1
|
Puig-Davi A, Martinez-Horta S, Pérez-Carasol L, Horta-Barba A, Ruiz-Barrio I, Aracil-Bolaños I, Pérez-González R, Rivas-Asensio E, Sampedro F, Campolongo A, Pagonabarraga J, Kulisevsky J. Prediction of Cognitive Heterogeneity in Parkinson's Disease: A 4-Year Longitudinal Study Using Clinical, Neuroimaging, Biological and Electrophysiological Biomarkers. Ann Neurol 2024; 96:981-993. [PMID: 39099459 DOI: 10.1002/ana.27035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024]
Abstract
OBJECTIVE Cognitive impairment in Parkinson's disease (PD) can show a very heterogeneous trajectory among patients. Here, we explored the mechanisms involved in the expression and prediction of different cognitive phenotypes over 4 years. METHODS In 2 independent cohorts (total n = 475), we performed a cluster analysis to identify trajectories of cognitive progression. Baseline and longitudinal level II neuropsychological assessments were conducted, and baseline structural magnetic resonance imaging, resting electroencephalogram and neurofilament light chain plasma quantification were carried out. Linear mixed-effects models were used to study longitudinal changes. Risk of mild cognitive impairment and dementia were estimated using multivariable hazard regression. Spectral power density from the electroencephalogram at baseline and source localization were computed. RESULTS Two cognitive trajectories were identified. Cluster 1 presented stability (PD-Stable) over time, whereas cluster 2 showed progressive cognitive decline (PD-Progressors). The PD-Progressors group showed an increased risk for evolving to PD mild cognitive impairment (HR 2.09; 95% CI 1.11-3.95) and a marked risk for dementia (HR 4.87; 95% CI 1.34-17.76), associated with progressive worsening in posterior-cortical-dependent cognitive processes. Both clusters showed equivalent clinical and sociodemographic characteristics, structural magnetic resonance imaging, and neurofilament light chain levels at baseline. Conversely, the PD-Progressors group showed a fronto-temporo-occipital and parietal slow-wave power density increase, that was in turn related to worsening at 2 and 4 years of follow-up in different cognitive measures. INTERPRETATION In the absence of differences in baseline cognitive function and typical markers of neurodegeneration, the further development of an aggressive cognitive decline in PD is associated with increased slow-wave power density and with a different profile of worsening in several posterior-cortical-dependent tasks. ANN NEUROL 2024;96:981-993.
Collapse
Affiliation(s)
- Arnau Puig-Davi
- Institute of Neuroscience, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - Saul Martinez-Horta
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Laura Pérez-Carasol
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - Andrea Horta-Barba
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Iñigo Ruiz-Barrio
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - Ignacio Aracil-Bolaños
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Rocío Pérez-González
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Alicante, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Elisa Rivas-Asensio
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Frederic Sampedro
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Antonia Campolongo
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Javier Pagonabarraga
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Jaime Kulisevsky
- Institute of Neuroscience, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| |
Collapse
|
2
|
Pang H, Li X, Yu Z, Yu H, Bu S, Wang J, Zhao M, Liu Y, Jiang Y, Fan G. Disentangling gray matter atrophy and its neurotransmitter architecture in drug-naïve Parkinson's disease: an atlas-based correlation analysis. Cereb Cortex 2024; 34:bhae420. [PMID: 39420471 DOI: 10.1093/cercor/bhae420] [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: 06/14/2024] [Revised: 08/19/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
Parkinson's disease is characterized by multiple neurotransmitter systems beyond the traditional dopaminergic pathway, yet their influence on volumetric alterations is not well comprehended. We included 72 de novo, drug-naïve Parkinson's disease patients and 61 healthy controls. Voxel-wise gray matter volume was evaluated between Parkinson's disease and healthy controls, as well as among Parkinson's disease subgroups categorized by clinical manifestations. The Juspace toolbox was utilized to explore the spatial relationship between gray matter atrophy and neurotransmitter distribution. Parkinson's disease patients exhibited widespread GM atrophy in the cerebral and cerebellar regions, with spatial correlations with various neurotransmitter receptors (FDR-P < 0.05). Cognitively impaired Parkinson's disease patients showed gray matter atrophy in the left middle temporal atrophy, which is associated with serotoninergic, dopaminergic, cholinergic, and glutamatergic receptors (FDR-P < 0.05). Postural and gait disorder patients showed atrophy in the right precuneus, which is correlated with serotoninergic, dopaminergic, gamma-aminobutyric acid, and opioid receptors (FDR-P < 0.05). Patients with anxiety showed atrophy in the right superior orbital frontal region; those with depression showed atrophy in the left lingual and right inferior occipital regions. Both conditions were linked to serotoninergic and dopaminergic receptors (FDR-P < 0.05). Parkinson's disease patients exhibited regional gray matter atrophy with a significant distribution of specific neurotransmitters, which might provide insights into the underlying pathophysiology of clinical manifestations and develop targeted intervention strategies.
Collapse
Affiliation(s)
- Huize Pang
- Department of Radiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province, 110001, China
| | - Xiaolu Li
- Department of Radiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province, 110001, China
| | - Ziyang Yu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, 38 Zheda Road, Xihu District, Hangzhou, Zhejiang Province, 310027, China
| | - Hongmei Yu
- Department of Neurology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province, 110001, China
| | - Shuting Bu
- Department of Radiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province, 110001, China
| | - Juzhou Wang
- Department of Radiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province, 110001, China
| | - Mengwan Zhao
- Department of Radiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province, 110001, China
| | - Yu Liu
- Department of Radiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province, 110001, China
| | - Yueluan Jiang
- MR Research Collaboration, Siemens Healthineers, 7 Wangjing Zhonghuan South Road, Chaoyang District, Beijing, 100102, China
| | - Guoguang Fan
- Department of Radiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province, 110001, China
| |
Collapse
|
3
|
Li X, Pang H, Bu S, Zhao M, Wang J, Liu Y, Yu H, Fan G. Stage-dependent differential impact of network communication on cognitive function across the continuum of cognitive decline in Parkinson's disease. Neurobiol Dis 2024; 199:106578. [PMID: 38925316 DOI: 10.1016/j.nbd.2024.106578] [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: 04/02/2024] [Revised: 06/04/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024] Open
Abstract
OBJECTIVE Our objective was to explore the patterns of resting-state network (RSN) connectivity alterations and investigate how the influences of individual-level network connections on cognition varied across clinical stages without assuming a constant relationship. METHODS 108 PD patients with continuum of cognitive decline (PD-NC = 46, PD-MCI = 43, PDD = 19) and 34 healthy controls (HCs) underwent resting-state functional MRI and neuropsychological tests. Independent component analysis (ICA) and graph theory analyses (GTA) were employed to explore RSN connection changes. Additionally, stage-dependent differential impact of network communication on cognitive performance were examined using sparse varying coefficient modeling. RESULTS Compared to HCs, the dorsal attention network (DAN) and dorsal sensorimotor network (dSMN) were central networks with decreased connections in PD-NC and PD-MCI stage, while the lateral visual network (LVN) emerged as a central network in patients with dementia. Additionally, connectivity of the cerebellum network (CBN) increased in the PD-NC and PD-MCI stages. GTA demonstrated decreased nodal metrics for DAN and dSMN, coupled with an increase for CBN. Moreover, the degree centrality (DC) values of DAN and dSMN exhibited a stage-dependent differential impact on cognitive performance across the continuum of cognitive decline. CONCLUSION Our findings suggest that across the progression of cognitive impairment, the LVN gradually transitions into a core node with reduced connectivity, while the enhancement of connections in CBN diminishes. Furthermore, the non-linear relationship between the DC values of RSNs and cognitive decline indicates the potential for tailored interventions targeting specific stages.
Collapse
Affiliation(s)
- Xiaolu Li
- Department of Radiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Huize Pang
- Department of Radiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Shuting Bu
- Department of Radiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Mengwan Zhao
- Department of Radiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Juzhou Wang
- Department of Radiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yu Liu
- Department of Radiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Hongmei Yu
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Guoguang Fan
- Department of Radiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
4
|
Brandão PR, Pereira DA, Grippe TC, Bispo DDDC, Maluf FB, Titze-de-Almeida R, de Almeida e Castro BM, Munhoz RP, Tavares MCH, Cardoso F. Mapping brain morphology to cognitive deficits: a study on PD-CRS scores in Parkinson's disease with mild cognitive impairment. Front Neuroanat 2024; 18:1362165. [PMID: 39206076 PMCID: PMC11349662 DOI: 10.3389/fnana.2024.1362165] [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: 12/27/2023] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Background The Parkinson's Disease-Cognitive Rating Scale (PD-CRS) is a widely used tool for detecting mild cognitive impairment (MCI) in Parkinson's Disease (PD) patients, however, the neuroanatomical underpinnings of this test's outcomes require clarification. This study aims to: (a) investigate cortical volume (CVol) and cortical thickness (CTh) disparities between PD patients exhibiting mild cognitive impairment (PD-MCI) and those with preserved cognitive abilities (PD-IC); and (b) identify the structural correlates in magnetic resonance imaging (MRI) of overall PD-CRS performance, including its subtest scores, within a non-demented PD cohort. Materials and methods This study involved 51 PD patients with Hoehn & Yahr stages I-II, categorized into two groups: PD-IC (n = 36) and PD-MCI (n = 15). Cognitive screening evaluations utilized the PD-CRS and the Montreal Cognitive Assessment (MoCA). PD-MCI classification adhered to the Movement Disorder Society Task Force criteria, incorporating extensive neuropsychological assessments. The interrelation between brain morphology and cognitive performance was determined using FreeSurfer. Results Vertex-wise analysis of the entire brain demonstrated a notable reduction in CVol within a 2,934 mm2 cluster, encompassing parietal and temporal regions, in the PD-MCI group relative to the PD-IC group. Lower PD-CRS total scores correlated with decreased CVol in the middle frontal, superior temporal, inferior parietal, and cingulate cortices. The PD-CRS subtests for Sustained Attention and Clock Drawing were associated with cortical thinning in distinct regions: the Clock Drawing subtest correlated with changes in the parietal lobe, insula, and superior temporal cortex morphology; while the PD-CRS frontal-subcortical scores presented positive correlations with CTh in the transverse temporal, medial orbitofrontal, superior temporal, precuneus, fusiform, and supramarginal regions. Additionally, PD-CRS subtests for Semantic and Alternating verbal fluency were linked to CTh changes in orbitofrontal, temporal, fusiform, insula, and precentral regions. Conclusion PD-CRS performance mirrors neuroanatomical changes across extensive fronto-temporo-parietal areas, covering both lateral and medial cortical surfaces, in PD patients without dementia. The observed changes in CVol and CTh associated with this cognitive screening tool suggest their potential as surrogate markers for cognitive decline in PD. These findings warrant further exploration and validation in multicenter studies involving independent patient cohorts.
Collapse
Affiliation(s)
- Pedro Renato Brandão
- Neuroscience and Behavior Lab, Biological Sciences Institute, University of Brasília (UnB), Brasília, Brazil
- Hospital Sírio-Libanês, Instituto de Ensino e Pesquisa, Brasília, Brazil
| | - Danilo Assis Pereira
- Brazilian Institute of Neuropsychology and Cognitive Sciences (IBNeuro), Brasília, Brazil
| | - Talyta Cortez Grippe
- Movement Disorders Centre, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
| | - Diógenes Diego de Carvalho Bispo
- Radiology Department, Brasilia University Hospital (HUB-UnB), University of Brasília (UnB), Brasília, Brazil
- Radiology Department, Santa Marta Hospital, Taguatinga, Brazil
| | | | - Ricardo Titze-de-Almeida
- Central Institute of Sciences, Research Center for Major Themes – Neurodegenerative disorders, University of Brasília, Brasília, Brazil
| | - Brenda Macedo de Almeida e Castro
- Neuroscience and Behavior Lab, Biological Sciences Institute, University of Brasília (UnB), Brasília, Brazil
- Hospital Sírio-Libanês, Instituto de Ensino e Pesquisa, Brasília, Brazil
| | - Renato Puppi Munhoz
- Movement Disorders Centre, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
| | | | - Francisco Cardoso
- Internal Medicine, Neurology Service, Movement Disorder Centre, The Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
5
|
Chun MY, Lee T, Kim SH, Lee HS, Kim YJ, Lee PH, Sohn YH, Jeong Y, Chung SJ. Hypoperfusion in Alzheimer's Disease-Prone Regions and Dementia Conversion in Parkinson's Disease. Clin Nucl Med 2024; 49:521-528. [PMID: 38584352 DOI: 10.1097/rlu.0000000000005211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
PURPOSE OF THE REPORT Although early detection of individuals at risk of dementia conversion is important in patients with Parkinson's disease (PD), there is still no consensus on neuroimaging biomarkers for predicting future cognitive decline. We aimed to investigate whether cerebral perfusion patterns on early-phase 18 F-N-(3-fluoropropyl)-2β-carboxymethoxy-3β-(4-iodophenyl) nortropane ( 18 F-FP-CIT) PET have the potential to serve as a neuroimaging predictor for early dementia conversion in patients with PD. MATERIALS AND METHODS In this retrospective analysis, we enrolled 187 patients with newly diagnosed PD who underwent dual-phase 18 F-FP-CIT PET at initial assessment and serial cognitive assessments during the follow-up period (>5 years). Patients with PD were classified into 2 groups: the PD with dementia (PDD)-high-risk (PDD-H; n = 47) and the PDD-low-risk (PDD-L; n = 140) groups according to dementia conversion within 5 years of PD diagnosis. We explored between-group differences in the regional uptake in the early-phase 18 F-FP-CIT PET images. We additionally performed a linear discriminant analysis to develop a prediction model for early PDD conversion. RESULTS The PDD-H group exhibited hypoperfusion in Alzheimer's disease (AD)-prone regions (inferomedial temporal and posterior cingulate cortices, and insula) compared with the PDD-L group. A prediction model using regional uptake in the right entorhinal cortex, left amygdala, and left isthmus cingulate cortex could optimally distinguish the PDD-H group from the PDD-L group. CONCLUSIONS Regional hypoperfusion in the AD-prone regions on early-phase 18 F-FP-CIT PET can be a useful biomarker for predicting early dementia conversion in patients with PD.
