1
|
Khalil I, Sayad R, Kedwany AM, Sayed HH, Caprara ALF, Rissardo JP. Cardiovascular dysautonomia and cognitive impairment in Parkinson's disease (Review). MEDICINE INTERNATIONAL 2024; 4:70. [PMID: 39355336 PMCID: PMC11443310 DOI: 10.3892/mi.2024.194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/03/2024] [Indexed: 10/03/2024]
Abstract
Cognitive impairment is a prevalent non-motor symptom of Parkinson's disease (PD), which can result in significant disability and distress for patients and caregivers. There is a marked variation in the timing, characteristics and rate at which cognitive decline occurs in patients with PD. This decline can vary from normal cognition to mild cognitive impairment and dementia. Cognitive impairment is associated with several pathophysiological mechanisms, including the accumulation of β-amyloid and tau in the brain, oxidative stress and neuroinflammation. Cardiovascular autonomic dysfunctions are commonly observed in patients with PD. These dysfunctions play a role in the progression of cognitive impairment, the incidents of falls and even in mortality. The majority of symptoms of dysautonomia arise from changes in the peripheral autonomic nervous system, including both the sympathetic and parasympathetic nervous systems. Cardiovascular changes, including orthostatic hypotension, supine hypertension and abnormal nocturnal blood pressure (BP), can occur in both the early and advanced stages of PD. These changes tend to increase as the disease advances. The present review aimed to describe the cognitive changes in the setting of cardiovascular dysautonomia and to discuss strategies through which these changes can be modified and managed. It is a multifactorial process usually involving decreased blood flow to the brain, resulting in the development of cerebral ischemic lesions, an increased presence of abnormal white matter signals in the brain, and a potential influence on the process of neurodegeneration in PD. Another possible explanation is this association being independent observations of PD progression. Patients with clinical symptoms of dysautonomia should undergo 24-h ambulatory BP monitoring, as they are frequently subtle and underdiagnosed.
Collapse
Affiliation(s)
- Ibrahim Khalil
- Faculty of Medicine, Alexandria University, Alexandria 5372066, Egypt
| | - Reem Sayad
- Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | | | - Hager Hamdy Sayed
- Department of Nuclear Medicine, Assuit University, Assuit 71515, Egypt
| | | | | |
Collapse
|
2
|
Carvalho de Abreu DC, Pieruccini-Faria F, Son S, Montero-Odasso M, Camicioli R. Is white matter hyperintensity burden associated with cognitive and motor impairment in patients with parkinson's disease? A systematic review and meta-analysis. Neurosci Biobehav Rev 2024; 161:105677. [PMID: 38636832 DOI: 10.1016/j.neubiorev.2024.105677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/08/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024]
Abstract
White matter damage quantified as white matter hyperintensities (WMH) may aggravate cognitive and motor impairments, but whether and how WMH burden impacts these problems in Parkinson's disease (PD) is not fully understood. This study aimed to examine the association between WMH and cognitive and motor performance in PD through a systematic review and meta-analysis. We compared the WMH burden across the cognitive spectrum (cognitively normal, mild cognitive impairment, dementia) in PD including controls. Motor signs were compared in PD with low/negative and high/positive WMH burden. We compared baseline WMH burden of PD who did and did not convert to MCI or dementia. MEDLINE and EMBASE databases were used to conduct the literature search resulting in 50 studies included for data extraction. Increased WMH burden was found in individuals with PD compared with individuals without PD (i.e. control) and across the cognitive spectrum in PD (i.e. PD, PD-MCI, PDD). Individuals with PD with high/positive WMH burden had worse global cognition, executive function, and attention. Similarly, PD with high/positive WMH presented worse motor signs compared with individuals presenting low/negative WMH burden. Only three longitudinal studies were retrieved from our search and they showed that PD who converted to MCI or dementia, did not have significantly higher WMH burden at baseline, although no data was provided on WMH burden changes during the follow up. We conclude, based on cross-sectional studies, that WMH burden appears to increase with PD worse cognitive and motor status in PD.
Collapse
Affiliation(s)
- Daniela Cristina Carvalho de Abreu
- Post-doctoral fellow at Gait and Brain Lab, University of Western Ontario, Canada, and Associated Professor of Physiotherapy Course, Department of Health Sciences, Rehabilitation and Functional Performance Program, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.
| | - Frederico Pieruccini-Faria
- Deparment of Medicine, Schulich School of Medicine and Dentistry, The University of Western Ontario, Lawson Health Research Institute, St. Josephs Health Care, Parkwood Institute, Deputy Director of the Gait & Brain Lab, Canada
| | - Surim Son
- Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, University of Western Ontario, Statistician, Departments of Medicine, University of Western Ontario, Canada, Parkwood Institute, Lawson Health Research Institute, Canada
| | - Manuel Montero-Odasso
- Departments of Medicine, and Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, The University of Western Ontario, Canada Director of Gait and Brain Lab, Parkwood Institute, Lawson Health Research Institute, Canada
| | - Richard Camicioli
- Department of Medicine, Division of Neurology, University of Alberta, Canada
| |
Collapse
|
3
|
Paola Caminiti S, Gallo S, Menegon F, Naldi A, Comi C, Tondo G. Lifestyle Modulators of Neuroplasticity in Parkinson's Disease: Evidence in Human Neuroimaging Studies. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:602-613. [PMID: 37326116 DOI: 10.2174/1871527322666230616121213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/25/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by both motor and non-motor symptoms. A progressive neuronal loss and the consequent clinical impairment lead to deleterious effects on daily living and quality of life. Despite effective symptomatic therapeutic approaches, no disease-modifying therapies are currently available. Emerging evidence suggests that adopting a healthy lifestyle can improve the quality of life of PD patients. In addition, modulating lifestyle factors can positively affect the microstructural and macrostructural brain levels, corresponding to clinical improvement. Neuroimaging studies may help to identify the mechanisms through which physical exercise, dietary changes, cognitive enrichment, and exposure to substances modulate neuroprotection. All these factors have been associated with a modified risk of developing PD, with attenuation or exacerbation of motor and non-motor symptomatology, and possibly with structural and molecular changes. In the present work, we review the current knowledge on how lifestyle factors influence PD development and progression and the neuroimaging evidence for the brain structural, functional, and molecular changes induced by the adoption of positive or negative lifestyle behaviours.
Collapse
Affiliation(s)
| | - Silvia Gallo
- Neurology Unit, Department of Translational Medicine, Movement Disorders Centre, University of Piemonte Orientale, 28100 Novara, Italy
| | - Federico Menegon
- Neurology Unit, Department of Translational Medicine, Movement Disorders Centre, University of Piemonte Orientale, 28100 Novara, Italy
| | - Andrea Naldi
- Neurology Unit, San Giovanni Bosco Hospital, 10154 Turin, Italy
| | - Cristoforo Comi
- Neurology Unit, Department of Translational Medicine, S. Andrea Hospital, University of Piemonte Orientale, 13100 Vercelli, Italy
| | - Giacomo Tondo
- Neurology Unit, Department of Translational Medicine, S. Andrea Hospital, University of Piemonte Orientale, 13100 Vercelli, Italy
| |
Collapse
|
4
|
Walker L, Attems J. Prevalence of Concomitant Pathologies in Parkinson's Disease: Implications for Prognosis, Diagnosis, and Insights into Common Pathogenic Mechanisms. JOURNAL OF PARKINSON'S DISEASE 2024; 14:35-52. [PMID: 38143370 PMCID: PMC10836576 DOI: 10.3233/jpd-230154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/11/2023] [Indexed: 12/26/2023]
Abstract
Pathologies characteristic of Alzheimer's disease (i.e., hyperphosphorylated tau and amyloid-β (Aβ) plaques), cardiovascular disease, and limbic predominant TDP-43 encephalopathy (LATE) often co-exist in patients with Parkinson's disease (PD), in addition to Lewy body pathology (α-synuclein). Numerous studies point to a putative synergistic relationship between hyperphosphorylation tau, Aβ, cardiovascular lesions, and TDP-43 with α-synuclein, which may alter the stereotypical pattern of pathological progression and accelerate cognitive decline. Here we discuss the prevalence and relationships between common concomitant pathologies observed in PD. In addition, we highlight shared genetic risk factors and developing biomarkers that may provide better diagnostic accuracy for patients with PD that have co-existing pathologies. The tremendous heterogeneity observed across the PD spectrum is most likely caused by the complex interplay between pathogenic, genetic, and environmental factors, and increasing our understanding of how these relate to idiopathic PD will drive research into finding accurate diagnostic tools and disease modifying therapies.
Collapse
Affiliation(s)
- Lauren Walker
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Johannes Attems
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK
| |
Collapse
|
5
|
Carlisle TC, Medina LD, Holden SK. Original research: initial development of a pragmatic tool to estimate cognitive decline risk focusing on potentially modifiable factors in Parkinson's disease. Front Neurosci 2023; 17:1278817. [PMID: 37942138 PMCID: PMC10628974 DOI: 10.3389/fnins.2023.1278817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/03/2023] [Indexed: 11/10/2023] Open
Abstract
Introduction Cognitive decline is common in Parkinson's disease (PD). Calculating personalized risk of cognitive decline in PD would allow for appropriate counseling, early intervention with available treatments, and inclusion in disease-modifying trials. Methods Data were from the Parkinson's Progression Markers Initiative de novo cohort. Baseline scores were calculated for Lifestyle for Brain Health (LIBRA) and the Montreal Parkinson Risk of Dementia Scale (MoPaRDS) per prior literature and preliminary Parkinson's disease Risk Estimator for Decline In Cognition Tool (pPREDICT) by attributing a point for fourteen posited risk factors. Baseline and 5-year follow-up composite cognitive scores (CCSs) were calculated from a neuropsychological battery and used to define cognitive decliners (PD-decline) versus maintainers (PD-maintain). Results The PD-decline group (n = 44) had higher LIBRA (6.76 ± 0.57, p < 0.05), MoPaRDS (2.45 ± 1.41, p < 0.05) and pPREDICT (4.52 ± 1.66, p < 0.05) scores compared to the PD-maintain group (n = 263; LIBRA 4.98 ± 0.20, MoPaRDS 1.68 ± 1.16, pPREDICT 3.38 ± 1.69). Area-under-the-curve (AUC) for LIBRA was 0.64 (95% confidence interval [CI], 0.55-0.73), MoPaRDS was 0.66 (95% CI, 0.58-0.75) and for pPREDICT was 0.68 (95% CI, 0.61-0.76). In linear regression analyses, LIBRA (p < 0.05), MoPaRDS (p < 0.05) and pPREDICT (p < 0.05) predicted change in CCS. Only age stratified by sex (p < 0.05) contributed significantly to the model for LIBRA. Age and presence of hallucinations (p < 0.05) contributed significantly to the model for MoPaRDS. Male sex, older age, excessive daytime sleepiness, and moderate-severe motor symptoms (all p < 0.05) contributed significantly to the model for pPREDICT. Conclusion Although MoPaRDS is a PD-specific tool for predicting cognitive decline relying on only clinical features, it does not focus on potentially modifiable risk factors. LIBRA does focus on potentially modifiable risk factors and is associated with prediction of all-cause dementia in some populations, but pPREDICT potentially demonstrates improved performance in cognitive decline risk calculation in individuals with PD and may identify actionable risk factors. As pPREDICT incorporates multiple potentially modifiable risk factors that can be obtained easily in the clinical setting, it is a first step in developing an easily assessable tool for a personalized approach to reduce dementia risk in people with PD.
Collapse
Affiliation(s)
- Tara C. Carlisle
- Department of Neurology, Behavioral Neurology Section, University of Colorado School of Medicine, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, Aurora, CO, United States
- University of Colorado Movement Disorders Center, Aurora, CO, United States
| | - Luis D. Medina
- Department of Psychology, University of Houston, Houston, TX, United States
| | - Samantha K. Holden
- Department of Neurology, Behavioral Neurology Section, University of Colorado School of Medicine, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, Aurora, CO, United States
- University of Colorado Movement Disorders Center, Aurora, CO, United States
- Department of Neurology, Movement Disorders Section, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
6
|
Buongiorno M, Marzal C, Fernandez M, Cullell N, de Mena L, Sánchez-Benavides G, de la Sierra A, Krupinski J, Compta Y. Altered sleep and neurovascular dysfunction in alpha-synucleinopathies: the perfect storm for glymphatic failure. Front Aging Neurosci 2023; 15:1251755. [PMID: 37693650 PMCID: PMC10484002 DOI: 10.3389/fnagi.2023.1251755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 08/04/2023] [Indexed: 09/12/2023] Open
Abstract
Clinical and cognitive progression in alpha-synucleinopathies is highly heterogeneous. While some patients remain stable over long periods of time, other suffer early dementia or fast motor deterioration. Sleep disturbances and nocturnal blood pressure abnormalities have been identified as independent risk factors for clinical progression but a mechanistic explanation linking both aspects is lacking. We hypothesize that impaired glymphatic system might play a key role on clinical progression. Glymphatic system clears brain waste during specific sleep stages, being blood pressure the motive force that propels the interstitial fluid through brain tissue to remove protein waste. Thus, the combination of severe sleep alterations, such as REM sleep behavioral disorder, and lack of the physiological nocturnal decrease of blood pressure due to severe dysautonomia may constitute the perfect storm for glymphatic failure, causing increased abnormal protein aggregation and spreading. In Lewy body disorders (Parkinson's disease and dementia with Lewy bodies) the increment of intraneuronal alpha-synuclein and extracellular amyloid-β would lead to cognitive deterioration, while in multisystemic atrophy, increased pathology in oligodendroglia would relate to the faster and malignant motor progression. We present a research model that may help in developing studies aiming to elucidate the role of glymphatic function and associated factors mainly in alpha-synucleinopathies, but that could be relevant also for other protein accumulation-related neurodegenerative diseases. If the model is proven to be useful could open new lines for treatments targeting glymphatic function (for example through control of nocturnal blood pressure) with the objective to ameliorate cognitive and motor progression in alpha-synucleinopathies.
