1
|
Labrador-Espinosa MA, Silva-Rodríguez J, Mir P, Grothe MJ. Data-driven characterization of distinct cognitive subtypes in Parkinson's disease dementia. NPJ Parkinsons Dis 2025; 11:119. [PMID: 40346094 PMCID: PMC12064668 DOI: 10.1038/s41531-025-00970-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/17/2025] [Indexed: 05/11/2025] Open
Abstract
Individual cognitive profiles of patients with Parkinson's disease dementia (PDD) are highly heterogeneous, suggesting possible biological subtypes. We studied 75 PD patients who developed dementia in the course of the Parkinson's Progression Markers Initiative study to investigate data-driven evidence for the existence of distinct cognitive subtypes of PDD. Using Ward's hierarchical clustering on neuropsychological test data, we identified two distinct cognitive subtypes. Despite similar dementia severity (MoCA: 20.6 vs 20.0), cluster-A exhibited more pronounced memory deficits (n = 50), whereas cluster-B showed greater visuospatial impairments (n = 25). The subtypes did not differ in demographic, motor, or MRI-based neurodegeneration measures. However, the visuospatial-predominant cluster-B had a higher prevalence of GBA mutations (p = 0.003) and hallucinations (p = 0.009). No differences were found in APOE-ε4 prevalence or cerebrospinal fluid biomarkers of Alzheimer's pathology. These findings reveal distinct memory-predominant and visuospatial-predominant PDD subtypes, which associate with different clinical and genetic features but are independent of comorbid Alzheimer's pathology.
Collapse
Affiliation(s)
- Miguel A Labrador-Espinosa
- Department of Psychiatry and Neurochemistry, Institute of Physiology and Neuroscience, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | | | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain.
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Seville, Spain.
| | - Michel J Grothe
- Reina Sofia Alzheimer Center, CIEN Foundation, ISCIII, Madrid, Spain.
| |
Collapse
|
2
|
Martín-Bórnez M, Shar N, Nour MA, Murphy D, Elsayed I, Megha SN, Nwaokorie F, Olusanya A, Kuznetsov N, Bandres-Ciga S, Noyce AJ, Iwaki H, Jones L, Gómez-Garre P, Mir P, Periñan MT. Does COMT Play a Role in Parkinson's Disease Susceptibility Across Diverse Ancestral Populations? MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.11.25325572. [PMID: 40297458 PMCID: PMC12036390 DOI: 10.1101/2025.04.11.25325572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Background The catechol-O-methyltransferase (COMT) gene is involved in brain catecholamine metabolism, but its association with Parkinson's disease (PD) risk remains unclear. Objective To investigate the relationship between COMT genetic variants and PD risk across diverse ancestries. Methods We analyzed COMT variants in 2,251 PD patients and 2,835 controls of European descent using whole-genome sequencing from the Accelerating Medicines Partnership-Parkinson Disease (AMP-PD), along with 20,427 PD patients and 11,837 controls from 10 ancestries using genotyping data from the Global Parkinson's Genetics Program (GP2). Results Utilizing the largest case-control datasets to date, no significant enrichment of COMT risk alleles in PD patients was observed across any ancestry group after correcting for multiple testing. Among Europeans, no correlations with cognitive decline, motor function, motor complications, or time to LID onset were observed. Conclusions These findings emphasize the need for larger, diverse cohorts to confirm the role of COMT in PD development and progression.
Collapse
Affiliation(s)
- Miguel Martín-Bórnez
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Nisar Shar
- Department of clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- NED University of Engineering & Technology, Karachi, Pakistan
| | | | - David Murphy
- Department of clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Inas Elsayed
- Faculty of Pharmacy, University of Gezira, Wadmedani, Sudan
- Systems Biology Ireland, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Shri N Megha
- Department of Neurology, National Institute of Mental Health and Neurosciences, India
| | - Francisca Nwaokorie
- Department of Medical Laboratory Science, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, Nigeria
| | - Adedunni Olusanya
- Department of Pharmacology, Therapeutics and Toxicology, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, Nigeria
| | - Nicole Kuznetsov
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica, Washington DC, USA
| | - Sara Bandres-Ciga
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Alastair J Noyce
- Center for Preventive Neurology, Wolfson Institute of Population Health, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Hirotaka Iwaki
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica, Washington DC, USA
| | - Lietsel Jones
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica, Washington DC, USA
| | - Pilar Gómez-Garre
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Maria Teresa Periñan
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
- Center for Preventive Neurology, Wolfson Institute of Population Health, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
3
|
Tong S, Wang R, Li H, Tong Z, Geng D, Zhang X, Ren C. Executive dysfunction in Parkinson's disease: From neurochemistry to circuits, genetics and neuroimaging. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111272. [PMID: 39880275 DOI: 10.1016/j.pnpbp.2025.111272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/18/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
Cognitive decline is one of the most significant non-motor symptoms of Parkinson's disease (PD), with executive dysfunction (EDF) being the most prominent characteristic of PD-associated cognitive deficits. Currently, lack of uniformity in the conceptualization and assessment scales for executive functions impedes the early and accurate diagnosis of EDF in PD. The neurobiological mechanisms of EDF in PD remain poorly understood. Moreover, the treatment of cognitive impairment in PD has progressed slowly and with limited efficacy. Thus, this review explores the characteristics and potential mechanisms of EDF in PD from multiple perspectives, including the concept of executive function, commonly used neuropsychological tests, neurobiochemistry, genetics, electroencephalographic activity and neuroimaging. The available evidence indicates that degeneration of the frontal-striatal circuit, along with mutations in the Catechol-O-methyltransferase (COMT) gene and Leucine-rich repeat kinase 2 (LRRK2) gene, may contribute to EDF in patients with PD. The increase in theta and delta waves, along with the decrease in alpha waves, offers potential biomarkers for the early identification and monitoring of EDF, as well as the development of dementia in patients with PD. The PD cognition-related pattern (PDCP) pattern may serve as a tool for monitoring and assessing cognitive function progression in these patients and is anticipated to become a biomarker for cognitive disorders associated with PD. The aim is to provide new insights for the early and precise diagnosis and treatment of EDF in PD.
Collapse
Affiliation(s)
- Shuyan Tong
- Department of Geriatric Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China; Department of Neurology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ruiwen Wang
- Department of Anesthesiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Huihua Li
- Department of Psychiatry, Zhenjiang Mental Health Center, Zhenjiang, Jiangsu, China
| | - Zhu Tong
- The Affiliated Xuzhou Oriental Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiangrong Zhang
- Department of Geriatric Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China; The Affiliated Xuzhou Oriental Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Chao Ren
- Department of Neurology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China.
| |
Collapse
|
4
|
Rosal AE, Martin SL, Strafella AP. The role of Apolipoprotein E4 on cognitive impairment in Parkinson's disease and Parkinsonisms. Front Neurosci 2025; 19:1515374. [PMID: 40052092 PMCID: PMC11882537 DOI: 10.3389/fnins.2025.1515374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Cognitive impairment is a prevalent non-motor symptom of Parkinson's disease (PD), increasing the risk of dementia as the disease progresses. Despite its clinical significance, the etiology of cognitive impairment in PD remains unclear. Apolipoprotein E4 (APOE4), a well-known genetic risk factor of Alzheimer's disease, has been studied for its potential role in PD-related cognitive impairment. However, findings have been conflicting and thus inconclusive, highlighting a need to critically evaluate the current research. Several studies using neuroimaging modalities have explored the brains of individuals with PD and atypical parkinsonian disorders who have APOE4. Some of these studies have identified distinct neuropathological changes that have been previously reported to be associated with cognitive impairments in those with Parkinsonisms. Here, we review the role of APOE4 on cognitive impairment in PD and atypical Parkinsonisms using neuroimaging evidence. We will examine how APOE4 may contribute to pathological changes within the brain and its association with cognitive impairment.
Collapse
Affiliation(s)
- Angenelle Eve Rosal
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Sarah L. Martin
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Translation and Computational Neurosciences Unit (TCNU), Faculty of Health and Education, Manchester Metropolitan University, Manchester, United Kingdom
| | - Antonio P. Strafella
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Edmond J. Safra Parkinson Disease Program, Neurology Division, Toronto Western Hospital and Krembil Brain Institute, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
5
|
Poplawska-Domaszewicz K, Qamar MA, Falup Pecurariu C, Chaudhuri KR. Recognition and characterising non-motor profile in early onset Parkinson's disease (EOPD). Parkinsonism Relat Disord 2024; 129:107123. [PMID: 39489627 DOI: 10.1016/j.parkreldis.2024.107123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/15/2024] [Accepted: 08/31/2024] [Indexed: 11/05/2024]
Abstract
Early onset Parkinson's disease (EOPD) has been recently defined as a clinical entity with subjects presenting with Parkinson's disease (PD) between the ages of 21-50 and replaces the term Young Onset PD (YOPD). Presentations in this age group are somewhat different to the typical Late Onset sporadic PD (LOPD) and genetic basis may play an important role. The presentations are however, to be differentiated from other causes of juvenile onset or early onset parkinsonism, which are often driven by rare genetic, brain metal deposition, or metabolic progressive disorders with a levolevodopa unresponsive or poorly responsive phenotype. Specific genetic mutations can also underpin EOPD and include nonmotor symptoms of EOPD, which have not been studied extensively. However, some real-life comparator studies with LOPD suggest a nonmotor profile in EOPD dominated by neuropsychiatric symptoms (anxiety), pain, sexual dysfunction, and a higher risk of impulse control disorders and segregation to the recently described noradrenergic and Park-sleep nonmotor endophenotypes may occur. Awareness of the phenotypic variants and nonmotor expression will pave the way for future precision and personalised medicine.
Collapse
Affiliation(s)
- Karolina Poplawska-Domaszewicz
- Department of Neurology, Poznan University of Medical Sciences, 60-355, Poznan, Poland; Parkinson's Foundation Centre of Excellence, King's College Hospital, Denmark Hill, London, SE5 9RS, UK
| | - Mubasher A Qamar
- Parkinson's Foundation Centre of Excellence, King's College Hospital, Denmark Hill, London, SE5 9RS, UK; Basic and Clinical Neuroscience Department, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London SE5 9RX, UK
| | - Cristian Falup Pecurariu
- Parkinson's Foundation Centre of Excellence, King's College Hospital, Denmark Hill, London, SE5 9RS, UK; Department of Neurology, County Clinic Hospital, Faculty of Medicine, Transilvania University Brasov, 500019, Brasov, Romania; Department of Neurology, Transilvania University Brasov, Brașov, Romania
| | - K Ray Chaudhuri
- Parkinson's Foundation Centre of Excellence, King's College Hospital, Denmark Hill, London, SE5 9RS, UK; Basic and Clinical Neuroscience Department, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London SE5 9RX, UK.
| |
Collapse
|
6
|
Khalil I, Sayad R, Kedwany AM, Sayed HH, Caprara ALF, Rissardo JP. Cardiovascular dysautonomia and cognitive impairment in Parkinson's disease (Review). MEDICINE INTERNATIONAL 2024; 4:70. [PMID: 39355336 PMCID: PMC11443310 DOI: 10.3892/mi.2024.194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/03/2024] [Indexed: 10/03/2024]
Abstract
Cognitive impairment is a prevalent non-motor symptom of Parkinson's disease (PD), which can result in significant disability and distress for patients and caregivers. There is a marked variation in the timing, characteristics and rate at which cognitive decline occurs in patients with PD. This decline can vary from normal cognition to mild cognitive impairment and dementia. Cognitive impairment is associated with several pathophysiological mechanisms, including the accumulation of β-amyloid and tau in the brain, oxidative stress and neuroinflammation. Cardiovascular autonomic dysfunctions are commonly observed in patients with PD. These dysfunctions play a role in the progression of cognitive impairment, the incidents of falls and even in mortality. The majority of symptoms of dysautonomia arise from changes in the peripheral autonomic nervous system, including both the sympathetic and parasympathetic nervous systems. Cardiovascular changes, including orthostatic hypotension, supine hypertension and abnormal nocturnal blood pressure (BP), can occur in both the early and advanced stages of PD. These changes tend to increase as the disease advances. The present review aimed to describe the cognitive changes in the setting of cardiovascular dysautonomia and to discuss strategies through which these changes can be modified and managed. It is a multifactorial process usually involving decreased blood flow to the brain, resulting in the development of cerebral ischemic lesions, an increased presence of abnormal white matter signals in the brain, and a potential influence on the process of neurodegeneration in PD. Another possible explanation is this association being independent observations of PD progression. Patients with clinical symptoms of dysautonomia should undergo 24-h ambulatory BP monitoring, as they are frequently subtle and underdiagnosed.
Collapse
Affiliation(s)
- Ibrahim Khalil
- Faculty of Medicine, Alexandria University, Alexandria 5372066, Egypt
| | - Reem Sayad
- Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | | | - Hager Hamdy Sayed
- Department of Nuclear Medicine, Assuit University, Assuit 71515, Egypt
| | | | | |
Collapse
|
7
|
Li C, Hou Y, Ou R, Wei Q, Zhang L, Liu K, Lin J, Chen X, Song W, Zhao B, Wu Y, Shang H. GWAS Identifies DPP6 as Risk Gene of Cognitive Decline in Parkinson's Disease. J Gerontol A Biol Sci Med Sci 2024; 79:glae155. [PMID: 38875006 PMCID: PMC11272050 DOI: 10.1093/gerona/glae155] [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: 02/26/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Cognitive decline is among the most common non-motor symptoms in Parkinson's disease (PD), while its physiological mechanisms remain poorly understood. Genetic factors constituted a fundamental determinant in the heterogeneity of cognitive decline among PD patients. However, the underlying genetic background was still less studied. METHODS To explore the genetic determinants contributing to cognitive decline in PD, we performed genome-wide survival analysis using a Cox proportional hazards model in a longitudinal cohort of 450 Chinese patients with PD, and further explored the functional effect of the target variant. Additionally, we built a clinical-genetic model by incorporating clinical characteristics and polygenic risk score (PRS) to predict cognitive decline in PD. RESULTS The cohort was followed up for an average of 5.25 (SE = 2.46) years, with 95 incidents of cognitive impairment. We identified significant association between locus rs75819919 (DPP6) and accelerated cognitive decline (p = 8.63E-09, beta = 1.74, SE = 0.30). Dual-luciferase reporter assay suggested this locus might be involved in the regulation of DPP6 expression. Using data set from the UK Biobank, we identified rs75819919 was associated with cognitive performance in the general population. Incorporation of PRS increased the model's predictability, achieving an average AUC of 75.6% through fivefold cross-validation in 1 000 iterations. CONCLUSIONS These findings improve the current understanding of the genetic etiology of cognitive impairment in PD, and provide a novel target DPP6 to explore therapeutic options. Our results also demonstrate the potential to develop clinical-genetic model to identify patients susceptible to cognitive impairment and thus provide personalized clinical guidance.
Collapse
Affiliation(s)
- Chunyu Li
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yanbing Hou
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Ruwei Ou
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Qianqian Wei
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Lingyu Zhang
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Kuncheng Liu
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Junyu Lin
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xueping Chen
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Song
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Bi Zhao
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Wu
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Huifang Shang
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Wang Q, Gu X, Yang L, Jiang Y, Zhang J, He J. Emerging perspectives on precision therapy for Parkinson's disease: multidimensional evidence leading to a new breakthrough in personalized medicine. Front Aging Neurosci 2024; 16:1417515. [PMID: 39026991 PMCID: PMC11254646 DOI: 10.3389/fnagi.2024.1417515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
PD is a prevalent and progressive neurodegenerative disorder characterized by both motor and non-motor symptoms. Genes play a significant role in the onset and progression of the disease. While the complexity and pleiotropy of gene expression networks have posed challenges for gene-targeted therapies, numerous pathways of gene variant expression show promise as therapeutic targets in preclinical studies, with some already in clinical trials. With the recognition of the numerous genes and complex pathways that can influence PD, it may be possible to take a novel approach to choose a treatment for the condition. This approach would be based on the symptoms, genomics, and underlying mechanisms of the disease. We discuss the utilization of emerging genetic and pathological knowledge of PD patients to categorize the disease into subgroups. Our long-term objective is to generate new insights for the therapeutic approach to the disease, aiming to delay and treat it more effectively, and ultimately reduce the burden on individuals and society.
