51
|
Cukier HN, Kim H, Griswold AJ, Codreanu SG, Prince LM, Sherrod SD, McLean JA, Dykxhoorn DM, Ess KC, Hedera P, Bowman AB, Neely MD. Genomic, transcriptomic, and metabolomic profiles of hiPSC-derived dopamine neurons from clinically discordant brothers with identical PRKN deletions. NPJ Parkinsons Dis 2022; 8:84. [PMID: 35768426 PMCID: PMC9243035 DOI: 10.1038/s41531-022-00346-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 05/27/2022] [Indexed: 11/25/2022] Open
Abstract
We previously reported on two brothers who carry identical compound heterozygous PRKN mutations yet present with significantly different Parkinson's Disease (PD) clinical phenotypes. Juvenile cases demonstrate that PD is not necessarily an aging-associated disease. Indeed, evidence for a developmental component to PD pathogenesis is accumulating. Thus, we hypothesized that the presence of additional genetic modifiers, including genetic loci relevant to mesencephalic dopamine neuron development, could potentially contribute to the different clinical manifestations of the two brothers. We differentiated human-induced pluripotent stem cells (hiPSCs) derived from the two brothers into mesencephalic neural precursor cells and early postmitotic dopaminergic neurons and performed wholeexome sequencing and transcriptomic and metabolomic analyses. No significant differences in the expression of canonical dopamine neuron differentiation markers were observed. Yet our transcriptomic analysis revealed a significant downregulation of the expression of three neurodevelopmentally relevant cell adhesion molecules, CNTN6, CNTN4 and CHL1, in the cultures of the more severely affected brother. In addition, several HLA genes, known to play a role in neurodevelopment, were differentially regulated. The expression of EN2, a transcription factor crucial for mesencephalic dopamine neuron development, was also differentially regulated. We further identified differences in cellular processes relevant to dopamine metabolism. Lastly, wholeexome sequencing, transcriptomics and metabolomics data all revealed differences in glutathione (GSH) homeostasis, the dysregulation of which has been previously associated with PD. In summary, we identified genetic differences which could potentially, at least partially, contribute to the discordant clinical PD presentation of the two brothers.
Collapse
Affiliation(s)
- Holly N Cukier
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Hyunjin Kim
- School of Health Sciences, Purdue University, West Lafayette, Indiana, IN, USA
| | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Simona G Codreanu
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Lisa M Prince
- School of Health Sciences, Purdue University, West Lafayette, Indiana, IN, USA
| | - Stacy D Sherrod
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - John A McLean
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Derek M Dykxhoorn
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kevin C Ess
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Peter Hedera
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Neurology, University of Louisville, Louisville, KY, USA
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, Indiana, IN, USA.
| | - M Diana Neely
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
52
|
Sergi CM. Epigallocatechin Gallate (EGCG) for Parkinson's Disease. Clin Exp Pharmacol Physiol 2022; 49:1029-1041. [PMID: 35748799 DOI: 10.1111/1440-1681.13691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 01/03/2022] [Accepted: 06/19/2022] [Indexed: 11/28/2022]
Abstract
In the last couple of decades, we have experienced increased use of nutraceuticals worldwide with a demand for organic foods, which has been elevated to an extent probably unmatched with other periods of our civilization. One of the nutraceuticals that gained attention is epigallocatechin gallate (EGCG), a polyphenol in green tea. It has been suggested that diseases of the central nervous system (CNS) can benefit from consuming some antioxidants, despite current results showing little evidence for their use in preventing and treating these diseases. ECGC may be beneficial in delaying the neurodegeneration of the substantia nigra (SN) regardless of the origin of Parkinson's disease (PD). This review covers the effect of EGCG on vitro and animal models of PD, the potential mechanisms of neuroprotection involved and summaries recent clinical trials in human PD. This review also aims to provide an investigative analysis of the current knowledge in this field and identify putative crucial issues. Environmental factors such as dietary habits, drug use, and social interaction are all factors that influence the evolution of neurodegenerative diseases. Therefore, the use of nutraceuticals requires further investigation. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Consolato M Sergi
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, Hubei, China.,Anatomic Pathology, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON, Canada.,Department of Orthopedics, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Laboratory Medicine and Pathology, University of Alberta, AB, Canada
| |
Collapse
|
53
|
Angelopoulou E, Bougea A, Papageorgiou SG, Villa C. Psychosis in Parkinson's Disease: A Lesson from Genetics. Genes (Basel) 2022; 13:genes13061099. [PMID: 35741861 PMCID: PMC9222985 DOI: 10.3390/genes13061099] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/17/2022] [Accepted: 06/18/2022] [Indexed: 02/06/2023] Open
Abstract
Psychosis in Parkinson's disease (PDP) represents a common and debilitating condition that complicates Parkinson's disease (PD), mainly in the later stages. The spectrum of psychotic symptoms are heterogeneous, ranging from minor phenomena of mild illusions, passage hallucinations and sense of presence to severe psychosis consisting of visual hallucinations (and rarely, auditory and tactile or gustatory) and paranoid delusions. PDP is associated with increased caregiver stress, poorer quality of life for patients and carers, reduced survival and risk of institutionalization with a significant burden on the healthcare system. Although several risk factors for PDP development have been identified, such as aging, sleep disturbances, long history of PD, cognitive impairment, depression and visual disorders, the pathophysiology of psychosis in PD is complex and still insufficiently clarified. Additionally, several drugs used to treat PD can aggravate or even precipitate PDP. Herein, we reviewed and critically analyzed recent studies exploring the genetic architecture of psychosis in PD in order to further understand the pathophysiology of PDP, the risk factors as well as the most suitable therapeutic strategies.
Collapse
Affiliation(s)
- Efthalia Angelopoulou
- Department of Neurology, Eginition University Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (E.A.); (A.B.); (S.G.P.)
| | - Anastasia Bougea
- Department of Neurology, Eginition University Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (E.A.); (A.B.); (S.G.P.)
| | - Sokratis G. Papageorgiou
- Department of Neurology, Eginition University Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (E.A.); (A.B.); (S.G.P.)
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Correspondence: ; Tel.: +39-02-6448-8138
| |
Collapse
|
54
|
Gao XY, Yang T, Gu Y, Sun XH. Mitochondrial Dysfunction in Parkinson’s Disease: From Mechanistic Insights to Therapy. Front Aging Neurosci 2022; 14:885500. [PMID: 35795234 PMCID: PMC9250984 DOI: 10.3389/fnagi.2022.885500] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/30/2022] [Indexed: 12/02/2022] Open
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative movement disorders worldwide. There are currently no cures or preventative treatments for PD. Emerging evidence indicates that mitochondrial dysfunction is closely associated with pathogenesis of sporadic and familial PD. Because dopaminergic neurons have high energy demand, cells affected by PD exhibit mitochondrial dysfunction that promotes the disease-defining the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). The mitochondrion has a particularly important role as the cellular “powerhouse” of dopaminergic neurons. Therefore, mitochondria have become a promising therapeutic target for PD treatments. This review aims to describe mitochondrial dysfunction in the pathology of PD, outline the genes associated with familial PD and the factors related to sporadic PD, summarize current knowledge on mitochondrial quality control in PD, and give an overview of therapeutic strategies for targeting mitochondria in neuroprotective interventions in PD.
Collapse
Affiliation(s)
- Xiao-Yan Gao
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Science Experiment Center, China Medical University, Shenyang, China
| | - Tuo Yang
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Ying Gu
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiao-Hong Sun
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Science Experiment Center, China Medical University, Shenyang, China
- *Correspondence: Xiao-Hong Sun,
| |
Collapse
|
55
|
Moksnes MR, Graham SE, Wu KH, Hansen AF, Gagliano Taliun SA, Zhou W, Thorstensen K, Fritsche LG, Gill D, Mason A, Cucca F, Schlessinger D, Abecasis GR, Burgess S, Åsvold BO, Nielsen JB, Hveem K, Willer CJ, Brumpton BM. Genome-wide meta-analysis of iron status biomarkers and the effect of iron on all-cause mortality in HUNT. Commun Biol 2022; 5:591. [PMID: 35710628 PMCID: PMC9203493 DOI: 10.1038/s42003-022-03529-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 05/24/2022] [Indexed: 01/19/2023] Open
Abstract
Iron is essential for many biological processes, but iron levels must be tightly regulated to avoid harmful effects of both iron deficiency and overload. Here, we perform genome-wide association studies on four iron-related biomarkers (serum iron, serum ferritin, transferrin saturation, total iron-binding capacity) in the Trøndelag Health Study (HUNT), the Michigan Genomics Initiative (MGI), and the SardiNIA study, followed by their meta-analysis with publicly available summary statistics, analyzing up to 257,953 individuals. We identify 123 genetic loci associated with iron traits. Among 19 novel protein-altering variants, we observe a rare missense variant (rs367731784) in HUNT, which suggests a role for DNAJC13 in transferrin recycling. We further validate recently published results using genetic risk scores for each biomarker in HUNT (6% variance in serum iron explained) and present linear and non-linear Mendelian randomization analyses of the traits on all-cause mortality. We find evidence of a harmful effect of increased serum iron and transferrin saturation in linear analyses that estimate population-averaged effects. However, there was weak evidence of a protective effect of increasing serum iron at the very low end of its distribution. Our findings contribute to our understanding of the genes affecting iron status and its consequences on human health.
Collapse
Affiliation(s)
- Marta R Moksnes
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Trondheim, Norway.
| | - Sarah E Graham
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Kuan-Han Wu
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Ailin Falkmo Hansen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Sarah A Gagliano Taliun
- Department of Medicine and Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
- Montréal Heart Institute, Montréal, QC, Canada
| | - Wei Zhou
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ketil Thorstensen
- Department of Clinical Chemistry, St. Olavs hospital Trondheim University Hospital, Trondheim, Norway
| | - Lars G Fritsche
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Clinical Pharmacology and Therapeutics Section, Institute for Infection and Immunity, St George's, University of London, London, UK
- Clinical Pharmacology Group, Pharmacy and Medicines Directorate, St George's University Hospitals NHS Foundation Trust, London, UK
- Novo Nordisk Research Centre Oxford, Old Road Campus, Oxford, UK
| | - Amy Mason
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Cagliari, Italy
- Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy
| | - David Schlessinger
- Laboratory of Genetics, National Institute on Aging, US National Institutes of Health, Baltimore, MD, USA
| | - Gonçalo R Abecasis
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Stephen Burgess
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Bjørn Olav Åsvold
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, Clinic of Medicine, St. Olavs hospital Trondheim University Hospital, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Levanger, Norway
| | - Jonas B Nielsen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Levanger, Norway
| | - Cristen J Willer
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Montréal Heart Institute, Montréal, QC, Canada
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Ben M Brumpton
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Trondheim, Norway.
- HUNT Research Centre, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Levanger, Norway.
- Clinic of Medicine, St. Olavs hospital Trondheim University Hospital, Trondheim, Norway.
| |
Collapse
|
56
|
Amin R, Quispe C, Docea AO, Alibek Y, Kulbayeva M, Durna Daştan S, Calina D, Sharifi-Rad J. The role of Tumour Necrosis Factor in neuroinflammation associated with Parkinson's disease and targeted therapies. Neurochem Int 2022; 158:105376. [PMID: 35667491 DOI: 10.1016/j.neuint.2022.105376] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/29/2022] [Accepted: 05/30/2022] [Indexed: 12/21/2022]
Abstract
Neurodegenerative disorders Parkinson's disease is a progressive neurodegenerative disorder associated with neuroinflammatory responses that lead to the neurodegeneration of the dopaminergic neurons. These neuroinflammatory mechanisms involve various cytokines produced by the activated glial cells. Tumour Necrosis factor α (TNF α) is one of the major mediators of the neuroinflammation associated with neurodegeneration. TNF α has a dual role of neuroprotection and neurotoxicity in the brain. The effective pathways of TNF involve various signalling pathways transduced by the receptors TNFR1 and TNFR2. Effective therapeutic strategies have been produced targeting the neurotoxic behaviour of the Tumour Necrosis Factor and the associated neurodegeneration which includes the use of Dominant Negative Tumour Necrosis Factor (DN-TNF) inhibitors like XENP 345 and XPro®1595 and peroxisome proliferator receptor gamma (PPAR-γ) agonists.
Collapse
Affiliation(s)
- Ruhul Amin
- Faculty of Pharmaceutical Science, Assam Down Town University, Panikhaiti, Guwahati, Assam, India.
| | - Cristina Quispe
- Facultad de Ciencias de la Salud, Universidad Arturo Prat, Avda. Arturo Prat 2120, Iquique, 1110939, Chile.