Collapse
Affiliation(s)
- Mariateresa Buongiorno
- Hospital Universitari MútuaTerrassa/Fundacio Docència i Recerca MútuaTerrassa, Terrassa, Spain
| | - Clara Marzal
- Hospital Universitari MútuaTerrassa/Fundacio Docència i Recerca MútuaTerrassa, Terrassa, Spain
| | - Manel Fernandez
- Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders, Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Institut de Neurociències (UBNeuro), Universitat de Barcelona, Barcelona, Spain
| | - Natalia Cullell
- Hospital Universitari MútuaTerrassa/Fundacio Docència i Recerca MútuaTerrassa, Terrassa, Spain
| | - Lorena de Mena
- Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders, Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Institut de Neurociències (UBNeuro), Universitat de Barcelona, Barcelona, Spain
| | - Gonzalo Sánchez-Benavides
- Barcelonaβeta Brain Research Center, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Alejandro de la Sierra
- Hospital Universitari MútuaTerrassa/Fundacio Docència i Recerca MútuaTerrassa, Terrassa, Spain
| | - Jerzy Krupinski
- Hospital Universitari MútuaTerrassa/Fundacio Docència i Recerca MútuaTerrassa, Terrassa, Spain
- Department of Life Sciences John Dalton Building, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Yaroslau Compta
- Parkinson’s Disease and Movement Disorders Unit, Neurology Service, Hospital Clínic i Universitari de Barcelona, CIBERNED (CB06/05/0018-ISCIII), ERN-RND, UBNeuro Institut Clínic de Neurociències (Maria de Maeztu Excellence Centre), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
7
|
Ryman SG, Shaff N, Dodd A, Nitschke S, Wertz C, Julio K, Suarez Cedeno G, Deligtisch A, Erhardt E, Lin H, Vakhtin A, Poston KL, Tarawneh R, Pirio Richardson S, Mayer A. Reduced and Delayed Cerebrovascular Reactivity in Patients with Parkinson's Disease. Mov Disord 2023; 38:1262-1272. [PMID: 37157056 PMCID: PMC10524339 DOI: 10.1002/mds.29429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND Cerebrovascular dysfunction in Parkinson's disease (PD) is heterogeneous and may contribute to disease pathophysiology or progression. There is a need to understand the mechanisms by which cerebrovascular dysfunction is altered in participants with PD. OBJECTIVES The objective of this study is to test the hypothesis that participants with PD exhibit a significant reduction in the ability of the cerebral vessels to dilate in response to vasoactive challenges relative to healthy controls (HC). METHODS The current study uses a vasodilatory challenge while participants undergo functional magnetic resonance imaging to quantify the amplitude and delay of cerebrovascular reactivity in participants with PD relative to age and sex-matched HC. An analysis of covariance was used to evaluate differences in cerebrovascular reactivity amplitude and latency between PD participants and HC. RESULTS A significant main effect of group was observed for whole-brain cerebrovascular reactivity amplitude (F(1, 28) = 4.38, p = 0.046, Hedge's g = 0.73) and latency (F(1, 28) = 16.35, p < 0.001, Hedge's g = 1.42). Participants with PD exhibited reduced whole-brain amplitude and increased latencies in cerebrovascular reactivity relative to HC. The evaluation of regional effects indicates that the largest effects were observed in the cuneus, precuneus, and parietal regions. CONCLUSIONS PD participants exhibited reduced and delayed cerebrovascular reactivity. This dysfunction may play an important role in chronic hypoxia, neuroinflammation, and protein aggregation, mechanisms that could lead to disease progression. Cerebrovascular reactivity may serve as an important biomarker and target for future interventions. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sephira G Ryman
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Nicholas Shaff
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Andrew Dodd
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Stephanie Nitschke
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Christopher Wertz
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Kayla Julio
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Gerson Suarez Cedeno
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Amanda Deligtisch
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Erik Erhardt
- Department of Mathematics and Statistics, University of New Mexico, Albuquerque, New Mexico, USA
| | - Henry Lin
- Department of Medicine, University of New Mexico, Albuquerque, New Mexico, USA
- Neurology|Medicine, New Mexico VA Health Care System, Albuquerque, New Mexico, USA
| | - Andrei Vakhtin
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Kathleen L Poston
- Movement Disorders Division in the Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, USA
| | - Rawan Tarawneh
- Memory and Aging Center, Department of Neurology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Sarah Pirio Richardson
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, New Mexico, USA
- Neurology|Medicine, New Mexico VA Health Care System, Albuquerque, New Mexico, USA
| | - Andrew Mayer
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| |
Collapse
|
8
|
Weintraub D, Picillo M, Cho HR, Caspell‐Garcia C, Blauwendraat C, Brown EG, Chahine LM, Coffey CS, Dobkin RD, Foroud T, Galasko D, Kieburtz K, Marek K, Merchant K, Mollenhauer B, Poston KL, Simuni T, Siderowf A, Singleton A, Seibyl J, Tanner CM. Impact of the Dopamine System on Long-Term Cognitive Impairment in Parkinson Disease: An Exploratory Study. Mov Disord Clin Pract 2023; 10:943-955. [PMID: 37332638 PMCID: PMC10272925 DOI: 10.1002/mdc3.13751] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/09/2023] [Accepted: 04/02/2023] [Indexed: 06/20/2023] Open
Abstract
Background Little is known about the impact of the dopamine system on development of cognitive impairment (CI) in Parkinson disease (PD). Objectives We used data from a multi-site, international, prospective cohort study to explore the impact of dopamine system-related biomarkers on CI in PD. Methods PD participants were assessed annually from disease onset out to 7 years, and CI determined by applying cut-offs to four measures: (1) Montreal Cognitive Assessment; (2) detailed neuropsychological test battery; (3) Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS) cognition score; and (4) site investigator diagnosis of CI (mild cognitive impairment or dementia). The dopamine system was assessed by serial Iodine-123 Ioflupane dopamine transporter (DAT) imaging, genotyping, and levodopa equivalent daily dose (LEDD) recorded at each assessment. Multivariate longitudinal analyses, with adjustment for multiple comparisons, determined the association between dopamine system-related biomarkers and CI, including persistent impairment. Results Demographic and clinical variables associated with CI were higher age, male sex, lower education, non-White race, higher depression and anxiety scores and higher MDS-UPDRS motor score. For the dopamine system, lower baseline mean striatum dopamine transporter values (P range 0.003-0.005) and higher LEDD over time (P range <0.001-0.01) were significantly associated with increased risk for CI. Conclusions Our results provide preliminary evidence that alterations in the dopamine system predict development of clinically-relevant, cognitive impairment in Parkinson's disease. If replicated and determined to be causative, they demonstrate that the dopamine system is instrumental to cognitive health status throughout the disease course. TRIAL REGISTRATION Parkinson's Progression Markers Initiative is registered with ClinicalTrials.gov (NCT01141023).
Collapse
Affiliation(s)
- Daniel Weintraub
- Department of PsychiatryPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Marina Picillo
- Assistant Professor in Neurology at the Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”University of SalernoItaly
| | - Hyunkeun Ryan Cho
- Department of Biostatistics, College of Public HealthUniversity of IowaIowa CityIowaUSA
| | | | - Cornelis Blauwendraat
- Center for Alzheimer's and Related Dementias, and the Integrative Neurogenomics Unit, Laboratory of NeurogeneticsNational Institute on Aging, National Institutes of HealthBethesdaMarylandUSA
| | - Ethan G. Brown
- Department of NeurologyWeill Institute for Neurosciences, University of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Lana M. Chahine
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Christopher S. Coffey
- Department of Biostatistics, College of Public HealthUniversity of IowaIowa CityIowaUSA
| | - Roseanne D. Dobkin
- Department of PsychiatryRutgers University, Robert Wood Johnson Medical SchoolPiscatawayNew JerseyUSA
| | - Tatiana Foroud
- Department of Medical and Molecular GeneticsIndiana UniversityIndianapolisIndianaUSA
| | - Doug Galasko
- Department of NeurologyUniversity of CaliforniaSan DiegoCaliforniaUSA
| | - Karl Kieburtz
- Department of NeurologyUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - Kenneth Marek
- Institute for Neurodegenerative DisordersNew HavenConnecticutUSA
| | - Kalpana Merchant
- Department of NeurologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Brit Mollenhauer
- Department of NeurologyUniversity Medical Center GoettingenGoettingenGermany
| | - Kathleen L. Poston
- Department of Neurology and Neurological SciencesStanford UniversityStanfordCaliforniaUSA
| | - Tanya Simuni
- Department of NeurologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Andrew Siderowf
- Department of NeurologyPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Andrew Singleton
- Center for Alzheimer's and Related Dementias, and the Molecular Genetics SectionLaboratory of Neurogenetics, National Institute on Aging, National Institutes of HealthBethesdaMarylandUSA
| | - John Seibyl
- Institute for Neurodegenerative DisordersNew HavenConnecticutUSA
| | - Caroline M. Tanner
- Department of NeurologyWeill Institute for Neurosciences, University of California, San FranciscoSan FranciscoCaliforniaUSA
| | | |
Collapse
|
9
|
Endothelial LRP1-ICD Accelerates Cognition-Associated Alpha-Synuclein Pathology and Neurodegeneration through PARP1 Activation in a Mouse Model of Parkinson's Disease. Mol Neurobiol 2023; 60:979-1003. [PMID: 36394710 DOI: 10.1007/s12035-022-03119-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 11/03/2022] [Indexed: 11/19/2022]
Abstract
Parkinson's disease (PD) is characterized by progressive loss of dopaminergic neurons and accumulation of misfolded alpha-synuclein (αSyn) into Lewy bodies. In addition to motor impairment, PD commonly presents with cognitive impairment, a non-motor symptom with poor outcome. Cortical αSyn pathology correlates closely with vascular risk factors and vascular degeneration in cognitive impairment. However, how the brain microvasculature regulates αSyn pathology and neurodegeneration remains unclear. Here, we constructed a rapidly progressive PD model by injecting alpha-synuclein preformed fibrils (αSyn PFFs) into the cerebral cortex and striatum. Brain capillaries in mice with cognitive impairment showed a reduction in diameter and length after 6 months, along with string vessel formation. The intracellular domain of low-density lipoprotein receptor-related protein-1 (LRP1-ICD) was upregulated in brain microvascular endothelium. LRP1-ICD promoted αSyn PFF uptake and exacerbated endothelial damage and neuronal apoptosis. Then, we overexpressed LRP1-ICD in brain capillaries using an adeno-associated virus carrying an endothelial-specific promoter. Endothelial LRP1-ICD worsened αSyn PFF-induced vascular damage, αSyn pathology, or neuron death in the cortex and hippocampus, resulting in severe motor and cognitive impairment. LRP1-ICD increased the synthesis of poly(adenosine 5'-diphosphate-ribose) (PAR) in the presence of αSyn PFFs. Inhibition of PAR polymerase 1 (PARP1) prevented vascular-derived injury, as did loss of PARP1 in the endothelium, which was further implicated in endothelial cell proliferation and inflammation. Together, we demonstrate a novel vascular mechanism of cognitive impairment in PD. These findings support a role for endothelial LRP1-ICD/PARP1 in αSyn pathology and neurodegeneration, and provide evidence for vascular protection strategies in PD therapy.
