1
|
Barthelemy OJ, Shirey AJ, Anakwe S, Neargarder S, DeGutis J, Cronin-Golomb A. Positive Associations between the Personality Trait of Openness and Verbal Learning and Memory in Individuals with Parkinson's Disease: A Pilot Study. Arch Clin Neuropsychol 2025:acaf044. [PMID: 40420367 DOI: 10.1093/arclin/acaf044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/28/2025] Open
Abstract
OBJECTIVE Individuals with idiopathic Parkinson's disease (PD) often experience difficulties with verbal learning and memory, even in the absence of dementia. Higher levels of the personality trait of openness predict better learning and memory in other older adult populations, but openness's contributions in PD are unknown. Lower openness and alterations in openness's neural substrates in PD suggest that openness may have strong associations with memory in PD. METHOD We used the Big Five Inventory-2 (BFI-2) personality self-rating questionnaire and the Rey Auditory Verbal Learning Test (RAVLT) in a cross-sectional sample of 33 persons with PD (PwPD; 17 men), 26 healthy older adults (OA; 14 men), and 37 healthy younger adults (YA; 19 men). Correlation analysis examined relations between openness (BFI-2 open-mindedness) and verbal learning and memory (RAVLT performances). Correlation and regression analysis controlled for psychosocial and cognitive factors and examined possible moderators and mediators. RESULTS Significant, positive correlations between openness and RAVLT scores occurred in PwPD but not in OA or YA. Among PwPD, openness independently predicted most RAVLT scores in regression models. Its associations were not explained by PD duration, disease severity, disease stage, or sex. PwPD low in openness performed worse than OA. Among OA, older age predicted significantly more positive association between openness and memory. CONCLUSIONS Openness is positively associated with verbal memory in PwPD, as well as in healthy older adults (depending on age), with implications for the relevance of personality factors in cognition.
Collapse
Affiliation(s)
- Olivier J Barthelemy
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
| | - Alexandria J Shirey
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
| | - Stephanie Anakwe
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
| | - Sandy Neargarder
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
- Department of Psychology, Bridgewater State University, Bridgewater, MA, USA
| | - Joseph DeGutis
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
- Boston Attention and Learning Laboratory, Boston Division VA Healthcare System, Boston, MA, USA
- Geriatric Research Education and Clinical Center (GRECC), Boston Division VA Healthcare System, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Alice Cronin-Golomb
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
| |
Collapse
|
2
|
Silva-Rodríguez J, Labrador-Espinosa MÁ, Castro-Labrador S, Muñoz-Delgado L, Franco-Rosado P, Castellano-Guerrero AM, Macías-García D, Jesús S, Adarmes-Gómez AD, Carrillo F, Martín-Rodríguez JF, García-Solís D, Roldán-Lora F, Mir P, Grothe MJ. Imaging biomarkers of cortical neurodegeneration underlying cognitive impairment in Parkinson's disease. Eur J Nucl Med Mol Imaging 2025; 52:2002-2014. [PMID: 39888421 PMCID: PMC12014801 DOI: 10.1007/s00259-025-07070-z] [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: 06/13/2024] [Accepted: 12/30/2024] [Indexed: 02/01/2025]
Abstract
PURPOSE Imaging biomarkers bear great promise for improving the diagnosis and prognosis of cognitive impairment in Parkinson's disease (PD). We compared the ability of three commonly used neuroimaging modalities to detect cortical changes in PD patients with mild cognitive impairment (PD-MCI) and dementia (PDD). METHODS 53 cognitively normal PD patients (PD-CN), 32 PD-MCI, and 35 PDD underwent concurrent structural MRI (sMRI), diffusion-weighted MRI (dMRI), and [18F]FDG PET. We extracted grey matter volumes (sMRI), mean diffusivity (MD, dMRI), and standardized uptake value ratios ([18F]FDG PET) for 52 cortical regions included in a neuroanatomical atlas. We assessed group differences using ANCOVA models and further applied a cross-validated machine learning approach to identify the modality-specific brain regions that are most indicative of dementia status and assessed their diagnostic accuracy for group separation using receiver operating characteristic analyses. RESULTS In sMRI, atrophy of temporal and posterior-parietal areas allowed separating PDD from PD-CN (AUC = 0.77 ± 0.07), but diagnostic accuracy was poor for separating PD-MCI from PD-CN (0.57 ± 0.10). dMRI showed most pronounced diffusivity changes in the medial temporal lobe, which provided excellent diagnostic performance for PDD (AUC = 0.87 ± 0.06), and a more modest but still significant performance for PD-MCI (AUC = 0.71 ± 0.09). Finally, [18F]FDG PET revealed pronounced hypometabolism in posterior-occipital regions, which provided the highest diagnostic accuracies for both PDD (AUC = 0.89 ± 0.05) and PD-MCI (AUC = 0.78 ± 0.05). In statistical comparisons, both [18F]FDG PET (p < 0.001) and dMRI (p < 0.031) outperformed sMRI for detecting PDD and PD-MCI. CONCLUSION Among the tested modalities, [18F]FDG PET was most accurate for detecting cortical changes associated with cognitive impairment in PD, especially at early stages. Diffusion measurements may represent a promising MRI-based alternative.
Collapse
Affiliation(s)
- Jesús Silva-Rodríguez
- Reina Sofia Alzheimer Center, CIEN Foundation, ISCIII, Madrid, Spain
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Miguel Ángel Labrador-Espinosa
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
- Department of Psychiatry and Neurochemistry, Institute of Physiology and Neuroscience, University of Gothenburg, Gothenburg, Sweden
| | - Sandra Castro-Labrador
- Reina Sofia Alzheimer Center, CIEN Foundation, ISCIII, Madrid, Spain
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Laura Muñoz-Delgado
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Pablo Franco-Rosado
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Ana María Castellano-Guerrero
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Daniel Macías-García
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Silvia Jesús
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Astrid D Adarmes-Gómez
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Fátima Carrillo
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Juan Francisco Martín-Rodríguez
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
- Departamento de Psicología Experimental, Facultad de Psicología, Universidad de Sevilla, Sevilla, Spain
| | - David García-Solís
- Servicio de Medicina Nuclear, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Florinda Roldán-Lora
- Unidad de Radiodiagnóstico, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain.
- Unidad de Trastornos del Movimiento, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío, Avda. Manuel Siurot s/n, Seville, 41013, Spain.
| | - Michel J Grothe
- Reina Sofia Alzheimer Center, CIEN Foundation, ISCIII, Madrid, Spain.
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain.
- Fundación CIEN, Centro Alzheimer Reina Sofía, C. de Valderrebollo, 5, Vallecas, Madrid, 28031, Spain.
| |
Collapse
|
3
|
Yang L, Wang Y. New method for diffusion-weighted images denoising based on patch-matching with higher-order singular value decomposition. JOURNAL OF X-RAY SCIENCE AND TECHNOLOGY 2025; 33:526-539. [PMID: 40343883 DOI: 10.1177/08953996241313321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
BackgroundDiffusion-weighted imaging (DWI) is an important technique to study brain microstructure. However, diffusion-weighted (DW) images suffer from severe low signal-to-noise ratio (SNR) problem, affecting subsequent diffusion analysis.ObjectiveThe goal of this paper is to develop advanced DWI denoising technique to effectively reduce noise while improving the accuracy and reliability of subsequent diffusion model fitting and diffusion analysis, thereby facilitating the research and analysis of brain science.MethodsWe propose a new method for denoising DW images based on patch-matching with higher-order singular value decomposition (HOSVD) by combined with the variance-stabilizing transformation technique. It starts with introducing a novel non-local mean algorithm as a prefiltering stage, and then denoises the noisy data using a local HOSVD algorithm based on the HOSVD bases learned from prefiltered images.ResultsExperiments are performed on simulation, HCP and in vivo brain DWI datasets. Results show that the proposed method significantly reduces spatially invariant and variant noise, improving the most reliable diffusion analysis compared with the different denoising methods.ConclusionsThe proposed method achieves state-of-the-art performance which can improve image quality and enable accurate diffusion analysis.
Collapse
Affiliation(s)
- Liming Yang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yuanjun Wang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
4
|
Pan L, Yang L, Ding W, Hu Y, Yang W, Wang J, Zhang Z, Fan K, Sun Z, Liang Y, Lin X, Chen J, Zhang Y. Integrated genetic analysis and single cell-RNA sequencing for brain image-derived phenotypes and Parkinson's disease. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111317. [PMID: 40081564 DOI: 10.1016/j.pnpbp.2025.111317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Previous studies have reported Parkinson's disease (PD) patients usually have changes in brain image-derived phenotypes (IDPs). However, the role of genetic factors in their association and biological mechanism remains unclear. We aimed to unveil genetic and biological links between brain IDPs and PD. METHODS Using genome-wide association study (GWAS) summary statistics and single-cell RNA sequencing (scRNA-seq) data, we performed a comprehensive analysis between 624 brain IDPs and PD. The genetic correlations and causality were examined by linkage disequilibrium score regression (LDSC), two-sample bidirectional Mendelian randomization (MR) and meta-analysis. Potential shared genes were identified using MAGMA and PLACO. Finally, pathway enrichment using FUMA and Metascape, and scRNA-seq analysis were performed to determine biological mechanisms and gene expression atlas across various cell types in brain tissue. RESULTS LDSC revealed that 50 brain IDPs were genetically correlated with PD (P < 0.05), in which 5 IDPs, exhibited putative causality on PD through MR (P < 0.05). For instance, we identified that the increased volume of the right thalamus (IVW: OR = 2.08, 95 % CI: 1.33 to 3.25, PFDR = 0.03) was positively correlated with the risk of PD, which was also supported by replicated MR (IVW: OR = 1.63, 95 % CI: 1.17-2.26, PFDR = 0.02) in FinnGen and meta-analysis (OR = 1.78, 95 % CI: 1.36-2.31, PFDR = 5.00 × 10-4). Additionally, we identified 56 unique pleiotropic genes, such as FAM13A, with notable enrichment in neuronal cells. Biological mechanism analysis revealed these genes were enriched in brain tissues and a variety of pathways such as negative regulation of neuron apoptotic processes. CONCLUSION We indicated the shared genetic architecture and biological mechanisms between brain IDPs and PD. These findings might provide insights on the therapeutic intervention and early prediction of PD at the brain imaging level.
Collapse
Affiliation(s)
- Lin Pan
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China
| | - Laiyu Yang
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Weijie Ding
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China
| | - Yongfei Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Dongfeng Road East 651, Guangzhou 510060, China
| | - Wenzhuo Yang
- Department of Neurosurgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jingning Wang
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China
| | - Zhiyun Zhang
- Department of Plastic Surgery, The First Hospital of Jilin University, Changchun 130000, China
| | - Kangli Fan
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China
| | - Zhihui Sun
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China
| | - Yue Liang
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China
| | - Xiaoyue Lin
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China
| | - Jun Chen
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China.
| | - Ying Zhang
- Department of Neurology, Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
5
|
Tong S, Wang R, Li H, Tong Z, Geng D, Zhang X, Ren C. Executive dysfunction in Parkinson's disease: From neurochemistry to circuits, genetics and neuroimaging. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111272. [PMID: 39880275 DOI: 10.1016/j.pnpbp.2025.111272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/18/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
Cognitive decline is one of the most significant non-motor symptoms of Parkinson's disease (PD), with executive dysfunction (EDF) being the most prominent characteristic of PD-associated cognitive deficits. Currently, lack of uniformity in the conceptualization and assessment scales for executive functions impedes the early and accurate diagnosis of EDF in PD. The neurobiological mechanisms of EDF in PD remain poorly understood. Moreover, the treatment of cognitive impairment in PD has progressed slowly and with limited efficacy. Thus, this review explores the characteristics and potential mechanisms of EDF in PD from multiple perspectives, including the concept of executive function, commonly used neuropsychological tests, neurobiochemistry, genetics, electroencephalographic activity and neuroimaging. The available evidence indicates that degeneration of the frontal-striatal circuit, along with mutations in the Catechol-O-methyltransferase (COMT) gene and Leucine-rich repeat kinase 2 (LRRK2) gene, may contribute to EDF in patients with PD. The increase in theta and delta waves, along with the decrease in alpha waves, offers potential biomarkers for the early identification and monitoring of EDF, as well as the development of dementia in patients with PD. The PD cognition-related pattern (PDCP) pattern may serve as a tool for monitoring and assessing cognitive function progression in these patients and is anticipated to become a biomarker for cognitive disorders associated with PD. The aim is to provide new insights for the early and precise diagnosis and treatment of EDF in PD.
Collapse
Affiliation(s)
- Shuyan Tong
- Department of Geriatric Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China; Department of Neurology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ruiwen Wang
- Department of Anesthesiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Huihua Li
- Department of Psychiatry, Zhenjiang Mental Health Center, Zhenjiang, Jiangsu, China
| | - Zhu Tong
- The Affiliated Xuzhou Oriental Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiangrong Zhang
- Department of Geriatric Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China; The Affiliated Xuzhou Oriental Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Chao Ren
- Department of Neurology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China.
| |
Collapse
|
6
|
Laurell AAS, Mak E, O'Brien JT. A systematic review of diffusion tensor imaging and tractography in dementia with Lewy bodies and Parkinson's disease dementia. Neurosci Biobehav Rev 2025; 169:106007. [PMID: 39793681 DOI: 10.1016/j.neubiorev.2025.106007] [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: 08/21/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
We reviewed studies using diffusion tensor imaging (DTI) and tractography to characterise white matter changes in Dementia with Lewy Bodies (DLB) and Parkinson's Disease Dementia (PDD). The search included MEDLINE and EMBASE, and we used a narrative strategy to synthesise the evidence. Data was extracted from 57 studies, of which the majority were considered 'good quality'. Subjects with DLB and PDD had widespread white matter changes compared to healthy controls and Parkinson's disease without cognitive impairment, with a relative sparing of the hippocampus. Compared to subjects with Alzheimer's disease (AD), DLB had greater changes in thalamic connectivity and in the nigroputaminal tract, while AD had greater changes in the parahippocampal white matter and fornix. Cognition was associated with widespread white matter changes, visual hallucinations with thalamic and cholinergic connectivity, and parkinsonism with changes in structures involved in motor control. DTI and tractography may therefore be well suited for discriminating DLB and PDD from other types of dementia, and for studying the aetiology of common symptoms.
