Parkinson & Neurodegeneration
Neuro Wednesday, 19 May 2021

Oral Session - Parkinson & Neurodegeneration
Neuro
Wednesday, 19 May 2021 14:00 - 16:00
  • Twelve-Year Microstructural Changes in The Deep Gray Nuclei in Parkinson’s Disease: A Serial Diffusion Tensor Imaging Study
    Yao-Chia Shih1,2, Qi Rong Leon Ooi3, Septian Hartono2,3, Thomas Welton2,3, Hui-Hua Li2,4, John Carson Allen2, Eng King Tan2,3, and Ling Ling Chan1,2
    1Department of Diagnostic Radiology, Singapore General Hospital, Singapore, Singapore, 2Duke-NUS Medical School, Singapore, Singapore, 3Department of Neurology, National Neuroscience Institute (Outram-campus), Singapore, Singapore, 4Health Services Research Unit, Singapore General Hospital, Singapore, Singapore
    Widespread deep nuclear neurodegeneration as evidenced by MD increases is seen in late PD stages after 12 years, while FA changes suggested more complex interplay of iron deposition effects and functional re-organization in the putamen and thalamus.
    Fig. 1 Schematic of semi-automated tissue segmentation pipeline.
    Fig. 2 Temporal FA and MD profiles in the four deep gray nuclei across 12 years. In each line chart, the blue solid line indicates mean DTI metric in healthy controls, whereas the red dotted line indicates mean DTI metrics in PD patients. The upper and row charts refer to FA and MD changes respectively.
  • Characterizing white matter microstructural alterations in de novo Parkinson’s disease using diffusion MRI
    Yiming Xiao1,2, Terry M Peters3,4,5, and Ali R Khan3,4,5,6
    1PERFORM Centre, Concordia University, Montreal, QC, Canada, 2Computer Science and Software Engineering, Concordia University, Montreal, QC, Canada, 3Robarts Research Institute, Western University, London, ON, Canada, 4Department of Medical Biophysics, Western University, London, ON, Canada, 5School of Biomedical Engineering, Western University, London, ON, Canada, 6The Brain and Mind Institute, Western University, Lonon, ON, Canada
    DTI measures and fixel-based analysis revealed strengthened and weakened white matter integrity, which is subject to laterality of motor symptoms in de novo Parkinson’s disease, suggesting both functional degeneration and compensatory network creation.
    Figure 1. Investigation of white matter tract changes between Parkinson’s and healthy subjects, using fixel-based analysis (FBA). The white matter tracts that are statistically significant (p<0.05) in fiber cross-section (FC), fiber density (FD) and fiber density and cross-section (FDC) are shown overlaid on a group-averaged template with a heat map. The cross-hair in each sub-figure points at the same location.
    Figure 2. Investigation of white matter changes between Parkinson’s and healthy subjects, using voxel-based analysis with fractional anisotropy (FA) and mean diffusivity (MD). The regions that are statistically significant (p<0.05) are shown overlaid on a group-averaged template with a heat map. The cross-hair in each sub-figure points at the same location.
  • Progressive microstructural alterations in subcortical nuclei in Parkinson's disease: a diffusion magnetic resonance imaging study
    Xueqin Bai1, Tao Guo1, Xiaojun Guan 1, Cheng Zhou1, Jingjing Wu1, Xiaocao Liu1, Ting Gao1, Luyan Gu1, Xiaojun Xu1, Peiyu Huang1, and Minming Zhang1
    1The second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
    In this study, we employed diffusion kurtosis imaging (DKI) and diffusion tensor imaging (DTI) to measure the microstructural alterations in subcortical nuclei across PD patients at different disease stages. Individual diagnostic model was constructed to test the performance of diffusion metrics in identifying PD patients at different stages.  We found that PD patients at different stages have progressive microstructural alterations in the main nuclei of widely acknowledged nigral-pallidal and thalamo-cortical pathways. DKI is sensitive to detect microstructural changes in GP and thalamus between early stage PD and moderate-late stage PD patients. The combination of kurtosis and tensor metrics can achieve a good performance in diagnosing PD.
    Figure 2. Mean diffusion metrics in healthy controls (HC), early-stage Parkinson’s disease (EPD) group, and moderate-late-stage Parkinson’s disease (MLPD) group. (a) Mean diffusion metrics in substantia nigra (SN) in HC, EPD, and MLPD. (b) Mean diffusion metrics in globus pallidus (GP) in HC, EPD, and MLPD. (c) Mean diffusion metrics in thalamus in HC, EPD, and MLPD. * and※ indicate p < 0.05 and p < 0.01.
    Figure 4.Receiver operating characteristic curves of the diagnostic performance of diffusion metrics in substantia nigra(SN), globus pallidus(GP), thalamus and their combinations for discriminating Parkinson’s disease (PD) patients from healthy controls (HC) or between early-stage Parkinson’s disease group (EPD) (and moderate-late-stage Parkinson’s disease group (MLPD)) and HC. Plots of the FA, AD, MD, MK, AK in the SN and MK, RK in the GP and FA, MK and RK values in the thalamus and their combinations for diagnosing PD (a), EPD (b), and MLPD (c).
  • Microstructure of grey matter nuclei in early Parkinson’s disease: longitudinal study using diffusion kurtosis imaging
    Thomas Welton1,2, Septian Hartono3, Yao-Chia Shih3, Samuel Y-E Ng1, Nicole S Y Chia1, Weiling Lee3, Say Lee Chong3, Eng-King Tan1,2,3, Ling-Ling Chan1,2,3, and Louis CS Tan1,2
    1National Neuroscience Institute, Singapore, Singapore, 2Duke-NUS Medical School, Singapore, Singapore, 3Singapore General Hospital, Singapore, Singapore
    We found elevated mean kurtosis in specific basal ganglia regions, which was maintained over two years and negatively correlated with worsening motor function in Parkinson’s disease.
    Figure 1. (A) Substantia nigra mask (orange region) from the CIT168 atlas. (B) Mean kurtosis in each region of interest compared between groups. Bars show the raw uncorrected data while p-values and adjusted R2 values given are for the corrected model controlling for age, sex and education. (C) Radial plots of diffusion kurtosis indices for each region and both groups. (D) Plots of the estimated marginal means from repeated-measures ANOVA. Error bars show 95%CI. * Significant for p<0.05.
  • Tract Density Imaging in Patients with Parkinson’s Disease Before and After Magnetic Resonance-guided Focused Ultrasound
    Yu Shen1, Xianchang Zhang2, Yan Bai1, Rui Zhang1, Rushi Chen1, Wei Wei1, Menghuan Zhang1, and Meiyun Wang1
    1Department of Medical Imaging, Henan Provincial People’s Hospital & Zhengzhou University, Zhengzhou, China, 2MR Collaboration, Siemens Healthcare Ltd. Beijing China, Beijing, China
    Tract density imaging values were significantly decreased after magnetic resonance-guided focused ultrasound in the genu of the corpus callosum and left globus pallidus of seven patients with Parkinson’s disease.
    Figure 3. The brain areas with significantly decreased track density values postoperatively, overlaying on the T1 template. A) Left globus pallidus and B) genu of the corpus callosum. The color bar represents the t-value of the T-test.
    Figure 1. The A) fractional anisotropy (FA) and B) track density image (TDI) from the same patient. TDI has a higher spatial resolution than that of the FA image (1 × 1 × 1 mm3 vs 2.2 × 2.2 × 2.2 mm3, respectively).
  • Investigating Spatiotemporal Changes in Dopamine, Neuromelanin and Iron in the Nigrostriatal System in Parkinson's Disease
    Emma Biondetti1,2,3, Mathieu D. Santin1,2, Romain Valabrègue1,2, Graziella Mangone1,4, Rahul Gaurav1,2,3, Nadya Pyatigorskaya1,3,5, Matthew Hutchison6, Lydia Yahia-Cherif1,2, Nicolas Villain1,7, Marie-Odile Habert8, Isabelle Arnulf1,3,9, Smaranda Leu-Semenescu9, Pauline Dodet9, Jean-Christophe Corvol1,4,7, Marie Vidailhet1,3,7, and Stéphane Lehéricy1,2,3,5
    1Institut du Cerveau – ICM, INSERM U 1127, CNRS UMR 7225, Sorbonne Université, Paris, France, 2ICM, Centre de NeuroImagerie de Recherche – CENIR, Paris, France, 3ICM, Team “Movement Investigations and Therapeutics” (MOV’IT), Paris, France, 4National Institute of Health and Medical Research - INSERM, Clinical Investigation Centre, Pitié-Salpêtrière Hospital, Paris, France, 5Department of Neuroradiology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France, 6Biogen Inc., Cambridge, MA, United States, 7Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France, 8Department of Nuclear Medicine, Pitié-Salpêtrière Hospital, AP-HP, Paris, France, 9National Reference Center for Rare Hypersomnias, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
    Dopamine, neuromelanin and iron undergo neurodegeneration in Parkinson’s disease. We investigated the spatial evolution of these processes in the nigrostriatal system, along with their interrelationship, time of onset and relative temporal ordering.
    Figure 2: Change over disease duration in striatal DaT, SNc NM and SNc iron in PDs relative to HCs. For all PDs relative to HCs, in each functional subregion of the striatum (A-C) and SNc (D, E) in the most affected brain hemisphere, the mono-exponential fitting functions for the percent ratios of DaT striatal binding ratio (SBR), NM SNR and QSM are plotted against disease duration
    Figure 1: Group differences in striatal DaT and SNc NM and iron levels. Functional subregions in the striatum (A) and the SNc (D). The panels show the results of the statistical comparison between groups in the clinically most (B, E) and least affected brain hemispheres (C, F). Each panel shows the inter-group comparison between iRBDs and HCs (left column), PDs and HCs (centre column), or PDs and iRBDs (right column). P-values were converted into z-scores
  • Tracking serial Parkinson’s related changes in the substantia nigra using Neuromelanin MRI and free-water diffusion MRI
    Yue Xing1,2,3, Saadnah Naidu1,2,3, Halim Abdul-Sapuan1,2,3, Ali-Reza Mohammadi-Nejad2,3, Jonathan Evans4, Ofer Pasternak5, Stamatios Sotiropoulos2,3, Christopher R. Tench1,3, and Dorothee P. Auer1,2,3
