1
|
Khani M, Cerquera-Cleves C, Kekenadze M, Crea PAW, Singleton AB, Bandres-Ciga S. Towards a Global View of Parkinson's Disease Genetics. Ann Neurol 2024; 95:831-842. [PMID: 38557965 PMCID: PMC11060911 DOI: 10.1002/ana.26905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 04/04/2024]
Abstract
Parkinson's disease (PD) is a global health challenge, yet historically studies of PD have taken place predominantly in European populations. Recent genetics research conducted in non-European populations has revealed novel population-specific genetic loci linked to PD risk, highlighting the importance of studying PD globally. These insights have broadened our understanding of PD etiology, which is crucial for developing disease-modifying interventions. This review comprehensively explores the global genetic landscape of PD, emphasizing the scientific rationale for studying underrepresented populations. It underscores challenges, such as genotype-phenotype heterogeneity and inclusion difficulties for non-European participants, emphasizing the ongoing need for diverse and inclusive research in PD. ANN NEUROL 2024;95:831-842.
Collapse
Affiliation(s)
- Marzieh Khani
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Catalina Cerquera-Cleves
- Pontificia Universidad Javeriana, San Ignacio Hospital, Neurology Unit, Bogotá, Colombia
- CHU de Québec Research Center, Axe Neurosciences, Laval University. Quebec City, Canada
| | - Mariam Kekenadze
- Tbilisi State Medical University, Tbilisi, 0141, Georgia
- University College London, Queen Square Institute of Neurology , WC1N 3BG, London, UK
| | - Peter A. Wild Crea
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Andrew B. Singleton
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Sara Bandres-Ciga
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Doskas T, Vadikolias K, Ntoskas K, Vavougios GD, Tsiptsios D, Stamati P, Liampas I, Siokas V, Messinis L, Nasios G, Dardiotis E. Neurocognitive Impairment and Social Cognition in Parkinson's Disease Patients. Neurol Int 2024; 16:432-449. [PMID: 38668129 PMCID: PMC11054167 DOI: 10.3390/neurolint16020032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/06/2024] [Accepted: 04/11/2024] [Indexed: 04/29/2024] Open
Abstract
In addition to motor symptoms, neurocognitive impairment (NCI) affects patients with prodromal Parkinson's disease (PD). NCI in PD ranges from subjective cognitive complaints to dementia. The purpose of this review is to present the available evidence of NCI in PD and highlight the heterogeneity of NCI phenotypes as well as the range of factors that contribute to NCI onset and progression. A review of publications related to NCI in PD up to March 2023 was performed using PubMed/Medline. There is an interconnection between the neurocognitive and motor symptoms of the disease, suggesting a common underlying pathophysiology as well as an interconnection between NCI and non-motor symptoms, such as mood disorders, which may contribute to confounding NCI. Motor and non-motor symptom evaluation could be used prognostically for NCI onset and progression in combination with imaging, laboratory, and genetic data. Additionally, the implications of NCI on the social cognition of afflicted patients warrant its prompt management. The etiology of NCI onset and its progression in PD is multifactorial and its effects are equally grave as the motor effects. This review highlights the importance of the prompt identification of subjective cognitive complaints in PD patients and NCI management.
Collapse
Affiliation(s)
- Triantafyllos Doskas
- Department of Neurology, Athens Naval Hospital, 11521 Athens, Greece;
- Department of Neurology, General University Hospital of Alexandroupoli, 68100 Alexandroupoli, Greece; (K.V.); (D.T.)
| | - Konstantinos Vadikolias
- Department of Neurology, General University Hospital of Alexandroupoli, 68100 Alexandroupoli, Greece; (K.V.); (D.T.)
| | | | - George D. Vavougios
- Department of Neurology, Athens Naval Hospital, 11521 Athens, Greece;
- Department of Neurology, Faculty of Medicine, University of Cyprus, 1678 Lefkosia, Cyprus
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece
| | - Dimitrios Tsiptsios
- Department of Neurology, General University Hospital of Alexandroupoli, 68100 Alexandroupoli, Greece; (K.V.); (D.T.)
| | - Polyxeni Stamati
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece; (P.S.); (I.L.); (V.S.); (E.D.)
| | - Ioannis Liampas
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece; (P.S.); (I.L.); (V.S.); (E.D.)
| | - Vasileios Siokas
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece; (P.S.); (I.L.); (V.S.); (E.D.)
| | - Lambros Messinis
- School of Psychology, Laboratory of Neuropsychology and Behavioural Neuroscience, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Grigorios Nasios
- Department of Speech and Language Therapy, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece;
| | - Efthimios Dardiotis
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece; (P.S.); (I.L.); (V.S.); (E.D.)
| |
Collapse
|
3
|
Sun X, Dou K, Xue L, Xie Y, Yang Y, Xie A. Comprehensive analysis of clinical and biological features in Parkinson's disease associated with the LRRK2 G2019S mutation: Data from the PPMI study. Clin Transl Sci 2024; 17:e13720. [PMID: 38266062 PMCID: PMC10804919 DOI: 10.1111/cts.13720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/03/2024] [Accepted: 01/03/2024] [Indexed: 01/26/2024] Open
Abstract
The Parkinson's Progression Marker Initiative (PPMI) aims to identify biomarkers for Parkinson's disease (PD) risk, onset, and progression. This study focuses on the G2019S missense mutation in the LRRK2 gene, which is associated with hereditary and sporadic PD. Utilizing data from the PPMI database, we conducted an analysis of baseline clinical characteristics, as well as serum and cerebrospinal fluid levels in two groups: patients with PD with the G2019S mutation (PD + G2019S) and patients with PD without the mutation (PD-G2019S). Multiple linear regression and longitudinal analysis were performed, controlling for confounding factors. Compared to the PD-G2019S group, the PD + G2019S group showed more obvious initial motor dysfunction-higher baseline Movement Disorder Society-Sponsored Revision of the Unified Parkinson Disease Rating Scale (MDS-UPDRS) scores (false discovery rate [FDR]-adjusted p < 0.001), but progressed more slowly. Mechanism of Coordinated Access and activities of daily living (ADL) scores were lower at baseline (FDR-adjusted p < 0.001), whereas Scales for Outcomes of Parkinson's Disease (SCOPA)-Thermoregulatory (FDR-adjusted p = 0.015) scores were higher, emphasizing the increase of non-motor symptoms associated with LRRK2-G2019S mutation. During the follow-up period, the motor and non-motor symptoms changed dynamically with time, and there were longitudinal differences in the scores of MDS-UPDRS (FDR-adjusted PI = 0.013, PII = 0.008, PIV < 0.001), Questionnaire for Impulsive-Compulsive Disorders in Parkinson's Disease (FDR-adjusted p = 0.027), SCOPA-Thermoregulatory (FDR-adjusted p = 0.021), and ADL (FDR-adjusted p = 0.027) scale scores. PD associated with the LRRK2 G2019S mutation demonstrated more severe symptoms at baseline but slower progression. Motor complications and thermoregulatory disorders were more pronounced.
Collapse
Affiliation(s)
- Xiaohui Sun
- Department of NeurologyAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Kaixin Dou
- Department of NeurologyAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Li Xue
- Recording RoomThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Yijie Xie
- Clinical Laboratory, Central LaboratoryQingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital)QingdaoChina
| | - Yong Yang
- Department of NeurologyAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Anmu Xie
- Department of NeurologyAffiliated Hospital of Qingdao UniversityQingdaoChina
- Cerebral Vascular Disease Institute, Affiliated Hospital of Qingdao UniversityQingdaoChina
| |
Collapse
|
4
|
Shkury E, Danziger-Schragenheim S, Katzir Z, Ezra Y, Giladi N, Mirelman A, Maidan I. Differences in EEG Event-Related Potentials during Dual Task in Parkinson's Disease Carriers and Non-Carriers of the G2019S-LRRK2 Mutation. SENSORS (BASEL, SWITZERLAND) 2023; 23:8266. [PMID: 37837096 PMCID: PMC10575245 DOI: 10.3390/s23198266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/28/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND The G2019S-LRRK2 gene mutation is a common cause of hereditary Parkinson's disease (PD), associated with a higher frequency of the postural instability gait difficulty (PIGD) motor phenotype yet with preserved cognition. This study investigated neurophysiological changes during motor and cognitive tasks in PD patients with and without the G2019S-LRRK2 mutation. METHODS 33 iPD patients and 22 LRRK2-PD patients performed the visual Go/NoGo task (VGNG) during sitting (single-task) and walking (dual-task) while wearing a 64-channel EEG cap. Event-related potentials (ERP) from Fz and Pz, specifically N200 and P300, were extracted and analyzed to quantify brain activity patterns. RESULTS The LRRK2-PD group performed better in the VGNG than the iPD group (group*task; p = 0.05). During Go, the iPD group showed reduced N2 amplitude and prolonged N2 latency during walking, whereas the LRRK2-PD group showed only shorter latency (group*task p = 0.027). During NoGo, opposite patterns emerged; the iPD group showed reduced N2 and increased P3 amplitudes during walking while the LRRK2-PD group demonstrated increased N2 and reduced P3 (N2: group*task, p = 0.010, P3: group*task, p = 0.012). CONCLUSIONS The LRRK2-PD group showed efficient early cognitive processes, reflected by N2, resulting in greater neural synchronization and prominent ERPs. These processes are possibly the underlying mechanisms for the observed better cognitive performance as compared to the iPD group. As such, future applications of intelligent medical sensing should be capable of capturing these electrophysiological patterns in order to enhance motor-cognitive functions.
Collapse
Affiliation(s)
- Eden Shkury
- Laboratory of Early Markers of Neurodegeneration, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (E.S.); (S.D.-S.); (Z.K.); (Y.E.); (N.G.); (A.M.)
- School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shani Danziger-Schragenheim
- Laboratory of Early Markers of Neurodegeneration, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (E.S.); (S.D.-S.); (Z.K.); (Y.E.); (N.G.); (A.M.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Zoya Katzir
- Laboratory of Early Markers of Neurodegeneration, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (E.S.); (S.D.-S.); (Z.K.); (Y.E.); (N.G.); (A.M.)
- School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Yael Ezra
- Laboratory of Early Markers of Neurodegeneration, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (E.S.); (S.D.-S.); (Z.K.); (Y.E.); (N.G.); (A.M.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Nir Giladi
- Laboratory of Early Markers of Neurodegeneration, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (E.S.); (S.D.-S.); (Z.K.); (Y.E.); (N.G.); (A.M.)
- School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Anat Mirelman
- Laboratory of Early Markers of Neurodegeneration, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (E.S.); (S.D.-S.); (Z.K.); (Y.E.); (N.G.); (A.M.)