Collapse
Affiliation(s)
- Axel A S Laurell
- Department of Psychiatry, University of Cambridge, Level E4, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2QQ, United Kingdom.
| | - Elijah Mak
- Department of Psychiatry, University of Cambridge, Level E4, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2QQ, United Kingdom
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge, Level E4, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2QQ, United Kingdom
| |
Collapse
|
7
|
Chen J, Wu J, Zhou C, Wu H, Guo T, Bai X, Wu C, Wen J, Qin J, Duanmu X, Tan S, Yuan W, Zheng Q, Zhang B, Guan X, Xu X, Zhang M. Microstructural Degeneration of the Corpus Callosum in Parkinson's Disease with Unilateral Onset: A Free-Water Imaging Study. ACS Chem Neurosci 2025; 16:161-170. [PMID: 39723476 DOI: 10.1021/acschemneuro.4c00598] [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] [Indexed: 12/28/2024] Open
Abstract
Background: Motor symptom laterality is an important clinical feature of PD that not only manifests as lateral limb dysfunction but also affects the nonmotor symptoms and the prognosis in PD patients. Previous studies suggested that the compensatory mechanisms in the dominant hemisphere of the brain may be an underlying explanation. The corpus callosum (CC) is the largest fiber connecting the two hemispheres of the brain. Considering the CC as the pointcut may help elucidate the mechanisms underlying how motor symptom laterality affects nonmotor symptoms and prognosis in PD patients. Purpose: To explore microstructural degeneration of the CC in PD patients with unilateral motor symptom onset and evaluate its relationship with motor and nonmotor performance. Methods: In this study, 201 right-handed PD patients with unilateral motor symptom onset (91 patients with left-onset [LPD] and 110 with right-onset [RPD]) and 100 right-handed healthy controls (HC) were included. A bitensor model of diffusion tensor imaging was applied to analyze free water (FW) as well as fractional anisotropy (FAT) and mean diffusivity (MDT) of the tissue compartment after correcting FW. These provide noninvasive in vivo measures of white matter integrity and pathological processes including atrophy, edema, and neuroinflammation. The CC was divided into halves along the median sagittal line, and each half was manually divided into five functional segments. A total of 10 subregions were obtained and numbered in sequence. The laterality index was calculated to quantify the asymmetry of the CC and its segments. A general linear model was used to compare among groups, and partial correlation analysis was performed to explore the relationships between the diffusion parameters of CC subregions and clinical manifestations. Results: Compared with HC, FW and FAT of the bilateral CC were decreased in the LPD group, whereas MDT in the right hemisphere was increased. In the LPD group, FAT of all CC subregions except for subregions 1, 3, and 6 was significantly lower than HC, and MDT in the anterior and posterior segments of the CC (CC subregions 1, 5-7, and 10) was significantly higher than HC. In the RPD group, FAT of subregion 7 was significantly decreased and MDT was increased than HC. Laterality index analysis of the CC indicated significant interhemispheric FAT asymmetry in the anterior and middle in the RPD group, with a more significant reduction in the right CC. Moreover, degeneration of the CC and its subregions was related to motor and nonmotor symptoms in PD. Conclusions: More extensive CC damage was observed in the LPD group than in the RPD group. Additional, asymmetrical damage was observed in the anterior and middle segments of the CC in the RPD group, suggesting that differences in callosal degeneration patterns may be a potential mechanism underlying how asymmetrical motor symptoms affect the nonmotor symptoms and prognosis in PD.
Collapse
Affiliation(s)
- Jingwen Chen
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou 310009, China
| | - Jingjing Wu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou 310009, China
| | - Cheng Zhou
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou 310009, China
| | - Haoting Wu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou 310009, China
| | - Tao Guo
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou 310009, China
| | - Xueqin Bai
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou 310009, China
| | - Chenqing Wu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou 310009, China
| | - Jiaqi Wen
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou 310009, China
| | - Jianmei Qin
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou 310009, China
| | - Xiaojie Duanmu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou 310009, China
| | - Sijia Tan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou 310009, China
| | - Weijin Yuan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou 310009, China
| | - Qianshi Zheng
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou 310009, China
| | - Baorong Zhang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou 310009, China
| | - Xiaojun Guan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou 310009, China
| | - Xiaojun Xu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou 310009, China
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou 310009, China
| |
Collapse
|
8
|
Sun L, He S, Cheng B, Shen Y, Zhao W, Tu R, Zhang S. White Matter Microstructure Alteration in Patients with Drug-Induced Parkinsonism: A Diffusion Tensor Imaging Study with Tract-Based Spatial Statistics. J Integr Neurosci 2024; 23:202. [PMID: 39613471 DOI: 10.31083/j.jin2311202] [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: 04/27/2024] [Revised: 08/05/2024] [Accepted: 08/15/2024] [Indexed: 12/01/2024] Open
Abstract
INTRODUCTION This research aimed to investigate the pathophysiological mechanism of how drug-induced parkinsonism (DIP) affects the integrity of the white matter (WM) fiber microstructure as measured by magnetic resonance diffusion tensor image (DTI) fractional anisotropy (FA) and mean diffusivity (MD). METHODS We recruited 17 participants diagnosed with DIP, 20 Parkinson's disease (PD) patients, and 16 normal controls (NCs) with a similar age, gender, and years of education. Subsequently, all participants underwent DTI magnetic resonance imaging scanning. To analyze the data, we utilized the software packages Functional MRI of the Brain Centre (FMRIB) Diffusion Toolbox (FDT), developed by the FMRIB laboratory at Oxford University, and tract-based spatial statistics (TBSS). RESULTS The Argentina Hyposmia Rating Scale (AHRS) scores of patients in DIP group were markedly higher than those in PD patients group. Compared with the NC group, the FA values in the genu and body of the corpus callosum (CC), anterior limb of the right internal capsule, bilateral anterior corona radiata, bilateral superior corona radiata, right external capsule, and right superior fronto-occipital fasciculus (could be a part of the anterior internal capsule) were significantly decreased in the DIP group; however, no significant cluster was found in MD. CONCLUSIONS The present study provides novel insights into the alterations in WM microstructure among DIP patients, suggesting that these methodologies have the potential to aid in the early diagnosis and treatment of DIP.
Collapse
Affiliation(s)
- Ling Sun
- Department of Geriatrics, Nanchong Central Hospital, 637000 Nanchong, Sichuan, China
| | - Shijia He
- Department of Neurology, Meishan People's Hospital, 620010 Meishan, Sichuan, China
| | - Bo Cheng
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
| | - Yao Shen
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
| | - Wenhao Zhao
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
| | - Rong Tu
- Department of Neurology, Nanchong Central Hospital, 637000 Nanchong, Sichuan, China
| | - Shushan Zhang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
| |
Collapse
|
9
|
Scorziello A, Sirabella R, Sisalli MJ, Tufano M, Giaccio L, D’Apolito E, Castellano L, Annunziato L. Mitochondrial Dysfunction in Parkinson's Disease: A Contribution to Cognitive Impairment? Int J Mol Sci 2024; 25:11490. [PMID: 39519043 PMCID: PMC11546611 DOI: 10.3390/ijms252111490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/05/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Among the non-motor symptoms associated with Parkinson's disease (PD), cognitive impairment is one of the most common and disabling. It can occur either early or late during the disease, and it is heterogeneous in terms of its clinical manifestations, such as Subjective Cognitive Dysfunction (SCD), Mild Cognitive Impairment (MCI), and Parkinson's Disease Dementia (PDD). The aim of the present review is to delve deeper into the molecular mechanisms underlying cognitive decline in PD. This is extremely important to delineate the guidelines for the differential diagnosis and prognosis of the dysfunction, to identify the molecular and neuronal mechanisms involved, and to plan therapeutic strategies that can halt cognitive impairment progression. Specifically, the present review will discuss the pathogenetic mechanisms involved in the progression of cognitive impairment in PD, with attention to mitochondria and their contribution to synaptic dysfunction and neuronal deterioration in the brain regions responsible for non-motor manifestations of the disease.
Collapse
Affiliation(s)
- Antonella Scorziello
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Rossana Sirabella
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Maria Josè Sisalli
- Department of Translational Medicine, Federico II University of Naples, 80138 Napoli, Italy;
| | - Michele Tufano
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Lucia Giaccio
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Elena D’Apolito
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Lorenzo Castellano
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | | |
Collapse
|
10
|
Owens-Walton C, Nir TM, Al-Bachari S, Ambrogi S, Anderson TJ, Aventurato ÍK, Cendes F, Chen YL, Ciullo V, Cook P, Dalrymple-Alford JC, Dirkx MF, Druzgal J, Emsley HCA, Guimarães R, Haroon HA, Helmich RC, Hu MT, Johansson ME, Kim HB, Klein JC, Laansma M, Lawrence KE, Lochner C, Mackay C, McMillan CT, Melzer TR, Nabulsi L, Newman B, Opriessnig P, Parkes LM, Pellicano C, Piras F, Piras F, Pirpamer L, Pitcher TL, Poston KL, Roos A, Silva LS, Schmidt R, Schwingenschuh P, Shahid-Besanti M, Spalletta G, Stein DJ, Thomopoulos SI, Tosun D, Tsai CC, van den Heuvel OA, van Heese E, Vecchio D, Villalón-Reina JE, Vriend C, Wang JJ, Wu YR, Yasuda CL, Thompson PM, Jahanshad N, van der Werf Y. A worldwide study of white matter microstructural alterations in people living with Parkinson's disease. NPJ Parkinsons Dis 2024; 10:151. [PMID: 39128907 PMCID: PMC11317500 DOI: 10.1038/s41531-024-00758-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 07/22/2024] [Indexed: 08/13/2024] Open
Abstract
The progression of Parkinson's disease (PD) is associated with microstructural alterations in neural pathways, contributing to both motor and cognitive decline. However, conflicting findings have emerged due to the use of heterogeneous methods in small studies. Here we performed a large diffusion MRI study in PD, integrating data from 17 cohorts worldwide, to identify stage-specific profiles of white matter differences. Diffusion-weighted MRI data from 1654 participants diagnosed with PD (age: 20-89 years; 33% female) and 885 controls (age: 19-84 years; 47% female) were analyzed using the ENIGMA-DTI protocol to evaluate white matter microstructure. Skeletonized maps of fractional anisotropy (FA) and mean diffusivity (MD) were compared across Hoehn and Yahr (HY) disease groups and controls to reveal the profile of white matter alterations at different stages. We found an enhanced, more widespread pattern of microstructural alterations with each stage of PD, with eventually lower FA and higher MD in almost all regions of interest: Cohen's d effect sizes reached d = -1.01 for FA differences in the fornix at PD HY Stage 4/5. The early PD signature in HY stage 1 included higher FA and lower MD across the entire white matter skeleton, in a direction opposite to that typical of other neurodegenerative diseases. FA and MD were associated with motor and non-motor clinical dysfunction. While overridden by degenerative changes in the later stages of PD, early PD is associated with paradoxically higher FA and lower MD in PD, consistent with early compensatory changes associated with the disorder.
Collapse
Grants
- R01 AG058854 NIA NIH HHS
- P41 EB015922 NIBIB NIH HHS
- R01NS107513 U.S. Department of Health & Human Services | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- R01 MH117601 NIMH NIH HHS
- R01 NS107513 NINDS NIH HHS
- U19 AG062418 NIA NIH HHS
- F32 MH122057 NIMH NIH HHS
- R01 AG059874 NIA NIH HHS
- U.S. Alzheimer’s Association (AARG-23-1149996)
- Health Research Council of New Zealand (20/538; 21/165)
- São Paulo Research Foundation FAPESP-BRAINN Grants# 2013-07559-3 / FAPESP #2022-1178-4
- São Paulo Research Foundation FAPESP-BRAINN Grant # 2013–07559-3.
- Health Research Council of New Zealand (20/538); Marsden Fund New Zealand (UOC2105); Neurological Foundation of New Zealand (2232 PRG); Research and Education Trust Pacific Radiology (MRIJDA).
- Grant from ParkinsonNL (P2023-14); Honoraria from Movement Disorders Society Quebec.
- NINDS R01NS107513
- Engineering and Physical Sciences Research Council (EPSRC) UK
- Parkinson's UK, Cure Parkinsons Trust, Oxford Biomedical Research Centre, GSK-Oxford IMCM.
- JK is supported by the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), and the NIHR Oxford Health Clinical Research Facility. The views expressed are those of the authors and not necessarily those of the NHS, the NIHR or the Department of Health.
- NIMH 32MH122057
- U19 AG062418
- Health Research Council of New Zealand (20/538); Neurological Foundation of New Zealand (2232 PRG); Research and Education Trust Pacific Radiology (MRIJDA).
- EPSRC UK, MRC UK, GE medical systems, Academy of Medical Sciences UK
- Italian Ministry of Health, grant number RF-2019-12370182
- Health Research Council of New Zealand (21/165)
- Personal fees from Bial, AbbVie and Boston Scientific.
- NIH/NIA
- São Paulo Research Foundation FAPESP-BRAINN Grant # 2013–07559-3; CNPQ (#315953/2021-7) National Council for Scientific and Technological Development
- U.S. Department of Health & Human Services | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- R01AG059874, R01MH117601, R01NS107513, R01AG058854, P41EB015922
Collapse
Affiliation(s)
- Conor Owens-Walton
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA.
