1
|
Li J, Chen D, Tang Y, Chen Z, Zhou M, Wan L, Xiao L, Fu Y, He Z, Tang Z, Hu Z, Yuan X, Yang J, Zhu S, Guo X, Ouyang R, Qiu R, Tang B, Guo J, Jiang H, Hu S. Synaptic Density Reductions in MSA: A Potential Biomarker Identified Through [ 18F]SynVesT-1 PET Imaging. Ann Neurol 2025; 97:879-893. [PMID: 39829068 DOI: 10.1002/ana.27179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 12/03/2024] [Accepted: 12/30/2024] [Indexed: 01/22/2025]
Abstract
OBJECTIVE The objective of this study was to delineate synaptic density alterations in multiple system atrophy (MSA) and explore its potential role as a biomarker for MSA diagnosis and disease severity monitoring using [18F]SynVesT-1 positron emission tomography / computed tomography (PET CT). METHODS In this prospective study, 60 patients with MSA (30 patients with MSA-parkinsonian [MSA-P] subtype and 30 patients with MSA-cerebellar [MSA-C] subtype), 30 patients with Parkinson's disease (PD), and 30 age-matched healthy controls (HCs) underwent [18F]SynVesT-1 PET/CT for synaptic density assessment. Visual, voxel, and volumetric region of interest (VOI) analyses were used to elucidate synaptic density patterns in the MSA brain and establish diagnostic criteria. The diagnostic performances of both visual and VOI-based diagnostics were evaluated using receiver operating characteristic (ROC) analysis. Spearman correlation analyses were conducted to investigate the relationship between brain synaptic density and disease severity RESULTS: Patients with MSA displayed extensive reductions in synaptic density throughout the brain, notably affecting both primary VOIs (the cerebellum and putamen) and secondary VOIs including the medulla oblongata, ventral tegmental area, and pons. Notably, patients with MSA-C exhibited a remarkable decrease in cerebellar synaptic density, whereas patients with MSA-P demonstrated significant synaptic loss within the posterior putamen. Compared with patients with PD, the patients with MSA show a more pronounced reduction in synaptic density in infratentorial brain regions. VOI-based diagnosis significantly outperformed visual analysis in diagnosing and differentiating MSA and its subtypes. Synaptic density in primary and multiple secondary VOIs correlated significantly with motor scales in patients with MSA. INTERPRETATION Our study identified widespread synaptic density reductions in MSA, particularly in the basal ganglia and infratentorial region, suggesting [18F]SynVesT-1 PET as a potential biomarker for diagnosing and evaluating the disease, and guiding synaptic restoration trials. ANN NEUROL 2025;97:879-893.
Collapse
Affiliation(s)
- Jian Li
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Daji Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Yongxiang Tang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Zhao Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, People's Republic of China
| | - Ming Zhou
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Linlin Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, People's Republic of China
| | - Ling Xiao
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - You Fu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Zhiyou He
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Zhichao Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Zhengqun Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Xinrong Yuan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Jinhui Yang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Sudan Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Xuan Guo
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Riwei Ouyang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Rong Qiu
- School of Computer Science and Engineering, Central South University, Changsha, People's Republic of China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, People's Republic of China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, People's Republic of China
- National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, People's Republic of China
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Key Laboratory of Biological Nanotechnology, Changsha, People's Republic of China
| |
Collapse
|
2
|
Everix L, Elvas F, Miranda Menchaca A, Khetarpal V, Liu L, Bard J, Staelens S, Bertoglio D. Preclinical validation and kinetic modelling of the SV2A PET ligand [ 18F]UCB-J in mice. J Cereb Blood Flow Metab 2025; 45:920-931. [PMID: 39628318 PMCID: PMC11615906 DOI: 10.1177/0271678x241304923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/19/2024] [Accepted: 11/15/2024] [Indexed: 12/06/2024]
Abstract
Synaptic vesicle protein 2A (SV2A) is ubiquitously expressed in presynaptic terminals where it functions as a neurotransmission regulator protein. Synaptopathy has been reported during healthy ageing and in a variety of neurodegenerative diseases. Positron emission tomography (PET) imaging of SV2A can be used to evaluate synaptic density. The PET ligand [11C]UCB-J has high binding affinity and selectivity for SV2A but has a short physical half-life due to the 11C isotope. Here we report the characterization and validation of its 18F-labeled equivalent, [18F]UCB-J, in terms of specificity, reproducibility and stability in C57BL/6J mice. Plasma analysis revealed at least one polar radiometabolite. Kinetic modelling was performed using a population-based metabolite corrected image-derived input function (IDIF). [18F]UCB-J showed relatively fast kinetics and a reliable measure of the IDIF-based volume of distribution (VT(IDIF)). [18F]UCB-J specificity for SV2A was confirmed through a levetiracetam blocking assay (50 to 200 mg/kg). Reproducibility of the VT(IDIF) was determined through test-retest analysis, revealing significant correlation (r2 = 0.773, p < 0.0001). Time-stability analyses indicate a scan duration of 60 min to be sufficient to obtain a reliable VT(IDIF). In conclusion, [18F]UCB-J is a selective SV2A ligand with optimal kinetics in mice. Further investigation is warranted for (pre)clinical applicability of [18F]UCB-J in synaptopathies.
Collapse
Affiliation(s)
- Liesbeth Everix
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
| | - Filipe Elvas
- Molecular Imaging and Radiology (MIRA), Wilrijk, Belgium
| | | | - Vinod Khetarpal
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc., Princeton, NJ, USA
| | - Longbin Liu
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc., Princeton, NJ, USA
| | - Jonathan Bard
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc., Princeton, NJ, USA
| | - Steven Staelens
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
| | - Daniele Bertoglio
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
- Bio-Imaging Lab, University of Antwerp, Wilrijk, Belgium
- µNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
3
|
Accioly ARM, Menezes VO, Calixto LH, Bispo DPCF, Lachmann M, Mourato FA, Machado MAD, Diniz PRB. Machine Learning-Based Diagnostic Prediction Model Using T1-Weighted Striatal Magnetic Resonance Imaging for Early-Stage Parkinson's Disease Detection. Acad Radiol 2025:S1076-6332(25)00300-9. [PMID: 40253220 DOI: 10.1016/j.acra.2025.04.001] [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: 01/02/2025] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/21/2025]
Abstract
RATIONALE AND OBJECTIVES Diagnosing Parkinson's disease (PD) typically relies on clinical evaluations, often detecting it in advanced stages. Recently, artificial intelligence has increasingly been applied to imaging for neurodegenerative disorders. This study aims to develop a diagnostic prediction model using T1-weighted magnetic resonance imaging (T1-MRI) data from the caudate and putamen in individuals with early-stage PD. MATERIALS AND METHODS This retrospective case-control study included 69 early-stage PD patients and 22 controls, recruited through the Parkinson's Progression Markers Initiative. T1-MRI scans were acquired using a 3-tesla system. 432 radiomic features were extracted from images of the segmented caudate and putâmen in an automated way. Feature selection was performed using Pearson's correlation and recursive feature elimination to identify the most relevant variables. Three machine learning algorithms-random forest (RF), support vector machine and logistic regression-were evaluated for diagnostic prediction effectiveness using a cross-validation method. The Shapley Additive Explanations technique identified the most significant features distinguishing between the groups. The metrics used to evaluate the performance were discrimination, expressed in area under the ROC curve (AUC), sensitivity and specificity; and calibration, expressed as accuracy. RESULTS The RF algorithm showed superior performance with an average accuracy of 92.85%, precision of 100.00%, sensitivity of 86.66%, specificity of 96.65% and AUC of 0.93. The three most influential features were contrast, elongation, and gray-level non-uniformity, all from the putamen. CONCLUSION Machine learning-based models can differentiate early-stage PD from controls using T1-weighted MRI radiomic features.
Collapse
Affiliation(s)
- Alicia R M Accioly
- Medical Science Center, Federal University of Pernambuco, Recife, Brazil (A.R.M.A., L.H.C., D.P.C.F.B.).
| | - Vinícius O Menezes
- Nuclear Medicine, Clinical Hospital of Federal University of Pernambuco, Recife, Brazil (V.O.M., F.A.M.)
| | - Lucas H Calixto
- Medical Science Center, Federal University of Pernambuco, Recife, Brazil (A.R.M.A., L.H.C., D.P.C.F.B.)
| | - Dharah P C F Bispo
- Medical Science Center, Federal University of Pernambuco, Recife, Brazil (A.R.M.A., L.H.C., D.P.C.F.B.)
| | - Maximilian Lachmann
- Center for Exact and Natural Sciences, Federal University of Pernambuco, Recife, Brazil (M.L.)
| | - Felipe A Mourato
- Nuclear Medicine, Clinical Hospital of Federal University of Pernambuco, Recife, Brazil (V.O.M., F.A.M.)
| | - Marcos A D Machado
- Nuclear Medicine, Clinical Hospital of Federal University of Bahia, Salvador, Brazil (M.A.D.M.)
| | - Paula R B Diniz
- Telehealth Unit, Medical Science Center, Federal University of Pernambuco, Recife, Brazil (P.R.B.D.)
| |
Collapse
|
4
|
Tang C, Vanderlinden G, Schroyen G, Deprez S, Van Laere K, Koole M. A support vector machine-based approach to guide the selection of a pseudo-reference region for brain PET quantification. J Cereb Blood Flow Metab 2025; 45:568-577. [PMID: 39397394 PMCID: PMC11563559 DOI: 10.1177/0271678x241290912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 08/07/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024]
Abstract
A Support Vector Machine (SVM) based approach was developed to identify a pseudo-reference region for brain PET scans with the aim of reducing interscan and intersubject variability. By training a binary linear SVM classifier with PET datasets from two different groups, potential pseudo-reference regions were identified by considering their regional average or total contribution to the classification score. This approach was evaluated in three cohorts with different brain PET tracers: (1) 11C-PiB PET scans of Alzheimer's disease (AD) patients and age-matched controls (OC); (2) baseline and blocking scans of an 11C-UCB-J PET occupancy study; and (3) 18F-DPA-714 PET scans for healthy controls (HC) and chemo-treated women with breast cancer (BC). In the first cohort, cerebellum, brainstem, and subcortical white matter were confirmed as pseudo-reference regions. The same regions were identified for the second cohort using either the VT maps or the SUV images. In the third cohort, cerebellum and brainstem were identified as pseudo-reference regions, alongside subcortical white matter and temporal cortex. In addition, the SVM-based approach demonstrated robust performance even with a reduced number of subjects, therefore confirming its applicability in identifying pseudo-reference regions without a priori assumptions and with only limited data across different PET tracers.
Collapse
Affiliation(s)
- Chunmeng Tang
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Greet Vanderlinden
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Gwen Schroyen
- Translational MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Sabine Deprez
- Translational MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Division of Nuclear Medicine, University Hospitals UZ Leuven, Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| |
Collapse
|
5
|
Wang W, Wang Y, Xu L, Liu X, Hu Y, Li J, Huang Q, Ren S, Huang Y, Guan Y, Li Y, Hua F, Ye Q, Xie F. Presynaptic terminal integrity is associated with glucose metabolism in Parkinson's disease. Eur J Nucl Med Mol Imaging 2025; 52:1510-1519. [PMID: 39572432 DOI: 10.1007/s00259-024-06993-3] [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: 09/16/2024] [Accepted: 11/13/2024] [Indexed: 02/20/2025]
Abstract
OBJECTIVE To investigate the relationship of synaptic loss with glucose metabolism and dopaminergic transporters in Parkinson's disease (PD) patients. METHODS A total of 16 patients with PD and 11 age-matched healthy controls underwent positron emission tomography (PET) with the tracers [18F]SynVesT-1, a ligand for the presynaptic terminal marker synaptic vesicle protein 2 A (SV2A), and FDG. PD patients also underwent PET with the dopamine transporter (DAT) ligand [18F]FP-CIT. The difference in synaptic density between PD patients and age-matched normal controls(NCs) was determined in the selected regions of interest, and the correlations of the [18F]SynVesT-1 PET SUVRs with [18F]FP-CIT PET SUVRs and [18F]FDG PET SUVRs were evaluated. RESULTS Compared with that in the NC group, the synaptic density in the caudate region was significantly lower in the PD group (SUVR: 2.51 ± 0.36 vs. 3.18 ± 0.32, p < 0.001), especially in the pre-commissural caudate and post-commissural caudate (SUVR: 2.42 ± 0.29 vs. 2.63 ± 0.32, p < 0.01; 0.76 ± 0.31 vs. 0.97 ± 0.33, p < 0.001). A reduced synaptic density was significantly correlated with DAT (r = 0.61, p < 0.001) and glucose metabolism (r = 0.73, p < 0.001) in the post-commissural caudate. In the post-commissural regions of the caudate, there was a partial mediating effect of synaptic density on the relationship between glucose metabolism and DAT availability (indirect effect: β4 = 0.039, p = 0.024). CONCLUSION [18F]SynVesT-1 binds specifically to SV2A, reflecting synaptic density, and there is a positive correlation metabolic pattern related to the changes reflected by [18F]SynVesT-1 and [18F]FDG.
Collapse
Affiliation(s)
- Weiyi Wang
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yanru Wang
- Department of Nuclear Medicine, Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China
| | - Limin Xu
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China
| | - Xueling Liu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuqing Hu
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China
| | - Junpeng Li
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Qi Huang
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Shuhua Ren
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yuxin Li
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Fengchun Hua
- Department of Nuclear Medicine, Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China.
| | - Qing Ye
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China.
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
6
|
Kiersnowski OC, Mattioli P, Argenti L, Avanzino L, Calizzano F, Diociasi A, Falcitano L, Liu C, Losa M, Massa F, Morbelli S, Orso B, Pelosin E, Raffa S, Pardini M, Arnaldi D, Roccatagliata L, Costagli M. Magnetic susceptibility components reveal different aspects of neurodegeneration in alpha-synucleinopathies. Sci Rep 2025; 15:4186. [PMID: 39905067 PMCID: PMC11794440 DOI: 10.1038/s41598-024-83593-z] [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: 09/20/2024] [Accepted: 12/16/2024] [Indexed: 02/06/2025] Open
Abstract
Nigrostriatal dopaminergic degeneration in alpha-synucleinopathies is indirectly reflected by low dopamine transporter (DaT) uptake through [123I]FP-CIT-SPECT. Bulk magnetic susceptibility (χ) in the substantia nigra, from MRI-based quantitative susceptibility mapping (QSM), is a potential biomarker of nigrostriatal degeneration, however, QSM cannot disentangle paramagnetic (e.g. iron) and diamagnetic (e.g. myelin) sources. Using the susceptibility source-separation technique DECOMPOSE, paramagnetic component susceptibility (PCS) and diamagnetic component susceptibility (DCS) were studied in prodromal and overt alpha-synucleinopathies, and their relationships with DaT-SPECT specific binding ratio (SBR) and clinical scores. 78 participants were included (23 controls, 30 prodromal and 25 overt alpha-synucleinopathies). Prodromal patients were subdivided into groups with positive or negative DaT-SPECT (SBR Z-scores below or above -1, respectively). Correlations of putamen and caudate SBR Z-scores with PCS and DCS in the substantia nigra, putamen, and caudate were investigated. Increased PCS was observed in the substantia nigra of prodromal alpha-synucleinopathy patients with positive DaT-SPECT compared to controls and prodromal patients with negative DaT-SPECT. SBR Z-scores in the putamen correlated with increased PCS in the substantia nigra and reduced |DCS| in the putamen, which may reflect dopaminergic degeneration ascribable to iron accumulation and nigrostriatal neuron axonal loss, respectively.
