1
|
Vermeulen I, Vandenbosch M, Viot D, Mercier J, Cabañas DAW, Martinez-Martinez P, Barton P, Heeren RM, Cillero-Pastor B. Spatial Distribution of Brain PET Tracers by MALDI Imaging. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2025; 36:688-698. [PMID: 40073292 PMCID: PMC11969657 DOI: 10.1021/jasms.4c00307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 02/16/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025]
Abstract
Evaluating tissue distribution of Positron Emission Tomography (PET) tracers during their development conventionally involves autoradiography techniques, where radioactive compounds are used for ex vivo visualization and quantification in tissues during preclinical development stages. Mass Spectrometry Imaging (MSI) offers a potential alternative, providing spatial information without the need for radioactivity with a similar spatial resolution. This study aimed to optimize a MSI sample preparation protocol for assessing PET tracer candidates ex vivo with a focus on two compounds: UCB-J and UCB2400. We tested different matrices and introduced washing steps to improve PET tracer detection. Tissue homogenates were prepared to construct calibration curves for quantification. The incorporation of a washing step into the MSI sample preparation protocol enhanced the signal of both PET tracers. Our findings highlight MSI's potential as a cost-effective and efficient method for the evaluation of PET tracer distribution. The optimized approach offered here can provide a protocol that enhances the signal and minimizes ion suppression effect, which can be valuable for future evaluation of PET tracers in MSI studies.
Collapse
Affiliation(s)
- Isabeau Vermeulen
- The Maastricht
MultiModal Molecular Imaging Institute (M4i), Division of Imaging
Mass Spectrometry, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Michiel Vandenbosch
- The Maastricht
MultiModal Molecular Imaging Institute (M4i), Division of Imaging
Mass Spectrometry, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Delphine Viot
- Translational
Science, DMPK, UCB Biopharma SRL, Chemin du Foriest, B1420 Braine-l’Alleud, Belgium
| | - Joel Mercier
- Discovery
Chemistry BE, UCB Biopharma SRL, Chemin du Foriest, B1420 Braine-l’Alleud, Belgium
| | - Diego Asensio-Wandosell Cabañas
- Department
of Psychiatry and Neuropsychology, Maastricht
University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Pilar Martinez-Martinez
- Department
of Psychiatry and Neuropsychology, Maastricht
University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Patrick Barton
- Translational
Science, DMPK, UCB Celltech, Branch of UCB Pharma S.A., 208 Bath
Road, Slough, Berkshire SL1 3WE, United
Kingdom
| | - Ron M.A. Heeren
- The Maastricht
MultiModal Molecular Imaging Institute (M4i), Division of Imaging
Mass Spectrometry, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Berta Cillero-Pastor
- The Maastricht
MultiModal Molecular Imaging Institute (M4i), Division of Imaging
Mass Spectrometry, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
- Cell Biology-Inspired
Tissue Engineering (cBITE), MERLN, Maastricht
University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
2
|
Berckmans L, Schrauwen C, Miranda A, Staelens S, Bertoglio D. Assessing non-invasive quantitative methods for [ 18F]SynVesT-1 PET imaging of synaptic vesicle glycoprotein 2A in the rat brain. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07170-w. [PMID: 40032689 DOI: 10.1007/s00259-025-07170-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: 01/16/2025] [Accepted: 02/17/2025] [Indexed: 03/05/2025]
Abstract
PURPOSE Synaptic vesicle glycoprotein 2A (SV2A) is a critical biomarker for evaluating synaptic density in neurological research. Among available radioligands, [18F]SynVesT-1 is increasingly used in PET research because of its extended half-life, while having comparable pharmacokinetic properties to the widely used [11C]UCB-J. However, quantitative application in rat models remains unexplored for [18F]SynVesT-1. This study aims to validate quantitative kinetic modelling methods for [18F]SynVesT-1 and develop non-invasive quantification methods for synaptic density in rats. METHODS First, blood analysis of [18F]SynVesT-1 was performed to generate metabolite-corrected plasma input functions. Then, kinetic modelling was evaluated using compartmental analysis approaches, as well as Logan plot. Furthermore, non-invasive image-derived input functions (IDIF), with and without non-negative matrix factorization (NMF) were compared against the arterial input function (AIF). RESULTS Blood analysis showed that the parent fraction of the tracer decreased over time following a sigmoid curve, while the plasma-to-whole blood ratio remained stable over time (0.89 ± 0.02). The two-tissue compartmental model (2TCM) and Logan plot were determined to be the most accurate methods for quantification of [18F]SynVesT-1 kinetics in rats. Additionally, the results demonstrated strong agreement between AIF-derived and image-derived volume of distribution (VT) values, with both image-derived input approaches (IDIF and IDIF-NMF) performing equally well. CONCLUSION These findings validate kinetic modelling methods for [18F]SynVesT-1 PET, enabling their application in further rat studies for preclinical neuroscience research and prove that image-derived input functions are reliable non-invasive alternatives to AIF.
