1
|
van der Horn HJ, Vakhtin AA, Julio K, Nitschke S, Shaff N, Dodd AB, Erhardt E, Phillips JP, Pirio Richardson S, Deligtisch A, Stewart M, Suarez Cedeno G, Meles SK, Mayer AR, Ryman SG. Parkinson's disease cerebrovascular reactivity pattern: A feasibility study. J Cereb Blood Flow Metab 2024; 44:1774-1786. [PMID: 38578669 PMCID: PMC11494834 DOI: 10.1177/0271678x241241895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 04/06/2024]
Abstract
A mounting body of research points to cerebrovascular dysfunction as a fundamental element in the pathophysiology of Parkinson's disease (PD). In the current feasibility study, blood-oxygen-level-dependent (BOLD) MRI was used to measure cerebrovascular reactivity (CVR) in response to hypercapnia in 26 PD patients and 16 healthy controls (HC), and aimed to find a multivariate pattern specific to PD. Whole-brain maps of CVR amplitude (i.e., magnitude of response to CO2) and latency (i.e., time to reach maximum amplitude) were computed, which were further analyzed using scaled sub-profile model principal component analysis (SSM-PCA) with leave-one-out cross-validation. A meaningful pattern based on CVR latency was identified, which was named the PD CVR pattern (PD-CVRP). This pattern was characterized by relatively increased latency in basal ganglia, sensorimotor cortex, supplementary motor area, thalamus and visual cortex, as well as decreased latency in the cerebral white matter, relative to HC. There were no significant associations with clinical measures, though sample size may have limited our ability to detect significant associations. In summary, the PD-CVRP highlights the importance of cerebrovascular dysfunction in PD, and may be a potential biomarker for future clinical research and practice.
Collapse
Affiliation(s)
- Harm Jan van der Horn
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Andrei A Vakhtin
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Kayla Julio
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Stephanie Nitschke
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Nicholas Shaff
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Andrew B Dodd
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Erik Erhardt
- Department of Mathematics and Statistics, University of New Mexico, Albuquerque, NM, USA
| | - John P Phillips
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Sarah Pirio Richardson
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, NM, USA
- New Mexico VA Health Care System, Albuquerque, NM, USA
| | - Amanda Deligtisch
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Melanie Stewart
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Gerson Suarez Cedeno
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Sanne K Meles
- Department of Neurology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Andrew R Mayer
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Sephira G Ryman
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
2
|
He C, Yang R, Rong S, Zhang P, Chen X, Qi Q, Gao Z, Li Y, Li H, de Leeuw FE, Tuladhar AM, Duering M, Helmich RC, van der Vliet R, Darweesh SKL, Liu Z, Wang L, Cai M, Zhang Y. Temporal evolution of microstructural integrity in cerebellar peduncles in Parkinson's disease: Stage-specific patterns and dopaminergic correlates. Neuroimage Clin 2024; 44:103679. [PMID: 39366283 PMCID: PMC11489329 DOI: 10.1016/j.nicl.2024.103679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/25/2024] [Accepted: 09/28/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Previous research revealed differences in cerebellar white matter integrity by disease stages, indicating a compensatory role in Parkinson's disease (PD). However, the temporal evolution of cerebellar white matter microstructure in patients with PD (PwPD) remains unclear. OBJECTIVE To unravel temporal evolution of cerebellar white matter and its dopaminergic correlates in PD. METHODS We recruited 124 PwPD from the PPMI study. The participants were divided into two subsets: Subset 1 (n = 41) had three MRI scans (baseline, 2 years, and 4 years), and Subset 2 (n = 106) had at least two MRI scans at baseline, 1 year, and/or 2 years. Free water-corrected diffusion metrics were used to measure the microstructural integrity in cerebellar peduncles (CP), the main white matter tracts connecting to and from the cerebellum. The ACAPULCO processing pipeline was used to assess cerebellar lobules volumes. Linear mixed-effect models were used to study longitudinal changes. We also examined the relationships between microstructural integrity in CP, striatal dopamine transporter specific binding ratio (SBR), and clinical symptoms. RESULTS Microstructural changes in CP showed a non-linear pattern in PwPD. Free water-corrected fractional anisotropy (FAt) increased in the first two years but declined from 2 to 4 years, while free water-corrected mean diffusivity exhibited the opposite trend. The initial increased FAt in CP correlated with cerebellar regional volume atrophy, striatal dopaminergic SBR decline, and worsening clinical symptoms, but this correlation varied across disease stages. CONCLUSIONS Our findings suggest a non-linear evolution of microstructural integrity in CP throughout the course of PD, indicating the adaptive structural reorganization of the cerebellum simultaneously with progressive striatal dopaminergic degeneration in PD.
Collapse
Affiliation(s)
- Chentao He
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province 510080, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Rui Yang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province 510080, China
| | - Siming Rong
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province 510080, China
| | - Piao Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province 510080, China
| | - Xi Chen
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province 510080, China
| | - Qi Qi
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province 510080, China
| | - Ziqi Gao
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province 510080, China
| | - Yan Li
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province 510080, China
| | - Hao Li
- Radboud University Medical Center, Nijmegen, Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, the Netherlands
| | - Frank-Erik de Leeuw
- Radboud University Medical Center, Nijmegen, Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, the Netherlands
| | - Anil M Tuladhar
- Radboud University Medical Center, Nijmegen, Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, the Netherlands
| | - Marco Duering
- Medical Image Analysis Center (MIAC AG) and Department of Biomedical Engineering, University of Basel, Basel, Switzerland; Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Germany
| | - Rick C Helmich
- Radboud University Medical Center, Nijmegen, Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, the Netherlands
| | - Rick van der Vliet
- Department of Neurology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Sirwan K L Darweesh
- Radboud University Medical Center, Nijmegen, Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, the Netherlands
| | - Zaiyi Liu
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China; Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province 510080, China
| | - Lijuan Wang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province 510080, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Mengfei Cai
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province 510080, China; Radboud University Medical Center, Nijmegen, Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, the Netherlands.
| | - Yuhu Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province 510080, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.
| |
Collapse
|
3
|
Du X, Zhao H, Li Y, Dai Y, Gao L, Li Y, Fan K, Sun Z, Zhang Y. The value of PET/CT in the diagnosis and differential diagnosis of Parkinson's disease: a dual-tracer study. NPJ Parkinsons Dis 2024; 10:171. [PMID: 39256393 PMCID: PMC11387816 DOI: 10.1038/s41531-024-00786-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
Positron emission tomography/computed tomography (PET/CT) is a molecular imaging method commonly used to diagnose and differentiate Parkinson's disease (PD). This study aimed to evaluate the performance of PET/CT with 11C-2β-Carbomethoxy-3β-(4-fluorophenyl) tropane (11C-CFT) and 18F-fluorodeoxyglucose (18F-FDG) tracers in the differential diagnosis between PD, multiple system atrophy parkinsonian type (MSA-P), progressive supranuclear palsy (PSP) and vascular parkinsonism (VP) using the data of 220 patients with clinical PD-like symptoms. Of the 220 enrolled patients, 166 (PD, n = 80; MSA-P, n = 54; PSP, n = 15; VP, n = 17) completed the motor, cognitive and PET/CT assessment and were included in this study. 11C-CFT and 18F-FDG PET/CT images were analyzed using the SNBPI toolbox and CortexID Suite software. The uptake values of 11C-CFT and 18F-FDG PET/CT were compared among the groups after controlling for covariates using generalized linear models. Receiver operating characteristic (ROC) curves were generated to estimate the diagnostic values. Patients with PSP showed the most significant reduction on 11C-CFT PET/CT, while patients with PD and MSA-P showed similar reductions, and patients with VP did not show any significant reduction in 11C-CFT uptake. The areas under the curve (AUCs) for 11C-CFT PET/CT for distinguishing PD from VP, PSP, and MSA-P were 0.902, 0.830, and 0.580, respectively, and 0.728 for distinguishing advanced-stage PD from PSP. On 18F-FDG PET/CT, the AUCs for distinguishing PD from PSP and MSA-P were 0.968 and 0.963, respectively. These results suggest that 11C-CFT and 18F-FDG PET/CT complement each other in improving the accuracy in differential diagnosis of PD.