Collapse
Affiliation(s)
- Qiaoli Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xuan Gu
- Department of Trauma center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Le Yang
- Department of Endocrinology, The People’s Hospital of Jilin Province, Changchun, China
| | - Yan Jiang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiao Zhang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jinting He
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
9
|
Cao LX, Kong WL, Chan P, Zhang W, Morris MJ, Huang Y. Assessment tools for cognitive performance in Parkinson's disease and its genetic contributors. Front Neurol 2024; 15:1413187. [PMID: 38988604 PMCID: PMC11233456 DOI: 10.3389/fneur.2024.1413187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/14/2024] [Indexed: 07/12/2024] Open
Abstract
Background We have shown that genetic factors associating with motor progression of Parkinson's disease (PD), but their roles in cognitive function is poorly understood. One reason is that while cognitive performance in PD can be evaluated by various cognitive scales, there is no definitive guide indicating which tool performs better. Methods Data were obtained from the Parkinson's Progression Markers Initiative, where cognitive performance was assessed using five cognitive screening tools, including Symbol Digit Modalities Test (SDMT), Montreal Cognitive Assessment, Benton Judgment of Line Orientation, Modified Semantic Fluency Test, and Letter Number Sequencing Test, at baseline and subsequent annual follow-up visit for 5 years. Genetic data including ApoE and other PD risk genetic information were also obtained. We used SPSS-receiver operating characteristic and ANOVA repeated measures to evaluate which cognitive assessment is the best reflecting cognitive performance in PD at early stage and over time. Logistic regression analyses were used to determine the genetic associations with the rapidity of cognitive decline in PD. Results SDMT performed better in detecting mild cognitive impairment at baseline (AUC = 0.763), and SDMT was the only tool showing a steady cognitive decline during longitudinal observation. Multigenetic factors significantly associated with cognitive impairment at early stage of the disease (AUC = 0.950) with IP6K2 rs12497850 more evident, and a significantly faster decline (AUC = 0.831) within 5 years after motor onset, particularly in those carrying FGF20 rs591323. Conclusion SDMT is a preferable cognitive assessment tool for PD and genetic factors synergistically contribute to the cognitive dysfunction in PD.
Collapse
Affiliation(s)
- Ling-Xiao Cao
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wee Lee Kong
- Pharmacology Department, School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Piu Chan
- Department of Neurobiology, Neurology and Geriatrics, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wei Zhang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Margaret J. Morris
- Pharmacology Department, School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Yue Huang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Pharmacology Department, School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
10
|
Faouzi J, Tan M, Casse F, Lesage S, Tesson C, Brice A, Mangone G, Mariani LL, Iwaki H, Colliot O, Pihlstrøm L, Corvol JC. Proxy-analysis of the genetics of cognitive decline in Parkinson's disease through polygenic scores. NPJ Parkinsons Dis 2024; 10:8. [PMID: 38177146 PMCID: PMC10767119 DOI: 10.1038/s41531-023-00619-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 12/08/2023] [Indexed: 01/06/2024] Open
Abstract
Cognitive decline is common in Parkinson's disease (PD) and its genetic risk factors are not well known to date, besides variants in the GBA and APOE genes. However, variation in complex traits is caused by numerous variants and is usually studied with genome-wide association studies (GWAS), requiring a large sample size, which is difficult to achieve for outcome measures in PD. Taking an alternative approach, we computed 100 polygenic scores (PGS) related to cognitive, dementia, stroke, and brain anatomical phenotypes and investigated their association with cognitive decline in six longitudinal cohorts. The analysis was adjusted for age, sex, genetic ancestry, follow-up duration, GBA and APOE status. Then, we meta-analyzed five of these cohorts, comprising a total of 1702 PD participants with 6156 visits, using the Montreal Cognitive Assessment as a cognitive outcome measure. After correction for multiple comparisons, we found four PGS significantly associated with cognitive decline: intelligence (p = 5.26e-13), cognitive performance (p = 1.46e-12), educational attainment (p = 8.52e-10), and reasoning (p = 3.58e-5). Survival analyses highlighted an offset of several years between the first and last quartiles of PGS, with significant differences for the PGS of cognitive performance (5 years) and educational attainment (7 years). In conclusion, we found four PGS associated with cognitive decline in PD, all associated with general cognitive phenotypes. This study highlights the common genetic factors between cognitive decline in PD and the general population, and the importance of the participant's cognitive reserve for cognitive outcome in PD.
Collapse
Affiliation(s)
- Johann Faouzi
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, F-75013, Paris, France
- Univ Rennes, Ensai, CNRS, CREST-UMR 9194, F-35000, Rennes, France
| | - Manuela Tan
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Fanny Casse
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Suzanne Lesage
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Christelle Tesson
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Alexis Brice
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, DMU Neurosciences, Département de Génétique, F-75013, Paris, France
| | - Graziella Mangone
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, DMU Neurosciences, Département de Neurologie, F-75013, Paris, France
- Department of Neurology, Movement Disorder Division, Rush University Medical Center, 1725 W. Harrison Street, Chicago, IL, 60612, USA
| | - Louise-Laure Mariani
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, DMU Neurosciences, Département de Neurologie, F-75013, Paris, France
| | - Hirotaka Iwaki
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International LLC, Washington, DC, USA
| | - Olivier Colliot
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, F-75013, Paris, France
| | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Jean-Christophe Corvol
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, DMU Neurosciences, Département de Neurologie, F-75013, Paris, France.
| |
Collapse
|
11
|
Lin J, Ou R, Li C, Hou Y, Zhang L, Wei Q, Liu K, Jiang Q, Yang T, Xiao Y, Pang D, Zhao B, Chen X, Yang J, Shang H. Evolution and Predictive Role of Plasma Alzheimer's Disease-related Pathological Biomarkers in Parkinson's Disease. J Gerontol A Biol Sci Med Sci 2023; 78:2203-2213. [PMID: 37560912 DOI: 10.1093/gerona/glad189] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Indexed: 08/11/2023] Open
Abstract
Plasma Alzheimer's disease-related pathological biomarkers' role in Parkinson's disease (PD) remains unknown. We aimed to determine whether plasma Alzheimer's disease-related biomarkers can predict PD progression. A total of 184 PD patients and 86 healthy controls were included and followed up for 5 years. Plasma phosphorylated tau181 (p-tau181), Aβ40, and Aβ42 were measured at baseline and the 1- and 2-year follow-ups using the Quanterix-single-molecule array. Global cognitive function and motor symptoms were assessed using the Montreal Cognitive Assessment and Unified Parkinson's Disease Rating Scale part III. Genetic analyses were conducted to identify APOE and MAPT genotypes. Plasma p-tau181 levels were higher in PD than healthy controls. APOE-ε4 carriers had lower plasma Aβ42 levels and Aβ42/Aβ40 ratio. The linear mixed-effects models showed that Montreal Cognitive Assessment scores were associated with plasma p-tau181/Aβ42 ratio (β -1.719 [-3.398 to -0.040], p = .045). Higher baseline plasma p-tau181 correlated with faster cognitive decline and motor symptoms deterioration in total patients (β -0.170 [-0.322 to -0.018], p = .029; β 0.329 [0.032 to 0.626], p = .030) and APOE-ε4 carriers (β -0.318 [-0.602 to -0.034], p = .030; β 0.632 [0.017 to 1.246], p = .046), but not in the noncarriers. Higher baseline plasma Aβ40 correlated with faster cognitive decline in total patients (β -0.007 [-0.015 to -0.0001], p = .047) and faster motor symptoms deterioration in total patients (β 0.026 [0.010 to 0.041], p = .001) and APOE-ε4 carriers (β 0.044 [-0.026 to 0.049], p = .020), but not in the noncarriers. The plasma p-tau181/Aβ2 ratio monitors the cognitive status of PD. Higher baseline plasma p-tau181 and Aβ40 predict faster cognitive decline and motor symptoms deterioration in PD, especially in APOE-ε4 carriers.
Collapse
Affiliation(s)
- Junyu Lin
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruwei Ou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chunyu Li
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanbing Hou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lingyu Zhang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qianqian Wei
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kuncheng Liu
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qirui Jiang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tianmi Yang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Xiao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dejiang Pang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bi Zhao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xueping Chen
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jing Yang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huifang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Paslawski W, Svenningsson P. Elevated ApoE, ApoJ and lipoprotein-bound α-synuclein levels in cerebrospinal fluid from Parkinson's disease patients - Validation in the BioFIND cohort. Parkinsonism Relat Disord 2023; 116:105765. [PMID: 37479568 PMCID: PMC11140586 DOI: 10.1016/j.parkreldis.2023.105765] [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: 03/17/2023] [Revised: 06/12/2023] [Accepted: 07/11/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND The progressive accumulation, aggregation, and spread of α-synuclein (aSN) are common hallmarks of Parkinson's disease (PD) pathology. The genotype of apolipoprotein E (ApoE) influences PD progression. Recently we found that aSN co-localize with apolipoproteins on lipoprotein vesicles. We reported an increased level of ApoE, ApoJ and lipoprotein-bound aSN in CSF from early PD patients compared to matched controls. We also found reduced plasma ApoAI in PD patients. OBJECTIVE In this study, we used the same approach on the BioFIND cohort to validate our previous results and extended the studies to examine correlations with ApoE genotype, demographic variables, clinical symptoms and other biochemical findings reported in the BioFIND cohort. METHODS For the assessment, we used Western-Blot (WB) technique for apolipoproteins measurements in CSF and plasma from PD patients and healthy controls. Further, for measurement of aSN bound to lipoproteins, we combined immunodepletion with the enzyme-linked immunosorbent assay (ELISA). RESULTS Levels of ApoE, ApoJ and lipoprotein bound aSN were significantly increased in CSF from PD patients compared to controls. We also observed decreased levels of ApoAI and ApoJ in plasma from PD patients compared to controls. CONCLUSIONS Concluding, the present data validated our previous findings. Altered lipoproteins appear to be important in early PD pathology and may be involved in mechanisms underlying aSN cell-to-cell transfer in the nervous system and be developed in algorithms for early diagnosis of PD.
Collapse
Affiliation(s)
- Wojciech Paslawski
- Laboratory of Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Per Svenningsson
- Laboratory of Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
| |
Collapse
|
13
|
Lloyd GM, Long B, Quintin S, Sorrentino ZA, Gorion KMM, Bell BM, Carrillo D, Sullivan P, Borchelt D, Giasson BI. Carboxyl truncation of α-synuclein occurs early and is influenced by human APOE genotype in transgenic mouse models of α-synuclein pathogenesis. Acta Neuropathol Commun 2023; 11:119. [PMID: 37482615 PMCID: PMC10363304 DOI: 10.1186/s40478-023-01623-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/15/2023] [Indexed: 07/25/2023] Open
Abstract
Post-translational modifications to the carboxyl (C) terminus domain of α-synuclein can play an important role in promoting the pathologic aggregation of α-synuclein. Various cleavages that diminish this highly charged, proline-rich region can result in exposure of hydrophobic, aggregation-prone regions, thereby accelerating the aggregation kinetics of α-synuclein into misfolded, pathologic forms. C-terminally truncated forms of α-synuclein are abundant in human diseased brains compared to controls, suggesting a role in disease pathogenesis. Factors that alter the homeostatic proteolytic processing of α-synuclein may ultimately tip the balance towards a progressive disease state. Apolipoprotein E (APOE) has been implicated in the acceleration of cognitive impairment in patients with Lewy body diseases. The APOE4 isoform has been found to cause dysregulation in the endosomal-lysosomal pathway, which could result in altered α-synuclein degradation as a potential mechanism for promoting its pathologic misfolding. Herein, we investigate the spatiotemporal accumulation of C-terminally truncated α-synuclein in a seeded and progressive mouse model of synucleinopathy. Furthermore, we study how this process is influenced in the context of mice that are altered to express either the human APOE3 or APOE4 isoforms. We found that specific C-terminal truncation of α-synuclein occurs at early stages of pathogenesis. We also found that proteolytic processing of this domain differs across various brain regions and is influenced by the presence of different human APOE isoforms. Our data demonstrate an early pathogenic role for C-terminally truncated α-synuclein, and highlight the influence of APOE isoforms in modulating its impact.
Collapse
Affiliation(s)
- Grace M Lloyd
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Brooke Long
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
| | - Stephan Quintin
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Zachary A Sorrentino
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Kimberly-Marie M Gorion
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Brach M Bell
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Denise Carrillo
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
| | - Patrick Sullivan
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - David Borchelt
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Benoit I Giasson
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA.
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
14
|
Liu T, Zuo H, Ma D, Song D, Zhao Y, Cheng O. Cerebrospinal fluid GFAP is a predictive biomarker for conversion to dementia and Alzheimer's disease-associated biomarkers alterations among de novo Parkinson's disease patients: a prospective cohort study. J Neuroinflammation 2023; 20:167. [PMID: 37475029 PMCID: PMC10357612 DOI: 10.1186/s12974-023-02843-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/27/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Dementia is a prevalent non-motor manifestation among individuals with advanced Parkinson's disease (PD). Glial fibrillary acidic protein (GFAP) is an inflammatory marker derived from astrocytes. Research has demonstrated the potential of plasma GFAP to forecast the progression to dementia in PD patients with mild cognitive impairment (PD-MCI). However, the predictive role of cerebrospinal fluid (CSF) GFAP on future cognitive transformation and alterations in Alzheimer's disease (AD)-associated CSF biomarkers in newly diagnosed PD patients has not been investigated. METHODS 210 de novo PD patients from the Parkinson's Progression Markers Initiative were recruited. Cognitive progression in PD participants was evaluated using Cox regression. Cross-sectional and longitudinal associations between baseline CSF GFAP and cognitive function and AD-related CSF biomarkers were evaluated using multiple linear regression and generalized linear mixed model. RESULTS At baseline, the mean age of PD participants was 60.85 ± 9.78 years, including 142 patients with normal cognition (PD-NC) and 68 PD-MCI patients. The average follow-up time was 6.42 ± 1.69 years. A positive correlation was observed between baseline CSF GFAP and age (β = 0.918, p < 0.001). There was no statistically significant difference in baseline CSF GFAP levels between PD-NC and PD-MCI groups. Higher baseline CSF GFAP predicted greater global cognitive decline over time in early PD patients (Montreal Cognitive Assessment, β = - 0.013, p = 0.014). Furthermore, Cox regression showed that high baseline CSF GFAP levels were associated with a high risk of developing dementia over an 8-year period in the PD-NC group (adjusted HR = 3.070, 95% CI 1.119-8.418, p = 0.029). In addition, the baseline CSF GFAP was positively correlated with the longitudinal changes of not only CSF α-synuclein (β = 0.313, p < 0.001), but also CSF biomarkers associated with AD, namely, amyloid-β 42 (β = 0.147, p = 0.034), total tau (β = 0.337, p < 0.001) and phosphorylated tau (β = 0.408, p < 0.001). CONCLUSIONS CSF GFAP may be a valuable prognostic tool that can predict the severity and progression of cognitive deterioration, accompanied with longitudinal changes in AD-associated pathological markers in early PD.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Hongzhou Zuo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Di Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Dan Song
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Yuying Zhao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Oumei Cheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| |
Collapse
|
15
|
Ghanem A, Berry DS, Farrell K, Cosentino S, Crary JF, Louis ED. Cognitive Performance as a Function of MAPT Haplotype: A Prospective Longitudinal Study of an Essential Tremor Cohort. Tremor Other Hyperkinet Mov (N Y) 2023; 13:19. [PMID: 37214541 PMCID: PMC10198222 DOI: 10.5334/tohm.768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/20/2023] [Indexed: 05/24/2023] Open
Abstract
Background Cognitive impairment is a feature of essential tremor (ET). There are no studies of the genetic drivers of this association. We examined whether the microtubule-associated protein tau (MAPT) H1 haplotype is associated with cognitive performance in ET. Methods ET cases genotyped for the MAPT H1 and H2 haplotypes completed a battery of neuropsychological tests at baseline and four follow-up evaluations. Chi-square, t-tests, and analyses of covariance examined associations between the presence of the MAPT H1 haplotype, cognitive diagnoses of normal, mild cognitive impairment (MCI), and dementia, and performance in specific cognitive domains. Results We observed no evidence of cognitive differences as a function of the presence of the MAPT H1 haplotype. Specifically, cases with (n = 57) and without (n = 42) this haplotype did not differ with respect to the prevalence of diagnoses of MCI or dementia, p ≥ 0.87. Moreover, cases with vs without this haplotype did not differ in either the age or point in the disease course at which observed conversions to MCI or dementia occurred, p's ≥ 0.51. Finally, no haplotype-related differences were observed in performance in the cognitive domains of attention, executive function, language, memory, visuospatial or global ability, p's ≥ 0.21, or in changes in performance in these domains across time, p's ≥ 0.08. Discussion The study in an ET cohort revealed no influence of MAPT haplotypes on cognitive performance. This study serves as a valuable foundation for future studies to expand our understanding of the genetic drivers of cognitive impairment in ET. Highlights This study found no evidence of cognitive differences between individuals with and without the MAPT H1 haplotype. Our work provides a valuable foundation for future work to expand our knowledge of the genetic drivers of cognitive impairment in ET.