Collapse
Affiliation(s)
- Lori Berckmans
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, Antwerp, Belgium.
- µNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium.
| | - Claudia Schrauwen
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, Antwerp, Belgium
- µNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium
- URPhyM-NARILIS, University of Namur, Namur, Belgium
| | - Alan Miranda
- µNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp, Belgium
| | - Steven Staelens
- µNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp, Belgium
| | - Daniele Bertoglio
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, Antwerp, Belgium
- µNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
3
|
Khanppnavar B, Leka O, Pal SK, Korkhov VM, Kammerer RA. Cryo-EM structure of the botulinum neurotoxin A/SV2B complex and its implications for translocation. Nat Commun 2025; 16:1224. [PMID: 39934119 PMCID: PMC11814414 DOI: 10.1038/s41467-025-56304-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 01/14/2025] [Indexed: 02/13/2025] Open
Abstract
Botulinum neurotoxin A1 (BoNT/A1) belongs to the most potent toxins and is used as a major therapeutic agent. Neurotoxin conformation is crucial for its translocation to the neuronal cytosol, a key process for intoxication that is only poorly understood. To gain molecular insights into the steps preceding toxin translocation, we determine cryo-EM structures of BoNT/A1 alone and in complex with its receptor synaptic vesicle glycoprotein 2B (SV2B). In solution, BoNT/A1 adopts a unique, semi-closed conformation. The toxin changes its structure into an open state upon receptor binding with the translocation domain (HN) and the catalytic domain (LC) remote from the membrane, suggesting translocation incompatibility. Under acidic pH conditions, where translocation is initiated, receptor-bound BoNT/A1 switches back into a semi-closed conformation. This conformation brings the LC and HN close to the membrane, suggesting that a translocation-competent state of the toxin is required for successful LC transport into the neuronal cytosol.
Collapse
Affiliation(s)
| | - Oneda Leka
- PSI Center for Life Sciences, Villigen, Switzerland
| | | | - Volodymyr M Korkhov
- PSI Center for Life Sciences, Villigen, Switzerland.
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland.
| | | |
Collapse
|
4
|
Asch RH, Abdallah CG, Carson RE, Esterlis I. Challenges and rewards of in vivo synaptic density imaging, and its application to the study of depression. Neuropsychopharmacology 2024; 50:153-163. [PMID: 39039139 PMCID: PMC11525584 DOI: 10.1038/s41386-024-01913-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/14/2024] [Accepted: 06/26/2024] [Indexed: 07/24/2024]
Abstract
The development of novel radiotracers for Positron Emission Tomography (PET) imaging agents targeting the synaptic vesicle glycoprotein 2 A (SV2A), an integral glycoprotein present in the membrane of all synaptic vesicles throughout the central nervous system, provides a method for the in vivo quantification of synaptic density. This is of particular interest in neuropsychiatric disorders given that synaptic alterations appear to underlie disease progression and symptom severity. In this review, we briefly describe the development of these SV2A tracers and the evaluation of quantification methods. Next, we discuss application of SV2A PET imaging to the study of depression, including a review of our findings demonstrating lower SV2A synaptic density in people with significant depressive symptoms and the use of a ketamine drug challenge to examine synaptogenesis in vivo. We then highlight the importance of performing translational PET imaging in animal models in conjunction with clinical imaging. We consider the ongoing challenges, possible solutions, and present preliminary findings from our lab demonstrating the translational benefit and potential of in vivo SV2A imaging in animal models of chronic stress. Finally, we discuss methodological improvements and future directions for SV2A imaging, potentially in conjunction with other neural markers.
Collapse
Affiliation(s)
- Ruth H Asch
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Chadi G Abdallah
- Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale School of Engineering, New Haven, CT, USA
| | - Irina Esterlis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale School of Medicine, New Haven, CT, USA.