Collapse
|
10
|
Oosterwegel MJ, Krijthe JH, den Brok MGHE, van den Heuvel L, Richard E, Heskes T, Bloem BR, Evers LJW. The effect of cardiovascular risk on disease progression in de novo Parkinson's disease patients: An observational analysis. Front Neurol 2023; 14:1138546. [PMID: 37122316 PMCID: PMC10130532 DOI: 10.3389/fneur.2023.1138546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/20/2023] [Indexed: 05/02/2023] Open
Abstract
Background Currently available treatment options for Parkinson's disease are symptomatic and do not alter the course of the disease. Recent studies have raised the possibility that cardiovascular risk management may slow the progression of the disease. Objectives We estimated the effect of baseline cardiovascular risk factors on the progression of Parkinson's disease, using measures for PD-specific motor signs and cognitive functions. Methods We used data from 424 de novo Parkinson's disease patients and 199 age-matched controls from the observational, multicenter Parkinson's Progression Markers Initiative (PPMI) study, which included follow-up of up to 9 years. The primary outcome was the severity of PD-specific motor signs, assessed with the MDS-UPDRS part III in the "OFF"-state. The secondary outcome was cognitive function, measured with the Montreal Cognitive Assessment, Symbol Digit Modalities Test, and Letter-Number Sequencing task. Exposures of interest were diabetes mellitus, hypertension, body mass index, cardiovascular event history and hypercholesterolemia, and a modified Framingham risk score, measured at baseline. The effect of each of these exposures on disease progression was modeled using linear mixed models, including adjustment for identified confounders. A secondary analysis on the Tracking Parkinson's cohort including 1,841 patients was performed to validate our findings in an independent patient cohort. Results Mean age was 61.4 years, and the average follow-up was 5.5 years. We found no statistically significant effect of any individual cardiovascular risk factor on the MDS-UPDRS part III progression (all 95% confidence intervals (CIs) included zero), with one exception: in the PD group, the estimated effect of a one-point increase in body mass index was 0.059 points on the MDS-UPDRS part III per year (95% CI: 0.017 to 0.102). We found no evidence for an effect of any of the exposures on the rate of change in cognitive functioning in the PD group. Similar results were observed for the Tracking Parkinson's cohort (all 95% CIs overlapped with PPMI), but the 95% CI of the effect of body mass index on the MDS-UPDRS part III progression included zero. Conclusions Based on this analysis of two large cohorts of de novo PD patients, we found no evidence to support clinically relevant effects of cardiovascular risk factors on the clinical progression of Parkinson's disease.
Collapse
Affiliation(s)
- Max J. Oosterwegel
- Center of Expertise for Parkinson and Movement Disorders, Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Data Science, Institute for Computing and Information Sciences, Radboud University, Nijmegen, Netherlands
- *Correspondence: Max J. Oosterwegel
| | - Jesse H. Krijthe
- Department of Intelligent Systems, Delft University of Technology, Delft, Netherlands
| | - Melina G. H. E. den Brok
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Neurology, Amsterdam University Medical Center, Location AMC, Amsterdam, Netherlands
| | - Lieneke van den Heuvel
- Center of Expertise for Parkinson and Movement Disorders, Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
| | - Edo Richard
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Neurology, Amsterdam University Medical Center, Location AMC, Amsterdam, Netherlands
| | - Tom Heskes
- Department of Data Science, Institute for Computing and Information Sciences, Radboud University, Nijmegen, Netherlands
| | - Bastiaan R. Bloem
- Center of Expertise for Parkinson and Movement Disorders, Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
| | - Luc J. W. Evers
- Center of Expertise for Parkinson and Movement Disorders, Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Data Science, Institute for Computing and Information Sciences, Radboud University, Nijmegen, Netherlands
| |
Collapse
|
11
|
Sinani O, Dadouli K, Ntellas P, Kapsalaki EZ, Vlychou M, Raptis DG, Marogianni C, Markou K, Dardiotis E, Xiromerisiou G. Association between white matter lesions and Parkinson's disease: an impact on Postural/Gait difficulty phenotype and cognitive performance. Neurol Res 2022; 44:1122-1131. [PMID: 35994524 DOI: 10.1080/01616412.2022.2112378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND White matter hyperintensities (WMHs) may be observed on Magnetic Resonance Imaging (MRI) in patients with Parkinson disease with or without vascular risk factors. Whether WMHs may influence motor and non-motor aspects of Parkinson disease is a subject of debate. The aim of this study is to evaluate the impact of WMH severity on various aspects of Parkinson disease in combination to the estimation of the impact of cerebrovascular risk factors. MATERIALS AND METHODS We included a cohort of patients with Parkinson's disease who underwent MRI examination. The Fazekas visual rating scale was used to assess the severity and location of WMHs, and patient clinical characteristics were correlated with MRI data. RESULTS All vascular risk factors were associated with higher Fazekas score in both periventricular and deep white matter. Periventricular white matter hyperintensities (PWMHs) and deep white matter hyperintensities (DWMHs) were associated with lower scores in the ACE-R cognitive assessment scale (p < 0.001). Furthermore, PWMHs and DWMHs severity was associated with higher UPDRS motor score (p < 0.001), while the Postural Instability Gait Difficulty (PIGD) phenotype was correlated with higher burden of WMHs. CONCLUSIONS Comorbid WMHs may contribute to multi-dimension dysfunction in patients with Parkinson disease and consequently the management of vascular risk factors may be crucial to maintain motor and non-motor functions in PD.
Collapse
Affiliation(s)
- Olga Sinani
- Faculty of Medicine, University of Thessaly, Larisa, Greece
| | - Katerina Dadouli
- Laboratory of Hygiene and Epidemiology, Faculty of Medicine, University of Thessaly, Larisa, Greece
| | - Panagiotis Ntellas
- Department of Medical Oncology, University Hospital of Ioannina, Ioannina, Greece
| | - Eftychia Z Kapsalaki
- Department of Diagnostic Radiology, Faculty of Medicine, University of Thessaly, Larisa, Greece
| | - Marianna Vlychou
- Department of Diagnostic Radiology, Faculty of Medicine, University of Thessaly, Larisa, Greece
| | - Dimitrios G Raptis
- Respiratory Medicine Department, Faculty of Medicine, University of Thessaly, Larisa, Greece
| | | | - Katerina Markou
- Department of Neurology, University Hospital of Larisa, Larisa, Greece
| | | | - Georgia Xiromerisiou
- Department of Neurology, University Hospital of Larisa, Larisa, Greece.,Faculty of Medicine, School of Health Sciences, University of Thessaly, Volos, Greece
| |
Collapse
|
12
|
Chronic Cerebral Hypoperfusion Aggravates Parkinson’s Disease Dementia-Like Symptoms and Pathology in 6-OHDA-Lesioned Rat through Interfering with Sphingolipid Metabolism. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5392966. [PMID: 35979400 PMCID: PMC9377946 DOI: 10.1155/2022/5392966] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/20/2022] [Indexed: 11/17/2022]
Abstract
Chronic cerebral hypoperfusion (CCH) is a cardinal risk factor for Parkinson's disease dementia (PDD), but this potential causality lacks mechanistic evidence. We selected bilateral common carotid artery occlusion (BCCAO) to simulate chronic cerebral hypoperfusion in the rat model of PD induced by typical neurotoxin 6-hydroxy dopamine (6-OHDA). Four weeks after unilateral injection of 6-OHDA into the medial forebrain bundle, rats underwent BCCAO. Male Sprague-Dawley rats were divided into five groups of ten, including sham, PD+BCCAO 2 weeks, PD+BCCAO 1 week, PD, and BCCAO 2 weeks. Then, open field test (OFT) and Morris water maze test (MWM) were used to assess the PDD-like symptoms in rats. Also, the pathological manifestations and mechanisms of BCCAO impairing cognitive functions have been explored via hematoxylin-eosin staining, Nissl staining, immunohistochemistry, immunofluorescence, RNA sequencing analysis, lipidomics, and quantitative real-time polymerase chain reaction. In this study, we found that CCH could aggravate PDD-like cognitive symptoms (i.e., learning memory and spatial cognition) and PDD-like pathology (higher expression of α-Syn and Aβ in prefrontal cortex and striatum). Moreover, a potential relationship between differentially expressed mRNAs and lipid metabolism was revealed by RNA sequencing analysis. Lipidomics showed that CCH could affect the intensity of 5 lipids, including sphingomyelin (SM 9:0;2O/26:2; SM 8:1;2O/25:0; and SM 8:0;2O/28:4), cardiolipin, lysophosphatidylcholine, cholesteryl ester, and triacylglycerol. Interestingly, the KEGG pathway analysis of both RNA sequencing analysis and lipidomics suggested that CCH leaded to learning impairment by affecting sphingolipid metabolism. Finally, we found that CCH disrupts the sphingolipid metabolism by affecting the mRNA expression of SMPD1 and SMS2, leading to the accumulation of sphingomyelin in the prefrontal cortex. In summary, CCH, an independent exacerbating reason for impairment in learning and memory within the pathopoiesis of PD, aggravates Parkinson's disease dementia-like symptoms and pathology in 6-OHDA-lesioned rat through interfering with sphingolipid metabolism.
Collapse
|
13
|
Meira B, Fernandes M, Salavisa M, Saraiva M, Conceição L, Borbinha C, Ladeira F, Marto JP, Barbosa R, Mendonça M, Bugalho P. Obstructive Sleep Apnea and Other Vascular Risk factors' Impact on Non‐Motor Symptoms in Parkinson's Disease. Mov Disord Clin Pract 2022; 9:785-798. [DOI: 10.1002/mdc3.13504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 04/16/2022] [Accepted: 05/22/2022] [Indexed: 11/10/2022] Open
Affiliation(s)
- Bruna Meira
- Neurology Department, Hospital Egas Moniz Centro Hospitalar de Lisboa Ocidental Lisbon Portugal
| | - Marco Fernandes
- Neurology Department, Hospital Egas Moniz Centro Hospitalar de Lisboa Ocidental Lisbon Portugal
| | - Manuel Salavisa
- Neurology Department, Hospital Egas Moniz Centro Hospitalar de Lisboa Ocidental Lisbon Portugal
| | - Marlene Saraiva
- Neurology Department, Hospital Egas Moniz Centro Hospitalar de Lisboa Ocidental Lisbon Portugal
| | - Laurete Conceição
- Neurology Department, Hospital Egas Moniz Centro Hospitalar de Lisboa Ocidental Lisbon Portugal
| | - Cláudia Borbinha
- Neurology Department, Hospital Egas Moniz Centro Hospitalar de Lisboa Ocidental Lisbon Portugal
| | - Filipa Ladeira
- Neurology Department, Hospital Egas Moniz Centro Hospitalar de Lisboa Ocidental Lisbon Portugal
| | - João Pedro Marto
- Neurology Department, Hospital Egas Moniz Centro Hospitalar de Lisboa Ocidental Lisbon Portugal
- NOVA Medical School/Faculdade de Ciências Médicas Universidade Nova de Lisboa Lisbon Portugal
| | - Raquel Barbosa
- Neurology Department, Hospital Egas Moniz Centro Hospitalar de Lisboa Ocidental Lisbon Portugal
- NOVA Medical School/Faculdade de Ciências Médicas Universidade Nova de Lisboa Lisbon Portugal
| | - Marcelo Mendonça
- Neurology Department, Hospital Egas Moniz Centro Hospitalar de Lisboa Ocidental Lisbon Portugal
- NOVA Medical School/Faculdade de Ciências Médicas Universidade Nova de Lisboa Lisbon Portugal
- Champalimaud Research and Clinical Centre Champalimaud Foundation Lisbon Portugal
| | - Paulo Bugalho
- Neurology Department, Hospital Egas Moniz Centro Hospitalar de Lisboa Ocidental Lisbon Portugal
- NOVA Medical School/Faculdade de Ciências Médicas Universidade Nova de Lisboa Lisbon Portugal
| |
Collapse
|
14
|
Morphological basis of Parkinson disease-associated cognitive impairment: an update. J Neural Transm (Vienna) 2022; 129:977-999. [PMID: 35726096 DOI: 10.1007/s00702-022-02522-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022]
Abstract
Cognitive impairment is one of the most salient non-motor symptoms of Parkinson disease (PD) that poses a significant burden on the patients and carers as well as being a risk factor for early mortality. People with PD show a wide spectrum of cognitive dysfunctions ranging from subjective cognitive decline and mild cognitive impairment (MCI) to frank dementia. The mean frequency of PD with MCI (PD-MCI) is 25.8% and the pooled dementia frequency is 26.3% increasing up to 83% 20 years after diagnosis. A better understanding of the underlying pathological processes will aid in directing disease-specific treatment. Modern neuroimaging studies revealed considerable changes in gray and white matter in PD patients with cognitive impairment, cortical atrophy, hypometabolism, dopamine/cholinergic or other neurotransmitter dysfunction and increased amyloid burden, but multiple mechanism are likely involved. Combined analysis of imaging and fluid markers is the most promising method for identifying PD-MCI and Parkinson disease dementia (PDD). Morphological substrates are a combination of Lewy- and Alzheimer-associated and other concomitant pathologies with aggregation of α-synuclein, amyloid, tau and other pathological proteins in cortical and subcortical regions causing destruction of essential neuronal networks. Significant pathological heterogeneity within PD-MCI reflects deficits in various cognitive domains. This review highlights the essential neuroimaging data and neuropathological changes in PD with cognitive impairment, the amount and topographical distribution of pathological protein aggregates and their pathophysiological relevance. Large-scale clinicopathological correlative studies are warranted to further elucidate the exact neuropathological correlates of cognitive impairment in PD and related synucleinopathies as a basis for early diagnosis and future disease-modifying therapies.