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania
| | - Ydyrys Alibek
- Biomedical Research Centre, Al-Farabi Kazakh National University, Al-Farabi av. 71, 050040, Almaty, Kazakhstan.
| | - Marzhan Kulbayeva
- Department of Biophysics, Biomedicine and Neuroscience, Al-Farabi Kazakh National University, Al-Farabi av. 71, 050040, Almaty, Kazakhstan.
| | - Sevgi Durna Daştan
- Department of Biology, Faculty of Science, Sivas Cumhuriyet University, 58140, Sivas, Turkey; Beekeeping Development Application and Research Center, Sivas Cumhuriyet University, 58140, Sivas, Turkey.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | | |
Collapse
|
57
|
Hua P, Zhao Y, Zeng Q, Li L, Ren J, Guo J, Tang B, Liu W. Genetic Analysis of Patients With Early-Onset Parkinson’s Disease in Eastern China. Front Aging Neurosci 2022; 14:849462. [PMID: 35645773 PMCID: PMC9131032 DOI: 10.3389/fnagi.2022.849462] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/04/2022] [Indexed: 11/23/2022] Open
Abstract
Background Genetic factors play an important role in the pathogenesis of early-onset Parkinson’s disease (EOPD). To date, more than 20 pathogenic genes associated with Parkinson’s disease (PD) have been identified. This study aims to explore the mutation spectrum of EOPD and the clinical characteristics of mutation carriers in eastern China. Methods We recruited 155 unrelated EOPD patients, including 8 familial and 147 sporadic EOPD (age at onset ≤ 50 years). Overall, 24 known PD-associated genes were detected by whole exome sequencing and multiplex ligation-dependent probe amplification (MLPA) from patient samples. The genetic and clinical characteristics of pathogenic/likely pathogenic (P/LP) loci in this cohort were analyzed. Results Overall, 14 (9.03%) patients were detected with P/LP variants distributed in seven genes. The most frequent mutation occurred in PRKN (7/155, 4.52%), followed by LRRK2 (2/155, 1.29%), SNCA, CHCHD2, TMEM230, DNAJC13 and PLA2G6 (1/155, 0.64%, respectively). Exon rearrangement mutations accounted for 57.9% (11/19) of all mutations in PRKN. Four novel variants were detected: c.14T > C (p.M5T) in SNCA, c.297C > A (p.Y99X) in CHCHD2, c.2578C > T (p.R860C) in DNAJC13 and c.4C > T (p.Q2X) in TMEM230. We found the first case of LRRK2 c.6055G > A (p.G2019S) mutation in Chinese population. The median onset age of patients with P/LP mutations in autosomal recessive genes (PRKN and PLA2G6) was about 18.0 years earlier than patients without mutation. The proportion of patients with mutations were 63.64%, 27.03% and 9.68% when patients were stratified according to the age of onset at ≤ 30, ≤ 40 and ≤ 50 years, respectively. Conclusion Early-onset Parkinson’s disease patients from eastern China present a regional specific mutation spectrum. Analysis of larger patient cohorts is required to support these findings, and mechanistic studies of the four novel missense/non-sense mutations will clarify their role in the pathogenicity of EOPD.
Collapse
Affiliation(s)
- Ping Hua
- Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Yuwen Zhao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qian Zeng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lanting Li
- Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Jingru Ren
- Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Medical Genetics, Centre for Medical Genetics, Xiangya Hospital, School of Life Sciences, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Medical Genetics, Centre for Medical Genetics, Xiangya Hospital, School of Life Sciences, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Beisha Tang,
| | - Weiguo Liu
- Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Weiguo Liu,
| |
Collapse
|
58
|
Lange LM, Gonzalez-Latapi P, Rajalingam R, Tijssen MAJ, Ebrahimi-Fakhari D, Gabbert C, Ganos C, Ghosh R, Kumar KR, Lang AE, Rossi M, van der Veen S, van de Warrenburg B, Warner T, Lohmann K, Klein C, Marras C. Nomenclature of Genetic Movement Disorders: Recommendations of the International Parkinson and Movement Disorder Society Task Force - An Update. Mov Disord 2022; 37:905-935. [PMID: 35481685 DOI: 10.1002/mds.28982] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/28/2022] [Accepted: 02/14/2022] [Indexed: 12/13/2022] Open
Abstract
In 2016, the Movement Disorder Society Task Force for the Nomenclature of Genetic Movement Disorders presented a new system for naming genetically determined movement disorders and provided a criterion-based list of confirmed monogenic movement disorders. Since then, a substantial number of novel disease-causing genes have been described, which warrant classification using this system. In addition, with this update, we further refined the system and propose dissolving the imaging-based categories of Primary Familial Brain Calcification and Neurodegeneration with Brain Iron Accumulation and reclassifying these genetic conditions according to their predominant phenotype. We also introduce the novel category of Mixed Movement Disorders (MxMD), which includes conditions linked to multiple equally prominent movement disorder phenotypes. In this article, we present updated lists of newly confirmed monogenic causes of movement disorders. We found a total of 89 different newly identified genes that warrant a prefix based on our criteria; 6 genes for parkinsonism, 21 for dystonia, 38 for dominant and recessive ataxia, 5 for chorea, 7 for myoclonus, 13 for spastic paraplegia, 3 for paroxysmal movement disorders, and 6 for mixed movement disorder phenotypes; 10 genes were linked to combined phenotypes and have been assigned two new prefixes. The updated lists represent a resource for clinicians and researchers alike and they have also been published on the website of the Task Force for the Nomenclature of Genetic Movement Disorders on the homepage of the International Parkinson and Movement Disorder Society (https://www.movementdisorders.org/MDS/About/Committees--Other-Groups/MDS-Task-Forces/Task-Force-on-Nomenclature-in-Movement-Disorders.htm). © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson Movement Disorder Society.
Collapse
Affiliation(s)
- Lara M Lange
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Paulina Gonzalez-Latapi
- The Edmond J. Safra Program in Parkinson's Disease and The Morton and Gloria Shulman Movement Disorder Clinic, Toronto Western Hospital, University of Toronto, Toronto, Canada.,Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Rajasumi Rajalingam
- The Edmond J. Safra Program in Parkinson's Disease and The Morton and Gloria Shulman Movement Disorder Clinic, Toronto Western Hospital, University of Toronto, Toronto, Canada
| | - Marina A J Tijssen
- UMCG Expertise Centre Movement Disorders, Department of Neurology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Darius Ebrahimi-Fakhari
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Carolin Gabbert
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Christos Ganos
- Department of Neurology, Charité University Hospital Berlin, Berlin, Germany
| | - Rhia Ghosh
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Kishore R Kumar
- Molecular Medicine Laboratory and Department of Neurology, Concord Repatriation General Hospital, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Anthony E Lang
- The Edmond J. Safra Program in Parkinson's Disease and The Morton and Gloria Shulman Movement Disorder Clinic, Toronto Western Hospital, University of Toronto, Toronto, Canada
| | - Malco Rossi
- Movement Disorders Section, Neuroscience Department, Raul Carrea Institute for Neurological Research (FLENI), Buenos Aires, Argentina
| | - Sterre van der Veen
- UMCG Expertise Centre Movement Disorders, Department of Neurology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Bart van de Warrenburg
- Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Center of Expertise for Parkinson and Movement Disorders, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Tom Warner
- Department of Clinical & Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Connie Marras
- The Edmond J. Safra Program in Parkinson's Disease and The Morton and Gloria Shulman Movement Disorder Clinic, Toronto Western Hospital, University of Toronto, Toronto, Canada
| | | |
Collapse
|
59
|
Xu J, Minobe E, Kameyama M. Ca2+ Dyshomeostasis Links Risk Factors to Neurodegeneration in Parkinson’s Disease. Front Cell Neurosci 2022; 16:867385. [PMID: 35496903 PMCID: PMC9050104 DOI: 10.3389/fncel.2022.867385] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/23/2022] [Indexed: 12/06/2022] Open
Abstract
Parkinson’s disease (PD), a common neurodegenerative disease characterized by motor dysfunction, results from the death of dopaminergic neurons in the substantia nigra pars compacta (SNc). Although the precise causes of PD are still unknown, several risk factors for PD have been determined, including aging, genetic mutations, environmental factors, and gender. Currently, the molecular mechanisms underlying risk factor-related neurodegeneration in PD remain elusive. Endoplasmic reticulum stress, excessive reactive oxygen species production, and impaired autophagy have been implicated in neuronal death in the SNc in PD. Considering that these pathological processes are tightly associated with intracellular Ca2+, it is reasonable to hypothesize that dysregulation of Ca2+ handling may mediate risk factors-related PD pathogenesis. We review the recent findings on how risk factors cause Ca2+ dyshomeostasis and how aberrant Ca2+ handling triggers dopaminergic neurodegeneration in the SNc in PD, thus putting forward the possibility that manipulation of specific Ca2+ handling proteins and subcellular Ca2+ homeostasis may lead to new promising strategies for PD treatment.
Collapse
|
60
|
Genetics of cognitive dysfunction in Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2022; 269:195-226. [PMID: 35248195 DOI: 10.1016/bs.pbr.2022.01.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Presentation and progression of cognitive symptoms in Parkinson's disease are highly variable. PD is a genetically complex disorder with multiple genetic risk factors and understanding the role that genes play in cognitive outcomes is important for patient counseling and treatment. Currently, there are seven well-described genes that increase the risk for PD, with variable levels of penetrance: SNCA, LRRK2, VPS35, PRKN, PINK1, DJ1 and GBA. In addition, large, genome-wide association studies have identified multiple loci in our DNA which increase PD risk. In this chapter, we summarize what is currently known about each of the seven strongly-associated PD genes and select PD risk variants, including PITX3, TMEM106B, SNCA Rep1, APOɛ4, COMT and MAPT H1/H1, along with their respective relationships to cognition.
Collapse
|
61
|
Hemmings SMJ, Swart P, Womersely JS, Ovenden ES, van den Heuvel LL, McGregor NW, Meier S, Bardien S, Abrahams S, Tromp G, Emsley R, Carr J, Seedat S. RNA-seq analysis of gene expression profiles in posttraumatic stress disorder, Parkinson's disease and schizophrenia identifies roles for common and distinct biological pathways. DISCOVER MENTAL HEALTH 2022; 2:6. [PMID: 37861850 PMCID: PMC10501040 DOI: 10.1007/s44192-022-00009-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/14/2022] [Indexed: 10/21/2023]
Abstract
Evidence suggests that shared pathophysiological mechanisms in neuropsychiatric disorders (NPDs) may contribute to risk and resilience. We used single-gene and network-level transcriptomic approaches to investigate shared and disorder-specific processes underlying posttraumatic stress disorder (PTSD), Parkinson's disease (PD) and schizophrenia in a South African sample. RNA-seq was performed on blood obtained from cases and controls from each cohort. Gene expression and weighted gene correlation network analyses (WGCNA) were performed using DESeq2 and CEMiTool, respectively. Significant differences in gene expression were limited to the PTSD cohort. However, WGCNA implicated, amongst others, ribosomal expression, inflammation and ubiquitination as key players in the NPDs under investigation. Differential expression in ribosomal-related pathways was observed in the PTSD and PD cohorts, and focal adhesion and extracellular matrix pathways were implicated in PD and schizophrenia. We propose that, despite different phenotypic presentations, core transdiagnostic mechanisms may play important roles in the molecular aetiology of NPDs.
Collapse
Affiliation(s)
- Sian M J Hemmings
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa.
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa.
| | - Patricia Swart
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa
| | - Jacqueline S Womersely
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa
| | - Ellen S Ovenden
- Systems Genetics Working Group, Department of Genetics, Stellenbosch University, Stellenbosch, South Africa
| | - Leigh L van den Heuvel
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa
| | - Nathaniel W McGregor
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa
- Systems Genetics Working Group, Department of Genetics, Stellenbosch University, Stellenbosch, South Africa
| | - Stuart Meier
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Tuberculosis Bioinformatics Initiative, Stellenbosch University, Cape Town, South Africa
| | - Soraya Bardien
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
| | - Shameemah Abrahams
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gerard Tromp
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Tuberculosis Bioinformatics Initiative, Stellenbosch University, Cape Town, South Africa
- Centre for Bioinformatics and Computational Biology, Stellenbosch University, Stellenbosch, South Africa
| | - Robin Emsley
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa
| | - Jonathan Carr
- Division of Neurology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Soraya Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
62
|
Golan H, Volkov O, Shalom E. Nuclear imaging in Parkinson's disease: The past, the present, and the future. J Neurol Sci 2022; 436:120220. [DOI: 10.1016/j.jns.2022.120220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 02/15/2022] [Accepted: 03/02/2022] [Indexed: 01/15/2023]
|
63
|
Gouda NA, Elkamhawy A, Cho J. Emerging Therapeutic Strategies for Parkinson’s Disease and Future Prospects: A 2021 Update. Biomedicines 2022; 10:biomedicines10020371. [PMID: 35203580 PMCID: PMC8962417 DOI: 10.3390/biomedicines10020371] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 11/16/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder pathologically distinguished by degeneration of dopaminergic neurons in the substantia nigra pars compacta. Muscle rigidity, tremor, and bradykinesia are all clinical motor hallmarks of PD. Several pathways have been implicated in PD etiology, including mitochondrial dysfunction, impaired protein clearance, and neuroinflammation, but how these factors interact remains incompletely understood. Although many breakthroughs in PD therapy have been accomplished, there is currently no cure for PD, only trials to alleviate the related motor symptoms. To reduce or stop the clinical progression and mobility impairment, a disease-modifying approach that can directly target the etiology rather than offering symptomatic alleviation remains a major unmet clinical need in the management of PD. In this review, we briefly introduce current treatments and pathophysiology of PD. In addition, we address the novel innovative therapeutic targets for PD therapy, including α-synuclein, autophagy, neurodegeneration, neuroinflammation, and others. Several immunomodulatory approaches and stem cell research currently in clinical trials with PD patients are also discussed. Moreover, preclinical studies and clinical trials evaluating the efficacy of novel and repurposed therapeutic agents and their pragmatic applications with encouraging outcomes are summarized. Finally, molecular biomarkers under active investigation are presented as potentially valuable tools for early PD diagnosis.
Collapse
Affiliation(s)
- Noha A. Gouda
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang 10326, Korea; (N.A.G.); (A.E.)
| | - Ahmed Elkamhawy
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang 10326, Korea; (N.A.G.); (A.E.)
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Jungsook Cho
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang 10326, Korea; (N.A.G.); (A.E.)