Collapse
Affiliation(s)
- Xiaoxiao Du
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Hongguang Zhao
- Nuclear Medicine Department, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yinghua Li
- Nuclear Medicine Department, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yuyin Dai
- Nuclear Medicine Department, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Lulu Gao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yi Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Kangli Fan
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhihui Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ying Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
4
|
Chen B, Chen X, Peng L, Liu S, Tang Y, Gao X. Metabolic network connectivity disturbances in Parkinson's disease: a novel imaging biomarker. Cereb Cortex 2024; 34:bhae355. [PMID: 39329355 DOI: 10.1093/cercor/bhae355] [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: 04/15/2024] [Revised: 07/20/2024] [Accepted: 08/14/2024] [Indexed: 09/28/2024] Open
Abstract
The diagnosis of Parkinson's Disease (PD) presents ongoing challenges. Advances in imaging techniques like 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) have highlighted metabolic alterations in PD, yet the dynamic network interactions within the metabolic connectome remain elusive. To this end, we examined a dataset comprising 49 PD patients and 49 healthy controls. By employing a personalized metabolic connectome approach, we assessed both within- and between-network connectivities using Standard Uptake Value (SUV) and Jensen-Shannon Divergence Similarity Estimation (JSSE). A random forest algorithm was utilized to pinpoint key neuroimaging features differentiating PD from healthy states. Specifically, the results revealed heightened internetwork connectivity in PD, specifically within the somatomotor (SMN) and frontoparietal (FPN) networks, persisting after multiple comparison corrections (P < 0.05, Bonferroni adjusted for 10% and 20% sparsity). This altered connectivity effectively distinguished PD patients from healthy individuals. Notably, this study utilizes 18F-FDG PET imaging to map individual metabolic networks, revealing enhanced connectivity in the SMN and FPN among PD patients. This enhanced connectivity may serve as a promising imaging biomarker, offering a valuable asset for early PD detection.
Collapse
Affiliation(s)
- Bei Chen
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No. 172, Tongzipo Road, Changsha City, Hunan Province, Changsha 410008, China
| | - Xiran Chen
- College of Mathematics and Statistics, Chongqing Jiaotong University, Xuefu Road No. 66, Chongqing 400074, China
| | - Liling Peng
- Department of PET/MR, Shanghai Universal Medical Imaging Diagnostic Center, Guilin Road No. 406, Shanghai 200233 China
- Hubei Province Key Laboratory of Molecular Imaging, Jiefang Road No. 1277, Wuhan 430022 China
| | - Shiqi Liu
- College of Mathematics and Statistics, Chongqing Jiaotong University, Xuefu Road No. 66, Chongqing 400074, China
| | - Yongxiang Tang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No. 172, Tongzipo Road, Changsha City, Hunan Province, Changsha 410008, China
| | - Xin Gao
- Department of PET/MR, Shanghai Universal Medical Imaging Diagnostic Center, Guilin Road No. 406, Shanghai 200233 China
| |
Collapse
|
5
|
Chen F, Chen L, Cai G, Wang Y, Li Y, Xu H, Song W, Jian J, Chen X, Ye Q. Association of synuclein alpha ( SNCA) gene polymorphisms with spontaneous brain activity in patients with Parkinson's disease. Quant Imaging Med Surg 2024; 14:6806-6819. [PMID: 39281177 PMCID: PMC11400684 DOI: 10.21037/qims-24-14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/10/2024] [Indexed: 09/18/2024]
Abstract
Background The synuclein alpha (SNCA) gene responsible for encoding alpha-synuclein, is believed to play a crucial role in the pathogenesis of Parkinson's disease (PD). However, the specific impact of SNCA gene single-nucleotide polymorphisms (SNPs) on brain function in PD remains unclear. Therefore, this cross-sectional retrospective study, particularly through use of imaging analysis, aimed to characterize the relationship between SNCA gene SNPs and spontaneous brain activity in PD in order to enhance our understanding of the mechanisms underlying PD pathogenesis. Methods A total of 63 patients with PD and 73 sex- and age-matched healthy control (HC) participants were recruited from outpatient and inpatient clinics at Fujian Medical University Union Hospital from August 2017 to November 2019, and all underwent a resting-state functional magnetic resonance imaging (rs-fMRI) scanning. All participants were also examined to determine the correlation of different genotypes with regional brain activity measured by rs-fMRI using amplitude of low-frequency fluctuation (ALFF) analysis. Multivariate regression analysis was used to calculate the correlation between the brain function data and clinical features. All rs-fMRI data were analyzed with the SPM12 software and adjusted according to the false discovery rate (FDR) at the cluster level. Results This study included 63 patients with PD and 73 sex- and age-matched healthy participants were included in the study. The spontaneous brain activity in the right superior cerebellum (Cerebelum_Crus1_R), vermis (Vermis_7), and left supplementary motor area (Supp_Motor_Area_L) of patients in the PD group was weak compared to that in the HC group. The z-score ALFF of left central posterior gyrus was positively correlated with the Mini-Mental State Examination score (r=0.542; P<0.001) in the PD group. For rs11931074, the main genotypic effects were found in the left inferior cerebellum (Cerebellum_9_L) and right anterior cingulate and paracingulate gyri (Cingulum_Ant_R); for rs356219 and rs356165, the main genotypic effects were found in the left caudate nucleus (Caudate_L). An interaction effect of disease with genotype was found in the right inferior parietal gyrus (Parietal_Inf_R) only for rs356219. Conclusions Our study found a correlation of the SNCA SNPs rs11931074, rs356219, and rs356165 with brain functional alterations in patients with PD. Furthermore, an interaction effect was found in the right inferior parietal gyrus only for rs356219. This study may contribute to furthering the understanding of the influence of SNCA gene SNPs on brain function in patients with PD.
Collapse
Affiliation(s)
- Fengxian Chen
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Neurology, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, China
| | - Lina Chen
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Guoen Cai
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yingqing Wang
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yunjing Li
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Haoling Xu
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Wenjing Song
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jing Jian
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaochun Chen
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
- Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Qinyong Ye
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
- Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
6
|
Tang J, Liu C, Liu C, Hu Q, Fang Y, Chen Z. Evaluation of damage discrimination in dopaminergic neurons using dopamine transporter PET tracer [ 18F]FECNT-d 4. EJNMMI Res 2024; 14:78. [PMID: 39210186 PMCID: PMC11362440 DOI: 10.1186/s13550-024-01140-3] [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: 04/13/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a prevalent neurodegenerative disorder worldwide, diagnosed based on classic symptoms like motor dysfunction and cognitive impairments. With the development of various radioactive ligands, positron emission tomography (PET) imaging combined with specific radiolabelling probes has proven to be effective in aiding clinical PD diagnosis. Among these probes, 2β-Carbomethoxy-3β-(4-chlorophenyl)-8-(2-[18F]-fluoroethyl) nortropane ([18F]FECNT) has been utilized as a PET tracer to image dopamine transporter (DAT) integrity in striatal presynaptic dopaminergic terminals. However, the presence of brain-penetrant radioactive metabolites produced by [18F]FECNT may impact the accuracy of PET imaging. In previous research, we developed 2β-Carbomethoxy-3β-(4-chlorophenyl)-8-(2-[18F]-fluoroethyl-1,1,2,2-d4) nortropane ([18F]FECNT-d4), a deuterated derivative with enhanced stability in plasma and the striatum, along with a slower washout rate. In this study, we further investigated the potential of [18F]FECNT-d4 to detect dopaminergic neuron degeneration in Parkinson's disease. This involved PET imaging in unilaterally-lesioned PD model rats and in vitro autoradiography conducted on postmortem brain sections. RESULTS PET images revealed reduced specific uptake in the ipsilateral striatum of rats stereotactically injected with 6-hydroxydopamine hydrochloride (6-OHDA). Compared to the sham group, the ratio of standardized uptake value (SUV) in the ipsilateral to contralateral striatum decreased by 13%, 23%, and 63% in the mild, moderate, and severe lesioned groups, respectively. Dopaminergic denervation observed in PET imaging was further supported by behavioral assessments, immunostaining, and monoamine concentration tests. Moreover, the microPET results exhibited positive correlations with these measurements, except for the apomorphine-induced rotational behavior test, which showed a negative correlation. Additionally, [18F]FECNT-d4 uptake was approximately 40% lower in the postmortem striatal sections of a PD patient compared to a healthy subject. Furthermore, estimated human dosimetry (effective dose equivalent: 5.06 E-03 mSv/MBq), extrapolated from rat biodistribution data, remained below the current Food and Drug Administration limit for radiation exposure. CONCLUSION Our findings demonstrate that [18F]FECNT-d4 accurately estimates levels of dopaminergic neuron degeneration in the 6-OHDA-induced PD rat model and effectively distinguishes between PD patients and healthy individuals. This highly sensitive and safe PET probe holds promising potential for clinical application in the diagnosis and monitoring of Parkinson's disease.
