/05/2024]
Abstract
Background and Objectives Evidence suggests that either family history or polygenic risk score (PRS) is associated with developing Parkinson disease (PD). However, little is known about the longitudinal prognosis of PD according to family history and higher PRS. Methods From the Parkinson's Progression Markers Initiative database, 395 patients with PD who followed up for more than 2 years were grouped into those with family history within first-degree, second-degree, and third-degree relatives (N = 127 [32.2%]) vs those without (N = 268 [67.8%]). The PRS of 386 patients was computed using whole-genome sequencing data. Longitudinal assessment of motor, cognition, and imaging based on dopaminergic degeneration was conducted during the regular follow-up period. Effects of family history, PRS, or both on longitudinal changes of cognition, motor severity, and nigrostriatal degeneration were tested using a linear mixed model. The risk of freezing of gait (FOG) according to family history was assessed using the Kaplan-Meier analysis and Cox regression models. Results During a median follow-up of 9.1 years, PD with positive family history showed a slower decline of caudate dopamine transporter uptake (β estimate of family history × time = 0.02, 95% CI = 0.002-0.036, p = 0.027). Family history of PD and higher PRS were independently associated with a slower decline of Montreal Cognitive Assessment (β estimate of family history × time = 0.12, 95% CI = 0.02-0.22, p = 0.017; β estimate of PRS × time = 0.09, 95% CI = 0.03-0.16, p = 0.006). In those 364 patients without FOG at baseline, PD with positive family history had a lower risk of FOG (hazard ratio of family history = 0.57, 95% CI = 0.38-0.84, p = 0.005). Discussion Having a family history of PD predicts slower progression of cognitive decline and caudate dopaminergic degeneration, and less FOG compared with those without a family history independent of PRS. Taken together, information on family history could be used as a proxy for the clinical heterogeneity of PD. Trial Registration Information The study was registered at clinicaltrials.gov (NCT01141023), and the enrollment began June 1, 2010.
Collapse
Affiliation(s)
- Mincheol Park
- From the Department of Neurology (M.P.), Gwangmyeong Hospital, Chung-Ang University College of Medicine; and Department of Neurology (Y.L.), Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| | - Young-Gun Lee
- From the Department of Neurology (M.P.), Gwangmyeong Hospital, Chung-Ang University College of Medicine; and Department of Neurology (Y.L.), Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| |
Collapse
|
7
|
Bharat V, Durairaj AS, Vanhauwaert R, Li L, Muir CM, Chandra S, Kwak CS, Le Guen Y, Nandakishore P, Hsieh CH, Rensi SE, Altman RB, Greicius MD, Feng L, Wang X. A mitochondrial inside-out iron-calcium signal reveals drug targets for Parkinson's disease. Cell Rep 2023; 42:113544. [PMID: 38060381 PMCID: PMC10804639 DOI: 10.1016/j.celrep.2023.113544] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/11/2023] [Accepted: 11/17/2023] [Indexed: 12/30/2023] Open
Abstract
Dysregulated iron or Ca2+ homeostasis has been reported in Parkinson's disease (PD) models. Here, we discover a connection between these two metals at the mitochondria. Elevation of iron levels causes inward mitochondrial Ca2+ overflow, through an interaction of Fe2+ with mitochondrial calcium uniporter (MCU). In PD neurons, iron accumulation-triggered Ca2+ influx across the mitochondrial surface leads to spatially confined Ca2+ elevation at the outer mitochondrial membrane, which is subsequently sensed by Miro1, a Ca2+-binding protein. A Miro1 blood test distinguishes PD patients from controls and responds to drug treatment. Miro1-based drug screens in PD cells discover Food and Drug Administration-approved T-type Ca2+-channel blockers. Human genetic analysis reveals enrichment of rare variants in T-type Ca2+-channel subtypes associated with PD status. Our results identify a molecular mechanism in PD pathophysiology and drug targets and candidates coupled with a convenient stratification method.
Collapse
Affiliation(s)
- Vinita Bharat
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aarooran S Durairaj
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Roeland Vanhauwaert
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Li Li
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Colin M Muir
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Graduate Program of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sujyoti Chandra
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chulhwan S Kwak
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yann Le Guen
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Institut du Cerveau - Paris Brain Institute - ICM, 75013 Paris, France
| | | | - Chung-Han Hsieh
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stefano E Rensi
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Russ B Altman
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Liang Feng
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xinnan Wang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
8
|
Zhou C, Wang L, Cheng W, Lv J, Guan X, Guo T, Wu J, Zhang W, Gao T, Liu X, Bai X, Wu H, Cao Z, Gu L, Chen J, Wen J, Huang P, Xu X, Zhang B, Feng J, Zhang M. Two distinct trajectories of clinical and neurodegeneration events in Parkinson's disease. NPJ Parkinsons Dis 2023; 9:111. [PMID: 37443179 PMCID: PMC10344958 DOI: 10.1038/s41531-023-00556-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Increasing evidence suggests that Parkinson's disease (PD) exhibits disparate spatial and temporal patterns of progression. Here we used a machine-learning technique-Subtype and Stage Inference (SuStaIn) - to uncover PD subtypes with distinct trajectories of clinical and neurodegeneration events. We enrolled 228 PD patients and 119 healthy controls with comprehensive assessments of olfactory, autonomic, cognitive, sleep, and emotional function. The integrity of substantia nigra (SN), locus coeruleus (LC), amygdala, hippocampus, entorhinal cortex, and basal forebrain were assessed using diffusion and neuromelanin-sensitive MRI. SuStaIn model with above clinical and neuroimaging variables as input was conducted to identify PD subtypes. An independent dataset consisting of 153 PD patients and 67 healthy controls was utilized to validate our findings. We identified two distinct PD subtypes: subtype 1 with rapid eye movement sleep behavior disorder (RBD), autonomic dysfunction, and degeneration of the SN and LC as early manifestations, and cognitive impairment and limbic degeneration as advanced manifestations, while subtype 2 with hyposmia, cognitive impairment, and limbic degeneration as early manifestations, followed later by RBD and degeneration of the LC in advanced disease. Similar subtypes were shown in the validation dataset. Moreover, we found that subtype 1 had weaker levodopa response, more GBA mutations, and poorer prognosis than subtype 2. These findings provide new insights into the underlying disease biology and might be useful for personalized treatment for patients based on their subtype.
Collapse
Affiliation(s)
- Cheng Zhou
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000, Hangzhou, China
| | - Linbo Wang
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, 200433, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Zhangjiang Fudan International Innovation Center, Shanghai, China
| | - Wei Cheng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, 200433, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Zhangjiang Fudan International Innovation Center, Shanghai, China.
- Department of Computer Science, University of Warwick, Coventry, CV4 7AL, United Kingdom.
| | - JinChao Lv
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, 200433, Shanghai, China
| | - Xiaojun Guan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000, Hangzhou, China
| | - Tao Guo
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000, Hangzhou, China
| | - Jingjing Wu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000, Hangzhou, China
| | - Wei Zhang
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, 200433, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Zhangjiang Fudan International Innovation Center, Shanghai, China
| | - Ting Gao
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000, Hangzhou, China
| | - Xiaocao Liu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000, Hangzhou, China
| | - Xueqin Bai
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000, Hangzhou, China
| | - Haoting Wu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000, Hangzhou, China
| | - Zhengye Cao
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000, Hangzhou, China
| | - Luyan Gu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000, Hangzhou, China
| | - Jingwen Chen
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000, Hangzhou, China
| | - Jiaqi Wen
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000, Hangzhou, China
| | - Peiyu Huang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000, Hangzhou, China
| | - Xiaojun Xu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000, Hangzhou, China
| | - Baorong Zhang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000, Hangzhou, China
| | - Jianfeng Feng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, 200433, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Zhangjiang Fudan International Innovation Center, Shanghai, China.
- Department of Computer Science, University of Warwick, Coventry, CV4 7AL, United Kingdom.
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310000, Hangzhou, China.
| |
Collapse
|
9
|
Chen S, Li Z, Liu L, Wen Y. The systematic comparison between Gaussian mirror and Model-X knockoff models. Sci Rep 2023; 13:5478. [PMID: 37015993 PMCID: PMC10073103 DOI: 10.1038/s41598-023-32605-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/30/2023] [Indexed: 04/06/2023] Open
Abstract
While the high-dimensional biological data have provided unprecedented data resources for the identification of biomarkers, consensus is still lacking on how to best analyze them. The recently developed Gaussian mirror (GM) and Model-X (MX) knockoff-based methods have much related model assumptions, which makes them appealing for the detection of new biomarkers. However, there are no guidelines for their practical use. In this research, we systematically compared the performance of MX-based and GM methods, where the impacts of the distribution of explanatory variables, their relatedness and the signal-to-noise ratio were evaluated. MX with knockoff generated using the second-order approximates (MX-SO) has the best performance as compared to other MX-based methods. MX-SO and GM have similar levels of power and computational speed under most of the simulations, but GM is more robust in the control of false discovery rate (FDR). In particular, MX-SO can only control the FDR well when there are weak correlations among explanatory variables and the sample size is at least moderate. On the contrary, GM can have the desired FDR as long as explanatory variables are not highly correlated. We further used GM and MX-based methods to detect biomarkers that are associated with the Alzheimer's disease-related PET-imaging trait and the Parkinson's disease-related T-tau of cerebrospinal fluid. We found that MX-based and GM methods are both powerful for the analysis of big biological data. Although genes selected from MX-based methods are more similar as compared to those from the GM method, both MX-based and GM methods can identify the well-known disease-associated genes for each disease. While MX-based methods can have a slightly higher power than that of the GM method, it is less robust, especially for data with small sample sizes, unknown distributions, and high correlations.
Collapse
Affiliation(s)
- Shuai Chen
- Department of Health Statistics, School of Public Health, Shanxi Medical University, No 56 Xinjian South Road, Yingze District, Taiyuan, Shanxi Province, China
| | - Ziqi Li
- Department of Health Statistics, School of Public Health, Shanxi Medical University, No 56 Xinjian South Road, Yingze District, Taiyuan, Shanxi Province, China
| | - Long Liu
- Department of Health Statistics, School of Public Health, Shanxi Medical University, No 56 Xinjian South Road, Yingze District, Taiyuan, Shanxi Province, China.
| | - Yalu Wen
- Department of Health Statistics, School of Public Health, Shanxi Medical University, No 56 Xinjian South Road, Yingze District, Taiyuan, Shanxi Province, China.
- Department of Statistics, University of Auckland, 38 Princes Street, Auckland Central, Auckland, New Zealand, 1010.
| |
Collapse
|
10
|
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: 1.5] [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
|
11
|
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: 2.0] [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
|
12
|
Sandor C, Millin S, Dahl A, Schalkamp AK, Lawton M, Hubbard L, Rahman N, Williams N, Ben-Shlomo Y, Grosset DG, Hu MT, Marchini J, Webber C. Universal clinical Parkinson's disease axes identify a major influence of neuroinflammation. Genome Med 2022; 14:129. [PMID: 36384636 PMCID: PMC9670420 DOI: 10.1186/s13073-022-01132-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 10/21/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND There is large individual variation in both clinical presentation and progression between Parkinson's disease patients. Generation of deeply and longitudinally phenotyped patient cohorts has enormous potential to identify disease subtypes for prognosis and therapeutic targeting. METHODS Replicating across three large Parkinson's cohorts (Oxford Discovery cohort (n = 842)/Tracking UK Parkinson's study (n = 1807) and Parkinson's Progression Markers Initiative (n = 472)) with clinical observational measures collected longitudinally over 5-10 years, we developed a Bayesian multiple phenotypes mixed model incorporating genetic relationships between individuals able to explain many diverse clinical measurements as a smaller number of continuous underlying factors ("phenotypic axes"). RESULTS When applied to disease severity at diagnosis, the most influential of three phenotypic axes "Axis 1" was characterised by severe non-tremor motor phenotype, anxiety and depression at diagnosis, accompanied by faster progression in cognitive function measures. Axis 1 was associated with increased genetic risk of Alzheimer's disease and reduced CSF Aβ1-42 levels. As observed previously for Alzheimer's disease genetic risk, and in contrast to Parkinson's disease genetic risk, the loci influencing Axis 1 were associated with microglia-expressed genes implicating neuroinflammation. When applied to measures of disease progression for each individual, integration of Alzheimer's disease genetic loci haplotypes improved the accuracy of progression modelling, while integrating Parkinson's disease genetics did not. CONCLUSIONS We identify universal axes of Parkinson's disease phenotypic variation which reveal that Parkinson's patients with high concomitant genetic risk for Alzheimer's disease are more likely to present with severe motor and non-motor features at baseline and progress more rapidly to early dementia.
Collapse
Affiliation(s)
- Cynthia Sandor
- UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK.
| | - Stephanie Millin
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Andrew Dahl
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | | | - Michael Lawton
- School of Social and Community Medicine, University of Bristol, Bristol, BS8 1TH, UK
| | - Leon Hubbard
- MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Nabila Rahman
- UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Nigel Williams
- MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Yoav Ben-Shlomo
- School of Social and Community Medicine, University of Bristol, Bristol, BS8 1TH, UK
| | - Donald G Grosset
- Department of Neurology, Institute of Neurological Sciences, Queen Elizabeth University Hospital, G51 4LB, Glasgow, UK
| | - Michele T Hu
- Department of Physiology, Anatomy and Genetics, Le Gros Clark Building, Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3PT, UK
- Nuffield Department of Clinical Neurosciences, Division of Clinical Neurology, University of Oxford, Oxford, OX3 7LF, UK
| | - Jonathan Marchini
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Department of Statistics, University of Oxford, Oxford, OX1, UK
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - Caleb Webber
- UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK.
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK.
| |
Collapse
|
13
|
Harvey J, Reijnders RA, Cavill R, Duits A, Köhler S, Eijssen L, Rutten BPF, Shireby G, Torkamani A, Creese B, Leentjens AFG, Lunnon K, Pishva E. Machine learning-based prediction of cognitive outcomes in de novo Parkinson's disease. NPJ Parkinsons Dis 2022; 8:150. [PMID: 36344548 PMCID: PMC9640625 DOI: 10.1038/s41531-022-00409-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 10/11/2022] [Indexed: 11/09/2022] Open
Abstract
Cognitive impairment is a debilitating symptom in Parkinson's disease (PD). We aimed to establish an accurate multivariate machine learning (ML) model to predict cognitive outcome in newly diagnosed PD cases from the Parkinson's Progression Markers Initiative (PPMI). Annual cognitive assessments over an 8-year time span were used to define two cognitive outcomes of (i) cognitive impairment, and (ii) dementia conversion. Selected baseline variables were organized into three subsets of clinical, biofluid and genetic/epigenetic measures and tested using four different ML algorithms. Irrespective of the ML algorithm used, the models consisting of the clinical variables performed best and showed better prediction of cognitive impairment outcome over dementia conversion. We observed a marginal improvement in the prediction performance when clinical, biofluid, and epigenetic/genetic variables were all included in one model. Several cerebrospinal fluid measures and an epigenetic marker showed high predictive weighting in multiple models when included alongside clinical variables.
Collapse
Affiliation(s)
- Joshua Harvey
- Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Rick A Reijnders
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Rachel Cavill
- Department of Advanced Computing Sciences, FSE, Maastricht University, Maastricht, The Netherlands
| | - Annelien Duits
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Medical Psychology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sebastian Köhler
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Lars Eijssen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Bioinformatics-BiGCaT, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Gemma Shireby
- Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Ali Torkamani
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, 92037, USA
| | - Byron Creese
- Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Albert F G Leentjens
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Katie Lunnon
- Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Ehsan Pishva
- Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK.