- Correspondence:
| |
Collapse
|
64
|
Huang Y, He S, Chen Y, Sheng J, Fu Y, Du X, Yang Y, Liu H, Han Z, Huang Y, Wen Q, Zhou C, Zhou X, Hu S, Ma L. UCHL1 Promoted Polarization of M1 Macrophages by Regulating the PI3K/AKT Signaling Pathway. J Inflamm Res 2022; 15:735-746. [PMID: 35153498 PMCID: PMC8824699 DOI: 10.2147/jir.s343487] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Affiliation(s)
- Yulan Huang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Shitong He
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Yitian Chen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Junli Sheng
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Yuling Fu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Xialin Du
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Yalong Yang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Honglin Liu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Zhenyu Han
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Yingqi Huang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Qian Wen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Chaoying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Xinying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Shengfeng Hu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Li Ma
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Correspondence: Li Ma; Shengfeng Hu, Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China, Email ;
| |
Collapse
|
65
|
Li C, Ou R, Gu X, Hou Y, Chen Y, Wei Q, Zhang L, Lin J, Liu K, Huang J, Chen X, Song W, Zhao B, Wu Y, Shang H. ANXA1 and the risk for early-onset Parkinson's disease. Neurobiol Aging 2022; 112:212-214. [DOI: 10.1016/j.neurobiolaging.2022.01.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/15/2022] [Accepted: 01/26/2022] [Indexed: 01/01/2023]
|
66
|
Raket LL, Oudin Åström D, Norlin JM, Kellerborg K, Martinez-Martin P, Odin P. Impact of age at onset on symptom profiles, treatment characteristics and health-related quality of life in Parkinson's disease. Sci Rep 2022; 12:526. [PMID: 35017548 PMCID: PMC8752787 DOI: 10.1038/s41598-021-04356-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/20/2021] [Indexed: 12/30/2022] Open
Abstract
Parkinson's disease (PD) is typically considered an age-related disease, but the age at disease onset can vary by decades between patients. Aging and aging-associated diseases can affect the movement system independently of PD, and advanced age has previously been proposed to be associated with a more severe PD phenotype with accelerated progression. In this work, we investigated how interactions between PD progression and aging affect a wide range of outcomes related to PD motor and nonmotor symptoms as well as Health Related Quality of Life (HRQoL) and treatment characteristics. This population-based cohort study is based on 1436 PD patients from southern Sweden followed longitudinally for up to approximately 7.5 years from enrollment (3470 visits covering 2285 patient years, average follow-up time 1.7 years). Higher age at onset was generally associated with faster progression of motor symptoms, with a notable exception of dyskinesia and other levodopa-associated motor fluctuations that had less severe trajectories for patients with higher age at onset. Mixed results were observed for emergence of non-motor symptoms, while higher age at onset was generally associated with worse HRQoL trajectories. Accounting for these identified age-associated differences in disease progression could positively impact patient management and drug development efforts.
Collapse
Affiliation(s)
- Lars Lau Raket
- H. Lundbeck A/S, Valby, Denmark
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | | | - Jenny M Norlin
- The Swedish Institute for Health Economics, Lund, Sweden
| | | | - Pablo Martinez-Martin
- Center of Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), Carlos III Institute of Health, Madrid, Spain
| | - Per Odin
- Division of Neurology, Department of Clinical Sciences, Lund University, Lund, Sweden.
- Forskningsenhet Neurologi, Skåne University Hospital, Wigerthuset, Remissgatan 4, pl 1, 222 42, Lund, Sweden.
| |
Collapse
|
67
|
Emekli I, Tepgeç F, Samancı B, Toksoy G, Hasanoğulları Kına G, Tüfekçioğlu Z, Başaran S, Bilgiç B, Gürvit IH, Emre M, Uyguner ZO, Hanagasi HA. Clinical and molecular genetic findings of hereditary Parkinson's patients from Turkey. Parkinsonism Relat Disord 2021; 93:35-39. [PMID: 34781237 DOI: 10.1016/j.parkreldis.2021.10.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/28/2021] [Accepted: 10/23/2021] [Indexed: 11/16/2022]
Abstract
INTRODUCTION The majority of Parkinson's disease (PD) ensue late-onset with a complex spectrum of environmental and genetic risk factors. Awareness of genetic causes in patients with PD is essential for genetic counseling and future genotype-oriented therapeutic developments. METHODS Large pathogenic changes in eight PD-related genes and small pathogenic sequence variants in 22 PD-related genes were investigated simultaneously in 82 PD patients from 79 families where clinical evaluations were performed. The phenotypic characteristics of the patients with molecular changes were examined for genotype-phenotype relations. RESULTS Pathogenic variants in SNCA, PRKN, DJ-1, FBXO7, and GBA genes were determined in 25 patients from 24 families (24/79, 30%). Associated variants were found in PRKN in 14, SNCA in three, FBXO7 in two, and DJ-1 in one patient. A novel homozygous deletion (c.491delT, p.(V164Dfs*13) (SCV001733595)) leading to protein truncation in the PRKN gene was identified in two patients from the same family. Furthermore, heterozygous GBA gene variants were detected in five patients from different families. CONCLUSION It has been shown that the most common cause of genetically transmitted PD is the PRKN gene, while LRRK2 does not play an essential role in this selected population. It has been suggested that even if the autosomal recessive inheritance is expected, genes with autosomal dominant effects such as SNCA should not be overlooked and suggested for investigation. Our study is also the first for evaluating the pathogenic GBA variants' frequency in PD patients from Turkey.
Collapse
Affiliation(s)
- Inci Emekli
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Turkey.
| | - Fatih Tepgeç
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | - Bedia Samancı
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | - Güven Toksoy
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | | | - Zeynep Tüfekçioğlu
- Department of Neurology, Faculty of Medicine, Istanbul Aydin University, Istanbul, Turkey
| | - Seher Başaran
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | - Başar Bilgiç
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | - I Hakan Gürvit
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | - Murat Emre
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | - Zehra Oya Uyguner
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | - Hasmet A Hanagasi
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Turkey
| |
Collapse
|
68
|
Cook L, Schulze J, Verbrugge J, Beck JC, Marder KS, Saunders-Pullman R, Klein C, Naito A, Alcalay RN. The commercial genetic testing landscape for Parkinson's disease. Parkinsonism Relat Disord 2021; 92:107-111. [PMID: 34696975 PMCID: PMC8633166 DOI: 10.1016/j.parkreldis.2021.10.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/29/2021] [Accepted: 10/02/2021] [Indexed: 12/12/2022]
Abstract
INTRODUCTION There have been no specific guidelines regarding which genes should be tested in the clinical setting for Parkinson's disease (PD) or parkinsonism. We evaluated the types of clinical genetic testing offered for PD as the first step of our gene curation. METHODS The National Institutes of Health (NIH) Genetic Testing Registry (GTR) was queried on 12/7/2020 to identify current commercial PD genetic test offerings by clinical laboratories, internationally. RESULTS We identified 502 unique clinical genetic tests for PD, from 28 Clinical Laboratory Improvement Amendments (CLIA)-approved clinical laboratories. These included 11 diagnostic PD panels. The panels were notable for their differences in size, ranging from 5 to 62 genes. Five genes for variant query were included in all panels (SNCA, PRKN, PINK-1, PARK7 (DJ1), and LRRK2). Notably, the addition of the VPS35 and GBA genes was variable. Panel size differences stemmed from inclusion of genes linked to atypical parkinsonism and dystonia disorders, and genes in which the link to PD causation is controversial. CONCLUSION There is an urgent need for expert opinion regarding which genes should be included in a commercial laboratory multi-gene panel for PD.
Collapse
Affiliation(s)
- Lola Cook
- Department of Medical and Molecular Genetics (LC, JS, TF), Indiana University School of Medicine, Indianapolis, USA.
| | - Jeanine Schulze
- Department of Medical and Molecular Genetics (LC, JS, TF), Indiana University School of Medicine, Indianapolis, USA
| | - Jennifer Verbrugge
- Department of Medical and Molecular Genetics (LC, JS, TF), Indiana University School of Medicine, Indianapolis, USA
| | | | - Karen S Marder
- Department of Neurology, Columbia University College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | | | - Christine Klein
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | | | - Roy N Alcalay
- Department of Neurology, Columbia University College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
69
|
Elsayed I, Martinez-Carrasco A, Cornejo-Olivas M, Bandres-Ciga S. Mapping the Diverse and Inclusive Future of Parkinson's Disease Genetics and Its Widespread Impact. Genes (Basel) 2021; 12:1681. [PMID: 34828286 PMCID: PMC8624537 DOI: 10.3390/genes12111681] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/27/2022] Open
Abstract
Over the last decades, genetics has been the engine that has pushed us along on our voyage to understand the etiology of Parkinson's disease (PD). Although a large number of risk loci and causative mutations for PD have been identified, it is clear that much more needs to be done to solve the missing heritability mystery. Despite remarkable efforts, as a field, we have failed in terms of diversity and inclusivity. The vast majority of genetic studies in PD have focused on individuals of European ancestry, leading to a gap of knowledge on the existing genetic differences across populations and PD as a whole. As we move forward, shedding light on the genetic architecture contributing to PD in non-European populations is essential, and will provide novel insight into the generalized genetic map of the disease. In this review, we discuss how better representation of understudied ancestral groups in PD genetics research requires addressing and resolving all the challenges that hinder the inclusion of these populations. We further provide an overview of PD genetics in the clinics, covering the current challenges and limitations of genetic testing and counseling. Finally, we describe the impact of worldwide collaborative initiatives in the field, shaping the future of the new era of PD genetics as we advance in our understanding of the genetic architecture of PD.
Collapse
Affiliation(s)
- Inas Elsayed
- Faculty of Pharmacy, University of Gezira, Wad Medani P.O. Box 20, Sudan;
- International Parkinson Disease Genomics Consortium (IPDGC)-Africa, University of Gezira, Wad Medani P.O. Box 20, Sudan
| | | | - Mario Cornejo-Olivas
- Neurogenetics Research Center, Instituto Nacional de Ciencias Neurológicas, Lima 15003, Peru;
- Center for Global Health, Universidad Peruana Cayetano Heredia, Lima 15103, Peru
| | - Sara Bandres-Ciga
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
70
|
Hernandez SM, Tikhonova EB, Baca KR, Zhao F, Zhu X, Karamyshev AL. Unexpected Implication of SRP and AGO2 in Parkinson's Disease: Involvement in Alpha-Synuclein Biogenesis. Cells 2021; 10:2792. [PMID: 34685771 PMCID: PMC8534902 DOI: 10.3390/cells10102792] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/09/2021] [Accepted: 10/12/2021] [Indexed: 01/21/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder classified by the loss of dopaminergic neurons in the substantia nigra pars compacta, the region of the brain that is responsible for motor control. Surviving neurons in this region contain aggregated protein alpha-Synuclein (αSyn) in the form of cytoplasmic inclusions, referred to as Lewy bodies. Changes in αSyn expression are also associated with PD and its progression. Previously, we demonstrated that signal recognition particle (SRP) and Argonaute 2 (AGO2) proteins are involved in protein quality control at the ribosome during translation. We also demonstrated that SRP has an mRNA protection function in addition to a protein targeting function, thus controlling mRNA and protein expression. In this study, we tested involvement of these factors in αSyn biogenesis. We hypothesize that loss of these factors may interfere with αSyn expression, and subsequently, be associated with PD. Using depletion assays in human cell culture and analysis of these proteins in the brains of deceased PD patients, we demonstrate that SRP and AGO2 are involved in the control of αSyn expression and AGO2 has reduced expression in PD. We show for the first time that SRP is involved in mRNA protection of αSyn, a protein that does not have a signal sequence or transmembrane span. Our findings suggest that SRP may interact with a hydrophobic domain in the middle of αSyn during translation. Understanding the molecular mechanisms controlling αSyn biogenesis in cells is vital to developing preventative therapies against PD.
Collapse
Affiliation(s)
- Sarah M. Hernandez
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (S.M.H.); (E.B.T.); (K.R.B.)
| | - Elena B. Tikhonova
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (S.M.H.); (E.B.T.); (K.R.B.)
| | - Kristen R. Baca
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (S.M.H.); (E.B.T.); (K.R.B.)
- Center for the Integration of STEM Education and Research (CISER), Texas Tech University, Lubbock, TX 79409, USA
| | - Fanpeng Zhao
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA; (F.Z.); (X.Z.)
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA; (F.Z.); (X.Z.)
| | - Andrey L. Karamyshev
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (S.M.H.); (E.B.T.); (K.R.B.)
| |
Collapse
|
71
|
Dopaminergic Axons: Key Recitalists in Parkinson's Disease. Neurochem Res 2021; 47:234-248. [PMID: 34637100 DOI: 10.1007/s11064-021-03464-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 12/20/2022]
Abstract
Parkinson's disease (PD) is associated with dopamine depletion in the striatum owing to the selective and progressive loss of the nigrostriatal dopaminergic neurons, which results in motor dysfunction and secondary clinical manifestations. The dopamine level in the striatum is preserved because of the innervation of the substantia nigra (SN) dopaminergic neurons into it. Therefore, protection of the SN neurons is crucial for maintaining the dopamine level in the striatum and for ensuring the desired motor coordination. Several strategies have been devised to protect the degenerating dopaminergic neurons or to restore the dopamine levels for treating PD. Most of the methods focus exclusively on preventing cell body death in the neurons. Although advances have been made in understanding the disease, the search for disease-modifying drugs is an ongoing process. The present review describes the evidence from studies involving patients with PD as well as PD models that axon terminals are highly vulnerable to exogenous and endogenous insults and degenerate at the early stage of the disease. Impairment of mitochondrial dynamics, Ca2+ homeostasis, axonal transport, and loss of plasticity of axon terminals appear before the neuronal degeneration in PD. Furthermore, distortion of synaptic morphology and reduction of postsynaptic dendritic spines are the neuropathological hallmarks of early-stage disease. Thus, the review proposes a shift in focus from discerning the mechanism of neuronal cell body loss and targeting it to an entirely different approach of preventing axonal degeneration. The review also suggests appropriate strategies to prevent the loss of synaptic terminals, which could induce regrowth of the axon and its auxiliary fibers and might offer relief from the symptomatic features of PD.