Collapse
Affiliation(s)
- Jie Tang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, No. 20 Qianrong Road, Binhu District, Wuxi, 214063, China
| | - Congjin Liu
- Department of Nuclear Medicine, Huashan Hospital, Fudan University, No. 12 Wulumuqi Middle Road, Jing'an District, Shanghai, 200040, China
| | - Chunyi Liu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, No. 20 Qianrong Road, Binhu District, Wuxi, 214063, China
| | - Qianyue Hu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, No. 20 Qianrong Road, Binhu District, Wuxi, 214063, China
| | - Yi Fang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, No. 20 Qianrong Road, Binhu District, Wuxi, 214063, China
| | - Zhengping Chen
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, No. 20 Qianrong Road, Binhu District, Wuxi, 214063, China.
| |
Collapse
|
7
|
Vroom MM, Dodart JC. Active Immunotherapy for the Prevention of Alzheimer's and Parkinson's Disease. Vaccines (Basel) 2024; 12:973. [PMID: 39340005 PMCID: PMC11435640 DOI: 10.3390/vaccines12090973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Neurodegenerative diseases (ND) give rise to significant declines in motor, autonomic, behavioral, and cognitive functions. Of these conditions, Alzheimer's disease (AD) and Parkinson's disease (PD) are the most prevalent, impacting over 55 million people worldwide. Given the staggering financial toll on the global economy and their widespread manifestation, NDs represent a critical issue for healthcare systems worldwide. Current treatment options merely seek to provide symptomatic relief or slow the rate of functional decline and remain financially inaccessible to many patients. Indeed, no therapy has yet demonstrated the potential to halt the trajectory of NDs, let alone reverse them. It is now recognized that brain accumulation of pathological variants of AD- or PD-associated proteins (i.e., amyloid-β, Tau, α-synuclein) begins years to decades before the onset of clinical symptoms. Accordingly, there is an urgent need to pursue therapies that prevent the neurodegenerative processes associated with pathological protein aggregation long before a clinical diagnosis can be made. These therapies must be safe, convenient, and affordable to ensure broad coverage in at-risk populations. Based on the need to intervene long before clinical symptoms appear, in this review, we present a rationale for greater investment to support the development of active immunotherapy for the prevention of the two most common NDs based on their safety profile, ability to specifically target pathological proteins, as well as the significantly lower costs associated with manufacturing and distribution, which stands to expand accessibility to millions of people globally.
Collapse
Affiliation(s)
- Madeline M Vroom
- Vaxxinity, Inc., Space Life Sciences Lab, 505 Odyssey Way, Merritt Island, FL 32953, USA
| | - Jean-Cosme Dodart
- Vaxxinity, Inc., Space Life Sciences Lab, 505 Odyssey Way, Merritt Island, FL 32953, USA
| |
Collapse
|
8
|
Duță C, Muscurel C, Dogaru CB, Stoian I. Ferroptosis-A Shared Mechanism for Parkinson's Disease and Type 2 Diabetes. Int J Mol Sci 2024; 25:8838. [PMID: 39201524 PMCID: PMC11354749 DOI: 10.3390/ijms25168838] [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: 06/30/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Type 2 diabetes (T2D) and Parkinson's disease (PD) are the two most frequent age-related chronic diseases. There are many similarities between the two diseases: both are chronic diseases; both are the result of a decrease in a specific substance-insulin in T2D and dopamine in PD; and both are caused by the destruction of specific cells-beta pancreatic cells in T2D and dopaminergic neurons in PD. Recent epidemiological and experimental studies have found that there are common underlying mechanisms in the pathophysiology of T2D and PD: chronic inflammation, mitochondrial dysfunction, impaired protein handling and ferroptosis. Epidemiological research has indicated that there is a higher risk of PD in individuals with T2D. Moreover, clinical studies have observed that the symptoms of Parkinson's disease worsen significantly after the onset of T2D. This article provides an up-to-date review on the intricate interplay between oxidative stress, reactive oxygen species (ROS) and ferroptosis in PD and T2D. By understanding the shared molecular pathways and how they can be modulated, we can develop more effective therapies, or we can repurpose existing drugs to improve patient outcomes in both disorders.
Collapse
|
9
|
Ghosh N, Sinha K, Sil PC. Pesticides and the Gut Microbiota: Implications for Parkinson's Disease. Chem Res Toxicol 2024; 37:1071-1085. [PMID: 38958636 DOI: 10.1021/acs.chemrestox.4c00057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Parkinson's disease (PD) affects more people worldwide than just aging alone can explain. This is likely due to environmental influences, genetic makeup, and changes in daily habits. The disease develops in a complex way, with movement problems caused by Lewy bodies and the loss of dopamine-producing neurons. Some research suggests Lewy bodies might start in the gut, hinting at a connection between these structures and gut health in PD patients. These patients often have different gut bacteria and metabolites. Pesticides are known to increase the risk of PD, with evidence showing they harm more than just dopamine neurons. Long-term exposure to pesticides in food might affect the gut barrier, gut bacteria, and the blood-brain barrier, but the exact link is still unknown. This review looks at how pesticides and gut bacteria separately influence PD development and progression, highlighting the harmful effects of pesticides and changes in gut bacteria. We have examined the interaction between pesticides and gut bacteria in PD patients, summarizing how pesticides cause imbalances in gut bacteria, the resulting changes, and their overall effects on the PD prognosis.
Collapse
Affiliation(s)
- Nabanita Ghosh
- Assistant Professor in Zoology, Maulana Azad College, Kolkata 700013, India
| | - Krishnendu Sinha
- Assistant Professor in Zoology, Jhargram Raj College, Jhargram 721507 India
| | - Parames C Sil
- Professor, Division of Molecular Medicine, Bose Institute, Kolkata 700054 India
| |
Collapse
|
10
|
Ghosh N, Sinha K, Sil PC. A review on the new age methodologies for early detection of Alzheimer's and Parkinson's disease. Basic Clin Pharmacol Toxicol 2024; 134:602-613. [PMID: 38482977 DOI: 10.1111/bcpt.14003] [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: 10/10/2023] [Revised: 02/18/2024] [Accepted: 02/26/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUNDS Neurodegenerative diseases (NDDs) such as Alzheimer's (AD) and Parkinson's (PD) are often diagnosed late, impeding effective treatment; therefore, early detection is imperative. Modern methodologies can serve a pivotal role in fulfilling the crucial need for timely detection and intervention in this context. OBJECTIVES Evaluate early detection's significance and summarize key technologies (biomarkers, neuroimaging, AI/ML, genetics, digital health) for enhanced diagnostic strategies in AD and PD. METHODS This study employs a focused descriptive review approach, encompassing analysis of peer-reviewed articles and clinical trials from existing literature, to provide a nuanced exploration of the subject matter. FINDINGS This review underscores the efficacy of non-invasive biomarkers, biosensors and emerging promising technologies for advancing early diagnosis of AD and PD. CONCLUSION The landscape of early NDD detection has been reshaped by technology, yet challenges persist, encompassing the domains of validation and ethics. A collaborative effort between medical professionals, researchers and technologists is imperative to effectively address and combat NDDs.
