1
|
Ahlskog JE. Parkinson's disease progression is multifaceted: Evidence for the underlying benchmarks. Parkinsonism Relat Disord 2024; 121:106037. [PMID: 38360506 DOI: 10.1016/j.parkreldis.2024.106037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 02/17/2024]
Abstract
Long-term Parkinson's disease (PD) progression is not a homogeneous process and manifests in diverse ways over the lifetime. Recognition of progression benchmarks and their substrates is important for treatment and addressing the expectations of patients, as well as for PD research planning.
Collapse
Affiliation(s)
- J Eric Ahlskog
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
2
|
Di Folco C, Couronné R, Arnulf I, Mangone G, Leu-Semenescu S, Dodet P, Vidailhet M, Corvol JC, Lehéricy S, Durrleman S. Charting Disease Trajectories from Isolated REM Sleep Behavior Disorder to Parkinson's Disease. Mov Disord 2024; 39:64-75. [PMID: 38006282 DOI: 10.1002/mds.29662] [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: 10/10/2022] [Revised: 10/03/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Clinical presentation and progression dynamics are variable in patients with Parkinson's disease (PD). Disease course mapping is an innovative disease modelling technique that summarizes the range of possible disease trajectories and estimates dimensions related to onset, sequence, and speed of progression of disease markers. OBJECTIVE To propose a disease course map for PD and investigate progression profiles in patients with or without rapid eye movement sleep behavioral disorders (RBD). METHODS Data of 919 PD patients and 88 isolated RBD patients from three independent longitudinal cohorts were analyzed (follow-up duration = 5.1; 95% confidence interval, 1.1-8.1] years). Disease course map was estimated by using eight clinical markers (motor and non-motor symptoms) and four imaging markers (dopaminergic denervation). RESULTS PD course map showed that the first changes occurred in the contralateral putamen 13 years before diagnosis, followed by changes in motor symptoms, dysautonomia, sleep-all before diagnosis-and finally cognitive decline at the time of diagnosis. The model showed earlier disease onset, earlier non-motor and later motor symptoms, more rapid progression of cognitive decline in PD patients with RBD than PD patients without RBD. This pattern was even more pronounced in patients with isolated RBD with early changes in sleep, followed by cognition and non-motor symptoms and later changes in motor symptoms. CONCLUSIONS Our findings are consistent with the presence of distinct patterns of progression between patients with and without RBD. Understanding heterogeneity of PD progression is key to decipher the underlying pathophysiology and select homogeneous subgroups of patients for precision medicine. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Cécile Di Folco
- Inria, Centre de Paris, Paris, France
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Raphaël Couronné
- Inria, Centre de Paris, Paris, France
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Isabelle Arnulf
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Graziella Mangone
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Smaranda Leu-Semenescu
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Pauline Dodet
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Marie Vidailhet
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Jean-Christophe Corvol
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Stéphane Lehéricy
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Stanley Durrleman
- Inria, Centre de Paris, Paris, France
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| |
Collapse
|
3
|
Molcho L, Maimon NB, Hezi N, Zeimer T, Intrator N, Gurevich T. Evaluation of Parkinson's disease early diagnosis using single-channel EEG features and auditory cognitive assessment. Front Neurol 2023; 14:1273458. [PMID: 38174098 PMCID: PMC10762798 DOI: 10.3389/fneur.2023.1273458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 11/29/2023] [Indexed: 01/05/2024] Open
Abstract
Background Parkinson's disease (PD) often presents with subtle early signs, making diagnosis difficult. F-DOPA PET imaging provides a reliable measure of dopaminergic function and is a primary tool for early PD diagnosis. This study aims to evaluate the ability of machine-learning (ML) extracted EEG features to predict F-DOPA results and distinguish between PD and non-PD patients. These features, extracted using a single-channel EEG during an auditory cognitive assessment, include EEG feature A0 associated with cognitive load in healthy subjects, and EEG feature L1 associated with cognitive task differentiation. Methods Participants in this study are comprised of cognitively healthy patients who had undergone an F-DOPA PET scan as a part of their standard care (n = 32), and cognitively healthy controls (n = 20). EEG data collected using the Neurosteer system during an auditory cognitive task, was decomposed using wavelet-packet analysis and machine learning methods for feature extraction. These features were used in a connectivity analysis that was applied in a similar manner to fMRI connectivity. A preliminary model that relies on the features and their connectivity was used to predict initially unrevealed F-DOPA test results. Then, generalized linear mixed models (LMM) were used to discern between PD and non-PD subjects based on EEG variables. Results The prediction model correctly classified patients with unrevealed scores as positive F-DOPA. EEG feature A0 and the Delta band revealed distinct activity patterns separating between study groups, with controls displaying higher activity than PD patients. In controls, EEG feature L1 showed variations between resting state and high-cognitive load, an effect lacking in PD patients. Conclusion Our findings exhibit the potential of single-channel EEG technology in combination with an auditory cognitive assessment to distinguish positive from negative F-DOPA PET scores. This approach shows promise for early PD diagnosis. Additional studies are needed to further verify the utility of this tool as a potential biomarker for PD.
Collapse
Affiliation(s)
- Lior Molcho
- Neurosteer Inc., New York, NY, United States
| | - Neta B. Maimon
- Neurosteer Inc., New York, NY, United States
- Department of Musicology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Neomi Hezi
- Movement Disorders Unit, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | | | - Nathan Intrator
- Neurosteer Inc., New York, NY, United States
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Tanya Gurevich
- Movement Disorders Unit, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
4
|
Beauchamp LC, Dore V, Villemagne VL, Xu S, Finkelstein D, Barnham KJ, Rowe C. Using 18F-AV-133 VMAT2 PET Imaging to Monitor Progressive Nigrostriatal Degeneration in Parkinson Disease. Neurology 2023; 101:e2314-e2324. [PMID: 37816639 PMCID: PMC10727223 DOI: 10.1212/wnl.0000000000207748] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/11/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND AND OBJECTIVES There are limited validated biomarkers in Parkinson disease (PD) which substantially hinders the ability to monitor disease progression and consequently measure the efficacy of disease-modifying treatments. Imaging biomarkers, such as vesicular monoamine transporter type 2 (VMAT2) PET, enable enhanced diagnostic accuracy and detect early neurodegenerative changes associated with prodromal PD. This study sought to assess whether 18F-AV-133 VMAT2 PET is sensitive enough to monitor and quantify disease progression over a 2-year window. METHODS 18F-AV-133 PET scans were performed on participants with PD and REM sleep behavior disorder (RBD) and neurologic controls (NC). All participants were scanned twice ∼26 months apart. Regional tracer retention was calculated with a primary visual cortex reference region and expressed as the standard uptake volume ratio. Regions of interest included caudate, anterior, and posterior putamen. At the time of scanning, participants underwent clinical evaluation including UPDRSMOTOR test, Sniffin' Sticks, and Hospital Anxiety and Depression Score. RESULTS Over the 26-month interval, a significant decline in PET signal was observed in all 3 regions in participants with PD (N = 26) compared with NC (N = 12), consistent with a decrease in VMAT2 level and ongoing neurodegeneration. Imaging trajectory calculations suggest that the neurodegeneration in PD occurs over ∼33 years [CI: 27.2-39.5], with ∼10.5 years [CI: 9.1-11.3] of degeneration in the posterior putamen before it becomes detectable on a VMAT2 PET scan, a further ∼6.5 years [CI: 1.6-12.7] until symptom onset, and a further ∼3 years [CI: 0.3-8.7] until clinical diagnosis. DISCUSSION Over a 2-year period, 18F-AV-133 VMAT2 PET was able to detect progression of nigrostriatal degeneration in participants with PD, and it represents a sensitive tool to identify individuals at risk of progression to PD, which are currently lacking using clinical readouts. Trajectory models propose that there is nigrostriatal degeneration occurring for 20 years before clinical diagnosis. These data demonstrate that VMAT2 PET provides a sensitive measure to monitor neurodegenerative progression of PD which has implications for PD diagnostics and subsequently clinical trial patient stratification and monitoring. CLASSIFICATION OF EVIDENCE This study provides Class IV evidence that VMAT2 PET can detect patients with Parkinson disease and quantify progression over a 2-year window.
Collapse
Affiliation(s)
- Leah C Beauchamp
- From the The Florey Institute of Neuroscience and Mental Health (L.C.B., D.F., K.J.B.); Health & Biosecurity Flagship (V.D.), The Australian eHealth Research Centre, The Commonwealth Scientific and Industrial Research Organisation; Department of Psychiatry (V.L.V.), University of Pittsburgh, PA; Department of Neurology (S.X.), Austin Health, Melbourne; The University of Melbourne (D.F.); Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne, Australia
| | - Vincent Dore
- From the The Florey Institute of Neuroscience and Mental Health (L.C.B., D.F., K.J.B.); Health & Biosecurity Flagship (V.D.), The Australian eHealth Research Centre, The Commonwealth Scientific and Industrial Research Organisation; Department of Psychiatry (V.L.V.), University of Pittsburgh, PA; Department of Neurology (S.X.), Austin Health, Melbourne; The University of Melbourne (D.F.); Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne, Australia
| | - Victor L Villemagne
- From the The Florey Institute of Neuroscience and Mental Health (L.C.B., D.F., K.J.B.); Health & Biosecurity Flagship (V.D.), The Australian eHealth Research Centre, The Commonwealth Scientific and Industrial Research Organisation; Department of Psychiatry (V.L.V.), University of Pittsburgh, PA; Department of Neurology (S.X.), Austin Health, Melbourne; The University of Melbourne (D.F.); Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne, Australia
| | - SanSan Xu
- From the The Florey Institute of Neuroscience and Mental Health (L.C.B., D.F., K.J.B.); Health & Biosecurity Flagship (V.D.), The Australian eHealth Research Centre, The Commonwealth Scientific and Industrial Research Organisation; Department of Psychiatry (V.L.V.), University of Pittsburgh, PA; Department of Neurology (S.X.), Austin Health, Melbourne; The University of Melbourne (D.F.); Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne, Australia
| | - David Finkelstein
- From the The Florey Institute of Neuroscience and Mental Health (L.C.B., D.F., K.J.B.); Health & Biosecurity Flagship (V.D.), The Australian eHealth Research Centre, The Commonwealth Scientific and Industrial Research Organisation; Department of Psychiatry (V.L.V.), University of Pittsburgh, PA; Department of Neurology (S.X.), Austin Health, Melbourne; The University of Melbourne (D.F.); Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne, Australia
| | - Kevin J Barnham
- From the The Florey Institute of Neuroscience and Mental Health (L.C.B., D.F., K.J.B.); Health & Biosecurity Flagship (V.D.), The Australian eHealth Research Centre, The Commonwealth Scientific and Industrial Research Organisation; Department of Psychiatry (V.L.V.), University of Pittsburgh, PA; Department of Neurology (S.X.), Austin Health, Melbourne; The University of Melbourne (D.F.); Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne, Australia.