Collapse
Affiliation(s)
- Ali Ghanem
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Diane S. Berry
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kurt Farrell
- Department of Pathology, Department of Artificial Intelligence & Human Health, Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer’s Disease, Friedman Brain Institute, Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Stephanie Cosentino
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, New York, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - John F. Crary
- Department of Pathology, Department of Artificial Intelligence & Human Health, Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer’s Disease, Friedman Brain Institute, Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Elan D. Louis
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
16
|
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
|
17
|
Dulski J, Uitti RJ, Ross OA, Wszolek ZK. Genetic architecture of Parkinson’s disease subtypes – Review of the literature. Front Aging Neurosci 2022; 14:1023574. [PMID: 36337703 PMCID: PMC9632166 DOI: 10.3389/fnagi.2022.1023574] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/03/2022] [Indexed: 11/13/2022] Open
Abstract
The heterogeneity of Parkinson’s disease (PD) has been recognized since its description by James Parkinson over 200 years ago. The complexity of motor and non-motor PD manifestations has led to many attempts of PD subtyping with different prognostic outcomes; however, the pathophysiological foundations of PD heterogeneity remain elusive. Genetic contributions to PD may be informative in understanding the underpinnings of PD subtypes. As such, recognizing genotype-phenotype associations may be crucial for successful gene therapy. We review the state of knowledge on the genetic architecture underlying PD subtypes, discussing the monogenic forms, as well as oligo- and polygenic risk factors associated with various PD subtypes. Based on our review, we argue for the unification of PD subtyping classifications, the dichotomy of studies on genetic factors and genetic modifiers of PD, and replication of results from previous studies.
Collapse
Affiliation(s)
- Jarosław Dulski
- Department of Neurology, Mayo Clinic, Jacksonville, FL, United States
- Division of Neurological and Psychiatric Nursing, Faculty of Health Sciences, Medical University of Gdańsk, Gdańsk, Poland
- Department of Neurology, St. Adalbert Hospital, Copernicus PL Ltd., Gdańsk, Poland
| | - Ryan J. Uitti
- Department of Neurology, Mayo Clinic, Jacksonville, FL, United States
| | - Owen A. Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Zbigniew K. Wszolek
- Department of Neurology, Mayo Clinic, Jacksonville, FL, United States
- *Correspondence: Zbigniew K. Wszolek,
| |
Collapse
|
18
|
Li J, Yi M, Li B, Yin S, Zhang Y, Huang Z, Shu L, Zhang Y. Polymorphism of neurodegeneration-related genes associated with Parkinson’s disease risk. Neurol Sci 2022; 43:5301-5312. [DOI: 10.1007/s10072-022-06192-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/29/2022] [Indexed: 12/23/2022]
|
19
|
Hill EJ, Robak LA, Al-Ouran R, Deger J, Fong JC, Vandeventer PJ, Schulman E, Rao S, Saade H, Savitt JM, von Coelln R, Desai N, Doddapaneni H, Salvi S, Dugan-Perez S, Muzny DM, McGuire AL, Liu Z, Gibbs RA, Shaw C, Jankovic J, Shulman LM, Shulman JM. Genome Sequencing in the Parkinson Disease Clinic. Neurol Genet 2022; 8:e200002. [PMID: 35747619 PMCID: PMC9210549 DOI: 10.1212/nxg.0000000000200002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 04/22/2022] [Indexed: 11/18/2022]
Abstract
Background and Objectives Genetic variants affect both Parkinson disease (PD) risk and manifestations. Although genetic information is of potential interest to patients and clinicians, genetic testing is rarely performed during routine PD clinical care. The goal of this study was to examine interest in comprehensive genetic testing among patients with PD and document reactions to possible findings from genome sequencing in 2 academic movement disorder clinics. Methods In 203 subjects with PD (age = 63 years, 67% male), genome sequencing was performed and filtered using a custom panel, including 49 genes associated with PD, parkinsonism, or related disorders, as well as a 90-variant PD genetic risk score. Based on the results, 231 patients (age = 67 years, 63% male) were surveyed on interest in genetic testing and responses to vignettes covering (1) familial risk of PD (LRRK2); (2) risk of PD dementia (GBA); (3) PD genetic risk score; and (4) secondary, medically actionable variants (BRCA1). Results Genome sequencing revealed a LRRK2 variant in 3% and a GBA risk variant in 10% of our clinical sample. The genetic risk score was normally distributed, identifying 41 subjects with a high risk of PD. Medically actionable findings were discovered in 2 subjects (1%). In our survey, the majority (82%) responded that they would share a LRRK2 variant with relatives. Most registered unchanged or increased interest in testing when confronted with a potential risk for dementia or medically actionable findings, and most (75%) expressed interest in learning their PD genetic risk score. Discussion Our results highlight broad interest in comprehensive genetic testing among patients with PD and may facilitate integration of genome sequencing in clinical practice.
Collapse
Affiliation(s)
| | | | - Rami Al-Ouran
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Jennifer Deger
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Jamie C. Fong
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Paul Jerrod Vandeventer
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Emily Schulman
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Sindhu Rao
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Hiba Saade
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Joseph M. Savitt
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Rainer von Coelln
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Neeja Desai
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Harshavardhan Doddapaneni
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Sejal Salvi
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Shannon Dugan-Perez
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Donna M. Muzny
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Amy L. McGuire
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Zhandong Liu
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Richard A. Gibbs
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Chad Shaw
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Joseph Jankovic
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Lisa M. Shulman
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Joshua M. Shulman
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| |
Collapse
|
20
|
Reyes-González J, Barajas-Olmos F, García-Ortiz H, Magraner-Pardo L, Pons T, Moreno S, Aguirre-Cruz L, Reyes-Abrahantes A, Martínez-Hernández A, Contreras-Cubas C, Barrios-Payan J, Ruiz-Garcia H, Hernandez-Pando R, Quiñones-Hinojosa A, Orozco L, Abrahantes-Pérez MDC. Brain radiotoxicity-related 15CAcBRT gene expression signature predicts survival prognosis of glioblastoma patients. Neuro Oncol 2022; 25:303-314. [PMID: 35802478 PMCID: PMC9925695 DOI: 10.1093/neuonc/noac171] [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: 11/13/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Glioblastoma is the most common and devastating primary brain cancer. Radiotherapy is standard of care; however, it is associated with brain radiation toxicity (BRT). This study used a multi-omics approach to determine whether BRT-related genes (RGs) harbor survival prognostic value and whether their encoded proteins represent novel therapeutic targets for glioblastoma. METHODS RGs were identified through analysis of single-nucleotide variants associated with BRT (R-SNVs). Functional relationships between RGs were established using Protein-Protein Interaction networks. The influence of RGs and their functional groups on glioblastoma prognosis was evaluated using clinical samples from the Glioblastoma Bio-Discovery Portal database and validated using the Chinese Glioma Genome Atlas dataset. The identification of clusters of radiotoxic and putative pathogenic variants in proteins encoded by RGs was achieved by computational 3D structural analysis. RESULTS We identified the BRT-related 15CAcBRT molecular signature with prognostic value in glioblastoma, by analysis of the COMT and APOE protein functional groups. Its external validation confirmed clinical relevance independent of age, MGMT promoter methylation status, and IDH mutation status. Interestingly, the genes IL6, APOE, and MAOB documented significant gene expression levels alteration, useful for drug repositioning. Biological networks associated with 15CAcBRT signature involved pathways relevant to cancer and neurodegenerative diseases. Analysis of 3D clusters of radiotoxic and putative pathogenic variants in proteins coded by RGs unveiled potential novel therapeutic targets in neuro-oncology. CONCLUSIONS 15CAcBRT is a BRT-related molecular signature with prognostic significance for glioblastoma patients and represents a hub for drug repositioning and development of novel therapies.
Collapse
Affiliation(s)
| | | | - Humberto García-Ortiz
- Immunogenomics and Metabolic Diseases Laboratory, National Institute of Genomic Medicine, Mexico City, Mexico
| | | | - Tirso Pons
- Department of Immunology and Oncology, National Center for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, Spain
| | - Sergio Moreno
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery;Mexico City, Mexico
| | - Lucinda Aguirre-Cruz
- Neuroendocrinology Laboratory, National Institute of Neurology and Neurosurgery; Mexico City, Mexico
| | - Andy Reyes-Abrahantes
- Precision Translational Oncology Laboratory, National Institute of Genomic Medicine, Mexico City, Mexico
| | - Angélica Martínez-Hernández
- Immunogenomics and Metabolic Diseases Laboratory, National Institute of Genomic Medicine, Mexico City, Mexico
| | - Cecilia Contreras-Cubas
- Immunogenomics and Metabolic Diseases Laboratory, National Institute of Genomic Medicine, Mexico City, Mexico
| | - Jorge Barrios-Payan
- Department of Pathology, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, Mexico
| | - Henry Ruiz-Garcia
- Department of Neurosurgery and Brain Tumor Stem Cell Research Laboratory, Mayo Clinic, Jacksonville, Florida,USA
| | - Rogelio Hernandez-Pando
- Department of Pathology, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, Mexico
| | - Alfredo Quiñones-Hinojosa
- Department of Neurosurgery and Brain Tumor Stem Cell Research Laboratory, Mayo Clinic, Jacksonville, Florida,USA
| | - Lorena Orozco
- Immunogenomics and Metabolic Diseases Laboratory, National Institute of Genomic Medicine, Mexico City, Mexico
| | - María del Carmen Abrahantes-Pérez
- Corresponding Author: María del Carmen Abrahantes-Pérez, PhD, Precision Translational Oncology Laboratory, National Institute of Genomic Medicine, Periférico Sur 4809, Tlalpan, Mexico City C.P. 14610, Mexico ()
| |
Collapse
|
21
|
Szwedo AA, Dalen I, Pedersen KF, Camacho M, Bäckström D, Forsgren L, Tzoulis C, Winder-Rhodes S, Hudson G, Liu G, Scherzer CR, Lawson RA, Yarnall AJ, Williams-Gray CH, Macleod AD, Counsell CE, Tysnes OB, Alves G, Maple-Grødem J, Parkinson’s Incidence Cohorts Collaboration. GBA and APOE Impact Cognitive Decline in Parkinson's Disease: A 10-Year Population-Based Study. Mov Disord 2022; 37:1016-1027. [PMID: 35106798 PMCID: PMC9362732 DOI: 10.1002/mds.28932] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/30/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Common genetic variance in apolipoprotein E (APOE), β-glucocerebrosidase (GBA), microtubule-associated protein tau (MAPT), and α-synuclein (SNCA) has been linked to cognitive decline in Parkinson's disease (PD), although studies have yielded mixed results. OBJECTIVES To evaluate the effect of genetic variants in APOE, GBA, MAPT, and SNCA on cognitive decline and risk of dementia in a pooled analysis of six longitudinal, non-selective, population-based cohorts of newly diagnosed PD patients. METHODS 1002 PD patients, followed for up to 10 years (median 7.2 years), were genotyped for at least one of APOE-ε4, GBA mutations, MAPT H1/H2, or SNCA rs356219. We evaluated the effect of genotype on the rate of cognitive decline (Mini-Mental State Examanation, MMSE) using linear mixed models and the development of dementia (diagnosed using standardized criteria) using Cox regression; multiple comparisons were accounted for using Benjamini-Hochberg corrections. RESULTS Carriers of APOE-ε4 (n = 281, 29.7%) and GBA mutations (n = 100, 10.3%) had faster cognitive decline and were at higher risk of progression to dementia (APOE-ε4, HR 3.57, P < 0.001; GBA mutations, HR 1.76, P = 0.001) than non-carriers. The risk of cognitive decline and dementia (HR 5.19, P < 0.001) was further increased in carriers of both risk genotypes (n = 23). No significant effects were observed for MAPT or SNCA rs356219. CONCLUSIONS GBA and APOE genotyping could improve the prediction of cognitive decline in PD, which is important to inform the clinical trial selection and potentially to enable personalized treatment © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Aleksandra A. Szwedo
- The Norwegian Center for Movement Disorders, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
| | - Ingvild Dalen
- Department of Research, Section of Biostatistics, Stavanger University Hospital, Stavanger, Norway
| | - Kenn Freddy Pedersen
- The Norwegian Center for Movement Disorders, Stavanger University Hospital, Stavanger, Norway
- Department of Neurology, Stavanger University Hospital, Stavanger, Norway
| | - Marta Camacho
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - David Bäckström
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
- Department of Neurology, and Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lars Forsgren
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Charalampos Tzoulis
- Department of Neurology, Haukeland University Hospital, University of Bergen, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | | | - Gavin Hudson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
- Wellcome Centre for Mitochondrial Research, Newcastle University, Newcastle upon Tyne, UK
| | - Ganqiang Liu
- Neurobiology Research Center, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Clemens R. Scherzer
- Center for Advanced Parkinson Research, Harvard Medical School, Brigham and Women’s Hospital, Boston, USA
| | - Rachael A. Lawson
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Alison J. Yarnall
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Angus D. Macleod
- Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - Carl E. Counsell
- Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - Ole-Bjørn Tysnes
- Department of Neurology, Haukeland University Hospital, University of Bergen, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Guido Alves
- The Norwegian Center for Movement Disorders, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
- Department of Neurology, Stavanger University Hospital, Stavanger, Norway
| | - Jodi Maple-Grødem
- The Norwegian Center for Movement Disorders, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
| | | |
Collapse
|
22
|
Genetics of cognitive dysfunction in Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2022; 269:195-226. [PMID: 35248195 DOI: 10.1016/bs.pbr.2022.01.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Presentation and progression of cognitive symptoms in Parkinson's disease are highly variable. PD is a genetically complex disorder with multiple genetic risk factors and understanding the role that genes play in cognitive outcomes is important for patient counseling and treatment. Currently, there are seven well-described genes that increase the risk for PD, with variable levels of penetrance: SNCA, LRRK2, VPS35, PRKN, PINK1, DJ1 and GBA. In addition, large, genome-wide association studies have identified multiple loci in our DNA which increase PD risk. In this chapter, we summarize what is currently known about each of the seven strongly-associated PD genes and select PD risk variants, including PITX3, TMEM106B, SNCA Rep1, APOɛ4, COMT and MAPT H1/H1, along with their respective relationships to cognition.
Collapse
|
23
|
Wei X, Shen Q, Litvan I, Huang M, Lee RR, Harrington DL. Internetwork Connectivity Predicts Cognitive Decline in Parkinson’s and Is Altered by Genetic Variants. Front Aging Neurosci 2022; 14:853029. [PMID: 35418853 PMCID: PMC8996114 DOI: 10.3389/fnagi.2022.853029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/02/2022] [Indexed: 12/30/2022] Open
Abstract
In Parkinson’s disease (PD) functional changes in the brain occur years before significant cognitive symptoms manifest yet core large-scale networks that maintain cognition and predict future cognitive decline are poorly understood. The present study investigated internetwork functional connectivity of visual (VN), anterior and posterior default mode (aDMN, pDMN), left/right frontoparietal (LFPN, RFPN), and salience (SN) networks in 63 cognitively normal PD (PDCN) and 43 healthy controls who underwent resting-state functional MRI. The functional relevance of internetwork coupling topologies was tested by their correlations with baseline cognitive performance in each group and with 2-year cognitive changes in a PDCN subsample. To disentangle heterogeneity in neurocognitive functioning, we also studied whether α-synuclein (SNCA) and microtubule-associated protein tau (MAPT) variants alter internetwork connectivity and/or accelerate cognitive decline. We found that internetwork connectivity was largely preserved in PDCN, except for reduced pDMN-RFPN/LFPN couplings, which correlated with poorer baseline global cognition. Preserved internetwork couplings also correlated with domain-specific cognition but differently for the two groups. In PDCN, stronger positive internetwork coupling topologies correlated with better cognition at baseline, suggesting a compensatory mechanism arising from less effective deployment of networks that supported cognition in healthy controls. However, stronger positive internetwork coupling topologies typically predicted greater longitudinal decline in most cognitive domains, suggesting that they were surrogate markers of neuronal vulnerability. In this regard, stronger aDMN-SN, LFPN-SN, and/or LFPN-VN connectivity predicted longitudinal decline in attention, working memory, executive functioning, and visual cognition, which is a risk factor for dementia. Coupling strengths of some internetwork topologies were altered by genetic variants. PDCN carriers of the SNCA risk allele showed amplified anticorrelations between the SN and the VN/pDMN, which supported cognition in healthy controls, but strengthened pDMN-RFPN connectivity, which maintained visual memory longitudinally. PDCN carriers of the MAPT risk allele showed greater longitudinal decline in working memory and increased VN-LFPN connectivity, which in turn predicted greater decline in visuospatial processing. Collectively, the results suggest that cognition is maintained by functional reconfiguration of large-scale internetwork communications, which are partly altered by genetic risk factors and predict future domain-specific cognitive progression.