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
14
|
Dou K, Ma J, Zhang X, Shi W, Tao M, Xie A. Multi-predictor modeling for predicting early Parkinson’s disease and non-motor symptoms progression. Front Aging Neurosci 2022; 14:977985. [PMID: 36092799 PMCID: PMC9459236 DOI: 10.3389/fnagi.2022.977985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022] Open
Abstract
Background Identifying individuals with high-risk Parkinson’s disease (PD) at earlier stages is an urgent priority to delay disease onset and progression. In the present study, we aimed to develop and validate clinical risk models using non-motor predictors to distinguish between early PD and healthy individuals. In addition, we constructed prognostic models for predicting the progression of non-motor symptoms [cognitive impairment, Rapid-eye-movement sleep Behavior Disorder (RBD), and depression] in de novo PD patients at 5 years of follow-up. Methods We retrieved the data from the Parkinson’s Progression Markers Initiative (PPMI) database. After a backward variable selection approach to identify predictors, logistic regression analyses were applied for diagnosis model construction, and cox proportional-hazards models were used to predict non-motor symptom progression. The predictive models were internally validated by correcting measures of predictive performance for “optimism” or overfitting with the bootstrap resampling approach. Results For constructing diagnostic models, the final model reached a high accuracy with an area under the curve (AUC) of 0.93 (95% CI: 0.91–0.96), which included eight variables (age, gender, family history, University of Pennsylvania Smell Inventory Test score, Montreal Cognitive Assessment score, RBD Screening Questionnaire score, levels of cerebrospinal fluid α-synuclein, and SNCA rs356181 polymorphism). For the construction of prognostic models, our results showed that the AUC of the three prognostic models improved slightly with increasing follow-up time. The overall AUCs fluctuated around 0.70. The model validation established good discrimination and calibration for predicting PD onset and progression of non-motor symptoms. Conclusion The findings of our study facilitate predicting the individual risk at an early stage based on the predictors derived from these models. These predictive models provide relatively reliable information to prevent PD onset and progression. However, future validation analysis is still needed to clarify these findings and provide more insight into the predictive models over more extended periods of disease progression in more diverse samples.
Collapse
|
15
|
Yang NN, Sang SS, Peng T, lu H. SNCA rs3910105 Is Associated With Development of Rapid Eye Movement Sleep Behavior Disorder in Parkinson’s Disease. Front Neurosci 2022; 16:832550. [PMID: 35310107 PMCID: PMC8927062 DOI: 10.3389/fnins.2022.832550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background and Purpose Rapid eye movement (REM) Rapid eye movement sleep behavior disorder (RBD) is a common non-motor symptom of PD. However, the association between the SNCA rs3910105 genotype and RBD in Parkinson’s disease (PD) remains unclear. Methods This study used Parkinson’s Progression Markers Initiative (PPMI) data and included 270 patients with newly diagnosed PD without RBD who were divided into SNCA rs3910105 C carriers (CC+CT; n = 187) and TT carriers (n = 83). They were followed up for 5 years to identify the development of RBD. To investigate the influence of cerebrospinal fluid (CSF) alpha-synuclein (α-syn) and β-amyloid 1–42 (Aβ42) in the association between rs3910105 and RBD, the patients were additionally classified into “high-level” and “low-level” groups using cutoff values for CSF α-syn and Aβ42 levels. Results At baseline, the rs3910105 C allele group had lower CSF α-syn and Aβ42 levels than the TT group. During the 5.0-year follow-up, the rs3910105 C allele group had a higher incidence of RBD than the TT group. In the subgroup analyses, the effect of the rs3910105 C allele was not found in the “low-level” group. However, in the “high-level” group, the rs3910105 C allele independently increased the risk of RBD. Conclusion The SNCA rs3910105 C allele might be a novel genetic risk factor for RBD development in PD, α-syn pathways might have a role in this association and more basic research would be needed to elucidate the mechanism in the future.
Collapse
Affiliation(s)
- Nan-nan Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Nan-nan Yang,
| | - Shu-shan Sang
- Department of Otolaryngology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tao Peng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hong lu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Hong lu,
| |
Collapse
|
16
|
Pal G, Mangone G, Hill EJ, Ouyang B, Liu Y, Lythe V, Ehrlich D, Saunders-Pullman R, Shanker V, Bressman S, Alcalay RN, Garcia P, Marder KS, Aasly J, Mouradian MM, Link S, Rosenbaum M, Anderson S, Bernard B, Wilson R, Stebbins G, Nichols WC, Welter ML, Sani S, Afshari M, Verhagen L, de Bie RM, Foltynie T, Hall D, Corvol JC, Goetz CG. Parkinson Disease and Subthalamic Nucleus Deep Brain Stimulation: Cognitive Effects in GBA Mutation Carriers. Ann Neurol 2022; 91:424-435. [PMID: 34984729 PMCID: PMC8857042 DOI: 10.1002/ana.26302] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/01/2022] [Accepted: 01/03/2022] [Indexed: 12/16/2022]
Abstract
OBJECTIVE This study was undertaken to compare the rate of change in cognition between glucocerebrosidase (GBA) mutation carriers and noncarriers with and without subthalamic nucleus deep brain stimulation (STN-DBS) in Parkinson disease. METHODS Clinical and genetic data from 12 datasets were examined. Global cognition was assessed using the Mattis Dementia Rating Scale (MDRS). Subjects were examined for mutations in GBA and categorized as GBA carriers with or without DBS (GBA+DBS+, GBA+DBS-), and noncarriers with or without DBS (GBA-DBS+, GBA-DBS-). GBA mutation carriers were subcategorized according to mutation severity (risk variant, mild, severe). Linear mixed modeling was used to compare rate of change in MDRS scores over time among the groups according to GBA and DBS status and then according to GBA severity and DBS status. RESULTS Data were available for 366 subjects (58 GBA+DBS+, 82 GBA+DBS-, 98 GBA-DBS+, and 128 GBA-DBS- subjects), who were longitudinally followed (range = 36-60 months after surgery). Using the MDRS, GBA+DBS+ subjects declined on average 2.02 points/yr more than GBA-DBS- subjects (95% confidence interval [CI] = -2.35 to -1.69), 1.71 points/yr more than GBA+DBS- subjects (95% CI = -2.14 to -1.28), and 1.49 points/yr more than GBA-DBS+ subjects (95% CI = -1.80 to -1.18). INTERPRETATION Although not randomized, this composite analysis suggests that the combined effects of GBA mutations and STN-DBS negatively impact cognition. We advise that DBS candidates be screened for GBA mutations as part of the presurgical decision-making process. We advise that GBA mutation carriers be counseled regarding potential risks associated with STN-DBS so that alternative options may be considered. ANN NEUROL 2022;91:424-435.
Collapse
Affiliation(s)
- Gian Pal
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Graziella Mangone
- Sorbonne Université, Assistance Publique Hôpitaux de Paris, Inserm, CNRS, Institut du Cerveau – Paris Brain Institute – ICM, Pitié-Salpêtrière Hospital, Department of Neurology, Centre d’Investigation Clinique Neurosciences, Paris, France
| | - Emily J. Hill
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Bichun Ouyang
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Yuanqing Liu
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Vanessa Lythe
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology, London, UK
| | - Debra Ehrlich
- Parkinson’s Disease Clinic, Office of the Clinical Director, NIH/NINDS, Bethesda, MD, USA
| | - Rachel Saunders-Pullman
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Vicki Shanker
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Susan Bressman
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Roy N. Alcalay
- Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Priscilla Garcia
- Department of Neurology, New York Medical College, Valhalla, NY, USA
| | - Karen S. Marder
- Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Jan Aasly
- Department of Neurology, St. Olavs Hospital and Department of Neuroscience, Norwegian University of Science and Technology, Trondheim, 7030, Norway
| | - M. Maral Mouradian
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| | - Samantha Link
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Marc Rosenbaum
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Sharlet Anderson
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Bryan Bernard
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Robert Wilson
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Glenn Stebbins
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - William C. Nichols
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Marie-Laure Welter
- Sorbonne Université, Assistance Publique Hôpitaux de Paris, Inserm, CNRS, Institut du Cerveau – Paris Brain Institute – ICM, Pitié-Salpêtrière Hospital, Department of Neurology, Centre d’Investigation Clinique Neurosciences, Paris, France
- Normandie Univ, CHU Rouen, Department of Neurophysiology, Rouen, France
| | - Sepehr Sani
- Department of Neurosurgery, Rush University Medical Center, Chicago, IL, USA
| | - Mitra Afshari
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Leo Verhagen
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Rob M.A. de Bie
- Amsterdam University Medical Centers, University of Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Tom Foltynie
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology, London, UK
| | - Deborah Hall
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Jean-Christophe Corvol
- Sorbonne Université, Assistance Publique Hôpitaux de Paris, Inserm, CNRS, Institut du Cerveau – Paris Brain Institute – ICM, Pitié-Salpêtrière Hospital, Department of Neurology, Centre d’Investigation Clinique Neurosciences, Paris, France
| | - Christopher G. Goetz
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
17
|
Pihlstrøm L, Fan CC, Frei O, Blauwendraat C, Bandres-Ciga S, Dale AM, Seibert TM, Andreassen OA, Dale AM, Seibert TM, Andreassen OA. Genetic Stratification of Age-Dependent Parkinson's Disease Risk by Polygenic Hazard Score. Mov Disord 2022; 37:62-69. [PMID: 34612543 PMCID: PMC9843635 DOI: 10.1002/mds.28808] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 09/08/2021] [Accepted: 09/13/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a highly age-related disorder, where common genetic risk variants affect both disease risk and age at onset. A statistical approach that integrates these effects across all common variants may be clinically useful for individual risk stratification. A polygenic hazard score methodology, leveraging a time-to-event framework, has recently been successfully applied in other age-related disorders. OBJECTIVES We aimed to develop and validate a polygenic hazard score model in sporadic PD. METHODS Using a Cox regression framework, we modeled the polygenic hazard score in a training data set of 11,693 PD patients and 9841 controls. The score was then validated in an independent test data set of 5112 PD patients and 5372 controls and a small single-study sample of 360 patients and 160 controls. RESULTS A polygenic hazard score predicts the onset of PD with a hazard ratio of 3.78 (95% confidence interval 3.49-4.10) when comparing the highest to the lowest risk decile. Combined with epidemiological data on incidence rate, we apply the score to estimate genetically stratified instantaneous PD risk across age groups. CONCLUSIONS We demonstrate the feasibility of a polygenic hazard approach in PD, integrating the genetic effects on disease risk and age at onset in a single model. In combination with other predictive biomarkers, the approach may hold promise for risk stratification in future clinical trials of disease-modifying therapies, which aim at postponing the onset of PD. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, Oslo, Norway,Corresponding authors at: Department of Neurology, Oslo University Hospital, PO Box 4950 Nydalen, 0424 Oslo, Norway. , NORMENT Centre, Oslo University Hospital, Ullevål, PO Box 4956 Nydalen, 0424 Oslo, Norway.
| | - Chun Chieh Fan
- Department of Cognitive Science, University of California San Diego, La Jolla, CA, USA,Center for Multimodal Imaging and Genetics, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Oleksandr Frei
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Center for Bioinformatics, Department of Informatics, University of Oslo
| | - Cornelis Blauwendraat
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MY, USA
| | - Sara Bandres-Ciga
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MY, USA
| | | | - Anders M. Dale
- Department of Cognitive Science, University of California San Diego, La Jolla, CA, USA,Department of Radiology, University of California San Diego, La Jolla, CA, USA,Department of Neurosciences, University of California, San Diego, La Jolla, California, United States of America
| | - Tyler M. Seibert
- Department of Radiology, University of California San Diego, La Jolla, CA, USA,Department of Radiation Medicine and Applied Sciences, University of California, San Diego, La Jolla, CA, USA,Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Ole A. Andreassen
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Anders M Dale
- Department of Cognitive Science, University of California San Diego, La Jolla, California, USA.,Center for Multimodal Imaging and Genetics, School of Medicine, University of California San Diego, La Jolla, California, USA.,NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Radiology, University of California San Diego, La Jolla, California, USA.,Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Tyler M Seibert
- Center for Multimodal Imaging and Genetics, School of Medicine, University of California San Diego, La Jolla, California, USA.,NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, USA.,Department of Radiology, University of California San Diego, La Jolla, California, USA.,Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Ole A Andreassen
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
18
|
Pak K, Lee MJ, Kim K, Kim IJ. No effect of Parkinson's disease-polygenic load on striatal density of dopaminergic neuron in healthy subjects. Ann Nucl Med 2021; 35:1187-1192. [PMID: 34287783 DOI: 10.1007/s12149-021-01657-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 07/16/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE There has been increasing evidence to support the role of genetic factors in Parkinson's disease (PD). 123I-FP-CIT single-photon emission computed tomography (SPECT) enables in vivo visualization of the striatal density of dopaminergic neuron. METHODS We investigated the association between PD-associated polygenic load and striatal density of dopaminergic neuron in healthy subjects. Data used in the preparation of this article were obtained from Parkinson's Progression Markers Initiative database. 123I-FP-CIT SPECT was performed for all subjects. Specific binding ratios (SBRs) were calculated from the ventral striatum, caudate nucleus, and putamen with reference to cerebellum. Singe nucleotide polymorphism (SNP) genotyping from the PPMI database was adopted in calculating genetic risk score (GRS). GRS was defined as the sum of the number of risk alleles weighted by log odds ratios for PD. We calculated three GRSs using three different sets of SNPs. RESULT A total of 151 subjects were included in this study (101 males, 50 females). GRS1, GRS2 and GRS3 were significantly different with the highest scores of GRS1. Multiple regression was done to investigate whether striatal SBRs are influenced by GRSs after adjusting for age and sex. However, none of GRSs were associated with SBRs of the ventral striatum, caudate nucleus and putamen. CONCLUSION PD risk SNPs weighted by odds ratio for PD were not associated with SBRs measured from SPECT in healthy subjects. Therefore, there is no effect of PD-associated polygenic load on striatal density of dopaminergic neuron in healthy subjects.