| | - Christopher Rowe
- From the The Florey Institute of Neuroscience and Mental Health (L.C.B., D.F., K.J.B.); Health & Biosecurity Flagship (V.D.), The Australian eHealth Research Centre, The Commonwealth Scientific and Industrial Research Organisation; Department of Psychiatry (V.L.V.), University of Pittsburgh, PA; Department of Neurology (S.X.), Austin Health, Melbourne; The University of Melbourne (D.F.); Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne, Australia
| |
Collapse
|
5
|
Davis J, Kolaski E, Babcock DT. Vexed mutations promote degeneration of dopaminergic neurons through excessive activation of the innate immune response. NPJ Parkinsons Dis 2022; 8:147. [PMID: 36323700 PMCID: PMC9630459 DOI: 10.1038/s41531-022-00417-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/14/2022] [Indexed: 11/11/2022] Open
Abstract
The hallmark of Parkinson's disease (PD) is the loss of dopaminergic (DA) neurons in the brain. However, little is known about why DA neurons are selectively vulnerable to PD. We previously completed a screen identifying genes associated with the progressive degeneration of DA neurons. Here we describe the role of a previously uncharacterized gene, CG42339, in the loss of DA neurons using Drosophila Melanogaster. CG42339 mutants display a progressive loss of DA neurons and locomotor dysfunction, along with an accumulation of advanced glycation end products (AGEs) in the brain. Based on this phenotype, we refer to CG42339 as vexed. We demonstrate that vexed is specifically required within cortex glia to maintain neuronal viability. Loss of vexed function results in excessive activation of the innate immune response in the brain, leading to loss of DA neurons. We show that activation of the innate immune response leads to increased nitric oxide signaling and accumulation of AGEs, which ultimately result in neurodegeneration. These results provide further insight into the relationship between the role of the immune response in the central nervous system and how this impacts neuronal viability.
Collapse
Affiliation(s)
- Jacinta Davis
- grid.259029.50000 0004 1936 746XDepartment of Biological Sciences, Lehigh University, Bethlehem, PA USA
| | - Elizabeth Kolaski
- grid.259029.50000 0004 1936 746XDepartment of Biological Sciences, Lehigh University, Bethlehem, PA USA
| | - Daniel T. Babcock
- grid.259029.50000 0004 1936 746XDepartment of Biological Sciences, Lehigh University, Bethlehem, PA USA
| |
Collapse
|
6
|
Nishikawa N, Murata M, Hatano T, Mukai Y, Saitoh Y, Sakamoto T, Hanakawa T, Kamei Y, Tachimori H, Hatano K, Matsuda H, Taruno Y, Sawamoto N, Kajiyama Y, Ikenaka K, Kawabata K, Nakamura T, Iwaki H, Kadotani H, Sumi Y, Inoue Y, Hayashi T, Ikeuchi T, Shimo Y, Mochizuki H, Watanabe H, Hattori N, Takahashi Y, Takahashi R. Idiopathic rapid eye movement sleep behavior disorder in Japan: An observational study. Parkinsonism Relat Disord 2022; 103:129-135. [PMID: 36113390 DOI: 10.1016/j.parkreldis.2022.08.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/12/2022] [Accepted: 08/10/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Idiopathic rapid eye movement sleep behavior disorder (iRBD) is one of the most specific prodromal symptoms of synucleinopathies, including Parkinson's disease (PD) and multiple system atrophy. The Japan Parkinson's Progression Markers Initiative (J-PPMI) was a prospective cohort study conducted in Japanese patients with iRBD to investigate biomarkers for prodromal synucleinopathies. We carried out an initial assessment of the J-PPMI study to reveal the factors correlated with dopamine transporter single-photon emission computed tomography (DaT) and 123I-meta-iodobenzylguanidine (MIBG) myocardial scintigraphy. METHODS This cross-sectional study was conducted in 108 patients with iRBD, selected from the J-PPMI study. We divided the patients into four groups based on the MIBG and DaT results. We also recorded the patients' demographics and clinical data. Following PD probability calculation, we examined the biomarkers associated with DaT and MIBG. RESULTS Ninety-five of the enrolled patients (88%) met the diagnostic criteria for prodromal PD based on the probability score. Only five patients had normal MIBG and DaT. We identified 29 cases with decreased DaT and MIBG, all of whom met the above diagnostic criteria. Both DaT and MIBG were significantly correlated with the Japanese version of the Montreal Cognitive Assessment (MoCA-J) score. CONCLUSION Both DaT and MIBG are important biomarkers for confirming synucleinopathies and/or staging disease progression. Although 95% of iRBD patients were consistent with the body-first subtype concept, alpha-synuclein pathologies of iRBD might have widespread systemic involvement rather than being confined to the lower brainstem, particularly in patients with reduced MoCA-J scores.
Collapse
Affiliation(s)
- Noriko Nishikawa
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Miho Murata
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Taku Hatano
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yohei Mukai
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yuji Saitoh
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Takashi Sakamoto
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Takashi Hanakawa
- Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yuichi Kamei
- Department of Sleep-Wake Disorder, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Psychiatry, Kamisuwa Hospital, Nagano, Japan
| | - Hisateru Tachimori
- Department of Clinical Data Science, Clinical Research & Education Promotion Division, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kenji Hatano
- Department of Clinical Data Science, Clinical Research & Education Promotion Division, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Hiroshi Matsuda
- Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yosuke Taruno
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Nobukatsu Sawamoto
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuta Kajiyama
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kensuke Ikenaka
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuya Kawabata
- Brain and Mind Research Centre, Nagoya University, Nagoya, Japan; Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomohiko Nakamura
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan; Department of Neurology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | | | - Hiroshi Kadotani
- Department of Psychiatry, Shiga University of Medical Science, Shiga, Japan
| | - Yukiyoshi Sumi
- Department of Psychiatry, Shiga University of Medical Science, Shiga, Japan
| | - Yuichi Inoue
- Japan Somnology Center, Neuropsychiatric Research Institute, Tokyo, Japan; Department of Somnology, Tokyo Medical University, Tokyo, Japan
| | - Toshihiro Hayashi
- Department of Neurology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan; Department of Physiology, Teikyo University School of Medicine, Tokyo, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yasushi Shimo
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Neurology, Juntendo University Nerima Hospital, Tokyo, Japan
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hirohisa Watanabe
- Brain and Mind Research Centre, Nagoya University, Nagoya, Japan; Department of Neurology, School of Medicine, Fujita Health University, Aichi, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuji Takahashi
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | |
Collapse
|
7
|
Gamma camera imaging in movement disorders. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00193-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
8
|
Kim K, Kim S, Myung W, Shim I, Lee H, Kim B, Cho SK, Yoon J, Kim DK, Won HH. Shared Genetic Background between Parkinson's Disease and Schizophrenia: A Two-Sample Mendelian Randomization Study. Brain Sci 2021; 11:1042. [PMID: 34439661 PMCID: PMC8393703 DOI: 10.3390/brainsci11081042] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 12/25/2022] Open
Abstract
Background and objectives: Parkinson's disease (PD) and schizophrenia often share symptomatology. Psychotic symptoms are prevalent in patients with PD, and similar motor symptoms with extrapyramidal signs are frequently observed in antipsychotic-naïve patients with schizophrenia as well as premorbid families. However, few studies have examined the relationship between PD and schizophrenia. We performed this study to evaluate whether genetic variants which increase PD risk influence the risk of developing schizophrenia, and vice versa. Materials and Methods: Two-sample Mendelian randomization (TSMR) with summary statistics from large-scale genome-wide association studies (GWAS) was applied. Summary statistics were extracted for these instruments from GWAS of PD and schizophrenia; Results: We found an increase in the risk of schizophrenia per one-standard deviation (SD) increase in the genetically-predicted PD risk (inverse-variance weighted method, odds ratio = 1.10; 95% confidence interval, 1.05-1.15; p = 3.49 × 10-5). The association was consistent in sensitivity analyses, including multiple TSMR methods, analysis after removing outlier variants with potential pleiotropic effects, and analysis after applying multiple GWAS subthresholds. No relationships were evident between PD and smoking or other psychiatric disorders, including attention deficit hyperactivity disorder, autism spectrum disorder, bipolar affective disorder, major depressive disorder, Alzheimer's disease, or alcohol dependence. However, we did not find a reverse relationship; genetic variants increasing schizophrenia risk did not alter the risk of PD; Conclusions: Overall, our findings suggest that increased genetic risk of PD can be associated with increased risk of schizophrenia. This association supports the intrinsic nature of the psychotic symptom in PD rather than medication or environmental effects. Future studies for possible comorbidities and shared genetic structure between the two diseases are warranted.
