108 results on '"Clemens R Scherzer"'
Search Results
2. Disease progression strikingly differs in research and real-world Parkinson’s populations
- Author
-
Brett K. Beaulieu-Jones, Francesca Frau, Sylvie Bozzi, Karen J. Chandross, M. Judith Peterschmitt, Caroline Cohen, Catherine Coulovrat, Dinesh Kumar, Mark J. Kruger, Scott L. Lipnick, Lane Fitzsimmons, Isaac S. Kohane, and Clemens R. Scherzer
- Subjects
Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Characterization of Parkinson’s disease (PD) progression using real-world evidence could guide clinical trial design and identify subpopulations. Efforts to curate research populations, the increasing availability of real-world data, and advances in natural language processing, particularly large language models, allow for a more granular comparison of populations than previously possible. This study includes two research populations and two real-world data-derived (RWD) populations. The research populations are the Harvard Biomarkers Study (HBS, N = 935), a longitudinal biomarkers cohort study with in-person structured study visits; and Fox Insights (N = 36,660), an online self-survey-based research study of the Michael J. Fox Foundation. Real-world cohorts are the Optum Integrated Claims-electronic health records (N = 157,475), representing wide-scale linked medical and claims data and de-identified data from Mass General Brigham (MGB, N = 22,949), an academic hospital system. Structured, de-identified electronic health records data at MGB are supplemented using a manually validated natural language processing with a large language model to extract measurements of PD progression. Motor and cognitive progression scores change more rapidly in MGB than HBS (median survival until H&Y 3: 5.6 years vs. >10, p
- Published
- 2024
- Full Text
- View/download PDF
3. Network analysis identifies SOD2 mRNA as a potential biomarker for Parkinson's disease.
- Author
-
Jose A Santiago, Clemens R Scherzer, and Judith A Potashkin
- Subjects
Medicine ,Science - Abstract
Increasing evidence indicates that Parkinson's disease (PD) and type 2 diabetes (T2DM) share dysregulated molecular networks. We identified 84 genes shared between PD and T2DM from curated disease-gene databases. Nitric oxide biosynthesis, lipid and carbohydrate metabolism, insulin secretion and inflammation were identified as common dysregulated pathways. A network prioritization approach was implemented to rank genes according to their distance to seed genes and their involvement in common biological pathways. Quantitative polymerase chain reaction assays revealed that a highly ranked gene, superoxide dismutase 2 (SOD2), is upregulated in PD patients compared to healthy controls in 192 whole blood samples from two independent clinical trials, the Harvard Biomarker Study (HBS) and the Diagnostic and Prognostic Biomarkers in Parkinson's disease (PROBE). The results from this study reinforce the idea that shared molecular networks between PD and T2DM provides an additional source of biologically meaningful biomarkers. Evaluation of this biomarker in de novo PD patients and in a larger prospective longitudinal study is warranted.
- Published
- 2014
- Full Text
- View/download PDF
4. Lysosomal dysfunction promotes cleavage and neurotoxicity of tau in vivo.
- Author
-
Vikram Khurana, Ilan Elson-Schwab, Tudor A Fulga, Katherine A Sharp, Carin A Loewen, Erin Mulkearns, Jaana Tyynelä, Clemens R Scherzer, and Mel B Feany
- Subjects
Genetics ,QH426-470 - Abstract
Expansion of the lysosomal system, including cathepsin D upregulation, is an early and prominent finding in Alzheimer's disease brain. Cell culture studies, however, have provided differing perspectives on the lysosomal connection to Alzheimer's disease, including both protective and detrimental influences. We sought to clarify and molecularly define the connection in vivo in a genetically tractable model organism. Cathepsin D is upregulated with age in a Drosophila model of Alzheimer's disease and related tauopathies. Genetic analysis reveals that cathepsin D plays a neuroprotective role because genetic ablation of cathepsin D markedly potentiates tau-induced neurotoxicity. Further, generation of a C-terminally truncated form of tau found in Alzheimer's disease patients is significantly increased in the absence of cathepsin D. We show that truncated tau has markedly increased neurotoxicity, while solubility of truncated tau is decreased. Importantly, the toxicity of truncated tau is not affected by removal of cathepsin D, providing genetic evidence that modulation of neurotoxicity by cathepsin D is mediated through C-terminal cleavage of tau. We demonstrate that removing cathepsin D in adult postmitotic neurons leads to aberrant lysosomal expansion and caspase activation in vivo, suggesting a mechanism for C-terminal truncation of tau. We also demonstrate that both cathepsin D knockout mice and cathepsin D-deficient sheep show abnormal C-terminal truncation of tau and accompanying caspase activation. Thus, caspase cleavage of tau may be a molecular mechanism through which lysosomal dysfunction and neurodegeneration are causally linked in Alzheimer's disease.
- Published
- 2010
- Full Text
- View/download PDF
5. Circular RNAs in the human brain are tailored to neuron identity and neuropsychiatric disease
- Author
-
Xianjun Dong, Yunfei Bai, Zhixiang Liao, David Gritsch, Xiaoli Liu, Tao Wang, Rebeca Borges-Monroy, Alyssa Ehrlich, Geidy E. Serrano, Mel B. Feany, Thomas G. Beach, and Clemens R. Scherzer
- Subjects
Science - Abstract
Abstract Little is known about circular RNAs (circRNAs) in specific brain cells and human neuropsychiatric disease. Here, we systematically identify over 11,039 circRNAs expressed in vulnerable dopamine and pyramidal neurons laser-captured from 190 human brains and non-neuronal cells using ultra-deep, total RNA sequencing. 1526 and 3308 circRNAs are custom-tailored to the cell identity of dopamine and pyramidal neurons and enriched in synapse pathways. 29% of Parkinson’s and 12% of Alzheimer’s disease-associated genes produced validated circRNAs. circDNAJC6, which is transcribed from a juvenile-onset Parkinson’s gene, is already dysregulated during prodromal, onset stages of common Parkinson’s disease neuropathology. Globally, addiction-associated genes preferentially produce circRNAs in dopamine neurons, autism-associated genes in pyramidal neurons, and cancers in non-neuronal cells. This study shows that circular RNAs in the human brain are tailored to neuron identity and implicate circRNA-regulated synaptic specialization in neuropsychiatric diseases.
- Published
- 2023
- Full Text
- View/download PDF
6. Plasma biomarkers for diagnosis of Alzheimer's disease and prediction of cognitive decline in individuals with mild cognitive impairment
- Author
-
Pia Kivisäkk, Becky C. Carlyle, Thadryan Sweeney, Bianca A. Trombetta, Kathryn LaCasse, Leena El-Mufti, Idil Tuncali, Lori B. Chibnik, Sudeshna Das, Clemens R. Scherzer, Keith A. Johnson, Bradford C. Dickerson, Teresa Gomez-Isla, Deborah Blacker, Derek H. Oakley, Matthew P. Frosch, Bradley T. Hyman, Anahit Aghvanyan, Pradeepthi Bathala, Christopher Campbell, George Sigal, Martin Stengelin, and Steven E. Arnold
- Subjects
biomarker ,plasma ,Alzheimer's disease ,mild cognitive impairment ,pTau181 ,neurofilament light (NfL) ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
BackgroundThe last few years have seen major advances in blood biomarkers for Alzheimer's Disease (AD) with the development of ultrasensitive immunoassays, promising to transform how we diagnose, prognose, and track progression of neurodegenerative dementias.MethodsWe evaluated a panel of four novel ultrasensitive electrochemiluminescence (ECL) immunoassays against presumed CNS derived proteins of interest in AD in plasma [phosphorylated-Tau181 (pTau181), total Tau (tTau), neurofilament light (NfL), and glial fibrillary acidic protein (GFAP)]. Two sets of banked plasma samples from the Massachusetts Alzheimer's Disease Research Center's longitudinal cohort study were examined: A longitudinal prognostic sample (n = 85) consisting of individuals with mild cognitive impairment (MCI) and 4 years of follow-up and a cross-sectional sample (n = 238) consisting of individuals with AD, other neurodegenerative diseases (OND), and normal cognition (CN).ResultsParticipants with MCI who progressed to dementia due to probable AD during follow-up had higher baseline plasma concentrations of pTau181, NfL, and GFAP compared to non-progressors. The best prognostic discrimination was observed with pTau181 (AUC = 0.83, 1.7-fold increase) and GFAP (AUC = 0.83, 1.6-fold increase). Participants with autopsy- and/or biomarker verified AD had higher plasma levels of pTau181, tTau and GFAP compared to CN and OND, while NfL was elevated in AD and further increased in OND. The best diagnostic discrimination was observed with pTau181 (AD vs CN: AUC = 0.90, 2-fold increase; AD vs. OND: AUC = 0.84, 1.5-fold increase) but tTau, NfL, and GFAP also showed good discrimination between AD and CN (AUC = 0.81–0.85; 1.5–2.2 fold increase).ConclusionsThese new ultrasensitive ECL plasma assays for pTau181, tTau, NfL, and GFAP demonstrated diagnostic utility for detection of AD. Moreover, the absolute baseline plasma levels of pTau181 and GFAP reflect cognitive decline over the next 4 years, providing prognostic information that may have utility in both clinical practice and clinical trial populations.
- Published
- 2023
- Full Text
- View/download PDF
7. DNA methylation changes associated with Parkinson’s disease progression: outcomes from the first longitudinal genome-wide methylation analysis in blood
- Author
-
Adrienne Henderson-Smith, Kathleen M. Fisch, Jianping Hua, Ganqiang Liu, Eugenia Ricciardelli, Kristen Jepsen, Mathew Huentelman, Gabriel Stalberg, Steven D. Edland, Clemens R. Scherzer, Travis Dunckley, and Paula Desplats
- Subjects
parkinson’s disease ,dna methylation ,longitudinal ,blood epigenome ,disease progression ,biomarkers ,dopamine replacement therapy ,one-carbon metabolism ,Genetics ,QH426-470 - Abstract
Parkinson’s Disease (PD) is a common neurodegenerative disorder currently diagnosed based on the presentation of characteristic movement symptoms. Unfortunately, patients exhibiting these symptoms have already undergone significant dopaminergic neuronal loss. Earlier diagnosis, aided by molecular biomarkers specific to PD, would improve overall patient care. Epigenetic mechanisms, which are modified by both environment and disease pathophysiology, are emerging as important components of neurodegeneration. Alterations to the PD methylome have been reported in epigenome-wide association studies. However, the extent to which methylation changes correlate with disease progression has not yet been reported; nor the degree to which methylation is affected by PD medication. We performed a longitudinal genome-wide methylation study surveying ~850,000 CpG sites in whole blood from 189 well-characterized PD patients and 191 control individuals obtained at baseline and at a follow-up visit ~2 y later. We identified distinct patterns of methylation in PD cases versus controls. Importantly, we identified genomic sites where methylation changes longitudinally as the disease progresses. Moreover, we identified methylation changes associated with PD pathology through the analysis of PD cases that were not exposed to anti-parkinsonian therapy. In addition, we identified methylation sites modulated by exposure to dopamine replacement drugs. These results indicate that DNA methylation is dynamic in PD and changes over time during disease progression. To the best of our knowledge, this is the first longitudinal epigenome-wide methylation analysis for Parkinson’s disease and reveals changes associated with disease progression and in response to dopaminergic medications in the blood methylome.
- Published
- 2019
- Full Text
- View/download PDF
8. Differential blood DNA methylation across Lewy body dementias
- Author
-
Chanond A. Nasamran, Anubhav Nikunj Singh Sachan, Jennifer Mott, Yuliya I. Kuras, Clemens R. Scherzer, Harvard Biomarkers Study, Eugenia Ricciardelli, Kristen Jepsen, Steven D. Edland, Kathleen M. Fisch, and Paula Desplats
- Subjects
biomarker ,blood ,cognitive scores ,dementia ,dementia with Lewy bodies (DLB) ,DNA methylation ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Introduction Dementia with Lewy bodies (DLB) and Parkinson's disease dementia (PDD) are characterized by cognitive alterations, visual hallucinations, and motor impairment. Diagnosis is based on type and timing of clinical manifestations; however, determination of clinical subtypes is challenging. The utility of blood DNA methylation as a biomarker for Lewy body disorders (LBD) is mostly unexplored. Methods We performed a cross‐sectional analysis of blood methylation in 42 DLB and 50 PDD cases applying linear models to compare groups and logistic least absolute shrinkage and selection operator regression to explore the discriminant power of methylation signals. Results DLB blood shows differential methylation compared to PDD. Some methylation changes associate with core features of LBD. Sets of probes show high predictive value to discriminate between variants. Discussion Our study is the first to explore LBD blood methylation. Despite overlapping clinical presentation, we detected differential epigenetic signatures that, if confirmed in independent cohorts, could be developed into useful biomarkers.
- Published
- 2021
- Full Text
- View/download PDF
9. Genome-wide structural variant analysis identifies risk loci for non-Alzheimer's dementias
- Author
-
Karri Kaivola, Ruth Chia, Jinhui Ding, Memoona Rasheed, Masashi Fujita, Vilas Menon, Ronald L. Walton, Ryan L. Collins, Kimberley Billingsley, Harrison Brand, Michael Talkowski, Xuefang Zhao, Ramita Dewan, Ali Stark, Anindita Ray, Sultana Solaiman, Pilar Alvarez Jerez, Laksh Malik, Ted M. Dawson, Liana S. Rosenthal, Marilyn S. Albert, Olga Pletnikova, Juan C. Troncoso, Mario Masellis, Julia Keith, Sandra E. Black, Luigi Ferrucci, Susan M. Resnick, Toshiko Tanaka, Eric Topol, Ali Torkamani, Pentti Tienari, Tatiana M. Foroud, Bernardino Ghetti, John E. Landers, Mina Ryten, Huw R. Morris, John A. Hardy, Letizia Mazzini, Sandra D'Alfonso, Cristina Moglia, Andrea Calvo, Geidy E. Serrano, Thomas G. Beach, Tanis Ferman, Neill R. Graff-Radford, Bradley F. Boeve, Zbigniew K. Wszolek, Dennis W. Dickson, Adriano Chiò, David A. Bennett, Philip L. De Jager, Owen A. Ross, Clifton L. Dalgard, J. Raphael Gibbs, Bryan J. Traynor, Sonja W. Scholz, Anthony R. Soltis, Coralie Viollet, Gauthaman Sukumar, Camille Alba, Nathaniel Lott, Elisa McGrath Martinez, Meila Tuck, Jatinder Singh, Dagmar Bacikova, Xijun Zhang, Daniel N. Hupalo, Adelani Adeleye, Matthew D. Wilkerson, Harvey B. Pollard, Ziv Gan-Or, Ekaterina Rogaeva, Alexis Brice, Suzanne Lesage, Georgia Xiromerisiou, Antonio Canosa, Adriano Chio, Giancarlo Logroscino, Gabriele Mora, Reijko Krüger, Patrick May, Daniel Alcolea, Jordi Clarimon, Juan Fortea, Isabel Gonzalez-Aramburu, Jon Infante, Carmen Lage, Alberto Lleó, Pau Pastor, Pascual Sanchez-Juan, Francesca Brett, Dag Aarsland, Safa Al-Sarraj, Johannes Attems, Steve Gentleman, Angela K. Hodges, Seth Love, Ian G. McKeith, Christopher M. Morris, Laura Palmer, Stuart Pickering-Brown, Alan J. Thomas, Claire Troakes, Matthew J. Barrett, Lynn M. Bekris, Kelley Faber, Margaret E. Flanagan, Alison Goate, David S. Goldstein, Horacio Kaufmann, Walter A. Kukull, James B. Leverenz, Grisel Lopez, Qinwen Mao, Eliezer Masliah, Edwin Monuki, Kathy L. Newell, Jose-Alberto Palma, Matthew Perkins, Alan E. Renton, Clemens R. Scherzer, Vikram G. Shakkottai, Ellen Sidransky, Nahid Tayebi, Randy Woltjer, Robert H. Baloh, Robert Bowser, James Broach, William Camu, John Cooper-Knock, Carsten Drepper, Vivian E. Drory, Travis L. Dunckley, Eva Feldman, Pietro Fratta, Glenn Gerhard, Summer B. Gibson, Jonathan D. Glass, Matthew B. Harms, Terry D. Heiman-Patterson, Lilja Jansson, Janine Kirby, Justin Kwan, Hannu Laaksovirta, Francesco Landi, Isabelle Le Ber, Serge Lumbroso, Daniel J.L. MacGowan, Nicholas J. Maragakis, Kevin Mouzat, Liisa Myllykangas, Richard W. Orrell, Lyle W. Ostrow, Roger Pamphlett, Erik Pioro, Stefan M. Pulst, John M. Ravits, Wim Robberecht, Jeffrey D. Rothstein, Michael Sendtner, Pamela J. Shaw, Katie C. Sidle, Zachary Simmons, Thor Stein, David J. Stone, Pentti J. Tienari, Miko Valori, Philip Van Damme, Vivianna M. Van Deerlin, Ludo Van Den Bosch, Lorne Zinman, Fonds National de la Recherche - FnR [sponsor], Luxembourg Centre for Systems Biomedicine (LCSB): Bioinformatics Core (R. Schneider Group) [research center], Luxembourg Centre for Systems Biomedicine (LCSB): Clinical & Experimental Neuroscience (Krüger Group) [research center], and Luxembourg Institute of Health - LIH [research center]
- Subjects
amyotrophic lateral sclerosis ,Neurologie [D14] [Sciences de la santé humaine] ,genome-wide association study ,Neurology [D14] [Human health sciences] ,case-control study ,Non-Alzheimer dementia ,resource ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,frontotemporal dementia ,structural variant ,Genetics ,non–Alzheimer's dementia ,Genetics & genetic processes [F10] [Life sciences] ,Lewy body dementia ,Structural variants ,Génétique & processus génétiques [F10] [Sciences du vivant] - Abstract
We characterized the role of structural variants, a largely unexplored type of genetic variation, in two non-Alzheimer’s dementias, namely Lewy body dementia (LBD) and frontotemporal dementia (FTD)/amyotrophic lateral sclerosis (ALS). To do this, we applied an advanced structural variant calling pipeline (GATK-SV) to short-read whole-genome sequence data from 5,213 European-ancestry cases and 4,132 controls. We discovered, replicated, and validated a deletion in TPCN1 as a novel risk locus for LBD and detected the known structural variants at the C9orf72 and MAPT loci as associated with FTD/ALS. We also identified rare pathogenic structural variants in both LBD and FTD/ALS. Finally, we assembled a catalog of structural variants that can be mined for new insights into the pathogenesis of these understudied forms of dementia.
