1
|
Chahine LM, Lafontant DE, Choi SH, Iwaki H, Blauwendraat C, Singleton AB, Brumm MC, Alcalay RN, Merchant K, Nudelman KNH, Dagher A, Vo A, Tao Q, Venuto CS, Kieburtz K, Poston KL, Bressman S, Gonzalez-Latapi P, Avants B, Coffey C, Jennings D, Tolosa E, Siderowf A, Marek K, Simuni T. LRRK2-associated parkinsonism with and without in vivo evidence of alpha-synuclein aggregates: longitudinal clinical and biomarker characterization. Brain Commun 2025; 7:fcaf103. [PMID: 40114783 PMCID: PMC11925012 DOI: 10.1093/braincomms/fcaf103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/17/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
Among LRRK2-associated parkinsonism cases with nigral degeneration, over two-thirds demonstrate evidence of pathologic alpha-synuclein, but many do not. Understanding the clinical phenotype and underlying biology in such individuals is critical for therapeutic development. Our objective was to compare clinical and biomarker features, and rate of progression over 4 years of follow-up, among LRRK2-associated parkinsonism cases with and without in vivo evidence of alpha-synuclein aggregates. Data were from the Parkinson's Progression Markers Initiative, a multicentre prospective cohort study. The sample included individuals diagnosed with Parkinson disease with pathogenic variants in LRRK2. Presence of CSF alpha-synuclein aggregation was assessed with seed amplification assay. A range of clinician- and patient-reported outcome assessments were administered. Biomarkers included dopamine transporter scan, CSF amyloid-beta1-42, total tau, phospho-tau181, urine bis(monoacylglycerol)phosphate levels and serum neurofilament light chain. Linear mixed-effects (LMMs) models examined differences in trajectory in CSF-negative and CSF-positive groups. A total of 148 LRRK2 parkinsonism cases (86% with G2019S variant), 46 negative and 102 positive for CSF alpha-synuclein seed amplification assay, were included. At baseline, the negative group was older than the positive group [median (inter-quartile range) 69.1 (65.2-72.3) versus 61.5 (55.6-66.9) years, P < 0.001] and a greater proportion were female [28 (61%) versus 43 (42%), P = 0.035]. Despite being older, the negative group had similar duration since diagnosis and similar motor rating scale [16 (11-23) versus 16 (10-22), P = 0.480] though lower levodopa equivalents. Only 13 (29%) of the negative group were hyposmic, compared with 75 (77%) of the positive group. The negative group, compared with the positive group, had higher per cent-expected putamenal dopamine transporter binding for their age and sex [0.36 (0.29-0.45) versus 0.26 (0.22-0.37), P < 0.001]. Serum neurofilament light chain was higher in the negative group compared with the positive group [17.10 (13.60-22.10) versus 10.50 (8.43-14.70) pg/mL; age-adjusted P-value = 0.013]. In terms of longitudinal change, the negative group remained stable in functional rating scale score in contrast to the positive group who had a significant increase (worsening) of 0.729 per year (P = 0.037), but no other differences in trajectory were found. Among individuals diagnosed with Parkinson disease with pathogenic variants in the LRRK2 gene, we found clinical and biomarker differences in cases without versus with in vivo evidence of CSF alpha-synuclein aggregates. LRRK2 parkinsonism cases without evidence of alpha-synuclein aggregates as a group exhibit less severe motor manifestations and decline. The underlying biology in LRRK2 parkinsonism cases without evidence of alpha-synuclein aggregates requires further investigation.
Collapse
Affiliation(s)
- Lana M Chahine
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - David-Erick Lafontant
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA 55848, USA
| | - Seung Ho Choi
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA 55848, USA
| | - Hirotaka Iwaki
- DataTecnica LLC, Washington, DC 20037, USA
- Center for Alzheimer’s and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA
- National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Neurogenetics, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cornelis Blauwendraat
- Center for Alzheimer’s and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA
- National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Neurogenetics, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew B Singleton
- Center for Alzheimer’s and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA
- National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Neurogenetics, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael C Brumm
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA 55848, USA
| | - Roy N Alcalay
- Tel Aviv Sourasky Medical Center, 64239 Tel-Aviv, Israel
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kalpana Merchant
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | - Alain Dagher
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada H3A 2B4
| | - Andrew Vo
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada H3A 2B4
| | - Qin Tao
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada H3A 2B4
| | - Charles S Venuto
- Department of Neurology, Center for Health and Technology, University of Rochester Medical Center, Rochester, NY 14642,USA
| | - Karl Kieburtz
- Department of Neurology, Center for Health and Technology, University of Rochester Medical Center, Rochester, NY 14642,USA
| | - Kathleen L Poston
- Department of Neurology, Stanford University School of Medicine, Palo Alto, 94304 CA, USA
| | - Susan Bressman
- Department of Neurology, Icahn School of Medicine, Mount Sinai Beth Israel, New York City, NY 10029, USA
| | - Paulina Gonzalez-Latapi
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | - Christopher Coffey
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA 55848, USA
| | - Danna Jennings
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Eduardo Tolosa
- Parkinson’s Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, 08028 Barcelona, Spain
| | - Andrew Siderowf
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ken Marek
- Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA
| | - Tatyana Simuni
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
2
|
Carricarte Naranjo C, Marras C, Visanji NP, Cornforth DJ, Sanchez-Rodriguez L, Schüle B, Goldman SM, Estévez M, Stein PK, Jelinek HF, Lang AE, Machado A. Heartbeat signature for predicting motor and non-motor involvement among nonparkinsonian LRRK2 G2019S mutation carriers. Clin Auton Res 2025:10.1007/s10286-024-01104-6. [PMID: 39969690 DOI: 10.1007/s10286-024-01104-6] [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: 07/11/2024] [Accepted: 12/23/2024] [Indexed: 02/20/2025]
Abstract
PURPOSE Increased beat-to-beat heart rate variability (HRV) is a feature of patients with Parkinson's disease (PD) who carry the G2019S mutation in the LRRK2 gene (LRRK2-PD). Since LRRK2 mutations have incomplete penetrance, HRV changes preceding PD conversion would likely be observed only in a subset of LRRK2 non-manifesting carriers (NMC). We aimed to assess HRV in a subgroup of NMC with distinctive characteristics of LRRK2-PD, identified through clustering analysis. METHODS HRV measures derived from 300 normal heartbeat intervals extracted from the electrocardiograms of 25 NMC, 32 related non-carriers (RNC), 27 unrelated healthy controls, and 14 patients with LRRK2-PD were analyzed. Clinical symptoms were evaluated using questionnaires and scales, and three NMC subgroups were identified using a k-means cluster analysis on the basis of the deceleration capacity of heart rate (DC) and Rényi entropy. Standard and advanced HRV measures were compared using multiple regression analysis, controlling for age, sex, and mean heart rate. RESULTS Beat-to-beat HRV markers were significantly increased in a subgroup of seven NMC (NMC2, 28%) compared with RNC and controls. Increased irregularity and DC were also verified in the NMC2 compared with controls, and were typical traits in both the NMC2 and RNC. Overall, the HRV profile of NMC2 was comparable to that of patients with LRRK2-PD. NMC2 further exhibited greater motor and non-motor traits than the other NMC, RNC, and controls. CONCLUSIONS Our results confirmed that HRV characteristics of LRRK2-PD are also found in a subset of NMC displaying clinical traits of LRRK2-PD. Further research is needed to clarify whether higher HRV represents a LRRK2-PD prodromal manifestation.
Collapse
Affiliation(s)
| | - Connie Marras
- Edmond J Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON, Canada
| | - Naomi P Visanji
- Edmond J Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON, Canada
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - David J Cornforth
- Member of the National Coalition of Independent Scholars (NCIS), Brattleboro, VT, USA
| | | | - Birgitt Schüle
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - Samuel M Goldman
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Mario Estévez
- Departamento de Neurofisiología Clínica, Instituto de Neurología y Neurocirugía, La Habana, Cuba
| | - Phyllis K Stein
- School of Medicine, Washington University, St. Louis, MO, USA
| | - Herbert F Jelinek
- Department of Medical Sciences and Biotechnology Center, Khalifa University, Abu Dhabi, UAE
| | - Anthony E Lang
- Edmond J Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON, Canada
| | - Andrés Machado
- Facultad de Biología, Universidad de La Habana, La Habana, Cuba
| |
Collapse
|
3
|
Bonato G, Antonini A, Pistonesi F, Campagnolo M, Guerra A, Biundo R, Pilleri M, Bertolin C, Salviati L, Carecchio M. Genetic mutations in Parkinson's disease: screening of a selected population from North-Eastern Italy. Neurol Sci 2025; 46:165-174. [PMID: 39034353 PMCID: PMC11698772 DOI: 10.1007/s10072-024-07690-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Parkinson's disease (PD) is a progressive neurodegenerative disorder with a multifactorial pathogenesis. Several genetic variants increase the risk of PD and about 5-10% of cases are monogenic. This study aims to define the genetic bases and clinical features of PD in a cohort of patients from Northeastern Italy, a peculiar geographical area previously not included in genetic screenings. METHODS Using an NGS multigenic panel, 218 PD patients were tested based on age at onset, family history and development of atypical features. RESULTS A total of 133 genetic variants were found in 103 patients. Monogenic PD was diagnosed in 43 patients (20% of the cohort); 28 (12.8%) carried mutations in GBA1, 10 in LRRK2 (4.6%) and 5 in PRKN (2.3%). In 17% of patients the genetic defect remained of uncertain interpretation. The selection criterion "age of onset < 55 years" was a significant predictor of a positive genetic test (OR 3.8, p 0.0037). GBA1 patients showed more severe symptoms and a higher burden of motor and non-motor complications compared to negative patients (dyskinesias OR 3, sleep disturbances OR 2.8, cognitive deficits OR 3.6; p < 0.05), with greater autonomic dysfunction (COMPASS-31 score 34.1 vs 20.2, p 0.03). CONCLUSIONS Applying simple clinical criteria for genetic testing allows to increase the probability to identify patients with monogenic PD and better allocate resources. This process is critical to widen the understanding of disease mechanisms and to increase the individuation of patients potentially benefitting from future disease-modifying therapies.
Collapse
Affiliation(s)
- Giulia Bonato
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padua, Italy
| | - Angelo Antonini
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padua, Italy
| | - Francesca Pistonesi
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padua, Italy
- Department of General Psychology, University of Padova, Padua, Italy
| | - Marta Campagnolo
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padua, Italy
| | - Andrea Guerra
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padua, Italy
| | - Roberta Biundo
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padua, Italy
- Department of General Psychology, University of Padova, Padua, Italy
| | | | - Cinzia Bertolin
- Department of Woman and Children's Health, Genetic Unit, University of Padova, Padua, Italy
| | - Leonardo Salviati
- Department of Woman and Children's Health, Genetic Unit, University of Padova, Padua, Italy
| | - Miryam Carecchio
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, Via Giustiniani 2, 35128, Padua, Italy.
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padua, Italy.
| |
Collapse
|
4
|
Song T, Zhou X, Wang C, Wu H, Yan X, Guo J, Tang B, Lei L, Xu Q. Clinical features and progression of Parkinson's disease with LRRK2 variants: A prospective study. Ann Clin Transl Neurol 2025; 12:34-42. [PMID: 39529459 PMCID: PMC11752092 DOI: 10.1002/acn3.52244] [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: 07/28/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE We established a prospective cohort study to investigate the differences in motor and non-motor symptoms between idiopathic Parkinson's disease (IPD) and Parkinson's disease in carriers of leucine-rich repeat kinase 2 (LRRK2) gene risk variants (LRRK2-PD). METHODS The study included 1407 individuals with IPD and 649 individuals with LRRK2-PD (comprising 304 with LRRK2-G2385R, 220 with LRRK2-R1628P, and 105 with LRRK2-A419V). Differences in symptoms between LRRK2-PD and IPD were analyzed using LCMM modeling and Cox regression analysis. RESULTS The LRRK2-G2385R variant showed slower progression in tremor symptoms and excessive daytime sleepiness compared with IPD. In contrast, symptoms associated with LRRK2-R1628P and LRRK2-A419V were more similar to those of IPD. Survival analysis revealed that LRRK2-PD does not affect life expectancy compared with IPD. INTERPRETATION Our extended longitudinal follow-up of LRRK2-PD in the Chinese population provided valuable insights, further confirming the clinical characteristics of the three LRRK2 variants.