- School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Inbal Maidan
- Laboratory of Early Markers of Neurodegeneration, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (E.S.); (S.D.-S.); (Z.K.); (Y.E.); (N.G.); (A.M.)
- School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
5
|
Pajarillo E, Kim S, Digman A, Dutton M, Son DS, Aschner M, Lee E. The role of microglial LRRK2 kinase in manganese-induced inflammatory neurotoxicity via NLRP3 inflammasome and RAB10-mediated autophagy dysfunction. J Biol Chem 2023; 299:104879. [PMID: 37269951 PMCID: PMC10331485 DOI: 10.1016/j.jbc.2023.104879] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/12/2023] [Accepted: 05/20/2023] [Indexed: 06/05/2023] Open
Abstract
Chronic manganese (Mn) exposure can lead to manganism, a neurological disorder sharing common symptoms with Parkinson's disease (PD). Studies have shown that Mn can increase the expression and activity of leucine-rich repeat kinase 2 (LRRK2), leading to inflammation and toxicity in microglia. LRRK2 G2019S mutation also elevates LRRK2 kinase activity. Thus, we tested if Mn-increased microglial LRRK2 kinase is responsible for Mn-induced toxicity, and exacerbated by G2019S mutation, using WT and LRRK2 G2019S knock-in mice and BV2 microglia. Mn (30 mg/kg, nostril instillation, daily for 3 weeks) caused motor deficits, cognitive impairments, and dopaminergic dysfunction in WT mice, which were exacerbated in G2019S mice. Mn induced proapoptotic Bax, NLRP3 inflammasome, IL-1β, and TNF-α in the striatum and midbrain of WT mice, and these effects were more pronounced in G2019S mice. BV2 microglia were transfected with human LRRK2 WT or G2019S, followed by Mn (250 μM) exposure to better characterize its mechanistic action. Mn increased TNF-α, IL-1β, and NLRP3 inflammasome activation in BV2 cells expressing WT LRRK2, which was elevated further in G2019S-expressing cells, while pharmacological inhibition of LRRK2 mitigated these effects in both genotypes. Moreover, the media from Mn-treated G2019S-expressing BV2 microglia caused greater toxicity to the cath.a-differentiated (CAD) neuronal cells compared to media from microglia expressing WT. Mn-LRRK2 activated RAB10 which was exacerbated in G2019S. RAB10 played a critical role in LRRK2-mediated Mn toxicity by dysregulating the autophagy-lysosome pathway and NLRP3 inflammasome in microglia. Our novel findings suggest that microglial LRRK2 via RAB10 plays a critical role in Mn-induced neuroinflammation.
Collapse
Affiliation(s)
- Edward Pajarillo
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Sanghoon Kim
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Alexis Digman
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Matthew Dutton
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Deok-Soo Son
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, Tennessee, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Eunsook Lee
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA.
| |
Collapse
|
6
|
Ito G, Utsunomiya-Tate N. Overview of the Impact of Pathogenic LRRK2 Mutations in Parkinson's Disease. Biomolecules 2023; 13:biom13050845. [PMID: 37238714 DOI: 10.3390/biom13050845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/25/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a large protein kinase that physiologically phosphorylates and regulates the function of several Rab proteins. LRRK2 is genetically implicated in the pathogenesis of both familial and sporadic Parkinson's disease (PD), although the underlying mechanism is not well understood. Several pathogenic mutations in the LRRK2 gene have been identified, and in most cases the clinical symptoms that PD patients with LRRK2 mutations develop are indistinguishable from those of typical PD. However, it has been shown that the pathological manifestations in the brains of PD patients with LRRK2 mutations are remarkably variable when compared to sporadic PD, ranging from typical PD pathology with Lewy bodies to nigral degeneration with deposition of other amyloidogenic proteins. The pathogenic mutations in LRRK2 are also known to affect the functions and structure of LRRK2, the differences in which may be partly attributable to the variations observed in patient pathology. In this review, in order to help researchers unfamiliar with the field to understand the mechanism of pathogenesis of LRRK2-associated PD, we summarize the clinical and pathological manifestations caused by pathogenic mutations in LRRK2, their impact on the molecular function and structure of LRRK2, and their historical background.
Collapse
Affiliation(s)
- Genta Ito
- Department of Biomolecular Chemistry, Faculty of Pharma-Sciences, Teikyo University, Tokyo 173-8605, Japan
| | - Naoko Utsunomiya-Tate
- Department of Biomolecular Chemistry, Faculty of Pharma-Sciences, Teikyo University, Tokyo 173-8605, Japan
| |
Collapse
|
7
|
Pajarillo E, Kim SH, Digman A, Dutton M, Son DS, Aschner M, Lee E. The role of microglial LRRK2 in manganese-induced inflammatory neurotoxicity via NLRP3 inflammasome and RAB10-mediated autophagy dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.535418. [PMID: 37066140 PMCID: PMC10103982 DOI: 10.1101/2023.04.03.535418] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Chronic exposure to manganese (Mn) can lead to manganism, a neurological disorder sharing common symptoms with Parkinson's disease (PD). Studies have shown that Mn can increase the expression and activity of leucine-rich repeat kinase 2 (LRRK2), leading to inflammation and toxicity in microglia. LRRK2 G2019S mutation also elevates LRRK2 kinase activity. Thus, we tested if Mn-increased microglial LRRK2 kinase is responsible for Mn-induced toxicity, and exacerbated by G2019S mutation, using WT and LRRK2 G2019S knock-in mice, and BV2 microglia. Mn (30 mg/kg, nostril instillation, daily for 3 weeks) caused motor deficits, cognitive impairments, and dopaminergic dysfunction in WT mice, which were exacerbated in G2019S mice. Mn induced proapoptotic Bax, NLRP3 inflammasome, IL-1β and TNF-α in the striatum and midbrain of WT mice, and these effects were exacerbated in G2019S mice. BV2 microglia were transfected with human LRRK2 WT or G2019S, followed by Mn (250 μM) exposure to better characterize its mechanistic action. Mn increased TNF-α, IL-1β, and NLRP3 inflammasome activation in BV2 cells expressing WT LRRK2, which was exacerbated in G2019S-expressing cells, while pharmacological inhibition of LRRK2 mitigated these effects in both genotypes. Moreover, the media from Mn-treated BV2 microglia expressing G2019S caused greater toxicity to cath.a-differentiated (CAD) neuronal cells compared to media from microglia expressing WT. Mn-LRRK2 activated RAB10, which was exacerbated in G2019S. RAB10 played a critical role in LRRK2-mediated Mn toxicity by dysregulating the autophagy-lysosome pathway, and NLRP3 inflammasome in microglia. Our novel findings suggest that microglial LRRK2 via RAB10 plays a critical role in Mn-induced neuroinflammation.
Collapse
|
8
|
Mata I, Salles P, Cornejo-Olivas M, Saffie P, Ross OA, Reed X, Bandres-Ciga S. LRRK2: Genetic mechanisms vs genetic subtypes. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:133-154. [PMID: 36803807 DOI: 10.1016/b978-0-323-85555-6.00018-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
In 2004, the identification of pathogenic variants in the LRRK2 gene across several families with autosomal dominant late-onset Parkinson's disease (PD) revolutionized our understanding of the role of genetics in PD. Previous beliefs that genetics in PD was limited to rare early-onset or familial forms of the disease were quickly dispelled. Currently, we recognize LRRK2 p.G2019S as the most common genetic cause of both sporadic and familial PD, with more than 100,000 affected carriers across the globe. The frequency of LRRK2 p.G2019S is also highly variable across populations, with some regions of Asian or Latin America reporting close to 0%, contrasting to Ashkenazi Jews or North African Berbers reporting up to 13% and 40%, respectively. Patients with LRRK2 pathogenic variants are clinically and pathologically heterogeneous, highlighting the age-related variable penetrance that also characterizes LRRK2-related disease. Indeed, the majority of patients with LRRK2-related disease are characterized by a relatively mild Parkinsonism with less motor symptoms with variable presence of α-synuclein and/or tau aggregates, with pathologic pleomorphism widely described. At a functional cellular level, it is likely that pathogenic variants mediate a toxic gain-of-function of the LRRK2 protein resulting in increased kinase activity perhaps in a cell-specific manner; by contrast, some LRRK2 variants appear to be protective reducing PD risk by decreasing the kinase activity. Therefore, employing this information to define appropriate patient populations for clinical trials of targeted kinase LRRK2 inhibition strategies is very promising and demonstrates a potential future application for PD using precision medicine.
Collapse
Affiliation(s)
- Ignacio Mata
- Genomic Medicine Institute (GMI), Cleveland Clinic, Cleveland, OH, United States.
| | - Philippe Salles
- Corporación Centro de Trastornos del Movimiento (CETRAM), Lo Espejo, Santiago, Chile
| | - Mario Cornejo-Olivas
- Neurogenetics Research Center, Instituto Nacional de Ciencias Neurológicas, Lima, Peru
| | - Paula Saffie
- Corporación Centro de Trastornos del Movimiento (CETRAM), Lo Espejo, Santiago, Chile
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Xylena Reed
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
| | - Sara Bandres-Ciga
- Laboratory of Neurogenetics and Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
9
|
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: 2.5] [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
|
10
|
Cognitive Impairment in Genetic Parkinson's Disease. PARKINSON'S DISEASE 2022; 2021:8610285. [PMID: 35003622 PMCID: PMC8739522 DOI: 10.1155/2021/8610285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 12/08/2021] [Indexed: 11/24/2022]
Abstract
Cognitive impairment is common in idiopathic Parkinson's disease (PD). Knowledge of the contribution of genetics to cognition in PD is increasing in the last decades. Monogenic forms of genetic PD show distinct cognitive profiles and rate of cognitive decline progression. Cognitive impairment is higher in GBA- and SNCA-associated PD, lower in Parkin- and PINK1-PD, and possibly milder in LRRK2-PD. In this review, we summarize data regarding cognitive function on clinical studies, neuroimaging, and biological markers of cognitive decline in autosomal dominant PD linked to mutations in LRRK2 and SNCA, autosomal recessive PD linked to Parkin and PINK1, and also PD linked to GBA mutations.