- Published
- 2023
10. Correction to: Clinical trial-ready patient cohorts for multiple system atrophy: coupling biospecimen and iPSC banking to longitudinal deep-phenotyping
- Author
-
Alain Ndayisaba, Ariana T. Pitaro, Andrew S. Willett, Kristie A. Jones, Claudio Melo de Gusmao, Abby L. Olsen, Jisoo Kim, Eero Rissanen, Jared K. Woods, Sharan R. Srinivasan, Anna Nagy, Amanda Nagy, Merlyne Mesidor, Steven Cicero, Viharkumar Patel, Derek H. Oakley, Idil Tuncali, Katherine Taglieri-Noble, Emily C. Clark, Jordan Paulson, Richard C. Krolewski, Gary P. Ho, Albert Y. Hung, Anne-Marie Wills, Michael T. Hayes, Jason P. Macmore, Luigi Warren, Pamela G. Bower, Carol B. Langer, Lawrence R. Kellerman, Christopher W. Humphreys, Bonnie I. Glanz, Elodi J. Dielubanza, Matthew P. Frosch, Roy L. Freeman, Christopher H. Gibbons, Nadia Stefanova, Tanuja Chitnis, Howard L. Weiner, Clemens R. Scherzer, Sonja W. Scholz, Dana Vuzman, Laura M. Cox, Gregor Wenning, Jeremy D. Schmahmann, Anoopum S. Gupta, Peter Novak, Geoffrey S. Young, Mel B. Feany, Tarun Singhal, and Vikram Khurana
- Subjects
Neurology ,Neurology (clinical) - Published
- 2022
- Full Text
- View/download PDF
11. Clinical Trial-Ready Patient Cohorts for Multiple System Atrophy: Coupling Biospecimen and iPSC Banking to Longitudinal Deep-Phenotyping
- Author
-
Alain Ndayisaba, Ariana T. Pitaro, Andrew S. Willett, Kristie A. Jones, Claudio Melo de Gusmao, Abby L. Olsen, Jisoo Kim, Eero Rissanen, Jared K. Woods, Sharan R. Srinivasan, Anna Nagy, Amanda Nagy, Merlyne Mesidor, Steven Cicero, Viharkumar Patel, Derek H. Oakley, Idil Tuncali, Katherine Taglieri-Noble, Emily C. Clark, Jordan Paulson, Richard C. Krolewski, Gary P. Ho, Albert Y. Hung, Anne-Marie Wills, Michael T. Hayes, Jason P. Macmore, Luigi Warren, Pamela G. Bower, Carol B. Langer, Lawrence R. Kellerman, Christopher W. Humphreys, Bonnie I. Glanz, Elodi J. Dielubanza, Matthew P. Frosch, Roy L. Freeman, Christopher H. Gibbons, Nadia Stefanova, Tanuja Chitnis, Howard L. Weiner, Clemens R. Scherzer, Sonja W. Scholz, Dana Vuzman, Laura M. Cox, Gregor Wenning, Jeremy D. Schmahmann, Anoopum S. Gupta, Peter Novak, Geoffrey S. Young, Mel B. Feany, Tarun Singhal, and Vikram Khurana
- Subjects
Neurology ,Neurology (clinical) - Abstract
Multiple system atrophy (MSA) is a fatal neurodegenerative disease of unknown etiology characterized by widespread aggregation of the protein alpha-synuclein in neurons and glia. Its orphan status, biological relationship to Parkinson’s disease (PD), and rapid progression have sparked interest in drug development. One significant obstacle to therapeutics is disease heterogeneity. Here, we share our process of developing a clinical trial-ready cohort of MSA patients (69 patients in 2 years) within an outpatient clinical setting, and recruiting 20 of these patients into a longitudinal “n-of-few” clinical trial paradigm. First, we deeply phenotype our patients with clinical scales (UMSARS, BARS, MoCA, NMSS, and UPSIT) and tests designed to establish early differential diagnosis (including volumetric MRI, FDG-PET, MIBG scan, polysomnography, genetic testing, autonomic function tests, skin biopsy) or disease activity (PBR06-TSPO). Second, we longitudinally collect biospecimens (blood, CSF, stool) and clinical, biometric, and imaging data to generate antecedent disease-progression scores. Third, in our Mass General Brigham SCiN study (stemcellsinneurodegeneration), we generate induced pluripotent stem cell (iPSC) models from our patients, matched to biospecimens, including postmortem brain. We present 38 iPSC lines derived from MSA patients and relevant disease controls (spinocerebellar ataxia and PD, including alpha-synuclein triplication cases), 22 matched to whole-genome sequenced postmortem brain. iPSC models may facilitate matching patients to appropriate therapies, particularly in heterogeneous diseases for which patient-specific biology may elude animal models. We anticipate that deeply phenotyped and genotyped patient cohorts matched to cellular models will increase the likelihood of success in clinical trials for MSA.
- Published
- 2022
- Full Text
- View/download PDF
12. Plasma biomarkers for prognosis of cognitive decline in patients with mild cognitive impairment
- Author
-
Pia Kivisäkk, Colin Magdamo, Bianca A Trombetta, Ayush Noori, Yi kai E Kuo, Lori B Chibnik, Becky C Carlyle, Alberto Serrano-Pozo, Clemens R Scherzer, Bradley T Hyman, Sudeshna Das, and Steven E Arnold
- Subjects
General Engineering - Abstract
Plasma-based biomarkers present a promising approach in the research and clinical practice of Alzheimer's disease as they are inexpensive, accessible and minimally invasive. In particular, prognostic biomarkers of cognitive decline may aid in planning and management of clinical care. Although recent studies have demonstrated the prognostic utility of plasma biomarkers of Alzheimer pathology or neurodegeneration, such as pTau-181 and NF-L, whether other plasma biomarkers can further improve prediction of cognitive decline is undetermined. We conducted an observational cohort study to determine the prognostic utility of plasma biomarkers in predicting progression to dementia for individuals presenting with mild cognitive impairment due to probable Alzheimer's disease. We used the Olink™ Proximity Extension Assay technology to measure the level of 460 circulating proteins in banked plasma samples of all participants. We used a discovery data set comprised 60 individuals with mild cognitive impairment (30 progressors and 30 stable) and a validation data set consisting of 21 stable and 21 progressors. We developed a machine learning model to distinguish progressors from stable and used 44 proteins with significantly different plasma levels in progressors versus stable along with age, sex, education and baseline cognition as candidate features. A model with age, education, APOE genotype, baseline cognition, plasma pTau-181 and 12 plasma Olink protein biomarker levels was able to distinguish progressors from stable with 86.7% accuracy (mean area under the curve = 0.88). In the validation data set, the model accuracy was 78.6%. The Olink proteins selected by the model included those associated with vascular injury and neuroinflammation (e.g. IL-8, IL-17A, TIMP-4, MMP7). In addition, to compare these prognostic biomarkers to those that are altered in Alzheimer's disease or other types of dementia relative to controls, we analyzed samples from 20 individuals with Alzheimer, 30 with non-Alzheimer dementias and 34 with normal cognition. The proteins NF-L and PTP-1B were significantly higher in both Alzheimer and non-Alzheimer dementias compared with cognitively normal individuals. Interestingly, the prognostic markers of decline at the mild cognitive impairment stage did not overlap with those that differed between dementia and control cases. In summary, our findings suggest that plasma biomarkers of inflammation and vascular injury are associated with cognitive decline. Developing a plasma biomarker profile could aid in prognostic deliberations and identify individuals at higher risk of dementia in clinical practice.
- Published
- 2022
- Full Text
- View/download PDF
13. Accelerating Medicines Partnership: Parkinson's Disease. Genetic Resource
- Author
-
Mahdiar Sadeghi, Bradford Casey, David Vismer, Sonja W. Scholz, Mary B. Makarious, Andrew B. Singleton, Mike A. Nalls, J. Raphael Gibbs, Shameek Biswas, Barry Landin, Clemens R. Scherzer, Hampton L. Leonard, Dena G. Hernandez, Matt Bookman, Daniel Vitale, Leonie Misquitta, Dinesh Kumar, Hirotaka Iwaki, Xianjun Dong, Cornelis Blauwendraat, Clifton L. Dalgard, and Yeajin Song
- Subjects
0301 basic medicine ,Gerontology ,Open science ,Parkinson's disease ,Movement disorders ,Population ,Regular Issue Articles ,Disease ,clinical ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,open science ,medicine ,Humans ,genetics ,education ,Genotyping ,Research Articles ,education.field_of_study ,business.industry ,Parkinson Disease ,medicine.disease ,LRRK2 ,030104 developmental biology ,Neurology ,Mutation ,Neurology (clinical) ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Research Article ,Cohort study - Abstract
Background Whole‐genome sequencing data are available from several large studies across a variety of diseases and traits. However, massive storage and computation resources are required to use these data, and to achieve sufficient power for discoveries, harmonization of multiple cohorts is critical. Objectives The Accelerating Medicines Partnership Parkinson's Disease program has developed a research platform for Parkinson's disease (PD) that integrates the storage and analysis of whole‐genome sequencing data, RNA expression data, and clinical data, harmonized across multiple cohort studies. Methods The version 1 release contains whole‐genome sequencing data derived from 3941 participants from 4 cohorts. Samples underwent joint genotyping by the TOPMed Freeze 9 Variant Calling Pipeline. We performed descriptive analyses of these whole‐genome sequencing data using the Accelerating Medicines Partnership Parkinson's Disease platform. Results The clinical diagnosis of participants in version 1 release includes 2005 idiopathic PD patients, 963 healthy controls, 64 prodromal subjects, 62 clinically diagnosed PD subjects without evidence of dopamine deficit, and 705 participants of genetically enriched cohorts carrying PD risk‐associated GBA variants or LRRK2 variants, of whom 304 were affected. We did not observe significant enrichment of pathogenic variants in the idiopathic PD group, but the polygenic risk score was higher in PD both in nongenetically enriched cohorts and genetically enriched cohorts. The population analysis showed a correlation between genetically enriched cohorts and Ashkenazi Jewish ancestry. Conclusions We describe the genetic component of the Accelerating Medicines Partnership Parkinson's Disease platform, a solution to democratize data access and analysis for the PD research community. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society. This article is a U.S. Government work and is in the public domain in the USA.
- Published
- 2021
- Full Text
- View/download PDF
14. Age-associated insolubility of parkin in human midbrain is linked to redox balance and sequestration of reactive dopamine metabolites
- Author
-
Qiubo Jiang, Brian O'Nuallain, Clemens R. Scherzer, Eve C. Tsai, Jennifer A. Chan, Peggy Taylor, John Woulfe, An Tran, Ming Jin, Steve M. Callaghan, Gary S. Shaw, Masashi Takanashi, Bojan Shutinoski, Mei Zhang, Jacqueline M. Tokarew, Jasmine M. Khan, Nobutaka Hattori, Daniel N. El-Kodsi, Luigi Zecca, Alexandre Prat, Andrew B. West, Andy C. H. Ng, Xiajun Dong, Juan Li, Travis K. Fehr, Liqun Wang, Nathalie A. Lengacher, Angela P. Nguyen, David S. Park, Doo Soon Im, Julianna J. Tomlinson, Gergely Tóth, Michael G. Schlossmacher, Kathryn R. Barber, Lawrence G. Puente, Arne Holmgren, Stephanie Zandee, and Rajib Sengupta
- Subjects
Adult ,Male ,Aging ,Adolescent ,Dopamine ,Ubiquitin-Protein Ligases ,Substantia nigra ,Oxidative phosphorylation ,Parkinsonism ,medicine.disease_cause ,Parkin ,Neuromelanin ,Anti-oxidant ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,Mice ,Young Adult ,Mesencephalon ,Dopaminergic Cell ,PRKN/PARK2 gene ,medicine ,Animals ,Humans ,Young-onset Parkinson disease ,Child ,Aged ,Aged, 80 and over ,Original Paper ,Redox chemistry ,Chemistry ,Neurodegeneration ,Middle Aged ,medicine.disease ,Cell biology ,nervous system diseases ,Mice, Inbred C57BL ,Child, Preschool ,Nerve Degeneration ,Dopamine metabolism ,Female ,Neurology (clinical) ,Oxidation-Reduction ,Oxidative stress ,medicine.drug - Abstract
The mechanisms by which parkin protects the adult human brain from Parkinson disease remain incompletely understood. We hypothesized that parkin cysteines participate in redox reactions and that these are reflected in its posttranslational modifications. We found that in post mortem human brain, including in the Substantia nigra, parkin is largely insoluble after age 40 years; this transition is linked to its oxidation, such as at residues Cys95 and Cys253. In mice, oxidative stress induces posttranslational modifications of parkin cysteines that lower its solubility in vivo. Similarly, oxidation of recombinant parkin by hydrogen peroxide (H2O2) promotes its insolubility and aggregate formation, and in exchange leads to the reduction of H2O2. This thiol-based redox activity is diminished by parkin point mutants, e.g., p.C431F and p.G328E. In prkn-null mice, H2O2 levels are increased under oxidative stress conditions, such as acutely by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxin exposure or chronically due to a second, genetic hit; H2O2 levels are also significantly increased in parkin-deficient human brain. In dopamine toxicity studies, wild-type parkin, but not disease-linked mutants, protects human dopaminergic cells, in part through lowering H2O2. Parkin also neutralizes reactive, electrophilic dopamine metabolites via adduct formation, which occurs foremost at the primate-specific residue Cys95. Further, wild-type but not p.C95A-mutant parkin augments melanin formation in vitro. By probing sections of adult, human midbrain from control individuals with epitope-mapped, monoclonal antibodies, we found specific and robust parkin reactivity that co-localizes with neuromelanin pigment, frequently within LAMP-3/CD63+ lysosomes. We conclude that oxidative modifications of parkin cysteines are associated with protective outcomes, which include the reduction of H2O2, conjugation of reactive dopamine metabolites, sequestration of radicals within insoluble aggregates, and increased melanin formation. The loss of these complementary redox effects may augment oxidative stress during ageing in dopamine-producing cells of mutant PRKN allele carriers, thereby enhancing the risk of Parkinson’s-linked neurodegeneration.
- Published
- 2021
15. The Mutation Matters: <scp>CSF</scp> Profiles of <scp>GCase</scp> , Sphingolipids, α‐Synuclein in <scp> PD GBA </scp>
- Author
-
Christian Deuschle, Inga Liepelt-Scarfone, Gerrit Machetanz, Michelle M. Mielke, Ingolf Lachmann, Ruby Chiang, Claudia Schulte, Hyejung Park, Katharina Waniek, Walter Maetzler, Ingeborg Krägeloh-Mann, Stefanie Lerche, S. Pablo Sardi, Clemens R. Scherzer, Daniela Berg, Kathrin Brockmann, Xuan Mai Petterson, Isabel Wurster, Ann Kathrin Hauser, Douglas Galasko, Brit Mollenhauer, Milan Zimmermann, Benjamin Roeben, Judith Böhringer, Samantha J. Hutten, Thomas Gasser, and Bing Wang
- Subjects
0301 basic medicine ,medicine.medical_specialty ,genetics [Mutation] ,CSF ,Biology ,medicine.disease_cause ,genetics [Glucosylceramidase] ,03 medical and health sciences ,α-synuclein ,0302 clinical medicine ,genetics [Parkinson Disease] ,Internal medicine ,medicine ,Humans ,GCase ,ddc:610 ,Sphingolipids ,Mutation ,ceramides ,Wild type ,Parkinson Disease ,Sphingolipid ,030104 developmental biology ,Endocrinology ,Neurology ,genetics [alpha-Synuclein] ,alpha-Synuclein ,Glucosylceramidase ,Biomarker (medicine) ,GBA ,Neurology (clinical) ,Lactosylceramides ,Sphingomyelin ,Glucocerebrosidase ,Glucosylceramides ,030217 neurology & neurosurgery - Abstract
Background With pathway-specific trials in PD associated with variants in the glucocerebrosidase gene (PDGBA ) under way, we need markers that confirm the impact of genetic variants in patient-derived biofluids in order to allow patient stratification merely based on genetics and that might serve as biochemical read-out for target engagement. Objective To explore GBA-pathway-specific biomarker profiles cross-sectionally (TUEPAC-MIGAP, PPMI) and longitudinally (PPMI). Methods We measured enzyme activity of the lysosomal glucocerebrosidase, CSF levels of glucosylceramides (upstream substrate of glucocerebrosidase), CSF levels of ceramides (downstream product of glucocerebrosidase), lactosylceramides, sphingosines, sphingomyelin (by-products) and CSF levels of total α-synuclein in PDGBA patients compared to PDGBA_wildtype patients. Results Cross-sectionally in both cohorts and longitudinally in PPMI: (1) glucocerebrosidase activity was significantly lower in PDGBA compared to PDGBA_wildtype . (2) CSF levels of upstream substrates (glucosylceramides species) were higher in PDGBA compared to PDGBA_wildtype . (3) CSF levels of total α-synuclein were lower in PDGBA compared to PDGBA_wildtype . All of these findings were most pronounced in PDGBA with severe mutations (PDGBA_severe ). Cross-sectionally in TUEPAC-MIGAP and longitudinally in PPMI, CSF levels of downstream-products (ceramides) were higher in PDGBA_severe . Cross-sectionally in TUEPAC-MIGAP by-products sphinganine and sphingosine-1-phosphate and longitudinally in PPMI species of by-products lactosylceramides and sphingomyelin were higher in PDGBA_severe . Interpretation These findings confirm that GBA mutations have a relevant functional impact on biomarker profiles in patients. Bridging the gap between genetics and biochemical profiles now allows patient stratification for clinical trials merely based on mutation status. Importantly, all findings were most prominent in PDGBA with severe variants. © 2021 International Parkinson and Movement Disorder Society.