    1Division of Clinical Neuroscience, Queen’s Medical Centre, University of Nottingham, Nottingham, United Kingdom, 2Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, United Kingdom, 3NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, United Kingdom, 4Department of Neurology, Nottingham University Hospital Trust, Nottingham, United Kingdom, 5Departments of Psychiatry and Radiology (O.P.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
    Longitudinal NM signal-loss and FW increase was seen in PD throughout the SN with significant acceleration compared to controls in the ventral-SN, but there was no between-metrics correlation, suggesting that these promising serial biomarkers may track different aspects of PD progression.
    Figure 1. Illustration of the analysis pipeline: first, an age-specific healthy control’s mean brainstem template was generated, along with their respective spatial prior probability maps with three classes: brainstem outside of the SN (BS), SN, and irrelevant background (BG and it is not shown here). The same procedure was performed to transform NM-MRIs in the same space as the prior probability maps. Finally, the Bayesian classification was applied for partitioning the SN from the brainstem in both image intensity and in space and the mean intensity of the brainstem was extracted.
    Figure 2. The annualized %change of MRI metrics (a. % increase Free-water and b. % reduction of NM contrast to background ratio) of ventral and dorsal SN in PD and controls. c. The scatter plot of annualized % increase of FW values versus the annualized % reduction of NM contrast ratio in PD. Error bars show SEM.
  • Distinct cognitive and anthropometric functional connectivity traits of cognitive decline in Parkinson’s disease using partial least squares.
    Vicente Jose Ferrer-Gallardo1, Thomas Bolton2, Manuel Delgado3, Pedro M. Paz-Alonso1, Maricuz Rodriguez-Oroz4, and César Caballero-Gaudes1
    1Basque Center on Cognition, Brain and Language, Donostia, Spain, 2Medical image processing, Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland, 3Department of Neurology, Sierrallana Hospital, Torrelavega, Spain, 4Neurology Department, Clinica Universidad de Navarra, Pamplona, Spain
    Our data underscore two types of whole-brain functional connectomes that are differentially related to cognitive abilities and anthropometric measures in Parkinson Disease patients with mild cognitive impairment using a partial least squares regression approach. 
    Figure 4. Significant FC-traits of PLS component 1 (shown in blue in Figure 3) Each FC-trait is represented by its FC circular plot thresholded to 1% of the strongest connections grouped by functional networks organization, and the map of FC node degree (i.e. sum of functional connections from and to each brain region). The variance explained by each FC-trait is also indicated.
    Figure 5. Significant FC-traits of PLS component 2 (shown in yellow in Figure 3). Each FC-trait is represented by its FC circular plot thresholded to 1% of the strongest connections grouped by functional networks organization, and the map of FC node degree (i.e. sum of functional connections from and to each brain region). The variance explained by each FC-trait is also indicated.
  • Dysfunction of Olfactory Resting-State Functional network in Early-onset Early-stage Parkinson’s Disease
    Jianli Wang1, Rachel Stanford1, Lauren Spreen2, Jeffrey Vesek2, Christopher Sica1, Thyagarajan Subramanian3, and Qing X Yang4
    1Radiology, Penn State College of Medicine, HERSHEY, PA, United States, 2Molecular Biology, Penn State College of Medicine, HERSHEY, PA, United States, 3Neurology, Penn State College of Medicine, HERSHEY, PA, United States, 4Neurosurgery, Penn State College of Medicine, HERSHEY, PA, United States
    In this study, we tested the hypothesis that there are PD-related dysfunctions in the central olfactory functional network at the early stage of Parkinson's disease.
    Figure 1. Significantly weaker FC with bilateral POCs in the left substantia nigra (SN), temporal pole, interior temporal gyrus, insular cortex, and posterior hippocampus and parahippocampal gyrus of PDs (2-sample t-test, uncorrected, p < 0.005, extent threshold = 6 voxels)
    Values are presented in mean ± SD. *, p < 0.05, MoCA, Montreal Cognitive Assessment; UPDRS, Unified Parkinson’s Disease Rating Scale; N/A, not applicable.
  • Ultra-high field imaging of deep brain stimulation at 7T: The first study of RF safety, displacement force and image artifact
    Bhumi Bhusal1, Jason Stockmann2, Azma Mareyam2, John Kirsch2, Lawrence L Wald2, Mark J Nolt1, Joshua Rosenow1, Roberto Lopez-Rosado1, Behzad Elahi1, and Laleh Golestanirad1
    1Northwestern University, Chicago, IL, United States, 2Massachusetts General Hospital, Charlestown, MA, United States
    7T MRI may be safely performed in patients with deep brain stimulation implants with careful evaluation of implant and MRI hardware.
    Figure 2. (A) 3D-rendered view of postoperative CT images of DBS patients showing extracranial trajectories of leads and extensions. (B) Schematic of the experimental phantom showing a full DBS system as well as examples of some trajectories used during the RF heating experiments. (C) Setup for the measurement of the magnetic force on the IPG.
    Figure 3. Steps of DBS implantation and artifact measurement. (A) Formalin fixed cadaveric brain placed inside a 3D printed skull. (B) 1.5T MRI of the cadaveric brain were transferred to BrainLAB iPlan server for localizing subthalamic nucleus. (C) Leksell G Frame and stereotactic arc attached to the skull for electrode implantation. (D) Burr hole cover placed to fixate the lead in position. (E) Artifact was quantified on transverse and coronal planes for different imaging sequences.
Back to Top
Digital Poster Session - Brain Stem & Cerebellar Diseases
Neuro
Wednesday, 19 May 2021 15:00 - 16:00
  • Cerebellar changes in Spinocerebellar Ataxia Type 2 and 12 in comparison with healthy controls
    Pankaj pankaj1, S Senthil Kumaran1, and Achal Kumar Srivastava2
    1NMR, All India Institute of Medical Sciences, New Delhi, India, 2Neurology, All India Institute of Medical Sciences, New Delhi, India
    White and grey matter atrophy in SCA2 with respect to SCA12, and bilateral cerebellar atrophy in SCA in comparison with healthy controls correlate with tremor and gait abnormality.
    Figure 1: Cerebellum atrophy in comparison of(a) HC and SCA2 (b)HC and SCA12 (c) SCA2 and SCA12 at the significant level of p<0.001, uncorrected (t-test) displayed on SUIT Flatmapview(color map: jet and threshold 0:0.1)
    Table 1. Significant difference in the white matter between SCA2 and SCA12 (analysis using SUIT, one way ANOVA, p<0.05 FWE, voxel threshold (k)≥5)
  • Cerebro-cerebellar impact on brain dynamics in a single-subject with cerebellar ataxia
    Silvia Maria Marchese1, Fulvia Palesi2,3, Mariagrazia Bruzzone4, Anna Nigri4, Stefano D'Arrigo5, Chiara Pantaleoni5, Claudia AM Gandini Wheeler-Kingshott2,3,6, Egidio D'Angelo2,3, and Paolo Cavallari1
    1Human Physiology Section of the DePT, Università degli Studi di Milano, Milano, Italy, 2Department of Brain and Behavioral Science, Università degli Studi di Pavia, Pavia, Italy, 3Brain Connectivity Center Research Department, IRCCS Mondino Foundation, Pavia, Italy, 4Neuroradiology Department, Fondazione IRCCS Istituto Neurologico "C. Besta", Milano, Italy, 5Developmental Neurology Department, Fondazione IRCCS Istituto Neurologico "C. Besta", Milano, Italy, 6Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, NMR Research Unit, Queen Square MS Centre, London, United Kingdom
    Brain dynamics simulated in a subject with Joubert syndrome highlighted the impact of excluding cerebellum from brain network in cerebellar dysfunction. Pearson correlation coefficient is strongly decreased when cerebro-cerebellar connectivity is excluded from the simulation. 
    Table 1: Parameters forwhole-brain network, cerebral subnetwork and embedded cerebro-cerebellar subnetwork.Optimal value for global coupling (Gcoupl) and Pearson correlation coefficients (PCC) between structural connectivity (SC), experimental (expFC) and simulated functional connectivity (simFC) are reported.
    Figure 1: A) Experimental matrices of structural connectivity (expSC) and functional connectivity (expFC) in the patient with Joubert syndrome. B) Simulated functional connectivity (simFC) matrices for whole-brain network (left), cerebral subnetwork (center) and embedded cerebro-cerebellar subnetwork (right). In each matrix, rows and columns represent a specific brain region (node), while each intersection point represents a connection, weighted by the number of streamlines, between two nodes (edge).
  • Genetic impacts on nigral iron deposition in Parkinson’s disease
    Jing jing Wu1, Xiao jun Guan2, Tao Guo2, Cheng Zhou2, Ting Gao3, Xue qin Bai2, Xiao cao Liu3, Lu yan Gu3, Pei yu Huang3, Xiao jun Xu3, and Min ming Zhang2
    1Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, HangZhou, China, 2Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 3The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
    Two variants, rs602201 and rs198440, were found to have a positive impact on nigral iron deposition in PD. Specifically, patients with rs602201 polymorphism are particularly vulnerable to iron deposition in SN.
    Figure 1 Data processing procedures. A. The labels of subcortical nuclei in processed QSM images. B. The framework of quality control steps.
    Figure 2 The association between genetic variations and imaging phenotypes. A. Heat map with significant associations between SNP and QT at p<10-5 (blocks labeled with “X”). Color key on the top left coded the magnitude of -log10(p-values). B, C. Manhattan and Q-Q plot of the most significant association (p<10-6) (rs602201-R_SN). The horizontal line displayed the cutoff for p<10-6. Shown on (C) is the Q-Q plot of the distribution of the observed p-values (-log10(observed p-value)) versus the expected p-values (-log10(expected p-value)) under the null hypothesis of no association.
  • The presumed structure alterations of Spinocerebellar Ataxias 3: from presymptomatic to the symptomatic stage
    Haishan Qiu1, Jing Zhao1, Manshi Hu1, Mengzhu Wang2, and Jianping Chu1
    1Sun Yat-sen University, Guangzhou, China, 2Simens Healthcare, Guangzhou, China