Collapse
Affiliation(s)
| | | | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| |
Collapse
|
11
|
Tassan Mazzocco M, Serra M, Maspero M, Coliva A, Presotto L, Casu MA, Morelli M, Moresco RM, Belloli S, Pinna A. Positive relation between dopamine neuron degeneration and metabolic connectivity disruption in the MPTP plus probenecid mouse model of Parkinson's disease. Exp Neurol 2024; 374:114704. [PMID: 38281587 DOI: 10.1016/j.expneurol.2024.114704] [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: 10/11/2023] [Revised: 01/15/2024] [Accepted: 01/25/2024] [Indexed: 01/30/2024]
Abstract
The clinical manifestation of Parkinson's disease (PD) appears when neurodegeneration is already advanced, compromising the efficacy of disease-modifying treatment approaches. Biomarkers to identify the early stages of PD are therefore of paramount importance for the advancement of the therapy of PD. In the present study, by using a mouse model of PD obtained by subchronic treatment with the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and the clearance inhibitor probenecid (MPTPp), we identified prodromal markers of PD by combining in vivo positron emission tomography (PET) imaging and ex vivo immunohistochemistry. Longitudinal PET imaging of the dopamine transporter (DAT) by [18F]-N-(3-fluoropropyl)-2β-carboxymethoxy-3β-(4-iodophenyl) nortropane ([18F]-FP-CIT), and brain glucose metabolism by 2-deoxy-2-[18F]-fluoroglucose ([18F]-FDG) were performed before MPTPp treatment and after 1, 3, and 10 MPTPp administrations, in order to assess relation between dopamine neuron integrity and brain connectivity. The results show that in vivo [18F]-FP-CIT in the dorsal striatum was not modified after the first administration of MPTPp, tended to decrease after 3 administrations, and significantly decreased after 10 MPTPp administrations. Post-mortem immunohistochemical analyses of DAT and tyrosine hydroxylase (TH) in the striatum showed a positive correlation with [18F]-FP-CIT, confirming the validity of repeated MPTPp-treated mice as a model that can reproduce the progressive pathological changes in the early phases of PD. Analysis of [18F]-FDG uptake in several brain areas connected to the striatum showed that metabolic connectivity was progressively disrupted, starting from the first MPTPp administration, and that significant connections between cortical and subcortical regions were lost after 10 MPTPp administrations, suggesting an association between dopamine neuron degeneration and connectivity disruption in this PD model. The results of this study provide a relevant model, where new drugs that can alleviate neurodegeneration in PD could be evaluated preclinically.
Collapse
Affiliation(s)
- Margherita Tassan Mazzocco
- PhD Program in Neuroscience, Medicine and Surgery Department, University of Milano-Bicocca, Monza, Italy; Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Marcello Serra
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Marco Maspero
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy; National Research Council of Italy, Institute of Molecular Bioimaging and Physiology, UOS of Segrate, Italy
| | - Angela Coliva
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Luca Presotto
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy; Department of Physics "G. Occhialini", University of Milano - Bicocca, Milan, Italy
| | - Maria Antonietta Casu
- National Research Council of Italy, Institute of Translational Pharmacology, UOS of Cagliari, Scientific and Technological Park of Sardinia POLARIS, Pula, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy; National Research Council of Italy, Neuroscience Institute, UOS of Cagliari, Italy
| | - Rosa Maria Moresco
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy; National Research Council of Italy, Institute of Molecular Bioimaging and Physiology, UOS of Segrate, Italy; School of Medicine and Surgery, University of Milano - Bicocca, Monza, Italy.
| | - Sara Belloli
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy; National Research Council of Italy, Institute of Molecular Bioimaging and Physiology, UOS of Segrate, Italy
| | - Annalisa Pinna
- National Research Council of Italy, Neuroscience Institute, UOS of Cagliari, Italy
| |
Collapse
|
12
|
Ryu DW, Yoo SW, Choi KE, Oh YS, Kim JS. Correlation of olfactory function factors with cardiac sympathetic denervation in Parkinson's disease. J Neurol 2024; 271:1397-1407. [PMID: 37940708 DOI: 10.1007/s00415-023-12080-8] [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: 08/10/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Hyposmia is a common nonmotor symptom of Parkinson's disease (PD) and reportedly associated with dysautonomia in PD. The smell identification test for measuring olfactory function consists of multiple items to discriminate specific scents. In the present study, factor analysis of the smell identification test was performed, and the correlation of extracted factors with cardiac sympathetic denervation (CSD) in patients with PD was investigated. METHODS The present study included 183 early PD patients who underwent the Cross-Cultural Smell Identification Test (CC-SIT) and 123I-meta-iodobenzylguanidine (123I-MIBG) myocardial scintigraphy. Factor analysis of 12 items on the CC-SIT was performed using the computed correlation matrix for the binary items, and five smell factors were extracted. Multiple linear regression was performed to determine the correlation of olfactory function with late heart-to-mediastinum (H/M) ratio of 123I-MIBG uptake. RESULTS The mean CC-SIT score was 6.1 ± 2.6, and 133 patients (72.7%) had CSD. The CC-SIT score and five smell factors were not associated with dopamine transporter uptake or cognitive functions. However, the CC-SIT score significantly correlated with age (P < 0.001) and late H/M ratio (P < 0.001). Factors 1 and 5 showed an increasing trend with larger H/M ratio, although it was not statistically significant (β = 0.203, P = 0.085 and β = 0.230, P = 0.085, respectively). Factor 5 significantly correlated with the H/M ratio in PD patients with CSD (β = 0.676, P = 0.036). DISCUSSION The results showed olfactory dysfunction to be selectively associated with cardiac sympathetic burden in PD. The correlation of certain factors with CSD indicates the possibility of selective hyposmia in PD patients.
Collapse
Affiliation(s)
- Dong-Woo Ryu
- Department of Neurology, College of Medicine, The Catholic University of Korea, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Sang-Won Yoo
- Department of Neurology, College of Medicine, The Catholic University of Korea, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Ko-Eun Choi
- Department of Neurology, College of Medicine, The Catholic University of Korea, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Yoon-Sang Oh
- Department of Neurology, College of Medicine, The Catholic University of Korea, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Joong-Seok Kim
- Department of Neurology, College of Medicine, The Catholic University of Korea, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| |
Collapse
|
13
|
Wang Y, Xiao Y, Xing Y, Yu M, Wang X, Ren J, Liu W, Zhong Y. Morphometric similarity differences in drug-naive Parkinson's disease correlate with transcriptomic signatures. CNS Neurosci Ther 2024; 30:e14680. [PMID: 38529533 PMCID: PMC10964038 DOI: 10.1111/cns.14680] [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: 10/30/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Differences in cortical morphology have been reported in individuals with Parkinson's disease (PD). However, the pathophysiological mechanism of transcriptomic vulnerability in local brain regions remains unclear. OBJECTIVE This study aimed to characterize the morphometric changes of brain regions in early drug-naive PD patients and uncover the brain-wide gene expression correlates. METHODS The morphometric similarity (MS) network analysis was used to quantify the interregional structural similarity from multiple magnetic resonance imaging anatomical indices measured in each brain region of 170 early drug-naive PD patients and 123 controls. Then, we applied partial least squares regression to determine the relationship between regional changes in MS and spatial transcriptional signatures from the Allen Human Brain Atlas dataset, and identified the specific genes related to MS differences in PD. We further investigated the biological processes by which the PD-related genes were enriched and the cellular characterization of these genes. RESULTS Our results showed that MS was mainly decreased in cingulate, frontal, and temporal cortical areas and increased in parietal and occipital cortical areas in early drug-naive PD patients. In addition, genes whose expression patterns were associated with regional MS changes in PD were involved in astrocytes, excitatory, and inhibitory neurons and were functionally enriched in neuron-specific biological processes related to trans-synaptic signaling and nervous system development. CONCLUSIONS These findings advance our understanding of the microscale genetic and cellular mechanisms driving macroscale morphological abnormalities in early drug-naive PD patients and provide potential targets for future therapeutic trials.
Collapse
Affiliation(s)
- Yajie Wang
- Department of NeurologyThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina
- Department of NeurologyThe First People's Hospital of YanchengYanchengChina
| | - Yiwen Xiao
- School of PsychologyNanjing Normal UniversityNanjingChina
| | - Yi Xing
- Department of NeurologyThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina
| | - Miao Yu
- Department of NeurologyThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina
| | - Xiao Wang
- Department of RadiologyThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina
| | - Jingru Ren
- Department of NeurologyThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina
| | - Weiguo Liu
- Department of NeurologyThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina
| | - Yuan Zhong
- School of PsychologyNanjing Normal UniversityNanjingChina
| |
Collapse
|
14
|
Das S, Ramteke H. A Comprehensive Review of the Role of Biomarkers in Early Diagnosis of Parkinson's Disease. Cureus 2024; 16:e54337. [PMID: 38500934 PMCID: PMC10945043 DOI: 10.7759/cureus.54337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 02/16/2024] [Indexed: 03/20/2024] Open
Abstract
Parkinson's disease (PD) is a complex neurological, degenerative clinical condition depicted by the advancing loss of dopaminergic neurons in the substantia nigra pars compacta, which manifests itself as a myriad of sensorimotor and non-motor signs in patients. The disease occurs due to the reduced levels of the neurotransmitter dopamine in the brain, which is primarily associated with functional characteristics regarding mobility and cognition. The basal ganglion is mainly involved in the generation of cognitive functions and therefore is the most significantly associated area in PD. Since the classical diagnosis and assessment of PD depends majorly on the appearance of motor characteristics, which only arise when ~60-80% of the dopamine neuronal cell death has already occurred, it is imperative we focus on identifying biomarkers that can help us assess and diagnose PD in the earlier stages of disease progression, thus providing a better prognosis for the patients. This review article will focus on the different biomarkers that are currently available and in use, divided under the headings of clinical, biological, imaging, and genetic biomarkers, and assess their specificity and sensitivity toward providing an early assessment of Parkinson's for the patients and the future of preclinical diagnostics using molecular biomarkers. PD affects over 1% of the population worldwide and only ranks second to Alzheimer's disease in the context of its incidence and consequent socioeconomic burden. While recent breakthroughs in biomarkers have dramatically improved patients' odds of survival and prognosis, it still remains primarily a symptomatic diagnostic tool. It is an area of research that requires to focus on creating more advanced approaches toward diagnosing PD early, involving clinical diagnostics, neuroimaging technology, and molecular biology collaborations to provide the highest degree of care and quality of life that a Parkinson's patient deserves.