- Published
- 2021
- Full Text
- View/download PDF
16. Sensitive ELISA-based detection method for the mitophagy marker p-S65-Ub in human cells, autopsy brain, and blood samples
- Author
-
Xu Hou, Fabienne C. Fiesel, Jens O. Watzlawik, Michael G. Heckman, Wolfdieter Springer, Owen A. Ross, Matthew S. Goldberg, Zbigniew K. Wszolek, Michael DeTure, Sandeep Kumar Barodia, Dennis W. Dickson, Joanna Siuda, Chloe Ramnarine, Tania F. Gendron, Guojun Bu, Dominika Fricova, and Clemens R. Scherzer
- Subjects
0301 basic medicine ,autophagy ,Ubiquitin-Protein Ligases ,Autopsy ,PINK1 ,Enzyme-Linked Immunosorbent Assay ,Disease ,Parkin ,03 medical and health sciences ,Mice ,Ubiquitin ,Mitophagy ,medicine ,Animals ,Humans ,PRKN ,Molecular Biology ,030102 biochemistry & molecular biology ,biology ,Autophagy ,Brain ,Cell Biology ,medicine.disease ,nervous system diseases ,Parkinson disease ,030104 developmental biology ,Cancer research ,biology.protein ,Alzheimer's disease ,Alzheimer disease ,Toolbox ,Protein Kinases ,Research Article - Abstract
Mitochondrial dysfunction is an early, imminent event in neurodegenerative disorders including Parkinson disease (PD) and Alzheimer disease (AD). The enzymatic pair PINK1 and PRKN/Parkin recognize and transiently label damaged mitochondria with ubiquitin (Ub) phosphorylated at Ser65 (p-S65-Ub) as a signal for degradation via the autophagy-lysosome system (mitophagy). Despite its discovery in cell culture several years ago, robust and quantitative detection of altered mitophagy in vivo has remained challenging. Here we developed a sandwich ELISA targeting p-S65-Ub with the goal to assess mitophagy levels in mouse brain and in human clinical and pathological samples. We characterized five total Ub and four p-S65-Ub antibodies by several techniques and found significant differences in their ability to recognize phosphorylated Ub. The most sensitive antibody pair detected recombinant p-S65-Ub chains in the femtomolar to low picomolar range depending on the poly-Ub chain linkage. Importantly, this ELISA was able to assess very low baseline mitophagy levels in unstressed human cells and in brains from wild-type and prkn knockout mice as well as elevated p-S65-Ub levels in autopsied frontal cortex from AD patients vs. control cases. Moreover, the assay allowed detection of p-S65-Ub in blood plasma and was able to discriminate between PINK1 mutation carriers and controls. In summary, we developed a robust and sensitive tool to measure mitophagy levels in cells, tissue, and body fluids. Our data strongly support the idea that the stress-activated PINK1-PRKN mitophagy pathway is constitutively active in mice and humans under unstimulated, physiological and elevated in diseased, pathological conditions. Abbreviations: Ab: antibody; AD: Alzheimer disease; AP: alkaline phosphatase; CV: coefficient of variation; ECL: electrochemiluminescence; KO: knockout; LoB: Limit of Blank; LoD: Limit of Detection; LoQ: Limit of Quantification; MSD: meso scale discovery; PD: Parkinson disease; p-S65-PRKN: phosphorylated PRKN at serine 65; p-S65-Ub: phosphorylated ubiquitin at serine 65; Std.Dev.: standard deviation; Ub: ubiquitin; WT: wild type
- Published
- 2020
17. Longitudinal Measurements of Glucocerebrosidase activity in Parkinson’s patients
- Author
-
Pavlina Wolf, Samantha J. Hutten, Clemens R. Scherzer, Ziv Gan-Or, Ming Sum Ruby Chiang, Dandi Zheng, Sergio Pablo Sardi, Tanya Simuni, Kalpana Merchant, Chelsea Caspell-Garcia, Tatiana Foroud, Xiaokui Kate Zhang, Roy N. Alcalay, Parkinson's Progression Markers Initiative, Kelly N.H. Nudelman, Cornelis Blauwendraat, Oren A. Levy, Karolina Helesicova, Gen Li, Lana M. Chahine, and Michael J Fox Foundation
- Subjects
Male ,Gastroenterology ,DISEASE ,Glucocerebrosidase activity ,Genotype ,Dried blood ,Research Articles ,Sanger sequencing ,General Neuroscience ,Parkinson Disease ,Middle Aged ,Mental Status and Dementia Tests ,medicine.anatomical_structure ,Phenotype ,Cohort ,LYSOSOMAL STORAGE DISORDERS ,symbols ,Disease Progression ,Glucosylceramidase ,Female ,Life Sciences & Biomedicine ,RC321-571 ,Research Article ,Adult ,medicine.medical_specialty ,Heterozygote ,Clinical Neurology ,Neurosciences. Biological psychiatry. Neuropsychiatry ,symbols.namesake ,Internal medicine ,White blood cell ,medicine ,Humans ,Parkinson’s Progression Markers Initiative ,RC346-429 ,Science & Technology ,business.industry ,MUTATIONS ,Neurosciences ,1103 Clinical Sciences ,Wbc count ,PERFORMANCE ,Mutation ,Dementia ,Neurology (clinical) ,Neurology. Diseases of the nervous system ,Neurosciences & Neurology ,business ,1109 Neurosciences ,Glucocerebrosidase - Abstract
Author(s): Alcalay, Roy N; Wolf, Pavlina; Chiang, Ming Sum Ruby; Helesicova, Karolina; Zhang, Xiaokui Kate; Merchant, Kalpana; Hutten, Samantha J; Scherzer, Clemens; Caspell-Garcia, Chelsea; Blauwendraat, Cornelis; Foroud, Tatiana; Nudelman, Kelly; Gan-Or, Ziv; Simuni, Tanya; Chahine, Lana M; Levy, Oren; Zheng, Dandi; Li, Gen; Sardi, Sergio Pablo; Parkinson’s Progression Markers Initiative | Abstract: ObjectiveReduction in glucocerebrosidase (GCase; encoded by GBA) enzymatic activity has been linked to Parkinson's disease (PD). Here, we correlated GCase activity and PD phenotype in the Parkinson's Progression Markers Initiative (PPMI) cohort.MethodsWe measured GCase activity in dried blood spots from 1559 samples of participants in the inception PPMI cohort, collected in four annual visits (from baseline visit to Year-3). Participants (PD, nn=n392; controls, nn=n175) were fully sequenced for GBA variants by means of genome-wide genotyping arrays, whole-exome sequencing, whole-genome sequencing, Sanger sequencing, and RNA-sequencing.ResultsFifty-two PD participants (13.4%) and 13 (7.4%) controls carried a GBA variant. GBA status was strongly associated with GCase activity. Among noncarriers, GCase activity was similar between PD and controls. Among GBA p.E326K carriers (PD, nn=n20; controls, nn=n5), activity was significantly lower in PD carriers than control carriers (9.53nµmol/L/h vs. 11.68nµmol/L/h, Pn=n0.035). Glucocerebrosidase activity was moderately (rn=n0.45) associated with white blood cell (WBC) count. Next, we divided the noncarriers with PD to tertiles based on WBC count-corrected enzymatic activity. Members of the lower tertile had higher MDS-Unified Parkinson's Disease Rating Scale motor score in the "off" medication examination at year-III exam. Longitudinal analyses demonstrated slight reduction of activity in samples collected earlier on in the study, likely because of longer storage time.InterpretationGCase activity is associated with GBA genotype, WBC count, and among p.E326K variant carriers, with PD status. Reduced activity may also be associated with worse phenotype but longer follow up is required to confirm this observation.
- Published
- 2020
18. Large-scale pathway specific polygenic risk and transcriptomic community network analysis identifies novel functional pathways in Parkinson disease
- Author
-
Anthony R. Soltis, Ziv Gan-Or, Clifton L. Dalgard, Debra Ehrlich, Leonard H, Sara Saez-Atienzar, Cornelis Blauwendraat, Ali Torkamani, J. R. Gibbs, Sonja W. Scholz, Bryan J. Traynor, Clemens R. Scherzer, Jonggeol Jeff Kim, Jinhui Ding, Mark R. Cookson, Juan A. Botía, Matt Bookman, Andrew B. Singleton, Mike A. Nalls, Sara Bandres-Ciga, Monica Diez-Fairen, Hirotaka Iwaki, Lasse Pihlstrøm, Alastair J. Noyce, Dena G. Hernandez, Mina Ryten, Mary B. Makarious, and Faraz Faghri
- Subjects
Original Paper ,Polygenic risk ,Transcriptome community maps ,Context (language use) ,Computational biology ,Disease ,Quantitative trait locus ,Biology ,Chromatin remodeling ,Pathology and Forensic Medicine ,Parkinson disease ,Transcriptome ,Cellular and Molecular Neuroscience ,Mendelian randomization ,Neurology (clinical) ,Allele ,Signal transduction ,Gene - Abstract
Polygenic inheritance plays a central role in Parkinson disease (PD). A priority in elucidating PD etiology lies in defining the biological basis of genetic risk. Unraveling how risk leads to disruption will yield disease-modifying therapeutic targets that may be effective. Here, we utilized a high-throughput and hypothesis-free approach to determine biological processes underlying PD using the largest currently available cohorts of genetic and gene expression data from International Parkinson’s Disease Genetics Consortium (IPDGC) and the Accelerating Medicines Partnership-Parkinson’s disease initiative (AMP-PD), among other sources. We applied large-scale gene-set specific polygenic risk score (PRS) analyses to assess the role of common variation on PD risk focusing on publicly annotated gene sets representative of curated pathways. We nominated specific molecular sub-processes underlying protein misfolding and aggregation, post-translational protein modification, immune response, membrane and intracellular trafficking, lipid and vitamin metabolism, synaptic transmission, endosomal–lysosomal dysfunction, chromatin remodeling and apoptosis mediated by caspases among the main contributors to PD etiology. We assessed the impact of rare variation on PD risk in an independent cohort of whole-genome sequencing data and found evidence for a burden of rare damaging alleles in a range of processes, including neuronal transmission-related pathways and immune response. We explored enrichment linked to expression cell specificity patterns using single-cell gene expression data and demonstrated a significant risk pattern for dopaminergic neurons, serotonergic neurons, hypothalamic GABAergic neurons, and neural progenitors. Subsequently, we created a novel way of building de novo pathways by constructing a network expression community map using transcriptomic data derived from the blood of PD patients, which revealed functional enrichment in inflammatory signaling pathways, cell death machinery related processes, and dysregulation of mitochondrial homeostasis. Our analyses highlight several specific promising pathways and genes for functional prioritization and provide a cellular context in which such work should be done. Electronic supplementary material The online version of this article (10.1007/s00401-020-02181-3) contains supplementary material, which is available to authorized users.
- Published
- 2020
- Full Text
- View/download PDF
19. Associations of Lower Caffeine Intake and Plasma Urate Levels with Idiopathic Parkinson’s Disease in the Harvard Biomarkers Study
- Author
-
Clemens R. Scherzer, Anne-Marie Wills, Bradley T. Hyman, Michael A. Schwarzschild, Stephen N. Gomperts, Eric A. Macklin, Rachit Bakshi, Michael T. Hayes, Alberto Ascherio, Albert Y. Hung, and John H. Growdon
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Longitudinal study ,Parkinson's disease ,Gastroenterology ,Article ,Eating ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Sex Factors ,0302 clinical medicine ,Caffeine ,Internal medicine ,Humans ,Medicine ,Aged ,business.industry ,Parkinson Disease ,Middle Aged ,medicine.disease ,Uric Acid ,Cross-Sectional Studies ,030104 developmental biology ,chemistry ,Case-Control Studies ,Cohort ,Uric acid ,Biomarker (medicine) ,Female ,Neurology (clinical) ,Caffeine intake ,business ,Body mass index ,Biomarkers ,030217 neurology & neurosurgery - Abstract
Two purines, caffeine and urate, have been associated with a reduced risk of idiopathic Parkinson's disease (PD) in multiple cohorts and populations. The Harvard Biomarkers Study (HBS) is a longitudinal study designed to accelerate the discovery and validation of molecular diagnostic and progression markers of early-stage PD. To investigate whether these 'reduced risk' factors are associated with PD within this cohort, we conducted a cross-sectional, case-control study in 566 subjects consisting of idiopathic PD patients and healthy controls. Caffeine intake as assessed by a validated questionnaire was significantly lower in idiopathic PD patients compared to healthy controls in males (mean difference -125 mg/day, p < 0.001) but not in females (mean difference -30 mg/day, p = 0.29). A strong inverse association was also observed with plasma urate levels both in males (mean difference -0.46 mg/dL, p = 0.017) and females (mean difference -0.45 mg/dL, p = 0.001). Both analyses stratified for sex and adjusted for age, body mass index, and either urate level or caffeine consumption, respectively. These results highlight the robustness of caffeine intake and urate as factors inversely associated with idiopathic PD.
- Published
- 2020
- Full Text
- View/download PDF
20. Cerebrospinal fluid proteomics implicates the granin family in Parkinson’s disease
- Author
-
Monica Lane, Clemens R. Scherzer, Kate Zhang, Wenfei Zhang, Catherine Viel, Petra Oliva, S. Pablo Sardi, Roy N. Alcalay, Lamya S. Shihabuddin, Pavlina Wolf, Melissa S. Rotunno, and Anne-Marie Wills
- Subjects
Male ,Proteomics ,Oncology ,medicine.medical_specialty ,Parkinson's disease ,lcsh:Medicine ,Disease ,Article ,03 medical and health sciences ,0302 clinical medicine ,Cerebrospinal fluid ,Tandem Mass Spectrometry ,Internal medicine ,Chromogranins ,medicine ,Humans ,Neurodegeneration ,lcsh:Science ,Aged ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,business.industry ,lcsh:R ,Granin ,Parkinson Disease ,Middle Aged ,medicine.disease ,3. Good health ,Cohort ,Etiology ,Biomarker (medicine) ,Female ,lcsh:Q ,business ,Biomarkers ,030217 neurology & neurosurgery - Abstract
Parkinson’s disease, the most common age-related movement disorder, is a progressive neurodegenerative disease with unclear etiology. Better understanding of the underlying disease mechanism(s) is an urgent need for the development of disease-modifying therapeutics. Limited studies have been performed in large patient cohorts to identify protein alterations in cerebrospinal fluid (CSF), a proximal site to pathology. We set out to identify disease-relevant protein changes in CSF to gain insights into the etiology of Parkinson’s disease and potentially assist in disease biomarker identification. In this study, we used liquid chromatography-tandem mass spectrometry in data-independent acquisition (DIA) mode to identify Parkinson’s-relevant biomarkers in cerebrospinal fluid. We quantified 341 protein groups in two independent cohorts (n = 196) and a longitudinal cohort (n = 105 samples, representing 40 patients) consisting of Parkinson’s disease and healthy control samples from three different sources. A first cohort of 53 Parkinson’s disease and 72 control samples was analyzed, identifying 53 proteins with significant changes (p
- Published
- 2020
- Full Text
- View/download PDF
21. Australian Parkinson's Genetics Study (APGS): pilot (n=1532)
- Author
-
Svetlana Bivol, George D Mellick, Jacob Gratten, Richard Parker, Aoibhe Mulcahy, Philip E Mosley, Peter C Poortvliet, Adrian I Campos, Brittany L Mitchell, Luis M Garcia-Marin, Simone Cross, Mary Ferguson, Penelope A Lind, Danuta Z Loesch, Peter M Visscher, Sarah E Medland, Clemens R Scherzer, Nicholas G Martin, and Miguel E Rentería
- Subjects
Male ,Surveys and Questionnaires ,Australia ,Humans ,Parkinson Disease ,General Medicine ,Anxiety ,Constipation - Abstract
PurposeParkinson’s disease (PD) is a neurodegenerative disorder associated with progressive disability. While the precise aetiology is unknown, there is evidence of significant genetic and environmental influences on individual risk. The Australian Parkinson’s Genetics Study seeks to study genetic and patient-reported data from a large cohort of individuals with PD in Australia to understand the sociodemographic, genetic and environmental basis of PD susceptibility, symptoms and progression.ParticipantsIn the pilot phase reported here, 1819 participants were recruited through assisted mailouts facilitated by Services Australia based on having three or more prescriptions for anti-PD medications in their Pharmaceutical Benefits Scheme records. The average age at the time of the questionnaire was 64±6 years. We collected patient-reported information and sociodemographic variables via an online (93% of the cohort) or paper-based (7%) questionnaire. One thousand five hundred and thirty-two participants (84.2%) met all inclusion criteria, and 1499 provided a DNA sample via traditional post.Findings to date65% of participants were men, and 92% identified as being of European descent. A previous traumatic brain injury was reported by 16% of participants and was correlated with a younger age of symptom onset. At the time of the questionnaire, constipation (36% of participants), depression (34%), anxiety (17%), melanoma (16%) and diabetes (10%) were the most reported comorbid conditions.Future plansWe plan to recruit sex-matched and age-matched unaffected controls, genotype all participants and collect non-motor symptoms and cognitive function data. Future work will explore the role of genetic and environmental factors in the aetiology of PD susceptibility, onset, symptoms, and progression, including as part of international PD research consortia.
- Published
- 2022
22. Parkinson-causing mutations in LRRK2 impair the physiological tetramerization of endogenous α-synuclein in human neurons
- Author
-
Luis Fonseca-Ornelas, Jonathan M. S. Stricker, Stephanie Soriano-Cruz, Beatrice Weykopf, Ulf Dettmer, Christina R. Muratore, Clemens R. Scherzer, and Dennis J. Selkoe
- Subjects
Cellular and Molecular Neuroscience ,Neurology ,Neurology (clinical) - Abstract
α-Synuclein (αSyn) aggregation in Lewy bodies and neurites defines both familial and ‘sporadic’ Parkinson’s disease. We previously identified α-helically folded αSyn tetramers, in addition to the long-known unfolded monomers, in normal cells. PD-causing αSyn mutations decrease the tetramer:monomer (T:M) ratio, associated with αSyn hyperphosphorylation and cytotoxicity in neurons and a motor syndrome of tremor and gait deficits in transgenic mice that responds in part to L-DOPA. Here, we asked whether LRRK2 mutations, the most common genetic cause of cases previously considered sporadic PD, also alter tetramer homeostasis. Patient neurons carrying G2019S, the most prevalent LRRK2 mutation, or R1441C each had decreased T:M ratios and pSer129 hyperphosphorylation of their endogenous αSyn along with increased phosphorylation of Rab10, a widely reported substrate of LRRK2 kinase activity. Two LRRK2 kinase inhibitors normalized the T:M ratio and the hyperphosphorylation in the G2019S and R1441C patient neurons. An inhibitor of stearoyl-CoA desaturase, the rate-limiting enzyme for monounsaturated fatty acid synthesis, also restored the αSyn T:M ratio and reversed pSer129 hyperphosphorylation in both mutants. Coupled with the recent discovery that PD-causing mutations of glucocerebrosidase in Gaucher’s neurons also decrease T:M ratios, our findings indicate that three dominant genetic forms of PD involve life-long destabilization of αSyn physiological tetramers as a common pathogenic mechanism that can occur upstream of progressive neuronal synucleinopathy. Based on αSyn’s finely-tuned interaction with certain vesicles, we hypothesize that the fatty acid composition and fluidity of membranes regulate αSyn’s correct binding to highly curved membranes and subsequent assembly into metastable tetramers.
- Published
- 2022
23. Proteome profiling of cerebrospinal fluid reveals biomarker candidates for Parkinson's disease
- Author
-
Ozge Karayel, Sebastian Virreira Winter, Shalini Padmanabhan, Yuliya I. Kuras, Duc Tung Vu, Idil Tuncali, Kalpana Merchant, Anne-Marie Wills, Clemens R. Scherzer, and Matthias Mann
- Subjects
Proteomics ,Heterozygote ,Proteome ,Parkinson's disease ,Parkinson Disease ,CSF ,LRRK2 ,General Biochemistry, Genetics and Molecular Biology ,proteomics ,DIA ,Humans ,biomarker ,Biomarkers ,mass spectrometry - Abstract
Parkinson's disease (PD) is a growing burden worldwide, and there is no reliable biomarker used in clinical routines to date. Cerebrospinal fluid (CSF) is routinely collected in patients with neurological symptoms and should closely reflect alterations in PD patients’ brains. Here, we describe a scalable and sensitive mass spectrometry (MS)-based proteomics workflow for CSF proteome profiling. From two independent cohorts with over 200 individuals, our workflow reproducibly quantifies over 1,700 proteins from minimal CSF amounts. Machine learning determines OMD, CD44, VGF, PRL, and MAN2B1 to be altered in PD patients or to significantly correlate with clinical scores. We also uncover signatures of enhanced neuroinflammation in LRRK2 G2019S carriers, as indicated by increased levels of CTSS, PLD4, and HLA proteins. A comparison with our previously acquired urinary proteomes reveals a large overlap in PD-associated changes, including lysosomal proteins, opening up new avenues to improve our understanding of PD pathogenesis.