Collapse
|
11
|
Gorzkowska A, Cholewa J, Cholewa J, Wilk A, Klimkowicz-Mrowiec A. Risk Factors for Apathy in Polish Patients with Parkinson's Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph181910196. [PMID: 34639517 PMCID: PMC8507785 DOI: 10.3390/ijerph181910196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/15/2021] [Accepted: 09/22/2021] [Indexed: 12/05/2022]
Abstract
Apathy, a feeling of indifference or a general lack of interest and motivation to engage in activity, is one of the most common neuropsychiatric symptoms in Parkinson’s disease (PD). The large variation in prevalence and the underlying pathophysiological processes remain unclear due to heterogeneous PD populations. The purpose of this study was to identify risk factors for apathy, the modification or treatment of which may be clinically relevant and improve quality of life and caregiver burden for patients with Parkinson’s disease. Caucasian subjects with Parkinson’s disease were included in the study. Baseline demographics, neurological deficit, medications taken, cognitive and neuropsychiatric status, and the polymorphisms in the brain-derived neurotrophic factor gene were assessed. Apathy was diagnosed in 53 (50.5%) patients. They were less educated (OR 0.76 CI 0.64–0.89; p = 0.001), more frequently depressed (OR 1.08 CI 1.01–1.15; p = 0.018), and less frequently treated with inhibitors of monoamine oxidase-B (MAOB-I) (OR 0.07 CI 0.01–0.69; p = 0.023). Although apathetic patients were more likely to carry the Met/Met genotype, differences in the brain-derived neurotrophic factor BDNF rs6265 polymorphism between apathetic and non-apathetic PD patients were not statistically significant in multivariate analysis. Some risk factors for apathy may be clinically modifiable. Further studies are needed to assess whether modeling modifiable apathy risk factors will affect the prevalence of this neuropsychiatric symptom in patients with Parkinson’s disease.
Collapse
Affiliation(s)
- Agnieszka Gorzkowska
- Department of Neurorehabilitation, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Joanna Cholewa
- Department of Physical Education and Adapted Physical Activity, The Jerzy Kukuczka Academy of Physical Education in Katowice, 40-065 Katowice, Poland;
| | - Jaroslaw Cholewa
- Department of Health Related Physical Activity and Tourism, The Jerzy Kukuczka Academy of Physical Education in Katowice, 40-065 Katowice, Poland
- Correspondence: ; Tel.: +48-601-560-011
| | - Aleksander Wilk
- Department of Neurosurgery, University Hospital, 31-501 Krakow, Poland;
| | - Aleksandra Klimkowicz-Mrowiec
- Department of Internal Medicine and Gerontology, Faculty of Medicine, Medical College, Jagiellonian University, 31-008 Krakow, Poland;
| |
Collapse
|
12
|
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: 7.7] [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
|
13
|
Yakhine-Diop SMS, Rodríguez-Arribas M, Canales-Cortés S, Martínez-Chacón G, Uribe-Carretero E, Blanco-Benítez M, Duque-González G, Paredes-Barquero M, Alegre-Cortés E, Climent V, Aiastui A, López de Munain A, Bravo-San Pedro JM, Niso-Santano M, Fuentes JM, González-Polo RA. The parkinsonian LRRK2 R1441G mutation shows macroautophagy-mitophagy dysregulation concomitant with endoplasmic reticulum stress. Cell Biol Toxicol 2021; 38:889-911. [PMID: 34060004 DOI: 10.1007/s10565-021-09617-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 05/12/2021] [Indexed: 12/15/2022]
Abstract
Autophagy is a mechanism responsible for the degradation of cellular components to maintain their homeostasis. However, autophagy is commonly altered and compromised in several diseases, including neurodegenerative disorders. Parkinson's disease (PD) can be considered a multifactorial disease because environmental factors, genetic factors, and aging are involved. Several genes are involved in PD pathology, among which the LRRK2 gene and its mutations, inherited in an autosomal dominant manner, are responsible for most genetic PD cases. The R1441G LRRK2 mutation is, after G2019S, the most important in PD pathogenesis. Our results demonstrate a relationship between the R1441G LRRK2 mutation and a mechanistic dysregulation of autophagy that compromises cell viability. This altered autophagy mechanism is associated with organellar stress including mitochondrial (which induces mitophagy) and endoplasmic reticulum (ER) stress, consistent with the fact that patients with this mutation are more vulnerable to toxins related to PD, such as MPP+.
Collapse
Affiliation(s)
- Sokhna M S Yakhine-Diop
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain.,Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain.,Instituto de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - Mario Rodríguez-Arribas
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain.,Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain.,Instituto de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - Saray Canales-Cortés
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain
| | - Guadalupe Martínez-Chacón
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain.,Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain.,Instituto de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - Elisabet Uribe-Carretero
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain.,Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain.,Instituto de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - Mercedes Blanco-Benítez
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain
| | - Gema Duque-González
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain
| | - Marta Paredes-Barquero
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain
| | - Eva Alegre-Cortés
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain
| | - Vicente Climent
- Departamento de Anatomía Y Embriología Humana, Facultad de Medicina, Universidad de Extremadura, Badajoz, Spain
| | - Ana Aiastui
- Cell Culture Platform, Donostia University Hospital, San Sebastián, Spain.,Neuroscience Area of Biodonostia Health Research Institute, Donostia University Hospital, San Sebastián, Spain
| | - Adolfo López de Munain
- Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain.,Department of Neurology, Donostia University Hospital, San Sebastian, Spain.,Ilundain Foundation, San Sebastian, Spain.,Department of Neurosciences, University of the Basque Country UPV-EHU, San Sebastián, Spain
| | - José M Bravo-San Pedro
- Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain.,Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Mireia Niso-Santano
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain. .,Instituto de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain.
| | - José M Fuentes
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain. .,Instituto de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain.
| | - Rosa A González-Polo
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain. .,Instituto de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain.
| |
Collapse
|
14
|
Vinagre-Aragón A, Campo-Caballero D, Mondragón-Rezola E, Pardina-Vilella L, Hernandez Eguiazu H, Gorostidi A, Croitoru I, Bergareche A, Ruiz-Martinez J. A More Homogeneous Phenotype in Parkinson's Disease Related to R1441G Mutation in the LRRK2 Gene. Front Neurol 2021; 12:635396. [PMID: 33763016 PMCID: PMC7982912 DOI: 10.3389/fneur.2021.635396] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/13/2021] [Indexed: 11/27/2022] Open
Abstract
Parkinson's disease (PD) is characterized by a great clinical heterogeneity. Nevertheless, the biological drivers of this heterogeneity have not been completely elucidated and are likely to be complex, arising from interactions between genetic, epigenetic, and environmental factors. Despite this heterogeneity, the clinical patterns of monogenic forms of PD have usually maintained a good clinical correlation with each mutation once a sufficient number of patients have been studied. Mutations in LRRK2 are the most commonly known genetic cause of autosomal dominant PD known to date. Furthermore, recent genome-wide association studies have revealed variations in LRRK2 as significant risk factors also for the development of sporadic PD. The LRRK2-R1441G mutation is especially frequent in the population of Basque ascent based on a possible founder effect, being responsible for almost 50% of cases of familial PD in our region, with a high penetrance. Curiously, Lewy bodies, considered the neuropathological hallmark of PD, are absent in a significant subset of LRRK2-PD cases. Indeed, these cases appear to be associated with a less aggressive primarily pure motor phenotype. The aim of our research is to examine the clinical phenotype of R1441G-PD patients, more homogeneous when we compare it with sporadic PD patients or with patients carrying other LRRK2 mutations, and reflect on the value of the observed correlation in the genetic forms of PD. The clinical heterogeneity of PD leads us to think that there may be as many different diseases as the number of people affected. Undoubtedly, genetics constitutes a relevant key player, as it may significantly influence the phenotype, with differences according to the mutation within the same gene, and not only in familial PD but also in sporadic forms. Thus, extending our knowledge regarding genetic forms of PD implies an expansion of knowledge regarding sporadic forms, and this may be relevant due to the future therapeutic implications of all forms of PD.
Collapse
Affiliation(s)
- Ana Vinagre-Aragón
- Department of Neurology, Hospital Universitario Donostia, San Sebastián, Spain.,Neuroscience Area, Biodonostia Health Research Institute, San Sebastián, Spain
| | - David Campo-Caballero
- Department of Neurology, Hospital Universitario Donostia, San Sebastián, Spain.,Neuroscience Area, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Elisabet Mondragón-Rezola
- Department of Neurology, Hospital Universitario Donostia, San Sebastián, Spain.,Neuroscience Area, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Lara Pardina-Vilella
- Department of Neurology, Hospital Universitario Donostia, San Sebastián, Spain.,Neuroscience Area, Biodonostia Health Research Institute, San Sebastián, Spain
| | | | - Ana Gorostidi
- Neuroscience Area, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Ioana Croitoru
- Neuroscience Area, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Alberto Bergareche
- Department of Neurology, Hospital Universitario Donostia, San Sebastián, Spain.,Neuroscience Area, Biodonostia Health Research Institute, San Sebastián, Spain.,Centro de Investigación Biomédica en Red para Enfermedades Neurodegenerativas (CIBERNED), Carlos III Health Institute, Madrid, Spain
| | - Javier Ruiz-Martinez
- Department of Neurology, Hospital Universitario Donostia, San Sebastián, Spain.,Neuroscience Area, Biodonostia Health Research Institute, San Sebastián, Spain.,Centro de Investigación Biomédica en Red para Enfermedades Neurodegenerativas (CIBERNED), Carlos III Health Institute, Madrid, Spain
| |
Collapse
|
15
|
Donahue EK, Murdos A, Jakowec MW, Sheikh-Bahaei N, Toga AW, Petzinger GM, Sepehrband F. Global and Regional Changes in Perivascular Space in Idiopathic and Familial Parkinson's Disease. Mov Disord 2021; 36:1126-1136. [PMID: 33470460 DOI: 10.1002/mds.28473] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 11/23/2020] [Accepted: 12/14/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The glymphatic system, including the perivascular space (PVS), plays a critical role in brain homeostasis. Although mounting evidence from Alzheimer's disease has supported the potential role of PVS in neurodegenerative disorders, its contribution in Parkinson's disease (PD) has not been fully elucidated. Although idiopathic (IPD) and familial PD (FPD) share similar pathophysiology in terms of protein aggregation, the differential impact of PVS on PD subtypes remains unknown. Our objective was to examine the differences in PVS volume fraction in IPD and FPD compared to healthy controls (HCs) and nonmanifest carriers (NMCs). METHODS A total of 470 individuals were analyzed from the Parkinson's Progression Markers Initiative database, including (1) IPD (n = 179), (2) FPD (LRRK2 [leucine-rich repeat kinase 2], glucocerebrosidase, or α-synuclein) (n = 67), (3) NMC (n = 101), and (4) HCs (n = 84). Total PVS volume fraction (%) was compared using parcellation and quantitation within greater white matter volume at global and regional levels in all cortical and subcortical white matter. RESULTS There was a significant increase in global and regional PVS volume fraction in PD versus non-PD, particularly in FPD versus NMC and LRRK2 FPD versus NMC. Regionally, FPD and NMC differed in the medial orbitofrontal region, as did LRRK2 FPD versus NMC. Non-PD and PD differed in the medial orbitofrontal region and the banks of the superior temporal regions. IPD and FPD differed in the cuneus and lateral occipital regions. CONCLUSIONS Our findings support the role of PVS in PD and highlight a potentially significant contribution of PVS to the pathophysiology of FPD, particularly LRRK2. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Erin K Donahue
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Neuroscience Graduate Program, University of Southern California, Los Angeles, California, USA
| | - Amjad Murdos
- Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Michael W Jakowec
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Neuroscience Graduate Program, University of Southern California, Los Angeles, California, USA
| | - Nasim Sheikh-Bahaei
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Arthur W Toga
- Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Giselle M Petzinger
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Neuroscience Graduate Program, University of Southern California, Los Angeles, California, USA
| | - Farshid Sepehrband
- Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
16
|
Dwyer Z, Rudyk C, Thompson A, Farmer K, Fenner B, Fortin T, Derksen A, Sun H, Hayley S. Leucine-rich repeat kinase-2 (LRRK2) modulates microglial phenotype and dopaminergic neurodegeneration. Neurobiol Aging 2020; 91:45-55. [PMID: 32247534 DOI: 10.1016/j.neurobiolaging.2020.02.017] [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: 09/19/2019] [Revised: 01/23/2020] [Accepted: 02/15/2020] [Indexed: 12/21/2022]
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a common gene implicated in Parkinson's disease and many inflammatory processes. Thus, we assessed the role of LRRK2 in the context of endotoxin (lipopolysaccharide, LPS)-induced inflammation of the substantia nigra together with the environmental toxicant, paraquat, that has been implicated in PD. Here we found that LRRK2 ablation prevented the loss of dopaminergic neurons and behavioral deficits (motor) induced by LPS priming followed by paraquat exposure. The LRRK2 ablation also provoked a phenotypic shift in LPS-primed microglia cells. The LRRK2 deficiency reduced their "activated" morphology and upregulation of the inflammatory phagocytic regulator, WAVE2 (critical for actin remodeling), while the chemokine receptor, CX3CR1, was elevated in isolated CD11b+ myeloid cells. Furthermore, LRRK2 knockout attenuated the signs of oxidative stress and morphological changes induced in primary microglia by LPS treatment. However, induced WAVE2 expression together with LPS exposure in microglia overcame the inhibitory effects of LRRK2 knockout, suggesting WAVE2 may be acting downstream of LRRK2. Neither WAVE2 nor did LRRK2 knockout influence LPS-induced cytokine elevations in the microglia. We are the first to show the importance of LRRK2 in neurodegenerative and inflammatory processes in this multi-hit toxin model of PD. These data are consistent with the proposition that LRRK2 and WAVE2 are useful therapeutic targets for PD or other conditions with a prominent neuroinflammatory component.