Collapse
Affiliation(s)
| | - Pietro Mattioli
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Lucia Argenti
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Laura Avanzino
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Francesco Calizzano
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | | | | | - Chunlei Liu
- University of California Berkeley, Berkeley, United States of America
| | - Mattia Losa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Federico Massa
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Silvia Morbelli
- Department of Nuclear Medicine, University of Turin, Turin, Italy
| | - Beatrice Orso
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Elisa Pelosin
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Stefano Raffa
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Matteo Pardini
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Dario Arnaldi
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Luca Roccatagliata
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.
- Department of Health Sciences, University of Genova, Genova, Italy.
| | - Mauro Costagli
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| |
Collapse
|
7
|
Binda KH, Real CC, Simonsen MT, Grove EK, Bender D, Gjedde A, Brooks DJ, Landau AM. Acute transcutaneous auricular vagus nerve stimulation modulates presynaptic SV2A density in healthy rat brain: An in vivo microPET study. Psychophysiology 2025; 62:e14709. [PMID: 39428713 PMCID: PMC11775880 DOI: 10.1111/psyp.14709] [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: 03/31/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/22/2024]
Abstract
Vagus nerve stimulation (VNS) is the subject of exploration as an adjunct treatment for neurological disorders such as epilepsy, chronic migraine, pain, and depression. A non-invasive form of VNS is transcutaneous auricular VNS (taVNS). Combining animal models and positron emission tomography (PET) may lead to a better understanding of the elusive mechanisms of taVNS. We evaluated the acute effect of electrical stimulation of the left vagus nerve via the ear on brain synaptic vesicle glycoprotein 2A (SV2A) as a measure of presynaptic density and glucose metabolism in naïve rats. Female Sprague-Dawley rats were imaged with [11C]UCB-J (n = 11) or [18F]fluorodeoxyglucose ([18F]FDG) PET (n = 13) on two separate days, (1) at baseline, and (2) after acute unilateral left taVNS or sham stimulation (30 min). We calculated the regional volume of distribution (VT) for [11C]UCB-J and standard uptake values (SUV) for [18F]FDG. We observed regional reductions of [11C]UCB-J binding in response to taVNS ranging from 36% to 59%. The changes in taVNS compared to baseline were significantly larger than those induced by sham stimulation. The differences were observed bilaterally in the frontal cortex, striatum, and midbrain. The [18F]FDG PET uptake remained unchanged following acute taVNS or sham stimulation compared to baseline values. This proof-of-concept study shows for the first time that acute taVNS for 30 min can modulate in vivo synaptic SV2A density in cortical and subcortical regions of healthy rats. Preclinical disease models and PET ligands of different targets can be a powerful combination to assess the therapeutic potential of taVNS.
Collapse
Affiliation(s)
- Karina H. Binda
- Translational Neuropsychiatry Unit, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Caroline C. Real
- Department of Nuclear Medicine and PET, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Mette T. Simonsen
- Department of Nuclear Medicine and PET, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Ebbe K. Grove
- Department of Nuclear Medicine and PET, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Dirk Bender
- Department of Nuclear Medicine and PET, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Albert Gjedde
- Translational Neuropsychiatry Unit, Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of NeuroscienceUniversity of CopenhagenCopenhagenDenmark
- Department of Neurology and NeurosurgeryMcGill UniversityMontrealQuebecCanada
| | - David J. Brooks
- Department of Nuclear Medicine and PET, Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Institute of Translational and Clinical Research, University of Newcastle upon TyneNewcastle Upon TyneUK
| | - Anne M. Landau
- Translational Neuropsychiatry Unit, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| |
Collapse
|
8
|
Howes O, Marcinkowska J, Turkheimer FE, Carr R. Synaptic changes in psychiatric and neurological disorders: state-of-the art of in vivo imaging. Neuropsychopharmacology 2024; 50:164-183. [PMID: 39134769 PMCID: PMC11525650 DOI: 10.1038/s41386-024-01943-x] [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: 03/27/2024] [Revised: 07/03/2024] [Accepted: 07/19/2024] [Indexed: 11/01/2024]
Abstract
Synapses are implicated in many neuropsychiatric illnesses. Here, we provide an overview of in vivo techniques to index synaptic markers in patients. Several positron emission tomography (PET) tracers for synaptic vesicle glycoprotein 2 A (SV2A) show good reliability and selectivity. We review over 50 clinical studies including over 1700 participants, and compare findings in healthy ageing and across disorders, including addiction, schizophrenia, depression, posttraumatic stress disorder, and neurodegenerative disorders, including tauopathies, Huntington's disease and α-synucleinopathies. These show lower SV2A measures in cortical brain regions across most of these disorders relative to healthy volunteers, with the most well-replicated findings in tauopathies, whilst changes in Huntington's chorea, Parkinson's disease, corticobasal degeneration and progressive supranuclear palsy are predominantly subcortical. SV2A PET measures are correlated with functional connectivity across brain networks, and a number of other measures of brain function, including glucose metabolism. However, the majority of studies found no relationship between grey matter volume measured with magnetic resonance imaging and SV2A PET measures. Cognitive dysfunction, in domains including working memory and executive function, show replicated inverse relationships with SV2A measures across diagnoses, and initial findings also suggest transdiagnostic relationships with mood and anxiety symptoms. This suggests that synaptic abnormalities could be a common pathophysiological substrate underlying cognitive and, potentially, affective symptoms. We consider limitations of evidence and future directions; highlighting the need to develop postsynaptic imaging markers and for longitudinal studies to test causal mechanisms.
Collapse
Affiliation(s)
- Oliver Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England.
- South London & the Maudsley NHS Trust, London, England.
- London Institute of Medical Sciences, London, England.
| | - Julia Marcinkowska
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England
| | - Federico E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England
| | - Richard Carr
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England
- South London & the Maudsley NHS Trust, London, England
- London Institute of Medical Sciences, London, England
| |
Collapse
|
9
|
Yao H, Tong W, Song Y, Li R, Xiang X, Cheng W, Zhou Y, He Y, Yang Y, Liu Y, Li S, Jin L. Exercise training upregulates CD55 to suppress complement-mediated synaptic phagocytosis in Parkinson's disease. J Neuroinflammation 2024; 21:246. [PMID: 39342308 PMCID: PMC11439226 DOI: 10.1186/s12974-024-03234-0] [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: 07/08/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024] Open
Abstract
The primary pathological change in Parkinson's disease (PD) is the progressive degeneration of dopaminergic neurons in the substantia nigra. Additionally, excessive microglial activation and synaptic loss are also typical features observed in PD samples. Exercise trainings have been proven to improve PD symptoms, delay the disease progression as well as affect excessive microglial synaptic phagocytosis. In this study, we established a mouse model of PD by injecting mouse-derived α-synuclein preformed fibrils (M-α-syn PFFs) into the substantia nigra, and demonstrated that treadmill exercise inhibits microglial activation and synaptic phagocytosis in striatum. Using RNA-Seq and proteomics, we also found that PD involves excessive activation of the complement pathway which is closely related to over-activation of microglia and abnormal synaptic function. More importantly, exercise training can inhibit complement levels and complement-mediated microglial phagocytosis of synapses. It is probably triggered by CD55, as we observed that CD55 in the striatum significantly increased after exercise training and up-regulation of that molecule rescued motor deficits of PD mice, accompanied with reduced microglial synaptic phagocytosis in the striatum. This research elucidated the interplay among microglia, complement, and synapses, and analyzed the effects of exercise training on these factors. Our work also suggested CD55 as a complement-relevant candidate molecule for developing therapeutic strategies of PD.
Collapse
Affiliation(s)
- Hongkai Yao
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
- Neurotoxin Research Center, Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weifang Tong
- Neurotoxin Research Center, Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yunping Song
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Ruoyu Li
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Xuerui Xiang
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Wen Cheng
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
- Neurotoxin Research Center, Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yunjiao Zhou
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Yijing He
- Neurotoxin Research Center, Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi Yang
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Yunxi Liu
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Siguang Li
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Lingjing Jin
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
10
|
Chen L, Li X, Ge Y, Li H, Li R, Song X, Liang J, Zhang W, Li X, Wang X, Wang Y, Wu Y, Bai Y, Wang M. GMP-compliant automated radiosynthesis of [ 18F] SynVesT-1 for PET imaging of synaptic vesicle glycoprotein 2 A (SV2A). EJNMMI Radiopharm Chem 2024; 9:66. [PMID: 39254802 PMCID: PMC11387577 DOI: 10.1186/s41181-024-00284-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 07/09/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND A novel positron emission tomography (PET) imaging tracer, [18F] SynVesT-1, targeting synaptic vesicle glycoprotein 2 (SV2A), has been developed to meet clinical demand. Utilizing the Trasis AllinOne-36 (AIO) module, we've automated synthesis to Good Manufacturing Practice (GMP) standards, ensuring sterile, pyrogen-free production. The fully GMP-compliant robust synthesis of [18F] SynVesT-1 boosting reliability and introducing a significant degree of simplicity and its comprehensive validation for routine human use. RESULTS [18F] SynVesT-1 was synthesized by small modifications to the original [18F] SynVesT-1 synthesis protocol to better fit AIO module using an in-house designed cassette and sequence. With a relatively small precursor load of 5 mg, [18F] SynVesT-1 was obtained with consistently high radiochemical yields (RCY) of 20.6 ± 1.2% (the decay-corrected RCY, n = 3) at end of synthesis. Each of the final formulated batches demonstrated radiochemical purity (RCP) and enantiomeric purity surpassing 99%. The entire synthesis process was completed within a timeframe of 80 min (75 ± 3.1 min, n = 3), saves 11 min compared to reported GMP automated synthesis procedures. The in-human PET imaging of total body PET/CT and time-of-flight (TOF) PET/MR showed that [18F] SynVesT-1 is an excellent tracer for SV2A. It is advantageous for decentralized promotion and application in multi-center studies. CONCLUSION The use of AIO synthesizer maintains high production yields and increases reliability, reduces production time and allows rapid training of production staff. Besides, the as-prepared [18F] SynVesT-1 displays excellent in vivo binding properties in humans and holds great potential for the imaging and quantification of synaptic density in vivo.
Collapse
Affiliation(s)
- Lijuan Chen
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Xiaochen Li
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Yao Ge
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Huiqiang Li
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Ruili Li
- School of Clinical Medicine, Henan University, Kaifeng, 475004, China
| | - Xiaosheng Song
- Institute of Biomedicine, Henan Academy of Sciences, Zhengzhou, 450046, China
| | | | - Weifeng Zhang
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Xiaona Li
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Xiaoqi Wang
- School of Clinical Medicine, Henan University, Kaifeng, 475004, China
| | - Yunjuan Wang
- Institute of Biomedicine, Henan Academy of Sciences, Zhengzhou, 450046, China
- School of life sciences, Henan University, Kaifeng, 475004, China
| | - Yaping Wu
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Yan Bai
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Meiyun Wang
- Department of Medical Imaging, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, China.
- Institute of Biomedicine, Henan Academy of Sciences, Zhengzhou, 450046, China.
| |
Collapse
|
11
|
Fernandes EFA, Palner M, Raval NR, Jeppesen TE, Danková D, Bærentzen SL, Werner C, Eilts J, Maric HM, Doose S, Aripaka SS, Kaalund SS, Aznar S, Kjaer A, Schlosser A, Haugaard-Kedström LM, Knudsen GM, Herth MM, Stro Mgaard K. Development of Peptide-Based Probes for Molecular Imaging of the Postsynaptic Density in the Brain. J Med Chem 2024; 67:11975-11988. [PMID: 38981131 DOI: 10.1021/acs.jmedchem.4c00615] [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: 07/11/2024]
Abstract
The postsynaptic density (PSD) comprises numerous scaffolding proteins, receptors, and signaling molecules that coordinate synaptic transmission in the brain. Postsynaptic density protein 95 (PSD-95) is a master scaffold protein within the PSD and one of its most abundant proteins and therefore constitutes a very attractive biomarker of PSD function and its pathological changes. Here, we exploit a high-affinity inhibitor of PSD-95, AVLX-144, as a template for developing probes for molecular imaging of the PSD. AVLX-144-based probes were labeled with the radioisotopes fluorine-18 and tritium, as well as a fluorescent tag. Tracer binding showed saturable, displaceable, and uneven distribution in rat brain slices, proving effective in quantitative autoradiography and cell imaging studies. Notably, we observed diminished tracer binding in human post-mortem Parkinson's disease (PD) brain slices, suggesting postsynaptic impairment in PD. We thus offer a suite of translational probes for visualizing and understanding PSD-related pathologies.
Collapse
Affiliation(s)
- Eduardo F A Fernandes
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen DK-2100, Denmark
| | - Mikael Palner
- Neurobiology Research Unit, Rigshospitalet, Blegdamsvej 9, Copenhagen DK-2100, Denmark
| | - Nakul Ravi Raval
- Neurobiology Research Unit, Rigshospitalet, Blegdamsvej 9, Copenhagen DK-2100, Denmark
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3, Copenhagen DK-2200, Denmark
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Troels E Jeppesen
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen DK-2200, Denmark
| | - Daniela Danková
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen DK-2100, Denmark
| | - Simone L Bærentzen
- Neurobiology Research Unit, Rigshospitalet, Blegdamsvej 9, Copenhagen DK-2100, Denmark
| | - Christian Werner
- Department of Biotechnology and Biophysics, Biocenter, Julius-Maximilians-University, Am Hubland, Würzburg D-97074, Germany
| | - Janna Eilts
- Department of Biotechnology and Biophysics, Biocenter, Julius-Maximilians-University, Am Hubland, Würzburg D-97074, Germany
| | - Hans M Maric
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen DK-2100, Denmark
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-University, Josef-Schneider-Str. 2, Würzburg 97080, Germany
| | - Sören Doose
- Department of Biotechnology and Biophysics, Biocenter, Julius-Maximilians-University, Am Hubland, Würzburg D-97074, Germany
| | - Sanjay Sagar Aripaka
- Neurobiology Research Unit, Rigshospitalet, Blegdamsvej 9, Copenhagen DK-2100, Denmark
| | - Sanne Simone Kaalund
- Center for Neuroscience and Stereology, Bispebjerg University Hospital, Nielsine Nielsens Vej 6B, Copenhagen DK-2400, Denmark
| | - Susana Aznar
- Center for Neuroscience and Stereology, Bispebjerg University Hospital, Nielsine Nielsens Vej 6B, Copenhagen DK-2400, Denmark
- Center for Translational Research, Bispebjerg University Hospital, Nielsine Nielsens Vej 4B, Copenhagen DK-2400, Denmark
| | - Andreas Kjaer
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3, Copenhagen DK-2200, Denmark
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen DK-2200, Denmark
| | - Andreas Schlosser
- Department of Biotechnology and Biophysics, Biocenter, Julius-Maximilians-University, Am Hubland, Würzburg D-97074, Germany
| | - Linda M Haugaard-Kedström
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen DK-2100, Denmark
| | - Gitte M Knudsen
- Neurobiology Research Unit, Rigshospitalet, Blegdamsvej 9, Copenhagen DK-2100, Denmark
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3, Copenhagen DK-2200, Denmark
| | - Matthias M Herth
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen DK-2100, Denmark
- Neurobiology Research Unit, Rigshospitalet, Blegdamsvej 9, Copenhagen DK-2100, Denmark
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen DK-2200, Denmark
| | - Kristian Stro Mgaard
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen DK-2100, Denmark
| |
Collapse
|
12
|
Bavarsad MS, Grinberg LT. SV2A PET imaging in human neurodegenerative diseases. Front Aging Neurosci 2024; 16:1380561. [PMID: 38699560 PMCID: PMC11064927 DOI: 10.3389/fnagi.2024.1380561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/20/2024] [Indexed: 05/05/2024] Open
Abstract
This manuscript presents a thorough review of synaptic vesicle glycoprotein 2A (SV2A) as a biomarker for synaptic integrity using Positron Emission Tomography (PET) in neurodegenerative diseases. Synaptic pathology, characterized by synaptic loss, has been linked to various brain diseases. Therefore, there is a need for a minimally invasive approach to measuring synaptic density in living human patients. Several radiotracers targeting synaptic vesicle protein 2A (SV2A) have been created and effectively adapted for use in human subjects through PET scans. SV2A is an integral glycoprotein found in the membranes of synaptic vesicles in all synaptic terminals and is widely distributed throughout the brain. The review delves into the development of SV2A-specific PET radiotracers, highlighting their advancements and limitations in neurodegenerative diseases. Among these tracers, 11C-UCB-J is the most used so far. We summarize and discuss an increasing body of research that compares measurements of synaptic density using SV2A PET with other established indicators of neurodegenerative diseases, including cognitive performance and radiological findings, thus providing a comprehensive analysis of SV2A's effectiveness and reliability as a diagnostic tool in contrast to traditional markers. Although the literature overall suggests the promise of SV2A as a diagnostic and therapeutic monitoring tool, uncertainties persist regarding the superiority of SV2A as a biomarker compared to other available markers. The review also underscores the paucity of studies characterizing SV2A distribution and loss in human brain tissue from patients with neurodegenerative diseases, emphasizing the need to generate quantitative neuropathological maps of SV2A density in cases with neurodegenerative diseases to fully harness the potential of SV2A PET imaging in clinical settings. We conclude by outlining future research directions, stressing the importance of integrating SV2A PET imaging with other biomarkers and clinical assessments and the need for longitudinal studies to track SV2A changes throughout neurodegenerative disease progression, which could lead to breakthroughs in early diagnosis and the evaluation of new treatments.