Collapse
|
15
|
Neuropathological substrates of cognition in Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2022; 269:177-193. [PMID: 35248194 DOI: 10.1016/bs.pbr.2022.01.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Autopsy validation is still required for a definitive diagnosis of Parkinson's disease (Postuma et al., 2015), where the presence of Lewy bodies and Lewy neurites, composed primarily of alpha-synuclein, are observed in stereotyped patterns throughout regions of the brainstem, limbic, and neocortical regions of the brain (Braak et al., 2003). In spite of these relatively reliable observed patterns of alpha-synuclein pathology, there is a large degree of heterogeneity in the timing and features of neuropsychiatric and cognitive dysfunction in Parkinson's disease (Fereshtehnejad et al., 2015; Selikhova et al., 2009; Williams-Gray et al., 2013). Detailed studies of their neuropathological substrates of cognitive dysfunction and their associations with a variety of in vivo biomarkers have begun to disentangle this complex relationship, but ongoing multicentered, longitudinal studies of well-characterized and autopsy validated cases are still required.
Collapse
|
16
|
Hou Y, Shang H. Magnetic Resonance Imaging Markers for Cognitive Impairment in Parkinson’s Disease: Current View. Front Aging Neurosci 2022; 14:788846. [PMID: 35145396 PMCID: PMC8821910 DOI: 10.3389/fnagi.2022.788846] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 01/03/2022] [Indexed: 12/24/2022] Open
Abstract
Cognitive impairment (CI) ranging from mild cognitive impairment (MCI) to dementia is a common and disturbing complication in patients with Parkinson’s disease (PD). Numerous studies have focused on neuropathological mechanisms underlying CI in PD, along with the identification of specific biomarkers for CI. Magnetic resonance imaging (MRI), a promising method, has been adopted to examine the changes in the brain and identify the candidate biomarkers associated with CI. In this review, we have summarized the potential biomarkers for CI in PD which have been identified through multi-modal MRI studies. Structural MRI technology is widely used in biomarker research. Specific patterns of gray matter atrophy are promising predictors of the evolution of CI in patients with PD. Moreover, other MRI techniques, such as MRI related to small-vessel disease, neuromelanin-sensitive MRI, quantitative susceptibility mapping, MR diffusion imaging, MRI related to cerebrovascular abnormality, resting-state functional MRI, and proton magnetic resonance spectroscopy, can provide imaging features with a good degree of prediction for CI. In the future, novel combined biomarkers should be developed using the recognized analysis tools and predictive algorithms in both cross-sectional and longitudinal studies.
Collapse
|
17
|
Metabolic Syndrome, Cognitive Impairment and the Role of Diet: A Narrative Review. Nutrients 2022; 14:nu14020333. [PMID: 35057514 PMCID: PMC8780484 DOI: 10.3390/nu14020333] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/04/2022] [Accepted: 01/10/2022] [Indexed: 01/27/2023] Open
Abstract
Background: This narrative review presents the association between metabolic syndrome (MetS), along with its components, and cognition-related disorders, as well as the potential reversal role of diet against cognitive impairment by modulating MetS. Methods: An electronic research in Medline (Pubmed) and Scopus was conducted. Results: MetS and cognitive decline share common cardiometabolic pathways as MetS components can trigger cognitive impairment. On the other side, the risk factors for both MetS and cognitive impairment can be reduced by optimizing the nutritional intake. Clinical manifestations such as dyslipidemia, hypertension, diabetes and increased central body adiposity are nutrition-related risk factors present during the prodromal period before cognitive impairment. The Mediterranean dietary pattern stands among the most discussed predominantly plant-based diets in relation to cardiometabolic disorders that may prevent dementia, Alzheimer’s disease and other cognition-related disorders. In addition, accumulating evidence suggests that the consumption of specific dietary food groups as a part of the overall diet can improve cognitive outcomes, maybe due to their involvement in cardiometabolic paths. Conclusions: Early MetS detection may be helpful to prevent or delay cognitive decline. Moreover, this review highlights the importance of healthy nutritional habits to reverse such conditions and the urgency of early lifestyle interventions.
Collapse
|
18
|
Increased homocysteine levels correlate with cortical structural damage in Parkinson's disease. J Neurol Sci 2022; 434:120148. [DOI: 10.1016/j.jns.2022.120148] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/07/2021] [Accepted: 01/05/2022] [Indexed: 12/12/2022]
|
19
|
Picillo M, LaFontant DE, Bressman S, Caspell-Garcia C, Coffey C, Cho HR, Burghardt EL, Dahodwala N, Saunders-Pullman R, Tanner CM, Amara AW. Sex-Related Longitudinal Change of Motor, Non-Motor, and Biological Features in Early Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:421-436. [PMID: 34744052 PMCID: PMC8842783 DOI: 10.3233/jpd-212892] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/03/2021] [Indexed: 01/26/2023]
Abstract
BACKGROUND Investigation of sex-related motor and non-motor differences and biological markers in Parkinson's disease (PD) may improve precision medicine approach. OBJECTIVE To examine sex-related longitudinal changes in motor and non-motor features and biologic biomarkers in early PD. METHODS We compared 5-year longitudinal changes in de novo, untreated PD men and women (at baseline N = 423; 65.5%male) of the Parkinson's Progression Markers Initiative (PPMI), assessing motor and non-motor manifestations of disease; and biologic measures in cerebrospinal fluid (CSF) and dopamine transporter deficit on DaTscanTM uptake. RESULTS Men experienced greater longitudinal decline in self-reported motor (p < 0.001) and non-motor (p = 0.009) aspects of experiences of daily living, such that men had a yearly increase in MDS-UPDRS part II by a multiplicative factor of 1.27 compared to women at 0.7, while men had a yearly increase in MDS-UPDRS part I by a multiplicative factor of 0.98, compared to women at 0.67. Compared to women, men had more longitudinal progression in clinician-assessed motor features in the ON medication state (p = 0.010) and required higher dopaminergic medication dosages over time (p = 0.014). Time to reach specific disease milestones and longitudinal changes in CSF biomarkers and DaTscanTM uptake were not different by sex. CONCLUSION Men showed higher self-assessed motor and non-motor burden of disease, with possible contributions from suboptimal dopaminergic therapeutic response in men. However, motor features of disease evaluated with clinician-based scales in the OFF medication state, as well as biological biomarkers do not show specific sex-related progression patterns.
Collapse
Affiliation(s)
- Marina Picillo
- Center for Neurodegenerative Diseases (CEMAND), Department of Medicine, Surgery and Dentistry, Neuroscience Section, University of Salerno, Italy
| | | | - Susan Bressman
- Department of Neurology, Icahn School of Medicine at Mount Sinai and Mount Sinai Beth Israel, New York, NY, USA
| | | | | | - Hyunkeun Ryan Cho
- Department of Biostatistics, The University of Iowa, Iowa City, IA, USA
| | | | - Nabila Dahodwala
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel Saunders-Pullman
- Department of Neurology, Icahn School of Medicine at Mount Sinai and Mount Sinai Beth Israel, New York, NY, USA
| | - Caroline M. Tanner
- Weill Institute for Neuroscience, Department of Neurology, University of California–San Francisco, & Parkinson’s Disease Research Education and Clinical Center, San Francisco Veterans Affairs Health Care System, San Francisco, CA, USA
| | - Amy W. Amara
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | |
Collapse
|
20
|
Ge YL, Gong SY, Wang PZ, Yan JH, Li W, Zhang JR, Jin H, Zhuang S, Hu L, Ding CW, Yang YP, Wang F, Li D, Chen J, Mao CJ, Zhang YC, Li K, Liu CF. Cognitive Performance is Associated with Altered Cerebral Hemodynamics Assessed by Transcranial Ultrasound in Parkinson's Disease. Neuropsychiatr Dis Treat 2022; 18:1421-1431. [PMID: 35855751 PMCID: PMC9288215 DOI: 10.2147/ndt.s358150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/18/2022] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Cognitive impairment (CI) is a common but debilitating non-motor symptom in Parkinson's disease (PD). Although cerebrovascular functions are related to cognitive performance in healthy individuals, such a relation in PD remains elusive. This study aims to assess the association between cerebrovascular function and cognitive performance in PD individuals. PATIENTS AND METHODS Two-hundred-and-one PD individuals were retrospectively included. They were subsequently divided into two groups: PD with normal cognition (PD-NC) and PD with CI (PD-CI). Cerebral hemodynamic characteristics of the middle cerebral arteries were assessed by transcranial ultrasound. The association between scores in each cognitive domain and cerebral hemodynamic parameters was further analyzed using regression analyses. Additionally, a binary logistic regression model with backward stepwise procedure was applied to build the model for discriminating CI in PD individuals. An independent dataset of additional 46 PD individuals was used further. RESULTS The PD-CI group showed a relatively lower end-diastolic blood flow velocity (EDV, p < 0.05) and a higher resistive index (RI, p < 0.05) compared to the PD-NC group. RI showed significant associations with the memory item score of Montreal Cognitive Assessment (p < 0.05). A model combining clinical and hemodynamic variables was established with optimal efficiency (area under the curve, AUC = 0.651). Further replication of the model in an independent dataset yielded a great consistency (AUC = 0.704). CONCLUSION In our study, cerebrovascular functions were significantly associated with the cognitive performance in PD individuals, especially with the memory task. The established model was effective in identifying CI in PD individuals, which might be a potentially useful tool to screen the cognitive decline in PD individuals at an early stage of the disease. Further studies with larger sample sizes in different populations are warranted.
Collapse
Affiliation(s)
- Yi-Lun Ge
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Si-Yi Gong
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Pu-Zhi Wang
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Jia-Hui Yan
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Wen Li
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Jin-Ru Zhang
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Hong Jin
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Sheng Zhuang
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Lei Hu
- Department of Ultrasound, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Chang-Wei Ding
- Department of Ultrasound, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Ya-Ping Yang
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Fen Wang
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Dan Li
- Department of Neurology, Suqian First Hospital, Suqian, People's Republic of China
| | - Jing Chen
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Cheng-Jie Mao
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China.,Department of Neurology, Suqian First Hospital, Suqian, People's Republic of China
| | - Ying-Chun Zhang
- Department of Ultrasound, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Kai Li
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Chun-Feng Liu
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China.,Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, People's Republic of China.,Department of Neurology, Suqian First Hospital, Suqian, People's Republic of China
| |
Collapse
|
21
|
Scamarcia PG, Agosta F, Spinelli EG, Basaia S, Stojković T, Stankovic I, Sarasso E, Canu E, Markovic V, Petrović I, Stefanova E, Pagani E, Kostic VS, Filippi M. Longitudinal White Matter Damage Evolution in Parkinson's Disease. Mov Disord 2021; 37:315-324. [PMID: 34806799 DOI: 10.1002/mds.28864] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND White matter hyperintensities (WMHs) have a role in cognitive impairment in normal brain aging, while the effect on Parkinson's disease (PD) progression is still controversial. OBJECTIVE To investigate the longitudinal evolution of micro- and macrostructural damage of cerebral white matter (WM) and its relationship with the clinical picture in PD. METHODS A total of 154 PD patients underwent clinical, cognitive, and magnetic resonance imaging (MRI) assessment once a year for up to 4 years. Sixty healthy controls underwent the same protocol at baseline. WMHs were identified and total WMH volume was measured. WMHs were also used as exclusion masks to define normal-appearing white matter (NAWM). Using tract-based spatial statistics, diffusion tensor (DT) MRI metrics of whole-brain WM and NAWM were obtained. Linear mixed-effects models defined the longitudinal evolution and association between variables. WM alterations were tested as risk factors of disease progression using linear regression and Cox proportional hazards models. RESULTS At baseline, PD patients showed alterations of all DT MRI measures compared to controls. Longitudinally, DT MRI measures did not vary significantly and no association with clinical variables was found. WMH volume changed over time and was associated with impairment in global cognition, executive functions, and language. Baseline WMH volume was a moderate risk factor for progression to mild cognitive impairment. CONCLUSIONS Our study suggests an association between WMHs and cognitive deterioration in PD, whereas WM microstructural damage is a negligible contributor to clinical deterioration. WMHs assessed by MRI can provide an important tool for monitoring the development of cognitive impairment in PD patients. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Pietro Giuseppe Scamarcia
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federica Agosta
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Edoardo Gioele Spinelli
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Silvia Basaia
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Tanja Stojković
- Clinic of Neurology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Iva Stankovic
- Clinic of Neurology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Elisabetta Sarasso
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Canu
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vladana Markovic
- Clinic of Neurology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Igor Petrović
- Clinic of Neurology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Elka Stefanova
- Clinic of Neurology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Elisabetta Pagani
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vladimir S Kostic
- Clinic of Neurology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy.,Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
22
|
Narasimhan M, Schwartz R, Halliday G. Parkinsonism and cerebrovascular disease. J Neurol Sci 2021; 433:120011. [PMID: 34686356 DOI: 10.1016/j.jns.2021.120011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/01/2021] [Accepted: 09/29/2021] [Indexed: 11/27/2022]
Abstract
The relationship between cerebrovascular disease and parkinsonism is commonly seen in everyday clinical practice but remains ill-defined and under-recognised with little guidance for the practising neurologist. We attempt to define this association and to illustrate key clinical, radiological and pathological features of the syndrome of Vascular Parkinsonism (VaP). VaP is a major cause of morbidity in the elderly associated with falls, hip fractures and cognitive impairment. Although acute parkinsonism is reported in the context of an acute cerebrovascular event, the vast majority of VaP presents as an insidious syndrome usually in the context of vascular risk factors and radiological evidence of small vessel disease. There may be an anatomic impact on basal ganglia neuronal networks, however the effect of small vessel disease (SVD) on these pathways is not clear. There are now established reporting standards for radiological features of SVD on MRI. White matter hyperintensities and lacunes have been thought to be the representative radiological features of SVD but other features such as the perivascular space are gaining more importance, especially in context of the glymphatic system. It is important to consider VaP in the differential diagnosis of Parkinson disease (PD) and in these situations, neuroimaging may offer diagnostic benefit especially in those patients with atypical presentations or refractoriness to levodopa. Proactive management of vascular risk factors, monitoring of bone density and an exercise program may offer easily attainable therapeutic targets in PD and VaP. Levodopa therapy should be considered in patients with VaP, however the dose and effect may be different from use in PD. This article is part of the Special Issue "Parkinsonism across the spectrum of movement disorders and beyond" edited by Joseph Jankovic, Daniel D. Truong and Matteo Bologna.
