101
|
Periñán MT, Macías-García D, Labrador-Espinosa MÁ, Jesús S, Buiza-Rueda D, Adarmes-Gómez AD, Muñoz-Delgado L, Gómez-Garre P, Mir P. Association of PICALM with Cognitive Impairment in Parkinson's Disease. Mov Disord 2020; 36:118-123. [PMID: 32914893 DOI: 10.1002/mds.28283] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/07/2020] [Accepted: 08/17/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Cognitive impairment is one of the most disabling nonmotor symptoms in Parkinson's disease (PD). Recently, a genome-wide association study in Alzheimer's disease has identified the PICALM rs3851179 polymorphism as one of the most significant susceptibility genes for Alzheimer's disease after APOE. The aim of this study was to determine the potential role of PICALM and its genetic interaction with APOE in the development of cognitive decline in PD. METHODS A discovery cohort of 712 patients with PD were genotyped for PICALM (rs3851179) and APOE (rs429358 and rs7412) polymorphisms. The association of PICALM and APOE-PICALM genetic interaction with cognitive dysfunction in PD was studied using logistic regression models, and the relationship of PICALM with cognitive decline onset was assessed with Cox regression analysis. PICALM effect was then replicated in an international, independent cohort (Parkinson's Progression Markers Initiative, N = 231). RESULTS PICALM rs3851179 TT genotype was significantly associated with a decreased risk of cognitive impairment in PD (TT vs. CC + CT, P = 0.041, odds ratio = 0.309). Replication studies further demonstrated its protective effect on cognitive impairment in PD. In addition, the protective effect of the PICALM rs3851179 TT genotype was more pronounced in the APOE ε4 (-) carriers from the discovery cohort (P = 0.037, odds ratio = 0.241), although these results were not replicated in the Parkinson's Progression Markers Initiative cohort. CONCLUSIONS Our results support the fact that PICALM is associated with cognitive impairment in PD. The understanding of its contribution to cognitive decline in PD could provide new targets for the development of novel therapies. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- María Teresa Periñán
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del, Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Daniel Macías-García
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del, Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Miguel Ángel Labrador-Espinosa
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del, Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Silvia Jesús
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del, Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Dolores Buiza-Rueda
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del, Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Astrid D Adarmes-Gómez
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del, Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Laura Muñoz-Delgado
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del, Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Seville, Spain
| | - Pilar Gómez-Garre
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del, Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del, Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
102
|
Abraham DS, Pham Nguyen TP, Hennessy S, Weintraub D, Gray SL, Xie D, Willis AW. Frequency of and risk factors for potentially inappropriate medication use in Parkinson's disease. Age Ageing 2020; 49:786-792. [PMID: 32255485 DOI: 10.1093/ageing/afaa033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/16/2019] [Accepted: 01/01/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND impairments in neurotransmitter pathways put Parkinson's disease (PD) patients at risk for drug-disease interactions and adverse medication events. OBJECTIVE to determine the prevalence and risk factors for potentially inappropriate medication (PIM) prescriptions, as defined by the 2015 Beers List, in PD. METHODS cross-sectional analysis was conducted on 2014 Medicare beneficiaries with PD who had parts A, B and D coverage. The prevalence of PIM prescriptions for older adults was determined overall, and specifically for medications that can exacerbate motor symptoms or cognitive impairment in PD. Logistic regression models were constructed to determine the association between age, sex, race, geography and poverty with PIM prescriptions. RESULTS the final sample included 458,086 beneficiaries. In 2014, 35.8% of beneficiaries with PD filled a prescription for at least one PIM for older adults. In total, 8.7% of beneficiaries received a PIM that could exacerbate motor symptoms and 29.0% received a PIM that could worsen cognitive impairment. After adjustment, in all models, beneficiaries who were younger, female, white, urban-dwelling and eligible for Medicaid benefits were more likely to receive a PIM. CONCLUSION PIM prescriptions are not uncommon in PD, particularly for medications that can exacerbate cognitive impairment. Future research will examine underlying drivers of sex and other disparities in PIM prescribing. Additional studies are needed to understand the impact of PIMs on disease symptoms, healthcare utilisation and patient outcomes.
Collapse
Affiliation(s)
- Danielle S Abraham
- Department of Neurology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Department of Neurology Translational Center for Excellence for Neuroepidemiology and Neurological Outcomes Research, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Department of Biostatics, Epidemiology, and Informatics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Thanh Phuong Pham Nguyen
- Department of Neurology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Department of Neurology Translational Center for Excellence for Neuroepidemiology and Neurological Outcomes Research, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Department of Biostatics, Epidemiology, and Informatics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Sean Hennessy
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Department of Biostatics, Epidemiology, and Informatics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Daniel Weintraub
- Department of Neurology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Parkinson’s Disease Research, Education and Clinical Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Shelly L Gray
- Department of Pharmacy, University of Washington School of Pharmacy, Seattle, WA, USA
| | - Dawei Xie
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Department of Biostatics, Epidemiology, and Informatics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Allison W Willis
- Department of Neurology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Department of Neurology Translational Center for Excellence for Neuroepidemiology and Neurological Outcomes Research, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Department of Biostatics, Epidemiology, and Informatics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
103
|
Cao X, Wang X, Xue C, Zhang S, Huang Q, Liu W. A Radiomics Approach to Predicting Parkinson's Disease by Incorporating Whole-Brain Functional Activity and Gray Matter Structure. Front Neurosci 2020; 14:751. [PMID: 32760248 PMCID: PMC7373781 DOI: 10.3389/fnins.2020.00751] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/25/2020] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease (PD) is a progressive, chronic, and neurodegenerative disorder that is primarily diagnosed by clinical examinations and magnetic resonance imaging (MRI). In this study, we proposed a machine learning based radiomics method to predict PD. Fifty healthy controls (HC) along with 70 PD patients underwent resting-state magnetic resonance imaging (rs-fMRI). For all subjects, we extracted five types of 6664 features, including mean amplitude of low-frequency fluctuation (mALFF), mean regional homogeneity (mReHo), resting-state functional connectivity (RSFC), voxel-mirrored homotopic connectivity (VMHC) and gray matter (GM) volume. After conducting dimension reduction utilizing Least absolute shrinkage and selection operator (LASSO), fifty-three radiomic features including 46 RSFCs, 1 mALFF, 3 mReHos, 1 VMHC, 2 GM volumes and 1 clinical factor were retained. The selected features also indicated the most discriminative regions for PD. We further conducted model fitting procedure for classifying subjects in the training set employing random forest and support volume machine (SVM) to evaluate the performance of the two methods. After cross-validation, both methods achieved 100% accuracy and area under curve (AUC) for distinguishing between PD and HC in the training set. In the testing set, SVM performed better than random forest with the accuracy, true positive rate (TPR) and AUC being 85%, 1 and 0.97, respectively. These findings demonstrate the radiomics technique has the potential to support radiological diagnosis and to achieve high classification accuracy for clinical diagnostic systems for patients with PD.
Collapse
Affiliation(s)
- Xuan Cao
- Division of Statistics and Data Science, Department of Mathematical Sciences, University of Cincinnati, Cincinnati, OH, United States
| | - Xiao Wang
- Department of Radiology, Affiliated Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Chen Xue
- Department of Radiology, Affiliated Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Shaojun Zhang
- Department of Statistics, University of Florida, Gainesville, FL, United States
| | - Qingling Huang
- Department of Radiology, Affiliated Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Weiguo Liu
- Department of Neurology, Affiliated Brain Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
104
|
Marschallinger J, Altendorfer B, Rockenstein E, Holztrattner M, Garnweidner-Raith J, Pillichshammer N, Leister I, Hutter-Paier B, Strempfl K, Unger MS, Chishty M, Felder T, Johnson M, Attems J, Masliah E, Aigner L. The Leukotriene Receptor Antagonist Montelukast Reduces Alpha-Synuclein Load and Restores Memory in an Animal Model of Dementia with Lewy Bodies. Neurotherapeutics 2020; 17:1061-1074. [PMID: 32072462 PMCID: PMC7609773 DOI: 10.1007/s13311-020-00836-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Dementia with Lewy bodies (DLB) represents a huge medical need as it accounts for up to 30% of all dementia cases, and there is no cure available. The underyling spectrum of pathology is complex and creates a challenge for targeted molecular therapies. We here tested the hypothesis that leukotrienes are involved in the pathology of DLB and that blocking leukotrienes through Montelukast, a leukotriene receptor antagonist and approved anti-asthmatic drug, might alleviate pathology and restore cognitive functions. Expression of 5-lipoxygenase, the rate-limiting enzyme for leukotriene production, was indeed elevated in brains with DLB. Treatment of cognitively deficient human alpha-synuclein overexpressing transgenic mice with Montelukast restored memory. Montelukast treatment resulted in modulation of beclin-1 expression, a marker for autophagy, and in a reduction in the human alpha-synulcein load in the transgenic mice. Reducing the protein aggregation load in neurodegenerative diseases might be a novel model of action of Montelukast. Moreover, this work presents leukotriene signaling as a potential drug target for DLB and shows that Montelukast might be a promising drug candidate for future DLB therapy development.
Collapse
Affiliation(s)
- Julia Marschallinger
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Barbara Altendorfer
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Edward Rockenstein
- Department of Neuroscience, School of Medicine, University of California San Diego, San Diego, USA
| | - Miriam Holztrattner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Julia Garnweidner-Raith
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Nadine Pillichshammer
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Iris Leister
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | | | - Katharina Strempfl
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
- QPS Austria GmbH, Neuropharmacology, Grambach, Austria
| | - Michael S Unger
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | | | - Thomas Felder
- Department of Laboratory Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Mary Johnson
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Johannes Attems
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Eliezer Masliah
- Department of Neuroscience, School of Medicine, University of California San Diego, San Diego, USA
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria.
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria.
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| |
Collapse
|
105
|
Schierding W, Farrow S, Fadason T, Graham OEE, Pitcher TL, Qubisi S, Davidson AJ, Perry JK, Anderson TJ, Kennedy MA, Cooper A, O'Sullivan JM. Common Variants Coregulate Expression of GBA and Modifier Genes to Delay Parkinson's Disease Onset. Mov Disord 2020; 35:1346-1356. [PMID: 32557794 PMCID: PMC7496525 DOI: 10.1002/mds.28144] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/02/2020] [Accepted: 05/20/2020] [Indexed: 12/29/2022] Open
Abstract
Background GBA mutations are numerically the most significant genetic risk factor for Parkinson's disease (PD), yet these mutations have low penetrance, suggesting additional mechanisms. Objectives The objective of this study was to determine if the penetrance of GBA in PD can be explained by regulatory effects on GBA and modifier genes. Methods Genetic variants associated with the regulation of GBA were identified by screening 128 common single nucleotide polymorphisms (SNPs) in the GBA locus for spatial cis‐expression quantitative trail locus (supported by chromatin interactions). Results We identified common noncoding SNPs within GBA that (1) regulate GBA expression in peripheral tissues, some of which display α‐synuclein pathology and (2) coregulate potential modifier genes in the central nervous system and/or peripheral tissues. Haplotypes based on 3 of these SNPs delay disease onset by 5 years. In addition, SNPs on 6 separate chromosomes coregulate GBA expression specifically in either the substantia nigra or cortex, and their combined effect potentially modulates motor and cognitive symptoms, respectively. Conclusions This work provides a new perspective on the haplotype‐specific effects of GBA and the genetic etiology of PD, expanding the role of GBA from the gene encoding the β‐glucocerebrosidase (GCase) to that of a central regulator and modifier of PD onset, with GBA expression itself subject to distant regulation. Some idiopathic patients might possess insufficient GBA‐encoded GCase activity in the substantia nigra as the result of distant regulatory variants and therefore might benefit from GBA‐targeting therapeutics. The SNPs’ regulatory impacts provide a plausible explanation for the variable phenotypes also observed in GBA‐centric Gaucher's disease and dementia with Lewy bodies. © 2020 The Authors. Movement Disorders published by Wiley Periodicals, LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Sophie Farrow
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Tayaza Fadason
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Oscar E E Graham
- Gene Structure and Function Laboratory, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Toni L Pitcher
- Department of Medicine, University of Otago, Christchurch, New Zealand.,Brain Research New Zealand, The University of Auckland, Auckland, New Zealand.,New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Sara Qubisi
- Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Alan J Davidson
- Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Jo K Perry
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Tim J Anderson
- Department of Medicine, University of Otago, Christchurch, New Zealand.,Brain Research New Zealand, The University of Auckland, Auckland, New Zealand.,New Zealand Brain Research Institute, Christchurch, New Zealand.,Neurology Department, Christchurch Hospital, Canterbury District Health Board, Christchurch, New Zealand
| | - Martin A Kennedy
- Gene Structure and Function Laboratory, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand.,New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Antony Cooper
- Australian Parkinsons Mission, Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Justin M O'Sullivan
- Liggins Institute, The University of Auckland, Auckland, New Zealand.,Brain Research New Zealand, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
106
|
Boertien JM, van der Zee S, Chrysou A, Gerritsen MJJ, Jansonius NM, Spikman JM, van Laar T. Study protocol of the DUtch PARkinson Cohort (DUPARC): a prospective, observational study of de novo Parkinson's disease patients for the identification and validation of biomarkers for Parkinson's disease subtypes, progression and pathophysiology. BMC Neurol 2020; 20:245. [PMID: 32534583 PMCID: PMC7293131 DOI: 10.1186/s12883-020-01811-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
Background Parkinson’s Disease (PD) is a heterogeneous, progressive neurodegenerative disorder which is characterized by a variety of motor and non-motor symptoms. To date, no disease modifying treatment for PD exists. Here, the study protocol of the Dutch Parkinson Cohort (DUPARC) is described. DUPARC is a longitudinal cohort study aimed at deeply phenotyping de novo PD patients who are treatment-naïve at baseline, to discover and validate biomarkers for PD progression, subtypes and pathophysiology. Methods/design DUPARC is a prospective cohort study in which 150 de novo PD subjects will be recruited through a collaborative network of PD treating neurologists in the northern part of the Netherlands (Parkinson Platform Northern Netherlands, PPNN). Participants will receive follow-up assessments after 1 year and 3 years, with the intention of an extended follow-up with 3 year intervals. Subjects are extensively characterized to primarily assess objectives within three major domains of PD: cognition, gastrointestinal function and vision. This includes brain magnetic resonance imaging (MRI); brain cholinergic PET-imaging with fluoroethoxybenzovesamicol (FEOBV-PET); brain dopaminergic PET-imaging with fluorodopa (FDOPA-PET); detailed neuropsychological assessments, covering all cognitive domains; gut microbiome composition; intestinal wall permeability; optical coherence tomography (OCT); genotyping; motor and non-motor symptoms; overall clinical status and lifestyle factors, including a dietary assessment; storage of blood and feces for additional analyses of inflammation and metabolic parameters. Since the start of the inclusion, at the end of 2017, over 100 PD subjects with a confirmed dopaminergic deficit on FDOPA-PET have been included. Discussion DUPARC is the first study to combine data within, but not limited to, the non-motor domains of cognition, gastrointestinal function and vision in PD subjects over time. As a de novo PD cohort, with treatment naïve subjects at baseline, DUPARC provides a unique opportunity for biomarker discovery and validation without the possible confounding influences of dopaminergic medication. Trial registration NCT04180865; registered retrospectively, November 28th 2019.
