1
|
Xiong R, Fleming E, Caldwell R, Vernon SD, Kozhaya L, Gunter C, Bateman L, Unutmaz D, Oh J. BioMapAI: Artificial Intelligence Multi-Omics Modeling of Myalgic Encephalomyelitis / Chronic Fatigue Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600378. [PMID: 38979186 PMCID: PMC11230215 DOI: 10.1101/2024.06.24.600378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Chronic diseases like ME/CFS and long COVID exhibit high heterogeneity with multifactorial etiology and progression, complicating diagnosis and treatment. To address this, we developed BioMapAI, an explainable Deep Learning framework using the richest longitudinal multi-'omics dataset for ME/CFS to date. This dataset includes gut metagenomics, plasma metabolome, immune profiling, blood labs, and clinical symptoms. By connecting multi-'omics to asymptom matrix, BioMapAI identified both disease- and symptom-specific biomarkers, reconstructed symptoms, and achieved state-of-the-art precision in disease classification. We also created the first connectivity map of these 'omics in both healthy and disease states and revealed how microbiome-immune-metabolome crosstalk shifted from healthy to ME/CFS. Thus, we proposed several innovative mechanistic hypotheses for ME/CFS: Disrupted microbial functions - SCFA (butyrate), BCAA (amino acid), tryptophan, benzoate - lost connection with plasma lipids and bile acids, and activated inflammatory and mucosal immune cells (MAIT, γδT cells) with INFγ and GzA secretion. These abnormal dynamics are linked to key disease symptoms, including gastrointestinal issues, fatigue, and sleep problems.
Collapse
Affiliation(s)
- Ruoyun Xiong
- The Jackson Laboratory, Farmington, Connecticut, USA. 06032
- The University of Connecticut Health Center, Farmington, Connecticut, USA. 06030
| | | | - Ryan Caldwell
- The Jackson Laboratory, Farmington, Connecticut, USA. 06032
| | | | - Lina Kozhaya
- The Jackson Laboratory, Farmington, Connecticut, USA. 06032
| | - Courtney Gunter
- The Jackson Laboratory, Farmington, Connecticut, USA. 06032
- The University of Connecticut Health Center, Farmington, Connecticut, USA. 06030
| | | | - Derya Unutmaz
- The Jackson Laboratory, Farmington, Connecticut, USA. 06032
| | - Julia Oh
- The Jackson Laboratory, Farmington, Connecticut, USA. 06032
| |
Collapse
|
2
|
Mata I, Salles P, Cornejo-Olivas M, Saffie P, Ross OA, Reed X, Bandres-Ciga S. LRRK2: Genetic mechanisms vs genetic subtypes. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:133-154. [PMID: 36803807 DOI: 10.1016/b978-0-323-85555-6.00018-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
In 2004, the identification of pathogenic variants in the LRRK2 gene across several families with autosomal dominant late-onset Parkinson's disease (PD) revolutionized our understanding of the role of genetics in PD. Previous beliefs that genetics in PD was limited to rare early-onset or familial forms of the disease were quickly dispelled. Currently, we recognize LRRK2 p.G2019S as the most common genetic cause of both sporadic and familial PD, with more than 100,000 affected carriers across the globe. The frequency of LRRK2 p.G2019S is also highly variable across populations, with some regions of Asian or Latin America reporting close to 0%, contrasting to Ashkenazi Jews or North African Berbers reporting up to 13% and 40%, respectively. Patients with LRRK2 pathogenic variants are clinically and pathologically heterogeneous, highlighting the age-related variable penetrance that also characterizes LRRK2-related disease. Indeed, the majority of patients with LRRK2-related disease are characterized by a relatively mild Parkinsonism with less motor symptoms with variable presence of α-synuclein and/or tau aggregates, with pathologic pleomorphism widely described. At a functional cellular level, it is likely that pathogenic variants mediate a toxic gain-of-function of the LRRK2 protein resulting in increased kinase activity perhaps in a cell-specific manner; by contrast, some LRRK2 variants appear to be protective reducing PD risk by decreasing the kinase activity. Therefore, employing this information to define appropriate patient populations for clinical trials of targeted kinase LRRK2 inhibition strategies is very promising and demonstrates a potential future application for PD using precision medicine.
Collapse
Affiliation(s)
- Ignacio Mata
- Genomic Medicine Institute (GMI), Cleveland Clinic, Cleveland, OH, United States.
| | - Philippe Salles
- Corporación Centro de Trastornos del Movimiento (CETRAM), Lo Espejo, Santiago, Chile
| | - Mario Cornejo-Olivas
- Neurogenetics Research Center, Instituto Nacional de Ciencias Neurológicas, Lima, Peru
| | - Paula Saffie
- Corporación Centro de Trastornos del Movimiento (CETRAM), Lo Espejo, Santiago, Chile
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Xylena Reed
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
| | - Sara Bandres-Ciga
- Laboratory of Neurogenetics and Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
3
|
Cognition as a mediator for gait and balance impairments in GBA-related Parkinson's disease. NPJ Parkinsons Dis 2022; 8:78. [PMID: 35725575 PMCID: PMC9209443 DOI: 10.1038/s41531-022-00344-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 05/26/2022] [Indexed: 11/18/2022] Open
Abstract
The extent to which the heterogeneity of gait and balance problems in PD may be explained by genetic variation is unknown. Variants in the glucocerebrosidase (GBA) gene are the strongest known genetic risk factor for PD and are associated with greater motor and cognitive severity. However, the impact of GBA variants on comprehensive measures of gait and balance and their relationship to cognition remains unknown. We aimed to determine differences in gait and balance impairments in those with and without GBA variants (mutation carriers and E326K polymorphism) and explore direct and indirect effects of GBA status on gait, balance, and cognition. 332 participants, 43 of whom had GBA variants, were recruited. Participants completed a comprehensive, objective assessment of gait and standing balance using body-worn inertial sensors. Group differences in gait and balance between PD with and without GBA variants were assessed with linear regression, adjusting for age, gender, clinical testing site, disease duration, and apolipoprotein E (APOE) ɛ4 status. Structural equation modeling (SEM) explored direct relationships between GBA status and gait and balance and indirect relationships between GBA status and gait and balance via cognition. The GBA variant group had more impaired gait (pace and variability) and balance (sway area/jerk and sway velocity), than the non-GBA variant group. SEM demonstrated cognition as a mediator of GBA status on gait and balance. The close relationships among GBA, gait/balance, and cognition suggest potential for novel therapeutics to target the GBA pathway and/or cognition to improve mobility in PD GBA variants.