Collapse
Affiliation(s)
- Zach Dwyer
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.
| | - Chris Rudyk
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Ashley Thompson
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Kyle Farmer
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Barbara Fenner
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Teresa Fortin
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Alexa Derksen
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Hongyu Sun
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.
| | -
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
17
|
Simuni T, Brumm MC, Uribe L, Caspell-Garcia C, Coffey CS, Siderowf A, Alcalay RN, Trojanowski JQ, Shaw LM, Seibyl J, Singleton A, Toga AW, Galasko D, Foroud T, Nudelman K, Tosun-Turgut D, Poston K, Weintraub D, Mollenhauer B, Tanner CM, Kieburtz K, Chahine LM, Reimer A, Hutten S, Bressman S, Marek K. Clinical and Dopamine Transporter Imaging Characteristics of Leucine Rich Repeat Kinase 2 (LRRK2) and Glucosylceramidase Beta (GBA) Parkinson's Disease Participants in the Parkinson's Progression Markers Initiative: A Cross-Sectional Study. Mov Disord 2020; 35:833-844. [PMID: 32073681 DOI: 10.1002/mds.27989] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND There are limited data on the phenotypic and dopamine transporter (DAT) imaging characterization of the Parkinson's disease (PD) patients with leucine rich kinase 2 (LRRK2) and glucosylceramidase beta (GBA) mutations. OBJECTIVE The objective of this study was to examine baseline clinical and DAT imaging characteristics in GBA and LRRK2 mutation carriers with early PD compared with sporadic PD. METHODS The Parkinson's Progression Markers Initiative is an ongoing observational longitudinal study that enrolled participants with sporadic PD, LRRK2 and GBA PD carriers from 33 sites worldwide. All participants are assessed annually with a battery of motor and nonmotor scales, 123-I Ioflupane DAT imaging, and biologic variables. RESULTS We assessed 158 LRRK2 (89% G2019S), 80 GBA (89 %N370S), and 361 sporadic PD participants with the mean (standard deviation) disease duration of 2.9 (1.9), 3.1 (2.0), and 2.6 (0.6) years, respectively. When compared with sporadic PD, the GBA PD patients had no difference in any motor, cognitive, or autonomic features. The LRRK2 PD patients had less motor disability and lower rapid eye movement behavior disorder questionnaire scores, but no meaningful difference in cognitive or autonomic features. Both genetic cohorts had a higher score on the impulse control disorders scale when compared with sporadic PD, but no difference in other psychiatric features. Both genetic PD cohorts had less loss of dopamine transporter on DAT imaging when compared with sporadic PD. CONCLUSIONS We confirm previous reports of milder phenotype associated with LRRK2-PD. A previously reported more aggressive phenotype in GBA-PD is not evident early in the disease in N370s carriers. This observation identifies a window for potential disease-modifying interventions. Longitudinal data will be essential to define the slope of progression for both genetic cohorts. TRIAL REGISTRATION ClinicalTrials.gov (NCT01141023). © 2020 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Tanya Simuni
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Michael C Brumm
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa, USA
| | - Liz Uribe
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa, USA
| | - Chelsea Caspell-Garcia
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa, USA
| | - Christopher S Coffey
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa, USA
| | - Andrew Siderowf
- Departments of Neurology Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Roy N Alcalay
- Department of Neurology, The Taub Institite for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA
| | - John Q Trojanowski
- Departments of Pathology and Laboratory Medicine Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Leslie M Shaw
- Departments of Pathology and Laboratory Medicine Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John Seibyl
- Institute for Neurodegenerative Disorders, New Haven, Connecticut, USA
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, Maryland, USA
| | - Arthur W Toga
- Laboratory of Neuroimaging (LONI), University of Southern California, Los Angeles, California, USA
| | - Doug Galasko
- Department of Neurology, University of California, San Diego, California, USA
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana, USA
| | - Kelly Nudelman
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana, USA
| | - Duygu Tosun-Turgut
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Kathleen Poston
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, USA
| | - Daniel Weintraub
- Departments of Psychiatry and Neurology Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany and Paracelsus-Elena-Klinik, Kassel, Germany
| | - Caroline M Tanner
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Karl Kieburtz
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
| | - Lana M Chahine
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alyssa Reimer
- The Michael J. Fox Foundation for Parkinson's Research, New York, New York, USA
| | - Samantha Hutten
- The Michael J. Fox Foundation for Parkinson's Research, New York, New York, USA
| | - Susan Bressman
- Icahn School of Medicine, Mount Sinai, New York, New York, USA
| | - Kenneth Marek
- Institute for Neurodegenerative Disorders, New Haven, Connecticut, USA
| | | |
Collapse
|
18
|
Systematic review of genetic variants associated with cognitive impairment and depressive symptoms in Parkinson's disease. Acta Neuropsychiatr 2020; 32:10-22. [PMID: 31292011 DOI: 10.1017/neu.2019.28] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Cognitive impairment and depression are among the most prevalent and most disabling non-motor symptoms in Parkinson's disease (PD). The genetic factors that are associated with these symptoms remain uncertain. This systematic review aims to summarise the prevailing evidence from all genetic association studies investigating the genetic variants associated with cognitive impairment and depressive symptoms in people with PD. METHOD A systematic review using five online databases: PubMed, PsycINFO, CINAHL, EMBASE and OpenGrey (PROSPERO protocol: CRD42017067431). We completed the quality assessment using the Q-Genie tool. RESULTS 2353 articles were screened, and 43 articles were found to be eligible to be included. A meta-analysis of studies investigating LRRK2 rs34637584 confirmed that the minor allele carriers had significantly less cognitive impairment (p = 0.015). Further meta-analyses showed that GBA variants rs76763715 (p < 0.001) and rs421016 (p = 0.001) were significantly associated with more cognitive impairment in people with PD. Minor alleles of GBA variants rs76763715, rs421016, rs387906315 and rs80356773 were associated with more depressive symptoms in PD. Moreover, APOE ε4 allele has been associated with more cognitive impairment in PD. BDNF (rs6265) and CRY1 (rs2287161) variants have been associated with more depressive symptoms in people with PD. CONCLUSIONS PD carriers of GBA variants are at high risk for cognitive decline and depression. Screening for these variants may facilitate early identification and effective management of these non-motor symptoms. The molecular mechanisms underlying favourable cognitive functioning in LRRK2 rs34637584 variant carriers warrant further investigation.
Collapse
|
19
|
Tolosa E, Vila M, Klein C, Rascol O. LRRK2 in Parkinson disease: challenges of clinical trials. Nat Rev Neurol 2020; 16:97-107. [PMID: 31980808 DOI: 10.1038/s41582-019-0301-2] [Citation(s) in RCA: 268] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2019] [Indexed: 12/27/2022]
Abstract
One of the most common monogenic forms of Parkinson disease (PD) is caused by mutations in the LRRK2 gene that encodes leucine-rich repeat kinase 2 (LRRK2). LRRK2 mutations, and particularly the most common mutation Gly2019Ser, are observed in patients with autosomal dominant PD and in those with apparent sporadic PD, who are clinically indistinguishable from those with idiopathic PD. The discoveries that pathogenic mutations in the LRRK2 gene increase LRRK2 kinase activity and that small-molecule LRRK2 kinase inhibitors can be neuroprotective in preclinical models of PD have placed LRRK2 at the centre of disease modification efforts in PD. Recent investigations also suggest that LRRK2 has a role in the pathogenesis of idiopathic PD and that LRRK2 therapies might, therefore, be beneficial in this common subtype of PD. In this Review, we describe the characteristics of LRRK2-associated PD that are most relevant to the development of LRRK2-targeted therapies and the design and implementation of clinical trials. We highlight strategies for correcting the effects of mutations in the LRRK2 gene, focusing on how to identify which patients are the optimal candidates and how to decide on the timing of such trials. In addition, we discuss challenges in implementing trials of disease-modifying treatment in people who carry LRRK2 mutations.