    43 gene confirmed SCA3 patients, including 37 symptomatic and 15 presymptomatic, and 35 health controls were prospectively included, and VBM and TBSS were used to investigate the differences of GM and WM. Our study indicates that there is an evolving history of structural images with SCA3 patients in different disease stages. The WM damage starts with the impairment of ICP and goes through SCP extends to the midbrain, then widespread to the whole brain. The alteration of GMV does not occur until the arise of ataxia symptom, then began to involve the medulla, cerebellum, and pons, and developed to involve basal ganglion, after the decompensation of bilateral dorsal thalamus, finally affect the cortical cortex. The impairment of WM tracts precedes the GM atrophy and, irrespective of the patients with or without clinical manifestation, the identified WM damage was significantly correlated with SARA.
    Tract-based statistical analysis of DKT/DTI metrics between symptomatic SCA3 patients and health controls. Red-yellow highlights show areas of decreased DKI/DTI metrics values in symptomatic SCA3. Blue highlights show areas of increased MD value in symptomatic SCA3.(P<0.05, TFCE correction)
    Tract-based statistical analysis of DKT/DTI metrics between symptomatic SCA3 patients and health controls. Red-yellow highlights show areas of decreased DKI/DTI metrics values in symptomatic SCA3. Blue highlights show areas of increased MD value in symptomatic SCA3.(P<0.05, TFCE correction)
  • Volumetric estimation of various brain parts in Gluten Ataxia patients: A quantitative MRI study
    Uma Sharma1, Vishwa Rawat1, Prasenjit Das 2, Achal Kumar Srivastava3, and Govind Makharia4
    1Nuclear Magnetic Resonance and MRI Facility, All India Institute of Medical Sciences, New Delhi, India, 2Pathology, All India Institute of Medical Sciences, New Delhi, India, 3Neurology, All India Institute of Medical Sciences, New Delhi, India, 4Gasteroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India
    Volumetric analysis of whole brain in gluten ataxia (GA) patients using MRI revealed significantly low brain and cerebellar volumes along the lobules which form part of vermis while cerebrum volume is not linked to GA.
    Figure 2: Representative CERES image of cerebellum taken from Healthy control (A) and GA patient (B).
    Table 2: Volumetric estimation of Brain structures of GA patients and Healthy Controls.
  • Evaluating normative Cerebellum radiomics on FLAIR images
    Umang Pandey1, Jitender Saini2, Manoj Kumar2, Rakesh Gupta3, and Madhura Ingalhalika1
    1Symbiosis Centre for Medical Image Analysis, Symbiosis International University, Pune, India, 2Department of Radiology, National Institute of Mental Health and Neurosciences, Bengaluru, India, 3Department of Radiology,Fortis Memorial Research Institute, Gurgaon, India
    Cerebellum and Cerebrum radiomics vary significantly. Presence of non -robust features across timepoints/scanners suggest that care must be taken while interpreting these features for pathological inferences.
    Figure 4: ICC score boxplots for cerebellum pacellations for A) intra-scanner, B) Pre-harmonization inter-scanner and C) Post-harmonized inter-scanner.
    Figure 2: Boxplot of ICC scores for GM/WM hemispheric masks of Cerebrum & Cerebellum for inter- and intra-scanner. The ICC score is interpreted on a scale of 0 to 1, with 1 indicating absolute repeatability and anything below 0.5 considered poor for reproducibility.
  • Spatial changes of neuromelanin and iron content in substantial nigra pars compacta in early-stage idiopathic Parkinson’s disease
    Zenghui Cheng1, Bin Xiao2, Naying He3, Dinggang Shen4, Qian Wang5, Feng Shi4, Youmin Zhang3, Pei Huang3, Yan Li3, Sean K Sethi6, Kiarash Ghassaban7, Shengdi Chen3, Fuhua Yan3, and Ewart Mark Haacke7
    1Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, 2Medical Imaging Technology, 、School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China, 3Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, 4Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China, 5Medical Imaging Technology, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China, 6Magnetic Resonance Innovations, Inc, Bingham Farms, MI, United States, 7Wayne State University, Detroit, MI, United States
    Iron deposition and neuromelanin-containing neuron loss is prominent in the ventral and medial part of SNpc in early-stage PD. This region may correspond to nigrosome 2.
    Figure 1. Flow of chart illustrating the voxel-wised analysis. PD- Parkinson’s disease, HC-healthy control, QSM- quantitative susceptibility mapping, NMCNR- contrast to noise of neuromelanin.
    Figure 4. Negative Pearson’s correlation map of substantia nigra pars compacta (QSM-NMCNR overlap) PD- Parkinson’s disease, HC-healthy control, QSM- quantitative susceptibility mapping, NMCNR- contrast to noise of neuromelanin.
  • Automatic Detection of the Neuromelanin, Substantia Nigra, Red Nucleus and Subthalamic Nucleus using a High Resolution Brain Template
    Mojtaba Jokar1, Ying Wang1,2, Zhijia Jin3, Yan Li3, Zenghui Cheng3, Yu Liu3, Naying He3, Fuhua Yan3, and E. Mark Haacke1,2,3,4,5
    1Magnetic Resonance Innovations, Inc., Bingham Farms, MI, United States, 2Department of Radiology, Wayne State University, Detroit, MI, United States, 3Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, 4Department of Biomedical Engineering, Wayne State University, Detroit, MI, United States, 5Department of Neurology, Wayne State University, Detroit, MI, United States
    Comparing the template results to the manual data, yielded excellent DICE similarity coefficients and volume ratios for the four structures of interest and good agreement between the manual and template measurements for SN iron content and the NM background mean.
    Figure 1. The stages of mapping the boundaries from the template space to the original space for neuromelanin. Each column represents a different slice. A) NM template; B) the transformed NM template; C) the same boundaries superimposed on the original midbrain images; and D) final boundaries after DPA was used to refine the boundaries.
    Figure 2. The stages of mapping the boundaries from the template space to the original space for the QSM data. Each column represents a different slice. A) the QSM template; B) the transformed QSM template; C) the same boundaries superimposed on the original midbrain images; and D) final boundaries after the DPA was used to refine the boundaries. The fourth and fifth columns show the SN boundary in red and the STN boundary in orange. The second and third rows show the RN boundary in light green.
  • Neuromelanin Sensitive MRI and QSM of the Substantia Nigra in Parkinson’s-Linked Asian LRRK2 Carriers
    Septian Hartono1,2, An Sen Tan3, Weiling Lee4, Joey Oh4, Kuan Jin Lee5, Jongho Lee6, Eng King Tan1,2, and Ling Ling Chan2,4
    1National Neuroscience Institute, Singapore, Singapore, 2Duke-NUS Medical School, Singapore, Singapore, 3Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore, 4Singapore General Hospital, Singapore, Singapore, 5Singapore BioImaging Consortium, Singapore, Singapore, 6Seoul National University, Seoul, Korea, Republic of
    Neuromelanin-sensitive MRI showed no significant differences between PD LRRK2 carriers and non-carriers. Quantitative susceptibility mapping was able to distinguish the two groups, with higher substantia nigra (SN) iron deposition and larger high-iron area of the SN in PD LRRK2 carriers.
    SN ROI derived from (1) semi-automated segmentation on SMWI images (left) by thresholding for voxels with high iron deposition containing signal intensity 7 standard deviations less than background in and (2) NMS (right) by thresholding for voxels with signal intensity 4 standard deviations higher than background in PD patient LRRK2 risk-variant carriers (top row) and non-carriers (bottom row).
    ROC analysis of QSM susceptibility, high-iron SN size derived from SMWI, and combination model of QSM susceptibility and high-iron SN size to classify LRRK2 risk-variant carriers and non-carriers in PD patients.
  • Altered cortico-cerebellar functional connectivity of language processing in congenital blind children
    A Ankeeta1, S Senthil Kumaran1, and Rohit Saxena2
    1Department of NMR & MRI Facility, All India Institute of Medical Sciences, Delhi, India, 2Dr RP Centre of Ophthalmology, All India Institute of Medical Sciences, Delhi, India
    Congenitally blind subjects conscript visual cortex and cerebellum on functional response to haptic language processing. Improved grey matter volume and duration of Braille reading influences the functional connectivity of language network with cerebellum.
    Figure 1. Illustration of (A) independent component analysis for calculation of cortico-cerebellum during semantic word recognition task in congenital blind relative to sighted control. (B) Difference in cerebellar areas effect size and (C) estimation of gray matter volume.
    Table 2. Significant functional connectivity of cerebellum and language integration estimated by using independent component analysis for congenital blind children group as compared to sighted control group.
  • Substantia Nigra Neuromelanin-sensitive Imaging Biomarker to Differentiate Between Atypical Parkinsonian Syndromes
    Rahul Gaurav1,2,3, Emina Arsovic1,4, Lydia Chougar1,4, Nadya Pyatigorskaya1,2,3,4, Marie Vidailhet2,3,5, and Stephane Lehericy1,2,3,4
    1CENIR, ICM Paris, Paris, France, 2Paris Brain Institute (ICM), Sorbonne University, UPMC Univ Paris 06, Inserm U1127, CNRS UMR 7225, Paris, France, 3ICM Team “Movement Investigations and Therapeutics” (MOV’IT), Paris, France, 4Department of Neuroradiology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France, 5Department of Neurology, APHP, Pitié-Salpêtrière Hospital, Paris, France
    Using neuromelanin-sensitive T1-weighted MRI, we found a significant decrease in substantia nigra pars compacta of atypical Parkinsonian disorders compared to healthy volunteers.
    Figure 1: Substantia Nigra pars compacta regions of interest (left in yellow and right in pink) based on neuromelanin-sensitive imaging of a representative healthy volunteer.