| | - Talia M Nir
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | | | - Sonia Ambrogi
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Tim J Anderson
- Department of Medicine, University of Otago, Christchurch, Christchurch, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
- Neurology Department, Te Whatu Ora-Health New Zealand Waitaha Canterbury, Christchurch, New Zealand
| | - Ítalo Karmann Aventurato
- Department of Neurology, University of Campinas-UNICAMP, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Fernando Cendes
- Department of Neurology, University of Campinas-UNICAMP, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Yao-Liang Chen
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Keelung, Taiwan, ROC
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan, ROC
| | - Valentina Ciullo
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Phil Cook
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - John C Dalrymple-Alford
- New Zealand Brain Research Institute, Christchurch, New Zealand
- Te Kura Mahi ā- Hirikapo | School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
| | - Michiel F Dirkx
- Department of Neurology and Center of Expertise for Parkinson & Movement Disorders, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Jason Druzgal
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Hedley C A Emsley
- Lancaster Medical School, Lancaster University, Lancaster, UK
- Department of Neurology, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, UK
| | - Rachel Guimarães
- Department of Neurology, University of Campinas-UNICAMP, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Hamied A Haroon
- Division of Psychology, Communication & Human Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Rick C Helmich
- Department of Neurology and Center of Expertise for Parkinson & Movement Disorders, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
- Centre for Cognitive Neuroimaging, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Michele T Hu
- Oxford Parkinson's Disease Centre, Nuffield, Department of Clinical Neurosciences, Division of Clinical Neurology, University of Oxford, Oxford, UK
| | - Martin E Johansson
- Department of Neurology and Center of Expertise for Parkinson & Movement Disorders, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
- Centre for Cognitive Neuroimaging, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Ho Bin Kim
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Johannes C Klein
- Oxford Parkinson's Disease Centre, Nuffield, Department of Clinical Neurosciences, Division of Clinical Neurology, University of Oxford, Oxford, UK
| | - Max Laansma
- Amsterdam UMC, Dept. Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Katherine E Lawrence
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Christine Lochner
- SA MRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry, Stellenbosch University, Cape Town, South Africa
| | - Clare Mackay
- Oxford Parkinson's Disease Centre, Nuffield, Department of Clinical Neurosciences, Division of Clinical Neurology, University of Oxford, Oxford, UK
| | - Corey T McMillan
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Tracy R Melzer
- Department of Medicine, University of Otago, Christchurch, Christchurch, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
- Te Kura Mahi ā- Hirikapo | School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
| | - Leila Nabulsi
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Ben Newman
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Peter Opriessnig
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University Graz, Graz, Austria
| | - Laura M Parkes
- Division of Psychology, Communication & Human Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Clelia Pellicano
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Fabrizio Piras
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Federica Piras
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Lukas Pirpamer
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University Graz, Graz, Austria
| | - Toni L Pitcher
- Department of Medicine, University of Otago, Christchurch, Christchurch, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Kathleen L Poston
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Annerine Roos
- SA MRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Lucas Scárdua Silva
- Department of Neurology, University of Campinas-UNICAMP, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Reinhold Schmidt
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University Graz, Graz, Austria
| | - Petra Schwingenschuh
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University Graz, Graz, Austria
| | - Marian Shahid-Besanti
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | | | - Dan J Stein
- SA MRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Sophia I Thomopoulos
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Chih-Chien Tsai
- Healthy Aging Research Center, Chang Gung University, Taoyuan City, Taiwan, ROC
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan City, Taiwan, ROC
| | - Odile A van den Heuvel
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, USA
- Amsterdam UMC, Dept. Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC, Department of Psychiatry, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Eva van Heese
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, USA
- Amsterdam UMC, Dept. Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Daniela Vecchio
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Julio E Villalón-Reina
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Chris Vriend
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, USA
- Amsterdam UMC, Department of Psychiatry, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging program, Amsterdam, The Netherlands
| | - Jiun-Jie Wang
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Keelung, Taiwan, ROC
- Healthy Aging Research Center, Chang Gung University, Taoyuan City, Taiwan, ROC
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan City, Taiwan, ROC
- Department of Chemical Engineering, Ming-Chi University of Technology, New Taipei City, Taiwan, ROC
| | - Yih-Ru Wu
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan, ROC
- Department of Neurology, College of Medicine, Chang Gung University, Taoyuan City, Taiwan, ROC
| | - Clarissa Lin Yasuda
- Department of Neurology, University of Campinas-UNICAMP, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Ysbrand van der Werf
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, USA
- Amsterdam UMC, Dept. Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Gao B, Qu M, Jiang Y, Li W, Wang M, Pei C, Zheng D, Yang C, Miao Y. Fractional Anisotropy is a More Sensitive Diagnostic Biomarker Than Mean Kurtosis for Patients with Parkinson Disease with Cognitive Dysfunction: A Diffusional Kurtosis Map Tract-Based Spatial Statistics Study. AJNR Am J Neuroradiol 2024; 45:1098-1105. [PMID: 38991767 PMCID: PMC11383405 DOI: 10.3174/ajnr.a8297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 03/16/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND AND PURPOSE There is heterogeneity of white matter damage in Parkinson's disease patients with different cognitive states. Our aim was to find sensitive diffusional kurtosis imaging biomarkers to differentiate the white matter damage pattern of mild cognitive impairment and dementia. MATERIALS AND METHODS Nineteen patients with Parkinson disease with mild cognitive impairment and 18 patients with Parkinson disease with dementia were prospectively enrolled. All participants underwent MR examination with 3D-T1-weighted image and diffusional kurtosis imaging sequences. Demographic data were compared between the 2 groups. Voxelwise statistical analyses of diffusional kurtosis imaging parameters were performed using tract-based spatial statistics. The receiver operator characteristic curve of significantly different metrics was graphed. The correlation of significantly different metrics with global cognitive status was analyzed. RESULTS Compared with the Parkinson disease with mild cognitive impairment group, the fractional anisotropy and mean kurtosis values decreased in 4 independent clusters in the forceps minor, forceps major, inferior fronto-occipital fasciculus, and the inferior and superior longitudinal fasciculus in patients with Parkinson disease with dementia; the mean diffusivity decreased in 1 cluster in the forceps minor. The fractional anisotropy value in the inferior fronto-occipital fasciculus and inferior longitudinal fasciculus would be the diffusional kurtosis imaging marker for the differential diagnosis of Parkinson disease with mild cognitive impairment and patients with Parkinson disease with dementia, with the best diagnostic efficiency of 0.853. The fractional anisotropy values in the forceps minor (β = 84.20, P < .001) and years of education (β = 0.38, P = .014) were positively correlated with the Montreal Cognitive Assessment. CONCLUSIONS The diffusional kurtosis imaging-derived fractional anisotropy and mean kurtosis can detect the different white matter damage patterns of Parkinson disease with mild cognitive impairment and Parkinson disease with dementia. Fractional anisotropy is more sensitive than mean kurtosis in the differential diagnosis; fractional anisotropy derived from diffusional kurtosis imaging could become a promising imaging marker for the differential diagnosis of Parkinson disease with mild cognitive impairment and Parkinson disease with dementia.
Collapse
Affiliation(s)
- Bingbing Gao
- From the Department of Radiology (B.G., M.Q., Y.J., W.l., M.W., C.Y., Y.M), The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Mingrui Qu
- From the Department of Radiology (B.G., M.Q., Y.J., W.l., M.W., C.Y., Y.M), The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuhan Jiang
- From the Department of Radiology (B.G., M.Q., Y.J., W.l., M.W., C.Y., Y.M), The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wanyao Li
- From the Department of Radiology (B.G., M.Q., Y.J., W.l., M.W., C.Y., Y.M), The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Man Wang
- From the Department of Radiology (B.G., M.Q., Y.J., W.l., M.W., C.Y., Y.M), The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Chenhui Pei
- Department of Neurology (C.P.), The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dandan Zheng
- Philips Healthcare, Clinical &Technique Support (D.Z.), Beijing, China
| | - Chao Yang
- From the Department of Radiology (B.G., M.Q., Y.J., W.l., M.W., C.Y., Y.M), The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yanwei Miao
- From the Department of Radiology (B.G., M.Q., Y.J., W.l., M.W., C.Y., Y.M), The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
12
|
Albadawi EA. Microstructural Changes in the Corpus Callosum in Neurodegenerative Diseases. Cureus 2024; 16:e67378. [PMID: 39310519 PMCID: PMC11413839 DOI: 10.7759/cureus.67378] [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] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
The corpus callosum, the largest white matter structure in the brain, plays a crucial role in interhemispheric communication and cognitive function. This review examines the microstructural changes observed in the corpus callosum across various neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis (ALS). New neuroimaging studies, mainly those that use diffusion tensor imaging (DTI) and advanced tractography methods, were put together to show how changes have happened in the organization of white matter and the connections between them. Some of the most common ways the corpus callosum breaks down are discussed, including less fractional anisotropy, higher mean diffusivity, and atrophy in certain regions. The relationship between these microstructural changes and cognitive decline, motor dysfunction, and disease progression is explored. Additionally, we consider the potential of corpus callosum imaging as a biomarker for early disease detection and monitoring. Studies show that people with these disorders have lower fractional anisotropy and higher mean diffusivity in the corpus callosum, often in ways that are specific to the disease. These changes often happen before gray matter atrophy and are linked to symptoms, which suggests that the corpus callosum could be used as an early sign of neurodegeneration. The review also highlights the implications of these findings for understanding disease mechanisms and developing therapeutic strategies. Future directions, including the application of advanced imaging techniques and longitudinal studies, are discussed to elucidate the role of corpus callosum degeneration in neurodegenerative processes. This review underscores the importance of the corpus callosum in understanding the pathophysiology of neurodegenerative diseases and its potential as a target for therapeutic interventions.
Collapse
Affiliation(s)
- Emad A Albadawi
- Department of Basic Medical Sciences, College of Medicine, Taibah Univeristy, Madinah, SAU
| |
Collapse
|
13
|
Pang H, Yu Z, Yu H, Li X, Bu S, Liu Y, Wang J, Zhao M, Fan G. Advanced Cognitive Patterns in Multiple System Atrophy Compared to Parkinson's Disease: An Individual Diffusion Tensor Imaging Study. Acad Radiol 2024; 31:2897-2909. [PMID: 38220569 DOI: 10.1016/j.acra.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/27/2023] [Accepted: 01/04/2024] [Indexed: 01/16/2024]
Abstract
RATIONALE AND OBJECTIVES Although both Multiple system atrophy (MSA) and Parkinson's disease (PD) belong to alpha-synucleinopathy, they have divergent clinical courses and prognoses. The degeneration of white matter has a considerable impact on cognitive performance, yet it is uncertain how PD and MSA affect its functioning in a similar or different manner. METHODS In this study, a total of 116 individuals (37 PD with mild cognitive impairment (PD-MCI), 37 MSA (parkinsonian variant) with mild cognitive impairment (MSA-MCI), and 42 healthy controls) underwent diffusion tensor imaging (DTI) and cognitive assessment. Utilizing probabilistic fiber tracking, association fibers, projection fibers, and thalamic fibers were reconstructed. Subsequently, regression, support vector machine, and SHAP (Shapley Addictive exPlanations) analyzes were conducted to evaluate the association between microstructural diffusion metrics and multiple cognitive domains, thus determining the white matter predictors of MCI. RESULTS MSA-MCI patients exhibited distinct white matter impairment extending to the middle cerebellar peduncle, corticospinal tract, and cingulum bundle. Furthermore, the fractional anisotropy (FA) and mean diffusivity (MD)values of the right anterior thalamic radiation were significantly associated with global efficiency (FA: B = 0.69, P < 0.001, VIF = 1.31; MD: B = -0.53, P = 0.02, VIF = 2.50). The diffusion metrics of white matter between PD-MCI and MSA-MCI proved to be an effective predictor of the MCI, with an accuracy of 0.73 (P < 0.01), and the most predictive factor being the MD of the anterior thalamic radiation. CONCLUSIONS Our results demonstrated that MSA-MCI had a more noticeable deterioration in white matter, which potentially linked to various cognitive domain connections. Diffusion MRI could be a useful tool in comprehending the neurological basis of cognitive impairment in Parkinsonian disorders.
Collapse
Affiliation(s)
- Huize Pang
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.P., Z.Y., X.L., S.B., Y.L., J.W., M.Z., G.F.)
| | - Ziyang Yu
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.P., Z.Y., X.L., S.B., Y.L., J.W., M.Z., G.F.)
| | - Hongmei Yu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.Y.)
| | - Xiaolu Li
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.P., Z.Y., X.L., S.B., Y.L., J.W., M.Z., G.F.)
| | - Shuting Bu
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.P., Z.Y., X.L., S.B., Y.L., J.W., M.Z., G.F.)
| | - Yu Liu
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.P., Z.Y., X.L., S.B., Y.L., J.W., M.Z., G.F.)
| | - Juzhou Wang
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.P., Z.Y., X.L., S.B., Y.L., J.W., M.Z., G.F.)
| | - Mengwan Zhao
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.P., Z.Y., X.L., S.B., Y.L., J.W., M.Z., G.F.)
| | - Guoguang Fan
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.P., Z.Y., X.L., S.B., Y.L., J.W., M.Z., G.F.).
| |
Collapse
|
14
|
Patil P, Ford WR. Parkinson's Disease Recognition Using Decorrelated Convolutional Neural Networks: Addressing Imbalance and Scanner Bias in rs-fMRI Data. BIOSENSORS 2024; 14:259. [PMID: 38785733 PMCID: PMC11117585 DOI: 10.3390/bios14050259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/11/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative and progressive disease that impacts the nerve cells in the brain and varies from person to person. The exact cause of PD is still unknown, and the diagnosis of PD does not include a specific objective test with certainty. Although deep learning has made great progress in medical neuroimaging analysis, these methods are very susceptible to biases present in neuroimaging datasets. An innovative decorrelated deep learning technique is introduced to mitigate class bias and scanner bias while simultaneously focusing on finding distinguishing characteristics in resting-state functional MRI (rs-fMRI) data, which assists in recognizing PD with good accuracy. The decorrelation function reduces the nonlinear correlation between features and bias in order to learn bias-invariant features. The publicly available Parkinson's Progression Markers Initiative (PPMI) dataset, referred to as a single-scanner imbalanced dataset in this study, was used to validate our method. The imbalanced dataset problem affects the performance of the deep learning framework by overfitting to the majority class. To resolve this problem, we propose a new decorrelated convolutional neural network (DcCNN) framework by applying decorrelation-based optimization to convolutional neural networks (CNNs). An analysis of evaluation metrics comparisons shows that integrating the decorrelation function boosts the performance of PD recognition by removing class bias. Specifically, our DcCNN models perform significantly better than existing traditional approaches to tackle the imbalance problem. Finally, the same framework can be extended to create scanner-invariant features without significantly impacting the performance of a model. The obtained dataset is a multiscanner dataset, which leads to scanner bias due to the differences in acquisition protocols and scanners. The multiscanner dataset is a combination of two publicly available datasets, namely, PPMI and FTLDNI-the frontotemporal lobar degeneration neuroimaging initiative (NIFD) dataset. The results of t-distributed stochastic neighbor embedding (t-SNE) and scanner classification accuracy of our proposed feature extraction-DcCNN (FE-DcCNN) model validated the effective removal of scanner bias. Our method achieves an average accuracy of 77.80% on a multiscanner dataset for differentiating PD from a healthy control, which is superior to the DcCNN model trained on a single-scanner imbalanced dataset.
Collapse
Affiliation(s)
- Pranita Patil
- Department of Analytics, Harrisburg University of Science and Technology, Harrisburg, PA 17101, USA;
| | | |
Collapse
|
15
|
Jiang S, Wang Y, Pei H, Li H, Chen J, Yao Y, Li Q, Yao D, Luo C. Brain activation and connection across resting and motor-task states in patients with generalized tonic-clonic seizures. CNS Neurosci Ther 2024; 30:e14672. [PMID: 38644561 PMCID: PMC11033329 DOI: 10.1111/cns.14672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 02/08/2024] [Accepted: 02/17/2024] [Indexed: 04/23/2024] Open
Abstract
AIMS Motor abnormalities have been identified as one common symptom in patients with generalized tonic-clonic seizures (GTCS) inspiring us to explore the disease in a motor execution condition, which might provide novel insight into the pathomechanism. METHODS Resting-state and motor-task fMRI data were collected from 50 patients with GTCS, including 18 patients newly diagnosed without antiepileptic drugs (ND_GTCS) and 32 patients receiving antiepileptic drugs (AEDs_GTCS). Motor activation and its association with head motion and cerebral gradients were assessed. Whole-brain network connectivity across resting and motor states was further calculated and compared between groups. RESULTS All patients showed over-activation in the postcentral gyrus and the ND_GTCS showed decreased activation in putamen. Specifically, activation maps of ND_GTCS showed an abnormal correlation with head motion and cerebral gradient. Moreover, we detected altered functional network connectivity in patients within states and across resting and motor states by using repeated-measures analysis of variance. Patients did not show abnormal connectivity in the resting state, while distributed abnormal connectivity in the motor-task state. Decreased across-state network connectivity was also found in all patients. CONCLUSION Convergent findings suggested the over-response of activation and connection of the brain to motor execution in GTCS, providing new clues to uncover motor susceptibility underlying the disease.