Collapse
Affiliation(s)
- Tingwei Song
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xiaoxia Zhou
- Department of Nuclear Medicine, The Third Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Chunyu Wang
- Department of Neurology, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Heng Wu
- Department of NeurologyThe First Affiliated Hospital of University of South ChinaHengyangHunanChina
| | - Xinxiang Yan
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Jifeng Guo
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Hunan Province in Neurodegenerative DisordersChangshaHunanChina
- National Clinical Research Centre for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Beisha Tang
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Hunan Province in Neurodegenerative DisordersChangshaHunanChina
- National Clinical Research Centre for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Lifang Lei
- Department of Neurology, The Third Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Qian Xu
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Hunan Province in Neurodegenerative DisordersChangshaHunanChina
- National Clinical Research Centre for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | | |
Collapse
|
5
|
Gómez‐Garre P, Martín‐Bórnez M, Muñoz‐Delgado L, Díaz‐Belloso R, Periñán MT, Bonilla‐Toribio M, Buiza‐Rueda D, Macías‐García D, Jesús S, Adarmes‐Gómez A, Ojeda E, Luque‐Ambrosiani A, García‐Díaz S, Sánchez RP, Carrillo F, Mir P. Understanding Parkinson disease in Spain: Genetic and clinical insights. Eur J Neurol 2025; 32:e16499. [PMID: 39498811 PMCID: PMC11622318 DOI: 10.1111/ene.16499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 11/07/2024]
Abstract
BACKGROUND AND PURPOSE Parkinson disease (PD) is a complex and heterogeneous neurodegenerative disorder with a broad spectrum of clinical manifestations, determined by a complex interplay of environmental and genetic factors. This study aimed to investigate genetic variants associated with PD and assess their impact on the disease phenotype through genotype-phenotype correlations. METHODS We employed a targeted resequencing panel to analyze 27 genes linked to PD in a cohort of 1185 PD patients from southern Spain. Variants were categorized based on the American College of Medical Genetics and Genomics pathogenicity criteria. Demographic and clinical data were also collected. RESULTS Among the patients analyzed, 13.5% carried potential disease-causing pathogenic or likely pathogenic variants in 12 different genes, indicating significant genetic heterogeneity. The most frequently affected genes were LRRK2, PRKN, and GBA1 (accounting for 72.1% of positive cases). Sex-specific differences were observed, with a higher proportion of female patients carrying LRRK2 variants. Differences in age at onset and clinical features were also observed among the different mutated genes. Notably, variants in genes associated with atypical parkinsonism presented distinct clinical presentations, highlighting the importance of genetic factors in the differential diagnosis. CONCLUSIONS Our study provides valuable information on the genetic landscape of PD and its clinical manifestations. The observed genotype-phenotype correlations, along with sex-specific differences, emphasize the complexity of PD pathogenesis, underlining the importance of personalized approaches to PD diagnosis and treatment. Further investigations into genetic interactions and population-specific effects are warranted to enhance our understanding of PD etiology and improve patient care.
Collapse
Affiliation(s)
- Pilar Gómez‐Garre
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasInstituto de Salud Carlos IIIMadridSpain
| | - Miguel Martín‐Bórnez
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
| | - Laura Muñoz‐Delgado
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasInstituto de Salud Carlos IIIMadridSpain
| | - Rafael Díaz‐Belloso
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasInstituto de Salud Carlos IIIMadridSpain
- Departamento de Medicina, Facultad de MedicinaUniversidad de SevillaSevilleSpain
| | - María Teresa Periñán
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasInstituto de Salud Carlos IIIMadridSpain
- Centre for Preventive Neurology Unit, Wolfson Institute of Population HealthQueen Mary University of LondonLondonUK
| | - Marta Bonilla‐Toribio
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasInstituto de Salud Carlos IIIMadridSpain
| | - Dolores Buiza‐Rueda
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasInstituto de Salud Carlos IIIMadridSpain
| | - Daniel Macías‐García
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasInstituto de Salud Carlos IIIMadridSpain
| | - Silvia Jesús
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasInstituto de Salud Carlos IIIMadridSpain
| | - Astrid Adarmes‐Gómez
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasInstituto de Salud Carlos IIIMadridSpain
| | - Elena Ojeda
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasInstituto de Salud Carlos IIIMadridSpain
| | - Antonio Luque‐Ambrosiani
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
| | - Sergio García‐Díaz
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
| | - Rocío Pineda Sánchez
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasInstituto de Salud Carlos IIIMadridSpain
| | - Fátima Carrillo
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasInstituto de Salud Carlos IIIMadridSpain
- Departamento de Medicina, Facultad de MedicinaUniversidad de SevillaSevilleSpain
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasInstituto de Salud Carlos IIIMadridSpain
- Departamento de Medicina, Facultad de MedicinaUniversidad de SevillaSevilleSpain
| |
Collapse
|
6
|
Towns C, Fang ZH, Tan MMX, Jasaityte S, Schmaderer TM, Stafford EJ, Pollard M, Tilney R, Hodgson M, Wu L, Labrum R, Hehir J, Polke J, Lange LM, Schapira AHV, Bhatia KP, Singleton AB, Blauwendraat C, Klein C, Houlden H, Wood NW, Jarman PR, Morris HR, Real R. Parkinson's families project: a UK-wide study of early onset and familial Parkinson's disease. NPJ Parkinsons Dis 2024; 10:188. [PMID: 39420034 PMCID: PMC11487259 DOI: 10.1038/s41531-024-00778-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 08/12/2024] [Indexed: 10/19/2024] Open
Abstract
The Parkinson's Families Project is a UK-wide study aimed at identifying genetic variation associated with familial and early-onset Parkinson's disease (PD). We recruited individuals with a clinical diagnosis of PD and age at motor symptom onset ≤45 years and/or a family history of PD in up to third-degree relatives. Where possible, we also recruited affected and unaffected relatives. We analysed DNA samples with a combination of single nucleotide polymorphism (SNP) array genotyping, multiplex ligation-dependent probe amplification (MLPA), and whole-genome sequencing (WGS). We investigated the association between identified pathogenic mutations and demographic and clinical factors such as age at motor symptom onset, family history, motor symptoms (MDS-UPDRS) and cognitive performance (MoCA). We performed baseline genetic analysis in 718 families, of which 205 had sporadic early-onset PD (sEOPD), 113 had familial early-onset PD (fEOPD), and 400 had late-onset familial PD (fLOPD). 69 (9.6%) of these families carried pathogenic variants in known monogenic PD-related genes. The rate of a molecular diagnosis increased to 28.1% in PD with motor onset ≤35 years. We identified pathogenic variants in LRRK2 in 4.2% of families, and biallelic pathogenic variants in PRKN in 3.6% of families. We also identified two families with SNCA duplications and three families with a pathogenic repeat expansion in ATXN2, as well as single families with pathogenic variants in VCP, PINK1, PNPLA6, PLA2G6, SPG7, GCH1, and RAB32. An additional 73 (10.2%) families were carriers of at least one pathogenic or risk GBA1 variant. Most early-onset and familial PD cases do not have a known genetic cause, indicating that there are likely to be further monogenic causes for PD.
Collapse
Affiliation(s)
- Clodagh Towns
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Zih-Hua Fang
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Manuela M X Tan
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Simona Jasaityte
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Theresa M Schmaderer
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Eleanor J Stafford
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Miriam Pollard
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Russel Tilney
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Megan Hodgson
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
| | - Lesley Wu
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Robyn Labrum
- Neurogenetics Laboratory, National Hospital for Neurology & Neurosurgery, Queen Square, London, UK
| | - Jason Hehir
- Neurogenetics Laboratory, National Hospital for Neurology & Neurosurgery, Queen Square, London, UK
| | - James Polke
- Neurogenetics Laboratory, National Hospital for Neurology & Neurosurgery, Queen Square, London, UK
| | - Lara M Lange
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Neurology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Kailash P Bhatia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
| | - 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
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Cornelis Blauwendraat
- 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
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Nicholas W Wood
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Paul R Jarman
- National Hospital for Neurology & Neurosurgery, Queen Square, London, UK
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.
- UCL Movement Disorders Centre, University College London, London, UK.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Raquel Real
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.
- UCL Movement Disorders Centre, University College London, London, UK.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
7
|
Sosero YL, Bandres-Ciga S, Ferwerda B, Tocino MTP, Belloso DR, Gómez-Garre P, Faouzi J, Taba P, Pavelka L, Marques TM, Gomes CPC, Kolodkin A, May P, Milanowski LM, Wszolek ZK, Uitti RJ, Heutink P, van Hilten JJ, Simon DK, Eberly S, Alvarez I, Krohn L, Yu E, Freeman K, Rudakou U, Ruskey JA, Asayesh F, Menéndez-Gonzàlez M, Pastor P, Ross OA, Krüger R, Corvol JC, Koks S, Mir P, De Bie RM, Iwaki H, Gan-Or Z. Dopamine Pathway and Parkinson's Risk Variants Are Associated with Levodopa-Induced Dyskinesia. Mov Disord 2024; 39:1773-1783. [PMID: 39132902 PMCID: PMC11490412 DOI: 10.1002/mds.29960] [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: 09/20/2023] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND Levodopa-induced dyskinesia (LID) is a common adverse effect of levodopa, one of the main therapeutics used to treat the motor symptoms of Parkinson's disease (PD). Previous evidence suggests a connection between LID and a disruption of the dopaminergic system as well as genes implicated in PD, including GBA1 and LRRK2. OBJECTIVES Our goal was to investigate the effects of genetic variants on risk and time to LID. METHODS We performed a genome-wide association study (GWAS) and analyses focused on GBA1 and LRRK2 variants. We also calculated polygenic risk scores (PRS) including risk variants for PD and variants in genes involved in the dopaminergic transmission pathway. To test the influence of genetics on LID risk we used logistic regression, and to examine its impact on time to LID we performed Cox regression including 1612 PD patients with and 3175 without LID. RESULTS We found that GBA1 variants were associated with LID risk (odds ratio [OR] = 1.65; 95% confidence interval [CI], 1.21-2.26; P = 0.0017) and LRRK2 variants with reduced time to LID onset (hazard ratio [HR] = 1.42; 95% CI, 1.09-1.84; P = 0.0098). The fourth quartile of the PD PRS was associated with increased LID risk (ORfourth_quartile = 1.27; 95% CI, 1.03-1.56; P = 0.0210). The third and fourth dopamine pathway PRS quartiles were associated with a reduced time to development of LID (HRthird_quartile = 1.38; 95% CI, 1.07-1.79; P = 0.0128; HRfourth_quartile = 1.38; 95% CI = 1.06-1.78; P = 0.0147). CONCLUSIONS This study suggests that variants implicated in PD and in the dopaminergic transmission pathway play a role in the risk/time to develop LID. Further studies will be necessary to examine how these findings can inform clinical care. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Yuri L. Sosero
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - 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 on Health, Bethesda, MD, USA
| | - Bart Ferwerda
- Department of Clinical Epidemiology and Biostatistics, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Maria T. P. Tocino
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Dìaz R. Belloso
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Pilar Gómez-Garre
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Johann Faouzi
- Sorbonne Université, Paris Brain Institute – ICM, Inserm, CNRS, Assistance Publique Hôpitaux de Paris, Department of Neurology, Pitié-Salpêtrière Hospital, Paris, France
- CREST, ENSAI, Campus de Ker-Lann, 51 Rue Blaise Pascal – BP 37203 35172 Bruz Cedex, France
| | - Pille Taba
- Department of Neurology and Neurosurgery, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
| | - Lukas Pavelka
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Centre Hospitalier de Luxembourg (CHL), Strassen, Luxembourg
| | - Tainà M. Marques
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | - Clarissa P. C. Gomes
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Alexey Kolodkin
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | - Patrick May
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Centre Hospitalier de Luxembourg (CHL), Strassen, Luxembourg
| | - Lukasz M Milanowski
- Department of Neurology Faculty of Health Science, Medical University of Warsaw, Warsaw, Poland
- Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida, USA
| | | | - Ryan J. Uitti
- Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida, USA
| | | | | | - David K. Simon
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School
| | - Shirley Eberly
- Department of Biostatistics and Computational Biology at the University of Rochester School of Medicine and Dentistry
| | - Ignacio Alvarez
- Department of Neurology, Hospital Universitari Mutua de Terrassa, Barcelona, Spain
| | - Lynne Krohn
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Eric Yu
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Kathryn Freeman
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Uladzislau Rudakou
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Jennifer A. Ruskey
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Farnaz Asayesh
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Manuel Menéndez-Gonzàlez
- Facultad de Medicina y Ciencias de la Salud, Universidad de Oviedo, Calle Julián Clavería s/n, 33006 Oviedo, Spain
- Department of Neurology, Hospital Universitario Central de Asturias, Avenida Roma s/n, 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida Roma s/n, 33011 Oviedo, Spain
| | - Pau Pastor
- Department of Neurology, Hospital Universitari Mutua de Terrassa, Barcelona, Spain
- Unit of Neurodegenerative Diseases, Department of Neurology, University Hospital Germans Trias i Pujol and The Germans Trias i Pujol Research Institute (IGTP) Badalona, Barcelona, Spain
| | - Owen A. Ross
- Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Rejko Krüger
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Centre Hospitalier de Luxembourg (CHL), Strassen, Luxembourg
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jean-Christophe Corvol
- CREST, ENSAI, Campus de Ker-Lann, 51 Rue Blaise Pascal – BP 37203 35172 Bruz Cedex, France
| | - Sulev Koks
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Australia
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Rob M.A. De Bie
- Department of Neurology and Clinical Neurophysiology, Amsterdam University Medical Centers, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Hirotaka Iwaki
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes on Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, District of Columbia, USA
| | - Ziv Gan-Or
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| |
Collapse
|
8
|
Chahine LM, Lafontant DE, Ho Choi S, Iwaki H, Blauwendraat C, Singleton AB, Brumm MC, Alcalay RN, Merchant K, Nudelman KNH, Dagher A, Vo A, Tao Q, Venuto CS, Kieburtz K, Poston KL, Bressman S, Gonzalez-Latapi P, Avants B, Coffey C, Jennings D, Tolosa E, Siderowf A, Marek K, Simuni T. LRRK2-Associated Parkinsonism With and Without In Vivo Evidence of Alpha-Synuclein Aggregates. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.22.24310806. [PMID: 39108519 PMCID: PMC11302724 DOI: 10.1101/2024.07.22.24310806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Background Among LRRK2-associated parkinsonism cases with nigral degeneration, over two-thirds demonstrate evidence of pathologic alpha-synuclein, but many do not. Understanding the clinical phenotype and underlying biology in such individuals is critical for therapeutic development. Our objective was to compare clinical and biomarker features, and rate of progression over 4 years follow-up, among LRRK2-associated parkinsonism cases with and without in vivo evidence of alpha-synuclein aggregates. Methods Data were from the Parkinson's Progression Markers Initiative, a multicenter prospective cohort study. The sample included individuals diagnosed with Parkinson disease with pathogenic variants in LRRK2. Presence of CSF alpha-synuclein aggregation was assessed with seed amplification assay. A range of clinician- and patient- reported outcome assessments were administered. Biomarkers included dopamine transporter SPECT scan, CSF amyloid-beta1-42, total tau, phospho-tau181, urine bis(monoacylglycerol)phosphate levels, and serum neurofilament light chain. Linear mixed effects models examined differences in trajectory in CSF negative and positive groups. Results 148 LRRK2-parkinsonism cases (86% with G2019S variant), 46 negative and 102 positive for CSF alpha-synuclein seed amplification assay were included. At baseline, the negative group were older than the positive group (median [interquartile range] 69.1 [65.2-72.3] vs 61.5 [55.6-66.9] years, p<0.001) and a greater proportion were female (28 (61%) vs 43 (42%), p=0.035). Despite being older, the negative group had similar duration since diagnosis, and similar motor rating scale (16 [11-23] vs 16 [10-22], p=0.480) though lower levodopa equivalents. Only 13 (29%) of the negative group were hyposmic, compared to 75 (77%) of the positive group. Lowest putamen dopamine transporter binding expected for age and sex was greater in the negative vs positive groups (0.36 [0.29-0.45] vs 0.26 [0.22-0.37], p<0.001). Serum neurofilament light chain was higher in the negative group compared to the positive group (17.10 [13.60-22.10] vs 10.50 [8.43-14.70]; age-adjusted p-value=0.013). In terms of longitudinal change, the negative group remained stable in functional rating scale score in contrast to the positive group who had a significant increase (worsening) of 0.729 per year (p=0.037), but no other differences in trajectory were found. Conclusion Among individuals diagnosed with Parkinson disease with pathogenic variants in the LRRK2 gene, we found clinical and biomarker differences in cases without versus with in vivo evidence of CSF alpha-synuclein aggregates. LRRK2 parkinsonism cases without evidence of alpha-synuclein aggregates as a group exhibit less severe motor manifestations and decline may have more significant cognitive dysfunction. The underlying biology in LRRK2-parkinsonism cases without evidence of alpha-synuclein aggregates requires further investigation.
Collapse
Affiliation(s)
- Lana M Chahine
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA, 15213
| | - David-Erick Lafontant
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Seung Ho Choi
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Hirotaka Iwaki
- DataTecnica LLC, Washington, District of Columbia, USA. (2) Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
- Center for Alzheimer's and Related Dementias, National Institute on Aging and Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Cornelis Blauwendraat
- Center for Alzheimer's and Related Dementias, National Institute on Aging and Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrew B Singleton
- Center for Alzheimer's and Related Dementias, National Institute on Aging and Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael C Brumm
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Roy N Alcalay
- Neurological Institute, Tel-Aviv Medical Center, Tel-Aviv, Israel and Department of Neurology; Columbia University Irving Medical Center
| | - Kalpana Merchant
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Alain Dagher
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Andrew Vo
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Qin Tao
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Charles S Venuto
- Department of Neurology, Center for Health and Technology, University of Rochester Medical Center, Rochester, NY
| | - Karl Kieburtz
- Department of Neurology, Center for Health and Technology, University of Rochester Medical Center, Rochester, NY
| | - Kathleen L Poston
- Department of Neurology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Susan Bressman
- Department of Neurology, Mount Sinai Beth Israel and Icahn School of Medicine, Mount Sinai, New York City, New York, USA
| | - Paulina Gonzalez-Latapi
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Christopher Coffey
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | | | - Eduard Tolosa
- Parkinson's disease & Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona. Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII) Barcelona, Spain
| | - Andrew Siderowf
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ken Marek
- Institute for Neurodegenerative Disorders, New Haven, CT, USA
| | - Tanya Simuni
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
9
|
Bhatia P, Bickle M, Agrawal AA, Truss B, Nikolaidi A, Brockmann K, Reinhardt L, Vogel S, Szegoe EM, Pal A, Hermann A, Mikicic I, Yun M, Falkenburger B, Sterneckert J. Axonal Lysosomal Assays for Characterizing the Effects of LRRK2 G2019S. BIOLOGY 2024; 13:58. [PMID: 38275734 PMCID: PMC10813644 DOI: 10.3390/biology13010058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024]
Abstract
The degeneration of axon terminals before the soma, referred to as "dying back", is a feature of Parkinson's disease (PD). Axonal assays are needed to model early PD pathogenesis as well as identify protective therapeutics. We hypothesized that defects in axon lysosomal trafficking as well as injury repair might be important contributing factors to "dying back" pathology in PD. Since primary human PD neurons are inaccessible, we developed assays to quantify axonal trafficking and injury repair using induced pluripotent stem cell (iPSC)-derived neurons with LRRK2 G2019S, which is one of the most common known PD mutations, and isogenic controls. We observed a subtle axonal trafficking phenotype that was partially rescued by a LRRK2 inhibitor. Mutant LRRK2 neurons showed increased phosphorylated Rab10-positive lysosomes, and lysosomal membrane damage increased LRRK2-dependent Rab10 phosphorylation. Neurons with mutant LRRK2 showed a transient increase in lysosomes at axotomy injury sites. This was a pilot study that used two patient-derived lines to develop its methodology; we observed subtle phenotypes that might correlate with heterogeneity in LRRK2-PD patients. Further analysis using additional iPSC lines is needed. Therefore, our axonal lysosomal assays can potentially be used to characterize early PD pathogenesis and test possible therapeutics.
Collapse
Affiliation(s)
- Priyanka Bhatia
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, 01307 Dresden, Germany; (P.B.)
| | - Marc Bickle
- Roche Institute of Human Biology, 4070 Basel, Switzerland
| | - Amay A. Agrawal
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, 01307 Dresden, Germany; (P.B.)
| | - Buster Truss
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, 01307 Dresden, Germany; (P.B.)
| | - Aikaterina Nikolaidi
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, 01307 Dresden, Germany; (P.B.)
| | - Kathrin Brockmann
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - Lydia Reinhardt
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, 01307 Dresden, Germany; (P.B.)
| | - Stefanie Vogel
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, 01307 Dresden, Germany; (P.B.)
| | - Eva M. Szegoe
- Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany
| | - Arun Pal
- Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany
| | - Andreas Hermann
- Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock/Greifswald, 18147 Rostock, Germany
| | - Ivan Mikicic
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, 01307 Dresden, Germany; (P.B.)
| | - Maximina Yun
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, 01307 Dresden, Germany; (P.B.)
- Max Planck Institute of Molecular Cellular Biology and Genetics, 01307 Dresden, Germany
- Physics of Life Excellence Cluster, 01307 Dresden, Germany
| | - Björn Falkenburger
- Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany
| | - Jared Sterneckert
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, 01307 Dresden, Germany; (P.B.)
- Medical Faculty Carl Gustav Carus of TU Dresden, 01307 Dresden, Germany
| |
Collapse
|
10
|
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
|
11
|
Sosero YL, Bandres-Ciga S, Ferwerda B, Tocino MTP, Belloso DR, Gómez-Garre P, Faouzi J, Taba P, Pavelka L, Marques TM, Gomes CPC, Kolodkin A, May P, Milanowski LM, Wszolek ZK, Uitti RJ, Heutink P, van Hilten JJ, Simon DK, Eberly S, Alvarez I, Krohn L, Yu E, Freeman K, Rudakou U, Ruskey JA, Asayesh F, Menéndez-Gonzàlez M, Pastor P, Ross OA, Krüger R, Corvol JC, Koks S, Mir P, De Bie RMA, Iwaki H, Gan-Or Z. Dopamine pathway and Parkinson's risk variants are associated with levodopa-induced dyskinesia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.08.28.23294610. [PMID: 37790572 PMCID: PMC10543218 DOI: 10.1101/2023.08.28.23294610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Background Levodopa-induced dyskinesia (LID) is a common adverse effect of levodopa, one of the main therapeutics used to treat the motor symptoms of Parkinson's disease (PD). Previous evidence suggests a connection between LID and a disruption of the dopaminergic system as well as genes implicated in PD, including GBA1 and LRRK2. Objectives To investigate the effects of genetic variants on risk and time to LID. Methods We performed a genome-wide association study (GWAS) and analyses focused on GBA1 and LRRK2 variants. We also calculated polygenic risk scores including risk variants for PD and variants in genes involved in the dopaminergic transmission pathway. To test the influence of genetics on LID risk we used logistic regression, and to examine its impact on time to LID we performed Cox regression including 1,612 PD patients with and 3,175 without LID. Results We found that GBA1 variants were associated with LID risk (OR=1.65, 95% CI=1.21-2.26, p=0.0017) and LRRK2 variants with reduced time to LID onset (HR=1.42, 95% CI=1.09-1.84, p=0.0098). The fourth quartile of the PD PRS was associated with increased LID risk (ORfourth_quartile=1.27, 95% CI=1.03-1.56, p=0.0210). The third and fourth dopamine pathway PRS quartiles were associated with a reduced time to development of LID (HRthird_quartile=1.38, 95% CI=1.07-1.79, p=0.0128; HRfourth_quartile=1.38, 95% CI=1.06-1.78, p=0.0147). Conclusions This study suggests that variants implicated in PD and in the dopaminergic transmission pathway play a role in the risk/time to develop LID. Further studies will be necessary to examine how these findings can inform clinical care.