Collapse
Affiliation(s)
- Eduardo Tolosa
- Parkinson and Movement Disorders Unit, Neurology Service, Hospital Clinic of Barcelona, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), University of Barcelona, Barcelona, Spain. .,Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.
| | - Miquel Vila
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.,Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR), Autonomous University of Barcelona, Barcelona, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Olivier Rascol
- Clinical Investigation Center CIC1436, Departments of Clinical Pharmacology and Neurosciences, NS-Park/FCRIN network and NeuroToul Center of Excellence for Neurodegeneration, INSERM, University Hospital of Toulouse and University of Toulouse, Toulouse, France
| |
Collapse
|
20
|
Kim T, Song S, Park Y, Kang S, Seo H. HDAC Inhibition by Valproic Acid Induces Neuroprotection and Improvement of PD-like Behaviors in LRRK2 R1441G Transgenic Mice. Exp Neurobiol 2019; 28:504-515. [PMID: 31495079 PMCID: PMC6751862 DOI: 10.5607/en.2019.28.4.504] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 12/15/2022] Open
Abstract
Parkinson’s disease (PD) is one of the late-onset neurodegenerative movement disorder. Major pathological markers of PD include progressive loss of dopaminergic neurons, Lewy body formation, genetic mutations, and environmental factors. Epigenetic regulation of specific gene expression via impaired histone acetylation is associated with neuronal dysfunction in various neurodegenerative diseases. In this study, we hypothesized that histone deacetylase (HDAC) inhibitor, valproic acid (VPA), can improve motor function by enhancing cell survival in PD genetic model mice with LRRK2 R1441G mutation. To address this question, we administered VPA in LRRK2 R1441G transgenic mice to determine whether VPA affects 1) histone acetylation and HDAC expression, 2) dopaminergic neuron survival, 3) inflammatory responses, 4) motor or non-motor symptoms. As results, VPA administration increased histone acetylation level and the number of tyrosine hydroxylase (TH) positive neurons in substantia nigra of LRRK2 R1441G mice. VPA reduced iba-1 positive activated microglia and the mRNA levels of pro-inflammatory marker genes in LRRK2 R1441G mice. In addition, VPA induced the improvement of PD-like motor and non-motor behavior in LRRK2 R1441G mice. These data suggest that the inhibition of HDAC can be further studied as potential future therapeutics for PD.
Collapse
Affiliation(s)
- Taewoo Kim
- Department of Molecular & Life Sciences, Hanyang University, Ansan 15588, Korea
| | - Seohoe Song
- Department of Molecular & Life Sciences, Hanyang University, Ansan 15588, Korea
| | - Yeongwon Park
- Department of Molecular & Life Sciences, Hanyang University, Ansan 15588, Korea
| | - Sinil Kang
- Department of Molecular & Life Sciences, Hanyang University, Ansan 15588, Korea
| | - Hyemyung Seo
- Department of Molecular & Life Sciences, Hanyang University, Ansan 15588, Korea
| |
Collapse
|
21
|
Stemmer A, Galili T, Kozlovski T, Zeevi Y, Marcus-Kalish M, Benjamini Y, Mitelpunkt A. Current and Potential Approaches for Defining Disease Signatures: a Systematic Review. J Mol Neurosci 2019; 67:550-558. [PMID: 30778835 DOI: 10.1007/s12031-019-01269-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 01/22/2019] [Indexed: 01/07/2023]
Abstract
Identifying disease signatures in order to facilitate accurate diagnosis/treatment has been the focus of research efforts in the last decade. However, the term "disease signature" has not been properly defined, resulting in inconsistencies between studies, as well as limited ability to fully utilize the tools/information available in the evolving field of healthcare big data. Research was conducted according to the Preferred Reporting Items for Systematic Review and Meta-analyses (PRISMA) guidelines. The search (in PubMed, Cochrane, and Web of Science) was limited to English articles published up to 31/12/2016. The search string was "disease signature" OR "disease signatures" OR "disease fingerprint" OR "disease fingerprints" OR "subtype signature" OR "subtype signatures" OR "subgroup signature" OR "subgroup signatures." The full text of the articles was reviewed to determine the meaning of the phrase "disease signature" as well as the context of its use. Of 285 articles identified in the search, 129 were included in the final analysis. The term disease signature was first found in an article from 2001. In the last 10 years, the use of the term increased by approximately ninefold, which is double the general increase in the number of published articles. Only one article attempted to define the term. The two major medical fields where the term was used were oncology (31%) and neurology (20%); 71% of the identified articles used a single biomarker to define the term, 13% of the articles used a pair of biomarkers, and 16% used signatures with multiple biomarker; in 42% of the identified articles, genomic biomarkers were used for the signature, in 17% measurements of biochemical compounds in body fluids, and in 10%, changes in imaging studies were used for the signature. Our findings identified a lack of consistency in defining the term disease signature. We suggest a novel hierarchical multidimensional concept for this term that would combine both current approaches for identifying diseases (one focusing on undesired effects of the disease and the other on its causes). This model can improve disease signature definition consistency which will enable to generalize and classify diseases, resulting in more precise treatments and better outcomes. Ultimately, this model could lead to developing a statistical confidence in a disease signature that would allow physicians/patients to estimate the precision of the diagnosis, which, in turn, may have important implications on patients' prognosis and treatment.
Collapse
Affiliation(s)
- Amos Stemmer
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Tal Galili
- Department of Statistics and Operations Research, Tel Aviv University, Tel Aviv, Israel
| | - Tal Kozlovski
- Department of Statistics and Operations Research, Tel Aviv University, Tel Aviv, Israel
| | - Yoav Zeevi
- The Sagol School for Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Mira Marcus-Kalish
- Department of Statistics and Operations Research, Tel Aviv University, Tel Aviv, Israel
- The Sagol School for Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Yoav Benjamini
- Department of Statistics and Operations Research, Tel Aviv University, Tel Aviv, Israel
- The Sagol School for Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Alexis Mitelpunkt
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Statistics and Operations Research, Tel Aviv University, Tel Aviv, Israel
- Pediatric Neurology, Dana-Dwek Children's Hospital, Tel Aviv Medical Center, Tel Aviv, Israel
| |
Collapse
|
22
|
Ben Romdhan S, Farhat N, Nasri A, Lesage S, Hdiji O, Ben Djebara M, Landoulsi Z, Stevanin G, Brice A, Damak M, Gouider R, Mhiri C. LRRK2 G2019S Parkinson's disease with more benign phenotype than idiopathic. Acta Neurol Scand 2018; 138:425-431. [PMID: 29989150 DOI: 10.1111/ane.12996] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 06/21/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVES The LRRK2-G2019S mutation is the most common cause of Parkinson's disease (PD) in North Africa. G2019S-PD has been described as similar to idiopathic with minor clinical differences. The aim of this study was to determine the G2019S-related phenotype and to investigate gender and gene dosage effects on clinical features of G2019S carriers. PATIENTS AND METHODS The G2019S mutation was screened in 250 Tunisian patients with PD. Twenty-four patients carrying mutations in other PD genes were excluded. Logistic regression models were used to compare clinical features between the studied groups. RESULTS G2019S carriers (107 cases) and non-carriers (119 cases) were similar in disease duration, levodopa doses, and gender and phenotype distributions. However, carriers had a younger age at examination, higher level of education, and were more likely to report family history of PD and to develop PD at earlier age (P = 0.017). Adjusted for age, sex, disease duration, levodopa-equivalent dose and educational level, MMSE scores remained significantly higher (adjust P = 0.019) and UPDRS-III scores were lower (adjust P = 0.012) in the G2019S carriers than non-carriers. Demographic characteristics of men and women with G2019S mutation were similar, but men had higher level of education, better cognition (adjust P-value for educational level = 0.042) and less tendency towards depression than females (adjust P = 0.046). Furthermore, PD phenotype did not differ between the homozygous and heterozygous G2019S carriers. CONCLUSION In this study, G2019S carriers had a more benign phenotype than non-carriers. Cognitive impairment and depression were less common in G2019S male carriers compared with females. In addition, we found that LRRK2 gene dosage does not influence the severity of PD.
Collapse
Affiliation(s)
- Sawssan Ben Romdhan
- Laboratory of Neurogenetics; Parkinson's Disease and Cerebrovascular Disease; University Hospital Habib Bourguiba; Sfax Tunisia
- Institut du Cerveau et de la Moelle épinière; INSERM U1127; Sorbonne Université; UPMC Paris VI Univ. UMR_S1127; CNRS UMR 7225; Paris France
- École Pratique des Hautes Études EPHE; PSL Research University; Paris France
| | - Nouha Farhat
- Laboratory of Neurogenetics; Parkinson's Disease and Cerebrovascular Disease; University Hospital Habib Bourguiba; Sfax Tunisia
| | - Amina Nasri
- Department of Neurology; University Hospital Razi; Tunis; Mannouba Tunisia
| | - Suzanne Lesage
- Institut du Cerveau et de la Moelle épinière; INSERM U1127; Sorbonne Université; UPMC Paris VI Univ. UMR_S1127; CNRS UMR 7225; Paris France
| | - Olfa Hdiji
- Laboratory of Neurogenetics; Parkinson's Disease and Cerebrovascular Disease; University Hospital Habib Bourguiba; Sfax Tunisia
| | - Mouna Ben Djebara
- Department of Neurology; University Hospital Razi; Tunis; Mannouba Tunisia
| | - Zied Landoulsi
- Department of Neurology; University Hospital Razi; Tunis; Mannouba Tunisia
| | - Giovanni Stevanin
- Institut du Cerveau et de la Moelle épinière; INSERM U1127; Sorbonne Université; UPMC Paris VI Univ. UMR_S1127; CNRS UMR 7225; Paris France
- École Pratique des Hautes Études EPHE; PSL Research University; Paris France
| | - Alexis Brice
- Institut du Cerveau et de la Moelle épinière; INSERM U1127; Sorbonne Université; UPMC Paris VI Univ. UMR_S1127; CNRS UMR 7225; Paris France
| | - Mariem Damak
- Laboratory of Neurogenetics; Parkinson's Disease and Cerebrovascular Disease; University Hospital Habib Bourguiba; Sfax Tunisia
| | - Riadh Gouider
- Department of Neurology; University Hospital Razi; Tunis; Mannouba Tunisia
| | - Chokri Mhiri
- Laboratory of Neurogenetics; Parkinson's Disease and Cerebrovascular Disease; University Hospital Habib Bourguiba; Sfax Tunisia
| |
Collapse
|
23
|
Shu L, Zhang Y, Pan H, Xu Q, Guo J, Tang B, Sun Q. Clinical Heterogeneity Among LRRK2 Variants in Parkinson's Disease: A Meta-Analysis. Front Aging Neurosci 2018; 10:283. [PMID: 30283330 PMCID: PMC6156433 DOI: 10.3389/fnagi.2018.00283] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 08/30/2018] [Indexed: 12/14/2022] Open
Abstract
Background: Parkinson's disease (PD) is one of the most common neurodegenerative diseases. Variants in the LRRK2 gene have been shown to be associated with PD. However, the clinical characteristics of LRRK2-related PD are heterogeneous. In our study, we performed a comprehensive pooled analysis of the association between specific LRRK2 variants and clinical features of PD. Methods: Articles from the Medline, Embase, and Cochrane databases were included in the meta-analysis. Strict inclusion criteria were applied, and detailed information was extracted from the final original articles included. Revman 5.3 software was used for publication biases and pooled and sensitivity analyses. Results: In all, 66 studies having the clinical manifestations of PD patients with G2019S, G2385R, R1628P, and R1441G were included for the final analysis. The prominent clinical features of LRRK2-G2019S-related PD patients were female sex, higher rates of early-onset PD (EOPD), and family history (OR: 0.77 [male], 1.37, 2.62; p < 0.00001, 0.02, < 0.00001). PD patients with G2019S were more likely to have high scores of Schwab & England (MD: 1.49; p < 0.00001), low GDS scores, high UPSIT scores (MD: 0.43, 4.70; p = 0.01, < 0.00001), and good response to L-dopa (OR: 2.33; p < 0.0001). Further, G2019S carriers had higher LEDD (MD: 115.20; p < 0.00001) and were more likely to develop motor complications, such as dyskinesia and motor fluctuations (OR: 2.18, 2.02; p < 0.00001, 0.04) than non-carriers. G2385R carriers were more likely to have family history (OR: 2.10; p = 0.007) than non-G2385R carriers and lower H-Y and higher MMSE scores (MD: −0.13, 1.02; p = 0.02, 0.0007). G2385R carriers had higher LEDD and tended to develop motor complications, such as motor fluctuations (MD: 53.22, OR: 3.17; p = 0.01, < 0.00001) than non-carriers. Other clinical presentations did not feature G2019S or G2385R. We observed no distinct clinical features for R1628P or R1441G. Our subgroup analyses in different ethnic group for specific variant also presented with relevant clinical characteristics of PD patients. Conclusions: Clinical heterogeneity was observed among LRRK2-associated PD in different variants in total and in different ethnic groups, especially for G2019S and G2385R.