Collapse
|
72
|
Guadagnolo D, Piane M, Torrisi MR, Pizzuti A, Petrucci S. Genotype-Phenotype Correlations in Monogenic Parkinson Disease: A Review on Clinical and Molecular Findings. Front Neurol 2021; 12:648588. [PMID: 34630269 PMCID: PMC8494251 DOI: 10.3389/fneur.2021.648588] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/08/2021] [Indexed: 12/30/2022] Open
Abstract
Parkinson disease (PD) is a complex neurodegenerative disorder, usually with multifactorial etiology. It is characterized by prominent movement disorders and non-motor symptoms. Movement disorders commonly include bradykinesia, rigidity, and resting tremor. Non-motor symptoms can include behavior disorders, sleep disturbances, hyposmia, cognitive impairment, and depression. A fraction of PD cases instead is due to Parkinsonian conditions with Mendelian inheritance. The study of the genetic causes of these phenotypes has shed light onto common pathogenetic mechanisms underlying Parkinsonian conditions. Monogenic Parkinsonisms can present autosomal dominant, autosomal recessive, or even X-linked inheritance patterns. Clinical presentations vary from forms indistinguishable from idiopathic PD to severe childhood-onset conditions with other neurological signs. We provided a comprehensive description of each condition, discussing current knowledge on genotype-phenotype correlations. Despite the broad clinical spectrum and the many genes involved, the phenotype appears to be related to the disrupted cell function and inheritance pattern, and several assumptions about genotype-phenotype correlations can be made. The interest in these assumptions is not merely speculative, in the light of novel promising targeted therapies currently under development.
Collapse
Affiliation(s)
- Daniele Guadagnolo
- Department of Experimental Medicine, Policlinico Umberto i Hospital, Sapienza University of Rome, Rome, Italy
| | - Maria Piane
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy.,Medical Genetics and Advanced Cell Diagnostics Unit, S. Andrea University Hospital, Rome, Italy
| | - Maria Rosaria Torrisi
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy.,Medical Genetics and Advanced Cell Diagnostics Unit, S. Andrea University Hospital, Rome, Italy
| | - Antonio Pizzuti
- Department of Experimental Medicine, Policlinico Umberto i Hospital, Sapienza University of Rome, Rome, Italy
| | - Simona Petrucci
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy.,Medical Genetics and Advanced Cell Diagnostics Unit, S. Andrea University Hospital, Rome, Italy
| |
Collapse
|
73
|
Meng J, Wang F, Ji L, Liang Y, Nian W, Song L, Zhu A. Comprehensive methylation profile of CSF cfDNA revealed pathogenesis and diagnostic markers for early-onset Parkinson's disease. Epigenomics 2021; 13:1637-1651. [PMID: 34664993 DOI: 10.2217/epi-2021-0176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/16/2021] [Indexed: 02/08/2023] Open
Abstract
Background: Early-onset Parkinson's disease (EOPD) is one uncommon Parkinson's disease subtype with characteristic clinicopathological features. The full epigenomic profile of EOPD is largely unknown. Methods: We performed the first study to investigate the EOPD full methylation profile of cerebrospinal fluid (CSF) cell-free DNA (cfDNA) from 26 EOPD patients and 10 control patients. Results: 2220 differentially methylated genes were identified in EOPD. Hypermethylation far outweighed hypomethylation in gene numbers. Clustering and enrichment analyses identified aberrant neuronal function and immune response. Weighted correlation network analysis demonstrated significant correlation between methylation signatures and clock drawing test (CDT), mini-mental state examination (MMSE), education, working status, alcohol drinking history and Hamilton anxiety scale (HAMA). Several key networking genes in EOPD aberrant methylation were also identified. Conclusions: The methylation profile and signatures of CSF cfDNA were revealed for the first time in EOPD. Aberrant methylation signatures were correlated with education, working status, alcohol drinking history, CDT, MMSE and HAMA.
Collapse
Affiliation(s)
- Jie Meng
- Institution of Geriatric, Qinghai Provincial People's Hospital, Xining, 810007, PR China
- Department of Neurology & State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, & Collaborative Innovation Center for Biotherapy, Chengdu, 610041, PR China
| | - Fenglin Wang
- Department of genetics and cell biology, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Lei Ji
- Institution of Geriatric, Qinghai Provincial People's Hospital, Xining, 810007, PR China
| | - Yuhua Liang
- Institution of Geriatric, Qinghai Provincial People's Hospital, Xining, 810007, PR China
| | - Wei Nian
- Institution of Geriatric, Qinghai Provincial People's Hospital, Xining, 810007, PR China
| | - Lele Song
- Institution of Geriatric, Qinghai Provincial People's Hospital, Xining, 810007, PR China
- Department of Radiotherapy, The Eighth Medical Center of the Chinese PLA General Hospital, Beijing, 100091, PR China
| | - Aiqin Zhu
- Institution of Geriatric, Qinghai Provincial People's Hospital, Xining, 810007, PR China
| |
Collapse
|
74
|
Muzio L, Viotti A, Martino G. Microglia in Neuroinflammation and Neurodegeneration: From Understanding to Therapy. Front Neurosci 2021; 15:742065. [PMID: 34630027 PMCID: PMC8497816 DOI: 10.3389/fnins.2021.742065] [Citation(s) in RCA: 261] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/25/2021] [Indexed: 12/19/2022] Open
Abstract
Microglia are the resident macrophages of the central nervous system (CNS) acting as the first line of defense in the brain by phagocytosing harmful pathogens and cellular debris. Microglia emerge from early erythromyeloid progenitors of the yolk sac and enter the developing brain before the establishment of a fully mature blood-brain barrier. In physiological conditions, during brain development, microglia contribute to CNS homeostasis by supporting cell proliferation of neural precursors. In post-natal life, such cells contribute to preserving the integrity of neuronal circuits by sculpting synapses. After a CNS injury, microglia change their morphology and down-regulate those genes supporting homeostatic functions. However, it is still unclear whether such changes are accompanied by molecular and functional modifications that might contribute to the pathological process. While comprehensive transcriptome analyses at the single-cell level have identified specific gene perturbations occurring in the "pathological" microglia, still the precise protective/detrimental role of microglia in neurological disorders is far from being fully elucidated. In this review, the results so far obtained regarding the role of microglia in neurodegenerative disorders will be discussed. There is solid and sound evidence suggesting that regulating microglia functions during disease pathology might represent a strategy to develop future therapies aimed at counteracting brain degeneration in multiple sclerosis, Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Luca Muzio
- Neuroimmunology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | | | | |
Collapse
|
75
|
Riboldi GM, Frattini E, Monfrini E, Frucht SJ, Fonzo AD. A Practical Approach to Early-Onset Parkinsonism. JOURNAL OF PARKINSONS DISEASE 2021; 12:1-26. [PMID: 34569973 PMCID: PMC8842790 DOI: 10.3233/jpd-212815] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Early-onset parkinsonism (EO parkinsonism), defined as subjects with disease onset before the age of 40 or 50 years, can be the main clinical presentation of a variety of conditions that are important to differentiate. Although rarer than classical late-onset Parkinson’s disease (PD) and not infrequently overlapping with forms of juvenile onset PD, a correct diagnosis of the specific cause of EO parkinsonism is critical for offering appropriate counseling to patients, for family and work planning, and to select the most appropriate symptomatic or etiopathogenic treatments. Clinical features, radiological and laboratory findings are crucial for guiding the differential diagnosis. Here we summarize the most important conditions associated with primary and secondary EO parkinsonism. We also proposed a practical approach based on the current literature and expert opinion to help movement disorders specialists and neurologists navigate this complex and challenging landscape.
Collapse
Affiliation(s)
- Giulietta M Riboldi
- The Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, Department of Neurology, NYU Langone Health, New York, NY, USA
| | - Emanuele Frattini
- IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy.,Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation , University of Milan, Milan, Italy
| | - Edoardo Monfrini
- IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy.,Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation , University of Milan, Milan, Italy
| | - Steven J Frucht
- The Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, Department of Neurology, NYU Langone Health, New York, NY, USA
| | - Alessio Di Fonzo
- IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy
| |
Collapse
|
76
|
Gu X, Hou Y, Chen Y, Ou R, Cao B, Wei Q, Zhang L, Song W, Zhao B, Wu Y, Li C, Shang H. Enrichment of rare variants in E3 ubiquitin ligase genes in Early onset Parkinson's disease. Neurobiol Aging 2021; 109:273-278. [PMID: 34544586 DOI: 10.1016/j.neurobiolaging.2021.08.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/15/2021] [Accepted: 08/18/2021] [Indexed: 02/08/2023]
Abstract
Altered ubiquitin signaling and disrupted protein quality control have been implicated in the pathogenesis of PD. The aim of the study was to systematically examine the overlaps between E3 ubiquitin ligase genes and early onset PD (EOPD). A total of 695 EOPD patients were analyzed aggregate burden for rare variants (MAF <0.001 and MAF <0.0001) in a total of 44 E3 ubiquitin ligase genes causing disorders involved in the nervous system. There was significant enrichment of the rare and rare damaging variants in the E3 ubiquitin ligase genes in EOPD patients. Detailly, in the gene-based level, the strongest associations were found in HERC1, IRF2BPL, KMT2D, RAPSN, RLIM, RNF168 and RNF216. Our findings highlighted the importance of UPS mechanism in the pathogenesis of PD from the genetic perspective. Moreover, our study also expanded the susceptible gene spectrum for PD.
Collapse
Affiliation(s)
- Xiaojing Gu
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanbing Hou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yongping Chen
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruwei Ou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bei Cao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qianqian Wei
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lingyu Zhang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei Song
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bi Zhao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ying Wu
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chunyu Li
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huifang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
77
|
Chang KH, Huang CY, Ou-Yang CH, Ho CH, Lin HY, Hsu CL, Chen YT, Chou YC, Chen YJ, Chen Y, Lin JL, Wang JK, Lin PW, Lin YR, Lin MH, Tseng CK, Lin CH. In vitro genome editing rescues parkinsonism phenotypes in induced pluripotent stem cells-derived dopaminergic neurons carrying LRRK2 p.G2019S mutation. Stem Cell Res Ther 2021; 12:508. [PMID: 34551822 PMCID: PMC8456557 DOI: 10.1186/s13287-021-02585-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/27/2021] [Indexed: 12/26/2022] Open
Abstract
Background The c.G6055A (p.G2019S) mutation in leucine-rich repeat kinase 2 (LRRK2) is the most prevalent genetic cause of Parkinson’s disease (PD). CRISPR/Cas9-mediated genome editing by homology-directed repair (HDR) has been applied to correct the mutation but may create small insertions and deletions (indels) due to double-strand DNA breaks. Adenine base editors (ABEs) could convert targeted A·T to G·C in genomic DNA without double-strand breaks. However, the correction efficiency of ABE in LRRK2 c.G6055A (p.G2019S) mutation remains unknown yet. This study aimed to compare the mutation correction efficiencies and off-target effects between HDR and ABEs in induced pluripotent stem cells (iPSCs) carrying LRRK2 c.G6055A (p.G2019S) mutation. Methods A set of mutation-corrected isogenic lines by editing the LRRK2 c.G6055A (p.G2019S) mutation in a PD patient-derived iPSC line using HDR or ABE were established. The mutation correction efficacies, off-target effects, and indels between HDR and ABE were compared. Comparative transcriptomic and proteomic analyses between the LRRK2 p.G2019S iPSCs and isogenic control cells were performed to identify novel molecular targets involved in LRRK2-parkinsonism pathways. Results ABE had a higher correction rate (13/53 clones, 24.5%) than HDR (3/47 clones, 6.4%). Twenty-seven HDR clones (57.4%), but no ABE clones, had deletions, though 14 ABE clones (26.4%) had off-target mutations. The corrected isogenic iPSC-derived dopaminergic neurons exhibited reduced LRRK2 kinase activity, decreased phospho-α-synuclein expression, and mitigated neurite shrinkage and apoptosis. Comparative transcriptomic and proteomic analysis identified different gene expression patterns in energy metabolism, protein degradation, and peroxisome proliferator-activated receptor pathways between the mutant and isogenic control cells. Conclusions The results of this study envision that ABE could directly correct the pathogenic mutation in iPSCs for reversing disease-related phenotypes in neuropathology and exploring novel pathophysiological targets in PD.
Collapse
Affiliation(s)
- Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital-Linkou Medical Center, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Cheng-Yen Huang
- The First Core Laboratory, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chih-Hsin Ou-Yang
- Department of Neurology, National Taiwan University Hospital and School of Medicine, Taipei, 100, Taiwan
| | - Chang-Han Ho
- Department of Neurology, National Taiwan University Hospital and School of Medicine, Taipei, 100, Taiwan
| | - Han-Yi Lin
- Department of Neurology, National Taiwan University Hospital and School of Medicine, Taipei, 100, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - You-Tzung Chen
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Chi Chou
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Yi-Jing Chen
- Department of Neurology, Chang Gung Memorial Hospital-Linkou Medical Center, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Ying Chen
- Department of Neurology, Chang Gung Memorial Hospital-Linkou Medical Center, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Jia-Li Lin
- Department of Neurology, Chang Gung Memorial Hospital-Linkou Medical Center, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Ji-Kuan Wang
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Wen Lin
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ying-Ru Lin
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Miao-Hsia Lin
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chi-Kang Tseng
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital and School of Medicine, Taipei, 100, Taiwan.
| |
Collapse
|
78
|
UQCRC1 engages cytochrome c for neuronal apoptotic cell death. Cell Rep 2021; 36:109729. [PMID: 34551295 DOI: 10.1016/j.celrep.2021.109729] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 07/15/2021] [Accepted: 08/27/2021] [Indexed: 12/14/2022] Open
Abstract
Human ubiquinol-cytochrome c reductase core protein 1 (UQCRC1) is an evolutionarily conserved core subunit of mitochondrial respiratory chain complex III. We recently identified the disease-associated variants of UQCRC1 from patients with familial parkinsonism, but its function remains unclear. Here we investigate the endogenous function of UQCRC1 in the human neuronal cell line and the Drosophila nervous system. Flies with neuronal knockdown of uqcrc1 exhibit age-dependent parkinsonism-resembling defects, including dopaminergic neuron reduction and locomotor decline, and are ameliorated by UQCRC1 expression. Lethality of uqcrc1-KO is also rescued by neuronally expressing UQCRC1, but not the disease-causing variant, providing a platform to discern the pathogenicity of this mutation. Furthermore, UQCRC1 associates with the apoptosis trigger cytochrome c (cyt-c), and uqcrc1 deficiency increases cyt-c in the cytoplasmic fraction and activates the caspase cascade. Depleting cyt-c or expression of the anti-apoptotic p35 ameliorates uqcrc1-mediated neurodegeneration. Our findings identify a role for UQCRC1 in regulating cyt-c-induced apoptosis.