Collapse
Affiliation(s)
- Xiangyu Wei
- Research and Radiology Services, VA San Diego Healthcare System, San Diego, CA, United States
- Revelle College, University of California San Diego, La Jolla, CA, United States
| | - Qian Shen
- Research and Radiology Services, VA San Diego Healthcare System, San Diego, CA, United States
- Department of Radiology, University of California San Diego, La Jolla, CA, United States
| | - Irene Litvan
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States
| | - Mingxiong Huang
- Research and Radiology Services, VA San Diego Healthcare System, San Diego, CA, United States
- Department of Radiology, University of California San Diego, La Jolla, CA, United States
| | - Roland R. Lee
- Research and Radiology Services, VA San Diego Healthcare System, San Diego, CA, United States
- Department of Radiology, University of California San Diego, La Jolla, CA, United States
| | - Deborah L. Harrington
- Research and Radiology Services, VA San Diego Healthcare System, San Diego, CA, United States
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States
- *Correspondence: Deborah L. Harrington,
| |
Collapse
|
24
|
Chung J, Ushakova A, Doitsidou M, Tzoulis C, Tysnes OB, Dalen I, Pedersen KF, Alves G, Maple-Grødem J. The impact of common genetic variants in cognitive decline in the first seven years of Parkinson's disease: A longitudinal observational study. Neurosci Lett 2021; 764:136243. [PMID: 34509566 DOI: 10.1016/j.neulet.2021.136243] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Cognitive impairment is a common feature of Parkinson's disease and is a significant determinant of patients' quality of life and dependence. The pattern and progression of cognitive symptoms vary greatly between individuals, and genetic biomarkers may help to predict the severity and trajectory of cognitive impairment in groups of patients. METHODS The study included 171 patients from a longitudinal population-based incident Parkinson's disease study from South Western Norway. All participants were followed from the time of diagnosis for up to seven years, undertaking repeated batteries of clinical and neuropsychological tests, measuring global cognitive impairment, executive function, attention, verbal learning and memory, and visuospatial skills. We used linear mixed regression analyses to explore associations between the function in specific cognitive domains over time and common genetic variants in APOE, MAPT, COMT and BDNF. RESULTS The COMT158Val/Val allele wasassociatedwith faster decline in executive function (p = 0.028), verbal learning and memory (p = 0.029), and visuospatial skills (p = 0.027). The BDNF, MAPT and APOE genotypes were not significantly associated with longitudinal changes in individual cognitive domains, however carriers of the APOE-ε4 allele were shown to be at increased risk of mild cognitive impairment and dementia within the study period (OR3.03; p = 0.006). CONCLUSIONS This population-based study of newly diagnosed patients provides new evidence that COMTVal158Met effects cognitive outcomes limited to discrete domains and APOE-ε4 status predicts a poor overall cognitive prognosis. Together, these data contribute to our understanding of the biology underlying the heterogeneity observed in the progression of PD.
Collapse
Affiliation(s)
- Janete Chung
- The Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway
| | - Anastasia Ushakova
- Department of Research, Section of Biostatistics, Stavanger University Hospital, Stavanger, Norway
| | - Maria Doitsidou
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | | | - Ole-Bjørn Tysnes
- Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Norway
| | - Ingvild Dalen
- Department of Research, Section of Biostatistics, Stavanger University Hospital, Stavanger, Norway
| | - Kenn Freddy Pedersen
- The Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway; Department of Neurology, Stavanger University Hospital, Stavanger, Norway
| | - Guido Alves
- The Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway; Department of Neurology, Stavanger University Hospital, Stavanger, Norway; Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
| | - Jodi Maple-Grødem
- The Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway; Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway.
| |
Collapse
|
25
|
Kusters CDJ, Paul KC, Folle AD, Keener AM, Bronstein JM, Bertram L, Hansen J, Horvath S, Sinsheimer JS, Lill CM, Ritz BR. Increased Menopausal Age Reduces the Risk of Parkinson's Disease: A Mendelian Randomization Approach. Mov Disord 2021; 36:2264-2272. [PMID: 34426982 PMCID: PMC8530889 DOI: 10.1002/mds.28760] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 07/15/2021] [Accepted: 07/26/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Studies of Parkinson's disease (PD) and the association with age at menarche or menopause have reported inconsistent findings. Mendelian randomization (MR) may address measurement errors because of difficulties accurately reporting the age these life events occur. OBJECTIVE We used MR to assess the association between age at menopause and age at menarche with PD risk. METHODS We performed inverse variant-weighted (IVW) MR analysis using external genome-wide association study (GWAS) summary data from the United Kingdom biobank, and the effect estimates between genetic variants and PD among two population-based studies (Parkinson's disease in Denmark (PASIDA) study, Denmark, and Parkinson's Environment and Gene study [PEG], United States) that enrolled 1737 female and 2430 male subjects of European ancestry. We, then, replicated our findings for age at menopause using summary statistics from the PD consortium (19 773 women), followed by a meta-analysis combining all summary statistics. RESULTS For each year increase in age at menopause, the risk for PD decreased (odds ration [OR], 0.84; 95% confidence interval [CI], 0.73-0.98; P = 0.03) among women in our study, whereas there was no association among men (OR, 0.98; 95% CI, 0.85-1.11; P = 0.71). A replication using summary statistics from the PD consortium estimated an OR of 0.94 (95% CI, 0.90-0.99; P = 0.01), and we calculated a meta-analytic OR of 0.93 (95% CI, 0.89-0.98; P = 0.003). There was no indication for an association between age at menarche and PD (OR, 0.75; 95% CI, 0.44-1.29; P = 0.29). CONCLUSIONS A later age at menopause was associated with a decreased risk of PD in women, supporting the hypothesis that sex hormones or other factors related to late menopause may be neuroprotective in PD. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Cynthia DJ Kusters
- Department of Human Genetics, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Kimberly C Paul
- Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Aline Duarte Folle
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Adrienne M Keener
- Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA,Parkinson’s Disease Research, Education, and Clinical Center, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, California, USA
| | - Jeff M. Bronstein
- Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA,Brain Research Institute, University of California, Los Angeles, CA, USA
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, University of Lübeck, Lübeck, Germany,Department of Psychology, Centre for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway
| | - Johnni Hansen
- Danish Cancer Society Research Center, Danish Cancer Society, Copenhagen, Denmark
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, Los Angeles, CA, USA,Department of Biostatistics, School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA
| | - Janet S. Sinsheimer
- Department of Human Genetics, David Geffen School of Medicine, Los Angeles, CA, USA,Department of Biostatistics, School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA,Department of Computational Medicine, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Christina M. Lill
- Translational Epidemiology Group, Lübeck Interdisciplinary Platform for Genome Analytics, University of Lübeck, Lübeck, Germany,Ageing Epidemiology Research Unit, School of Public Health, Imperial College, London, United Kingdom
| | - Beate R. Ritz
- Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA,Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA,Department of Environmental Health, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| |
Collapse
|
26
|
Paul KC, Binder AM, Horvath S, Kusters C, Yan Q, Rosario ID, Yu Y, Bronstein J, Ritz B. Accelerated hematopoietic mitotic aging measured by DNA methylation, blood cell lineage, and Parkinson's disease. BMC Genomics 2021; 22:696. [PMID: 34565328 PMCID: PMC8474781 DOI: 10.1186/s12864-021-08009-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 09/13/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Aging and inflammation are important components of Parkinson's disease (PD) pathogenesis and both are associated with changes in hematopoiesis and blood cell composition. DNA methylation (DNAm) presents a mechanism to investigate inflammation, aging, and hematopoiesis in PD, using epigenetic mitotic aging and aging clocks. Here, we aimed to define the influence of blood cell lineage on epigenetic mitotic age and then investigate mitotic age acceleration with PD, while considering epigenetic age acceleration biomarkers. RESULTS We estimated epigenetic mitotic age using the "epiTOC" epigenetic mitotic clock in 10 different blood cell populations and in a population-based study of PD with whole-blood. Within subject analysis of the flow-sorted purified blood cell types DNAm showed a clear separation of epigenetic mitotic age by cell lineage, with the mitotic age significantly lower in myeloid versus lymphoid cells (p = 2.1e-11). PD status was strongly associated with accelerated epigenetic mitotic aging (AccelEpiTOC) after controlling for cell composition (OR = 2.11, 95 % CI = 1.56, 2.86, p = 1.6e-6). AccelEpiTOC was also positively correlated with extrinsic epigenetic age acceleration, a DNAm aging biomarker related to immune system aging (with cell composition adjustment: R = 0.27, p = 6.5e-14), and both were independently associated with PD. Among PD patients, AccelEpiTOC measured at baseline was also associated with longitudinal motor and cognitive symptom decline. CONCLUSIONS The current study presents a first look at epigenetic mitotic aging in PD and our findings suggest accelerated hematopoietic cell mitosis, possibly reflecting immune pathway imbalances, in early PD that may also be related to motor and cognitive progression.
Collapse
Affiliation(s)
- Kimberly C Paul
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.
| | - Alexandra M Binder
- Population Sciences in the Pacific Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Cynthia Kusters
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Qi Yan
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Irish Del Rosario
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Yu Yu
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Jeff Bronstein
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Beate Ritz
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| |
Collapse
|
27
|
Leveille E, Ross OA, Gan-Or Z. Tau and MAPT genetics in tauopathies and synucleinopathies. Parkinsonism Relat Disord 2021; 90:142-154. [PMID: 34593302 PMCID: PMC9310195 DOI: 10.1016/j.parkreldis.2021.09.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/25/2021] [Accepted: 09/09/2021] [Indexed: 10/20/2022]
Abstract
MAPT encodes the microtubule-associated protein tau, which is the main component of neurofibrillary tangles (NFTs) and found in other protein aggregates. These aggregates are among the pathological hallmarks of primary tauopathies such as frontotemporal dementia (FTD). Abnormal tau can also be observed in secondary tauopathies such as Alzheimer's disease (AD) and synucleinopathies such as Parkinson's disease (PD). On top of pathological findings, genetic data also links MAPT to these disorders. MAPT variations are a cause or risk factors for many tauopathies and synucleinopathies and are associated with certain clinical and pathological features in affected individuals. In addition to clinical, pathological, and genetic overlap, evidence also suggests that tau and alpha-synuclein may interact on the molecular level, and thus might collaborate in the neurodegenerative process. Understanding the role of MAPT variations in tauopathies and synucleinopathies is therefore essential to elucidate the role of tau in the pathogenesis and phenotype of those disorders, and ultimately to develop targeted therapies. In this review, we describe the role of MAPT genetic variations in tauopathies and synucleinopathies, several genotype-phenotype and pathological features, and discuss their implications for the classification and treatment of those disorders.
Collapse
Affiliation(s)
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA; Department of Clinical Genomics, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Ziv Gan-Or
- The Neuro (Montreal Neurological Institute-hospital), McGill University, Montréal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada; Department of Human Genetics, McGill University, Montréal, QC, Canada.
| |
Collapse
|
28
|
Davis AA, Inman CE, Wargel ZM, Dube U, Freeberg BM, Galluppi A, Haines JN, Dhavale DD, Miller R, Choudhury FA, Sullivan PM, Cruchaga C, Perlmutter JS, Ulrich JD, Benitez BA, Kotzbauer PT, Holtzman DM. APOE genotype regulates pathology and disease progression in synucleinopathy. Sci Transl Med 2021; 12:12/529/eaay3069. [PMID: 32024799 DOI: 10.1126/scitranslmed.aay3069] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022]
Abstract
Apolipoprotein E (APOE) ε4 genotype is associated with increased risk of dementia in Parkinson's disease (PD), but the mechanism is not clear, because patients often have a mixture of α-synuclein (αSyn), amyloid-β (Aβ), and tau pathologies. APOE ε4 exacerbates brain Aβ pathology, as well as tau pathology, but it is not clear whether APOE genotype independently regulates αSyn pathology. In this study, we generated A53T αSyn transgenic mice (A53T) on Apoe knockout (A53T/EKO) or human APOE knockin backgrounds (A53T/E2, E3, and E4). At 12 months of age, A53T/E4 mice accumulated higher amounts of brainstem detergent-insoluble phosphorylated αSyn compared to A53T/EKO and A53T/E3; detergent-insoluble αSyn in A53T/E2 mice was undetectable. By immunohistochemistry, A53T/E4 mice displayed a higher burden of phosphorylated αSyn and reactive gliosis compared to A53T/E2 mice. A53T/E2 mice exhibited increased survival and improved motor performance compared to other APOE genotypes. In a complementary model of αSyn spreading, striatal injection of αSyn preformed fibrils induced greater accumulation of αSyn pathology in the substantia nigra of A53T/E4 mice compared to A53T/E2 and A53T/EKO mice. In two separate cohorts of human patients with PD, APOE ε4/ε4 individuals showed the fastest rate of cognitive decline over time. Our results demonstrate that APOE genotype directly regulates αSyn pathology independent of its established effects on Aβ and tau, corroborate the finding that APOE ε4 exacerbates pathology, and suggest that APOE ε2 may protect against αSyn aggregation and neurodegeneration in synucleinopathies.
Collapse
Affiliation(s)
- Albert A Davis
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA. .,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Casey E Inman
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Zachary M Wargel
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Umber Dube
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Psychiatry, Washington University, St. Louis, MO 63110, USA
| | - Brittany M Freeberg
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Alexander Galluppi
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Jessica N Haines
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Dhruva D Dhavale
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Rebecca Miller
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Fahim A Choudhury
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Patrick M Sullivan
- Department of Medicine, Duke University Medical Center, Durham VAMC and Geriatric Research Clinical Center, Durham, NC 27705, USA
| | - Carlos Cruchaga
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Psychiatry, Washington University, St. Louis, MO 63110, USA
| | - Joel S Perlmutter
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA.,Departments of Neuroscience and Radiology, Programs in Physical and Occupational Therapy, Washington University, St. Louis, MO 63110, USA
| | - Jason D Ulrich
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Bruno A Benitez
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Psychiatry, Washington University, St. Louis, MO 63110, USA
| | - Paul T Kotzbauer
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - David M Holtzman
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA. .,Department of Neurology, Washington University, St. Louis, MO 63110, USA.,Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA
| |
Collapse
|
29
|
Salles PA, Mata IF, Fernandez HH. Should we start integrating genetic data in decision-making on device-aided therapies in Parkinson disease? A point of view. Parkinsonism Relat Disord 2021; 88:51-57. [PMID: 34119931 DOI: 10.1016/j.parkreldis.2021.05.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 04/26/2021] [Accepted: 05/12/2021] [Indexed: 12/14/2022]
Abstract
Parkinson disease (PD) is a complex heterogeneous neurodegenerative disorder. Association studies have revealed numerous genetic risk loci and variants, and about 5-10% suffer from a monogenic form. Because the presentation and course of PD is unique to each patient, personalized symptomatic treatment should ideally be offered to treat the most disabling motor and non-motor symptoms. Indeed, clinical milestones and treatment complications that appear during disease progression are influenced by the genetic imprint. With recent advances in PD, more patients live longer to become eligible for device-aided therapies, such as apomorphine continuous subcutaneous infusion, levodopa duodenal gel infusion, and deep brain stimulation surgery, each with its own inclusion and exclusion criteria, advantages and disadvantages. Because genetic variants influence the expression of particular clinical profiles, factors for better or worse outcomes for device-aided therapies may then be proactively identified. For example, mutations in PRKN, LRRK2 and GBA express phenotypes that favor suitability for different device therapies, although with marked differences in the therapeutic window; whereas multiplications of SNCA express phenotypes that make them less desirable for device therapies.