Collapse
Affiliation(s)
| | - Lea T. Grinberg
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco (UCSF), San Francisco, CA, United States
| |
Collapse
|
13
|
Wang X, Zhang X, Liu J, Zhang J, Liu C, Cui Y, Song Q, Hou Y, Wang Y, Zhang Q, Zhang Y, Fan Y, Jia J, Wang P. Synaptic vesicle glycoprotein 2 A in serum is an ideal biomarker for early diagnosis of Alzheimer's disease. Alzheimers Res Ther 2024; 16:82. [PMID: 38615037 PMCID: PMC11015666 DOI: 10.1186/s13195-024-01440-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/26/2024] [Indexed: 04/15/2024]
Abstract
BACKGROUND Previous studies have demonstrated that early intervention was the best plan to inhibit the progression of Alzheimer's disease (AD), which relied on the discovery of early diagnostic biomarkers. In this study, synaptic vesicle glycoprotein 2 A (SV2A) was examined to improve the early diagnostic efficiency in AD. METHODS In this study, biomarker testing was performed through the single-molecule array (Simoa). A total of 121 subjects including cognitively unimpaired controls, amnestic mild cognitive impairment (aMCI), AD and other types of dementia underwent cerebrospinal fluid (CSF) SV2A testing; 430 subjects including health controls, aMCI, AD and other types of dementia underwent serum SV2A, glial fibrillary acidic protein (GFAP), neurofilament light chain (NfL) and p-tau217 testing; 92 subjects including aMCI and AD underwent both CSF SV2A and serum SV2A testing; 115 cognitively unimpaired subjects including APOE ε4 carriers and APOE ε4 non-carriers were tested for serum SV2A, GFAP, NfL and p-tau217. Then, the efficacy of SV2A for the early diagnosis of AD and its ability to identify those at high risk of AD from a cognitively unimpaired population were further analyzed. RESULTS Both CSF and serum SV2A significantly and positively correlated with cognitive performance in patients with AD, and their levels gradually decreased with the progression of AD. Serum SV2A demonstrated excellent diagnostic efficacy for aMCI, with a sensitivity of 97.8%, which was significantly higher than those of NfL, GFAP, and p-tau217. The SV2A-positive rates ranged from 92.86 to 100% in aMCI cases that were negative for the above three biomarkers. Importantly, of all the biomarkers tested, serum SV2A had the highest positivity rate (81.82%) in individuals at risk for AD. CONCLUSIONS Serum SV2A was demonstrated to be a novel and ideal biomarker for the early diagnosis of AD, which can effectively distinguish those at high risk of AD in cognitively unimpaired populations.
Collapse
Affiliation(s)
- Xiaoling Wang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Xiaomin Zhang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Jing Liu
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Jingjing Zhang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Congcong Liu
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Yuting Cui
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Qiao Song
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Yuli Hou
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Yaqi Wang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Qian Zhang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Yingzhen Zhang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Yujian Fan
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Jianping Jia
- National Clinical Research Center for Geriatric Disorders, 45 Changchun Street, Beijing, 100053, China.
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.
- Beijing Key Laboratory of Geriatric Cognitive Disorders, 45 Changchun Street, Beijing, 100053, China.
- Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.
- Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, 45 Changchun Street, Beijing, 100053, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, 45 Changchun Street, Beijing, 100053, China.
| | - Peichang Wang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.
- National Clinical Research Center for Geriatric Disorders, 45 Changchun Street, Beijing, 100053, China.
| |
Collapse
|
14
|
Martin SL, Uribe C, Strafella AP. PET imaging of synaptic density in Parkinsonian disorders. J Neurosci Res 2024; 102:e25253. [PMID: 37814917 DOI: 10.1002/jnr.25253] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/31/2023] [Accepted: 09/21/2023] [Indexed: 10/11/2023]
Abstract
Synaptic dysfunction and altered synaptic pruning are present in people with Parkinsonian disorders. Dopamine loss and alpha-synuclein accumulation, two hallmarks of Parkinson's disease (PD) pathology, contribute to synaptic dysfunction and reduced synaptic density in PD. Atypical Parkinsonian disorders are likely to have unique spatiotemporal patterns of synaptic density, differentiating them from PD. Therefore, quantification of synaptic density has the potential to support diagnoses, monitor disease progression, and treatment efficacy. Novel radiotracers for positron emission tomography which target the presynaptic vesicle protein SV2A have been developed to quantify presynaptic density. The radiotracers have successfully investigated synaptic density in preclinical models of PD and people with Parkinsonian disorders. Therefore, this review will summarize the preclinical and clinical utilization of SV2A radiotracers in people with Parkinsonian disorders. We will evaluate how SV2A abundance is associated with other imaging modalities and the considerations for interpreting SV2A in Parkinsonian pathology.
Collapse
Affiliation(s)
- Sarah L Martin
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Carme Uribe
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Unitat de Psicologia Medica, Departament de Medicina, Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
| | - Antonio P Strafella
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Edmond J. Safra Parkinson Disease Program, Neurology Division, Toronto Western Hospital & Krembil Brain Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
15
|
Holmes SE, Honhar P, Tinaz S, Naganawa M, Hilmer AT, Gallezot JD, Dias M, Yang Y, Toyonaga T, Esterlis I, Mecca A, Van Dyck C, Henry S, Ropchan J, Nabulsi N, Louis ED, Comley R, Finnema SJ, Carson RE, Matuskey D. Synaptic loss and its association with symptom severity in Parkinson's disease. NPJ Parkinsons Dis 2024; 10:42. [PMID: 38402233 PMCID: PMC10894197 DOI: 10.1038/s41531-024-00655-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/06/2024] [Indexed: 02/26/2024] Open
Abstract
Parkinson's disease (PD) is the fastest growing neurodegenerative disease, but at present there is no cure, nor any disease-modifying treatments. Synaptic biomarkers from in vivo imaging have shown promise in imaging loss of synapses in PD and other neurodegenerative disorders. Here, we provide new clinical insights from a cross-sectional, high-resolution positron emission tomography (PET) study of 30 PD individuals and 30 age- and sex-matched healthy controls (HC) with the radiotracer [11C]UCB-J, which binds to synaptic vesicle glycoprotein 2A (SV2A), and is therefore, a biomarker of synaptic density in the living brain. We also examined a measure of relative brain perfusion from the early part of the same PET scan. Our results provide evidence for synaptic density loss in the substantia nigra that had been previously reported, but also extend this to other early-Braak stage regions known to be affected in PD (brainstem, caudate, olfactory cortex). Importantly, we also found a direct association between synaptic density loss in the nigra and severity of symptoms in patients. A greater extent and wider distribution of synaptic density loss in PD patients with longer illness duration suggests that [11C]UCB-J PET can be used to measure synapse loss with disease progression. We also demonstrate lower brain perfusion in PD vs. HC groups, with a greater extent of abnormalities in those with longer duration of illness, suggesting that [11C]UCB-J PET can simultaneously provide information on changes in brain perfusion. These results implicate synaptic imaging as a useful PD biomarker for future disease-modifying interventions.
Collapse
Affiliation(s)
- Sophie E Holmes
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Praveen Honhar
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale School of Engineering and Applied Sciences, Yale University, New Haven, CT, USA
| | - Sule Tinaz
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Mika Naganawa
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Ansel T Hilmer
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale School of Engineering and Applied Sciences, Yale University, New Haven, CT, USA
| | | | - Mark Dias
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Yanghong Yang
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Takuya Toyonaga
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Irina Esterlis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Adam Mecca
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | | | - Shannan Henry
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Jim Ropchan
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Nabeel Nabulsi
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Elan D Louis
- Department of Neurology, University of Texas Southwestern Medical Center, New Haven, CT, USA
| | | | | | - Richard E Carson
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale School of Engineering and Applied Sciences, Yale University, New Haven, CT, USA
| | - David Matuskey
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
16
|
Drake LR, Wu Y, Naganawa M, Asch R, Zheng C, Najafzadeh S, Pracitto R, Lindemann M, Li S, Ropchan J, Labaree D, Emery PR, Dias M, Henry S, Nabulsi N, Matuskey D, Hillmer AT, Gallezot JD, Carson RE, Cai Z, Huang Y. First-in-Human Study of 18F-SynVesT-2: An SV2A PET Imaging Probe with Fast Brain Kinetics and High Specific Binding. J Nucl Med 2024; 65:jnumed.123.266470. [PMID: 38360052 PMCID: PMC10924160 DOI: 10.2967/jnumed.123.266470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 02/17/2024] Open
Abstract
PET imaging of synaptic vesicle glycoprotein 2A allows for noninvasive quantification of synapses. This first-in-human study aimed to evaluate the kinetics, test-retest reproducibility, and extent of specific binding of a recently developed synaptic vesicle glycoprotein 2A PET ligand, (R)-4-(3-(18F-fluoro)phenyl)-1-((3-methylpyridin-4-yl)methyl)pyrrolidine-2-one (18F-SynVesT-2), with fast brain kinetics. Methods: Nine healthy volunteers participated in this study and were scanned on a High Resolution Research Tomograph scanner with 18F-SynVesT-2. Five volunteers were scanned twice on 2 different days. Five volunteers were rescanned with preinjected levetiracetam (20 mg/kg, intravenously). Arterial blood was collected to calculate the plasma free fraction and generate the arterial input function. Individual MR images were coregistered to a brain atlas to define regions of interest for generating time-activity curves, which were fitted with 1- and 2-tissue-compartment (1TC and 2TC) models to derive the regional distribution volume (V T). The regional nondisplaceable binding potential (BP ND) was calculated from 1TC V T, using the centrum semiovale (CS) as the reference region. Results: 18F-SynVesT-2 was synthesized with high molar activity (187 ± 69 MBq/nmol, n = 19). The parent fraction of 18F-SynVesT-2 in plasma was 28% ± 8% at 30 min after injection, and the plasma free fraction was high (0.29 ± 0.04). 18F-SynVesT-2 entered the brain quickly, with an SUVpeak of 8 within 10 min after injection. Regional time-activity curves fitted well with both the 1TC and the 2TC models; however, V T was estimated more reliably using the 1TC model. The 1TC V T ranged from 1.9 ± 0.2 mL/cm3 in CS to 7.6 ± 0.8 mL/cm3 in the putamen, with low absolute test-retest variability (6.0% ± 3.6%). Regional BP ND ranged from 1.76 ± 0.21 in the hippocampus to 3.06 ± 0.29 in the putamen. A 20-min scan was sufficient to provide reliable V T and BP ND Conclusion: 18F-SynVesT-2 has fast kinetics, high specific uptake, and low nonspecific uptake in the brain. Consistent with the nonhuman primate results, the kinetics of 18F-SynVesT-2 is faster than the kinetics of 11C-UCB-J and 18F-SynVesT-1 in the human brain and enables a shorter dynamic scan to derive physiologic information on cerebral blood flow and synapse density.
Collapse
Affiliation(s)
| | - Yanjun Wu
- Yale PET Center, New Haven, Connecticut
| | | | - Ruth Asch
- Yale PET Center, New Haven, Connecticut
| | | | | | | | | | - Songye Li
- Yale PET Center, New Haven, Connecticut
| | | | | | | | - Mark Dias
- Yale PET Center, New Haven, Connecticut
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Leung IHK, Strudwick MW. A systematic review of the challenges, emerging solutions and applications, and future directions of PET/MRI in Parkinson's disease. EJNMMI REPORTS 2024; 8:3. [PMID: 38748251 PMCID: PMC10962627 DOI: 10.1186/s41824-024-00194-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 12/26/2023] [Indexed: 05/19/2024]
Abstract
PET/MRI is a hybrid imaging modality that boasts the simultaneous acquisition of high-resolution anatomical data and metabolic information. Having these exceptional capabilities, it is often implicated in clinical research for diagnosing and grading, as well as tracking disease progression and response to interventions. Despite this, its low level of clinical widespread use is questioned. This is especially the case with Parkinson's disease (PD), the fastest progressively disabling and neurodegenerative cause of death. To optimise the clinical applicability of PET/MRI for diagnosing, differentiating, and tracking PD progression, the emerging novel uses, and current challenges must be identified. This systematic review aimed to present the specific challenges of PET/MRI use in PD. Further, this review aimed to highlight the possible resolution of these challenges, the emerging applications and future direction of PET/MRI use in PD. EBSCOHost (indexing CINAHL Plus, PsycINFO) Ovid (Medline, EMBASE) PubMed, Web of Science, and Scopus from 2006 (the year of first integrated PET/MRI hybrid system) to 30 September 2022 were used to search for relevant primary articles. A total of 933 studies were retrieved and following the screening procedure, 18 peer-reviewed articles were included in this review. This present study is of great clinical relevance and significance, as it informs the reasoning behind hindered widespread clinical use of PET/MRI for PD. Despite this, the emerging applications of image reconstruction developed by PET/MRI research data to the use of fully automated systems show promising and desirable utility. Furthermore, many of the current challenges and limitations can be resolved by using much larger-sampled and longitudinal studies. Meanwhile, the development of new fast-binding tracers that have specific affinity to PD pathological processes is warranted.
Collapse
|
18
|
Kang S, Jeon S, Lee YG, Ye BS. Dopamine transporter positron emission tomography in patients with Alzheimer's disease with Lewy body disease features. Neurobiol Aging 2024; 134:57-65. [PMID: 37992545 DOI: 10.1016/j.neurobiolaging.2023.10.008] [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: 02/22/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 11/24/2023]
Abstract
In 36 normal controls (NC), 37 patients with Alzheimer's disease (AD) without parkinsonism (ADP-), 31 AD with parkinsonism (ADP+), and 40 AD with dementia with Lewy bodies (ADDLB), dual-phase dopamine transporter (DAT) positron emission tomography (PET) were performed to evaluate the diagnostic performance of DAT and early-to-delayed uptake ratios (E/Ds) in the anterior caudate (AC), posterior caudate (PC), anterior putamen (AP), posterior putamen (PP), and substantia nigra (SN) to differentiate ADP+/ADDLB from NC, and their effects on parkinsonism and cognition. DAT-SN and E/D-PP showed higher accuracies to differentiate ADP+/ADDLB from NC than DAT-PP. Among AD patients, lower DAT in the putamen and PC and higher E/Ds in the striatum were associated with severe parkinsonism, while higher E/Ds in the putamen, PC, and SN were associated with executive dysfunction. Our results suggest that decreased DAT-SN and increased E/D-PP could be biomarkers differentiating ADP+/ADDLB from pure AD and controls. Meanwhile, increased E/Ds in the putamen could reflect the severity of DLB presenting with parkinsonism and executive dysfunction among AD patients.