Collapse
|
79
|
Courtin T, Brice A. Monogenic PD in Brazil: a step towards precision medicine. ARQUIVOS DE NEURO-PSIQUIATRIA 2021; 79:563-564. [PMID: 34231651 DOI: 10.1590/0004-282x-anp-2021-e007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/01/2021] [Indexed: 11/21/2022]
Affiliation(s)
- Thomas Courtin
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, INSERM, CNRS, Département de Génétique, Hôpital Pitié-Salpêtrière, APHP, Paris, France
| | - Alexis Brice
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, INSERM, CNRS, Département de Génétique, Hôpital Pitié-Salpêtrière, APHP, Paris, France
| |
Collapse
|
80
|
Santos-Lobato BL, Schumacher-Schuh A, Mata IF, Letro GH, Braga-Neto P, Brandão PRP, Godeiro-Junior CO, Coletta MVD, Camargos ST, Borges V, Rieder CRM, Tumas V. Genetics of Parkinson's disease in Brazil: a systematic review of monogenic forms. ARQUIVOS DE NEURO-PSIQUIATRIA 2021; 79:612-623. [PMID: 34468500 DOI: 10.1590/0004-282x-anp-2020-0409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 09/17/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Increasing numbers of mutations causing monogenic forms of Parkinson's disease (PD) have been described, mostly among patients in Europe and North America. Since genetic architecture varies between different populations, studying the specific genetic profile of Brazilian patients is essential for improving genetic counseling and for selecting patients for clinical trials. OBJECTIVE We conducted a systematic review to identify genetic studies on Brazilian patients and to set a background for future studies on monogenic forms of PD in Brazil. METHODS We searched MEDLINE, EMBASE and Web of Science from inception to December 2019 using terms for "Parkinson's disease", "genetics" and "Brazil". Two independent reviewers extracted the data. For the genes LRRK2 and PRKN, the estimated prevalence was calculated for each study, and a meta-analysis was performed. RESULTS A total of 32 studies were included, comprising 94 Brazilian patients with PD with a causative mutation, identified from among 2,872 screened patients (3.2%). PRKN mutations were causative of PD in 48 patients out of 576 (8.3%). LRRK2 mutations were identified in 40 out of 1,556 patients (2.5%), and p.G2019S was the most common mutation (2.2%). CONCLUSIONS PRKN is the most common autosomal recessive cause of PD, and LRRK2 is the most common autosomal dominant form. We observed that there was a lack of robust epidemiological studies on PD genetics in Brazil and, especially, that the diversity of Brazil's population had not been considered.
Collapse
Affiliation(s)
- Bruno L Santos-Lobato
- Universidade Federal do Pará, Laboratório de Neuropatologia Experimental, Belém PA, Brazil.,Hospital Ophir Loyola, Serviço de Neurologia, Belém PA, Brazil
| | - Artur Schumacher-Schuh
- Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Porto Alegre RS, Brazil.,Universidade Federal do Rio Grande do Sul, Departamento de Farmacologia, Porto Alegre RS, Brazil
| | - Ignacio F Mata
- Lerner Research Institute, Genomic Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Grace H Letro
- Pontifícia Universidade Católica de Campinas, Centro de Ciências da Vida, Campinas SP, Brazil
| | - Pedro Braga-Neto
- Universidade Federal do Ceará, Departamento de Medicina Clínica, Serviço de Neurologia e Neurocirurgia, Fortaleza CE, Brazil
| | - Pedro R P Brandão
- Universidade de Brasília, Laboratório de Neurociências e Comportamento, Brasília DF, Brazil
| | - Clécio O Godeiro-Junior
- Universidade Federal do Rio Grande do Norte, Departamento de Medicina Integrada, Natal RN, Brazil
| | | | - Sarah T Camargos
- Universidade Federal de Minas Gerais, Departamento de Medicina Interna, Belo Horizonte MG, Brazil
| | - Vanderci Borges
- Universidade Federal de São Paulo, Departamento de Neurologia e Neurocirurgia, Setor de Transtornos de Movimento, São Paulo SP, Brazil
| | - Carlos R M Rieder
- Universidade Federal de Ciências da Saúde de Porto Alegre, Departamento de Neurologia, Porto Alegre RS, Brazil
| | - Vitor Tumas
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Departamento de Neurociências e Ciências do Comportamento, Ribeirão Preto SP, Brazil
| |
Collapse
|
81
|
Heurtaux T, Kirchmeyer M, Koncina E, Felten P, Richart L, Uriarte Huarte O, Schohn H, Mittelbronn M. Apomorphine Reduces A53T α-Synuclein-Induced Microglial Reactivity Through Activation of NRF2 Signalling Pathway. Cell Mol Neurobiol 2021; 42:2673-2695. [PMID: 34415465 PMCID: PMC9560932 DOI: 10.1007/s10571-021-01131-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 07/18/2021] [Indexed: 12/19/2022]
Abstract
The chiral molecule, apomorphine, is currently used for the treatment of Parkinson’s disease (PD). As a potent dopamine receptor agonist, this lipophilic compound is especially effective for treating motor fluctuations in advanced PD patients. In addition to its receptor-mediated actions, apomorphine has also antioxidant and free radical scavenger activities. Neuroinflammation, oxidative stress, and microglia reactivity have emerged as central players in PD. Thus, modulating microglia activation in PD may be a valid therapeutic strategy. We previously reported that murine microglia are strongly activated upon exposure to A53T mutant α-synuclein. The present study was designed to investigate whether apomorphine enantiomers could modulate this A53T-induced microglial activation. Taken together, the results provided evidence that apomorphine enantiomers decrease A53T-induced microgliosis, through the activation of the NRF2 signalling pathway, leading to a lower pro-inflammatory state and restoring the phagocytic activity. Suppressing NRF2 recruitment (trigonelline exposure) or silencing specifically Nfe2l2 gene (siRNA treatment) abolished or strongly decreased the anti-inflammatory activity of apomorphine. In conclusion, apomorphine, which is already used in PD patients to mimic dopamine activity, may also be suitable to decrease α-synuclein-induced microglial reactivity.
Collapse
Affiliation(s)
- Tony Heurtaux
- Faculty of Science, Technology and Medicine, University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg.
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, 7, Avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg.
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg.
| | - Melanie Kirchmeyer
- Faculty of Science, Technology and Medicine, University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, 7, Avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Eric Koncina
- Faculty of Science, Technology and Medicine, University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, 7, Avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Paul Felten
- Faculty of Science, Technology and Medicine, University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, 7, Avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Lorraine Richart
- Faculty of Science, Technology and Medicine, University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
- Department of Oncology (DONC), Luxembourg Institute of Health (LIH), L-1526, Strassen, Luxembourg
| | - Oihane Uriarte Huarte
- Faculty of Science, Technology and Medicine, University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Herve Schohn
- CNRS, CRAN, Université de Lorraine, 54000, Nancy, France
| | - Michel Mittelbronn
- Faculty of Science, Technology and Medicine, University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, 7, Avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
- Department of Oncology (DONC), Luxembourg Institute of Health (LIH), L-1526, Strassen, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), L-3555, Dudelange, Luxembourg
| |
Collapse
|
82
|
The Impact of SNCA Variations and Its Product Alpha-Synuclein on Non-Motor Features of Parkinson's Disease. Life (Basel) 2021; 11:life11080804. [PMID: 34440548 PMCID: PMC8401994 DOI: 10.3390/life11080804] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson’s disease (PD) is a common and progressive neurodegenerative disease, caused by the loss of dopaminergic neurons in the substantia nigra pars compacta in the midbrain, which is clinically characterized by a constellation of motor and non-motor manifestations. The latter include hyposmia, constipation, depression, pain and, in later stages, cognitive decline and dysautonomia. The main pathological features of PD are neuronal loss and consequent accumulation of Lewy bodies (LB) in the surviving neurons. Alpha-synuclein (α-syn) is the main component of LB, and α-syn aggregation and accumulation perpetuate neuronal degeneration. Mutations in the α-syn gene (SNCA) were the first genetic cause of PD to be identified. Generally, patients carrying SNCA mutations present early-onset parkinsonism with severe and early non-motor symptoms, including cognitive decline. Several SNCA polymorphisms were also identified, and some of them showed association with non-motor manifestations. The functional role of these polymorphisms is only partially understood. In this review we explore the contribution of SNCA and its product, α-syn, in predisposing to the non-motor manifestations of PD.
Collapse
|
83
|
Hustad E, Myklebust TÅ, Gulati S, Aasly JO. Increased Mortality in Young-Onset Parkinson's Disease. J Mov Disord 2021; 14:214-220. [PMID: 34315208 PMCID: PMC8490197 DOI: 10.14802/jmd.21029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 04/28/2021] [Indexed: 11/27/2022] Open
Abstract
Objective
Few studies have followed Parkinson’s disease (PD) patients from the time of diagnosis to the date of death. This study compared mortality in the Trondheim PD cohort to the general population, investigated causes of death and analyzed the associations between mortality and age at disease onset (AAO) and cognitive decline defined as Montreal Cognitive Assessment (MoCA) score below 26. Methods
The cohort was followed longitudinally from 1997. By the end of January 2020, 587 patients had died. Comparisons to the Norwegian population were performed by calculating standardized mortality ratios (SMRs). Survival curves were estimated using the standard Kaplan-Meier estimator, and multivariable Cox proportional hazard models were estimated to investigate associations. Results
SMR was 2.28 [95% confidence interval (CI): 2.13–2.44] for the whole cohort. For participants with AAO 20–39 years, the SMR was 5.55 (95% CI: 3.38–8.61). Median survival was 15 years (95% CI: 14.2–15.5) for the whole cohort. Early-onset PD (EOPD) patients (AAO < 50 years) had the longest median survival time. For all groups, there was a significant shortening in median survival time and an almost 3-fold higher age- and sex-adjusted hazard ratio for death when the MoCA score decreased below 26. Conclusion
PD patients with an AAO before 40 years had a more than fivefold higher mortality rate compared to a similar general population. EOPD patients had the longest median survival; however, their life expectancy was reduced to a greater degree than that of late-onset PD patients. Cognitive impairment was strongly associated with mortality in PD.
Collapse
Affiliation(s)
- Eldbjørg Hustad
- Department of Neuromedicine and Movement Science (INB), NTNU, Faculty of Medicine and Health Sciences, Trondheim, Norway
| | - Tor Åge Myklebust
- Department of Research and Innovation, More and Romsdal Hospital Trust, Ålesund, Norway
| | - Sasha Gulati
- Department of Neuromedicine and Movement Science (INB), NTNU, Faculty of Medicine and Health Sciences, Trondheim, Norway.,Department of Neurosurgery, St. Olavs Hospital, Trondheim, Norway
| | - Jan O Aasly
- Department of Neuromedicine and Movement Science (INB), NTNU, Faculty of Medicine and Health Sciences, Trondheim, Norway.,Department of Neurology, St. Olavs Hospital, Trondheim, Norway
| |
Collapse
|
84
|
Cui X, Xu C, Zhang L, Wang Y. Identification of Parkinson's Disease-Causing Genes via Omics Data. Front Genet 2021; 12:712164. [PMID: 34394198 PMCID: PMC8355633 DOI: 10.3389/fgene.2021.712164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/02/2021] [Indexed: 01/13/2023] Open
Abstract
Parkinson's disease (PD) is the second most frequent neurogenic disease after Alzheimer's disease. The clinical manifestations include mostly motor disorders, such as bradykinesia, myotonia, and static tremors. Since the cause of this pathological features remain unclear, there is currently no radical treatment for PD. Environmental and genetic factors are thought to contribute to the pathology of PD. To identify the genetic factors, some studies employed the Genome-Wide Association Studies (GWAS) method and detected certain genes closely related to PD. However, the functions of these gene mutants in the development of PD are unknown. Combining GWAS and expression Quantitative Trait Loci (eQTL) analysis, the biological meaning of mutation could be explained to some extent. Therefore, the present investigation used Summary data-based Mendelian Randomization (SMR) analysis to integrate of two PD GWAS datasets and four eQTL datasets with the objective of identifying casual genes. Using this strategy, we found six Single Nucleotide Polymorphism (SNP) loci which could cause the development of PD through altering the susceptibility gene expression, and three risk genes: Synuclein Alpha (SNCA), Mitochondrial Poly(A) Polymerase (MTPAP), and RP11-305E6.4. We proved the accuracy of results through case studies and inferred the functions of these genes in PD. Overall, this study provides insights into the genetic mechanism behind PD, which is crucial for the study of the development of this disease and its diagnosis and treatment.