Collapse
Affiliation(s)
- Kyoungjune Pak
- Department of Nuclear Medicine and Biomedical Research Institute, Pusan National University Hospital, 179 Gudeok-ro, Seo-gu, Busan, 49241, Republic of Korea.
| | - Myung Jun Lee
- Department of Neurology and Biomedical Research Institute, Pusan National University Hospital, 179 Gudeok-ro, Seo-gu, Busan, 49241, Republic of Korea.
| | - Keunyoung Kim
- Department of Nuclear Medicine and Biomedical Research Institute, Pusan National University Hospital, 179 Gudeok-ro, Seo-gu, Busan, 49241, Republic of Korea
| | - In Joo Kim
- Department of Nuclear Medicine and Biomedical Research Institute, Pusan National University Hospital, 179 Gudeok-ro, Seo-gu, Busan, 49241, Republic of Korea.
| |
Collapse
|
19
|
Ye G, Li Y, Zhou L, Zhang Y, Zhu L, Zhao A, Kang W, Liu J. Predictors of Conversion to α-Synucleinopathy Diseases in Idiopathic Rapid Eye Movement Sleep Behavior Disorder. JOURNAL OF PARKINSONS DISEASE 2021; 10:1443-1455. [PMID: 32986685 DOI: 10.3233/jpd-202243] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Idiopathic rapid eye movement sleep behavior disorder (iRBD) often precedes the development of α-synucleinopathy diseases. OBJECTIVE We aimed to assess the predictive value of clinical variables and biomarkers for the early development of α-synucleinopathy diseases in subjects with iRBD. METHODS 56 patients with RBD Screening Questionnaire (RBDSQ) scores ≥5 at baseline and subsequent visit were enrolled as probable iRBD from the Parkinson's Progression Markers Initiative (PPMI) database. Baseline clinical data and biomarkers were analyzed. The endpoint was defined as disease progression to α-synucleinopathy diseases. Cox proportional hazard and Kaplan-Meier analyses were used to evaluate the predictive values of the indicators. RESULTS During a mean follow-up duration of 5.1 years, 15 of 56 patients (26.8%) developed α-synucleinopathy diseases. Baseline clinical variables, including University of Pennsylvania Smell Identification Test (UPSIT, HR = 26.18, p = 0.004), 15-item Geriatric Depression Scale (GDS, HR = 14.26, p = 0.001), Montreal Cognitive Assessment (MoCA, HR = 3.56, p = 0.025), and Hopkins Verbal Learning Test Total recall (HVLT-TR, HR = 3.70, p = 0.014); genotype status of TMEM175 (HR = 3.74, p = 0.017), SCN3A (HR = 5.81, p = 0.022) and NUCKS1 (HR = 0.342, p = 0.049); ratio of phosphorylated tau to total tau (p-tau/t-tau, HR = 8.36, p = 0.001) in cerebrospinal fluid; and gray matter atrophy in inferior frontal gyrus (IFG, HR = 15.49, p = 0.001) were associated with phenoconversion to α-synucleinopathy diseases. A model combined the three independent variables (UPSIT, TMEM175 and gray matter atrophy in IFG) exhibited significantly improved predictive performance. CONCLUSION For patients with iRBD, progression to α-synucleinopathy diseases can be predicted with good accuracy using a model combining clinical variables and biomarkers, which could form a basis for future disease prevention.
Collapse
Affiliation(s)
- Guanyu Ye
- Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Neurology & Institute of Neurology, Ruijin Hospital/Lu Wan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyuan Li
- Department of Neurology & Institute of Neurology, Ruijin Hospital/Lu Wan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liche Zhou
- Department of Neurology & Institute of Neurology, Ruijin Hospital/Lu Wan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yichi Zhang
- Department of Neurology & Institute of Neurology, Ruijin Hospital/Lu Wan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Zhu
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aonan Zhao
- Department of Neurology & Institute of Neurology, Ruijin Hospital/Lu Wan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenyan Kang
- Department of Neurology, Ruijin Hospital/North Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Liu
- Department of Neurology & Institute of Neurology, Ruijin Hospital/Lu Wan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Ruijin Hospital/Lu Wan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Iwaki H, Leonard HL, Makarious MB, Bookman M, Landin B, Vismer D, Casey B, Gibbs JR, Hernandez DG, Blauwendraat C, Vitale D, Song Y, Kumar D, Dalgard CL, Sadeghi M, Dong X, Misquitta L, Scholz SW, Scherzer CR, Nalls MA, Biswas S, Singleton AB. Accelerating Medicines Partnership: Parkinson's Disease. Genetic Resource. Mov Disord 2021; 36:1795-1804. [PMID: 33960523 PMCID: PMC8453903 DOI: 10.1002/mds.28549] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/20/2021] [Accepted: 02/11/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Whole-genome sequencing data are available from several large studies across a variety of diseases and traits. However, massive storage and computation resources are required to use these data, and to achieve sufficient power for discoveries, harmonization of multiple cohorts is critical. OBJECTIVES The Accelerating Medicines Partnership Parkinson's Disease program has developed a research platform for Parkinson's disease (PD) that integrates the storage and analysis of whole-genome sequencing data, RNA expression data, and clinical data, harmonized across multiple cohort studies. METHODS The version 1 release contains whole-genome sequencing data derived from 3941 participants from 4 cohorts. Samples underwent joint genotyping by the TOPMed Freeze 9 Variant Calling Pipeline. We performed descriptive analyses of these whole-genome sequencing data using the Accelerating Medicines Partnership Parkinson's Disease platform. RESULTS The clinical diagnosis of participants in version 1 release includes 2005 idiopathic PD patients, 963 healthy controls, 64 prodromal subjects, 62 clinically diagnosed PD subjects without evidence of dopamine deficit, and 705 participants of genetically enriched cohorts carrying PD risk-associated GBA variants or LRRK2 variants, of whom 304 were affected. We did not observe significant enrichment of pathogenic variants in the idiopathic PD group, but the polygenic risk score was higher in PD both in nongenetically enriched cohorts and genetically enriched cohorts. The population analysis showed a correlation between genetically enriched cohorts and Ashkenazi Jewish ancestry. CONCLUSIONS We describe the genetic component of the Accelerating Medicines Partnership Parkinson's Disease platform, a solution to democratize data access and analysis for the PD research community. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society. This article is a U.S. Government work and is in the public domain in the USA.
Collapse
Affiliation(s)
- Hirotaka Iwaki
- Data Tecnica InternationalGlen EchoMarylandUSA
- Center for Alzheimer's and Related DementiasNational Institute on AgingBethesdaMarylandUSA
- Laboratory of NeurogeneticsNational Institute on AgingBethesdaMarylandUSA
| | - Hampton L. Leonard
- Data Tecnica InternationalGlen EchoMarylandUSA
- Center for Alzheimer's and Related DementiasNational Institute on AgingBethesdaMarylandUSA
- Laboratory of NeurogeneticsNational Institute on AgingBethesdaMarylandUSA
| | - Mary B. Makarious
- Laboratory of NeurogeneticsNational Institute on AgingBethesdaMarylandUSA
| | | | | | | | - Bradford Casey
- The Michael J. Fox Foundation for Parkinson's ResearchNew YorkNew YorkUSA
| | - J. Raphael Gibbs
- Laboratory of NeurogeneticsNational Institute on AgingBethesdaMarylandUSA
| | - Dena G. Hernandez
- Laboratory of NeurogeneticsNational Institute on AgingBethesdaMarylandUSA
| | | | - Daniel Vitale
- Data Tecnica InternationalGlen EchoMarylandUSA
- Center for Alzheimer's and Related DementiasNational Institute on AgingBethesdaMarylandUSA
- Laboratory of NeurogeneticsNational Institute on AgingBethesdaMarylandUSA
| | - Yeajin Song
- Data Tecnica InternationalGlen EchoMarylandUSA
- Center for Alzheimer's and Related DementiasNational Institute on AgingBethesdaMarylandUSA
- Laboratory of NeurogeneticsNational Institute on AgingBethesdaMarylandUSA
| | | | - Clifton L. Dalgard
- Department of Anatomy, Physiology & GeneticsUniformed Services University of the Health SciencesBethesdaMarylandUSA
- The American Genome CenterUniformed Services University of the Health SciencesBethesdaMarylandUSA
| | - Mahdiar Sadeghi
- SanofiFraminghamMassachusettsUSA
- Northeastern UniversityBostonMassachusettsUSA
| | - Xianjun Dong
- Harvard Medical SchoolBrigham and Women's HospitalBostonMassachusettsUSA
| | | | - Sonja W. Scholz
- National Institute of Neurological Disorders and StrokeBethesdaMarylandUSA
- Department of NeurologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | | | - Mike A. Nalls
- Data Tecnica InternationalGlen EchoMarylandUSA
- Center for Alzheimer's and Related DementiasNational Institute on AgingBethesdaMarylandUSA
- Laboratory of NeurogeneticsNational Institute on AgingBethesdaMarylandUSA
| | | | - Andrew B. Singleton
- Center for Alzheimer's and Related DementiasNational Institute on AgingBethesdaMarylandUSA
- Laboratory of NeurogeneticsNational Institute on AgingBethesdaMarylandUSA
| | | | | | | |
Collapse
|
21
|
Craig DW, Hutchins E, Violich I, Alsop E, Gibbs JR, Levy S, Robison M, Prasad N, Foroud T, Crawford KL, Toga AW, Whitsett TG, Kim S, Casey B, Reimer A, Hutten SJ, Frasier M, Kern F, Fehlman T, Keller A, Cookson MR, Van Keuren-Jensen K. RNA sequencing of whole blood reveals early alterations in immune cells and gene expression in Parkinson's disease. NATURE AGING 2021; 1:734-747. [PMID: 37117765 DOI: 10.1038/s43587-021-00088-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 06/21/2021] [Indexed: 04/30/2023]
Abstract
Changes in the blood-based RNA transcriptome have the potential to inform biomarkers of Parkinson's disease (PD) progression. Here we sequenced a discovery set of whole-blood RNA species in 4,871 longitudinally collected samples from 1,570 clinically phenotyped individuals from the Parkinson's Progression Marker Initiative (PPMI) cohort. Samples were sequenced to an average of 100 million read pairs to create a high-quality transcriptome. Participants with PD in the PPMI had significantly altered RNA expression (>2,000 differentially expressed genes), including an early and persistent increase in neutrophil gene expression, with a concomitant decrease in lymphocyte cell counts. This was validated in a cohort from the Parkinson's Disease Biomarkers Program (PDBP) consisting of 1,599 participants and by alterations in immune cell subtypes. This publicly available transcriptomic dataset, coupled with available detailed clinical data, provides new insights into PD biological processes impacting whole blood and new paths for developing diagnostic and prognostic PD biomarkers.
Collapse
Affiliation(s)
- David W Craig
- Institute of Translational Genomics, University of Southern California, Los Angeles, CA, USA
| | - Elizabeth Hutchins
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Ivo Violich
- Institute of Translational Genomics, University of Southern California, Los Angeles, CA, USA
| | - Eric Alsop
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - J Raphael Gibbs
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Shawn Levy
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Madison Robison
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Nripesh Prasad
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | | | - Karen L Crawford
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Arthur W Toga
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Timothy G Whitsett
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Seungchan Kim
- Center for Computational Systems Biology, Department of Electrical and Computer Engineering, Roy G. Perry College of Engineering, Prairie View A&M University, Prairie View, TX, USA
| | - Bradford Casey
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Alyssa Reimer
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Samantha J Hutten
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Mark Frasier
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Fabian Kern
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Tobias Fehlman
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Mark R Cookson
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
22
|
Chen F, Li Y, Ye G, Zhou L, Bian X, Liu J. Development and Validation of a Prognostic Model for Cognitive Impairment in Parkinson's Disease With REM Sleep Behavior Disorder. Front Aging Neurosci 2021; 13:703158. [PMID: 34322014 PMCID: PMC8311737 DOI: 10.3389/fnagi.2021.703158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/21/2021] [Indexed: 12/29/2022] Open
Abstract
The presentation and progression of Parkinson’s disease (PD) are not uniform, but the presence of rapid eye movement sleep behavior disorder (RBD) in PD patients may indicate a worse prognosis than isolated PD. Increasing evidence suggests that patients with comorbid PD and RBD (PD-RBD) are more likely to develop cognitive impairment (CI) than those with isolated PD; however, the predictors of CI in PD-RBD patients are not well understood. This study aimed to develop a prognostic model for predicting mild cognitive impairment (MCI) in PD-RBD patients. The data of PD-RBD patients were extracted from the Parkinson’s Progression Markers Initiative study (PPMI), and the sample was randomly divided into a training set (n = 96) and a validation set (n = 24). PD-MCI as defined by the level II Movement Disorder Society (MDS) diagnostic criteria was the outcome of interest. The demographic features, clinical assessments, dopamine transporter (DAT) imaging data, cerebrospinal fluid (CSF) analyses and genetic data of PD patients were considered candidate predictors. We found that performance on the University of Pennsylvania Smell Identification Test (UPSIT), the mean signal and asymmetry index of the putamen on DAT imaging, p-tau/α-syn and p-tau in CSF, and rs55785911 genotype were predictors of PD-MCI in PD-RBD patients. A C-index of 0.81 was obtained with this model, and a C-index of 0.73 was obtained in the validation set. Favorable results of calibrations and decision curve analysis demonstrated the efficacy and feasibility of this model. In conclusion, we developed a prognostic model for predicting MCI in PD-RBD patients; the model displayed good discrimination and calibration and may be a convenient tool for clinical application. Larger samples and external validation sets are needed to validate this model.
Collapse
Affiliation(s)
- Fangzheng Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyuan Li
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanyu Ye
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liche Zhou
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolan Bian
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
23
|
Le Guen Y, Napolioni V, Belloy ME, Yu E, Krohn L, Ruskey JA, Gan-Or Z, Kennedy G, Eger SJ, Greicius MD. Common X-Chromosome Variants Are Associated with Parkinson Disease Risk. Ann Neurol 2021; 90:22-34. [PMID: 33583074 PMCID: PMC8601399 DOI: 10.1002/ana.26051] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 02/12/2021] [Accepted: 02/12/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The objective of this study was to identify genetic variants on the X-chromosome associated with Parkinson disease (PD) risk. METHODS We performed an X-chromosome-wide association study (XWAS) of PD risk by meta-analyzing results from sex-stratified analyses. To avoid spurious associations, we designed a specific harmonization pipeline for the X-chromosome and focused on a European ancestry sample. We included 11,142 cases, 280,164 controls, and 5,379 proxy cases, based on parental history of PD. Additionally, we tested the association of significant variants with (1) PD risk in an independent replication with 1,561 cases and 2,465 controls and (2) putamen volume in 33,360 individuals from the UK Biobank. RESULTS In the discovery meta-analysis, we identified rs7066890 (odds ratio [OR] = 1.10, 95% confidence interval [CI] = 1.06-1.14, p = 2.2 × 10-9 ), intron of GPM6B, and rs28602900 (OR = 1.10, 95% CI = 1.07-1.14, p = 1.6 × 10-8 ) in a high gene density region including RPL10, ATP6A1, FAM50A, and PLXNA3. The rs28602900 association with PD was replicated (OR = 1.16, 95% CI = 1.03-1.30, p = 0.016) and shown to colocalize with a significant expression quantitative locus (eQTL) regulating RPL10 expression in the putamen and other brain tissues in the Genotype-Tissue Expression Project. Additionally, the rs28602900 locus was found to be associated with reduced brain putamen volume. No results reached genome-wide significance in the sex-stratified analyses. INTERPRETATION We report the first XWAS of PD and identify 2 genome-wide significant loci. The rs28602900 association was replicated in an independent PD dataset and showed concordant effects in its association with putamen volume. Critically, rs26802900 is a significant eQTL of RPL10. These results support a role for ribosomal proteins in PD pathogenesis and show that the X-chromosome contributes to PD genetic risk. ANN NEUROL 2021;90:22-34.