Collapse
Affiliation(s)
- Philippe A Salles
- Center for Neurological Restoration, Cleveland Clinic Neurological Institute, OH, USA; Movement Disorders Center, CETRAM, Santiago, Chile.
| | - Ignacio F Mata
- Lerner Research Institute, Genomic Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA.
| | - Hubert H Fernandez
- Center for Neurological Restoration, Cleveland Clinic Neurological Institute, OH, USA.
| |
Collapse
|
30
|
Gonzalez-Latapi P, Bayram E, Litvan I, Marras C. Cognitive Impairment in Parkinson's Disease: Epidemiology, Clinical Profile, Protective and Risk Factors. Behav Sci (Basel) 2021; 11:bs11050074. [PMID: 34068064 PMCID: PMC8152515 DOI: 10.3390/bs11050074] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 02/07/2023] Open
Abstract
Cognitive impairment is a common non-motor symptom in Parkinson's Disease (PD) and an important source of patient disability and caregiver burden. The timing, profile and rate of cognitive decline varies widely among individuals with PD and can range from normal cognition to mild cognitive impairment (PD-MCI) and dementia (PDD). Beta-amyloid and tau brain accumulation, oxidative stress and neuroinflammation are reported risk factors for cognitive impairment. Traumatic brain injury and pesticide and tobacco exposure have also been described. Genetic risk factors including genes such as COMT, APOE, MAPT and BDNF may also play a role. Less is known about protective factors, although the Mediterranean diet and exercise may fall in this category. Nonetheless, there is conflicting evidence for most of the factors that have been studied. The use of inconsistent criteria and lack of comprehensive assessment in many studies are important methodological issues. Timing of exposure also plays a crucial role, although identification of the correct time window has been historically difficult in PD. Our understanding of the mechanism behind these factors, as well as the interactions between gene and environment as determinants of disease phenotype and the identification of modifiable risk factors will be paramount, as this will allow for potential interventions even in established PD.
Collapse
Affiliation(s)
- Paulina Gonzalez-Latapi
- Edmond J. Safra Program in Parkinson’s Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, University Health Network, Toronto, ON M5T2S8, Canada;
| | - Ece Bayram
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA; (E.B.); (I.L.)
| | - Irene Litvan
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA; (E.B.); (I.L.)
| | - Connie Marras
- Edmond J. Safra Program in Parkinson’s Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, University Health Network, Toronto, ON M5T2S8, Canada;
- Correspondence:
| |
Collapse
|
31
|
Oh YS, Yoo SW, Lyoo CH, Yoo JY, Yoon H, Ha S, Lee KS, Kim JS. The Association of β-Amyloid with Cognition and Striatal Dopamine in Early, Non-Demented Parkinson's Disease. JOURNAL OF PARKINSONS DISEASE 2021; 11:605-613. [PMID: 33646180 DOI: 10.3233/jpd-202496] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Co-occurrence of β-amyloid (Aβ) pathology has been reported in Parkinson's disease (PD), and Aβ deposition in the brain may contribute to cognitive decline in patients with PD. Whether striatal dopamine uptake and cognitive status differ with amyloid deposition has been reported in only a few studies. OBJECTIVE The purpose of this study was to investigate the association among striatal dopaminergic availability, Aβ-positivity, and motor and cognitive status in early and non-demented PD. METHODS A total of 98 newly-diagnosed, non-medicated, and non-demented patients with PD were included in this study. Cognitive status was assessed using neuropsychological testing. Patients with mild cognitive impairment (MCI) were stratified into two groups: amnestic MCI (aMCI) and non-amnestic MCI (naMCI). Patient motor status was examined using the Unified Parkinson's Disease Rating Scale (UPDRS) and positron emission tomography (PET) with 18F-N-(3-fluoropropyl)-2beta-carbon ethoxy-3beta-(4-iodophenyl) nortropane (18F-FP-CIT). All patients also underwent 18F-florbetaben (18F-FBB) PET and were divided based on the results into Aβ-positive and Aβ-negative groups. RESULTS Eighteen patients had Aβ-positivity in 18F-FBB PET and 67 had MCI. Sixteen of 18 with Aβ-positive patients had MCI. The Aβ-positive group had higher frequency of MCI, especially amnestic-type, and lower dopaminergic activities in the left ventral striatum, but not with UPDRS motor score. CONCLUSION Amyloid pathology was associated with MCI, especially amnestic-subtype, in early and non-demented PD patients and with low dopaminergic activities in the left ventral striatum. This finding suggests that PD patients with Aβ-positivity have AD-related cognitive pathophysiology in PD and associated impaired dopaminergic availability in the ventral striatum can affect the pathophysiology in various ways.
Collapse
Affiliation(s)
- Yoon-Sang Oh
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang-Won Yoo
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji-Yeon Yoo
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyukjin Yoon
- Department of Nuclear Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seunggyun Ha
- Department of Nuclear Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kwang-Soo Lee
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Joong-Seok Kim
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
32
|
Ma SX, Lim SB. Single-Cell RNA Sequencing in Parkinson's Disease. Biomedicines 2021; 9:368. [PMID: 33916045 PMCID: PMC8066089 DOI: 10.3390/biomedicines9040368] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/28/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
Single-cell and single-nucleus RNA sequencing (sc/snRNA-seq) technologies have enhanced the understanding of the molecular pathogenesis of neurodegenerative disorders, including Parkinson's disease (PD). Nonetheless, their application in PD has been limited due mainly to the technical challenges resulting from the scarcity of postmortem brain tissue and low quality associated with RNA degradation. Despite such challenges, recent advances in animals and human in vitro models that recapitulate features of PD along with sequencing assays have fueled studies aiming to obtain an unbiased and global view of cellular composition and phenotype of PD at the single-cell resolution. Here, we reviewed recent sc/snRNA-seq efforts that have successfully characterized diverse cell-type populations and identified cell type-specific disease associations in PD. We also examined how these studies have employed computational and analytical tools to analyze and interpret the rich information derived from sc/snRNA-seq. Finally, we highlighted important limitations and emerging technologies for addressing key technical challenges currently limiting the integration of new findings into clinical practice.
Collapse
Affiliation(s)
- Shi-Xun Ma
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | - Su Bin Lim
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 16499, Korea
| |
Collapse
|
33
|
Tunold JA, Geut H, Rozemuller JMA, Henriksen SP, Toft M, van de Berg WDJ, Pihlstrøm L. APOE and MAPT Are Associated With Dementia in Neuropathologically Confirmed Parkinson's Disease. Front Neurol 2021; 12:631145. [PMID: 33613437 PMCID: PMC7892776 DOI: 10.3389/fneur.2021.631145] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/11/2021] [Indexed: 01/24/2023] Open
Abstract
Introduction: Cognitive decline and dementia are common and debilitating non-motor phenotypic features of Parkinson's disease with a variable severity and time of onset. Common genetic variation of the Apolipoprotein E (APOE) and micro-tubule associated protein tau (MAPT) loci have been linked to cognitive decline and dementia in Parkinson's disease, although studies have yielded mixed results. To further elucidate the influence of APOE and MAPT variability on dementia in Parkinson's disease, we genotyped postmortem brain tissue samples of clinically and pathologically well-characterized Parkinson's donors and performed a survival analysis of time to dementia. Methods: We included a total of 152 neuropathologically confirmed Parkinson's disease donors with or without clinical dementia during life. We genotyped known risk variants tagging the APOE ε4 allele and MAPT H1/H2 inversion haplotype. Cox proportional hazards regression analyses adjusted for age at onset, sex and genetic principal components were performed to assess the association between the genetic variants and time from motor onset to onset of dementia. Results: We found that both the APOE ε4 allele (HR 1.82, 95 % CI 1.16–2.83, p = 0.009) and MAPT H1-haplotype (HR 1.71, 95 % CI 1.06–2.78, p = 0.03) were associated with earlier development of dementia in patients with Parkinson's disease. Conclusion: Our results provide further support for the importance of APOE ε4 and MAPT H1-haplotype in the etiology of Parkinson's disease dementia, with potential future relevance for risk stratification and patient selection for clinical trials of therapies targeting cognitive decline in Parkinson's disease.
Collapse
Affiliation(s)
- Jon-Anders Tunold
- Department of Neurology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Hanneke Geut
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - J M Annemieke Rozemuller
- Department of Pathology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | | | - Mathias Toft
- Department of Neurology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Wilma D J van de Berg
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
34
|
Wang FY, Wang P, Zhao DF, Gonzalez FJ, Fan YF, Xia YL, Ge GB, Yang L. Analytical methodologies for sensing catechol- O-methyltransferase activity and their applications. J Pharm Anal 2021; 11:15-27. [PMID: 33717608 PMCID: PMC7930641 DOI: 10.1016/j.jpha.2020.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/21/2020] [Accepted: 03/30/2020] [Indexed: 01/02/2023] Open
Abstract
Mammalian catechol-O-methyltransferases (COMT) are an important class of conjugative enzymes, which play a key role in the metabolism and inactivation of catechol neurotransmitters, catechol estrogens and a wide range of endobiotics and xenobiotics that bear the catechol group. Currently, COMT inhibitors are used in combination with levodopa for the treatment of Parkinson's disease in clinical practice. The crucial role of COMT in human health has raised great interest in the development of more practical assays for highly selective and sensitive detection of COMT activity in real samples, as well as for rapid screening and characterization of COMT inhibitors as drug candidates. This review summarizes recent advances in analytical methodologies for sensing COMT activity and their applications. Several lists of biochemical assays for measuring COMT activity, including the probe substrates, along with their analytical conditions and kinetic parameters, are presented. Finally, the challenges and future perspectives in the field, such as visualization of COMT activity in vivo and in situ, are highlighted. Collectively, this review article overviews the practical assays for measuring COMT activities in complex biological samples, which will strongly facilitate the investigations on the relevance of COMT to human diseases and promote the discovery of COMT inhibitors via high-throughput screening.
Collapse
Affiliation(s)
- Fang-Yuan Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ping Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dong-Fang Zhao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yu-Fan Fan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yang-Liu Xia
- School of Life Science and Medicine, Dalian University of Technology, Panjin, 124221, China
| | - Guang-Bo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ling Yang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
35
|
Haider S, Madiha S, Batool Z. Amelioration of motor and non-motor deficits and increased striatal APoE levels highlight the beneficial role of pistachio supplementation in rotenone-induced rat model of PD. Metab Brain Dis 2020; 35:1189-1200. [PMID: 32529399 DOI: 10.1007/s11011-020-00584-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/01/2020] [Indexed: 12/21/2022]
Abstract
Pistachio contains polyphenolic compounds including flavonoids and anthocyanins which have antioxidant and antiinflammatory activity. Present study was aimed to evaluate the protective effects of pistachio on neurobehavioral and neurochemical changes in rats with Parkinson's disease (PD). Animal model of PD was induced by the injection of rotenone (1.5 mg/kg/day, s.c.) for 8 days. Pistachio (800 mg/kg/day, p.o.) was given for two weeks in both pre- and post-treatment. At the end of treatment brain was dissected out and striatum was isolated for biochemical and neurochemical analysis. Memory was assessed by Morris water maze (MWM) and novel object recognition (NOR) test while open field test (OFT), Kondziela inverted screen test (KIST), pole test (PT), beam walking test (BWT), inclined plane test (IPT) and footprint (FP) test were used to observe motor behavior. Rotenone administration significantly (p < 0.01) impaired the memory but pistachio in both pre- and post-treatment groups significantly (p < 0.01) improved memory performance. Rotenone-induced motor deficits were significantly attenuated in both pre- and post-pistachio treatment. Increased oxidative stress and decreased DA and 5-HT levels induced by rotenone were also significantly attenuated by pistachio supplementation. Furthermore, raised apolipoprotein E (APoE) levels in rotenone injected rats were also normalized following treatment with pistachio. Present findings show that pistachio possesses neuroprotective effects and improves memory and motor deficits via increasing DA levels and improving oxidative status in brain.
Collapse
Affiliation(s)
- Saida Haider
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, 75270, Karachi, Pakistan.
| | - Syeda Madiha
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, 75270, Karachi, Pakistan
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| |
Collapse
|
36
|
Shang S, Chen YC, Zhang H, Dou W, Qian L, Yin X, Wu J. Mapping the Interactive Effects of ApoE Gene Polymorphism on Caudate Functional Connectivity in Mild Cognitive Impairment Associated With Parkinson's Disease. Front Neurosci 2020; 14:857. [PMID: 33041748 PMCID: PMC7527607 DOI: 10.3389/fnins.2020.00857] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/22/2020] [Indexed: 11/17/2022] Open
Abstract
Introduction Cognitive impairment (CI) is a frequent non-motor symptom of Parkinson’s disease (PD). Caudate and Apolipoprotein E (ApoE) are biomarkers linked to CI in PD. There is little known about whether ApoE affects caudate in mild CI of PD (PD-MCI). We investigated the possible interactive effect of ApoE genotypes on caudate functional connectivity (FC) in PD-MCI. Methods A total of 95 PD-MCI patients and 99 matched healthy controls underwent extensive neuropsychological assessment and magnetic resonance imaging. The two groups were separated into three subgroups according to their genotyping. Functional data were analyzed with FC analysis. Results Decreased FC between the caudate and the bilateral inferior orbit frontal gyrus and bilateral middle occipital gyrus (MOG) was found between groups, along with poor performance in general, executive, episodic memory, language, and visual–spatial function. Decreased FC between the caudate and right MOG, right middle temporal gyrus, and right superior occipital gyrus was found as an interaction effect. The FC values of ε4 carriers with PD-MCI were much lower than the other carriers, and FC was positively correlated with the impairment of global and language function. Conclusion These results support the idea that altered FC between the bilateral caudate and posterior cortical regions was interactively influenced by ApoE genotype and PD-MCI status, and the ε4 subtype associated with underlying pathology of global cognitive decline and semantic fluency impairment in an interactive manner. Gene-based imaging approaches might strengthen the credibility in imaging genetic associations, which might provide new powerful insights into the neural mechanisms underlying PD-MCI.
Collapse
Affiliation(s)
- Song'an Shang
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yu-Chen Chen
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hongying Zhang
- Department of Radiology, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Weiqiang Dou
- GE Healthcare, MR Research China, Beijing, China
| | - Long Qian
- GE Healthcare, MR Research China, Beijing, China
| | - Xindao Yin
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jingtao Wu
- Department of Radiology, Clinical Medical College, Yangzhou University, Yangzhou, China
| |
Collapse
|
37
|
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
|
38
|
Su C, Tong J, Wang F. Mining genetic and transcriptomic data using machine learning approaches in Parkinson's disease. NPJ PARKINSONS DISEASE 2020; 6:24. [PMID: 32964109 PMCID: PMC7481248 DOI: 10.1038/s41531-020-00127-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 08/13/2020] [Indexed: 01/08/2023]
Abstract
High-throughput techniques have generated abundant genetic and transcriptomic data of Parkinson’s disease (PD) patients but data analysis approaches such as traditional statistical methods have not provided much in the way of insightful integrated analysis or interpretation of the data. As an advanced computational approach, machine learning, which enables people to identify complex patterns and insight from data, has consequently been harnessed to analyze and interpret large, highly complex genetic and transcriptomic data toward a better understanding of PD. In particular, machine learning models have been developed to integrate patient genotype data alone or combined with demographic, clinical, neuroimaging, and other information, for PD outcome study. They have also been used to identify biomarkers of PD based on transcriptomic data, e.g., gene expression profiles from microarrays. This study overviews the relevant literature on using machine learning models for genetic and transcriptomic data analysis in PD, points out remaining challenges, and suggests future directions accordingly. Undoubtedly, the use of machine learning is amplifying PD genetic and transcriptomic achievements for accelerating the study of PD. Existing studies have demonstrated the great potential of machine learning in discovering hidden patterns within genetic or transcriptomic information and thus revealing clues underpinning pathology and pathogenesis. Moving forward, by addressing the remaining challenges, machine learning may advance our ability to precisely diagnose, prognose, and treat PD.
Collapse
Affiliation(s)
- Chang Su
- Department of Population Health Sciences, Weill Cornell Medical College, Cornell University, New York, NY USA
| | - Jie Tong
- Department of Mechanical and Aerospace Engineering, New York University, New York, NY USA
| | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medical College, Cornell University, New York, NY USA
| |
Collapse
|
39
|
Torres ERS, Weber Boutros S, Meshul CK, Raber J. ApoE isoform-specific differences in behavior and cognition associated with subchronic MPTP exposure. ACTA ACUST UNITED AC 2020; 27:372-379. [PMID: 32817303 PMCID: PMC7433653 DOI: 10.1101/lm.052126.120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/07/2020] [Indexed: 12/25/2022]
Abstract
Parkinson's disease (PD) is characterized clinically by progressive motor dysfunction; overt parkinsonism is often preceded by prodromal symptoms including disturbances in the sleep–wake cycle. Up to 80% of patients with PD also develop dementia. In humans, there are three major apolipoprotein E isoforms: E2, E3, and E4. Increased rate of dementia in PD may be associated with E4 isoform. To better understand prodromal changes associated with E4, we exposed young (3–5 mo) male and female mice expressing E3 or E4 via targeted replacement to a subchronic dosage of the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). We hypothesized that E4 mice would be more susceptible to MPTP-related behavioral and cognitive changes. MPTP-treated E4 mice explored novel objects longer than genotype-matched saline-treated mice. In contrast, saline-treated E3 mice preferentially explored the novel object whereas MPTP-treated E3 mice did not and showed impaired object recognition. MPTP treatment altered swim speed of E4, but not E3, mice in the water maze compared to controls. Thus, E4 carriage may influence the preclinical symptoms associated with PD. Increased efforts are warranted to study early time points in this disease model.