    Figure 2: Box plot of Substantia Nigra pars compacta volume in Atypical Parkinsonian along with healthy volunteers. HV is Healthy Volunteers, PD is Parkinson's Disease, PSP is Progressive supranuclear palsy, DCB is CorticoBasal Degeneration, MSA is Multiple System Atrophy, with its cerebellar (MSAc) and Parkinsonian (MSAp) subtypes, and DCL is dementia with Lewy body.

  • Fibre-specific white matter reduction in patients with multiple system atrophy: comparison of parkinsonian and cerebellar subtypes
    Po-Yuan Chen1, Chih-Chien Tsai1, Chin-Song Lu2, Yi-Hsin Weng2, Yi-Ming Wu3, and Jiun-Jie Wang1
    1Chang Gung University, Taoyuan, Taiwan, 2Chang Gung Memorial Hospital, Taoyuan, Taoyuan, Taiwan, 3Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
    This study provides evidence for the subtype-specific white matter differences in patients with MSA. Importantly, early diagnosis for parkinsonian or cerebellar subtype of MSA is possible with the white matter pattern by fixel-based analysis.
    Figure 2. Fixels with significant (p < 0.05, FWE-corrected) decrease in fixel-based metrics. (a) MSA-P versus controls. Subtle changes were observed in the cohorts with disease duration≦3 years; on the contrary, FC and FDC were significantly decreased in the (b) MSA-C versus controls. Streamlines were colored by direction (anterior-posterior: green; superior-inferior: blue; left-right: red).
    Figure 1. Significant changes in fixel-based metrics in patients with MSA compared to healthy control subjects. Regions of significant changes in FD, FC and FDC were displayed stereoscopically in the (a) sagittal, (b) superior left frontal and (c) inferior right occipital view. Streamlines corresponding to significant fixels (family-wise error corrected p < 0.05) were illustrated and colored according to p values. Cold color represented reduction, whereas warm color represented increase.
  • Substantia Nigra Susceptibility Features Derived by Radiomics Predict Motor Outcome for STN-DBS in Parkinson’s Disease
    Naying He1, Yu Liu1, Bin Xiao2, Junchen Li3, Chencheng Zhang4, Yijie Lai4, Feng Shi5, Dinggang Shen5, Yan Li1, Hongjiang Wei6, Ewart Mark Haacke1,7, Weibo Chen8, Qian Wang2, Dianyou Li4, and Fuhua Yan1
    1Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, 2Institute for Medical Imaging Technology, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China, Shanghai, China, 3Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, No. 6 Huanghe Road, Changshu, China, Changshu, China, 4Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, Shanghai, China, 5Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China, Shanghai, China, 6Institute for Medical Imaging Technology, Department of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China, Shanghai, China, 7Department of Radiology, Wayne State University, Detroit, Michigan, USA, Detroit, MI, United States, 8Philips Healthcare,Shanghai,China, Shanghai, China
    This QSM based radiomics model performed best with an AUC of 0.897 to predict the STN-DBS motor outcome in PD. In addition, the threshold probability of the RA-ML model can differentiate surgical responders and non-responders.
    Fig. 2. Graph shows receiver operating characteristic curve to assess the utility of two different models with the clinical variables included or not (with and w/o clinical information) for predicting global motor outcome.
    Fig. 1. Illustration of the processing pipeline of the radiomics model with machine learning (RA-ML). RF=radiomics feature; RFE= recursive feature elimination
  • Locus coeruleus degeneration associated with less levodopa responsiveness in Parkinson’s Disease
    Cheng Zhou1 and Minming Zhang1
    1Zhejiang University, Hangzhou, China
    LC degeneration was an indicator for less levodopa responsiveness. LC integrity evaluation might be an alternative tool in predicting disease prognosis and stratifying patients into clinical trials for improving the efficacy of levodopa.
    A, B, and C: Relationships between LC integrity, the change rate of UPDRS III score, and the change rate of somatomotor network synchronization in PD group. D: The difference of somatomotor network synchronization among HC, PD during OFF and ON.
    Comparison of CNRLC between the HC and PD groups. (A) Signal intensity measurements of the LC (two small red circles) and pontine (a big red circles) from a HC; (B) and (C) The location of LC in continuous layers (red arrow); (D) Significantly decreased CNRLC was found in PD group when compared with HC group.
  • Automatic assessment of motion artifact on Nigrosome 1 visualization protocol using CNN-LSTM
    Junghwa Kang1, Na Young Shin2, and Yoonho Nam1,2
    1Division of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin, South Korea, yongin, Korea, Republic of, 2Seoul St.Mary’s Hospital, Department of Radiology, The Catholic University of Korea, Seoul, South Korea, Seoul, Korea, Republic of
    We proposed to evaluate the degree of motion artifact on high-resolution magnetic susceptibility contrast images for N1 visualization that is sensitive to patient’s motions. We introduced deep CNN-LSTM network. The proposed method could be helpful in clinical use.
    Figure 3. The architecture of CNN-LSTM for motion assessment
    Figure 4. Confusion Matrix and ROC analysis (validation set) (a), (c) CNN , (b), (d) CNN-LSTM.
  • Quantitative mapping of substantia nigra iron and neuromelanin in Parkinson’s Disease
    Jiahao Li1,2, Kelly Gillen1, Ilhami Kovanlikaya1, Thanh Nguyen1, Alexey Dimov1, Kailyn Li1, Weiyuan Huang1, Xianfu Luo1, Carly Skudin1, Eileen Chang1, Alexander Shtilbans1,3, and Yi Wang1,2
    1Weill Cornell Medicine, New York, NY, United States, 2Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States, 3Hospital for Special Surgery, New York, NY, United States
    There is an increase in susceptibility but a decrease in neuromelanin in the SN of PD subjects as compared to controls.
    Figure 2. Representative QSM images from control, RBD, PD Stage 1 and PD Stage 2 subjects. Note increase in susceptibility in substantia nigra (indicated by red arrows) in PD Stage 1 and PD Stage 2 compared to healthy controls or RBD. Scale bar in ppm (parts per million); RBD, REM-sleep behavior disorder
    Figure 1. Average susceptibility in the substantia nigra (SN; left) and red nucleus (RN; right) across all cohorts. Left: there is significant increase in susceptibility PD as compared to controls. Right: there are no statistically significant differences in susceptibility in the RN across all cohorts. RBD, REM-sleep behavior disorder; ppm, parts per million; *, p < 0.05 (multiple comparison t-test, Bonferroni correction)
  • Substantia nigra magnetic resonance spectrum in differentiating tremor-dominant Parkinson’s disease from essential tremors
    Rushi Chen1, Yan Bai1, Qin Feng1, Menghuan Zhang1, Xianchang Zhang2, and Meiyun Wang1
    1Henan provincial people's hospital, Zhengzhou, China, 2MR Collaboration, Siemens Healthcare Ltd, Beijing, China, Beijing, China
    The NAA/Cr ratio derived from MRS in the contralateral SN was significantly higher in tremor-dominant PD than in ET.
    Figure 1. NAA/Cr was significantly decreased in the contralateral SN of a patient with tremor-dominant Parkinson’s disease (PD) (A) compared with that of a patient with essential tremors (ET).
  • Feasibility of a short but comprehensive MRI protocol for quantitative characterization of progressive neurodegeneration in Friedreich ataxia
    Koene R.A. Van Dijk1, Courtney A. Bishop2, James O’Callaghan2, James A. Goodman1, Laigao Chen1, Peter T. Loudon3, Lawrence Charnas4, Eugenii A. Rabiner2, and Richard Festenstein5
    1Digital Medicine and Translational Imaging, Early Clinical Development, Pfizer, Cambridge, MA, United States, 2Invicro, London, United Kingdom, 3Clinical Sciences, Early Clinical Development, Pfizer, Cambridge, United Kingdom, 4Rare Disease Research Unit, Pfizer, Cambridge, MA, United States, 5Department of Brain Sciences, Imperial Clinical Research Facility and BRC (NIHR), Imperial College London, London, United Kingdom
    We found good quality data for the majority of sequences and patients in a scan session lasting less than 60 minutes and show sample imaging and spectroscopy data focusing on the dentate nucleus, a structure involved in planning, initiating, and modifying voluntary movements.
    Figure 2. Sample dentate nucleus ROIs and contrast of mpMRI data at 3T. T1w: blue indicates the location of the participants’ left dentate nucleus; QSM: heatmap indicates estimated iron content in the left dentate nucleus mask as measured via R2* mapping (color scale shows rendering of values 5.0-40.0 s-1); DWI: dominant fiber orientation coded as follows: red=left-right, green=anterior-posterior, blue=superior-inferior. DD=disease duration. SARA=Scale for the Assessment and Rating of Ataxia.
    Figure 3. Sample spectroscopy data. Green circles indicate the location of the Myo-inositol peak (3.5ppm) for FA patient #4 (left panel) and a healthy volunteer (HV; center panel). The white square shows the location of the MRS volume of interest placed over the right dentate nucleus (right panel).
  • Swallow tail sign and nigrosome 1  -  close but not quite the same
    Malte Brammerloh1,2, Evgeniya Kirilina1,3, Anneke Alkemade4, Pierre-Louis Bazin1,4, Caroline Jantzen1, Carsten Jäger1,5, Andreas Herrler6, Kerrin J. Pine1, Penny Gowland7, Markus Morawski5, Birte Forstmann4, and Nikolaus Weiskopf1,2
    1Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany, 2Faculty of Physics and Earth Sciences, Leipzig University, Leipzig, Germany, 3Center for Cognitive Neuroscience Berlin, Freie Universität Berlin, Berlin, Germany, 4Integrative Model-based Cognitive Neuroscience Research Unit, University of Amsterdam, Amsterdam, Netherlands, 5Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany, 6Department of Anatomy and Embryology, Maastricht University, Maastricht, Netherlands, 7Sir Peter Mansfield Imaging Center, University of Nottingham, Nottingham, United Kingdom
    The swallow tail sign in T2*-weighted MR images of the substantia nigra does not show a one-to-one correspondence to nigrosome 1, as demonstrated by an overlay of in vivo and postmortem MRI and 3D histology.
    Combining in vivo (A) and postmortem (B) MRI with 3D immunohistochemistry (D, E) to study the anatomical underpinning of the swallow tail sign (ST). The ST was segmented (C) as a bright stripe in SN on in vivo MRI (A). N1 was segmented (F) as dark-pigmented areas on grayscale BF images (D) verified by calbindin immunohistochemistry (E). Co-registration (G) of 3D immunohistochemistry, postmortem MRI and in vivo MRI revealed contrast mechanisms of N1 and its relation to the swallow tail sign.
    Masks of the ST and N1 for three randomly assigned pairs of in vivo and postmortem datasets (A, B, C) overlaid over BF. In all cases, ST covered a large part of N1. While N1 consistently showed a narrow width, ST was approximately twice as wide as N1. The ST only covered the superior-posterior-lateral portion of N1, while it did not match the ventromedial part of the rostral extent of N1. The anatomical medial (M), lateral (L), superior (S), and inferior (I) directions are illustrated in A.
Back to Top
Digital Poster Session - Parkinson's Disease & Non-PD Dementia
Neuro
Wednesday, 19 May 2021 15:00 - 16:00
  • Subcortical grey matter changes associated with motor skills in Parkinson Disease: a longitudinal study
    Céline Charroud1 and Luca Turella1
    1Center for Mind/Brain Sciences - CIMeC, Mattarello, Italy
    An atrophy in all subcortical regions was observed over time in Parkinson Disease. A reduced volume in thalamus and an increased volume in pallidum may be related to the decline in motor skills. VBM and volumetry methods should be used jointly.