Collapse
Affiliation(s)
- Yann Le Guen
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Valerio Napolioni
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Michael E Belloy
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Eric Yu
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Lynne Krohn
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Jennifer A Ruskey
- Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Ziv Gan-Or
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Gabriel Kennedy
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Sarah J Eger
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| |
Collapse
|
24
|
Ortega RA, Wang C, Raymond D, Bryant N, Scherzer CR, Thaler A, Alcalay RN, West AB, Mirelman A, Kuras Y, Marder KS, Giladi N, Ozelius LJ, Bressman SB, Saunders-Pullman R. Association of Dual LRRK2 G2019S and GBA Variations With Parkinson Disease Progression. JAMA Netw Open 2021; 4:e215845. [PMID: 33881531 PMCID: PMC8060834 DOI: 10.1001/jamanetworkopen.2021.5845] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Importance Despite a hypothesis that harboring a leucine-rich repeat kinase 2(LRRK2) G2019S variation and a glucocerebrosidase (GBA) variant would have a combined deleterious association with disease pathogenesis, milder clinical phenotypes have been reported in dual LRRK2 and GBA variations Parkinson disease (PD) than in GBA variation PD alone. Objective To evaluate the association of LRRK2 G2019S and GBA variants with longitudinal cognitive and motor decline in PD. Design, Setting, and Participants This longitudinal cohort study of continuous measures in LRRK2 PD, GBA PD, LRRK2/GBA PD, and wild-type idiopathic PD used pooled annual visit data ranging from 2004 to 2019 from the Mount Sinai Beth Israel, Parkinson Disease Biomarker Program, Harvard Biomarkers Study, Ashkenazi Jewish-LRRK2-Consortium, Parkinson Progression Marker Initiative, and SPOT-PD studies. Patients who were screened for GBA and LRRK2 variations and completed either a motor or cognitive assessment were included. Data were analyzed from May to July 2020. Main Outcomes and Measures The associations of LRRK2 G2019S and GBA genotypes on the rate of decline in Montreal Cognitive Assessment (MoCA) and Movement Disorders Society-Unified Parkinson Disease Rating Scale-Part III scores were examined using linear mixed effects models with PD duration as the time scale. Results Among 1193 individuals with PD (mean [SD] age, 66.6 [9.9] years; 490 [41.2%] women), 128 (10.7%) had GBA PD, 155 (13.0%) had LRRK2 PD, 21 (1.8%) had LRRK2/GBA PD, and 889 (74.5%) had idiopathic PD. Patients with GBA PD had faster decline in MoCA than those with LRRK2/GBA PD (B [SE], -0.31 [0.09] points/y; P < .001), LRRK2 PD (B [SE], -0.33 [0.09] points/y; P < .001), or idiopathic PD (B [SE], -0.23 [0.08] points/y; P = .005). There was a LRRK2 G2019S × GBA interaction in MoCA decline (B [SE], 0.22 [0.11] points/y; P = .04), but not after excluding severe GBA variations (B [SE], 0.12 [0.11] points/y; P = .28). Patients with GBA PD had significantly worse motor progression compared with those with idiopathic PD (B [SE], 0.49 [0.22] points/y; P = .03) or LRRK2 PD (B [SE], 0.77 [0.26] points/y; P = .004). Conclusions and Relevance These findings suggest that longitudinal cognitive decline in patients with GBA PD was more severe than in those with LRRK2/GBA PD, which more closely resembled LRRK2 PD. This further supports the notion of a dominant association of LRRK2 on GBA in individuals who carry both and raises the possibility of an LRRK2 × GBA interaction. However, the biological basis of a dominant association or interaction is not clear and is apparently contrary to basic investigations. Study of a larger cohort of individuals with severe GBA variation is warranted.
Collapse
Affiliation(s)
- Roberto A Ortega
- Department of Neurology, Mount Sinai Beth Israel, and Icahn School of Medicine, Mount Sinai, New York, New York
| | - Cuiling Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Yeshiva University, Bronx, New York
- Department of Neurology, Albert Einstein College of Medicine, Yeshiva University, Bronx, New York
| | - Deborah Raymond
- Department of Neurology, Mount Sinai Beth Israel, and Icahn School of Medicine, Mount Sinai, New York, New York
| | - Nicole Bryant
- Duke Center for Neurodegeneration and Neurotherapeutics, Duke University, Durham, North Carolina
| | - Clemens R Scherzer
- Center for Advanced Parkinson Research and Precision Neurology Program, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts
| | - Avner Thaler
- Laboratory for Early Markers of Neurodegeneration, Center for the Study of Movement, Cognition, and Mobility, Neurological Institute, Tel Aviv Medical Center, Sackler School of Medicine, Sagol School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Roy N Alcalay
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York
| | - Andrew B West
- Duke Center for Neurodegeneration and Neurotherapeutics, Duke University, Durham, North Carolina
| | - Anat Mirelman
- Laboratory for Early Markers of Neurodegeneration, Center for the Study of Movement, Cognition, and Mobility, Neurological Institute, Tel Aviv Medical Center, Sackler School of Medicine, Sagol School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Yuliya Kuras
- Center for Advanced Parkinson Research and Precision Neurology Program, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts
| | - Karen S Marder
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York
| | - Nir Giladi
- Laboratory for Early Markers of Neurodegeneration, Center for the Study of Movement, Cognition, and Mobility, Neurological Institute, Tel Aviv Medical Center, Sackler School of Medicine, Sagol School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Susan B Bressman
- Department of Neurology, Mount Sinai Beth Israel, and Icahn School of Medicine, Mount Sinai, New York, New York
| | - Rachel Saunders-Pullman
- Department of Neurology, Mount Sinai Beth Israel, and Icahn School of Medicine, Mount Sinai, New York, New York
| |
Collapse
|
25
|
Kim R, Park S, Yoo D, Jun JS, Jeon B. Association of Physical Activity and APOE Genotype With Longitudinal Cognitive Change in Early Parkinson Disease. Neurology 2021; 96:e2429-e2437. [PMID: 33790041 DOI: 10.1212/wnl.0000000000011852] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 02/10/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine whether greater physical activity could modify the negative association of APOE ε4 with longitudinal cognitive changes in early Parkinson disease (PD) and to uncover the disease-specific mechanism for explaining such benefits of physical activity. METHODS We used data from the Parkinson's Progression Markers Initiative cohort. Because self-reported physical activity, measured by the Physical Activity Scale of the Elderly, was initiated at 2 years after enrollment, this longitudinal analysis was based on assessments performed at years 2, 3, and 4. Cognitive function was measured annually with the Montreal Cognitive Assessment (MoCA). Dopamine transporter (DAT) imaging was performed at years 2 and 4. We assessed the interactive associations between physical activity and the APOE ε4 allele on the longitudinal changes in MoCA scores and striatal DAT activities. RESULTS A total of 173 patients with early PD (age 63.3 ± 10.0 years, 27% APOE ε4 carriers) were included. The APOE ε4 allele showed a steeper rate of cognitive decline than the non-APOE ε4 allele (estimate -1.33, 95% confidence interval [CI] -2.12 to -0.47, p = 0.002). However, there was a significant interaction between physical activity and APOE ε4 such that higher physical activity was related to slower APOE ε4-related cognitive decline (estimate 0.007, 95% CI 0.003-0.011, p = 0.001). No significant interaction was found between physical activity and the APOE ε4 allele regarding the change in striatal DAT activities. CONCLUSION Increased physical activity attenuated APOE ε4-related vulnerability to early cognitive decline in patients with PD. This protective effect did not appear to be mediated by striatal dopaminergic function. TRIAL REGISTRATION INFORMATION ClinicalTrials.gov Identifier: NCT01141023. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that increased physical activity was associated with decreased APOE ε4-related early cognitive decline in patients with PD.
Collapse
Affiliation(s)
- Ryul Kim
- From the Department of Neurology (R.K.), Inha University Hospital, Incheon; Department of Neurology (S.P.), School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu; Department of Neurology (D.Y.), Kyung Hee University Hospital; Department of Neurology (J.-S.J.), Kangnam Sacred Heart Hospital, Hallym University College of Medicine; and Department of Neurology (B.J.), College of Medicine, Seoul National University Hospital, Korea
| | - Sangmin Park
- From the Department of Neurology (R.K.), Inha University Hospital, Incheon; Department of Neurology (S.P.), School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu; Department of Neurology (D.Y.), Kyung Hee University Hospital; Department of Neurology (J.-S.J.), Kangnam Sacred Heart Hospital, Hallym University College of Medicine; and Department of Neurology (B.J.), College of Medicine, Seoul National University Hospital, Korea
| | - Dallah Yoo
- From the Department of Neurology (R.K.), Inha University Hospital, Incheon; Department of Neurology (S.P.), School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu; Department of Neurology (D.Y.), Kyung Hee University Hospital; Department of Neurology (J.-S.J.), Kangnam Sacred Heart Hospital, Hallym University College of Medicine; and Department of Neurology (B.J.), College of Medicine, Seoul National University Hospital, Korea
| | - Jin-Sun Jun
- From the Department of Neurology (R.K.), Inha University Hospital, Incheon; Department of Neurology (S.P.), School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu; Department of Neurology (D.Y.), Kyung Hee University Hospital; Department of Neurology (J.-S.J.), Kangnam Sacred Heart Hospital, Hallym University College of Medicine; and Department of Neurology (B.J.), College of Medicine, Seoul National University Hospital, Korea.
| | - Beomseok Jeon
- From the Department of Neurology (R.K.), Inha University Hospital, Incheon; Department of Neurology (S.P.), School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu; Department of Neurology (D.Y.), Kyung Hee University Hospital; Department of Neurology (J.-S.J.), Kangnam Sacred Heart Hospital, Hallym University College of Medicine; and Department of Neurology (B.J.), College of Medicine, Seoul National University Hospital, Korea
| |
Collapse
|
26
|
Kim R, Park S, Yoo D, Jun JS, Jeon B. Impact of the apolipoprotein E ε4 allele on early Parkinson's disease progression. Parkinsonism Relat Disord 2021; 83:66-70. [PMID: 33484977 DOI: 10.1016/j.parkreldis.2021.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/11/2020] [Accepted: 01/05/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Emerging evidence shows that apolipoprotein E (APOE) ε4 exacerbates alpha-synuclein pathology. We aimed to investigate whether the APOE ε4 allele contributes to early Parkinson's disease (PD) progression. METHODS This cohort study included 361 early PD patients who were classified as APOE ε4 carriers (n = 90) and noncarriers (n = 271). The patients underwent yearly motor and nonmotor assessments covering neuropsychiatric, sleep-related, and autonomic symptoms over 5 years of follow-up. Dopamine transporter (DAT) imaging was conducted at baseline and the 1-, 2-, and 4-year follow-up visits. RESULTS The APOE ε4 carriers had steeper declines in the Montreal Cognitive Assessment score (p=0.005) and the semantic fluency test score (p=0.012) than the noncarriers. No significant between-group differences in the longitudinal changes in motor, other nonmotor, and DAT imaging variables were observed. CONCLUSIONS Our exploratory analyses show that only cognitive performance was negatively affected by the APOE ε4 allele in the progression of early PD. More specifically, this allele was associated with poorer performance in semantic verbal fluency among cognitive domains.
Collapse
Affiliation(s)
- Ryul Kim
- Department of Neurology, Inha University Hospital, Incheon, South Korea
| | - Sangmin Park
- Department of Neurology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | - Dallah Yoo
- Department of Neurology, Kyung Hee University Medical Center, Seoul, South Korea
| | - Jin-Sun Jun
- Department of Neurology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, South Korea.
| | - Beomseok Jeon
- Department of Neurology, College of Medicine, Seoul National University Hospital, Seoul, South Korea
| |
Collapse
|
27
|
Zhou X, Li YYT, Fu AKY, Ip NY. Polygenic Score Models for Alzheimer's Disease: From Research to Clinical Applications. Front Neurosci 2021; 15:650220. [PMID: 33854414 PMCID: PMC8039467 DOI: 10.3389/fnins.2021.650220] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/09/2021] [Indexed: 12/13/2022] Open
Abstract
The high prevalence of Alzheimer's disease (AD) among the elderly population and its lack of effective treatments make this disease a critical threat to human health. Recent epidemiological and genetics studies have revealed the polygenic nature of the disease, which is possibly explainable by a polygenic score model that considers multiple genetic risks. Here, we systemically review the rationale and methods used to construct polygenic score models for studying AD. We also discuss the associations of polygenic risk scores (PRSs) with clinical outcomes, brain imaging findings, and biochemical biomarkers from both the brain and peripheral system. Finally, we discuss the possibility of incorporating polygenic score models into research and clinical practice along with potential challenges.
Collapse
Affiliation(s)
- Xiaopu Zhou
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen, China
| | - Yolanda Y. T. Li
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Amy K. Y. Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen, China
| | - Nancy Y. Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen, China
- *Correspondence: Nancy Y. Ip,
| |
Collapse
|
28
|
Kim R, Park S, Yoo D, Ju Suh Y, Jun JS, Jeon B. Potential Sex-Specific Effects of Apolipoprotein E ɛ4 on Cognitive Decline in Early Parkinson's Disease. JOURNAL OF PARKINSONS DISEASE 2020; 11:497-505. [PMID: 33325396 DOI: 10.3233/jpd-202288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND/OBJECTIVE To compare the longitudinal trajectories of cognition according to the presence of the apolipoprotein E (APOE) ɛ4 allele in male and female Parkinson's disease (PD) patients. METHODS This study included a total of 361 patients with recently diagnosed de novo PD (mean age [standard deviation], 61.4 [9.8] years). The patients were classified into the following groups: APOEɛ4 + /M (n = 65), APOEɛ4-/M (n = 173), APOEɛ4 + /F (n = 25), and APOEɛ4-/F (n = 98). Cognitive decline was assessed annually over 5 years of follow-up using the Montreal Cognitive Assessment (MoCA). To assess the sex-specific impacts of the APOEɛ4 status on cognitive decline, we used generalized linear mixed effects (GLME) models separately for men, women, and the two sexes combined. RESULTS In the sex-stratified GLME models adjusted for covariates, the interaction results showed that the males with APOEɛ4 had a steeper rate of cognitive decline than those without APOEɛ4. In contrast, there was no significant interaction between APOEɛ4 and time on longitudinal MoCA performance in the females. The main effect of APOEɛ4 on the change in the MoCA score was not significant for either men or women. When the data from both men and women were used, the APOEɛ4 + /M group exhibited a steeper rate of cognitive decline than did the APOEɛ4 + /F and APOEɛ4-/F groups. These results were consistent with those of sensitivity analyses. CONCLUSION Sex may be considered when APOEɛ4-related vulnerability to early cognitive decline is evaluated in PD patients.
Collapse
Affiliation(s)
- Ryul Kim
- Department of Neurology, Inha University Hospital, Incheon, Korea
| | - Sangmin Park
- Department of Neurology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
| | - Dallah Yoo
- Department of Neurology, Kyung Hee University Medical Center, Seoul, Korea
| | - Young Ju Suh
- Department of Biomedical Sciences, Inha University College of Medicine, Incheon, Korea
| | - Jin-Sun Jun
- Department of Neurology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Beomseok Jeon
- Department of Neurology, College of Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
29
|
Apolipoprotein E ε4 genotype and risk of freezing of gait in Parkinson's disease. Parkinsonism Relat Disord 2020; 81:173-178. [DOI: 10.1016/j.parkreldis.2020.10.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/14/2020] [Accepted: 10/19/2020] [Indexed: 11/22/2022]
|
30
|
Botelho J, Mascarenhas P, Mendes JJ, Machado V. Network Protein Interaction in Parkinson's Disease and Periodontitis Interplay: A Preliminary Bioinformatic Analysis. Genes (Basel) 2020; 11:E1385. [PMID: 33238395 PMCID: PMC7700320 DOI: 10.3390/genes11111385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/19/2020] [Accepted: 11/21/2020] [Indexed: 12/19/2022] Open
Abstract
Recent studies supported a clinical association between Parkinson's disease (PD) and periodontitis. Hence, investigating possible interactions between proteins associated to these two conditions is of interest. In this study, we conducted a protein-protein network interaction analysis with recognized genes encoding proteins with variants strongly associated with PD and periodontitis. Genes of interest were collected via the Genome-Wide Association Studies (GWAS) database. Then, we conducted a protein interaction analysis, using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database, with a highest confidence cutoff of 0.9 and sensitivity analysis with confidence cutoff of 0.7. Our protein network casts a comprehensive analysis of potential protein-protein interactions between PD and periodontitis. This analysis may underpin valuable information for new candidate molecular mechanisms between PD and periodontitis and may serve new potential targets for research purposes. These results should be carefully interpreted, giving the limitations of this approach.