Collapse
Affiliation(s)
- Li Shu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Hongxu Pan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qian Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,Parkinson's Disease Center of Beijing Institute for Brain Disorders, Beijing, China.,Collaborative Innovation Center for Brain Science, Shanghai, China.,Collaborative Innovation Center for Genetics and Development, Shanghai, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,Parkinson's Disease Center of Beijing Institute for Brain Disorders, Beijing, China.,Collaborative Innovation Center for Brain Science, Shanghai, China.,Collaborative Innovation Center for Genetics and Development, Shanghai, China.,Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China.,Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Qiying Sun
- National Clinical Research Center for Geriatric Disorders, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
24
|
Gunzler SA, Riley DE, Chen SG, Tatsuoka CM, Johnson WM, Mieyal JJ, Walter EM, Whitney CM, Feng IJ, Owusu-Dapaah H, Mittal SO, Wilson-Delfosse AL. Motor and non-motor features of Parkinson's disease in LRRK2 G2019S carriers versus matched controls. J Neurol Sci 2018; 388:203-207. [PMID: 29627023 DOI: 10.1016/j.jns.2018.03.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/04/2018] [Accepted: 03/14/2018] [Indexed: 11/17/2022]
Abstract
INTRODUCTION LRRK2 G2019S mutation carriers with Parkinson's disease (PD) have been generally indistinguishable from those with idiopathic PD, with the exception of variable differences in some motor and non-motor domains, including cognition, gait, and balance. LRRK2 G2019S is amongst the most common genetic etiologies for PD, particularly in Ashkenazi Jewish (AJ) populations. METHODS This cross-sectional data collection study sought to clarify the phenotype of LRRK2 G2019S mutation carriers with PD. Primary endpoints were the Movement Disorder Society Unified Parkinson Disease Rating Scale (MDS-UPDRS) and Montreal Cognitive Assessment (MoCA). Other motor and non-motor data were also assessed. The Mann-Whitney U Test was utilized to compare LRRK2 G2019S carriers with PD (LRRK2+) with non-carrier PD controls who were matched for age, gender, education, and PD duration. Survival analyses and log rank tests were utilized to compare interval from onset of PD to development of motor and non-motor complications. RESULTS We screened 251 subjects and 231 completed the study, of whom 9 were LRRK2+, including 7 AJ subjects. 22.73% of AJ subjects with a family history of PD (FH) and 12.96% of AJ subjects without a FH were LRRK2+. There were no significant differences between the 9 LRRK2+ subjects and 19 matched PD controls in MDS-UPDRS, MoCA, or other motor and non-motor endpoints. CONCLUSION Prevalence of the LRRK2 G2019S mutation in AJ and non-AJ subjects in our study population in Cleveland, Ohio was comparable to other clinical studies. There were no significant motor or non-motor differences between LRRK2+ PD and matched PD controls.
Collapse
Affiliation(s)
- Steven A Gunzler
- Parkinson's and Movement Disorders Center, Neurological Institute, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, 11100 Euclid Avenue, HAN5040, Cleveland, OH 44106, USA.
| | - David E Riley
- InMotion, 4829 Galaxy Parkway, Suite M, Warrensville Heights, OH 44128, USA
| | - Shu G Chen
- Department of Pathology, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| | - Curtis M Tatsuoka
- Biostatistics, Neurological Institute, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, 11100 Euclid Avenue, HAN5040, Cleveland, OH 44106, USA.
| | - William M Johnson
- Department of Ophthalmology, Duke University School of Medicine, 2608 Erwin Rd, Durham, NC 27705, USA.
| | - John J Mieyal
- Department of Pharmacology, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| | - Ellen M Walter
- Parkinson's and Movement Disorders Center, Neurological Institute, University Hospitals Cleveland Medical Center, 11100 Euclid Avenue, HAN5040, Cleveland, OH 44106, USA.
| | - Christina M Whitney
- Parkinson's and Movement Disorders Center, Neurological Institute, University Hospitals Cleveland Medical Center (retired), 11100 Euclid Avenue, HAN5040, Cleveland, OH 44106, USA
| | - I Jung Feng
- Epidemiology & Biostatistics, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| | - Harry Owusu-Dapaah
- Department of Internal Medicine, MetroHealth Medical Center, 2500 MetroHealth Drive, Cleveland, OH 44109, USA.
| | - Shivam O Mittal
- Neurology, Columbia Asia Hospitals, Sarjapur Road, Bangalore 560035, India
| | - Amy L Wilson-Delfosse
- Department of Pharmacology, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| |
Collapse
|
25
|
De Rosa A, Peluso S, De Lucia N, Russo P, Annarumma I, Esposito M, Manganelli F, Brunetti A, De Michele G, Pappatà S. Cognitive profile and 18F-fluorodeoxyglucose PET study in LRRK2 -related Parkinson's disease. Parkinsonism Relat Disord 2018; 47:80-83. [DOI: 10.1016/j.parkreldis.2017.12.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/23/2017] [Accepted: 12/08/2017] [Indexed: 11/28/2022]
|
26
|
da Silva CP, de M Abreu G, Cabello Acero PH, Campos M, Pereira JS, de A Ramos SR, Nascimento CM, Voigt DD, Rosso AL, Araujo Leite MA, Vasconcellos LFR, Nicaretta DH, Della Coletta MV, da Silva DJ, Gonçalves AP, Dos Santos JM, Calassara V, Valença DCT, de M Martins CJ, Santos-Rebouças CB, Pimentel MMG. Clinical profiles associated with LRRK2 and GBA mutations in Brazilians with Parkinson's disease. J Neurol Sci 2017; 381:160-164. [PMID: 28991672 DOI: 10.1016/j.jns.2017.08.3249] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/11/2017] [Accepted: 08/23/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative disorder characterized by remarkable phenotypic variability. Accumulated evidence points that the manifestation of PD clinical signs might be differentially modified by genetic factors, as mutations in LRRK2 and GBA genes. In this sense, the clarification of the genotype-phenotype correlations in PD has important implications in predicting prognosis and can contribute to the development of specific therapeutic approaches. METHODS Here, we conducted the first comparative analysis of motor and non-motor features in 17 LRRK2 and 22 GBA mutation carriers and 93 non-carriers unrelated PD patients from Brazil, a highly admixed population. RESULTS Significant differences were found between the three groups. LRRK2 PD patients presented more occurrence of familiar history. Resting tremor was observed in a lower frequency in GBA mutation carries. In contrast, gait freezing and dysautonomia was present in lower frequencies in LRRK2 carriers. Besides that, LRRK2 and GBA mutation carriers showed a higher incidence of depressive symptoms and a younger age at onset, when compared to non-carriers. CONCLUSION Our results suggest that specific mutations in GBA and LRRK2 influence the clinical signs of the disease, with significant implications for handling of specific patient groups.
Collapse
Affiliation(s)
- Camilla P da Silva
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabriella de M Abreu
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Pedro H Cabello Acero
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Laboratory of Genetics, School of Health Science, University of Grande Rio, Rio de Janeiro, Brazil
| | - Mário Campos
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - João S Pereira
- Movement Disorders Section, Neurology Service, Pedro Ernesto University Hospital, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sarah R de A Ramos
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Caroline M Nascimento
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danielle D Voigt
- Laboratory of Genetics, School of Health Science, University of Grande Rio, Rio de Janeiro, Brazil
| | - Ana Lucia Rosso
- University Hospital Clementino Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marco A Araujo Leite
- Movement Disorders Unit, Division of Neurology, Hospital Antônio Pedro, Fluminense Federal University, Brazil
| | - Luiz Felipe R Vasconcellos
- Institute of Neurology Deolindo Couto, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Federal Hospital of Servidores do Estado, Rio de Janeiro, Brazil
| | | | | | - Delson José da Silva
- Neuroscience Core, Hospital Clinics, Federal University of Goiás, Brazil; Integrated Neurosciences Institute, Goiás, Brazil
| | - Andressa P Gonçalves
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jussara M Dos Santos
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Veluma Calassara
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Débora Cristina T Valença
- Laboratory of Clinical and Experimental Pathophysiology (CLINEX), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Cyro J de M Martins
- Laboratory of Clinical and Experimental Pathophysiology (CLINEX), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Cíntia B Santos-Rebouças
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Márcia M G Pimentel
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
27
|
Chang A, Fox SH. Psychosis in Parkinson's Disease: Epidemiology, Pathophysiology, and Management. Drugs 2017; 76:1093-118. [PMID: 27312429 DOI: 10.1007/s40265-016-0600-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Psychotic symptoms are common in Parkinson's disease (PD) and are associated with poorer quality of life and increased caregiver burden. PD psychosis is correlated with several factors, such as more advanced disease, cognitive impairment, depression, and sleep disorders. The underlying causes of psychosis in PD thus involve a complex interplay between exogenous (e.g., drugs, intercurrent illnesses) and endogenous (e.g., PD disease pathology) factors. Current theories of the pathophysiology of PD psychosis have come from several neuropathological and neuroimaging studies that implicate pathways involving visual processing and executive function, including temporo-limbic structures and neocortical gray matter with altered neurotransmitter functioning (e.g., dopamine, serotonin, and acetylcholine). Treatment of PD psychosis requires a step-wise process, including initial careful investigation of treatable triggering conditions and a comprehensive evaluation with adjustment of PD medications and/or initiation of specific antipsychotic therapies. Clozapine remains the only recommended drug for the treatment of PD psychosis; however, because of regular blood monitoring, quetiapine is usually first-line therapy, although less efficacious. Emerging studies have focused on agents involving other neurotransmitters, including the serotonin 5-HT2A receptor inverse agonist pimavanserin, cholinesterase inhibitors, and antidepressants and anxiolytics.