Collapse
Affiliation(s)
- Jeffrey M Boertien
- Department of Neurology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB, Groningen, The Netherlands.,Parkinson Expertise Center Groningen, Groningen, the Netherlands
| | - Sygrid van der Zee
- Department of Neurology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB, Groningen, The Netherlands.,Parkinson Expertise Center Groningen, Groningen, the Netherlands
| | - Asterios Chrysou
- Department of Neurology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB, Groningen, The Netherlands.,Parkinson Expertise Center Groningen, Groningen, the Netherlands
| | - Marleen J J Gerritsen
- Department of Neuropsychology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Nomdo M Jansonius
- Department of Ophthalmology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jacoba M Spikman
- Department of Neuropsychology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Teus van Laar
- Department of Neurology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB, Groningen, The Netherlands. .,Parkinson Expertise Center Groningen, Groningen, the Netherlands.
| | | |
Collapse
|
107
|
Gonçalves AR, Mendes A, Vila-Chã N, Damásio J, Fernandes J, Cavaco SM. Past appendectomy may be related to early cognitive dysfunction in Parkinson's disease. Neurol Sci 2020; 42:123-130. [PMID: 32529319 DOI: 10.1007/s10072-020-04507-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 05/30/2020] [Indexed: 12/18/2022]
Abstract
INTRODUCTION The vermiform appendix is a potential site of initiation of Parkinson's disease (PD) pathology. We hypothesized that the appendectomy earlier in life may alter the clinical expression of PD. OBJECTIVE To explore the effects of appendectomy prior to onset of PD motor symptoms on patients' symptoms, in particular on cognitive dysfunction. METHODS Two hundred and sixty-two consecutive PD patients were asked about past history of appendectomy and underwent an evaluation, which included the Unified Parkinson's Disease Rating Scale (UPDRS), Hoehn & Yahr scale (H&Y), Schwab & England Independence Scale (S&E), Dementia Rating Scale-2 (DRS-2), Apathy Evaluation Scale, Hospital Anxiety and Depression Scale, and Brief Smell Identification Test. Motor symptoms were evaluated in OFF and ON states. Non-parametric group comparisons and logistic regressions were used for data analyses. RESULTS Thirty-one patients (11.8%) had history of appendectomy prior to PD onset. These patients had more severe motor symptoms (UPDRS-III and H&Y) and lower functional independence (S&E) in ON and had higher frequency of cognitive dysfunction (DRS-2 Initiation/Perseveration, Conceptualization, and Memory subscales) (p < 0.05). The association between history of appendectomy and cognitive dysfunction was evident only in patients with late onset PD (≥ 55 years) and with disease duration ≤ 5 years. History of appendectomy remained statistically associated with impairment on DRS-2 Conceptualization and Memory subscales, when demographic and clinical variables were considered. CONCLUSION History of appendectomy appears to alter the clinical expression of late onset PD, with early cognitive impairment, more severe motor symptoms in ON, and poorer functional independence under anti-parkinsonian medication.
Collapse
Affiliation(s)
- Alexandra R Gonçalves
- Department of Neurology, Centro Hospitalar Universitário do Porto, Largo Prof. Abel Salazar, 4099-001, Porto, Portugal. .,Faculty of Medicine, University of Porto, Porto, Portugal.
| | - Alexandre Mendes
- Department of Neurology, Centro Hospitalar Universitário do Porto, Largo Prof. Abel Salazar, 4099-001, Porto, Portugal.,Abel Salazar Biomedical Sciences Institute, University of Porto, Porto, Portugal
| | - Nuno Vila-Chã
- Department of Neurology, Centro Hospitalar Universitário do Porto, Largo Prof. Abel Salazar, 4099-001, Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| | - Joana Damásio
- Department of Neurology, Centro Hospitalar Universitário do Porto, Largo Prof. Abel Salazar, 4099-001, Porto, Portugal.,Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - Joana Fernandes
- DEFI, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Sara M Cavaco
- Department of Neurology, Centro Hospitalar Universitário do Porto, Largo Prof. Abel Salazar, 4099-001, Porto, Portugal.,Abel Salazar Biomedical Sciences Institute, University of Porto, Porto, Portugal
| |
Collapse
|
108
|
Stock L, Krüger-Zechlin C, Deeb Z, Timmermann L, Waldthaler J. Natural Reading in Parkinson's Disease With and Without Mild Cognitive Impairment. Front Aging Neurosci 2020; 12:120. [PMID: 32528271 PMCID: PMC7258085 DOI: 10.3389/fnagi.2020.00120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 04/08/2020] [Indexed: 11/21/2022] Open
Abstract
Background: Patients with Parkinson’s disease (PD) show eye movement abnormalities and frequently complain about difficulties in reading. So far, it is unclear whether basal ganglia dysfunction or cognitive impairment has a greater impact on eye movements during reading. Objective: To analyze eye movement behavior during a natural reading task with respect to cognitive state and dopaminergic therapy in PD and healthy controls. Methods: Eye movements of 59 PD patients and 29 age- and education-matched healthy controls were recorded during mute, self-paced reading of a text. 25 cognitively normal PD patients performed the task additionally in off medication state. Clinical assessment included a comprehensive neuropsychological test battery and the motor section of MDS—Unified Parkinson’s Disease Rating Scale (MDS-UPDRS). Results: PD-mild cognitive impairment (MCI) was diagnosed in 21 patients. Reading speed was significantly reduced in PD-MCI compared to healthy controls and PD patients without MCI due to higher numbers of progressive saccades. Cognitively intact PD patients showed no significant alterations of reading speed or eye movement pattern during reading. The fixation duration tended to be prolonged in PD compared to healthy controls and decreased significantly after levodopa intake. Scores for executive functions, attention, and language correlated with reading speed in the PD group. Conclusion: The present study is the first to reveal (1) reduced reading speed with altered reading pattern in PD with MCI and (2) a relevant impact of levodopa on fixation duration during reading in PD. Further research is needed to determine whether therapeutic interventions, e.g., levodopa or neuropsychological training, improve the subjective reading experience for patients with PD.
Collapse
Affiliation(s)
- Lena Stock
- Department of Neurology, University Hospital Marburg, Marburg, Germany
| | | | - Zain Deeb
- Department of Neurology, University Hospital Marburg, Marburg, Germany
| | - Lars Timmermann
- Department of Neurology, University Hospital Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior - CMBB, Universities Marburg and Gießen, Marburg, Germany
| | - Josefine Waldthaler
- Department of Neurology, University Hospital Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior - CMBB, Universities Marburg and Gießen, Marburg, Germany
| |
Collapse
|
109
|
Lee K, Cao X. Bayesian group selection in logistic regression with application to MRI data analysis. Biometrics 2020; 77:391-400. [PMID: 32365231 DOI: 10.1111/biom.13290] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/22/2022]
Abstract
We consider Bayesian logistic regression models with group-structured covariates. In high-dimensional settings, it is often assumed that only a small portion of groups are significant, and thus, consistent group selection is of significant importance. While consistent frequentist group selection methods have been proposed, theoretical properties of Bayesian group selection methods for logistic regression models have not been investigated yet. In this paper, we consider a hierarchical group spike and slab prior for logistic regression models in high-dimensional settings. Under mild conditions, we establish strong group selection consistency of the induced posterior, which is the first theoretical result in the Bayesian literature. Through simulation studies, we demonstrate that the proposed method outperforms existing state-of-the-art methods in various settings. We further apply our method to a magnetic resonance imaging data set for predicting Parkinson's disease and show its benefits over other contenders.
Collapse
Affiliation(s)
- Kyoungjae Lee
- Department of Statistics, Inha University, Incheon, South Korea
| | - Xuan Cao
- Department of Mathematical Sciences, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
110
|
Urban P, Falkenburger B, Jost WH, Ransmayr G, Riederer P, Winkler C. [Structure and efferences of the substantia nigra pars compacta in Parkinson's disease]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2020; 88:591-599. [PMID: 32396943 DOI: 10.1055/a-1149-9280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
There is consensus that the neuropathological characteristic of Parkinson's disease (PD) is the neuronal cell loss of the substantia nigra pars compacta (SNc) in connection with a Lewy pathology. The transsynaptic spread of Lewy pathology is considered essential in PD pathogenesis. Therefore, the knowledge of pre-existing neuroanatomical connections of the SNc is essential. We describe recent animal experiments on the afferent and efferent projections of the SNc and discuss the evidence for and against the sequential transsynaptic spread of Lewy pathology in the pathogenesis of PD.
Collapse
Affiliation(s)
- Peter Urban
- Abteilung für Neurologie, Asklepios Klinik Barmbek
| | | | | | - Gerhard Ransmayr
- Klinik für Neurologie 2, Kepler Universitätsklinikum, Linz/Austria
| | - Peter Riederer
- Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie, Universitätsklinikum Würzburg
| | | |
Collapse
|
111
|
Vriend C, van Balkom TD, van Druningen C, Klein M, van der Werf YD, Berendse HW, van den Heuvel OA. Processing speed is related to striatal dopamine transporter availability in Parkinson's disease. Neuroimage Clin 2020; 26:102257. [PMID: 32344372 PMCID: PMC7186552 DOI: 10.1016/j.nicl.2020.102257] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/27/2020] [Accepted: 04/02/2020] [Indexed: 01/14/2023]
Abstract
BACKGROUND Parkinson's disease (PD) affects the integrity of the dopamine and serotonin system, and is characterized by a plethora of different symptoms, including cognitive impairments of which the pathophysiology is not yet fully elucidated. OBJECTIVES Investigate the role of the integrity of the dopaminergic and serotonergic system in cognitive functioning in early-stage PD using Single Photon Emission Computed Tomography (SPECT) combined with the radiotracer 123I-N-ω-fluoropropyl-2β-carbomethoxy-3β-(4-iodophenyl)nortropane (123I-FP-CIT). METHODS We studied the association between cognitive functions and dopamine transporter (DAT) availability in the caudate nucleus and putamen - as a proxy for striatal dopaminergic integrity - and serotonin transporter (SERT) availability as a proxy for serotonergic integrity in the thalamus and hippocampus using bootstrapped multiple regression. One-hundred-and-twenty-nine (129) PD patients underwent a 123I-FP-CIT SPECT scan and a neuropsychological assessment. RESULTS We showed a positive association between DAT availability in the head of the caudate nucleus and the Stroop Color Word Task - card I (reading words; β = 0.32, P = 0.001) and a positive association between DAT availability in the anterior putamen and the Trail Making Test part A (connecting consecutively numbered circles; β = 0.25, P = 0.02). These associations remained after adjusting for motor symptom severity or volume of the region-of-interest and were most pronounced in medication-naïve PD patients. There were no associations between cognitive performance and SERT availability in the thalamus or hippocampus. CONCLUSIONS We interpret these results as a role for striatal dopamine - and its PD-related decline - in aspects of processing speed.