Collapse
Affiliation(s)
- Sungwoo Kang
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seun Jeon
- Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young-Gun Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byoung Seok Ye
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
19
|
Visser M, O'Brien JT, Mak E. In vivo imaging of synaptic density in neurodegenerative disorders with positron emission tomography: A systematic review. Ageing Res Rev 2024; 94:102197. [PMID: 38266660 DOI: 10.1016/j.arr.2024.102197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Positron emission tomography (PET) with radiotracers that bind to synaptic vesicle glycoprotein 2 A (SV2A) enables quantification of synaptic density in the living human brain. Assessing the regional distribution and severity of synaptic density loss will contribute to our understanding of the pathological processes that precede atrophy in neurodegeneration. In this systematic review, we provide a discussion of in vivo SV2A PET imaging research for quantitative assessment of synaptic density in various dementia conditions: amnestic Mild Cognitive Impairment and Alzheimer's disease, Frontotemporal dementia, Progressive supranuclear palsy and Corticobasal degeneration, Parkinson's disease and Dementia with Lewy bodies, Huntington's disease, and Spinocerebellar Ataxia. We discuss the main findings concerning group differences and clinical-cognitive correlations, and explore relations between SV2A PET and other markers of pathology. Additionally, we touch upon synaptic density in healthy ageing and outcomes of radiotracer validation studies. Studies were identified on PubMed and Embase between 2018 and 2023; last searched on the 3rd of July 2023. A total of 36 studies were included, comprising 5 on normal ageing, 21 clinical studies, and 10 validation studies. Extracted study characteristics were participant details, methodological aspects, and critical findings. In summary, the small but growing literature on in vivo SV2A PET has revealed different spatial patterns of synaptic density loss among various neurodegenerative disorders that correlate with cognitive functioning, supporting the potential role of SV2A PET imaging for differential diagnosis. SV2A PET imaging shows tremendous capability to provide novel insights into the aetiology of neurodegenerative disorders and great promise as a biomarker for synaptic density reduction. Novel directions for future synaptic density research are proposed, including (a) longitudinal imaging in larger patient cohorts of preclinical dementias, (b) multi-modal mapping of synaptic density loss onto other pathological processes, and (c) monitoring therapeutic responses and assessing drug efficacy in clinical trials.
Collapse
Affiliation(s)
- Malouke Visser
- Department of Psychiatry, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, United Kingdom; Neuropsychology and Rehabilitation Psychology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - John T O'Brien
- Department of Psychiatry, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, United Kingdom
| | - Elijah Mak
- Department of Psychiatry, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, United Kingdom.
| |
Collapse
|
20
|
Holmes S, Tinaz S. Neuroimaging Biomarkers in Parkinson's Disease. ADVANCES IN NEUROBIOLOGY 2024; 40:617-663. [PMID: 39562459 DOI: 10.1007/978-3-031-69491-2_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Idiopathic Parkinson's disease (PD) is a neurodegenerative disorder that affects multiple systems in the body and is characterized by a variety of motor and non-motor (e.g., psychiatric, autonomic) symptoms. As the fastest growing neurological disorder expected to affect over 12 million people globally by 2040 (Dorsey, Bloem JAMA Neurol 75(1):9-10. https://doi.org/10.1001/jamaneurol.2017.3299 . PMID: 29131880, 2018), PD poses an enormous individual and public health burden. Currently, there are no therapies that can slow down the disease progression in PD, and existing therapies are limited to symptomatic treatment. Importantly, people in the prodromal phase who are at high risk of developing PD can now be identified, which makes disease prevention an achievable goal. An in-depth understanding of the pathological processes in PD is crucial for prevention and treatment development. Advanced multimodal neuroimaging techniques provide unique biomarkers that can further our understanding of PD at multiple levels ranging from neurotransmitters to neural networks. These neuroimaging biomarkers also have value in clinical application, for example, in the differential diagnosis of PD. As the field continues to advance, neuroimaging biomarkers are expected to become more specific, more widely accessible, and can be readily incorporated into translational research for treatment development in PD.
Collapse
Affiliation(s)
- Sophie Holmes
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Sule Tinaz
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
21
|
Millevert C, Vidas-Guscic N, Vanherp L, Jonckers E, Verhoye M, Staelens S, Bertoglio D, Weckhuysen S. Resting-State Functional MRI and PET Imaging as Noninvasive Tools to Study (Ab)Normal Neurodevelopment in Humans and Rodents. J Neurosci 2023; 43:8275-8293. [PMID: 38073598 PMCID: PMC10711730 DOI: 10.1523/jneurosci.1043-23.2023] [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: 09/18/2023] [Revised: 06/09/2023] [Accepted: 09/13/2023] [Indexed: 12/18/2023] Open
Abstract
Neurodevelopmental disorders (NDDs) are a group of complex neurologic and psychiatric disorders. Functional and molecular imaging techniques, such as resting-state functional magnetic resonance imaging (rs-fMRI) and positron emission tomography (PET), can be used to measure network activity noninvasively and longitudinally during maturation in both humans and rodent models. Here, we review the current knowledge on rs-fMRI and PET biomarkers in the study of normal and abnormal neurodevelopment, including intellectual disability (ID; with/without epilepsy), autism spectrum disorder (ASD), and attention deficit hyperactivity disorder (ADHD), in humans and rodent models from birth until adulthood, and evaluate the cross-species translational value of the imaging biomarkers. To date, only a few isolated studies have used rs-fMRI or PET to study (abnormal) neurodevelopment in rodents during infancy, the critical period of neurodevelopment. Further work to explore the feasibility of performing functional imaging studies in infant rodent models is essential, as rs-fMRI and PET imaging in transgenic rodent models of NDDs are powerful techniques for studying disease pathogenesis, developing noninvasive preclinical imaging biomarkers of neurodevelopmental dysfunction, and evaluating treatment-response in disease-specific models.
Collapse
Affiliation(s)
- Charissa Millevert
- Applied & Translational Neurogenomics Group, Vlaams Instituut voor Biotechnology (VIB) Center for Molecular Neurology, VIB, Antwerp 2610, Belgium
- Department of Neurology, University Hospital of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Nicholas Vidas-Guscic
- Bio-Imaging Lab, University of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Liesbeth Vanherp
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Elisabeth Jonckers
- Bio-Imaging Lab, University of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Marleen Verhoye
- Bio-Imaging Lab, University of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Daniele Bertoglio
- Bio-Imaging Lab, University of Antwerp, Antwerp 2610, Belgium
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Sarah Weckhuysen
- Applied & Translational Neurogenomics Group, Vlaams Instituut voor Biotechnology (VIB) Center for Molecular Neurology, VIB, Antwerp 2610, Belgium
- Department of Neurology, University Hospital of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
- Translational Neurosciences, Faculty of Medicine and Health Science, University of Antwerp, Antwerp 2610, Belgium
| |
Collapse
|
22
|
Vijiaratnam N, Foltynie T. How should we be using biomarkers in trials of disease modification in Parkinson's disease? Brain 2023; 146:4845-4869. [PMID: 37536279 PMCID: PMC10690028 DOI: 10.1093/brain/awad265] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/18/2023] [Accepted: 07/22/2023] [Indexed: 08/05/2023] Open
Abstract
The recent validation of the α-synuclein seed amplification assay as a biomarker with high sensitivity and specificity for the diagnosis of Parkinson's disease has formed the backbone for a proposed staging system for incorporation in Parkinson's disease clinical studies and trials. The routine use of this biomarker should greatly aid in the accuracy of diagnosis during recruitment of Parkinson's disease patients into trials (as distinct from patients with non-Parkinson's disease parkinsonism or non-Parkinson's disease tremors). There remain, however, further challenges in the pursuit of biomarkers for clinical trials of disease modifying agents in Parkinson's disease, namely: optimizing the distinction between different α-synucleinopathies; the selection of subgroups most likely to benefit from a candidate disease modifying agent; a sensitive means of confirming target engagement; and the early prediction of longer-term clinical benefit. For example, levels of CSF proteins such as the lysosomal enzyme β-glucocerebrosidase may assist in prognostication or allow enrichment of appropriate patients into disease modifying trials of agents with this enzyme as the target; the presence of coexisting Alzheimer's disease-like pathology (detectable through CSF levels of amyloid-β42 and tau) can predict subsequent cognitive decline; imaging techniques such as free-water or neuromelanin MRI may objectively track decline in Parkinson's disease even in its later stages. The exploitation of additional biomarkers to the α-synuclein seed amplification assay will, therefore, greatly add to our ability to plan trials and assess the disease modifying properties of interventions. The choice of which biomarker(s) to use in the context of disease modifying clinical trials will depend on the intervention, the stage (at risk, premotor, motor, complex) of the population recruited and the aims of the trial. The progress already made lends hope that panels of fluid biomarkers in tandem with structural or functional imaging may provide sensitive and objective methods of confirming that an intervention is modifying a key pathophysiological process of Parkinson's disease. However, correlation with clinical progression does not necessarily equate to causation, and the ongoing validation of quantitative biomarkers will depend on insightful clinical-genetic-pathophysiological comparisons incorporating longitudinal biomarker changes from those at genetic risk with evidence of onset of the pathophysiology and those at each stage of manifest clinical Parkinson's disease.
Collapse
Affiliation(s)
- Nirosen Vijiaratnam
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Thomas Foltynie
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| |
Collapse
|
23
|
Van Cauwenberge MGA, Delva A, Vande Casteele T, Laroy M, Radwan A, Vansteelandt K, Van den Stock J, Bouckaert F, Van Laere K, Emsell L, Vandenberghe W, Vandenbulcke M. Mild Motor Signs in Healthy Aging Are Associated with Lower Synaptic Density in the Brain. Mov Disord 2023; 38:1786-1794. [PMID: 37574924 DOI: 10.1002/mds.29570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/17/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
OBJECTIVE To investigate whether mild motor signs (MMS) in old age correlate with synaptic density in the brain. BACKGROUND Normal aging is associated with a decline in movement quality and quantity, commonly termed "mild parkinsonian signs" or more recently MMS. Whether MMS stem from global brain aging or pathology within motor circuits remains unresolved. The synaptic vesicle glycoprotein 2A positron emission tomography (PET) ligand 11 C-UCB-J allows the investigation of brain-motor associations at the synaptic level in vivo. METHOD Fifty-eight healthy older adults (≥50 years) were included from two monocentric control cohorts. Brain magnetic resonance imaging and 11 C-UCB-J PET data were available in 54 participants. 11 C-UCB-J PET binding was quantified by standardized uptake value ratio (SUVR) values in grey matter (GM) volumes of interest (VOIs): caudate, putamen, globus pallidus, substantia nigra, thalamus, cerebellum, and the frontal, parietal, temporal, and occipital cortex. Multiple linear regression analyses were performed with Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS) part III score measuring MMS as the dependent variable and mean SUVR values in each VOI as the independent variable with age, Fazekas score (white matter lesion [WML] load), VOI and cohort as covariates. RESULTS Participants (68 ± 7.5 years; 52% female) had an average MDS-UPDRS part III score of 3.3 ± 2.8. The MDS-UPDRS part III score was inversely associated with synaptic density, independently of WML load or GM volume, in the caudate, substantia nigra, thalamus, cerebellum, and parietal, occipital, temporal cortex. Cohen's f2 showed moderate effect sizes for subcortical (range, 0.30-0.35), cortical (0.28-0.35) and cerebellar VOIs (0.31). CONCLUSION MMS in healthy aging are associated with lower synaptic density throughout the brain. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Margot G A Van Cauwenberge
- Department of Neurosciences, Neuropsychiatry, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Aline Delva
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
- Department of Neurosciences, Laboratory for Parkinson Research, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Thomas Vande Casteele
- Department of Neurosciences, Neuropsychiatry, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Maarten Laroy
- Department of Neurosciences, Neuropsychiatry, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Ahmed Radwan
- Department of Imaging and Pathology, Translational MRI, KU Leuven, Leuven, Belgium
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | - Kristof Vansteelandt
- Department of Neurosciences, Neuropsychiatry, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Jan Van den Stock
- Department of Neurosciences, Neuropsychiatry, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Filip Bouckaert
- Department of Neurosciences, Neuropsychiatry, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- Division of Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, Leuven, Belgium
| | - Louise Emsell
- Department of Neurosciences, Neuropsychiatry, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
- Department of Imaging and Pathology, Translational MRI, KU Leuven, Leuven, Belgium
| | - Wim Vandenberghe
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
- Department of Neurosciences, Laboratory for Parkinson Research, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Department of Neurosciences, Neuropsychiatry, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
| |
Collapse
|
24
|
Smart K, Uribe C, Desmond KL, Martin SL, Vasdev N, Strafella AP. Preliminary Assessment of Reference Region Quantification and Reduced Scanning Times for [ 18F]SynVesT-1 PET in Parkinson's Disease. Mol Imaging 2023; 2023:1855985. [PMID: 37622164 PMCID: PMC10445483 DOI: 10.1155/2023/1855985] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/02/2023] [Accepted: 07/26/2023] [Indexed: 08/26/2023] Open
Abstract
Synaptic density in the central nervous system can be measured in vivo using PET with [18F]SynVesT-1. While [18F]SynVesT-1 has been proven to be a powerful radiopharmaceutical for PET imaging of neurodegenerative disorders such as Parkinson's disease (PD), its currently validated acquisition and quantification protocols are invasive and technically challenging in these populations due to the arterial sampling and relatively long scanning times. The objectives of this work were to evaluate a noninvasive (reference tissue) quantification method for [18F]SynVesT-1 in PD patients and to determine the minimum scan time necessary for accurate quantification. [18F]SynVesT-1 PET scans were acquired in 5 patients with PD and 3 healthy control subjects for 120 min with arterial blood sampling. Quantification was performed using the one-tissue compartment model (1TCM) with arterial input function, as well as with the simplified reference tissue model (SRTM) to estimate binding potential (BPND) using centrum semiovale (CS) as a reference region. The SRTM2 method was used with k2' fixed to either a sample average value (0.037 min-1) or a value estimated first through coupled fitting across regions for each participant. Direct SRTM estimation and the Logan reference region graphical method were also evaluated. There were no significant group differences in CS volume, radiotracer uptake, or efflux (ps > 0.47). Each fitting method produced BPND estimates in close agreement with those derived from the 1TCM (subject R2s > 0.98, bias < 10%), with no difference in bias between the control and PD groups. With SRTM2, BPND estimates from truncated scan data as short as 80 min produced values in excellent agreement with the data from the full 120 min scans (bias < 6%). While these are preliminary results from a small sample of patients with PD (n = 5), this work suggests that accurate synaptic density quantification may be performed without blood sampling and with scan time under 90 minutes. If further validated, these simplified procedures for [18F]SynVesT-1 PET quantification can facilitate its application as a clinical research imaging technology and allow for larger study samples and include a broader scope of patients including those with neurodegenerative diseases.