Collapse
|
4
|
Peterschmitt MJ, Saiki H, Hatano T, Gasser T, Isaacson SH, Gaemers SJM, Minini P, Saubadu S, Sharma J, Walbillic S, Alcalay RN, Cutter G, Hattori N, Höglinger GU, Marek K, Schapira AHV, Scherzer CR, Simuni T, Giladi N, Sardi SP, Fischer TZ. Safety, Pharmacokinetics, and Pharmacodynamics of Oral Venglustat in Patients with Parkinson's Disease and a GBA Mutation: Results from Part 1 of the Randomized, Double-Blinded, Placebo-Controlled MOVES-PD Trial. JOURNAL OF PARKINSON'S DISEASE 2021; 12:557-570. [PMID: 34897099 PMCID: PMC8925113 DOI: 10.3233/jpd-212714] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Glucocerebrosidase gene (GBA) mutations influence risk and prognosis of Parkinson’s disease (PD), possibly through accumulation of glycosphingolipids, including glucosylceramide (GL-1). Venglustat is a novel, brain penetrant glucosylceramide synthase inhibitor. Objective: Evaluate venglustat pharmacology, safety, and tolerability in patients with PD and GBA mutations (GBA-PD). Methods: Part 1 of the phase 2 MOVES-PD trial (NCT02906020) was a randomized, double-blinded, placebo-controlled, dose-escalation study performed in six countries. Eligible participants included Japanese and non-Japanese patients aged 18–80 years with PD diagnosis and heterozygous GBA mutation. Participants were randomized to three doses of once-daily oral venglustat or placebo and were followed up to 36 weeks (Japanese participants: 52 weeks). Primary endpoint was venglustat safety and tolerability versus placebo. Secondary and exploratory endpoints included venglustat pharmacokinetics and pharmacodynamics. Results: Participants (N = 29) received venglustat (Japanese, n = 9; non-Japanese, n = 13) or placebo (n = 3; n = 4). Eight (89%) Japanese and 12 (92%) non-Japanese venglustat-treated participants experienced at least one adverse event (AE) versus two (67%) and four (100%) participants from the respective placebo groups. Most AEs were mild or moderate; no serious AEs or deaths occurred. Two venglustat-treated non-Japanese participants discontinued due to AEs (confusional state and panic attack). Over 4 weeks, venglustat exposure in plasma and cerebrospinal fluid (CSF) increased, and GL-1 levels in plasma and CSF decreased, both in a dose-dependent manner. At the highest dose, CSF GL-1 decreased by 72.0% in Japanese and 74.3% in non-Japanese participants. Conclusion: Venglustat showed favorable safety and tolerability in MOVES-PD Part 1 and target engagement was achieved in CSF.
Collapse
Affiliation(s)
| | - Hidemoto Saiki
- Kitano Hospital, The Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | | | - Thomas Gasser
- Neurologische Universitätsklinik, Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Stuart H Isaacson
- Parkinson's Disease and Movement Disorders Center, Boca Raton, FL, USA
| | | | | | | | | | | | - Roy N Alcalay
- Department of Neurology and the Taub Institute, Columbia University Medical Center, New York, NY, USA
| | - Gary Cutter
- University of Alabama at Birmingham, School of Public Health, Birmingham, AL, USA
| | | | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, and Department of Neurology, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany.,Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Kenneth Marek
- Institute for Neurodegenerative Disorders, New Haven, CT, USA
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | | | - Nir Giladi
- Neurological Institute, Tel Aviv Medical Center, Sackler School of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | | | | | | |
Collapse
|
5
|
Menozzi E, Schapira AHV. Exploring the Genotype-Phenotype Correlation in GBA-Parkinson Disease: Clinical Aspects, Biomarkers, and Potential Modifiers. Front Neurol 2021; 12:694764. [PMID: 34248830 PMCID: PMC8264189 DOI: 10.3389/fneur.2021.694764] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/18/2021] [Indexed: 01/01/2023] Open
Abstract
Variants in the glucocerebrosidase (GBA) gene are the most common genetic risk factor for Parkinson disease (PD). These include pathogenic variants causing Gaucher disease (GD) (divided into “severe,” “mild,” or “complex”—resulting from recombinant alleles—based on the phenotypic effects in GD) and “risk” variants, which are not associated with GD but nevertheless confer increased risk of PD. As a group, GBA-PD patients have more severe motor and nonmotor symptoms, faster disease progression, and reduced survival compared with noncarriers. However, different GBA variants impact variably on clinical phenotype. In the heterozygous state, “complex” and “severe” variants are associated with a more aggressive and rapidly progressive disease. Conversely, “mild” and “risk” variants portend a more benign course. Homozygous or compound heterozygous carriers usually display severe phenotypes, akin to heterozygous “complex” or “severe” variants carriers. This article reviews genotype–phenotype correlations in GBA-PD, focusing on clinical and nonclinical aspects (neuroimaging and biochemical markers), and explores other disease modifiers that deserve consideration in the characterization of these patients.