Collapse
Affiliation(s)
- Sisi Jiang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of ChinaChengduP. R. China
- Research Unit of NeuroInformationChinese Academy of Medical SciencesChengduP. R. China
- High‐Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan ProvinceCenter for Information in MedicineUniversity of Electronic Science and Technology of ChinaChengduP. R. China
| | - Yuehan Wang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of ChinaChengduP. R. China
| | - Haonan Pei
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of ChinaChengduP. R. China
| | - Hechun Li
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of ChinaChengduP. R. China
| | - Junxia Chen
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of ChinaChengduP. R. China
| | - Yutong Yao
- Department of NeurosurgeySichuan Provincial People's Hospital, University of Electronic Science and Technology of ChinaChengduP. R. China
| | - Qifu Li
- Department of NeurologyHainan Medical UniversityHainanP. R. China
| | - Dezhong Yao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of ChinaChengduP. R. China
- Research Unit of NeuroInformationChinese Academy of Medical SciencesChengduP. R. China
- High‐Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan ProvinceCenter for Information in MedicineUniversity of Electronic Science and Technology of ChinaChengduP. R. China
| | - Cheng Luo
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of ChinaChengduP. R. China
- Research Unit of NeuroInformationChinese Academy of Medical SciencesChengduP. R. China
- High‐Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan ProvinceCenter for Information in MedicineUniversity of Electronic Science and Technology of ChinaChengduP. R. China
| |
Collapse
|
16
|
Chen CL, Cheng SY, Montaser-Kouhsari L, Wu WC, Hsu YC, Tai CH, Tseng WYI, Kuo MC, Wu RM. Advanced brain aging in Parkinson's disease with cognitive impairment. NPJ Parkinsons Dis 2024; 10:62. [PMID: 38493188 PMCID: PMC10944471 DOI: 10.1038/s41531-024-00673-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 02/29/2024] [Indexed: 03/18/2024] Open
Abstract
Patients with Parkinson's disease and cognitive impairment (PD-CI) deteriorate faster than those without cognitive impairment (PD-NCI), suggesting an underlying difference in the neurodegeneration process. We aimed to verify brain age differences in PD-CI and PD-NCI and their clinical significance. A total of 94 participants (PD-CI, n = 27; PD-NCI, n = 34; controls, n = 33) were recruited. Predicted age difference (PAD) based on gray matter (GM) and white matter (WM) features were estimated to represent the degree of brain aging. Patients with PD-CI showed greater GM-PAD (7.08 ± 6.64 years) and WM-PAD (8.82 ± 7.69 years) than those with PD-NCI (GM: 1.97 ± 7.13, Padjusted = 0.011; WM: 4.87 ± 7.88, Padjusted = 0.049) and controls (GM: -0.58 ± 7.04, Padjusted = 0.004; WM: 0.88 ± 7.45, Padjusted = 0.002) after adjusting demographic factors. In patients with PD, GM-PAD was negatively correlated with MMSE (Padjusted = 0.011) and MoCA (Padjusted = 0.013) and positively correlated with UPDRS Part II (Padjusted = 0.036). WM-PAD was negatively correlated with logical memory of immediate and delayed recalls (Padjusted = 0.003 and Padjusted < 0.001). Also, altered brain regions in PD-CI were identified and significantly correlated with brain age measures, implicating the neuroanatomical underpinning of neurodegeneration in PD-CI. Moreover, the brain age metrics can improve the classification between PD-CI and PD-NCI. The findings suggest that patients with PD-CI had advanced brain aging that was associated with poor cognitive functions. The identified neuroimaging features and brain age measures can serve as potential biomarkers of PD-CI.
Collapse
Affiliation(s)
- Chang-Le Chen
- Institute of Medical Device and Imaging, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shao-Ying Cheng
- Department of Neurology, National Taiwan University Hospital Bei-Hu Branch, Taipei, Taiwan
| | | | - Wen-Chao Wu
- Institute of Medical Device and Imaging, National Taiwan University College of Medicine, Taipei, Taiwan
| | | | - Chun-Hwei Tai
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Yih Isaac Tseng
- Institute of Medical Device and Imaging, National Taiwan University College of Medicine, Taipei, Taiwan.
- Acroviz Inc, Taipei, Taiwan.
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan.
| | - Ming-Che Kuo
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.
- Department of Medicine, National Taiwan University Cancer Center, Taipei, Taiwan.
- Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan.
| | - Ruey-Meei Wu
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
17
|
Liu L, Shen Q, Zhang D, Bao Y, Xu F, Huang H, Xu Y. Mendelian Randomization Analysis to Assess Whether Magnetic Resonance Imaging Signs of Cerebral Small Vessel Disease Can Cause Cognitive Decline and Dementia. J Prev Alzheimers Dis 2024; 11:1390-1396. [PMID: 39350385 DOI: 10.14283/jpad.2024.95] [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] [Indexed: 03/17/2025]
Abstract
OBJECTIVE Cognitive decline and dementia have been linked to cerebral small vessel disease, so we explored using Mendelian randomization whether cerebral small vessel disease visible as 10 neuroimaging signs may cause cognitive decline and dementia. METHODS We analyzed publicly available data from genome-wide association studies using two-sample Mendelian randomization involving inverse variance weighting, weighted median, MR-Egger, and MR-PRESSO approaches. RESULTS Mendelian randomization suggested that cognitive decline can be caused by lacunar stroke (inverse variance weighting, β = -0.012, 95% CI -0.024 to -0.001, P = 0.033). Furthermore, an elevated burden of white matter hyperintensities was associated with an increased risk of Dementia due to Parkinson's disease (inverse variance weighting, OR 2.035, 95% CI 1.105 to 3.745, P = 0.023). Notably, no significant associations were observed between neuroimaging markers of Cerebral Small Vessel Disease and other types of dementia. CONCLUSION This Mendelian randomization study provides evidence that lacunar stroke and white matter lesions can cause cognitive decline, and that white matter hyperintensity may increase risk of dementia due to Parkinson's disease. These results underscore the need for further investigations into the neurocognitive effects of cerebral small vessel disease.
Collapse
Affiliation(s)
- L Liu
- Yanming Xu, Sichuan University West China Hospital, West China Hospital of Sichuan University, China,
| | | | | | | | | | | | | |
Collapse
|
18
|
Jia Y, Sun H, Sun L, Wang Y, Xu Q, Liu Y, Chang X, He Y, Guo D, Shi M, Chen GC, Zheng J, Zhang Y, Zhu Z. Mendelian randomization analysis implicates bidirectional associations between brain imaging-derived phenotypes and ischemic stroke. Cereb Cortex 2023; 33:10848-10857. [PMID: 37697910 DOI: 10.1093/cercor/bhad329] [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: 07/24/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/13/2023] Open
Abstract
Brian imaging-derived phenotypes (IDPs) have been suggested to be associated with ischemic stroke, but the causality between them remains unclear. In this bidirectional two-sample Mendelian randomization (MR) study, we explored the potential causal relationship between 461 imaging-derived phenotypes (n = 33,224, UK Biobank) and ischemic stroke (n = 34,217 cases/406,111 controls, Multiancestry Genome-Wide Association Study of Stroke). Forward MR analyses identified five IDPs associated with ischemic stroke, including mean diffusivity (MD) in the right superior fronto-occipital fasciculus (1.22 [95% CI, 1.11-1.34]), MD in the left superior fronto-occipital fasciculus (1.30 [1.17-1.44]), MD in the anterior limb of the right internal capsule (1.36 [1.22-1.51]), MD in the right anterior thalamic radiation (1.17 [1.09-1.26]), and MD in the right superior thalamic radiation (1.23 [1.11-1.35]). In the reverse MR analyses, ischemic stroke was identified to be associated with three IDPs, including high isotropic or free water volume fraction in the body of corpus callosum (beta, 0.189 [95% confidence interval, 0.107-0.271]), orientation dispersion index in the pontine crossing tract (0.175 [0.093-0.257]), and volume of the third ventricle (0.219 [0.138-0.301]). This bidirectional two-sample MR study suggested five predictors and three diagnostic markers for ischemic stroke at the brain-imaging level. Further studies are warranted to replicate our findings and clarify underlying mechanisms.
Collapse
Affiliation(s)
- Yiming Jia
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Ministry of Education (MOE) Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Hongyan Sun
- Department of Medical Imaging, The Affiliated Guangji Hospital of Soochow University, Suzhou 215137, China
| | - Lulu Sun
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Ministry of Education (MOE) Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Yinan Wang
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Ministry of Education (MOE) Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Qingyun Xu
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Ministry of Education (MOE) Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Yi Liu
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Ministry of Education (MOE) Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Xinyue Chang
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Ministry of Education (MOE) Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Yu He
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Ministry of Education (MOE) Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Daoxia Guo
- School of Nursing, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Mengyao Shi
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Ministry of Education (MOE) Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Guo-Chong Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Jin Zheng
- Department of Neurology, Minhang Hospital, Fudan University, Shanghai 200433, China
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Ministry of Education (MOE) Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Zhengbao Zhu
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Ministry of Education (MOE) Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| |
Collapse
|
19
|
Neophytou K, Wiley R, Litovsky C, Tsapkini K, Rapp B. The right hemisphere's capacity for language: evidence from primary progressive aphasia. Cereb Cortex 2023; 33:9971-9985. [PMID: 37522277 PMCID: PMC10502784 DOI: 10.1093/cercor/bhad258] [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: 01/06/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 08/01/2023] Open
Abstract
The role of the right hemisphere (RH) in core language processes is still a matter of intense debate. Most of the relevant evidence has come from studies of gray matter, with relatively little research on RH white matter (WM) connectivity. Using Diffusion Tensor Imaging-based tractography, the current work examined the role of the two hemispheres in language processing in 33 individuals with Primary Progressive Aphasia (PPA), aiming to better characterize the contribution of the RH to language processing in the context of left hemisphere (LH) damage. The findings confirm the impact of PPA on the integrity of the WM language tracts in the LH. Additionally, an examination of the relationship between tract integrity and language behaviors provides robust evidence of the involvement of the WM language tracts of both hemispheres in language processing in PPA. Importantly, this study provides novel evidence of a unique contribution of the RH to language processing (i.e. a contribution independent from that of the language-dominant LH). Finally, we provide evidence that the RH contribution is specific to language processing rather than being domain general. These findings allow us to better characterize the role of RH in language processing, particularly in the context of LH damage.
Collapse
Affiliation(s)
- Kyriaki Neophytou
- Department of Neurology, Johns Hopkins Medicine, Baltimore, MD, United States
| | - Robert Wiley
- Department of Psychology, University of North Carolina at Greensboro, Greensboro, NC, United States
- Department of Cognitive Science, Johns Hopkins University, Baltimore, MD, United States
| | - Celia Litovsky
- Department of Cognitive Science, Johns Hopkins University, Baltimore, MD, United States
| | - Kyrana Tsapkini
- Department of Neurology, Johns Hopkins Medicine, Baltimore, MD, United States
- Department of Cognitive Science, Johns Hopkins University, Baltimore, MD, United States
| | - Brenda Rapp
- Department of Cognitive Science, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
20
|
Elmers J, Colzato LS, Akgün K, Ziemssen T, Beste C. Neurofilaments - Small proteins of physiological significance and predictive power for future neurodegeneration and cognitive decline across the life span. Ageing Res Rev 2023; 90:102037. [PMID: 37619618 DOI: 10.1016/j.arr.2023.102037] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/15/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023]
Abstract
Neurofilaments (NFs) are not only important for axonal integrity and nerve conduction in large myelinated axons but they are also thought to be crucial for receptor and synaptic functioning. Therefore, NFs may play a critical role in cognitive functions, as cognitive processes are known to depend on synaptic integrity and are modulated by dopaminergic signaling. Here, we present a theory-driven interdisciplinary approach that NFs may link inflammation, neurodegeneration, and cognitive functions. We base our hypothesis on a wealth of evidence suggesting a causal link between inflammation and neurodegeneration and between these two and cognitive decline (see Fig. 1), also taking dopaminergic signaling into account. We conclude that NFs may not only serve as biomarkers for inflammatory, neurodegenerative, and cognitive processes but also represent a potential mechanical hinge between them, moreover, they may even have predictive power regarding future cognitive decline. In addition, we advocate the use of both NFs and MRI parameters, as their synthesis offers the opportunity to individualize medical treatment by providing a comprehensive view of underlying disease activity in neurological diseases. Since our society will become significantly older in the upcoming years and decades, maintaining cognitive functions and healthy aging will play an important role. Thanks to technological advances in recent decades, NFs could serve as a rapid, noninvasive, and relatively inexpensive early warning system to identify individuals at increased risk for cognitive decline and could facilitate the management of cognitive dysfunctions across the lifespan.
Collapse
Affiliation(s)
- Julia Elmers
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Lorenza S Colzato
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan, China.
| | - Katja Akgün
- Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan, China.
| |
Collapse
|
21
|
Xu C, Neuroth T, Fujiwara T, Liang R, Ma KL. A Predictive Visual Analytics System for Studying Neurodegenerative Disease Based on DTI Fiber Tracts. IEEE TRANSACTIONS ON VISUALIZATION AND COMPUTER GRAPHICS 2023; 29:2020-2035. [PMID: 34965212 DOI: 10.1109/tvcg.2021.3137174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Diffusion tensor imaging (DTI) has been used to study the effects of neurodegenerative diseases on neural pathways, which may lead to more reliable and early diagnosis of these diseases as well as a better understanding of how they affect the brain. We introduce a predictive visual analytics system for studying patient groups based on their labeled DTI fiber tract data and corresponding statistics. The system's machine-learning-augmented interface guides the user through an organized and holistic analysis space, including the statistical feature space, the physical space, and the space of patients over different groups. We use a custom machine learning pipeline to help narrow down this large analysis space and then explore it pragmatically through a range of linked visualizations. We conduct several case studies using DTI and T1-weighted images from the research database of Parkinson's Progression Markers Initiative.
Collapse
|
22
|
Brown G, Hakun J, Lewis MM, De Jesus S, Du G, Eslinger PJ, Kong L, Huang X. Frontostriatal and limbic contributions to cognitive decline in Parkinson's disease. J Neuroimaging 2023; 33:121-133. [PMID: 36068704 PMCID: PMC9840678 DOI: 10.1111/jon.13045] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND AND PURPOSE The circuitry underlying heterogenous cognitive profiles in Parkinson's disease (PD) remains unclear. The purpose of this study is to investigate whether structural changes in frontostriatal and limbic pathways contribute to different cognitive trajectories in PD. METHODS We obtained clinical and multimodal MRI data from 120 control and 122 PD subjects without dementia or severe motor disability. T1/T2-weighted images estimated volume, and diffusion imaging evaluated fractional anisotropy (FA) of frontostriatal (striatum and frontostriatal white matter [FSWM]) and limbic (hippocampus and fornix) structures. Montreal Cognitive Assessment (MoCA) gauged total and domain-specific (attention/executive and memory) cognitive function. Linear mixed-effects models were used to compare MRI and cognitive progression over 4.5 years between controls and PD and evaluate associations between baseline MRI and cognitive changes in PD. RESULTS At baseline, control and PD groups were comparable, except PD participants had smaller striatal volume (p < 0.001). Longitudinally, PD showed faster decline in hippocampal volume, FSWM FA, and fornix FA (ps < .016), but not striatal volume (p = .218). Total and domain-specific MoCA scores declined faster in PD (ps < .030). In PD, lower baseline hippocampal volume (p = .005) and fornix FA (p = .032), but not striatal volume (p = .662) or FSWM FA (p = .143), were associated with faster total MoCA decline. Baseline frontostriatal metrics of striatal volume and FSWM FA were associated with faster attention/executive decline (p < .038), whereas lower baseline hippocampal volume was associated with faster memory decline (p = .005). CONCLUSION In PD, frontostriatal structural metrics are associated with attention/executive tasks, whereas limbic changes correlated with faster global cognitive decline, particularly in memory tasks.