Collapse
Affiliation(s)
- Kiwon Kim
- Department of Psychiatry, Kangdong Sacred Heart Hospital, College of Medicine, Hallym University, Sungan-ro, Kangdong-gu, Seoul 05355, Korea;
| | - Soyeon Kim
- Samsung Medical Center, Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Korea; (S.K.); (I.S.); (B.K.)
| | - Woojae Myung
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam 13620, Korea;
| | - Injeong Shim
- Samsung Medical Center, Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Korea; (S.K.); (I.S.); (B.K.)
| | - Hyewon Lee
- Department of Health Administration and Management, College of Medical Sciences, Soonchunhyang University, Asan 31538, Korea;
- Department of Software Convergence, Graduate School, Soonchunhyang University, Asan 31538, Korea
| | - Beomsu Kim
- Samsung Medical Center, Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Korea; (S.K.); (I.S.); (B.K.)
| | - Sung Kweon Cho
- Department of Pharmacology, School of Medicine, Ajou University, Worldcup-ro, Yeongtong-gu, Suwon 16499, Korea;
| | - Joohyun Yoon
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam 13620, Korea;
| | - Doh Kwan Kim
- Samsung Medical Center, Department of Psychiatry, School of Medicine, Sungkyunkwan University, Seoul 06351, Korea;
| | - Hong-Hee Won
- Samsung Medical Center, Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Korea; (S.K.); (I.S.); (B.K.)
| |
Collapse
|
9
|
Ma X, Li S, Li C, Wang R, Chen M, Chen H, Su W. Total Cerebral Small Vessel Score Association With Hoehn and Yahr Stage in Parkinson's Disease. Front Aging Neurosci 2021; 13:682776. [PMID: 34122053 PMCID: PMC8192831 DOI: 10.3389/fnagi.2021.682776] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
Background: This study aimed to evaluate the total cerebral small vessel disease (CSVD) score in patients with Parkinson’s disease (PD) at different stages and related factors. Methods: A 100 and seven patients with idiopathic PD and 62 normal controls (NCs) who underwent brain magnetic resonance imaging (MRI) were enrolled. PD patients were divided into two groups: early PD [(Hoehn and Yahr (H&Y) 1–1.5, n = 36)] and advanced PD (H&Y 2–4, n = 71) groups. We calculated the total CSVD score for each participant based on lacunes, high-grade white matter hyperintensities (WMH), enlarged perivascular spaces (EPVS), and cerebral microbleeds (CMBs). Differences in total CSVD score between the PD and NCs and between the two subgroups were compared. In addition, a multivariate logistic regression analysis was conducted to investigate the association between CSVD markers and clinical variables in PD. Results: Lacunes were found in 9.3% of patients with PD, periventricular WMH (PVWMH) in 89.7%, deep WMH (DWMH) in 81.3%, EPVS in 85%, and CMBs in 2.8%. Compared with NCs, patients with PD showed higher PVWMH and DWMH scores. Advanced PD patients exhibited greater PVWMH (P = 0.041), DWMH (P = 0.046), and total CSVD score (P = 0.044) than the early PD group. After adjusting for multiple variables, higher H&Y stage was independently correlated with increased total CSVD score (OR = 2.667, 95% CI 1.154–2.266) and PVWMH score (OR = 2.237, 95% CI 1.084–1.696). Conclusions: CSVD may play a critical role in patients with PD. The total CSVD score is a potential neuroimaging marker for monitoring the progression of PD.
Collapse
Affiliation(s)
- Xinxin Ma
- Department of Neurology, Parkinson's Disease and Extra Pyramidal Disease Diagnosis and Treatment Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuhua Li
- Department of Neurology, Parkinson's Disease and Extra Pyramidal Disease Diagnosis and Treatment Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Chunmei Li
- Department of Radiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Rui Wang
- Department of Radiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Min Chen
- Department of Radiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Haibo Chen
- Department of Neurology, Parkinson's Disease and Extra Pyramidal Disease Diagnosis and Treatment Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Wen Su
- Department of Neurology, Parkinson's Disease and Extra Pyramidal Disease Diagnosis and Treatment Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Valli M, Cho SS, Masellis M, Chen R, Koshimori Y, Diez-Cirarda M, Mihaescu A, Christopher L, Strafella AP. Extra-striatal dopamine in Parkinson's disease with rapid eye movement sleep behavior disorder. J Neurosci Res 2021; 99:1177-1187. [PMID: 33470445 DOI: 10.1002/jnr.24779] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 11/05/2022]
Abstract
Rapid eye movement sleep behavior disorder (RBD) is a common condition found in more than 50% of the patients with Parkinson's disease (PD). Molecular imaging shows that PD with RBD (PD-RBD+) have lower striatal dopamine transporter activity within the caudate and putamen relative to PD without RBD (PD-RBD-). However, the characterization of the extra-striatal dopamine within the mesocortical and mesolimbic pathways remains unknown. We aim to elucidate this with PET imaging in 15 PD-RBD+ and 15 PD-RBD- patients, while having 15 age-matched healthy controls (HC). Each participant underwent a single PET scan with [11 C]FLB-457 to detect the D2 receptor availability within the extra-striatal regions of interest (ROI), including the prefrontal, temporal, and limbic areas. [11 C]FLB-457 retention was expressed as the nondisplaceable binding potential. Our results reveal that relative to HC, PD-RBD+ and PD-RBD- patients have lower levels of D2 receptor availability within the uncus parahippocampus, superior, lateral, and inferior temporal cortex. PD-RBD+ showed steep decline in D2 receptors within the left uncus parahippocampus with increasing disease severity, but this was not observed for PD-RBD- patients. Findings imply that extra-striatal dopaminergic system may play a role in contributing to symptomatic progress in PD patients with RBD. However, validation with more advanced PD patients are needed.
Collapse
Affiliation(s)
- Mikaeel Valli
- Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, ON, Canada.,Krembil Research Institute, University Health Network, University of Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, ON, Canada
| | - Sang Soo Cho
- Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, ON, Canada.,Krembil Research Institute, University Health Network, University of Toronto, ON, Canada
| | - Mario Masellis
- Institute of Medical Science, University of Toronto, ON, Canada.,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Robert Chen
- Krembil Research Institute, University Health Network, University of Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, ON, Canada.,Morton and Gloria Shulman Movement Disorder Unit & E.J. Safra Parkinson Disease Program, Neurology Division, Department of Medicine, Toronto Western Hospital, UHN, University of Toronto, ON, Canada
| | - Yuko Koshimori
- Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, ON, Canada.,Music and Health Research Collaboratory (MaRC), Faculty of Music, University of Toronto, Toronto, ON, Canada
| | - Maria Diez-Cirarda
- Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, ON, Canada.,Krembil Research Institute, University Health Network, University of Toronto, ON, Canada.,Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Alexander Mihaescu
- Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, ON, Canada.,Krembil Research Institute, University Health Network, University of Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, ON, Canada
| | - Leigh Christopher
- Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, ON, Canada.,Krembil Research Institute, University Health Network, University of Toronto, ON, Canada
| | - Antonio P Strafella
- Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, ON, Canada.,Krembil Research Institute, University Health Network, University of Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, ON, Canada.,Morton and Gloria Shulman Movement Disorder Unit & E.J. Safra Parkinson Disease Program, Neurology Division, Department of Medicine, Toronto Western Hospital, UHN, University of Toronto, ON, Canada
| |
Collapse
|
11
|
The Effect of Caffeine on the Risk and Progression of Parkinson's Disease: A Meta-Analysis. Nutrients 2020; 12:nu12061860. [PMID: 32580456 PMCID: PMC7353179 DOI: 10.3390/nu12061860] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 12/21/2022] Open
Abstract
Coffee and caffeine are speculated to be associated with the reduced risk of Parkinson's disease (PD). The present study aimed to investigate the disease-modifying potential of caffeine on PD, either for healthy people or patients, through a meta-analysis. The electronic databases were searched using terms related to PD and coffee and caffeinated food products. Articles were included only upon fulfillment of clear diagnostic criteria for PD and details regarding their caffeine content. Reference lists of relevant articles were reviewed to identify eligible studies not shortlisted using these terms. In total, the present study enrolled 13 studies, nine were categorized into a healthy cohort and the rest into a PD cohort. The individuals in the healthy cohort with regular caffeine consumption had a significantly lower risk of PD during follow-up evaluation (hazard ratio (HR) = 0.797, 95% CI = 0.748-0.849, p < 0.001). The outcomes of disease progression in PD cohorts included dyskinesia, motor fluctuation, symptom onset, and levodopa initiation. Individuals consuming caffeine presented a significantly lower rate of PD progression (HR = 0.834, 95% CI = 0.707-0.984, p = 0.03). In conclusion, caffeine modified disease risk and progression in PD, among both healthy individuals or those with PD. Potential biological benefits, such as those obtained from adenosine 2A receptor antagonism, may require further investigation for designing new drugs.
Collapse
|
12
|
Lopez G, Eisenberg DP, Gregory MD, Ianni AM, Grogans SE, Masdeu JC, Kim J, Groden C, Sidransky E, Berman KF. Longitudinal Positron Emission Tomography of Dopamine Synthesis in Subjects with GBA1 Mutations. Ann Neurol 2020; 87:652-657. [PMID: 32030791 DOI: 10.1002/ana.25692] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/22/2020] [Accepted: 02/02/2020] [Indexed: 11/11/2022]
Abstract
Mutations in GBA1, the gene mutated in Gaucher disease, are a common genetic risk factor for Parkinson disease, although the penetrance is low. We performed [18 F]-fluorodopa positron emission tomography studies of 57 homozygous and heterozygous GBA1 mutation carriers (15 with parkinsonism) and 98 controls looking for early indications of dopamine loss using voxelwise analyses to identify group differences in striatal [18 F]-fluorodopa uptake (Ki ). Forty-eight subjects were followed longitudinally. Cross-sectional and longitudinal comparisons of Ki and Ki change found significant effects of Parkinson disease. However, at baseline and over time, striatal [18 F]-fluorodopa uptake in mutation carriers without parkinsonism did not significantly differ from controls. ANN NEUROL 2020;87:652-657.