Collapse
Affiliation(s)
- Yuri L Sosero
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - 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 on Health, Bethesda, MD, USA
| | - Bart Ferwerda
- Department of Clinical Epidemiology and Biostatistics, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Maria T P Tocino
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Dìaz R Belloso
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Pilar Gómez-Garre
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Johann Faouzi
- Sorbonne Université, Paris Brain Institute - ICM, Inserm, CNRS, Assistance Publique Hôpitaux de Paris, Department of Neurology, Pitié-Salpêtrière Hospital, Paris, France
- CREST, ENSAI, Campus de Ker-Lann, 51 Rue Blaise Pascal - BP 37203 35172 Bruz Cedex, France
| | - Pille Taba
- Department of Neurology and Neurosurgery, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
| | - Lukas Pavelka
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Centre Hospitalier de Luxembourg (CHL), Strassen, Luxembourg
| | - Tainà M Marques
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | - Clarissa P C Gomes
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Alexey Kolodkin
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | - Patrick May
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Centre Hospitalier de Luxembourg (CHL), Strassen, Luxembourg
| | - Lukasz M Milanowski
- Department of Neurology Faculty of Health Science, Medical University of Warsaw, Warsaw, Poland
- Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Zbigniew K Wszolek
- Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Ryan J Uitti
- Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida, USA
| | | | | | - David K Simon
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School
| | - Shirley Eberly
- Department of Biostatistics and Computational Biology at the University of Rochester School of Medicine and Dentistry
| | - Ignacio Alvarez
- Department of Neurology, Hospital Universitari Mutua de Terrassa, Barcelona, Spain
| | - Lynne Krohn
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Eric Yu
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Kathryn Freeman
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Uladzislau Rudakou
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Jennifer A Ruskey
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Farnaz Asayesh
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Manuel Menéndez-Gonzàlez
- Facultad de Medicina y Ciencias de la Salud, Universidad de Oviedo, Calle Julián Clavería s/n, 33006 Oviedo, Spain
- Department of Neurology, Hospital Universitario Central de Asturias, Avenida Roma s/n, 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida Roma s/n, 33011 Oviedo, Spain
| | - Pau Pastor
- Department of Neurology, Hospital Universitari Mutua de Terrassa, Barcelona, Spain
- Unit of Neurodegenerative Diseases, Department of Neurology, University Hospital Germans Trias i Pujol and The Germans Trias i Pujol Research Institute (IGTP) Badalona, Barcelona, Spain
| | - Owen A Ross
- Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Rejko Krüger
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Centre Hospitalier de Luxembourg (CHL), Strassen, Luxembourg
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jean-Christophe Corvol
- CREST, ENSAI, Campus de Ker-Lann, 51 Rue Blaise Pascal - BP 37203 35172 Bruz Cedex, France
| | - Sulev Koks
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Australia
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Rob M A De Bie
- Department of Neurology and Clinical Neurophysiology, Amsterdam University Medical Centers, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Hirotaka Iwaki
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes on Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, District of Columbia, USA
| | - Ziv Gan-Or
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| |
Collapse
|
12
|
Raheel K, Deegan G, Di Giulio I, Cash D, Ilic K, Gnoni V, Chaudhuri KR, Drakatos P, Moran R, Rosenzweig I. Sex differences in alpha-synucleinopathies: a systematic review. Front Neurol 2023; 14:1204104. [PMID: 37545736 PMCID: PMC10398394 DOI: 10.3389/fneur.2023.1204104] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/13/2023] [Indexed: 08/08/2023] Open
Abstract
Background Past research indicates a higher prevalence, incidence, and severe clinical manifestations of alpha-synucleinopathies in men, leading to a suggestion of neuroprotective properties of female sex hormones (especially estrogen). The potential pathomechanisms of any such effect on alpha-synucleinopathies, however, are far from understood. With that aim, we undertook to systematically review, and to critically assess, contemporary evidence on sex and gender differences in alpha-synucleinopathies using a bench-to-bedside approach. Methods In this systematic review, studies investigating sex and gender differences in alpha-synucleinopathies (Rapid Eye Movement (REM) Behavior Disorder (RBD), Parkinson's Disease (PD), Dementia with Lewy Bodies (DLB), Multiple System Atrophy (MSA)) from 2012 to 2022 were identified using electronic database searches of PubMed, Embase and Ovid. Results One hundred sixty-two studies were included; 5 RBD, 6 MSA, 20 DLB and 131 PD studies. Overall, there is conclusive evidence to suggest sex-and gender-specific manifestation in demographics, biomarkers, genetics, clinical features, interventions, and quality of life in alpha-synucleinopathies. Only limited data exists on the effects of distinct sex hormones, with majority of studies concentrating on estrogen and its speculated neuroprotective effects. Conclusion Future studies disentangling the underlying sex-specific mechanisms of alpha-synucleinopathies are urgently needed in order to enable novel sex-specific therapeutics.
Collapse
Affiliation(s)
- Kausar Raheel
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Gemma Deegan
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- BRAIN, Imaging Centre, CNS, King’s College London, London, United Kingdom
| | - Irene Di Giulio
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- School of Basic and Medical Biosciences, Faculty of Life Science and Medicine, King’s College London, London, United Kingdom
| | - Diana Cash
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- BRAIN, Imaging Centre, CNS, King’s College London, London, United Kingdom
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Katarina Ilic
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- BRAIN, Imaging Centre, CNS, King’s College London, London, United Kingdom
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Valentina Gnoni
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro, Lecce, Italy
| | - K. Ray Chaudhuri
- Movement Disorders Unit, King’s College Hospital and Department of Clinical and Basic Neurosciences, Institute of Psychiatry, Psychology and Neuroscience and Parkinson Foundation Centre of Excellence, King’s College London, London, United Kingdom
| | - Panagis Drakatos
- School of Basic and Medical Biosciences, Faculty of Life Science and Medicine, King’s College London, London, United Kingdom
- Sleep Disorders Centre, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Rosalyn Moran
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Ivana Rosenzweig
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- Sleep Disorders Centre, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
13
|
Rao SC, Li Y, Lapin B, Pattipati S, Ghosh Galvelis K, Naito A, Gutierrez N, Leal TP, Salim A, Salles PA, De Leon M, Mata IF. Association of women-specific health factors in the severity of Parkinson's disease. NPJ Parkinsons Dis 2023; 9:86. [PMID: 37277346 PMCID: PMC10241917 DOI: 10.1038/s41531-023-00524-x] [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: 10/17/2022] [Accepted: 05/15/2023] [Indexed: 06/07/2023] Open
Abstract
Parkinson's disease (PD) is an age-related neurological disorder known for the observational differences in its risk, progression, and severity between men and women. While estrogen has been considered to be a protective factor in the development of PD, there is little known about the role that fluctuations in hormones and immune responses from sex-specific health experiences have in the disease's development and severity. We sought to identify women-specific health experiences associated with PD severity, after adjusting for known PD factors, by developing and distributing a women-specific questionnaire across the United States and creating multivariable models for PD severity. We created a questionnaire that addresses women's specific experiences and their PD clinical history and deployed it through The Parkinson's Foundation: PD Generation. To determine the association between women-specific health factors and PD severity, we constructed multivariable logistic regression models based on the MDS-UPDRS scale and the participants' questionnaire responses, genetics, and clinical data. For our initial launch in November 2021, we had 304 complete responses from PD GENEration. Univariate and multivariate logistic modeling found significant associations between major depressive disorder, perinatal depression, natural childbirth, LRRK2 genotype, B12 deficiency, total hysterectomy, and increased PD severity. This study is a nationally available questionnaire for women's health and PD. It shifts the paradigm in understanding PD etiology and acknowledging how sex-specific experiences may contribute to PD severity. In addition, the work in this study sets the foundation for future research to investigate the factors behind sex differences in PD.
Collapse
Affiliation(s)
- Shilpa C Rao
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yadi Li
- Center for Outcomes Research and Evaluation, Cleveland Clinic, Cleveland, OH, USA
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Brittany Lapin
- Center for Outcomes Research and Evaluation, Cleveland Clinic, Cleveland, OH, USA
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Sreya Pattipati
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | | | | | | | - Amira Salim
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Philippe A Salles
- Center for Movement Disorders CETRAM, University of Santiago de Chile, Santiago, Chile
| | - Maria De Leon
- DefeatParkinsons, Houston, TX, USA
- De Leon Enterprises, Houston, TX, USA
| | - Ignacio F Mata
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
14
|
Jagota P, Lim S, Pal PK, Lee J, Kukkle PL, Fujioka S, Shang H, Phokaewvarangkul O, Bhidayasiri R, Mohamed Ibrahim N, Ugawa Y, Aldaajani Z, Jeon B, Diesta C, Shambetova C, Lin C. Genetic Movement Disorders Commonly Seen in Asians. Mov Disord Clin Pract 2023; 10:878-895. [PMID: 37332644 PMCID: PMC10272919 DOI: 10.1002/mdc3.13737] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 02/27/2023] [Accepted: 03/21/2023] [Indexed: 11/21/2023] Open
Abstract
The increasing availability of molecular genetic testing has changed the landscape of both genetic research and clinical practice. Not only is the pace of discovery of novel disease-causing genes accelerating but also the phenotypic spectra associated with previously known genes are expanding. These advancements lead to the awareness that some genetic movement disorders may cluster in certain ethnic populations and genetic pleiotropy may result in unique clinical presentations in specific ethnic groups. Thus, the characteristics, genetics and risk factors of movement disorders may differ between populations. Recognition of a particular clinical phenotype, combined with information about the ethnic origin of patients could lead to early and correct diagnosis and assist the development of future personalized medicine for patients with these disorders. Here, the Movement Disorders in Asia Task Force sought to review genetic movement disorders that are commonly seen in Asia, including Wilson's disease, spinocerebellar ataxias (SCA) types 12, 31, and 36, Gerstmann-Sträussler-Scheinker disease, PLA2G6-related parkinsonism, adult-onset neuronal intranuclear inclusion disease (NIID), and paroxysmal kinesigenic dyskinesia. We also review common disorders seen worldwide with specific mutations or presentations that occur frequently in Asians.
Collapse
Affiliation(s)
- Priya Jagota
- Chulalongkorn Centre of Excellence for Parkinson's Disease and Related Disorders, Department of Medicine, Faculty of MedicineChulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross SocietyBangkokThailand
| | - Shen‐Yang Lim
- Division of Neurology, Department of Medicine, Faculty of MedicineUniversity of MalayaKuala LumpurMalaysia
- The Mah Pooi Soo & Tan Chin Nam Centre for Parkinson's & Related Disorders, Faculty of MedicineUniversity of MalayaKuala LumpurMalaysia
| | - Pramod Kumar Pal
- Department of NeurologyNational Institute of Mental Health & Neurosciences (NIMHANS)BengaluruIndia
| | - Jee‐Young Lee
- Department of NeurologySeoul Metropolitan Government‐Seoul National University Boramae Medical Center & Seoul National University College of MedicineSeoulRepublic of Korea
| | - Prashanth Lingappa Kukkle
- Center for Parkinson's Disease and Movement DisordersManipal HospitalBangaloreIndia
- Parkinson's Disease and Movement Disorders ClinicBangaloreIndia
| | - Shinsuke Fujioka
- Department of Neurology, Fukuoka University, Faculty of MedicineFukuokaJapan
| | - Huifang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases CenterWest China Hospital, Sichuan UniversityChengduChina
| | - Onanong Phokaewvarangkul
- Chulalongkorn Centre of Excellence for Parkinson's Disease and Related Disorders, Department of Medicine, Faculty of MedicineChulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross SocietyBangkokThailand
| | - Roongroj Bhidayasiri
- Chulalongkorn Centre of Excellence for Parkinson's Disease and Related Disorders, Department of Medicine, Faculty of MedicineChulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross SocietyBangkokThailand
- The Academy of Science, The Royal Society of ThailandBangkokThailand
| | - Norlinah Mohamed Ibrahim
- Neurology Unit, Department of Medicine, Faculty of MedicineUniversiti Kebangsaan MalaysiaKuala LumpurMalaysia
| | - Yoshikazu Ugawa
- Deprtment of Human Neurophysiology, Faculty of MedicineFukushima Medical UniversityFukushimaJapan
| | - Zakiyah Aldaajani
- Neurology Unit, King Fahad Military Medical ComplexDhahranSaudi Arabia
| | - Beomseok Jeon
- Department of NeurologySeoul National University College of MedicineSeoulRepublic of Korea
- Movement Disorder CenterSeoul National University HospitalSeoulRepublic of Korea
| | - Cid Diesta
- Section of Neurology, Department of NeuroscienceMakati Medical Center, NCRMakatiPhilippines
| | | | - Chin‐Hsien Lin
- Department of NeurologyNational Taiwan University HospitalTaipeiTaiwan
| |
Collapse
|
15
|
Sosero YL, Gan‐Or Z. LRRK2 and Parkinson's disease: from genetics to targeted therapy. Ann Clin Transl Neurol 2023; 10:850-864. [PMID: 37021623 PMCID: PMC10270275 DOI: 10.1002/acn3.51776] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/07/2023] [Accepted: 03/27/2023] [Indexed: 04/07/2023] Open
Abstract
LRRK2 variants are implicated in both familial and sporadic PD. LRRK2-PD has a generally benign clinical presentation and variable pathology, with inconsistent presence of Lewy bodies and marked Alzheimer's disease pathology. The mechanisms underlying LRRK2-PD are still unclear, but inflammation, vesicle trafficking, lysosomal homeostasis, and ciliogenesis have been suggested, among others. As novel therapies targeting LRRK2 are under development, understanding the role and function of LRRK2 in PD is becoming increasingly important. Here, we outline the epidemiological, pathophysiological, and clinical features of LRRK2-PD, and discuss the arising therapeutic approaches targeting LRRK2 and possible future directions for research.