Collapse
Affiliation(s)
- Gregory Brown
- Department of Neurology, Penn State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Jonathan Hakun
- Department of Neurology, Penn State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Mechelle M. Lewis
- Department of Neurology, Penn State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Sol De Jesus
- Department of Neurology, Penn State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Guangwei Du
- Department of Pharmacology, Penn State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Paul J. Eslinger
- Department of Neurology, Penn State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Lan Kong
- Department of Public Health Sciences, Penn State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Xuemei Huang
- Department of Neurology, Penn State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Department of Public Health Sciences, Penn State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Department of Neurosurgery, Penn State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Department of Kinesiology, Penn State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| |
Collapse
|
23
|
Bergamino M, Keeling EG, Ray NJ, Macerollo A, Silverdale M, Stokes AM. Structural connectivity and brain network analyses in Parkinson's disease: A cross-sectional and longitudinal study. Front Neurol 2023; 14:1137780. [PMID: 37034088 PMCID: PMC10076650 DOI: 10.3389/fneur.2023.1137780] [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: 01/04/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Parkinson's disease (PD) is an idiopathic disease of the central nervous system characterized by both motor and non-motor symptoms. It is the second most common neurodegenerative disease. Magnetic resonance imaging (MRI) can reveal underlying brain changes associated with PD. Objective In this study, structural connectivity and white matter networks were analyzed by diffusion MRI and graph theory in a cohort of patients with PD and a cohort of healthy controls (HC) obtained from the Parkinson's Progression Markers Initiative (PPMI) database in a cross-sectional analysis. Furthermore, we investigated longitudinal changes in the PD cohort over 36 months. Result Compared with the control group, participants with PD showed lower structural connectivity in several brain areas, including the corpus callosum, fornix, and uncinate fasciculus, which were also confirmed by a large effect-size. Additionally, altered connectivity between baseline and after 36 months was found in different network paths inside the white matter with a medium effect-size. Network analysis showed trends toward lower network density in PD compared with HC at baseline and after 36 months, though not significant after correction. Significant differences were observed in nodal degree and strength in several nodes. Conclusion In conclusion, altered structural and network metrics in several brain regions, such as corpus callosum, fornix, and cingulum were found in PD, compared to HC. We also report altered connectivity in the PD group after 36 months, reflecting the impact of both PD pathology and aging processes. These results indicate that structural and network metrics might yield insight into network reorganization that occurs in PD.
Collapse
Affiliation(s)
- Maurizio Bergamino
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, AZ, United States
- *Correspondence: Maurizio Bergamino
| | - Elizabeth G. Keeling
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, AZ, United States
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Nicola J. Ray
- Health, Psychology and Communities Research Centre, Department of Psychology, Manchester Metropolitan University, Manchester, United Kingdom
| | - Antonella Macerollo
- Neurology Department, The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom
- Institute of Systems, Molecular and Integrative Biology, School of Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Monty Silverdale
- Manchester Centre for Clinical Neurosciences, University of Manchester, Manchester, United Kingdom
| | - Ashley M. Stokes
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, AZ, United States
| |
Collapse
|
24
|
Harvey J, Reijnders RA, Cavill R, Duits A, Köhler S, Eijssen L, Rutten BPF, Shireby G, Torkamani A, Creese B, Leentjens AFG, Lunnon K, Pishva E. Machine learning-based prediction of cognitive outcomes in de novo Parkinson's disease. NPJ Parkinsons Dis 2022; 8:150. [PMID: 36344548 PMCID: PMC9640625 DOI: 10.1038/s41531-022-00409-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 10/11/2022] [Indexed: 11/09/2022] Open
Abstract
Cognitive impairment is a debilitating symptom in Parkinson's disease (PD). We aimed to establish an accurate multivariate machine learning (ML) model to predict cognitive outcome in newly diagnosed PD cases from the Parkinson's Progression Markers Initiative (PPMI). Annual cognitive assessments over an 8-year time span were used to define two cognitive outcomes of (i) cognitive impairment, and (ii) dementia conversion. Selected baseline variables were organized into three subsets of clinical, biofluid and genetic/epigenetic measures and tested using four different ML algorithms. Irrespective of the ML algorithm used, the models consisting of the clinical variables performed best and showed better prediction of cognitive impairment outcome over dementia conversion. We observed a marginal improvement in the prediction performance when clinical, biofluid, and epigenetic/genetic variables were all included in one model. Several cerebrospinal fluid measures and an epigenetic marker showed high predictive weighting in multiple models when included alongside clinical variables.
Collapse
Affiliation(s)
- Joshua Harvey
- Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Rick A Reijnders
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Rachel Cavill
- Department of Advanced Computing Sciences, FSE, Maastricht University, Maastricht, The Netherlands
| | - Annelien Duits
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Medical Psychology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sebastian Köhler
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Lars Eijssen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Bioinformatics-BiGCaT, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Gemma Shireby
- Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Ali Torkamani
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, 92037, USA
| | - Byron Creese
- Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Albert F G Leentjens
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Katie Lunnon
- Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Ehsan Pishva
- Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK.
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
25
|
Zhang J, Zhou C, Xiao X, Chen W, Jiang Y, Zhu R, Xin T. Magnetic resonance imaging image analysis of the therapeutic effect and neuroprotective effect of deep brain stimulation in Parkinson's disease based on a deep learning algorithm. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2022; 38:e3642. [PMID: 36054274 PMCID: PMC9786712 DOI: 10.1002/cnm.3642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 06/15/2023]
Abstract
In order to study the therapeutic neuroprotective effect of deep brain stimulation (DBS) in Parkinson's disease (PD), based on the deep learning algorithm, this study combines with magnetic resonance imaging (MRI) image analysis technology to study the clinical efficacy of DBS in the surgical treatment of PD and the neuroprotective and neurological recovery effects after surgery. Establish a deep learning algorithm model based on MRI image analysis technology, comparison of UPDRS motor status assessment and the improvement of daily life ability before and after DBS surgery, evaluate the accuracy rate and the detection speed of the model. The models constructed in this study have an accuracy rate of more than 90% in the PD detection test, and the detection speed of the algorithm model under the condition of big data is between 60 and 200 ms. DBS significantly improve a series of clinical symptoms in patients with PD. The deep learning algorithm model based on MRI image analysis technology in this paper has a certain effect. DBS operation can improve the symptoms of PD, and has the effect of neuroprotection and neurological recovery.
Collapse
Affiliation(s)
- Jianzhong Zhang
- Department of NeurosurgeryThe First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| | - Chaoyang Zhou
- Department of NeurosurgeryThe First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| | - Xiang Xiao
- Department of NeurosurgeryThe First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| | - Weihua Chen
- Department of ImagingThe First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| | - Yi Jiang
- Network Information CenterThe First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| | - Ronglan Zhu
- Department of NeurosurgeryThe First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| | - Tao Xin
- Department of NeurosurgeryThe First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| |
Collapse
|
26
|
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: 21] [Impact Index Per Article: 7.0] [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
|
27
|
Chung SJ, Kim YJ, Kim YJ, Lee HS, Yun M, Lee PH, Jeong Y, Sohn YH. Potential Link Between Cognition and Motor Reserve in Patients With Parkinson's Disease. J Mov Disord 2022; 15:249-257. [PMID: 36065615 DOI: 10.14802/jmd.22063] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/17/2022] [Indexed: 11/24/2022] Open
Abstract
Objective To investigate whether there is a link between cognitive function and motor reserve (i.e., individual capacity to cope with nigrostriatal dopamine depletion) in patients with newly diagnosed Parkinson's disease (PD). Methods A total of 163 patients with drug-naïve PD who underwent 18F-FP-CIT PET, brain MRI, and a detailed neuropsychological test were enrolled. We estimated individual motor reserve based on initial motor deficits and striatal dopamine depletion using a residual model. We performed correlation analyses between motor reserve estimates and cognitive composite scores. Diffusion connectometry analysis was performed to map the white matter fiber tracts, of which fractional anisotropy (FA) values were well correlated with motor reserve estimates. Additionally, Cox regression analysis was used to assess the effect of initial motor reserve on the risk of dementia conversion. Results The motor reserve estimate was positively correlated with the composite score of the verbal memory function domain (γ = 0.246) and with the years of education (γ = 0.251). Connectometry analysis showed that FA values in the left fornix were positively correlated with the motor reserve estimate, while no fiber tracts were negatively correlated with the motor reserve estimate. Cox regression analysis demonstrated that higher motor reserve estimates tended to be associated with a lower risk of dementia conversion (hazard ratio, 0.781; 95% confidence interval, 0.576-1.058). Conclusion The present study demonstrated that the motor reserve estimate was well correlated with verbal memory function and with white matter integrity in the left fornix, suggesting a possible link between cognition and motor reserve in patients with PD.
Collapse
Affiliation(s)
- Seok Jong Chung
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea.,Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, Korea.,YONSEI BEYOND LAB, Yongin, Korea
| | - Yae Ji Kim
- Program of Brain and Cognitive Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea.,KI for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Yun Joong Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea.,Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, Korea.,YONSEI BEYOND LAB, Yongin, Korea
| | - Hye Sun Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Korea
| | - Mijin Yun
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Yong Jeong
- YONSEI BEYOND LAB, Yongin, Korea.,Program of Brain and Cognitive Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea.,Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Young H Sohn
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
28
|
Tamura Y, Shimoji K, Ishikawa J, Murao Y, Yorikawa F, Kodera R, Oba K, Toyoshima K, Chiba Y, Tokumaru AM, Araki A. Association between white matter alterations on diffusion tensor imaging and incidence of frailty in older adults with cardiometabolic diseases. Front Aging Neurosci 2022; 14:912972. [PMID: 35966786 PMCID: PMC9363893 DOI: 10.3389/fnagi.2022.912972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/30/2022] [Indexed: 11/22/2022] Open
Abstract
Diffusion tensor imaging (DTI) can be used for the early detection of abnormal changes in the integrity of cerebral white matter tracts, and we have previously reported that these changes are associated with indices of early atherosclerotic lesions. Although these changes have been demonstrated to be associated with the incidence of frailty in older adults, no studies have investigated this relationship in patients at high risk for vascular disease. In this longitudinal study, we followed outpatients with cardiometabolic diseases for a maximum of 6 years (median, 3 years) and evaluated the association of baseline DTI data of seven white matter tracts with the incidence of frailty. The modified version of the Cardiovascular Health Study criteria and the Kihon Checklist were used as indices of frailty; fractional anisotropy (FA) and mean diffusivity (MD) were used as indices of white matter changes. Patients who developed frailty based on both indices had low FA and high MD in many of the tracts tested, with the most significant difference found in the MD of the anterior thalamic radiation (ATR). Cox proportional hazard model analysis revealed a significantly high risk of frailty defined by both indices in the groups with high MD values in the left ATR. Similar results were found in patients with diabetes mellitus but not in those without diabetes mellitus. Therefore, abnormalities in the integrity of the left ATR could be associated with the progression of frailty in older adults with cardiometabolic disease, particularly those with diabetes mellitus.
Collapse
Affiliation(s)
- Yoshiaki Tamura
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
- The Center for Comprehensive Care and Research for Prefrailty, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
- *Correspondence: Yoshiaki Tamura
| | - Keigo Shimoji
- Department of Diagnostic Radiology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Joji Ishikawa
- The Center for Comprehensive Care and Research for Prefrailty, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
- Department of Cardiology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Yuji Murao
- The Center for Comprehensive Care and Research for Prefrailty, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Fumino Yorikawa
- The Center for Comprehensive Care and Research for Prefrailty, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Remi Kodera
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Kazuhito Oba
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
- The Center for Comprehensive Care and Research for Prefrailty, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Kenji Toyoshima
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
- The Center for Comprehensive Care and Research for Prefrailty, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Yuko Chiba
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Aya M. Tokumaru
- Department of Diagnostic Radiology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Atsushi Araki
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
- The Center for Comprehensive Care and Research for Prefrailty, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| |
Collapse
|
29
|
Chen J, Jiang X, Wu J, Wu H, Zhou C, Guo T, Bai X, Liu X, Wen J, Cao Z, Gu L, Yang W, Pu J, Guan X, Xu X, Zhang B, Zhang M. Gray and white matter alterations in different predominant side and type of motor symptom in Parkinson's disease. CNS Neurosci Ther 2022; 28:1372-1379. [PMID: 35673762 PMCID: PMC9344082 DOI: 10.1111/cns.13877] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/12/2022] [Accepted: 05/20/2022] [Indexed: 11/29/2022] Open
Abstract
Background Parkinson's disease (PD) is highly heterogeneous reflected by different affected side of body and type of motor symptom. We aim to explore clinical characteristics and underlying brain structure alterations in PD with different predominant sides and motor types. Methods We recruited 161 PD patients and 50 healthy controls (HC). Patients were classified into four subtypes according to their predominant side and motor type: left akinetic/rigid‐dominant (LAR), left tremor‐dominant (LTD), right akinetic/rigid‐dominant (RAR), and right tremor‐dominant (RTD). All participants assessed motor and cognitive performances, then underwent T1‐weighted and diffusion tensor imaging scanning. A general linear model was used to compare neuroimaging parameters among five groups. Results Among four PD subtypes, patients of LAR subtype experienced the worst motor impairment, and only this subtype showed worse cognitive performance compared with HC. Compared with HC and other subtypes, LAR subtype showed a significant reduction in cortical thickness of the right caudal‐anterior‐cingulate gyrus and fractional anisotropy of the right cingulum bundle. Conclusions We demonstrated that LAR subtype had the worst clinical performance, which the severer damage in the right cingulate region might be the underlying mechanism. This study underscores the importance of classifying PD subtypes based on both the side and type of motor symptom for clinical intervention and research to optimize behavioral outcomes in the future.
Collapse
Affiliation(s)
- Jingwen Chen
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou, China
| | - Xianchen Jiang
- Quzhou Center for Disease Control and Prevention, Quzhou, China
| | - Jingjing Wu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou, China
| | - Haoting Wu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou, China
| | - Cheng Zhou
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou, China
| | - Tao Guo
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou, China
| | - Xueqin Bai
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou, China
| | - Xiaocao Liu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou, China
| | - Jiaqi Wen
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou, China
| | - Zhengye Cao
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou, China
| | - Luyan Gu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou, China
| | - Wenyi Yang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou, China
| | - Jiali Pu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou, China
| | - Xiaojun Guan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou, China
| | - Xiaojun Xu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou, China
| | - Baorong Zhang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou, China
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University of Medicine, Hangzhou, China
| |
Collapse
|
30
|
Birk F, Glang F, Loktyushin A, Birkl C, Ehses P, Scheffler K, Heule R. High-resolution neural network-driven mapping of multiple diffusion metrics leveraging asymmetries in the balanced steady-state free precession frequency profile. NMR IN BIOMEDICINE 2022; 35:e4669. [PMID: 34964998 DOI: 10.1002/nbm.4669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/04/2021] [Accepted: 12/06/2021] [Indexed: 06/14/2023]
Abstract
We propose to utilize the rich information content about microstructural tissue properties entangled in asymmetric balanced steady-state free precession (bSSFP) profiles to estimate multiple diffusion metrics simultaneously by neural network (NN) parameter quantification. A 12-point bSSFP phase-cycling scheme with high-resolution whole-brain coverage is employed at 3 and 9.4 T for NN input. Low-resolution target diffusion data are derived based on diffusion-weighted spin-echo echo-planar-imaging (SE-EPI) scans, that is, mean, axial, and radial diffusivity (MD, AD, and RD), fractional anisotropy (FA), as well as the spherical coordinates (azimuth Φ and inclination ϴ) of the principal diffusion eigenvector. A feedforward NN is trained with incorporated probabilistic uncertainty estimation. The NN predictions yielded highly reliable results in white matter (WM) and gray matter structures for MD. The quantification of FA, AD, and RD was overall in good agreement with the reference but the dependence of these parameters on WM anisotropy was somewhat biased (e.g. in corpus callosum). The inclination ϴ was well predicted for anisotropic WM structures, while the azimuth Φ was overall poorly predicted. The findings were highly consistent across both field strengths. Application of the optimized NN to high-resolution input data provided whole-brain maps with rich structural details. In conclusion, the proposed NN-driven approach showed potential to provide distortion-free high-resolution whole-brain maps of multiple diffusion metrics at high to ultrahigh field strengths in clinically relevant scan times.