- Published
- 2022
- Full Text
- View/download PDF
24. powerEQTL: an R package and shiny application for sample size and power calculation of bulk tissue and single-cell eQTL analysis
- Author
-
Tzuu-Wang Chang, Weiliang Qiu, Xianjun Dong, Clemens R. Scherzer, Xiaoqi Li, and Scott T. Weiss
- Subjects
Statistics and Probability ,0303 health sciences ,Source code ,business.industry ,Computer science ,media_common.quotation_subject ,Genome-wide association study ,Computational biology ,Biochemistry ,Statistical power ,Computer Science Applications ,Minor allele frequency ,03 medical and health sciences ,Computational Mathematics ,0302 clinical medicine ,Computational Theory and Mathematics ,Sample size determination ,Expression quantitative trait loci ,Web application ,business ,Molecular Biology ,Allele frequency ,030217 neurology & neurosurgery ,030304 developmental biology ,media_common - Abstract
Summary Genome-wide association studies (GWAS) have revealed thousands of genetic loci for common diseases. One of the main challenges in the post-GWAS era is to understand the causality of the genetic variants. Expression quantitative trait locus (eQTL) analysis is an effective way to address this question by examining the relationship between gene expression and genetic variation in a sufficiently powered cohort. However, it is frequently a challenge to determine the sample size at which a variant with a specific allele frequency will be detected to associate with gene expression with sufficient power. This is a particularly difficult task for single-cell RNAseq studies. Therefore, a user-friendly tool to estimate statistical power for eQTL analyses in both bulk tissue and single-cell data is needed. Here, we presented an R package called powerEQTL with flexible functions to estimate power, minimal sample size or detectable minor allele frequency for both bulk tissue and single-cell eQTL analysis. A user-friendly, program-free web application is also provided, allowing users to calculate and visualize the parameters interactively. Availability and implementation The powerEQTL R package source code and online tutorial are freely available at CRAN: https://cran.r-project.org/web/packages/powerEQTL/. The R shiny application is publicly hosted at https://bwhbioinfo.shinyapps.io/powerEQTL/. Supplementary information Supplementary data are available at Bioinformatics online.
- Published
- 2021
- Full Text
- View/download PDF
25. Proteome Profiling of Cerebrospinal Fluid Reveals Novel Biomarker Candidates for Parkinson’s Disease
- Author
-
Idil Tuncali, Matthias Mann, Clemens R. Scherzer, Shalini Padmanabhan, Kalpana Merchant, Sebastian Virreira Winter, Yuliya I. Kuras, Anne-Marie Wills, Duc Tung Vu, and Ozge Karayel
- Subjects
Parkinson's disease ,business.industry ,Proteome ,Biomarker (medicine) ,Medicine ,Disease ,Biomarker discovery ,business ,Bioinformatics ,medicine.disease ,Proteomics ,LRRK2 ,CSF albumin - Abstract
SUMMARYParkinson’s disease (PD) is a growing burden worldwide, and despite ongoing efforts to find reliable biomarkers for early and differential diagnosis, prognosis and disease monitoring, there is no biofluid biomarker used in clinical routine to date. Cerebrospinal fluid (CSF) is collected often and should closely reflect structural and functional alterations in PD patients’ brains. Here we describe a scalable and sensitive mass spectrometry (MS)-based proteomics workflow for CSF proteome profiling to find specific biomarkers and identify disease-related changes in CSF protein levels in PD. From two independent cohorts consisting of more than 200 individuals, our workflow reproducibly quantified over 1,700 proteins from minimal sample amounts. Combined with machine learning, this identified a group of several proteins, including OMD, CD44, VGF, PRL, and MAN2B1 that were altered in PD patients or significantly correlate with clinical scores, indicative of disease progression. Interestingly, we uncovered signatures of enhanced neuroinflammation in patients with familial PD (LRRK2 G2019S carriers) as indicated by increased levels of CTSS, PLD4, HLA-DRA, HLA-DRB1, and HLA-DPA1. A comparison with urinary proteome changes in PD patients revealed a large overlap in protein composition PD-associated changes in these body fluids, including lysosomal factors like CTSS. Our results validate MS-based proteomics of CSF as a valuable strategy for biomarker discovery and patient stratification in a neurodegenerative disease like PD. Consistent proteomic signatures across two independent CSF cohorts and previously acquired urinary proteome profiles open up new avenues to improve our understanding of PD pathogenesis.
- Published
- 2021
- Full Text
- View/download PDF
26. Genome-wide survival study identifies a novel synaptic locus and polygenic score for cognitive progression in Parkinson's disease
- Author
-
Roger A. Barker, Claudia Trenkwalder, Sami S. Amr, Anne-Marie Wills, Christine Klein, Meike Kasten, Florence Cormier-Dequaire, Thomas G. Beach, Todd M. Herrington, Ganqiang Liu, Suzanne Lesage, Ole-Bjørn Tysnes, Clemens R. Scherzer, Jacobus J. van Hilten, Michael A. Schwarzschild, Meghan C. Campbell, Jodi Maple-Grødem, Albert Y. Hung, Zhixiang Liao, Michael T. Hayes, Jean-Christophe Corvol, Alexis Elbaz, Guido Alves, Frank Zhu, John H. Growdon, Pille Taba, Peter Heutink, Johan Marinus, Brit Mollenhauer, Joseph J. Locascio, Caroline H. Williams-Gray, Xianjun Dong, Joel S. Perlmutter, Bernard Ravina, Graziella Mangone, Alexis Brice, Ira Shoulson, Sulev Kõks, Jiajie Peng, Liu, Ganqiang [0000-0002-1921-9542], Dong, Xianjun [0000-0002-8052-9320], Maple-Grødem, Jodi [0000-0001-7142-0078], Elbaz, Alexis [0000-0001-9724-5490], Hung, Albert Y [0000-0003-3658-571X], Kõks, Sulev [0000-0001-6087-6643], Alves, Guido [0000-0003-0630-2870], Heutink, Peter [0000-0001-5218-1737], Scherzer, Clemens R [0000-0002-0567-9193], and Apollo - University of Cambridge Repository
- Subjects
Oncology ,Multifactorial Inheritance ,Parkinson's disease ,Apolipoprotein E4 ,Genome-wide association study ,Disease ,genetics [Cognition Disorders] ,genetics [Glucosylceramidase] ,Cognition ,0302 clinical medicine ,genetics [Parkinson Disease] ,Risk Factors ,Longitudinal Studies ,GBA protein, human ,genetics [Multifactorial Inheritance] ,genetics [Apolipoprotein E4] ,0303 health sciences ,education.field_of_study ,Hazard ratio ,Parkinson Disease ,genetics [Synapses] ,Disease Progression ,Glucosylceramidase ,physiopathology [Parkinson Disease] ,medicine.medical_specialty ,Population ,genetics [Mutation] ,Biology ,Article ,03 medical and health sciences ,ddc:570 ,Internal medicine ,Genetics ,medicine ,Humans ,Dementia ,Genetic Predisposition to Disease ,education ,Survival analysis ,Proportional Hazards Models ,030304 developmental biology ,Proportional hazards model ,medicine.disease ,Survival Analysis ,pathology [Parkinson Disease] ,Genetic Loci ,Mutation ,Synapses ,Cognition Disorders ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
A key driver of patients' well-being and clinical trials for Parkinson's disease (PD) is the course that the disease takes over time (progression and prognosis). To assess how genetic variation influences the progression of PD over time to dementia, a major determinant for quality of life, we performed a longitudinal genome-wide survival study of 11.2 million variants in 3,821 patients with PD over 31,053 visits. We discover RIMS2 as a progression locus and confirm this in a replicate population (hazard ratio (HR) = 4.77, P = 2.78 x 10(-11)), identify suggestive evidence for TMEM108 (HR = 2.86, P = 2.09 x 10(-8)) and WWOX (HR = 2.12, P = 2.37 x 10(-8)) as progression loci, and confirm associations for GBA (HR = 1.93, P = 0.0002) and APOE (HR = 1.48, P = 0.001). Polygenic progression scores exhibit a substantial aggregate association with dementia risk, while polygenic susceptibility scores are not predictive. This study identifies a novel synaptic locus and polygenic score for cognitive disease progression in PD and proposes diverging genetic architectures of progression and susceptibility.A genome-wide survival study identifies variants at RIMS2 associated with progression of Parkinson's disease to dementia and highlights divergence in the genetic architecture of disease onset and progression.
- Published
- 2021
- Full Text
- View/download PDF
27. Association of Dual LRRK2 G2019S and GBA Variations With Parkinson Disease Progression
- Author
-
Clemens R. Scherzer, Nir Giladi, Rachel Saunders-Pullman, Susan Bressman, Roberto A. Ortega, Avner Thaler, Anat Mirelman, Laurie J. Ozelius, Yuliya I. Kuras, Cuiling Wang, Andrew B. West, Deborah Raymond, Karen Marder, Nicole Bryant, and Roy N. Alcalay
- Subjects
Male ,medicine.medical_specialty ,Movement disorders ,Genotype ,Disease ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Internal medicine ,medicine ,Humans ,Cognitive Dysfunction ,Longitudinal Studies ,Cognitive decline ,Aged ,Original Investigation ,business.industry ,Research ,Montreal Cognitive Assessment ,Cognition ,Parkinson Disease ,General Medicine ,Middle Aged ,LRRK2 ,nervous system diseases ,Online Only ,Neurology ,Cohort ,Mutation ,Disease Progression ,Glucosylceramidase ,Female ,medicine.symptom ,business ,Glucocerebrosidase - Abstract
Key Points Question What are the associations of concurrent LRRK2 G2019S and GBA variations with clinical progression of Parkinson disease (PD)? Findings In this cohort study combining data for 1193 participants with PD from multiple studies, individuals with dual LRRK2 G2019S and GBA variation PD had a slower rate of cognitive decline than those with GBA PD alone, and this was not different from individuals with LRRK2 G2019S PD alone, supporting the notion that there is a dominant association of the LRRK2 gene in individuals with both variations. There was also a novel statistical interaction between LRRK2 G2019S and GBA variations in cognitive decline. Meaning These findings suggest that there was not a convergent deleterious association of LRRK2 and GBA variations in PD progression, as would be expected based on prior cellular studies., This cohort study examines the associations of LRRK2 G2019S and GBA variants with longitudinal cognitive and motor decline in Parkinson disease., Importance Despite a hypothesis that harboring a leucine-rich repeat kinase 2(LRRK2) G2019S variation and a glucocerebrosidase (GBA) variant would have a combined deleterious association with disease pathogenesis, milder clinical phenotypes have been reported in dual LRRK2 and GBA variations Parkinson disease (PD) than in GBA variation PD alone. Objective To evaluate the association of LRRK2 G2019S and GBA variants with longitudinal cognitive and motor decline in PD. Design, Setting, and Participants This longitudinal cohort study of continuous measures in LRRK2 PD, GBA PD, LRRK2/GBA PD, and wild-type idiopathic PD used pooled annual visit data ranging from 2004 to 2019 from the Mount Sinai Beth Israel, Parkinson Disease Biomarker Program, Harvard Biomarkers Study, Ashkenazi Jewish-LRRK2-Consortium, Parkinson Progression Marker Initiative, and SPOT-PD studies. Patients who were screened for GBA and LRRK2 variations and completed either a motor or cognitive assessment were included. Data were analyzed from May to July 2020. Main Outcomes and Measures The associations of LRRK2 G2019S and GBA genotypes on the rate of decline in Montreal Cognitive Assessment (MoCA) and Movement Disorders Society-Unified Parkinson Disease Rating Scale–Part III scores were examined using linear mixed effects models with PD duration as the time scale. Results Among 1193 individuals with PD (mean [SD] age, 66.6 [9.9] years; 490 [41.2%] women), 128 (10.7%) had GBA PD, 155 (13.0%) had LRRK2 PD, 21 (1.8%) had LRRK2/GBA PD, and 889 (74.5%) had idiopathic PD. Patients with GBA PD had faster decline in MoCA than those with LRRK2/GBA PD (B [SE], −0.31 [0.09] points/y; P
- Published
- 2021
28. The Australian Parkinson’s Genetics Study (APGS) - pilot (N = 1,532)
- Author
-
Danuta Z. Loesch, Miguel E. Rentería, Clemens R. Scherzer, Gratten J, Aoibhe Mulcahy, Philip E. Mosley, Poortvliet Pc, N. G. Martin, Adrian I. Campos, Luis M. García-Marín, George D. Mellick, Brittany L. Mitchell, Penelope A. Lind, Ferguson M, Bivol S, Simone M. Cross, Richard Parker, Peter M. Visscher, and Sarah E. Medland
- Subjects
Genetics ,business.industry ,Recall bias ,Cohort ,Etiology ,Medicine ,Anxiety ,Disease ,Medical prescription ,medicine.symptom ,Family history ,business ,Depression (differential diagnoses) - Abstract
PurposeParkinson’s disease (PD) is a neurodegenerative disorder associated with progressive disability. While the precise aetiology is unknown, there is evidence of significant genetic and environmental influences on individual risk. The Australian Parkinson’s Genetics Study (APGS) seeks to study genetic and patient-reported data from a large cohort of individuals with PD in Australia to understand the sociodemographic, genetic, and environmental basis of PD susceptibility, symptoms and progression.ParticipantsIn the pilot phase reported here, 1,819 participants were recruited through assisted mailouts facilitated by Services Australia based on having three or more prescriptions for anti-PD medications in their Pharmaceutical Benefits Scheme (PBS) records. The average age at the time of the questionnaire was 64 ± 6 years. We collected patient-reported PD information and socio-demographic variables via an online (93% of the cohort) or paper-based (7%) questionnaire. One thousand five hundred thirty-two participants (84.2%) met all inclusion criteria, and 1,499 provided a DNA sample via traditional post.Findings to date65% of participants were male, and 92% identified as being of European descent. A previous traumatic brain injury was reported by 16% of participants and was correlated with a younger age of symptom onset. At the time of the questionnaire, constipation (36% of participants), depression (34%), anxiety (17%), melanoma (16%) and diabetes (10%) were the most reported comorbid conditions.Future plansWe plan to recruit sex- and age-matched unaffected controls, genotype all participants, and collect non-motor symptoms and cognitive function data. Future work will explore the role of genetic and environmental factors in the aetiology of PD susceptibility, onset, symptoms, and progression, including as part of international PD research consortia.Article summaryStrengths and limitations of this studyWe used a time- and cost-effective recruitment method that enabled us to reach out to a random sample of individuals who have been prescribed medications for Parkinson’s disease across all over Australia to invite them to participate in this study.The identities of letter recipients remained private and confidential and were not shared with the researchers. However, those recipients who were interested and willing to participate were directed to a website where they could sign up and provide informed consent.The source database only captures individuals who have been prescribed medications to treat Parkinson’s disease in Australia and who are eligible for Medicare. Those without an official diagnosis, not receiving treatment, or not eligible for government subsidies are not included.We collected a wide range of patient-reported variables relevant to disease onset, diagnosis, symptoms, medical comorbidities, lifestyle, and family history in a large cohort of participants. However, some variables might not be as accurate as when measured by a specialist clinician.Given the 9% response rate to our single-letter invitation, there is a substantial risk of self-selection bias. Thus, patient characteristics for this cohort might differ from those of the typical population of individuals with Parkinson’s disease in Australia.
- Published
- 2021
- Full Text
- View/download PDF
29. Differential blood DNA methylation across Lewy body dementias
- Author
-
Eugenia Ricciardelli, Chanond A Nasamran, Yuliya I. Kuras, Clemens R. Scherzer, Anubhav Nikunj Singh Sachan, Steven D. Edland, Kathleen M. Fisch, Paula Desplats, Harvard Biomarkers Study, Jennifer Mott, and Kristen Jepsen
- Subjects
Aging ,REM sleep behavior disorder ,Disease ,Neurodegenerative ,Bioinformatics ,0302 clinical medicine ,Medicine ,s disease dementia ,Lewy body diseases (LBD) ,Alzheimer's Disease Related Dementias (ADRD) ,0303 health sciences ,Parkinson's Disease ,DNA methylation ,REM sleep behavior disorder (RBD) ,Methylation ,Lewy body diseases ,Psychiatry and Mental health ,Blood‐based Biomarkers ,Parkinson's disease dementia (PDD) ,Neurological ,Biomarker (medicine) ,biomarker ,dementia with Lewy bodies ,Research Article ,Lewy Body Dementia ,Parkinson's disease dementia ,03 medical and health sciences ,Clinical Research ,blood ,mental disorders ,Genetics ,Acquired Cognitive Impairment ,Dementia ,Epigenetics ,RC346-429 ,030304 developmental biology ,dementia with Lewy bodies (DLB) ,Lewy body ,epigenetics ,business.industry ,Dementia with Lewy bodies ,Human Genome ,cognitive scores ,RC952-954.6 ,Neurosciences ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Parkinson&apos ,medicine.disease ,Brain Disorders ,nervous system diseases ,Geriatrics ,Neurology (clinical) ,Neurology. Diseases of the nervous system ,business ,030217 neurology & neurosurgery ,dementia - Abstract
Introduction Dementia with Lewy bodies (DLB) and Parkinson's disease dementia (PDD) are characterized by cognitive alterations, visual hallucinations, and motor impairment. Diagnosis is based on type and timing of clinical manifestations; however, determination of clinical subtypes is challenging. The utility of blood DNA methylation as a biomarker for Lewy body disorders (LBD) is mostly unexplored. Methods We performed a cross‐sectional analysis of blood methylation in 42 DLB and 50 PDD cases applying linear models to compare groups and logistic least absolute shrinkage and selection operator regression to explore the discriminant power of methylation signals. Results DLB blood shows differential methylation compared to PDD. Some methylation changes associate with core features of LBD. Sets of probes show high predictive value to discriminate between variants. Discussion Our study is the first to explore LBD blood methylation. Despite overlapping clinical presentation, we detected differential epigenetic signatures that, if confirmed in independent cohorts, could be developed into useful biomarkers.