Collapse
Affiliation(s)
- Eileen Ruth S Torres
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, 97239 Oregon, USA
| | - Sydney Weber Boutros
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, 97239 Oregon, USA
| | - Charles K Meshul
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, 97239 Oregon, USA.,Portland VA Medical Center, Portland, 97239 Oregon, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, 97239 Oregon, USA.,Departments of Neurology and Radiation Medicine and Division of Neuroscience, ONPRC, Oregon Health and Science University, Portland, Oregon 97239, USA
| |
Collapse
|
40
|
Kusters CDJ, Paul KC, Duarte Folle A, Keener AM, Bronstein JM, Dobricic V, Tysnes OB, Bertram L, Alves G, Sinsheimer JS, Lill CM, Maple-Grødem J, Ritz BR. Genetic risk scores and hallucinations in patients with Parkinson disease. NEUROLOGY-GENETICS 2020; 6:e492. [PMID: 32802953 PMCID: PMC7413629 DOI: 10.1212/nxg.0000000000000492] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/26/2020] [Indexed: 01/01/2023]
Abstract
Objective We examine the hypothesized overlap of genetic architecture for Alzheimer disease (AD), schizophrenia (SZ), and Parkinson disease (PD) through the use of polygenic risk scores (PRSs) with the occurrence of hallucinations in PD. Methods We used 2 population-based studies (ParkWest, Norway, and Parkinson's Environment and Gene, USA) providing us with 399 patients with PD with European ancestry and a PD diagnosis after age 55 years to assess the associations between 4 PRSs and hallucinations after 5 years of mean disease duration. Based on the existing genome-wide association study of other large consortia, 4 PRSs were created: one each using AD, SZ, and PD cohorts and another PRS for height, which served as a negative control. Results A higher prevalence of hallucinations was observed with each SD increase of the AD-PRS (odds ratio [OR]: 1.37, 95% confidence interval [CI]: 1.03-1.83). This effect was mainly driven by APOE (OR: 1.92, 95% CI: 1.14-3.22). In addition, a suggestive decrease and increase, respectively, in hallucination prevalence were observed with the SZ-PRS and the PD-PRS (OR: 0.77, 95% CI: 0.59-1.01; and OR: 1.29, 95% CI: 0.95-1.76, respectively). No association was observed with the height PRS. Conclusions These results suggest that mechanisms for hallucinations in PD may in part be driven by the same genetic architecture that leads to cognitive decline in AD, especially by APOE.
Collapse
Affiliation(s)
- Cynthia D J Kusters
- Department of Epidemiology (C.D.J.K., K.C.P., A.D.F., B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Human Genetics (C.D.J.K., J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Department of Neurology (A.M.K., J.M.B., B.R.R.), David Geffen School of Medicine, Los Angeles, CA; Parkinson's Disease Research (A.M.K.), Education, and Clinical Center, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, CA; Brain Research Institute (J.M.B.), University of California, Los Angeles, CA; Lübeck Interdisciplinary Platform for Genome Analytics (V.D., L.B.), Institutes of Neurogenetics & Cardiogenetics, University of Lübeck, Lübeck, Germany; Department of Neurology (O.-B.T.), Haukeland University Hospital, Bergen, Norway and University of Bergen, Bergen, Norway; Department of Clinical Medicine (O.-B.T.), University of Bergen, Bergen, Norway; Department of Psychology (L.B.), Centre for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway; The Norwegian Center for Movement Disorders (G.A., J.M.-G.), Stavanger University Hospital, Stavanger, Norway; Department of Neurology (G.A.), Stavanger University Hospital, Stavanger, Norway; Department of Chemistry (G.A., J.M.-G.), Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway; Department of Biostatistics (J.S.S.), UCLA Fielding School of Public Health, Los Angeles, CA; Department of Computational Medicine (J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Section for Translational Surgical Oncology and Biobanking (C.M.L.), Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck; Ageing Epidemiology Research Unit (C.M.L.), School of Public Health, Imperial College, London, United Kingdom; and Department of Environmental Health (B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA
| | - Kimberly C Paul
- Department of Epidemiology (C.D.J.K., K.C.P., A.D.F., B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Human Genetics (C.D.J.K., J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Department of Neurology (A.M.K., J.M.B., B.R.R.), David Geffen School of Medicine, Los Angeles, CA; Parkinson's Disease Research (A.M.K.), Education, and Clinical Center, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, CA; Brain Research Institute (J.M.B.), University of California, Los Angeles, CA; Lübeck Interdisciplinary Platform for Genome Analytics (V.D., L.B.), Institutes of Neurogenetics & Cardiogenetics, University of Lübeck, Lübeck, Germany; Department of Neurology (O.-B.T.), Haukeland University Hospital, Bergen, Norway and University of Bergen, Bergen, Norway; Department of Clinical Medicine (O.-B.T.), University of Bergen, Bergen, Norway; Department of Psychology (L.B.), Centre for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway; The Norwegian Center for Movement Disorders (G.A., J.M.-G.), Stavanger University Hospital, Stavanger, Norway; Department of Neurology (G.A.), Stavanger University Hospital, Stavanger, Norway; Department of Chemistry (G.A., J.M.-G.), Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway; Department of Biostatistics (J.S.S.), UCLA Fielding School of Public Health, Los Angeles, CA; Department of Computational Medicine (J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Section for Translational Surgical Oncology and Biobanking (C.M.L.), Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck; Ageing Epidemiology Research Unit (C.M.L.), School of Public Health, Imperial College, London, United Kingdom; and Department of Environmental Health (B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA
| | - Aline Duarte Folle
- Department of Epidemiology (C.D.J.K., K.C.P., A.D.F., B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Human Genetics (C.D.J.K., J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Department of Neurology (A.M.K., J.M.B., B.R.R.), David Geffen School of Medicine, Los Angeles, CA; Parkinson's Disease Research (A.M.K.), Education, and Clinical Center, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, CA; Brain Research Institute (J.M.B.), University of California, Los Angeles, CA; Lübeck Interdisciplinary Platform for Genome Analytics (V.D., L.B.), Institutes of Neurogenetics & Cardiogenetics, University of Lübeck, Lübeck, Germany; Department of Neurology (O.-B.T.), Haukeland University Hospital, Bergen, Norway and University of Bergen, Bergen, Norway; Department of Clinical Medicine (O.-B.T.), University of Bergen, Bergen, Norway; Department of Psychology (L.B.), Centre for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway; The Norwegian Center for Movement Disorders (G.A., J.M.-G.), Stavanger University Hospital, Stavanger, Norway; Department of Neurology (G.A.), Stavanger University Hospital, Stavanger, Norway; Department of Chemistry (G.A., J.M.-G.), Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway; Department of Biostatistics (J.S.S.), UCLA Fielding School of Public Health, Los Angeles, CA; Department of Computational Medicine (J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Section for Translational Surgical Oncology and Biobanking (C.M.L.), Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck; Ageing Epidemiology Research Unit (C.M.L.), School of Public Health, Imperial College, London, United Kingdom; and Department of Environmental Health (B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA
| | - Adrienne M Keener
- Department of Epidemiology (C.D.J.K., K.C.P., A.D.F., B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Human Genetics (C.D.J.K., J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Department of Neurology (A.M.K., J.M.B., B.R.R.), David Geffen School of Medicine, Los Angeles, CA; Parkinson's Disease Research (A.M.K.), Education, and Clinical Center, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, CA; Brain Research Institute (J.M.B.), University of California, Los Angeles, CA; Lübeck Interdisciplinary Platform for Genome Analytics (V.D., L.B.), Institutes of Neurogenetics & Cardiogenetics, University of Lübeck, Lübeck, Germany; Department of Neurology (O.-B.T.), Haukeland University Hospital, Bergen, Norway and University of Bergen, Bergen, Norway; Department of Clinical Medicine (O.-B.T.), University of Bergen, Bergen, Norway; Department of Psychology (L.B.), Centre for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway; The Norwegian Center for Movement Disorders (G.A., J.M.-G.), Stavanger University Hospital, Stavanger, Norway; Department of Neurology (G.A.), Stavanger University Hospital, Stavanger, Norway; Department of Chemistry (G.A., J.M.-G.), Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway; Department of Biostatistics (J.S.S.), UCLA Fielding School of Public Health, Los Angeles, CA; Department of Computational Medicine (J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Section for Translational Surgical Oncology and Biobanking (C.M.L.), Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck; Ageing Epidemiology Research Unit (C.M.L.), School of Public Health, Imperial College, London, United Kingdom; and Department of Environmental Health (B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA
| | - Jeff M Bronstein
- Department of Epidemiology (C.D.J.K., K.C.P., A.D.F., B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Human Genetics (C.D.J.K., J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Department of Neurology (A.M.K., J.M.B., B.R.R.), David Geffen School of Medicine, Los Angeles, CA; Parkinson's Disease Research (A.M.K.), Education, and Clinical Center, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, CA; Brain Research Institute (J.M.B.), University of California, Los Angeles, CA; Lübeck Interdisciplinary Platform for Genome Analytics (V.D., L.B.), Institutes of Neurogenetics & Cardiogenetics, University of Lübeck, Lübeck, Germany; Department of Neurology (O.-B.T.), Haukeland University Hospital, Bergen, Norway and University of Bergen, Bergen, Norway; Department of Clinical Medicine (O.-B.T.), University of Bergen, Bergen, Norway; Department of Psychology (L.B.), Centre for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway; The Norwegian Center for Movement Disorders (G.A., J.M.-G.), Stavanger University Hospital, Stavanger, Norway; Department of Neurology (G.A.), Stavanger University Hospital, Stavanger, Norway; Department of Chemistry (G.A., J.M.-G.), Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway; Department of Biostatistics (J.S.S.), UCLA Fielding School of Public Health, Los Angeles, CA; Department of Computational Medicine (J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Section for Translational Surgical Oncology and Biobanking (C.M.L.), Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck; Ageing Epidemiology Research Unit (C.M.L.), School of Public Health, Imperial College, London, United Kingdom; and Department of Environmental Health (B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA
| | - Valerija Dobricic
- Department of Epidemiology (C.D.J.K., K.C.P., A.D.F., B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Human Genetics (C.D.J.K., J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Department of Neurology (A.M.K., J.M.B., B.R.R.), David Geffen School of Medicine, Los Angeles, CA; Parkinson's Disease Research (A.M.K.), Education, and Clinical Center, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, CA; Brain Research Institute (J.M.B.), University of California, Los Angeles, CA; Lübeck Interdisciplinary Platform for Genome Analytics (V.D., L.B.), Institutes of Neurogenetics & Cardiogenetics, University of Lübeck, Lübeck, Germany; Department of Neurology (O.-B.T.), Haukeland University Hospital, Bergen, Norway and University of Bergen, Bergen, Norway; Department of Clinical Medicine (O.-B.T.), University of Bergen, Bergen, Norway; Department of Psychology (L.B.), Centre for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway; The Norwegian Center for Movement Disorders (G.A., J.M.-G.), Stavanger University Hospital, Stavanger, Norway; Department of Neurology (G.A.), Stavanger University Hospital, Stavanger, Norway; Department of Chemistry (G.A., J.M.-G.), Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway; Department of Biostatistics (J.S.S.), UCLA Fielding School of Public Health, Los Angeles, CA; Department of Computational Medicine (J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Section for Translational Surgical Oncology and Biobanking (C.M.L.), Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck; Ageing Epidemiology Research Unit (C.M.L.), School of Public Health, Imperial College, London, United Kingdom; and Department of Environmental Health (B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA
| | - Ole-Bjørn Tysnes
- Department of Epidemiology (C.D.J.K., K.C.P., A.D.F., B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Human Genetics (C.D.J.K., J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Department of Neurology (A.M.K., J.M.B., B.R.R.), David Geffen School of Medicine, Los Angeles, CA; Parkinson's Disease Research (A.M.K.), Education, and Clinical Center, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, CA; Brain Research Institute (J.M.B.), University of California, Los Angeles, CA; Lübeck Interdisciplinary Platform for Genome Analytics (V.D., L.B.), Institutes of Neurogenetics & Cardiogenetics, University of Lübeck, Lübeck, Germany; Department of Neurology (O.-B.T.), Haukeland University Hospital, Bergen, Norway and University of Bergen, Bergen, Norway; Department of Clinical Medicine (O.-B.T.), University of Bergen, Bergen, Norway; Department of Psychology (L.B.), Centre for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway; The Norwegian Center for Movement Disorders (G.A., J.M.-G.), Stavanger University Hospital, Stavanger, Norway; Department of Neurology (G.A.), Stavanger University Hospital, Stavanger, Norway; Department of Chemistry (G.A., J.M.-G.), Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway; Department of Biostatistics (J.S.S.), UCLA Fielding School of Public Health, Los Angeles, CA; Department of Computational Medicine (J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Section for Translational Surgical Oncology and Biobanking (C.M.L.), Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck; Ageing Epidemiology Research Unit (C.M.L.), School of Public Health, Imperial College, London, United Kingdom; and Department of Environmental Health (B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA
| | - Lars Bertram
- Department of Epidemiology (C.D.J.K., K.C.P., A.D.F., B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Human Genetics (C.D.J.K., J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Department of Neurology (A.M.K., J.M.B., B.R.R.), David Geffen School of Medicine, Los Angeles, CA; Parkinson's Disease Research (A.M.K.), Education, and Clinical Center, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, CA; Brain Research Institute (J.M.B.), University of California, Los Angeles, CA; Lübeck Interdisciplinary Platform for Genome Analytics (V.D., L.B.), Institutes of Neurogenetics & Cardiogenetics, University of Lübeck, Lübeck, Germany; Department of Neurology (O.-B.T.), Haukeland University Hospital, Bergen, Norway and University of Bergen, Bergen, Norway; Department of Clinical Medicine (O.-B.T.), University of Bergen, Bergen, Norway; Department of Psychology (L.B.), Centre for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway; The Norwegian Center for Movement Disorders (G.A., J.M.-G.), Stavanger University Hospital, Stavanger, Norway; Department of Neurology (G.A.), Stavanger University Hospital, Stavanger, Norway; Department of Chemistry (G.A., J.M.-G.), Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway; Department of Biostatistics (J.S.S.), UCLA Fielding School of Public Health, Los Angeles, CA; Department of Computational Medicine (J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Section for Translational Surgical Oncology and Biobanking (C.M.L.), Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck; Ageing Epidemiology Research Unit (C.M.L.), School of Public Health, Imperial College, London, United Kingdom; and Department of Environmental Health (B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA
| | - Guido Alves
- Department of Epidemiology (C.D.J.K., K.C.P., A.D.F., B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Human Genetics (C.D.J.K., J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Department of Neurology (A.M.K., J.M.B., B.R.R.), David Geffen School of Medicine, Los Angeles, CA; Parkinson's Disease Research (A.M.K.), Education, and Clinical Center, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, CA; Brain Research Institute (J.M.B.), University of California, Los Angeles, CA; Lübeck Interdisciplinary Platform for Genome Analytics (V.D., L.B.), Institutes of Neurogenetics & Cardiogenetics, University of Lübeck, Lübeck, Germany; Department of Neurology (O.-B.T.), Haukeland University Hospital, Bergen, Norway and University of Bergen, Bergen, Norway; Department of Clinical Medicine (O.-B.T.), University of Bergen, Bergen, Norway; Department of Psychology (L.B.), Centre for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway; The Norwegian Center for Movement Disorders (G.A., J.M.-G.), Stavanger University Hospital, Stavanger, Norway; Department of Neurology (G.A.), Stavanger University Hospital, Stavanger, Norway; Department of Chemistry (G.A., J.M.-G.), Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway; Department of Biostatistics (J.S.S.), UCLA Fielding School of Public Health, Los Angeles, CA; Department of Computational Medicine (J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Section for Translational Surgical Oncology and Biobanking (C.M.L.), Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck; Ageing Epidemiology Research Unit (C.M.L.), School of Public Health, Imperial College, London, United Kingdom; and Department of Environmental Health (B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA
| | - Janet S Sinsheimer
- Department of Epidemiology (C.D.J.K., K.C.P., A.D.F., B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Human Genetics (C.D.J.K., J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Department of Neurology (A.M.K., J.M.B., B.R.R.), David Geffen School of Medicine, Los Angeles, CA; Parkinson's Disease Research (A.M.K.), Education, and Clinical Center, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, CA; Brain Research Institute (J.M.B.), University of California, Los Angeles, CA; Lübeck Interdisciplinary Platform for Genome Analytics (V.D., L.B.), Institutes of Neurogenetics & Cardiogenetics, University of Lübeck, Lübeck, Germany; Department of Neurology (O.-B.T.), Haukeland University Hospital, Bergen, Norway and University of Bergen, Bergen, Norway; Department of Clinical Medicine (O.-B.T.), University of Bergen, Bergen, Norway; Department of Psychology (L.B.), Centre for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway; The Norwegian Center for Movement Disorders (G.A., J.M.-G.), Stavanger University Hospital, Stavanger, Norway; Department of Neurology (G.A.), Stavanger University Hospital, Stavanger, Norway; Department of Chemistry (G.A., J.M.-G.), Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway; Department of Biostatistics (J.S.S.), UCLA Fielding School of Public Health, Los Angeles, CA; Department of Computational Medicine (J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Section for Translational Surgical Oncology and Biobanking (C.M.L.), Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck; Ageing Epidemiology Research Unit (C.M.L.), School of Public Health, Imperial College, London, United Kingdom; and Department of Environmental Health (B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA
| | - Christina M Lill