Collapse
Affiliation(s)
| | | | | | - Hye Sun Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Phil Hyu Lee
- From the Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Young H Sohn
- From the Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | | | | |
Collapse
|
6
|
Basaia S, Agosta F, Sarasso E, Balestrino R, Stojković T, Stanković I, Tomić A, Marković V, Vignaroli F, Stefanova E, Kostić VS, Filippi M. Brain Connectivity Networks Constructed Using MRI for Predicting Patterns of Atrophy Progression in Parkinson Disease. Radiology 2024; 311:e232454. [PMID: 38916507 DOI: 10.1148/radiol.232454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Background Whether connectome mapping of structural and functional connectivity across the brain could be used to predict patterns of atrophy progression in patients with mild Parkinson disease (PD) has not been well studied. Purpose To assess the structural and functional connectivity of brain regions in healthy controls and its relationship with the spread of gray matter (GM) atrophy in patients with mild PD. Materials and Methods This prospective study included participants with mild PD and controls recruited from a single center between January 2012 and December 2023. Participants with PD underwent three-dimensional T1-weighted brain MRI, and the extent of regional GM atrophy was determined at baseline and every year for 3 years. The structural and functional brain connectome was constructed using diffusion tensor imaging and resting-state functional MRI in healthy controls. Disease exposure (DE) indexes-indexes of the pathology of each brain region-were defined as a function of the structural or functional connectivity of all the connected regions in the healthy connectome and the severity of atrophy of the connected regions in participants with PD. Partial correlations were tested between structural and functional DE indexes of each GM region at 1- or 2-year follow-up and atrophy progression at 2- or 3-year follow-up. Prediction models of atrophy at 2- or 3-year follow-up were constructed using exhaustive feature selection. Results A total of 86 participants with mild PD (mean age at MRI, 60 years ± 8 [SD]; 48 male) and 60 healthy controls (mean age at MRI, 62 years ± 9; 31 female) were included. DE indexes at 1 and 2 years were correlated with atrophy at 2 and 3 years (r range, 0.22-0.33; P value range, .002-.04). Models including DE indexes predicted GM atrophy accumulation over 3 years in the right caudate nucleus and some frontal, parietal, and temporal brain regions (R2 range, 0.40-0.61; all P < .001). Conclusion The structural and functional organization of the brain connectome plays a role in atrophy progression in the early stages of PD. © RSNA, 2024 Supplemental material is available for this article. See also the editorial by Yamada in this issue.
Collapse
Affiliation(s)
- Silvia Basaia
- From the Neuroimaging Research Unit, Division of Neuroscience (S.B., F.A., E. Sarasso, R.B., M.F.), Neurology Unit (F.A., M.F.), Department of Rehabilitation and Functional Recovery (E. Sarasso), Neurorehabilitation Unit (M.F.), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy (F.A., R.B., M.F.); Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Maternal Child Health, University of Genoa, Genoa, Italy (E. Sarasso); Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia (T.S., I.S., A.T., V.M., E. Stefanova, V.S.K.); and Neurology Unit, University Hospital Maggiore della Carità, Novara, Italy (F.V.)
| | - Federica Agosta
- From the Neuroimaging Research Unit, Division of Neuroscience (S.B., F.A., E. Sarasso, R.B., M.F.), Neurology Unit (F.A., M.F.), Department of Rehabilitation and Functional Recovery (E. Sarasso), Neurorehabilitation Unit (M.F.), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy (F.A., R.B., M.F.); Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Maternal Child Health, University of Genoa, Genoa, Italy (E. Sarasso); Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia (T.S., I.S., A.T., V.M., E. Stefanova, V.S.K.); and Neurology Unit, University Hospital Maggiore della Carità, Novara, Italy (F.V.)
| | - Elisabetta Sarasso
- From the Neuroimaging Research Unit, Division of Neuroscience (S.B., F.A., E. Sarasso, R.B., M.F.), Neurology Unit (F.A., M.F.), Department of Rehabilitation and Functional Recovery (E. Sarasso), Neurorehabilitation Unit (M.F.), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy (F.A., R.B., M.F.); Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Maternal Child Health, University of Genoa, Genoa, Italy (E. Sarasso); Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia (T.S., I.S., A.T., V.M., E. Stefanova, V.S.K.); and Neurology Unit, University Hospital Maggiore della Carità, Novara, Italy (F.V.)
| | - Roberta Balestrino
- From the Neuroimaging Research Unit, Division of Neuroscience (S.B., F.A., E. Sarasso, R.B., M.F.), Neurology Unit (F.A., M.F.), Department of Rehabilitation and Functional Recovery (E. Sarasso), Neurorehabilitation Unit (M.F.), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy (F.A., R.B., M.F.); Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Maternal Child Health, University of Genoa, Genoa, Italy (E. Sarasso); Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia (T.S., I.S., A.T., V.M., E. Stefanova, V.S.K.); and Neurology Unit, University Hospital Maggiore della Carità, Novara, Italy (F.V.)
| | - Tanja Stojković
- From the Neuroimaging Research Unit, Division of Neuroscience (S.B., F.A., E. Sarasso, R.B., M.F.), Neurology Unit (F.A., M.F.), Department of Rehabilitation and Functional Recovery (E. Sarasso), Neurorehabilitation Unit (M.F.), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy (F.A., R.B., M.F.); Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Maternal Child Health, University of Genoa, Genoa, Italy (E. Sarasso); Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia (T.S., I.S., A.T., V.M., E. Stefanova, V.S.K.); and Neurology Unit, University Hospital Maggiore della Carità, Novara, Italy (F.V.)
| | - Iva Stanković
- From the Neuroimaging Research Unit, Division of Neuroscience (S.B., F.A., E. Sarasso, R.B., M.F.), Neurology Unit (F.A., M.F.), Department of Rehabilitation and Functional Recovery (E. Sarasso), Neurorehabilitation Unit (M.F.), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy (F.A., R.B., M.F.); Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Maternal Child Health, University of Genoa, Genoa, Italy (E. Sarasso); Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia (T.S., I.S., A.T., V.M., E. Stefanova, V.S.K.); and Neurology Unit, University Hospital Maggiore della Carità, Novara, Italy (F.V.)
| | - Aleksandra Tomić
- From the Neuroimaging Research Unit, Division of Neuroscience (S.B., F.A., E. Sarasso, R.B., M.F.), Neurology Unit (F.A., M.F.), Department of Rehabilitation and Functional Recovery (E. Sarasso), Neurorehabilitation Unit (M.F.), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy (F.A., R.B., M.F.); Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Maternal Child Health, University of Genoa, Genoa, Italy (E. Sarasso); Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia (T.S., I.S., A.T., V.M., E. Stefanova, V.S.K.); and Neurology Unit, University Hospital Maggiore della Carità, Novara, Italy (F.V.)
| | - Vladana Marković
- From the Neuroimaging Research Unit, Division of Neuroscience (S.B., F.A., E. Sarasso, R.B., M.F.), Neurology Unit (F.A., M.F.), Department of Rehabilitation and Functional Recovery (E. Sarasso), Neurorehabilitation Unit (M.F.), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy (F.A., R.B., M.F.); Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Maternal Child Health, University of Genoa, Genoa, Italy (E. Sarasso); Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia (T.S., I.S., A.T., V.M., E. Stefanova, V.S.K.); and Neurology Unit, University Hospital Maggiore della Carità, Novara, Italy (F.V.)
| | - Francesca Vignaroli
- From the Neuroimaging Research Unit, Division of Neuroscience (S.B., F.A., E. Sarasso, R.B., M.F.), Neurology Unit (F.A., M.F.), Department of Rehabilitation and Functional Recovery (E. Sarasso), Neurorehabilitation Unit (M.F.), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy (F.A., R.B., M.F.); Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Maternal Child Health, University of Genoa, Genoa, Italy (E. Sarasso); Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia (T.S., I.S., A.T., V.M., E. Stefanova, V.S.K.); and Neurology Unit, University Hospital Maggiore della Carità, Novara, Italy (F.V.)
| | - Elka Stefanova
- From the Neuroimaging Research Unit, Division of Neuroscience (S.B., F.A., E. Sarasso, R.B., M.F.), Neurology Unit (F.A., M.F.), Department of Rehabilitation and Functional Recovery (E. Sarasso), Neurorehabilitation Unit (M.F.), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy (F.A., R.B., M.F.); Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Maternal Child Health, University of Genoa, Genoa, Italy (E. Sarasso); Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia (T.S., I.S., A.T., V.M., E. Stefanova, V.S.K.); and Neurology Unit, University Hospital Maggiore della Carità, Novara, Italy (F.V.)
| | - Vladimir S Kostić
- From the Neuroimaging Research Unit, Division of Neuroscience (S.B., F.A., E. Sarasso, R.B., M.F.), Neurology Unit (F.A., M.F.), Department of Rehabilitation and Functional Recovery (E. Sarasso), Neurorehabilitation Unit (M.F.), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy (F.A., R.B., M.F.); Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Maternal Child Health, University of Genoa, Genoa, Italy (E. Sarasso); Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia (T.S., I.S., A.T., V.M., E. Stefanova, V.S.K.); and Neurology Unit, University Hospital Maggiore della Carità, Novara, Italy (F.V.)
| | - Massimo Filippi
- From the Neuroimaging Research Unit, Division of Neuroscience (S.B., F.A., E. Sarasso, R.B., M.F.), Neurology Unit (F.A., M.F.), Department of Rehabilitation and Functional Recovery (E. Sarasso), Neurorehabilitation Unit (M.F.), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy (F.A., R.B., M.F.); Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Maternal Child Health, University of Genoa, Genoa, Italy (E. Sarasso); Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia (T.S., I.S., A.T., V.M., E. Stefanova, V.S.K.); and Neurology Unit, University Hospital Maggiore della Carità, Novara, Italy (F.V.)
| |
Collapse
|
7
|
Aslam S, Manfredsson F, Stokes A, Shill H. "Advanced" Parkinson's disease: A review. Parkinsonism Relat Disord 2024; 123:106065. [PMID: 38418318 DOI: 10.1016/j.parkreldis.2024.106065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/05/2024] [Accepted: 02/21/2024] [Indexed: 03/01/2024]
Abstract
There is no consensus driven definition of "advanced" Parkinson's disease (APD) currently. APD has been described in terms of emergence of specific clinical features and clinical milestones of the disease e.g., motor fluctuations, time to increasing falls, emergence of cognitive decline, etc. The pathological burden of disease has been used to characterize various stages of the disease. Imaging markers have been associated with various motor and nonmotor symptoms of advancing disease. In this review, we present an overview of clinical, pathologic, and imaging markers of APD. We also propose a model of disease definition involving longitudinal assessments of these markers as well as quality of life metrics to better understand and predict disease progression in those with Parkinson's disease.
Collapse
Affiliation(s)
- Sana Aslam
- Barrow Neurological Institute, Phoenix, AZ, United States.
| | | | - Ashley Stokes
- Barrow Neurological Institute, Phoenix, AZ, United States
| | - Holly Shill
- Barrow Neurological Institute, Phoenix, AZ, United States
| |
Collapse
|
8
|
Wang M, Tan C, Shen Q, Cai S, Liu Q, Liao H. Altered functional-structural coupling may predict Parkinson's patient's depression. Brain Struct Funct 2024; 229:897-907. [PMID: 38478052 DOI: 10.1007/s00429-024-02780-w] [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: 11/21/2023] [Accepted: 02/20/2024] [Indexed: 04/10/2024]
Abstract
We aimed to elucidate the neurobiological basis of depression in Parkinson's disease and identify potential imaging markers for depression in patients with Parkinson's disease. We recruited 43 normal controls (NC), 46 depressed Parkinson's disease patients (DPD) and 56 non-depressed Parkinson's disease (NDPD). All participants underwent routine T2-weighted, T2Flair, and resting-state scans on the same 3.0 T magnetic resonance imaging (MRI) scanner at our hospital. Pre-processing includes calculating surface-based Regional Homogeneity (2DReHo) and cortical thickness. Then we defined the correlation coefficient between 2DReHo and cortical thickness as the functional-structural coupling index. Between-group comparisons were conducted on the Fisher's Z-transformed correlation coefficients. To identify specific regions of decoupling, the 2DReHo for each participant were divided by cortical thickness at each vertex, followed by threshold-free cluster enhancement (TFCE) multiple comparison correction. Binary logistic regression analysis was performed with DPD as the dependent variable, and significantly altered indicators as the independent variables. Receiver operating characteristic curves were constructed to compare the diagnostic performance of individual predictors and combinations using R and MedCalc software. DPD patients exhibited a significantly lower whole-brain functional-structural coupling index than NDPD patients and NC. Abnormal functional-structural coupling was primarily observed in the left inferior parietal lobule and right primary and early visual cortices in DPD patients. Receiver operating characteristic analysis revealed that the combination of cortical functional-structural coupling, surface-based ReHo, and thickness had the best diagnostic performance, achieving a sensitivity of 65% and specificity of 77.7%. This is the first study to explore the relationship between functional and structural changes in DPD patients and evaluate the diagnostic performance of these altered correlations to predict depression in Parkinson's disease patients. We posit that these changes in functional-structural relationships may serve as imaging biomarkers for depression in Parkinson's disease patients, potentially aiding in the classification and diagnosis of Parkinson's disease. Additionally, our findings provide functional and structural imaging evidence for exploring the neurobiological basis of depression in Parkinson's disease.