    Figure 2: Statistical parametric maps showing positive and negative correlation between grey matter volume and UPDRS-III scores at 48 months using voxel-based morphometry analyses.

    Results showed a positive association between grey matter volume and UPDRS-III in right pallidum at 48 months (p<0.05 TFCE, FWE corrected, age, sex and total intracranial volume adjusted). Conversely, a negative association was identified in bilateral thalamus at 48 months. No findings were significant at baseline.

    Figure 4: Correlation between grey matter volume and UPDRS-III scores at baseline and 48 months.

    At baseline, one positive correlation was identified between grey matter volume and UPDRS-III scores in the right pallidum. At 48 months, a positive correlation was observed between grey matter volume and UPDRS-III scores in bilateral pallidum whereas a negative correlation was identified in bilateral thalamus. Pearson correlations are reported with a statistical threshold of p<0.05.

  • The perfusion deficits in general cognitive, executive, and visual dysfunction in Parkinson's disease measured by arterial spin labeling MRI
    Dilek Betul Arslan1, Hakan Ibrahim Gurvit2, Ozan Genc1, Sevim Cengiz1, Ani Kicik3,4, Kardelen Eryurek4,5, Emel Erdogdu4,6, Zerrin Yildirim2, Zeynep Tufekcioglu2, Aziz Mufit Ulug1,7, Basar Bilgic2, Hasmet Hanagasi2, Tamer Demiralp4,8, and Esin Ozturk-Isik1
    1Biomedical Engineering, Biomedical Engineering Institution, Bogazici University, Istanbul, Turkey, 2Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey, 3Department of Physiology, Faculty of Medicine, Demiroglu Bilim University, Istanbul, Turkey, 4Neuroimaging Unit, Hulusi Behcet Life Sciences Research Center, Istanbul University, Istanbul, Turkey, 5Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey, 6Department of Psychology, Faculty of Arts and Sciences, Isik University, Istanbul, Turkey, 7CorTechs Labs, San Diego, CA, United States, 8Department of Physiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
    Hypoperfusion pattern related to cognitive and executive dysfunction was found in Parkinson's disease in line with  measured by Arterial Spin Labeling MRI
    Figure 1. Regions of significant CBF decreases of LP compared to HP in all of the participants according to general cognitive and executive domain scores. The color bars represent t values. R: Right, L: Left.

    Table 1. Regions of CBF differences between two groups defined based on composite z-scores in general cognitive domain and executive domain within the entire subject group.

    * The composite z-scores are the average of the z-converted raw scores of ACE-R, MMSE and MOCA. Median of the composite z-scores: - 1.22.

    ** The composite z-scores are the average of the z-converted raw scores of WCST percentage of perseverative responses score and Stroop interference duration score. Median of the composite z-scores: 0.56.