- U.S. Department of Veteran Affairs National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
5
|
Chang CH, Lim KL, Foo JN. Synaptic Vesicle Glycoprotein 2C: a role in Parkinson's disease. Front Cell Neurosci 2024; 18:1437144. [PMID: 39301216 PMCID: PMC11410587 DOI: 10.3389/fncel.2024.1437144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024] Open
Abstract
Synaptic Vesicle Glycoprotein 2C (SV2C), characterized by its selective expression in discrete brain regions such as the midbrain, has recently emerged as a promising player in Parkinson's Disease (PD) - a debilitating neurodegenerative disorder affecting millions worldwide. This review aims to consolidate our current understanding of SV2C's function, its involvement in PD pathogenesis, and to evaluate its potential as a therapeutic target. Integrating previous findings of SV2C, from genetics to molecular studies, and drawing on insights from the largest East Asian genome-wide association study that highlights SV2C as a novel risk factor for PD, we explore the potential pathways through which SV2C may influence the disease. Our discussion extends to the implications of SV2C's role in synaptic vesicle trafficking, neurotransmitter release, and α-synuclein homeostasis, thereby laying the groundwork for future investigations that could pave the way for novel therapeutic strategies in combating PD.
Collapse
Affiliation(s)
- Chu Hua Chang
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Interdisciplinary Graduate Programme (IGP-Neuroscience), Nanyang Technological University, Singapore, Singapore
| | - Kah Leong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Jia Nee Foo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
6
|
Li J, Zou R, Varrone A, Nag S, Halldin C, Ågren H. Exploring the Interactions between two Ligands, UCB-J and UCB-F, and Synaptic Vesicle Glycoprotein 2 Isoforms. ACS Chem Neurosci 2024; 15:2018-2027. [PMID: 38701380 PMCID: PMC11099911 DOI: 10.1021/acschemneuro.4c00029] [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: 01/13/2024] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 05/05/2024] Open
Abstract
In silico modeling was applied to study the efficiency of two ligands, namely, UCB-J and UCB-F, to bind to isoforms of the synaptic vesicle glycoprotein 2 (SV2) that are involved in the regulation of synaptic function in the nerve terminals, with the ultimate goal to understand the selectivity of the interaction between UCB-J and UCB-F to different isoforms of SV2. Docking and large-scale molecular dynamics simulations were carried out to unravel various binding patterns, types of interactions, and binding free energies, covering hydrogen bonding and nonspecific hydrophobic interactions, water bridge, π-π, and cation-π interactions. The overall preference for bonding types of UCB-J and UCB-F with particular residues in the protein pockets can be disclosed in detail. A unique interaction fingerprint, namely, hydrogen bonding with additional cation-π interaction with the pyridine moiety of UCB-J, could be established as an explanation for its high selectivity over the SV2 isoform A (SV2A). Other molecular details, primarily referring to the presence of π-π interactions and hydrogen bonding, could also be analyzed as sources of selectivity of the UCB-F tracer for the three isoforms. The simulations provide atomic details to support future development of new selective tracers targeting synaptic vesicle glycoproteins and their associated diseases.