Collapse
Affiliation(s)
- Anna Chang
- Morton and Gloria Shulman Movement Disorder Clinic, University of Toronto, Toronto Western Hospital, 7th Floor, McLaughlin Pavilion, 399 Bathurst Street, Toronto, ON, M5T 2S8, Canada.,Department of Neurology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Susan H Fox
- Morton and Gloria Shulman Movement Disorder Clinic, University of Toronto, Toronto Western Hospital, 7th Floor, McLaughlin Pavilion, 399 Bathurst Street, Toronto, ON, M5T 2S8, Canada.
| |
Collapse
|
28
|
Fagan ES, Pihlstrøm L. Genetic risk factors for cognitive decline in Parkinson's disease: a review of the literature. Eur J Neurol 2017; 24:561-e20. [DOI: 10.1111/ene.13258] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/11/2017] [Indexed: 01/18/2023]
Affiliation(s)
- E. S. Fagan
- Institute of Clinical Medicine; University of Oslo; Oslo Norway
| | - L. Pihlstrøm
- Institute of Clinical Medicine; University of Oslo; Oslo Norway
- Department of Neurology; Oslo University Hospital; Oslo Norway
| |
Collapse
|
29
|
Abstract
LRRK2 mutations are present in 1% of all sporadic Parkinson's disease (PD) cases and 5% of all familial PD cases. Several mutations in the LRRK2 gene are associated with PD, the most common of which is the Gly2019Ser mutation. In the following review, we summarize the demographics and motor and non-motor symptoms of LRRK2 carriers with PD, as well as symptoms in non-manifesting carriers. The clinical features of LRRK2-associated PD are often indistinguishable from those of idiopathic PD on an individual basis. However, LRRK2 PD patients are likely to have less non-motor symptoms compared to idiopathic PD patients, including less olfactory and cognitive impairment. LRRK2-associated PD patients are less likely to report REM sleep behavior disorder (RBD) than noncarriers. In addition, it is possible that carriers are more prone to cancer than noncarriers with PD, but larger studies are required to confirm this observation. Development of more sensitive biomarkers to identify mutation carriers at risk of developing PD, as well as biomarkers of disease progression among LRRK2 carriers with PD, is required. Such biomarkers would help evaluate interventions, which may prevent PD among non-manifesting carriers, or slow down disease progression among carriers with PD.
Collapse
|
30
|
Hong JH, Kim YK, Park JS, Lee JE, Oh MS, Chung EJ, Kim JY, Sung YH, Lyoo CH, Lee JH, Kwon DY, Kim HS, Shin HW, Park SA, Park IS, Kim JS, Lee PH, Koh SB, Baik JS, Kim SJ, Ma HI, Kim JW, Kim YJ. Lack of association between LRRK2 G2385R and cognitive dysfunction in Korean patients with Parkinson's disease. J Clin Neurosci 2016; 36:108-113. [PMID: 27839916 DOI: 10.1016/j.jocn.2016.10.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 10/15/2016] [Indexed: 01/22/2023]
Abstract
Aside from the glucocerebrosidase gene, the genetic risk factors for cognitive decline in Parkinson's disease (PD) are controversial. We investigated whether the G2385R polymorphism in leucine-rich repeat kinase 2 gene (LRRK2), a risk variant for the development of PD in East Asians, is associated with cognitive dysfunction in PD. We recruited 299 PD patients, consisting of 23 carriers and 276 non-carriers of LRRK2 G2385R, from 14 centers. Global cognitive function was assessed using the Mini-Mental State Examination (MMSE) or the Montreal Cognitive Assessment (MoCA). PD with cognitive dysfunction was defined as an MMSE Z score that, adjusting for age at study entry and years of education, was below -1.0 standard deviations. In multivariate analysis, PD duration, age at study entry and depression were significant risk factors for cognitive dysfunction as assessed by MMSE performance or the MoCA. In linear regression analysis of the association between MMSE Z scores and PD duration, there was no significant difference associated with the LRRK2 G2385R genotype. The interaction terms between PD duration and the LRRK2 G2385R genotype were not significant for the MMSE Z score but were significant for the MoCA. In conclusion, the LRRK2 G2385R genotype may not be associated with cognitive dysfunction in PD.
Collapse
Affiliation(s)
- Jeong Hoon Hong
- ILSONG Institute of Life Science, Hallym University, Anyang, South Korea
| | - Yue Kyung Kim
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, South Korea
| | - Jae Seol Park
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, South Korea
| | - Ji Eun Lee
- Department of Neurology, National Health Insurance Service Ilsan Hospital, Ilsan, South Korea
| | - Mi Sun Oh
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, South Korea
| | - Eun Joo Chung
- Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, South Korea
| | - Jeong-Yeon Kim
- Department of Neurology, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, South Korea
| | - Young-Hee Sung
- Department of Neurology, Gachon University College of Medicine, Incheon, South Korea
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae Hyeok Lee
- Department of Neurology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Do-Young Kwon
- Department of Neurology, Korea University College of Medicine, Ansan Hospital, Ansan, South Korea
| | - Hyun Sook Kim
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Hae-Won Shin
- Department of Neurology, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Sun Ah Park
- Department of Neurology, Soonchunhyang University Bucheon Hospital, Bucheon, South Korea
| | - In-Seok Park
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Joong-Seok Kim
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Seong-Beom Koh
- Department of Neurology, Korea University College of Medicine at Guro Hospital, Seoul, South Korea
| | - Jong Sam Baik
- Department of Neurology, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, South Korea
| | - Sang Jin Kim
- Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, South Korea
| | - Hyeo-Il Ma
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, South Korea
| | - Jae Woo Kim
- Department of Neurology, Dong-A University School of Medicine, Busan, South Korea
| | - Yun Joong Kim
- ILSONG Institute of Life Science, Hallym University, Anyang, South Korea; Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, South Korea; Hallym Institute of Translational Genomics and Bioinformatics, Hallym University Medical Center, Anyang, South Korea.
| |
Collapse
|
31
|
Weil RS, Schrag AE, Warren JD, Crutch SJ, Lees AJ, Morris HR. Visual dysfunction in Parkinson's disease. Brain 2016; 139:2827-2843. [PMID: 27412389 PMCID: PMC5091042 DOI: 10.1093/brain/aww175] [Citation(s) in RCA: 247] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 05/23/2016] [Accepted: 06/05/2016] [Indexed: 01/09/2023] Open
Abstract
Patients with Parkinson's disease have a number of specific visual disturbances. These include changes in colour vision and contrast sensitivity and difficulties with complex visual tasks such as mental rotation and emotion recognition. We review changes in visual function at each stage of visual processing from retinal deficits, including contrast sensitivity and colour vision deficits to higher cortical processing impairments such as object and motion processing and neglect. We consider changes in visual function in patients with common Parkinson's disease-associated genetic mutations including GBA and LRRK2 . We discuss the association between visual deficits and clinical features of Parkinson's disease such as rapid eye movement sleep behavioural disorder and the postural instability and gait disorder phenotype. We review the link between abnormal visual function and visual hallucinations, considering current models for mechanisms of visual hallucinations. Finally, we discuss the role of visuo-perceptual testing as a biomarker of disease and predictor of dementia in Parkinson's disease.