Collapse
Affiliation(s)
- Kelly Smart
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, Canada M5T 1R8
- Department of Psychiatry, University of Toronto, 250 College St., Toronto, ON, Canada M5T 1R8
| | - Carme Uribe
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, Canada M5T 1R8
- Unitat de Psicologia Medica, Departament de Medicina, Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
| | - Kimberly L. Desmond
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, Canada M5T 1R8
- Department of Psychiatry, University of Toronto, 250 College St., Toronto, ON, Canada M5T 1R8
| | - Sarah L. Martin
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, Canada M5T 1R8
| | - Neil Vasdev
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, Canada M5T 1R8
- Department of Psychiatry, University of Toronto, 250 College St., Toronto, ON, Canada M5T 1R8
| | - Antonio P. Strafella
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, Canada M5T 1R8
- Edmond J. Safra Parkinson Disease Program, Neurology Division, Toronto Western Hospital & Krembil Brain Institute, University Health Network, University of Toronto, 399 Bathurst Street, Toronto, ON, Canada M5T 2S8
| |
Collapse
|
25
|
O'Dell RS, Higgins-Chen A, Gupta D, Chen MK, Naganawa M, Toyonaga T, Lu Y, Ni G, Chupak A, Zhao W, Salardini E, Nabulsi NB, Huang Y, Arnsten AFT, Carson RE, van Dyck CH, Mecca AP. Principal component analysis of synaptic density measured with [ 11C]UCB-J PET in early Alzheimer's disease. Neuroimage Clin 2023; 39:103457. [PMID: 37422964 PMCID: PMC10338149 DOI: 10.1016/j.nicl.2023.103457] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 05/01/2023] [Accepted: 06/19/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND Synaptic loss is considered an early pathological event and major structural correlate of cognitive impairment in Alzheimer's disease (AD). We used principal component analysis (PCA) to identify regional patterns of covariance in synaptic density using [11C]UCB-J PET and assessed the association between principal components (PC) subject scores with cognitive performance. METHODS [11C]UCB-J binding was measured in 45 amyloid + participants with AD and 19 amyloid- cognitively normal participants aged 55-85. A validated neuropsychological battery assessed performance across five cognitive domains. PCA was applied to the pooled sample using distribution volume ratios (DVR) standardized (z-scored) by region from 42 bilateral regions of interest (ROI). RESULTS Parallel analysis determined three significant PCs explaining 70.2% of the total variance. PC1 was characterized by positive loadings with similar contributions across the majority of ROIs. PC2 was characterized by positive and negative loadings with strongest contributions from subcortical and parietooccipital cortical regions, respectively, while PC3 was characterized by positive and negative loadings with strongest contributions from rostral and caudal cortical regions, respectively. Within the AD group, PC1 subject scores were positively correlated with performance across all cognitive domains (Pearson r = 0.24-0.40, P = 0.06-0.006), PC2 subject scores were inversely correlated with age (Pearson r = -0.45, P = 0.002) and PC3 subject scores were significantly correlated with CDR-sb (Pearson r = 0.46, P = 0.04). No significant correlations were observed between cognitive performance and PC subject scores in CN participants. CONCLUSIONS This data-driven approach defined specific spatial patterns of synaptic density correlated with unique participant characteristics within the AD group. Our findings reinforce synaptic density as a robust biomarker of disease presence and severity in the early stages of AD.
Collapse
Affiliation(s)
- Ryan S O'Dell
- Alzheimer's Disease Research Unit, Yale University School of Medicine, One Church Street, 8(th) Floor, New Haven, CT 06510, USA; Department of Psychiatry, Yale University School of Medicine, 300 George Street, New Haven, CT 06510, USA.
| | - Albert Higgins-Chen
- Alzheimer's Disease Research Unit, Yale University School of Medicine, One Church Street, 8(th) Floor, New Haven, CT 06510, USA; Department of Psychiatry, Yale University School of Medicine, 300 George Street, New Haven, CT 06510, USA; Pain Research, Informatics, Multi-morbidities, and Education Center, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Dhruva Gupta
- Alzheimer's Disease Research Unit, Yale University School of Medicine, One Church Street, 8(th) Floor, New Haven, CT 06510, USA
| | - Ming-Kai Chen
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, P.O. Box 208048, New Haven, CT 06520, USA
| | - Mika Naganawa
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, P.O. Box 208048, New Haven, CT 06520, USA
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, P.O. Box 208048, New Haven, CT 06520, USA
| | - Yihuan Lu
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, P.O. Box 208048, New Haven, CT 06520, USA
| | - Gessica Ni
- Alzheimer's Disease Research Unit, Yale University School of Medicine, One Church Street, 8(th) Floor, New Haven, CT 06510, USA; Department of Psychiatry, Yale University School of Medicine, 300 George Street, New Haven, CT 06510, USA
| | - Anna Chupak
- Alzheimer's Disease Research Unit, Yale University School of Medicine, One Church Street, 8(th) Floor, New Haven, CT 06510, USA; Department of Psychiatry, Yale University School of Medicine, 300 George Street, New Haven, CT 06510, USA
| | - Wenzhen Zhao
- Alzheimer's Disease Research Unit, Yale University School of Medicine, One Church Street, 8(th) Floor, New Haven, CT 06510, USA; Department of Psychiatry, Yale University School of Medicine, 300 George Street, New Haven, CT 06510, USA
| | - Elaheh Salardini
- Alzheimer's Disease Research Unit, Yale University School of Medicine, One Church Street, 8(th) Floor, New Haven, CT 06510, USA; Department of Psychiatry, Yale University School of Medicine, 300 George Street, New Haven, CT 06510, USA
| | - Nabeel B Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, P.O. Box 208048, New Haven, CT 06520, USA
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, P.O. Box 208048, New Haven, CT 06520, USA
| | - Amy F T Arnsten
- Department of Neuroscience, Yale University School of Medicine, P.O. Box 208001, New Haven, CT 06520, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, P.O. Box 208048, New Haven, CT 06520, USA
| | - Christopher H van Dyck
- Alzheimer's Disease Research Unit, Yale University School of Medicine, One Church Street, 8(th) Floor, New Haven, CT 06510, USA; Department of Psychiatry, Yale University School of Medicine, 300 George Street, New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, P.O. Box 208001, New Haven, CT 06520, USA; Department of Neurology, Yale University School of Medicine, P.O. Box 208018, New Haven, CT 06520, USA
| | - Adam P Mecca
- Alzheimer's Disease Research Unit, Yale University School of Medicine, One Church Street, 8(th) Floor, New Haven, CT 06510, USA; Department of Psychiatry, Yale University School of Medicine, 300 George Street, New Haven, CT 06510, USA.
| |
Collapse
|
26
|
Xiong M, Roshanbin S, Sehlin D, Hansen HD, Knudsen GM, Rokka J, Eriksson J, Syvänen S. Synaptic density in aging mice measured by [ 18F]SynVesT-1 PET. Neuroimage 2023:120230. [PMID: 37355199 DOI: 10.1016/j.neuroimage.2023.120230] [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: 12/03/2022] [Revised: 05/03/2023] [Accepted: 06/14/2023] [Indexed: 06/26/2023] Open
Abstract
Synaptic alterations in certain brain structures are related to cognitive decline in neurodegeneration and in aging. Synaptic loss in many neurodegenerative diseases can be visualized by positron emission tomography (PET) imaging of synaptic vesicle glycoprotein 2A (SV2A). However, the use of SV2A PET for studying synaptic changes during aging is not particularly explored. Thus, in the present study, PET ligand [18F]SynVesT-1, which binds to SV2A, was used to investigate synaptic density at different ages in healthy mice. Wild type C57BL/6 mice divided into three age groups (4-5 months (n = 7), 12-14 months (n = 11), 17-19 months (n = 7)) were PET scanned with [18F]SynVesT-1. Brain retention of [18F]SynVesT-1 expressed as the volume of distribution (VIDIF) was calculated using an image-derived input function. Estimates of VIDIF were derived using either a one-tissue compartment model (1TCM), a two-tissue compartment model (2TCM), or the Logan plot with blood input to find the best-fit model for [18F]SynVesT-1. After the PET scans, tissue sections were immunostained for the detection of SV2A and neuronal markers. We found that [18F]SynVesT-1 data acquired 60 min post intravenously injection and analyzed with 1TCM described the brain pharmacokinetics of the radioligand in mice well. [18F]SynVesT-1 brain retention was lower in the oldest group of mice, indicating a decrease in synaptic density in this age group. However, no gradual age-dependent decrease in synaptic density at a region-specific level was observed. Immunostaining indicated that SV2A expression and neuron numbers were similar across all three age groups. In general, these data obtained in healthy aging mice are consistent with previous findings in humans where synaptic density appeared stable during aging up to a certain age, after which a small decrease is observed.
Collapse
Affiliation(s)
- Mengfei Xiong
- Molecular Geriatrics, Department of Public Health and Caring Sciences, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Sahar Roshanbin
- Molecular Geriatrics, Department of Public Health and Caring Sciences, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Dag Sehlin
- Molecular Geriatrics, Department of Public Health and Caring Sciences, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Hanne D Hansen
- Neurobiology Research Unit, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark
| | - Gitte M Knudsen
- Neurobiology Research Unit, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Johanna Rokka
- Molecular Geriatrics, Department of Public Health and Caring Sciences, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Jonas Eriksson
- PET Centre, Uppsala University Hospital, SE-751 85 Uppsala, Sweden; Department of Medicinal Chemistry, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Stina Syvänen
- Molecular Geriatrics, Department of Public Health and Caring Sciences, Uppsala University, SE-751 85 Uppsala, Sweden.
| |
Collapse
|
27
|
Chen Z, Liao G, Wan N, He Z, Chen D, Tang Z, Long Z, Zou G, Peng L, Wan L, Wang C, Peng H, Shi Y, Tang Y, Li J, Li Y, Long T, Hou X, He L, Qiu R, Chen D, Wang J, Guo J, Shen L, Huang Y, Ashizawa T, Klockgether T, Tang B, Zhou M, Hu S, Jiang H. Synaptic Loss in Spinocerebellar Ataxia Type 3 Revealed by SV2A Positron Emission Tomography. Mov Disord 2023; 38:978-989. [PMID: 37023261 DOI: 10.1002/mds.29395] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/16/2023] [Accepted: 03/15/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Severe reduced synaptic density was observed in spinocerebellar ataxia (SCA) in postmortem neuropathology, but in vivo assessment of synaptic loss remains challenging. OBJECTIVE SPINOCEREBELLAR ATAXIA TYPE 3: The objective of this study was to assess in vivo synaptic loss and its clinical correlates in spinocerebellar ataxia type 3 (SCA3) patients by synaptic vesicle glycoprotein 2A (SV2A)-positron emission tomography (PET) imaging. METHODS We recruited 74 SCA3 individuals including preataxic and ataxic stages and divided into two cohorts. All participants received SV2A-PET imaging using 18 F-SynVesT-1 for synaptic density assessment. Specifically, cohort 1 received standard PET procedure and quantified neurofilament light chain (NfL), and cohort 2 received simplified PET procedure for exploratory purpose. Bivariate correlation was performed between synaptic loss and clinical as well as genetic assessments. RESULTS In cohort 1, significant reductions of synaptic density were observed in cerebellum and brainstem in SCA3 ataxia stage compared to preataxic stage and controls. Vermis was found significantly involved in preataxic stage compared to controls. Receiver operating characteristic (ROC) curves highlighted SV2A of vermis, pons, and medulla differentiating preataxic stage from ataxic stage, and SV2A combined with NfL improved the performance. Synaptic density was significantly negatively correlated with disease severity in cerebellum and brainstem (International Co-operative Ataxia Rating Scale: ρ ranging from -0.467 to -0.667, P ≤ 0.002; Scale of Assessment and Rating of Ataxia: ρ ranging from -0.465 to -0.586, P ≤ 0.002). SV2A reduction tendency of cerebellum and brainstem identified in cohort 1 was observed in cohort 2 with simplified PET procedure. CONCLUSIONS We first identified in vivo synaptic loss was related to disease severity of SCA3, suggesting SV2A PET could be a promising clinical biomarker for disease progression of SCA3. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Zhao Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| | - Guang Liao
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Na Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhiyou He
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Daji Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhichao Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhe Long
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guangdong Zou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Linliu Peng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Linlin Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chunrong Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huirong Peng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuting Shi
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yongxiang Tang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Li
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yulai Li
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tingting Long
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xuan Hou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lang He
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Rong Qiu
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China
| | - Dengming Chen
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Junling Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| | - Yiyun Huang
- Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Tetsuo Ashizawa
- Neuroscience Research Program, Department of Neurology, Houston Methodist Research Institute, Weil Cornell Medical College, Houston, Texas, USA
| | - Thomas Klockgether
- Department of Neurology, University Hospital of Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| | - Ming Zhou
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuo Hu
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- School of Basic Medical Science, Central South University, Changsha, Hunan, China
- National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, Hunan, China
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
28
|
Bademosi AT, Decet M, Kuenen S, Calatayud C, Swerts J, Gallego SF, Schoovaerts N, Karamanou S, Louros N, Martin E, Sibarita JB, Vints K, Gounko NV, Meunier FA, Economou A, Versées W, Rousseau F, Schymkowitz J, Soukup SF, Verstreken P. EndophilinA-dependent coupling between activity-induced calcium influx and synaptic autophagy is disrupted by a Parkinson-risk mutation. Neuron 2023; 111:1402-1422.e13. [PMID: 36827984 PMCID: PMC10166451 DOI: 10.1016/j.neuron.2023.02.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 11/09/2022] [Accepted: 01/31/2023] [Indexed: 02/26/2023]
Abstract
Neuronal activity causes use-dependent decline in protein function. However, it is unclear how this is coupled to local quality control mechanisms. We show in Drosophila that the endocytic protein Endophilin-A (EndoA) connects activity-induced calcium influx to synaptic autophagy and neuronal survival in a Parkinson disease-relevant fashion. Mutations in the disordered loop, including a Parkinson disease-risk mutation, render EndoA insensitive to neuronal stimulation and affect protein dynamics: when EndoA is more flexible, its mobility in membrane nanodomains increases, making it available for autophagosome formation. Conversely, when EndoA is more rigid, its mobility reduces, blocking stimulation-induced autophagy. Balanced stimulation-induced autophagy is required for dopagminergic neuron survival, and a variant in the human ENDOA1 disordered loop conferring risk to Parkinson disease also blocks nanodomain protein mobility and autophagy both in vivo and in human-induced dopaminergic neurons. Thus, we reveal a mechanism that neurons use to connect neuronal activity to local autophagy and that is critical for neuronal survival.