Collapse
Affiliation(s)
- Grisel Lopez
- Section on Molecular Neurogenetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Daniel P Eisenberg
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD
| | - Michael D Gregory
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD
| | - Angela M Ianni
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD
| | - Shannon E Grogans
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD
| | - Joseph C Masdeu
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD
| | - Jenny Kim
- Section on Molecular Neurogenetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Catherine Groden
- Section on Molecular Neurogenetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Ellen Sidransky
- Section on Molecular Neurogenetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Karen F Berman
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD
| |
Collapse
|
13
|
Miyamoto M, Miyamoto T, Saitou J, Sato T. Longitudinal study of striatal aromatic l-amino acid decarboxylase activity in patients with idiopathic rapid eye movement sleep behavior disorder. Sleep Med 2020; 68:50-56. [PMID: 32028226 DOI: 10.1016/j.sleep.2019.09.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/15/2019] [Accepted: 09/20/2019] [Indexed: 12/20/2022]
Abstract
STUDY OBJECTIVES To determine if nigrostriatal dopaminergic system function, evaluated by aromatic l-amino acid decarboxylase (AADC) activity using 6-[18F]fluoro-meta-tyrosine brain positron emission tomography (FMT-PET) can accurately and efficiently identify idiopathic rapid-eye-movement behavior disorder (IRBD) individuals at risk for conversion to a clinical diagnosis of Parkinson's disease (PD) or dementia with Lewy bodies (DLB). METHODS We assessed prospectively striatal aromatic l-amino acid decarboxylase activity using FMT brain PET imaging in IRBD patients who were followed systematically every 1-3 months for 1-10 years. IRBD patients (n = 27) were enrolled in this prospective cohort study starting in 2009. Those who underwent follow-up scans between January 2011 and September 2014 (n = 24) were analyzed in the present study. RESULTS Of the 24 IRBD patients with baseline and follow-up FMT-PET scans, 11 (45.8%) developed PD (n = 6) or DLB (n = 5). Compared to IRBD patients who were still disease-free, those who developed PD (n = 5) or DLB with parkinsonism (n = 1) had significantly reduced bilateral putaminal FMT uptake during the follow-up. Furthermore, the rate of FMT decline between baseline and follow-up scans was higher in all converted patients, even for those with DLB without parkinsonism, than in IRBD patients who remained disease-free. CONCLUSIONS FMT-PET, which represents a dynamic change in AADC activity over time, may also be a useful predictor for the risk of conversion to PD or DLB over short-term clinical follow-up periods, or when testing neuroprotective and restorative strategies in the prodromal phases of PD or DLB.
Collapse
Affiliation(s)
- Masayuki Miyamoto
- Department of Neurology, Center of Sleep Medicine, Dokkyo Medical University, Japan
| | - Tomoyuki Miyamoto
- Department of Neurology, Dokkyo Medical University Saitama Medical Center, Japan.
| | | | | |
Collapse
|
14
|
Abstract
Positron emission tomography (PET) has revealed key insights into the pathophysiology of movement disorders. This paper will focus on how PET investigations of pathophysiology are particularly relevant to Parkinson disease, a neurodegenerative condition usually starting later in life marked by a varying combination of motor and nonmotor deficits. Various molecular imaging modalities help to determine what changes in brain herald the onset of pathology; can these changes be used to identify presymptomatic individuals who may be appropriate for to-be-developed treatments that may forestall onset of symptoms or slow disease progression; can PET act as a biomarker of disease progression; can molecular imaging help enrich homogenous cohorts for clinical studies; and what other pathophysiologic mechanisms relate to nonmotor manifestations. PET methods include measurements of regional cerebral glucose metabolism and blood flow, selected receptors, specific neurotransmitter systems, postsynaptic signal transducers, and abnormal protein deposition. We will review each of these methodologies and how they are relevant to important clinical issues pertaining to Parkinson disease.
Collapse
Affiliation(s)
- Baijayanta Maiti
- Department of Neurology, Washington University in St. Louis, St Louis, MO.
| | - Joel S Perlmutter
- Department of Neurology, Washington University in St. Louis, St Louis, MO; Department of Radiology, Washington University in St. Louis, St Louis, MO; Department of Neuroscience, Washington University in St. Louis, St Louis, MO; Department of Physical Therapy, Washington University in St. Louis, St Louis, MO; Department of Occupational Therapy, Washington University in St. Louis, St Louis, MO
| |
Collapse
|
15
|
Differential effects of levodopa and apomorphine on neuronal population oscillations in the cortico-basal ganglia loop circuit in vivo in experimental parkinsonism. Exp Neurol 2017; 298:122-133. [PMID: 28893517 DOI: 10.1016/j.expneurol.2017.09.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/03/2017] [Accepted: 09/07/2017] [Indexed: 11/21/2022]
Abstract
The current pharmacotherapy of Parkinson's disease (PD) is primarily based on two classes of drugs: dopamine precursors, namely levodopa, and dopamine receptor agonists, such as apomorphine. Although both types of agents exert their beneficial clinical effects on motor and non-motor symptoms in PD via dopamine receptors, clinical efficiency and side effects differ substantially between levodopa and apomorphine. Levodopa can provide a greater symptomatic relief than dopamine receptor agonists. However, because long-term levodopa use is associated with early debilitating motor fluctuations, dopamine receptor agonists are often recommended in younger patients. The pharmacodynamic basis of these profound differences is incompletely understood. It has been hypothesized that levodopa and dopamine receptor agonists may have diverging effects on beta and gamma oscillations that have been shown to be of importance for the pathophysiology of PD. Here, we used electrophysiological recordings in anesthetized dopamine-intact and dopamine-depleted rats to systemically compare the impact of levodopa or apomorphine on neuronal population oscillations in three nodes of the cortico-basal ganglia loop circuit. Our results showed that levodopa had a higher potency than apomorphine to suppress the abnormal beta oscillations often associated with bradykinesia while simultaneously enhancing the gamma oscillations often associated with increased movement. Our data suggests that the higher clinical efficacy of levodopa as well as some of its side effects, as e.g. dyskinesias may be based on its characteristic ability to modulate beta-/gamma-oscillation dynamics in the cortico-basal ganglia loop circuit.
Collapse
|
16
|
Sung C, Lee JH, Oh JS, Oh M, Lee SJ, Oh SJ, Chung SJ, Lee CS, Kim JS. Longitudinal Decline of Striatal Subregional [ 18F]FP-CIT Uptake in Parkinson's Disease. Nucl Med Mol Imaging 2017; 51:304-313. [PMID: 29242724 DOI: 10.1007/s13139-017-0481-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 03/20/2017] [Accepted: 03/24/2017] [Indexed: 10/19/2022] Open
Abstract
Purpose Dopamine transporter imaging is suggested to be a useful imaging biomarker for Parkinson's disease (PD) progression and monitoring drug effects. We investigated the longitudinal decline characteristics of striatal [18F]FP-CIT uptake in PD. Methods We retrospectively reviewed 35 PD patients and 9 non-PD patients. All patients underwent [18F]FP-CIT PET at the initial diagnosis and follow-up. PET images were spatially normalized and analyzed with eight striatal and one occipital VOI templates. We measured the specific to non-specific binding ratio (SNBR) of the striatal subregions and calculated the absolute annual reduction (AAR) and relative annual reduction (%RAR) of the SNBRs. Results Total striatal SNBRs in PD patients were significantly lower than those in non-PD patients, with the most significant difference in the posterior putamen. Both AAR (0.26 ± 0.14 vs. 0.09 ± 0.19, p < 0.05) and %RAR (6.9 ± 3.5 vs. 1.2 ± 2.7, p < 0.001) of total striatal SNBRs were significantly greater in PD than non-PD patients. There were no significant differences in the AAR and %RAR of total striatal SNBRs between elderly and young onset PD. The AARs of the posterior putamen were higher in early PD than in advanced PD. Conversely, the %RARs were not significantly different between early and more advanced PD. The disease duration was significantly negatively correlated with the AAR but not with the %RAR of the posterior putamen. Conclusions The longitudinal decline of striatal [18F]FP-CIT uptake in PD was nonlinear and significantly faster than that in non-PD, with a different rate of decline among the striatal subregions.
Collapse
Affiliation(s)
- Changhwan Sung
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympicro 43-gil, Songpa-gu, Seoul, 05505 South Korea
| | - Jai Hyuen Lee
- Department of Nuclear Medicine, Dankook University College of Medicine, Cheonan, South Korea
| | - Jungsu S Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympicro 43-gil, Songpa-gu, Seoul, 05505 South Korea
| | - Minyoung Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympicro 43-gil, Songpa-gu, Seoul, 05505 South Korea
| | - Sang Ju Lee
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympicro 43-gil, Songpa-gu, Seoul, 05505 South Korea
| | - Seung Jun Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympicro 43-gil, Songpa-gu, Seoul, 05505 South Korea
| | - Sun Ju Chung
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Chong Sik Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jae Seung Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympicro 43-gil, Songpa-gu, Seoul, 05505 South Korea
| |
Collapse
|
17
|
Abstract
In recent decades, identification of the dopaminergic deficit in Parkinson's disease has spawned an explo sion of research in the molecular neurobiology of the basal ganglia. In vivo imaging has provided a tool to bridge developments in basic neuroscience and clinicial neurology. Imaging studies have opened a unique window on the neurochemical pathophysiology of Parkinson's disease and more specifically on the onset, progression and physiology of the degenerative process. As we are poised on the brink of new protective and restorative therapies for Parkinson's disease, the potential of imaging to teach us about in vivo brain neurochemistry offers both promise and challenge. NEUROSCIENTIST 5:333-340, 1999
Collapse
Affiliation(s)
- Kenneth Marek
- Department of Neurology Yale University School of Medicine
New Haven, Connecticut
| |
Collapse
|
18
|
Zou J, Weng RH, Chen ZY, Wei XB, Wang R, Chen D, Xia Y, Wang Q. Position Emission Tomography/Single-Photon Emission Tomography Neuroimaging for Detection of Premotor Parkinson's Disease. CNS Neurosci Ther 2016; 22:167-77. [PMID: 26776081 DOI: 10.1111/cns.12493] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 11/12/2015] [Accepted: 11/14/2015] [Indexed: 12/21/2022] Open
Abstract
Premotor Parkinson's disease (PD) refers to a prodromal stage of Parkinson's disease (PD) during which nonmotor clinical features may be present. Currently, it is difficult to make an early diagnosis for premotor PD. Molecular imaging with position emission tomography (PET) or single-photon emission tomography (SPECT) offers a wide variety of tools for overcoming this difficulty. Indeed, molecular imaging techniques may play a crucial role in diagnosing, monitoring and evaluating the individuals with the risk for PD. For example, dopaminergic dysfunctions can be identified by detecting the expression of vesicular monoamine transporter (VMAT2) and aromatic amino acid decarboxylase (AADC) to evaluate the conditions of dopaminergic terminals functions in high-risk individuals of PD. This detection provides a sensitive and specific measurement of nonmotor symptoms (NMS) such as olfactory dysfunction, sleep disorders, and psychiatric symptoms in the high-risk patients, especially at the premotor phase. Molecular imaging technique is capable of detecting the dysfunction of serotonergic, noradrenergic, and cholinergic systems that are typically associated with premotor manifestations. This review discusses the importance of SPECT/PET applications in the detection of premotor markers preceding motor abnormalities with highlighting their great potential for early and accurate diagnosis of premotor symptoms of PD and its scientific significance.