Collapse
Affiliation(s)
- João Botelho
- Periodontology Department, Clinical Research Unit (CRU), Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Universitário Egas Moniz (IUEM), 2829-511 Caparica, Portugal;
- Evidence-Based Hub, Clinical Research Unit (CRU), Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Universitário Egas Moniz (IUEM), 2829-511 Caparica, Portugal; (P.M.); (J.J.M.)
| | - Paulo Mascarenhas
- Evidence-Based Hub, Clinical Research Unit (CRU), Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Universitário Egas Moniz (IUEM), 2829-511 Caparica, Portugal; (P.M.); (J.J.M.)
- Center for Medical Genetics and Pediatric Nutrition Egas Moniz, Instituto Universitário Egas Moniz (IUEM), 2829-511 Caparica, Portugal
| | - José João Mendes
- Evidence-Based Hub, Clinical Research Unit (CRU), Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Universitário Egas Moniz (IUEM), 2829-511 Caparica, Portugal; (P.M.); (J.J.M.)
| | - Vanessa Machado
- Periodontology Department, Clinical Research Unit (CRU), Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Universitário Egas Moniz (IUEM), 2829-511 Caparica, Portugal;
- Evidence-Based Hub, Clinical Research Unit (CRU), Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Universitário Egas Moniz (IUEM), 2829-511 Caparica, Portugal; (P.M.); (J.J.M.)
| |
Collapse
|
31
|
Sampedro F, Marín-Lahoz J, Martínez-Horta S, Camacho V, Lopez-Mora DA, Pagonabarraga J, Kulisevsky J. Extrastriatal SPECT-DAT uptake correlates with clinical and biological features of de novo Parkinson's disease. Neurobiol Aging 2020; 97:120-128. [PMID: 33212336 DOI: 10.1016/j.neurobiolaging.2020.10.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 10/09/2020] [Accepted: 10/17/2020] [Indexed: 10/23/2022]
Abstract
Striatal dopamine transporter (DAT) uptake assessment through I123-Ioflupane Single-Pphoton Emission Computed Tomography (SPECT) provides valuable information about the dopaminergic denervation occurring in Parkinson's disease (PD). However, little is known about the clinical or biological relevance of extrastriatal DAT uptake in PD. Here, from the Parkinson's Progression Markers Initiative, we studied 623 participants (431 PD and 192 healthy controls) with available SPECT data. Even though striatal denervation was undoubtedly the imaging hallmark of PD, extrastriatal DAT uptake was also reduced in patients with PD. Topographically, widespread frontal but also temporal and posterior cortical regions showed lower DAT uptake in PD patients with respect to healthy controls. Importantly, a longitudinal voxelwise analysis confirmed an active one-year loss of extrastriatal DAT uptake within the PD group. Extrastriatal DAT uptake also correlated with the severity of motor symptoms, cognitive performance, and cerebrospinal fluid α-synuclein levels. In addition, we found an association between the Catechol-O-methyltransferase val158met genotype and extrastriatal DAT uptake. These results highlight the clinical and biological relevance of extrastriatal SPECT-DAT uptake in PD.
Collapse
Affiliation(s)
- Frederic Sampedro
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Juan Marín-Lahoz
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Saul Martínez-Horta
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Faculty of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Valle Camacho
- Nuclear Medicine Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Javier Pagonabarraga
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Faculty of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Jaime Kulisevsky
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Faculty of Medicine, Autonomous University of Barcelona, Barcelona, Spain.
| |
Collapse
|
32
|
Periñán MT, Macías-García D, Labrador-Espinosa MÁ, Jesús S, Buiza-Rueda D, Adarmes-Gómez AD, Muñoz-Delgado L, Gómez-Garre P, Mir P. Association of PICALM with Cognitive Impairment in Parkinson's Disease. Mov Disord 2020; 36:118-123. [PMID: 32914893 DOI: 10.1002/mds.28283] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/07/2020] [Accepted: 08/17/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Cognitive impairment is one of the most disabling nonmotor symptoms in Parkinson's disease (PD). Recently, a genome-wide association study in Alzheimer's disease has identified the PICALM rs3851179 polymorphism as one of the most significant susceptibility genes for Alzheimer's disease after APOE. The aim of this study was to determine the potential role of PICALM and its genetic interaction with APOE in the development of cognitive decline in PD. METHODS A discovery cohort of 712 patients with PD were genotyped for PICALM (rs3851179) and APOE (rs429358 and rs7412) polymorphisms. The association of PICALM and APOE-PICALM genetic interaction with cognitive dysfunction in PD was studied using logistic regression models, and the relationship of PICALM with cognitive decline onset was assessed with Cox regression analysis. PICALM effect was then replicated in an international, independent cohort (Parkinson's Progression Markers Initiative, N = 231). RESULTS PICALM rs3851179 TT genotype was significantly associated with a decreased risk of cognitive impairment in PD (TT vs. CC + CT, P = 0.041, odds ratio = 0.309). Replication studies further demonstrated its protective effect on cognitive impairment in PD. In addition, the protective effect of the PICALM rs3851179 TT genotype was more pronounced in the APOE ε4 (-) carriers from the discovery cohort (P = 0.037, odds ratio = 0.241), although these results were not replicated in the Parkinson's Progression Markers Initiative cohort. CONCLUSIONS Our results support the fact that PICALM is associated with cognitive impairment in PD. The understanding of its contribution to cognitive decline in PD could provide new targets for the development of novel therapies. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- María Teresa Periñán
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del, Rocío/Consejo Superior de Investigaciones Científicas (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/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Miguel Ángel Labrador-Espinosa
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del, Rocío/Consejo Superior de Investigaciones Científicas (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/Consejo Superior de Investigaciones Científicas (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/Consejo Superior de Investigaciones Científicas (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/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Laura Muñoz-Delgado
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del, Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Seville, 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/Consejo Superior de Investigaciones Científicas (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/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
33
|
Alcalay RN, Wolf P, Chiang MSR, Helesicova K, Zhang XK, Merchant K, Hutten SJ, Scherzer C, Caspell-Garcia C, Blauwendraat C, Foroud T, Nudelman K, Gan-Or Z, Simuni T, Chahine LM, Levy O, Zheng D, Li G, Sardi SP. Longitudinal Measurements of Glucocerebrosidase activity in Parkinson's patients. Ann Clin Transl Neurol 2020; 7:1816-1830. [PMID: 32888397 PMCID: PMC7545591 DOI: 10.1002/acn3.51164] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 07/30/2020] [Indexed: 12/14/2022] Open
Abstract
Objective Reduction in glucocerebrosidase (GCase; encoded by GBA) enzymatic activity has been linked to Parkinson’s disease (PD). Here, we correlated GCase activity and PD phenotype in the Parkinson’s Progression Markers Initiative (PPMI) cohort. Methods We measured GCase activity in dried blood spots from 1559 samples of participants in the inception PPMI cohort, collected in four annual visits (from baseline visit to Year‐3). Participants (PD, n = 392; controls, n = 175) were fully sequenced for GBA variants by means of genome‐wide genotyping arrays, whole‐exome sequencing, whole‐genome sequencing, Sanger sequencing, and RNA‐sequencing. Results Fifty‐two PD participants (13.4%) and 13 (7.4%) controls carried a GBA variant. GBA status was strongly associated with GCase activity. Among noncarriers, GCase activity was similar between PD and controls. Among GBA p.E326K carriers (PD, n = 20; controls, n = 5), activity was significantly lower in PD carriers than control carriers (9.53 µmol/L/h vs. 11.68 µmol/L/h, P = 0.035). Glucocerebrosidase activity was moderately (r = 0.45) associated with white blood cell (WBC) count. Next, we divided the noncarriers with PD to tertiles based on WBC count‐corrected enzymatic activity. Members of the lower tertile had higher MDS‐Unified Parkinson’s Disease Rating Scale motor score in the “off” medication examination at year‐III exam. Longitudinal analyses demonstrated slight reduction of activity in samples collected earlier on in the study, likely because of longer storage time. Interpretation GCase activity is associated with GBA genotype, WBC count, and among p.E326K variant carriers, with PD status. Reduced activity may also be associated with worse phenotype but longer follow up is required to confirm this observation.
Collapse
Affiliation(s)
- Roy N Alcalay
- Department of Neurology, Columbia University Irving Medical Center New York, New York, USA
| | - Pavlina Wolf
- Translational Sciences, Sanofi, Framingham, Massachusetts, USA
| | - Ming Sum Ruby Chiang
- Rare and Neurological Diseases Therapeutic Area, Sanofi, Framingham, Massachusetts, USA
| | | | | | - Kalpana Merchant
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Samantha J Hutten
- The Michael J. Fox Foundation for Parkinson's Research, New York, New York, USA
| | - Clemens Scherzer
- Advanced Center for Parkinson's Disease Research of Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Precision Neurology Program, Harvard Medical School, Brigham & Women's Hospital, Boston, Massachusetts, USA.,Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Chelsea Caspell-Garcia
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa, USA
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Tatiana Foroud
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kelly Nudelman
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ziv Gan-Or
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada.,Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada.,Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Tanya Simuni
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Lana M Chahine
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Oren Levy
- Department of Neurology, Columbia University Irving Medical Center New York, New York, USA
| | - Dandi Zheng
- Department of Biostatistics, Mailman School of Public Health, Columbia University Irving Medical Center New York, New York, USA
| | - Gen Li
- Department of Biostatistics, Mailman School of Public Health, Columbia University Irving Medical Center New York, New York, USA
| | - Sergio Pablo Sardi
- Rare and Neurological Diseases Therapeutic Area, Sanofi, Framingham, Massachusetts, USA
| | | |
Collapse
|
34
|
Koros C, Simitsi AM, Prentakis A, Papagiannakis N, Bougea A, Pachi I, Papadimitriou D, Beratis I, Papageorgiou SG, Stamelou M, Trapali XG, Stefanis L. DaTSCAN (123I-FP-CIT SPECT) imaging in early versus mid and late onset Parkinson's disease: Longitudinal data from the PPMI study. Parkinsonism Relat Disord 2020; 77:36-42. [PMID: 32615498 DOI: 10.1016/j.parkreldis.2020.06.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 06/10/2020] [Accepted: 06/19/2020] [Indexed: 10/24/2022]
Abstract
INTRODUCTION It has been reported that early onset Parkinson's Disease (PD) patients have a less profound dopaminergic degeneration. The aim of the current study was to determine whether there are longitudinal differences in dopaminergic denervation [signal reduction in 123I-FP-CIT SPECT] in early versus mid and late onset PD. METHODS DaTSCAN (123I-FP-CIT SPECT) imaging was acquired at Parkinson's Progression Markers Initiative (PPMI) imaging centers and sent to the imaging core for calculation of striatal binding ratios. Data from the PPMI database of 58 early de novo PD patients (age ≤ 50 years) were compared to those of 362 mid and late onset PD patients (age > 50 years). RESULTS Although raw striatal binding ratios were higher in early onset versus mid/late onset PD, especially on the ipsilateral side, such differences were not observed, and were in fact reversed in the contralateral putamen, after age correction. The rate of signal decline was similar between the two groups. Interestingly, based on both raw and age-adjusted data, caudate nucleus and putamen asymmetry (contralateral/ipsilateral ratio) was more pronounced in early onset PD. Striatal asymmetry also significantly correlated with age at onset as a continuous variable. CONCLUSION Early onset PD patients exhibited similar rates of decline of dopaminergic denervation compared to mid/late onset PD. These results are not supportive of a more benign disease in this subgroup. The more pronounced asymmetry in early onset PD may however signify a qualitatively different pattern of neurodegeneration compared to mid/late onset PD.
Collapse
Affiliation(s)
- Christos Koros
- 1st Neurology Clinic, Eginition Hospital, Athens University Medical School, Athens, Greece; 2nd Neurology Clinic, Attikon Hospital, Athens University Medical School, Athens, Greece
| | - Athina-Maria Simitsi
- 1st Neurology Clinic, Eginition Hospital, Athens University Medical School, Athens, Greece
| | - Andreas Prentakis
- 1st Neurology Clinic, Eginition Hospital, Athens University Medical School, Athens, Greece; Nuclear Medicine Unit, Attikon Hospital, Athens, Greece
| | - Nikolaos Papagiannakis
- 1st Neurology Clinic, Eginition Hospital, Athens University Medical School, Athens, Greece
| | - Anastasia Bougea
- 1st Neurology Clinic, Eginition Hospital, Athens University Medical School, Athens, Greece
| | - Ioanna Pachi
- 1st Neurology Clinic, Eginition Hospital, Athens University Medical School, Athens, Greece; 2nd Neurology Clinic, Attikon Hospital, Athens University Medical School, Athens, Greece
| | | | - Ion Beratis
- 2nd Neurology Clinic, Attikon Hospital, Athens University Medical School, Athens, Greece
| | | | - Maria Stamelou
- 1st Neurology Clinic, Eginition Hospital, Athens University Medical School, Athens, Greece; Neurology Clinic, Philipps University, Marburg, Germany; Parkinson's Disease and Movement Disorders Dept., HYGEIA Hospital, Athens, Greece
| | | | - Leonidas Stefanis
- 1st Neurology Clinic, Eginition Hospital, Athens University Medical School, Athens, Greece.
| |
Collapse
|
35
|
Bobbili DR, Banda P, Krüger R, May P. Excess of singleton loss-of-function variants in Parkinson's disease contributes to genetic risk. J Med Genet 2020; 57:617-623. [PMID: 32054687 PMCID: PMC7476273 DOI: 10.1136/jmedgenet-2019-106316] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 12/04/2019] [Accepted: 01/20/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative disorder with complex genetic architecture. Besides rare mutations in high-risk genes related to monogenic familial forms of PD, multiple variants associated with sporadic PD were discovered via association studies. METHODS We studied the whole-exome sequencing data of 340 PD cases and 146 ethnically matched controls from the Parkinson's Progression Markers Initiative (PPMI) and performed burden analysis for different rare variant classes. Disease prediction models were built based on clinical, non-clinical and genetic features, including both common and rare variants, and two machine learning methods. RESULTS We observed a significant exome-wide burden of singleton loss-of-function variants (corrected p=0.037). Overall, no exome-wide burden of rare amino acid changing variants was detected. Finally, we built a disease prediction model combining singleton loss-of-function variants, a polygenic risk score based on common variants, and family history of PD as features and reached an area under the curve of 0.703 (95% CI 0.698 to 0.708). By incorporating a rare variant feature, our model increased the performance of the state-of-the-art classification model for the PPMI dataset, which reached an area under the curve of 0.639 based on common variants alone. CONCLUSION The main finding of this study is to highlight the contribution of singleton loss-of-function variants to the complex genetics of PD and that disease risk prediction models combining singleton and common variants can improve models built solely on common variants.