Collapse
Affiliation(s)
- Xinran Cui
- Center for Bioinformatics, School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Chen Xu
- Center for Bioinformatics, School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Liyuan Zhang
- Center for Bioinformatics, School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yadong Wang
- Center for Bioinformatics, School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
85
|
Di Lazzaro G, Magrinelli F, Estevez-Fraga C, Valente EM, Pisani A, Bhatia KP. X-Linked Parkinsonism: Phenotypic and Genetic Heterogeneity. Mov Disord 2021; 36:1511-1525. [PMID: 33960519 DOI: 10.1002/mds.28565] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 02/06/2023] Open
Abstract
X-linked parkinsonism encompasses rare heterogeneous disorders mainly inherited as a recessive trait, therefore being more prevalent in males. Recent developments have revealed a complex underlying panorama, including a spectrum of disorders in which parkinsonism is variably associated with additional neurological and non-neurological signs. In particular, a childhood-onset encephalopathy with epilepsy and/or cognitive disability is the most common feature. Their genetic basis is also heterogeneous, with many causative genes and different mutation types ranging from "classical" coding variants to intronic repeat expansions. In this review, we provide an updated overview of the phenotypic and genetic spectrum of the most relevant X-linked parkinsonian syndromes, namely X-linked dystonia-parkinsonism (XDP, Lubag disease), fragile X-associated tremor/ataxia syndrome (FXTAS), beta-propeller protein-associated neurodegeneration (BPAN, NBIA/PARK-WDR45), Fabry disease, Waisman syndrome, methyl CpG-binding protein 2 (MeCP2) spectrum disorder, phosphoglycerate kinase-1 deficiency syndrome (PGK1) and X-linked parkinsonism and spasticity (XPDS). All clinical and radiological features reported in the literature have been reviewed. Epilepsy occasionally represents the symptom of onset, predating parkinsonism even by a few years; action tremor is another common feature along with akinetic-rigid parkinsonism. A focus on the genetic background and its pathophysiological implications is provided. The pathogenesis of these disorders ranges from well-defined metabolic alterations (PGK1) to non-specific lysosomal dysfunctions (XPDS) and vesicular trafficking alterations (Waisman syndrome). However, in other cases it still remains poorly defined. Recognition of the phenotypic and genetic heterogeneity of X-linked parkinsonism has important implications for diagnosis, management, and genetic counseling. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Giulia Di Lazzaro
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Francesca Magrinelli
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Carlos Estevez-Fraga
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Enza M Valente
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- IRCCS Mondino Foundation, Pavia, Italy
| | - Antonio Pisani
- IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | | |
Collapse
|
86
|
Khan A, Johnson R, Wittmer C, Maile M, Tatsukawa K, Wong JL, Gill MB, Stocking EM, Natala SR, Paulino AD, Bowden-Verhoek JK, Wrasidlo W, Masliah E, Bonhaus DW, Price DL. NPT520-34 improves neuropathology and motor deficits in a transgenic mouse model of Parkinson's disease. Brain 2021; 144:3692-3709. [PMID: 34117864 DOI: 10.1093/brain/awab214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/12/2021] [Accepted: 05/11/2021] [Indexed: 12/09/2022] Open
Abstract
NPT520-34 is a clinical-stage, small molecule being developed for the treatment of Parkinson's disease and other neurodegenerative disorders. The therapeutic potential of NPT520-34 was first suggested by findings from cell-based assays of alpha-synuclein (ASYN) clearance. As reported here, NPT520-34 was subsequently evaluated for therapeutically relevant actions in a transgenic animal model of Parkinson's disease that overexpresses human ASYN and in an acute lipopolysaccharide (LPS)-challenge model using wild-type mice. Daily administration of NPT520-34 to mThy1-ASYN (Line 61) transgenic mice for one or three months resulted in reduced ASYN pathology, reduced expression of markers of neuroinflammation, and improvements in multiple indices of motor function. In an LPS-challenge model using wild-type mice, a single-dose of NPT520-34 reduced LPS-evoked increases in the expression of several pro-inflammatory cytokines in plasma. These findings demonstrate the beneficial effects of NPT520-34 on both inflammation and protein-pathology endpoints, with consequent improvements in motor function in an animal model of Parkinson's disease. These findings further suggest that NPT520-34 may have two complementary actions: (1) to increase the clearance of neurotoxic protein aggregates and (2) to directly attenuate inflammation. NPT520-34 treatment may thereby address two of the predominate underlying pathophysiological aspects of neurodegenerative disorders such as Parkinson's disease.
Collapse
Affiliation(s)
- Asma Khan
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Robert Johnson
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Carrie Wittmer
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Michelle Maile
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Keith Tatsukawa
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Julian L Wong
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Martin B Gill
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Emily M Stocking
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Srinivasa R Natala
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Amy D Paulino
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Jon K Bowden-Verhoek
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Wolfgang Wrasidlo
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Eliezer Masliah
- Departments of Neuroscience and Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Douglas W Bonhaus
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Diana L Price
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| |
Collapse
|
87
|
Cacabelos R, Carrera I, Martínez O, Alejo R, Fernández-Novoa L, Cacabelos P, Corzo L, Rodríguez S, Alcaraz M, Nebril L, Tellado I, Cacabelos N, Pego R, Naidoo V, Carril JC. Atremorine in Parkinson's disease: From dopaminergic neuroprotection to pharmacogenomics. Med Res Rev 2021; 41:2841-2886. [PMID: 34106485 DOI: 10.1002/med.21838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 02/11/2021] [Accepted: 05/21/2021] [Indexed: 12/15/2022]
Abstract
Atremorine is a novel bioproduct obtained by nondenaturing biotechnological processes from a genetic species of Vicia faba. Atremorine is a potent dopamine (DA) enhancer with powerful effects on the neuronal dopaminergic system, acting as a neuroprotective agent in Parkinson's disease (PD). Over 97% of PD patients respond to a single dose of Atremorine (5 g, p.o.) 1 h after administration. This response is gender-, time-, dose-, and genotype-dependent, with optimal doses ranging from 5 to 20 g/day, depending upon disease severity and concomitant medication. Drug-free patients show an increase in DA levels from 12.14 ± 0.34 pg/ml to 6463.21 ± 1306.90 pg/ml; and patients chronically treated with anti-PD drugs show an increase in DA levels from 1321.53 ± 389.94 pg/ml to 16,028.54 ± 4783.98 pg/ml, indicating that Atremorine potentiates the dopaminergic effects of conventional anti-PD drugs. Atremorine also influences the levels of other neurotransmitters (adrenaline, noradrenaline) and hormones which are regulated by DA (e.g., prolactin, PRL), with no effect on serotonin or histamine. The variability in Atremorine-induced DA response is highly attributable to pharmacogenetic factors. Polymorphic variants in pathogenic (SNCA, NUCKS1, ITGA8, GPNMB, GCH1, BCKDK, APOE, LRRK2, ACMSD), mechanistic (DRD2), metabolic (CYP2D6, CYP2C9, CYP2C19, CYP3A4/5, NAT2), transporter (ABCB1, SLC6A2, SLC6A3, SLC6A4) and pleiotropic genes (APOE) influence the DA response to Atremorine and its psychomotor and brain effects. Atremorine enhances DNA methylation and displays epigenetic activity via modulation of the pharmacoepigenetic network. Atremorine is a novel neuroprotective agent for dopaminergic neurons with potential prophylactic and therapeutic activity in PD.
Collapse
Affiliation(s)
- Ramón Cacabelos
- Department of Genomic Medicine, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Iván Carrera
- Department of Health Biotechnology, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Olaia Martínez
- Department of Medical Epigenetics, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | | | | | - Pablo Cacabelos
- Department of Digital Diagnosis, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Lola Corzo
- Department of Medical Biochemistry, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Susana Rodríguez
- Department of Medical Biochemistry, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Margarita Alcaraz
- Department of Genomic Medicine, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Laura Nebril
- Department of Genomic Medicine, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Iván Tellado
- Department of Digital Diagnosis, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Natalia Cacabelos
- Department of Medical Documentation, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Rocío Pego
- Department of Neuropsychology, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Vinogran Naidoo
- Department of Neuroscience, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Juan C Carril
- Department of Genomics & Pharmacogenomics, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| |
Collapse
|
88
|
Estrada E. Cascading from SARS-CoV-2 to Parkinson's Disease through Protein-Protein Interactions. Viruses 2021; 13:897. [PMID: 34066091 PMCID: PMC8150712 DOI: 10.3390/v13050897] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 04/21/2021] [Accepted: 05/06/2021] [Indexed: 12/18/2022] Open
Abstract
Extensive extrapulmonary damages in a dozen of organs/systems, including the central nervous system (CNS), are reported in patients of the coronavirus disease 2019 (COVID-19). Three cases of Parkinson's disease (PD) have been reported as a direct consequence of COVID-19. In spite of the scarce data for establishing a definitive link between COVID-19 and PD, some hypotheses have been proposed to explain the cases reported. They, however, do not fit well with the clinical findings reported for COVID-19 patients, in general, and for the PD cases reported, in particular. Given the importance of this potential connection, we present here a molecular-level mechanistic hypothesis that explains well these findings and will serve to explore the potential CNS damage in COVID-19 patients. The model explaining the cascade effects from COVID-19 to CNS is developed by using bioinformatic tools. It includes the post-translational modification of host proteins in the lungs by viral proteins, the transport of modified host proteins via exosomes out the lungs, and the disruption of protein-protein interaction in the CNS by these modified host proteins. Our hypothesis is supported by finding 44 proteins significantly expressed in the CNS which are associated with PD and whose interactions can be perturbed by 24 host proteins significantly expressed in the lungs. These 24 perturbators are found to interact with viral proteins and to form part of the cargoes of exosomes in human tissues. The joint set of perturbators and PD-vulnerable proteins form a tightly connected network with significantly more connections than expected by selecting a random cluster of proteins of similar size from the human proteome. The molecular-level mechanistic hypothesis presented here provides several routes for the cascading of effects from the lungs of COVID-19 patients to PD. In particular, the disruption of autophagy/ubiquitination processes appears as an important mechanism that triggers the generation of large amounts of exosomes containing perturbators in their cargo, which would insult several PD-vulnerable proteins, potentially triggering Parkinsonism in COVID-19 patients.
Collapse
Affiliation(s)
- Ernesto Estrada
- Institute of Mathematics and Applications, University of Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain;
- ARAID Foundation, Government of Aragon, 50018 Zaragoza, Spain
- Institute for Cross-Disciplinary Physics and Complex Systems (IFISC, UIB-CSIC), Campus Universitat de les Illes Balears, E-07122 Palma de Mallorca, Spain
| |
Collapse
|
89
|
Pérez-Santamarina E, García-Ruiz P, Martínez-Rubio D, Ezquerra M, Pla-Navarro I, Puente J, Martí MJ, Palau F, Hoenicka J. Regulatory rare variants of the dopaminergic gene ANKK1 as potential risk factors for Parkinson's disease. Sci Rep 2021; 11:9879. [PMID: 33972609 PMCID: PMC8110570 DOI: 10.1038/s41598-021-89300-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 04/22/2021] [Indexed: 12/22/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by cerebral dopamine depletion that causes motor and cognitive deficits. The dopamine-related gene ANKK1 has been associated with neuropsychiatric disorders with a dopaminergic deficiency in the striatum. This study aims to define the contribution of ANKK1 rare variants in PD. We found in 10 out of 535 PD patients 6 ANKK1 heterozygous rare alleles located at the 5′UTR, the first exon, intron 1, and the nearby enhancer located 2.6 kb upstream. All 6 ANKK1 single nucleotide variants were located in conserved regulatory regions and showed significant allele-dependent effects on gene regulation in vitro. ANKK1 variant carriers did not show other PD-causing Mendelian mutations. Nevertheless, four patients were heterozygous carriers of rare variants of ATP7B gene, which is related to catecholamines. We also found an association between the polymorphic rs7107223 of the ANKK1 enhancer and PD in two independent clinical series (P = 0.007 and 0.021). rs7107223 functional analysis showed significant allele-dependent effects on both gene regulation and dopaminergic response. In conclusion, we have identified in PD patients functional variants at the ANKK1 locus highlighting the possible relevance of rare variants and non-coding regulatory regions in both the genetics of PD and the dopaminergic vulnerability of this disease.
Collapse
Affiliation(s)
- Estela Pérez-Santamarina
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain.,Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain.,University of East Anglia, Norwich, UK
| | - Pedro García-Ruiz
- Unit of Movement Disorders, Department of Neurology, Fundación Jimenez Díaz, Madrid, Spain
| | - Dolores Martínez-Rubio
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain.,Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain.,Unit of Rare Neurodegenerative Diseases, CIPF, Valencia, Spain.,Rare Diseases Joint Units, CIPF-IIS La Fe and INCLIVA, Valencia, Spain
| | - Mario Ezquerra
- Laboratory of Neurodegenerative Disorders, Department of Neurology, Hospital Clínic of Barcelona, IDIBAPS, Barcelona, Spain
| | | | | | - María José Martí
- Movement Disorders Unit, Department of Neurology, Hospital Clínic of Barcelona, IDIBAPS, Barcelona, Spain
| | - Francesc Palau
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain.,Laboratory of Neurogenetics and Molecular Medicine, Neurogenetics and Molecular Medicine Research Group, Institut de Recerca Sant Joan de Déu, C/ Santa Rosa 39-57, Esplugues de Llobrega, 08950, tBarcelona, Spain.,Department of Genetic Medicine, Hospital Sant Joan de Déu, Barcelona, Spain.,ICMID, Hospital Clínic, and Division of Pediatrics, University of Barcelona School of Medicine and Health Sciences, Barcelona, Spain
| | - Janet Hoenicka
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain. .,Laboratory of Neurogenetics and Molecular Medicine, Neurogenetics and Molecular Medicine Research Group, Institut de Recerca Sant Joan de Déu, C/ Santa Rosa 39-57, Esplugues de Llobrega, 08950, tBarcelona, Spain.
| |
Collapse
|
90
|
Na + leak-current channel (NALCN) at the junction of motor and neuropsychiatric symptoms in Parkinson's disease. J Neural Transm (Vienna) 2021; 128:749-762. [PMID: 33961117 DOI: 10.1007/s00702-021-02348-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/30/2021] [Indexed: 12/27/2022]
Abstract
Parkinson's disease (PD) is a debilitating movement disorder often accompanied by neuropsychiatric symptoms that stem from the loss of dopaminergic function in the basal ganglia and altered neurotransmission more generally. Akinesia, postural instability, tremors and frozen gait constitute the major motor disturbances, whereas neuropsychiatric symptoms include altered circadian rhythms, disordered sleep, depression, psychosis and cognitive impairment. Evidence is emerging that the motor and neuropsychiatric symptoms may share etiologic factors. Calcium/ion channels (CACNA1C, NALCN), synaptic proteins (SYNJ1) and neuronal RNA-binding proteins (RBFOX1) are among the risk genes that are common to PD and various psychiatric disorders. The Na+ leak-current channel (NALCN) is the focus of this review because it has been implicated in dystonia, regulation of movement, cognitive impairment, sleep and circadian rhythms. It regulates the resting membrane potential in neurons, mediates pace-making activity, participates in synaptic vesicle recycling and is functionally co-localized to the endoplasmic reticulum (ER)-several of the major processes adversely affected in PD. Here, we summarize the literature on mechanisms and pathways that connect the motor and neuropsychiatric symptoms of PD with a focus on recurring relationships to the NALCN. It is hoped that the various connections outlined here will stimulate further discussion, suggest additional areas for exploration and ultimately inspire novel treatment strategies.