Collapse
Affiliation(s)
- Manisha Narasimhan
- Brain and Mind Centre and Faculty of Health and Medical Sciences, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia.
| | - Raymond Schwartz
- Brain and Mind Centre and Faculty of Health and Medical Sciences, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Glenda Halliday
- Brain and Mind Centre and Faculty of Health and Medical Sciences, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
23
|
Cui W, Fu W, Lin Y, Zhang T. Application of Nanomaterials in Neurodegenerative Diseases. Curr Stem Cell Res Ther 2021; 16:83-94. [PMID: 32213159 DOI: 10.2174/1574888x15666200326093410] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/07/2020] [Accepted: 02/04/2020] [Indexed: 02/08/2023]
Abstract
Neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Huntington's disease are very harmful brain lesions. Due to the difficulty in obtaining therapeutic drugs, the best treatment for neurodegenerative diseases is often not available. In addition, the bloodbrain barrier can effectively prevent the transfer of cells, particles and macromolecules (such as drugs) in the brain, resulting in the failure of the traditional drug delivery system to provide adequate cellular structure repair and connection modes, which are crucial for the functional recovery of neurodegenerative diseases. Nanomaterials are designed to carry drugs across the blood-brain barrier for targets. Nanotechnology uses engineering materials or equipment to interact with biological systems at the molecular level to induce physiological responses through stimulation, response and target site interactions, while minimizing the side effects, thus revolutionizing the treatment and diagnosis of neurodegenerative diseases. Some magnetic nanomaterials play a role as imaging agents or nanoprobes for Magnetic Resonance Imaging to assist in the diagnosis of neurodegenerative diseases. Although the current research on nanomaterials is not as useful as expected in clinical applications, it achieves a major breakthrough and guides the future development direction of nanotechnology in the application of neurodegenerative diseases. This review briefly discusses the application and advantages of nanomaterials in neurodegenerative diseases. Data for this review were identified by searches of PubMed, and references from relevant articles published in English between 2015 and 2019 using the search terms "nanomaterials", "neurodegenerative diseases" and "blood-brain barrier".
Collapse
Affiliation(s)
- Weitong Cui
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wei Fu
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tianxu Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Liu H, Deng B, Xie F, Yang X, Xie Z, Chen Y, Yang Z, Huang X, Zhu S, Wang Q. The influence of white matter hyperintensity on cognitive impairment in Parkinson's disease. Ann Clin Transl Neurol 2021; 8:1917-1934. [PMID: 34310081 PMCID: PMC8419402 DOI: 10.1002/acn3.51429] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/05/2021] [Accepted: 07/02/2021] [Indexed: 01/11/2023] Open
Abstract
The aim of this meta‐analysis was to review systematically and to identify the relationship between the severity and location of white matter hyperintensities (WMHs) and the degree of cognitive decline in patients with Parkinson’s disease (PD). We searched the PubMed, EMBASE, Web of Science, Ovid, and Cochrane Library databases for clinical trials of the severity and location of WMHs on the degree of cognitive impairment in PD through October 2020. We conducted the survey to compare the association of WMH burden in patients with PD with mild cognitive impairment (PD‐MCI) versus those with normal cognition (PD‐NC) and in patients with PD with dementia (PDD) versus those with PD without dementia (PD‐ND). Nine studies with PD‐MCI versus PD‐NC and 10 studies with PDD versus PD‐ND comparisons were included. The WMH burden in PD‐MCI patients was significantly different compared to that in PD‐NC patients (standard mean difference, SMD = 0.39, 95% CI: 0.12 to 0.66, p = 0.005), while there was no correlation shown in the age‐matched subgroup of the comparison. In addition, PDD patients had a significantly higher burden of WMHs (SMD = 0.8, 95% CI: 0.44 to 1.71, p < 0.0001), especially deep white matter hyperintensities (SMD = 0.54, 95% CI: 0.36 to 0.73, p < 0.00001) and periventricular hyperintensities (SMD = 0.70, 95% CI: 0.36 to 1.04, p < 0.0001), than PD‐NC patients, regardless of the adjustment of age. WMHs might be imaging markers for cognitive impairment in PDD but not in PD‐MCI, regardless of age, vascular risk factors, or race. Further prospective studies are needed to validate the conclusions.
Collapse
Affiliation(s)
- Hailing Liu
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, P.R. China.,Department of Neurology, Maoming People's Hospital, Maoming, Guangdong, China
| | - Bin Deng
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, P.R. China
| | - Fen Xie
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, P.R. China
| | - Xiaohua Yang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, P.R. China
| | - Zhenchao Xie
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, P.R. China
| | - Yonghua Chen
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, P.R. China
| | - Zhi Yang
- Department of Neurology, Maoming People's Hospital, Maoming, Guangdong, China
| | - Xiyan Huang
- Department of Rehabilitation, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, P.R. China
| | - Shuzhen Zhu
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, P.R. China
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, P.R. China
| |
Collapse
|
25
|
Aarsland D, Batzu L, Halliday GM, Geurtsen GJ, Ballard C, Ray Chaudhuri K, Weintraub D. Parkinson disease-associated cognitive impairment. Nat Rev Dis Primers 2021; 7:47. [PMID: 34210995 DOI: 10.1038/s41572-021-00280-3] [Citation(s) in RCA: 438] [Impact Index Per Article: 146.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/27/2021] [Indexed: 02/08/2023]
Abstract
Parkinson disease (PD) is the second most common neurodegenerative disorder, affecting >1% of the population ≥65 years of age and with a prevalence set to double by 2030. In addition to the defining motor symptoms of PD, multiple non-motor symptoms occur; among them, cognitive impairment is common and can potentially occur at any disease stage. Cognitive decline is usually slow and insidious, but rapid in some cases. Recently, the focus has been on the early cognitive changes, where executive and visuospatial impairments are typical and can be accompanied by memory impairment, increasing the risk for early progression to dementia. Other risk factors for early progression to dementia include visual hallucinations, older age and biomarker changes such as cortical atrophy, as well as Alzheimer-type changes on functional imaging and in cerebrospinal fluid, and slowing and frequency variation on EEG. However, the mechanisms underlying cognitive decline in PD remain largely unclear. Cortical involvement of Lewy body and Alzheimer-type pathologies are key features, but multiple mechanisms are likely involved. Cholinesterase inhibition is the only high-level evidence-based treatment available, but other pharmacological and non-pharmacological strategies are being tested. Challenges include the identification of disease-modifying therapies as well as finding biomarkers to better predict cognitive decline and identify patients at high risk for early and rapid cognitive impairment.
Collapse
Affiliation(s)
- Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK. .,Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway.
| | - Lucia Batzu
- Parkinson's Foundation Centre of Excellence, King's College Hospital and Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Glenda M Halliday
- Brain and Mind Centre and Faculty of Medicine and Health School of Medical Sciences, University of Sydney, Sydney, New South Wales, Australia
| | - Gert J Geurtsen
- Amsterdam UMC, University of Amsterdam, Department of Medical Psychology, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | | | - K Ray Chaudhuri
- Parkinson's Foundation Centre of Excellence, King's College Hospital and Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Daniel Weintraub
- Departments of Psychiatry and Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Parkinson's Disease Research, Education and Clinical Center (PADRECC), Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
| |
Collapse
|
26
|
Gonzalez-Latapi P, Bayram E, Litvan I, Marras C. Cognitive Impairment in Parkinson's Disease: Epidemiology, Clinical Profile, Protective and Risk Factors. Behav Sci (Basel) 2021; 11:bs11050074. [PMID: 34068064 PMCID: PMC8152515 DOI: 10.3390/bs11050074] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 02/07/2023] Open
Abstract
Cognitive impairment is a common non-motor symptom in Parkinson's Disease (PD) and an important source of patient disability and caregiver burden. The timing, profile and rate of cognitive decline varies widely among individuals with PD and can range from normal cognition to mild cognitive impairment (PD-MCI) and dementia (PDD). Beta-amyloid and tau brain accumulation, oxidative stress and neuroinflammation are reported risk factors for cognitive impairment. Traumatic brain injury and pesticide and tobacco exposure have also been described. Genetic risk factors including genes such as COMT, APOE, MAPT and BDNF may also play a role. Less is known about protective factors, although the Mediterranean diet and exercise may fall in this category. Nonetheless, there is conflicting evidence for most of the factors that have been studied. The use of inconsistent criteria and lack of comprehensive assessment in many studies are important methodological issues. Timing of exposure also plays a crucial role, although identification of the correct time window has been historically difficult in PD. Our understanding of the mechanism behind these factors, as well as the interactions between gene and environment as determinants of disease phenotype and the identification of modifiable risk factors will be paramount, as this will allow for potential interventions even in established PD.
Collapse
Affiliation(s)
- Paulina Gonzalez-Latapi
- Edmond J. Safra Program in Parkinson’s Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, University Health Network, Toronto, ON M5T2S8, Canada;
| | - Ece Bayram
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA; (E.B.); (I.L.)
| | - Irene Litvan
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA; (E.B.); (I.L.)
| | - Connie Marras
- Edmond J. Safra Program in Parkinson’s Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, University Health Network, Toronto, ON M5T2S8, Canada;
- Correspondence:
| |
Collapse
|
27
|
Lee HM, Han KD, Suh JD, Cho JH. Uvulopalatopharyngoplasty may decrease the incidence of Parkinson's disease associated with obstructive sleep apnea. Sci Rep 2021; 11:9608. [PMID: 33953319 PMCID: PMC8100168 DOI: 10.1038/s41598-021-89205-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
Abstract
The purpose of this study was to investigate whether the incidence of Parkinson’s disease (PD) is increased among patients with obstructive sleep apnea (OSA) and whether surgical treatment can prevent such an increase. This was a retrospective cohort study. We analysed the claims data from the Korea National Health Insurance Service. A total of 202,726 patients who were newly diagnosed with OSA between 2007 and 2014 were included. The patients were divided into two groups: patients who underwent uvulopalatopharyngoplasty (surgery group, n = 22,742) and those who did not (conservative group, n = 179,984). The control group (n = 1,013,630) was selected by propensity score matching. They were tracked until 31st December 2015. The hazard ratio of PD diagnosis (95% confidence interval) in the OSA group with respect to the control group was calculated using the Cox proportional hazard model. In the conservative group, the incidence of PD (hazard ratio 2.57 [2.32–2.85]) was significantly higher than that in the control group, while the incidence of PD in the surgery group was similar to that in the control group (hazard ratio 1.45 [0.89–2.22]). Patients with OSA are at an increased risk of developing PD, and uvulopalatopharyngoplasty may mitigate this risk.