Collapse
Affiliation(s)
- Chris Vriend
- Amsterdam UMC, Vrije Universiteit Amsterdam, Psychiatry, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Anatomy and Neurosciences, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam 1007 MB, the Netherlands.
| | - Tim D van Balkom
- Amsterdam UMC, Vrije Universiteit Amsterdam, Psychiatry, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Corné van Druningen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Psychiatry, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Martin Klein
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Psychology, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Ysbrand D van der Werf
- Amsterdam UMC, Vrije Universiteit Amsterdam, Anatomy and Neurosciences, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam 1007 MB, the Netherlands
| | - Henk W Berendse
- Amsterdam UMC, Vrije Universiteit Amsterdam, Neurology, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Odile A van den Heuvel
- Amsterdam UMC, Vrije Universiteit Amsterdam, Psychiatry, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Anatomy and Neurosciences, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam 1007 MB, the Netherlands
| |
Collapse
|
112
|
Cognition Deficits in Parkinson's Disease: Mechanisms and Treatment. PARKINSONS DISEASE 2020; 2020:2076942. [PMID: 32269747 PMCID: PMC7128056 DOI: 10.1155/2020/2076942] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 02/08/2020] [Accepted: 02/26/2020] [Indexed: 12/22/2022]
Abstract
Parkinson's disease (PD) is the second most common progressive neurodegenerative disorder mainly in middle-elderly population, which represents diverse nonmotor symptoms (NMS) besides such well-documented motor symptoms as bradykinesia, resting tremor, rigidity, and postural instability. With the advancement of aging trend worldwide, the global prevalence of PD is mounting up year after year. Nowadays, accumulating lines of studies have given a comprehensive and thorough coverage of motor symptoms in PD. Yet much less attention as compared has been paid to the nonmotor symptoms of PD, such as cognition deficits. Of note, a patient with PD who suffers from cognitive impairment may harbour a statistically significantly higher risk of progressing toward dementia, which negatively affects their life expectancy and daily functioning and overall lowers the global quality of life. Furthermore, it is a widely held view that cognitive dysfunction does not just occur in the late stage of PD. On the basis of numerous studies, mild cognitive impairment (MCI) is a harbinger of dementia in PD, which is observed as an intermediate state with considerable variability; some patients remain stable and some even revert to normal cognition. Considered that the timing, profile, and rate of cognitive impairment vary greatly among PD individuals, it is extremely urgent for researchers and clinicians alike to identify and predict future cognitive decline in this population. Simultaneously, early screening and canonical management of PD with cognitive deficits are very imperative to postpone the disease progression and improve the prognosis of patients. In our review, we focus on a description of cognitive decline in PD, expound emphatically the pathological mechanisms underlying cognition deficits in PD, then give a comprehensive overview of specific therapeutic strategies, and finally dissect what fresh insights may bring new exciting prospect for the subfield.
Collapse
|
113
|
Svenningsson P, Odin P, Dizdar N, Johansson A, Grigoriou S, Tsitsi P, Wictorin K, Bergquist F, Nyholm D, Rinne J, Hansson F, Sonesson C, Tedroff J, Andersson K, Sundgren M, Duzynski W, Carlström C. A Phase 2a Trial Investigating the Safety and Tolerability of the Novel Cortical Enhancer IRL752 in Parkinson's Disease Dementia. Mov Disord 2020; 35:1046-1054. [DOI: 10.1002/mds.28020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 12/14/2022] Open
Affiliation(s)
- Per Svenningsson
- Section of Neurology, Department of Clinical NeuroscienceKarolinska Institutet Stockholm Sweden
| | - Per Odin
- Division of Neurology, Department of Clinical Sciences LundLund University Lund Sweden
| | - Nil Dizdar
- Department of Clinical and Experimental MedicineLinköping University Linköping Sweden
| | - Anders Johansson
- Section of Neurology, Department of Clinical NeuroscienceKarolinska Institutet Stockholm Sweden
| | - Sotirios Grigoriou
- Division of Neurology, Department of Clinical Sciences LundLund University Lund Sweden
| | - Panagiota Tsitsi
- Section of Neurology, Department of Clinical NeuroscienceKarolinska Institutet Stockholm Sweden
| | - Klas Wictorin
- Department of NeurologyHelsingborg Hospital Helsingborg Sweden
| | - Filip Bergquist
- Department of Pharmacology, Sahlgrenska AcademyUniversity of Gothenburg Gothenburg Sweden
| | - Dag Nyholm
- Department of Neuroscience, NeurologyUppsala University Uppsala Sweden
| | - Juha Rinne
- Clinical Research Services Turku Oy Turku Finland
- Division of Clinical NeurosciencesTurku University Hospital Turku Finland
| | | | - Clas Sonesson
- Integrative Research Laboratories AB Göteborg Sweden
| | - Joakim Tedroff
- Section of Neurology, Department of Clinical NeuroscienceKarolinska Institutet Stockholm Sweden
- Integrative Research Laboratories AB Göteborg Sweden
| | | | | | | | | | | |
Collapse
|
114
|
Orgeta V, McDonald KR, Poliakoff E, Hindle JV, Clare L, Leroi I. Cognitive training interventions for dementia and mild cognitive impairment in Parkinson's disease. Cochrane Database Syst Rev 2020; 2:CD011961. [PMID: 32101639 PMCID: PMC7043362 DOI: 10.1002/14651858.cd011961.pub2] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Approximately 60% to 80% of people with Parkinson's disease (PD) experience cognitive impairment that impacts on their quality of life. Cognitive decline is a core feature of the disease and can often present before the onset of motor symptoms. Cognitive training may be a useful non-pharmacological intervention that could help to maintain or improve cognition and quality of life for people with PD dementia (PDD) or PD-related mild cognitive impairment (PD-MCI). OBJECTIVES To determine whether cognitive training (targeting single or multiple domains) improves cognition in people with PDD and PD-MCI or other clearly defined forms of cognitive impairment in people with PD. SEARCH METHODS We searched the Cochrane Dementia and Cognitive Improvement Group Trials Register (8 August 2019), the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, CINAHL, and PsycINFO. We searched reference lists and trial registers, searched relevant reviews in the area and conference proceedings. We also contacted experts for clarifications on data and ongoing trials. SELECTION CRITERIA We included randomised controlled trials where the participants had PDD or PD-MCI, and where the intervention was intended to train general or specific areas of cognitive function, targeting either a single domain or multiple domains of cognition, and was compared to a control condition. Multicomponent interventions that also included motor or other elements were considered eligible. DATA COLLECTION AND ANALYSIS Two review authors independently screened titles, abstracts, and full-text articles for inclusion in the review. Two review authors also independently undertook extraction of data and assessment of methodological quality. We used GRADE methods to assess the overall quality of the evidence. MAIN RESULTS Seven studies with a total of 225 participants met the inclusion criteria for this review. All seven studies compared the effects of a cognitive training intervention to a control intervention at the end of treatment periods lasting four to eight weeks. Six studies included people with PD living in the community. These six studies recruited people with single-domain (executive) or multiple-domain mild cognitive impairment in PD. Four of these studies identified participants with MCI using established diagnostic criteria, and two included both people with PD-MCI and people with PD who were not cognitively impaired. One study recruited people with a diagnosis of PD dementia who were living in long-term care settings. The cognitive training intervention in three studies targeted a single cognitive domain, whilst in four studies multiple domains of cognitive function were targeted. The comparison groups either received no intervention or took part in recreational activities (sports, music, arts), speech or language exercises, computerised motor therapy, or motor rehabilitation combined with recreational activity. We found no clear evidence that cognitive training improved global cognition. Although cognitive training was associated with higher scores on global cognition at the end of treatment, the result was imprecise and not statistically significant (6 trials, 178 participants, standardised mean difference (SMD) 0.28, 95% confidence interval (CI) -0.03 to 0.59; low-certainty evidence). There was no evidence of a difference at the end of treatment between cognitive training and control interventions on executive function (5 trials, 112 participants; SMD 0.10, 95% CI -0.28 to 0.48; low-certainty evidence) or visual processing (3 trials, 64 participants; SMD 0.30, 95% CI -0.21 to 0.81; low-certainty evidence). The evidence favoured the cognitive training group on attention (5 trials, 160 participants; SMD 0.36, 95% CI 0.03 to 0.68; low-certainty evidence) and verbal memory (5 trials, 160 participants; SMD 0.37, 95% CI 0.04 to 0.69; low-certainty evidence), but these effects were less certain in sensitivity analyses that excluded a study in which only a minority of the sample were cognitively impaired. There was no evidence of differences between treatment and control groups in activities of daily living (3 trials, 67 participants; SMD 0.03, 95% CI -0.47 to 0.53; low-certainty evidence) or quality of life (5 trials, 147 participants; SMD -0.01, 95% CI -0.35 to 0.33; low-certainty evidence). There was very little information on adverse events. We considered the certainty of the evidence for all outcomes to be low due to risk of bias in the included studies and imprecision of the results. We identified six ongoing trials recruiting participants with PD-MCI, but no ongoing trials of cognitive training for people with PDD. AUTHORS' CONCLUSIONS This review found no evidence that people with PD-MCI or PDD who receive cognitive training for four to eight weeks experience any important cognitive improvements at the end of training. However, this conclusion was based on a small number of studies with few participants, limitations of study design and execution, and imprecise results. There is a need for more robust, adequately powered studies of cognitive training before conclusions can be drawn about the effectiveness of cognitive training for people with PDD and PD-MCI. Studies should use formal criteria to diagnose cognitive impairments, and there is a particular need for more studies testing the efficacy of cognitive training in people with PDD.
Collapse
Affiliation(s)
- Vasiliki Orgeta
- University College LondonDivision of Psychiatry6th Floor, Maple House,149 Tottenham Court Road,LondonUKW1T 7NF
| | - Kathryn R McDonald
- University of ManchesterDivision of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health3.306, Jean McFarlane Building, Oxford RoadManchesterUKM13 9PL
| | - Ellen Poliakoff
- Division of Neuroscience and Experimental Psychology, School of Biological SciencesUniversity of ManchesterManchesterUKM13 9PL
| | - John Vincent Hindle
- Llandudno Hospital, Betsi Cadwaladr University Health BoardCare of the Elderly DepartmentHospital RoadLlandudnoConwyUKLL30 1LB
| | - Linda Clare
- University of ExeterREACH: The Centre for Research in Ageing and Cognitive HealthPerry RoadExeterUKEX4 4QG
| | - Iracema Leroi
- Trinity College DublinGlobal Brain Health InstituteDublinIreland
| | | |
Collapse
|
115
|
Azam S, Haque ME, Jakaria M, Jo SH, Kim IS, Choi DK. G-Protein-Coupled Receptors in CNS: A Potential Therapeutic Target for Intervention in Neurodegenerative Disorders and Associated Cognitive Deficits. Cells 2020; 9:cells9020506. [PMID: 32102186 PMCID: PMC7072884 DOI: 10.3390/cells9020506] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/15/2020] [Accepted: 02/18/2020] [Indexed: 12/17/2022] Open
Abstract
Neurodegenerative diseases are a large group of neurological disorders with diverse etiological and pathological phenomena. However, current therapeutics rely mostly on symptomatic relief while failing to target the underlying disease pathobiology. G-protein-coupled receptors (GPCRs) are one of the most frequently targeted receptors for developing novel therapeutics for central nervous system (CNS) disorders. Many currently available antipsychotic therapeutics also act as either antagonists or agonists of different GPCRs. Therefore, GPCR-based drug development is spreading widely to regulate neurodegeneration and associated cognitive deficits through the modulation of canonical and noncanonical signals. Here, GPCRs’ role in the pathophysiology of different neurodegenerative disease progressions and cognitive deficits has been highlighted, and an emphasis has been placed on the current pharmacological developments with GPCRs to provide an insight into a potential therapeutic target in the treatment of neurodegeneration.
Collapse
Affiliation(s)
- Shofiul Azam
- Department of Applied Life Science & Integrated Bioscience, Graduate School, Konkuk University, Chungju 27478, Korea; (S.A.); (M.E.H.); (M.J.); (S.-H.J.)
| | - Md. Ezazul Haque
- Department of Applied Life Science & Integrated Bioscience, Graduate School, Konkuk University, Chungju 27478, Korea; (S.A.); (M.E.H.); (M.J.); (S.-H.J.)
| | - Md. Jakaria
- Department of Applied Life Science & Integrated Bioscience, Graduate School, Konkuk University, Chungju 27478, Korea; (S.A.); (M.E.H.); (M.J.); (S.-H.J.)
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Song-Hee Jo
- Department of Applied Life Science & Integrated Bioscience, Graduate School, Konkuk University, Chungju 27478, Korea; (S.A.); (M.E.H.); (M.J.); (S.-H.J.)
| | - In-Su Kim
- Department of Integrated Bioscience & Biotechnology, College of Biomedical and Health Science, and Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Korea
- Correspondence: (I.-S.K.); (D.-K.C.); Tel.: +82-010-3876-4773 (I.-S.K.); +82-43-840-3610 (D.-K.C.); Fax: +82-43-840-3872 (D.-K.C.)
| | - Dong-Kug Choi
- Department of Applied Life Science & Integrated Bioscience, Graduate School, Konkuk University, Chungju 27478, Korea; (S.A.); (M.E.H.); (M.J.); (S.-H.J.)