Collapse
Affiliation(s)
- Jing Zou
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Rui-Hui Weng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhao-Yu Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiao-Bo Wei
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Rui Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Dan Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Ying Xia
- Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, TX, USA
| | - Qing Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Skoloudík D, Jelínková M, Blahuta J, Cermák P, Soukup T, Bártová P, Langová K, Herzig R. Transcranial sonography of the substantia nigra: digital image analysis. AJNR Am J Neuroradiol 2014; 35:2273-8. [PMID: 25059698 DOI: 10.3174/ajnr.a4049] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Increased echogenicity of the substantia nigra is a typical transcranial sonography finding in Parkinson disease. Experimental software for digital analysis of the echogenic substantia nigra area has been developed. The aim of this study was to compare the evaluation of substantia nigra echogenicity by using digital analysis with a manual measurement in patients with Parkinson disease and healthy volunteers. MATERIALS AND METHODS One hundred thirteen healthy volunteers were enrolled in the derivation cohort, and 50 healthy volunteers and 30 patients with Parkinson disease, in the validation cohort. The substantia nigra was imaged from the right and left temporal bone window by using transcranial sonography. All subjects were examined twice by using different sonographic machines by an experienced sonographer. DICOM images of the substantia nigra were encoded; then, digital analysis and manual measurement of the substantia nigra were performed. The 90th percentile of the derivation cohort values was used as a cut-point for the evaluation of the hyperechogenic substantia nigra in the validation cohort. The Spearman coefficient was used for assessment of the correlation between both measurements. The Cohen κ coefficient was used for the assessment of the correlation between both measurements and Parkinson disease diagnosis. RESULTS The Spearman coefficient between measurements by using different machines was 0.686 for digital analysis and 0.721 for manual measurement (P < .0001). Hyperechogenic substantia nigra was detected in the same 26 (86.7%) patients with Parkinson disease by using both measurements. Cohen κ coefficients for digital analysis and manual measurement were 0.787 and 0.762, respectively (P < .0001). CONCLUSIONS The present study showed comparable results when measuring the substantia nigra features conventionally and by using the developed software.
Collapse
Affiliation(s)
- D Skoloudík
- From the Faculty of Health Science (D.Š.), Palacký University Olomouc, Olomouc, Czech Republic Department of Neurology (D.Š., M.J., P.B.), University Hospital Ostrava, Ostrava, Czech Republic
| | - M Jelínková
- Department of Neurology (D.Š., M.J., P.B.), University Hospital Ostrava, Ostrava, Czech Republic Department of Neurology (M.J.), Hospital, Karviná-Ráj, Karviná, Czech Republic
| | - J Blahuta
- Institute of Computer Science (J.B., P.Č., T.S.), Faculty of Philosophy and Science, Silesian University in Opava, Opava, Czech Republic
| | - P Cermák
- Institute of Computer Science (J.B., P.Č., T.S.), Faculty of Philosophy and Science, Silesian University in Opava, Opava, Czech Republic
| | - T Soukup
- Institute of Computer Science (J.B., P.Č., T.S.), Faculty of Philosophy and Science, Silesian University in Opava, Opava, Czech Republic
| | - P Bártová
- Department of Neurology (D.Š., M.J., P.B.), University Hospital Ostrava, Ostrava, Czech Republic
| | - K Langová
- Department of Biophysics (K.L.), Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacký University, Olomouc, Czech Republic
| | - R Herzig
- Department of Neurology (R.H.), Charles University Faculty of Medicine and University Hospital Hradec Králové, Hradec Králové, Czech Republic
| |
Collapse
|
20
|
Kim E, Howes OD, Kapur S. Molecular imaging as a guide for the treatment of central nervous system disorders. DIALOGUES IN CLINICAL NEUROSCIENCE 2014. [PMID: 24174903 PMCID: PMC3811103 DOI: 10.31887/dcns.2013.15.3/ekim] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Molecular imaging techniques have a number of advantages for research into the pathophysiology and treatment of central nervous system (CNS) disorders. Firstly, they provide a noninvasive means of characterizing physiological processes in the living brain, enabling molecular alterations to be linked to clinical changes. Secondly, the pathophysiological target in a given CNS disorder can be measured in animal models and in experimental human models in the same way, which enables translational research. Moreover, as molecular imaging facilitates the detection of functional change which precedes gross pathology, it is particularly useful for the early diagnosis and treatment of CNS disorders. This review considers the application of molecular imaging to CNS disorders focusing on its potential to inform the development and evaluation of treatments. We focus on schizophrenia, Parkinson's disease, depression, and dementia as major CNS disorders. We also review the potential of molecular imaging to guide new drug development for CNS disorders.
Collapse
Affiliation(s)
- Euitae Kim
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Gyeonggi-do 463-707, Korea
| | | | | |
Collapse
|
21
|
Krismer F, Jellinger KA, Scholz SW, Seppi K, Stefanova N, Antonini A, Poewe W, Wenning GK. Multiple system atrophy as emerging template for accelerated drug discovery in α-synucleinopathies. Parkinsonism Relat Disord 2014; 20:793-9. [PMID: 24894118 PMCID: PMC4141743 DOI: 10.1016/j.parkreldis.2014.05.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 04/27/2014] [Accepted: 05/07/2014] [Indexed: 12/21/2022]
Abstract
There is evidence that the α-synucleinopathies Parkinson's disease (PD) and the Parkinson variant of multiple system atrophy (MSA-P) overlap at multiple levels. Both disorders are characterized by deposition of abnormally phosphorylated fibrillar α-synuclein within the central nervous system suggesting shared pathophysiological mechanisms. Despite the considerable clinical overlap in the early disease stages, MSA-P, in contrast to PD, is fatal and rapidly progressive. Moreover recent clinical studies have shown that surrogate markers of disease progression can be quantified easily and may reliably depict the rapid course of MSA. We therefore posit that, MSA-P may be exploited as a filter barrier in the development of disease-modifying therapeutic strategies targeting common pathophysiological mechanisms of α-synucleinopathies. This approach might reduce the number of negative phase III clinical trials, and, in turn, shift the available resources to earlier development stages, thereby increasing the number of candidate compounds validated. α-synucleinopathies overlap at multiple levels. α-synucleinopathies are characterized by an abnormal deposition of α-synuclein. Validated surrogate markers in MSA reliably monitor disease progression. MSA may serve as a template disease for other α-synucleinopathies.
Collapse
Affiliation(s)
- Florian Krismer
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria.
| | | | - Sonja W Scholz
- Department of Neurology, The Johns Hopkins Hospital, Baltimore, MD 21287, USA.
| | - Klaus Seppi
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria.
| | - Nadia Stefanova
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria.
| | - Angelo Antonini
- Department of Parkinson's Disease and Movement Disorders, IRCCS San Camillo, Venice, Italy.
| | - Werner Poewe
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria.
| | - Gregor K Wenning
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria.
| |
Collapse
|
22
|
Felicio AC, Dinelle K, Agarwal PA, McKenzie J, Heffernan N, Road JD, Appel-Cresswell S, Wszolek ZK, Farrer MJ, Schulzer M, Sossi V, Stoessl AJ. In vivo dopaminergic and serotonergic dysfunction in DCTN1 gene mutation carriers. Mov Disord 2014; 29:1197-201. [PMID: 24797316 DOI: 10.1002/mds.25893] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 03/10/2014] [Accepted: 03/25/2014] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION We used positron emission tomography (PET) to assess dopaminergic and serotonergic terminal density in three subjects carrying a mutation in the DCT1 gene, two clinically affected with Perry syndrome. METHODS All subjects had brain imaging using 18F-6-fluoro-l-dopa (FDOPA, dopamine synthesis and storage), (+)-11C-dihydrotetrabenazine (DTBZ, vesicular monoamine transporter type 2), and 11C-raclopride (RAC, dopamine D2/D3 receptors). One subject also underwent PET with 11C-3-amino-4-(2-dimethylaminomethyl-phenylsulfanyl)-benzonitrile (DASB, serotonin transporter). RESULTS FDOPA-PET and DTBZ-PET in the affected individuals showed a reduction of striatal tracer uptake. Also, RAC-PET showed higher uptake in these area. DASB-PET showed significant uptake changes in left orbitofrontal cortex, bilateral anterior insula, left dorsolateral prefrontal cortex, left orbitofrontal cortex, left posterior cingulate cortex, left caudate, and left ventral striatum. CONCLUSIONS Our data showed evidence of both striatal dopaminergic and widespread cortical/subcortical serotonergic dysfunctions in individuals carrying a mutation in the DCTN1 gene.