Collapse
Affiliation(s)
- Somdutta Das
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Harshal Ramteke
- General Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
15
|
Liu Y, Lin W, Liu J, Zhu H. Structural and temporal dynamics analysis of neural circuit from 2002 to 2022: A bibliometric analysis. Heliyon 2024; 10:e24649. [PMID: 38298625 PMCID: PMC10828061 DOI: 10.1016/j.heliyon.2024.e24649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 01/05/2024] [Accepted: 01/11/2024] [Indexed: 02/02/2024] Open
Abstract
Background In the pursuit of causal insights into neural circuit functionality, various interventions, including electrical, genetic, and pharmacological approaches, have been applied over recent decades. This study employs a comprehensive bibliometric perspective to explore the field of neural circuits. Methods Reviews and articles on neural circuits were obtained from the Web of Science Core Collection (WOSCC) database on Apr. 12, 2023. In this article, co-authorship analysis, co-occurrence analysis, citation analysis, bibliographic analysis, and co-citation analysis were used to clarify the authors, journals, institutions, countries, topics, and internal associations between them. Results More than 2000 organizations from 52 different countries published 3975 articles in the field of "neural circuit" were used to analysis. Luo liqun emerged as the most prolific author, and Deisseroth Karl garners the highest co-citations (3643). The Journal of Neuroscience leaded in publications, while Nature toped in citations. Chinese Academy of Science recorded the highest article count institutionally, with Stanford University ranking first with 14,350 citations. Since 2020, neurodynamic, anxiety-related mechanisms, and GABAergic neurons have gained prominence, shaping the trajectory of neural circuitry research. Conclusions Our investigation has discerned a paradigmatic reorientation towards neurodynamic processes, anxiety-related mechanisms, and GABAergic neurons within the domain of neural circuit research. This identification intimates a prospective trajectory for the field. In the future, it is imperative for research endeavors to accord priority to the translational application of these discernments, with the aim of materializing tangible clinical solutions.
Collapse
Affiliation(s)
- Yuan Liu
- Cancer Research Center Nantong, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Wei Lin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
- Department of Pediatrics, The First Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Jie Liu
- Department of Orthopedics, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Haixia Zhu
- Cancer Research Center Nantong, Affiliated Tumor Hospital of Nantong University, Nantong, China
| |
Collapse
|
16
|
Shi Q, Kang W, Liu Z, Zhu X. The role of exosomes in the diagnosis of Parkinson's disease. Heliyon 2023; 9:e20595. [PMID: 37928387 PMCID: PMC10622621 DOI: 10.1016/j.heliyon.2023.e20595] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 07/22/2023] [Accepted: 09/29/2023] [Indexed: 11/07/2023] Open
Abstract
Parkinson's disease is a common neurodegenerative disease characterized by intracellular aggregation of misfolded α-synuclein as a major pathological hallmark. Exosomes are cell-derived lipid bilayer membrane vesicles with various components, including proteins, RNA, and lipids, that mediate intercellular communication. Currently, exosomes are found to be responsible for transporting misfolded proteins from unhealthy neurons to nearby cells, spreading the disease from cell to cell. Such exosomes can also be found in the cerebrospinal fluid and blood. Thus, exosomes may serve as a potential tool to detect the pathology of Parkinson's disease for clinical diagnosis. In this article, the role and challenges of exosomes in the diagnosis of Parkinson's disease are outlined.
Collapse
Affiliation(s)
- Qingqing Shi
- Tianjin Medical University, General Hospital, 300000, Tianjin, China
| | - Wei Kang
- Beijing Conga Technology Co., LTD., Tianjin Branch, 300000, Tianjin, China
| | - Zhijun Liu
- Beijing Conga Technology Co., LTD., Tianjin Branch, 300000, Tianjin, China
| | - Xiaodong Zhu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University, General Hospital, 300000, Tianjin, China
| |
Collapse
|
17
|
Carli G, Meles SK, Janzen A, Sittig E, Kogan RV, Perani D, Oertel WH, Leenders KL. Occipital hypometabolism is a risk factor for conversion to Parkinson's disease in isolated REM sleep behaviour disorder. Eur J Nucl Med Mol Imaging 2023; 50:3290-3301. [PMID: 37310428 PMCID: PMC10542098 DOI: 10.1007/s00259-023-06289-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 05/30/2023] [Indexed: 06/14/2023]
Abstract
PURPOSE Isolated REM sleep behaviour disorder (iRBD) patients are at high risk of developing clinical syndromes of the α-synuclein spectrum. Progression markers are needed to determine the neurodegenerative changes and to predict their conversion. Brain imaging with 18F-FDG PET in iRBD is promising, but longitudinal studies are scarce. We investigated the regional brain changes in iRBD over time, related to phenoconversion. METHODS Twenty iRBD patients underwent two consecutive 18F-FDG PET brain scans and clinical assessments (3.7 ± 0.6 years apart). Seventeen patients also underwent 123I-MIBG and 123I-FP-CIT SPECT scans at baseline. Four subjects phenoconverted to Parkinson's disease (PD) during follow-up. 18F-FDG PET scans were compared to controls with a voxel-wise single-subject procedure. The relationship between regional brain changes in metabolism and PD-related pattern scores (PDRP) was investigated. RESULTS Individual hypometabolism t-maps revealed three scenarios: (1) normal 18F-FDG PET scans at baseline and follow-up (N = 10); (2) normal scans at baseline but occipital or occipito-parietal hypometabolism at follow-up (N = 4); (3) occipital hypometabolism at baseline and follow-up (N = 6). All patients in the last group had pathological 123I-MIBG and 123I-FP-CIT SPECT. iRBD converters (N = 4) showed occipital hypometabolism at baseline (third scenario). At the group level, hypometabolism in the frontal and occipito-parietal regions and hypermetabolism in the cerebellum and limbic regions were progressive over time. PDRP z-scores increased over time (0.54 ± 0.36 per year). PDRP expression was driven by occipital hypometabolism and cerebellar hypermetabolism. CONCLUSIONS Our results suggest that occipital hypometabolism at baseline in iRBD implies a short-term conversion to PD. This might help in stratification strategies for disease-modifying trials.