- Department of Integrated Bioscience & Biotechnology, College of Biomedical and Health Science, and Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Korea
- Correspondence: (I.-S.K.); (D.-K.C.); Tel.: +82-010-3876-4773 (I.-S.K.); +82-43-840-3610 (D.-K.C.); Fax: +82-43-840-3872 (D.-K.C.)
| |
Collapse
|
116
|
Knox MG, Adler CH, Shill HA, Driver-Dunckley E, Mehta SA, Belden C, Zamrini E, Serrano G, Sabbagh MN, Caviness JN, Sue LI, Davis KJ, Dugger BN, Beach TG. Neuropathological Findings in Parkinson's Disease With Mild Cognitive Impairment. Mov Disord 2020; 35:845-850. [PMID: 32034933 DOI: 10.1002/mds.27991] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 01/14/2020] [Accepted: 01/17/2020] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE There are few neuropathological studies on Parkinson's disease with mild cognitive impairment (PD-MCI). Those published reveal coexisting Lewy body and Alzheimer's disease pathology. Our objective is to determine the pathology that underlies PD-MCI. METHODS We used data from the Arizona Study of Aging and Neurodegenerative Disorders, a longitudinal clinicopathological study. Of 736 autopsied subjects with standardized movement and cognitive assessments, 25 had PD-MCI. Neuropathological findings, including Lewy body and Alzheimer's disease pathology, were compared in PD subjects with amnestic MCI (A-MCI) and nonamnestic MCI (NA-MCI). RESULTS Significant pathological heterogeneity within PD-MCI was found. This included varying Lewy body stages, Alzheimer's disease pathology, and cerebral amyloid angiopathy. There was a significant increase in the severity of Lewy body pathology (meeting The Unified Staging System for Lewy Body disorders neocortical stage) in nonamnestic MCI (7/1, 63%) when compared with amnestic MCI (3/14, 21%, P = 0.032). CONCLUSION Although a small study, distinct pathological changes may contribute to PD-MCI phenotype. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Molly G Knox
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Holly A Shill
- Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Erika Driver-Dunckley
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Shyamal A Mehta
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Christine Belden
- Cleo Roberts Center, Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Edward Zamrini
- Cleo Roberts Center, Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Geidy Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - John N Caviness
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Lucia I Sue
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Kathryn J Davis
- Cleo Roberts Center, Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Brittany N Dugger
- Department of Pathology and Laboratory Medicine, University of California-Davis, Davis, California, USA
| | - Thomas G Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona, USA
| |
Collapse
|
117
|
Zhao N, Attrebi ON, Ren Y, Qiao W, Sonustun B, Martens YA, Meneses AD, Li F, Shue F, Zheng J, Van Ingelgom AJ, Davis MD, Kurti A, Knight JA, Linares C, Chen Y, Delenclos M, Liu CC, Fryer JD, Asmann YW, McLean PJ, Dickson DW, Ross OA, Bu G. APOE4 exacerbates α-synuclein pathology and related toxicity independent of amyloid. Sci Transl Med 2020; 12:eaay1809. [PMID: 32024798 PMCID: PMC8309690 DOI: 10.1126/scitranslmed.aay1809] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 12/03/2019] [Indexed: 12/17/2022]
Abstract
The apolipoprotein E (APOE) ε4 allele is the strongest genetic risk factor for late-onset Alzheimer's disease mainly by driving amyloid-β pathology. Recently, APOE4 has also been found to be a genetic risk factor for Lewy body dementia (LBD), which includes dementia with Lewy bodies and Parkinson's disease dementia. How APOE4 drives risk of LBD and whether it has a direct effect on α-synuclein pathology are not clear. Here, we generated a mouse model of synucleinopathy using an adeno-associated virus gene delivery of α-synuclein in human APOE-targeted replacement mice expressing APOE2, APOE3, or APOE4. We found that APOE4, but not APOE2 or APOE3, increased α-synuclein pathology, impaired behavioral performances, worsened neuronal and synaptic loss, and increased astrogliosis at 9 months of age. Transcriptomic profiling in APOE4-expressing α-synuclein mice highlighted altered lipid and energy metabolism and synapse-related pathways. We also observed an effect of APOE4 on α-synuclein pathology in human postmortem brains with LBD and minimal amyloid pathology. Our data demonstrate a pathogenic role of APOE4 in exacerbating α-synuclein pathology independent of amyloid, providing mechanistic insights into how APOE4 increases the risk of LBD.
Collapse
Affiliation(s)
- Na Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Olivia N Attrebi
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yingxue Ren
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Wenhui Qiao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Berkiye Sonustun
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yuka A Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Axel D Meneses
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Fuyao Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Jiaying Zheng
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Mary D Davis
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Aishe Kurti
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Joshua A Knight
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Cynthia Linares
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yixing Chen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Marion Delenclos
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - John D Fryer
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yan W Asmann
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Pamela J McLean
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
118
|
Liu Y, Tong S, Ding L, Liu N, Gao D. Serum levels of glial cell line-derived neurotrophic factor and multiple neurotransmitters: In relation to cognitive performance in Parkinson's disease with mild cognitive impairment. Int J Geriatr Psychiatry 2020; 35:153-162. [PMID: 31650626 DOI: 10.1002/gps.5222] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 09/24/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Mild cognitive impairment is a common non-motor feature of Parkinson's disease, termed PD-MCI. But there is a scarcity of data on the role of glial cell line-derived neurotrophic factor (GDNF) and neurotransmitters in pathogenesis of PD-MCI. The aim of this project was to detect the serum levels of GDNF and multiple neurotranmitters and explore their relationships with cognitive performance in PD-MCI patients. METHODS Neuropsychological testing was administered to PD patients and healthy controls to investigate different domains of cognitive function. Serum levels of GDNF and four cognition-related neurotransmitters including Dopamine metabolites Homovanillic acid (HVA), acetylcholine (Ach), γ-aminobutyric acid (GABA) and 5-hydroxytryptamine (5-HT) were detected by enzyme-linked immunosorbent assay and liquid chromatography-electrospray ionization tandem mass spectrometry analysis respectively. RESULTS The more serious cognitive impairment of PD, the lower levels of GDNF, HVA and 5-HT. In PD-MCI patients, the levels of GDNF, HVA, Ach, 5-HT, and GABA had a significant positive correlation with Digit span backward test (DSB-T) scores and negative correlation with the scores of Trail Making Test A (TMT-A) and Trail Making Test B (TMT-B) respectively. Effect size analysis showed that GDNF and GDNF*Ach have a significant effect on DSB-T, TMT-A and TMT-B respectively; GDNF*HVA, GDNF*5-HT and GDNF*GABA play important part in Auditory Verbal Learning Test separately. CONCLUSIONS Serum GDNF may be involved in the impairment of attention, memory and executive function of PD-MCI patients, by acting alone or in conjunction with neurotransmitters (HVA, 5-HT, GABA, and Ach).
Collapse
Affiliation(s)
- Yi Liu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University. 209 Tongshan Road, Xuzhou, PR China
| | - Shuyan Tong
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University. 209 Tongshan Road, Xuzhou, PR China
| | - Li Ding
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University. 209 Tongshan Road, Xuzhou, PR China
| | - Na Liu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University. 209 Tongshan Road, Xuzhou, PR China
| | - Dianshuai Gao
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University. 209 Tongshan Road, Xuzhou, PR China
| |
Collapse
|
119
|
Ye R, Touroutoglou A, Brickhouse M, Katz S, Growdon JH, Johnson KA, Dickerson BC, Gomperts SN. Topography of cortical thinning in the Lewy body diseases. NEUROIMAGE-CLINICAL 2020; 26:102196. [PMID: 32059167 PMCID: PMC7016450 DOI: 10.1016/j.nicl.2020.102196] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 11/15/2022]
Abstract
Objective Regional cortical thinning in dementia with Lewy bodies (DLB) and Parkinson disease dementia (PDD) may underlie some aspect of their clinical impairments; cortical atrophy likely reflects extensive Lewy body pathology with alpha-synuclein deposits, as well as associated Alzheimer's disease co-pathologies, when present. Here we investigated the topographic distribution of cortical thinning in these Lewy body diseases compared to cognitively normal PD and healthy non-PD control subjects, explored the association of regional thinning with clinical features and evaluated the impact of amyloid deposition. Methods Twenty-one participants with dementia with Lewy bodies (DLB), 16 with Parkinson disease (PD) - associated cognitive impairment (PD-MCI and PDD), and 24 cognitively normal participants with PD underwent MRI, PiB PET, and clinical evaluation. Cortical thickness across the brain and in regions of interest (ROIs) was compared across diagnostic groups and across subgroups stratified by amyloid status, and was related to clinical and cognitive measures. Results DLB and PD-impaired groups shared a similar distribution of cortical thinning that included regions characteristic of AD, as well as the fusiform, precentral, and paracentral gyri. Elevated PiB retention in DLB and PD-impaired but not in PD-normal participants was associated with more extensive and severe cortical thinning, in an overlapping topography that selectively affected the medial temporal lobe in DLB participants. In DLB, greater thinning in AD signature and fusiform regions was associated with greater cognitive impairment. Conclusions The pattern of cortical thinning is similar in DLB and PD-associated cognitive impairment, overlapping with and extending beyond AD signature regions to involve fusiform, precentral, and paracentral regions. Cortical thinning in AD signature and fusiform regions in these diseases reflects cognitive impairment and is markedly accentuated by amyloid co-pathology. Further work will be required to determine whether the distinct topography of cortical thinning in DLB and PD-associated cognitive impairment might have value as a diagnostic and/ or outcome biomarker in clinical trials.
Collapse
Affiliation(s)
- Rong Ye
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Alexandra Touroutoglou
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Michael Brickhouse
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Samantha Katz
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - John H Growdon
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Bradford C Dickerson
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Stephen N Gomperts
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA.
| |
Collapse
|
120
|
Yoo SW, Oh YS, Yoo JY, Ryu DW, Lee KS, Shin NY, Kim JS. Intervening Effects of Orthostatic Blood Pressure Change on Subcortical Atrophy and Cognition in De Novo and Drug-Naïve Parkinson’s Disease. JOURNAL OF PARKINSONS DISEASE 2020; 10:153-160. [DOI: 10.3233/jpd-191748] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Sang-Won Yoo
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoon-Sang Oh
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji-Yeon Yoo
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dong-Woo Ryu
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kwang-Soo Lee
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Na-Young Shin
- Department of Radiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Joong-Seok Kim
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
121
|
Tajiri Y, Wada-Isoe K, Tanaka K, Adachi T, Hanajima R, Nakashima K. A Single-institution Study on Predictors of Short-term Progression from Mild Cognitive Impairment in Parkinson's Disease to Parkinson's Disease with Dementia. Yonago Acta Med 2020; 63:28-33. [PMID: 32158330 DOI: 10.33160/yam.2020.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 12/20/2019] [Indexed: 11/05/2022]
Abstract
Background Patients with non-demented Parkinson's disease (PD) sometime have mild cognitive impairment (MCI), and mild cognitive impairment in Parkinson's disease (PD-MCI) may convert to Parkinson's disease with dementia (PDD) within several years. Cognitive impairment also occurs in the early stages of the disease, gradually progressing to lower quality of life and instrumental activities of daily living. It is important to elucidate the predictors of progression from PD-MCI to PDD via longitudinal studies. Methods This was a single center, case-control study. We analysed data from 49 patients with PD-MCI diagnosed as level I using the Movement Disorder Society PD-MCI criteria at baseline who had completed 1.5 years of follow-up. We defined patients who progressed to PDD as patients with progressive PD-MCI and patients who did not progress to PDD as patients with non-progressive PD-MCI. Depression, apathy, sleep disorders, constipation, light-headedness, hallucinations, impulse control disorders (ICDs) and impulsive-compulsive behaviors (ICBs) at baseline were statistically analysed as predictors of progression. Results Of the 49 PD-MCI patients, 33 did not convert to PDD (non-progressive PD-MCI), and 16 converted to PDD (progressive PD-MCI). The Mini-Mental State Examination (MMSE) score, light-headedness and ICDs were elucidated as predictors of progressive PD-MCI via a multivariate logistic regression model. The adjusted odds ratios (ORs) and 95% confidence intervals (CIs) for each item were MMSE score, OR 0.324, 95% CI 0.119-0.882, P = 0.027; light-headedness, OR 27.665, 95% CI 2.263-338.185, P= 0.009; and ICDs, OR 53.451, 95% CI 2.298-291.085, P = 0.010. Conclusion Cognitive function, ICDs and light-headedness may be risk factors for the development of PDD in PD-MCI patients.
Collapse
Affiliation(s)
- Yuki Tajiri
- Division of Neurology, Department of Brain and Neurosciences, Tottori University Faculty of Medicine, Yonago 683-8504, Japan
| | - Kenji Wada-Isoe
- Division of Neurology, Department of Brain and Neurosciences, Tottori University Faculty of Medicine, Yonago 683-8504, Japan.,Department of Dementia Research, Kawasaki Medical School, Okayama 700-8505, Japan
| | - Kenichiro Tanaka
- Division of Neurology, Department of Brain and Neurosciences, Tottori University Faculty of Medicine, Yonago 683-8504, Japan
| | - Tadashi Adachi
- Division of Neurology, Department of Brain and Neurosciences, Tottori University Faculty of Medicine, Yonago 683-8504, Japan
| | - Ritsuko Hanajima
- Division of Neurology, Department of Brain and Neurosciences, Tottori University Faculty of Medicine, Yonago 683-8504, Japan
| | - Kenji Nakashima
- National Hospital Organization Matsue Medical Center, Matsue 690-8556, Japan
| |
Collapse
|
122
|
Dementia in long-term Parkinson's disease patients: a multicentre retrospective study. NPJ PARKINSONS DISEASE 2020; 6:2. [PMID: 31934610 PMCID: PMC6946687 DOI: 10.1038/s41531-019-0106-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 12/10/2019] [Indexed: 01/12/2023]
Abstract
While several studies have investigated the clinical progression of cognitive decline in Parkinson’s disease (PD) patients, there has been a paucity of data on specifically evaluating PD patients with a disease duration of over 20 years. This study retrospectively investigated the frequency of dementia in PD (PDD) patients with a disease duration of over 20 years assessed in research clinics across the UK and Australia. Data from 2327 PD patients meeting the United Kingdom Parkinson’s Disease Society Brain Bank Criteria was pooled. A diagnosis of probable PDD was made according to the Movement Disorder Society Level 1 criteria. Thirty-six participants had a disease duration of at least 20 years. Of the 36 patients, only 7 (19%) were classified as probable PDD. Compared to PD patients without dementia, those with dementia had lower levels of educational attainment and exhibited more severe motor features. Additionally, 34 out of the 36 patients (94%) exhibited a non-tremor dominant phenotype. No significant differences in age, age onset, disease duration, dopaminergic medication use, and sex distribution were observed between PD patients with and without dementia. Findings from the present study suggest that the prevalence of dementia in long-term PD patients may be lower than anticipated and suggest that the trajectory of cognitive decline in PD patients can be different. These findings highlight the need to investigate factors that might affect the outcome of cognitive decline in long-term PD patients, which may lead to the determination of potential modulating factors in the development of dementia in these patients.