Collapse
Affiliation(s)
- Yuri L. Sosero
- Montreal Neurological InstituteMcGill UniversityMontréalQuébecH3A 1A1Canada
- Department of Human GeneticsMcGill UniversityMontréalQuébecH3A 1A1Canada
| | - Ziv Gan‐Or
- Montreal Neurological InstituteMcGill UniversityMontréalQuébecH3A 1A1Canada
- Department of Human GeneticsMcGill UniversityMontréalQuébecH3A 1A1Canada
- Department of Neurology and NeurosurgeryMcGill UniversityMontréalQuébecH3A 0G4Canada
| |
Collapse
|
16
|
Kolicheski A, Turcano P, Tamvaka N, McLean PJ, Springer W, Savica R, Ross OA. Early-Onset Parkinson's Disease: Creating the Right Environment for a Genetic Disorder. JOURNAL OF PARKINSON'S DISEASE 2022; 12:2353-2367. [PMID: 36502340 PMCID: PMC9837689 DOI: 10.3233/jpd-223380] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) by its common understanding is a late-onset sporadic movement disorder. However, there is a need to recognize not only the fact that PD pathogenesis expands beyond (or perhaps to) the brain but also that many early-onset patients develop motor signs before the age of 50 years. Indeed, studies have shown that it is likely the protein aggregation observed in the brains of patients with PD precedes the motor symptoms by perhaps a decade. Studies on early-onset forms of PD have shown it to be a heterogeneous disease with multiple genetic and environmental factors determining risk of different forms of disease. Genetic and neuropathological evidence suggests that there are α-synuclein centric forms (e.g., SNCA genomic triplication), and forms that are driven by a breakdown in mitochondrial function and specifically in the process of mitophagy and clearance of damaged mitochondria (e.g., PARKIN and PINK1 recessive loss-of-function mutations). Aligning genetic forms with recognized environmental influences will help better define patients, aid prognosis, and hopefully lead to more accurately targeted clinical trial design. Work is now needed to understand the cross-talk between these two pathomechanisms and determine a sense of independence, it is noted that autopsies studies for both have shown the presence or absence of α-synuclein aggregation. The integration of genetic and environmental data is critical to understand the etiology of early-onset forms of PD and determine how the different pathomechanisms crosstalk.
Collapse
Affiliation(s)
- Ana Kolicheski
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Pierpaolo Turcano
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA,
Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Nicole Tamvaka
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA,
Mayo Graduate School, Neuroscience Track, Mayo Clinic, Jacksonville, FL, USA
| | - Pamela J. McLean
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA,
Mayo Graduate School, Neuroscience Track, Mayo Clinic, Jacksonville, FL, USA
| | - Wolfdieter Springer
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA,
Mayo Graduate School, Neuroscience Track, Mayo Clinic, Jacksonville, FL, USA
| | - Rodolfo Savica
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Owen A. Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA,
Mayo Graduate School, Neuroscience Track, Mayo Clinic, Jacksonville, FL, USA,
Department of Medicine, University College Dublin, Dublin, Ireland,
Department of Clinical Genomics, Mayo Clinic, Jacksonville, FL, USA,Department of Biology, University of NorthFlorida, Jacksonville, FL, USA,Correspondence to: Owen A. Ross, PhD, Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Road, Jacksonville, FL 32224, USA. Tel.: +1 904 953 6280; Fax: +1 904 953 7370; E-mail:
| |
Collapse
|
17
|
Xenias HS, Chen C, Kang S, Cherian S, Situ X, Shanmugasundaram B, Liu G, Scesa G, Chan CS, Parisiadou L. R1441C and G2019S LRRK2 knockin mice have distinct striatal molecular, physiological, and behavioral alterations. Commun Biol 2022; 5:1211. [PMID: 36357506 PMCID: PMC9649688 DOI: 10.1038/s42003-022-04136-8] [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: 07/27/2020] [Accepted: 10/20/2022] [Indexed: 11/12/2022] Open
Abstract
LRRK2 mutations are closely associated with Parkinson's disease (PD). Convergent evidence suggests that LRRK2 regulates striatal function. Here, by using knock-in mouse lines expressing the two most common LRRK2 pathogenic mutations-G2019S and R1441C-we investigated how LRRK2 mutations altered striatal physiology. While we found that both R1441C and G2019S mice displayed reduced nigrostriatal dopamine release, hypoexcitability in indirect-pathway striatal projection neurons, and alterations associated with an impaired striatal-dependent motor learning were observed only in the R1441C mice. We also showed that increased synaptic PKA activities in the R1441C and not G2019S mice underlie the specific alterations in motor learning deficits in the R1441C mice. In summary, our data argue that LRRK2 mutations' impact on the striatum cannot be simply generalized. Instead, alterations in electrochemical, electrophysiological, molecular, and behavioral levels were distinct between LRRK2 mutations. Our findings offer mechanistic insights for devising and optimizing treatment strategies for PD patients.
Collapse
Affiliation(s)
- Harry S Xenias
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Chuyu Chen
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Shuo Kang
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Suraj Cherian
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Xiaolei Situ
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Guoxiang Liu
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Giuseppe Scesa
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - C Savio Chan
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Loukia Parisiadou
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
18
|
Periñán MT, Brolin K, Bandres‐Ciga S, Blauwendraat C, Klein C, Gan‐Or Z, Singleton A, Gomez‐Garre P, Swanberg M, Mir P, Noyce A. Effect Modification between Genes and Environment and Parkinson's Disease Risk. Ann Neurol 2022; 92:715-724. [PMID: 35913124 PMCID: PMC9588606 DOI: 10.1002/ana.26467] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 01/11/2023]
Abstract
Parkinson's disease (PD) is a complex neurodegenerative condition in which genetic and environmental factors interact to contribute to its etiology. Remarkable progress has been made in deciphering disease etiology through genetic approaches, but there is limited data about how environmental and genetic factors interact to modify penetrance, risk, and disease severity. Here, we provide insights into environmental modifiers of PD, discussing precedents from other neurological and non-neurological conditions. Based on these examples, we outline genetic and environmental factors contributing to PD and review potential environmental modifiers of penetrance and clinical variability in monogenic and idiopathic PD. We also highlight the potential challenges and propose how future studies might tackle these important questions. ANN NEUROL 2022;92:715-724.
Collapse
Affiliation(s)
- Maria Teresa Periñán
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de SevillaHospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaMadridSpain
| | - Kajsa Brolin
- Translational Neurogenetics Unit, Wallenberg Neuroscience Center, Department of Experimental Medical ScienceLund UniversityLundSweden
| | - Sara Bandres‐Ciga
- Laboratory of Neurogenetics, Molecular Genetics Section, National Institute on AgingNational Institutes of HealthBethesdaMarylandUSA
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, Molecular Genetics Section, National Institute on AgingNational Institutes of HealthBethesdaMarylandUSA
| | - Christine Klein
- Institute of Neurogenetics and Department of NeurologyUniversity of Lübeck and University Hospital Schleswig‐HolsteinLübeckGermany
| | - Ziv Gan‐Or
- The Neuro (Montreal Neurological Institute‐Hospital)McGill UniversityMontrealQuebecCanada,Department of Neurology and NeurosurgeryMcGill UniversityMontrealQuebecCanada,Department of Human GeneticsMcGill UniversityMontrealQuebecCanada
| | - Andrew Singleton
- Laboratory of Neurogenetics, Molecular Genetics Section, National Institute on AgingNational Institutes of HealthBethesdaMarylandUSA
| | - Pilar Gomez‐Garre
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de SevillaHospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaMadridSpain
| | - Maria Swanberg
- Translational Neurogenetics Unit, Wallenberg Neuroscience Center, Department of Experimental Medical ScienceLund UniversityLundSweden
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de SevillaHospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaMadridSpain
| | - Alastair Noyce
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUK,Preventive Neurology Unit, Centre for Prevention, Detection and Diagnosis, Wolfson Institute of Population HealthQueen Mary University of LondonLondonUK
| |
Collapse
|
19
|
Niotis K, West AB, Saunders-Pullman R. Who to Enroll in Parkinson Disease Prevention Trials? The Case for Genetically At-Risk Cohorts. Neurology 2022; 99:10-18. [PMID: 35970585 DOI: 10.1212/wnl.0000000000200812] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 04/15/2022] [Indexed: 12/15/2022] Open
Abstract
Therapies that prevent the occurrence of Parkinson disease (PD) (primary prevention) or mitigate the progression of symptoms in those with early disease (secondary prevention) are a critical unmet need in disease management. Despite great promise, PD prevention trials have not yet demonstrated success. Initiation of treatment too late in the disease course and the heterogeneity of disease are obstacles that may have contributed to the failure. Genetically stratified groups offer many advantages to primary and secondary prevention trials. In addition to their ease of identification, they decrease disease heterogeneity on several levels. Particularly, they comprise a phenotypically and pathologically enriched group with defined clinical features, pathogenic mechanisms and associated proteins that may serve as specific trial endpoints, therapeutic targets and biomarkers for disease state, and pharmacodynamic and pharmacokinetic status. However, challenges arise from genetic variant heterogeneity, from reduced penetrance whereby many carriers will not develop PD, and in recruiting a population that will meet the desired outcome in the proposed study duration. In this review, we discussed the opportunities afforded by the enrollment of genetically stratified cohorts (i.e., leucine-rich repeat kinase 2 and glucocerebrosidase 1) into prevention trials with a primary focus on primary prevention trials. We also outlined challenges surrounding the enrollment of these cohorts and offered suggestions to leverage their many advantages.
Collapse
Affiliation(s)
- Kellyann Niotis
- From the Department of Neurology (K.N., R.S.-P.), Mount Sinai Beth Israel Medical Center; Department of Neurology (K.N., R.S.-P.), Icahn School of Medicine at Mount Sinai, New York; and Duke Center for Neurodegeneration Research (A.B.W.), Departments of Pharmacology and Cancer Biology, Neurology, and Neurobiology, Duke University, Durham, NC
| | - Andrew B West
- From the Department of Neurology (K.N., R.S.-P.), Mount Sinai Beth Israel Medical Center; Department of Neurology (K.N., R.S.-P.), Icahn School of Medicine at Mount Sinai, New York; and Duke Center for Neurodegeneration Research (A.B.W.), Departments of Pharmacology and Cancer Biology, Neurology, and Neurobiology, Duke University, Durham, NC
| | - Rachel Saunders-Pullman
- From the Department of Neurology (K.N., R.S.-P.), Mount Sinai Beth Israel Medical Center; Department of Neurology (K.N., R.S.-P.), Icahn School of Medicine at Mount Sinai, New York; and Duke Center for Neurodegeneration Research (A.B.W.), Departments of Pharmacology and Cancer Biology, Neurology, and Neurobiology, Duke University, Durham, NC.
| |
Collapse
|
20
|
Simpson C, Vinikoor-Imler L, Nassan FL, Shirvan J, Lally C, Dam T, Maserejian N. Prevalence of ten LRRK2 variants in Parkinson's disease: A comprehensive review. Parkinsonism Relat Disord 2022; 98:103-113. [PMID: 35654702 DOI: 10.1016/j.parkreldis.2022.05.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Variants in the leucine-rich repeat kinase 2 gene (LRRK2) are risk factors for Parkinson's disease (PD), but their prevalence varies geographically, reflecting the locations of founder events and dispersion of founders' descendants. METHODS A comprehensive literature review was conducted to identify studies providing prevalence estimates for any of ten variants in LRRK2 (G2019S, R1441C, R1441G, R1441H, I2020T, N1437H, Y1699C, S1761R, G2385R, R1628P) among individuals with PD globally. We calculated crude country-specific variant prevalence estimates and, when possible, adjusted estimates for ethno-racial composition. For clinic-based studies, probands were used over other familial cases, whereas for population-based studies, all PD cases were used. RESULTS The analysis included 161 articles from 52 countries yielding 581 prevalence estimates across the ten variants. G2019S was the most common variant, exceeding 1.0% in 26 of 51 countries with estimates. The other variants were far less common. G2385R and R1628P were observed almost exclusively in East Asian countries, where they were found in ∼5-10% of cases. All prevalence estimates adjusted for ethno-racial composition were lower than their unadjusted counterparts, although data permitting this adjustment was only available for six countries. CONCLUSIONS Except for G2019S, the LRRK2 variants covered in this review were uncommon in most countries studied. However, there were countries with higher prevalence for some variants, reflecting the uneven geographic distribution of LRRK2 variants. The fact that ethno-racial group‒adjusted estimates were lower than crude estimates suggests that estimates derived largely from clinic-based studies may overstate the true prevalence of some LRRK2 variants in PD.