Collapse
Affiliation(s)
- Adekunle T Bademosi
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven 3000, Belgium; Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia Campus, Brisbane, QLD 4072, Australia
| | - Marianna Decet
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven 3000, Belgium
| | - Sabine Kuenen
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven 3000, Belgium
| | - Carles Calatayud
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven 3000, Belgium
| | - Jef Swerts
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven 3000, Belgium
| | - Sandra F Gallego
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven 3000, Belgium
| | - Nils Schoovaerts
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven 3000, Belgium
| | - Spyridoula Karamanou
- KU Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Leuven 3000, Belgium
| | - Nikolaos Louros
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
| | - Ella Martin
- VIB-VUB Center for Structural Biology, Brussels 1050, Belgium; Department of Structural Biology Brussels, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | - Jean-Baptiste Sibarita
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, F-33000 Bordeaux, France
| | - Katlijn Vints
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven 3000, Belgium; VIB Bio Core, KU Leuven, Leuven 3000, Belgium
| | - Natalia V Gounko
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven 3000, Belgium; VIB Bio Core, KU Leuven, Leuven 3000, Belgium
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia Campus, Brisbane, QLD 4072, Australia; School of Biomedical Sciences, The University of Queensland, St Lucia Campus, Brisbane, QLD 4072, Australia
| | - Anastassios Economou
- KU Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Leuven 3000, Belgium
| | - Wim Versées
- VIB-VUB Center for Structural Biology, Brussels 1050, Belgium; Department of Structural Biology Brussels, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | - Frederic Rousseau
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
| | - Joost Schymkowitz
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
| | | | - Patrik Verstreken
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven 3000, Belgium.
| |
Collapse
|
29
|
Lopresti BJ, Royse SK, Mathis CA, Tollefson SA, Narendran R. Beyond monoamines: I. Novel targets and radiotracers for Positron emission tomography imaging in psychiatric disorders. J Neurochem 2023; 164:364-400. [PMID: 35536762 DOI: 10.1111/jnc.15615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 10/18/2022]
Abstract
With the emergence of positron emission tomography (PET) in the late 1970s, psychiatry had access to a tool capable of non-invasive assessment of human brain function. Early applications in psychiatry focused on identifying characteristic brain blood flow and metabolic derangements using radiotracers such as [15 O]H2 O and [18 F]FDG. Despite the success of these techniques, it became apparent that more specific probes were needed to understand the neurochemical bases of psychiatric disorders. The first neurochemical PET imaging probes targeted sites of action of neuroleptic (dopamine D2 receptors) and psychoactive (serotonin receptors) drugs. Based on the centrality of monoamine dysfunction in psychiatric disorders and the measured success of monoamine-enhancing drugs in treating them, the next 30 years witnessed the development of an armamentarium of PET radiopharmaceuticals and imaging methodologies for studying monoamines. Continued development of monoamine-enhancing drugs over this time however was less successful, realizing only modest gains in efficacy and tolerability. As patent protection for many widely prescribed and profitable psychiatric drugs lapsed, drug development pipelines shifted away from monoamines in search of novel targets with the promises of improved efficacy, or abandoned altogether. Over this period, PET radiopharmaceutical development activities closely paralleled drug development priorities resulting in the development of new PET imaging agents for non-monoamine targets. Part one of this review will briefly survey novel PET imaging targets with relevance to the field of psychiatry, which include the metabotropic glutamate receptor type 5 (mGluR5), purinergic P2 X7 receptor, type 1 cannabinoid receptor (CB1 ), phosphodiesterase 10A (PDE10A), and describe radiotracers developed for these and other targets that have matured to human subject investigations. Current limitations of the targets and techniques will also be discussed.
Collapse
Affiliation(s)
- Brian J Lopresti
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sarah K Royse
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Chester A Mathis
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Savannah A Tollefson
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rajesh Narendran
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Departments of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
30
|
Real CC, Binda KH, Thomsen MB, Lillethorup TP, Brooks DJ, Landau AM. Selecting the Best Animal Model of Parkinson's Disease for Your Research Purpose: Insight from in vivo PET Imaging Studies. Curr Neuropharmacol 2023; 21:1241-1272. [PMID: 36797611 PMCID: PMC10286593 DOI: 10.2174/1570159x21666230216101659] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 02/18/2023] Open
Abstract
Parkinson's disease (PD) is a debilitating neurodegenerative multisystem disorder leading to motor and non-motor symptoms in millions of individuals. Despite intense research, there is still no cure, and early disease biomarkers are lacking. Animal models of PD have been inspired by basic elements of its pathogenesis, such as dopamine dysfunction, alpha-synuclein accumulation, neuroinflammation and disruption of protein degradation, and these have been crucial for a deeper understanding of the mechanisms of pathology, the identification of biomarkers, and evaluation of novel therapies. Imaging biomarkers are non-invasive tools to assess disease progression and response to therapies; their discovery and validation have been an active field of translational research. Here, we highlight different considerations of animal models of PD that can be applied to future research, in terms of their suitability to answer different research questions. We provide the reader with important considerations of the best choice of model to use based on the disease features of each model, including issues related to different species. In addition, positron emission tomography studies conducted in PD animal models in the last 5 years are presented. With a variety of different species, interventions and genetic information, the choice of the most appropriate model to answer research questions can be daunting, especially since no single model recapitulates all aspects of this complex disorder. Appropriate animal models in conjunction with in vivo molecular imaging tools, if selected properly, can be a powerful combination for the assessment of novel therapies and developing tools for early diagnosis.
Collapse
Affiliation(s)
- Caroline Cristiano Real
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Karina Henrique Binda
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Majken Borup Thomsen
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Thea Pinholt Lillethorup
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - David James Brooks
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Institute of Translational and Clinical Research, University of Newcastle, Upon Tyne, UK
| | - Anne Marlene Landau
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
31
|
Akkermans J, Zajicek F, Miranda A, Adhikari MH, Bertoglio D. Identification of pre-synaptic density networks using [ 11C]UCB-J PET imaging and ICA in mice. Neuroimage 2022; 264:119771. [PMID: 36436710 DOI: 10.1016/j.neuroimage.2022.119771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/28/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Synaptic vesicle glycoprotein 2A (SV2A) is a vesicle glycoprotein involved in neurotransmitter release. SV2A is located on the pre-synaptic terminals of neurons and visualized using the radioligand [11C]UCB-J and positron emission tomography (PET) imaging. Thus, SV2A PET imaging can provide a proxy for pre-synaptic density in health and disease. This study aims to apply independent component analysis (ICA) to SV2A PET data acquired in mice to identify pre-synaptic density networks (pSDNs), explore how ageing affects these pSDNs, and determine the impact of a neurological disorder on these networks. METHODS We used [11C]UCB-J PET imaging data (n = 135) available at different ages (3, 7, 10, and 16 months) in wild-type (WT) C57BL/6J mice and in diseased mice (mouse model of Huntington's disease, HD) with reported synaptic deficits. First, ICA was performed on a healthy dataset after it was split into two equal-sized samples (n = 36 each) and the analysis was repeated 50 times in different partitions. We tested different model orders (8, 12, and 16) and identified the pSDNs. Next, we investigated the effect of age on the loading weights of the identified pSDNs. Additionally, the identified pSDNs were compared to those of diseased mice to assess the impact of disease on each pSDNs. RESULTS Model order 12 resulted in the preferred choice to provide six reliable and reproducible independent components (ICs) as supported by the cluster-quality index (IQ) and regression coefficients (β) values. Temporal analysis showed age-related statistically significant changes on the loading weights in four ICs. ICA in an HD model revealed a statistically significant disease-related effect on the loading weights in several pSDNs in line with the progression of the disease. CONCLUSION This study validated the use of ICA on SV2A PET data acquired with [11C]UCB-J for the identification of cerebral pre-synaptic density networks in mice in a rigorous and reproducible manner. Furthermore, we showed that different pSDNs change with age and are affected in a disease condition. These findings highlight the potential value of ICA in understanding pre-synaptic density networks in the mouse brain.
Collapse
Affiliation(s)
- Jordy Akkermans
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Belgium
| | - Franziska Zajicek
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Belgium
| | - Alan Miranda
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Belgium
| | | | - Daniele Bertoglio
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Belgium; Bio-Imaging Lab, University of Antwerp, Belgium.
| |
Collapse
|
32
|
Du J, Zhou X, Liang Y, Zhao L, Dai C, Zhong Y, Liu H, Liu G, Mo L, Tan C, Liu X, Chen L. Levodopa responsiveness and white matter alterations in Parkinson's disease: A DTI-based study and brain network analysis: A cross-sectional study. Brain Behav 2022; 12:e2825. [PMID: 36423257 PMCID: PMC9759147 DOI: 10.1002/brb3.2825] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/24/2022] [Accepted: 11/01/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Patients with Parkinson's disease (PD) present various responsiveness to levodopa, but the cause of such differences in levodopa responsiveness is unclear. Previous studies related the damage of brain white matter (WM) to levodopa responsiveness in PD patients, but no study investigated the relationship between the structural brain network change in PD patients and their levodopa responsiveness. METHODS PD patients were recruited and evaluated using the Unified Parkinson's Disease Rating Scale (UPDRS). Each patient received a diffusion tensor imaging (DTI) scan and an acute levodopa challenge test. The improvement rate of UPDRS-III was calculated. PD patients were grouped into irresponsive group (improvement rate < 30%) and responsive group (improvement rate ≥ 30%). Tract-based spatial statistics (TBSS), deterministic tracing (DT), region of interest (ROI) analysis, and automatic fiber identification (AFQ) analyses were performed. The structural brain network was also constructed and the topological parameters were calculated. RESULTS Fifty-four PD patients were included. TBSS identified significant differences in fractional anisotropy (FA) values in the corpus callosum and other regions of the brain. DT and ROI analysis of the corpus callosum found a significant difference in FA between the two groups. Graph theory analysis showed statistical differences in global efficiency, local efficiency, and characteristic path length. CONCLUSION PD patients with poor responsiveness to levodopa had WM damage in multiple brain areas, especially the corpus callosum, which might cause disruption of information integration of the structural brain network.
Collapse
Affiliation(s)
- Juncong Du
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Xuan Zhou
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Yi Liang
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Lili Zhao
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Chengcheng Dai
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Yuke Zhong
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Hang Liu
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Guohui Liu
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Lijuan Mo
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Changhong Tan
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Xi Liu
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Lifen Chen
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| |
Collapse
|
33
|
Disentangling nigral and putaminal contribution to motor impairment and levodopa response in Parkinson's disease. NPJ Parkinsons Dis 2022; 8:132. [PMID: 36241644 PMCID: PMC9568583 DOI: 10.1038/s41531-022-00401-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 09/28/2022] [Indexed: 11/08/2022] Open
Abstract
The extent to which the degeneration of the substantia nigra (SN) and putamen each contribute to motor impairment in Parkinson's disease (PD) is unclear, as they are usually investigated using different imaging modalities. To examine the pathophysiological significance of the SN and putamen in both motor impairment and the levodopa response in PD using diffusion microstructure imaging (DMI). In this monocentric retrospective cross-sectional study, DMI parameters from 108 patients with PD and 35 healthy controls (HC) were analyzed using a voxel- and region-based approach. Linear models were applied to investigate the association between individual DMI parameters and Movement Disorder Society Unified Parkinson's Disease Rating Scale-Part 3 performance in ON- and OFF-states, as well as the levodopa response, controlling for age and sex. Voxel- and region-based group comparisons of DMI parameters between PD and HC revealed significant differences in the SN and putamen. In PD, a poorer MDS-UPDRS-III performance in the ON-state was associated with increased free fluid in the SN (b-weight = 65.79, p = 0.004) and putamen (b-weight = 86.00, p = 0.006), and contrariwise with the demise of cells in both structures. The levodopa response was inversely associated with free fluid both in the SN (b-weight = -83.61, p = 0.009) and putamen (b-weight = -176.56, p < 0.001). Interestingly, when the two structures were assessed together, the integrity of the putamen, but not the SN, served as a predictor for the levodopa response (b-weight = -158.03, p < 0.001). Structural alterations in the SN and putamen can be measured by diffusion microstructure imaging in PD. They are associated with poorer motor performance in the ON-state, as well as a reduced response to levodopa. While both nigral and putaminal integrity are required for good performance in the ON-state, it is putaminal integrity alone that determines the levodopa response. Therefore, the structural integrity of the putamen is crucial for the improvement of motor symptoms to dopaminergic medication, and might therefore serve as a promising biomarker for motor staging.
Collapse
|
34
|
Small soluble α-synuclein aggregates are the toxic species in Parkinson's disease. Nat Commun 2022; 13:5512. [PMID: 36127374 PMCID: PMC9489799 DOI: 10.1038/s41467-022-33252-6] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 09/08/2022] [Indexed: 11/08/2022] Open
Abstract
Soluble α-synuclein aggregates varying in size, structure, and morphology have been closely linked to neuronal death in Parkinson's disease. However, the heterogeneity of different co-existing aggregate species makes it hard to isolate and study their individual toxic properties. Here, we show a reliable non-perturbative method to separate a heterogeneous mixture of protein aggregates by size. We find that aggregates of wild-type α-synuclein smaller than 200 nm in length, formed during an in vitro aggregation reaction, cause inflammation and permeabilization of single-liposome membranes and that larger aggregates are less toxic. Studying soluble aggregates extracted from post-mortem human brains also reveals that these aggregates are similar in size and structure to the smaller aggregates formed in aggregation reactions in the test tube. Furthermore, we find that the soluble aggregates present in Parkinson's disease brains are smaller, largely less than 100 nm, and more inflammatory compared to the larger aggregates present in control brains. This study suggests that the small non-fibrillar α-synuclein aggregates are the critical species driving neuroinflammation and disease progression.
Collapse
|
35
|
Marner L, Korsholm K, Anderberg L, Lonsdale MN, Jensen MR, Brødsgaard E, Denholt CL, Gillings N, Law I, Friberg L. [ 18F]FE-PE2I PET is a feasible alternative to [ 123I]FP-CIT SPECT for dopamine transporter imaging in clinically uncertain parkinsonism. EJNMMI Res 2022; 12:56. [PMID: 36070114 PMCID: PMC9452620 DOI: 10.1186/s13550-022-00930-x] [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: 03/01/2022] [Accepted: 08/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Dopamine transporter (DAT) imaging of striatum is clinically used in Parkinson's disease (PD) and neurodegenerative parkinsonian syndromes (PS) especially in the early disease stages. The aim of the present study was to evaluate the diagnostic performance of the recently developed tracer for DAT imaging [18F]FE-PE2I PET/CT to the reference standard [123I]FP-CIT SPECT. METHODS Ninety-eight unselected patients referred for DAT imaging were included prospectively and consecutively and evaluated with [18F]FE-PE2I PET/CT and [123I]FP-CIT SPECT on two separate days. PET and SPECT scans were categorized independently by two blinded expert readers as either normal, vascular changes, or mixed. Semiquantitative values were obtained for each modality and compared regarding effect size using Glass' delta. RESULTS Fifty-six of the [123I]FP-CIT SPECT scans were considered abnormal (52 caused by PS, 4 by infarctions). Using [18F]FE-PE2I PET/CT, 95 of the 98 patients were categorized identically to SPECT as PS or non-PS with a sensitivity of 0.94 [0.84-0.99] and a specificity of 1.00 [0.92-1.00]. Inter-reader agreement for [18F]FE-PE2I PET with a kappa of 0.97 [0.89-1.00] was comparable to the agreement for [123I]FP-CIT SPECT of 0.96 [0.76-1.00]. Semiquantitative values for short 10-min reconstructions of [18F]FE-PE2I PET/CT were comparable to longer reconstructions. The effect size for putamen/caudate nucleus ratio was significantly increased using PET compared to SPECT. CONCLUSIONS The high correspondence of [18F]FE-PE2I PET compared to reference standard [123I]FP-CIT SPECT establishes [18F]FE-PE2I PET as a feasible PET tracer for clinical use with favourable scan logistics.