- Published
- 2021
30. Safety, Pharmacokinetics, and Pharmacodynamics of Oral Venglustat in Patients With Parkinson's Disease and a GBA Mutation: Results From Part 1 of the Randomised, Double-Blinded, Placebo-Controlled MOVES-PD Trial
- Author
-
Judith Peterschmitt, Hidemoto Saiki, Taku Hatano, Thomas Gasser, Stuart H. Isaacson, Sebastiaan JM Gaemers, Pascal Minini, Stéphane Saubadu, Jyoti Sharma, Samantha Walbillic, Roy N. Alcalay, Gary Cutter, Nobutaka Hattori, Günter U. Höglinger, Kenneth Marek, Anthony Schapira, Clemens R. Scherzer, Tanya Simuni, Nir Giladi, Sergio Pablo Sardi, Tanya Z. Fischer, and MOVES-PD Investigators Group
- Subjects
medicine.medical_specialty ,business.industry ,Double blinded ,Placebo ,Informed consent ,Family medicine ,Good clinical practice ,Medicine ,media_common.cataloged_instance ,In patient ,Oversight Committee ,European union ,business ,Declaration of Helsinki ,media_common - Abstract
Background: Glucocerebrosidase gene (GBA) mutations influence the risk and prognosis of Parkinson’s disease (PD), possibly through accumulation of glycosphingolipids, including glucosylceramide (GL-1). Venglustat, a glucosylceramide synthase inhibitor, is investigated in patients with PD and GBA mutations. Methods: Part 1 of the phase 2 MOVES-PD trial (NCT02906020) is a randomised, double-blinded, placebo-controlled, dose-escalation study performed at 13 sites in six countries. Eligible participants were 18–80 years old and had PD diagnosis and heterozygous GBA mutation. Per regulatory agency request, Japanese participants were evaluated separately from those outside Japan. Participants were randomised to three doses of once-daily orally administered venglustat or placebo by an interactive voice-response system, in a sequential cohort design, and were followed up to 36 weeks (52 weeks for Japanese participants). The primary endpoint was safety and tolerability of venglustat versus placebo. Secondary and exploratory endpoints included venglustat pharmacokinetics and pharmacodynamics. Findings: Between Jan 26, 2017, and Oct 17, 2018, 29 participants were randomised to venglustat (Japanese, n=9; non-Japanese, n=13) or placebo (n=3; n=4). Eight (89%) Japanese and 12 (92%) non-Japanese venglustat-treated participants experienced at least one adverse event (AE) versus two (67%) and four (100%) participants from the respective placebo groups. Most AEs were mild or moderate, and no serious AEs or deaths occurred. Two venglustat-treated non-Japanese participants discontinued due to AEs (confusional state and panic attack). Over 4 weeks, venglustat exposure in plasma and cerebrospinal fluid (CSF) increased, and GL-1 levels in plasma and CSF decreased, both in a dose-dependent manner. At the highest dose, CSF GL-1 decreased by 72·0% in Japanese and 74·3% in non-Japanese participants. Interpretation: Venglustat showed favourable safety and tolerability in MOVES-PD Part 1 and its target engagement was achieved in the CSF. Part 2 will assess the efficacy of venglustat at the dose selected in Part 1. Trial Registration: This trial is registered with ClinicalTrials.gov, number NCT02906020. Funding Statement: Supported by Sanofi. Declaration of Interests: MJP, SJMG, PM, SS, JS, SW, and SPS report receiving personal compensation as employees of Sanofi. HS reports receiving grant support from Boehringer Japan, Dainippon Sumitomo, Kyowa Hakko-Kirin, and Otsuka, and honoraria from Dainippon Sumitomo, Kyowa Hakko-Kirin, Medtronic, Novartis Pharma KK, Otsuka FP, and Takeda. TH reports receiving speaker’s honoraria and research funding from Dainippon Sumitomo, Eisai Co, Ltd, Kyowa Hakko-Kirin, Novartis Pharma KK, Otsuka, Sanofi, and Takeda, and grant support from Dainippon Sumitomo, Eisai Co, Ltd, the Setsuro Fujii Memorial, the Osaka Foundation for Promotion of Fundamental Medical Research, and the Japan Agency for Medical Research and Development under grant number 19dm0107156, the Setsuro Fujii Memorial. TG reports receiving speaker’s honoraria from MedUpdate, Novartis, Teva, and UCB Pharma, and grant support from the German Research Foundation, the German Federal Ministry of Education and Research, the European Commission, the Helmholtz Association, and the Michael J Fox Foundation; he also serves as chairman of the Scientific Advisory Board of the “Joint Programming for Neurodegenerative Diseases” program, funded by the European Commission. SHI has received honoraria for CME services, consulting, research grants, and/or promotional speaking on behalf of AbbVie, Acadia, Acorda, Adamas, Addex, Affiris, Alexva, Allergan, Amarantus, Amneal, Aptinyx, Axial, Axovant, Benevolent, Biogen, Britannia, Cadent, Cala, Cerecor, Cerevel, Cipla, Eli Lilly, Enterin, GE Healthcare, Global Kinetics, Impax, Impel, Intec Pharma, Ipsen, Jazz, Kyowa, Lundbeck, Merz, the Michael J Fox Foundation, Mitsubishi Tanabe, Neuralys, Neurocrine, Neuroderm, Parkinson Study Group, Pharma2B Prilenia, Promentis, Revance, Roche, Sanofi, Sunovion, Sun Pharma, Teva, Theravance, UCB, US WorldMeds and Zambon. RNA reports research support from the NIH, the Department of Defense, the Parkinson’s Foundation, and the Michael J Fox Foundation, and receives consultation fees from Janssen, Restorbio, Roche, and Sanofi. GC reports participating on Data and Safety Monitoring Boards for Avexis Pharmaceuticals, Biolinerx, Brainstorm Cell Therapeutics, CSL Behring, Galmed Pharmaceuticals, Horizon Pharmaceuticals, Hisun Pharmaceuticals, Mapi Pharmaceuticals, Merck, Merck/Pfizer, Neurim, Novartis, Ophazyme, Opko Biologics, Reata Pharmaceuticals, Receptos/Celgene, Sanofi-Aventis, Teva Pharmaceuticals, Vivus, NHLBI(Protocol Review Committee), and NICHD (OPRU oversight committee); reports receiving consulting fees or participating on advisory boards from Biogen, Click Therapeutics, Genentech, Genzyme, Gilgamesh Pharmaceuticals, GW Pharmaceuticals, Klein-Buendel Incorporated, Medimmune, MedDay, Novartis, Osmotica Pharmaceuticals, Perception Neurosciences, Recursion Pharmaceuticals, Roche, Somahlution, and TG Therapeutics; is employed by the University of Alabama at Birmingham; and is President of Pythagoras, Inc, a private consulting company located in Birmingham, AL, USA. NH reports receiving consulting fees for participating in advisory boards from Dainippon Sumitomo, Eisai, FP, Kyowa Hakko-Kirin, Ono, Otsuka, Tanabe-Mitsubishi, and; grant support from Biogen, Boehringer Japan, Boston Scientific, Eisai, Medtronic, Meiji, Otsuka, and Takeda; and honoraria from Dainippon, FP, Novartis, Otsuka, and Takeda. GUH reports receiving consulting fees or participating on advisory boards from AbbVie, Alzprotect, Asceneuron, Biogen, Biohaven, Lundbeck, Novartis, Retrotope, Roche, Sanofi, UCB, and the Weston Brain Institute, and has received honoraria for scientific presentations from AbbVie, Biogen, Roche, Teva, UCB, and Zambon. KM reports receiving consulting fees from Ceraspir, GE Healthcare, Invicro, LTI, Lundbeck, the Michael J Fox Foundation, Neuroderm, Neuron23, Proclara, Roche, Takeda, and UBC. AHVS reports receiving consulting fees from Inflammazome, Kyowa, Prevail, and Sanofi. CRS is named as co-inventor on a US patent application on sphingolipids biomarkers that is jointly held by Brigham & Women’s Hospital and Sanofi; has consulted for Sanofi; has collaborated with Genzyme, Lysosomal Therapies, Opko, Pfizer, and Proteome Sciences; is on the Scientific Advisory Board of the American Parkinson Disease Association; has served as Advisor to the Michael J Fox Foundation, NIH, Department of Defense, and Google; and has received funding from the NIH, the US Department of Defense, the Michael J Fox Foundation, and the American Parkinson Disease Association. TS reports receiving consulting fees and honoraria from Acadia, Adamas, Teva, and UCB Pharma; consulting fees from AbbVie, Acorda, Anavex, Allergan, NeuroDerm, PhotoPharmics, Revance, Sanofi, Sunovion, Voyager, US WorldMeds, and the Michael J Fox Foundation; and research funding from Biogen, NeuroDerm, Roche, Sanofi, NINDS, the Michael J Fox Foundation, and the Parkinson Foundation. NG reports owning stock from BOL, Lysosomal Therapeutic Ltd and Vibrant; has served as a consultant for AbbVie, Biogen, BOL, Denali, NeuroDerm, Pharma2B, Sanofi Genzyme and Vibrant; has participated in advisory boards for Biogen, Denali, NeuroDerm, Sanofi Genzyme and Sionara; has received honoraria from AbbVie, Sanofi Genzyme and the Movement Disorder Society; and has received grants from Biogen, Ionis, the European Union, the Israel Science Foundation, the Michael J Fox Foundation and the National Parkinson Foundation. TZF received compensation as an employee of Sanofi at the time the study was conducted, and is currently employed by Alnylam Pharmaceuticals (Cambridge, MA, USA). Ethics Approval Statement: This study was conducted in accordance with international ethics guidelines, including the Declaration of Helsinki and the International Council for Harmonisation guidelines for good clinical practice, and all applicable laws, rules, and regulations. All study procedures were approved by local institutional ethics review boards of participating sites, and participants provided written informed consent.
- Published
- 2021
- Full Text
- View/download PDF
31. Pathogenic huntingtin repeat expansions in patients with frontotemporal dementia and amyotrophic lateral sclerosis
- Author
-
Ramita Dewan, Ruth Chia, Jinhui Ding, Richard A. Hickman, Thor D. Stein, Yevgeniya Abramzon, Sarah Ahmed, Marya S. Sabir, Makayla K. Portley, Arianna Tucci, Kristina Ibáñez, F.N.U. Shankaracharya, Pamela Keagle, Giacomina Rossi, Paola Caroppo, Fabrizio Tagliavini, Maria L. Waldo, Per M. Johansson, Christer F. Nilsson, James B. Rowe, Luisa Benussi, Giuliano Binetti, Roberta Ghidoni, Edwin Jabbari, Coralie Viollet, Jonathan D. Glass, Andrew B. Singleton, Vincenzo Silani, Owen A. Ross, Mina Ryten, Ali Torkamani, Toshiko Tanaka, Luigi Ferrucci, Susan M. Resnick, Stuart Pickering-Brown, Christopher B. Brady, Neil Kowal, John A. Hardy, Vivianna Van Deerlin, Jean Paul Vonsattel, Matthew B. Harms, Huw R. Morris, Raffaele Ferrari, John E. Landers, Adriano Chiò, J. Raphael Gibbs, Clifton L. Dalgard, Sonja W. Scholz, Bryan J. Traynor, Adelani Adeleye, Camille Alba, Dagmar Bacikova, Daniel N. Hupalo, Elisa McGrath Martinez, Harvey B. Pollard, Gauthaman Sukumar, Anthony R. Soltis, Meila Tuck, Xijun Zhang, Matthew D. Wilkerson, Bradley N. Smith, Nicola Ticozzi, Claudia Fallini, Athina Soragia Gkazi, Simon D. Topp, Jason Kost, Emma L. Scotter, Kevin P. Kenna, Jack W. Miller, Cinzia Tiloca, Caroline Vance, Eric W. Danielson, Claire Troakes, Claudia Colombrita, Safa Al-Sarraj, Elizabeth A. Lewis, Andrew King, Daniela Calini, Viviana Pensato, Barbara Castellotti, Jacqueline de Belleroche, Frank Baas, Anneloor L.M.A. ten Asbroek, Peter C. Sapp, Diane McKenna-Yasek, Russell L. McLaughlin, Meraida Polak, Seneshaw Asress, Jesús Esteban-Pérez, José Luis Muñoz-Blanco, Zorica Stevic, Sandra D’Alfonso, Letizia Mazzini, Giacomo P. Comi, Roberto Del Bo, Mauro Ceroni, Stella Gagliardi, Giorgia Querin, Cinzia Bertolin, Wouter van Rheenen, Frank P. Diekstra, Rosa Rademakers, Marka van Blitterswijk, Kevin B. Boylan, Giuseppe Lauria, Stefano Duga, Stefania Corti, Cristina Cereda, Lucia Corrado, Gianni Sorarù, Kelly L. Williams, Garth A. Nicholson, Ian P. Blair, Claire Leblond-Manry, Guy A. Rouleau, Orla Hardiman, Karen E. Morrison, Jan H. Veldink, Leonard H. van den Berg, Ammar Al-Chalabi, Hardev Pall, Pamela J. Shaw, Martin R. Turner, Kevin Talbot, Franco Taroni, Alberto García-Redondo, Zheyang Wu, Cinzia Gellera, Antonia Ratti, Robert H. Brown, Christopher E. Shaw, John C. Ambrose, Prabhu Arumugam, Emma L. Baple, Marta Bleda, Freya Boardman-Pretty, Jeanne M. Boissiere, Christopher R. Boustred, H. Brittain, Mark J. Caulfield, Georgia C. Chan, Clare E.H. Craig, Louise C. Daugherty, Anna de Burca, Andrew Devereau, Greg Elgar, Rebecca E. Foulger, Tom Fowler, Pedro Furió-Tarí, Joanne M. Hackett, Dina Halai, Angela Hamblin, Shirley Henderson, James E. Holman, Tim J.P. Hubbard, Rob Jackson, Louise J. Jones, Dalia Kasperaviciute, Melis Kayikci, Lea Lahnstein, Kay Lawson, Sarah E.A. Leigh, Ivonne U.S. Leong, Javier F. Lopez, Fiona Maleady-Crowe, Joanne Mason, Ellen M. McDonagh, Loukas Moutsianas, Michael Mueller, Nirupa Murugaesu, Anna C. Need, Chris A. Odhams, Christine Patch, Daniel Perez-Gil, Dimitris Polychronopoulos, John Pullinger, Tahrima Rahim, Augusto Rendon, Pablo Riesgo-Ferreiro, Tim Rogers, Kevin Savage, Kushmita Sawant, Richard H. Scott, Afshan Siddiq, Alexander Sieghart, Damian Smedley, Katherine R. Smith, Alona Sosinsky, William Spooner, Helen E. Stevens, Alexander Stuckey, Razvan Sultana, Ellen R.A. Thomas, Simon R. Thompson, Carolyn Tregidgo, Emma Walsh, Sarah A. Watters, Matthew J. Welland, Eleanor Williams, Katarzyna Witkowska, Suzanne M. Wood, Magdalena Zarowiecki, Sampath Arepalli, Pavan Auluck, Robert H. Baloh, Robert Bowser, Alexis Brice, James Broach, William Camu, John Cooper-Knock, Philippe Corcia, Carsten Drepper, Vivian E. Drory, Travis L. Dunckley, Faraz Faghri, Jennifer Farren, Eva Feldman, Mary Kay Floeter, Pietro Fratta, Glenn Gerhard, Summer B. Gibson, Stephen A. Goutman, Terry D. Heiman-Patterson, Dena G. Hernandez, Ben Hoover, Lilja Jansson, Freya Kamel, Janine Kirby, Neil W. Kowall, Hannu Laaksovirta, Francesco Landi, Isabelle Le Ber, Serge Lumbroso, Daniel JL. MacGowan, Nicholas J. Maragakis, Gabriele Mora, Kevin Mouzat, Liisa Myllykangas, Mike A. Nalls, Richard W. Orrell, Lyle W. Ostrow, Roger Pamphlett, Erik Pioro, Stefan M. Pulst, John M. Ravits, Alan E. Renton, Wim Robberecht, Ian Robey, Ekaterina Rogaeva, Jeffrey D. Rothstein, Michael Sendtner, Katie C. Sidle, Zachary Simmons, David J. Stone, Pentti J. Tienari, John Q. Trojanowski, Juan C. Troncoso, Miko Valori, Philip Van Damme, Ludo Van Den Bosch, Lorne Zinman, Diego Albani, Barbara Borroni, Alessandro Padovani, Amalia Bruni, Jordi Clarimon, Oriol Dols-Icardo, Ignacio Illán-Gala, Alberto Lleó, Adrian Danek, Daniela Galimberti, Elio Scarpini, Maria Serpente, Caroline Graff, Huei-Hsin Chiang, Behzad Khoshnood, Linn Öijerstedt, Christopher M. Morris, Benedetta Nacmias, Sandro Sorbi, Jorgen E. Nielsen, Lynne E. Hjermind, Valeria Novelli, Annibale A. Puca, Pau Pastor, Ignacio Alvarez, Monica Diez-Fairen, Miquel Aguilar, Robert Perneczky, Janine Diehl-Schimd, Mina Rossi, Agustin Ruiz, Mercè Boada, Isabel Hernández, Sonia Moreno-Grau, Johannes C. Schlachetzki, Dag Aarsland, Marilyn S. Albert, Johannes Attems, Matthew J. Barrett, Thomas G. Beach, Lynn M. Bekris, David A. Bennett, Lilah M. Besser, Eileen H. Bigio, Sandra E. Black, Bradley F. Boeve, Ryan C. Bohannan, Francesca Brett, Maura Brunetti, Chad A. Caraway, Jose-Alberto Palma, Andrea Calvo, Antonio Canosa, Dennis Dickson, Charles Duyckaerts, Kelley Faber, Tanis Ferman, Margaret E. Flanagan, Gianluca Floris, Tatiana M. Foroud, Juan Fortea, Ziv Gan-Or, Steve Gentleman, Bernardino Ghetti, Jesse Raphael Gibbs, Alison Goate, David Goldstein, Isabel González-Aramburu, Neill R. Graff-Radford, Angela K. Hodges, Heng-Chen Hu, Daniel Hupalo, Jon Infante, Alex Iranzo, Scott M. Kaiser, Horacio Kaufmann, Julia Keith, Ronald C. Kim, Gregory Klein, Rejko Krüger, Walter Kukull, Amanda Kuzma, Carmen Lage, Suzanne Lesage, James B. Leverenz, Giancarlo Logroscino, Grisel Lopez, Seth Love, Qinwen Mao, Maria Jose Marti, Elisa Martinez-McGrath, Mario Masellis, Eliezer Masliah, Patrick May, Ian McKeith, Marek-Marsel Mesulam, Edwin S. Monuki, Kathy L. Newell, Lucy Norcliffe-Kaufmann, Laura Palmer, Matthew Perkins, Olga Pletnikova, Laura Molina-Porcel, Regina H. Reynolds, Eloy Rodríguez-Rodríguez, Jonathan D. Rohrer, Pascual Sanchez-Juan, Clemens R. Scherzer, Geidy E. Serrano, Vikram Shakkottai, Ellen Sidransky, Nahid Tayebi, Alan J. Thomas, Bension S. Tilley, Ronald L. Walton, Randy Woltjer, Zbigniew K. Wszolek, Georgia Xiromerisiou, Chiara Zecca, Hemali Phatnani, Justin Kwan, Dhruv Sareen, James R. Broach, Ximena Arcila-Londono, Edward B. Lee, Neil A. Shneider, Ernest Fraenkel, Noah Zaitlen, James D. Berry, Andrea Malaspina, Gregory A. Cox, Leslie M. Thompson, Steve Finkbeiner, Efthimios Dardiotis, Timothy M. Miller, Siddharthan Chandran, Suvankar Pal, Eran Hornstein, Daniel J. MacGowan, Terry Heiman-Patterson, Molly G. Hammell, Nikolaos.A. Patsopoulos, Oleg Butovsky, Joshua Dubnau, Avindra Nath, Matt Harms, Eleonora Aronica, Mary Poss, Jennifer Phillips-Cremins, John Crary, Nazem Atassi, Dale J. Lange, Darius J. Adams, Leonidas Stefanis, Marc Gotkine, Suma Babu, Towfique Raj, Sabrina Paganoni, Ophir Shalem, Colin Smith, Bin Zhang, Brent Harris, Iris Broce, Vivian Drory, John Ravits, Corey McMillan, Vilas Menon, Lani Wu, Steven Altschuler, Khaled Amar, Neil Archibald, Oliver Bandmann, Erica Capps, Alistair Church, Jan Coebergh, Alyssa Costantini, Peter Critchley, Boyd CP. Ghosh, Michele T.M. Hu, Christopher Kobylecki, P. Nigel Leigh, Carl Mann, Luke A. Massey, Uma Nath, Nicola Pavese, Dominic Paviour, Jagdish Sharma, Jenny Vaughan, HUS Neurocenter, Neurologian yksikkö, Department of Neurosciences, Clinicum, Pentti Tienari / Principal Investigator, Parkinson's UK, Human Genetics, ARD - Amsterdam Reproduction and Development, ANS - Complex Trait Genetics, Pathology, ANS - Cellular & Molecular Mechanisms, AII - Inflammatory diseases, Universidad de Cantabria, Rowe, James [0000-0001-7216-8679], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Huntington's Disease ,Pathology ,amyotrophic lateral sclerosis ,Huntingtin ,Neurology ,1702 Cognitive Sciences ,International ALS/FTD Genomics Consortium ,Neurodegenerative ,frontotemporal dementia ,3124 Neurology and psychiatry ,0302 clinical medicine ,Medicine ,2.1 Biological and endogenous factors ,Psychology ,Amyotrophic lateral sclerosis ,Aetiology ,Alzheimer's Disease Related Dementias (ADRD) ,NYGC ALS Consortium ,Huntingtin Protein ,DNA Repeat Expansion ,General Neuroscience ,Frontotemporal Dementia (FTD) ,International FTD Genetics Consortium ,whole-genome sequencing ,Frontotemporal Dementia ,Neurological ,Cognitive Sciences ,Lewy body dementia ,huntingtin ,repeat expansions ,Amyotrophic Lateral Sclerosis ,Humans ,Mutation ,Whole Genome Sequencing ,Frontotemporal dementia ,Huntington’s disease ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,FALS Sequencing Consortium ,Article ,03 medical and health sciences ,Atrophy ,Rare Diseases ,American Genome Center ,Clinical Research ,mental disorders ,Genetics ,Acquired Cognitive Impairment ,Dementia ,PROSPECT Consortium ,Neurology & Neurosurgery ,Lewy body ,business.industry ,International LBD Genomics Consortium ,Neurosciences ,3112 Neurosciences ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,nutritional and metabolic diseases ,medicine.disease ,Brain Disorders ,nervous system diseases ,030104 developmental biology ,Genomics England Research Consortium ,1701 Psychology ,ALS ,business ,1109 Neurosciences ,030217 neurology & neurosurgery - Abstract