Collapse
|
123
|
Alirezaei Z, Pourhanifeh MH, Borran S, Nejati M, Mirzaei H, Hamblin MR. Neurofilament Light Chain as a Biomarker, and Correlation with Magnetic Resonance Imaging in Diagnosis of CNS-Related Disorders. Mol Neurobiol 2020; 57:469-491. [PMID: 31385229 PMCID: PMC6980520 DOI: 10.1007/s12035-019-01698-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/09/2019] [Indexed: 12/11/2022]
Abstract
The search for diagnostic and prognostic biomarkers for neurodegenerative conditions is of high importance, since these disorders may present difficulties in differential diagnosis. Biomarkers with high sensitivity and specificity are required. Neurofilament light chain (NfL) is a unique biomarker related to axonal damage and neural cell death, which is elevated in a number of neurological disorders, and can be detected in cerebrospinal fluid (CSF), as well as blood, serum, or plasma samples. Although the NfL concentration in CSF is higher than that in blood, blood measurement may be easier in practice due to its lesser invasiveness, reproducibility, and convenience. Many studies have investigated NfL in both CSF and serum/plasma as a potential biomarker of neurodegenerative disorders. Neuroimaging biomarkers can also potentially improve detection of CNS-related disorders at an early stage. Magnetic resonance imaging (MRI) and diffusion tensor imaging (DTI) are sensitive techniques to visualize neuroaxonal loss. Therefore, investigating the combination of NfL levels with indices extracted from MRI and DTI scans could potentially improve diagnosis of CNS-related disorders. This review summarizes the evidence for NfL being a reliable biomarker in the early detection and disease management in several CNS-related disorders. Moreover, we highlight the correlation between MRI and NfL and ask whether they can be combined.
Collapse
Affiliation(s)
- Zahra Alirezaei
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Sarina Borran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Nejati
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, 40 Blossom Street, Boston, MA, 02114, USA.
| |
Collapse
|
124
|
Behavioral defects associated with amygdala and cortical dysfunction in mice with seeded α-synuclein inclusions. Neurobiol Dis 2019; 134:104708. [PMID: 31837424 PMCID: PMC7206936 DOI: 10.1016/j.nbd.2019.104708] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 11/20/2019] [Accepted: 12/08/2019] [Indexed: 12/20/2022] Open
Abstract
Parkinson’s disease (PD) is defined by motor symptoms such as tremor at rest, bradykinesia, postural instability, and stiffness. In addition to the classical motor defects that define PD, up to 80% of patients experience cognitive changes and psychiatric disturbances, referred to as PD dementia (PDD). Pathologically, PD is characterized by loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and intracellular inclusions, called Lewy bodies and Lewy neurites, composed mostly of α-synuclein. Much of PD research has focused on the role of α-synuclein aggregates in degeneration of SNpc dopamine neurons because of the impact of loss of striatal dopamine on the classical motor phenotypes. However, abundant Lewy pathology is also found in other brain regions including the cortex and limbic brain regions such as the amygdala, which may contribute to non-motor phenotypes. Little is known about the consequences of α-synuclein inclusions in these brain regions, or in neuronal subtypes other than dopamine neurons. This project expands knowledge on how α-synuclein inclusions disrupt behavior, specifically non-motor symptoms of synucleinopathies. We show that bilateral injections of fibrils into the striatum results in robust bilateral α-synuclein inclusion formation in the cortex and amygdala. Inclusions in the amygdala and prefrontal cortex primarily localize to excitatory neurons, but unbiased stereology shows no significant loss of neurons in the amygdala or cortex. Fibril injected mice show defects in a social dominance behavioral task and fear conditioning, tasks that are associated with prefrontal cortex and amygdala function. Together, these observations suggest that seeded α-synuclein inclusion formation impairs behaviors associated with cortical and amygdala function, without causing cell loss, in brain areas that may play important roles in the complex cognitive features of PDD
Collapse
|
125
|
Chen F, Wu T, Luo Y, Li Z, Guan Q, Meng X, Tao W, Zhang H. Amnestic mild cognitive impairment in Parkinson's disease: White matter structural changes and mechanisms. PLoS One 2019; 14:e0226175. [PMID: 31830080 PMCID: PMC6907797 DOI: 10.1371/journal.pone.0226175] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022] Open
Abstract
Mild cognitive impairment (MCI) is a heterogeneous cognitive disorder that is often comorbid with Parkinson's diseases (PD). The amnestic subtype of PD-MCI (PD-aMCI) has a higher risk to develop dementia. However, there is a lack of studies on the white matter (WM) structural changes of PD-aMCI. We characterized the WM structural changes of PD-aMCI (n = 17) with cognitively normal PD (PD-CN, n = 19) and normal controls (n = 20), using voxel-based and tract-based spatial statistics (TBSS) analyses on fractional anisotropy (FA) axial diffusivity (AD), and radial diffusivity (RD). By excluding and then including the motor performance as a covariate in the comparison analysis between PD-aMCI and PD-CN, we attempted to discern the influences of two neuropathological mechanisms on the WM structural changes of PD-aMCI. The correlation analyses between memory and voxel-based WM measures in all PD patients were also performed (n = 36). The results showed that PD-aMCI had smaller FA values than PD-CN in the diffuse WM areas, and PD-CN had higher AD and RD values than normal controls in the right caudate. Most FA difference between PD-aMCI and PD-CN could be weakened by the motor adjustment. The FA differences between PD-aMCI and PD-CN were largely spatially overlapped with the memory-correlated FA values. Our findings demonstrated that the WM structural differences between PD-aMCI and PD-CN were mainly memory-related, and the influence of motor adjustment might indicate a common mechanism underlying both motor and memory impairment in PD-aMCI, possibly reflecting a predominant influence of dopaminergic neuropathology.
Collapse
Affiliation(s)
- Fuyong Chen
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong Province, China
- Shenzhen University Clinical Research Center for Neurological Diseases, Shenzhen, Guangdong Province, China
- Department of Neurosurgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Tao Wu
- Department of Neurology, National Clinical Research Center for Geriatric Disorders, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory on Parkinson's Disease, Parkinson Disease Center of Beijing Institute for Brain Disorders, Beijing, China
| | - Yuejia Luo
- School of Psychology, Shenzhen University, Shenzhen, Guangdong Province, China
- Shenzhen Key Laboratory of Affective and Social Cognitive Science, Shenzhen, Guangdong Province, China
| | - Zhihao Li
- School of Psychology, Shenzhen University, Shenzhen, Guangdong Province, China
- Shenzhen Key Laboratory of Affective and Social Cognitive Science, Shenzhen, Guangdong Province, China
| | - Qing Guan
- School of Psychology, Shenzhen University, Shenzhen, Guangdong Province, China
- Shenzhen Key Laboratory of Affective and Social Cognitive Science, Shenzhen, Guangdong Province, China
| | - Xianghong Meng
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong Province, China
- Shenzhen University Clinical Research Center for Neurological Diseases, Shenzhen, Guangdong Province, China
| | - Wei Tao
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong Province, China
- Shenzhen University Clinical Research Center for Neurological Diseases, Shenzhen, Guangdong Province, China
| | - Haobo Zhang
- School of Psychology, Shenzhen University, Shenzhen, Guangdong Province, China
- Shenzhen Key Laboratory of Affective and Social Cognitive Science, Shenzhen, Guangdong Province, China
- Center for Emotion and Brain, Shenzhen Institute of Neuroscience, Shenzhen, Guangdong Province, China
- * E-mail:
| |
Collapse
|
126
|
Vila M. Neuromelanin, aging, and neuronal vulnerability in Parkinson's disease. Mov Disord 2019; 34:1440-1451. [PMID: 31251435 PMCID: PMC7079126 DOI: 10.1002/mds.27776] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 12/22/2022] Open
Abstract
Neuromelanin, a dark brown intracellular pigment, has long been associated with Parkinson's disease (PD). In PD, neuromelanin-containing neurons preferentially degenerate, tell-tale neuropathological inclusions form in close association with this pigment, and neuroinflammation is restricted to neuromelanin-containing areas. In humans, neuromelanin accumulates with age, which in turn is the main risk factor for PD. The potential contribution of neuromelanin to PD pathogenesis remains unknown because, in contrast to humans, common laboratory animals lack neuromelanin. The recent introduction of a rodent model exhibiting an age-dependent production of human-like neuromelanin has allowed, for the first time, for the consequences of progressive neuromelanin accumulation-up to levels reached in elderly human brains-to be assessed in vivo. In these animals, intracellular neuromelanin accumulation above a specific threshold compromises neuronal function and triggers a PD-like pathology. As neuromelanin levels reach this threshold in PD patients and presymptomatic PD patients, the modulation of neuromelanin accumulation could provide a therapeutic benefit for PD patients and delay brain aging. © 2019 The Author. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Miquel Vila
- Neurodegenerative Diseases Research GroupVall d'Hebron Research Institute–Center for Networked Biomedical Research on Neurodegenerative DiseasesBarcelonaSpain
- Department of Biochemistry and Molecular BiologyAutonomous University of BarcelonaBarcelonaSpain
- Catalan Institution for Research and Advanced StudiesBarcelonaSpain
| |
Collapse
|
127
|
Behnke S, Pilotto A, Liepelt-Scarfone I, Yilmaz R, Pausch C, Dieterich S, Bürmann J, Spiegel J, Dillmann U, Unger M, Posner I, Berg D. Third ventricular width assessed by transcranial ultrasound correlates with cognitive performance in Parkinson's disease. Parkinsonism Relat Disord 2019; 66:68-73. [DOI: 10.1016/j.parkreldis.2019.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/08/2019] [Accepted: 07/06/2019] [Indexed: 12/14/2022]
|
128
|
Seeding of protein aggregation causes cognitive impairment in rat model of cortical synucleinopathy. Mov Disord 2019; 34:1699-1710. [DOI: 10.1002/mds.27810] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 07/09/2019] [Accepted: 07/11/2019] [Indexed: 12/15/2022] Open
|
129
|
Hassani SA, Lendor S, Boyaci E, Pawliszyn J, Womelsdorf T. Multineuromodulator measurements across fronto-striatal network areas of the behaving macaque using solid-phase microextraction. J Neurophysiol 2019; 122:1649-1660. [PMID: 31433731 DOI: 10.1152/jn.00321.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Different neuromodulators rarely act independent from each other to modify neural processes but are instead coreleased, gated, or modulated. To understand this interdependence of neuromodulators and their collective influence on local circuits during different brain states, it is necessary to reliably extract local concentrations of multiple neuromodulators in vivo. Here we describe results using solid-phase microextraction (SPME), a method providing sensitive, multineuromodulator measurements. SPME is a sampling method that is coupled with mass spectrometry to quantify collected analytes. Reliable measurements of glutamate, dopamine, acetylcholine, and choline were made simultaneously within frontal cortex and striatum of two macaque monkeys (Macaca mulatta) during goal-directed behavior. We find glutamate concentrations several orders of magnitude higher than acetylcholine and dopamine in all brain regions. Dopamine was reliably detected in the striatum at tenfold higher concentrations than acetylcholine. Acetylcholine and choline concentrations were detected with high consistency across brain areas within monkeys and between monkeys. These findings illustrate that SPME microprobes provide a versatile novel tool to characterize multiple neuromodulators across different brain areas in vivo to understand the interdependence and covariation of neuromodulators during goal-directed behavior. Such data would be important to better distinguish between different behavioral states and characterize dysfunctional brain states that may be evident in psychiatric disorders.NEW & NOTEWORTHY Our paper reports a reliable and sensitive novel method for measuring the absolute concentrations of glutamate, acetylcholine, choline, dopamine, and serotonin in brain circuits in vivo. We show that this method reliably samples multiple neurochemicals in three brain areas simultaneously while nonhuman primates are engaged in goal-directed behavior. We further describe how the methodology we describe here may be used by electrophysiologists as a low-barrier-to-entry tool for measuring multiple neurochemicals.