  • Disrupted Functional Connectivity of the Nucleus basalis of Meynert in Parkinson's Disease patients with Mild Cognitive Impairment
    Chao Zhang1, Weiqiang Dou2, and Kai Xu1
    1Department of Radiology, Xuzhou Medical University, Xuzhou, China, 2GE Healthcare,MR Research China, Beijing, China
    Our study provides new insights into the neural basis of cognitive dysfunction in PD patients. We also found that BNM-FC can be an effective feature to distinguish PD-MCI from PD-NC.
    Figure 1. Functional Connectivity (FC) between the basal nucleus of Meynert (BNM) and whole brain. (A–C): BNM-FC was evaluated in healthy control (HC) (A), Parkinson’s disease with normal cognition (PD-NC) (B), and Parkinson’s disease with mild cognitive impairment (PD-MCI) (C) groups. (D): BNM-FC was decreased in the right superior parietal lobe (SPL) of the PD-MCI group compared to the PD-NC and HC groups.
    Figure 2. Classification accuracy of functional connectivity (FC) in the basal nucleus of Meynert (BNM) (BNM-FC) in the right superior parietal lobe (SPL) determined by the leave-one-out cross-validation (LOOCV) method in distinguishing Parkinson’s disease with mild cognitive impairment (PD-MCI) from Parkinson’s disease with normal cognition (PD-NC) patient (A–C). Box plots with whiskers (min–max) show BNM-FC in the right SPL of the three groups (D).
  • Automated Magnetic Resonance Parkinsonism Index: Test-Retest Reliability and 10 Year Changes in Aging and Parkinson’s Disease
    Yao-Chia Shih1,2, Bénédicte Maréchal3,4,5, Ricardo Corredor Jerez3,4,5, Septian Hartono2,6, Hui-Hua Li2,7, Isabel Hui Min Chew1, Eng King Tan2,6, and Ling Ling Chan1,2
    1Department of Diagnostic Radiology, Singapore General Hospital, Singapore, Singapore, 2Duke-NUS Medical School, Singapore, Singapore, 3Advanced Clinical Imaging Technology, Siemens Healthcare AG, Lausanne, Switzerland, 4Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland, 5LTS5, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland, 6Department of Neurology, National Neuroscience Institute (Outram-campus), Singapore, Singapore, 7Health Services Research Unit, Singapore General Hospital, Singapore, Singapore
    The automated MRPI using the proposed algorithm was expeditious (averaging 25s ± 2s per case) and reliable (average coefficient of variance <18%) as measured on >2000 brain scans across different 1.5T and 3T MRI systems, subject cohorts and time-points.
    Fig. 1 Identical paired standard T1-weighted images of a healthy 84 year-old male control illustrating automated anatomical placement of color-coded landmarks for midbrain (yellow) and pons (blue) areas (first row), followed by left and right SCP and MCP widths respectively. Abbreviation: MCP = Middle Cerebellar Peduncle, SCP = Superior Cerebellar Peduncle.
    Fig. 3 Bar charts of coefficients of variance (CV, y-axes) of values and subcomponents of MRPI (x-axes) from (A) manual and automated MRPI derived from longitudinal SGH-PD cohorts, and (B) automated MRPI derived from single TP ADNI1 (1.5T) and ADNI 2/GO (3T) cohorts. Abbreviation: MCP = Middle Cerebellar Peduncle, SCP = Superior Cerebellar Peduncle, TP = Time Point.
  • Magnetic resonance characteristics of iron and neuromelanin in early Parkinson’s disease: correlation with vesicular monoamine transporter 2
    xueling Liu1, Pu-Yeh Wu2, and Yuxin Li1
    1Department of Radiology, Huashan hospital affiliated to Fudan University, Shanghai, China, 2GE Healthcare, Beijing, China, Shang Hai, China
    Our results suggested that changes of neuromelanin and susceptibility on QSM images could quantitatively reflect the pathology of EPD and could be used as imaging biomarkers for diagnosing of EPD. The SUR measured by 18F-DTBZ images could reflect the lateralization in EPD patients.
    Fig 1. The volume and contrast ratio (CR) of neuromelanin, and susceptibility in the substantia nigra pars compacta in HC and EPD
    Fig 2. Correlations between the contrast ratio (CR) of neuromelanin and susceptibility in the substantia nigra pars compacta in HC and EPD groups (adjusted for age and gender)
  • Value of magnetic resonance fingerprinting for differentiating tremor-dominant Parkinson’s disease from essential tremors
    Yan Bai1,2, Rushi Chen1,2, Wei Wei1,2, Rui Zhang1,2, Zhun Huang2,3, Xianchang Zhang4, Mathias Nittka5, Gregor Koerzdoerfer5, and Meiyun Wang1,2
    1Department of Radiology, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Academy of Medical Sciences., Zhengzhou, China, 2Henan Key Laboratory for Medical Imaging of Neurological Diseases, Zhengzhou, Henan, China, ZhengZhou, China, 3Department of Radiology, Henan University People’s Hospital & Henan Provincial People’s Hospital, School of Basic Medicine., Zhengzhou, China, 4MR Collaboration, Siemens Healthcare Ltd., Beijing, China, BeiJing, China, 5MR Pre-development, Siemens Healthcare GmbH, Erlangen, Germany, Erlangen, Germany
    Conventional magnetic resonance imaging cannot reliably differentiate Parkinson’s disease (PD) from essential tremors (ET). Magnetic resonance fingerprinting (MRF) can simultaneously acquire T1 and T2 relaxometry. This study utilized MRF to obtain T1 and T2 values in substantia nigra (SN) of patients with tremor-dominant PD and ET. The T1 values of SN were significantly higher in patients with tremor-dominant PD than those with ET, whereas the T2 values showed no significant differences between groups. The findings suggest that MRF T1 mapping of the SN can potentially differentiate tremor-dominant PD from ET
    Figure 1. (A) T1 map and (B) T2 map from a patient with tremor-dominant Parkinson’s disease (PD). (C) T1 map and (D) T2 map from a patient with essential tremors (ET). The T1 value in the substantia nigra is significantly higher in the patient with tremor-dominant PD than in the patient with ET.
  • STN/GP-nets: Fully automatic deep-learning based segmentation for DBS applications using ultra-high 7 Tesla MRI
    Oren Solomon1, Tara Palnitkar1,2, Rémi Patriat1, Henry Braun1, Joshua Aman2, Michael C Park2,3, Guillermo Sapiro4, Jerrold Vitek2, and Noam Harel1,3
    1Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, United States, 2Department of Neurology, University of Minnesota, Minneapolis, MN, United States, 3Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States, 4Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Department of Computer Science, Department of Mathematics, Duke University, Durham, NC, United States
    Deep-learning based segmentation of subcortical structures from 7 Tesla MRI allows for patient-specific, robust and accurate deep brain stimulation surgery planning and post-operative lead location assessment. 
    Figure 3: (A) 3D reconstruction of a DBS electrode placement with respect to the manually delineated GPe (green)/GPi (yellow) for a specific PD patient. (B) 3D reconstruction of the same patient’s data showing the DBS electrode with respect to GP-net’s segmentation of the GPe (orange)/GPi (blue). Green arrow – anterior (A), blue arrow – superior (S) and red arrow – right (R).
    Figure 4: (A) 3D reconstruction of a DBS electrode placement with respect to the manually delineated STN for a specific PD patient. (B) 3D reconstruction of the same patient’s data showing the DBS electrode with respect to STN-net’s segmentation of the STN. Green arrow – anterior (A), blue arrow – superior (S) and red arrow – right (R).