Collapse
Affiliation(s)
- Junhao Li
- Department
of Physics and Astronomy, Uppsala University, Box 516, Uppsala SE-751 20, Sweden
| | - Rongfeng Zou
- Department
of Physics and Astronomy, Uppsala University, Box 516, Uppsala SE-751 20, Sweden
| | - Andrea Varrone
- Department
of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm Health Care Services, Stockholm 171 77, Sweden
| | - Sangram Nag
- Department
of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm Health Care Services, Stockholm 171 77, Sweden
| | - Christer Halldin
- Department
of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm Health Care Services, Stockholm 171 77, Sweden
| | - Hans Ågren
- Department
of Physics and Astronomy, Uppsala University, Box 516, Uppsala SE-751 20, Sweden
| |
Collapse
|
7
|
Yamagata A, Ito K, Suzuki T, Dohmae N, Terada T, Shirouzu M. Structural basis for antiepileptic drugs and botulinum neurotoxin recognition of SV2A. Nat Commun 2024; 15:3027. [PMID: 38637505 PMCID: PMC11026379 DOI: 10.1038/s41467-024-47322-4] [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: 07/12/2023] [Accepted: 03/26/2024] [Indexed: 04/20/2024] Open
Abstract
More than one percent of people have epilepsy worldwide. Levetiracetam (LEV) is a successful new-generation antiepileptic drug (AED), and its derivative, brivaracetam (BRV), shows improved efficacy. Synaptic vesicle glycoprotein 2a (SV2A), a putative membrane transporter in the synaptic vesicles (SVs), has been identified as a target of LEV and BRV. SV2A also serves as a receptor for botulinum neurotoxin (BoNT), which is the most toxic protein and has paradoxically emerged as a potent reagent for therapeutic and cosmetic applications. Nevertheless, no structural analysis on AEDs and BoNT recognition by full-length SV2A has been available. Here we describe the cryo-electron microscopy structures of the full-length SV2A in complex with the BoNT receptor-binding domain, BoNT/A2 HC, and either LEV or BRV. The large fourth luminal domain of SV2A binds to BoNT/A2 HC through protein-protein and protein-glycan interactions. LEV and BRV occupy the putative substrate-binding site in an outward-open conformation. A propyl group in BRV creates additional contacts with SV2A, explaining its higher binding affinity than that of LEV, which was further supported by label-free spectral shift assay. Numerous LEV derivatives have been developed as AEDs and positron emission tomography (PET) tracers for neuroimaging. Our work provides a structural framework for AEDs and BoNT recognition of SV2A and a blueprint for the rational design of additional AEDs and PET tracers.
Collapse
Affiliation(s)
- Atsushi Yamagata
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, Japan.
| | - Kaori Ito
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, Japan
| | - Takehiro Suzuki
- Biomolecular Characterization Unit, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama, Japan
| | - Naoshi Dohmae
- Biomolecular Characterization Unit, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama, Japan
| | - Tohru Terada
- Department of Biotechnology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, Japan
| | - Mikako Shirouzu
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, Japan
| |
Collapse
|
8
|
Fang XT, Raval NR, O’Dell RS, Naganawa M, Mecca AP, Chen MK, van Dyck CH, Carson RE. Synaptic density patterns in early Alzheimer's disease assessed by independent component analysis. Brain Commun 2024; 6:fcae107. [PMID: 38601916 PMCID: PMC11004947 DOI: 10.1093/braincomms/fcae107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 02/23/2024] [Accepted: 03/25/2024] [Indexed: 04/12/2024] Open
Abstract
Synaptic loss is a primary pathology in Alzheimer's disease and correlates best with cognitive impairment as found in post-mortem studies. Previously, we observed in vivo reductions of synaptic density with [11C]UCB-J PET (radiotracer for synaptic vesicle protein 2A) throughout the neocortex and medial temporal brain regions in early Alzheimer's disease. In this study, we applied independent component analysis to synaptic vesicle protein 2A-PET data to identify brain networks associated with cognitive deficits in Alzheimer's disease in a blinded data-driven manner. [11C]UCB-J binding to synaptic vesicle protein 2A was measured in 38 Alzheimer's disease (24 mild Alzheimer's disease dementia and 14 mild cognitive impairment) and 19 cognitively normal participants. [11C]UCB-J distribution volume ratio values were calculated with a whole cerebellum reference region. Principal components analysis was first used to extract 18 independent components to which independent component analysis was then applied. Subject loading weights per pattern were compared between groups using Kruskal-Wallis tests. Spearman's rank correlations were used to assess relationships between loading weights and measures of cognitive and functional performance: Logical Memory II, Rey Auditory Verbal Learning Test-long delay, Clinical Dementia Rating sum of boxes and Mini-Mental State Examination. We observed significant differences in loading weights among cognitively normal, mild cognitive impairment and mild Alzheimer's disease dementia groups in 5 of the 18 independent components, as determined by Kruskal-Wallis tests. Only Patterns 1 and 2 demonstrated significant differences in group loading weights after correction for multiple comparisons. Excluding the cognitively normal group, we observed significant correlations between the loading weights for Pattern 1 (left temporal cortex and the cingulate gyrus) and Clinical Dementia Rating sum of boxes (r = -0.54, P = 0.0019), Mini-Mental State Examination (r = 0.48, P = 0.0055) and Logical Memory II score (r = 0.44, P = 0.013). For Pattern 2 (temporal cortices), significant associations were demonstrated between its loading weights and Logical Memory II score (r = 0.34, P = 0.0384). Following false discovery rate correction, only the relationship between the Pattern 1 loading weights with Clinical Dementia Rating sum of boxes (r = -0.54, P = 0.0019) and Mini-Mental State Examination (r = 0.48, P = 0.0055) remained statistically significant. We demonstrated that independent component analysis could define coherent spatial patterns of synaptic density. Furthermore, commonly used measures of cognitive performance correlated significantly with loading weights for two patterns within only the mild cognitive impairment/mild Alzheimer's disease dementia group. This study leverages data-centric approaches to augment the conventional region-of-interest-based methods, revealing distinct patterns that differentiate between mild cognitive impairment and mild Alzheimer's disease dementia, marking a significant advancement in the field.