Collapse
Affiliation(s)
- Elisa Menozzi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom
| |
Collapse
|
6
|
Salles PA, Mata IF, Fernandez HH. Should we start integrating genetic data in decision-making on device-aided therapies in Parkinson disease? A point of view. Parkinsonism Relat Disord 2021; 88:51-57. [PMID: 34119931 DOI: 10.1016/j.parkreldis.2021.05.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 04/26/2021] [Accepted: 05/12/2021] [Indexed: 12/14/2022]
Abstract
Parkinson disease (PD) is a complex heterogeneous neurodegenerative disorder. Association studies have revealed numerous genetic risk loci and variants, and about 5-10% suffer from a monogenic form. Because the presentation and course of PD is unique to each patient, personalized symptomatic treatment should ideally be offered to treat the most disabling motor and non-motor symptoms. Indeed, clinical milestones and treatment complications that appear during disease progression are influenced by the genetic imprint. With recent advances in PD, more patients live longer to become eligible for device-aided therapies, such as apomorphine continuous subcutaneous infusion, levodopa duodenal gel infusion, and deep brain stimulation surgery, each with its own inclusion and exclusion criteria, advantages and disadvantages. Because genetic variants influence the expression of particular clinical profiles, factors for better or worse outcomes for device-aided therapies may then be proactively identified. For example, mutations in PRKN, LRRK2 and GBA express phenotypes that favor suitability for different device therapies, although with marked differences in the therapeutic window; whereas multiplications of SNCA express phenotypes that make them less desirable for device therapies.
Collapse
Affiliation(s)
- Philippe A Salles
- Center for Neurological Restoration, Cleveland Clinic Neurological Institute, OH, USA; Movement Disorders Center, CETRAM, Santiago, Chile.
| | - Ignacio F Mata
- Lerner Research Institute, Genomic Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA.
| | - Hubert H Fernandez
- Center for Neurological Restoration, Cleveland Clinic Neurological Institute, OH, USA.
| |
Collapse
|
7
|
Olszewska DA, McCarthy A, Soto-Beasley AI, Walton RL, Magennis B, McLaughlin RL, Hardiman O, Ross OA, Lynch T. Association Between Glucocerebrosidase Mutations and Parkinson's Disease in Ireland. Front Neurol 2020; 11:527. [PMID: 32714263 PMCID: PMC7344206 DOI: 10.3389/fneur.2020.00527] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 05/13/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple studies implicate heterozygous GBA mutations as a major genetic risk factor for Parkinson's disease (PD); however, the frequency of mutations has never been examined in PD patients from the Irish population. We prospectively recruited 314 unrelated Irish PD patients (UK Brain Bank Criteria) and 96 Irish healthy controls (without any signs or family history of parkinsonism) attending. The Dublin Neurological Institute (DNI). Complete exon GBA Sanger sequencing analysis with flanking intronic regions was performed. The GBA carrier frequency was 8.3% in PD and 3.1% in controls. We identified a number of potentially pathogenic mutations including a p.G195E substitution and a p.G377C variant, previously described in a case study of Gaucher's disease in Ireland. On genotype–phenotype assessment hallucinations, dyskinesia, and dystonia were more prevalent in GBA-PD. The genetic etiology of PD in Ireland differs from the continental Europe as seen with the lower LRRK2 and higher than in most European countries GBA mutation frequency. Determining genetic risk factors in different ethnicities will be critical for future personalized therapeutic approach.
Collapse
Affiliation(s)
- Diana A Olszewska
- The Dublin Neurological Institute at the Mater Misericordiae University Hospital, Dublin, Ireland.,Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, United States.,School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Allan McCarthy
- The Dublin Neurological Institute at the Mater Misericordiae University Hospital, Dublin, Ireland.,Department of Neurology, The Adelaide and Meath Hospital, Dublin, Ireland
| | | | - Ronald L Walton
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, United States
| | - Brian Magennis
- The Dublin Neurological Institute at the Mater Misericordiae University Hospital, Dublin, Ireland
| | | | - Orla Hardiman
- Department of Neurology, Beaumont Hospital, Dublin, Ireland.,Academic Unit of Neurology, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, United States.,School of Medicine and Medical Science, University College Dublin, Dublin, Ireland.,Department of Clinical Genomics, Mayo Clinic, Jacksonville, FL, United States
| | - Tim Lynch
- The Dublin Neurological Institute at the Mater Misericordiae University Hospital, Dublin, Ireland.,School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
8
|
Stute L, Krüger R. [Emerging concepts for precision medicine in Parkinson's disease with focus on genetics]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2020; 88:558-566. [PMID: 32485745 DOI: 10.1055/a-1149-2204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The diverse and highly individual presentations of Parkinson's disease (PD) as a complex combination of motor and non-motor symptoms are being increasingly well characterised not least through large patient cohorts applying deep phenotyping. However, in terms of treatment of PD, the approach is uniform and purely symptomatic. Better stratification strategies with better precision medicine approaches offer opportunities to improve symptomatic treatment, define first causative therapies and provide more patient-centred care. Insight from targeted therapies for monogenic forms of PD aiming at neuroprotection may pave the way for new mechanism-based interventions also for the more common idiopathic PD. Improved stratification of patients may support symptomatic treatments by predicting treatment efficacy and long-term benefit of current pharmacological or neuromodulatory therapies, e.g. in the context of emerging pharmacogenomic knowledge. Based on asymptomatic carriers with monogenic PD or patients with REM sleep behaviour disorder (RBD), first options for applying preventive treatments emerge. The implications of these treatment strategies in relation to disease progression, and the prospects of their implementation in clinical practice need to be addressed.