Hannu Laaksovirta konsortion jäsenenä. The Genomics England Research Consortium, The International ALS/FTD Genomics Consortium (iAFGC), The International FTD Genetics Consortium (IFGC), The International LBD Genomics Consortium (iLBDGC), The NYGC ALS Consortium, The PROSPECT Consortium,17 James B. Rowe,17 Luisa Benussi,18 Giuliano Binetti,18,19 Roberta Ghidoni,18 Edwin Jabbari,20,21 Coralie Viollet,22 Jonathan D. Glass,23 Andrew B. Singleton,24 Vincenzo Silani,25,26 Owen A. Ross,27 Mina Ryten,8,28,29 Ali Torkamani,30 Toshiko Tanaka,31 Luigi Ferrucci,31 Susan M. Resnick,32 We examined the role of repeat expansions in the pathogenesis of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) by analyzing whole-genome sequence data from 2,442 FTD/ALS patients, 2,599 Lewy body dementia (LBD) patients, and 3,158 neurologically healthy subjects. Pathogenic expansions (range, 40?64 CAG repeats) in the huntingtin (HTT) gene were found in three (0.12%) patients diagnosed with pure FTD/ALS syndromes but were not present in the LBD or healthy cohorts. We replicated our findings in an independent collection of 3,674 FTD/ALS patients. Postmortem evaluations of two patients revealed the classical TDP-43 pathology of FTD/ALS, as well as huntingtin-positive, ubiquitin-positive aggregates in the frontal cortex. The neostriatal atrophy that pathologically defines Huntington?s disease was absent in both cases. Our findings reveal an etiological relationship between HTT repeat expansions and FTD/ALS syndromes and indicate that genetic screening of FTD/ALS patients for HTT repeat expansions should be considered. We examined the role of repeat expansions in the pathogenesis of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) by analyzing whole-genome sequence data from 2,442 FTD/ALS patients, 2,599 Lewy body dementia (LBD) patients, and 3,158 neurologically healthy subjects. Pathogenic expansions (range, 40?64 CAG repeats) in the huntingtin (HTT) gene were found in three (0.12%) patients diagnosed with pure FTD/ALS syndromes but were not present in the LBD or healthy cohorts. We replicated our findings in an independent collection of 3,674 FTD/ALS patients. Postmortem evaluations of two patients revealed the classical TDP-43 pathology of FTD/ALS, as well as huntingtin-positive, ubiquitin-positive aggregates in the frontal cortex. The neostriatal atrophy that pathologically defines Huntington?s disease was absent in both cases. Our findings reveal an etiological relationship between HTT repeat expansions and FTD/ALS syndromes and indicate that genetic screening of FTD/ALS patients for HTT repeat expansions should be considered.
- Published
- 2020
- Full Text
- View/download PDF
32. Accelerating Medicines Partnership: Parkinson’s Disease. Genetic Resource
- Author
-
Andrew B. Singleton, Xianjun Dong, David Vismer, Matt Bookman, Leonie Misquitta, Cornelis Blauwendraat, Mahdiar Sadeghi, Yeajin Song, Sonja W. Scholz, Barry Landin, Hirotaka Iwaki, J. Raphael Gibbs, Clifton L. Dalgard, Mary B. Makarious, Mike A. Nalls, Hampton L. Leonard, Dinesh Kumar, Shameek Biswas, Daniel Vitale, Clemens R. Scherzer, Bradford Casey, and Dena G. Hernandez
- Subjects
Oncology ,medicine.medical_specialty ,education.field_of_study ,Parkinson's disease ,business.industry ,Population ,Disease ,medicine.disease ,LRRK2 ,Genetic resources ,Clinical diagnosis ,Internal medicine ,medicine ,business ,education ,Genotyping ,Cohort study - Abstract
BackgroundWhole-genome sequencing (WGS) data is available from several large studies across a variety of diseases and traits. However, massive storage and computation resources are required to use these data, and, to achieve the sufficient power for discoveries, harmonization of multiple cohorts is critical.ObjectivesThe Accelerating Medicines Partnership Parkinson’s Disease (AMP PD) program has developed a research platform for Parkinson’s disease (PD) which integrates the storage and analysis of WGS data, RNA expression data, and clinical data, harmonized across multiple cohort studies.MethodsThe version 1 release contains WGS data derived from 3,941 participants from 4 cohorts. Samples underwent joint genotyping by the TOPMed Freeze 9 Variant Calling Pipeline. We performed descriptive analyses of these WGS data using the AMP PD platform.ResultsThe clinical diagnosis of participants in version 1 release includes 2,005 idiopathic PD patients, 963 healthy controls, 64 prodromal subjects, 62 clinically diagnosed PD subjects without evidence of dopamine deficit (SWEDD) and 705 participants of genetically enriched cohorts carrying PD risk associated GBA variants or LRRK2 variants in which 304 were affected. We did not observe a significant enrichment of pathogenic variants in the idiopathic PD group, but the polygenic risk score (PRS) was higher in PD both in non-genetically enriched cohorts and genetically enriched cohorts. The population analysis showed a correlation between genetically enriched cohorts and Ashkenazi Jewish ancestry.ConclusionsWe describe the genetic component of the AMP PD platform, a solution to democratise data access and analysis for the PD research community.(d) Financial Disclosure/CoI
- Published
- 2020
- Full Text
- View/download PDF
33. Understanding the effect of smoking and drinking behavior on Parkinson's disease risk: a Mendelian randomization study
- Author
-
Clemens R. Scherzer, Xianjun Dong, Carmen Domínguez-Baleón, Jue-Sheng Ong, and Miguel E. Rentería
- Subjects
0301 basic medicine ,Data Analysis ,medicine.medical_specialty ,Alcohol Drinking ,Science ,Parkinson's disease ,Protective factor ,Drinking Behavior ,Genome-wide association study ,Disease ,Polymorphism, Single Nucleotide ,Article ,Nicotine ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Mendelian randomization ,medicine ,Odds Ratio ,Tobacco Smoking ,Genetics ,Humans ,Genetic Predisposition to Disease ,Alleles ,Multidisciplinary ,business.industry ,Confounding ,ADH1B ,Parkinson Disease ,Mendelian Randomization Analysis ,Computational biology and bioinformatics ,030104 developmental biology ,Risk factors ,Medicine ,Observational study ,Disease Susceptibility ,business ,030217 neurology & neurosurgery ,Algorithms ,medicine.drug ,Genome-Wide Association Study - Abstract
Previous observational studies have identified correlations between Parkinson’s disease (PD) risk and lifestyle factors. However, whether or not those associations are causal remains unclear. To infer causality between PD risk and smoking or alcohol intake, we conducted a two-sample Mendelian randomization study using genome-wide association study summary statistics from the GWAS & Sequencing Consortium of Alcohol and Nicotine use study (1.2 million participants) and the latest meta-analysis from the International Parkinson’s Disease Genomics Consortium (37,688 PD cases and 18,618 proxy-cases). We performed sensitivity analyses, including testing for pleiotropy with MR-Egger and MR-PRESSO, and multivariable MR modeling to account for the genetic effects of competing substance use traits on PD risk. Our results revealed causal associations of alcohol intake (OR 0.79; 95% CI 0.65–0.96; p = 0.021) and smoking continuation (which compares current vs. former smokers) (OR 0.64; 95% CI 0.46–0.89; p = 0.008) with lower PD risk. Multivariable MR analyses showed that the causal association between drinks per week and PD is unlikely due to confounding by smoking behavior. Finally, frailty analyses suggested that the causal effects of both alcohol intake and smoking continuation on PD risk estimated from MR analysis are not explained by the presence of survival bias alone. Our findings support the role of smoking as a protective factor against PD, but only when comparing current vs. former smokers. Similarly, increased alcohol intake had a protective effect over PD risk, with the alcohol dehydrogenase 1B (ADH1B) locus as a potential candidate for further investigation of the mechanisms underlying this association.
- Published
- 2020
34. Genome sequencing analysis identifies new loci associated with Lewy body dementia and provides insights into the complex genetic architecture
- Author
-
Pentti J. Tienari, James B. Leverenz, Nahid Tayebi, Gabriele Mora, Bradley F. Boeve, Laura Palmer, Steve M. Gentleman, Ellen Sidransky, Pau Pastor, Liana S. Rosenthal, G. Xiromerisiou, Sara Saez-Atienzar, Francesco Landi, Scott M. Kaiser, Qinwen Mao, Claire Troakes, Peter St George-Hyslop, Andrea Calvo, Suzanne Lesage, Mario Masellis, Randy Woltjer, Marilyn S. Albert, Thomas T. Warner, Lorraine N. Clark, Gregory Klein, Charles Duyckaerts, Seth Love, Ed Monuki, Lawrence S. Honig, Kelley Faber, Dennis W. Dickson, Lucy Norcliffe-Kaufmann, Cornelis Blauwendraat, Ronald C. Kim, Kevin Morgan, Clifton L. Dalgard, Joshua T. Geiger, Ali Torkamani, Jinhui Ding, Juan Fortea, Eliezer Masliah, Ekaterina Rogaeva, Matthew H. Perkins, Clemens R. Scherzer, John Q. Trojanowski, Zbigniew K. Wszolek, Glenda M. Halliday, Jordi Clarimón, Sonja W. Scholz, Olaf Ansorge, Makayla K. Portley, Toshiko Tanaka, Mary B. Makarious, Safa Al-Sarraj, Giancarlo Logroscino, John D. Eicher, Neill R. Graff-Radford, Carmen Lage, Ziv Gan-Or, Francesca Brett, Alison Goate, Raffaele Ferrari, John C. Morris, J. Raphael Gibbs, Lynn M. Bekris, Jose-Alberto Palma, Angela K. Hodges, Regina H. Reynolds, Alexis Brice, Monica Diez-Fairen, Coralie Viollet, Patrick May, Minna Oinas, Erika Salvi, Vivianna M. Van Deerlin, Estrella Morenas-Rodríguez, Anni Moore, Zane Jaunmuktane, Eileen H. Bigio, Daniele Cusi, Douglas Galasko, Ruth Chia, Kathy L. Newell, Isabel Santana, Claudia Schulte, David Goldstein, Thomas Gasser, Owen A. Ross, Walter A. Kukull, Tatiana Foroud, Chad A. Caraway, David A. Bennett, Samreen Ahmed, Lilah M. Besser, Antonio Canosa, Daniel Alcolea, Yevgeniya Abramzon, Elisabet Londos, Laura Parkkinen, Sandra E. Black, Eric Topol, Marya S. Sabir, Olga Pletnikova, Grisel Lopez, Tanis J. Ferman, Johannes Attems, Matthew J. Barrett, Margaret E. Flanagan, Horacio Kaufmann, Stuart Pickering Brown, Jon Infante, Ryan C. Bohannan, Alberto Lleó, Eloy Rodríguez-Rodríguez, Huw R. Morris, Gianluca Floris, Ted M. Dawson, Maura Brunetti, Alan E. Renton, Andrew B. Singleton, Karen Marder, Alan J. Thomas, Pascual Sanchez-Juan, Adriano Chiò, Nigel J. Cairns, David J. Stone, Tammaryn Lashley, Mike A. Nalls, Bernardino Ghetti, Sara Bandres-Ciga, Zalak Shah, Ian G. McKeith, Susan M. Resnick, Julia Keith, Liisa Myllykangas, Diego Albani, Christopher M. Morris, Vikram Shakkottai, M. Ryten, Ronald L. Walton, Isabel González-Aramburu, Luigi Ferrucci, Bryan J. Traynor, Amanda B. Kuzma, Afina W. Lemstra, Thomas G. Beach, Juan C. Troncoso, Emil K. Gustavsson, Maurizio Grassano, John Hardy, Geidy E. Serrano, Rejko Krüger, Dag Aarsland, Bension S. Tilley, and Dena G. Hernandez
- Subjects
0303 health sciences ,Lewy body ,Disease ,Computational biology ,Biology ,medicine.disease ,DNA sequencing ,Genetic architecture ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Dementia ,Genetic risk ,Gene ,030217 neurology & neurosurgery ,030304 developmental biology ,Genetic association - Abstract
The genetic basis of Lewy body dementia (LBD) is not well understood. Here, we performed whole-genome sequencing in large cohorts of LBD cases and neurologically healthy controls to study the genetic architecture of this understudied form of dementia and to generate a resource for the scientific community. Genome-wide association analysis identified five independent risk loci, whereas genome-wide gene-aggregation tests implicated mutations in the gene GBA. Genetic risk scores demonstrate that LBD shares risk profiles and pathways with Alzheimer’s and Parkinson’s disease, providing a deeper molecular understanding of the complex genetic architecture of this age-related neurodegenerative condition.
- Published
- 2020
- Full Text
- View/download PDF
35. Plasma-borne indicators of inflammasome activity in Parkinson's disease patients
- Author
-
Clemens R. Scherzer, Alison L. Young, Katharine M. von Herrmann, William F. Hickey, Matthew C. Havrda, Angeline S. Andrew, Stephen L. Lee, Faith L Anderson, and Yuliya I. Kuras
- Subjects
0301 basic medicine ,Programmed cell death ,Parkinson's disease ,Substantia nigra ,Inflammation ,Article ,Proinflammatory cytokine ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,medicine ,RC346-429 ,business.industry ,Pyroptosis ,Inflammasome ,medicine.disease ,030104 developmental biology ,Neurology ,Immunology ,Neurology. Diseases of the nervous system ,Neurology (clinical) ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Intracellular ,Biomarkers ,medicine.drug - Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder characterized by motor and non-motor symptoms and loss of dopaminergic neurons of the substantia nigra. Inflammation and cell death are recognized aspects of PD suggesting that strategies to monitor and modify these processes may improve the management of the disease. Inflammasomes are pro-inflammatory intracellular pattern recognition complexes that couple these processes. The NLRP3 inflammasome responds to sterile triggers to initiate pro-inflammatory processes characterized by maturation of inflammatory cytokines, cytoplasmic membrane pore formation, vesicular shedding, and if unresolved, pyroptotic cell death. Histologic analysis of tissues from PD patients and individuals with nigral cell loss but no diagnosis of PD identified elevated expression of inflammasome-related proteins and activation-related “speck” formation in degenerating mesencephalic tissues compared with controls. Based on previous reports of circulating inflammasome proteins in patients suffering from heritable syndromes caused by hyper-activation of the NLRP3 inflammasome, we evaluated PD patient plasma for evidence of inflammasome activity. Multiple circulating inflammasome proteins were detected almost exclusively in extracellular vesicles indicative of ongoing inflammasome activation and pyroptosis. Analysis of plasma obtained from a multi-center cohort identified elevated plasma-borne NLRP3 associated with PD status. Our findings are consistent with others indicating inflammasome activity in neurodegenerative disorders. Findings suggest mesencephalic inflammasome protein expression as a histopathologic marker of early-stage nigral degeneration and suggest plasma-borne inflammasome-related proteins as a potentially useful class of biomarkers for patient stratification and the detection and monitoring of inflammation in PD.