Collapse
Affiliation(s)
- Dheeraj Reddy Bobbili
- Bioinformatics Core, Luxembourg Centre for Systems Biomedicine (LCSB), Belvaux, Luxembourg .,MeGeno S.A, Esch-sur-Alzette, Luxembourg
| | - Peter Banda
- Bioinformatics Core, Luxembourg Centre for Systems Biomedicine (LCSB), Belvaux, Luxembourg
| | - Rejko Krüger
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), Belvaux, Luxembourg.,Parkinson Research Clinic, Centre Hospitalier de Luxemborg (CHL), Luxembourg, Luxembourg.,Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | - Patrick May
- Bioinformatics Core, Luxembourg Centre for Systems Biomedicine (LCSB), Belvaux, Luxembourg
| |
Collapse
|
36
|
Parkinson's disease prognostic scores for progression of cognitive decline. Sci Rep 2019; 9:17485. [PMID: 31767922 PMCID: PMC6877592 DOI: 10.1038/s41598-019-54029-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 11/08/2019] [Indexed: 11/08/2022] Open
Abstract
Clinical and biochemical diversity of Parkinson’s disease (PD) presents a major challenge for accurate diagnosis and prediction of its progression. We propose, develop and optimize PD clinical scores as efficient integrated progression biomarkers for prediction of the likely rate of cognitive decline in PD patients. We considered 269 drug-naïve participants from the Parkinson’s Progression Marker Initiative database, diagnosed with idiopathic PD and observed between 4 and 6 years. Nineteen baseline clinical and pathological measures were systematically considered. Relative variable importance and logistic regressions were used to optimize combinations of significant baseline measures as integrated biomarkers. Parkinson’s disease cognitive decline scores were designed as new clinical biomarkers using optimally categorized baseline measures. Specificities and sensitivities of the biomarkers reached ~93% for prediction of severe rate of cognitive decline (with more than 5 points decline in 4 years on the Montreal Cognitive Assessment scale), and up to ~73% for mild-to-moderate decline (between 1 and 5 points decline). The developed biomarkers and clinical scores could resolve the long-standing clinical problem about reliable prediction of PD progression into cognitive deterioration. The outcomes also provide insights into the contributions of individual clinical and pathological measures to PD progression, and will assist with better-targeted treatment regiments, stratification of clinical trial and their evaluation.
Collapse
|
37
|
Mollenhauer B, Caspell-Garcia CJ, Coffey CS, Taylor P, Singleton A, Shaw LM, Trojanowski JQ, Frasier M, Simuni T, Iranzo A, Oertel W, Siderowf A, Weintraub D, Seibyl J, Toga AW, Tanner CM, Kieburtz K, Chahine LM, Marek K, Galasko D. Longitudinal analyses of cerebrospinal fluid α-Synuclein in prodromal and early Parkinson's disease. Mov Disord 2019; 34:1354-1364. [PMID: 31361367 PMCID: PMC7098385 DOI: 10.1002/mds.27806] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/17/2019] [Accepted: 07/08/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Aggregation of α-synuclein is central to the pathophysiology of PD. Biomarkers related to α-synuclein may be informative for PD diagnosis/progression. OBJECTIVES To analyze α-synuclein in CSF in drug-naïve PD, healthy controls, and prodromal PD in the Parkinson's Progression Markers Initiative. METHODS Over up to 36-month follow-up, CSF total α-synuclein and its association with MDS-UPDRS motor scores, cognitive assessments, and dopamine transporter imaging were assessed. RESULTS The inception cohort included PD (n = 376; age [mean {standard deviation} years]: 61.7 [9.62]), healthy controls (n = 173; age, 60.9 [11.3]), hyposmics (n = 16; age, 68.3 [6.15]), and idiopathic rapid eye movement sleep behavior disorder (n = 32; age, 69.3 [4.83]). Baseline CSF α-synuclein was lower in manifest and prodromal PD versus healthy controls. Longitudinal α-synuclein decreased significantly in PD at 24 and 36 months, did not change in prodromal PD over 12 months, and trended toward an increase in healthy controls. The decrease in PD was not shown when CSF samples with high hemoglobin concentration were removed from the analysis. CSF α-synuclein changes did not correlate with longitudinal MDS-UPDRS motor scores or dopamine transporter scan. CONCLUSIONS CSF α-synuclein decreases early in the disease, preceding motor PD. CSF α-synuclein does not correlate with progression and therefore does not reflect ongoing dopaminergic neurodegeneration. Decreased CSF α-synuclein may be an indirect index of changes in the balance between α-synuclein secretion, solubility, or aggregation in the brain, reflecting its overall turnover. Additional biomarkers more directly related to α-synuclein pathophysiology and disease progression and other markers to be identified by, for example, proteomics and metabolomics are needed. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Brit Mollenhauer
- Department of Neurology, University Medical Center Goettingen, Göttingen, Germany; and Paracelsus-Elena Klinik, Kassel, Germany
| | | | - Christopher S. Coffey
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa, USA
| | | | - Andy Singleton
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Leslie M. Shaw
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John Q. Trojanowski
- Center for Neurodegenerative Disease Research, Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Morris K. Udall Center of Excellence for Parkinson’s Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mark Frasier
- The Michael J. Fox Foundation for Parkinson’s Research, New York, New York, USA
| | - Tanya Simuni
- Parkinson’s Disease and Movement Disorders Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Alex Iranzo
- Neurological Service, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Wolfgang Oertel
- Department of Neurology, Philipps University Marburg, Marburg, Germany
| | - Andrew Siderowf
- Morris K. Udall Center of Excellence for Parkinson’s Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Neurology Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel Weintraub
- Department of Neurology Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John Seibyl
- Institute for Neurodegenerative Disorders, New Haven, Connecticut, USA
| | - Arthur W. Toga
- University of Southern California, Laboratory of Neuro Imaging, Los Angeles, California, USA
| | - Caroline M. Tanner
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Karl Kieburtz
- Clinical Trials Coordination Center, University of Rochester Medical Center, Rochester, New York, USA
| | - Lana M. Chahine
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kenneth Marek
- Institute for Neurodegenerative Disorders, New Haven, Connecticut, USA
| | - Douglas Galasko
- Department of Neurosciences, University of California, San Diego, San Diego, California, USA
| |
Collapse
|
38
|
Bloem BR, Marks WJ, Silva de Lima AL, Kuijf ML, van Laar T, Jacobs BPF, Verbeek MM, Helmich RC, van de Warrenburg BP, Evers LJW, intHout J, van de Zande T, Snyder TM, Kapur R, Meinders MJ. The Personalized Parkinson Project: examining disease progression through broad biomarkers in early Parkinson's disease. BMC Neurol 2019; 19:160. [PMID: 31315608 PMCID: PMC6636112 DOI: 10.1186/s12883-019-1394-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/04/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Our understanding of the etiology, pathophysiology, phenotypic diversity, and progression of Parkinson's disease has stagnated. Consequently, patients do not receive the best care, leading to unnecessary disability, and to mounting costs for society. The Personalized Parkinson Project (PPP) proposes an unbiased approach to biomarker development with multiple biomarkers measured longitudinally. Our main aims are: (a) to perform a set of hypothesis-driven analyses on the comprehensive dataset, correlating established and novel biomarkers to the rate of disease progression and to treatment response; and (b) to create a widely accessible dataset for discovery of novel biomarkers and new targets for therapeutic interventions in Parkinson's disease. METHODS/DESIGN This is a prospective, longitudinal, single-center cohort study. The cohort will comprise 650 persons with Parkinson's disease. The inclusion criteria are purposely broad: age ≥ 18 years; and disease duration ≤5 years. Participants are followed for 2 years, with three annual assessments at the study center. Outcomes include a clinical assessment (including motor and neuro-psychological tests), collection of biospecimens (stool, whole blood, and cerebrospinal fluid), magnetic resonance imaging (both structural and functional), and ECG recordings (both 12-lead and Holter). Additionally, collection of physiological and environmental data in daily life over 2 years will be enabled through the Verily Study Watch. All data are stored with polymorphic encryptions and pseudonyms, to guarantee the participants' privacy on the one hand, and to enable data sharing on the other. The data and biospecimens will become available for scientists to address Parkinson's disease-related research questions. DISCUSSION The PPP has several distinguishing elements: all assessments are done in a single center; inclusion of "real life" subjects; deep and repeated multi-dimensional phenotyping; and continuous monitoring with a wearable device for 2 years. Also, the PPP is powered by privacy and security by design, allowing for data sharing with scientists worldwide respecting participants' privacy. The data are expected to open the way for important new insights, including identification of biomarkers to predict differences in prognosis and treatment response between patients. Our long-term aim is to improve existing treatments, develop new therapeutic approaches, and offer Parkinson's disease patients a more personalized disease management approach. TRIAL REGISTRATION Clinical Trials NCT03364894 . Registered December 6, 2017 (retrospectively registered).
Collapse
Affiliation(s)
- B. R. Bloem
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - W. J. Marks
- Verily Life Sciences, South San Francisco, CA USA
| | - A. L. Silva de Lima
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- CAPES Foundation, Ministry of Education of Brazil, Brasília/DF, Brazil
| | - M. L. Kuijf
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - T. van Laar
- Department of Neurology, Universtity Medical Center Groningen, Groningen, The Netherlands
| | - B. P. F. Jacobs
- Faculty of Science, University of Nijmegen, Nijmegen, The Netherlands
| | - M. M. Verbeek
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Donders Institute for Brain, Cognition and Behaviour, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - R. C. Helmich
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - B. P. van de Warrenburg
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - L. J. W. Evers
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Computing and Information Sciences, Radboud University, Nijmegen, The Netherlands
| | - J. intHout
- Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - T. van de Zande
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - T. M. Snyder
- Verily Life Sciences, South San Francisco, CA USA
| | - R. Kapur
- Neurology Platform, Verily Life Sciences, South San Francisco, CA USA
| | - M. J. Meinders
- Scientific Center for Quality of Healthcare, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
39
|
Lee MJ, Pak K, Kim JH, Kim YJ, Yoon J, Lee J, Lyoo CH, Park HJ, Lee JH, Jung NY. Effect of polygenic load on striatal dopaminergic deterioration in Parkinson disease. Neurology 2019; 93:e665-e674. [PMID: 31289143 DOI: 10.1212/wnl.0000000000007939] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 03/21/2019] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To investigate the effect of polygenic load on the progression of striatal dopaminergic dysfunction in patients with Parkinson disease (PD). METHODS Using data from 335 patients with PD in the Parkinson's Progression Markers Initiative (PPMI) database, we investigated the longitudinal association of PD-associated polygenic load with changes in striatal dopaminergic activity as measured by 123I-N-3-fluoropropyl-2-β-carboxymethoxy-3β-(4-iodophenyl) nortropane (123I-FP-CIT) SPECT over 4 years. PD-associated polygenic load was estimated by calculating weighted genetic risk scores (GRS) using 1) all available 27 PD-risk single nucleotide polymorphisms (SNPs) in the PPMI database (GRS1) and 2) 23 SNPs with minor allele frequency >0.05 (GRS2). RESULTS GRS1 and GRS2 were correlated with younger age at onset in patients with PD (GRS1, Spearman ρ = -0.128, p = 0.019; GRS2, Spearman ρ = -0.109, p = 0.047). Although GRS1 did not show an association with changes in striatal 123I-FP-CIT availability, GRS2 was associated with a slower decline of striatal dopaminergic activity (interactions with disease duration in linear mixed model; caudate nucleus, estimate = 0.399, SE = 0.165, p = 0.028; putamen, estimate = 0.396, SE = 0.137, p = 0.016). CONCLUSIONS Our results suggest that genetic factors for PD risk may have heterogeneous effects on striatal dopaminergic degeneration, and some factors may be associated with a slower decline of dopaminergic activity. Composition of PD progression-specific GRS may be useful in predicting disease progression in patients.
Collapse
Affiliation(s)
- Myung Jun Lee
- From the Departments of Neurology (M.J.L.) and Nuclear Medicine (K.P.), Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Busan; Department of Neurology (J.H.K.), National Health Insurance Service Ilsan Hospital, Goyang; Department of Neurology (Y.J.K.), Hallym University College of Medicine, Anyang; Department of Computer Engineering (J.Y., J.L.), Hallym University, Chuncheon; Department of Neurology (C.H.L.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul; Department of Neurology (H.J.P.), Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung; and Department of Neurology (J.-H.L., N.-Y.J.), Pusan National University Yangsan Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Yangsan, Republic of Korea.
| | - Kyoungjune Pak
- From the Departments of Neurology (M.J.L.) and Nuclear Medicine (K.P.), Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Busan; Department of Neurology (J.H.K.), National Health Insurance Service Ilsan Hospital, Goyang; Department of Neurology (Y.J.K.), Hallym University College of Medicine, Anyang; Department of Computer Engineering (J.Y., J.L.), Hallym University, Chuncheon; Department of Neurology (C.H.L.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul; Department of Neurology (H.J.P.), Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung; and Department of Neurology (J.-H.L., N.-Y.J.), Pusan National University Yangsan Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Yangsan, Republic of Korea
| | - Jong Hun Kim
- From the Departments of Neurology (M.J.L.) and Nuclear Medicine (K.P.), Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Busan; Department of Neurology (J.H.K.), National Health Insurance Service Ilsan Hospital, Goyang; Department of Neurology (Y.J.K.), Hallym University College of Medicine, Anyang; Department of Computer Engineering (J.Y., J.L.), Hallym University, Chuncheon; Department of Neurology (C.H.L.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul; Department of Neurology (H.J.P.), Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung; and Department of Neurology (J.-H.L., N.-Y.J.), Pusan National University Yangsan Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Yangsan, Republic of Korea
| | - Yun Joong Kim
- From the Departments of Neurology (M.J.L.) and Nuclear Medicine (K.P.), Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Busan; Department of Neurology (J.H.K.), National Health Insurance Service Ilsan Hospital, Goyang; Department of Neurology (Y.J.K.), Hallym University College of Medicine, Anyang; Department of Computer Engineering (J.Y., J.L.), Hallym University, Chuncheon; Department of Neurology (C.H.L.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul; Department of Neurology (H.J.P.), Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung; and Department of Neurology (J.-H.L., N.-Y.J.), Pusan National University Yangsan Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Yangsan, Republic of Korea
| | - Jeehee Yoon
- From the Departments of Neurology (M.J.L.) and Nuclear Medicine (K.P.), Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Busan; Department of Neurology (J.H.K.), National Health Insurance Service Ilsan Hospital, Goyang; Department of Neurology (Y.J.K.), Hallym University College of Medicine, Anyang; Department of Computer Engineering (J.Y., J.L.), Hallym University, Chuncheon; Department of Neurology (C.H.L.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul; Department of Neurology (H.J.P.), Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung; and Department of Neurology (J.-H.L., N.-Y.J.), Pusan National University Yangsan Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Yangsan, Republic of Korea
| | - Jinwoo Lee
- From the Departments of Neurology (M.J.L.) and Nuclear Medicine (K.P.), Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Busan; Department of Neurology (J.H.K.), National Health Insurance Service Ilsan Hospital, Goyang; Department of Neurology (Y.J.K.), Hallym University College of Medicine, Anyang; Department of Computer Engineering (J.Y., J.L.), Hallym University, Chuncheon; Department of Neurology (C.H.L.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul; Department of Neurology (H.J.P.), Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung; and Department of Neurology (J.-H.L., N.-Y.J.), Pusan National University Yangsan Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Yangsan, Republic of Korea.