- Department of Epidemiology (C.D.J.K., K.C.P., A.D.F., B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Human Genetics (C.D.J.K., J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Department of Neurology (A.M.K., J.M.B., B.R.R.), David Geffen School of Medicine, Los Angeles, CA; Parkinson's Disease Research (A.M.K.), Education, and Clinical Center, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, CA; Brain Research Institute (J.M.B.), University of California, Los Angeles, CA; Lübeck Interdisciplinary Platform for Genome Analytics (V.D., L.B.), Institutes of Neurogenetics & Cardiogenetics, University of Lübeck, Lübeck, Germany; Department of Neurology (O.-B.T.), Haukeland University Hospital, Bergen, Norway and University of Bergen, Bergen, Norway; Department of Clinical Medicine (O.-B.T.), University of Bergen, Bergen, Norway; Department of Psychology (L.B.), Centre for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway; The Norwegian Center for Movement Disorders (G.A., J.M.-G.), Stavanger University Hospital, Stavanger, Norway; Department of Neurology (G.A.), Stavanger University Hospital, Stavanger, Norway; Department of Chemistry (G.A., J.M.-G.), Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway; Department of Biostatistics (J.S.S.), UCLA Fielding School of Public Health, Los Angeles, CA; Department of Computational Medicine (J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Section for Translational Surgical Oncology and Biobanking (C.M.L.), Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck; Ageing Epidemiology Research Unit (C.M.L.), School of Public Health, Imperial College, London, United Kingdom; and Department of Environmental Health (B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA
| | - Jodi Maple-Grødem
- Department of Epidemiology (C.D.J.K., K.C.P., A.D.F., B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Human Genetics (C.D.J.K., J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Department of Neurology (A.M.K., J.M.B., B.R.R.), David Geffen School of Medicine, Los Angeles, CA; Parkinson's Disease Research (A.M.K.), Education, and Clinical Center, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, CA; Brain Research Institute (J.M.B.), University of California, Los Angeles, CA; Lübeck Interdisciplinary Platform for Genome Analytics (V.D., L.B.), Institutes of Neurogenetics & Cardiogenetics, University of Lübeck, Lübeck, Germany; Department of Neurology (O.-B.T.), Haukeland University Hospital, Bergen, Norway and University of Bergen, Bergen, Norway; Department of Clinical Medicine (O.-B.T.), University of Bergen, Bergen, Norway; Department of Psychology (L.B.), Centre for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway; The Norwegian Center for Movement Disorders (G.A., J.M.-G.), Stavanger University Hospital, Stavanger, Norway; Department of Neurology (G.A.), Stavanger University Hospital, Stavanger, Norway; Department of Chemistry (G.A., J.M.-G.), Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway; Department of Biostatistics (J.S.S.), UCLA Fielding School of Public Health, Los Angeles, CA; Department of Computational Medicine (J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Section for Translational Surgical Oncology and Biobanking (C.M.L.), Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck; Ageing Epidemiology Research Unit (C.M.L.), School of Public Health, Imperial College, London, United Kingdom; and Department of Environmental Health (B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA
| | - Beate R Ritz
- Department of Epidemiology (C.D.J.K., K.C.P., A.D.F., B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Human Genetics (C.D.J.K., J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Department of Neurology (A.M.K., J.M.B., B.R.R.), David Geffen School of Medicine, Los Angeles, CA; Parkinson's Disease Research (A.M.K.), Education, and Clinical Center, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, CA; Brain Research Institute (J.M.B.), University of California, Los Angeles, CA; Lübeck Interdisciplinary Platform for Genome Analytics (V.D., L.B.), Institutes of Neurogenetics & Cardiogenetics, University of Lübeck, Lübeck, Germany; Department of Neurology (O.-B.T.), Haukeland University Hospital, Bergen, Norway and University of Bergen, Bergen, Norway; Department of Clinical Medicine (O.-B.T.), University of Bergen, Bergen, Norway; Department of Psychology (L.B.), Centre for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway; The Norwegian Center for Movement Disorders (G.A., J.M.-G.), Stavanger University Hospital, Stavanger, Norway; Department of Neurology (G.A.), Stavanger University Hospital, Stavanger, Norway; Department of Chemistry (G.A., J.M.-G.), Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway; Department of Biostatistics (J.S.S.), UCLA Fielding School of Public Health, Los Angeles, CA; Department of Computational Medicine (J.S.S.), David Geffen School of Medicine, Los Angeles, CA; Section for Translational Surgical Oncology and Biobanking (C.M.L.), Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck; Ageing Epidemiology Research Unit (C.M.L.), School of Public Health, Imperial College, London, United Kingdom; and Department of Environmental Health (B.R.R.), UCLA Fielding School of Public Health, Los Angeles, CA
| |
Collapse
|
41
|
Cai L, Tu L, Yang X, Zhang Q, Tian T, Gu R, Qu X, Wang Q, Tian J. HOTAIR Accelerates Dyskinesia in a MPTP-Lesioned Mouse Model of PD via SSTR1 Methylation-Mediated ERK1/2 Axis. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:140-152. [PMID: 32927363 PMCID: PMC7494946 DOI: 10.1016/j.omtn.2020.07.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 07/06/2020] [Accepted: 07/09/2020] [Indexed: 12/26/2022]
Abstract
Homeobox transcript antisense RNA (HOTAIR), has been associated with neuroprotective effects in Parkinson's disease (PD). However, the underlying mechanisms still remain unclear. Hence, this present study attempted to clarify the functional relevance of HOTAIR in PD. We established an in vivo mouse model of PD using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and an in vitro cell model of PD by treating dopaminergic neuron MN9D cells with 1-methyl-4-phenylpyridinium species (MPP+). The expressions of somatostatin receptor 1 (SSTR1) and HOTAIR were altered to examine their effects on MN9D cell viability and apoptosis, as well as on movement impairments in MPTP-induced PD mouse model. The results indicated that HOTAIR expression was upregulated and SSTR1 was downregulated in in vivo and in vitro PD models. HOTAIR could bind to the promoter region of SSTR1, resulting in an increase of SSTR1 methylation through the recruitment of DNA methyltransferases in PD cell models. Notably, overexpression of HOTAIR and silencing of SSTR1 enhanced dopaminergic neuron apoptosis in MN9D cells and exacerbated dyskinesia in MPTP-induced PD mouse model. Collectively, overexpressed HOTAIR stimulates DNA methylation of SSTR1 to reduce SSTR1 expression, thereby accelerating dyskinesia and facilitating dopaminergic neuron apoptosis in a MPTP-lesioned PD mouse model via activation of the ERK1/2 axis.
Collapse
Affiliation(s)
- Lijun Cai
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, P.R. China
| | - Li Tu
- Department of General Practice, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, P.R. China
| | - Xiulin Yang
- Emergency Department of Internal Medicine, Guizhou Provincial People's Hospital, Guiyang 550002, P.R. China
| | - Qian Zhang
- Emergency Department of Internal Medicine, Guizhou Provincial People's Hospital, Guiyang 550002, P.R. China
| | - Tian Tian
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang 550002, P.R. China
| | - Rang Gu
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang 550002, P.R. China
| | - Xiang Qu
- Emergency Department of Internal Medicine, Guizhou Provincial People's Hospital, Guiyang 550002, P.R. China
| | - Qian Wang
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang 550002, P.R. China
| | - Jinyong Tian
- Emergency Department of Internal Medicine, Guizhou Provincial People's Hospital, Guiyang 550002, P.R. China.
| |
Collapse
|
42
|
Brunello CA, Merezhko M, Uronen RL, Huttunen HJ. Mechanisms of secretion and spreading of pathological tau protein. Cell Mol Life Sci 2020; 77:1721-1744. [PMID: 31667556 PMCID: PMC7190606 DOI: 10.1007/s00018-019-03349-1] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/10/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022]
Abstract
Accumulation of misfolded and aggregated forms of tau protein in the brain is a neuropathological hallmark of tauopathies, such as Alzheimer's disease and frontotemporal lobar degeneration. Tau aggregates have the ability to transfer from one cell to another and to induce templated misfolding and aggregation of healthy tau molecules in previously healthy cells, thereby propagating tau pathology across different brain areas in a prion-like manner. The molecular mechanisms involved in cell-to-cell transfer of tau aggregates are diverse, not mutually exclusive and only partially understood. Intracellular accumulation of misfolded tau induces several mechanisms that aim to reduce the cellular burden of aggregated proteins and also promote secretion of tau aggregates. However, tau may also be released from cells physiologically unrelated to protein aggregation. Tau secretion involves multiple vesicular and non-vesicle-mediated pathways, including secretion directly through the plasma membrane. Consequently, extracellular tau can be found in various forms, both as a free protein and in vesicles, such as exosomes and ectosomes. Once in the extracellular space, tau aggregates can be internalized by neighboring cells, both neurons and glial cells, via endocytic, pinocytic and phagocytic mechanisms. Importantly, accumulating evidence suggests that prion-like propagation of misfolding protein pathology could provide a general mechanism for disease progression in tauopathies and other related neurodegenerative diseases. Here, we review the recent literature on cellular mechanisms involved in cell-to-cell transfer of tau, with a particular focus in tau secretion.
Collapse
Affiliation(s)
- Cecilia A Brunello
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland
| | - Maria Merezhko
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland
| | - Riikka-Liisa Uronen
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland
| | - Henri J Huttunen
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland.
| |
Collapse
|
43
|
Iwaki H, Blauwendraat C, Leonard HL, Kim JJ, Liu G, Maple-Grødem J, Corvol JC, Pihlstrøm L, van Nimwegen M, Hutten SJ, Nguyen KDH, Rick J, Eberly S, Faghri F, Auinger P, Scott KM, Wijeyekoon R, Van Deerlin VM, Hernandez DG, Gibbs JR, International Parkinson’s Disease Genomics Consortium, Chitrala KN, Day-Williams AG, Brice A, Alves G, Noyce AJ, Tysnes OB, Evans JR, Breen DP, Estrada K, Wegel CE, Danjou F, Simon DK, Andreassen O, Ravina B, Toft M, Heutink P, Bloem BR, Weintraub D, Barker RA, Williams-Gray CH, van de Warrenburg BP, Van Hilten JJ, Scherzer CR, Singleton AB, Nalls MA. Genomewide association study of Parkinson's disease clinical biomarkers in 12 longitudinal patients' cohorts. Mov Disord 2019; 34:1839-1850. [PMID: 31505070 PMCID: PMC7017876 DOI: 10.1002/mds.27845] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/17/2019] [Accepted: 07/24/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Several reports have identified different patterns of Parkinson's disease progression in individuals carrying missense variants in GBA or LRRK2 genes. The overall contribution of genetic factors to the severity and progression of Parkinson's disease, however, has not been well studied. OBJECTIVES To test the association between genetic variants and the clinical features of Parkinson's disease on a genomewide scale. METHODS We accumulated individual data from 12 longitudinal cohorts in a total of 4093 patients with 22,307 observations for a median of 3.81 years. Genomewide associations were evaluated for 25 cross-sectional and longitudinal phenotypes. Specific variants of interest, including 90 recently identified disease-risk variants, were also investigated post hoc for candidate associations with these phenotypes. RESULTS Two variants were genomewide significant. Rs382940(T>A), within the intron of SLC44A1, was associated with reaching Hoehn and Yahr stage 3 or higher faster (hazard ratio 2.04 [1.58-2.62]; P value = 3.46E-8). Rs61863020(G>A), an intergenic variant and expression quantitative trait loci for α-2A adrenergic receptor, was associated with a lower prevalence of insomnia at baseline (odds ratio 0.63 [0.52-0.75]; P value = 4.74E-8). In the targeted analysis, we found 9 associations between known Parkinson's risk variants and more severe motor/cognitive symptoms. Also, we replicated previous reports of GBA coding variants (rs2230288: p.E365K; rs75548401: p.T408M) being associated with greater motor and cognitive decline over time, and an APOE E4 tagging variant (rs429358) being associated with greater cognitive deficits in patients. CONCLUSIONS We identified novel genetic factors associated with heterogeneity of Parkinson's disease. The results can be used for validation or hypothesis tests regarding Parkinson's disease. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Hirotaka Iwaki
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
- Data Tecnica International, Glen Echo, Maryland, USA
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Hampton L. Leonard
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
- Data Tecnica International, Glen Echo, Maryland, USA
| | - Jonggeol J. Kim
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Ganqiang Liu
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
- Advanced Center for Parkinson’s Disease Research, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Precision Neurology Program, Harvard Medical School, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Jodi Maple-Grødem
- The Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University in Stavanger, Stavanger, Norway
| | - Jean-Christophe Corvol
- Assistance-Publique Hôpitaux de Paris, ICM, INSERM UMRS 1127, CNRS 7225, ICM, Department of Neurology and CIC Neurosciences, Pitié-Salpêtrière Hospital, Paris, France
| | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Marlies van Nimwegen
- Radboud University Medical Centre, Donders Institute for Brain, Cognition, and Behaviour; Department of Neurology, Nijmegen, The Netherlands
| | - Samantha J. Hutten
- The Michael J. Fox Foundation for Parkinson’s Research, New York, New York, USA
| | | | - Jacqueline Rick
- Department of Neurology University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shirley Eberly
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, New York, USA
| | - Faraz Faghri
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
- Department of Computer Science, University of Illinois Urbana-Champaign, Champaign, Illinois, USA
| | - Peggy Auinger
- Department of Neurology, Center for Health + Technology, University of Rochester, Rochester, New York, USA
| | - Kirsten M. Scott
- Department of Clinical Neurosciences, University of Cambridge, John van Geest Centre for Brain Repair, Cambridge, United Kingdom
| | - Ruwani Wijeyekoon
- Department of Clinical Neurosciences, University of Cambridge, John van Geest Centre for Brain Repair, Cambridge, United Kingdom
| | - Vivianna M. Van Deerlin
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Parelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dena G. Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - J. Raphael Gibbs
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Kumaraswamy Naidu Chitrala
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Aaron G. Day-Williams
- Flagship Labs 60 Inc, Cambridge, Massachusetts, USA
- Statistical Genetics, Biogen, Cambridge, Massachusetts, USA
| | - Alexis Brice
- Institut du cerveau et de la moelle épinière ICM, Paris, France
- Sorbonne Université SU, Paris, France
- INSERM UMR1127, Paris, France
| | - Guido Alves
- The Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University in Stavanger, Stavanger, Norway
- Department of Neurology, Stavanger University Hospital, Stavanger, Norway
| | - Alastair J. Noyce
- Preventive Neurology Unit, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom
- Department of Clinical and Movement Neurosciences, UCL Institute of Neurology, London, United Kingdom
| | - Ole-Bjørn Tysnes
- Department of Neurology, Haukeland University Hospital, Bergen, Norway
- University of Bergen, Bergen, Norway
| | - Jonathan R. Evans
- Department of Neurology, Nottingham University NHS Trust, Nottingham, United Kingdom
| | - David P. Breen
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, Scotland
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland
| | - Karol Estrada
- Translational Genome Sciences, Biogen, Cambridge, Massachusetts, USA
| | - Claire E. Wegel
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana, USA
| | - Fabrice Danjou
- Institut du cerveau et de la moelle épinière ICM, Paris, France
| | - David K. Simon
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Ole Andreassen
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway, Norway
| | - Bernard Ravina
- Voyager Therapeutics, Cambridge, Massachusetts, USA
- Department of Neurology, University of Rochester School of Medicine, Rochester, New York, USA
| | - Mathias Toft
- Department of Neurology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Peter Heutink
- German Center for Neurodegenerative Diseases-Tubingen, Tuebingen, Germany
- HIH Tuebingen, Tubingen, Tuebingen, Germany
| | - Bastiaan R. Bloem
- Radboud University Medical Centre, Donders Institute for Brain, Cognition, and Behaviour; Department of Neurology, Nijmegen, The Netherlands
| | - Daniel Weintraub
- Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Veterans Affairs, Philadelphia, Pennsylvania, USA
| | - Roger A. Barker
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | | | - Bart P. van de Warrenburg
- Radboud University Medical Centre, Donders Institute for Brain, Cognition, and Behaviour; Department of Neurology, Nijmegen, The Netherlands
| | | | - Clemens R. Scherzer
- Advanced Center for Parkinson’s Disease Research, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Precision Neurology Program, Harvard Medical School, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Andrew B. Singleton
- 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
- Data Tecnica International, Glen Echo, Maryland, USA
| |
Collapse
|
44
|
Tang C, Wang W, Shi M, Zhang N, Zhou X, Li X, Ma C, Chen G, Xiang J, Gao D. Meta-Analysis of the Effects of the Catechol-O-Methyltransferase Val158/108Met Polymorphism on Parkinson's Disease Susceptibility and Cognitive Dysfunction. Front Genet 2019; 10:644. [PMID: 31354790 PMCID: PMC6639434 DOI: 10.3389/fgene.2019.00644] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 06/18/2019] [Indexed: 01/11/2023] Open
Abstract
Background: There is a continued debate and inconsistent findings in previous literature about the relationship of catechol-O-methyltransferase (COMT) and Parkinson’s disease (PD) susceptibility as well as cognitive dysfunction. To substantiate this existing gap, we comprehensively examine COMT genotype effects on the development of PD and test the hypothesis that the Met158 allele of the COMT gene is associated with cognitive dysfunction by conducting a meta-analysis review. Methods: PubMed/MEDLINE, Embase, Cochrane databases search (18/30/08) yielded 49 included studies. Data were extracted by two reviewers and included COMT genotype, publication year, diagnostic status, ancestry, the proportion of male participants, and whether genotype frequencies were consistent with Hardy–Weinberg equilibrium. Unadjusted odds ratios (ORs) were used to derive pooled estimates of PD risk overall and in subgroups defined by ethnicity, gender, and onset of disease. Moreover, the association of certain cognitive domains in PD and COMT gene type was explored. Meta-analyses were performed using random-effect models and p value–based methods. All statistical tests were two-sided. The present study was registered with PROSPERO (CRD42018087323). Results: In the current studies, we found no association between COMT Val158/108Met polymorphism and PD susceptibility. However, the gender-stratified analyses revealed marginally significant effects in heterozygote model analyses in women (P = 0.053). In addition, stratification according to onset of PD also shows significant effects of COMT Val158/108Met polymorphism on late-onset population both in recessive (P = 0.017) and allelic (P = 0.017) genetic models. For the intelligence quotient (IQ) score and Unified Parkinson Disease Rating Scale III (UPDRS III), there was no evidence for genetic association, except in subgroup analyses in Asian populations (IQ score, P = 0.016; UPDRS III, P < 0.001). Conclusion: The COMT Val158/108Met polymorphism is associated with the risk for PD in female or late-onset PD. Methionine/methionine carriers of Asian population performed significantly worse than the valine allele carriers in IQ score and UPDRS III.