- Published
- 2020
36. Large-scale pathway-specific polygenic risk, transcriptomic community networks and functional inferences in Parkinson disease
- Author
-
Mary B. Makarious, Andrew B. Singleton, Lasse Pihlstrøm, Mark R. Cookson, Mina Ryten, Alastair J. Noyce, Debra Ehrlich, Ziv Gan-Or, Jonggeol Jeff Kim, Jinhui Ding, Bryan J. Traynor, Hirotaka Iwaki, Cornelis Blauwendraat, Juan A. Botía, Dena G. Hernandez, Matthew Bookman, Ali Torkamani, Monica Diez-Fairen, Sara Saez-Atienzar, Mike A. Nalls, Sonja W. Scholz, Hampton L. Leonard, Sara Bandres-Ciga, Clemens R. Scherzer, Faraz Faghri, and Raphael Gibbs
- Subjects
0303 health sciences ,Context (language use) ,Computational biology ,Disease ,Quantitative trait locus ,Biology ,3. Good health ,Biological pathway ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Cohort ,Polygenic risk score ,Gene ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Polygenic inheritance plays a central role in Parkinson disease (PD). A priority in elucidating PD etiology lies in defining the biological basis of genetic risk. Unraveling how risk leads to disruption will yield disease-modifying therapeutic targets that may be effective. Here, we utilized a high-throughput and hypothesis-free approach to determine biological pathways underlying PD using the largest currently available cohorts of genetic data and gene expression data from International Parkinson’s Disease Genetics Consortium (IPDGC) and the Accelerating Medicines Partnership - Parkinson’s disease initiative (AMP-PD), among other sources. We placed these insights into a cellular context. We applied large-scale pathway-specific polygenic risk score (PRS) analyses to assess the role of common variation on PD risk in a cohort of 457,110 individuals by focusing on a compilation of 2,199 publicly annotated gene sets representative of curated pathways, of which we nominate 46 pathways associated with PD risk. We assessed the impact of rare variation on PD risk in an independent cohort of whole-genome sequencing data, including 4,331 individuals. We explored enrichment linked to expression cell specificity patterns using single-cell gene expression data and demonstrated a significant risk pattern for adult dopaminergic neurons, serotonergic neurons, and radial glia. Subsequently, we created a novel way of building de novo pathways by constructing a network expression community map using transcriptomic data derived from the blood of 1,612 PD patients, which revealed 54 connecting networks associated with PD. Our analyses highlight several promising pathways and genes for functional prioritization and provide a cellular context in which such work should be done.
- Published
- 2020
- Full Text
- View/download PDF
37. Genetic evidence for protective effects of smoking and drinking behavior on Parkinsons disease: A Mendelian Randomization study
- Author
-
Xianjun Dong, Jue-Sheng Ong, Miguel E. Rentería, Domínguez-Baleón C, and Clemens R. Scherzer
- Subjects
0303 health sciences ,medicine.medical_specialty ,Parkinson's disease ,business.industry ,medicine.medical_treatment ,Confounding ,Protective factor ,ADH1B ,Disease ,medicine.disease ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Mendelian randomization ,medicine ,Smoking cessation ,Observational study ,business ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
BackgroundObservational studies have identified correlations between environmental and lifestyle factors and Parkinson’s disease (PD). However, the causal direction of many of these relationships remains unclear.ObjectiveTo infer causal relationships between smoking and alcohol intake and PD.MethodsWe use a two-sample Mendelian randomization (MR) experimental design to infer causal relationships between smoking (initiation, age of initiation, heaviness, and cessation) and alcohol (drinks per week) consumption as exposure variables and PD as the health outcome. We also conduct sensitivity analyses, including testing for pleiotropic effects MR-Egger and MR-PRESSO, and multivariable MR to jointly model the effects of drinking and smoking behavior on PD risk.ResultsBoth alcohol intake (OR = 0.69; 95% CI 0.56-0.86; p=0.001). and smoking cessation (comparing current vs. former smokers) (IVW OR = 0.39; 95% CI 0.22 to 0.69; p=0.001) were causally associated with a reduced risk of PD. In addition, our multivariable MR results provide additional assurance that the causal association between drinks per week and PD is unlikely due to confounding by smoking behavior.ConclusionOur findings support the role of smoking as a protective factor against PD, but only when comparing current vs. former smokers. Increased alcohol intake also had a protective effect over PD risk, and the alcohol dehydrogenase 1B (ADH1B) locus is a candidate for further investigating the mechanisms underlying this association.
- Published
- 2020
- Full Text
- View/download PDF
38. Impact of GBA1 variants on long-term clinical progression and mortality in incident Parkinson’s disease
- Author
-
Jonathan Evans, Marta Camacho, David P. Breen, Ganqiang Liu, Roger A. Barker, Clemens R. Scherzer, Thomas B Stoker, Caroline H. Williams-Gray, Thomas Foltynie, Sophie Winder-Rhodes, Stoker, Thomas B [0000-0001-5186-7630], Foltynie, Thomas [0000-0003-0752-1813], and Apollo - University of Cambridge Repository
- Subjects
Oncology ,Male ,medicine.medical_specialty ,Parkinson's disease ,Disease ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Medicine ,Dementia ,Humans ,Risk factor ,Movement disorders ,Survival rate ,030304 developmental biology ,Aged ,0303 health sciences ,business.industry ,Proportional hazards model ,Parkinsonism ,Parkinson Disease ,Middle Aged ,medicine.disease ,3. Good health ,Survival Rate ,Psychiatry and Mental health ,Mutation ,Disease Progression ,Population study ,Glucosylceramidase ,Surgery ,Female ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Abstract
IntroductionVariants in the GBA1 gene have been identified as a common risk factor for Parkinson’s disease (PD). In addition to pathogenic mutations (those associated with Gaucher disease), a number of ‘non-pathogenic’ variants also occur at increased frequency in PD. Previous studies have reported that pathogenic variants adversely affect the clinical course of PD. The role of ‘non-pathogenic’ GBA1 variants on PD course is less clear. In this study, we report the effect of GBA1 variants in incident PD patients with long-term follow-up.MethodsThe study population consisted of patients in the Cambridgeshire Incidence of Parkinson’s disease from General Practice to Neurologist and Parkinsonism: Incidence, Cognition and Non-motor heterogeneity in Cambridgeshire cohorts. Patients were grouped into non-carriers, carriers of ‘non-pathogenic’ GBA1 variants and carriers of pathogenic GBA1 mutations. Survival analyses for time to development of dementia, postural instability and death were carried out. Cox regression analysis controlling for potential confounders were used to determine the impact of GBA1 variants on these outcome measures.ResultsGBA1 variants were identified in 14.4% of patients. Pathogenic and ‘non-pathogenic’ GBA1 variants were associated with the accelerated development of dementia and a more aggressive motor course. Pathogenic GBA1 variants were associated with earlier mortality in comparison with non-carriers, independent of the development of dementia.DiscussionGBA1 variants, including those not associated with Gaucher disease, are common in PD and result in a more aggressive disease course.
- Published
- 2020
- Full Text
- View/download PDF
39. Oxidative Modifications of Parkin Underlie its Selective Neuroprotection in Adult Human Brain
- Author
-
Jacqueline M. Tokarew, Daniel N. El-Kodsi, Jennifer A. Chan, Clemens R. Scherzer, Gary S. Shaw, Ming Jin, Liqun Wang, An Tran, Andy C. H. Ng, Mei Zhang, Eve C. Tsai, Qiubo Jiang, Xiajun Dong, John Woulfe, Masashi Takanashi, Brian O'Nuallain, Juan Li, Travis K. Fehr, Peggy Taylor, Nathalie A. Lengacher, Jasmine M. Khan, Andrew B. West, Angela P. Nguyen, Nobutaka Hattori, Alexandre Prat, Julianna J. Tomlinson, Gergely Tóth, Kathryn R. Barber, Michael G. Schlossmacher, Arne Holmgren, Stephanie Zandee, Rajib Sengupta, and Lawrence G. Puente
- Subjects
0303 health sciences ,Chemistry ,Substantia nigra ,Human brain ,Oxidative phosphorylation ,Neuroprotection ,Parkin ,Cell biology ,nervous system diseases ,Midbrain ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Neuromelanin ,Dopamine ,medicine ,030217 neurology & neurosurgery ,030304 developmental biology ,medicine.drug - Abstract
SUMMARYThe mechanisms by which Parkinson disease-linked parkin confers neuroprotection of human dopamine cells remain elusive. We hypothesized that its cysteines mediate multiple anti-oxidant effects in the midbrain. By studying >60 control specimens, we found that in adult human brain - but not in skeletal muscle- parkin is mostly aggregated and insoluble due to oxidative modifications, such as at C253. In vitro, parkin’s oxidation directly reduces hydrogen peroxide (H2O2) to water. In parkin-deficient human brain, H2O2 concentrations are elevated. In dopamine toxicity studies, wild-type parkin -but not disease-associated mutants-prevents neural death by lowering H2O2 and sequestering radicals within insoluble aggregates. Parkin conjugates dopamine metabolites at the human-specific residue C95 and augments melanin formation in vitro. Using epitope-mapped antibodies, we found that in adult Substantia nigra neurons parkin localizes to neuromelanin within LAMP-3/CD63-positive lysosomes. We conclude that parkin’s own oxidation, previously considered a loss-of-function event, underlies three neuroprotective effects in adult midbrain: its cysteines participate in H2O2 reduction, dopamine radical conjugation and the formation of neuromelanin.
- Published
- 2020
- Full Text
- View/download PDF
40. Parkinson's disease polygenic risk score is not associated with impulse control disorders: A longitudinal study
- Author
-
Vanessa Brochard, Fanny Artaud, Frédéric Bourdain, S. Sambin, Valérie Mesnage, O. Rascol, Clemens R. Scherzer, Fabienne Ory-Magne, Isabelle Rieu, Jean-Christophe Corvol, Fanny Pineau, Merry Mazmanian, Louise-Laure Mariani, Monica Roy, Hélène Bertrand, Marie Vidailhet, Graziella Mangone, Tiphaine Vidal, Benjamin Le Toullec, Bérangère Debilly, Sara Leder, Emmanuel Roze, Bertrand Degos, Samir Bekadar, Cecilia Bonnet, Genetic core: Alexis Brice, Jean-Philippe Brandel, Antoine Faurie-Grepon, Hana You, Monique Galitsky, Stephan Klebe, Julia Kraemmer, J. Ihle, Mostafa Hajji, Virginie Bayet, David Grabli, Richard Levy, P. Derkinderen, Hakima Manseur, Ll Mariani, Julie Socha, Suzanne Lesage, Florence Cormier-Dequaire, Statistical analyses, A. Marques, Fernando Pico, Khadija Tahiri, Andreas Hartmann, Stéphane Bernard, Olivier Rascol, Eve Benchetrit, Alexis Brice, Julia Muellner, F. Durif, Violaine Plante-Bordeneuve, Sponsor activities, J-C Corvol, Alain Mallet, Co-investigators, Coralie Villeret, Pascal Derkinderen, Franck Durif, Anne-Marie Bonnet, Neuropsychologists, Christine Brefel-Courbon, Mélissa Tir, A. Elbaz, Lucette Lacomblez, Alexis Elbaz, Elsa Pomies, F. Artaud, Principal investigators for sites, Service de neurologie 1 [CHU Pitié-Salpétrière], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Sorbonne Université (SU), NS-Park/FCRIN Network, UMS 015, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), CHU Toulouse [Toulouse], Centre d'investigation clinique de Toulouse (CIC 1436), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-CHU Toulouse [Toulouse]-Institut National de la Santé et de la Recherche Médicale (INSERM), Brigham & Women’s Hospital [Boston] (BWH), Harvard Medical School [Boston] (HMS), Centre de recherche en épidémiologie et santé des populations (CESP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université Paris-Sud - Paris 11 (UP11)-Hôpital Paul Brousse-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Université Paris-Sud - Paris 11 - Faculté de médecine (UP11 UFR Médecine), Université Paris-Sud - Paris 11 (UP11), CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Hôpital Foch [Suresnes], Fondation A. Rothschild, Centre hospitalier universitaire de Nantes (CHU Nantes), CHU Clermont-Ferrand, CHU Saint-Antoine [AP-HP], Centre Hospitalier de Versailles André Mignot (CHV), Hôpital Henri Mondor, Hopital Saint-Louis [AP-HP] (AP-HP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), ANR-10-IAIHU-06 Ministère des Affaires Sociales et de la Santé: AOR0810 Agence Nationale de Sécurité du Médicament et des Produits de Santé, ANSM: ANSM-2013, This project was funded by grants from the French Ministry of Health (Programme Hospitalier de Recherche Clinique, AOR0810 ) and from the French Drug Agency (Agence Nationale de Sécurité et des Médicaments, ANSM-2013 ). The research leading to these results has received funding from the program ' Investissements d’Avenir ' ANR-10-IAIHU-06 . We want to express our gratitude to all the participants who made this study possible., ANR-10-IAHU-0006,IHU-A-ICM,Institut de Neurosciences Translationnelles de Paris(2010), Service de Neurologie [CHU Pitié-Salpêtrière], IFR70-CHU Pitié-Salpêtrière [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), and Université de Versailles Saint-Quentin-en-Yvelines (UVSQ)-Université Paris-Sud - Paris 11 (UP11)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Paul Brousse-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Longitudinal study ,Multifactorial Inheritance ,Parkinson's disease ,Movement disorders ,Genome-wide association study ,Disease ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Genetic predisposition ,Humans ,Longitudinal Studies ,Age of Onset ,Adverse effect ,Generalized estimating equation ,Aged ,business.industry ,Parkinson Disease ,Middle Aged ,medicine.disease ,3. Good health ,Disruptive, Impulse Control, and Conduct Disorders ,030104 developmental biology ,Neurology ,Female ,[SDV.NEU]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,Neurology (clinical) ,France ,Geriatrics and Gerontology ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
International audience; Objective: To examine the relationship between a Parkinson's disease (PD) polygenic risk score (PRS) and impulse control disorders (ICDs) in PD. Background: Genome wide association studies (GWAS) have brought forth a PRS associated with increased risk of PD and younger disease onset. ICDs are frequent adverse effects of dopaminergic drugs and are also more frequent in patients with younger disease onset. It is unknown whether ICDs and PD share genetic susceptibility. Methods: We used data from a multicenter longitudinal cohort of PD patients with annual visits up to 6 years (DIG-PD). At each visit ICDs, defined as compulsive gambling, buying, eating, or sexual behavior were evaluated by movement disorders specialists. We genotyped DNAs using the Megachip assay (Illumina) and calculated a weighted PRS based on 90 SNPs associated with PD. We estimated the association between PRS and prevalence of ICDs at each visit using Poisson generalized estimating equations, adjusted for dopaminergic treatment and other known risk factors for ICDs. Results: Of 403 patients, 185 developed ICDs. Patients with younger age at onset had a higher prevalence of ICDs (p < 0.001) as well as higher PRS values (p = 0.06). At baseline, there was no association between the PRS and ICDs (overall, p = 0.84). The prevalence of ICDs increased over time similarly across the quartiles of the PRS (overall, p = 0.88; DA users, p = 0.99). Conclusion: Despite younger disease onset being associated with both higher PRS and ICD prevalence, our findings are not in favor of common susceptibility genes for PD and ICDs.
- Published
- 2020
- Full Text
- View/download PDF
41. Enhancers active in dopamine neurons are a primary link between genetic variation and neuropsychiatric disease
- Author
-
Clemens R. Scherzer, Ganqiang Liu, Ferenc Müller, David Gritsch, Patrizia Rizzu, Peter T. Nelson, Xianjun Dong, Cornelis Blauwendraat, Boris Guennewig, John S. Mattick, Joseph J. Locascio, Yunfei Bai, Zhixiang Liao, Peter Heutink, Tao Wang, Matthew P. Frosch, Charles H. Adler, Yavor Hadzhiev, Thomas G. Beach, Richard L.M. Faull, John C. Hedreen, and Antony A. Cooper
- Subjects
Male ,0301 basic medicine ,Quantitative Trait Loci ,Enhancer RNAs ,Biology ,genetics [Mental Disorders] ,Genome ,Article ,03 medical and health sciences ,0302 clinical medicine ,pathology [Brain] ,Dopamine ,ddc:570 ,medicine ,Animals ,Humans ,physiology [Dopaminergic Neurons] ,Enhancer ,Zebrafish ,Regulation of gene expression ,genetics [Quantitative Trait Loci] ,Dopaminergic Neurons ,Mental Disorders ,General Neuroscience ,Brain ,Genetic Variation ,pathology [Mental Disorders] ,genetics [Enhancer Elements, Genetic] ,Human brain ,biology.organism_classification ,genetics [Genetic Variation] ,3. Good health ,Enhancer Elements, Genetic ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Expression quantitative trait loci ,Female ,Neuroscience ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Enhancers function as DNA logic gates and may control specialized functions of billions of neurons. Here we show a tailored program of noncoding genome elements active in situ in physiologically distinct dopamine neurons of the human brain. We found 71,022 transcribed noncoding elements, many of which were consistent with active enhancers and with regulatory mechanisms in zebrafish and mouse brains. Genetic variants associated with schizophrenia, addiction, and Parkinson’s dis- ease were enriched in these elements. Expression quantitative trait locus analysis revealed that Parkinson’s disease-associated variants on chromosome 17q21 cis-regulate the expression of an enhancer RNA in dopamine neurons. This study shows that enhancers in dopamine neurons link genetic variation to neuropsychiatric traits.
- Published
- 2018
- Full Text
- View/download PDF
42. Genomewide association study of Parkinson's disease clinical biomarkers in 12 longitudinal patients' cohorts
- Author
-
Ole A. Andreassen, Vivianna M. Van Deerlin, David Simon, Claire E. Wegel, Guido Alves, Ruwani Wijeyekoon, Jodi Maple-Grødem, Alastair J. Noyce, Roger A. Barker, Bart P.C. van de Warrenburg, Peter Heutink, Shirley Eberly, J. Raphael Gibbs, Alexis Brice, Kumaraswamy Naidu Chitrala, Lasse Pihlstrøm, Marlies van Nimwegen, Khanh-Dung H. Nguyen, Ganqiang Liu, Bernard Ravina, Clemens R. Scherzer, Jean-Christophe Corvol, Bastiaan R. Bloem, Jacobus J. van Hilten, Karol Estrada, Faraz Faghri, Jonathan R. Evans, Daniel Weintraub, Ole-Bjørn Tysnes, Aaron G. Day-Williams, Hampton L. Leonard, Jonggeol J. Kim, Fabrice Danjou, David P. Breen, Samantha J. Hutten, Andrew B. Singleton, Hirotaka Iwaki, Peggy Auinger, Mike A. Nalls, Kirsten M. Scott, Dena G. Hernandez, Mathias Toft, Jacqueline Rick, Caroline H. Williams-Gray, and Cornelis Blauwendraat
- Subjects
0301 basic medicine ,Oncology ,Apolipoprotein E ,Male ,Parkinson's disease ,Disease ,genetics [Glucosylceramidase] ,etiology [Cognitive Dysfunction] ,Cohort Studies ,0302 clinical medicine ,genetics [Parkinson Disease] ,Medicine ,Longitudinal Studies ,Cognitive decline ,Aged, 80 and over ,Hazard ratio ,Parkinson Disease ,Middle Aged ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,LRRK2 ,Phenotype ,Neurology ,genetics [Organic Cation Transport Proteins] ,Disease Progression ,Glucosylceramidase ,GBA ,Female ,psychology [Cognitive Dysfunction] ,psychology [Parkinson Disease] ,Adult ,medicine.medical_specialty ,Organic Cation Transport Proteins ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Risk Assessment ,Article ,03 medical and health sciences ,Antigens, CD ,Internal medicine ,Humans ,Cognitive Dysfunction ,ddc:610 ,genomewide association study ,Aged ,business.industry ,genetics [Cognitive Dysfunction] ,Odds ratio ,medicine.disease ,genetics [Antigens, CD] ,030104 developmental biology ,Cross-Sectional Studies ,Expression quantitative trait loci ,genetics [Leucine-Rich Repeat Serine-Threonine Protein Kinase-2] ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Biomarkers ,Genome-Wide Association Study - Abstract
Item does not contain fulltext 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 alpha-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. (c) 2019 International Parkinson and Movement Disorder Society.