Collapse
Affiliation(s)
| | | | | | | | - Cathy Lally
- Epidemiology Research and Methods LLC, Atlanta, GA, USA.
| | | | | |
Collapse
|
21
|
Tönges L, Kwon EH, Klebe S. Monogenetic Forms of Parkinson’s Disease – Bridging the Gap Between Genetics and Biomarkers. Front Aging Neurosci 2022; 14:822949. [PMID: 35317530 PMCID: PMC8934414 DOI: 10.3389/fnagi.2022.822949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
The therapy of neurodegenerative diseases such as Parkinson’s disease (PD) is still limited to the treatment of symptoms and primarily aimed at compensating for dopaminergic hypofunction. Numerous disease-modifying therapies currently in the pipeline attempt to modify the underlying pathomechanisms. In recent decades, the results of molecular genetics and biomarker research have raised hopes of earlier diagnosis and new neuroprotective therapeutic approaches. As the disease-causing processes in monogenetic forms of PD are better understood than in sporadic PD, these disease subsets are likely to benefit first from disease-modifying therapies. Recent studies have suggested that disease-relevant changes found in genetically linked forms of PD (i.e., PARK-LRRK2, PARK-GBA) can also be reproduced in patients in whom no genetic cause can be found, i.e., those with sporadic PD. It can, therefore, be assumed that as soon as the first causal therapy for genetic forms of PD is approved, more patients with PD will undergo genetic testing and counseling. Regarding future neuroprotective trials in neurodegenerative diseases and objective parameters such as biomarkers with high sensitivity and specificity for the diagnosis and course of the disease are needed. These biomarkers will also serve to monitor treatment success in clinical trials. Promising examples in PD, such as alpha-synuclein species, lysosomal enzymes, markers of amyloid and tau pathology, and neurofilament light chain, are under investigation in blood and CSF. This paper provides an overview of the opportunities and current limitations of monogenetic diagnostic and biomarker research in PD and aims to build a bridge between current knowledge and association with PD genetics and biomarkers.
Collapse
Affiliation(s)
- Lars Tönges
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, Bochum, Germany
| | - Eun Hae Kwon
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Stephan Klebe
- Department of Neurology, University Hospital Essen, Essen, Germany
- *Correspondence: Stephan Klebe,
| |
Collapse
|
22
|
Vuletić V, Rački V, Papić E, Peterlin B. A Systematic Review of Parkinson's Disease Pharmacogenomics: Is There Time for Translation into the Clinics? Int J Mol Sci 2021; 22:ijms22137213. [PMID: 34281267 PMCID: PMC8268929 DOI: 10.3390/ijms22137213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is the second most frequent neurodegenerative disease, which creates a significant public health burden. There is a challenge for the optimization of therapies since patients not only respond differently to current treatment options but also develop different side effects to the treatment. Genetic variability in the human genome can serve as a biomarker for the metabolism, availability of drugs and stratification of patients for suitable therapies. The goal of this systematic review is to assess the current evidence for the clinical translation of pharmacogenomics in the personalization of treatment for Parkinson's disease. METHODS We performed a systematic search of Medline database for publications covering the topic of pharmacogenomics and genotype specific mutations in Parkinson's disease treatment, along with a manual search, and finally included a total of 116 publications in the review. RESULTS We analyzed 75 studies and 41 reviews published up to December of 2020. Most research is focused on levodopa pharmacogenomic properties and catechol-O-methyltransferase (COMT) enzymatic pathway polymorphisms, which have potential for clinical implementation due to changes in treatment response and side-effects. Likewise, there is some consistent evidence in the heritability of impulse control disorder via Opioid Receptor Kappa 1 (OPRK1), 5-Hydroxytryptamine Receptor 2A (HTR2a) and Dopa decarboxylase (DDC) genotypes, and hyperhomocysteinemia via the Methylenetetrahydrofolate reductase (MTHFR) gene. On the other hand, many available studies vary in design and methodology and lack in sample size, leading to inconsistent findings. CONCLUSIONS This systematic review demonstrated that the evidence for implementation of pharmacogenomics in clinical practice is still lacking and that further research needs to be done to enable a more personalized approach to therapy for each patient.
Collapse
Affiliation(s)
- Vladimira Vuletić
- Clinic of Neurology, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia; (V.R.); (E.P.)
- Department of Neurology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
- Correspondence:
| | - Valentino Rački
- Clinic of Neurology, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia; (V.R.); (E.P.)
- Department of Neurology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Eliša Papić
- Clinic of Neurology, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia; (V.R.); (E.P.)
| | - Borut Peterlin
- Clinical Institute of Medical Genetics, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia;
| |
Collapse
|
23
|
Zhang X, Guarin D, Mohammadzadehhonarvar N, Chen X, Gao X. Parkinson's disease and cancer: a systematic review and meta-analysis of over 17 million participants. BMJ Open 2021; 11:e046329. [PMID: 34215604 PMCID: PMC8256737 DOI: 10.1136/bmjopen-2020-046329] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE To systematically review and qualitatively evaluate epidemiological evidence on associations between Parkinson's disease (PD) and cancer via meta-analysis. DATA SOURCES MEDLINE via PubMed, Web of Science and EMBASE, until March 2021. STUDY SELECTION Included were publications that (1) were original epidemiological studies on PD and cancer; (2) reported risk estimates; (3) were in English. Exclusion criteria included: (1) review/comments; (2) biological studies; (3) case report/autopsy studies; (4) irrelevant exposure/outcome; (5) treated cases; (6) no measure of risk estimates; (7) no confidence intervals/exact p values and (8) duplicates. DATA EXTRACTION AND SYNTHESIS PRISMA and MOOSE guidelines were followed in data extraction. Two-step screening was performed by two authors blinded to each other. A random-effects model was used to calculate pooled relative risk (RR). MAIN OUTCOMES AND MEASURES We included publications that assessed the risk of PD in individuals with vs without cancer and the risk of cancer in individuals with vs without PD. RESULTS A total of 63 studies and 17 994 584 participants were included. Meta-analysis generated a pooled RR of 0.82 (n=33; 95% CI 0.76 to 0.88; p<0.001) for association between PD and total cancer, 0.76 (n=21; 95% CI 0.67 to 0.85; p<0.001) for PD and smoking-related cancer and 0.92 (n=19; 95% CI 0.84 to 0.99; p=0.03) for non-smoking-related cancer. PD was associated with an increased risk of melanoma (n=29; pooled RR=1.75; 95% CI 1.43 to 2.14; p<0.001) but not for other skin cancers (n=17; pooled RR=0.90; 95% CI 0.60 to 1.34; p=0.60). CONCLUSIONS PD and total cancer were inversely associated. This inverse association persisted for both smoking-related and non-smoking-related cancers. PD was positively associated with melanoma. These results provide evidence for further investigations for possible mechanistic associations between PD and cancer. PROSPERO REGISTRATION NUMBER CRD42020162103.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Nutritional Science, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - David Guarin
- Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Xiqun Chen
- Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Xiang Gao
- Nutritional Science, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
24
|
Day JO, Mullin S. The Genetics of Parkinson's Disease and Implications for Clinical Practice. Genes (Basel) 2021; 12:genes12071006. [PMID: 34208795 PMCID: PMC8304082 DOI: 10.3390/genes12071006] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/21/2021] [Accepted: 06/28/2021] [Indexed: 12/17/2022] Open
Abstract
The genetic landscape of Parkinson’s disease (PD) is characterised by rare high penetrance pathogenic variants causing familial disease, genetic risk factor variants driving PD risk in a significant minority in PD cases and high frequency, low penetrance variants, which contribute a small increase of the risk of developing sporadic PD. This knowledge has the potential to have a major impact in the clinical care of people with PD. We summarise these genetic influences and discuss the implications for therapeutics and clinical trial design.
Collapse
Affiliation(s)
- Jacob Oliver Day
- Faculty of Health, University of Plymouth, Plymouth PL4 8AA, UK;
| | - Stephen Mullin
- Faculty of Health, University of Plymouth, Plymouth PL4 8AA, UK;
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, London WC1N 3BG, UK
- Correspondence:
| |
Collapse
|
25
|
Sarangi SC, Sopory P, Reeta KH. Chronic Neurological Disorders: Genetic and Epigenetic Markers for Monitoring of Pharmacotherapy. Neurol India 2021; 69:252-259. [PMID: 33904433 DOI: 10.4103/0028-3886.314522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Introduction Chronic neurological diseases are a major cause of mortality and morbidity in the world. With increasing life expectancy in the developing world, the incidence and prevalence of these diseases are predicted to rise even further. This has also contributed to an increase in disability-adjusted life years (DALYs) for noncommunicable diseases. Treatment for such diseases also poses a challenge with multiple genetic and epigenetic factors leading to a varied outcome. Personalization of treatment is one way that treatment outcome/prognosis of disease can be improved, and pharmacogenomics plays a significant role in this context. Methodology This article reviewed the evidence pertaining to the association of genetic and epigenetic markers with major neurological disorders like multiple sclerosis (MS), Alzheimer's disease (AD), and Parkinson's disease (PD), which are a major source of burden among neurological disorders. Types of studies included are peer-reviewed original research articles from the PubMed database (1999-2018). Results This study compiled data regarding specific genetic and epigenetic markers with a significant correlation between the clinical diagnosis of the disease and prognosis of therapy from 65 studies. In a single platform, this review highlights the clues to some vital questions, such as why interferon beta (IFN-β) therapy fails to improve symptoms in all MS patients? why cholinesterase inhibitors fail to improve cognitive impairment in a subset of people suffering from AD? or why some individuals on levodopa (L-DOPA) for PD suffer from side-effects ranging from dyskinesia to hallucination while others do not? Conclusion This article summarizes the genetic and epigenetic factors that may either require monitoring or help in deciding future pharmacotherapy in a patient suffering from MS, AD, and PD. As the health care system develops and reaches newer heights, we expect more and more of these biomarkers to be used as pharmacotherapeutic outcome indicators.
Collapse
Affiliation(s)
| | - Pranav Sopory
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - K H Reeta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
26
|
Goveas L, Mutez E, Chartier-Harlin MC, Taymans JM. Mind the Gap: LRRK2 Phenotypes in the Clinic vs. in Patient Cells. Cells 2021; 10:981. [PMID: 33922322 PMCID: PMC8145309 DOI: 10.3390/cells10050981] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/14/2022] Open
Abstract
Mutations in the Parkinson's disease (PD) protein Leucine Rich Repeat Kinase 2 (LRRK2) have been under study for more than 15 years and our understanding of the cellular phenotypes for the pathogenic mutant forms of LRRK2 has significantly advanced. In parallel to research on LRRK2 mutations in experimental systems, clinical characterization of patients carrying LRRK2 mutations has advanced, as has the analysis of cells that are derived from these patients, including fibroblasts, blood-derived cells, or cells rendered pluripotent. Under the hypothesis that patient clinical phenotypes are a consequence of a cascade of underlying molecular mechanisms gone astray, we currently have a unique opportunity to compare findings from patients and patient-derived cells to ask the question of whether the clinical phenotype of LRRK2 Parkinson's disease and cellular phenotypes of LRRK2 patient-derived cells may be mutually informative. In this review, we aim to summarize the available information on phenotypes of LRRK2 mutations in the clinic, in patient-derived cells, and in experimental models in order to better understand the relationship between the three at the molecular and cellular levels and identify trends and gaps in correlating the data.
Collapse
Affiliation(s)
- Liesel Goveas
- UMR-S 1172—LilNCog—Lille Neuroscience & Cognition, Université de Lille, Inserm, CHU Lille, F-59000 Lille, France; (L.G.); (E.M.)
| | - Eugénie Mutez
- UMR-S 1172—LilNCog—Lille Neuroscience & Cognition, Université de Lille, Inserm, CHU Lille, F-59000 Lille, France; (L.G.); (E.M.)