  • Evaluation and Quantification of SWI MRA of Cerebral Vasculature in Parkinson’s Disease Patients
    Dmytro Pylypenko1, Yuhui Xiong1, Lanxin Ji1, Le He1, Yu Ma2, and Hua Guo1
    1Center for Biomedical Imaging Research, Department of Biomedical Engineering, Tsinghua University, Beijing, China, 2Department of Neurosurgery, Tsinghua University Yuquan Hospital, Beijing, China
    In this study, we performed a comparative study of the vascular geometry in PD patients vs. AMC to quantify the structural changes in the cerebral vasculature by analyzing SWI MRA data.
    Figure 1. The results of the Segmentation and Skeletonization for the PD patients. a) Male PD patient, 63 years old. The venous tree is visually incomplete, the superficial cortical veins are missing (red arrowheads); b) Male PD patient, 74 years old. Increased tortuosity and fractal dimension (red arrowheads) of the venous system.
    Figure 2. The results of the Segmentation and Skeletonization of the AMC, Female, 48 years old.
  • Cerebral hyper-perfusion associated with mild cognitive impairment in de novo Parkinson’s disease
    Yong Zhang1, Chang-Peng Wang2, Jian Wang2, Li-Rong Jin2, and Bing Wu3
    1GE Healthcare, Shanghai, China, 2Zhongshan Hospital, Shanghai, China, 3GE Healthcare, Beijing, China
    Increased CBF was detected in the early de novo PD patients with mild cognitive impairment as compared to controls, as well as higher perfusion relative to PD patients with normal cognition. Cerebral hyper-perfusion in frontal lobe might be associated with cognitive decline in early de novo PD.
    CBF comparison between PD-MCI and normal controls (Yellow: PD-MCI>NC, corrected p<0.05)
    CBF comparison between PD-MCI and PD-NC (Yellow: PD-MCI>PD-NC, corrected p<0.05)
  • Simultaneous multi-parameter mapping to characterize Parkinson’s Disease using strategically acquired gradient echo imaging
    Yu Shen1, Xianchang Zhang2, Yan Bai1, Yaping Wu1, Ewart Mark Haacke3, Bo Wu4, Yongsheng Chen4, Rui Zhang1, Rushi Chen1, Wei Wei1, and Meiyun Wang1
    1Department of Medical Imaging, Henan Provincial People’s Hospital & Zhengzhou University, Zhengzhou, China, 2MR Collaboration, Siemens Healthcare Ltd. Beijing China, Beijing, China, 3Wayne State University, Detroit, MI, United States, 4Shanghai Zhuyan Medical Technology Company, Shanghai, China
    Strategically acquired gradient echo imaging is a useful and accurate method to examine structural brain changes and iron accumulation in patients with Parkinson’s disease.
    Figure 2. Images showing brain regions with significant differences between PD and healthy controls (HC). (A) The left temporal fusiform cortex, posterior division (TFCp) is shown in red, right TFCp is shown in green. (B) The right parabrachial pigmented (PBP) nucleus is shown in blue. R2* of right TFCp and right PBP was significantly higher in PD than in HC; T2* of left TFCp, right TFCp and right PBP was significantly lower in PD than in HC; T1 mapping ofright PBP was significantly lower in PD than in HC.
    Figure 1. Images from T1-weighted enhanced imaging (T1WE), T1 mapping, T2*, R2*, and susceptibility-weighted imaging (SWI) mapping acquired using strategically acquired gradient echo imaging. T1WE shows improved image quality over the other methods.
  • Assessing the differences in diffusion measures of basal ganglia and basal ganglia circuitry between controls and Parkinson’s disease patients
    Jae-Hyuk Shim1 and Hyeon-Man Baek1
    1Gachon University, Incheon, Korea, Republic of
    Segmentation of basal ganglia structures and fiber tracking between basal ganglia segmentations for comparing diffusion measures such a between controls and Parkinson's disease patients shows potential biomarkers for diagnosing PD. 
    Figure 1. Results of MNI PD25 atlas segmentation using Lead-DBS SPM and ANTs co-registration, normalization. Left shows the 22 structures in 3D models overlaid on top of an example subject’s b0 FA image.
    Figure 2. Connectivity matrices of significant differences in qa, fa, md, ad, rd of MNI PD25 basal ganglia structure interconnectivity. Left column represents connectivity matrix of mean differences in qa, fa, md, ad and rd diffusion measures. Middle column represents p-values of student t-tests done for every connectivity matrix pair with significance at p < 0.05. Right column represents the adjusted p-values of t-tests using Benjamin-Hochberg procedure with p < 0.2.
  • Exposure to Traffic-Related Particulate Matter Impact Motor Function and White Mater Integrity in Elder Rodent Model
    Ting-Chieh Chen1, Yu-Chun Lo2, Yi-Chen Lin1, Ssu-Ju Li1, Ting-Chun Lin1, Ching-Wen Chang1, Yao-Wen Liang1, Hsiao-Chi Chuang3, and You-Yin Chen1,2
    1Biomedical Engineering, National Yang-Ming University, Taipei, Taiwan, 2Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taipei, Taiwan, 3School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
    Our findings provided the brain MRI tractographic and behavioral evidences indicating the negative impacts of the traffic-related PM on motor functions as the possible associations with the development of PD.
    Figure 1. Experimental design for investigating the effects of PM. According to the living condition, three experimental groups were established, including a control group, HEPA group and PM group. The tractography analysis and the rotarod performance test were conducted to respectively examine the effects of PM on the structural changes within rat brains and the motor-behavioral performances.
    Figure 2. Whole-brain in-vivo DTI tractography. Based on the function of the neural circuits, DTI tractography analysis was conducted with the ROIs divided into the following three groups, including (A) the olfactory circuit, (B) motor control circuit and (C) dopaminergic pathway.
  • Excessive Brain Iron Deposition Detected by QSM in Extrapyramidal System in Parkinson's with Type 2 Diabetes Melitus Patients
    Wanyao Li1, Yanwei Miao1, Bingbing Gao1, Yangyingqiu Liu1, Ailian Liu1, and Qingwei Song1
    1First Affiliated Hospital of Dalian Medical University, Dalian, China
    Our results display that diabetes can aggravate iron deposition due to the abnormal metabolism of dopamine in PD patients.
    Table 1 The clinical data of all groups
    Figure 1 ROI selection for gray matter nuclei
  • Brain white and gray matter alterations in early-stage Parkinson’s disease with GBA1 gene mutations evaluated using free water imaging
    Christina Andica1, Koji Kamagata1, Masahiro Abe1, Wataru Uchida1,2, Yuya Saito1, Hayato Nozaki1,2, Kaito Takabayashi1, Masaaki Hori3, and Shigeki Aoki1
    1Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan, 2Department of Radiological Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan, 3Department of Radiology, Toho University Omori Medical Center, Tokyo, Japan
    Using free water imaging, we showed the role of GBA1 mutations in early-stage drug-naïve Parkinson’s disease, including brain compensatory mechanisms and neuroinflammation, which provides opportunities for developing disease-modifying therapies and disease progression biomarkers.