Collapse
Affiliation(s)
- Heung Man Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Kyung-Do Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Korea
| | - Jeffrey D Suh
- Department of Head and Neck Surgery, UCLA School of Medicine, Los Angeles, CA, USA
| | - Jae Hoon Cho
- Department of Otorhinolaryngology-Head and Neck Surgery, Konkuk University College of Medicine, 4-12 Hwayang-dong, Gwangjin-gu, Seoul, 05030, Korea.
| |
Collapse
|
28
|
Butt A, Kamtchum-Tatuene J, Khan K, Shuaib A, Jickling GC, Miyasaki JM, Smith EE, Camicioli R. White matter hyperintensities in patients with Parkinson's disease: A systematic review and meta-analysis. J Neurol Sci 2021; 426:117481. [PMID: 33975191 DOI: 10.1016/j.jns.2021.117481] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/25/2021] [Accepted: 05/02/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Mechanisms driving neurodegeneration in Parkinson's disease (PD) are unclear and neurovascular dysfunction may be a contributing factor. White matter hyperintensities (WMH) are commonly found on brain MRI in patients with PD. It is controversial if they are more prevalent or more severe in PD compared with controls. This systematic review aims to answer this question. METHODS A systematic search of electronic databases was conducted for studies of WMH in patients with PD. A qualitative synthesis was done for studies reporting WMH prevalence or WMH scores on a visual rating scale (VRS). In studies reporting total WMH volume, the difference between patients with PD and controls was pooled using random effects meta-analysis. RESULTS Among 3860 subjects from 24 studies, 2360 were cases and 1500 controls. Fifteen studies reported WMH scores and four studies reported the prevalence of WMH. On VRS, five studies reported no difference in WMH scores, three found higher WMH scores in PD compared to controls, three reported increased WMH scores either in periventricular or deep white matter, and four reported higher scores only in PD with dementia. In studies reporting WMH volume, there was no difference between patients with PD and controls (pooled standardized mean difference = 0.1, 95%CI: -0.1-0.4, I2 = 81%). CONCLUSION WMH are not more prevalent or severe in patients with PD than in age-matched controls. PD dementia may have more severe WMH compared to controls and PD with normal cognition. Prospective studies using standardized methods of WMH assessment are needed.
Collapse
Affiliation(s)
- Asif Butt
- Department of Medicine, Division of Neurology, University of Alberta, 116 St & 85 Ave, Edmonton, AB T6G 2R3, Canada.
| | - Joseph Kamtchum-Tatuene
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Khurshid Khan
- Department of Medicine, Division of Neurology, University of Alberta, 116 St & 85 Ave, Edmonton, AB T6G 2R3, Canada
| | - Ashfaq Shuaib
- Department of Medicine, Division of Neurology, University of Alberta, 116 St & 85 Ave, Edmonton, AB T6G 2R3, Canada
| | - Glen C Jickling
- Department of Medicine, Division of Neurology, University of Alberta, 116 St & 85 Ave, Edmonton, AB T6G 2R3, Canada
| | - Janis M Miyasaki
- Department of Medicine, Division of Neurology, University of Alberta, 116 St & 85 Ave, Edmonton, AB T6G 2R3, Canada
| | - Eric E Smith
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, 2500 University Dr NW, Calgary, AB T2N 1N4, Canada
| | - Richard Camicioli
- Department of Medicine, Division of Neurology, University of Alberta, 116 St & 85 Ave, Edmonton, AB T6G 2R3, Canada
| |
Collapse
|
29
|
Amyloid related cerebral microbleed and plasma Aβ40 are associated with cognitive decline in Parkinson's disease. Sci Rep 2021; 11:7115. [PMID: 33782518 PMCID: PMC8007804 DOI: 10.1038/s41598-021-86617-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/25/2021] [Indexed: 02/01/2023] Open
Abstract
Cerebral microbleeds (MBs) have been found in patients with cognitive decline. We aimed to examine whether MBs are associated with motor or cognitive decline in patients with Parkinson’s disease (PD). We enrolled 135 PD patients and 34 healthy controls. All participants underwent brain MRI and plasma biomarker assays, including tau, Aβ42, Aβ40, and α-synuclein. PD with dementia (PDD) was operationally defined as Mini-Mental State Examination (MMSE) score < 26 and advanced motor stage was defined as Hoehn-Yahr stage ≥ 3 during “on” status. The association between MBs and disease severity was examined using multivariate logistic regression models. More lobar MBs were observed in PD patients than controls (20.7% vs. 3.3%, p = 0.031). PDD patients had more lobar MBs (33.3% vs. 15.6%, p = 0.034), more white matter hyperintensity (p = 0.021) and reduced hippocampal volume (p = 0.001) than PD with normal cognition. The presence of lobar MB (odds ratio = 2.83 [95% confidence interval 1.04–7.70], p = 0.042) and severe white matter hyperintensity (3.29 [1.21–8.96], p = 0.020) was independently associated with PDD after adjusting for vascular risk factors and other confounders. Furthermore, plasma Aβ40 levels were associated the MMSE score (p = 0.004) after adjusting for age and sex. Our findings demonstrated that lobar MBs, reduced hippocampal volume, and elevated plasma Aβ40 levels are associated with PDD.
Collapse
|
30
|
Dadar M, Miyasaki J, Duchesne S, Camicioli R. White matter hyperintensities mediate the impact of amyloid ß on future freezing of gait in Parkinson's disease. Parkinsonism Relat Disord 2021; 85:95-101. [PMID: 33770671 DOI: 10.1016/j.parkreldis.2021.02.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/17/2021] [Accepted: 02/28/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Freezing of gait (FOG) is a common symptom in Parkinson's Disease (PD) patients. Previous studies have reported relationships between FOG, substantia nigra (SN) degeneration, dopamine transporter (DAT) concentration, as well as amyloid β deposition. However, there is a paucity of research on the concurrent impact of white matter damage. OBJECTIVES To assess the inter-relationships between these different co-morbidities, their impact on future FOG and whether they act independently of each other. METHODS We used baseline MRI and longitudinal gait data from 423 de novo PD patients from the Parkinson's Progression Markers Initiative (PPMI). We used deformation based morphometry (DBM) from T1-weighted MRI to measure SN atrophy, and segmentation of white matter hyperintensities (WMH) as a measure of WM pathological load. Putamen and caudate DAT levels from SPECT as well as cerebrospinal fluid (CSF) amyloid β were obtained directly from the PPMI. Following correlation analyses, we investigated whether WMH burden mediates the impact of amyloid β on future FOG. RESULTS SN DBM, WMH load, putamen and caudate DAT activity and CSF amyloid β levels were significantly different between PD patients with and without future FOG (p < 0.008). Mediation analysis demonstrated an effect of CSF amyloid β levels on future FOG via WMH load, independent of SN atrophy and striatal DAT activity levels. CONCLUSIONS Amyloid β might impact future FOG in PD patients through an increase in WMH burden, in a pathway independent of Lewy body pathology.
Collapse
Affiliation(s)
- Mahsa Dadar
- CERVO Brain Research Center, Centre Intégré Universitaire Santé et Services Sociaux de La Capitale Nationale, Québec, QC, Canada.
| | - Janis Miyasaki
- Neuroscience and Mental Health Institute and Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Simon Duchesne
- CERVO Brain Research Center, Centre Intégré Universitaire Santé et Services Sociaux de La Capitale Nationale, Québec, QC, Canada; Department of Radiology and Nuclear Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Richard Camicioli
- Neuroscience and Mental Health Institute and Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
31
|
Pierzchlińska A, Kwaśniak-Butowska M, Sławek J, Droździk M, Białecka M. Arterial Blood Pressure Variability and Other Vascular Factors Contribution to the Cognitive Decline in Parkinson's Disease. Molecules 2021; 26:molecules26061523. [PMID: 33802165 PMCID: PMC8001922 DOI: 10.3390/molecules26061523] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 11/24/2022] Open
Abstract
Dementia is one of the most disabling non-motor symptoms in Parkinson’s disease (PD). Unlike in Alzheimer’s disease, the vascular pathology in PD is less documented. Due to the uncertain role of commonly investigated metabolic or vascular factors, e.g., hypertension or diabetes, other factors corresponding to PD dementia have been proposed. Associated dysautonomia and dopaminergic treatment seem to have an impact on diurnal blood pressure (BP) variability, which may presumably contribute to white matter hyperintensities (WMH) development and cognitive decline. We aim to review possible vascular and metabolic factors: Renin-angiotensin-aldosterone system, vascular endothelial growth factor (VEGF), hyperhomocysteinemia (HHcy), as well as the dopaminergic treatment, in the etiopathogenesis of PD dementia. Additionally, we focus on the role of polymorphisms within the genes for catechol-O-methyltransferase (COMT), apolipoprotein E (APOE), vascular endothelial growth factor (VEGF), and for renin-angiotensin-aldosterone system components, and their contribution to cognitive decline in PD. Determining vascular risk factors and their contribution to the cognitive impairment in PD may result in screening, as well as preventive measures.
Collapse
Affiliation(s)
- Anna Pierzchlińska
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, Aleja Powstańców Wlkp 72, 70-111 Szczecin, Poland;
- Correspondence: (A.P.); (M.D.)
| | - Magdalena Kwaśniak-Butowska
- Division of Neurological and Psychiatric Nursing, Medical University of Gdansk, Aleja Jana Pawła II 50, 80-462 Gdansk, Poland; (M.K.-B.); (J.S.)
- Department of Neurology, St Adalbert Hospital, Aleja Jana Pawła II 50, 80-462 Gdansk, Poland
| | - Jarosław Sławek
- Division of Neurological and Psychiatric Nursing, Medical University of Gdansk, Aleja Jana Pawła II 50, 80-462 Gdansk, Poland; (M.K.-B.); (J.S.)
- Department of Neurology, St Adalbert Hospital, Aleja Jana Pawła II 50, 80-462 Gdansk, Poland
| | - Marek Droździk
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Aleja Powstańców Wlkp 72, 70-111 Szczecin, Poland
- Correspondence: (A.P.); (M.D.)
| | - Monika Białecka
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, Aleja Powstańców Wlkp 72, 70-111 Szczecin, Poland;
| |
Collapse
|
32
|
Crowley SJ, Banan G, Amin M, Tanner JJ, Hizel L, Nguyen P, Brumback B, Rodriguez K, McFarland N, Bowers D, Ding M, Mareci TA, Price CC. Statistically Defined Parkinson's Disease Executive and Memory Cognitive Phenotypes: Demographic, Behavioral, and Structural Neuroimaging Comparisons. JOURNAL OF PARKINSONS DISEASE 2021; 11:283-297. [PMID: 33216042 DOI: 10.3233/jpd-202166] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Some individuals with Parkinson's disease (PD) experience working memory and inhibitory difficulties, others learning and memory difficulties, while some only minimal to no cognitive deficits for many years. OBJECTIVE To statistically derive PD executive and memory phenotypes, and compare PD phenotypes on disease and demographic variables, vascular risk factors, and specific neuroimaging variables with known associations to executive and memory function relative to non-PD peers. METHODS Non-demented individuals with PD (n = 116) and non-PD peers (n = 62) were recruited to complete neuropsychology measures, blood draw, and structural magnetic resonance imaging. Tests representing the cognitive domains of interest (4 executive function, 3 memory) were included in a k-means cluster analysis comprised of the PD participants. Resulting clusters were compared demographic and disease-related variables, vascular risk markers, gray/white regions of interest, and white matter connectivity between known regions involved in executive and memory functions (dorsolateral prefrontal cortices to caudate nuclei; entorhinal cortices to hippocampi). RESULTS Clusters showed: 1) PD Executive, n = 25; 2) PD Memory, n = 35; 3) PD Cognitively Well; n = 56. Even after disease variable corrections, PD Executive had less subcortical gray matter, white matter, and fewer bilateral dorsolateral-prefrontal cortex to caudate nucleus connections; PD Memory showed bilaterally reduced entorhinal-hippocampal connections. PD Cognitively Well showed only reduced putamen volume and right entorhinal cortex to hippocampi connections relative to non-PD peers. Groups did not statistically differ on cortical integrity measures or cerebrovascular disease markers. CONCLUSION PD cognitive phenotypes showed different structural gray and white matter patterns. We discuss data relative to phenotype demographics, cognitive patterns, and structural brain profiles.