Collapse
|
91
|
Franco R, Rivas-Santisteban R, Navarro G, Pinna A, Reyes-Resina I. Genes Implicated in Familial Parkinson's Disease Provide a Dual Picture of Nigral Dopaminergic Neurodegeneration with Mitochondria Taking Center Stage. Int J Mol Sci 2021; 22:4643. [PMID: 33924963 PMCID: PMC8124903 DOI: 10.3390/ijms22094643] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
The mechanism of nigral dopaminergic neuronal degeneration in Parkinson's disease (PD) is unknown. One of the pathological characteristics of the disease is the deposition of α-synuclein (α-syn) that occurs in the brain from both familial and sporadic PD patients. This paper constitutes a narrative review that takes advantage of information related to genes (SNCA, LRRK2, GBA, UCHL1, VPS35, PRKN, PINK1, ATP13A2, PLA2G6, DNAJC6, SYNJ1, DJ-1/PARK7 and FBXO7) involved in familial cases of Parkinson's disease (PD) to explore their usefulness in deciphering the origin of dopaminergic denervation in many types of PD. Direct or functional interactions between genes or gene products are evaluated using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database. The rationale is to propose a map of the interactions between SNCA, the gene encoding for α-syn that aggregates in PD, and other genes, the mutations of which lead to early-onset PD. The map contrasts with the findings obtained using animal models that are the knockout of one of those genes or that express the mutated human gene. From combining in silico data from STRING-based assays with in vitro and in vivo data in transgenic animals, two likely mechanisms appeared: (i) the processing of native α-syn is altered due to the mutation of genes involved in vesicular trafficking and protein processing, or (ii) α-syn mutants alter the mechanisms necessary for the correct vesicular trafficking and protein processing. Mitochondria are a common denominator since both mechanisms require extra energy production, and the energy for the survival of neurons is obtained mainly from the complete oxidation of glucose. Dopamine itself can result in an additional burden to the mitochondria of dopaminergic neurons because its handling produces free radicals. Drugs acting on G protein-coupled receptors (GPCRs) in the mitochondria of neurons may hopefully end up targeting those receptors to reduce oxidative burden and increase mitochondrial performance. In summary, the analysis of the data of genes related to familial PD provides relevant information on the etiology of sporadic cases and might suggest new therapeutic approaches.
Collapse
Affiliation(s)
- Rafael Franco
- Department Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain; (R.F.); (R.R.-S.); (I.R.-R.)
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain;
| | - Rafael Rivas-Santisteban
- Department Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain; (R.F.); (R.R.-S.); (I.R.-R.)
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain;
| | - Gemma Navarro
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain;
- Department Biochemistry and Physiology, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
| | - Annalisa Pinna
- National Research Council of Italy (CNR), Neuroscience Institute–Cagliari, Cittadella Universitaria, Blocco A, SP 8, Km 0.700, 09042 Monserrato (CA), Italy
| | - Irene Reyes-Resina
- Department Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain; (R.F.); (R.R.-S.); (I.R.-R.)
| |
Collapse
|
92
|
Li C, Ou R, Chen Y, Gu X, Wei Q, Cao B, Zhang L, Hou Y, Liu K, Chen X, Song W, Zhao B, Wu Y, Li T, Dong X, Shang H. Genetic Modifiers of Age at Onset for Parkinson's Disease in Asians: A Genome-Wide Association Study. Mov Disord 2021; 36:2077-2084. [PMID: 33884653 DOI: 10.1002/mds.28621] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/14/2021] [Accepted: 03/25/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Age at onset (AAO) is an essential feature of Parkinson's disease (PD) and can help predict disease progression and mortality. Identification of genetic variants influencing AAO of PD could lead to a better understanding of the disease's biological mechanism and provide clinical guidance. However, genetic determinants for AAO of PD remain mostly unknown, especially in the Asian population. OBJECTIVES To identify genetic determinants for AAO of PD in the Asian population. METHODS We performed a genome-wide association meta-analysis on AAO of PD in 5166 Chinese patients with PD (Ndiscovery = 3628, Nreplication = 1538). We then conducted a further cross-ethnic meta-analysis using our results and summary statistics for the AAO of PD from the European population. RESULTS The total heritability of AAO of PD was around 0.10 ~ 0.14, similar to that (~0.11) estimated in populations of European ancestry. One novel significant intergenic locus rs9783733 (NDN; PWRN4) was identified (P = 3.14E-09, beta = 2.30, SE = 0.39). Remarkably, this variant could delay AAO of PD by ~2.43 years, with a more considerable effect on males (~3.18 years) than females (~1.45 years). The variant was suggestively significant in the cross-ethnic meta-analysis and suggested a positive selection in the East Asian population. Additionally, cross-ethnic meta-analysis identified a significant locus rs356203 in SNCA (P = 2.35E-11, beta = -0.71, SE = 0.01). CONCLUSIONS These findings improve the current understanding of the genetic etiology of AAO of PD in different ethnic groups, and provide a new target for further research on PD pathogenesis and potential therapeutic options. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Chunyu Li
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Ruwei Ou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Yongping Chen
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Xiaojing Gu
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Qianqian Wei
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Bei Cao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Lingyu Zhang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Yanbing Hou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Kuncheng Liu
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Xueping Chen
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Wei Song
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Bi Zhao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Ying Wu
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Tao Li
- Psychiatric Laboratory and Mental Health Center, The State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China.,Huaxi Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| | - Xianjun Dong
- Genomics and Bioinformatics Hub, Harvard Medical School and Brigham & Women's Hospital, Boston, Massachusetts, USA.,Center for Advanced Parkinson Research, Harvard Medical School and Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Huifang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China.,Huaxi Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
93
|
Miller KM, Mercado NM, Sortwell CE. Synucleinopathy-associated pathogenesis in Parkinson's disease and the potential for brain-derived neurotrophic factor. NPJ PARKINSONS DISEASE 2021; 7:35. [PMID: 33846345 PMCID: PMC8041900 DOI: 10.1038/s41531-021-00179-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 03/17/2021] [Indexed: 12/12/2022]
Abstract
The lack of disease-modifying treatments for Parkinson’s disease (PD) is in part due to an incomplete understanding of the disease’s etiology. Alpha-synuclein (α-syn) has become a point of focus in PD due to its connection to both familial and idiopathic cases—specifically its localization to Lewy bodies (LBs), a pathological hallmark of PD. Within this review, we will present a comprehensive overview of the data linking synuclein-associated Lewy pathology with intracellular dysfunction. We first present the alterations in neuronal proteins and transcriptome associated with LBs in postmortem human PD tissue. We next compare these findings to those associated with LB-like inclusions initiated by in vitro exposure to α-syn preformed fibrils (PFFs) and highlight the profound and relatively unique reduction of brain-derived neurotrophic factor (BDNF) in this model. Finally, we discuss the multitude of ways in which BDNF offers the potential to exert disease-modifying effects on the basal ganglia. What remains unknown is the potential for BDNF to mitigate inclusion-associated dysfunction within the context of synucleinopathy. Collectively, this review reiterates the merit of using the PFF model as a tool to understand the physiological changes associated with LBs, while highlighting the neuroprotective potential of harnessing endogenous BDNF.
Collapse
Affiliation(s)
- Kathryn M Miller
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.,Neuroscience Graduate Program, College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Natosha M Mercado
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.,Neuroscience Graduate Program, College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Caryl E Sortwell
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA. .,Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, USA.
| |
Collapse
|
94
|
Lesage S, Mangone G, Tesson C, Bertrand H, Benmahdjoub M, Kesraoui S, Arezki M, Singleton A, Corvol JC, Brice A. Clinical Variability of SYNJ1-Associated Early-Onset Parkinsonism. Front Neurol 2021; 12:648457. [PMID: 33841314 PMCID: PMC8027075 DOI: 10.3389/fneur.2021.648457] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/25/2021] [Indexed: 11/29/2022] Open
Abstract
Autosomal recessive early-onset parkinsonism is clinically and genetically heterogeneous. Mutations of three genes, PRKN, PINK1, and DJ-1 cause pure phenotypes usually characterized by levodopa-responsive Parkinson's disease. By contrast, mutations of other genes, including ATP13A2, PLA2G6, FBXO7, DNAJC6, SYNJ1, VPS13C, and PTRHD1, cause rarer, more severe diseases with a poor response to levodopa, generally with additional atypical features. We performed data mining on a gene panel or whole-exome sequencing in 460 index cases with early-onset (≤ 40 years) Parkinson's disease, including 57 with autosomal recessive disease and 403 isolated cases. We identified two isolated cases carrying biallelic mutations of SYNJ1 (double-heterozygous p.D791fs/p.Y232H and homozygous p. Y832C mutations) and two siblings with the recurrent homozygous p.R258Q mutation. All four variants were absent or rare in the Genome Aggregation Database, were predicted to be deleterious on in silico analysis and were found to be highly conserved between species. The patient with both the previously unknown p.D791fs and p.Y232H mutations presented with dystonia-parkinsonism accompanied by a frontal syndrome and oculomotor disturbances at the age of 39. In addition, two siblings from an Algerian consanguineous family carried the homozygous p.R258Q mutation and presented generalized tonic-clonic seizures during childhood, with severe intellectual disability, followed by progressive parkinsonism during their teens. By contrast, the isolated patient with the homozygous p. Y832C mutation, diagnosed at the age of 20, had typical parkinsonism, with no atypical symptoms and slow disease progression. Our findings expand the mutational spectrum and phenotypic profile of SYNJ1-related parkinsonism.
Collapse
Affiliation(s)
- Suzanne Lesage
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, INSERM, CNRS, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, CIC Neurosciences, Paris, France
| | - Graziella Mangone
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, INSERM, CNRS, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, CIC Neurosciences, Paris, France
| | - Christelle Tesson
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, INSERM, CNRS, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, CIC Neurosciences, Paris, France
| | - Hélène Bertrand
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, INSERM, CNRS, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, CIC Neurosciences, Paris, France
| | | | | | | | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
| | - Jean-Christophe Corvol
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, INSERM, CNRS, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, CIC Neurosciences, Paris, France
| | - Alexis Brice
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, INSERM, CNRS, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, CIC Neurosciences, Paris, France
| |
Collapse
|
95
|
Caló L, Hidari E, Wegrzynowicz M, Dalley JW, Schneider BL, Podgajna M, Anichtchik O, Carlson E, Klenerman D, Spillantini MG. CSPα reduces aggregates and rescues striatal dopamine release in α-synuclein transgenic mice. Brain 2021; 144:1661-1669. [PMID: 33760024 PMCID: PMC8320296 DOI: 10.1093/brain/awab076] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 12/24/2020] [Accepted: 12/29/2020] [Indexed: 02/02/2023] Open
Abstract
α-Synuclein aggregation at the synapse is an early event in Parkinson's disease and is associated with impaired striatal synaptic function and dopaminergic neuronal death. The cysteine string protein (CSPα) and α-synuclein have partially overlapping roles in maintaining synaptic function and mutations in each cause neurodegenerative diseases. CSPα is a member of the DNAJ/HSP40 family of co-chaperones and like α-synuclein, chaperones the SNARE complex assembly and controls neurotransmitter release. α-Synuclein can rescue neurodegeneration in CSPαKO mice. However, whether α-synuclein aggregation alters CSPα expression and function is unknown. Here we show that α-synuclein aggregation at the synapse is associated with a decrease in synaptic CSPα and a reduction in the complexes that CSPα forms with HSC70 and STGa. We further show that viral delivery of CSPα rescues in vitro the impaired vesicle recycling in PC12 cells with α-synuclein aggregates and in vivo reduces synaptic α-synuclein aggregates increasing monomeric α-synuclein and restoring normal dopamine release in 1-120hαSyn mice. These novel findings reveal a mechanism by which α-synuclein aggregation alters CSPα at the synapse, and show that CSPα rescues α-synuclein aggregation-related phenotype in 1-120hαSyn mice similar to the effect of α-synuclein in CSPαKO mice. These results implicate CSPα as a potential therapeutic target for the treatment of early-stage Parkinson's disease.