Collapse
Affiliation(s)
- Giulia Carli
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Sanne K Meles
- Department of Neurology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Annette Janzen
- Department of Neurology, Philipps-Universität Marburg, Marburg, Germany
| | - Elisabeth Sittig
- Department of Neurology, Philipps-Universität Marburg, Marburg, Germany
| | - Rosalie V Kogan
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Internal Medicine, Sierra View Medical Center, Porterville, CA, USA
| | - Daniela Perani
- School of Psychology, Vita-Salute San Raffaele University, Milan, Italy
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Wolfgang H Oertel
- Department of Neurology, Philipps-Universität Marburg, Marburg, Germany
- Institute for Neurogenomics, Helmholtz Center for Health and Environment, Munich, Germany
| | - Klaus L Leenders
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Neurology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
18
|
Dong Z, Zuo R, Zhong X, Zhang C, Zou X, Tian Y, Zuo H, Du X, Yu Q, Cheng O. Antihypertensive drugs may not delay the symptom progression of Parkinson's disease: A 2-year follow-up study. Heliyon 2023; 9:e18538. [PMID: 37560660 PMCID: PMC10407028 DOI: 10.1016/j.heliyon.2023.e18538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is one of the most common neurodegenerative disease, and half of PD patients have hypertension as well. The effect of antihypertensive drugs on the progression of PD has been less studied. The focus of this study was on the changes in dopamine transporter (DAT) levels to assess the effect of antihypertensive drugs on the progression of PD. METHODS Data from 321 drug-naïve patients from the Parkinson's Disease Progression Marker Initiative (PPMI) were collected over a 2-year period. Patients were divided into the PD with arterial hypertension (AH) group (102 cases) with antihypertensive drugs, the PD with other cardiovascular risk factors (CVRFs) group (60 cases) with antidiabetic and/or lipid-lowering drugs, and the pure PD group (159 cases) without CVRFs. The Movement Disorder Society Sponsored Revision Unified Parkinson's Disease Rating Scale (MDS-UPDRS) and Hoehn-Yahr (H&Y) stage were used to assess progression. DAT semiquantitative values were used to evaluate damage to dopaminergic neurons in the substantia nigra, including the contralateral and ipsilateral count density ratio and asymmetry index. RESULTS There were no significant differences among the three groups in MDS-UPDRS score and H&Y stage. Changes in DAT levels among the three groups were without distinct differences in the first year and second year. In each group, DAT decreased more in the first year than in the second year. There was no decrease in DAT uptake in the PD with AH group compared with the other groups during the follow-up period. CONCLUSIONS There is no evidence that antihypertensive drugs can delay PD progression within 2 years.
Collapse
Affiliation(s)
- Zhaoying Dong
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 400016, Chongqing, China
| | - Rui Zuo
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, 400016, Chongqing, China
| | - Xiaoni Zhong
- School of Public Health and Management, Chongqing Medical University, 400016, Chongqing, China
| | - Changhong Zhang
- Chengdu Center For Disease Control and Prevention, 610041, Si Chuan, China
| | - Xiaoya Zou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 400016, Chongqing, China
| | - Yuan Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 400016, Chongqing, China
| | - Hongzhou Zuo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 400016, Chongqing, China
| | - Xinyi Du
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 400016, Chongqing, China
| | - Qian Yu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 400016, Chongqing, China
| | - Oumei Cheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 400016, Chongqing, China
| |
Collapse
|
19
|
Xin M, Li L, Wang C, Shao H, Liu J, Zhang C. Pilot study on 11C-CFT dynamic imaging using total-body PET/CT: biodistribution and radiation dosimetry in Parkinson's disease. Front Neurol 2023; 14:1153779. [PMID: 37260609 PMCID: PMC10227570 DOI: 10.3389/fneur.2023.1153779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/18/2023] [Indexed: 06/02/2023] Open
Abstract
Objective Total-body PET/CT equipment, uEXPLORER, is a newly developed imaging technology with a superior resolution, high sensitivity, and high signal-to-noise ratio, providing unique application advantages in the pharmacokinetic evaluation of positron tracers. While 11C-CFT PET/CT has been widely utilized in the early diagnosis of Parkinson's disease (PD), it is limited by the short half-life of the radionuclide and an incomplete understanding of its biological distribution in humans. This study aimed to use a total-body PET/CT dynamic scan with 11C-CFT imaging to describe the real-time internal biodistribution in PD patients and to obtain accurate radiation dosimetry. Methods Six male subjects with suspected PD underwent dynamic 11C-CFT total-body PET/CT. Following a bedside intravenous bolus injection of 373.3 ± 71.56 MBq of 11C-CFT, PET acquisition was performed synchronously for 75 min with a maximum axial field of view (AFOV) of 194 cm. Time-activity curves (TACs) were generated by delineating volumes of interest (VOIs) of the sourced organs using PMOD software. Tracer kinetics and cumulative organ activities were calculated, and absorbed doses were calculated and estimated using the OLINDA/EXM software. Results In the systemic TAC analysis of 11C-CFT, several unique types of distribution patterns were obtained among several major organs, including a "Fast-in Fast-out" pattern in the kidneys, lungs, spleen, and thyroid, a "Fast-in Slow-out" curve in the heart wall, a "Slow-in Slow-out" mode in the liver, a "Low-level extending" pattern in the whole brain and muscle, and a "Slow-in to plateau" trend in the striatum and bone. The effective dose of 11C-CFT was calculated to be 2.83E-03 mSv/MBq, which is only one-third of the literature value measured by the conventional method. Moreover, this dose is much lower compared to all other doses of DAT radioligands used in PET imaging. Conclusion This study is a pioneering application of total-body PET/CT to 11C-CFT dynamic imaging. Our results confirmed that 11C-CFT has a favorable total body biodistribution, an extremely low internal radiation dose, and high imaging quality, making it suitable for reasonable PD diagnosis in patients requiring multiple follow-up examinations.
Collapse
|
20
|
Kong L, He Q, Li Q, Schreiber R, Kaitin KI, Shao L. Rapid progress in neuroimaging technologies fuels central nervous system translational medicine. Drug Discov Today 2023; 28:103485. [PMID: 36623797 DOI: 10.1016/j.drudis.2023.103485] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/12/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023]
Abstract
Central nervous system (CNS) drug discovery suffers from high attrition rates; translational neuroscience approaches aiming to reduce these high rates include the use of brain imaging technologies. However, there is a need to better understand what methods are being used and for what diseases and purposes. Our analysis of the literature found that magnetic resonance imaging (MRI), positron emission tomography (PET), and single-photon emission computed tomography (SPECT) were the neuroimaging techniques used most often in clinical trials for the most prevalent CNS diseases: Alzheimer's disease (AD), Parkinson's disease (PD), depression, and schizophrenia. Moreover, the number of initiated clinical trials using MRI, PET, and SPECT increased over the period 1981-2021. Such insights indicate that the significant increase in the use of neuroimaging studies could decrease the attrition of novel drug candidates in late clinical development.
Collapse
Affiliation(s)
- Linghui Kong
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Zhangjiang Hi-tech Park, Pudong, Shanghai 201203, China
| | - Qian He
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Zhangjiang Hi-tech Park, Pudong, Shanghai 201203, China
| | - Qiu Li
- Shanghai Center for iDrug Discovery & Development, 826 Zhangheng Road, Zhangjiang Hi-tech Park, Pudong, Shanghai 201203, China
| | - Rudy Schreiber
- Faculty of Psychology and Neuroscience, Section Neuropsychology & Psychopharmacology, Universiteitssingel 40, Maastricht University, PO Box 616, 6229 ER Maastricht, the Netherlands
| | - Kenneth I Kaitin
- Tufts Center for the Study of Drug Development (CSDD), Tufts University School of Medicine, Boston, MA, USA
| | - Liming Shao
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Zhangjiang Hi-tech Park, Pudong, Shanghai 201203, China; Shanghai Center for iDrug Discovery & Development, 826 Zhangheng Road, Zhangjiang Hi-tech Park, Pudong, Shanghai 201203, China; State Key Laboratory of Medical Neurobiology, Fudan University, No. 138 Yixueyuan Road, Shanghai 200032, China.
| |
Collapse
|
21
|
Current Treatments and New, Tentative Therapies for Parkinson’s Disease. Pharmaceutics 2023; 15:pharmaceutics15030770. [PMID: 36986631 PMCID: PMC10051786 DOI: 10.3390/pharmaceutics15030770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/02/2023] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative pathology, the origin of which is associated with the death of neuronal cells involved in the production of dopamine. The prevalence of PD has increased exponentially. The aim of this review was to describe the novel treatments for PD that are currently under investigation and study and the possible therapeutic targets. The pathophysiology of this disease is based on the formation of alpha-synuclein folds that generate Lewy bodies, which are cytotoxic and reduce dopamine levels. Most pharmacological treatments for PD target alpha-synuclein to reduce the symptoms. These include treatments aimed at reducing the accumulation of alpha-synuclein (epigallocatechin), reducing its clearance via immunotherapy, inhibiting LRRK2, and upregulating cerebrosidase (ambroxol). Parkinson’s disease continues to be a pathology of unknown origin that generates a significant social cost for the patients who suffer from it. Although there is still no definitive cure for this disease at present, there are numerous treatments available aimed at reducing the symptomatology of PD in addition to other therapeutic alternatives that are still under investigation. However, the therapeutic approach to this pathology should include a combination of pharmacological and non-pharmacological strategies to maximise outcomes and improve symptomatological control in these patients. It is therefore necessary to delve deeper into the pathophysiology of the disease in order to improve these treatments and therefore the quality of life of the patients.