Collapse
Affiliation(s)
- Min Wang
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Changlian Tan
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qin Shen
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Sainan Cai
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qinru Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Haiyan Liao
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
9
|
Schneider I, Schönfeld R, Hanert A, Philippen S, Tödt I, Granert O, Mehdorn M, Becktepe J, Deuschl G, Berg D, Paschen S, Bartsch T. Deep brain stimulation of the subthalamic nucleus restores spatial reversal learning in patients with Parkinson's disease. Brain Commun 2024; 6:fcae068. [PMID: 38560516 PMCID: PMC10979721 DOI: 10.1093/braincomms/fcae068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/04/2024] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Spatial learning and navigation are supported by distinct memory systems in the human brain such as the hippocampus-based navigational system and the striatum-cortex-based system involved in motor sequence, habit and reversal learning. Here, we studied the role of subthalamic circuits in hippocampus-associated spatial memory and striatal-associated spatial reversal learning formation in patients with Parkinson's disease, who underwent a deep brain stimulation of the subthalamic nucleus. Deep brain stimulation patients (Parkinson's disease-subthalamic nucleus: n = 26) and healthy subjects (n = 15) were tested in a novel experimental spatial memory task based on the Morris water maze that assesses both hippocampal place memory as well as spatial reversal learning. All subjects were trained to navigate to a distinct spatial location hidden within the virtual environment during 16 learning trials in a subthalamic nucleus Stim-On condition. Patients were then randomized into two groups with either a deep brain stimulation On or Off condition. Four hours later, subjects were retested in a delayed recall and reversal learning condition. The reversal learning was realized with a new hidden location that should be memorized during six consecutive trials. The performance was measured by means of an index indicating the improvement during the reversal learning. In the delayed recall condition, neither patients, healthy subjects nor the deep brain stimulation On- versus Off groups showed a difference in place memory performance of the former trained location. In the reversal learning condition, healthy subjects (reversal index 2.0) and patients in the deep brain stimulation On condition (reversal index 1.6) showed a significant improvement. However, patients in the deep brain stimulation Off condition (reversal index 1.1) performed significantly worse and did not improve. There were no differences between all groups in a final visual guided navigation task with a visible target. These results suggest that deep brain stimulation of subthalamic nucleus restores spatial reversal learning in a virtual navigation task in patients with Parkinson's disease and gives insight into the neuromodulation effects on cognition of subthalamic circuits in Parkinson's disease.
Collapse
Affiliation(s)
- Isabel Schneider
- Memory Disorders and Plasticity Group, Department of Neurology, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Robby Schönfeld
- Institute of Psychology, Martin-Luther-University Halle-Wittenberg, Halle 06108, Germany
| | - Annika Hanert
- Memory Disorders and Plasticity Group, Department of Neurology, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Sarah Philippen
- Memory Disorders and Plasticity Group, Department of Neurology, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Inken Tödt
- Memory Disorders and Plasticity Group, Department of Neurology, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Oliver Granert
- Memory Disorders and Plasticity Group, Department of Neurology, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Maximilian Mehdorn
- Department of Neurosurgery, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Jos Becktepe
- Memory Disorders and Plasticity Group, Department of Neurology, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Günther Deuschl
- Memory Disorders and Plasticity Group, Department of Neurology, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Daniela Berg
- Memory Disorders and Plasticity Group, Department of Neurology, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Steffen Paschen
- Memory Disorders and Plasticity Group, Department of Neurology, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Thorsten Bartsch
- Memory Disorders and Plasticity Group, Department of Neurology, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| |
Collapse
|
10
|
Citro S, Lazzaro GD, Cimmino AT, Giuffrè GM, Marra C, Calabresi P. A multiple hits hypothesis for memory dysfunction in Parkinson disease. Nat Rev Neurol 2024; 20:50-61. [PMID: 38052985 DOI: 10.1038/s41582-023-00905-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2023] [Indexed: 12/07/2023]
Abstract
Cognitive disorders are increasingly recognized in Parkinson disease (PD), even in early disease stages, and memory is one of the most affected cognitive domains. Classically, hippocampal cholinergic system dysfunction was associated with memory disorders, whereas nigrostriatal dopaminergic system impairment was considered responsible for executive deficits. Evidence from PD studies now supports involvement of the amygdala, which modulates emotional attribution to experiences. Here, we propose a tripartite model including the hippocampus, striatum and amygdala as key structures for cognitive disorders in PD. First, the anatomo-functional relationships of these structures are explored and experimental evidence supporting their role in cognitive dysfunction in PD is summarized. We then discuss the potential role of α-synuclein, a pathological hallmark of PD, in the tripartite memory system as a key mechanism in the pathogenesis of memory disorders in the disease.
Collapse
Affiliation(s)
- Salvatore Citro
- Neurology Section, Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giulia Di Lazzaro
- Neurology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Angelo Tiziano Cimmino
- Neurology Section, Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Guido Maria Giuffrè
- Neurology Section, Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Neurology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Camillo Marra
- Neurology Section, Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Neurology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Paolo Calabresi
- Neurology Section, Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.
- Neurology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
11
|
Jellinger KA. Pathobiology of Cognitive Impairment in Parkinson Disease: Challenges and Outlooks. Int J Mol Sci 2023; 25:498. [PMID: 38203667 PMCID: PMC10778722 DOI: 10.3390/ijms25010498] [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: 11/23/2023] [Revised: 12/11/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Cognitive impairment (CI) is a characteristic non-motor feature of Parkinson disease (PD) that poses a severe burden on the patients and caregivers, yet relatively little is known about its pathobiology. Cognitive deficits are evident throughout the course of PD, with around 25% of subtle cognitive decline and mild CI (MCI) at the time of diagnosis and up to 83% of patients developing dementia after 20 years. The heterogeneity of cognitive phenotypes suggests that a common neuropathological process, characterized by progressive degeneration of the dopaminergic striatonigral system and of many other neuronal systems, results not only in structural deficits but also extensive changes of functional neuronal network activities and neurotransmitter dysfunctions. Modern neuroimaging studies revealed multilocular cortical and subcortical atrophies and alterations in intrinsic neuronal connectivities. The decreased functional connectivity (FC) of the default mode network (DMN) in the bilateral prefrontal cortex is affected already before the development of clinical CI and in the absence of structural changes. Longitudinal cognitive decline is associated with frontostriatal and limbic affections, white matter microlesions and changes between multiple functional neuronal networks, including thalamo-insular, frontoparietal and attention networks, the cholinergic forebrain and the noradrenergic system. Superimposed Alzheimer-related (and other concomitant) pathologies due to interactions between α-synuclein, tau-protein and β-amyloid contribute to dementia pathogenesis in both PD and dementia with Lewy bodies (DLB). To further elucidate the interaction of the pathomechanisms responsible for CI in PD, well-designed longitudinal clinico-pathological studies are warranted that are supported by fluid and sophisticated imaging biomarkers as a basis for better early diagnosis and future disease-modifying therapies.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, A-1150 Vienna, Austria
| |
Collapse
|
12
|
Huang X, He Q, Ruan X, Li Y, Kuang Z, Wang M, Guo R, Bu S, Wang Z, Yu S, Chen A, Wei X. Structural connectivity from DTI to predict mild cognitive impairment in de novo Parkinson's disease. Neuroimage Clin 2023; 41:103548. [PMID: 38061176 PMCID: PMC10755095 DOI: 10.1016/j.nicl.2023.103548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 01/01/2024]
Abstract
BACKGROUND Early detection of Parkinson's disease (PD) patients at high risk for mild cognitive impairment (MCI) can help with timely intervention. White matter structural connectivity is considered an early and sensitive indicator of neurodegenerative disease. OBJECTIVES To investigate whether baseline white matter structural connectivity features from diffusion tensor imaging (DTI) of de novo PD patients can help predict PD-MCI conversion at an individual level using machine learning methods. METHODS We included 90 de novo PD patients who underwent DTI and 3D T1-weighted imaging. Elastic net-based feature consensus ranking (ENFCR) was used with 1000 random training sets to select clinical and structural connectivity features. Linear discrimination analysis (LDA), support vector machine (SVM), K-nearest neighbor (KNN) and naïve Bayes (NB) classifiers were trained based on features selected more than 500 times. The area under the ROC curve (AUC), accuracy (ACC), sensitivity (SEN) and specificity (SPE) were used to evaluate model performance. RESULTS A total of 57 PD patients were classified as PD-MCI nonconverters, and 33 PD patients were classified as PD-MCI converters. The models trained with clinical data showed moderate performance (AUC range: 0.62-0.68; ACC range: 0.63-0.77; SEN range: 0.45-0.66; SPE range: 0.64-0.84). Models trained with structural connectivity (AUC range, 0.81-0.84; ACC range, 0.75-0.86; SEN range, 0.77-0.91; SPE range, 0.71-0.88) performed similar to models that were trained with both clinical and structural connectivity data (AUC range, 0.81-0.85; ACC range, 0.74-0.85; SEN range, 0.79-0.91; SPE range, 0.70-0.89). CONCLUSIONS Baseline white matter structural connectivity from DTI is helpful in predicting future MCI conversion in de novo PD patients.
Collapse
Affiliation(s)
- Xiaofei Huang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangdong, China
| | - Qing He
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangdong, China
| | - Xiuhang Ruan
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangdong, China
| | - Yuting Li
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangdong, China; Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), Guangdong, China
| | - Zhanyu Kuang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangdong, China
| | - Mengfan Wang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangdong, China
| | - Riyu Guo
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangdong, China
| | - Shuwen Bu
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangdong, China
| | - Zhaoxiu Wang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangdong, China
| | - Shaode Yu
- School of Information and Communication Engineering, Communication University of China, Beijing, China.
| | - Amei Chen
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangdong, China.
| | - Xinhua Wei
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangdong, China.
| |
Collapse
|
13
|
Labrador-Espinosa MA, Silva-Rodríguez J, Reina-Castillo MI, Mir P, Grothe MJ. Basal Forebrain Atrophy, Cortical Thinning, and Amyloid-β Status in Parkinson's disease-Related Cognitive Decline. Mov Disord 2023; 38:1871-1880. [PMID: 37492892 DOI: 10.1002/mds.29564] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 06/16/2023] [Accepted: 07/05/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND Degeneration of the cortically-projecting cholinergic basal forebrain (cBF) is a well-established pathologic correlate of cognitive decline in Parkinson's disease (PD). In Alzheimer's disease (AD) the effect of cBF degeneration on cognitive decline was found to be mediated by parallel atrophy of denervated cortical areas. OBJECTIVES To examine whether the association between cBF degeneration and cognitive decline in PD is mediated by parallel atrophy of cortical areas and whether these associations depend on the presence of comorbid AD pathology. METHODS We studied 162 de novo PD patients who underwent serial 3 T magnetic resonance imaging scanning (follow-up: 2.33 ± 1.46 years) within the Parkinson's Progression Markers Initiative. cBF volume and regional cortical thickness were automatically calculated using established procedures. Individual slopes of structural brain changes and cognitive decline were estimated using linear-mixed models. Associations between longitudinal cBF degeneration, regional cortical thinning, and cognitive decline were assessed using regression analyses and mediation effects were assessed using nonparametric bootstrap. Complementary analyses assessed the effect of amyloid-β biomarker positivity on these associations. RESULTS After controlling for global brain atrophy, longitudinal cBF degeneration was highly correlated with faster cortical thinning (PFDR < 0.05), and thinning in cBF-associated cortical areas mediated the association between cBF degeneration and cognitive decline (rcBF-MoCA = 0.30, P < 0.001). Interestingly, both longitudinal cBF degeneration and its association with cortical thinning were largely independent of amyloid-β status. CONCLUSIONS cBF degeneration in PD is linked to parallel thinning of cortical target areas, which mediate the effect on cognitive decline. These associations are independent of amyloid-β status, indicating that they reflect proper features of PD pathophysiology. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Miguel A Labrador-Espinosa
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Jesús Silva-Rodríguez
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - María Isabel Reina-Castillo
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Michel J Grothe
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
14
|
Yu Z, Pang H, Yu H, Wu Z, Ding Z, Fan G. Segmental disturbance of white matter microstructure in predicting mild cognitive impairment in idiopathic Parkinson's disease: An individualized study based on automated fiber quantification tractography. Parkinsonism Relat Disord 2023; 115:105802. [PMID: 37734997 DOI: 10.1016/j.parkreldis.2023.105802] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 08/01/2023] [Accepted: 08/05/2023] [Indexed: 09/23/2023]
Abstract
PURPOSE The neurobiological mechanisms and an early identification of MCI in idiopathic Parkinson's disease (IPD) remain unclear. To investigate the abnormalities of types of white matter (WM) fiber tracts segmentally and establish reliable indicator in IPD-MCI. METHODS Forty IPD with normal cognition (IPD-NCI), thirty IPD-MCI, and thirty healthy controls were included. Automated fiber quantification was applied to extract the fractional anisotropy (FA) and mean diffusivity (MD) values at 100 locations along the major fibers. Partial correlation was performed between diffusion values and cognitive performance. Furthermore, machine learning analyses were conducted to determine the imaging biomarker of MCI. Permutation tests were performed to evaluate the pointwise differences under the FWE correction. RESULTS IPD-MCI had similar but more severe and widespread WM degeneration in the association, projection, and commissural fibers compared with IPD-NCI. Meanwhile, IPD-MCI showed distinct degeneration pattern in the association fibers. The FA of the anterior segment of right inferior fronto-occipital fasciculus (IFOF) was positively correlated with MoCA (P < 0.05) and executive function (P < 0.05). The MD of the middle and posterior segment of left superior longitudinal fasciculus (SLF) was negatively correlated with MoCA P < 0.05), executive (P < 0.05), visuospatial function (P < 0.05). Furthermore, the AUC of support vector machine model was 0.80 in the validation dataset. The FA of anterior segment of right IFOF contribute the most. CONCLUSION This study demonstrated that regional tract-specific microstructural degeneration, especially the association fibers, can be used to predict MCI in IPD. Especially, the right IFOF may be a significant imaging biomarker in predicting IPD with MCI.
Collapse
Affiliation(s)
- Ziyang Yu
- School of Medicine, Xiamen University, Xiamen, Fujian Province, China; Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China.
| | - Huize Pang
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China.
| | - Hongmei Yu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China.
| | - Ziqian Wu
- School of Medicine, Xiamen University, Xiamen, Fujian Province, China.
| | - Zhi Ding
- School of Medicine, Xiamen University, Xiamen, Fujian Province, China.
| | - Guoguang Fan
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
15
|
Chen Y, Lyu S, Xiao W, Yi S, Liu P, Liu J. Sleep Traits Causally Affect the Brain Cortical Structure: A Mendelian Randomization Study. Biomedicines 2023; 11:2296. [PMID: 37626792 PMCID: PMC10452307 DOI: 10.3390/biomedicines11082296] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/01/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Background: Brain imaging results in sleep deprived patients showed structural changes in the cerebral cortex; however, the reasons for this phenomenon need to be further explored. Methods: This MR study evaluated causal associations between morningness, ease of getting up, insomnia, long sleep, short sleep, and the cortex structure. Results: At the functional level, morningness increased the surface area (SA) of cuneus with global weighted (beta(b) (95% CI): 32.63 (10.35, 54.90), p = 0.004). Short sleep increased SA of the lateral occipital with global weighted (b (95% CI): 394.37(107.89, 680.85), p = 0.007. Short sleep reduced cortical thickness (TH) of paracentral with global weighted (OR (95% CI): -0.11 (-0.19, -0.03), p = 0.006). Short sleep reduced TH of parahippocampal with global weighted (b (95% CI): -0.25 (-0.42, -0.07), p = 0.006). No pleiotropy was detected. However, none of the Bonferroni-corrected p values of the causal relationship between cortical structure and the five types of sleep traits met the threshold. Conclusions: Our results potentially show evidence of a higher risk association between neuropsychiatric disorders and not only paracentral and parahippocampal brain areas atrophy, but also an increase in the middle temporal zone. Our findings shed light on the associations of cortical structure with the occurrence of five types of sleep traits.