Collapse
Affiliation(s)
- Seyed-Alireza Hassani
- Department of Psychology, Vanderbilt University, Nashville, Tennessee.,Department of Biology, Centre for Vision Research, York University, Toronto, Ontario, Canada
| | - Sofia Lendor
- Department of Chemistry, University of Waterloo, Waterloo, Ontario, Canada
| | - Ezel Boyaci
- Department of Chemistry, University of Waterloo, Waterloo, Ontario, Canada
| | - Janusz Pawliszyn
- Department of Chemistry, University of Waterloo, Waterloo, Ontario, Canada
| | - Thilo Womelsdorf
- Department of Psychology, Vanderbilt University, Nashville, Tennessee.,Department of Biology, Centre for Vision Research, York University, Toronto, Ontario, Canada
| |
Collapse
|
130
|
Park YW, Shin N, Chung SJ, Kim J, Lim SM, Lee PH, Lee S, Ahn KJ. Magnetic Resonance Imaging–Visible Perivascular Spaces in Basal Ganglia Predict Cognitive Decline in Parkinson's Disease. Mov Disord 2019; 34:1672-1679. [DOI: 10.1002/mds.27798] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/20/2019] [Accepted: 06/25/2019] [Indexed: 01/17/2023] Open
Affiliation(s)
- Yae Won Park
- Department of Radiology Ewha Womans University College of Medicine Seoul Korea
- Department of Radiology and Research Institute of Radiological Science Yonsei University College of Medicine Seoul Korea
| | - Na‐Young Shin
- Department of Radiology Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea Seoul Korea
| | - Seok Jong Chung
- Department of Neurology Yonsei University College of Medicine Seoul Korea
| | - Jiwoong Kim
- Department of Radiology Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea Seoul Korea
| | - Soo Mee Lim
- Department of Radiology Ewha Womans University College of Medicine Seoul Korea
| | - Phil Hyu Lee
- Department of Neurology Yonsei University College of Medicine Seoul Korea
| | - Seung‐Koo Lee
- Department of Radiology and Research Institute of Radiological Science Yonsei University College of Medicine Seoul Korea
| | - Kook Jin Ahn
- Department of Radiology Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea Seoul Korea
| |
Collapse
|
131
|
Chen K, Yang G, So KF, Zhang L. Activation of Cortical Somatostatin Interneurons Rescues Synapse Loss and Motor Deficits after Acute MPTP Infusion. iScience 2019; 17:230-241. [PMID: 31307003 PMCID: PMC6629723 DOI: 10.1016/j.isci.2019.06.040] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/06/2019] [Accepted: 06/28/2019] [Indexed: 01/06/2023] Open
Abstract
Adult dendritic spines present structural and functional plasticity, which forms the basis of learning and memory. To provide in vivo evidence of spine plasticity under neurotoxicity, we generated an acute motor deficit model by single injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) into adult mice. Acute MPTP infusion impairs motor learnings across test paradigms. In vivo two-photon imaging further revealed MPTP-induced prominent dendritic spine loss and substantially increased calcium spikes in apical tufts of layer 5 pyramidal neurons in the motor cortex. MPTP infusion also decreased the activity of somatostatin (SST)-expressing inhibitory interneurons. Further chemogenetic re-activation of SST interneurons reversed MPTP-induced hyperactivation of dendrites, rescued spine loss, and enhanced motor learning. Taken together, our study reports MPTP-induced structural and functional deficits of dendritic spines and suggests the potency of modulating local inhibitory transmission to relieve neurological disorders. Acute MPTP infusion induces cortical spine loss and motor learning deficits MPTP hyperactivates dendritic Ca2+ spikes and suppresses SST-interneuron activity Chemogenetics activation of SST-interneuron corrects spine loss and motor deficits
Collapse
Affiliation(s)
- Kai Chen
- Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Guang Yang
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA.
| | - Kwok-Fai So
- Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; State Key Laboratory of Brain and Cognitive Science, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P. R. China; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, P. R. China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510530, P. R. China; Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou 510515, P. R. China.
| | - Li Zhang
- Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510530, P. R. China; Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou 510515, P. R. China.
| |
Collapse
|
132
|
Jellinger KA. Neuropathology and pathogenesis of extrapyramidal movement disorders: a critical update-I. Hypokinetic-rigid movement disorders. J Neural Transm (Vienna) 2019; 126:933-995. [PMID: 31214855 DOI: 10.1007/s00702-019-02028-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/05/2019] [Indexed: 02/06/2023]
Abstract
Extrapyramidal movement disorders include hypokinetic rigid and hyperkinetic or mixed forms, most of them originating from dysfunction of the basal ganglia (BG) and their information circuits. The functional anatomy of the BG, the cortico-BG-thalamocortical, and BG-cerebellar circuit connections are briefly reviewed. Pathophysiologic classification of extrapyramidal movement disorder mechanisms distinguish (1) parkinsonian syndromes, (2) chorea and related syndromes, (3) dystonias, (4) myoclonic syndromes, (5) ballism, (6) tics, and (7) tremor syndromes. Recent genetic and molecular-biologic classifications distinguish (1) synucleinopathies (Parkinson's disease, dementia with Lewy bodies, Parkinson's disease-dementia, and multiple system atrophy); (2) tauopathies (progressive supranuclear palsy, corticobasal degeneration, FTLD-17; Guamian Parkinson-dementia; Pick's disease, and others); (3) polyglutamine disorders (Huntington's disease and related disorders); (4) pantothenate kinase-associated neurodegeneration; (5) Wilson's disease; and (6) other hereditary neurodegenerations without hitherto detected genetic or specific markers. The diversity of phenotypes is related to the deposition of pathologic proteins in distinct cell populations, causing neurodegeneration due to genetic and environmental factors, but there is frequent overlap between various disorders. Their etiopathogenesis is still poorly understood, but is suggested to result from an interaction between genetic and environmental factors. Multiple etiologies and noxious factors (protein mishandling, mitochondrial dysfunction, oxidative stress, excitotoxicity, energy failure, and chronic neuroinflammation) are more likely than a single factor. Current clinical consensus criteria have increased the diagnostic accuracy of most neurodegenerative movement disorders, but for their definite diagnosis, histopathological confirmation is required. We present a timely overview of the neuropathology and pathogenesis of the major extrapyramidal movement disorders in two parts, the first one dedicated to hypokinetic-rigid forms and the second to hyperkinetic disorders.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|
133
|
Amin F, Khan MS, Bano B. Mammalian cystatin and protagonists in brain diseases. J Biomol Struct Dyn 2019; 38:2171-2196. [DOI: 10.1080/07391102.2019.1620636] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Fakhra Amin
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Bilqees Bano
- Department of Biochemistry, Faculty of Life Sciences, Aligarh MuslimUniversity, Aligarh, India
| |
Collapse
|
134
|
Resting-state fMRI in Parkinson's disease patients with cognitive impairment: A meta-analysis. Parkinsonism Relat Disord 2019; 62:16-27. [DOI: 10.1016/j.parkreldis.2018.12.016] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 10/29/2018] [Accepted: 12/15/2018] [Indexed: 12/14/2022]
|
135
|
Hanna-Pladdy B, Gullapalli R, Chen H. Functional Magnetic Resonance Imaging Biomarkers Predicting Cognitive Progression in Parkinson Disease: Protocol for a Prospective Longitudinal Cohort Study. JMIR Res Protoc 2019; 8:e12870. [PMID: 31033450 PMCID: PMC6660119 DOI: 10.2196/12870] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/02/2019] [Accepted: 03/03/2019] [Indexed: 01/06/2023] Open
Abstract
Background Cardinal features of Parkinson disease (PD) are motor symptoms, but nonmotor features such as mild cognitive impairment (MCI) are common early in the disease process. MCI can progress and convert to dementia in advanced stages, creating significant disability and reduced quality of life. The primary pathological substrate for cognitive decline in PD is unclear, and there are no reliable biomarkers predicting the risk of conversion to dementia. A subgroup of PD patients with visual hallucinations may display more rapid conversion to dementia, suggesting that regional markers of visuoperceptual dysfunction may be sensitive to pathologic density in posterior cortical regions. Objective The purpose of this project is to characterize PD-MCI and evaluate the utility of genetic and neuroimaging biomarkers in predicting cognitive outcomes with a prospective longitudinal study. We will evaluate whether accelerated cognitive progression may be reflected in biomarkers of early posterior cortical changes reflective of α-synuclein deposition. Methods We will evaluate a cohort of early-stage PD patients with the following methods to predict cognitive progression: (1) serial neuropsychological evaluations including detailed visuoperceptual functioning across 4 years; (2) genetic analysis of SNCA (α-synuclein), MAPT (microtubule-associated tau), and APOE (apolipoprotein E); (3) an event-related functional magnetic resonance imaging paradigm of object recognition memory; and (4) anatomical and regional brain activation changes (resting-state functional magnetic resonance imaging) across 4 years. Results The project received funding from the National Institutes of Health in August 2017, and data collection began in February 2018. Enrollment is ongoing, and subjects will be evaluated annually for 4 years extended across a 5-year project including data analysis and image processing. Conclusions Cognitive, genetic, and structural and functional magnetic resonance imaging will characterize neural network changes predictive of cognitive progression in PD across 4 years. Identification of biomarkers with sensitivity for early prediction and estimation of risk for conversion to dementia in PD will pave the way for effective intervention with neuroprotective therapies during the critical stage when treatment can have the greatest impact. International Registered Report Identifier (IRRID) DERR1-10.2196/12870
Collapse
Affiliation(s)
- Brenda Hanna-Pladdy
- Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rao Gullapalli
- Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Hegang Chen
- Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
136
|
Mild cognitive impairment in Parkinson's disease. J Neural Transm (Vienna) 2019; 126:897-904. [PMID: 30963293 DOI: 10.1007/s00702-019-02003-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/30/2019] [Indexed: 01/18/2023]
Abstract
Understanding on the clinical features and neural mechanisms leading to cognitive impairment and dementia in Parkinson's disease (PD) has notably increased. At time of diagnosis, nearly all PD patients present some degree of cognitive impairment not enough severe as to significantly affect functional independence. However, even mild cognitive changes have a measurable impact to functional capacity in PD. A clinically practical differentiation is based on the importance of executive deficits in the early phases of cognitive impairment in PD and on the evidence stressing the transitional role of posterior-cortical impairment on the progression of PD-MCI to dementia. However, the pattern of cognitive impairment in PD is variable not just to the extents on which are the affected cognitive domains, but also on which are those domains that became affected first. Specific diagnostic criteria for mild cognitive impairment associated with PD (PD-MCI) and dementia (PDD) and operative guidelines for the cognitive assessment have been developed. In the present review, we will describe general notions regarding the mechanisms and the profile of cognitive deterioration in PD, the diagnostic criteria for PD-MCI, and some of the currently recommended assessment approaches.
Collapse
|
137
|
Li Y, Wang C, Wang J, Zhou Y, Ye F, Zhang Y, Cheng X, Huang Z, Liu K, Fei G, Zhong C, Zeng M, Jin L. Mild cognitive impairment in de novo Parkinson's disease: A neuromelanin MRI study in locus coeruleus. Mov Disord 2019; 34:884-892. [PMID: 30938892 DOI: 10.1002/mds.27682] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 02/15/2019] [Accepted: 02/18/2019] [Indexed: 12/11/2022] Open
Affiliation(s)
- Yuanfang Li
- Department of NeurologyZhongshan Hospital, Fudan University Shanghai China
| | - Changpeng Wang
- Department of NeurologyZhongshan Hospital, Fudan University Shanghai China
| | - Jian Wang
- Department of RadiologyZhongshan Hospital, Fudan University Shanghai China
- Shanghai Medical Imaging Institute Shanghai China
| | - Ying Zhou
- Department of NeurologyZhongshan Hospital, Fudan University Shanghai China
| | - Fang Ye
- Department of RadiologyZhongshan Hospital, Fudan University Shanghai China
- Shanghai Medical Imaging Institute Shanghai China
| | - Yong Zhang
- MR Research, GE Healthcare Shanghai China
| | - Xiaoqin Cheng
- Department of NeurologyZhongshan Hospital, Fudan University Shanghai China
| | - Zhen Huang
- Department of NeurologyZhongshan Hospital, Fudan University Shanghai China
| | - Kai Liu
- Department of RadiologyZhongshan Hospital, Fudan University Shanghai China
- Shanghai Medical Imaging Institute Shanghai China
| | - Guoqiang Fei
- Department of NeurologyZhongshan Hospital, Fudan University Shanghai China
| | - Chunjiu Zhong
- Department of NeurologyZhongshan Hospital, Fudan University Shanghai China
| | - Mengsu Zeng
- Department of RadiologyZhongshan Hospital, Fudan University Shanghai China
- Shanghai Medical Imaging Institute Shanghai China
| | - Lirong Jin
- Department of NeurologyZhongshan Hospital, Fudan University Shanghai China
| |
Collapse
|
138
|
Lendor S, Hassani SA, Boyaci E, Singh V, Womelsdorf T, Pawliszyn J. Solid Phase Microextraction-Based Miniaturized Probe and Protocol for Extraction of Neurotransmitters from Brains in Vivo. Anal Chem 2019; 91:4896-4905. [DOI: 10.1021/acs.analchem.9b00995] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Sofia Lendor
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Seyed-Alireza Hassani
- Department of Psychology, Vanderbilt University, PMB 407817, 2301 Vanderbilt Place, Nashville, Tennessee 37240, United States
- Department of Biology, Centre for Vision Research, York University, Toronto, Ontario M6J 1P3, Canada
| | - Ezel Boyaci
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Varoon Singh
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Thilo Womelsdorf
- Department of Psychology, Vanderbilt University, PMB 407817, 2301 Vanderbilt Place, Nashville, Tennessee 37240, United States
- Department of Biology, Centre for Vision Research, York University, Toronto, Ontario M6J 1P3, Canada
| | - Janusz Pawliszyn
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| |
Collapse
|
139
|
Current treatment of behavioral and cognitive symptoms of Parkinson's disease. Parkinsonism Relat Disord 2019; 59:65-73. [PMID: 30852149 DOI: 10.1016/j.parkreldis.2019.02.042] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 02/12/2019] [Accepted: 02/25/2019] [Indexed: 12/19/2022]
Abstract
Cognitive and behavioral symptoms are common in Parkinson's disease, may occur even in the prodromal stages of the disease, worsen with disease progression, and surpass motor symptoms as the major factors affecting patient quality of life and caregiver burden. The symptoms may be caused by the disease pathology or they may represent adverse effects of treatment, or both etiological factors may contribute. Although many of these symptoms are related to dopaminergic dysfunction or dopaminergic medication, other neurotransmitters are involved as well. Behavioral symptoms including impulse control disorders, apathy, psychosis, as well as mild cognitive impairment and dementia are reviewed with a special focus on current treatment approaches.