Collapse
|
22
|
Perovnik M, Rus T, Schindlbeck KA, Eidelberg D. Functional brain networks in the evaluation of patients with neurodegenerative disorders. Nat Rev Neurol 2023; 19:73-90. [PMID: 36539533 DOI: 10.1038/s41582-022-00753-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2022] [Indexed: 12/24/2022]
Abstract
Network analytical tools are increasingly being applied to brain imaging maps of resting metabolic activity (PET) or blood oxygenation-dependent signals (functional MRI) to characterize the abnormal neural circuitry that underlies brain diseases. This approach is particularly valuable for the study of neurodegenerative disorders, which are characterized by stereotyped spread of pathology along discrete neural pathways. Identification and validation of disease-specific brain networks facilitate the quantitative assessment of pathway changes over time and during the course of treatment. Network abnormalities can often be identified before symptom onset and can be used to track disease progression even in the preclinical period. Likewise, network activity can be modulated by treatment and might therefore be used as a marker of efficacy in clinical trials. Finally, early differential diagnosis can be achieved by simultaneously measuring the activity levels of multiple disease networks in an individual patient's scans. Although these techniques were originally developed for PET, over the past several years analogous methods have been introduced for functional MRI, a more accessible non-invasive imaging modality. This advance is expected to broaden the application of network tools to large and diverse patient populations.
Collapse
Affiliation(s)
- Matej Perovnik
- Department of Neurology, University Medical Center Ljubljana, Ljubljana, Slovenia.,Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Tomaž Rus
- Department of Neurology, University Medical Center Ljubljana, Ljubljana, Slovenia.,Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | | | - David Eidelberg
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY, USA.
| |
Collapse
|
23
|
Searching for Biomarkers in the Blood of Patients at Risk of Developing Parkinson's Disease at the Prodromal Stage. Int J Mol Sci 2023; 24:ijms24031842. [PMID: 36768161 PMCID: PMC9915927 DOI: 10.3390/ijms24031842] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/02/2023] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
Parkinson's disease (PD) is diagnosed many years after its onset, under a significant degradation of the nigrostriatal dopaminergic system, responsible for the regulation of motor function. This explains the low effectiveness of the treatment of patients. Therefore, one of the highest priorities in neurology is the development of the early (preclinical) diagnosis of PD. The aim of this study was to search for changes in the blood of patients at risk of developing PD, which are considered potential diagnostic biomarkers. Out of 1835 patients, 26 patients were included in the risk group and 20 patients in the control group. The primary criteria for inclusion in a risk group were the impairment of sleep behavior disorder and sense of smell, and the secondary criteria were neurological and mental disorders. In patients at risk and in controls, the composition of plasma and the expression of genes of interest in lymphocytes were assessed by 27 indicators. The main changes that we found in plasma include a decrease in the concentrations of l-3,4-dihydroxyphenylalanine (L-DOPA) and urates, as well as the expressions of some types of microRNA, and an increase in the total oxidative status. In turn, in the lymphocytes of patients at risk, an increase in the expression of the DA D3 receptor gene and the lymphocyte activation gene 3 (LAG3), as well as a decrease in the expression of the Protein deglycase DJ-1 gene (PARK7), were observed. The blood changes we found in patients at risk are considered candidates for diagnostic biomarkers at the prodromal stage of PD.
Collapse
|
24
|
Zheng JH, Sun WH, Ma JJ, Wang ZD, Chang QQ, Dong LR, Shi XX, Li MJ, Gu Q, Chen SY. Structural and functional abnormalities in Parkinson's disease based on voxel-based morphometry and resting-state functional magnetic resonance imaging. Neurosci Lett 2022; 788:136835. [PMID: 35963477 DOI: 10.1016/j.neulet.2022.136835] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 07/25/2022] [Accepted: 08/07/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To explore differences in gray matter volume (GMV) and white matter volume (WMV) between patients with Parkinson's disease (PD) and healthy controls, and to examine whether the structural abnormalities correlate with functional abnormalities. METHODS T1-weighted magnetic resonance imaging and resting-state functional magnetic resonance imaging (fMRI) were performed on 180 patients with PD and 58 age- and sex-matched healthy controls. We used voxel-based morphometry (VBM) to compare GMV and WMV between groups, and resting-state fMRI to compare amplitudes of low-frequency fluctuations (ALFF) in the structurally abnormal brain regions. RESULTS Structural neuroimaging showed smaller whole-brain GMV, but not WMV, in patients. Furthermore, VBM revealed smaller GMV in the right superior temporal gyrus (STG) and left frontotemporal space in patients, after correction for multiple comparisons. Patients also showed significantly higher ALFF in the right STG. GMV in the right STG and left frontotemporal space in patients correlated negatively with age and scores on Part III of the Movement Disorder Society Unified Parkinson's Disease Rating Scale, but not with PD duration. CONCLUSIONS Structural atrophy in the frontotemporal lobe may be a useful imaging biomarker in PD, such as for detecting disease progression. Furthermore, this structural atrophy appears to correlate with enhanced spontaneous brain activity. This study associates particular structural and functional abnormalities with PD neuropathology.
Collapse
Affiliation(s)
- Jin Hua Zheng
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, Henan Province, China; Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China; Department of Neurology, People's Hospital of Henan University, Zhengzhou, Henan Province, China
| | - Wen Hua Sun
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, Henan Province, China; Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jian Jun Ma
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, Henan Province, China; Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China; Department of Neurology, People's Hospital of Henan University, Zhengzhou, Henan Province, China.
| | - Zhi Dong Wang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, Henan Province, China; Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Qing Qing Chang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, Henan Province, China; Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Lin Rui Dong
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, Henan Province, China; Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xiao Xue Shi
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, Henan Province, China; Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Ming Jian Li
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, Henan Province, China; Department of Neurology, People's Hospital of Henan University, Zhengzhou, Henan Province, China
| | - Qi Gu
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, Henan Province, China; Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China; Department of Neurology, People's Hospital of Henan University, Zhengzhou, Henan Province, China
| | - Si Yuan Chen
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, Henan Province, China; Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China; Department of Neurology, People's Hospital of Henan University, Zhengzhou, Henan Province, China
| |
Collapse
|
25
|
Ota Y, Kanel P, Bohnen N. Imaging of sleep disorders in pre-Parkinsonian syndromes. Curr Opin Neurol 2022; 35:443-452. [PMID: 35788559 PMCID: PMC9308698 DOI: 10.1097/wco.0000000000001084] [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] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Neuroimaging has been advanced in the last years and enabled clinicians to evaluate sleep disorders, especially isolated rapid eye movement sleep disorder (iRBD), which can be seen in alpha-synucleinopathies. iRBD is the best prodromal clinical marker for phenoconversion to these neurodegenerative diseases. This review aims to provide an update on advanced neuroimaging biomarkers in iRBD. RECENT FINDINGS Advanced structural MRI techniques, such as diffusion tensor imaging and functional MRI, neuromelanin-sensitive MRI, and scintigraphic neuroimaging such as cholinergic PET, dopamine transporter imaging - single-photon emission computerized tomography, perfusional single-photon emission computerized tomography, and cardiac metaiodobenzylguanidine can provide diagnostic and prognostic imaging biomarkers for iRBD, in isolation and more robustly when combined. SUMMARY New advanced neuroimaging can provide imaging biomarkers and aid in the appropriate clinical assessment and future therapeutic trials.