Collapse
Affiliation(s)
- Yanjing Chen
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha 410011, China; (Y.C.); (S.L.); (S.Y.); (P.L.)
| | - Shiyi Lyu
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha 410011, China; (Y.C.); (S.L.); (S.Y.); (P.L.)
| | - Wang Xiao
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha 410011, China;
| | - Sijie Yi
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha 410011, China; (Y.C.); (S.L.); (S.Y.); (P.L.)
| | - Ping Liu
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha 410011, China; (Y.C.); (S.L.); (S.Y.); (P.L.)
| | - Jun Liu
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha 410011, China; (Y.C.); (S.L.); (S.Y.); (P.L.)
- Clinical Research Center for Medical Imaging in Hunan Province, Changsha 410011, China
| |
Collapse
|
16
|
Chu C, Zhang Z, Wang J, Wang L, Shen X, Bai L, Li Z, Dong M, Liu C, Yi G, Zhu X. Evolution of brain network dynamics in early Parkinson's disease with mild cognitive impairment. Cogn Neurodyn 2023; 17:681-694. [PMID: 37265660 PMCID: PMC10229513 DOI: 10.1007/s11571-022-09868-1] [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: 06/27/2022] [Revised: 07/13/2022] [Accepted: 07/26/2022] [Indexed: 11/03/2022] Open
Abstract
How mild cognitive impairment (MCI) is instantiated in dynamically interacting and spatially distributed functional brain networks remains an unexplored mystery in early Parkinson's disease (PD). We applied a machine-learning technology based on personalized sliding-window algorithm to track continuously time-varying and overlapping subnetworks under the functional brain networks calculated form resting state electroencephalogram data within a sample of 33 early PD patients (13 early PD patients with MCI and 20 early PD patients without MCI). We decoded a set of subnetworks that captured surprisingly dynamically varying and integrated interactions among certain brain lobes. We observed that the master expressed subnetworks were particularly transient, and flexibly switching between high and low expression during integration into a dynamic brain network. This transience was particularly salient in a subnetwork predominantly linking temporal-parietal-occipital lobes, which decreases in both expression and flexibility in early PD patients with MCI and expresses their degree of cognitive impairment. Moreover, MCI induced a regularly interrupted, slow evolution of subnetworks in functional brain network dynamics in early PD at the individual level, and the dynamic expression characteristics of subnetworks also reflected the degree of cognitive impairment in patients with early PD. Collectively, these results provide novel and deeper insights regarding MCI-induced abnormal dynamical interaction and large-scale changes in functional brain network of early PD.
Collapse
Affiliation(s)
- Chunguang Chu
- School of Electrical and Information Engineering, Tianjin University, Tianjin, 300072 China
| | - Zhen Zhang
- School of Electrical and Information Engineering, Tianjin University, Tianjin, 300072 China
| | - Jiang Wang
- School of Electrical and Information Engineering, Tianjin University, Tianjin, 300072 China
| | - Liufang Wang
- School of Electrical and Information Engineering, Tianjin University, Tianjin, 300072 China
| | - Xiao Shen
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052 China
| | - Lipeng Bai
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052 China
| | - Zhuo Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052 China
| | - Mengmeng Dong
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052 China
| | - Chen Liu
- School of Electrical and Information Engineering, Tianjin University, Tianjin, 300072 China
| | - Guosheng Yi
- School of Electrical and Information Engineering, Tianjin University, Tianjin, 300072 China
| | - Xiaodong Zhu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052 China
| |
Collapse
|
17
|
Shen H, Song H, Wang S, Su D, Sun Q. NEAT1 enhances MPP + -induced pyroptosis in a cell model of Parkinson's disease via targeting miR-5047/YAF2 signaling. Immun Inflamm Dis 2023; 11:e817. [PMID: 37382256 PMCID: PMC10288836 DOI: 10.1002/iid3.817] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/11/2023] [Accepted: 03/07/2023] [Indexed: 06/30/2023] Open
Abstract
PURPOSE Parkinson's disease (PD) is the second most frequent neurodegenerative disease. The aim of our study is to explore the role and the regulatory mechanism of long noncoding RNA (lncRNA) NEAT1 in MPP+ -induced pyroptosis in a cell model of PD. MATERIALS AND METHODS MPP+ -treated SH-SY5Y cells were used as an in vitro model of dopaminergic neurons for PD. Expression levels of miR-5047 and YAF2 mRNA were determined through qRT-PCR. TUNEL staining was carried out to analyze neuronal apoptosis. Luciferase activity assay was accomplished to analyze the combination of miR-5047 with NEAT1 or YAF2 3'-UTR region. Besides, concentrations of IL-1β and IL-18 in supernatant samples were analyzed by using ELISA assay. Expression level of proteins were examined through Western blot. RESULTS NEAT1 and YAF2 expression were increased, while miR-5047 expression was declined in the SH-SY5Y cells treated with MPP+ . NEAT1 was a positively regulator to SH-SY5Y cells pyroptosis induced by MPP+ . In addition, YAF2 was a downstream target of miR-5047. NEAT1 promoted YAF2 expression via inhibiting miR-5047. Importantly, the promotion of NEAT1 to SH-SY5Y cells pyroptosis induced by MPP+ was rescued by miR-5047 mimic transfection or YAF2 downregulation. CONCLUSION In conclusion, NEAT1 was increased in MPP+ -induced SH-SY5Y cells, and it promoted MPP+ -induced pyroptosis through facilitating YAF2 expression by sponging miR-5047.
Collapse
Affiliation(s)
- Hong Shen
- Department of EncephalopathySecond People's HospitalSuzhou CityJiangsu ProvinceChina
| | - Hui Song
- Department of Neurology, TaiHe HospitalHubei University of MedicineShiyan CityHubei ProvinceChina
| | - Songlin Wang
- Department of Neurology, TaiHe HospitalHubei University of MedicineShiyan CityHubei ProvinceChina
| | - Daojing Su
- Department of Orthopaedic Rehabilitation, TaiHe HospitalHubei University of MedicineShiyan CityHubei ProvinceChina
| | - Qiang Sun
- Department of Neurology, TaiHe HospitalHubei University of MedicineShiyan CityHubei ProvinceChina
| |
Collapse
|
18
|
Pletcher C, Dabbs K, Barzgari A, Pozorski V, Haebig M, Wey S, Krislov S, Theisen F, Okonkwo O, Cary P, Oh J, Illingworth C, Wakely M, Law L, Gallagher CL. Cerebral cortical thickness and cognitive decline in Parkinson's disease. Cereb Cortex Commun 2023; 4:tgac044. [PMID: 36660417 PMCID: PMC9840947 DOI: 10.1093/texcom/tgac044] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/28/2022] [Accepted: 10/05/2022] [Indexed: 01/21/2023] Open
Abstract
In Parkinson's disease (PD), reduced cerebral cortical thickness may reflect network-based degeneration. This study performed cognitive assessment and brain MRI in 30 PD participants and 41 controls at baseline and 18 months later. We hypothesized that cerebral cortical thickness and volume, as well as change in these metrics, would differ between PD participants who remained cognitively stable and those who experienced cognitive decline. Dividing the participant sample into PD-stable, PD-decline, and control-stable groups, we compared mean cortical thickness and volume within segments that comprise the prefrontal cognitive-control, memory, dorsal spatial, and ventral object-based networks at baseline. We then compared the rate of change in cortical thickness and volume between the same groups using a vertex-wise approach. We found that the PD-decline group had lower cortical thickness within all 4 cognitive networks in comparison with controls, as well as lower cortical thickness within the prefrontal and medial temporal networks in comparison with the PD-stable group. The PD-decline group also experienced a greater rate of volume loss in the lateral temporal cortices in comparison with the control group. This study suggests that lower thickness and volume in prefrontal, medial, and lateral temporal regions may portend cognitive decline in PD.
Collapse
Affiliation(s)
- Colleen Pletcher
- William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
- Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Kevin Dabbs
- Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Amy Barzgari
- William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
- Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Vincent Pozorski
- William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
- Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Maureen Haebig
- William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
- Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Sasha Wey
- Medical College of Wisconsin, Milwaukee, WI, United States
| | - Stephanie Krislov
- Institute for Clinical and Translational Research, Madison, WI, United States
| | - Frances Theisen
- Cox Medical Centers, Department of Surgery, Springfield, MO, United States
| | - Ozioma Okonkwo
- William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
- Wisconsin Alzheimer’s Disease Research Center, Madison, WI, United States
| | - Paul Cary
- Wisconsin Alzheimer’s Disease Research Center, Madison, WI, United States
| | - Jennifer Oh
- William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
- Wisconsin Alzheimer’s Disease Research Center, Madison, WI, United States
| | - Chuck Illingworth
- William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
- Wisconsin Alzheimer’s Disease Research Center, Madison, WI, United States
| | - Michael Wakely
- Wisconsin Alzheimer’s Disease Research Center, Madison, WI, United States
| | - Lena Law
- Wisconsin Alzheimer’s Disease Research Center, Madison, WI, United States
| | - Catherine L Gallagher
- William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
- Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Wisconsin Alzheimer’s Disease Research Center, Madison, WI, United States
| |
Collapse
|
19
|
Deng JH, Zhang HW, Liu XL, Deng HZ, Lin F. Morphological changes in Parkinson's disease based on magnetic resonance imaging: A mini-review of subcortical structures segmentation and shape analysis. World J Psychiatry 2022; 12:1356-1366. [PMID: 36579355 PMCID: PMC9791612 DOI: 10.5498/wjp.v12.i12.1356] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/02/2022] [Accepted: 11/22/2022] [Indexed: 12/16/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder caused by the loss of dopaminergic neurons in the substantia nigra, resulting in clinical symptoms, including bradykinesia, resting tremor, rigidity, and postural instability. The pathophysiological changes in PD are inextricably linked to the subcortical structures. Shape analysis is a method for quantifying the volume or surface morphology of structures using magnetic resonance imaging. In this review, we discuss the recent advances in morphological analysis techniques for studying the subcortical structures in PD in vivo. This approach includes available pipelines for volume and shape analysis, focusing on the morphological features of volume and surface area.
Collapse
Affiliation(s)
- Jin-Huan Deng
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People’s Hospital, Shenzhen 518035, Guangdong Province, China
| | - Han-Wen Zhang
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People’s Hospital, Shenzhen 518035, Guangdong Province, China
| | - Xiao-Lei Liu
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People’s Hospital, Shenzhen 518035, Guangdong Province, China
| | - Hua-Zhen Deng
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People’s Hospital, Shenzhen 518035, Guangdong Province, China
| | - Fan Lin
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People’s Hospital, Shenzhen 518035, Guangdong Province, China
| |
Collapse
|
20
|
Wang Y, Ning H, Ren J, Pan C, Yu M, Xue C, Wang X, Zhou G, Chen Y, Liu W. Integrated Clinical Features with Plasma and Multi-modal Neuroimaging Biomarkers to Diagnose Mild Cognitive Impairment in Early Drug-Naive Parkinson's Disease. ACS Chem Neurosci 2022; 13:3523-3533. [PMID: 36417458 DOI: 10.1021/acschemneuro.2c00565] [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: 11/24/2022] Open
Abstract
The pathogenesis of cognitive impairment in Parkinson's disease (PD) patients remains unclear, and there is no ideal diagnostic tool available at present. We assessed integrated clinical features with plasma and multi-modal neuroimaging biomarkers to identify mild cognitive impairment (MCI) in early drug-naive PD patients. 49 early drug-naive PD patients, including 26 with MCI (PD-MCI) and 23 with normal cognition (PD-NC), and 20 controls were recruited. Plasma markers [α-synuclein, beta-amyloid 1-40 (Aβ40), beta-amyloid 1-42 (Aβ42), and phosphorylated Tau 181 (p-Tau181) levels], functional connectivity (FC) of the default mode network, and cortical thickness (CTh) were evaluated to identify PD-MCI. The PD-MCI group had significantly higher plasma p-Tau181 levels and p-Tau181/Aβ42 ratio and lower Aβ42/Aβ40 ratio compared to the PD-NC group. Compared to PD-NC, the PD-MCI group showed increased FC between left posterior cingulate cortex (pCC) and the left parahippocampal gyrus (PHG), and between the right hippocampal formation and the left anterior cingulate and paracingulate gyri, and the right middle temporal gyrus. Additionally, the PD-MCI group had thinner cortex thickness in the right lateral occipital and frontal pole compared to the PD-NC group. The final model combining clinical characteristics and several variables (age, sex, plasma p-Tau181 level, Aβ42/Aβ40 ratio, the right lateral occipital CTh, and the FC value between the left pCC and left PHG) had the highest diagnostic accuracy for PD-MCI (AUC = 0.987, 95% CI 0.903-1.000; p = 0.001 compared to age and sex alone). The combination of clinical features, plasma biomarkers, and multi-modal neuroimaging biomarkers can identify early cognitive decline in PD patients.