Collapse
|
140
|
Alwardat M, Schirinzi T, Di Lazzaro G, Sancesario GM, Franco D, Imbriani P, Sinibaldi Salimei P, Bernardini S, Mercuri NB, Pisani A. Association between physical activity and dementia's risk factors in patients with Parkinson's disease. J Neural Transm (Vienna) 2019; 126:319-325. [PMID: 30746564 DOI: 10.1007/s00702-019-01979-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 01/30/2019] [Indexed: 01/04/2023]
Abstract
Evidence suggests that physical activity (PA) exerts beneficial effects on neurodegenerative processes, either as symptomatic relief or disease-modifying strategy. Actually, it may represent a viable neuroprotective intervention in Parkinson's disease dementia (PDD), a severe, frequent, and untreatable complication of Parkinson's disease (PD). According to such hypothesis, this cross-sectional study tested, in PD patients, the association between levels of PA and well-known risk factors for PDD, such as mood disorders and amyloid-β42 CSF content. Amount of PA was measured by the International Physical Activity Questionnaires-Short Form (IPAQ-SF) in 128 cognitively intact PD patients and correlated with the Hamilton-Depression (HAM-D) and the Hamilton-Anxiety (HAM-A) scores; in a homogenous subgroup of 40 patients, it was further correlated with a panel of CSF biomarkers, including amyloid-β42, total α-synuclein, total, and phosphorylated tau. The statistical model was corrected for the main potential confounding factors (motor impairment, dopaminergic treatment, disease duration, age, and sex). Both the HAM-A and HAM-D scores, as well as the Aβ42 CSF content, improved in parallel with the increase of the total week amount of PA. Although with several limitations, we preliminarily demonstrated that a high level of PA is associated with a more favourable profile of PDD risk factors, in terms of both mood disturbances and CSF markers of neurodegeneration. However, confirmative studies are necessary to validate the efficacy of PA as protective intervention for PDD.
Collapse
Affiliation(s)
- Mohammad Alwardat
- Department of Systems Medicine, University of Roma "Tor Vergata", Via Montpellier 1, 00135, Rome, Italy
| | - Tommaso Schirinzi
- Department of Systems Medicine, University of Roma "Tor Vergata", Via Montpellier 1, 00135, Rome, Italy.
| | - Giulia Di Lazzaro
- Department of Systems Medicine, University of Roma "Tor Vergata", Via Montpellier 1, 00135, Rome, Italy
| | - Giulia Maria Sancesario
- Department of Experimental Medicine and Surgery, University of Roma "Tor Vergata", Via Montpellier 1, Rome, 00135, Italy
| | - Donatella Franco
- Department of Systems Medicine, University of Roma "Tor Vergata", Via Montpellier 1, 00135, Rome, Italy
| | - Paola Imbriani
- Department of Systems Medicine, University of Roma "Tor Vergata", Via Montpellier 1, 00135, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Paola Sinibaldi Salimei
- Department of Biomedicine and Prevention, University of Roma "Tor Vergata", Via Montpellier 1, Rome, 00135, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine and Surgery, University of Roma "Tor Vergata", Via Montpellier 1, Rome, 00135, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Roma "Tor Vergata", Via Montpellier 1, 00135, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Antonio Pisani
- Department of Systems Medicine, University of Roma "Tor Vergata", Via Montpellier 1, 00135, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
141
|
Silveira CRA, MacKinley J, Coleman K, Li Z, Finger E, Bartha R, Morrow SA, Wells J, Borrie M, Tirona RG, Rupar CA, Zou G, Hegele RA, Mahuran D, MacDonald P, Jenkins ME, Jog M, Pasternak SH. Ambroxol as a novel disease-modifying treatment for Parkinson's disease dementia: protocol for a single-centre, randomized, double-blind, placebo-controlled trial. BMC Neurol 2019; 19:20. [PMID: 30738426 PMCID: PMC6368728 DOI: 10.1186/s12883-019-1252-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 02/01/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Currently there are no disease-modifying treatments for Parkinson's disease dementia (PDD), a condition linked to aggregation of the protein α-synuclein in subcortical and cortical brain areas. One of the leading genetic risk factors for Parkinson's disease is being a carrier in the gene for β-Glucocerebrosidase (GCase; gene name GBA1). Studies in cell culture and animal models have shown that raising the levels of GCase can decrease levels of α-synuclein. Ambroxol is a pharmacological chaperone for GCase and is able to raise the levels of GCase and could therefore be a disease-modifying treatment for PDD. The aims of this trial are to determine if Ambroxol is safe and well-tolerated by individuals with PDD and if Ambroxol affects cognitive, biochemical, and neuroimaging measures. METHODS This is a phase II, single-centre, double-blind, randomized placebo-controlled trial involving 75 individuals with mild to moderate PDD. Participants will be randomized into Ambroxol high-dose (1050 mg/day), low-dose (525 mg/day), or placebo treatment arms. Assessments will be undertaken at baseline, 6-months, and 12-months follow up times. Primary outcome measures will be the Alzheimer's disease Assessment Scale-cognitive subscale (ADAS-Cog) and the ADCS Clinician's Global Impression of Change (CGIC). Secondary measures will include the Parkinson's disease Cognitive Rating Scale, Clinical Dementia Rating, Trail Making Test, Stroop Test, Unified Parkinson's disease Rating Scale, Purdue Pegboard, Timed Up and Go, and gait kinematics. Markers of neurodegeneration will include MRI and CSF measures. Pharmacokinetics and pharmacodynamics of Ambroxol will be examined through plasma levels during dose titration phase and evaluation of GCase activity in lymphocytes. DISCUSSION If found effective and safe, Ambroxol will be one of the first disease-modifying treatments for PDD. TRIAL REGISTRATION ClinicalTrials.gov NCT02914366, 26 Sep 2016/retrospectively registered.
Collapse
Affiliation(s)
- C R A Silveira
- Cognitive Neurology and Alzheimer's Disease Research Centre, Parkwood Institute - Main Building, Room A230, 550, Wellington Road, London, Ontario, N6G 0A7, Canada.,Lawson Health Research Institute, London, Ontario, Canada
| | - J MacKinley
- Cognitive Neurology and Alzheimer's Disease Research Centre, Parkwood Institute - Main Building, Room A230, 550, Wellington Road, London, Ontario, N6G 0A7, Canada.,Lawson Health Research Institute, London, Ontario, Canada
| | - K Coleman
- Cognitive Neurology and Alzheimer's Disease Research Centre, Parkwood Institute - Main Building, Room A230, 550, Wellington Road, London, Ontario, N6G 0A7, Canada.,Lawson Health Research Institute, London, Ontario, Canada
| | - Z Li
- Cognitive Neurology and Alzheimer's Disease Research Centre, Parkwood Institute - Main Building, Room A230, 550, Wellington Road, London, Ontario, N6G 0A7, Canada.,Lawson Health Research Institute, London, Ontario, Canada
| | - E Finger
- Cognitive Neurology and Alzheimer's Disease Research Centre, Parkwood Institute - Main Building, Room A230, 550, Wellington Road, London, Ontario, N6G 0A7, Canada.,Lawson Health Research Institute, London, Ontario, Canada.,Deparment of Clinical Neurological Science, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - R Bartha
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,Robarts Research Institute, Western University, London, Ontario, Canada
| | - S A Morrow
- Cognitive Neurology and Alzheimer's Disease Research Centre, Parkwood Institute - Main Building, Room A230, 550, Wellington Road, London, Ontario, N6G 0A7, Canada.,Lawson Health Research Institute, London, Ontario, Canada.,Deparment of Clinical Neurological Science, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - J Wells
- Lawson Health Research Institute, London, Ontario, Canada.,Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - M Borrie
- Lawson Health Research Institute, London, Ontario, Canada.,Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - R G Tirona
- Lawson Health Research Institute, London, Ontario, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - C A Rupar
- Lawson Health Research Institute, London, Ontario, Canada.,Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - G Zou
- Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,Robarts Research Institute, Western University, London, Ontario, Canada
| | - R A Hegele
- Lawson Health Research Institute, London, Ontario, Canada.,Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,Robarts Research Institute, Western University, London, Ontario, Canada
| | - D Mahuran
- Laboratory of Medicine and Pathobiology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - P MacDonald
- Deparment of Clinical Neurological Science, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - M E Jenkins
- Lawson Health Research Institute, London, Ontario, Canada.,Deparment of Clinical Neurological Science, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - M Jog
- Lawson Health Research Institute, London, Ontario, Canada.,Deparment of Clinical Neurological Science, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - S H Pasternak
- Cognitive Neurology and Alzheimer's Disease Research Centre, Parkwood Institute - Main Building, Room A230, 550, Wellington Road, London, Ontario, N6G 0A7, Canada. .,Lawson Health Research Institute, London, Ontario, Canada. .,Deparment of Clinical Neurological Science, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada. .,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada. .,Robarts Research Institute, Western University, London, Ontario, Canada.
| |
Collapse
|
142
|
Moretti DV. Available and future treatments for atypical parkinsonism. A systematic review. CNS Neurosci Ther 2019; 25:159-174. [PMID: 30294976 PMCID: PMC6488913 DOI: 10.1111/cns.13068] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 09/01/2018] [Accepted: 09/05/2018] [Indexed: 01/02/2023] Open
Abstract
AIMS Success in treating patients with atypical parkinsonian syndromes, namely progressive supranuclear palsy (PSP), cortico-basal degeneration (CBD), multiple system atrophy (MSA), Parkinson's disease with dementia (PDD), and Lewy body dementia with (LBD), remains exceedingly low. The present work overviews the most influential research literature collected on MEDLINE, ISI Web of Science, Cochrane Library, and Scopus for available treatment in atypical parkinsonisms without time restriction. DISCUSSION Transdermal rotigotine, autologous mesenchymal stem cells, tideglusib, and coenzyme Q10 along with donepezil, rivastigmine, memantine, and the deep brain stimulation have shown some benefits in alleviating symptoms in APS. Moreover, many new clinical trials are ongoing testing microtubule stabilizer, antitau monoclonal antibody, tau acetylation inhibition, cell replacement, selective serotonin reuptake inhibitor, active immunization, inhibition of toxic α-synuclein oligomers formation, and inhibition of microglia. CONCLUSION A detailed knowledge of the pathological mechanism underlying the disorders is needed, and disease-modifying therapies are required to offer better therapeutic options to physician and caregivers of APS patients.
Collapse
|
143
|
Li MG, Chen YY, Chen ZY, Feng J, Liu MY, Lou X, Shu SY, Wang ZF, Ma L. Altered functional connectivity of the marginal division in Parkinson's disease with mild cognitive impairment: A pilot resting-state fMRI study. J Magn Reson Imaging 2019; 50:183-192. [PMID: 30644620 DOI: 10.1002/jmri.26548] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/03/2018] [Accepted: 10/03/2018] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND The marginal division (MrD) is an important subcortical center involved in learning and memory. Mild cognitive impairment (MCI) is commonly seen in patients with Parkinson's disease (PD), but the neurobiological basis is yet to be elucidated. PURPOSE To use resting-state functional magnetic resonance imaging (rs-fMRI) to explore the altered functional connectivity (FC) of the MrD in patients with PD-MCI. STUDY TYPE Prospective pilot study. POPULATION Twenty-five patients with PD-MCI; 25 PD patients and no cognitive impairment (PD-NCI); and 25 healthy control (HC) participants. SEQUENCE 3.0 T GE Healthcare MRI scanner; three-dimensional T1 -weighted fast spoiled gradient recalled echo (3D T1 -FSPGR); rs-fMRI. ASSESSMENT The MrD was defined using manual delineation, which was the seed point to compute the FC to examine correlations between low-frequency fMRI signal fluctuations in MrD and the whole brain. STATISTICAL TESTS Between-group comparisons of the rs-fMRI data were computed using two-sample t-tests in a voxelwise manner after controlling for age and sex, to determine the brain regions that showed significant differences in FC with the bilateral MrDs. Correlation analyses were performed for FC values and cognitive abilities in patients with PD. RESULTS In the PD-MCI group, compared with the PD-NCI group, we observed lesser FC between the MrD bilaterally and right putamen, left insula, left cerebellum, and left thalamus; greater FC between the MrD bilaterally and left middle cingulate cortex, left middle frontal gyrus, left superior frontal gyrus, left supplementary motor area, and left middle/inferior occipital gyrus. Moreover, the strength of FC between the MrD and regions that showed differences between the PD-MCI and PD-NCI groups was significantly correlated with neuropsychological scores in patients with PD. DATA CONCLUSION The current study suggests that MrD dysfunction may contribute to MCI in PD. However, the mechanisms underlying this process require further investigation. Level of Evidence 1. Technical Efficacy Stage 2. J. Magn. Reson. Imaging 2019;50:183-192.