Collapse
Affiliation(s)
- Andre C Felicio
- Pacific Parkinson's Research Centre, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Potential impact of self-perceived prodromal symptoms on the early diagnosis of Parkinson’s disease. J Neurol 2013; 260:3077-85. [DOI: 10.1007/s00415-013-7125-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 09/16/2013] [Accepted: 09/17/2013] [Indexed: 10/26/2022]
|
24
|
Levodopa induces long-lasting modification in the functional activity of the nigrostriatal pathway. Neurobiol Dis 2013; 62:250-9. [PMID: 24076099 DOI: 10.1016/j.nbd.2013.09.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 09/13/2013] [Accepted: 09/17/2013] [Indexed: 12/29/2022] Open
Abstract
Much controversy exists concerning the effect of levodopa on striatal dopaminergic markers in Parkinson's disease (PD) and its influence on functional neuroimaging. To deal with this issue we studied the impact of neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and chronic levodopa administration on striatal (18)F-DOPA uptake (Ki) in an animal model of PD. The levels of several striatal dopaminergic markers and the number of surviving dopaminergic neurons in the substantia nigra (SN) were also assessed. Eleven Macaca fascicularis were included in the study. Eight animals received weekly intravenous injections of MPTP for 7weeks and 3 intact animals served as controls. MPTP-monkeys were divided in two groups. Group I was treated with placebo while Group II received levodopa. Both treatments were maintained for 11months and then followed by a washout period of 6months. (18)F-DOPA positron emission tomography (PET) scans were performed at baseline, after MPTP intoxication, following 11months of treatment, and after a washout period of 1, 3 and 6months. Monkeys were sacrificed 6months after concluding either placebo or levodopa treatment and immediately after the last (18)F-DOPA PET study. Striatal dopamine transporter (DAT) content, tyrosine hydroxylase (TH) content and aromatic l-amino acid decarboxylase (AADC) content were assessed. In Group II (18)F-DOPA PET studies performed at 3 and 6months after interrupting levodopa showed a significantly increased Ki in the anterior putamen as compared to Group I. Levodopa and placebo treated animals exhibited a similar number of surviving dopaminergic cells in the SN. Striatal DAT content was equally reduced in both groups of animals. Animals in Group I exhibited a significant decrease in TH protein content in all the striatal regions assessed. However, in Group II, TH levels were significantly reduced only in the anterior and posterior putamen. Surprisingly, in the levodopa-treated animals the TH levels in the posterior putamen were significantly lower than those in the placebo group. AADC levels in MPTP groups were similar to those of control animals in all striatal areas analyzed. This study shows that chronic levodopa administration to monkeys with partial nigrostriatal degeneration followed by a washout period induces modifications in the functional activity of the dopaminergic nigrostriatal pathway.
Collapse
|
25
|
Abstract
The use of functional imaging in neurodegenerative diseases has increased in recent years, with applications in research into the underlying pathophysiology, aiding in diagnosis, or evaluating new treatments. In Parkinson's disease (PD), these imaging methods have expanded our understanding of the disease beyond dopaminergic deficits. Moreover, functional imaging methods have described alterations in functional networks relating not only to the motor symptoms, but also to many nonmotor features of PD, such as cognitive dysfunction. From a clinical viewpoint, functional imaging methods can assist in monitoring disease progression, such as in the context of clinical trials, and holds the potential to aid in early diagnosis of PD and differentiation from other parkinsonian disorders.
Collapse
|
26
|
Substantia nigra hyperechogenicity in healthy controls: a [¹⁸Fluoro] Dopa-PET follow-up study. J Neurol 2013; 260:1907-11. [PMID: 23564333 DOI: 10.1007/s00415-013-6904-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Revised: 03/22/2013] [Accepted: 03/25/2013] [Indexed: 10/27/2022]
Abstract
In order to assess nigrostriatal function over time in healthy subjects with substantia nigra hyperechogenicity (SN+) believed to be at higher risk for Parkinson's disease (PD), ten healthy SN+ subjects underwent [(18)Fluoro] Dopa positron emission tomography (PET) twice-at baseline and after a mean of 7.3 years. Neurological examination took place at study inclusion followed by a structured telephone interview focusing on early Parkinsonian symptoms at the time point of second PET study and 3.5 years later. The [(18)Fluoro] Dopa uptake remained unchanged in eight of ten participants. In the other two subjects marked unilateral reduction of putaminal [(18)Fluoro] Dopa uptake ratios appeared over the time, followed by complaints of a clinically manifest PD in one. We suggest that the progressive pathological PET findings in 20 % and PD conversion in 10 % of our cohort may be in accordance with the presumed proportion of SN+ individuals eventually developing PD based on SN+ prevalence of 10 % within the healthy population, being ten times higher than PD prevalence in the age of over 60 years. Our findings hint towards a significance of SN+ indicating a high risk for PD in some extrapyramidally healthy SN+ individuals.
Collapse
|
27
|
Halliday GM, McRitchie DA, Cartwright H, Pamphlett R, Hely MA, Morris JG. Midbrain neuropathology in idiopathic Parkinson's disease and diffuse Lewy body disease. J Clin Neurosci 2012; 3:52-60. [PMID: 18644264 DOI: 10.1016/s0967-5868(96)90083-1] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/1995] [Accepted: 08/11/1995] [Indexed: 11/30/2022]
Abstract
We have quantified midbrain cell loss in idiopathic Parkinson's disease (PD) compared with controls; six patients had PD with onset before 70 years, five patients had late onset PD (>70 years) and nine patients had diffuse Lewy body disease. The pattern of cell loss in these last two groups has not been previously described. No age associated neuronal loss was seen in controls. There was cell loss and reduced area of the pars compacta in all cases but no difference in the pattern of cell loss, which was predominantly ventral. The amount of cell loss in the dorsolateral cluster correlated with the duration of Parkinsonian symptoms, while greater cell loss in the dorsomedial cluster correlated with the presence of tremor and the absence of early dementia. These results suggest that the topography of midbrain pathology does not assist in differentiating these overlapping syndromes.
Collapse
Affiliation(s)
- G M Halliday
- Prince of Wales Medical Research Institute, Randwick, Australia; Neuropathology Division, Department of Pathology, University of Sydney, Australia
| | | | | | | | | | | |
Collapse
|
28
|
Abstract
A 68-year-old, normal female volunteer was scanned by positron emission tomography (PET) with [(18)F]6-L-fluorodopa (FD). Striatal FD uptake was in the high normal range. Subsequently, she developed parkinsonism 3.7 years after the scan. A repeat FD PET scan revealed a significant reduction of FD uptake by 20% over the 5.2 year interval. Our observations suggest a relatively short presymptomatic period with fast initial losses of nigral neurons in idiopathic parkinsonism.
Collapse
|
29
|
Reichmann H, Emre M. Optimizing levodopa therapy to treat wearing-off symptoms in Parkinson's disease: focus on levodopa/carbidopa/entacapone. Expert Rev Neurother 2012; 12:119-31. [PMID: 22288667 DOI: 10.1586/ern.11.203] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Levodopa has been the mainstay of Parkinson's disease (PD) therapy for over 40 years, with its efficacy surpassing that of other antiparkinsonian medications. As such, most PD patients eventually require levodopa-based therapy during the course of the disease. However, despite its proven efficacy, long-term levodopa therapy is associated with motor complications, with wearing-off being the most prevalent. Wearing-off occurs, in part, as a result of the short half-life of levodopa, which leads to fluctuations in plasma levodopa levels. A pharmacokinetic profile characterized by a higher trough value of levodopa can be achieved by combining levodopa/carbidopa with entacapone, which inhibits the peripheral breakdown of levodopa, resulting in higher plasma levodopa levels. Here, we review the limitations of conventional levodopa and the clinical data for levodopa/carbidopa/entacapone in treating patients with wearing-off.
Collapse
Affiliation(s)
- Heinz Reichmann
- Department of Neurology, University of Dresden, Dresden, Germany.
| | | |
Collapse
|
30
|
Postuma RB, Gagnon JF, Montplaisir JY. REM sleep behavior disorder: From dreams to neurodegeneration. Neurobiol Dis 2012; 46:553-8. [DOI: 10.1016/j.nbd.2011.10.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 09/22/2011] [Accepted: 10/06/2011] [Indexed: 02/05/2023] Open
|
31
|
Postuma RB, Lang AE, Gagnon JF, Pelletier A, Montplaisir JY. How does parkinsonism start? Prodromal parkinsonism motor changes in idiopathic REM sleep behaviour disorder. Brain 2012; 135:1860-70. [PMID: 22561644 DOI: 10.1093/brain/aws093] [Citation(s) in RCA: 232] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Parkinsonism, as a gradually progressive disorder, has a prodromal interval during which neurodegeneration has begun but cardinal manifestations have not fully developed. A systematic direct assessment of this interval has never been performed. Since patients with idiopathic REM sleep behaviour disorder are at very high risk of parkinsonism, they provide a unique opportunity to observe directly the development of parkinsonism. Patients with idiopathic REM sleep behaviour disorder in an ongoing cohort study were evaluated annually with several quantitative motor measures, including the Unified Parkinson's Disease Rating Scale, Purdue Pegboard, alternate-tap test and timed up-and-go. Patients who developed parkinsonism were identified from this cohort and matched according to age to normal controls. Their results on motor testing from the preceding years were plotted, and then assessed with regression analysis, to determine when markers first deviated from normal values. Sensitivity and specificity of quantitative motor markers for diagnosing prodromal parkinsonism were assessed. Of 78 patients, 20 developed parkinsonism. On regression analysis, the Unified Parkinson's Disease Rating Scale first intersected normal values at an estimated 4.5 years before diagnosis. Voice and face akinesia intersected earliest (estimated prodromal interval = 9.8 years), followed by rigidity (4.4 years), gait abnormalities (4.4 years) and limb bradykinesia (4.2 years). Quantitative motor tests intersected normal values at longer prodromal intervals than subjective examination (Purdue Pegboard = 8.6 years, alternate-tap = 8.2, timed up-and-go = 6.3). Using Purdue Pegboard and the alternate-tap test, parkinsonism could be detected with 71-82% sensitivity and specificity 3 years before diagnosis, whereas a Unified Parkinson's Disease Rating Scale score >4 identified prodromal parkinsonism with 88% sensitivity and 94% specificity 2 years before diagnosis. Removal of action tremor scores improved sensitivity to 94% and specificity to 97% at 2 years before diagnosis (cut-off >3). Although distinction between conditions was often difficult, prodromal dementia with Lewy bodies appeared to have a slower progression than Parkinson's disease (prodromal interval = 6.0 versus 3.8 years). Using a cut-off of Unified Parkinson's Disease Rating Scale >3 (excluding action tremor), 25% of patients with 'still-idiopathic' REM sleep behaviour disorder demonstrated evidence of possible prodromal parkinsonism. Therefore, using direct assessment of motor examination before parkinsonism in a REM sleep behaviour disorder, we have estimated a prodromal interval of ∼4.5 years on the Unified Parkinson's Disease Rating Scale; other quantitative markers may detect parkinsonism earlier. Simple quantitative motor measures may be capable of reliably detecting parkinsonism, even before a clinical diagnosis can be made by experienced movement disorders neurologists.