Collapse
Affiliation(s)
- Lara Stute
- Parkinson Research Clinic, Centre Hospitalier de Luxembourg (CHL), Luxembourg, Luxembourg.,Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-Sur-Alzette, Luxembourg
| | - Rejko Krüger
- Parkinson Research Clinic, Centre Hospitalier de Luxembourg (CHL), Luxembourg, Luxembourg.,Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-Sur-Alzette, Luxembourg.,Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| |
Collapse
|
9
|
Genetic testing for Parkinson disease: current practice, knowledge, and attitudes among US and Canadian movement disorders specialists. Genet Med 2019; 22:574-580. [PMID: 31680121 PMCID: PMC7056638 DOI: 10.1038/s41436-019-0684-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/11/2019] [Accepted: 10/11/2019] [Indexed: 12/18/2022] Open
Abstract
Purpose Genetic testing for Parkinson disease (PD) has not been widely used
in clinical practice. In preparation for upcoming precision medicine–designed
clinical trials for GBA and LRRK2, we evaluated movement disorders specialists’
current practice, knowledge, attitudes, and barriers to genetic testing in
PD. Methods An anonymous questionnaire was sent to movement disorders
specialists at 146 Parkinson Study Group (PSG) sites in the United States
(n = 131) and Canada (n = 15) to assess their knowledge and attitudes
about genetic testing for PD. Results One hundred seventy-eight (47.6%) PSG clinicians completed the
questionnaire. Forty-one percent of respondents had not referred any PD patients
for genetic testing in the last year and >80% reported referring fewer than
11 patients over the same period. Most common reasons for not referring for
genetic testing included lack of insurance coverage/cost to the patient and lack
of perceived utility. On a scale of 0–100, the mean level of comfort in
respondents’ own ability to genetically counsel PD patients on GBA and LRRK2
was 52 (SD = 28). Sixty percent of clinicians correctly answered all questions
about the inheritance and penetrance of GBA
and LRRK2 variants. Conclusions There is an urgent need to increase knowledge and reduce practical
barriers to genetic counseling and testing in PD.
Collapse
|
10
|
Saunders-Pullman R, Mirelman A, Alcalay RN, Wang C, Ortega RA, Raymond D, Mejia-Santana H, Orbe-Reilly M, Johannes BA, Thaler A, Ozelius L, Orr-Urtreger A, Marder KS, Giladi N, Bressman SB. Progression in the LRRK2-Asssociated Parkinson Disease Population. JAMA Neurol 2019; 75:312-319. [PMID: 29309488 DOI: 10.1001/jamaneurol.2017.4019] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Few prospective longitudinal studies have evaluated the progression of Parkinson disease (PD) in patients with the leucine-rich repeat kinase 2 (LRRK2 [OMIM 609007]) mutation. Knowledge about such progression will aid clinical trials. Objective To determine whether the longitudinal course of PD in patients with the LRRK2 mutation differs from the longitudinal course of PD in patients without the mutation. Design, Setting, and Participants A prospective comprehensive assessment of a large cohort of patients from 3 sites with LRRK2 PD or with nonmutation PD was conducted from July 21, 2009, to September 30, 2016. All patients of Ashkenazi Jewish ancestry with PD were approached at each site; approximately 80% agreed to an initial visit. A total of 545 patients of Ashkenazi Jewish descent with PD who had 1 to 4 study visits were evaluated. A total of 144 patients (26.4%) had the LRRK2 G2019S mutation. Patients with GBA (OMIM 606463) mutations were excluded from the analysis. Main Outcomes and Measures Linear mixed-effects models for longitudinal motor scores were used to examine the association of LRRK2 mutation status with the rate of change in Unified Parkinson's Disease Rating Scale III scores using disease duration as the time scale, adjusting for sex, site, age, disease duration, cognitive score, and levodopa-equivalent dose at baseline. Mixed-effects models were used to assess change in cognition, as measured by Montreal Cognitive Assessment scores. Results Among the 545 participants, 233 were women, 312 were men, and the mean (SD) age was 68.2 (9.1) years for participants with the LRRK2 mutation and 67.8 (10.7) years for those without it. Seventy-two of 144 participants with the LRRK2 mutation and 161 of 401 participants with no mutation were women. The estimate (SE) of the rate of change in the Unified Parkinson's Disease Rating Scale III motor score per year among those with the LRRK2 mutation (0.689 [0.192] points per year) was less than among those without the mutation (1.056 [0.187] points per year; difference, -0.367 [0.149] points per year; P = .02). The estimate (SE) of the difference in the rate of change of the Montreal Cognitive Assessment score between those with the LRRK2 mutation (-0.096 [0.090] points per year) and those without the mutation (-0.192 [0.102] points per year) did not reach statistical significance (difference, 0.097 [0.055] points per year; P = .08). Conclusions and Relevance Prospective longitudinal follow-up of patients with PD with or without the LRRK2 G2019S mutation supports data from a cross-sectional study and demonstrates a slower decline in motor Unified Parkinson's Disease Rating Scale scores among those with LRRK2 G2019S-associated PD.