Collapse
Affiliation(s)
- Yoshiaki Ota
- The Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Prabesh Kanel
- The Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Morris K. Udall Center of Excellence for Parkinson’s Disease Research, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Parkinson’s Foundation Research Center of Excellence, Ann Arbor, MI, USA
| | - Nicolaas Bohnen
- The Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Morris K. Udall Center of Excellence for Parkinson’s Disease Research, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Parkinson’s Foundation Research Center of Excellence, Ann Arbor, MI, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- GRECC & Neurology Service, VAAAHS, Ann Arbor, MI, USA
| |
Collapse
|
26
|
Just MK, Gram H, Theologidis V, Jensen PH, Nilsson KPR, Lindgren M, Knudsen K, Borghammer P, Van Den Berge N. Alpha-Synuclein Strain Variability in Body-First and Brain-First Synucleinopathies. Front Aging Neurosci 2022; 14:907293. [PMID: 35693346 PMCID: PMC9178288 DOI: 10.3389/fnagi.2022.907293] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/02/2022] [Indexed: 12/15/2022] Open
Abstract
Pathogenic alpha-synuclein (asyn) aggregates are a defining feature of neurodegenerative synucleinopathies, which include Parkinson's disease, Lewy body dementia, pure autonomic failure and multiple system atrophy. Early accurate differentiation between these synucleinopathies is challenging due to the highly heterogeneous clinical profile at early prodromal disease stages. Therefore, diagnosis is often made in late disease stages when a patient presents with a broad range of motor and non-motor symptoms easing the differentiation. Increasing data suggest the clinical heterogeneity seen in patients is explained by the presence of distinct asyn strains, which exhibit variable morphologies and pathological functions. Recently, asyn seed amplification assays (PMCA and RT-QuIC) and conformation-specific ligand assays have made promising progress in differentiating between synucleinopathies in prodromal and advanced disease stages. Importantly, the cellular environment is known to impact strain morphology. And, asyn aggregate pathology can propagate trans-synaptically along the brain-body axis, affecting multiple organs and propagating through multiple cell types. Here, we present our hypothesis that the changing cellular environments, an asyn seed may encounter during its brain-to-body or body-to-brain propagation, may influence the structure and thereby the function of the aggregate strains developing within the different cells. Additionally, we aim to review strain characteristics of the different synucleinopathies in clinical and preclinical studies. Future preclinical animal models of synucleinopathies should investigate if asyn strain morphology is altered during brain-to-body and body-to-brain spreading using these seeding amplification and conformation-specific assays. Such findings would greatly deepen our understanding of synucleinopathies and the potential link between strain and phenotypic variability, which may enable specific diagnosis of different synucleinopathies in the prodromal phase, creating a large therapeutic window with potential future applications in clinical trials and personalized therapeutics.
Collapse
Affiliation(s)
- Mie Kristine Just
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| | - Hjalte Gram
- Department of Biomedicine, DANDRITE-Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark
| | - Vasileios Theologidis
- Department of Biomedicine, DANDRITE-Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark
| | - Poul Henning Jensen
- Department of Biomedicine, DANDRITE-Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark
| | - K. Peter R. Nilsson
- Division of Chemistry, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Mikael Lindgren
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Karoline Knudsen
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| | - Per Borghammer
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| | - Nathalie Van Den Berge
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
27
|
van der Horn HJ, Meles SK, Kok JG, Vergara VM, Qi S, Calhoun VD, Dalenberg JR, Siero JCW, Renken RJ, de Vries JJ, Spikman JM, Kremer HPH, De Jong BM. A resting-state fMRI pattern of spinocerebellar ataxia type 3 and comparison with 18F-FDG PET. Neuroimage Clin 2022; 34:103023. [PMID: 35489193 PMCID: PMC9062756 DOI: 10.1016/j.nicl.2022.103023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/25/2022] [Accepted: 04/24/2022] [Indexed: 11/17/2022]
Abstract
This is the first study identifying a resting-state fMRI pattern in SCA3. This pattern was closely associated with a metabolic (18F-FDG PET) counterpart. Pattern subject scores were highly correlated with ataxia severity.
Spinocerebellar ataxia type 3 (SCA3) is a rare genetic neurodegenerative disease. The neurobiological basis of SCA3 is still poorly understood, and up until now resting-state fMRI (rs-fMRI) has not been used to study this disease. In the current study we investigated (multi-echo) rs-fMRI data from patients with genetically confirmed SCA3 (n = 17) and matched healthy subjects (n = 16). Using independent component analysis (ICA) and subsequent regression with bootstrap resampling, we identified a pattern of differences between patients and healthy subjects, which we coined the fMRI SCA3 related pattern (fSCA3-RP) comprising cerebellum, anterior striatum and various cortical regions. Individual fSCA3-RP scores were highly correlated with a previously published 18F-FDG PET pattern found in the same sample (rho = 0.78, P = 0.0003). Also, a high correlation was found with the Scale for Assessment and Rating of Ataxia scores (r = 0.63, P = 0.007). No correlations were found with neuropsychological test scores, nor with levels of grey matter atrophy. Compared with the 18F-FDG PET pattern, the fSCA3-RP included a more extensive contribution of the mediofrontal cortex, putatively representing changes in default network activity. This rs-fMRI identification of additional regions is proposed to reflect a consequence of the nature of the BOLD technique, enabling measurement of dynamic network activity, compared to the more static 18F-FDG PET methodology. Altogether, our findings shed new light on the neural substrate of SCA3, and encourage further validation of the fSCA3-RP to assess its potential contribution as imaging biomarker for future research and clinical use.
Collapse
Affiliation(s)
- Harm J van der Horn
- Department of Neurology, University Medical Center Groningen, University of Groningen, the Netherlands.
| | - Sanne K Meles
- Department of Neurology, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Jelmer G Kok
- Department of Neurology, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Victor M Vergara
- Tri-institutional Center for Translational Research (TReNDS), Georgia State, Georgia Tech, Emory, Atlanta, GA, USA
| | - Shile Qi
- Tri-institutional Center for Translational Research (TReNDS), Georgia State, Georgia Tech, Emory, Atlanta, GA, USA
| | - Vince D Calhoun
- Tri-institutional Center for Translational Research (TReNDS), Georgia State, Georgia Tech, Emory, Atlanta, GA, USA
| | - Jelle R Dalenberg
- Department of Neurology, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Jeroen C W Siero
- Department of Radiology, Utrecht Center for Image Sciences, University Medical Center Utrecht, Utrecht, the Netherlands; Spinoza Centre for Neuroimaging Amsterdam, Amsterdam, the Netherlands
| | - Remco J Renken
- Department of Neuroscience, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Jeroen J de Vries
- Department of Neurology, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Jacoba M Spikman
- Department of Neuropsychology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Hubertus P H Kremer
- Department of Neurology, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Bauke M De Jong
- Department of Neurology, University Medical Center Groningen, University of Groningen, the Netherlands
| |
Collapse
|
28
|
Wang Y, Wei H, Du S, Yan H, Li X, Wu Y, Zhu J, Wang Y, Cai Z, Wang N. Functional Covariance Connectivity of Gray and White Matter in Olfactory-Related Brain Regions in Parkinson’s Disease. Front Neurosci 2022; 16:853061. [PMID: 35310108 PMCID: PMC8930839 DOI: 10.3389/fnins.2022.853061] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/14/2022] [Indexed: 01/13/2023] Open
Abstract
Before the onset of motor symptoms, Parkinson’s disease (PD) involves dysfunction of the anterior olfactory nucleus and olfactory bulb, causing olfactory disturbance, commonly resulting in hyposmia in the early stages of PD. Accumulating evidence has shown that blood oxygen level dependent (BOLD) signals in white matter are altered by olfactory disorders and related stimuli, and the signal changes in brain white matter pathways show a certain degree of specificity, which can reflect changes of early olfactory dysfunction in Parkinson’s disease. In this study, we apply the functional covariance connectivity (FCC) method to decode FCC of gray and white matter in olfactory-related brain regions in Parkinson’s disease. Our results show that the dorsolateral prefrontal, anterior entorhinal cortex and fronto-orbital cortices in the gray matter have abnormal connectivity with the posterior corona radiata and superior corona radiata in white matter in patients with Parkinson’s hyposmia. The functional covariance connection strength (FCS) of the right dorsolateral prefrontal cortex and white matter, and the covariance connection strength of the left superior corona radiata and gray matter function have potential diagnostic value. These results demonstrate that alterations in FCC of gray and white matter in olfactory-related brain regions can reflect the change of olfactory function in the early stages of Parkinson’s disease, indicating that it could be a potential neuroimaging marker for early diagnosis.
Collapse
Affiliation(s)
- Yiqing Wang
- Department of Neurology, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, China
- Department of Neurology, Gusu School, Nanjing Medical University, Suzhou, China
| | - Hongyu Wei
- Department of Neurology, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, China
| | - Shouyun Du
- Department of Neurology, Guanyun People’s Hospital, Lianyungang, China
| | - Hongjie Yan
- Department of Neurology, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, China
| | - Xiaojing Li
- Department of Neurology, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, China
| | - Yijie Wu
- Department of Neurology, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, China
| | - Jianbing Zhu
- Department of Radiology, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, China
| | - Yi Wang
- Department of Radiology, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, China
| | - Zenglin Cai
- Department of Neurology, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, China
- Department of Neurology, Gusu School, Nanjing Medical University, Suzhou, China
- *Correspondence: Zenglin Cai,
| | - Nizhuan Wang
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, China
- Nizhuan Wang,
| |
Collapse
|