Collapse
Affiliation(s)
- Ming-Ge Li
- School of Medicine, Nankai University, Tianjin, P.R. China.,Department of Radiology, Chinese PLA General Hospital, Beijing, P.R. China
| | | | - Zhi-Ye Chen
- Department of Radiology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Jie Feng
- Department of Radiology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Meng-Yu Liu
- Department of Radiology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Xin Lou
- Department of Radiology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Si-Yun Shu
- Institute of Cognitive Neuroscience, South China Normal University, Guangzhou, P.R. China
| | - Zhen-Fu Wang
- Department of Neurology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Lin Ma
- School of Medicine, Nankai University, Tianjin, P.R. China.,Department of Radiology, Chinese PLA General Hospital, Beijing, P.R. China
| |
Collapse
|
144
|
Geibl FF, Henrich MT, Oertel WH. Mesencephalic and extramesencephalic dopaminergic systems in Parkinson's disease. J Neural Transm (Vienna) 2019; 126:377-396. [PMID: 30643975 DOI: 10.1007/s00702-019-01970-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/08/2019] [Indexed: 12/13/2022]
Abstract
Neurodegeneration of the nigrostriatal dopaminergic system and concurrent dopamine (DA) deficiency in the basal ganglia represent core features of Parkinson's disease (PD). Despite the central role of DA in the pathogenesis of PD, dopaminergic systems outside of the midbrain have not been systematically investigated for Lewy body pathology or neurodegeneration. Dopaminergic neurons show a surprisingly rich neurobiological diversity, suggesting that there is not one general type of dopaminergic neuron, but rather a spectrum of different dopaminergic phenotypes. This heterogeneity on the cellular level could account for the observed differences in susceptibility of the dopaminergic systems to the PD disease process. In this review, we will summarize the long history from the first description of PD to the rationally derived DA replacement therapy, describe the basal neuroanatomical and neuropathological features of the different dopaminergic systems in health and PD, explore how neuroimaging techniques broadened our view of the dysfunctional dopaminergic systems in PD and discuss how dopaminergic replacement therapy ameliorates the classical motor symptoms but simultaneously induces a new set of hyperdopaminergic symptoms.
Collapse
Affiliation(s)
- Fanni F Geibl
- Department of Neurology, Philipps University Marburg, Baldingerstraße 1, 35043, Marburg, Germany.
| | - Martin T Henrich
- Department of Neurology, Philipps University Marburg, Baldingerstraße 1, 35043, Marburg, Germany
| | - Wolfgang H Oertel
- Department of Neurology, Philipps University Marburg, Baldingerstraße 1, 35043, Marburg, Germany
| |
Collapse
|
145
|
Novel Treatment Opportunities Against Cognitive Impairment in Parkinson's Disease with an Emphasis on Diabetes-Related Pathways. CNS Drugs 2019; 33:143-160. [PMID: 30687888 PMCID: PMC6373401 DOI: 10.1007/s40263-018-0601-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cognitive impairment is highly prevalent in patients with Parkinson's disease (PD) and causes adverse health outcomes. Novel procognitive therapies are needed to address this unmet need. It is now established that there is an increased risk of dementia in patients with type 2 diabetes mellitus (T2DM) and, moreover, T2DM and PD may have common underlying biological mechanisms. As such, T2DM medications are emerging as potential therapies in the context of PD dementia (PDD). In this review, we provide an update on pathophysiological mechanisms underlying cognitive impairments and PDD, focusing on diabetes-related pathways. Finally, we have conducted a review of ongoing clinical trials in PD patients with dementia, highlighting the multiple pharmacological mechanisms that are targeted to achieve cognitive enhancement.
Collapse
|
146
|
Schwartz RS, Halliday GM, Soh D, Cordato DJ, Kril JJ. Impact of small vessel disease on severity of motor and cognitive impairment in Parkinson’s disease. J Clin Neurosci 2018; 58:70-74. [DOI: 10.1016/j.jocn.2018.10.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 10/05/2018] [Indexed: 01/05/2023]
|
147
|
Delgado-Alvarado M, Dacosta-Aguayo R, Navalpotro-Gómez I, Gago B, Gorostidi A, Jiménez-Urbieta H, Quiroga-Varela A, Ruiz-Martínez J, Bergareche A, Rodríguez-Oroz MC. Ratios of proteins in cerebrospinal fluid in Parkinson's disease cognitive decline: prospective study. Mov Disord 2018; 33:1809-1813. [PMID: 30423201 DOI: 10.1002/mds.27518] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 08/10/2018] [Accepted: 08/22/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND There is a need for biomarkers of dementia in PD. OBJECTIVES To determine if the levels of the main CSF proteins and their ratios are associated with deterioration in cognition and progression to dementia in the short to mid term. METHODS The Parkinson's Progression Markers Initiative database was used as an exploratory cohort, and a center-based cohort was used as a replication cohort. Amyloid ß1-42, total tau, threonine-181 phosphorylated tau, and α-synuclein in the CSF and the ratios of these proteins were assessed. RESULTS In the Parkinson's Progression Markers Initiative cohort (n = 281), the total tau/amyloid ß1-42, total tau/α-synuclein, total tau/amyloid ß1-42+α-synuclein, and amyloid ß1-42/total tau ratios were associated with a risk of progression to dementia over a 3-year follow-up. In the replication cohort (n = 40), the total tau/α-synuclein and total tau/amyloid ß1-42+α-synuclein ratios were associated with progression to dementia over a 41-month follow-up. CONCLUSION Ratios of the main proteins found in PD patient brain inclusions that can be measured in the CSF appear to have value as short- to mid-term predictors of dementia. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Manuel Delgado-Alvarado
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Rosalía Dacosta-Aguayo
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute, San Sebastián, Spain.,Biomedical Research Networking Centre Consortium for the area of Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Irene Navalpotro-Gómez
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute, San Sebastián, Spain.,Biomedical Research Networking Centre Consortium for the area of Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Belén Gago
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute, San Sebastián, Spain.,Biomedical Research Networking Centre Consortium for the area of Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ana Gorostidi
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute, San Sebastián, Spain.,Biomedical Research Networking Centre Consortium for the area of Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Genomics Platform, Biodonostia Research Institute, San Sebastián, Spain
| | - Haritz Jiménez-Urbieta
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute, San Sebastián, Spain.,Biomedical Research Networking Centre Consortium for the area of Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ana Quiroga-Varela
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute, San Sebastián, Spain.,Biomedical Research Networking Centre Consortium for the area of Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Clinica Universidad de Navarra, Center for Applied Medical Research (CIMA)-Universidad de Navarra, Pamplona, Spain
| | - Javier Ruiz-Martínez
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute, San Sebastián, Spain.,Biomedical Research Networking Centre Consortium for the area of Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Movement Disorders Unit, Department of Neurology, University Hospital Donostia, San Sebastián, Spain
| | - Alberto Bergareche
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute, San Sebastián, Spain.,Biomedical Research Networking Centre Consortium for the area of Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Movement Disorders Unit, Department of Neurology, University Hospital Donostia, San Sebastián, Spain
| | - María C Rodríguez-Oroz
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute, San Sebastián, Spain.,Biomedical Research Networking Centre Consortium for the area of Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Clinica Universidad de Navarra, Center for Applied Medical Research (CIMA)-Universidad de Navarra, Pamplona, Spain.,Movement Disorders Unit, Department of Neurology, University Hospital Donostia, San Sebastián, Spain.,Ikerbasque (Basque Foundation for Science), Bilbao, Spain.,Basque Center on Cognition Brain and Language (BCBL), San Sebastián, Spain
| |
Collapse
|
148
|
Meta-Analysis of the Relationship between the APOE Gene and the Onset of Parkinson's Disease Dementia. PARKINSONS DISEASE 2018; 2018:9497147. [PMID: 30405900 PMCID: PMC6204165 DOI: 10.1155/2018/9497147] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 08/17/2018] [Accepted: 08/30/2018] [Indexed: 12/18/2022]
Abstract
Purpose To clarify the relationship between certain genotypes or alleles of the APOE gene and the onset risk of Parkinson's disease dementia (PDD). Methods The PubMed, Cochrane, Embase, CBM, CNKI, and Wanfang databases were searched to identify all case-control studies and cohort studies published before October 30, 2017, that investigated the association between the APOE gene and the onset of PDD. Manual information retrieval was also performed. All studies that met the quality requirements were included in a meta-analysis performed using RevMan 5.3 software. Results The meta-analysis included 17 studies, with a total of 820 patients in the PDD group and 1,922 in the non-PDD group. The influence of the APOE gene on PDD onset was analyzed from three aspects: five genotypes vs. ε3/3, ε2+/ε4+ vs. ε3/3, and ε4+ vs. ε4-. The risk factors for PDD may include the genotypes ε3/4 (OR 1.47, 95% CI 1.14-1.89) and ε4/4 (OR 2.93, 95% CI 1.20-7.14). In patients with PDD, there was no significant difference in the distribution of ε2+ vs. ε3/3 (OR 1.35, 95% CI 0.97-1.87, P=0.07). The risk of PDD was 1.61 times greater in ε4+ compared with ε3/3 (OR 1.61, 95% CI 1.24-2.08, P=0.0003). As the results indicated that ε2+ did not play a role as a risk factor or a protective factor, we divided the population into ε4+ and ε4- for the meta-analysis and found that, among patients with Parkinson's disease, the dementia risk of those with ε4+ was 1.72 times greater than that of those with ε4- (OR 1.72, 95% CI 1.41-2.10, P < 0.00001). Subgroup analysis in accordance with different geographical regions revealed that ε4+ was a risk factor for PDD in people from all regions. Conclusions Among the APOE genotypes, ε2+ is neither a risk factor nor a protective factor for PDD, while ε4+ is a risk factor for PDD. The present results are applicable to Asian, European, and American patients with Parkinson's disease. Regarding the single APOE genotypes, ε3/4 and ε4/4 may be risk factors for PDD; however, further studies with large sample sizes are needed to verify this.
Collapse
|
149
|
Dadar M, Zeighami Y, Yau Y, Fereshtehnejad SM, Maranzano J, Postuma RB, Dagher A, Collins DL. White matter hyperintensities are linked to future cognitive decline in de novo Parkinson's disease patients. NEUROIMAGE-CLINICAL 2018; 20:892-900. [PMID: 30292088 PMCID: PMC6176552 DOI: 10.1016/j.nicl.2018.09.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 08/14/2018] [Accepted: 09/25/2018] [Indexed: 11/15/2022]
Abstract
White Matter Hyperintensities (WMHs) are associated with cognitive decline in aging and Alzheimer's disease. However, the pathogenesis of cognitive decline in Parkinson's disease (PD) is not as clearly related to vascular causes, and therefore the role of WMHs as a marker of small-vessel disease (SVD) in PD is less clear. Currently, SVD in PD is assessed and treated independently of the disease. However, if WMH as the major MRI sign of SVD has a higher impact on cognitive decline in PD patients than in healthy controls, vascular pathology needs to be assessed and treated with a higher priority in this population. Here we investigate whether the presence of WMHs leads to increased cognitive decline in de novo PD, and if these effects relate to cortical atrophy. WMHs and cortical thickness were measured in de novo PD patients and age-matched controls (NPD = 365, NControl = 174) from Parkinson's Progression Markers Initiative (PPMI) to study the relationship between baseline WMHs, future cognitive decline (follow-up: 4.09 ± 1.14 years) and cortical atrophy (follow-up: 1.05 ± 0.10 years). PD subjects with high baseline WMH loads had significantly greater cognitive decline than i) PD subjects with low WMH load, and ii) control subjects with high WMH load. Furthermore, in PD subjects, high WMH load resulted in more cortical thinning in the right frontal lobe. Theses results show that the presence of WMHs in de novo PD patients predicts greater future cognitive decline and cortical atrophy than in normal aging.
Collapse
Affiliation(s)
- Mahsa Dadar
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada; NeuroImaging and Surgical Tools Laboratory, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| | - Yashar Zeighami
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| | - Yvonne Yau
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| | - Seyed-Mohammad Fereshtehnejad
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada; Division of Neurology, Department of Medicine, University of Ottawa and Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.
| | - Josefina Maranzano
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| | - Ronald B Postuma
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| | - Alain Dagher
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| | - D Louis Collins
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada; NeuroImaging and Surgical Tools Laboratory, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
150
|
Apathy in individuals with Parkinson's disease associated with mild cognitive impairment. A neuropsychological investigation. Neuropsychologia 2018; 118:4-11. [DOI: 10.1016/j.neuropsychologia.2018.05.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 05/18/2018] [Accepted: 05/18/2018] [Indexed: 12/31/2022]
|