Collapse
Affiliation(s)
- Yajie Wang
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Houxu Ning
- Department of Chinese Medicine, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jingru Ren
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chenxi Pan
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Miao Yu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chen Xue
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiao Wang
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Gaiyan Zhou
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yubing Chen
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Weiguo Liu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
21
|
Sampedro F, Puig-Davi A, Martinez-Horta S, Pagonabarraga J, Horta-Barba A, Aracil-Bolaños I, Kulisevsky J. Cortical macro and microstructural correlates of cognitive and neuropsychiatric symptoms in Parkinson's disease. Clin Neurol Neurosurg 2022; 224:107531. [PMID: 36455303 DOI: 10.1016/j.clineuro.2022.107531] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cognitive and neuropsychiatric disturbances in Parkinson's disease are as common and as disabling as its well-known motor symptoms. Even though several neural substrates for these symptoms have been suggested, to which extent these symptoms reflect cortical neurodegeneration in Parkinson's disease remains to be fully elucidated. METHODS In a representative sample of 44 Parkinson's disease patients, the data about the following symptoms was recorded: cognitive performance, apathy, depression and anxiety. Surface-based vertexwise multiple regression analyses were performed to investigate the cortical macro (cortical thinning) and microstructural (increased intracortical diffusivity) correlates of each symptom. A group of 18 healthy controls with similar sociodemographics was also included to assess the disease specificity of the neuroimaging results. RESULTS Compared to healthy controls, Parkinson's disease patients showed significantly increased scores in all the considered non-motor scales (p < 0.01). Within the Parkinson's disease group, increased scores in these scales were associated with cortical macro- and microstructural neurodegeneration (p < 0.05 corrected). Each of the considered non-motor scales was associated with a specific pattern of cortical degeneration. When observing both neuroimaging techniques, intracortical diffusivity revealed similar but extensive patterns of cortical compromise than cortical thickness for each symptom, with the exception of anxiety. CONCLUSIONS Cognitive and neuropsychiatric symptoms in Parkinson's disease reflect cortical degeneration. Increases in intracortical diffusivity were able to detect symptom-specific cortical microstructural damage in the absence of cortical thinning. A better understanding of this association may contribute to characterize the brain circuitry and the neurotransmitter pathways underlying these highly prevalent and debilitating symptoms in Parkinson's disease.
Collapse
Affiliation(s)
- Frederic Sampedro
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain; Radiology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Arnau Puig-Davi
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Saul Martinez-Horta
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Javier Pagonabarraga
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Andrea Horta-Barba
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain; Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain; Faculty of Health Sciences, Universitat Oberta de Catalunya (UOC), Barcelona, Spain
| | - Ignacio Aracil-Bolaños
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Jaime Kulisevsky
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain.
| |
Collapse
|
22
|
Bai X, Guo T, Chen J, Guan X, Zhou C, Wu J, Liu X, Wu H, Wen J, Gu L, Gao T, Xuan M, Huang P, Zhang B, Xu X, Zhang M. Microstructural but not macrostructural cortical degeneration occurs in Parkinson’s disease with mild cognitive impairment. NPJ Parkinsons Dis 2022; 8:151. [DOI: 10.1038/s41531-022-00416-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 10/14/2022] [Indexed: 11/11/2022] Open
Abstract
AbstractThis study aimed to investigate the cortical microstructural/macrostructural degenerative patterns in Parkinson’s disease (PD) patients with mild cognitive impairment (MCI). Overall, 38 PD patients with normal cognition (PD-NC), 38 PD-MCI, and 32 healthy controls (HC) were included. PD-MCI was diagnosed according to the MDS Task Force level II criteria. Cortical microstructural alterations were evaluated with Neurite Orientation Dispersion and Density Imaging. Cortical thickness analyses were derived from T1-weighted imaging using the FreeSurfer software. For cortical microstructural analyses, compared with HC, PD-NC showed lower orientation dispersion index (ODI) in bilateral cingulate and paracingulate gyri, supplementary motor area, right paracentral lobule, and precuneus (PFWE < 0.05); while PD-MCI showed lower ODI in widespread regions covering bilateral frontal, parietal, occipital, and right temporal areas and lower neurite density index in left frontal area, left cingulate, and paracingulate gyri (PFWE < 0.05). Furthermore, compared with PD-NC, PD-MCI showed reduced ODI in right frontal area and bilateral caudate nuclei (voxel P < 0.01 and cluster >100 voxels) and the ODI values were associated with the Montreal Cognitive Assessment scores (r = 0.440, P < 0.001) and the memory performance (r = 0.333, P = 0.004) in the PD patients. However, for cortical thickness analyses, there was no difference in the between-group comparisons. In conclusion, cortical microstructural alterations may precede macrostructural changes in PD-MCI. This study provides insightful evidence for the degenerative patterns in PD-MCI and contributes to our understanding of the latent biological basis of cortical neurite changes for early cognitive impairment in PD.
Collapse
|
23
|
Klostermann F, Wyrobnik M, Boll M, Ehlen F, Tiedt HO. Tracing embodied word production in persons with Parkinson's disease in distinct motor conditions. Sci Rep 2022; 12:16669. [PMID: 36198900 PMCID: PMC9534912 DOI: 10.1038/s41598-022-21106-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 09/22/2022] [Indexed: 11/09/2022] Open
Abstract
Embodied cognition theories posit direct interactions between sensorimotor and mental processing. Various clinical observations have been interpreted in this controversial framework, amongst others, low verb generation in word production tasks performed by persons with Parkinson's disease (PD). If this were the consequence of reduced motor simulation of prevalent action semantics in this word class, reduced PD pathophysiology should result in increased verb production and a general shift of lexical contents towards particular movement-related meanings. 17 persons with PD and bilateral deep brain stimulation (DBS) of the subhtalamic nucleus (STN) and 17 healthy control persons engaged in a semantically unconstrained, phonemic verbal fluency task, the former in both DBS-off and DBS-on states. The analysis referred to the number of words produced, verb use, and the occurrence of different dimensions of movement-related semantics in the lexical output. Persons with PD produced fewer words than controls. In the DBS-off, but not in the DBS-on condition, the proportion of verbs within this reduced output was lower than in controls. Lowered verb production went in parallel with a semantic shift: in persons with PD in the DBS-off, but not the DBS-on condition, the relatedness of produced words to own body-movement was lower than in controls. In persons with PD, DBS induced-changes of the motor condition appear to go along with formal and semantic shifts in word production. The results are compatible with the idea of some impact of motor system states on lexical processing.
Collapse
Affiliation(s)
- Fabian Klostermann
- Department of Neurology, Motor and Cognition Group, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12200, Berlin, Germany. .,Berlin School of Mind and Brain, Humboldt-Universität zu Berlin, Unter den Linden 6, 10099, Berlin, Germany.
| | - Michelle Wyrobnik
- Department of Neurology, Motor and Cognition Group, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12200, Berlin, Germany.,Berlin School of Mind and Brain, Humboldt-Universität zu Berlin, Unter den Linden 6, 10099, Berlin, Germany.,Institute of Psychology, Humboldt-Universität zu Berlin, Rudower Chaussee 18, 12489, Berlin, Germany
| | - Moritz Boll
- Department of Neurology, Motor and Cognition Group, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Felicitas Ehlen
- Department of Neurology, Motor and Cognition Group, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12200, Berlin, Germany.,Department of Psychiatry, Jüdisches Krankenhaus Berlin, Heinz-Galinski-Straße 1, 13347, Berlin, Germany
| | - Hannes Ole Tiedt
- Department of Neurology, Motor and Cognition Group, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12200, Berlin, Germany
| |
Collapse
|
24
|
Imaging the Limbic System in Parkinson's Disease-A Review of Limbic Pathology and Clinical Symptoms. Brain Sci 2022; 12:brainsci12091248. [PMID: 36138984 PMCID: PMC9496800 DOI: 10.3390/brainsci12091248] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/05/2022] [Accepted: 09/13/2022] [Indexed: 01/09/2023] Open
Abstract
The limbic system describes a complex of brain structures central for memory, learning, as well as goal directed and emotional behavior. In addition to pathological studies, recent findings using in vivo structural and functional imaging of the brain pinpoint the vulnerability of limbic structures to neurodegeneration in Parkinson's disease (PD) throughout the disease course. Accordingly, dysfunction of the limbic system is critically related to the symptom complex which characterizes PD, including neuropsychiatric, vegetative, and motor symptoms, and their heterogeneity in patients with PD. The aim of this systematic review was to put the spotlight on neuroimaging of the limbic system in PD and to give an overview of the most important structures affected by the disease, their function, disease related alterations, and corresponding clinical manifestations. PubMed was searched in order to identify the most recent studies that investigate the limbic system in PD with the help of neuroimaging methods. First, PD related neuropathological changes and corresponding clinical symptoms of each limbic system region are reviewed, and, finally, a network integration of the limbic system within the complex of PD pathology is discussed.
Collapse
|
25
|
Wang L, Wu P, Brown P, Zhang W, Liu F, Han Y, Zuo CT, Cheng W, Feng J. Association of Structural Measurements of Brain Reserve With Motor Progression in Patients With Parkinson Disease. Neurology 2022; 99:e977-e988. [PMID: 35667838 PMCID: PMC7613818 DOI: 10.1212/wnl.0000000000200814] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 04/19/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES To investigate the relationship between baseline structural measurements of brain reserve and clinical progression in Parkinson disease (PD). To further provide a possible underlying mechanism for structural measurements of brain reserve in PD, we combined functional and transcriptional data and investigated their relationship with progression-associated patterns derived from structural measurements and longitudinal clinical scores. METHODS This longitudinal study collected data from June 2010 to March 2019 from 2 datasets. The Parkinson's Progression Markers Initiative (PPMI) included controls and patients with newly diagnosed PD from 24 participating sites worldwide. Results were confirmed using data from the Huashan dataset (Shanghai, China), which included controls and patients with PD. Clinical symptoms were assessed with Movement Disorder Society-sponsored revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS) scores and Schwab & England activities of daily living (ADL). Both datasets were followed up to 5 years. Linear mixed-effects (LME) models were performed to examine whether changes in clinical scores over time differed as a function of brain structural measurements at baseline. RESULTS A total of 389 patients with PD (n = 346, age 61.3 ± 10.03, 35% female, PPMI dataset; n = 43, age 59.4 ± 7.3, 38.7% female, Huashan dataset) with T1-MRI and follow-up clinical assessments were included in this study. Results of LME models revealed significant interactions between baseline structural measurements of subcortical regions and time on longitudinal deterioration of clinical scores (MDS-UPDRS Part II, absolute β > 0.27; total MDS-UPDRS scores, absolute β > 1.05; postural instability-gait difficulty (PIGD) score, absolute β > 0.03; Schwab & England ADL, absolute β > 0.59; all p < 0.05, false discovery rate corrected). The interaction of baseline structural measurements of subcortical regions and time on longitudinal deterioration of the PIGD score was replicated using data from Huashan Hospital. Furthermore, the β-coefficients of these interactions recapitulated the spatial distribution of dopaminergic, metabolic, and structural changes between patients with PD and normal controls and the spatial distribution of expression of the α-synuclein gene (SNCA). DISCUSSION Patients with PD with greater brain resources (that is, higher deformation-based morphometry values) had greater compensatory capacity, which was associated with slower rates of clinical progression. This knowledge could be used to stratify and monitor patients for clinical trials.