Collapse
Affiliation(s)
- Rachel Saunders-Pullman
- Department of Neurology, Mount Sinai Beth Israel Medical Center, New York, New York.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Anat Mirelman
- Movement Disorders Unit, Neurological Institute, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler School of Medicine, Sagol School for Neuroscience, Tel Aviv University, Tel Aviv, Israel.,Department of Physical Therapy, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Roy N Alcalay
- Department of Neurology, College of Physicians and Surgeons, New York, New York
| | - Cuiling Wang
- Department of Neurology, College of Physicians and Surgeons, New York, New York.,Department of Epidemiology and Family Health, Albert Einstein College of Medicine, Yeshiva University, Bronx, New York.,Department of Neurology, Albert Einstein College of Medicine, Yeshiva University, Bronx, New York
| | - Roberto A Ortega
- Department of Neurology, Mount Sinai Beth Israel Medical Center, New York, New York.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Deborah Raymond
- Department of Neurology, Mount Sinai Beth Israel Medical Center, New York, New York.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | - Brooke A Johannes
- Department of Neurology, Mount Sinai Beth Israel Medical Center, New York, New York.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Avner Thaler
- Movement Disorders Unit, Neurological Institute, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler School of Medicine, Sagol School for Neuroscience, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Laurie Ozelius
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Avi Orr-Urtreger
- Movement Disorders Unit, Neurological Institute, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler School of Medicine, Sagol School for Neuroscience, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.,Genetic Institute, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Karen S Marder
- Department of Neurology, College of Physicians and Surgeons, New York, New York.,Taub Institute for Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York
| | - Nir Giladi
- Movement Disorders Unit, Neurological Institute, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler School of Medicine, Sagol School for Neuroscience, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Susan B Bressman
- Department of Neurology, Mount Sinai Beth Israel Medical Center, New York, New York.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York
| | | |
Collapse
|
11
|
de Oliveira LM, Barbosa ER, Aquino CC, Munhoz RP, Fasano A, Cury RG. Deep Brain Stimulation in Patients With Mutations in Parkinson's Disease-Related Genes: A Systematic Review. Mov Disord Clin Pract 2019; 6:359-368. [PMID: 31286005 DOI: 10.1002/mdc3.12795] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/07/2019] [Accepted: 05/05/2019] [Indexed: 12/11/2022] Open
Abstract
Background Deep brain stimulation (DBS) is an effective treatment for Parkinson's disease (PD), and careful selection of candidates is a key component of successful therapy. Although it is recognized that factors such as age, disease duration, and levodopa responsiveness can influence outcomes, it is unclear whether genetic background should also serve as a parameter. Objectives The aim of this systematic review is to explore studies that have evaluated DBS in patients with mutations in PD-related genes. Methods We performed a selective literature search for articles regarding the effects of DBS in autosomal dominant or recessive forms of PD or in PD patients with genetic risk factors. Data regarding changes in motor and nonmotor scores and the presence of adverse events after the stimulation were collected. Results A total of 25 studies were included in the systematic review, comprising 135 patients. In the shorter term, most patients showed marked or satisfactory response to subthalamic DBS, although leucine rich repeat kinase 2 carriers of R114G mutations had higher rates of unsatisfactory outcome. Longer term follow-up data were scarce but suggested that motor benefit is sustained. Patients with the glucosidase beta acid (GBA) mutation showed higher rates of cognitive decline after surgery. Motor outcome was scarce for pallidal DBS. Few adverse events were reported. Conclusions Subthalamic DBS results in positive outcomes in the short term in patients with Parkin, GBA, and leucine-rich repeat kinase 2 (non-R144G) mutations, although the small sample size limits the interpretation of our findings. Longer and larger cohorts of follow-up, with broader nonmotor symptom evaluations will be necessary to better customize DBS therapy in this population.
Collapse
Affiliation(s)
- Lais Machado de Oliveira
- The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic, University Health Network, Toronto Western Hospital, Division of Neurology University of Toronto Toronto Ontario Canada.,Movement Disorders Center, Department of Neurology, School of Medicine University of São Paulo São Paulo Brazil
| | - Egberto Reis Barbosa
- Movement Disorders Center, Department of Neurology, School of Medicine University of São Paulo São Paulo Brazil
| | - Camila Catherine Aquino
- Department Health Research Methods, Evidence, and Impact McMaster University Hamilton Ontario Canada
| | - Renato Puppi Munhoz
- The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic, University Health Network, Toronto Western Hospital, Division of Neurology University of Toronto Toronto Ontario Canada.,Krembil Brain Institute Toronto Ontario Canada
| | - Alfonso Fasano
- The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic, University Health Network, Toronto Western Hospital, Division of Neurology University of Toronto Toronto Ontario Canada.,Krembil Brain Institute Toronto Ontario Canada
| | - Rubens Gisbert Cury
- Movement Disorders Center, Department of Neurology, School of Medicine University of São Paulo São Paulo Brazil
| |
Collapse
|
12
|
Eisinger RS, Martinez-Ramirez D, Ramirez-Zamora A, Hess CW, Almeida L, Okun MS, Gunduz A. Parkinson's disease motor subtype changes during 20 years of follow-up. Parkinsonism Relat Disord 2019; 76:104-107. [PMID: 31129020 DOI: 10.1016/j.parkreldis.2019.05.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 05/06/2019] [Accepted: 05/15/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Robert S Eisinger
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, 32611, USA; Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32611, USA
| | - Daniel Martinez-Ramirez