Collapse
Affiliation(s)
- Xiaotian T Fang
- Yale PET Center, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Nakul R Raval
- Yale PET Center, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ryan S O’Dell
- Alzheimer’s Disease Research Unit, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Mika Naganawa
- Yale PET Center, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Adam P Mecca
- Alzheimer’s Disease Research Unit, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ming-Kai Chen
- Yale PET Center, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Christopher H van Dyck
- Alzheimer’s Disease Research Unit, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Richard E Carson
- Yale PET Center, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
9
|
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
|
10
|
Tan Z, Lin Y, Zhou M, Guo W, Qiu J, Ding L, Wu Z, Xu P, Chen X. Correlation of SV2C rs1423099 single nucleotide polymorphism with sporadic Parkinson's disease in Han population in Southern China. Neurosci Lett 2023; 813:137426. [PMID: 37544580 DOI: 10.1016/j.neulet.2023.137426] [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: 03/15/2023] [Revised: 07/18/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND The synaptic vesicle glycoprotein 2 (SV2) has been implicated in synaptic function throughout the brain. Accumulating evidence investigated that SV2C contributed to dopamine release and the disrupted expression of SV2C was considered to be a unique feature of PD that may facilitate dopaminergic neuron dysfunction. OBJECTIVE This study aimed to examine the relationship between the SV2C rs1423099 single nucleotide polymorphism and sporadic Parkinson's disease (PD) in the Chinese Han population. MATERIALS AND METHODS This study enrolled 351 patients with sporadic PD and 240 normal controls in Chinese Han population. Peripheral blood DNA was extracted by DNA extraction kits and the rs1423099 genotype was analyzed by Agena MassARRAY DNA mass spectrometry. The differences in genotype and allele distribution frequencies between PD patients and control groups were compared using chi-squared tests or Fisher's exact tests. RESULTS No statistical difference was revealed in age and sex distribution between the cases and control groups, and the distribution of genotype and allele frequencies was consistent with the Hardy-Weinberg equilibrium test. In SV2C rs1423099 dominant model, the frequency of the CC/CT genotype was significantly higher in the PD group compared to the control group (OR = 4.065,95% CI: 2.801-10.870, p = 0.002). Nevertheless, in the recessive model, CC or CT/TT genotypes have no statistical difference in the two groups (p = 0.09). Additionally, in allelic analysis, the C allele was investigated to increase the risk of PD (OR = 1.346, 95% CI: 1.036-1.745, p = 0.026); Furthermore, subgroup analysis suggested that those carrying the C allele in the male subgroup were at a higher risk to afflicted with PD (OR = 1.637, 95% CI: 1.147-2.336, p = 0.006). CONCLUSION SV2C rs1423099 single nucleotide polymorphism was associated with sporadic Parkinson's disease in the Chinese Han population, particularly in males.