Collapse
Affiliation(s)
- Lisbeth Marner
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Bispebjerg, Bispebjerg Bakke 23, Copenhagen, Denmark. .,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| | - Kirsten Korsholm
- grid.411702.10000 0000 9350 8874Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Bispebjerg, Bispebjerg Bakke 23, Copenhagen, Denmark ,grid.475435.4Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Lasse Anderberg
- grid.475435.4Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Markus N. Lonsdale
- grid.411702.10000 0000 9350 8874Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Bispebjerg, Bispebjerg Bakke 23, Copenhagen, Denmark
| | - Mads Radmer Jensen
- grid.411702.10000 0000 9350 8874Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Bispebjerg, Bispebjerg Bakke 23, Copenhagen, Denmark
| | - Eva Brødsgaard
- grid.411702.10000 0000 9350 8874Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Bispebjerg, Bispebjerg Bakke 23, Copenhagen, Denmark
| | - Charlotte L. Denholt
- grid.475435.4Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Nic Gillings
- grid.475435.4Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Ian Law
- grid.5254.60000 0001 0674 042XDepartment of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark ,grid.475435.4Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Lars Friberg
- grid.411702.10000 0000 9350 8874Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Bispebjerg, Bispebjerg Bakke 23, Copenhagen, Denmark
| |
Collapse
|
36
|
Prange S, Theis H, Banwinkler M, van Eimeren T. Molecular Imaging in Parkinsonian Disorders—What’s New and Hot? Brain Sci 2022; 12:brainsci12091146. [PMID: 36138882 PMCID: PMC9496752 DOI: 10.3390/brainsci12091146] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 12/02/2022] Open
Abstract
Highlights Abstract Neurodegenerative parkinsonian disorders are characterized by a great diversity of clinical symptoms and underlying neuropathology, yet differential diagnosis during lifetime remains probabilistic. Molecular imaging is a powerful method to detect pathological changes in vivo on a cellular and molecular level with high specificity. Thereby, molecular imaging enables to investigate functional changes and pathological hallmarks in neurodegenerative disorders, thus allowing to better differentiate between different forms of degenerative parkinsonism, improve the accuracy of the clinical diagnosis and disentangle the pathophysiology of disease-related symptoms. The past decade led to significant progress in the field of molecular imaging, including the development of multiple new and promising radioactive tracers for single photon emission computed tomography (SPECT) and positron emission tomography (PET) as well as novel analytical methods. Here, we review the most recent advances in molecular imaging for the diagnosis, prognosis, and mechanistic understanding of parkinsonian disorders. First, advances in imaging of neurotransmission abnormalities, metabolism, synaptic density, inflammation, and pathological protein aggregation are reviewed, highlighting our renewed understanding regarding the multiplicity of neurodegenerative processes involved in parkinsonian disorders. Consequently, we review the role of molecular imaging in the context of disease-modifying interventions to follow neurodegeneration, ensure stratification, and target engagement in clinical trials.
Collapse
Affiliation(s)
- Stéphane Prange
- Multimodal Neuroimaging Group, Department of Nuclear Medicine, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany
- Institut des Sciences Cognitives Marc Jeannerod, CNRS, UMR 5229, Université de Lyon, 69675 Bron, France
- Correspondence: (S.P.); (T.v.E.); Tel.: +49-221-47882843 (T.v.E.)
| | - Hendrik Theis
- Multimodal Neuroimaging Group, Department of Nuclear Medicine, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany
- Department of Neurology, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany
| | - Magdalena Banwinkler
- Multimodal Neuroimaging Group, Department of Nuclear Medicine, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany
| | - Thilo van Eimeren
- Multimodal Neuroimaging Group, Department of Nuclear Medicine, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany
- Department of Neurology, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany
- Correspondence: (S.P.); (T.v.E.); Tel.: +49-221-47882843 (T.v.E.)
| |
Collapse
|
37
|
A deep learning approach for parkinson’s disease severity assessment. HEALTH AND TECHNOLOGY 2022. [DOI: 10.1007/s12553-022-00698-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Abstract
Purpose
Parkinson’s Disease comes on top among neurodegenerative diseases affecting 10 million worldwide. To detect Parkinson’s Disease in a prior state, gait analysis is an effective choice. However, monitoring of Parkinson’s Disease using gait analysis is time consuming and exhaustive for patients and physicians. To assess severity of symptoms, a rating scale called Unified Parkinson's Disease Rating Scale is used. It determines mild and severe cases. Today, Parkinson’s Disease severity assessment is made in gait laboratories and by manual examination. These are time consuming and it is costly for health institutions to build and maintain laboratories. By using low-cost wearables and an effective model, aforementioned problems can be solved.
Methods
We provide a computerized solution for quantifiable assessment of Parkinson’s Disease symptoms severity. By using wearable sensors, our framework can predict exact symptom values to assess Parkinson’s Disease severity. We propose a deep learning approach that utilizes Ground Reaction Force sensors. From sensor signals, features are extracted and fed to a hybrid deep learning model. This model is the combination of Convolutional Neural Networks and Locally Weighted Random Forest.
Results
Proposed framework achieved 0.897, 3.009, 4.556 in terms of Correlation Coefficient, Mean Absolute Error and Root Mean Square Error, respectively. Proposed framework outperformed other machine and deep learning models. We also evaluated classification performance for disease detection. We outperformed most of the previous studies, achieving 99.5% accuracy, 98.7% sensitivity and 99.1% specificity.
Conclusion
This is the first study to use a deep learning regression approach to predict exact symptom value of Parkinson’s Disease patients. Results show that this approach can be effectively employed as a disease severity assessment tool using wearable sensors.
Collapse
|
38
|
Juengling FD, Wuest F, Kalra S, Agosta F, Schirrmacher R, Thiel A, Thaiss W, Müller HP, Kassubek J. Simultaneous PET/MRI: The future gold standard for characterizing motor neuron disease-A clinico-radiological and neuroscientific perspective. Front Neurol 2022; 13:890425. [PMID: 36061999 PMCID: PMC9428135 DOI: 10.3389/fneur.2022.890425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 07/20/2022] [Indexed: 01/18/2023] Open
Abstract
Neuroimaging assessment of motor neuron disease has turned into a cornerstone of its clinical workup. Amyotrophic lateral sclerosis (ALS), as a paradigmatic motor neuron disease, has been extensively studied by advanced neuroimaging methods, including molecular imaging by MRI and PET, furthering finer and more specific details of the cascade of ALS neurodegeneration and symptoms, facilitated by multicentric studies implementing novel methodologies. With an increase in multimodal neuroimaging data on ALS and an exponential improvement in neuroimaging technology, the need for harmonization of protocols and integration of their respective findings into a consistent model becomes mandatory. Integration of multimodal data into a model of a continuing cascade of functional loss also calls for the best attempt to correlate the different molecular imaging measurements as performed at the shortest inter-modality time intervals possible. As outlined in this perspective article, simultaneous PET/MRI, nowadays available at many neuroimaging research sites, offers the perspective of a one-stop shop for reproducible imaging biomarkers on neuronal damage and has the potential to become the new gold standard for characterizing motor neuron disease from the clinico-radiological and neuroscientific perspectives.
Collapse
Affiliation(s)
- Freimut D. Juengling
- Division of Oncologic Imaging, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Faculty of Medicine, University Bern, Bern, Switzerland
| | - Frank Wuest
- Division of Oncologic Imaging, University of Alberta, Edmonton, AB, Canada
| | - Sanjay Kalra
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Federica Agosta
- Division of Neuroscience, San Raffaele Scientific Institute, University Vita Salute San Raffaele, Milan, Italy
| | - Ralf Schirrmacher
- Division of Oncologic Imaging, University of Alberta, Edmonton, AB, Canada
- Medical Isotope and Cyclotron Facility, University of Alberta, Edmonton, AB, Canada
| | - Alexander Thiel
- Lady Davis Institute for Medical Research, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Wolfgang Thaiss
- Department of Nuclear Medicine, University of Ulm Medical Center, Ulm, Germany
- Department of Diagnostic and Interventional Radiology, University of Ulm Medical Center, Ulm, Germany
| | - Hans-Peter Müller
- Department of Neurology, Ulm University Medical Center, Ulm, Germany
| | - Jan Kassubek
- Department of Neurology, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
39
|
Delva A, Van Laere K, Vandenberghe W. Longitudinal Positron Emission Tomography Imaging of Presynaptic Terminals in Early Parkinson's Disease. Mov Disord 2022; 37:1883-1892. [PMID: 35819412 DOI: 10.1002/mds.29148] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/31/2022] [Accepted: 06/16/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Imaging tools that allow quantification of Parkinson's disease (PD) progression could facilitate the development of disease-modifying therapies. Cross-sectional studies have shown presynaptic terminal damage in PD patients, but longitudinal data are limited. OBJECTIVES The aim of this study was to longitudinally assess loss of presynaptic terminals in general and dopaminergic presynaptic terminals in particular as measures of disease progression in early PD. METHODS A total of 27 patients with early PD and 18 age- and sex-matched healthy controls underwent positron emission tomography (PET) with 11 C-UCB-J, a ligand for the brain-wide presynaptic terminal marker SV2A, and with 18 F-FE-PE2I, a highly selective dopamine transporter ligand, in combination with a comprehensive motor and non-motor clinical assessment at baseline (BL) and after 26.5 ± 2.1 months (Y2). SUVR-1 images were calculated and volumes of interest were delineated based on individual 3D T1 magnetic resonance imaging (MRI). RESULTS PD patients showed significant 2-year worsening of Movement Disorder Society-sponsored revision of the Unified Parkinson's Disease Rating Scale Part III (MDS-UPDRS-III) (off medication) scores, but not of non-motor scores. Motor and non-motor scores in controls did not change significantly over 2 years. 18 F-FE-PE2I binding in caudate and putamen showed significant 2-year decline in the PD group and remained unchanged in controls. Longitudinal decline of striatal 18 F-FE-PE2I binding in PD did not correlate with longitudinal changes in MDS-UPDRS-III scores. 11 C-UCB-J PET did not show any region with significant 2-year change in PD or controls. CONCLUSIONS 18 F-FE-PE2I PET showed robust 2-year decline in early PD, but 11 C-UCB-J PET did not. Longitudinal changes in 18 F-FE-PE2I binding did not correlate with clinical motor progression. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Aline Delva
- Department of Neurosciences, KU Leuven, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,Division of Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Wim Vandenberghe
- Department of Neurosciences, KU Leuven, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
40
|
Carson RE, Naganawa M, Toyonaga T, Koohsari S, Yang Y, Chen MK, Matuskey D, Finnema SJ. Imaging of Synaptic Density in Neurodegenerative Disorders. J Nucl Med 2022; 63:60S-67S. [PMID: 35649655 DOI: 10.2967/jnumed.121.263201] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/10/2022] [Indexed: 02/07/2023] Open
Abstract
PET technology has produced many radiopharmaceuticals that target specific brain proteins and other measures of brain function. Recently, a new approach has emerged to image synaptic density by targeting the synaptic vesicle protein 2A (SV2A), an integral glycoprotein in the membrane of synaptic vesicles and widely distributed throughout the brain. Multiple SV2A ligands have been developed and translated to human use. The most successful of these to date is 11C-UCB-J, because of its high uptake, moderate metabolism, and effective quantification with a 1-tissue-compartment model. Further, since SV2A is the target of the antiepileptic drug levetiracetam, human blocking studies have characterized specific binding and potential reference regions. Regional brain SV2A levels were shown to correlate with those of synaptophysin, another commonly used marker of synaptic density, providing the basis for SV2A PET imaging to have broad utility across neuropathologic diseases. In this review, we highlight the development of SV2A tracers and the evaluation of quantification methods, including compartment modeling and simple tissue ratios. Mouse and rat models of neurodegenerative diseases have been studied with small-animal PET, providing validation by comparison to direct tissue measures. Next, we review human PET imaging results in multiple neurodegenerative disorders. Studies on Parkinson disease and Alzheimer disease have progressed most rapidly at multiple centers, with generally consistent results of patterns of SV2A or synaptic loss. In Alzheimer disease, the synaptic loss patterns differ from those of amyloid, tau, and 18F-FDG, although intertracer and interregional correlations have been found. Smaller studies have been reported in other disorders, including Lewy body dementia, frontotemporal dementia, Huntington disease, progressive supranuclear palsy, and corticobasal degeneration. In conclusion, PET imaging of SV2A has rapidly developed, and qualified radioligands are available. PET studies on humans indicate that SV2A loss might be specific to disease-associated brain regions and consistent with synaptic density loss. The recent availability of new 18F tracers, 18F-SynVesT-1 and 18F-SynVesT-2, will substantially broaden the application of SV2A PET. Future studies are needed in larger patient cohorts to establish the clinical value of SV2A PET and its potential for diagnosis and progression monitoring of neurodegenerative diseases, as well as efficacy assessment of disease-modifying therapies.
Collapse
Affiliation(s)
- Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut;
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Mika Naganawa
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
| | - Sheida Koohsari
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
| | - Yanghong Yang
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
| | - Ming-Kai Chen
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
| | - David Matuskey
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut; and
| | - Sjoerd J Finnema
- Neuroscience Discovery Research, Translational Imaging, AbbVie, North Chicago, Illinois
| |
Collapse
|
41
|
Rossi R, Arjmand S, Bærentzen SL, Gjedde A, Landau AM. Synaptic Vesicle Glycoprotein 2A: Features and Functions. Front Neurosci 2022; 16:864514. [PMID: 35573314 PMCID: PMC9096842 DOI: 10.3389/fnins.2022.864514] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/05/2022] [Indexed: 01/05/2023] Open
Abstract
In recent years, the field of neuroimaging dramatically moved forward by means of the expeditious development of specific radioligands of novel targets. Among these targets, the synaptic vesicle glycoprotein 2A (SV2A) is a transmembrane protein of synaptic vesicles, present in all synaptic terminals, irrespective of neurotransmitter content. It is involved in key functions of neurons, focused on the regulation of neurotransmitter release. The ubiquitous expression in gray matter regions of the brain is the basis of its candidacy as a marker of synaptic density. Following the development of molecules derived from the structure of the anti-epileptic drug levetiracetam, which selectively binds to SV2A, several radiolabeled markers have been synthetized to allow the study of SV2A distribution with positron emission tomography (PET). These radioligands permit the evaluation of in vivo changes of SV2A distribution held to be a potential measure of synaptic density in physiological and pathological conditions. The use of SV2A as a biomarker of synaptic density raises important questions. Despite numerous studies over the last decades, the biological function and the expressional properties of SV2A remain poorly understood. Some functions of SV2A were claimed, but have not been fully elucidated. While the expression of SV2A is ubiquitous, stronger associations between SV2A and Υ amino butyric acid (GABA)-ergic rather than glutamatergic synapses were observed in some brain structures. A further issue is the unclear interaction between SV2A and its tracers, which reflects a need to clarify what really is detected with neuroimaging tools. Here, we summarize the current knowledge of the SV2A protein and we discuss uncertain aspects of SV2A biology and physiology. As SV2A expression is ubiquitous, but likely more strongly related to a certain type of neurotransmission in particular circumstances, a more extensive knowledge of the protein would greatly facilitate the analysis and interpretation of neuroimaging results by allowing the evaluation not only of an increase or decrease of the protein level, but also of the type of neurotransmission involved.
Collapse
Affiliation(s)
- Rachele Rossi
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Shokouh Arjmand
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Simone Larsen Bærentzen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Albert Gjedde
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.,Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Anne M Landau
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
42
|
Crișan G, Moldovean-Cioroianu NS, Timaru DG, Andrieș G, Căinap C, Chiș V. Radiopharmaceuticals for PET and SPECT Imaging: A Literature Review over the Last Decade. Int J Mol Sci 2022; 23:5023. [PMID: 35563414 PMCID: PMC9103893 DOI: 10.3390/ijms23095023] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Positron emission tomography (PET) uses radioactive tracers and enables the functional imaging of several metabolic processes, blood flow measurements, regional chemical composition, and/or chemical absorption. Depending on the targeted processes within the living organism, different tracers are used for various medical conditions, such as cancer, particular brain pathologies, cardiac events, and bone lesions, where the most commonly used tracers are radiolabeled with 18F (e.g., [18F]-FDG and NA [18F]). Oxygen-15 isotope is mostly involved in blood flow measurements, whereas a wide array of 11C-based compounds have also been developed for neuronal disorders according to the affected neuroreceptors, prostate cancer, and lung carcinomas. In contrast, the single-photon emission computed tomography (SPECT) technique uses gamma-emitting radioisotopes and can be used to diagnose strokes, seizures, bone illnesses, and infections by gauging the blood flow and radio distribution within tissues and organs. The radioisotopes typically used in SPECT imaging are iodine-123, technetium-99m, xenon-133, thallium-201, and indium-111. This systematic review article aims to clarify and disseminate the available scientific literature focused on PET/SPECT radiotracers and to provide an overview of the conducted research within the past decade, with an additional focus on the novel radiopharmaceuticals developed for medical imaging.