Collapse
Affiliation(s)
- Linbo Wang
- From the Institute of Science and Technology for Brain-inspired Intelligence (L.W., W.Z., W.C., J.F.), Fudan University; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University) (L.W., W.Z., W.C., J.F.), Ministry of Education; PET Center (P.W., C.-T.Z.), Huashan Hospital, Fudan University, Shanghai, China; Medical Research Council Brain Network Dynamics Unit (P.B.), and Nuffield Department of Clinical Neurosciences (P.B.), John Radcliffe Hospital, University of Oxford, United Kingdom; Department of Neurology (F.L., C.-T.Z.), Huashan Hospital North, Fudan University; Department of Neurology (Y.H.), Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine; Human Phenome Institute (C.-T.Z.), Fudan University; Zhangjiang Fudan International Innovation Center (W.C., J.F.), Shanghai, China; Department of Computer Science (W.C., J.F.), University of Warwick, Coventry, United Kingdom; and Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence (W.C., J.F.), Zhejiang Normal University, Jinhua, China
| | - Ping Wu
- From the Institute of Science and Technology for Brain-inspired Intelligence (L.W., W.Z., W.C., J.F.), Fudan University; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University) (L.W., W.Z., W.C., J.F.), Ministry of Education; PET Center (P.W., C.-T.Z.), Huashan Hospital, Fudan University, Shanghai, China; Medical Research Council Brain Network Dynamics Unit (P.B.), and Nuffield Department of Clinical Neurosciences (P.B.), John Radcliffe Hospital, University of Oxford, United Kingdom; Department of Neurology (F.L., C.-T.Z.), Huashan Hospital North, Fudan University; Department of Neurology (Y.H.), Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine; Human Phenome Institute (C.-T.Z.), Fudan University; Zhangjiang Fudan International Innovation Center (W.C., J.F.), Shanghai, China; Department of Computer Science (W.C., J.F.), University of Warwick, Coventry, United Kingdom; and Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence (W.C., J.F.), Zhejiang Normal University, Jinhua, China
| | - Peter Brown
- From the Institute of Science and Technology for Brain-inspired Intelligence (L.W., W.Z., W.C., J.F.), Fudan University; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University) (L.W., W.Z., W.C., J.F.), Ministry of Education; PET Center (P.W., C.-T.Z.), Huashan Hospital, Fudan University, Shanghai, China; Medical Research Council Brain Network Dynamics Unit (P.B.), and Nuffield Department of Clinical Neurosciences (P.B.), John Radcliffe Hospital, University of Oxford, United Kingdom; Department of Neurology (F.L., C.-T.Z.), Huashan Hospital North, Fudan University; Department of Neurology (Y.H.), Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine; Human Phenome Institute (C.-T.Z.), Fudan University; Zhangjiang Fudan International Innovation Center (W.C., J.F.), Shanghai, China; Department of Computer Science (W.C., J.F.), University of Warwick, Coventry, United Kingdom; and Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence (W.C., J.F.), Zhejiang Normal University, Jinhua, China
| | - Wei Zhang
- From the Institute of Science and Technology for Brain-inspired Intelligence (L.W., W.Z., W.C., J.F.), Fudan University; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University) (L.W., W.Z., W.C., J.F.), Ministry of Education; PET Center (P.W., C.-T.Z.), Huashan Hospital, Fudan University, Shanghai, China; Medical Research Council Brain Network Dynamics Unit (P.B.), and Nuffield Department of Clinical Neurosciences (P.B.), John Radcliffe Hospital, University of Oxford, United Kingdom; Department of Neurology (F.L., C.-T.Z.), Huashan Hospital North, Fudan University; Department of Neurology (Y.H.), Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine; Human Phenome Institute (C.-T.Z.), Fudan University; Zhangjiang Fudan International Innovation Center (W.C., J.F.), Shanghai, China; Department of Computer Science (W.C., J.F.), University of Warwick, Coventry, United Kingdom; and Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence (W.C., J.F.), Zhejiang Normal University, Jinhua, China
| | - Fengtao Liu
- From the Institute of Science and Technology for Brain-inspired Intelligence (L.W., W.Z., W.C., J.F.), Fudan University; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University) (L.W., W.Z., W.C., J.F.), Ministry of Education; PET Center (P.W., C.-T.Z.), Huashan Hospital, Fudan University, Shanghai, China; Medical Research Council Brain Network Dynamics Unit (P.B.), and Nuffield Department of Clinical Neurosciences (P.B.), John Radcliffe Hospital, University of Oxford, United Kingdom; Department of Neurology (F.L., C.-T.Z.), Huashan Hospital North, Fudan University; Department of Neurology (Y.H.), Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine; Human Phenome Institute (C.-T.Z.), Fudan University; Zhangjiang Fudan International Innovation Center (W.C., J.F.), Shanghai, China; Department of Computer Science (W.C., J.F.), University of Warwick, Coventry, United Kingdom; and Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence (W.C., J.F.), Zhejiang Normal University, Jinhua, China
| | - Yan Han
- From the Institute of Science and Technology for Brain-inspired Intelligence (L.W., W.Z., W.C., J.F.), Fudan University; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University) (L.W., W.Z., W.C., J.F.), Ministry of Education; PET Center (P.W., C.-T.Z.), Huashan Hospital, Fudan University, Shanghai, China; Medical Research Council Brain Network Dynamics Unit (P.B.), and Nuffield Department of Clinical Neurosciences (P.B.), John Radcliffe Hospital, University of Oxford, United Kingdom; Department of Neurology (F.L., C.-T.Z.), Huashan Hospital North, Fudan University; Department of Neurology (Y.H.), Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine; Human Phenome Institute (C.-T.Z.), Fudan University; Zhangjiang Fudan International Innovation Center (W.C., J.F.), Shanghai, China; Department of Computer Science (W.C., J.F.), University of Warwick, Coventry, United Kingdom; and Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence (W.C., J.F.), Zhejiang Normal University, Jinhua, China
| | - Chuan-Tao Zuo
- From the Institute of Science and Technology for Brain-inspired Intelligence (L.W., W.Z., W.C., J.F.), Fudan University; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University) (L.W., W.Z., W.C., J.F.), Ministry of Education; PET Center (P.W., C.-T.Z.), Huashan Hospital, Fudan University, Shanghai, China; Medical Research Council Brain Network Dynamics Unit (P.B.), and Nuffield Department of Clinical Neurosciences (P.B.), John Radcliffe Hospital, University of Oxford, United Kingdom; Department of Neurology (F.L., C.-T.Z.), Huashan Hospital North, Fudan University; Department of Neurology (Y.H.), Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine; Human Phenome Institute (C.-T.Z.), Fudan University; Zhangjiang Fudan International Innovation Center (W.C., J.F.), Shanghai, China; Department of Computer Science (W.C., J.F.), University of Warwick, Coventry, United Kingdom; and Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence (W.C., J.F.), Zhejiang Normal University, Jinhua, China
| | - Wei Cheng
- From the Institute of Science and Technology for Brain-inspired Intelligence (L.W., W.Z., W.C., J.F.), Fudan University; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University) (L.W., W.Z., W.C., J.F.), Ministry of Education; PET Center (P.W., C.-T.Z.), Huashan Hospital, Fudan University, Shanghai, China; Medical Research Council Brain Network Dynamics Unit (P.B.), and Nuffield Department of Clinical Neurosciences (P.B.), John Radcliffe Hospital, University of Oxford, United Kingdom; Department of Neurology (F.L., C.-T.Z.), Huashan Hospital North, Fudan University; Department of Neurology (Y.H.), Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine; Human Phenome Institute (C.-T.Z.), Fudan University; Zhangjiang Fudan International Innovation Center (W.C., J.F.), Shanghai, China; Department of Computer Science (W.C., J.F.), University of Warwick, Coventry, United Kingdom; and Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence (W.C., J.F.), Zhejiang Normal University, Jinhua, China
| | - Jianfeng Feng
- From the Institute of Science and Technology for Brain-inspired Intelligence (L.W., W.Z., W.C., J.F.), Fudan University; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University) (L.W., W.Z., W.C., J.F.), Ministry of Education; PET Center (P.W., C.-T.Z.), Huashan Hospital, Fudan University, Shanghai, China; Medical Research Council Brain Network Dynamics Unit (P.B.), and Nuffield Department of Clinical Neurosciences (P.B.), John Radcliffe Hospital, University of Oxford, United Kingdom; Department of Neurology (F.L., C.-T.Z.), Huashan Hospital North, Fudan University; Department of Neurology (Y.H.), Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine; Human Phenome Institute (C.-T.Z.), Fudan University; Zhangjiang Fudan International Innovation Center (W.C., J.F.), Shanghai, China; Department of Computer Science (W.C., J.F.), University of Warwick, Coventry, United Kingdom; and Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence (W.C., J.F.), Zhejiang Normal University, Jinhua, China.
| |
Collapse
|
26
|
Palmas MF, Etzi M, Pisanu A, Camoglio C, Sagheddu C, Santoni M, Manchinu MF, Pala M, Fusco G, De Simone A, Picci L, Mulas G, Spiga S, Scherma M, Fadda P, Pistis M, Simola N, Carboni E, Carta AR. The Intranigral Infusion of Human-Alpha Synuclein Oligomers Induces a Cognitive Impairment in Rats Associated with Changes in Neuronal Firing and Neuroinflammation in the Anterior Cingulate Cortex. Cells 2022; 11:cells11172628. [PMID: 36078036 PMCID: PMC9454687 DOI: 10.3390/cells11172628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/03/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Parkinson’s disease (PD) is a complex pathology causing a plethora of non-motor symptoms besides classical motor impairments, including cognitive disturbances. Recent studies in the PD human brain have reported microgliosis in limbic and neocortical structures, suggesting a role for neuroinflammation in the development of cognitive decline. Yet, the mechanism underlying the cognitive pathology is under investigated, mainly for the lack of a valid preclinical neuropathological model reproducing the disease’s motor and non-motor aspects. Here, we show that the bilateral intracerebral infusion of pre-formed human alpha synuclein oligomers (H-αSynOs) within the substantia nigra pars compacta (SNpc) offers a valid model for studying the cognitive symptoms of PD, which adds to the classical motor aspects previously described in the same model. Indeed, H-αSynOs-infused rats displayed memory deficits in the two-trial recognition task in a Y maze and the novel object recognition (NOR) test performed three months after the oligomer infusion. In the anterior cingulate cortex (ACC) of H-αSynOs-infused rats the in vivo electrophysiological activity was altered and the expression of the neuron-specific immediate early gene (IEG) Npas4 (Neuronal PAS domain protein 4) and the AMPA receptor subunit GluR1 were decreased. The histological analysis of the brain of cognitively impaired rats showed a neuroinflammatory response in cognition-related regions such as the ACC and discrete subareas of the hippocampus, in the absence of any evident neuronal loss, supporting a role of neuroinflammation in cognitive decline. We found an increased GFAP reactivity and the acquisition of a proinflammatory phenotype by microglia, as indicated by the increased levels of microglial Tumor Necrosis Factor alpha (TNF-α) as compared to vehicle-infused rats. Moreover, diffused deposits of phospho-alpha synuclein (p-αSyn) and Lewy neurite-like aggregates were found in the SNpc and striatum, suggesting the spreading of toxic protein within anatomically interconnected areas. Altogether, we present a neuropathological rat model of PD that is relevant for the study of cognitive dysfunction featuring the disease. The intranigral infusion of toxic oligomeric species of alpha-synuclein (α-Syn) induced spreading and neuroinflammation in distant cognition-relevant regions, which may drive the altered neuronal activity underlying cognitive deficits.
Collapse
Affiliation(s)
| | - Michela Etzi
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Augusta Pisanu
- National Research Council, Institute of Neuroscience, 09040 Cagliari, Italy
| | - Chiara Camoglio
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Claudia Sagheddu
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Michele Santoni
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Maria Francesca Manchinu
- Istituto Di Ricerca Genetica e Biomedica Del Consiglio Nazionale Delle Ricerche, 09040 Monserrato, Italy
| | - Mauro Pala
- Istituto Di Ricerca Genetica e Biomedica Del Consiglio Nazionale Delle Ricerche, 09040 Monserrato, Italy
| | - Giuliana Fusco
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Alfonso De Simone
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy
| | - Luca Picci
- Department of Life and Environmental Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Giovanna Mulas
- Department of Life and Environmental Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Saturnino Spiga
- Department of Life and Environmental Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Maria Scherma
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Paola Fadda
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Marco Pistis
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Nicola Simola
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Ezio Carboni
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Anna R. Carta
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
- Correspondence:
| |
Collapse
|
27
|
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
|
28
|
Pieperhoff P, Südmeyer M, Dinkelbach L, Hartmann CJ, Ferrea S, Moldovan AS, Minnerop M, Diaz-Pier S, Schnitzler A, Amunts K. Regional changes of brain structure during progression of idiopathic Parkinson’s disease – a longitudinal study using deformation based morphometry. Cortex 2022; 151:188-210. [DOI: 10.1016/j.cortex.2022.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 02/04/2022] [Accepted: 03/12/2022] [Indexed: 12/14/2022]
|
29
|
Huang R, Gao Y, Chen J, Duan Q, He P, Zhang J, Huang H, Zhang Q, Ma G, Zhang Y, Nie K, Wang L. TGR5 agonist INT-777 alleviates inflammatory neurodegeneration in parkinson’s disease mouse model by modulating mitochondrial dynamics in microglia. Neuroscience 2022; 490:100-119. [DOI: 10.1016/j.neuroscience.2022.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/16/2022] [Accepted: 02/25/2022] [Indexed: 11/24/2022]
|
30
|
Chen J, Zhou L, Jiang C, Chen Z, Zhang L, Zhou H, Kang W, Jiang X, Li Y, Luo N, Yao M, Niu M, Chen S, Zuo XN, Li L, Liu J. Impaired Ocular Tracking and Cortical Atrophy in Idiopathic Rapid Eye Movement Sleep Behavior Disorder. Mov Disord 2022; 37:972-982. [PMID: 35107831 DOI: 10.1002/mds.28931] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Idiopathic rapid eye movement sleep behavior disorder (iRBD) is a prodromal stage of synucleinopathies. Patients with synucleinopathies frequently display eye movement abnormalities. However, whether patients with iRBD have eye movement abnormalities remains unknown. OBJECTIVE The aim of this study was to assess eye movement abnormalities and related gray matter alterations and explore whether such abnormalities can serve as biomarkers to indicate phenoconversion to synucleinopathies in iRBD. METHODS Forty patients with iRBD with early disease progression and 35 healthy control subjects participated in a 15-minute ocular-tracking task that evaluated their control of eye movement abilities. They also underwent clinical assessments for olfactory function, nonmotor symptoms, and autonomic symptoms, all of which are biomarkers to predict phenoconversion to synucleinopathies in iRBD. A subgroup of the participants (20 patients with iRBD and 20 healthy control subjects) also participated in structural magnetic resonance imaging. RESULTS The ocular-tracking ability in patients with iRBD was inferior to that of healthy control subjects in two aspects: pursuit initiation and steady-state tracking. Cortical thinning in the right visual area V4 in patients with iRBD is coupled with impaired pursuit initiation. Furthermore, prolonged pursuit initiation in patients with iRBD exhibits a trend of correlation with olfactory loss, the earliest biomarker that develops prior to other prodromal biomarkers. CONCLUSIONS We found ocular-tracking abnormalities in patients with iRBD even early in their disease progression that have not been reported before. These abnormalities are coupled with atrophy of brain areas involved in the perception of object motion and might indicate phenoconversion to synucleinopathies in iRBD. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jing Chen
- Faculty of Arts and Science, New York University Shanghai, Shanghai, China
- NYU-ECNU Institute of Brain and Cognitive Science at New York University Shanghai, Shanghai, China
- Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai, China
| | - Liche Zhou
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Jiang
- Developmental Population Neuroscience Research Center, IDG/McGovern Institute for Brain Research, State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Zhichun Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lina Zhang
- Department of Biostatistics, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyan Zhou
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenyan Kang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xufeng Jiang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyuan Li
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ningdi Luo
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengsha Yao
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengyue Niu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xi-Nian Zuo
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Li
- Faculty of Arts and Science, New York University Shanghai, Shanghai, China
- NYU-ECNU Institute of Brain and Cognitive Science at New York University Shanghai, Shanghai, China
- Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai, China
| | - Jun Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
31
|
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
|
32
|
Wang L, Zhou C, Cheng W, Rolls ET, Huang P, Ma N, Liu Y, Zhang Y, Guan X, Guo T, Wu J, Gao T, Xuan M, Gu Q, Xu X, Zhang B, Gong W, Du J, Zhang W, Feng J, Zhang M. Dopamine depletion and subcortical dysfunction disrupt cortical synchronization and metastability affecting cognitive function in Parkinson's disease. Hum Brain Mapp 2021; 43:1598-1610. [PMID: 34904766 PMCID: PMC8886656 DOI: 10.1002/hbm.25745] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/28/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is primarily characterized by the loss of dopaminergic cells and atrophy in subcortical regions. However, the impact of these pathological changes on large-scale dynamic integration and segregation of the cortex are not well understood. In this study, we investigated the effect of subcortical dysfunction on cortical dynamics and cognition in PD. Spatiotemporal dynamics of the phase interactions of resting-state blood-oxygen-level-dependent signals in 159 PD patients and 152 normal control (NC) individuals were estimated. The relationships between subcortical atrophy, subcortical-cortical fiber connectivity impairment, cortical synchronization/metastability, and cognitive performance were then assessed. We found that cortical synchronization and metastability in PD patients were significantly decreased. To examine whether this is an effect of dopamine depletion, we investigated 45 PD patients both ON and OFF dopamine replacement therapy, and found that cortical synchronization and metastability are significantly increased in the ON state. The extent of cortical synchronization and metastability in the OFF state reflected cognitive performance and mediates the difference in cognitive performance between the PD and NC groups. Furthermore, both the thalamic volume and thalamocortical fiber connectivity had positive relationships with cortical synchronization and metastability in the dopaminergic OFF state, and mediate the difference in cortical synchronization between the PD and NC groups. In addition, thalamic volume also reflected cognitive performance, and cortical synchronization/metastability mediated the relationship between thalamic volume and cognitive performance in PD patients. Together, these results highlight that subcortical dysfunction and reduced dopamine levels are responsible for decreased cortical synchronization and metastability, further affecting cognitive performance in PD. This might lead to biomarkers being identified that can predict if a patient is at risk of developing dementia.