Collapse
Affiliation(s)
- Florian Birk
- High Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Felix Glang
- High Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Alexander Loktyushin
- High Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
- Empirical Inference, Max Planck Institute for Intelligent Systems, Tübingen, Germany
| | - Christoph Birkl
- Department of Neuroradiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Philipp Ehses
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Klaus Scheffler
- High Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
- Department of Biomedical Magnetic Resonance, University of Tübingen, Tübingen, Germany
| | - Rahel Heule
- High Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
- Department of Biomedical Magnetic Resonance, University of Tübingen, Tübingen, Germany
| |
Collapse
|
31
|
Narayanan NS, Albin RL. The way forward for cognition in Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2022; 269:457-462. [PMID: 35248206 DOI: 10.1016/bs.pbr.2022.01.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This is the concluding chapter in our volume on cognition in Parkinson's disease.
Collapse
Affiliation(s)
| | - Roger L Albin
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
32
|
Tian Q, Li Z, Fan Q, Polimeni JR, Bilgic B, Salat DH, Huang SY. SDnDTI: Self-supervised deep learning-based denoising for diffusion tensor MRI. Neuroimage 2022; 253:119033. [PMID: 35240299 DOI: 10.1016/j.neuroimage.2022.119033] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 02/15/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Diffusion tensor magnetic resonance imaging (DTI) is a widely adopted neuroimaging method for the in vivo mapping of brain tissue microstructure and white matter tracts. Nonetheless, the noise in the diffusion-weighted images (DWIs) decreases the accuracy and precision of DTI derived microstructural parameters and leads to prolonged acquisition time for achieving improved signal-to-noise ratio (SNR). Deep learning-based image denoising using convolutional neural networks (CNNs) has superior performance but often requires additional high-SNR data for supervising the training of CNNs, which reduces the feasibility of supervised learning-based denoising in practice. In this work, we develop a self-supervised deep learning-based method entitled "SDnDTI" for denoising DTI data, which does not require additional high-SNR data for training. Specifically, SDnDTI divides multi-directional DTI data into many subsets of six DWI volumes and transforms DWIs from each subset to along the same diffusion-encoding directions through the diffusion tensor model, generating multiple repetitions of DWIs with identical image contrasts but different noise observations. SDnDTI removes noise by first denoising each repetition of DWIs using a deep 3-dimensional CNN with the average of all repetitions with higher SNR as the training target, following the same approach as normal supervised learning based denoising methods, and then averaging CNN-denoised images for achieving higher SNR. The denoising efficacy of SDnDTI is demonstrated in terms of the similarity of output images and resultant DTI metrics compared to the ground truth generated using substantially more DWI volumes on two datasets with different spatial resolutions, b-values and numbers of input DWI volumes provided by the Human Connectome Project (HCP) and the Lifespan HCP in Aging. The SDnDTI results preserve image sharpness and textural details and substantially improve upon those from the raw data. The results of SDnDTI are comparable to those from supervised learning-based denoising and outperform those from state-of-the-art conventional denoising algorithms including BM4D, AONLM and MPPCA. By leveraging domain knowledge of diffusion MRI physics, SDnDTI makes it easier to use CNN-based denoising methods in practice and has the potential to benefit a wider range of research and clinical applications that require accelerated DTI acquisition and high-quality DTI data for mapping of tissue microstructure, fiber tracts and structural connectivity in the living human brain.
Collapse
Affiliation(s)
- Qiyuan Tian
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 13th Street, Charlestown, MA 02129, United States; Department of Radiology, Harvard Medical School, Boston, MA, United States.
| | - Ziyu Li
- Department of Biomedical Engineering, Tsinghua University, Beijing, PR China
| | - Qiuyun Fan
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 13th Street, Charlestown, MA 02129, United States; Department of Radiology, Harvard Medical School, Boston, MA, United States
| | - Jonathan R Polimeni
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 13th Street, Charlestown, MA 02129, United States; Department of Radiology, Harvard Medical School, Boston, MA, United States; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Berkin Bilgic
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 13th Street, Charlestown, MA 02129, United States; Department of Radiology, Harvard Medical School, Boston, MA, United States; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - David H Salat
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 13th Street, Charlestown, MA 02129, United States; Department of Radiology, Harvard Medical School, Boston, MA, United States
| | - Susie Y Huang
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 13th Street, Charlestown, MA 02129, United States; Department of Radiology, Harvard Medical School, Boston, MA, United States; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
33
|
Kan H, Uchida Y, Ueki Y, Arai N, Tsubokura S, Kunitomo H, Kasai H, Aoyama K, Matsukawa N, Shibamoto Y. R2* relaxometry analysis for mapping of white matter alteration in Parkinson's disease with mild cognitive impairment. Neuroimage Clin 2022; 33:102938. [PMID: 34998126 PMCID: PMC8741619 DOI: 10.1016/j.nicl.2022.102938] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/22/2021] [Accepted: 01/03/2022] [Indexed: 12/01/2022]
Abstract
R2* relaxometry analysis combined with QSM revealed detail of WM alteration in PD-MCI. R2* relaxometry analysis can detect slight demyelination in PD-MCI. R2* value shows potential for early evaluation of cognitive decline in PD.
Background R2* relaxometry analysis combined with quantitative susceptibility mapping (QSM), which has high sensitivity to iron deposition, can distinguish microstructural changes of the white matter (WM) and iron deposition, thereby providing a sensitive and biologically specific measure of the WM owing to the changes in myelin and its surrounding environment. This study aimed to explore the microstructural WM alterations associated with cognitive impairment in patients with Parkinson’s disease (PD) using R2* relaxometry analysis combined with QSM. Materials and methods We enrolled 24 patients with PD and mild cognitive impairment (PD-MCI), 22 patients with PD and normal cognition (PD-CN), and 19 age- and sex-matched healthy controls (HC). All participants underwent Montreal Cognitive Assessment (MoCA) and brain magnetic resonance imaging, including structural three-dimensional T1-weighted images and multiple spoiled gradient echo sequence (mGRE). The R2* and susceptibility maps were estimated from the multiple magnitude images of mGRE. The susceptibility maps were used for verifying iron deposition in the WM. The voxel-based R2* of the entire WM and its correlation with cognitive performance were analyzed. Results In the voxel-based group comparisons, the R2* in the PD-MCI group was lower in some WM regions, including the corpus callosum, than R2* in the PD-CN and HC groups. The mean susceptibility values in almost all brain regions were negative and close-to-zero values, indicating no detectable paramagnetic iron deposition in the WM of all subjects. There was a significant positive correlation between R2* and MoCA in some regions of the WM, mainly the corpus callosum and left hemisphere. Conclusion R2* relaxometry analysis for WM microstructural changes provided further biologic insights on demyelination and changes in the surrounding environment, supported by the QSM results demonstrating no iron existence. This analysis highlighted the potential for the early evaluation of cognitive decline in patients with PD.
Collapse
Affiliation(s)
- Hirohito Kan
- Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Japan; Department of Radiology, Nagoya City University, Graduate School of Medical Sciences, Japan.
| | - Yuto Uchida
- Department of Neurology, Nagoya City University, Graduate School of Medical Sciences, Japan; Department of Neurology, Toyokawa City Hospital, Japan.
| | - Yoshino Ueki
- Department of Rehabilitation Medicine, Nagoya City University, Graduate School of Medical Sciences, Japan.
| | - Nobuyuki Arai
- Department of Radiology, Suzuka University of Medical Science, Japan.
| | | | - Hiroshi Kunitomo
- Department of Radiology, Nagoya City University Hospital, Japan.
| | - Harumasa Kasai
- Department of Radiology, Nagoya City University Hospital, Japan
| | - Kiminori Aoyama
- Department of Rehabilitation Medicine, Nagoya City University, Graduate School of Medical Sciences, Japan
| | - Noriyuki Matsukawa
- Department of Neurology, Nagoya City University, Graduate School of Medical Sciences, Japan.
| | - Yuta Shibamoto
- Department of Radiology, Nagoya City University, Graduate School of Medical Sciences, Japan.
| |
Collapse
|
34
|
Arévalo-Sáenz A, López-Manzanares L, Navas-García M, Pastor J, Vega-Zelaya L, Torres CV. Deep brain stimulation in Parkinson's disease: analysis of brain fractional anisotropy differences in operated patients. Rev Neurol 2022; 74:125-134. [PMID: 35148421 PMCID: PMC11502176 DOI: 10.33588/rn.7404.2021196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Deep brain stimulation (DBS) of the subthalamic nucleus is currently an evidence-based therapeutic option for motor symptoms in patients with Parkinson's disease (PD), although other non-motor symptoms can be affected by stimulation. AIM Our objective is to evaluate the global changes in the connectivity of the large-scale structural network in PD patients that have obtained a benefit from subthalamic DBS. SUBJECTS AND METHODS Retrospective study of 31 subjects: 7 PD patients with subthalamic DBS (group A), 12 age and gender-matched non-operated PD (B) and 12 healthy controls (C). All subjects had undergone a 1.5 T brain MRI with DTI. DICOM images were processed with the FSL5.0 software and TBSS tool. RESULTS The study group comprised 23 men and 8 women. No statistically significant differences in age, gender, scores on the HandY scale and mean follow-up between group A and B were found, and in age and gender between groups A and C. Statistical analysis revealed differences in the fractional anisotropy of the different groups in certain areas: bilateral corticospinal tract, anterior thalamic radiations, bilateral fronto-occipital fascicle, both superior longitudinal fascicles, and left inferior longitudinal fascicle. CONCLUSIONS In our series, PD patients treated with bilateral subthalamic DBS showed a significantly higher fractional anisotropy in widespread areas of the cerebral white matter; suggesting that neuromodulation produces connectivity changes in different neural networks.
Collapse
Affiliation(s)
- Alejandra Arévalo-Sáenz
- Servicio de Neurocirugía. Hospital Clínico San Carlos (A. Arévalo-Sáenz)Hospital Clínico San CarlosHospital Clínico San CarlosMadridEspaña
| | - Lydia López-Manzanares
- Servicio de Neurología (L. López-Manzanares)Servicio de NeurologíaServicio de NeurologíaMadridEspaña
| | - Marta Navas-García
- Servicio de Neurocirugía (M. Navas, C.V. Torres)Servicio de NeurocirugíaServicio de NeurocirugíaMadridEspaña
| | - Jesús Pastor
- Servicio de Neurofisiología. Hospital Universitario de la Princesa. Madrid, España (J. Pastor, L. Vega-Zelaya)Hospital Universitario de la PrincesaHospital Universitario de la PrincesaMadridEspaña
| | - Lorena Vega-Zelaya
- Servicio de Neurofisiología. Hospital Universitario de la Princesa. Madrid, España (J. Pastor, L. Vega-Zelaya)Hospital Universitario de la PrincesaHospital Universitario de la PrincesaMadridEspaña
| | - Cristina V. Torres
- Servicio de Neurocirugía (M. Navas, C.V. Torres)Servicio de NeurocirugíaServicio de NeurocirugíaMadridEspaña
| |
Collapse
|
35
|
Uhr L, Tsolaki E, Pouratian N. Diffusion tensor imaging correlates of depressive symptoms in Parkinson disease. J Comp Neurol 2022; 530:1729-1738. [DOI: 10.1002/cne.25310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/30/2022] [Accepted: 02/03/2022] [Indexed: 11/10/2022]
Affiliation(s)
- Lauren Uhr
- Department of Neurosurgery David Geffen School of Medicine at UCLA Los Angeles California
| | - Evangelia Tsolaki
- Department of Neurosurgery David Geffen School of Medicine at UCLA Los Angeles California
| | - Nader Pouratian
- Department of Neurological Surgery UT Southwestern Medical Center Dallas Texas
| |
Collapse
|
36
|
Patriat R, Pisharady PK, Amundsen-Huffmaster S, Linn-Evans M, Howell M, Chung JW, Petrucci MN, Videnovic A, Holker E, De Kam J, Tuite P, Lenglet C, Harel N, MacKinnon CD. White matter microstructure in Parkinson's disease with and without elevated rapid eye movement sleep muscle tone. Brain Commun 2022; 4:fcac027. [PMID: 35310831 PMCID: PMC8924652 DOI: 10.1093/braincomms/fcac027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/06/2021] [Accepted: 02/07/2022] [Indexed: 11/28/2022] Open
Abstract
People with Parkinson's disease who have elevated muscle activity during rapid eye movement sleep (REM sleep without atonia) typically have a worse motor and cognitive impairment compared with those with normal muscle atonia during rapid eye movement sleep. This study used tract-based spatial statistics to compare diffusion MRI measures of fractional anisotropy, radial, mean and axial diffusivity (measures of axonal microstructure based on the directionality of water diffusion) in white matter tracts between people with Parkinson's disease with and without rapid eye movement sleep without atonia and controls and their relationship to measures of motor and cognitive function. Thirty-eight individuals with mild-to-moderate Parkinson's disease and 21 matched control subjects underwent ultra-high field MRI (7 T), quantitative motor assessments of gait and bradykinesia and neuropsychological testing. The Parkinson's disease cohort was separated post hoc into those with and without elevated chin or leg muscle activity during rapid eye movement sleep based on polysomnography findings. Fractional anisotropy was significantly higher, and diffusivity significantly lower, in regions of the corpus callosum, projection and association white matter pathways in the Parkinson's group with normal rapid eye movement sleep muscle tone compared with controls, and in a subset of pathways relative to the Parkinson's disease group with rapid eye movement sleep without atonia. The Parkinson's disease group with elevated rapid eye movement sleep muscle tone showed significant impairments in the gait and upper arm speed compared with controls and significantly worse scores in specific cognitive domains (executive function, visuospatial memory) compared with the Parkinson's disease group with normal rapid eye movement sleep muscle tone. Regression analyses showed that gait speed and step length in the Parkinson's disease cohort were predicted by measures of fractional anisotropy of the anterior corona radiata, whereas elbow flexion velocity was predicted by fractional anisotropy of the superior corona radiata. Visuospatial memory task performance was predicted by the radial diffusivity of the posterior corona radiata. These findings show that people with mild-to-moderate severity of Parkinson's disease who have normal muscle tone during rapid eye movement sleep demonstrate compensatory-like adaptations in axonal microstructure that are associated with preserved motor and cognitive function, but these adaptations are reduced or absent in those with increased rapid eye movement sleep motor tone.