    Figure 3. Box plots of FW for the HC, GBA-PD, and iPD groups in Braak stages III and IV. The bottom and top of the box are the first and third quartiles, and the thick band inside the box is the median. Whiskers represent maximum and minimum values of all data.

    Abbreviations: FW, free water, GBA-PD, Parkinson’s disease with GBA1 mutations; HC, healthy controls; and iPD, idiopathic Parkinson’s disease.

    Figure 2. Box plots and tract profiles of FAT, MDT, and FW for HC, GBA-PD, and iPD groups in the white matter pathways.

    Abbreviations: ATR, anterior thalamic radiation; CCG, cingulum cingulate gyrus; FAT, free water-corrected fractional anisotropy; Fmajor, forceps major; Fminor, forceps minor; FW, free water; GBA-PD, Parkinson’s disease with GBA1 mutations; HC, healthy controls; ILF, inferior longitudinal fasciculus; iPD, idiopathic Parkinson’s disease; MDT, free water-corrected mean diffusivity; UF, uncinate fasciculus.

  • Evidence on the association between executive functions and tractography-derived nodal properties in Parkinson's disease
    Giacomo Tomezzoli1,2, Lisa Novello2, Francesca Saviola2, Stefano Tambalo2, Beatrice Federica Luciani2, Enrica Pierotti2, Céline Charroud 2, Alessandro Gober2, Francesca Giacomoni2, Pamela Narduzzi2, Claudia Meli2, Marika Falla2, Alessandra Dodich2, Luca Turella2, Costanza Papagno2, and Jorge Jovicich2
    1DiPSCo, Department of Psychology and Cognitive Science, University of Trento, Rovereto, Italy, 2CIMeC, Center for Mind/Brain Sciences, University of Trento, Rovereto, Italy
    We provide initial evidence for associations between brain tractography-derived nodal properties and executive functions performance assessment in brain executive regions in a group of Parkinson’s patients.
    Figure 5: Betweenness Centrality and nodal Clustering Coefficient positive correlations (red nodes) and negative correlations (blue nodes) with EFs scores in nodes enrolled in the analysis. SFG: superior-frontal-gyrus; IPG: inferior-supramarginal-parietal-gyrus.
    Figure 3: Topological distribution of node degree (k); the size of each node represents k. SFG: superior-frontal-gyrus; OG: orbital-gyrus; SPG: superior-parietal-gyrus; IPG: inferior-supramarginal-parietal-gyrus
  • Patient-specific hyperdirect pathway activation in DBS for Parkinson’s Disease
    Alba Segura Amil1,2 and T. A. Khoa Nguyen1,2
    1Department of Neurosurgery, Inselspital, University Hospital Bern, Bern, Switzerland, 2ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
    The effect threshold was at 20% of hyperdirect pathway activation, while the side effect threshold was at 2% of corticospinal tract activation. 
    3D illustration of DBS leads and subcortical structures. Subthalamic nucleus in orange, volume of tissue activated in red and streamlines in dark blue.
    Hyperdirect pathway activation. In blue, points corresponding to not clinical effect; in orange, points corresponding to clinical effect. Correct predictions are marked as circles and incorrect predictions as crosses.
  • Brain structure network mediating the association between iron deposition and progression of Parkinson Disease
    Bingxue Cheng1, Chenfei Ye2, and Ting Ma1,2,3
    1Department of Information and Electronics, Harbin Institute of Technology at Shenzhen, GuangDong Province, China, 2Peng Cheng laboratory, Guangdong Province, China, 3National Clinical Research Center for Geriatric Disorders, Beijing, China
    The small-worldness and rich-club coefficients of brain structural network in PD significantly mediate the association between the mean QSM value in left putamen and H&Y scale.
    Table 2. Small-worldness mediation analysis results
    Table 3. Rich-club coefficients mediation analysis results
  • Abnormal intrinsic brain functional network dynamics associated with tremor in Parkinson’s disease
    Junlan Zhu1, Guanxun Cheng1, Qiaoling Zeng1, Chao Lai1, Jiao Li1, and Shuwen Dong1
    1Department of Medical Imaging, Peking University Shenzhen Hospital, Shenzhen, China
    Our results suggest that temporal variability of functional connectivity profile can detect the aberrant dynamic connectivity patterns, which were associated with the clinical deficits in Parkinson disease tremor and thus may deepen our understanding.
    Figure2:Cluster centroids of brain states
    Figure1: Independent functional components and corresponding static functional connectivity network
  • Motor Cerebro-Cerebellar Networks Breakdown Among Different Subtypes of Parkinson’s Disease
    Silvia Basaia1, Federica Agosta1,2,3, Alessandro Francia1, Camilla Cividini1,3, Tanja Stojkovic4, Iva Stankovic4, Rosita Di Micco1, Luigi Albano1, Elisabetta Sarasso1, Noemi Piramide1,3, Vladana Markovic4, Elka Stefanova4, Vladimir S. Kostic4, and Massimo Filippi1,2,3,5,6
    1Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy, 2Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy, 3Vita-Salute San Raffaele University, Milan, Italy, 4Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Italy, 5Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy, 6Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
    We investigated the functional neural organization of the motor cerebro-cerebellar system in Parkinson’s disease (PD) patients with tremor-dominant (TD) or postural instability and gait disorder (PIGD) variant.
  • Diffusion Tensor Imaging and Tractography in Deep Brain Stimulation Postoperative Patients with Parkinson's Disease
    Yan Li1, Yu Liu1, Naying He1, Chencheng Zhang2, Yijie Lai2, Hongyang Li2, Qing Li3, Caixia Fu4, Fuhua Yan1, and Ewart Mark Haacke1,5
    1Department of Radiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China, 2Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China, 3MR Collaborations, Siemens Healthcare Ltd., Shanghai, China, 4MR Application Development, Siemens Shenzhen Magnetic Resonance Ltd., Shenzhen, China, 5Department of Radiology, Wayne State University, Detroit, MI, United States
    Both DBS ON and OFF diffusion weighted imaging are safe and the tractography results are significantly comparable. DBS ON DTI is particularly fitting for those patients who cannot control their disease symptoms without stimulation but need MRI.
    Table 3 The track count number of nigrostriatal pathway of both left and right sides in DBS ON/OFF images and their comparisons.
    Figure 2 The deterministic tracking result of the same representative subject in two states: a) DBS ON, and b) DBS OFF.