Collapse
Affiliation(s)
- George Crișan
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | | | - Diana-Gabriela Timaru
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
| | - Gabriel Andrieș
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | - Călin Căinap
- The Oncology Institute “Prof. Dr. Ion Chiricuţă”, Republicii 34-36, 400015 Cluj-Napoca, Romania;
| | - Vasile Chiș
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Institute for Research, Development and Innovation in Applied Natural Sciences, Babeș-Bolyai University, Str. Fântânele 30, 400327 Cluj-Napoca, Romania
| |
Collapse
|
43
|
Michiels L, Mertens N, Thijs L, Radwan A, Sunaert S, Vandenbulcke M, Verheyden G, Koole M, Van Laere K, Lemmens R. Changes in synaptic density in the subacute phase after ischemic stroke: A 11C-UCB-J PET/MR study. J Cereb Blood Flow Metab 2022; 42:303-314. [PMID: 34550834 PMCID: PMC9122519 DOI: 10.1177/0271678x211047759] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Functional alterations after ischemic stroke have been described with Magnetic Resonance Imaging (MRI) and perfusion Positron Emission Tomography (PET), but no data on in vivo synaptic changes exist. Recently, imaging of synaptic density became available by targeting synaptic vesicle protein 2 A, a protein ubiquitously expressed in all presynaptic nerve terminals. We hypothesized that in subacute ischemic stroke loss of synaptic density can be evaluated with 11C-UCB-J PET in the ischemic tissue and that alterations in synaptic density can be present in brain regions beyond the ischemic core. We recruited ischemic stroke patients to undergo 11C-UCB-J PET/MR imaging 21 ± 8 days after stroke onset to investigate regional 11C-UCB-J SUVR (standardized uptake value ratio). There was a decrease (but residual signal) of 11C-UCB-J SUVR within the lesion of 16 stroke patients compared to 40 healthy controls (ratiolesion/controls = 0.67 ± 0.28, p = 0.00023). Moreover, 11C-UCB-J SUVR was lower in the non-lesioned tissue of the affected hemisphere compared to the unaffected hemisphere (ΔSUVR = -0.17, p = 0.0035). The contralesional cerebellar hemisphere showed a lower 11C-UCB-J SUVR compared to the ipsilesional cerebellar hemisphere (ΔSUVR = -0.14, p = 0.0048). In 8 out of 16 patients, the asymmetry index suggested crossed cerebellar diaschisis. Future research is required to longitudinally study these changes in synaptic density and their association with outcome.
Collapse
Affiliation(s)
- Laura Michiels
- Department of Neurosciences, KU Leuven, Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Nathalie Mertens
- Nuclear Medicine and Molecular Imaging, 26657KU Leuven, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Liselot Thijs
- Department of Rehabilitation Sciences, 26657KU Leuven, KU Leuven, Leuven, Belgium
| | - Ahmed Radwan
- Translational MRI, 26657KU Leuven, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Stefan Sunaert
- Translational MRI, 26657KU Leuven, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.,Department of Geriatric Psychiatry, University Psychiatric Centre, KU Leuven, Leuven, Belgium
| | - Geert Verheyden
- Department of Rehabilitation Sciences, 26657KU Leuven, KU Leuven, Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, 26657KU Leuven, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, 26657KU Leuven, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,Division of Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Robin Lemmens
- Department of Neurosciences, KU Leuven, Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
44
|
Cervantes González A, Belbin O. Fluid markers of synapse degeneration in synucleinopathies. J Neural Transm (Vienna) 2022; 129:187-206. [PMID: 35147800 DOI: 10.1007/s00702-022-02467-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/20/2022] [Indexed: 01/06/2023]
Abstract
The abnormal accumulation of α-synuclein in the brain is a common feature of Parkinson's disease (PD), PD dementia (PDD), dementia with Lewy bodies (DLB) and multiple system atrophy (MSA), and synucleinopathies that present with overlapping but distinct clinical symptoms that include motor and cognitive deficits. Synapse degeneration is the crucial neuropathological event in these synucleinopathies and the neuropathological correlate of connectome dysfunction. The cognitive and motor deficits resulting from the connectome dysfunction are currently measured by scalar systems that are limited in their sensitivity and largely subjective. Ideally, a marker of synapse degeneration would correlate with measures of cognitive or motor impairment, and could therefore be used as a more objective, surrogate biomarker of the core clinical features of these diseases. Furthermore, an objective surrogate biomarker that can detect and monitor the progression of synapse degeneration would improve patient management and clinical trial design, and could provide a measure of therapeutic response. Here, we review the published findings relating to candidate biomarkers of synapse degeneration in PD, PDD, DLB, and MSA patient-derived biofluids and discuss the findings in the context of the mechanisms associated with α-synuclein-mediated synapse degeneration. Understanding these mechanisms is essential not only for discovery of biomarkers, but also to improve our understanding of the earliest changes in disease pathogenesis of synucleinopathies.
Collapse
Affiliation(s)
- Alba Cervantes González
- Neurology Department, Biomedical Research Institute Sant Pau (IIB Sant Pau) and Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Olivia Belbin
- Neurology Department, Biomedical Research Institute Sant Pau (IIB Sant Pau) and Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.
| |
Collapse
|
45
|
Jordan S, Zielinski M, Kortylewski M, Kuhn T, Bystritsky A. Noninvasive Delivery of Biologicals to the Brain. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2022; 20:64-70. [PMID: 35746928 PMCID: PMC9063603 DOI: 10.1176/appi.focus.20210028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In the past, psychotherapy and neuropharmacological approaches have been the most common treatments for disordered thoughts, moods, and behaviors. One new path of brain therapeutics is in the deployment of noninvasive approaches designed to reprogram brain function at the cellular level. Treatment at the cellular level may be considered for a wide array of disorders, ranging from mood disorders to neurodegenerative disorders. Brain-targeted biological therapy may provide minimally invasive and accurate delivery of treatment. The present article discusses the hurdles and advances that characterize the pathway to this goal.
Collapse
|
46
|
Brumberg J, Varrone A. New PET radiopharmaceuticals for imaging CNS diseases. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
47
|
Decet M, Verstreken P. Presynaptic Autophagy and the Connection With Neurotransmission. Front Cell Dev Biol 2021; 9:790721. [PMID: 34988081 PMCID: PMC8722708 DOI: 10.3389/fcell.2021.790721] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/01/2021] [Indexed: 01/14/2023] Open
Abstract
Autophagy is an evolutionary conserved catabolic pathway essential for the maintenance of cellular homeostasis. Defective proteins and organelles are engulfed by autophagosomal membranes which fuse with lysosomes for cargo degradation. In neurons, the orchestrated progression of autophagosome formation and maturation occurs in distinct subcellular compartments. For synapses, the distance from the soma and the oxidative stress generated during intense neuronal activity pose a challenge to maintain protein homeostasis. Autophagy constitutes a crucial mechanism for proper functioning of this unique and vulnerable cellular compartment. We are now beginning to understand how autophagy is regulated at pre-synaptic terminals and how this pathway, when imbalanced, impacts on synaptic function and -ultimately- neuronal survival. We review here the current state of the art of "synaptic autophagy", with an emphasis on the biogenesis of autophagosomes at the pre-synaptic compartment. We provide an overview of the existing knowledge on the signals inducing autophagy at synapses, highlight the interplay between autophagy and neurotransmission, and provide perspectives for future research.
Collapse
Affiliation(s)
- Marianna Decet
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven, Belgium
| | - Patrik Verstreken
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven, Belgium
| |
Collapse
|
48
|
α-Synuclein Overexpression Increases Dopamine D2/3 Receptor Binding and Immune Activation in a Model of Early Parkinson’s Disease. Biomedicines 2021; 9:biomedicines9121876. [PMID: 34944691 PMCID: PMC8698691 DOI: 10.3390/biomedicines9121876] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/01/2021] [Accepted: 12/07/2021] [Indexed: 12/14/2022] Open
Abstract
Progressive degeneration of dopaminergic neurons, immune activation, and α-synuclein pathology characterize Parkinson’s disease (PD). We previously reported that unilateral intranigral injection of recombinant adeno-associated viral (rAAV) vectors encoding wild-type human α-synuclein produced a rat model of early PD with dopamine terminal dysfunction. Here we tested the hypothesis that decreases in dopamine result in increased postsynaptic dopamine D2/D3 receptor expression, neuroinflammation, and reduced synaptic vesicle glycoprotein 2A (SV2A) density. Rats were injected with rAAV encoding α-synuclein or green fluorescent protein and subjected to non-pharmacological motor tests, before euthanization at 12 weeks post-injection. We performed: (1) in situ hybridization of nigral tyrosine hydroxylase mRNA, (2) HPLC of striatal dopamine content, and (3) autoradiography with [3H]raclopride, [3H]DTBZ, [3H]GBR12935, [3H]PK11195, and [3H]UCB-J to measure binding at D2/3 receptors, vesicular monoamine transporter 2, dopamine transporters, mitochondrial translocator protein, and SV2A, respectively. rAAV-α-synuclein induced motor asymmetry and reduced tyrosine hydroxylase mRNA and dopamine content in ipsilateral brain regions. This was paralleled by elevated ipsilateral postsynaptic dopamine D2/3 receptor expression and immune activation, with no changes to synaptic SV2A density. In conclusion, α-synuclein overexpression results in dopaminergic degeneration that induced compensatory increases in D2/3 binding and immune activation, recapitulating many of the pathological characteristics of PD.
Collapse
|
49
|
Badanjak K, Mulica P, Smajic S, Delcambre S, Tranchevent LC, Diederich N, Rauen T, Schwamborn JC, Glaab E, Cowley SA, Antony PMA, Pereira SL, Venegas C, Grünewald A. iPSC-Derived Microglia as a Model to Study Inflammation in Idiopathic Parkinson's Disease. Front Cell Dev Biol 2021; 9:740758. [PMID: 34805149 PMCID: PMC8602578 DOI: 10.3389/fcell.2021.740758] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease with unknown cause in the majority of patients, who are therefore considered "idiopathic" (IPD). PD predominantly affects dopaminergic neurons in the substantia nigra pars compacta (SNpc), yet the pathology is not limited to this cell type. Advancing age is considered the main risk factor for the development of IPD and greatly influences the function of microglia, the immune cells of the brain. With increasing age, microglia become dysfunctional and release pro-inflammatory factors into the extracellular space, which promote neuronal cell death. Accordingly, neuroinflammation has also been described as a feature of PD. So far, studies exploring inflammatory pathways in IPD patient samples have primarily focused on blood-derived immune cells or brain sections, but rarely investigated patient microglia in vitro. Accordingly, we decided to explore the contribution of microglia to IPD in a comparative manner using, both, iPSC-derived cultures and postmortem tissue. Our meta-analysis of published RNAseq datasets indicated an upregulation of IL10 and IL1B in nigral tissue from IPD patients. We observed increased expression levels of these cytokines in microglia compared to neurons using our single-cell midbrain atlas. Moreover, IL10 and IL1B were upregulated in IPD compared to control microglia. Next, to validate these findings in vitro, we generated IPD patient microglia from iPSCs using an established differentiation protocol. IPD microglia were more readily primed as indicated by elevated IL1B and IL10 gene expression and higher mRNA and protein levels of NLRP3 after LPS treatment. In addition, IPD microglia had higher phagocytic capacity under basal conditions-a phenotype that was further exacerbated upon stimulation with LPS, suggesting an aberrant microglial function. Our results demonstrate the significance of microglia as the key player in the neuroinflammation process in IPD. While our study highlights the importance of microglia-mediated inflammatory signaling in IPD, further investigations will be needed to explore particular disease mechanisms in these cells.
Collapse
Affiliation(s)
- Katja Badanjak
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Patrycja Mulica
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Semra Smajic
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Sylvie Delcambre
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | | | - Nico Diederich
- Centre Hospitalier de Luxembourg (CHL), Luxembourg, Luxembourg
| | - Thomas Rauen
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Jens C. Schwamborn
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Sally A. Cowley
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Paul M. A. Antony
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
- Disease Modeling and Screening Platform (DMSP), Luxembourg Institute of Systems Biomedicine, University of Luxembourg and Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Sandro L. Pereira
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Carmen Venegas
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Anne Grünewald
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
50
|
Delva A, Michiels L, Koole M, Van Laere K, Vandenberghe W. Synaptic Damage and Its Clinical Correlates in People With Early Huntington Disease: A PET Study. Neurology 2021; 98:e83-e94. [PMID: 34663644 DOI: 10.1212/wnl.0000000000012969] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/04/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Synaptic damage has been proposed to play a major role in the pathophysiology of Huntington's disease (HD), but in vivo evidence in humans is lacking. We performed a PET imaging study to assess synaptic damage and its clinical correlates in early HD in vivo. METHODS: In this cross-sectional study, premanifest and early manifest (Shoulson-Fahn stage 1 and 2) HD mutation carriers and age- and gender-matched healthy controls underwent clinical assessment of motor and non-motor manifestations and time-of-flight PET with 11C-UCB-J, a radioligand targeting the ubiquitous presynaptic terminal marker SV2A. We also performed 18F-FDG PET in all subjects, as regional cerebral glucose consumption is thought to largely reflect synaptic activity. Volumes of interest were delineated based on individual 3D T1 MRI. Standardized uptake value ratio (SUVR)-1 images were calculated for 11C-UCB-J with the centrum semiovale as reference region. 18F-FDG PET activity was normalized to the pons. All PET data were corrected for partial volume effects. Volume of interest- and voxel-based analyses were performed. Correlations between clinical scores and 11C-UCB-J PET data were calculated. RESULTS 18 HD mutation carriers (51.4 ± 11.6 years; 6 female; 7 premanifest, 11 early manifest) and 15 healthy controls (52.3 ± 3.5 years; 4 female) were included. In the HD group, significant loss of SV2A binding was found in putamen, caudate, pallidum, cerebellum, parietal, temporal and frontal cortex, whereas reduced 18F-FDG uptake was restricted to caudate and putamen. In the premanifest subgroup, 11C-UCB-J and 18F-FDG PET showed significant reductions in putamen and caudate only. In the total HD group, SV2A loss in the putamen correlated with motor impairment. DISCUSSION Our data reveal loss of presynaptic terminal integrity in early HD, which begins in the striatum in the premanifest phase, spreads extensively to extrastriatal regions in the early manifest phase, and correlates with motor impairment. 11C-UCB-J PET is more sensitive than 18F-FDG PET for detection of extrastriatal changes in early HD. CLASSIFICATION OF EVIDENCE This study provides class III evidence that 11C-UCB-J PET accurately identifies HD from normal controls.
Collapse
Affiliation(s)
- Aline Delva
- Department of Neurosciences, KU Leuven, Belgium; .,Department of Neurology, University Hospitals Leuven, Belgium
| | - Laura Michiels
- Department of Neurosciences, KU Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Belgium.,VIB, Center for Brain & Disease Research, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Belgium.,Division of Nuclear Medicine, University Hospitals Leuven, Belgium
| | - Wim Vandenberghe
- Department of Neurosciences, KU Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Belgium
| |
Collapse
|