Collapse
Affiliation(s)
- Laura Caló
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge, UK,Dementia Research Institute, University of Cambridge, Cambridge, UK,Correspondence may also be addressed to: Dr Laura Caló E-mail:
| | - Eric Hidari
- Dementia Research Institute, University of Cambridge, Cambridge, UK,Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Michal Wegrzynowicz
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge, UK,Laboratory of Molecular Basis of Neurodegeneration, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Jeffrey W Dalley
- Department of Psychology, University of Cambridge, Cambridge, UK,Department of Psychiatry, Hershel Smith Building for Brain and Mind Sciences, University of Cambridge, Cambridge, UK
| | - Bernard L Schneider
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland,Bertarelli Platform for Gene Therapy, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1202 Geneva, Switzerland
| | - Martyna Podgajna
- Laboratory of Molecular Basis of Neurodegeneration, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Oleg Anichtchik
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge, UK
| | - Emma Carlson
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge, UK
| | - David Klenerman
- Dementia Research Institute, University of Cambridge, Cambridge, UK,Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Maria Grazia Spillantini
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge, UK,Correspondence to: Prof. Maria Grazia Spillantini Department of Clinical Neurosciences Clifford Allbutt Building, Hills Road, Cambridge CB2 0AH, UK E-mail:
| |
Collapse
|
96
|
Sassone J, Reale C, Dati G, Regoni M, Pellecchia MT, Garavaglia B. The Role of VPS35 in the Pathobiology of Parkinson's Disease. Cell Mol Neurobiol 2021; 41:199-227. [PMID: 32323152 PMCID: PMC11448700 DOI: 10.1007/s10571-020-00849-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/10/2020] [Indexed: 12/21/2022]
Abstract
The vacuolar protein sorting 35 (VPS35) gene located on chromosome 16 has recently emerged as a cause of late-onset familial Parkinson's disease (PD) (PARK17). The gene encodes a 796-residue protein nearly ubiquitously expressed in human tissues. The protein localizes on endosomes where it assembles with other peripheral membrane proteins to form the retromer complex. How VPS35 mutations induce dopaminergic neuron degeneration in humans is still unclear. Because the retromer complex recycles the receptors that mediate the transport of hydrolase to lysosome, it has been suggested that VPS35 mutations lead to impaired lysosomal and autophagy function. Recent studies also demonstrated that VPS35 and the retromer complex influence mitochondrial homeostasis, suggesting that VPS35 mutations elicit mitochondrial dysfunction. More recent studies have identified a key role of VPS35 in neurotransmission, whilst others reported a functional interaction between VPS35 and other genes associated with familial PD, including α-SYNUCLEIN-PARKIN-LRRK2. Here, we review the biological role of VPS35 protein, the VPS35 mutations identified in human PD patients, and the potential molecular mechanism by which VPS35 mutations can induce progressive neurodegeneration in PD.
Collapse
Affiliation(s)
- Jenny Sassone
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| | - Chiara Reale
- Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giovanna Dati
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, SA, Italy
| | - Maria Regoni
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Maria Teresa Pellecchia
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, SA, Italy
| | - Barbara Garavaglia
- Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
97
|
Differences in MTHFR and LRRK2 variant's association with sporadic Parkinson's disease in Mexican Mestizos correlated to Native American ancestry. NPJ Parkinsons Dis 2021; 7:13. [PMID: 33574311 PMCID: PMC7878860 DOI: 10.1038/s41531-021-00157-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 01/08/2021] [Indexed: 01/30/2023] Open
Abstract
Parkinson's disease (PD), a common neurodegenerative disorder, has a complex etiology where environmental and genetic factors intervene. While a number of genes and variants have been identified in recent decades as causative or protective agents of this condition, a limited number of studies have been conducted in mixed populations, such as Mexican Mestizos. The historical convergence of two founding groups and three ethnicities, and the increasing north-to-south gradient of Native American ancestry in Mexico resulted in a subpopulation structure with considerable genetic diversity. In this work, we investigate the influence of 21 known susceptibility variants for PD. Our case-control study, with a cohort of 311 Mexican Mestizo subjects, found a significant risk association for the variant rs1491942 in LRRK2. However, when stratification by ancestry was performed, a risk effect for MTHFR rs1801133 was observed only in the group with the highest percentage of European ancestry, and the PD risk effect for LRRK2 rs1491942 was significant in subjects with a higher ratio of Native American ancestry. Meta-analyses of these SNP revealed the effect of LRRK2 rs1491942 to be even more significant than previously described in populations of European descent. Although corroboration is necessary, our findings suggest that polymorphism rs1491942 may be useful as a risk marker of PD in Mexican Mestizos with greater Native American ancestry. The absence of associations with the remaining known risk factors is, in itself, a relevant finding and invites further research into the shared risk factors' role in the pathophysiological mechanisms of this neurodegenerative disorder.
Collapse
|
98
|
Sarihan EI, Pérez-Palma E, Niestroj LM, Loesch D, Inca-Martinez M, Horimoto AR, Cornejo-Olivas M, Torres L, Mazzetti P, Cosentino C, Sarapura-Castro E, Rivera-Valdivia A, Dieguez E, Raggio V, Lescano A, Tumas V, Borges V, Ferraz HB, Rieder CR, Schumacher-Schuh AF, Santos-Lobato BL, Velez-Pardo C, Jimenez-Del-Rio M, Lopera F, Moreno S, Chana-Cuevas P, Fernandez W, Arboleda G, Arboleda H, Arboleda-Bustos CE, Yearout D, Zabetian CP, Thornton TA, O’Connor TD, Lal D, Mata IF. Genome-Wide Analysis of Copy Number Variation in Latin American Parkinson's Disease Patients. Mov Disord 2021; 36:434-441. [PMID: 33150996 PMCID: PMC8059262 DOI: 10.1002/mds.28353] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/01/2020] [Accepted: 09/10/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Parkinson's disease is the second most common neurodegenerative disorder and affects people from all ethnic backgrounds, yet little is known about the genetics of Parkinson's disease in non-European populations. In addition, the overall identification of copy number variants at a genome-wide level has been understudied in Parkinson's patients. The objective of this study was to understand the genome-wide burden of copy number variants in Latinos and its association with Parkinson's disease. METHODS We used genome-wide genotyping data from 747 Parkinson's disease patients and 632 controls from the Latin American Research Consortium on the Genetics of Parkinson's disease. RESULTS Genome-wide copy number burden analysis showed that patients were significantly enriched for copy number variants overlapping known Parkinson's disease genes compared with controls (odds ratio, 3.97; 95%CI, 1.69-10.5; P = 0.018). PRKN showed the strongest copy number burden, with 20 copy number variant carriers. These patients presented an earlier age of disease onset compared with patients with other copy number variants (median age at onset, 31 vs 57 years, respectively; P = 7.46 × 10-7 ). CONCLUSIONS We found that although overall genome-wide copy number variant burden was not significantly different, Parkinson's disease patients were significantly enriched with copy number variants affecting known Parkinson's disease genes. We also identified that of 250 patients with early-onset disease, 5.6% carried a copy number variant on PRKN in our cohort. Our study is the first to analyze genome-wide copy number variant association in Latino Parkinson's disease patients and provides insights about this complex disease in this understudied population. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Elif Irem Sarihan
- Lerner Research Institute, Genomic Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Eduardo Pérez-Palma
- Lerner Research Institute, Genomic Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Douglas Loesch
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Program in Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Miguel Inca-Martinez
- Lerner Research Institute, Genomic Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Andrea R.V.R. Horimoto
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Mario Cornejo-Olivas
- Neurogenetics Research Center, Instituto Nacional de Ciencias Neurologicas, Lima, Peru
- Center for Global Health, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Luis Torres
- Movement Disorders Unit, Instituto Nacional de Ciencias Neurologicas, Lima, Peru
- School of Medicine, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Pilar Mazzetti
- Neurogenetics Research Center, Instituto Nacional de Ciencias Neurologicas, Lima, Peru
- School of Medicine, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Carlos Cosentino
- Movement Disorders Unit, Instituto Nacional de Ciencias Neurologicas, Lima, Peru
- School of Medicine, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | | | | | - Elena Dieguez
- Neurology Institute, Universidad de la República, Montevideo, Uruguay
| | - Victor Raggio
- Department of Genetics, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Andres Lescano
- Neurology Institute, Universidad de la República, Montevideo, Uruguay
| | - Vitor Tumas
- Ribeirão Preto Medical School, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Vanderci Borges
- Movement Disorders Unit, Department of Neurology and Neurosurgery, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Henrique B. Ferraz
- Movement Disorders Unit, Department of Neurology and Neurosurgery, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Carlos R. Rieder
- Departamento de Neurologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Artur F. Schumacher-Schuh
- Serviço de Neurologia, Hospital de Clínicas de Porto Alegre and Departamento de Farmacologia Universidade Federal do Rio Grande do Su, Porto Alegre, Brazil
| | | | - Carlos Velez-Pardo
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, Universidad de Antioquia (UdeA), Medellín, Colombia
| | - Marlene Jimenez-Del-Rio
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, Universidad de Antioquia (UdeA), Medellín, Colombia
| | - Francisco Lopera
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, Universidad de Antioquia (UdeA), Medellín, Colombia
| | - Sonia Moreno
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, Universidad de Antioquia (UdeA), Medellín, Colombia
| | - Pedro Chana-Cuevas
- CETRAM, Facultad de Ciencias Medicas, Universidad de Santiago de Chile, Santiago de Chile, Chile
| | - William Fernandez
- Neuroscience and Cell Death Research Groups, Medical School and Genetic Institute, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Gonzalo Arboleda
- Neuroscience and Cell Death Research Groups, Medical School and Genetic Institute, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Humberto Arboleda
- Neuroscience and Cell Death Research Groups, Medical School and Genetic Institute, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Carlos E. Arboleda-Bustos
- Neuroscience and Cell Death Research Groups, Medical School and Genetic Institute, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Dora Yearout
- Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA
- Department of Neurology, University of Washington, Seattle, Washington, USA
| | - Cyrus P. Zabetian
- Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA
- Department of Neurology, University of Washington, Seattle, Washington, USA
| | - Timothy A. Thornton
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Timothy D. O’Connor
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Program in Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Dennis Lal
- Lerner Research Institute, Genomic Medicine, Cleveland Clinic, Cleveland, Ohio, USA
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, Massachusetts, USA
- Epilepsy Center & Department of Neurology, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ignacio F. Mata
- Lerner Research Institute, Genomic Medicine, Cleveland Clinic, Cleveland, Ohio, USA
- Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA
- Department of Neurology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
99
|
Interaction between Parkin and α-Synuclein in PARK2-Mediated Parkinson's Disease. Cells 2021; 10:cells10020283. [PMID: 33572534 PMCID: PMC7911026 DOI: 10.3390/cells10020283] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Parkin and α-synuclein are two key proteins involved in the pathophysiology of Parkinson's disease (PD). Neurotoxic alterations of α-synuclein that lead to the formation of toxic oligomers and fibrils contribute to PD through synaptic dysfunction, mitochondrial impairment, defective endoplasmic reticulum and Golgi function, and nuclear dysfunction. In half of the cases, the recessively inherited early-onset PD is caused by loss of function mutations in the PARK2 gene that encodes the E3-ubiquitin ligase, parkin. Parkin is involved in the clearance of misfolded and aggregated proteins by the ubiquitin-proteasome system and regulates mitophagy and mitochondrial biogenesis. PARK2-related PD is generally thought not to be associated with Lewy body formation although it is a neuropathological hallmark of PD. In this review article, we provide an overview of post-mortem neuropathological examinations of PARK2 patients and present the current knowledge of a functional interaction between parkin and α-synuclein in the regulation of protein aggregates including Lewy bodies. Furthermore, we describe prevailing hypotheses about the formation of intracellular micro-aggregates (synuclein inclusions) that might be more likely than Lewy bodies to occur in PARK2-related PD. This information may inform future studies aiming to unveil primary signaling processes involved in PD and related neurodegenerative disorders.
Collapse
|
100
|
Pu JL, Gao T, Si XL, Zheng R, Jin CY, Ruan Y, Fang Y, Chen Y, Song Z, Yin XZ, Yan YP, Tian J, Zhang BR. Parkinson's Disease in Teneurin Transmembrane Protein 4 ( TENM4) Mutation Carriers. Front Genet 2021; 11:598064. [PMID: 33414808 PMCID: PMC7783409 DOI: 10.3389/fgene.2020.598064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/02/2020] [Indexed: 11/13/2022] Open
Abstract
Introduction Mutations in the teneurin transmembrane protein 4 (TENM4) gene, known to be involved in neuropsychiatric disorders, have been identified in three pedigree of essential tremor (ET) from Spain. ET has overlapping clinical manifestations and epidemiological symptoms with Parkinson’s disease (PD), suggesting these two disorders may reflect common genetic risk factors. In this study, we investigated clinical and genetic manifestations in four unrelated pedigrees with both ET and PD in which TENM4 variants were identified. Methods We subsequently explored whether TENM4 variants contributed to the risk of developing PD. The frequency of TENM4 variants was evaluated from four PD pedigrees and other 407 subjects. Results The results revealed 12 different novel heterozygous variants, all at low frequency. A clear general enrichment of TENM4 variants was detected in early onset PD patients (p < 0.001, OR = 5.264, 95% CI = 1.957–14.158). Conclusion The results indicate that rare TENM4 variants may be associated with an increased risk of PD.
Collapse
Affiliation(s)
- Jia-Li Pu
- Department of Neurology, College of Medicine, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Ting Gao
- Department of Neurology, College of Medicine, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Xiao-Li Si
- Department of Neurology, College of Medicine, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Ran Zheng
- Department of Neurology, College of Medicine, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Chong-Yao Jin
- Department of Neurology, College of Medicine, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Yang Ruan
- Department of Neurology, College of Medicine, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Yi Fang
- Department of Neurology, College of Medicine, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Ying Chen
- Department of Neurology, College of Medicine, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Zhe Song
- Department of Neurology, College of Medicine, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Xin-Zhen Yin
- Department of Neurology, College of Medicine, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Ya-Ping Yan
- Department of Neurology, College of Medicine, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Jun Tian
- Department of Neurology, College of Medicine, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Bao-Rong Zhang
- Department of Neurology, College of Medicine, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| |
Collapse
|