Collapse
Affiliation(s)
- Linbo Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China.,MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.,Zhangjiang Fudan International Innovation Center, Shanghai, China
| | - Cheng Zhou
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Cheng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China.,MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.,Zhangjiang Fudan International Innovation Center, Shanghai, China.,Department of Computer Science, University of Warwick, Coventry, UK
| | - Edmund T Rolls
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China.,MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.,Zhangjiang Fudan International Innovation Center, Shanghai, China.,Department of Computer Science, University of Warwick, Coventry, UK
| | - Peiyu Huang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ningning Ma
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
| | - Yuchen Liu
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
| | - Yajuan Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
| | - Xiaojun Guan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Guo
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingjing Wu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ting Gao
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Min Xuan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Quanquan Gu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojun Xu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Baorong Zhang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weikang Gong
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
| | - Jingnan Du
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China.,MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.,Zhangjiang Fudan International Innovation Center, Shanghai, China
| | - Wei Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China.,MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.,Zhangjiang Fudan International Innovation Center, Shanghai, China
| | - Jianfeng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China.,MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.,Zhangjiang Fudan International Innovation Center, Shanghai, China.,Department of Computer Science, University of Warwick, Coventry, UK
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
33
|
Chen X, Kong J, Pan J, Huang K, Zhou W, Diao X, Cai J, Zheng J, Yang X, Xie W, Yu H, Li J, Pei L, Dong W, Qin H, Huang J, Lin T. Kidney damage causally affects the brain cortical structure: A Mendelian randomization study. EBioMedicine 2021; 72:103592. [PMID: 34619639 PMCID: PMC8498227 DOI: 10.1016/j.ebiom.2021.103592] [Citation(s) in RCA: 132] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/30/2021] [Accepted: 09/07/2021] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Alterations in the brain cortical structures of patients with chronic kidney disease (CKD) have been reported; however, the cause has not been determined yet. Herein, we used Mendelian randomization (MR) to reveal the causal effect of kidney damage on brain cortical structure. METHODS Genome-wide association studies summary data of estimated glomerular filtration rate (eGFR) in 480,698 participants from the CKDGen Consortium were used to identify genetically predicted eGFR. Data from 567,460 individuals from the CKDGen Consortium were used to assess genetically determined CKD; 302,687 participants from the UK Biobank were used to evaluate genetically predicted albuminuria. Further, data from 51,665 patients from the ENIGMA Consortium were used to assess the relationship between genetic predisposition and reduced eGFR, CKD, and progressive albuminuria with alterations in cortical thickness (TH) or surficial area (SA) of the brain. Magnetic resonance imaging was used to measure the SA and TH globally and in 34 functional regions. Inverse-variance weighted was used as the primary estimate whereas MR Pleiotropy RESidual Sum and Outlier, MR-Egger and weighted median were used to detect heterogeneity and pleiotropy. FINDINGS At the global level, albuminuria decreased TH (β = -0.07 mm, 95% CI: -0.12 mm to -0.02 mm, P = 0.004); at the functional level, albuminuria reduced TH of pars opercularis gyrus without global weighted (β = -0.11 mm, 95% CI: -0.16 mm to -0.07 mm, P = 3.74×10-6). No pleiotropy was detected. INTERPRETATION Kidney damage causally influences the cortex structure which suggests the existence of a kidney-brain axis. FUNDING This study was supported by the Science and Technology Planning Project of Guangdong Province (Grant No. 2020A1515111119 and 2017B020227007), the National Key Research and Development Program of China (Grant No. 2018YFA0902803), the National Natural Science Foundation of China (Grant No. 81825016, 81961128027, 81772719, 81772728), the Key Areas Research and Development Program of Guangdong (Grant No. 2018B010109006), Guangdong Special Support Program (2017TX04R246), Grant KLB09001 from the Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, and Grants from the Guangdong Science and Technology Department (2020B1212060018).
Collapse
Affiliation(s)
- Xiong Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Department of Pediatric Urology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China
| | - Jianqiu Kong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Jiexin Pan
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Kai Huang
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, PR China
| | | | - Xiayao Diao
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Jiahao Cai
- Department of Pediatric Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China
| | - Junjiong Zheng
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Xuefan Yang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Weibin Xie
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Hao Yu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Jiande Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, PR China
| | - Lu Pei
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Wen Dong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Haide Qin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Jian Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China.
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China.
| |
Collapse
|
34
|
Tang C, Zhao X, Wu W, Zhong W, Wu X. An individualized prediction of time to cognitive impairment in Parkinson's disease: A combined multi-predictor study. Neurosci Lett 2021; 762:136149. [PMID: 34352339 DOI: 10.1016/j.neulet.2021.136149] [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: 02/24/2021] [Revised: 05/29/2021] [Accepted: 07/29/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Cognitive impairment (CI) is important for the prognosis of Parkinson's disease (PD). Early prediction whether and when cognitive decline from normal cognition (NC) will occur is crucial for preventing or delaying the progression timely. The current study aimed to provide a personalized risk assessment of CI by using baseline information and establishing a multi-predictor nomogram. METHODS 108 patients with PD were collected from the Parkinson's Progression Markers Initiative (PPMI), of whom 58 had progressed to CI and 50 remained NC during 5-year follow up. Radiomics signatures were obtained by using least absolute shrinkage and selection operator (LASSO) Cox regression algorithm. Clinical factors and laboratory biomarkers were selected by multivariate Cox regression analysis. The combined model of radiomics signatures and clinical risk factors was developed by a multivariate Cox proportional hazard model. A multi-predictor nomogram derived from the combined model was established for individualized estimation of time to progress (TTP) of CI. We analyzed the risk of two subgroups of the combined model by Kaplan-Meier (KM) analysis. RESULTS The combined model showed the best performance with a C-index of 0.988 and 0.926 in the training and validation datasets. KM analysis verified significant TTP of CI (P<0.05) between two subgroups stratified by the cutoff value (-0.058). CONCLUSION The combined model and its multi-predictor nomogram can be used to perfectly and individually predict the TTP of CI for patients with PD. Stratification of PD will benefit its timely clinical intervention and the delay and prevention of CI.
Collapse
Affiliation(s)
- Chunyan Tang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyan Zhao
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Wu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weijia Zhong
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Xiaojia Wu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
35
|
Longitudinal clinical, cognitive, and neuroanatomical changes over 5 years in GBA-positive Parkinson's disease patients. J Neurol 2021; 269:1485-1500. [PMID: 34297177 DOI: 10.1007/s00415-021-10713-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/23/2021] [Accepted: 07/11/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE To study the longitudinal disease course of Parkinson's disease (PD) patients with glucocerebrosidase (GBA) mutation (GBA-positive) compared to PD non-carriers (GBA-negative) along a 5-year follow-up, evaluating changes in clinical and cognitive outcomes, cortical thickness, and gray-matter (GM) volumes. METHODS Ten GBA-positive and 20 GBA-negative PD patients underwent clinical, neuropsychological, and MRI assessments (cortical thickness and subcortical, hippocampal, and amygdala volumes) at study entry and once a year for 5 years. At baseline and at the last visit, each group of patients was compared with 22 age-matched healthy controls. Clinical, cognitive, and MRI features were compared between groups at baseline and over time. RESULTS At baseline, GBA-positive and GBA-negative PD patients had similar clinical and cognitive profiles. Compared to GBA-negative and controls, GBA-positive patients showed cortical thinning of left temporal, parietal, and occipital gyri. Over time, compared to GBA-negative, GBA-positive PD patients progressed significantly in motor and cognitive symptoms, and showed a greater pattern of cortical thinning of posterior regions, and frontal and orbito-frontal cortices. After 5 years, compared to controls, GBA-negative PD patients showed a pattern of cortical thinning similar to that showed by GBA-positive cases at baseline. The two groups of patients showed similar patterns of subcortical, hippocampal, and amygdala volume loss over time. CONCLUSIONS Compared to GBA-negative PD, GBA-positive patients experienced a more rapid motor and cognitive decline together with a greater, earlier and faster cortical thinning. Cortical thickness measures may be a useful tool for monitoring and predicting PD progression in accordance with the genetic background.
Collapse
|
36
|
Bae YJ, Kim JM, Sohn CH, Choi JH, Choi BS, Song YS, Nam Y, Cho SJ, Jeon B, Kim JH. Imaging the Substantia Nigra in Parkinson Disease and Other Parkinsonian Syndromes. Radiology 2021; 300:260-278. [PMID: 34100679 DOI: 10.1148/radiol.2021203341] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Parkinson disease is characterized by dopaminergic cell loss in the substantia nigra of the midbrain. There are various imaging markers for Parkinson disease. Recent advances in MRI have enabled elucidation of the underlying pathophysiologic changes in the nigral structure. This has contributed to accurate and early diagnosis and has improved disease progression monitoring. This article aims to review recent developments in nigral imaging for Parkinson disease and other parkinsonian syndromes, including nigrosome imaging, neuromelanin imaging, quantitative iron mapping, and diffusion-tensor imaging. In particular, this article examines nigrosome imaging using 7-T MRI and 3-T susceptibility-weighted imaging. Finally, this article discusses volumetry and its clinical importance related to symptom manifestation. This review will improve understanding of recent advancements in nigral imaging of Parkinson disease. Published under a CC BY 4.0 license.
Collapse
Affiliation(s)
- Yun Jung Bae
- From the Departments of Radiology (Y.J.B., B.S.C., S.J.C., J.H.K.), Neurology (J.M.K., J.H.C.), and Nuclear Medicine (Y.S.S.), Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173-82 Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-707, Republic of Korea; Departments of Radiology (C.H.S.) and Neurology (B.J.), Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; and Division of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin, Republic of Korea (Y.N.)
| | - Jong-Min Kim
- From the Departments of Radiology (Y.J.B., B.S.C., S.J.C., J.H.K.), Neurology (J.M.K., J.H.C.), and Nuclear Medicine (Y.S.S.), Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173-82 Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-707, Republic of Korea; Departments of Radiology (C.H.S.) and Neurology (B.J.), Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; and Division of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin, Republic of Korea (Y.N.)
| | - Chul-Ho Sohn
- From the Departments of Radiology (Y.J.B., B.S.C., S.J.C., J.H.K.), Neurology (J.M.K., J.H.C.), and Nuclear Medicine (Y.S.S.), Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173-82 Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-707, Republic of Korea; Departments of Radiology (C.H.S.) and Neurology (B.J.), Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; and Division of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin, Republic of Korea (Y.N.)
| | - Ji-Hyun Choi
- From the Departments of Radiology (Y.J.B., B.S.C., S.J.C., J.H.K.), Neurology (J.M.K., J.H.C.), and Nuclear Medicine (Y.S.S.), Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173-82 Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-707, Republic of Korea; Departments of Radiology (C.H.S.) and Neurology (B.J.), Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; and Division of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin, Republic of Korea (Y.N.)
| | - Byung Se Choi
- From the Departments of Radiology (Y.J.B., B.S.C., S.J.C., J.H.K.), Neurology (J.M.K., J.H.C.), and Nuclear Medicine (Y.S.S.), Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173-82 Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-707, Republic of Korea; Departments of Radiology (C.H.S.) and Neurology (B.J.), Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; and Division of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin, Republic of Korea (Y.N.)
| | - Yoo Sung Song
- From the Departments of Radiology (Y.J.B., B.S.C., S.J.C., J.H.K.), Neurology (J.M.K., J.H.C.), and Nuclear Medicine (Y.S.S.), Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173-82 Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-707, Republic of Korea; Departments of Radiology (C.H.S.) and Neurology (B.J.), Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; and Division of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin, Republic of Korea (Y.N.)
| | - Yoonho Nam
- From the Departments of Radiology (Y.J.B., B.S.C., S.J.C., J.H.K.), Neurology (J.M.K., J.H.C.), and Nuclear Medicine (Y.S.S.), Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173-82 Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-707, Republic of Korea; Departments of Radiology (C.H.S.) and Neurology (B.J.), Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; and Division of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin, Republic of Korea (Y.N.)
| | - Se Jin Cho
- From the Departments of Radiology (Y.J.B., B.S.C., S.J.C., J.H.K.), Neurology (J.M.K., J.H.C.), and Nuclear Medicine (Y.S.S.), Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173-82 Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-707, Republic of Korea; Departments of Radiology (C.H.S.) and Neurology (B.J.), Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; and Division of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin, Republic of Korea (Y.N.)
| | - Beomseok Jeon
- From the Departments of Radiology (Y.J.B., B.S.C., S.J.C., J.H.K.), Neurology (J.M.K., J.H.C.), and Nuclear Medicine (Y.S.S.), Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173-82 Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-707, Republic of Korea; Departments of Radiology (C.H.S.) and Neurology (B.J.), Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; and Division of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin, Republic of Korea (Y.N.)
| | - Jae Hyoung Kim
- From the Departments of Radiology (Y.J.B., B.S.C., S.J.C., J.H.K.), Neurology (J.M.K., J.H.C.), and Nuclear Medicine (Y.S.S.), Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173-82 Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-707, Republic of Korea; Departments of Radiology (C.H.S.) and Neurology (B.J.), Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; and Division of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin, Republic of Korea (Y.N.)
| |
Collapse
|
37
|
Pourzinal D, Yang JHJ, Bakker A, McMahon KL, Byrne GJ, Pontone GM, Mari Z, Dissanayaka NN. Hippocampal correlates of episodic memory in Parkinson's disease: A systematic review of magnetic resonance imaging studies. J Neurosci Res 2021; 99:2097-2116. [PMID: 34075634 DOI: 10.1002/jnr.24863] [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: 02/25/2021] [Revised: 05/09/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022]
Abstract
The present review asks whether magnetic resonance imaging (MRI) studies are able to define neural correlates of episodic memory within the hippocampus in Parkinson's disease (PD). Systematic searches were performed in PubMed, Web of Science, Medline, CINAHL, and EMBASE using search terms related to structural and functional MRI (fMRI), the hippocampus, episodic memory, and PD. Risk of bias was assessed for each study using the Newtown-Ottawa Scale. Thirty-nine studies met inclusion criteria; eight fMRI, seven diffusion MRI (dMRI), and 24 structural MRI (14 exploring whole hippocampus and 10 exploring hippocampal subfields). Critical analysis of the literature revealed mixed evidence from functional and dMRI, but stronger evidence from sMRI of the hippocampus as a biomarker for episodic memory impairment in PD. Hippocampal subfield studies most often implicated CA1, CA3/4, and subiculum volume in episodic memory and cognitive decline in PD. Despite differences in imaging methodology, study design, and sample characteristics, MRI studies have helped elucidate an important neural correlate of episodic memory impairment in PD with both clinical and theoretical implications. Natural progression of this work encourages future research on hippocampal subfield function as a potential biomarker of, or therapeutic target for, episodic memory dysfunction in PD.