- Department of Neurology, University of Florida, Gainesville, FL, 32611, USA; Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32611, USA; Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo Leon, 64710, Mexico
| | - Adolfo Ramirez-Zamora
- Department of Neurology, University of Florida, Gainesville, FL, 32611, USA; Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32611, USA
| | - Christopher W Hess
- Department of Neurology, University of Florida, Gainesville, FL, 32611, USA; Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32611, USA
| | - Leonardo Almeida
- Department of Neurology, University of Florida, Gainesville, FL, 32611, USA; Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32611, USA
| | - Michael S Okun
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, 32611, USA; Department of Neurology, University of Florida, Gainesville, FL, 32611, USA; Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32611, USA
| | - Aysegul Gunduz
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, 32611, USA; Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32611, USA; J. Crayton Pruitt Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
13
|
Antonini A, Gentile G, Giglio M, Marcante A, Gage H, Touray MML, Fotiadis DI, Gatsios D, Konitsiotis S, Timotijevic L, Egan B, Hodgkins C, Biundo R, Pellicano C. Acceptability to patients, carers and clinicians of an mHealth platform for the management of Parkinson's disease (PD_Manager): study protocol for a pilot randomised controlled trial. Trials 2018; 19:492. [PMID: 30217235 PMCID: PMC6138904 DOI: 10.1186/s13063-018-2767-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 06/25/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Parkinson's disease is a degenerative neurological condition causing multiple motor and non-motor symptoms that have a serious adverse effect on quality of life. Management is problematic due to the variable and fluctuating nature of symptoms, often hourly and daily. The PD_Manager mHealth platform aims to provide a continuous feed of data on symptoms to improve clinical understanding of the status of any individual patient and inform care planning. The objectives of this trial are to (1) assess patient (and family carer) perspectives of PD_Manager regarding comfort, acceptability and ease of use; (2) assess clinician views about the utility of the data generated by PD_Manager for clinical decision making and the acceptability of the system in clinical practice. METHODS/DESIGN This trial is an unblinded, parallel, two-group, randomised controlled pilot study. A total of 200 persons with Parkinson's disease (Hoehn and Yahr stage 3, experiencing motor fluctuations at least 2 h per day), with primary family carers, in three countries (110 Rome, 50 Venice, Italy; 20 each in Ioannina, Greece and Surrey, England) will be recruited. Following informed consent, baseline information will be gathered, including the following: age, gender, education, attitudes to technology (patient and carer); time since Parkinson's diagnosis, symptom status and comorbidities (patient only). Randomisation will assign participants (1:1 in each country), to PD_Manager vs control, stratifying by age (1 ≤ 70 : 1 > 70) and gender (60% M: 40% F). The PD_Manager system captures continuous data on motor symptoms, sleep, activity, speech quality and emotional state using wearable devices (wristband, insoles) and a smartphone (with apps) for storing and transmitting the information. Control group participants will be asked to keep a symptom diary covering the same elements as PD_Manager records. After a minimum of two weeks, each participant will attend a consultation with a specialist doctor for review of the data gathered (by either means), and changes to management will be initiated as indicated. Patients, carers and clinicians will be asked for feedback on the acceptability and utility of the data collection methods. The PD_Manager intervention, compared to a symptom diary, will be evaluated in a cost-consequences framework. DISCUSSION Information gathered will inform further development of the PD_Manager system and a larger effectiveness trial. TRIAL REGISTRATION ISRCTN Registry, ISRCTN17396879 . Registered on 15 March 2017.
Collapse
Affiliation(s)
- Angelo Antonini
- Department of Neuroscience, University of Padua, Padua, Italy.,IRCCS San Camillo Hospital, Venice, Italy
| | | | | | - Andrea Marcante
- Department of Neuroscience, University of Padua, Padua, Italy.,IRCCS San Camillo Hospital, Venice, Italy
| | - Heather Gage
- Surrey Health Economics Centre, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Morro M L Touray
- Surrey Health Economics Centre, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK.
| | - Dimitrios I Fotiadis
- Department of Materials Science, Unit of Medical Technology and Intelligent Information Systems, University of Ioannina, Ioannina, Greece
| | - Dimitris Gatsios
- Department of Materials Science, Unit of Medical Technology and Intelligent Information Systems, University of Ioannina, Ioannina, Greece
| | - Spyridon Konitsiotis
- Department of Neurology, Medical School, University of Ioannina, Ioannina, Greece
| | - Lada Timotijevic
- Department of Psychology, University of Surrey, Guildford, England
| | - Bernadette Egan
- Department of Psychology, University of Surrey, Guildford, England
| | - Charo Hodgkins
- Department of Psychology, University of Surrey, Guildford, England
| | | | - Clelia Pellicano
- Fondazione Santa Lucia IRCCS, Via Ardeatina 306, 00179, Rome, Italy.,Department of Neuriscience, Mental Health and Sensory Organs, Sapienza University, Via di Grottarossa 1035, 00189, Rome, Italy
| | | |
Collapse
|
14
|
Abstract
PURPOSE OF REVIEW GBA mutations are the most common known genetic cause of Parkinson's disease (PD). Its biological pathway may be important in idiopathic PD, since activity of the enzyme encoded by GBA, glucocerebrosidase, is reduced even among PD patients without GBA mutations. This article describes the structure and function of GBA, reviews recent literature on the clinical phenotype of GBA PD, and suggests future directions for research, counseling, and treatment. RECENT FINDINGS Several longitudinal studies have shown that GBA PD has faster motor and cognitive progression than idiopathic PD and that this effect is dose dependent. New evidence suggests that GBA mutations may be important in multiple system atrophy. Further, new interventional studies focusing on GBA PD are described. These studies may increase the interest of PD patients and caregivers in genetic counseling. GBA mutation status may help clinicians estimate PD progression, though mechanisms underlying GBA and synucleinopathy require further understanding.