- Neurology and Movement Disorders Department, CHU Lille University Hospital, F-59000 Lille, France
| | - Marie-Christine Chartier-Harlin
- UMR-S 1172—LilNCog—Lille Neuroscience & Cognition, Université de Lille, Inserm, CHU Lille, F-59000 Lille, France; (L.G.); (E.M.)
| | - Jean-Marc Taymans
- UMR-S 1172—LilNCog—Lille Neuroscience & Cognition, Université de Lille, Inserm, CHU Lille, F-59000 Lille, France; (L.G.); (E.M.)
| |
Collapse
|
27
|
Chittoor-Vinod VG, Nichols RJ, Schüle B. Genetic and Environmental Factors Influence the Pleomorphy of LRRK2 Parkinsonism. Int J Mol Sci 2021; 22:1045. [PMID: 33494262 PMCID: PMC7864502 DOI: 10.3390/ijms22031045] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/16/2021] [Accepted: 01/17/2021] [Indexed: 12/25/2022] Open
Abstract
Missense mutations in the LRRK2 gene were first identified as a pathogenic cause of Parkinson's disease (PD) in 2004. Soon thereafter, a founder mutation in LRRK2, p.G2019S (rs34637584), was described, and it is now estimated that there are approximately 100,000 people worldwide carrying this risk variant. While the clinical presentation of LRRK2 parkinsonism has been largely indistinguishable from sporadic PD, disease penetrance and age at onset can be quite variable. In addition, its neuropathological features span a wide range from nigrostriatal loss with Lewy body pathology, lack thereof, or atypical neuropathology, including a large proportion of cases with concomitant Alzheimer's pathology, hailing LRRK2 parkinsonism as the "Rosetta stone" of parkinsonian disorders, which provides clues to an understanding of the different neuropathological trajectories. These differences may result from interactions between the LRRK2 mutant protein and other proteins or environmental factors that modify LRRK2 function and, thereby, influence pathobiology. This review explores how potential genetic and biochemical modifiers of LRRK2 function may contribute to the onset and clinical presentation of LRRK2 parkinsonism. We review which genetic modifiers of LRRK2 influence clinical symptoms, age at onset, and penetrance, what LRRK2 mutations are associated with pleomorphic LRRK2 neuropathology, and which environmental modifiers can augment LRRK2 mutant pathophysiology. Understanding how LRRK2 function is influenced and modulated by other interactors and environmental factors-either increasing toxicity or providing resilience-will inform targeted therapeutic development in the years to come. This will allow the development of disease-modifying therapies for PD- and LRRK2-related neurodegeneration.
Collapse
Affiliation(s)
| | - R. Jeremy Nichols
- Department Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Birgitt Schüle
- Department Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA;
| |
Collapse
|
28
|
Abstract
A number of studies reported the possible differences between men and women in movement disorders. Evidence shows that estrogens may have a neuroprotective effect and may modulate the neurodevelopment of the different brain structures. Movement disorders including Parkinson's disease, dementia with Lewy body, Huntington's disease, Tourette's syndrome, and dystonia among others display significant clinical differences between sexes, with structural differences in the dopaminergic pathways between men and women. Here we summarize the most relevant clinical aspects of some of the most common movement disorders, highlighting the differences in disease onset, clinical presentation, therapy, and outcomes. Increased recognition of these differences may help physicians better understand the pathophysiology of these conditions and provide a tailored therapeutic approach.
Collapse
Affiliation(s)
- Pierpaolo Turcano
- Department of Neurology, Mayo Clinic, Jacksonville, FL, United States.
| | - Rodolfo Savica
- Department of Neurology and Health Science Research, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
29
|
Lesage S, Houot M, Mangone G, Tesson C, Bertrand H, Forlani S, Anheim M, Brefel-Courbon C, Broussolle E, Thobois S, Damier P, Durif F, Roze E, Tison F, Grabli D, Ory-Magne F, Degos B, Viallet F, Cormier-Dequaire F, Ouvrard-Hernandez AM, Vidailhet M, Lohmann E, Singleton A, Corvol JC, Brice A. Genetic and Phenotypic Basis of Autosomal Dominant Parkinson's Disease in a Large Multi-Center Cohort. Front Neurol 2020; 11:682. [PMID: 32849182 PMCID: PMC7399219 DOI: 10.3389/fneur.2020.00682] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/08/2020] [Indexed: 12/20/2022] Open
Abstract
LRRK2, SNCA, and VPS35 are unequivocally associated with autosomal dominant Parkinson's disease (PD). We evaluated the prevalence of LRRK2, SNCA, and VPS35 mutations and associated clinical features in a large French multi-center cohort of PD patients. Demographic and clinical data were collected for 1,805 index cases (592 with autosomal dominant inheritance and 1,213 isolated cases) since 1990. All probands were screened with TaqMan assays for LRRK2 Gly2019Ser. In the absence of this mutation, the coding sequences of the three genes were analyzed by Sanger sequencing and/or next-generation sequencing. The data for the three genes were analyzed according to age at onset, family history, ethnic origin and clinical features. We identified 160 index cases (8.9%) with known pathogenic variants: 138 with pathogenic LRRK2 variants (7.6%), including 136 with the Gly2019Ser mutation, 19 with SNCA point mutations or genomic rearrangements (1.1%), and three with the VPS35 Asp620Asn mutation (0.16%). Mutation frequencies were higher in familial than isolated cases, consistent with autosomal dominant inheritance (12.0 vs. 7.3%; OR 1.7, 95% CI [1.2-2.4], p = 0.001). PD patients with LRRK2 variants were more likely to have higher rates of late-onset PD (>50 years; OR 1.5, 95% CI [1.0-2.1], p = 0.03), whereas those with SNCA mutations tended to have earlier age at onset disease (≤ 50 years, p = 0.06). The clinical features of LRRK2 carriers and those without any pathogenic variants in known PD-associated genes were similar. The likelihood of detecting disease-causing mutations was higher in cases compatible with autosomal dominant inheritance.
Collapse
Affiliation(s)
- Suzanne Lesage
- Sorbonne Université, Unité Mixte de Recherche (UMR) 1127, Paris, France
- Unité de Recherche U1127 à l'Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Unité de Recherche Unité Mixte de Recherche (UMR) 7225 au Centre National de la Recherche Scientifique (CNRS), Paris, France
- Institut du Cerveau (ICM), Paris, France
| | - Marion Houot
- Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Centre d'Excellence sur les Maladies Neurodégénératives (CoEN), Assistance Publique – Hôpitaux de Paris (AP-HP), Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, Université Paris 6, Paris, France
- Centre d'Investigation Clinique Pitié Neurosciences CIC-1422, Paris, France
| | - Graziella Mangone
- Sorbonne Université, Unité Mixte de Recherche (UMR) 1127, Paris, France
- Unité de Recherche U1127 à l'Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Unité de Recherche Unité Mixte de Recherche (UMR) 7225 au Centre National de la Recherche Scientifique (CNRS), Paris, France
- Institut du Cerveau (ICM), Paris, France
- Centre d'Investigation Clinique Pitié Neurosciences CIC-1422, Paris, France
| | - Christelle Tesson
- Sorbonne Université, Unité Mixte de Recherche (UMR) 1127, Paris, France
- Unité de Recherche U1127 à l'Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Unité de Recherche Unité Mixte de Recherche (UMR) 7225 au Centre National de la Recherche Scientifique (CNRS), Paris, France
- Institut du Cerveau (ICM), Paris, France
| | - Hélène Bertrand
- Sorbonne Université, Unité Mixte de Recherche (UMR) 1127, Paris, France
- Unité de Recherche U1127 à l'Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Unité de Recherche Unité Mixte de Recherche (UMR) 7225 au Centre National de la Recherche Scientifique (CNRS), Paris, France
- Institut du Cerveau (ICM), Paris, France
| | - Sylvie Forlani
- Sorbonne Université, Unité Mixte de Recherche (UMR) 1127, Paris, France
- Unité de Recherche U1127 à l'Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Unité de Recherche Unité Mixte de Recherche (UMR) 7225 au Centre National de la Recherche Scientifique (CNRS), Paris, France
- Institut du Cerveau (ICM), Paris, France
| | - Mathieu Anheim
- Département de Neurologie aux Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Christine Brefel-Courbon
- Service de Pharmacologie Clinique, Faculté de Médecine, Hôpital Universitaire, Toulouse, France
- Service de Neurologie B8, Hôpital Pierre Paul Riquet, Hôpital Universitaire, Toulouse, France
| | - Emmanuel Broussolle
- Université de Lyon, Institut des Sciences Cognitives Marc-Jeannerod, Unité Mixte de Recherche (UMR) 5229, Centre National de la Recherche Scientifique (CNRS), Bron, France
- Hospices Civils de Lyon, Hôpital Neurologique Pierre-Wertheimer, Département de Neurologie C, Bron, France
- Université de Lyon, Faculté de Médecine Lyon-Sud Charles-Mérieux, Oullins, France
| | - Stéphane Thobois
- Université de Lyon, Institut des Sciences Cognitives Marc-Jeannerod, Unité Mixte de Recherche (UMR) 5229, Centre National de la Recherche Scientifique (CNRS), Bron, France
- Hospices Civils de Lyon, Hôpital Neurologique Pierre-Wertheimer, Département de Neurologie C, Bron, France
- Université de Lyon, Faculté de Médecine Lyon-Sud Charles-Mérieux, Oullins, France
| | - Philippe Damier
- Centre Hospitalier Universitaire de Nantes, Centre d'Investigation Clinique, Nantes, France
| | - Franck Durif
- Département de Neurologie A, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Emmanuel Roze
- Sorbonne Université, Unité Mixte de Recherche (UMR) 1127, Paris, France
- Unité de Recherche U1127 à l'Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Unité de Recherche Unité Mixte de Recherche (UMR) 7225 au Centre National de la Recherche Scientifique (CNRS), Paris, France
- Institut du Cerveau (ICM), Paris, France
- Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - François Tison
- Institut des Maladies Neurodégénératives, Centre Hospitalier Universitaire et Université de Bordeaux, Bordeaux, France
| | - David Grabli
- Sorbonne Université, Unité Mixte de Recherche (UMR) 1127, Paris, France
- Unité de Recherche U1127 à l'Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Unité de Recherche Unité Mixte de Recherche (UMR) 7225 au Centre National de la Recherche Scientifique (CNRS), Paris, France
- Institut du Cerveau (ICM), Paris, France
- Centre d'Investigation Clinique Pitié Neurosciences CIC-1422, Paris, France
| | - Fabienne Ory-Magne
- Centre de Neuroimagerie de Toulouse, Université de Toulouse - Institut National de la Santé et de la Recherche Médicale (INSERM) - Université de Toulouse, Toulouse, France
- Centre des Neurosciences, Hôpital Universitaire de Toulouse, Toulouse, France
| | - Bertrand Degos
- Unité de Neurologie, Hôpital Universitaire Avicenne, Hôpitaux Universitaires de Paris-Seine Saint Denis, Assistance Publique – Hôpitaux de Paris (AP-HP), Sorbonne Paris Nord, Bobigny, France
- Equipe Dynamique et Physiopathologie des Réseaux Neuronaux, Centre pour la Recherche Interdisciplinaire en Biologie, Collège de France, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7241, Institut National de la Santé et de la Recherche Médicale (INSERM) U1050, Labex MemoLife, Paris, France
| | - François Viallet
- Département de Neurologie, Centre Hospitalier Intercommunal d'Aix-Pertuis, Aix-en-Provence, France
- Laboratoire Parole et Langage, Unité Mixte de Recherche (UMR) 7309, Centre National de la Recherche Scientifique (CNRS) et Université d'Aix-Marseille, Aix-en-Provence, France
| | - Florence Cormier-Dequaire
- Sorbonne Université, Unité Mixte de Recherche (UMR) 1127, Paris, France
- Unité de Recherche U1127 à l'Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Unité de Recherche Unité Mixte de Recherche (UMR) 7225 au Centre National de la Recherche Scientifique (CNRS), Paris, France
- Institut du Cerveau (ICM), Paris, France
- Centre d'Investigation Clinique Pitié Neurosciences CIC-1422, Paris, France
| | | | - Marie Vidailhet
- Sorbonne Université, Unité Mixte de Recherche (UMR) 1127, Paris, France
- Unité de Recherche U1127 à l'Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Unité de Recherche Unité Mixte de Recherche (UMR) 7225 au Centre National de la Recherche Scientifique (CNRS), Paris, France
- Institut du Cerveau (ICM), Paris, France
- Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Ebba Lohmann
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
| | - Jean-Christophe Corvol
- Sorbonne Université, Unité Mixte de Recherche (UMR) 1127, Paris, France
- Unité de Recherche U1127 à l'Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Unité de Recherche Unité Mixte de Recherche (UMR) 7225 au Centre National de la Recherche Scientifique (CNRS), Paris, France
- Institut du Cerveau (ICM), Paris, France
- Centre d'Investigation Clinique Pitié Neurosciences CIC-1422, Paris, France
| | - Alexis Brice
- Sorbonne Université, Unité Mixte de Recherche (UMR) 1127, Paris, France
- Unité de Recherche U1127 à l'Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Unité de Recherche Unité Mixte de Recherche (UMR) 7225 au Centre National de la Recherche Scientifique (CNRS), Paris, France
- Institut du Cerveau (ICM), Paris, France
| | | |
Collapse
|
30
|
Polissidis A, Petropoulou-Vathi L, Nakos-Bimpos M, Rideout HJ. The Future of Targeted Gene-Based Treatment Strategies and Biomarkers in Parkinson's Disease. Biomolecules 2020; 10:E912. [PMID: 32560161 PMCID: PMC7355671 DOI: 10.3390/biom10060912] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 12/13/2022] Open
Abstract
Biomarkers and disease-modifying therapies are both urgent unmet medical needs in the treatment of Parkinson's disease (PD) and must be developed concurrently because of their interdependent relationship: biomarkers for the early detection of disease (i.e., prior to overt neurodegeneration) are necessary in order for patients to receive maximal therapeutic benefit and vice versa; disease-modifying therapies must become available for patients whose potential for disease diagnosis and prognosis can be predicted with biomarkers. This review provides an overview of the milestones achieved to date in the therapeutic strategy development of disease-modifying therapies and biomarkers for PD, with a focus on the most common and advanced genetically linked targets alpha-synuclein (SNCA), leucine-rich repeat kinase-2 (LRRK2) and glucocerebrosidase (GBA1). Furthermore, we discuss the convergence of the different pathways and the importance of patient stratification and how these advances may apply more broadly to idiopathic PD. The heterogeneity of PD poses a challenge for therapeutic and biomarker development, however, the one gene- one target approach has brought us closer than ever before to an unprecedented number of clinical trials and biomarker advancements.