Collapse
Affiliation(s)
- R B Postuma
- Department of Neurology, McGill University, Montreal General Hospital, Montreal, QC H3G 1A4, Canada
| | | | | | | | | |
Collapse
|
32
|
Abstract
Parkinson's disease (PD) has a prodromal phase during which nonmotor clinical features as well as physiological abnormalities may be present. These premotor markers could be used to screen for PD before motor abnormalities are present. The technology to identify PD before it reaches symptomatic Braak Stage 3 (substantia nigra compacta [SNc] involvement) already exists. The current challenge is to define the appropriate scope of use of predictive testing for PD. Imaging technologies such as dopamine transporter imaging currently offer the highest degree of accuracy for identifying premotor PD, but they are expensive as screening tools, and abnormalities on these studies would only be evident at Braak Stage 3 or higher. Efficiency is greatly enhanced by combining imaging with a prescreening test such as olfactory testing. This 2-step process has the potential to greatly reduce costs while retaining diagnostic accuracy. Alternatively, or in concert with this approach, evaluating high-risk populations (eg, patients with rapid eye movement behavior disorder or LRRK2 mutations) would enrich the sample for cases with underlying PD. Ultimately, the role of preclinical detection of PD will be determined by the ability of emerging therapies to influence clinical outcomes. As such, implementation of large-scale screening strategies awaits the arrival of clearly safe and effective therapies that address the underlying pathogenesis of PD. Future research will establish more definitive biomarkers capable of revealing the presence of disease in advance of SNc involvement with the promise of the potential for introducing disease-modifying therapy even before the development of evidence of dopamine deficiency.
Collapse
Affiliation(s)
- Andrew Siderowf
- Parkinson's Disease and Mov Disord. Center, Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | | |
Collapse
|
33
|
|
34
|
Abstract
This paper reviews the clinically relevant determinants of levodopa peripheral pharmacokinetics and main observed changes in the levodopa concentration-effect relationship with Parkinson's disease (PD) progression. Available clinically practical strategies to optimise levodopa pharmacokinetics and pharmacodynamics are briefly discussed. Levodopa shows particular pharmacokinetics including an extensive presystemic metabolism, overcome by the combined use of extracerebral inhibitors of the enzyme L: -amino acid decarboxylase and rapid absorption in the proximal small bowel by a saturable facilitated transport system shared with other large neutral amino acids. Drug transport from plasma to the brain is mediated by the same carriers operating in the intestinal mucosa. The main strategies to assure reproducibility of both intestinal absorption and delivery to the brain, and the clinical effect include standardization of levodopa dosing with respect to meal times and a controlled dietary protein intake. Levodopa plasma half-life is very short, resulting in marked plasma drug concentration fluctuations which are matched, as the disease progresses, to swings in the therapeutic response ("wearing-off" phenomena). "Wearing-off" phenomena can also be associated, at the more advanced disease stages, with a "negative", both parkinsonism-exacerbating and dyskinetic effect of levodopa at low, subtherapeutic plasma concentrations. Dyskinesias may also be related to high-levodopa, excessive plasma concentrations. Recognition of the different levodopa toxic response patterns can be difficult on a clinical basis alone and simultaneous monitoring of the levodopa concentration-effect relationship may prove useful to disclose the underlying mechanism and in planning the correct management. Clinically practical strategies to optimise levodopa pharmacokinetics, and possibly its therapeutic response, include liquid drug solutions, controlled release formulations and the use of inhibitors of levodopa metabolism. Unfortunately, these attempts have proved so far only partly successful, due to the complex alterations in cerebral levodopa kinetics which accompany the progressive degeneration of the nigrostriatal dopaminergic system in PD patients.
Collapse
|
35
|
Abstract
In the last 25 years there have been enormous advances in brain imaging. In addition to utility in diagnosis, these have led to novel insights into the pathogenesis of basal ganglia disease and the role of dopamine and the basal ganglia in normal health. The authors review highlights of this work, with a focus on advances in Parkinson's disease, the dystonias, Huntington's disease, and the role of dopamine in cognition and reward signaling. Emerging areas for future development include studies of functional connectivity, the analysis of default mode networks, studies of novel neurochemical pathways, methods to study disease pathogenesis, and the application of imaging techniques to investigate animal models of disease.
Collapse
Affiliation(s)
- A Jon Stoessl
- Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, British Columbia, Canada.
| | | | | |
Collapse
|
36
|
Liepelt I, Behnke S, Schweitzer K, Wolf B, Godau J, Wollenweber F, Dillmann U, Gaenslen A, Di Santo A, Maetzler W, Berg D. Pre-motor signs of PD are related to SN hyperechogenicity assessed by TCS in an elderly population. Neurobiol Aging 2011; 32:1599-606. [DOI: 10.1016/j.neurobiolaging.2009.10.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Revised: 09/23/2009] [Accepted: 10/06/2009] [Indexed: 10/20/2022]
|
37
|
Booij J, Berendse HW. Monitoring therapeutic effects in Parkinson's disease by serial imaging of the nigrostriatal dopaminergic pathway. J Neurol Sci 2011; 310:40-3. [PMID: 21840542 DOI: 10.1016/j.jns.2011.07.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 06/03/2011] [Accepted: 07/19/2011] [Indexed: 10/16/2022]
Abstract
PET and SPECT are very sensitive techniques to detect in-vivo nigrostriatal degeneration in Parkinson's disease, even in the pre-motor phase of the disease. Furthermore, these techniques are able to measure disease progression. However, caution must be used in the interpretation of studies in which therapeutic effects in Parkinson's disease were also monitored by serial imaging of nigrostriatal neurons, as disparity between imaging and clinical outcomes has been reported in several clinical studies.
Collapse
Affiliation(s)
- Jan Booij
- Department of Nuclear Medicine, Academic Medical Center, University of Amsterdam, The Netherlands.
| | | |
Collapse
|
38
|
Progression of monoaminergic dysfunction in Parkinson's disease: a longitudinal 18F-dopa PET study. Neuroimage 2011; 56:1463-8. [PMID: 21396455 DOI: 10.1016/j.neuroimage.2011.03.012] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 02/28/2011] [Accepted: 03/03/2011] [Indexed: 11/21/2022] Open
Abstract
Post-mortem and neuroimaging studies in Parkinson's disease (PD) have shown involvement of the brain serotoninergic, noradrenergic and cholinergic pathways alongside the characteristic degeneration of nigrostriatal dopamine neurons. The rate of progression of the degenerative process in these extrastriatal areas is still unclear. We used (18)F-dopa PET, a marker of aromatic aminoacid decarboxylase activity in monoaminergic neurons, to assess longitudinal changes in tracer uptake in brain noradrenergic, serotoninergic and extrastriatal dopaminergic structures over a 3-year period in a group of early PD patients. Ten PD patients had (18)F-dopa PET twice: at baseline and again after 37.1±21.5 months follow up. A standard object map was used to extract tracer influx constants (Ki) in 11 striatal and extrastriatal regions. Progressive decreases in (18)F-dopa Ki occurred over the follow-up period in the majority of the investigated areas, the fastest annual declines occurring in putamen (8.1%), locus coeruleus (7.8%), and globus pallidus interna (7.7%). Caudate and hypothalamus showed 6.3% and 6.1% annual Ki declines, respectively. At baseline, some structures showed increased levels of (18)F-dopa uptake in PD compared to controls (internal pallidum, locus coeruleus), indicating possible compensatory upregulation of monoamine turnover. These increased levels had normalised (globus pallidus interna) or become subnormal (locus coeruleus) at follow-up suggesting exhaustion of these mechanisms within the first years of disease. Loss of monoaminergic function in extrastriatal regions, as reflected by(18)F-dopa PET, is delayed and occurs independently from nigrostriatal degeneration. When assessing the efficacy of novel neuroprotective agents on nigrostriatal dysfunction in PD, (18)F-dopa PET could provide supplementary information concerning function of extrastriatal monoaminergic structures.
Collapse
|
39
|
de la Fuente-Fernández R, Appel-Cresswell S, Doudet DJ, Sossi V. Functional neuroimaging in Parkinson's disease. ACTA ACUST UNITED AC 2011; 5:109-20. [PMID: 23480585 DOI: 10.1517/17530059.2011.554820] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Functional neuroimaging techniques have greatly contributed to improving our understanding of Parkinson's disease (PD) neurodegeneration and related compensatory mechanisms. AREAS COVERED In this paper, the authors analyze the role of functional neuroimaging as a diagnostic tool in PD and review functional neuroimaging studies on PD progression and compensatory adaptations. Through this, the article provides the reader with sensible approaches for the use of functional neuroimaging in the diagnosis of PD. The reader is also provided with knowledge on the time course of nigrostriatal dopamine dysfunction in PD as well as an overview of the potential beneficial and deleterious effects of increased dopamine turnover. Finally, the reader is provided with a critical discussion of the differential effects of levodopa and dopamine agonists on presynaptic dopamine markers and the implications for the interpretation of clinical trials. EXPERT OPINION Functional neuroimaging probably plays a limited role in the diagnosis of PD. Parkinson's disease pathology leads to an exponential decline in nigrostriatal dopamine function and a compensatory increase in dopamine turnover, which may help delay symptom onset. On the negative side, increased dopamine turnover contributes to the development of treatment-related motor complications. Presynaptic markers of dopamine function are subject to regulatory changes, compromising the direct interpretation of neuroimaging results in trials of neuroprotective therapies for PD.