Collapse
Affiliation(s)
- Samuel J Crowley
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA
| | - Guita Banan
- Department of Biochemistry and Molecular Biology, Gainesville, FL, USA.,McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Manish Amin
- Department of Biochemistry and Molecular Biology, Gainesville, FL, USA.,McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Jared J Tanner
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA.,McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Loren Hizel
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA
| | - Peter Nguyen
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA
| | - Babette Brumback
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Katie Rodriguez
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA
| | - Nikolaus McFarland
- Department of Neurology, Gainesville, FL, USA.,Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Dawn Bowers
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA.,Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Mingzhou Ding
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA.,Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Thomas A Mareci
- Department of Biochemistry and Molecular Biology, Gainesville, FL, USA.,McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Catherine C Price
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA.,Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| |
Collapse
|
33
|
Forbes E, Tropea TF, Mantri S, Xie SX, Morley JF. Modifiable Comorbidities Associated with Cognitive Decline in Parkinson's Disease. Mov Disord Clin Pract 2021; 8:254-263. [PMID: 33553496 DOI: 10.1002/mdc3.13143] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/23/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Background Cognitive impairment (CI) is one of the most feared and debilitating complications of PD. No therapy has been shown to slow or prevent CI in PD. Objective To determine associations between modifiable comorbidities, including cardiovascular disease risk factors, mood disorders, and sleep characteristics, and rate of cognitive decline in Parkinson's disease (PD). Methods Data from the Parkinson's Progression Markers Initiative (PPMI) cohort was queried for baseline cardiovascular disease risk factors, mood disorders, and sleep characteristics. Linear mixed- effects models (LME) were used to examine the association between baseline factors and change in cognition, evaluated by the Montreal Cognitive Assessment (MoCA) over time. Baseline comorbidities found to affect MoCA decline were assessed for an association with focal cognitive domains using LME. Results Higher Body Mass Index (BMI) (β = -0.009, P = 0.039), State Trait Anxiety Inventory (STAI) (β = -0.005, P < 0.001), Geriatric Depression Scale (GDS) (β = -0.034, P < 0.001), Epworth Sleepiness Scale (ESS) (β = -0.017, P = 0.003), and REM Sleep Behavior Disorder Screening Questionnaire (RBDSQ) (β = -0.037, P < 0.001) were associated with faster rates of MoCA decline. Using established cut-offs for clinically significant symptoms, being overweight, or the presence of depression, excessive day time sleepiness (EDS), and possible REM sleep behavior disorder (pRBD), were all associated with faster rate of cognitive decline. Conclusion Several modifiable baseline comorbidities are associated with faster rate of CI over time in patients with PD. These associations identify potential opportunities for early intervention that could influence CI in PD.
Collapse
Affiliation(s)
- Emily Forbes
- Parkinson's Disease Research, Education and Clinical Center, Michael J. Crescenz Veterans Affairs Medical Center Philadelphia Pennsylvania USA.,Department of Neurology Perelman School of Medicine, University of Pennsylvania Philadelphia Pennsylvania USA
| | - Thomas F Tropea
- Department of Neurology Perelman School of Medicine, University of Pennsylvania Philadelphia Pennsylvania USA
| | - Sneha Mantri
- Parkinson's Disease Research, Education and Clinical Center, Michael J. Crescenz Veterans Affairs Medical Center Philadelphia Pennsylvania USA.,Department of Neurology Duke University School of Medicine Durham North Carolina USA
| | - Sharon X Xie
- Department of Biostatistics, Epidemiology, and Informatics University of Pennsylvania School of Medicine Philadelphia Pennsylvania USA
| | - James F Morley
- Parkinson's Disease Research, Education and Clinical Center, Michael J. Crescenz Veterans Affairs Medical Center Philadelphia Pennsylvania USA.,Department of Neurology Perelman School of Medicine, University of Pennsylvania Philadelphia Pennsylvania USA
| |
Collapse
|
34
|
Paez AG, Gu C, Rajan S, Miao X, Cao D, Kamath V, Bakker A, Unschuld PG, Pantelyat AY, Rosenthal LS, Hua J. Differential Changes in Arteriolar Cerebral Blood Volume between Parkinson's Disease Patients with Normal and Impaired Cognition and Mild Cognitive Impairment (MCI) Patients without Movement Disorder - An Exploratory Study. Tomography 2020; 6:333-342. [PMID: 33364423 PMCID: PMC7744190 DOI: 10.18383/j.tom.2020.00033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cognitive impairment amongst Parkinson's disease (PD) patients is highly prevalent and associated with an increased risk of dementia. There is growing evidence that altered cerebrovascular functions contribute to cognitive impairment. Few studies have compared cerebrovascular changes in PD patients with normal and impaired cognition and those with mild-cognitive-impairment (MCI) without movement disorder. Here, we investigated arteriolar-cerebral-blood-volume (CBVa), an index reflecting the homeostasis of the most actively regulated segment in the microvasculature, using advanced MRI in various brain regions in PD and MCI patients and matched controls. Our goal is to find brain regions with altered CBVa that are specific to PD with normal and impaired cognition, and MCI-without-movement-disorder, respectively. In PD patients with normal cognition (n=10), CBVa was significantly decreased in the substantia nigra, caudate and putamen when compared to controls. In PD patients with impaired cognition (n=6), CBVa showed a decreasing trend in the substantia nigra, caudate and putamen, but was significantly increased in the presupplementary motor area and intracalcarine gyrus compared to controls. In MCI-patients-without-movement-disorder (n=18), CBVa was significantly increased in the caudate, putamen, hippocampus and lingual gyrus compared to controls. These findings provide important information for efforts towards developing biomarkers for the evaluation of potential risk of PD dementia (PDD) in PD patients. The current study is limited in sample size and therefore is exploratory in nature. The data from this pilot study will serve as the basis for power analysis for subsequent studies to further investigate and validate the current findings.
Collapse
Affiliation(s)
- Adrian G. Paez
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD
- Neurosection, Division of MR Research, Department of Radiology
| | - Chunming Gu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD
- Neurosection, Division of MR Research, Department of Radiology
| | - Suraj Rajan
- Department of Neurology; and
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD; and
| | - Xinyuan Miao
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD
- Neurosection, Division of MR Research, Department of Radiology
| | - Di Cao
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD
- Neurosection, Division of MR Research, Department of Radiology
- Department of Biomedical Engineering
| | - Vidyulata Kamath
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD; and
| | - Arnold Bakker
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD; and
| | - Paul G. Unschuld
- Department of Psychogeriatric Medicine, Psychiatric University Hospital Zurich, Zurich, Switzerland
| | | | | | - Jun Hua
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD
- Neurosection, Division of MR Research, Department of Radiology
| |
Collapse
|
35
|
Vascular risk factors, white matter lesions and cognitive impairment in Parkinson's disease: the PACOS longitudinal study. J Neurol 2020; 268:549-558. [PMID: 32865628 PMCID: PMC7880923 DOI: 10.1007/s00415-020-10189-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/18/2020] [Accepted: 08/24/2020] [Indexed: 11/24/2022]
Abstract
Background Vascular risk factors (VRFs) may be associated with cognitive decline in early Parkinson’s disease (PD) but results are inconclusive. The identification of modifiable risk factors is relevant for prevention and treatment. Methods Parkinson’s disease (PD) patients of the PACOS cohort who underwent a baseline and follow-up neuropsychological evaluation were enrolled in the study. PD with Mild Cognitive Impairment (MCI) and dementia (PDD) were diagnosed according to the MDS criteria. A Baseline 1.5 T brain MRI was used to calculate the white matter lesions (WMLs) burden using the Wahlund visual scale. Laboratory data, presence of hypertension, diabetes and use of anti-hypertensive drugs were collected and the Framingham Risk (FR) score was calculated. VRFs predicting PD-MCI and PDD were evaluated using Cox proportional hazard regression model. Results Out of 139 enrolled patients, 84 (60.4%) were classified as normal cognition (NC) and 55 (39.6%) as MCI at baseline. At follow-up 28 (33.3%) PD-NC developed MCI and 4 (4.8%) PDD (follow-up time 23.5 ± 10.3 months). Out of 55 PD-MCI patients at baseline, 14 (25.4%) converted to PDD. At multivariate analysis among PD-NC a systolic blood pressure (SBP) > 140 mmHg was the stronger predictor of MCI (adjHR 4.04; 95% CI 1.41–11.3) while the presence of MCI at baseline (adj HR 7.55; 95% CI 1.76–32.3) and a severe WMLs burden (adj HR 2.80; 95% CI 0.86–9.04) were the strongest predictors of PDD, even if this latter association has a trend towards significance. Conclusion Hypertension represents the most important modifiable risk factor for PD-MCI in the PACOS cohort, increasing the risk of about four times.
Collapse
|
36
|
Wang T, Yuan F, Chen Z, Zhu S, Chang Z, Yang W, Deng B, Que R, Cao P, Chao Y, Chan L, Pan Y, Wang Y, Xu L, Lyu Q, Chan P, Yenari MA, Tan EK, Wang Q. Vascular, inflammatory and metabolic risk factors in relation to dementia in Parkinson's disease patients with type 2 diabetes mellitus. Aging (Albany NY) 2020; 12:15682-15704. [PMID: 32805719 PMCID: PMC7467390 DOI: 10.18632/aging.103776] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 07/14/2020] [Indexed: 12/14/2022]
Abstract
There are limited data on vascular, inflammatory, metabolic risk factors of dementia in Parkinson’s disease (PD) with type 2 diabetes mellitus (DM) (PD-DM). In a study of 928 subjects comprising of 215 PD with DM (including 31 PD-DM with dementia, PD-DMD), 341 PD without DM (including 31 PD with dementia, PDD) and 372 DM without PD (including 35 DM with dementia, DMD) patients, we investigated if vascular, inflammatory, metabolic, and magnetic resonance imaging (MRI) markers were associated with dementia in PD-DM. Lower fasting blood glucose (FBG<5mmol/L, OR=4.380; 95%CI: 1.748-10.975; p=0.002), higher homocysteine (HCY>15μmol/L, OR=3.131; 95%CI: 1.243-7.888; p=0.015) and hyperlipidemia (OR=3.075; 95%CI: 1.142-8.277; p=0.026), increased age (OR=1.043; 95%CI: 1.003-1.084; p=0.034) were the most significant risk factors in PDD patients. Lower low-density lipoprotein cholesterol (LDL-C<2mmol/L, OR=4.499; 95%CI: 1.568-12.909; p=0.005) and higher fibrinogen (>4g/L, OR=4.066; 95%CI: 1.467-11.274; p=0.007) were the most significant risk factors in PD-DMD patients. The area under the curve (AUC) for fibrinogen and LDL-C was 0.717 (P=0.001), with a sensitivity of 80.0% for the prediction of PD-DMD. In summary, we identified several factors including LDL-C and fibrinogen as significant risk factors for PD-DMD and these may have prognostic and treatment implications.
Collapse
Affiliation(s)
- Ting Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Feilan Yuan
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Zhenze Chen
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Shuzhen Zhu
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Zihan Chang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Wanlin Yang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Bin Deng
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Rongfang Que
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Peihua Cao
- Clinical Research Center, ZhuJiang Hospital of Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Yinxia Chao
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Duke-NUS Medical School, Singapore
| | - Lingling Chan
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Duke-NUS Medical School, Singapore
| | - Ying Pan
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yanping Wang
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Linting Xu
- Department of Neurology, Puning People's Hospital, Puning, Guangdong, China
| | - Qiurong Lyu
- Department of Neurology, Guiping People's Hospital, Guangxi, China
| | - Piu Chan
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Midori A Yenari
- Department of Neurology, University of California, San Francisco and the San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Duke-NUS Medical School, Singapore
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
37
|
Kaufmann H, Palma JA. White Matter Hyperintensities in the Synucleinopathies: Orthostatic Hypotension, Supine Hypertension, or Both? Mov Disord Clin Pract 2020; 7:595-598. [PMID: 32775503 PMCID: PMC7396862 DOI: 10.1002/mdc3.13000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 06/10/2020] [Indexed: 11/10/2022] Open
Affiliation(s)
- Horacio Kaufmann
- Department of Neurology, Dysautonomia Center New York University School of Medicine New York New York USA
| | - Jose-Alberto Palma
- Department of Neurology, Dysautonomia Center New York University School of Medicine New York New York USA
| |
Collapse
|
38
|
Dadar M, Gee M, Shuaib A, Duchesne S, Camicioli R. Cognitive and motor correlates of grey and white matter pathology in Parkinson's disease. Neuroimage Clin 2020; 27:102353. [PMID: 32745994 PMCID: PMC7399172 DOI: 10.1016/j.nicl.2020.102353] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/25/2020] [Accepted: 07/15/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Previous studies have found associations between grey matter atrophy and white matter hyperintensities (WMH) of vascular origin with cognitive and motor deficits in Parkinson's disease (PD). Here we investigate these relationships in a sample of PD patients and age-matched healthy controls. METHODS Data included 50 PD patients and 45 age-matched controls with T1-weighted and FLAIR scans at baseline, 18-months, and 36-months follow-up. Deformation-based morphometry was used to measure grey matter atrophy. SNIPE (Scoring by Nonlocal Image Patch Estimator) was used to measure Alzheimer's disease-like textural patterns in the hippocampi. WMHs were segmented using T1-weighted and FLAIR images. The relationship between MRI features and clinical scores was assessed using mixed-effects models. The motor subscore of the Unified Parkinson's Disease Rating Scale (UPDRSIII), number of steps in a walking trial, and Dementia Rating Scale (DRS) were used respectively as measures of motor function, gait, and cognition. RESULTS Substantia nigra atrophy was significantly associated with motor deficits, with a greater impact in PDs (p < 0.05). Hippocampal SNIPE scores were associated with cognitve decline in both PD and controls (p < 0.01). WMH burden was significantly associated with cognitive decline and increased motor deficits in the PD group, and gait deficits in both PD and controls (p < 0.03). CONCLUSION While substantia nigra atrophy and WMH burden were significantly associated with additional motor deficits, WMH burden and hippocampal atrophy were associated with cognitive deficits in PD patients. These results suggest an additive contribution of both grey and white matter damage to the motor and cognitive deficits in PD.