| | - Chul Hyoung Lyoo
- From the Departments of Neurology (M.J.L.) and Nuclear Medicine (K.P.), Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Busan; Department of Neurology (J.H.K.), National Health Insurance Service Ilsan Hospital, Goyang; Department of Neurology (Y.J.K.), Hallym University College of Medicine, Anyang; Department of Computer Engineering (J.Y., J.L.), Hallym University, Chuncheon; Department of Neurology (C.H.L.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul; Department of Neurology (H.J.P.), Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung; and Department of Neurology (J.-H.L., N.-Y.J.), Pusan National University Yangsan Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Yangsan, Republic of Korea
| | - Hyung Jun Park
- From the Departments of Neurology (M.J.L.) and Nuclear Medicine (K.P.), Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Busan; Department of Neurology (J.H.K.), National Health Insurance Service Ilsan Hospital, Goyang; Department of Neurology (Y.J.K.), Hallym University College of Medicine, Anyang; Department of Computer Engineering (J.Y., J.L.), Hallym University, Chuncheon; Department of Neurology (C.H.L.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul; Department of Neurology (H.J.P.), Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung; and Department of Neurology (J.-H.L., N.-Y.J.), Pusan National University Yangsan Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Yangsan, Republic of Korea
| | - Jae-Hyeok Lee
- From the Departments of Neurology (M.J.L.) and Nuclear Medicine (K.P.), Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Busan; Department of Neurology (J.H.K.), National Health Insurance Service Ilsan Hospital, Goyang; Department of Neurology (Y.J.K.), Hallym University College of Medicine, Anyang; Department of Computer Engineering (J.Y., J.L.), Hallym University, Chuncheon; Department of Neurology (C.H.L.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul; Department of Neurology (H.J.P.), Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung; and Department of Neurology (J.-H.L., N.-Y.J.), Pusan National University Yangsan Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Yangsan, Republic of Korea.
| | - Na-Yeon Jung
- From the Departments of Neurology (M.J.L.) and Nuclear Medicine (K.P.), Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Busan; Department of Neurology (J.H.K.), National Health Insurance Service Ilsan Hospital, Goyang; Department of Neurology (Y.J.K.), Hallym University College of Medicine, Anyang; Department of Computer Engineering (J.Y., J.L.), Hallym University, Chuncheon; Department of Neurology (C.H.L.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul; Department of Neurology (H.J.P.), Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung; and Department of Neurology (J.-H.L., N.-Y.J.), Pusan National University Yangsan Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Yangsan, Republic of Korea
| |
Collapse
|
40
|
Tropea TF, Mak J, Guo MH, Xie SX, Suh E, Rick J, Siderowf A, Weintraub D, Grossman M, Irwin D, Wolk DA, Trojanowski JQ, Van Deerlin V, Chen-Plotkin AS. TMEM106B Effect on cognition in Parkinson disease and frontotemporal dementia. Ann Neurol 2019; 85:801-811. [PMID: 30973966 PMCID: PMC6953172 DOI: 10.1002/ana.25486] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 04/10/2019] [Accepted: 04/10/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Common variants near TMEM106B associate with risk of developing frontotemporal dementia (FTD). Emerging evidence suggests a role for TMEM106B in neurodegenerative processes beyond FTD. We evaluate the effect of TMEM106B genotype on cognitive decline across multiple neurogenerative diseases. METHODS We longitudinally followed 870 subjects with diagnoses of Parkinson disease (PD; n = 179), FTD (n = 179), Alzheimer disease (AD; n = 300), memory-predominant mild cognitive impairment (MCI; n = 75), or neurologically normal control subjects (NC; n = 137) at the University of Pennsylvania (UPenn). All participants had annual Mini-Mental State Examination (MMSE; median follow-up duration = 3.0 years) and were genotyped at TMEM106B index single nucleotide polymorphism rs1990622. Genotype effects on cognition were confirmed by extending analyses to additional cognitive instruments (Mattis Dementia Rating Scale-2 [DRS-2] and Montreal Cognitive Assessment [MoCA]) and to an international validation cohort (Parkinson's Progression Markers Initiative [PPMI], N = 371). RESULTS The TMEM106B rs1990622T allele, linked to increased risk of FTD, associated with greater MMSE decline over time in PD subjects but not in AD or MCI subjects. For FTD subjects, rs1990622T associated with more rapid decrease in MMSE only under the minor-allele, rs1990622C , dominant model. Among PD patients, rs1990622T carriers from the UPenn cohort demonstrated more rapid longitudinal decline in DRS-2 scores. Finally, in the PPMI cohort, TMEM106B risk allele carriers demonstrated more rapid longitudinal decline in MoCA scores. INTERPRETATION Irrespective of cognitive instrument or cohort assessed, TMEM106B acts as a genetic modifier for cognitive trajectory in PD. Our results implicate lysosomal dysfunction in the pathogenesis of cognitive decline in 2 different proteinopathies. ANN NEUROL 2019;85:801-811.
Collapse
Affiliation(s)
- Thomas F Tropea
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jordan Mak
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michael H Guo
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA
- Department of Medicine, University of North Carolina Hospitals, Chapel Hill, NC
| | - Sharon X Xie
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Eunran Suh
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jacqueline Rick
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Andrew Siderowf
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Daniel Weintraub
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Parkinson's Disease and Mental Illness Research, Education, and Clinical Centers, Philadelphia Veterans Affairs Medical Center, Philadelphia, PA
| | - Murray Grossman
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - David Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - David A Wolk
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Vivianna Van Deerlin
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Alice S Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
41
|
Selecting variants of unknown significance through network-based gene-association significantly improves risk prediction for disease-control cohorts. Sci Rep 2019; 9:3266. [PMID: 30824863 PMCID: PMC6397233 DOI: 10.1038/s41598-019-39796-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 01/31/2019] [Indexed: 12/12/2022] Open
Abstract
Variants of unknown/uncertain significance (VUS) pose a huge dilemma in current genetic variation screening methods and genetic counselling. Driven by methods of next generation sequencing (NGS) such as whole exome sequencing (WES), a plethora of VUS are being detected in research laboratories as well as in the health sector. Motivated by this overabundance of VUS, we propose a novel computational methodology, termed VariantClassifier (VarClass), which utilizes gene-association networks and polygenic risk prediction models to shed light into this grey area of genetic variation in association with disease. VarClass has been evaluated using numerous validation steps and proves to be very successful in assigning significance to VUS in association with specific diseases of interest. Notably, using VUS that are deemed significant by VarClass, we improved risk prediction accuracy in four large case-studies involving disease-control cohorts from GWAS as well as WES, when compared to traditional odds ratio analysis. Biological interpretation of selected high scoring VUS revealed interesting biological themes relevant to the diseases under investigation. VarClass is available as a standalone tool for large-scale data analyses, as well as a web-server with additional functionalities through a user-friendly graphical interface.
Collapse
|
42
|
Shahid M, Kim J, Leaver K, Hendershott T, Zhu D, Cholerton B, Henderson VW, Tian L, Poston KL. An increased rate of longitudinal cognitive decline is observed in Parkinson's disease patients with low CSF Aß42 and an APOE ε4 allele. Neurobiol Dis 2019; 127:278-286. [PMID: 30826425 DOI: 10.1016/j.nbd.2019.02.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 01/09/2019] [Accepted: 02/27/2019] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVE Low concentrations of cerebrospinal fluid (CSF) amyloid-beta (Aβ-42) are associated with increased risk of cognitive decline in Parkinson's disease (PD). We sought to determine whether APOE genotype modifies the rate of cognitive decline in PD patients with low CSF Aβ-42 compared to patients with normal levels. METHODS The Parkinson's Progression Markers Initiative is a longitudinal, ongoing study of de novo PD participants, which includes APOE genotyping, CSF Aβ-42 determinations, and neuropsychological assessments. We used linear mixed effects models in three PD groups (PD participants with low CSF Aβ at baseline, PD participants with normal CSF Aβ, and both groups combined). Having at least one copy of the APOE ɛ4 allele, time, and the interaction of APOE ɛ4 and time were predictor variables for cognitive change, adjusting for age, gender and education. RESULTS 423 de novo PD participants were followed up to 5 years with annual cognitive assessments. 103 participants had low baseline CSF Aβ-42 (39 APOE ε4+, 64 APOE ε4-). Compared to participants with normal CSF Aβ-42, those with low CSF Aβ-42 declined faster on most cognitive tests. Within the low CSF Aβ-42 group, APOE ε4+ participants had faster rates of decline on the Montreal Cognitive Assessment (primary outcome; 0.57 points annual decline, p = .005; 5-year standardized change of 1.2) and the Symbol Digit Modalities Test (1.4 points annual decline, p = .002; 5-year standardized change of 0.72). DISCUSSION PD patients with low CSF Aβ-42 and APOE ε4+ showed a higher rate of cognitive decline early in the disease. Tests of global cognition (Montreal Cognitive Assessment) and processing speed (Symbol Digit Modalities Test) were the most sensitive to early cognitive decline. Results suggest that CSF Aβ-42 and APOE ε4 might interact to promote early cognitive changes in PD patients.
Collapse
Affiliation(s)
- Marian Shahid
- Stanford University, Department of Neurology and Neurological Sciences, 300 Pasteur Dr. Room H3144, MC 5235, Stanford, CA 94305, United States of America
| | - Jeehyun Kim
- Stanford University, Department of Neurology and Neurological Sciences, 300 Pasteur Dr. Room H3144, MC 5235, Stanford, CA 94305, United States of America
| | - Katherine Leaver
- Stanford University, Department of Neurology and Neurological Sciences, 300 Pasteur Dr. Room H3144, MC 5235, Stanford, CA 94305, United States of America; Mount Sinai Beth Israel, Department of Neurology, 10 Union Square East, New York, NY 10003, United States of America
| | - Taylor Hendershott
- Stanford University, Department of Neurology and Neurological Sciences, 300 Pasteur Dr. Room H3144, MC 5235, Stanford, CA 94305, United States of America
| | - Delphine Zhu
- Stanford University, Department of Neurology and Neurological Sciences, 300 Pasteur Dr. Room H3144, MC 5235, Stanford, CA 94305, United States of America
| | - Brenna Cholerton
- Stanford University, Department of Pathology, 300 Pasteur Dr Rm L235, MC 5324, Stanford, CA 94305, United States of America
| | - Victor W Henderson
- Stanford University, Department of Neurology and Neurological Sciences, 300 Pasteur Dr. Room H3144, MC 5235, Stanford, CA 94305, United States of America; Stanford University, Department of Health Research and Policy (Epidemiology), 259 Campus Drive, MC 5405, Stanford, CA 94305, United States of America
| | - Lu Tian
- Stanford University, Department of Biomedical Data Science, 150 Governor's Lane, Room T160C, MC 5464, Stanford, CA 94305, United States of America
| | - Kathleen L Poston
- Stanford University, Department of Neurology and Neurological Sciences, 300 Pasteur Dr. Room H3144, MC 5235, Stanford, CA 94305, United States of America; Stanford University, Department of Neurosurgery, 300 Pasteur Dr. Room H3144, MC 5235, Stanford, CA 94305, United States of America.
| |
Collapse
|
43
|
Won JH, Kim M, Park BY, Youn J, Park H. Effectiveness of imaging genetics analysis to explain degree of depression in Parkinson's disease. PLoS One 2019; 14:e0211699. [PMID: 30742647 PMCID: PMC6370199 DOI: 10.1371/journal.pone.0211699] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 01/18/2019] [Indexed: 12/20/2022] Open
Abstract
Depression is one of the most common and important neuropsychiatric symptoms in Parkinson's disease and often becomes worse as Parkinson's disease progresses. However, the underlying mechanisms of depression in Parkinson's disease are not clear. The aim of our study was to find genetic features related to depression in Parkinson's disease using an imaging genetics approach and to construct an analytical model for predicting the degree of depression in Parkinson's disease. The neuroimaging and genotyping data were obtained from an openly accessible database. We computed imaging features through connectivity analysis derived from tractography of diffusion tensor imaging. The imaging features were used as intermediate phenotypes to identify genetic variants according to the imaging genetics approach. We then constructed a linear regression model using the genetic features from imaging genetics approach to describe clinical scores indicating the degree of depression. As a comparison, we constructed other models using imaging features and genetic features based on references to demonstrate the effectiveness of our imaging genetics model. The models were trained and tested in a five-fold cross-validation. The imaging genetics approach identified several brain regions and genes known to be involved in depression, with the potential to be used as meaningful biomarkers. Our proposed model using imaging genetic features predicted and explained the degree of depression in Parkinson's disease appropriately (adjusted R2 larger than 0.6 over five training folds) and with a lower error and higher correlation than with other models over five test folds.
Collapse
Affiliation(s)
- Ji Hye Won
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea
| | - Mansu Kim
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea
| | - Bo-yong Park
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea
| | - Jinyoung Youn
- Department of Neurology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Hyunjin Park
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea
- School of Electronic and Electrical Engineering, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
44
|
Kristiansen M, Maple-Grødem J, Alves G, Arepalli S, Hernandez DG, Iwaki H, Nalls MA, Singleton A, Tysnes OB, Toft M, Pihlstrøm L. A paradoxical relationship between family history, onset age, and genetic risk in Parkinson's disease. Mov Disord 2018; 34:298-299. [PMID: 30484896 DOI: 10.1002/mds.27555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 09/25/2018] [Accepted: 10/10/2018] [Indexed: 11/05/2022] Open
Affiliation(s)
| | - Jodi Maple-Grødem
- The Norwegian Center for Movement Disorders, Stavanger University Hospital, Stavanger, Norway.,The Centre for Organelle Research, University of Stavanger, Stavanger, Norway
| | - Guido Alves
- The Norwegian Center for Movement Disorders, Stavanger University Hospital, Stavanger, Norway.,Department of Neurology, Stavanger University Hospital, Stavanger, Norway.,Department of Mathematics and Natural Sciences, University of Stavanger, Stavanger, Norway
| | - Sampath Arepalli
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Dena G Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Hirotaka Iwaki
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA.,Founder/Consultant with Data Tecnica International, Glen Echo, Maryland, USA
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Ole-Bjørn Tysnes
- Department of Neurology, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Mathias Toft
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
45
|
Eusebi P, Romoli M, Paoletti FP, Tambasco N, Calabresi P, Parnetti L. Risk factors of levodopa-induced dyskinesia in Parkinson's disease: results from the PPMI cohort. NPJ Parkinsons Dis 2018; 4:33. [PMID: 30480086 PMCID: PMC6240081 DOI: 10.1038/s41531-018-0069-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/24/2018] [Indexed: 11/10/2022] Open
Abstract
Levodopa-induced dyskinesias (LID) negatively impact on the quality of life of patients with Parkinson's disease (PD). We assessed the risk factors for LID in a cohort of de-novo PD patients enrolled in the Parkinson's Progression Markers Initiative (PPMI). This retrospective cohort study included all PD patients enrolled in the PPMI cohort. Main outcome was the incidence rate of dyskinesia, defined as the first time the patient reported a non-zero score in the item "Time spent with dyskinesia" of the MDS-UPDRS part IV. Predictive value for LID development was assessed for clinical and demographical features, dopamine transporter imaging (DaTscan) pattern, cerebrospinal fluid (CSF) biomarkers (Aβ42, total tau, phosphorylated tau, total α synuclein) and genetic risk score for PD. Overall, data from 423 PD patients were analyzed. The cumulative incidence rate of LID was 27.4% (95% CI = 23.2-32.0%), with a mean onset time of 5.81 years from PD diagnosis. Multivariate Cox regression analysis showed several factors predicting LID development, including female gender (HR = 1.61, 95% CI = 1.05-2.47), being not completely functional independent as measured by the modified Schwab & England ADL scale (HR = 1.81, 95% CI = 0.98-3.38), higher MDS-UPDRS part III score (HR = 1.03, 95% CI = 1.00-1.05), postural instability gait disturbances or intermediate phenotypes (HR = 1.95, 95% CI = 1.28-2.96), higher DaTscan caudate asymmetry index (HR = 1.02, 95% CI = 1.00-1.03), higher polygenic genetic risk score (HR = 1.39, 95% CI = 1.08-1.78), and an anxiety trait (HR = 1.02, 95% CI = 1.00-1.04). In PD patients, cumulative levodopa exposure, female gender, severity of motor and functional impairment, non-tremor dominant clinical phenotype, genetic risk score, anxiety, and marked caudate asymmetric pattern at DaTscan at baseline represent independent risk factors for developing LID.