Collapse
|
15
|
Parkinson's disease phenotype is influenced by the severity of the mutations in the GBA gene. Parkinsonism Relat Disord 2018; 55:45-49. [PMID: 29784561 DOI: 10.1016/j.parkreldis.2018.05.009] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/04/2018] [Accepted: 05/09/2018] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Mutations in the glucocerebrosidase (GBA) gene are divided into mild and severe (mGBA, sGBA) based on their contribution to the phenotype of Gaucher disease (GD) among homozygotes. We conducted a longitudinal analysis of Parkinson's disease (PD) patients carrying mutations in the GBA gene to better characterize genotype-phenotype correlations. METHODS Patients underwent a comprehensive assessment of medical, neurological, cognitive and non-motor functions. Data from these patients was explored to evaluate differences in disease phenotype based on genotype. RESULTS A total of 355 PD patients participated in this study; 152 idiopathic PD patients, 139 mGBA, 48 sGBA and 16 GD-PD. Groups were similar in age, sex, years of education and age of onset. Both sGBA and GD-PD had higher Unified Parkinson Disease Rating Scale (UPDRS) scores (p = 0.041), higher frequencies of REM sleep behavior disorder (RBD) (p = 0.022) and hallucinations (p < 0.0001) compared to the other groups of patients. sGBA experienced more non-motor symptoms (p < 0.0001), depression (p < 0.001) and worse hyposmia (p = 0.010). Trail making test was significantly longer in GD-PD followed by sGBA, mGBA and iPD (p = 0.005). DISCUSSION Motor, cognitive, olfactory and psychiatric symptoms are more severe in sGBA and GD-PD compared to mGBA and iPD, reinforcing the notion that the severity of the PD phenotype is related to the severity of the mutation in the GBA gene.
Collapse
|
16
|
Dyskinesias and levodopa therapy: why wait? J Neural Transm (Vienna) 2018; 125:1119-1130. [PMID: 29428995 DOI: 10.1007/s00702-018-1856-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/05/2018] [Indexed: 12/28/2022]
Abstract
Throughout the years there has been a longstanding discussion on whether levodopa therapy in Parkinson's disease should be started in early vs. later stages, in order to prevent or delay motor complications such as fluctuations and dyskinesias. This controversial topic has been extensively debated for decades, and the prevailing view today is that levodopa should not be postponed. However, there is still fear associated with its use in early stages, especially in younger patients, who are more prone to develop dyskinesias. Even though dyskinesias are linked to levodopa use in Parkinson's disease, it has been shown that starting with a different medication (such as dopamine agonists) will not significantly delay their onset once levodopa is introduced. Since levodopa provides better symptomatic control, and other drugs may be associated with notable side effects, it is our view that there is insufficient evidence to justify levodopa-sparing strategies. The physician should try to assess each patient individually, taking into account motor and non-motor demands, as well as risk factors for potential complications, finding the optimum treatment strategy for each one. The following article provides an historical narrative perspective, as well as a literature review of those intrinsic and modifiable risk factors that have been associated with levodopa-induced dyskinesias, which should be taken into consideration when choosing the therapeutic strategy in individual Parkinson's disease patients.
Collapse
|
17
|
da Silva CP, de M Abreu G, Cabello Acero PH, Campos M, Pereira JS, de A Ramos SR, Nascimento CM, Voigt DD, Rosso AL, Araujo Leite MA, Vasconcellos LFR, Nicaretta DH, Della Coletta MV, da Silva DJ, Gonçalves AP, Dos Santos JM, Calassara V, Valença DCT, de M Martins CJ, Santos-Rebouças CB, Pimentel MMG. Clinical profiles associated with LRRK2 and GBA mutations in Brazilians with Parkinson's disease. J Neurol Sci 2017; 381:160-164. [PMID: 28991672 DOI: 10.1016/j.jns.2017.08.3249] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/11/2017] [Accepted: 08/23/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative disorder characterized by remarkable phenotypic variability. Accumulated evidence points that the manifestation of PD clinical signs might be differentially modified by genetic factors, as mutations in LRRK2 and GBA genes. In this sense, the clarification of the genotype-phenotype correlations in PD has important implications in predicting prognosis and can contribute to the development of specific therapeutic approaches. METHODS Here, we conducted the first comparative analysis of motor and non-motor features in 17 LRRK2 and 22 GBA mutation carriers and 93 non-carriers unrelated PD patients from Brazil, a highly admixed population. RESULTS Significant differences were found between the three groups. LRRK2 PD patients presented more occurrence of familiar history. Resting tremor was observed in a lower frequency in GBA mutation carries. In contrast, gait freezing and dysautonomia was present in lower frequencies in LRRK2 carriers. Besides that, LRRK2 and GBA mutation carriers showed a higher incidence of depressive symptoms and a younger age at onset, when compared to non-carriers. CONCLUSION Our results suggest that specific mutations in GBA and LRRK2 influence the clinical signs of the disease, with significant implications for handling of specific patient groups.