Collapse
|
40
|
Gaenslen A, Swid I, Liepelt-Scarfone I, Godau J, Berg D. The patients' perception of prodromal symptoms before the initial diagnosis of Parkinson's disease. Mov Disord 2011; 26:653-8. [PMID: 21370256 PMCID: PMC3130930 DOI: 10.1002/mds.23499] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Revised: 07/28/2010] [Accepted: 09/29/2010] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Before the occurrence of motor symptoms permits the clinical diagnosis of Parkinson's disease (PD), about or even more than 50% of the dopaminergic neurons of the substantia nigra have degenerated. This time be called the prodromal phase of PD. OBJECTIVE To evaluate the time span from onset of first prodromal symptoms to the initial diagnosis of PD as well as the order of symptom occurrence. METHODS Retrospective study of 93 consecutively interviewed PD patients without dementia and 93 sex and age matched controls free of neurodegenerative disorders. A standardized in-house telephone worksheet assessing 19 nonmotor and six early motor signs was used. RESULTS A total of 98.8% of all patients interviewed reported to have experienced prodromal symptoms prior to receiving the initial diagnosis of PD. Patients noticed an average of 7.6 different symptoms during this time interval. The mean time span between the recalled onset of any one symptom and PD diagnosis was 10.2 years. In both groups, the course of prodromal sign onset was associated with early neuropathological disease stages proposed by Braak. OUTLOOK These retrospectively gathered data confirm the existence of a long prodromal phase for PD that is consistent with neuropathological staging. A standardized questionnaire assessing such early symptoms may be helpful in identifying subjects at high risk for PD while they are still in the prodromal phase of the disorder.
Collapse
Affiliation(s)
- Alexandra Gaenslen
- Department of Neurodegeneration, Hertie Institute of Clinical Brain Research, Tübingen, Germany.
| | | | | | | | | |
Collapse
|
41
|
Pavese N, Kiferle L, Piccini P. Neuroprotection and imaging studies in Parkinson's disease. Parkinsonism Relat Disord 2010; 15 Suppl 4:S33-7. [PMID: 20123554 DOI: 10.1016/s1353-8020(09)70832-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The most challenging issue when testing putative neuroprotective agents for Parkinson's disease (PD) in clinical trials is the assessment of the effect of the treatment on the neurodegenerative process. By measuring changes in symptoms severity, clinical rating scales represent an important tool to rate the progression of the disease. However, the rating of clinical symptoms is dependent on the examiner and the neuroprotective effect can be masked by the symptomatic effect of the therapy. 18F-dopa PET and 123I-beta-CIT SPECT have been shown to be able to monitor the progressive loss of presynaptic nigrostriatal projections in PD and have been used as surrogate biomarkers of disease in several recent clinical trials. In this article the value of imaging as a biomarker for testing neuroprotective agents in PD is reviewed.
Collapse
Affiliation(s)
- Nicola Pavese
- Division of Neurosciences and Mental Health and MRC Clinical Sciences Centre, Faculty of Medicine, Hammersmith Hospital, Imperial College, London, UK.
| | | | | |
Collapse
|
42
|
Berg D, Seppi K, Liepelt I, Schweitzer K, Wollenweber F, Wolf B, Dillmann U, Stockner H, Godau J, Kiechl S, Gaenslen A, Willeit J, Santo AD, Maetzler W, Gasser T, Poewe W, Behnke S. Enlarged hyperechogenic substantia nigra is related to motor performance and olfaction in the elderly. Mov Disord 2010; 25:1464-9. [DOI: 10.1002/mds.23114] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
43
|
Dietary intake of folate, vitamin B6, vitamin B12 and riboflavin and risk of Parkinson's disease: a case-control study in Japan. Br J Nutr 2010; 104:757-64. [PMID: 20338075 DOI: 10.1017/s0007114510001005] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Increased homocysteine levels might accelerate dopaminergic cell death in Parkinson's disease (PD) through neurotoxic effects; thus, increasing intake of B vitamins involved in the regulation of homocysteine metabolism might decrease the risk of PD through decreasing plasma homocysteine. However, epidemiological evidence for the association of dietary B vitamins with PD is sparse, particularly in non-Western populations. We conducted a hospital-based case-control study in Japan to examine associations between dietary intake of folate, vitamin B6, vitamin B12 and riboflavin and the risk of PD. Patients with PD diagnosed using the UK PD Society Brain Bank criteria (n 249) and controls without neurodegenerative diseases (n 368) were recruited. Dietary intake during the preceding month was assessed at the time of study recruitment using a validated, self-administered, semi-quantitative, comprehensive diet history questionnaire. After adjustment for potential dietary and non-dietary confounding factors, intake of folate, vitamin B12 and riboflavin was not associated with the risk of PD (P for trend = 0.87, 0.70 and 0.11, respectively). However, low intake of vitamin B6 was associated with an increased risk of PD, independent of potential dietary and non-dietary confounders. Multivariate OR (95 % CI) for PD in the first, second, third and fourth quartiles of vitamin B6 were 1 (reference), 0.56 (0.33, 0.94), 0.69 (0.38, 1.25) and 0.48 (0.23, 0.99), respectively (P for trend = 0.10). In conclusion, in the present case-control study in Japan, low intake of vitamin B6, but not of folate, vitamin B12 or riboflavin, was independently associated with an increased risk of PD.
Collapse
|
44
|
Viallet F, Gayraud D, Bonnefoi B, Renie L, Aurenty R. Morbo di Parkinson idiopatico: aspetti clinici, diagnostici e terapeutici. Neurologia 2010. [DOI: 10.1016/s1634-7072(10)70494-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
45
|
Maetzler W, Liepelt I, Berg D. Progression of Parkinson's disease in the clinical phase: potential markers. Lancet Neurol 2009; 8:1158-71. [PMID: 19909914 DOI: 10.1016/s1474-4422(09)70291-1] [Citation(s) in RCA: 207] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Neuromodulatory or even neuroprotective therapy could soon be available for Parkinson's disease (PD), raising the question of how we should define and measure disease progression. Reported evidence suggests that several symptoms worsen with disease duration. Bradykinesia, rigidity, and activities of daily living deteriorate faster at the beginning of the disease, and this deterioration is paralleled by a decline in functional presynaptic dopaminergic activity, as shown by imaging techniques. Cognitive, speech, sleep, and gait difficulties might progress linearly in proportion to disease duration. Reduced variability in heart rate, orthostatic dysfunction, and visual hallucinations start to develop at mid-stage disease and are more common in late stages than earlier stages. In this Review, we summarise our current understanding of the progression of PD-associated symptoms and markers and conclude that an effective measurement of progression of PD must adapt to the different stages of the disease. In addition to routine clinical rating scales, new quantitative assessments of motor and non-motor symptoms, which should be more broadly available, reasonably priced, and easy-to-use, are needed.
Collapse
Affiliation(s)
- Walter Maetzler
- Center of Neurology, Department of Neurodegeneration and Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | | | | |
Collapse
|
46
|
|
47
|
|
48
|
|
49
|
Murakami K, Miyake Y, Sasaki S, Tanaka K, Fukushima W, Kiyohara C, Tsuboi Y, Yamada T, Oeda T, Miki T, Kawamura N, Sakae N, Fukuyama H, Hirota Y, Nagai M. Dietary glycemic index is inversely associated with the risk of Parkinson's disease: a case-control study in Japan. Nutrition 2009; 26:515-21. [PMID: 19628370 DOI: 10.1016/j.nut.2009.05.021] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Revised: 05/29/2009] [Accepted: 05/29/2009] [Indexed: 11/27/2022]
Abstract
OBJECTIVE High glycemic index (GI) or glycemic load (GL) carbohydrates might be expected to decrease the risk of Parkinson's disease (PD) by an insulin-induced increase in brain dopamine. We conducted a hospital-based case-control study in Japan to examine associations between dietary GI and GL and other dietary carbohydrate variables, including intake of available carbohydrate and dietary fiber, and PD. METHODS Patients with PD diagnosed using the U.K. Parkinson's Disease Society Brain Bank criteria (n=249) and controls without neurodegenerative diseases (n=368) were recruited. Dietary intake during the preceding month was assessed at the time of study recruitment using a validated, self-administered, semiquantitative, comprehensive diet history questionnaire. RESULTS After adjustment for potential dietary and non-dietary confounding factors, dietary GI was significantly inversely associated with the risk of PD. Multivariate odds ratios (95% confidence intervals) for PD in the first, second, third, and fourth quartiles of dietary GI were 1.00 (reference), 1.03 (0.64-1.66), 0.68 (0.41-1.15), and 0.61 (0.34-1.09), respectively (P for trend=0.04). Conversely, no significant association was observed for other dietary carbohydrates, including dietary GL (P for trend=0.77), available carbohydrate intake (P for trend=0.28), or dietary fiber intake (P for trend=0.73). CONCLUSION This preliminary case-control study based on current dietary habits found an independent inverse relation between dietary GI and PD. Considering the plausibility of the putative mechanism, further investigation using a case-control design with accurate assessment of past dietary habits or a prospective design is warranted.
Collapse
Affiliation(s)
- Kentaro Murakami
- Department of Social and Preventive Epidemiology, School of Public Health, the University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Puñal-Riobóo J, Serena-Puig A, Varela-Lema L, Álvarez-Páez A, Ruano-Ravina A. Utilidad clínica de la 18FDOPA-PET en trastornos del movimiento. Una revisión sistemática. ACTA ACUST UNITED AC 2009. [DOI: 10.1016/s0212-6982(09)71352-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|