Collapse
Affiliation(s)
- Zixin Tan
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Yuwan Lin
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Miaomiao Zhou
- Department of Neurology, Shanghai General Hospital, Shanghai 200940, China
| | - Wenyuan Guo
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Jiewen Qiu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Liuyan Ding
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Zhuohua Wu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| | - Pingyi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| | - Xiang Chen
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| |
Collapse
|
11
|
Tan X, Zhang CC, Lu JS, Li ZY, Li BL, Liu XY, Yu YZ, Xu Q. Biology activity and characterization of the functional L-HN fragment derivative of botulinum neurotoxin serotype E. Anaerobe 2023; 82:102764. [PMID: 37479022 DOI: 10.1016/j.anaerobe.2023.102764] [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: 01/19/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/23/2023]
Abstract
OBJECTIVES The mature botulinum neurotoxin (BoNT) is a long peptide chain consisting of a light chain (L) and a heavy chain (H) linked by a disulfide bond, where the heavy chain is divided into a translocation domain and an acceptor binding domain (Hc). In this study, we further explored the biology activity and characteristics of recombinant L-HN fragment (EL-HN) composed of the L and HN domains of BoNT/E in vivo and in vitro. METHODS Neurotoxicity of L-HN fragments from botulinum neurotoxins was assessed in mice. Cleavage of dichain EL-HN in vitro and in neuro-2a cells was assessed and compared with that of single chain EL-HN. Interaction of HN domain and the receptor synaptic vesicle glycoprotein 2C (SV2C) was explored in vitro and in neuro-2a cells only expressing SV2C. RESULTS We found that the 50% mouse lethal dose of the nicked dichain EL-HN fragment (EL-HN-DC) was 0.5 μg and its neurotoxicity was the highest among the L-HN's of the four serotypes of BoNT (A/B/E/F). The cleavage efficiency of EL-HN-DC toward synaptosome associated protein 25 (SNAP25) in vitro was 3-fold higher than that of the single chain at the cellular level, and showed 200-fold higher animal toxicity. The EL-HN-DC fragment might enter neuro-2a cells via binding to SV2C to efficiently cleave SNAP25. CONCLUSIONS The EL-HN fragment showed good biological activities in vivo and in vitro, and could be used as a drug screening model and to further explore the molecular mechanism of its transmembrane transport.
Collapse
Affiliation(s)
- Xiao Tan
- Institute of Life Science and Biotechnology, Beijing Jiaotong University, Beijing, 100044, China; Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Cong-Cong Zhang
- Institute of Life Science and Biotechnology, Beijing Jiaotong University, Beijing, 100044, China
| | - Jian-Sheng Lu
- Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Zhi-Ying Li
- Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Bo-Lin Li
- Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xu-Yang Liu
- Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yun-Zhou Yu
- Beijing Institute of Biotechnology, Beijing, 100071, China.
| | - Qing Xu
- Institute of Life Science and Biotechnology, Beijing Jiaotong University, Beijing, 100044, China.
| |
Collapse
|
12
|
Liu Z, Lee PG, Krez N, Lam KH, Liu H, Przykopanski A, Chen P, Yao G, Zhang S, Tremblay JM, Perry K, Shoemaker CB, Rummel A, Dong M, Jin R. Structural basis for botulinum neurotoxin E recognition of synaptic vesicle protein 2. Nat Commun 2023; 14:2338. [PMID: 37095076 PMCID: PMC10125960 DOI: 10.1038/s41467-023-37860-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 04/03/2023] [Indexed: 04/26/2023] Open
Abstract
Botulinum neurotoxin E (BoNT/E) is one of the major causes of human botulism and paradoxically also a promising therapeutic agent. Here we determined the co-crystal structures of the receptor-binding domain of BoNT/E (HCE) in complex with its neuronal receptor synaptic vesicle glycoprotein 2A (SV2A) and a nanobody that serves as a ganglioside surrogate. These structures reveal that the protein-protein interactions between HCE and SV2 provide the crucial location and specificity information for HCE to recognize SV2A and SV2B, but not the closely related SV2C. At the same time, HCE exploits a separated sialic acid-binding pocket to mediate recognition of an N-glycan of SV2. Structure-based mutagenesis and functional studies demonstrate that both the protein-protein and protein-glycan associations are essential for SV2A-mediated cell entry of BoNT/E and for its potent neurotoxicity. Our studies establish the structural basis to understand the receptor-specificity of BoNT/E and to engineer BoNT/E variants for new clinical applications.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, 92697, USA
| | - Pyung-Gang Lee
- Department of Urology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Nadja Krez
- Institute of Toxicology, Hannover Medical School, Hannover, 30623, Germany
| | - Kwok-Ho Lam
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, 92697, USA
| | - Hao Liu
- Department of Urology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Adina Przykopanski
- Institute of Toxicology, Hannover Medical School, Hannover, 30623, Germany
| | - Peng Chen
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, 92697, USA
| | - Guorui Yao
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, 92697, USA
| | - Sicai Zhang
- Department of Urology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | | | - Kay Perry
- NE-CAT and Department of Chemistry and Chemical Biology, Cornell University, Argonne National Laboratory, Argonne, IL, 60439, USA
| | | | - Andreas Rummel
- Institute of Toxicology, Hannover Medical School, Hannover, 30623, Germany
| | - Min Dong
- Department of Urology, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA.