Collapse
Affiliation(s)
- Rimona S. Weil
- 1 Institute of Neurology, University College London, London, UK
- 2 National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
- 3 Department of Clinical Neurosciences, Royal Free Hospital NHS Trust, London, UK
| | - Anette E. Schrag
- 1 Institute of Neurology, University College London, London, UK
- 2 National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Jason D. Warren
- 2 National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
- 4 Dementia Research Centre, UCL Institute of Neurology, University College London, London, UK
| | - Sebastian J. Crutch
- 4 Dementia Research Centre, UCL Institute of Neurology, University College London, London, UK
| | - Andrew J. Lees
- 1 Institute of Neurology, University College London, London, UK
- 2 National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Huw R. Morris
- 1 Institute of Neurology, University College London, London, UK
- 2 National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
- 3 Department of Clinical Neurosciences, Royal Free Hospital NHS Trust, London, UK
| |
Collapse
|
32
|
Mata IF, Davis MY, Lopez AN, Dorschner MO, Martinez E, Yearout D, Cholerton BA, Hu SC, Edwards KL, Bird TD, Zabetian CP. The discovery of LRRK2 p.R1441S, a novel mutation for Parkinson's disease, adds to the complexity of a mutational hotspot. Am J Med Genet B Neuropsychiatr Genet 2016; 171:925-30. [PMID: 27111571 PMCID: PMC5028305 DOI: 10.1002/ajmg.b.32452] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 04/08/2016] [Indexed: 12/16/2022]
Abstract
Mutations in the LRRK2 gene result in autosomal dominant, late onset Parkinson's disease (PD). Three such mutations (p.R1441C, p.R1441G, and p.R1441H) are known to occur within codon 1441, and haplotype analyses indicate that each one has arisen independently on multiple occasions. We sequenced the entire coding region of 18 casual genes for PD or other parkinsonian neurodegenerative disorders in the proband of a family with autosomal dominant PD. We discovered a new missense mutation in the LRRK2 gene, c.4321C>A (p.R1441S). The mutation was predicted to be highly deleterious in silico (Combined Annotation Dependent Depletion score of 25.5) and segregated with disease in the pedigree. The clinical characteristics of affected family members were similar to those described in PD families with other mutations in LRRK2 codon 1441 and included resting tremor, rigidity, bradykinesia, unilateral onset, and a good response to levodopa. Age at onset ranged from 41 to 76. Two of the affected members of the pedigree underwent detailed, longitudinal neuropsychological testing, and both displayed evidence of mild cognitive deficits at or slightly preceding the onset of motor symptoms. LRRK2 p.R1441S represents the fourth pathogenic mutation observed within codon 1441 and its discovery adds to the remarkable complexity of a mutational hotspot within the ROC domain of the LRRK2 protein. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ignacio F. Mata
- Veterans Affairs Puget Sound Health Care System, Seattle, WA,Department of Neurology, University of Washington School of Medicine, Seattle, WA
| | - Marie Y. Davis
- Veterans Affairs Puget Sound Health Care System, Seattle, WA,Department of Neurology, University of Washington School of Medicine, Seattle, WA
| | - Alexis N. Lopez
- Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Michael O. Dorschner
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA,Department of Pathology, University of Washington, Seattle, WA
| | - Erica Martinez
- Veterans Affairs Puget Sound Health Care System, Seattle, WA
| | - Dora Yearout
- Veterans Affairs Puget Sound Health Care System, Seattle, WA,Department of Neurology, University of Washington School of Medicine, Seattle, WA
| | - Brenna A. Cholerton
- Veterans Affairs Puget Sound Health Care System, Seattle, WA,Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA
| | - Shu-Ching Hu
- Veterans Affairs Puget Sound Health Care System, Seattle, WA,Department of Neurology, University of Washington School of Medicine, Seattle, WA
| | - Karen L. Edwards
- Department of Epidemiology, University of California, Irvine, CA
| | - Thomas D. Bird
- Veterans Affairs Puget Sound Health Care System, Seattle, WA,Department of Neurology, University of Washington School of Medicine, Seattle, WA
| | - Cyrus P. Zabetian
- Veterans Affairs Puget Sound Health Care System, Seattle, WA,Department of Neurology, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
33
|
Apathy associated with neurocognitive disorders: Recent progress and future directions. Alzheimers Dement 2016; 13:84-100. [PMID: 27362291 DOI: 10.1016/j.jalz.2016.05.008] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 05/10/2016] [Accepted: 05/22/2016] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Apathy is common in neurocognitive disorders (NCDs) such as Alzheimer's disease and mild cognitive impairment. Although the definition of apathy is inconsistent in the literature, apathy is primarily defined as a loss of motivation and decreased interest in daily activities. METHODS The Alzheimer's Association International Society to Advance Alzheimer's Research and Treatment (ISTAART) Neuropsychiatric Syndromes Professional Interest Area (NPS-PIA) Apathy workgroup reviewed the latest research regarding apathy in NCDs. RESULTS Progress has recently been made in three areas relevant to apathy: (1) phenomenology, including the use of diagnostic criteria and novel instruments for measurement, (2) neurobiology, including neuroimaging, neuropathological and biomarker correlates, and (3) interventions, including pharmacologic, nonpharmacologic, and noninvasive neuromodulatory approaches. DISCUSSION Recent progress confirms that apathy has a significant impact on those with major NCD and those with mild NCDs. As such, it is an important target for research and intervention.
Collapse
|
34
|
Giladi N, Mirelman A, Thaler A, Orr-Urtreger A. A Personalized Approach to Parkinson's Disease Patients Based on Founder Mutation Analysis. Front Neurol 2016; 7:71. [PMID: 27242656 PMCID: PMC4861838 DOI: 10.3389/fneur.2016.00071] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 04/25/2016] [Indexed: 12/20/2022] Open
Abstract
While the phenotype of Parkinson disease (PD) is heterogeneous, treatment approaches are mostly uniform. Personalized medicine aims to treat diseases with targeted therapies based on cumulative variables, including genotype. We believe that sufficient evidence has accumulated to warrant the initiation of personalized medicine in PD based on subjects genotype and provide examples for our reasoning from observations of GBA and LRRK2 mutations carriers. While PD patients who carry the G2019S mutation in the LRRK2 gene seem to develop relatively mild disease with more frequent postural instability gait disturbance phenotype, carriers of mutations in the GBA gene tend to have an early onset, rapidly deteriorating disease, with more pronounced cognitive and autonomic impairments. These characteristics have significant implications for treatment and outcome and should be addressed from an early stage in the attempt to improve the patient's quality of life.
Collapse
Affiliation(s)
- Nir Giladi
- Laboratory for Early Markers of Neurodegeneration, Department of Neurology, Tel-Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Anat Mirelman
- Laboratory for Early Markers of Neurodegeneration, Department of Neurology, Tel-Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Avner Thaler
- Laboratory for Early Markers of Neurodegeneration, Department of Neurology, Tel-Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Avi Orr-Urtreger
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Genetic Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| |
Collapse
|
35
|
Sloan M, Alegre-Abarrategui J, Potgieter D, Kaufmann AK, Exley R, Deltheil T, Threlfell S, Connor-Robson N, Brimblecombe K, Wallings R, Cioroch M, Bannerman DM, Bolam JP, Magill PJ, Cragg SJ, Dodson PD, Wade-Martins R. LRRK2 BAC transgenic rats develop progressive, L-DOPA-responsive motor impairment, and deficits in dopamine circuit function. Hum Mol Genet 2016; 25:951-63. [PMID: 26744332 PMCID: PMC4754049 DOI: 10.1093/hmg/ddv628] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 12/29/2015] [Indexed: 11/13/2022] Open
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) lead to late-onset, autosomal dominant Parkinson's disease, characterized by the degeneration of dopamine neurons of the substantia nigra pars compacta, a deficit in dopamine neurotransmission and the development of motor and non-motor symptoms. The most prevalent Parkinson's disease LRRK2 mutations are located in the kinase (G2019S) and GTPase (R1441C) encoding domains of LRRK2. To better understand the sequence of events that lead to progressive neurophysiological deficits in vulnerable neurons and circuits in Parkinson's disease, we have generated LRRK2 bacterial artificial chromosome transgenic rats expressing either G2019S or R1441C mutant, or wild-type LRRK2, from the complete human LRRK2 genomic locus, including endogenous promoter and regulatory regions. Aged (18-21 months) G2019S and R1441C mutant transgenic rats exhibit L-DOPA-responsive motor dysfunction, impaired striatal dopamine release as determined by fast-scan cyclic voltammetry, and cognitive deficits. In addition, in vivo recordings of identified substantia nigra pars compacta dopamine neurons in R1441C LRRK2 transgenic rats reveal an age-dependent reduction in burst firing, which likely results in further reductions to striatal dopamine release. These alterations to dopamine circuit function occur in the absence of neurodegeneration or abnormal protein accumulation within the substantia nigra pars compacta, suggesting that nigrostriatal dopamine dysfunction precedes detectable protein aggregation and cell death in the development of Parkinson's disease. In conclusion, our longitudinal deep-phenotyping provides novel insights into how the genetic burden arising from human mutant LRRK2 manifests as early pathophysiological changes to dopamine circuit function and highlights a potential model for testing Parkinson's therapeutics.
Collapse
Affiliation(s)
- Max Sloan
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics
| | | | - Dawid Potgieter
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics
| | - Anna-Kristin Kaufmann
- Oxford Parkinson's Disease Centre, Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology and
| | - Richard Exley
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics
| | - Thierry Deltheil
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics
| | - Sarah Threlfell
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics
| | | | | | - Rebecca Wallings
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics
| | - Milena Cioroch
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics
| | - David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - J Paul Bolam
- Oxford Parkinson's Disease Centre, Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology and
| | - Peter J Magill
- Oxford Parkinson's Disease Centre, Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology and
| | - Stephanie J Cragg
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics,
| | - Paul D Dodson
- Oxford Parkinson's Disease Centre, Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology and
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics,
| |
Collapse
|
36
|
Thaler A, Helmich RC, Or-Borichev A, van Nuenen BFL, Shapira-Lichter I, Gurevich T, Orr-Urtreger A, Marder K, Bressman S, Bloem BR, Giladi N, Hendler T, Mirelman A. Intact working memory in non-manifesting LRRK2 carriers--an fMRI study. Eur J Neurosci 2015; 43:106-12. [PMID: 26536050 DOI: 10.1111/ejn.13120] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 10/28/2015] [Accepted: 10/29/2015] [Indexed: 01/26/2023]
Abstract
Cognitive impairments are prevalent in patients with Parkinson's disease. Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common cause of genetic Parkinsonism. Non-manifesting carriers of the G2019S mutation in the LRRK2 gene were found to have lower executive functions as measured by the Stroop task. This exploratory study aimed to assess whether the cognitive impairment in non-manifesting carriers is specific for executive functions or includes other cognitive domains such as working memory. We recruited 77 non-manifesting first-degree relatives of Parkinson's disease patients (38 carriers). A block-design fMRI N-back task, with 0-back, 2-back and 3-back conditions, was used in order to assess working memory. Participants were well matched on the Montreal Cognitive Assessment, University of Pennsylvania Smell Identification Test, Unified Parkinson's Disease Rating Scale part III, digit span, age, gender and Beck Depression Inventory. The task achieved the overall expected effect in both groups with longer reaction times and lower accuracy rates with increasing task demands. However, no whole-brain or region-of-interest between-groups differences were found on any of the task conditions. These results indicate that non-manifesting carriers of the G2019S mutation in the LRRK2 gene have a specific cognitive profile with executive functions, as assessed by the Stroop task, demonstrating significant impairment but with working memory, as assessed with the N-back task, remaining relatively intact. These finding shed light on the pre-motor cognitive changes in this unique 'at risk' population and should enable more focused cognitive assessments of these cohorts.
Collapse
Affiliation(s)
- Avner Thaler
- Movement Disorders Unit, Department of Neurology, Tel-Aviv Sourasky Medical Center, 6 Weizman Street, Tel Aviv, 64239, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Functional Brain Center, Wohl Institute for Advanced Imaging, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Rick C Helmich
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Ayelet Or-Borichev
- Functional Brain Center, Wohl Institute for Advanced Imaging, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | | | - Irit Shapira-Lichter
- Functional Brain Center, Wohl Institute for Advanced Imaging, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Tanya Gurevich
- Movement Disorders Unit, Department of Neurology, Tel-Aviv Sourasky Medical Center, 6 Weizman Street, Tel Aviv, 64239, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Avi Orr-Urtreger
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Genetic Institute, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Karen Marder
- Columbia University Medical Center, Columbia University, New-York, NY, USA
| | | | - Bastiaan R Bloem
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Nir Giladi
- Movement Disorders Unit, Department of Neurology, Tel-Aviv Sourasky Medical Center, 6 Weizman Street, Tel Aviv, 64239, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Sieratzki Chair in Neurology, Tel Aviv University, Tel Aviv, Israel
| | - Talma Hendler
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Functional Brain Center, Wohl Institute for Advanced Imaging, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sagol School for Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Anat Mirelman
- Movement Disorders Unit, Department of Neurology, Tel-Aviv Sourasky Medical Center, 6 Weizman Street, Tel Aviv, 64239, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | |
Collapse
|