Collapse
Affiliation(s)
- Camilla P da Silva
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabriella de M Abreu
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Pedro H Cabello Acero
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Laboratory of Genetics, School of Health Science, University of Grande Rio, Rio de Janeiro, Brazil
| | - Mário Campos
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - João S Pereira
- Movement Disorders Section, Neurology Service, Pedro Ernesto University Hospital, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sarah R de A Ramos
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Caroline M Nascimento
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danielle D Voigt
- Laboratory of Genetics, School of Health Science, University of Grande Rio, Rio de Janeiro, Brazil
| | - Ana Lucia Rosso
- University Hospital Clementino Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marco A Araujo Leite
- Movement Disorders Unit, Division of Neurology, Hospital Antônio Pedro, Fluminense Federal University, Brazil
| | - Luiz Felipe R Vasconcellos
- Institute of Neurology Deolindo Couto, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Federal Hospital of Servidores do Estado, Rio de Janeiro, Brazil
| | | | | | - Delson José da Silva
- Neuroscience Core, Hospital Clinics, Federal University of Goiás, Brazil; Integrated Neurosciences Institute, Goiás, Brazil
| | - Andressa P Gonçalves
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jussara M Dos Santos
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Veluma Calassara
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Débora Cristina T Valença
- Laboratory of Clinical and Experimental Pathophysiology (CLINEX), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Cyro J de M Martins
- Laboratory of Clinical and Experimental Pathophysiology (CLINEX), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Cíntia B Santos-Rebouças
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Márcia M G Pimentel
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
18
|
Sokol LL, Shapiro D, Young MJ, Wise AH, Hadelsberg UP, Kaufman Y, Espay AJ, Merola A. The Parkinson Care Advocate: Integrating Care Delivery. Front Neurol 2017; 8:364. [PMID: 28798721 PMCID: PMC5529407 DOI: 10.3389/fneur.2017.00364] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 07/10/2017] [Indexed: 12/13/2022] Open
Affiliation(s)
- Leonard L Sokol
- University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Department of Neurology, James J and Joan A. Gardner Center for Parkinson's Disease and Movement Disorders, University of Cincinnati, Cincinnati, OH, United States
| | | | - Michael J Young
- Departments of Internal Medicine and Neurology, Massachusetts General Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Adina H Wise
- Columbia University Medical Center, New York, NY, United States
| | - Uri P Hadelsberg
- Department of Neurosurgery, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Yakir Kaufman
- Department of Neuropsychogeriatrics, Herzog Hospital, Jerusalem, Israel.,Faculty of Medicine, Hebrew University, Hadassah Medical Center, Jerusalem, Israel
| | - Alberto J Espay
- Department of Neurology, James J and Joan A. Gardner Center for Parkinson's Disease and Movement Disorders, University of Cincinnati, Cincinnati, OH, United States
| | - Aristide Merola
- Department of Neurology, James J and Joan A. Gardner Center for Parkinson's Disease and Movement Disorders, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
19
|
Eisinger RS, Hess CW, Martinez-Ramirez D, Almeida L, Foote KD, Okun MS, Gunduz A. Motor subtype changes in early Parkinson's disease. Parkinsonism Relat Disord 2017; 43:67-72. [PMID: 28754232 DOI: 10.1016/j.parkreldis.2017.07.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 07/12/2017] [Accepted: 07/19/2017] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Distinct motor subtypes of Parkinson's disease (PD) have been described through both clinical observation and through data-driven approaches. However, the extent to which motor subtypes change during disease progression remains unknown. Our objective was to determine motor subtypes of PD using an unsupervised clustering methodology and evaluate subtype changes with disease duration. METHODS The Parkinson's Progression Markers Initiative database of 423 newly diagnosed PD patients was utilized to retrospectively identify unique motor subtypes through a data-driven, hierarchical correlational clustering approach. For each patient, we assigned a subtype to each motor assessment at each follow-up visit (time points) and by using published criteria. We examined changes in PD subtype with disease duration using both qualitative and quantitative methods. RESULTS Five distinct motor subtypes were identified based on the motor assessment items and these included: Tremor Dominant (TD), Axial Dominant, Appendicular Dominant, Rigidity Dominant, and Postural and Instability Gait Disorder Dominant. About half of the patients had consistent subtypes at all time points. Most patients met criteria for TD subtype soon after diagnosis. For patients with inconsistent subtypes, there was an overall trend to shift away from a TD phenotype with disease duration, as shown by chi-squared test, p < 0.001, and linear regression analysis, p < 0.05. CONCLUSION These results strongly suggest that classification of motor subtypes in PD can shift with increasing disease duration. Shifting subtypes is a factor that should be accounted for in clinical practice or in clinical trials.
Collapse
Affiliation(s)
- Robert S Eisinger
- Department of Neuroscience, Center for Movement Disorders and Neurorestoration, 3450 Hull Road, University of Florida, Gainesville, FL 32607, United States.
| | - Christopher W Hess
- Department of Neurology, Center for Movement Disorders and Neurorestoration, 3450 Hull Road, University of Florida, Gainesville, FL 32607, United States.
| | - Daniel Martinez-Ramirez
- Department of Neurology, Center for Movement Disorders and Neurorestoration, University of Florida, Gainesville, FL, United States.
| | - Leonardo Almeida
- Department of Neurology, Center for Movement Disorders and Neurorestoration, University of Florida, Gainesville, FL, United States.
| | - Kelly D Foote
- Department of Neurosurgery, Center for Movement Disorders and Neurorestoration, McKnight Brain Institute, 3rd Floor, University of Florida, Gainesville, FL 32611, United States.
| | - Michael S Okun
- Department of Neurology, Center for Movement Disorders and Neurorestoration, University of Florida, Gainesville, FL, United States.
| | - Aysegul Gunduz
- J. Crayton Pruitt Family Department of Biomedical Engineering, Center for Movement Disorders and Neurorestoration, University of Florida, Gainesville, FL, United States.
| |
Collapse
|