Collapse
Affiliation(s)
- Chuanxi Tang
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, China
| | - Wei Wang
- Medical Technology School, Xuzhou Medical University, Xuzhou, China.,Department of Rehabilitation Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Mingyu Shi
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Na Zhang
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, China
| | - Xiaoyu Zhou
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xue Li
- School of Nursing, Xuzhou Medical University, Xuzhou, China
| | - Chengcheng Ma
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, China
| | - Gang Chen
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, China
| | - Jie Xiang
- Medical Technology School, Xuzhou Medical University, Xuzhou, China.,Department of Rehabilitation Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dianshuai Gao
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
45
|
α-synuclein-lipoprotein interactions and elevated ApoE level in cerebrospinal fluid from Parkinson's disease patients. Proc Natl Acad Sci U S A 2019; 116:15226-15235. [PMID: 31270237 PMCID: PMC6660770 DOI: 10.1073/pnas.1821409116] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Two of the most important issues in Parkinson’s disease (PD) research are the identification of mechanisms underlying α-synuclein cell-to-cell transfer in the nervous system and the discovery of early diagnostic biomarkers. Both of these issues are addressed in our current manuscript. Using multiple approaches, we present that α-synuclein interacts with lipoproteins within human cerebrospinal fluid and can be taken up by cells in such a state. Moreover, using cerebrospinal fluid samples from 3 large and independent cohorts of patients, we demonstrate that apolipoprotein E is elevated in early, not yet medicated, patients with PD. Finally, using postmortem brain tissue, we provide preliminary histological evidence that apolipoprotein E is enriched in a subpopulation of dopaminergic neurons of human substantia nigra. The progressive accumulation, aggregation, and spread of α-synuclein (αSN) are common hallmarks of Parkinson’s disease (PD) pathology. Moreover, numerous proteins interact with αSN species, influencing its toxicity in the brain. In the present study, we extended analyses of αSN-interacting proteins to cerebrospinal fluid (CSF). Using coimmunoprecipitation, followed by mass spectrometry, we found that αSN colocalize with apolipoproteins on lipoprotein vesicles. We confirmed these interactions using several methods, including the enrichment of lipoproteins with a recombinant αSN, and the subsequent uptake of prepared vesicles by human dopaminergic neuronal-like cells. Further, we report an increased level of ApoE in CSF from early PD patients compared with matched controls in 3 independent cohorts. Moreover, in contrast to controls, we observed the presence of ApoE-positive neuromelanin-containing dopaminergic neurons in substantia nigra of PD patients. In conclusion, the cooccurrence of αSN on lipoprotein vesicles, and their uptake by dopaminergic neurons along with an increase of ApoE in early PD, proposes a mechanism(s) for αSN spreading in the extracellular milieu of PD.
Collapse
|
46
|
Chen M, Xie M, Peng C, Long S. The absorption of apolipoprotein E by damaged neurons facilitates neuronal repair. Cell Biol Int 2019; 43:623-633. [PMID: 30958617 DOI: 10.1002/cbin.11135] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Min Chen
- Department of NeurologyThe First Affiliated Hospital of University of South China421001 Hengyang China
| | - Ming Xie
- Department of NeurologyThe First Affiliated Hospital of University of South China421001 Hengyang China
| | - Chao Peng
- Department of NeurologyThe First Affiliated Hospital of University of South China421001 Hengyang China
| | - Shuangqi Long
- Department of CardiologyThe Central Hospital of Yongzhou425000 Yongzhou China
| |
Collapse
|
47
|
Senkevich KA, Miliukhina IV, Pchelina SN. [The genetic predictors of cognitive impairment in Parkinson's disease]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 118:109-117. [PMID: 30251988 DOI: 10.17116/jnevro2018118081109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder that can be both sporadic and familial. A number of studies are devoted to the study of non-motor symptoms in PD today. Cognitive deficits, and especially dementia, are one of the most severe and disabling non-motor symptoms of PD. More than a quarter of patients in the early stages of PD have a moderate cognitive impairment, more than half of patients with PD develop dementia within 10 years from the date of diagnosis. Using genome-wide association studies (GWAS), a number of genes associated with cognitive impairment have been identified based on a comparison of genetic and clinical phenotypes. These genes can be divided into three groups: genes that lead to the development of PD and are inherited according to the laws of Mendel (SNCA), genes that are risk factors for PD development (GBA, MAPT) and genes associated with the development of cognitive impairment, but not with PD (COMT, APOE, BDNF). This review examines the effect of genetic variants in the above-mentioned genes on cognitive functions in patients with PD. The elucidation of the genetic basis of cognitive deficits in PD could help in choice of treatment tactics and in development of new therapeutic strategies.
Collapse
Affiliation(s)
- K A Senkevich
- Institute of Experimental Medicine, St. Petersburg, Russia; Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia; St. Petersburg Nuclear Physics Institute named by Konstantinov of NRC 'Kurchatov Institute', Gatchina, Russia
| | - I V Miliukhina
- Institute of Experimental Medicine, St. Petersburg, Russia; Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia
| | - S N Pchelina
- Institute of Experimental Medicine, St. Petersburg, Russia; Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia; St. Petersburg Nuclear Physics Institute named by Konstantinov of NRC 'Kurchatov Institute', Gatchina, Russia
| |
Collapse
|
48
|
Chuang YH, Paul KC, Sinsheimer JS, Bronstein JM, Bordelon YM, Ritz B. Genetic variants in nicotinic receptors and smoking cessation in Parkinson's disease. Parkinsonism Relat Disord 2019; 62:57-61. [PMID: 30777653 DOI: 10.1016/j.parkreldis.2019.01.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Negative associations between smoking and Parkinson's disease (PD) are well documented. While common biases may not explain this association, some studies have suggested reverse causality and ease of quitting might be an early sign of PD, possibly related to a reduced nicotinic response. We investigated nicotinic receptor (nAChR) genetics to add to our understanding of possible biologic mechanisms underlying the smoking-PD relationship. METHODS We relied on 612 patients and 691 controls enrolled in the PEG (Parkinson's Environment and Gene) study for whom we obtained information on smoking and quitting ease through interviews. Genotyping in the nAChR genes, i.e. CHRNA5-A3-B4 and CHRNB3-A6 gene regions that have been linked to smoking or quitting behaviors, were based on blood and saliva DNA samples. We assessed associations with logistic regression assuming logit-additive allelic effects and used product terms for genetic allele status and smoking or quitting assessing interactions. RESULTS As expected, we observed negative associations between smoking and PD that were strongest for current followed by former smokers. In former smokers, high quitting difficulty was negatively associated with PD risk (extremely hard vs. easy: OR = 0.62 [0.39-0.99], p = 0.05), meaning those who developed PD were able to quit smoking with less difficulty than controls. The CHRNA3 rs578776-A allele predicted quitting difficulty in smoking controls (OR = 0.53 [0.32-0.91], p = 0.02), but not in smoking PD patients (OR = 1.09 [0.61-1.95], p = 0.77). CONCLUSION Our study further corroborates previous findings that ease of quitting may be an early sign of PD onset related to a loss of nicotinic response in prodromal stages.
Collapse
Affiliation(s)
- Yu-Hsuan Chuang
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA.
| | - Kimberly C Paul
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA.
| | - Janet S Sinsheimer
- Department of Human Genetics and Biomathematics, UCLA David Geffen School of Medicine, Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, CA, USA.
| | - Jeff M Bronstein
- Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA.
| | - Yvette M Bordelon
- Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA.
| | - Beate Ritz
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA; Department of Environmental Health, UCLA Fielding School of Public Health, Los Angeles, CA, USA.
| |
Collapse
|
49
|
Paul KC, Chuang YH, Cockburn M, Bronstein JM, Horvath S, Ritz B. Organophosphate pesticide exposure and differential genome-wide DNA methylation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 645:1135-1143. [PMID: 30248838 PMCID: PMC6400463 DOI: 10.1016/j.scitotenv.2018.07.143] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 05/23/2023]
Abstract
BACKGROUND Organophosphates (OP) are widely used insecticides that acutely inhibit acetylcholinesterase enzyme activity. There is great interest in improving the understanding of molecular mechanisms related to chronic OP exposure induced toxicity. We aim to elucidate epigenetic changes associated with OP exposure, using untargeted analysis of genome-wide DNA methylation data. METHODS In a population-based case control study of Parkinson's disease (PD), we assessed ambient OP exposure via residential and workplace proximity to commercial applications. We investigated associations between OP exposure and genome-wide DNA methylation (Illumina 450 k) in 580 blood samples (342 PD patients, 238 controls) and 259 saliva samples (128 patients, 131 controls). To identify differential methylation related to OP exposure, we controlled for age, sex, European ancestry, and PD status; in addition, we stratified by disease status. RESULTS We identified 70 genome-wide significant CpGs, including cg01600516 in ALOX12 (cor = 0.27, p = 1.73E-11) and two CpGs in HLA genes, cg01655658 (cor = -0.24, p = 2.80E-09) in HLA-L (pseudogene) and cg15680603 (cor = 0.20, p = 7.94E-07) in HLA-DPA1. Among the 70 CpGs located in 41 genes, 14 were also differentially methylated in saliva samples. The most overrepresented pathway was the nicotinic acetylcholine receptor signaling pathway (fold enrichment = 15.63, p = 1.01E-03, FDR = 1.64E-01). Expanding to a larger number of genes (CpG p < 5E-04, FDR < 2.25E-01; 1077 CpGs, 662 genes), the most enriched pathway shifted to the muscarinic acetylcholine receptor 1 and 3 signaling pathway (p-value = 5.36E-04, FDR = 4.73E-02). When we stratified by PD status, results were similar. Of the 70 significant CpGs, 63 were detected among both patients and controls and 7 were only associated with OP exposure among patients. CONCLUSIONS This study finds chronic low-level OP exposure is associated with differential DNA methylation in blood and saliva, both in elderly population controls and PD patients. Our study results suggest that long-term sub-acute OP exposure influences methylation in genes enriched for muscarinic and nicotinic acetylcholine receptor pathways.
Collapse
Affiliation(s)
- Kimberly C Paul
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA.
| | - Yu-Hsuan Chuang
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Myles Cockburn
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, CA, USA
| | - Jeff M Bronstein
- Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Beate Ritz
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
50
|
Li J, Luo J, Liu L, Fu H, Tang L. The genetic association between apolipoprotein E gene polymorphism and Parkinson disease: A meta-Analysis of 47 studies. Medicine (Baltimore) 2018; 97:e12884. [PMID: 30412083 PMCID: PMC6221690 DOI: 10.1097/md.0000000000012884] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVE Although the relationship between apolipoprotein E (ApoE) gene polymorphisms and the risk of Parkinson disease (PD) has been established, the results were inconsistent and inconclusive. METHODS A comprehensive search examining the association between APOE polymorphisms and PD through PubMed, Embase, Chinese National Knowledge Infrastructure (CNKI), and Cochrane Library databases was performed without published year limited. RESULTS A total of 47 studies with 7533 cases and 14442 controls were included in present study. The results showed statistically significant association between risk factor ApoE ε4 allele and PD in Asian population (P = .003, odds ratio, OR [95% confidence interval, CI] = 1.43 [1.13,1.80]). Genotype ε2ε4 have significantly associated with PD in Asian population (P = .004, OR [95% CI] = 4.43 [1.62,12.10]). Genotype ε3ε4 was significantly associated with PD in Latin-American population (P = .01, OR [95% CI] = 1.44 [1.08,1.91]). In addition, the frequency of the genotype ε3ε4 is lower in PD group than that in the control group in Caucasian population, and the difference of genotype ε3ε4 is also statistically significant (P = .006, OR [95% CI] = 0.86 [0.77,0.96]). Although significant heterogeneity was observed among all studies, the results were shown to be stabilized by sensitive analysis. No publish bias was observed. CONCLUSIONS This meta-analysis suggests that the APOE ε4, but no ε2, might be a risk factor for PD in Asian population. Furthermore, the genotype ε2ε4 may be a susceptible factor for PD in Asian population, and the genotype ε3ε4 may be a susceptible factor for PD in both Caucasian and Latin-American populations.
Collapse
Affiliation(s)
- Jianming Li
- Department of Human Anatomy, Histology and Embryology, Institute of Neuroscience
- Department of Human Anatomy, School of Basic Medical Science, Changsha Medical University
- Department of Neurology, Xiang-ya Hospital, Central South University, Changsha, People's Republic of China
| | - Jia Luo
- Department of Human Anatomy, Histology and Embryology, Institute of Neuroscience
| | - Li Liu
- Department of Human Anatomy, Histology and Embryology, Institute of Neuroscience
| | - Hui Fu
- Department of Human Anatomy, Histology and Embryology, Institute of Neuroscience
- Department of Human Anatomy, School of Basic Medical Science, Changsha Medical University
| | - Liang Tang
- Department of Human Anatomy, Histology and Embryology, Institute of Neuroscience
| |
Collapse
|