Collapse
Affiliation(s)
- Rémi Patriat
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, USA
| | - Pramod K. Pisharady
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, USA
| | | | - Maria Linn-Evans
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Michael Howell
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | - Jae Woo Chung
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | | | | | - Erin Holker
- Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Joshua De Kam
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | - Paul Tuite
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | - Christophe Lenglet
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, USA
| | - Noam Harel
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, USA
| | | |
Collapse
|
37
|
Ni K, Liu Y, Zhu X, Tan H, Zeng Y, Guo Q, Xiao L, Yu B. Changed Cerebral White Matter Structural Network Topological Characters and Its Correlation with Cognitive Behavioral Abnormalities in Narcolepsy Type 1. Nat Sci Sleep 2022; 14:165-173. [PMID: 35140538 PMCID: PMC8818963 DOI: 10.2147/nss.s336967] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/19/2022] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE In the current study we investigated topological abnormalities of the cerebral white matter networks in narcolepsy type 1 (NT1) patients and its relationship with their cognitive abnormalities using diffusion tensor imaging (DTI) technology. METHODS DTI and the Beijing version of the Montreal Cognitive Assessment (MoCA-BJ) were applied to 30 NT1 patients and 30 age-matched healthy controls. DTI studies were also carried using the 3T MRI system. Next, DTI data was used to establish a cerebral white matter network for all subjects and graph theory was applied to analyze the topological characteristics of the white matter structural network. Topographical parameters (such as local efficiency (Eloc), global efficiency (Eglob) and small-world (σ)) between NT1 patients and controls were then compared. The correlation between MoCA-BJ scores and topological parameters was also analyzed. RESULTS MoCA-BJ scores in NT1 patients were lower than those in the healthy controls. Compared with healthy controls, the global efficiency of the white matter network and attributes of the small world network were significantly reduced in NT1 patients. Finally, the global efficiency of the white matter structural network was related to the MoCA-BJ score of NT1 patients. CONCLUSION The abnormal topological characteristics of the white matter structural network in NT1 patients may be associated with their cognitive impairment.
Collapse
Affiliation(s)
- Kunlin Ni
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People’s Republic of China
| | - Yishu Liu
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, People’s Republic of China
| | - Xiaoyu Zhu
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People’s Republic of China
| | - Huiwen Tan
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, People’s Republic of China
| | - Yin Zeng
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, People’s Republic of China
| | - Qiyong Guo
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People’s Republic of China
| | - Li Xiao
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, People’s Republic of China
- Sleep Medicine Center, Shengjing Hospital of China Medical University, Shenyang, 110004, People’s Republic of China
| | - Bing Yu
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People’s Republic of China
| |
Collapse
|
38
|
Sang T, He J, Wang J, Zhang C, Zhou W, Zeng Q, Yuan Y, Yu L, Feng Y. Alterations in white matter fiber in Parkinson's disease across different cognitive stages. Neurosci Lett 2021; 769:136424. [PMID: 34958911 DOI: 10.1016/j.neulet.2021.136424] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 02/03/2023]
Affiliation(s)
- Tian Sang
- Institute of Information Processing and Automation, College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, China; Zhejiang Provincial United Key Laboratory of Embedded Systems, Hangzhou 310023, China
| | - Jianzhong He
- Institute of Information Processing and Automation, College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, China; Zhejiang Provincial United Key Laboratory of Embedded Systems, Hangzhou 310023, China
| | - Jingqiang Wang
- Institute of Information Processing and Automation, College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, China; Zhejiang Provincial United Key Laboratory of Embedded Systems, Hangzhou 310023, China
| | - Chengzhe Zhang
- Institute of Information Processing and Automation, College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, China; Zhejiang Provincial United Key Laboratory of Embedded Systems, Hangzhou 310023, China
| | - Wenyang Zhou
- Institute of Information Processing and Automation, College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, China; Zhejiang Provincial United Key Laboratory of Embedded Systems, Hangzhou 310023, China
| | - Qingrun Zeng
- Institute of Information Processing and Automation, College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, China; Zhejiang Provincial United Key Laboratory of Embedded Systems, Hangzhou 310023, China
| | - Yuan Yuan
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou 310003, China
| | - Lihua Yu
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou 310003, China
| | - Yuanjing Feng
- Institute of Information Processing and Automation, College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, China; Zhejiang Provincial United Key Laboratory of Embedded Systems, Hangzhou 310023, China.
| |
Collapse
|
39
|
Dolatshahi M, Ashraf-Ganjouei A, Wu IW, Zhang Y, Aarabi MH, Tosun D. White matter changes in drug-naïve Parkinson's disease patients with impulse control & probable REM sleep behavior disorders. J Neurol Sci 2021; 430:120032. [PMID: 34688191 DOI: 10.1016/j.jns.2021.120032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 09/24/2021] [Accepted: 10/15/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND According to epidemiological studies, Parkinson's disease (PD) patients with probable REM sleep behavior disorder (pRBD) are more prone to develop impulse control disorders (ICDs), which is shown to be present in drug-naïve PD patients, and vice versa. OBJECTIVES To investigate white-matter integrity differences, with and without comorbid pRBD and ICDs. METHODS 149 de-novo PD patients and 30 age- and gender-matched controls from the Parkinson's Progression Markers Initiative were studied. PD subjects were categorized into four groups with and without these comorbidities. We investigated the white matter integrity differences between these groups. RESULTS PDs with only ICDs manifested greater fractional anisotropy (FA) and lower mean diffusivity (MD) in ipsilateral cerebellar connections when compared to controls and to Parkinson's with both comorbid disorders. In contrast, significantly lower FA and higher MD in the ipsilateral fornix-stria-terminalis was observed in PDs with only pRBD compared to controls and to PDs without either comorbid disorder. Also, PDs with only pRBD manifested greater FA in contralateral putamen when compared to controls. CONCLUSIONS Our results suggest the presence of an underlying neural network in PDs with ICDs, particularly involving cerebellar connections, which makes the subjects susceptible to pRBD. Lower white-matter integrity in the fornix of PDs with only pRBD suggests a neuropathological pathway specific to sleep behavior disorder, independent of impulse control disorders. Greater white-matter integrity observed in PDs without comorbid ICDs, regardless of their comorbid pRBD status, might reflect compensatory mechanisms. Targeted therapies for this particular neuropathology may help prevent these comorbidities.
Collapse
Affiliation(s)
- Mahsa Dolatshahi
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran; NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | | | - I-Wei Wu
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Yu Zhang
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
| | - Mohammad Hadi Aarabi
- Department of Neuroscience, Padova Neuroscience Center (PNC), University of Padova, Padova, Italy
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States.
| | | |
Collapse
|
40
|
Ferreira H, Amorim D, Lima AC, Pirraco RP, Costa-Pinto AR, Almeida R, Almeida A, Reis RL, Pinto-Ribeiro F, Neves NM. A biocompatible and injectable hydrogel to boost the efficacy of stem cells in neurodegenerative diseases treatment. Life Sci 2021; 287:120108. [PMID: 34717909 DOI: 10.1016/j.lfs.2021.120108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 01/03/2023]
Abstract
AIMS Stem cell therapies emerged as treatment modalities with potential to cure neurodegenerative diseases (NDs). However, despite high expectations, their clinical use is still limited. Critical issues in treatment outcomes may be related to stem cells formulation and administration route. We develop a hydrogel as a cell carrier, consisting of compounds (phospholipids and hyaluronic acid-HA) naturally present in the central nervous system (CNS). The HA-based hydrogel physically crosslinked with liposomes is designed for direct injection into the CNS to significantly increase the bone marrow mesenchymal stem cells (BMSCs) bioavailability. MATERIALS AND METHODS Hydrogel compatibility is confirmed in vitro with BMSCs and in vivo through its intracerebroventricular injection in rats. To assess its efficacy, the main cause of chronic neurologic disability in young adults is selected, namely multiple sclerosis (MS). The efficacy of the developed formulation containing a lower number of cells than previously reported is demonstrated using an experimental autoimmune encephalomyelitis (EAE) rat model. KEY FINDINGS The distribution of the engineered hydrogel into corpus callosum can be ideal for NDs treatment, since damage of this white matter structure is responsible for important neuronal deficits. Moreover, the BMSCs-laden hydrogel significantly decreases disease severity and maximum clinical score and eliminated the relapse. SIGNIFICANCE The engineering of advanced therapies using this natural carrier can result in efficacious treatments for MS and related debilitating conditions.
Collapse
Affiliation(s)
- Helena Ferreira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Diana Amorim
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Life and Health Sciences Research Institute, School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Ana Cláudia Lima
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rogério P Pirraco
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Rita Costa-Pinto
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui Almeida
- Neurosurgery Department, Hospital de Braga, Braga, Portugal
| | - Armando Almeida
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Life and Health Sciences Research Institute, School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Filipa Pinto-Ribeiro
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Life and Health Sciences Research Institute, School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Nuno M Neves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
41
|
Tamura Y, Shimoji K, Ishikawa J, Matsuo Y, Watanabe S, Takahashi H, Zen S, Tachibana A, Omura T, Kodera R, Oba K, Toyoshima K, Chiba Y, Tokumaru AM, Araki A. Subclinical Atherosclerosis, Vascular Risk Factors, and White Matter Alterations in Diffusion Tensor Imaging Findings of Older Adults With Cardiometabolic Diseases. Front Aging Neurosci 2021; 13:712385. [PMID: 34489681 PMCID: PMC8417784 DOI: 10.3389/fnagi.2021.712385] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/27/2021] [Indexed: 11/13/2022] Open
Abstract
White matter abnormalities may reflect cerebral microvessel disease. Diffusion tensor imaging (DTI) can help detect early changes in white matter integrity in each tract. However, studies investigating the relationship between subclinical atherosclerosis markers and white matter alterations in DTI findings are limited. This study aimed to examine associations between cardiovascular risk factors and indices of subclinical atherosclerosis-ankle brachial index (ABI), brachial-ankle pulse wave velocity (baPWV), and carotid artery intima-media thickness (IMT)-and altered white matter integrity in older patients. A total of 224 patients (aged ≥65 years) with cardiometabolic disease who underwent magnetic resonance imaging (MRI) and either plethysmography or cervical ultrasound at the start of the 3-year observational study period were included in this study. We measured fractional anisotropy (FA) and mean diffusivity (MD), which are indices of white matter integrity in seven white matter tracts. In a univariate analysis, lower ABI and higher baPWV values were associated with FA or MD abnormalities in several tracts, whereas IMT was scarcely associated with such change. In addition, high blood pressure and glycoalbumin/glycohemoglobin ratio (GA/HbA1c) and low body mass index (BMI) and triglyceride (TG) levels were associated with FA or MD abnormalities. In a multivariate analysis adjusted for age, sex, BMI, diastolic blood pressure, TG, and GA/HbA1c, the associations between ABI and FA or MD remained in all of either side of the following tracts: anterior thalamic radiation, forceps minor, inferior frontooccipital fasciculus (p < 0.001 for all) and superior longitudinal fasciculus (SLF; p < 0.05), whereas most of those between baPWV and FA or MD disappeared except for SLF (p < 0.05). These results indicate that low ABI could be an indicator of white matter abnormalities.
Collapse
Affiliation(s)
- Yoshiaki Tamura
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Keigo Shimoji
- Department of Diagnostic Radiology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Joji Ishikawa
- Department of Cardiology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Yoshinori Matsuo
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - So Watanabe
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Hisae Takahashi
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Shugo Zen
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Aya Tachibana
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Takuya Omura
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Remi Kodera
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Kazuhito Oba
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Kenji Toyoshima
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Yuko Chiba
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Aya M Tokumaru
- Department of Diagnostic Radiology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Atsushi Araki
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| |
Collapse
|
42
|
Yoo HS, Kwon H, Chung SJ, Sohn YH, Lee JM, Lee PH. Neural correlates of self-awareness of cognitive deficits in non-demented patients with Parkinson's disease. Eur J Neurol 2021; 28:4022-4030. [PMID: 34478599 DOI: 10.1111/ene.15095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND PURPOSE The aim was to investigate the neural correlates of impaired self-awareness of cognitive deficits (IACd) in non-demented patients with Parkinson's disease (PD). METHODS This cross-sectional study enrolled 153 drug-naïve and non-demented PD patients who underwent brain magnetic resonance imaging, dopamine transporter (DAT) positron emission tomography, detailed neuropsychological testing, and the Cognitive Complaints Interview at baseline. Based on the presence of mild cognitive impairment and subjective cognitive complaints, patients were grouped into those with IACd (PD-IACd+, n = 33) and those with normal recognition of cognitive function (n = 82) or underestimation of cognitive function (n = 38). Cortical thickness, white matter (WM) integrity, DAT availability and cognitive function were compared between the groups. RESULTS The prevalence of IACd was 21.6% in drug-naïve patients with PD. The PD-IACd+ group had a lower z-score in the Stroop color reading test than the other groups. Patients in the PD-IACd+ group had WM disintegrity, especially in the genu of the corpus callosum and anterior limb of the internal capsule, compared to those without IACd, whilst cortical thickness or striatal DAT availability was comparable regardless of the presence of IACd. Amongst patients with mild cognitive impairment, those with IACd had more severe WM disintegrity than those without IACd. CONCLUSION Structural connectivity between and from the frontal lobes is closely associated with self-awareness of cognitive deficits in PD. Evaluating frontal structural connectivity from the early stages of PD will be important in assessing the actual cognitive and daily life performance of patients with PD.
Collapse
Affiliation(s)
- Han Soo Yoo
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyeokjin Kwon
- Department of Electronic Engineering, Hanyang University, Seoul, South Korea
| | - Seok Jong Chung
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Young H Sohn
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Jong-Min Lee
- Department of Biomedical Engineering, Hanyang University, Seoul, South Korea
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
43
|
Yu B, Li L, Guan X, Xu X, Liu X, Yang Q, Wei H, Zuo C, Zhang Y. HybraPD atlas: Towards precise subcortical nuclei segmentation using multimodality medical images in patients with Parkinson disease. Hum Brain Mapp 2021; 42:4399-4421. [PMID: 34101297 PMCID: PMC8357000 DOI: 10.1002/hbm.25556] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 05/27/2021] [Accepted: 05/30/2021] [Indexed: 12/29/2022] Open
Abstract
Human brain atlases are essential for research and surgical treatment of Parkinson's disease (PD). For example, deep brain stimulation for PD often requires human brain atlases for brain structure identification. However, few atlases can provide disease-specific subcortical structures for PD, and most of them are based on T1w and T2w images. In this work, we construct a HybraPD atlas using fused quantitative susceptibility mapping (QSM) and T1w images from 87 patients with PD. The constructed HybraPD atlas provides a series of templates, that is, T1w, GRE magnitude, QSM, R2*, and brain tissue probabilistic maps. Then, we manually delineate a parcellation map with 12 bilateral subcortical nuclei, which are highly related to PD pathology, such as sub-regions in globus pallidus and substantia nigra. Furthermore, we build a whole-brain parcellation map by combining existing cortical parcellation and white-matter segmentation with the proposed subcortical nuclei map. Considering the multimodality of the HybraPD atlas, the segmentation accuracy of each nucleus is evaluated using T1w and QSM templates, respectively. The results show that the HybraPD atlas provides more accurate segmentation than existing atlases. Moreover, we analyze the metabolic difference in subcortical nuclei between PD patients and healthy control subjects by applying the HybraPD atlas to calculate uptake values of contrast agents on positron emission tomography (PET) images. The atlas-based analysis generates accurate disease-related brain nuclei segmentation on PET images. The newly developed HybraPD atlas could serve as an efficient template to study brain pathological alterations in subcortical regions for PD research.