- Published
- 2019
- Full Text
- View/download PDF
43. Reader response: Use of β2-adrenoreceptor agonist and antagonist drugs and risk of Parkinson disease
- Author
-
Joseph J. Locascio, Clemens R. Scherzer, and Trond Riise
- Subjects
Agonist ,medicine.medical_specialty ,COPD ,Essential tremor ,medicine.drug_class ,business.industry ,Inhaled corticosteroids ,Disease ,Propranolol ,medicine.disease ,Neuroprotection ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,030212 general & internal medicine ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
This Danish study1 “confirmed (β2-adrenoreceptor [β2AR]) agonist use to be associated with reduced Parkinson disease (PD) risk…,” replicating larger studies in Israel2 and Norway.3 However, the authors' speculations regarding causality, and that smoking may confound the association, conflict with their own study. First, they did not collect smoking data. In Israel,2 the association remained significant after directly adjusting for smoking (which was collected) and smoking-related covariates. Second, even after adjusting for COPD diagnosis, inhaled corticosteroids, and inhaled anticholinergics (their “markers of smoking”), β2AR agonists were significantly associated with reduced PD risk with an OR of 0.64 (0.42–0.98) compared with 0.66 (0.52–0.85) without adjusting (table 4).1 Adjusting for the smoking markers did not attenuate the OR. Once these covariates are statistically controlled, any remaining significant relation between β2AR agonists and reduced PD risk cannot be due to their mediating effects. Whether covariates themselves are related to PD risk is not directly relevant. Third, the beta-blocker propranolol, which increased neuronal SNCA,3 was associated with increased PD risk in Denmark (excluding essential tremor),1 Norway (restricted to cardiovascular indications),3 and Israel (5–8 years lag).2 Finally, β2AR agonists, associated with reduced PD risk after adjusting for smoking-related covariates in Norway,3 Israel,2 and Denmark,1 offered neuroprotection in 3 rodent models.3–5 Thankfully, rodents don't smoke.
- Published
- 2020
- Full Text
- View/download PDF
44. A common polymorphism in SNCA is associated with accelerated motor decline in GBA-Parkinson’s disease
- Author
-
Caroline H. Williams-Gray, Clemens R. Scherzer, Sophie Winder-Rhodes, Ganqiang Liu, Thomas Foltynie, Thomas B Stoker, Marta Camacho, Roger A. Barker, Stoker, Thomas B [0000-0001-5186-7630], Foltynie, Thomas [0000-0003-0752-1813], and Apollo - University of Cambridge Repository
- Subjects
Oncology ,Male ,medicine.medical_specialty ,Parkinson's disease ,Genotype ,Population ,Neurogenetics ,Disease ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Genetic predisposition ,medicine ,Humans ,education ,neurogenetics ,health care economics and organizations ,education.field_of_study ,Lewy body ,business.industry ,Parkinson Disease ,PostScript ,medicine.disease ,3. Good health ,Psychiatry and Mental health ,Mutation ,Disease Progression ,alpha-Synuclein ,movement disorders ,Glucosylceramidase ,Surgery ,Neurology (clinical) ,business ,Glucocerebrosidase ,030217 neurology & neurosurgery - Abstract
A growing number of genetic susceptibility factors have been identified for Parkinson’s disease (PD). The combination of inherited risk variants is likely to affect not only risk of developing PD but also its clinical course. Variants in the GBA gene are particularly common, being found in approximately 5% to 10% of patients, and they lead to more rapid disease progression.1 However, the effect of concomitant genetic risk factors on disease course in GBA -PD is not known. The aggregation of α-synuclein, encoded by the SNCA gene, is central to the pathogenesis of PD. The SNCA rs356219 A/G polymorphism alters the risk of developing PD, with homozygotes for guanine (G/G) having an increased risk compared with carriers of an adenine (G/A or A/A) at this locus.2 The relationship between glucocerebrosidase (the enzyme encoded by the GBA gene) and α-synuclein is complex. These proteins have been shown to interact directly in vitro, as well as to influence the intracellular levels and processing of each other, potentially in a bidirectional feedback loop.3 4 Interestingly, a recent genome-wide association study found that the presence of this SNCA polymorphism was associated with an increased likelihood of developing PD in GBA mutation carriers.5 We therefore hypothesised that the presence of the SNCA rs356219 polymorphism would accelerate the clinical course of GBA variant-associated PD. Here, we report on the effect of this SNCA polymorphism on clinical outcomes within the GBA -PD population. Longitudinal data from GBA -variant carriers were analysed from the community-based ‘Cambridgeshire Incidence of Parkinson’s disease from General Practice to Neurologist’ (CamPaIGN) cohort (n=142).6 This study was approved by the local ethics committee and written informed consent was obtained from all subjects. Newly diagnosed patients were followed up with assessments every 2 years for up to 18 years. Time to …
- Published
- 2020
45. β-Glucocerebrosidase activity in
- Author
-
Young Eun, Huh, Ming Sum Ruby, Chiang, Joseph J, Locascio, Zhixiang, Liao, Ganqiang, Liu, Karbi, Choudhury, Yuliya I, Kuras, Idil, Tuncali, Aleksandar, Videnovic, Ann L, Hunt, Michael A, Schwarzschild, Albert Y, Hung, Todd M, Herrington, Michael T, Hayes, Bradley T, Hyman, Anne-Marie, Wills, Stephen N, Gomperts, John H, Growdon, Sergio Pablo, Sardi, and Clemens R, Scherzer
- Subjects
Aged, 80 and over ,Male ,Neurologic Examination ,Quantitative Trait Loci ,Parkinson Disease ,Middle Aged ,Severity of Illness Index ,Article ,Cross-Sectional Studies ,Mutation ,Glucosylceramidase ,Humans ,Female ,Cognition Disorders ,Genetic Association Studies ,Aged ,Follow-Up Studies - Abstract
OBJECTIVE: To test the relationship between clinically relevant types of GBA mutations (none, risk variants, mild mutations, severe mutations) and β-glucocerebrosidase activity in patients with Parkinson disease (PD) in cross-sectional and longitudinal case-control studies. METHODS: A total of 481 participants from the Harvard Biomarkers Study (HBS) and the NIH Parkinson's Disease Biomarkers Program (PDBP) were analyzed, including 47 patients with PD carrying GBA variants (GBA-PD), 247 without a GBA variant (idiopathic PD), and 187 healthy controls. Longitudinal analysis comprised 195 participants with 548 longitudinal measurements over a median follow-up period of 2.0 years (interquartile range, 1–2 years). RESULTS: β-Glucocerebrosidase activity was low in blood of patients with GBA-PD compared to healthy controls and patients with idiopathic PD, respectively, in HBS (p < 0.001) and PDBP (p < 0.05) in multivariate analyses adjusting for age, sex, blood storage time, and batch. Enzyme activity in patients with idiopathic PD was unchanged. Innovative enzymatic quantitative trait locus (xQTL) analysis revealed a negative linear association between residual β-glucocerebrosidase activity and mutation type with p < 0.0001. For each increment in the severity of mutation type, a reduction of mean β-glucocerebrosidase activity by 0.85 μmol/L/h (95% confidence interval, −1.17, −0.54) was predicted. In a first longitudinal analysis, increasing mutation severity types were prospectively associated with steeper declines in β-glucocerebrosidase activity during a median 2 years of follow-up (p = 0.02). CONCLUSIONS: Residual activity of the β-glucocerebrosidase enzyme measured in blood inversely correlates with clinical severity types of GBA mutations in PD. β-Glucocerebrosidase activity is a quantitative endophenotype that can be monitored noninvasively and targeted therapeutically.
- Published
- 2019
46. Potential two-step proteomic signature for Parkinson's disease: Pilot analysis in the Harvard Biomarkers Study
- Author
-
Clemens R. Scherzer, Leigh A. Johnson, Fan Zhang, James Hall, Sid E. O'Bryant, Melissa Edwards, and Yuliya I. Kuras
- Subjects
Proteomics ,Parkinson's disease ,Two step ,Diagnostic accuracy ,Disease ,Computational biology ,lcsh:Geriatrics ,lcsh:RC346-429 ,Blood biomarkers ,03 medical and health sciences ,0302 clinical medicine ,medicine ,lcsh:Neurology. Diseases of the nervous system ,030304 developmental biology ,0303 health sciences ,Proteomic Profile ,Proteomic screening ,business.industry ,Precision medicine ,Special Section: Blood-Based Biomarkers for Alzheimer's Disease & Related Dementias. (Editor: Henrik Zetterberg) ,medicine.disease ,3. Good health ,lcsh:RC952-954.6 ,Psychiatry and Mental health ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Abstract
Introduction We sought to determine if our previously validated proteomic profile for detecting Alzheimer's disease would detect Parkinson's disease (PD) and distinguish PD from other neurodegenerative diseases. Methods Plasma samples were assayed from 150 patients of the Harvard Biomarkers Study (PD, n = 50; other neurodegenerative diseases, n = 50; healthy controls, n = 50) using electrochemiluminescence and Simoa platforms. Results The first step proteomic profile distinguished neurodegenerative diseases from controls with a diagnostic accuracy of 0.94. The second step profile distinguished PD cases from other neurodegenerative diseases with a diagnostic accuracy of 0.98. The proteomic profile differed in step 1 versus step 2, suggesting that a multistep proteomic profile algorithm to detecting and distinguishing between neurodegenerative diseases may be optimal. Discussion These data provide evidence of the potential use of a multitiered blood-based proteomic screening method for detecting individuals with neurodegenerative disease and then distinguishing PD from other neurodegenerative diseases.
- Published
- 2019
47. Genome-wide association study of Parkinson’s disease progression biomarkers in 12 longitudinal patients’ cohorts
- Author
-
David P. Breen, Samantha J. Hutten, Alastair J. Noyce, Dena G. Hernandez, Kumaraswamy Naidu Chitrala, Lasse Pihlstrøm, Jacqueline Rick, Mathias Toft, Mike A. Nalls, Ganqiang Liu, Karol Estrada, Jean-Christophe Corvol, Marlies van Nimwegen, Bernard Ravina, Shirley Eberly, Aaron G. Day-Williams, Jonathan R. Evans, Peter Heutink, Claire E. Wegel, Ole-Bjørn Tysnes, J. Raphael Gibbs, Bart P.C. van de Warrenburg, Peggy Auinger, Alexis Brice, Hirotaka Iwaki, Clemens R. Scherzer, Jonggeol J. Kim, Jodi Maple-Grødem, Kirsten M. Scott, Fabrice Danjou, Daniel Weintraub, H. Nguyen Khanh-Dung, Jacobus J. van Hilten, Roger A. Barker, Caroline H. Williams-Gray, Hampton L. Leonard, Cornelis Blauwendraat, David Simon, Andrew B. Singleton, Faraz Faghri, Guido Alves, Ruwani Wijeyekoon, Ole A. Andreassen, Vivianna M. Van Deerlin, and Bastiaan R. Bloem
- Subjects
Oncology ,medicine.medical_specialty ,Parkinson's disease ,business.industry ,Genome-wide association study ,Disease ,medicine.disease ,LRRK2 ,Clinical trial ,Internal medicine ,Expression quantitative trait loci ,medicine ,Missense mutation ,Cognitive decline ,business - Abstract
BackgroundSeveral reports have identified different patterns of Parkinson’s disease progression in individuals carrying missense variants in theGBAorLRRK2genes. The overall contribution of genetic factors to the severity and progression of Parkinson’s disease, however, has not been well studied.ObjectivesTo test the association between genetic variants and the clinical features and progression of Parkinson’s disease on a genome-wide scale.MethodsWe accumulated individual data from 12 longitudinal cohorts in a total of 4,093 patients with 25,254 observations over a median of 3.81 years. Genome-wide associations were evaluated for 25 cross-sectional and longitudinal phenotypes. Specific variants of interest, including 90 recently-identified disease risk variants, were also investigated for the associations with these phenotypes.ResultsTwo variants were genome-wide significant. Rs382940(T>A), within the intron ofSLC44A1, was associated with reaching Hoehn and Yahr stage 3 or higher faster (HR 2.04 [1.58, 2.62], P-value = 3.46E-8). Rs61863020(G>A), an intergenic variant and eQTL forADRA2A, was associated with a lower prevalence of insomnia at baseline (OR 0.63 [0,52, 0.75], P-value = 4.74E-8). In the targeted analysis, we found nine associations between known Parkinson’s risk variants and more severe motor/cognitive symptoms. Also, we replicated previous reports ofGBAcoding variants (rs2230288: p.E365K, rs75548401: p.T408M) being associated with greater motor and cognitive decline over time, andAPOEE4 tagging variant (rs429358) being associated with greater cognitive deficits in patients.ConclusionsWe identified novel genetic factors associated with heterogeneity of progression in Parkinson’s disease. The results provide new insights into the pathogenesis of Parkinson’s disease as well as patient stratification for clinical trials.
- Published
- 2019
- Full Text
- View/download PDF
48. Publisher Correction: Enhancers active in dopamine neurons are a primary link between genetic variation and neuropsychiatric disease
- Author
-
Charles H. Adler, Xianjun Dong, Cornelis Blauwendraat, Clemens R. Scherzer, David Gritsch, Richard L.M. Faull, Zhixiang Liao, John C. Hedreen, Patrizia Rizzu, Ganqiang Liu, Thomas G. Beach, John S. Mattick, Ferenc Müller, Matthew P. Frosch, Tao Wang, Yavor Hadzhiev, Joseph J. Locascio, Peter Heutink, Boris Guennewig, Yunfei Bai, Antony A. Cooper, and Peter T. Nelson
- Subjects
0301 basic medicine ,General Neuroscience ,Biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Dopamine ,ddc:570 ,Genetic variation ,medicine ,Enhancer ,Neuroscience ,030217 neurology & neurosurgery ,Neuropsychiatric disease ,medicine.drug - Abstract
In the version of this article initially published, the legends for Supplementary Figs. 4–8 and 10–14 contained errors. The Supplementary Figure legends have been corrected in the HTML and PDF versions of the article.
- Published
- 2019
- Full Text
- View/download PDF
49. Identification of genetic modifiers of age-at-onset for familial Parkinson’s disease
- Author
-
Joanna Siuda, Sepideh Zareparsi, Owen A. Ross, Clemens R. Scherzer, Alexandra I. Soto-Ortolaza, Timothy Lynch, Patrick Breheny, Haydeh Payami, Erin M. Hill-Burns, William T. Wissemann, Zbigniew K. Wszolek, Peter A. Silburn, Stewart A. Factor, Cyrus P. Zabetian, Beate Ritz, and George D. Mellick
- Subjects
0301 basic medicine ,Male ,Parkinson's disease ,Neurogenetics ,Genome-wide association study ,TPM1 ,Disease ,Tropomyosin ,Biology ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,Polymorphism (computer science) ,Genetics ,medicine ,Dementia ,Humans ,Genetic Predisposition to Disease ,Age of Onset ,Molecular Biology ,Genetics (clinical) ,Aged ,Aged, 80 and over ,Association Studies Articles ,Membrane Proteins ,Proteins ,Parkinson Disease ,General Medicine ,Middle Aged ,medicine.disease ,030104 developmental biology ,Female ,Age of onset ,Genome-Wide Association Study - Abstract
Parkinson’s disease (PD) is the most common cause of neurodegenerative movement disorder and the second most common cause of dementia. Genes are thought to have a stronger effect on age-at-onset of PD than on risk, yet there has been a phenomenal success in identifying risk loci but not age-at-onset modifiers. We conducted a genome-wide study for age-at-onset. We analysed familial and non-familial PD separately, per prior evidence for strong genetic effect on age-at-onset in familial PD. GWAS was conducted in 431 unrelated PD individuals with at least one affected relative (familial PD) and 1544 non-familial PD from the NeuroGenetics Research Consortium (NGRC); an additional 737 familial PD and 2363 non-familial PD were used for replication. In familial PD, two signals were detected and replicated robustly: one mapped to LHFPL2 on 5q14.1 (PNGRC = 3E-8, PReplication = 2E-5, PNGRC + Replication = 1E-11), the second mapped to TPM1 on 15q22.2 (PNGRC = 8E-9, PReplication = 2E-4, PNGRC + Replication = 9E-11). The variants that were associated with accelerated onset had low frequencies (
- Published
- 2016
50. A Feed-Forward Circuit of EndogenousPGC-1αandEstrogen Related Receptor αRegulates the Neuronal Electron Transport Chain
- Author
-
Zhixiang Liao, Clemens R. Scherzer, Rachit Bakshi, and Shuchi Mittal
- Subjects
0301 basic medicine ,Article Subject ,Neuroscience (miscellaneous) ,Regulator ,Endogeny ,Peroxisome ,Biology ,lcsh:RC346-429 ,Cell biology ,03 medical and health sciences ,Psychiatry and Mental health ,Estrogen-related receptor ,030104 developmental biology ,Biochemistry ,Transcription (biology) ,Coactivator ,Gene expression ,Neurology (clinical) ,Receptor ,lcsh:Neurology. Diseases of the nervous system ,Research Article - Abstract
Peroxisome proliferator-activated receptor γcoactivator 1α(PGC-1α) is a central regulator of cellular and mitochondrial metabolism. Cellular bioenergetics are critically important in “energy-guzzling” neurons, but the components and wiring of the transcriptional circuit through whichPGC-1αregulates the neuronal electron transport chain have not been established. This information may be vital for restoring neuronal bioenergetics gene expression that is compromised during incipient Parkinson’s neuropathology and in aging-dependent brain diseases. Here we delineate a neuronal transcriptional circuit controlled by endogenousPGC-1α. We show that a feed-forward circuit of endogenous neuronalPGC-1αand the orphan nuclear estrogen-related receptorα(ERRα) activates the nuclear-encoded mitochondrial electron transport chain.PGC-1αnot onlytrans-activated expression ofERRα, but also coactivatedERRαtarget genes in complexes I, II, IV, and V of the neuronal electron transport chain via association with evolutionary conservedERRαpromoter binding motifs. Chemical activation of this transcriptional program induced transcription of the neuronal electron transport chain. These data highlight a neuronal transcriptional circuit regulated byPGC-1αthat can be therapeutically targeted for Parkinson’s and other neurodegenerative diseases.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.