Collapse
Affiliation(s)
- Mahsa Dadar
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Laval University, Canada.
| | - Myrlene Gee
- Department of Medicine, Division of Neurology, University of Alberta, Canada.
| | - Ashfaq Shuaib
- Department of Medicine, Division of Neurology, University of Alberta, Canada.
| | - Simon Duchesne
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Laval University, Canada.
| | - Richard Camicioli
- Department of Medicine, Division of Neurology, University of Alberta, Canada.
| |
Collapse
|
39
|
Marras C, Mestre T, McDermott MP. Huntington's Disease and Hypertension: Sorting Out Mixed Messages. Mov Disord 2020; 35:915-917. [PMID: 32562461 DOI: 10.1002/mds.28076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 11/08/2022] Open
Affiliation(s)
- Connie Marras
- The Edmond J. Safra Program in Parkinson's Disease, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Tiago Mestre
- Parkinson's Disease and Movement Disorders Clinic, Division of Neurology, Department of Medicine, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Michael P McDermott
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, New York, USA
| |
Collapse
|
40
|
Imbriani P, D'Angelo V, Platania P, Di Lazzaro G, Scalise S, Salimei C, El Atiallah I, Colona VL, Mercuri NB, Bonsi P, Pisani A, Schirinzi T, Martella G. Ischemic injury precipitates neuronal vulnerability in Parkinson's disease: Insights from PINK1 mouse model study and clinical retrospective data. Parkinsonism Relat Disord 2020; 74:57-63. [PMID: 32335490 DOI: 10.1016/j.parkreldis.2020.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Increasing evidence demonstrates the relevant association between Parkinson's disease (PD) and vascular diseases/risk factors, as well as a worse clinico-pathological progression in those patients with vascular comorbidity. The mechanisms underlying this relationship have not been clarified yet, although their comprehension is critical in a perspective of disease-modifying treatments development or prevention. METHODS We performed an experimental protocol of ischemic injury (glucose-oxygen deprivation, OGD) on PTEN-induced kinase 1 knockout (PINK1-/-) mice, a well-established PD model, looking at both electrophysiological and morphological changes in basal ganglia. In addition, 253 PD patients were retrospectively analysed, to estimate the prevalence of vascular risk factors. RESULTS In PINK1-/- mice, the OGD protocol induced electrophysiological (prolonged depolarization) and morphological alterations (picnotic cells, cellular loss and swelling, thickening of nuclear chromatin) in striatal medium spiny neurons and nigral dopaminergic neurons. Vascular comorbidity occurred in 75% of PD patients. CONCLUSIONS The ischemic injury precipitates neuronal vulnerability in basal ganglia of PINK1-/- mice, probably through an impairment of mitochondrial metabolism and higher oxidative stress. These experimental data may provide a potential mechanistic explanation for both the association between vascular diseases and PD and their reciprocal interactions in determining the clinico-pathological burden of PD patients.
Collapse
Affiliation(s)
- Paola Imbriani
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy; IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Vincenza D'Angelo
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Paola Platania
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Giulia Di Lazzaro
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Simona Scalise
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Chiara Salimei
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Ilham El Atiallah
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy; IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Vito Luigi Colona
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy; IRCCS Fondazione Santa Lucia, Rome, Italy
| | | | - Antonio Pisani
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy; IRCCS Fondazione Santa Lucia, Rome, Italy.
| | - Tommaso Schirinzi
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Giuseppina Martella
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy; IRCCS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
41
|
Huang X, Wen MC, Ng SYE, Hartono S, Chia NSY, Choi X, Tay KY, Au WL, Chan LL, Tan EK, Tan LCS. Periventricular white matter hyperintensity burden and cognitive impairment in early Parkinson's disease. Eur J Neurol 2020; 27:959-966. [PMID: 32124496 DOI: 10.1111/ene.14192] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 02/20/2020] [Indexed: 01/29/2023]
Abstract
BACKGROUND AND PURPOSE This study quantified the total brain and periventricular white matter hyperintensity (WMH) burdens in patients with early Parkinson's disease (PD) and explored their associations with cardiovascular risk factors and cognitive performance. METHODS A total of 175 non-demented patients with early PD who had undergone baseline brain magnetic resonance imaging were included. Comprehensive neurocognitive testing was conducted to identify PD with mild cognitive impairment (PD-MCI) and to evaluate performances in individual cognitive domains. Cardiovascular risk was expressed as a modified Framingham 10-year cardiovascular risk score (mFRS). RESULTS A total of 53.7% of this early PD cohort fulfilled the diagnostic criteria for PD-MCI. An increase in mFRS was significantly associated with increases in the total brain WMH (P = 0.015) and periventricular WMH (P = 0.040) burden, independent of age and gender. The periventricular WMH burden was significantly associated with PD-MCI (P = 0.046) in early PD, independent of cardiovascular risk factors. Patients in the 5th quintile of periventricular WMH burden were 8.6 times more likely to have PD-MCI compared with patients in the 1st quintile of periventricular WMH burden (P = 0.004). However, total brain WMH burden was not associated with PD-MCI (P = 0.158). In individual cognitive domains, heavier periventricular WMH burden was associated with worse executive function and visuospatial function independent of cardiovascular risk factors. CONCLUSION Periventricular WMHs are a useful imaging biomarker for cognitive impairment in early PD. Cardiovascular risk factors, although associated with periventricular WMHs, were unable to fully explain the association between periventricular WMHs and cognitive impairment in early PD.
Collapse
Affiliation(s)
- X Huang
- Duke-NUS Medical School, Singapore, Singapore
| | - M-C Wen
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore.,Parkinson's Disease and Movement Disorders Centre, USA Parkinson Foundation International Center of Excellence, National Neuroscience Institute, Singapore, Singapore
| | - S Y-E Ng
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore.,Parkinson's Disease and Movement Disorders Centre, USA Parkinson Foundation International Center of Excellence, National Neuroscience Institute, Singapore, Singapore
| | - S Hartono
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore.,Parkinson's Disease and Movement Disorders Centre, USA Parkinson Foundation International Center of Excellence, National Neuroscience Institute, Singapore, Singapore
| | - N S-Y Chia
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore.,Parkinson's Disease and Movement Disorders Centre, USA Parkinson Foundation International Center of Excellence, National Neuroscience Institute, Singapore, Singapore
| | - X Choi
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore.,Parkinson's Disease and Movement Disorders Centre, USA Parkinson Foundation International Center of Excellence, National Neuroscience Institute, Singapore, Singapore
| | - K-Y Tay
- Duke-NUS Medical School, Singapore, Singapore.,Department of Neurology, National Neuroscience Institute, Singapore, Singapore.,Parkinson's Disease and Movement Disorders Centre, USA Parkinson Foundation International Center of Excellence, National Neuroscience Institute, Singapore, Singapore
| | - W-L Au
- Duke-NUS Medical School, Singapore, Singapore.,Department of Neurology, National Neuroscience Institute, Singapore, Singapore.,Parkinson's Disease and Movement Disorders Centre, USA Parkinson Foundation International Center of Excellence, National Neuroscience Institute, Singapore, Singapore
| | - L-L Chan
- Department of Radiology, Singapore General Hospital, Singapore, Singapore
| | - E-K Tan
- Duke-NUS Medical School, Singapore, Singapore.,Department of Neurology, National Neuroscience Institute, Singapore, Singapore.,Parkinson's Disease and Movement Disorders Centre, USA Parkinson Foundation International Center of Excellence, National Neuroscience Institute, Singapore, Singapore
| | - L C-S Tan
- Duke-NUS Medical School, Singapore, Singapore.,Department of Neurology, National Neuroscience Institute, Singapore, Singapore.,Parkinson's Disease and Movement Disorders Centre, USA Parkinson Foundation International Center of Excellence, National Neuroscience Institute, Singapore, Singapore
| |
Collapse
|
42
|
Gong L, Li H, Yang D, Peng Y, Liu D, Zhong M, Zhang B, Xu R, Kang J. Striatum Shape Hypertrophy in Early Stage Parkinson's Disease With Excessive Daytime Sleepiness. Front Neurosci 2020; 13:1353. [PMID: 31992965 PMCID: PMC6964599 DOI: 10.3389/fnins.2019.01353] [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: 09/08/2019] [Accepted: 12/02/2019] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Excessive daytime sleepiness (EDS) is one of the common and burdensome non-motor symptoms of Parkinson's disease (PD). However, the underlying neuropathology mechanism in PD patients with EDS (PD-EDS) remains unclear. The present study aims to delineate potential locations of structural alteration of subcortical regions in early stage and drug-naïve PD-EDS. METHODS The study had 252 patients with PD and 92 matched healthy controls (HC). EDS was estimated with the Epworth Sleepiness Scale, with a cutoff of 10. Ultimately, 59 patients were considered as PD-EDS. The remaining 193 were PD patients without EDS (PD-nEDS). FMRIB's Integrated Registration and Segmentation Tool (FIRST) was employed to assess the volumetric and surface alterations of subcortical nuclei in PD and PD-EDS. RESULTS Volumetric analyses found no difference in the subcortical nucleus volume between PD and HC, or PD-EDS and PD-nEDS groups. The shape analyses revealed the local atrophic changes in bilateral caudate and right putamen in patients with PD. In addition, the hypertrophic changes were located in the right putamen and left pallidum in PD-EDS than in PD-nEDS. CONCLUSION Our findings revealed the regional hypertrophy of the striatum in PD-EDS. Our results indicate that local hypertrophic striatum would be a valuable early biomarker for detecting the alteration in PD-EDS. The shape analysis contributes valuable information when investigating PD-EDS.
Collapse
Affiliation(s)
- Liang Gong
- Department of Neurology, Chengdu Second People’s Hospital, Chengdu, China
| | - Huaisu Li
- Department of Neurology, Chengdu Second People’s Hospital, Chengdu, China
| | - Dan Yang
- Department of Neurology, Chengdu Second People’s Hospital, Chengdu, China
| | - Yinwei Peng
- Department of Neurology, Chengdu Second People’s Hospital, Chengdu, China
| | - Duan Liu
- Department of Neurology, Chengdu Second People’s Hospital, Chengdu, China
| | - Ming Zhong
- Department of Neurology, Chengdu Second People’s Hospital, Chengdu, China
| | - Bei Zhang
- Department of Neurology, Chengdu Second People’s Hospital, Chengdu, China
| | - Ronghua Xu
- Department of Neurology, Chengdu Second People’s Hospital, Chengdu, China
| | - Jian Kang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
43
|
Weintraub D, Mamikonyan E. The Neuropsychiatry of Parkinson Disease: A Perfect Storm. Am J Geriatr Psychiatry 2019; 27:998-1018. [PMID: 31006550 PMCID: PMC7015280 DOI: 10.1016/j.jagp.2019.03.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/04/2019] [Accepted: 03/04/2019] [Indexed: 12/16/2022]
Abstract
Affective disorders, cognitive decline, and psychosis have long been recognized as common in Parkinson disease (PD), and other psychiatric disorders include impulse control disorders, anxiety symptoms, disorders of sleep and wakefulness, and apathy. Psychiatric aspects of PD are associated with numerous adverse outcomes, yet in spite of this and their frequent occurrence, there is incomplete understanding of epidemiology, presentation, risk factors, neural substrate, and management strategies. Psychiatric features are typically multimorbid, and there is great intra- and interindividual variability in presentation. The hallmark neuropathophysiological changes that occur in PD, plus the association between exposure to dopaminergic medications and certain psychiatric disorders, suggest a neurobiological basis for many psychiatric symptoms, although psychological factors are involved as well. There is evidence that psychiatric disorders in PD are still under-recognized and undertreated and although psychotropic medication use is common, controlled studies demonstrating efficacy and tolerability are largely lacking. Future research on neuropsychiatric complications in PD should be oriented toward determining modifiable correlates or risk factors and establishing efficacious and well-tolerated treatment strategies.
Collapse
Affiliation(s)
- Daniel Weintraub
- Perelman School of Medicine (DW, EM), University of Pennsylvania, Philadelphia; Parkinson's Disease Research, Education and Clinical Center (PADRECC) (DW), Philadelphia Veterans Affairs Medical Center, Philadelphia.
| | - Eugenia Mamikonyan
- Perelman School of Medicine (DW, EM), University of Pennsylvania, Philadelphia
| |
Collapse
|