Collapse
Affiliation(s)
- Boliang Yu
- School of Information Science and TechnologyShanghaiTech UniversityShanghaiChina
| | - Ling Li
- PET Center, Huashan HospitalFudan UniversityShanghaiChina
| | - Xiaojun Guan
- Department of Radiology, The Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Xiaojun Xu
- Department of Radiology, The Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Xueling Liu
- Department of Radiology, Huashan HospitalFudan UniversityShanghaiChina
| | - Qing Yang
- Institute of Brain‐Intelligence Technology, Zhangjiang LaboratoryShanghaiChina
| | - Hongjiang Wei
- Institute for Medicine Imaging Technology, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Chuantao Zuo
- PET Center, Huashan HospitalFudan UniversityShanghaiChina
| | - Yuyao Zhang
- School of Information Science and TechnologyShanghaiTech UniversityShanghaiChina
- Shanghai Engineering Research Center of Intelligent Vision and ImagingShanghaiTech UniversityShanghaiChina
| |
Collapse
|
44
|
Zhang Y, Huang B, Chen Q, Wang L, Zhang L, Nie K, Huang Q, Huang R. Altered microstructural properties of superficial white matter in patients with Parkinson's disease. Brain Imaging Behav 2021; 16:476-491. [PMID: 34410610 DOI: 10.1007/s11682-021-00522-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2021] [Indexed: 12/31/2022]
Abstract
Parkinson's disease (PD), a chronic neurodegenerative disease, is characterized by sensorimotor and cognitive deficits. Previous diffusion tensor imaging (DTI) studies found abnormal DTI metrics in white matter bundles, such as the corpus callosum, cingulate, and frontal-parietal bundles, in PD patients. These studies mainly focused on alterations in microstructural features of long-range bundles within the deep white matter (DWM) that connects pairs of distant cortical regions. However, less is known about the DTI metrics of the superficial white matter (SWM) that connects local cortical regions in PD patients. To determine whether the DTI metrics of the SWM were different between the PD patients and the healthy controls, we recruited DTI data from 34 PD patients and 29 gender- and age-matched healthy controls. Using a probabilistic tractographic approach, we first defined a population-based SWM mask across all the subjects. Using a tract-based spatial statistical (TBSS) analytic approach, we then identified the SWM bundles showing abnormal DTI metrics in the PD patients. We found that the PD patients showed significantly lower DTI metrics in the SWM bundles connecting the sensorimotor cortex, cingulate cortex, posterior parietal cortex (PPC), and parieto-occipital cortex than the healthy controls. We also found that the clinical measures in the PD patients was significantly negatively correlated with the fractional anisotropy in the SWM (FASWM) that connects core regions in the default mode network (DMN). The FASWM in the bundles that connected the PPC was significantly positively correlated with cognitive performance in the PD patients. Our findings suggest that SWM may serve as the brain structural basis underlying the sensorimotor deficits and cognitive degeneration in PD patients.
Collapse
Affiliation(s)
- Yichen Zhang
- Key Laboratory of Brain, Cognition and Education Sciences (South China Normal University), Ministry of Education, School of Psychology, Center for Studies of Psychological Application, Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, China
| | - Biao Huang
- Department of Radiology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, 510080 , China.
| | - Qinyuan Chen
- Key Laboratory of Brain, Cognition and Education Sciences (South China Normal University), Ministry of Education, School of Psychology, Center for Studies of Psychological Application, Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, China
| | - Lijuan Wang
- Department of Neurology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, 510080, China
| | - Lu Zhang
- Key Laboratory of Brain, Cognition and Education Sciences (South China Normal University), Ministry of Education, School of Psychology, Center for Studies of Psychological Application, Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, China
| | - Kun Nie
- Department of Neurology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, 510080, China
| | - Qinda Huang
- Key Laboratory of Brain, Cognition and Education Sciences (South China Normal University), Ministry of Education, School of Psychology, Center for Studies of Psychological Application, Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, China
| | - Ruiwang Huang
- Key Laboratory of Brain, Cognition and Education Sciences (South China Normal University), Ministry of Education, School of Psychology, Center for Studies of Psychological Application, Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
45
|
Thapaliya K, Marshall-Gradisnik S, Staines D, Barnden L. Diffusion tensor imaging reveals neuronal microstructural changes in myalgic encephalomyelitis/chronic fatigue syndrome. Eur J Neurosci 2021; 54:6214-6228. [PMID: 34355438 PMCID: PMC9291819 DOI: 10.1111/ejn.15413] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/01/2021] [Accepted: 08/02/2021] [Indexed: 11/26/2022]
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) patients suffer from a variety of physical and neurological complaints indicating the central nervous system plays a role in ME/CFS pathophysiology. Diffusion tensor imaging (DTI) has been used to study microstructural changes in neurodegenerative diseases. In this study, we evaluated DTI parameters to investigate microstructural abnormalities in ME/CFS patients. We estimated DTI parameters in 25 ME/CFS patients who met Fukuda criteria (ME/CFSFukuda ), 18 ME/CFS patients who met International Consensus Criteria (ICC) (ME/CFSICC ) only and 26 healthy control (HC) subjects. In addition to voxel-based DTI-parameter group comparisons, we performed voxel-based DTI-parameter interaction-with-group regressions with clinical and autonomic measures to test for abnormal regressions. Group comparisons between ME/CFSICC and HC detected significant clusters (a) with decreased axial diffusivity (p = .001) and mean diffusivity (p = .01) in the descending cortico-cerebellar tract in the midbrain and pons and (b) with increased transverse diffusivity in the medulla. The mode of anisotropy was significantly decreased (p = .001) in a cluster in the superior longitudinal fasciculus region. Voxel-based group comparisons between ME/CFSFukuda and HC did not detect significant clusters. For ME/CFSICC and HC, DTI parameter interaction-with-group regressions were abnormal for the clinical measures of information processing score, SF36 physical, sleep disturbance score and respiration rate in both grey and white matter regions. Our study demonstrated that DTI parameters are sensitive to microstructural changes in ME/CFSICC and could potentially act as an imaging biomarker of abnormal pathophysiology in ME/CFS. The study also shows that strict case definitions are essential in investigation of the pathophysiology of ME/CFS.
Collapse
Affiliation(s)
- Kiran Thapaliya
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Brisbane, Queensland, Australia.,Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland, Australia
| | - Sonya Marshall-Gradisnik
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Brisbane, Queensland, Australia
| | - Donald Staines
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Brisbane, Queensland, Australia
| | - Leighton Barnden
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Brisbane, Queensland, Australia
| |
Collapse
|
46
|
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: 7] [Impact Index Per Article: 1.8] [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
|
47
|
Role of Diffusion Tensor Imaging in Parkinson's Disease Diagnosis. ARCHIVES OF NEUROSCIENCE 2021. [DOI: 10.5812/ans.100174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Parkinson's disease (PD) is a chronic and progressive neurodegenerative disease that affects the dopamine-containing neurons. In this study, the role of the Diffusion Tensor imaging (DTI) method was investigated in the detection of PD. Objectives: The purpose of this study was to investigate the microstructural damage of the brain's white matter in PD using a non-invasive DTI technique. Methods: Twenty patients with PD were studied with comprehensive clinical assessments and DTI data. Also, 10 normal subjects were investigated. Fractional anisotropic (FA) and mean diffusivity (MD) values were calculated by drawing region of interest (ROI) on eight distinctive areas of the brain. Results: The level of FA and MD in substantia nigra (SN) was significantly different between the PD and healthy control (HC) groups. Also, differences were found in DTI parameters between PD and HC groups in some regions, such as genu, anterior limb of internal capsule (ALIC), splenium, and putamen. Conclusions: To summarize, DTI as a non-invasive method can be useful in the detection of Parkinson's disease.
Collapse
|
48
|
Hybrid diffusion imaging reveals altered white matter tract integrity and associations with symptoms and cognitive dysfunction in chronic traumatic brain injury. NEUROIMAGE-CLINICAL 2021; 30:102681. [PMID: 34215151 PMCID: PMC8102667 DOI: 10.1016/j.nicl.2021.102681] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 03/12/2021] [Accepted: 04/18/2021] [Indexed: 11/20/2022]
Abstract
Hybrid Diffusion Imaging (HYDI) detects white matter associations in patients with cTBI. The advanced diffusion model NODDI was more sensitive in detecting between-group differences than classic DTI. DTI appeared to be just as sensitive as NODDI for detecting white matter correlations with self-reported symptoms. This study highlights the advantages of acquiring both DTI and NODDI to fully characterize white matter microstructure in cTBI.
The detection and association of in vivo biomarkers in white matter (WM) pathology after acute and chronic mild traumatic brain injury (mTBI) are needed to improve care and develop therapies. In this study, we used the diffusion MRI method of hybrid diffusion imaging (HYDI) to detect white matter alterations in patients with chronic TBI (cTBI). 40 patients with cTBI presenting symptoms at least three months post injury, and 17 healthy controls underwent magnetic resonance HYDI. cTBI patients were assessed with a battery of neuropsychological tests. A voxel-wise statistical analysis within the white matter skeleton was performed to study between group differences in the diffusion models. In addition, a partial correlation analysis controlling for age, sex, and time after injury was performed within the cTBI cohort, to test for associations between diffusion metrics and clinical outcomes. The advanced diffusion modeling technique of neurite orientation dispersion and density imaging (NODDI) showed large clusters of between-group differences resulting in lower values in the cTBI across the brain, where the single compartment diffusion tensor model failed to show any significant results. However, the diffusion tensor model appeared to be just as sensitive in detecting self-reported symptoms in the cTBI population using a within-group correlation. To the best of our knowledge this study provides the first application of HYDI in evaluation of cTBI using combined DTI and NODDI, significantly enhancing our understanding of the effects of concussion on white matter microstructure and emphasizing the utility of full characterization of complex diffusion to diagnose, monitor, and treat brain injury.
Collapse
|
49
|
Zhao M, Liu J, Cai W, Li J, Zhu X, Yu D, Yuan K. Support vector machine based classification of smokers and nonsmokers using diffusion tensor imaging. Brain Imaging Behav 2021; 14:2242-2250. [PMID: 31428924 DOI: 10.1007/s11682-019-00176-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Despite significant progress in treatments for smoking cessation, smoking continues to be a significant public health concern, especially in young adulthood. Thus, developing a predictive model that can classify and characterize the brain-based biomarkers predicting smoking status would be imperative to improving treatment development. In this study, we applied a support vector machine-based classification method to discriminate 70 young male smokers and 70 matched nonsmokers using their diffusion tensor imaging (DTI) data. The classification procedure achieved an average accuracy of 88.6% and an average area under the curve of 0.95. The most discriminative features that contributed to the classification were primarily located in the sagittal stratum (SS), external capsule (EC), superior longitudinal fasciculus (SLF), anterior corona radiata (ACR) and inferior front-occipital fasciculus (IFOF). The following regression analysis showed a significant negatively correlation between the average RD values of the left ACR (r = -0.247, p = 0.039) and FTND. The average MD values in the right EC (r = -0.254, p = 0.034) and RD values in the right IFOF (r = -0.240, p = 0.046) were inversely associated with pack-years. Our findings indicate that the discriminative white matter (WM) features as brain biomarkers provide great predictive power for smoking status and suggest that machine learning techniques can reveal underlying smoking-related neurobiology.
Collapse
Affiliation(s)
- Meng Zhao
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710071, People's Republic of China.,Engineering Research Center of Molecular and Neuro Imaging Ministry of Education, Xi'an, People's Republic of China
| | - Jingjing Liu
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710071, People's Republic of China.,Engineering Research Center of Molecular and Neuro Imaging Ministry of Education, Xi'an, People's Republic of China
| | - Wanye Cai
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710071, People's Republic of China.,Engineering Research Center of Molecular and Neuro Imaging Ministry of Education, Xi'an, People's Republic of China
| | - Jun Li
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710071, People's Republic of China
| | - Xueling Zhu
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China.
| | - Dahua Yu
- Inner Mongolia Key Laboratory of Pattern Recognition and Intelligent Image Processing, School of Information Engineering, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, 014010, People's Republic of China.
| | - Kai Yuan
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710071, People's Republic of China. .,Engineering Research Center of Molecular and Neuro Imaging Ministry of Education, Xi'an, People's Republic of China. .,Inner Mongolia Key Laboratory of Pattern Recognition and Intelligent Image Processing, School of Information Engineering, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, 014010, People's Republic of China.
| |
Collapse
|
50
|
Yang Q, Nanivadekar S, Taylor PA, Dou Z, Lungu CI, Horovitz SG. Executive function network's white matter alterations relate to Parkinson's disease motor phenotype. Neurosci Lett 2021; 741:135486. [PMID: 33161103 PMCID: PMC7750296 DOI: 10.1016/j.neulet.2020.135486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/28/2020] [Accepted: 10/31/2020] [Indexed: 11/25/2022]
Abstract
Parkinson's disease (PD) patients with postural instability and gait disorder phenotype (PIGD) are at high risk of cognitive deficits compared to those with tremor dominant phenotype (TD). Alterations of white matter (WM) integrity can occur in patients with normal cognitive functions (PD-N). However, the alterations of WM integrity related to cognitive functions in PD-N, especially in these two motor phenotypes, remain unclear. Diffusion tensor imaging (DTI) is a non-invasive neuroimaging method to evaluate WM properties and by applying DTI tractography, one can identify WM tracts connecting functional regions. Here, we 1) compared the executive function (EF) in PIGD phenotype with normal cognitive functions (PIGD-N) and TD phenotype with normal cognitive functions (TD-N) phenotypes; 2) used DTI tractography to evaluated differences in WM alterations between these two phenotypes within a task-based functional network; and 3) examined the WM integrity alterations related to EF in a whole brain network for PD-N patients regardless of phenotypes. Thirty-four idiopathic PD-N patients were classified into two groups based on phenotypes: TD-N and PIGD-N, using an algorithm based on UPDRS part III. Neuropsychological tests were used to evaluate patients' EF, including the Trail making test part A and B, the Stroop color naming, the Stroop word naming, the Stroop color-word interference task, as well as the FAS verbal fluency task and the animal category fluency tasks. DTI measures were calculated among WM regions associated with the verbal fluency network defined from previous task fMRI studies and compared between PIGD-N and TD-N groups. In addition, the relationship of DTI measures and verbal fluency scores were evaluated for our full cohort of PD-N patients within the whole brain network. These values were also correlated with the scores of the FAS verbal fluency task. Only the FAS verbal fluency test showed significant group differences, having lower scores in PIGD-N when compared to TD-N phenotype (p < 0.05). Compared to the TD-N, PIGD-N group exhibited significantly higher MD and RD in the tracts connecting the left superior temporal gyrus and left insula, and those connecting the right pars opercularis and right insula. Moreover, compared to TD-N, PIGD-N group had significantly higher RD in the tracts connecting right pars opercularis and right pars triangularis, and the tracts connecting right inferior temporal gyrus and right middle temporal gyrus. For the entire PD-N cohort, FAS verbal fluency scores positively correlated with MD in the superior longitudinal fasciculus (SLF). This study confirmed that PIGD-N phenotype has more deficits in verbal fluency task than TD-N phenotype. Additionally, our findings suggest: (1) PIGD-N shows more microstructural changes related to FAS verbal fluency task when compared to TD-N phenotype; (2) SLF plays an important role in FAS verbal fluency task in PD-N patients regardless of motor phenotypes.
Collapse
Affiliation(s)
- Qinglu Yang
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States; The Third Affiliated Hospital of Sun Yat-sen University, Rehabilitation Department, Guangzhou, PR China
| | - Shruti Nanivadekar
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Paul A Taylor
- Scientific and Statistical Computing Core, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Zulin Dou
- The Third Affiliated Hospital of Sun Yat-sen University, Rehabilitation Department, Guangzhou, PR China
| | - Codrin I Lungu
- Parkinson Disease Clinic, OCD, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Silvina G Horovitz
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|