Collapse
Affiliation(s)
- Paolo Eusebi
- Neurology Clinic, Department of Medicine, University of Perugia, Ospedale S. Maria della Misericordia, Perugia, Italy
| | - Michele Romoli
- Neurology Clinic, Department of Medicine, University of Perugia, Ospedale S. Maria della Misericordia, Perugia, Italy
| | - Federico Paolini Paoletti
- Neurology Clinic, Department of Medicine, University of Perugia, Ospedale S. Maria della Misericordia, Perugia, Italy
| | - Nicola Tambasco
- Neurology Clinic, Department of Medicine, University of Perugia, Ospedale S. Maria della Misericordia, Perugia, Italy
| | - Paolo Calabresi
- Neurology Clinic, Department of Medicine, University of Perugia, Ospedale S. Maria della Misericordia, Perugia, Italy
- IRCCS Santa Lucia, Rome, Italy
| | - Lucilla Parnetti
- Neurology Clinic, Department of Medicine, University of Perugia, Ospedale S. Maria della Misericordia, Perugia, Italy
| |
Collapse
|
46
|
Berge-Seidl V, Pihlstrøm L, Wszolek ZK, Ross OA, Toft M. No evidence for DNM3 as genetic modifier of age at onset in idiopathic Parkinson's disease. Neurobiol Aging 2018; 74:236.e1-236.e5. [PMID: 30340792 DOI: 10.1016/j.neurobiolaging.2018.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 09/14/2018] [Accepted: 09/15/2018] [Indexed: 11/29/2022]
Abstract
Parkinson's disease (PD) is a disorder with highly variable clinical phenotype. The identification of genetic variants modifying age at onset and other traits is of great interest because it may provide insight into disease mechanisms and potential therapeutic targets. A variant in the DNM3 gene (rs2421947) has been reported as a genetic modifier of age at onset in LRRK2-associated PD. To test the possible effect of genetic variation in DNM3 on age at onset in idiopathic PD, we examined rs2421947 in a total of 5918 patients with PD from seven data sets. We also assessed the potential effect of all common variants in the DNM3 locus. There was no significant association between rs2421947 and age at onset in any of the individual studies. Meta-analysis of the seven studies was nonsignificant and the between-study heterogeneity was minimal. No other common variants within the DNM3 locus affected age at onset. In conclusion, we find no evidence of an association between DNM3 variants and age at onset in idiopathic PD.
Collapse
Affiliation(s)
- Victoria Berge-Seidl
- Department of Neurology, Oslo University Hospital, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | | | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Mathias Toft
- Department of Neurology, Oslo University Hospital, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
47
|
Pihlstrøm L, Blauwendraat C, Cappelletti C, Berge-Seidl V, Langmyhr M, Henriksen SP, van de Berg WDJ, Gibbs JR, Cookson MR, Singleton AB, Nalls MA, Toft M. A comprehensive analysis of SNCA-related genetic risk in sporadic parkinson disease. Ann Neurol 2018; 84:117-129. [PMID: 30146727 DOI: 10.1002/ana.25274] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/13/2018] [Accepted: 06/15/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The goal of this study was to refine our understanding of disease risk attributable to common genetic variation in SNCA, a major locus in Parkinson disease, with potential implications for clinical trials targeting α-synuclein. We aimed to dissect the multiple independent association signals, stratify individuals by SNCA-specific risk profiles, and explore expression quantitative trait loci. METHODS We analyzed participant-level data from 12,503 patients and 12,502 controls, optimizing a risk model and assessing SNCA-specific risk scores and haplotypes as predictors of individual risk. We also explored hypotheses about functional mechanisms and correlated risk variants to gene expression in human brain and protein levels in cerebrospinal fluid. RESULTS We report and replicate a novel, third independent association signal at genome-wide significance level downstream of SNCA (rs2870004, p = 3.0*10-8 , odds ratio [OR] = 0.88, 95% confidence interval [CI] = 0.84-0.92). SNCA risk score stratification showed a 2-fold difference in disease susceptibility between top and bottom quintiles (OR = 1.99, 95% CI = 1.78-2.23). Contrary to previous reports, we provide evidence supporting top variant rs356182 as functional in itself and associated with a specific SNCA 5' untranslated region transcript isoform in frontal cortex. INTERPRETATION The SNCA locus harbors a minimum of 3 independent association signals for Parkinson disease. We demonstrate a fine-grained stratification of α-synuclein-related genetic burden in individual patients of potential future clinical relevance. Further efforts to pinpoint the functional mechanisms are warranted, including studies of the likely causal top variant rs356182 and its role in regulating levels of specific SNCA mRNA transcript variants. Ann Neurol 2018;83:117-129.
Collapse
Affiliation(s)
- Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Cornelis Blauwendraat
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD.,Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD
| | | | | | | | | | - Wilma D J van de Berg
- Department of Anatomy and Neurosciences, Clinical Neuroanatomy Section, Amsterdam Neuroscience, VU Medical Center, Amsterdam, the Netherlands
| | - J Raphael Gibbs
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD
| | - Mark R Cookson
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD
| | | | | | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD.,Data Tecnica International, Glen Echo, MD
| | - Mathias Toft
- Department of Neurology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
48
|
Chen JA, Chen Z, Won H, Huang AY, Lowe JK, Wojta K, Yokoyama JS, Bensimon G, Leigh PN, Payan C, Shatunov A, Jones AR, Lewis CM, Deloukas P, Amouyel P, Tzourio C, Dartigues JF, Ludolph A, Boxer AL, Bronstein JM, Al-Chalabi A, Geschwind DH, Coppola G. Joint genome-wide association study of progressive supranuclear palsy identifies novel susceptibility loci and genetic correlation to neurodegenerative diseases. Mol Neurodegener 2018; 13:41. [PMID: 30089514 PMCID: PMC6083608 DOI: 10.1186/s13024-018-0270-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 06/29/2018] [Indexed: 11/21/2022] Open
Abstract
Background Progressive supranuclear palsy (PSP) is a rare neurodegenerative disease for which the genetic contribution is incompletely understood. Methods We conducted a joint analysis of 5,523,934 imputed SNPs in two newly-genotyped progressive supranuclear palsy cohorts, primarily derived from two clinical trials (Allon davunetide and NNIPPS riluzole trials in PSP) and a previously published genome-wide association study (GWAS), in total comprising 1646 cases and 10,662 controls of European ancestry. Results We identified 5 associated loci at a genome-wide significance threshold P < 5 × 10− 8, including replication of 3 loci from previous studies and 2 novel loci at 6p21.1 and 12p12.1 (near RUNX2 and SLCO1A2, respectively). At the 17q21.31 locus, stepwise regression analysis confirmed the presence of multiple independent loci (localized near MAPT and KANSL1). An additional 4 loci were highly suggestive of association (P < 1 × 10− 6). We analyzed the genetic correlation with multiple neurodegenerative diseases, and found that PSP had shared polygenic heritability with Parkinson’s disease and amyotrophic lateral sclerosis. Conclusions In total, we identified 6 additional significant or suggestive SNP associations with PSP, and discovered genetic overlap with other neurodegenerative diseases. These findings clarify the pathogenesis and genetic architecture of PSP. Electronic supplementary material The online version of this article (10.1186/s13024-018-0270-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jason A Chen
- Interdepartmental Program in Bioinformatics, University of California, Los Angeles, CA, 90095, USA
| | - Zhongbo Chen
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, King's College London, London, SE5 9RX, UK
| | - Hyejung Won
- Program in Neurogenetics, Department of Neurology and Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Alden Y Huang
- Interdepartmental Program in Bioinformatics, University of California, Los Angeles, CA, 90095, USA
| | - Jennifer K Lowe
- Program in Neurogenetics, Department of Neurology and Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Kevin Wojta
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, 90095, USA
| | - Jennifer S Yokoyama
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Gilbert Bensimon
- BESPIM, CHU-Nîmes, Nîmes, France.,Dept Pharmacologie Clinique, Pitié-Salpêtrière Hospital, AP-PH, Paris, France.,Pharmacology UPMC-Paris VI, Universite Paris-Sorbonne, Paris, France
| | - P Nigel Leigh
- Trafford Centre for Biomedical Research, Brighton and Sussex Medical School, University of Sussex, Falmer, Brighton, UK
| | - Christine Payan
- BESPIM, CHU-Nîmes, Nîmes, France.,Dept Pharmacologie Clinique, Pitié-Salpêtrière Hospital, AP-PH, Paris, France
| | - Aleksey Shatunov
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, King's College London, London, SE5 9RX, UK
| | - Ashley R Jones
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, King's College London, London, SE5 9RX, UK
| | - Cathryn M Lewis
- Medical Research Council Social, Genetic and Developmental Psychiatry Centre, and Department of Medical and Molecular Genetics, King's College London, London, SE5 8AF, UK
| | - Panagiotis Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Philippe Amouyel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factor and molecular determinants of aging diseases, Labex-Distalz, F-59000, Lille, France
| | - Christophe Tzourio
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, CHU Bordeaux, F-33000 Bordeaux, France
| | - Jean-Francois Dartigues
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, CHU Bordeaux, F-33000 Bordeaux, France
| | - Albert Ludolph
- Department of Neurology, University of Ulm, Oberer Eselsberg, Ulm, Germany
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Jeff M Bronstein
- Program in Neurogenetics, Department of Neurology and Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Ammar Al-Chalabi
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, King's College London, London, SE5 9RX, UK
| | - Daniel H Geschwind
- Interdepartmental Program in Bioinformatics, University of California, Los Angeles, CA, 90095, USA.,Program in Neurogenetics, Department of Neurology and Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Giovanni Coppola
- Interdepartmental Program in Bioinformatics, University of California, Los Angeles, CA, 90095, USA. .,Program in Neurogenetics, Department of Neurology and Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA. .,Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, 90095, USA. .,Departments of Psychiatry and Neurology, David Geffen School of Medicine, University of California, Los Angeles, 695 Charles E Young Dr. South, Gonda Bldg, Rm 1524, Los Angeles, CA, 90095, USA.
| |
Collapse
|
49
|
Koros C, Stamelou M, Simitsi A, Beratis I, Papadimitriou D, Papagiannakis N, Fragkiadaki S, Kontaxopoulou D, Papageorgiou SG, Stefanis L. Selective cognitive impairment and hyposmia in p.A53T SNCA PD vs typical PD. Neurology 2018; 90:e864-e869. [DOI: 10.1212/wnl.0000000000005063] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 12/04/2017] [Indexed: 02/07/2023] Open
Abstract
ObjectiveTo evaluate nonmotor symptoms in early SNCA/p.A53T Parkinson disease (PD) (A53T PD) compared to typical PD (tPD).MethodsThe presence of hyposmia, neuropsychiatric, dysautonomic, and sleep disturbances was assessed by standardized questionnaires and validated scales in 18 patients with A53T PD and 18 patients with tPD, matched for age, sex, and disease duration. All patients were enrolled into the Parkinson's Progression Markers Initiative study.ResultsThe levodopa equivalent daily dose was higher in the A53T PD (p = 0.018) group vs the tPD group. Scores on the University of Pennsylvania Smell Identification Test (p = 0.001), Benton Judgement of Line Orientation test (p = 0.001), Letter Number Sequencing Test (p = 0.002), and phonemic verbal fluency (p = 0.002) were lower in the A53T PD group vs the tPD group. In contrast, overall cognition, verbal memory, and semantic fluency were similar between groups.ConclusionThe observed selective cognitive impairment reflecting frontal-parietal network dysfunction, together with impaired olfaction, define a set of nonmotor dysfunctions related to A53T PD. These results have implications for the prognosis of patients with A53T PD. Moreover, as the archetypal α-synucleinopathy, such results may give insights into tPD.
Collapse
|
50
|
Ibanez L, Dube U, Saef B, Budde J, Black K, Medvedeva A, Del-Aguila JL, Davis AA, Perlmutter JS, Harari O, Benitez BA, Cruchaga C. Parkinson disease polygenic risk score is associated with Parkinson disease status and age at onset but not with alpha-synuclein cerebrospinal fluid levels. BMC Neurol 2017; 17:198. [PMID: 29141588 PMCID: PMC5688622 DOI: 10.1186/s12883-017-0978-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 11/05/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The genetic architecture of Parkinson's Disease (PD) is complex and not completely understood. Multiple genetic studies to date have identified multiple causal genes and risk loci. Nevertheless, most of the expected genetic heritability remains unexplained. Polygenic risk scores (PRS) may provide greater statistical power and inform about the genetic architecture of multiple phenotypes. The aim of this study was to test the association between PRS and PD risk, age at onset and cerebrospinal fluid (CSF) biomarkers (α-synuclein, Aβ1-42, t-tau and p-tau). METHODS The weighted PRS was created using the genome-wide loci from Nalls et al., 2014 PD GWAs meta-analysis. The PRS was tested for association with PD status, age at onset and CSF biomarker levels in 829 cases and 432 controls of European ancestry. RESULTS The PRS was associated with PD status (p = 5.83×10-08) and age at onset (p = 5.70×10-07). The CSF t-tau levels showed a nominal association with the PRS (p = 0.02). However, CSF α-synuclein, amyloid beta and phosphorylated tau were not found to be associated with the PRS. CONCLUSION Our study suggests that there is an overlap in the genetic architecture of PD risk and onset, although the different loci present different weights for those phenotypes. In our dataset we found a marginal association of the PRS with CSF t-tau but not with α-synuclein CSF levels, suggesting that the genetic architecture for the CSF biomarker levels is different from that of PD risk.
Collapse
Affiliation(s)
- Laura Ibanez
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA.,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Umber Dube
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA.,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA.,Medical Scientist Training Program, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Benjamin Saef
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA.,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA
| | - John Budde
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA.,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Kathleen Black
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA.,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Alexandra Medvedeva
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA.,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Jorge L Del-Aguila
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA.,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Albert A Davis
- Department of Neurology, School of Medicine, Washington University in St Louis, Saint Louis, MO, USA
| | - Joel S Perlmutter
- Department of Neurology, School of Medicine, Washington University in St Louis, Saint Louis, MO, USA
| | - Oscar Harari
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA.,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Bruno A Benitez
- Department of Medicine, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA. .,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA.
| |
Collapse
|