Collapse
Affiliation(s)
- Dana Pourzinal
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane & Women's Hospital, Brisbane, QLD, Australia
| | - Ji Hyun J Yang
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane & Women's Hospital, Brisbane, QLD, Australia
| | - Arnold Bakker
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Katie L McMahon
- School of Clinical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Gerard J Byrne
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane & Women's Hospital, Brisbane, QLD, Australia.,Mental Health Service, Royal Brisbane & Women's Hospital, Brisbane, QLD, Australia
| | - Gregory M Pontone
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Zoltan Mari
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Nadeeka N Dissanayaka
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane & Women's Hospital, Brisbane, QLD, Australia.,Department of Neurology, Royal Brisbane & Women's Hospital, Brisbane, QLD, Australia.,School of Psychology, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
38
|
Devignes Q, Viard R, Betrouni N, Carey G, Kuchcinski G, Defebvre L, Leentjens AFG, Lopes R, Dujardin K. Posterior Cortical Cognitive Deficits Are Associated With Structural Brain Alterations in Mild Cognitive Impairment in Parkinson's Disease. Front Aging Neurosci 2021; 13:668559. [PMID: 34054507 PMCID: PMC8155279 DOI: 10.3389/fnagi.2021.668559] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 03/22/2021] [Indexed: 12/16/2022] Open
Abstract
Context: Cognitive impairments are common in patients with Parkinson's disease (PD) and are heterogeneous in their presentation. The "dual syndrome hypothesis" suggests the existence of two distinct subtypes of mild cognitive impairment (MCI) in PD: a frontostriatal subtype with predominant attentional and/or executive deficits and a posterior cortical subtype with predominant visuospatial, memory, and/or language deficits. The latter subtype has been associated with a higher risk of developing dementia. Objective: The objective of this study was to identify structural modifications in cortical and subcortical regions associated with each PD-MCI subtype. Methods: One-hundred and fourteen non-demented PD patients underwent a comprehensive neuropsychological assessment as well as a 3T magnetic resonance imaging scan. Patients were categorized as having no cognitive impairment (n = 41) or as having a frontostriatal (n = 16), posterior cortical (n = 25), or a mixed (n = 32) MCI subtype. Cortical regions were analyzed using a surface-based Cortical thickness (CTh) method. In addition, the volumes, shapes, and textures of the caudate nuclei, hippocampi, and thalami were studied. Tractometric analyses were performed on associative and commissural white matter (WM) tracts. Results: There were no between-group differences in volumetric measurements and cortical thickness. Shape analyses revealed more abundant and more extensive deformations fields in the caudate nuclei, hippocampi, and thalami in patients with posterior cortical deficits compared to patients with no cognitive impairment. Decreased fractional anisotropy (FA) and increased mean diffusivity (MD) were also observed in the superior longitudinal fascicle, the inferior fronto-occipital fascicle, the striato-parietal tract, and the anterior and posterior commissural tracts. Texture analyses showed a significant difference in the right hippocampus of patients with a mixed MCI subtype. Conclusion: PD-MCI patients with posterior cortical deficits have more abundant and more extensive structural alterations independently of age, disease duration, and severity, which may explain why they have an increased risk of dementia.
Collapse
Affiliation(s)
- Quentin Devignes
- Lille Neuroscience and Cognition, Lille University, Inserm, Lille University Medical Centre, Lille, France
| | - Romain Viard
- US 41—UMS 2014—PLBS, Lille University, CNRS, Inserm, Lille University Medical Centre, Pasteur Institute, Lille, France
- Department of Neuroradiology, Lille University Medical Centre, Lille, France
| | - Nacim Betrouni
- Lille Neuroscience and Cognition, Lille University, Inserm, Lille University Medical Centre, Lille, France
| | - Guillaume Carey
- Lille Neuroscience and Cognition, Lille University, Inserm, Lille University Medical Centre, Lille, France
- Neurology and Movement Disorders Department, Lille University Medical Centre, Lille, France
| | - Gregory Kuchcinski
- Lille Neuroscience and Cognition, Lille University, Inserm, Lille University Medical Centre, Lille, France
- US 41—UMS 2014—PLBS, Lille University, CNRS, Inserm, Lille University Medical Centre, Pasteur Institute, Lille, France
- Department of Neuroradiology, Lille University Medical Centre, Lille, France
| | - Luc Defebvre
- Lille Neuroscience and Cognition, Lille University, Inserm, Lille University Medical Centre, Lille, France
- Neurology and Movement Disorders Department, Lille University Medical Centre, Lille, France
| | | | - Renaud Lopes
- Lille Neuroscience and Cognition, Lille University, Inserm, Lille University Medical Centre, Lille, France
- US 41—UMS 2014—PLBS, Lille University, CNRS, Inserm, Lille University Medical Centre, Pasteur Institute, Lille, France
- Department of Neuroradiology, Lille University Medical Centre, Lille, France
| | - Kathy Dujardin
- Lille Neuroscience and Cognition, Lille University, Inserm, Lille University Medical Centre, Lille, France
- Neurology and Movement Disorders Department, Lille University Medical Centre, Lille, France
| |
Collapse
|
39
|
Taximaimaiti R, Wang XP. Comparing the Clinical and Neuropsychological Characteristics of Parkinson's Disease With and Without Freezing of Gait. Front Neurosci 2021; 15:660340. [PMID: 33986641 PMCID: PMC8110824 DOI: 10.3389/fnins.2021.660340] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/23/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction Freezing of gait (FOG) is one of the most common walking problems in Parkinson’s disease (PD). Impaired cognitive function is believed to play an important role in developing and aggravating FOG in PD. But some evidence suggests that motor function discrepancy may affect testing results. Therefore, we think it is necessary for PD-FOG(+) and PD-FOG(−) patients to complete neuropsychological tests under similar motor conditions. Methods This study recruited 44 idiopathic PD patients [PD-FOG(+) n = 22, PD-FOG(−) n = 22] and 20 age-matched healthy controls (HC). PD-FOG(+) and PD-FOG(−) patients were matched for age, year of education, and Hoehn and Yahr score (H&Y). All participants underwent a comprehensive battery of neuropsychological assessment, and demographical and clinical information was also collected. Results PD patients showed poorer cognitive function, higher risks of depression and anxiety, and more neuropsychiatric symptoms compared with HC. When controlling for age, years of education, and H&Y, there were no statistical differences in cognitive function between PD-FOG(+) and PD-FOG(−) patients. But PD-FOG(+) patients had worse motor and non-motor symptoms than PD-FOG(−) patients. PD patients whose motor symptoms initiated with rigidity and initiated unilaterally were more likely to experience FOG. Conclusion Traditional neuropsychological testing may not be sensitive enough to detect cognitive impairment in PD. Motor symptoms initiated with rigidity and initiated unilaterally might be an important predictor of FOG.
Collapse
Affiliation(s)
- Reyisha Taximaimaiti
- Department of Neurology, Shanghai TongRen Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Ping Wang
- Department of Neurology, Shanghai TongRen Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
40
|
A selective NLRP3 inflammasome inhibitor attenuates behavioral deficits and neuroinflammation in a mouse model of Parkinson's disease. J Neuroimmunol 2021; 354:577543. [PMID: 33714750 DOI: 10.1016/j.jneuroim.2021.577543] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 12/11/2022]
Abstract
Nod-like receptor pyrin containing (NLRP)3 inflammasome-mediated neuroinflammation is involved in the pathology of Parkinson's disease (PD), but the roles of other inflammasomes in PD remain unclear. The NLRP3 inhibitor MCC950 exerts neuroprotective effects in several neurological diseases. Using a 1-methyl-4-phenyl-1,2,3,6-tetrahydro pyridine (MPTP)-induced mouse model with or without intraperitoneal MCC950 administration, we assessed whether specifically the NLRP3 inflammasome is activated in the nigrostriatal system and whether MCC950 has therapeutic potential in this PD model. Western blots were used to determine the nigrostriatal expression of inflammasome-specific proteins, including NLRP1, NLRP2, NLRP3, nod-like receptor CARD containing 4 (NLRC4), and absent in melanoma 2 (AIM2). The pole, hanging, and swimming tests were used to assess functional deficits, western blots and immunostainings were used to analyze dopaminergic neuronal degeneration, as well as activation of glial cells and the NLRP3 inflammasome. NLRP3 expression in the nigrostriatal system of MPTP-induced mice was significantly increased compared to control, whereas NLRP1, NLRP2, NLRC4, and AIM2 expression in the nigrostriatal system, as well as NLRP3 expression in the cerebral cortex and hippocampus, were similar in the two groups. Furthermore, MPTP-induced mice exhibited behavioral dysfunctions, dopaminergic neuronal degeneration, and activation of glial cells and the NLRP3 inflammasome. MCC950 treatment of MPTP-induced mice improved behavioral dysfunctions, reduced dopaminergic neuronal degeneration, and inhibited the activation of glial cells and the NLRP3 inflammasome. In conclusion, these findings indicated that NLRP3, not NLRP1, NLRP2, NLRC4, and AIM2, may be the key inflammasome that promotes MPTP-induced pathogenesis. MCC950 protects against MPTP-induced nigrostriatal damage and may be a novel promising therapeutic approach in treating MPTP-induced PD.
Collapse
|
41
|
Ruppert MC, Greuel A, Freigang J, Tahmasian M, Maier F, Hammes J, van Eimeren T, Timmermann L, Tittgemeyer M, Drzezga A, Eggers C. The default mode network and cognition in Parkinson's disease: A multimodal resting-state network approach. Hum Brain Mapp 2021; 42:2623-2641. [PMID: 33638213 PMCID: PMC8090788 DOI: 10.1002/hbm.25393] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/12/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022] Open
Abstract
Involvement of the default mode network (DMN) in cognitive symptoms of Parkinson's disease (PD) has been reported by resting-state functional MRI (rsfMRI) studies. However, the relation to metabolic measures obtained by [18F]-fluorodeoxyglucose positron emission tomography (FDG-PET) is largely unknown. We applied multimodal resting-state network analysis to clarify the association between intrinsic metabolic and functional connectivity abnormalities within the DMN and their significance for cognitive symptoms in PD. PD patients were classified into normal cognition (n = 36) and mild cognitive impairment (MCI; n = 12). The DMN was identified by applying an independent component analysis to FDG-PET and rsfMRI data of a matched subset (16 controls and 16 PD patients) of the total cohort. Besides metabolic activity, metabolic and functional connectivity within the DMN were compared between the patients' groups and healthy controls (n = 16). Glucose metabolism was significantly reduced in all DMN nodes in both patient groups compared to controls, with the lowest uptake in PD-MCI (p < .05). Increased metabolic and functional connectivity along fronto-parietal connections was identified in PD-MCI patients compared to controls and unimpaired patients. Functional connectivity negatively correlated with cognitive composite z-scores in patients (r = -.43, p = .005). The current study clarifies the commonalities of metabolic and hemodynamic measures of brain network activity and their individual significance for cognitive symptoms in PD, highlighting the added value of multimodal resting-state network approaches for identifying prospective biomarkers.
Collapse
Affiliation(s)
- Marina C Ruppert
- Department of Neurology, University Hospital of Marburg, Marburg, Germany.,Center for Mind, Brain, and Behavior-CMBB, Universities of Marburg and Gießen, Marburg, Germany
| | - Andrea Greuel
- Department of Neurology, University Hospital of Marburg, Marburg, Germany
| | - Julia Freigang
- Department of Neurology, University Hospital of Marburg, Marburg, Germany.,Center for Mind, Brain, and Behavior-CMBB, Universities of Marburg and Gießen, Marburg, Germany
| | - Masoud Tahmasian
- Institute of Medical Science and Technology, Shahid Beheshti University, Tehran, Iran
| | - Franziska Maier
- Medical Faculty, Department of Psychiatry, University Hospital Cologne, Cologne, Germany
| | - Jochen Hammes
- Multimodal Neuroimaging Group, Department of Nuclear Medicine, Medical Faculty and University Hospital Cologne, University Hospital Cologne, Cologne, Germany
| | - Thilo van Eimeren
- Multimodal Neuroimaging Group, Department of Nuclear Medicine, Medical Faculty and University Hospital Cologne, University Hospital Cologne, Cologne, Germany.,Department of Neurology, Medical Faculty and University Hospital Cologne, University Hospital Cologne, Cologne, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Lars Timmermann
- Department of Neurology, University Hospital of Marburg, Marburg, Germany.,Center for Mind, Brain, and Behavior-CMBB, Universities of Marburg and Gießen, Marburg, Germany
| | - Marc Tittgemeyer
- Max Planck Institute for Metabolism Research, Cologne, Germany.,Cluster of Excellence in Cellular Stress and Aging Associated Disease (CECAD), Cologne, Germany
| | - Alexander Drzezga
- Multimodal Neuroimaging Group, Department of Nuclear Medicine, Medical Faculty and University Hospital Cologne, University Hospital Cologne, Cologne, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-2), Jülich, Germany
| | - Carsten Eggers
- Department of Neurology, University Hospital of Marburg, Marburg, Germany.,Center for Mind, Brain, and Behavior-CMBB, Universities of Marburg and Gießen, Marburg, Germany
| |
Collapse
|