Collapse
Affiliation(s)
| | | | | | - Hardy J. Rideout
- Laboratory of Neurodegenerative Diseases, Centre for Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (A.P.); (L.P.-V.); (M.N.-B.)
| |
Collapse
|
31
|
Cerri S, Mus L, Blandini F. Parkinson's Disease in Women and Men: What's the Difference? JOURNAL OF PARKINSONS DISEASE 2020; 9:501-515. [PMID: 31282427 PMCID: PMC6700650 DOI: 10.3233/jpd-191683] [Citation(s) in RCA: 376] [Impact Index Per Article: 75.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Increasing evidence points to biological sex as an important factor in the development and phenotypical expression of Parkinson’s disease (PD). Risk of developing PD is twice as high in men than women, but women have a higher mortality rate and faster progression of the disease. Moreover, motor and nonmotor symptoms, response to treatments and disease risk factors differ between women and men. Altogether, sex-related differences in PD support the idea that disease development might involve distinct pathogenic mechanisms (or the same mechanism but in a different way) in male and female patients. This review summarizes the most recent knowledge concerning differences between women and men in PD clinical features, risk factors, response to treatments and mechanisms underlying the disease pathophysiology. Unraveling how the pathology differently affect the two sexes might allow the development of tailored interventions and the design of innovative programs that meet the distinct needs of men and women, improving patient care.
Collapse
Affiliation(s)
- Silvia Cerri
- Laboratory of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, Pavia, Italy
| | - Liudmila Mus
- Laboratory of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, Pavia, Italy
| | - Fabio Blandini
- Laboratory of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
32
|
Aasly JO. Long-Term Outcomes of Genetic Parkinson's Disease. J Mov Disord 2020; 13:81-96. [PMID: 32498494 PMCID: PMC7280945 DOI: 10.14802/jmd.19080] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/23/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disorder that affects 1–2% of people by the age of 70 years. Age is the most important risk factor, and most cases are sporadic without any known environmental or genetic causes. Since the late 1990s, mutations in the genes SNCA, PRKN, LRRK2, PINK1, DJ-1, VPS35, and GBA have been shown to be important risk factors for PD. In addition, common variants with small effect sizes are now recognized to modulate the risk for PD. Most studies in genetic PD have focused on finding new genes, but few have studied the long-term outcome of patients with the specific genetic PD forms. Patients with known genetic PD have now been followed for more than 20 years, and we see that they may have distinct and different prognoses. New therapeutic possibilities are emerging based on the genetic cause underlying the disease. Future medication may be based on the pathophysiology individualized to the patient’s genetic background. The challenge is to find the biological consequences of different genetic variants. In this review, the clinical patterns and long-term prognoses of the most common genetic PD variants are presented.
Collapse
Affiliation(s)
- Jan O Aasly
- Department of Neurology, St. Olav's Hospital, Trondheim, Norway.,Department of Neuroscience, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
33
|
Abstract
In a range of neurological conditions, including movement disorders, sex-related differences are emerging not only in brain anatomy and function, but also in pathogenesis, clinical features and response to treatment. In Parkinson disease (PD), for example, oestrogens can influence the severity of motor symptoms, whereas elevation of androgens can exacerbate tic disorders. Nevertheless, the real impact of sex differences in movement disorders remains under-recognized. In this article, we provide an up-to-date review of sex-related differences in PD and the most common hyperkinetic movement disorders, namely, essential tremor, dystonia, Huntington disease and other chorea syndromes, and Tourette syndrome and other chronic tic disorders. We highlight the most relevant clinical aspects of movement disorders that differ between men and women. Increased recognition of these differences and their impact on patient care could aid the development of tailored approaches to the management of movement disorders and enable the optimization of preclinical research and clinical studies.
Collapse
|
34
|
Chen W, Yan X, Lv H, Liu Y, He Z, Luo X. Gender differences in prevalence of LRRK2-associated Parkinson disease: A meta-analysis of observational studies. Neurosci Lett 2019; 715:134609. [PMID: 31698024 DOI: 10.1016/j.neulet.2019.134609] [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: 07/18/2019] [Revised: 10/21/2019] [Accepted: 11/01/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND The gender effect in the prevalence of leucine-rich repeat kinase 2 (LRRK2) associated Parkinson disease (PD) remains controversial. Herein, we conducted a meta-analysis to investigate the gender effect among these patients. METHODS PubMed and EMBASE databases were searched to identify the potential related studies published before December 2017. Case-control studies with separated data of sex and mutation status were included in further analyses. We pooled relative risk (RR) using fixed-effect model. The publication bias and sensitivity analyses were also performed. RESULTS Sixty-four studies with 32452 patients diagnosed with PD were included. Higher prevalence of female patients with LRRK2-associated PD was observed with a pooled RR of 1.22 (95% CI 1.14-1.30, P<0.001). Further subgroup analyses showed that higher prevalence of female patients was only obtained in G2019S mutation patients (RR = 1.32, 95% CI 1.23-1.43, P<0.001), but not in G2385R variant patients (RR = 1.03, 95% CI 0.91-1.17, P = 0.651). No significant heterogeneity and publication bias were observed in additional analyses. CONCLUSIONS Higher female prevalence of LRRK2 mutation suggests roles of gender-related risk factors in PD patients, especially who carried G2019S mutation. Contrary to idiopathic PD, no sex difference was observed in prevalence of patients carried G2385R variant.
Collapse
Affiliation(s)
- Weiyao Chen
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Xuejing Yan
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Hong Lv
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Yang Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Zhiyi He
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China.
| | - Xiaoguang Luo
- Shen Zhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen 518020, Guangdong, People's Republic of China.
| |
Collapse
|
35
|
Frequency of the LRRK2 G2019S mutation in South African patients with Parkinson’s disease. Neurogenetics 2019; 20:215-218. [DOI: 10.1007/s10048-019-00588-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/27/2019] [Indexed: 12/14/2022]
|
36
|
Ciani M, Bonvicini C, Scassellati C, Carrara M, Maj C, Fostinelli S, Binetti G, Ghidoni R, Benussi L. The Missing Heritability of Sporadic Frontotemporal Dementia: New Insights from Rare Variants in Neurodegenerative Candidate Genes. Int J Mol Sci 2019; 20:ijms20163903. [PMID: 31405128 PMCID: PMC6721049 DOI: 10.3390/ijms20163903] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/02/2019] [Accepted: 08/08/2019] [Indexed: 12/13/2022] Open
Abstract
Frontotemporal dementia (FTD) is a common form of dementia among early-onset cases. Several genetic factors for FTD have been revealed, but a large proportion of FTD cases still have an unidentified genetic origin. Recent studies highlighted common pathobiological mechanisms among neurodegenerative diseases. In the present study, we investigated a panel of candidate genes, previously described to be associated with FTD and/or other neurodegenerative diseases by targeted next generation sequencing (NGS). We focused our study on sporadic FTD (sFTD), devoid of disease-causing mutations in GRN, MAPT and C9orf72. Since genetic factors have a substantially higher pathogenetic contribution in early onset patients than in late onset dementia, we selected patients with early onset (<65 years). Our study revealed that, in 50% of patients, rare missense potentially pathogenetic variants in genes previously associated with Alzheimer's disease, Parkinson disease, amyotrophic lateral sclerosis and Lewy body dementia (GBA, ABCA7, PARK7, FUS, SORL1, LRRK2, ALS2), confirming genetic pleiotropy in neurodegeneration. In parallel, a synergic genetic effect on FTD is suggested by the presence of variants in five different genes in one single patient. Further studies employing genome-wide approaches might highlight pathogenic variants in novel genes that explain the still missing heritability of FTD.
Collapse
Affiliation(s)
- Miriam Ciani
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Cristian Bonvicini
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Catia Scassellati
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Matteo Carrara
- Service of Statistics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Carlo Maj
- Institute of Genomic Statistics and Bioinformatics, University of Bonn, 53127 Bonn, Germany
| | - Silvia Fostinelli
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Giuliano Binetti
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy.
| |
Collapse
|
37
|
Shu L, Zhang Y, Sun Q, Pan H, Tang B. A Comprehensive Analysis of Population Differences in LRRK2 Variant Distribution in Parkinson's Disease. Front Aging Neurosci 2019; 11:13. [PMID: 30760999 PMCID: PMC6363667 DOI: 10.3389/fnagi.2019.00013] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/14/2019] [Indexed: 11/13/2022] Open
Abstract
Background:LRRK2 variants have been demonstrated to have distinct distributions in different populations. However, researchers have thus far chosen to focus on relatively few variants, such as R1628P, G2019S, and G2385R. We therefore investigated the relationship between common LRRK2 variants and PD risk in various populations. Methods: Using a set of strict inclusion criteria, six databases were searched, resulting in the selection of 94 articles covering 49,299 cases and 47,319 controls for final pooled analysis and frequency analysis. Subgroup analysis were done for Africans, European/West Asians, Hispanics, East Asians, and mixed populations. Statistical analysis was carried out using the Mantel-Haenszel approach to determine the relationship between common LRRK2 variants and PD risk, with the significance level set at p < 0.05. Results: In the absence of obvious heterogeneities and publication biases among the included studies, we concluded that A419V, R1441C/G/H, R1628P, G2019S, and G2385R were associated with increased PD risk (p: 0.001, 0.0004, < 0.00001, < 0.00001, and < 0.00001, respectively), while R1398H was associated with decreased risk (p: < 0.00001). In East Asian populations, A419V, R1628P, and G2385R increased risk (p: 0.001, < 0.00001, < 0.00001), while R1398H had the opposite effect (p: 0.0005). G2019S increased PD risk in both European/West Asian and mixed populations (p: < 0.00001, < 0.00001), while R1441C/G/H increased risk in European/West Asian populations only (p: 0.0004). Conclusions: We demonstrated that LRRK2 variant distribution is different among various populations, which should inform decisions regarding the development of future genetic screening strategies.
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
| | - Qiying Sun
- Department of Geriatrics, 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
| | - Hongxu Pan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 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
| |
Collapse
|