| | - Rongsheng Jin
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
13
|
Isingrini E, Guinaudie C, Perret L, Guma E, Gorgievski V, Blum ID, Colby-Milley J, Bairachnaya M, Mella S, Adamantidis A, Storch KF, Giros B. Behavioral and Transcriptomic Changes Following Brain-Specific Loss of Noradrenergic Transmission. Biomolecules 2023; 13:biom13030511. [PMID: 36979445 PMCID: PMC10046559 DOI: 10.3390/biom13030511] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/25/2023] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
Noradrenaline (NE) plays an integral role in shaping behavioral outcomes including anxiety/depression, fear, learning and memory, attention and shifting behavior, sleep-wake state, pain, and addiction. However, it is unclear whether dysregulation of NE release is a cause or a consequence of maladaptive orientations of these behaviors, many of which associated with psychiatric disorders. To address this question, we used a unique genetic model in which the brain-specific vesicular monoamine transporter-2 (VMAT2) gene expression was removed in NE-positive neurons disabling NE release in the entire brain. We engineered VMAT2 gene splicing and NE depletion by crossing floxed VMAT2 mice with mice expressing the Cre-recombinase under the dopamine β-hydroxylase (DBH) gene promotor. In this study, we performed a comprehensive behavioral and transcriptomic characterization of the VMAT2DBHcre KO mice to evaluate the role of central NE in behavioral modulations. We demonstrated that NE depletion induces anxiolytic and antidepressant-like effects, improves contextual fear memory, alters shifting behavior, decreases the locomotor response to amphetamine, and induces deeper sleep during the non-rapid eye movement (NREM) phase. In contrast, NE depletion did not affect spatial learning and memory, working memory, response to cocaine, and the architecture of the sleep-wake cycle. Finally, we used this model to identify genes that could be up- or down-regulated in the absence of NE release. We found an up-regulation of the synaptic vesicle glycoprotein 2c (SV2c) gene expression in several brain regions, including the locus coeruleus (LC), and were able to validate this up-regulation as a marker of vulnerability to chronic social defeat. The NE system is a complex and challenging system involved in many behavioral orientations given it brain wide distribution. In our study, we unraveled specific role of NE neurotransmission in multiple behavior and link it to molecular underpinning, opening future direction to understand NE role in health and disease.
Collapse
Affiliation(s)
- Elsa Isingrini
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
- Faculté des Sciences Fondamentales et Biomédicales, Université Paris Cité, INCC UMR 8002, CNRS, F-75006 Paris, France
| | - Chloé Guinaudie
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
| | - Léa Perret
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
| | - Elisa Guma
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
| | - Victor Gorgievski
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
| | - Ian D. Blum
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
| | - Jessica Colby-Milley
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
| | - Maryia Bairachnaya
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
| | - Sébastien Mella
- Cytometry and Biomarkers Platform, Unit of Technology and Service, Institut Pasteur, Université de Paris, F-75015 Paris, France
- Bioinformatics and Biostatistics Hub Platform, Institut Pasteur, Université de Paris, F-75015 Paris, France
| | - Antoine Adamantidis
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, 3010 Bern, Switzerland
- Department of Biomedical Research, University of Bern, 3008 Bern, Switzerland
| | - Kai-Florian Storch
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
| | - Bruno Giros
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
- Faculté des Sciences Fondamentales et Biomédicales, Université Paris Cité, INCC UMR 8002, CNRS, F-75006 Paris, France
- Correspondence:
| |
Collapse
|
14
|
Fang XT, Volpi T, Holmes SE, Esterlis I, Carson RE, Worhunsky PD. Linking resting-state network fluctuations with systems of coherent synaptic density: A multimodal fMRI and 11C-UCB-J PET study. Front Hum Neurosci 2023; 17:1124254. [PMID: 36908710 PMCID: PMC9995441 DOI: 10.3389/fnhum.2023.1124254] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/31/2023] [Indexed: 02/25/2023] Open
Abstract
Introduction: Resting-state network (RSN) connectivity is a widely used measure of the brain's functional organization in health and disease; however, little is known regarding the underlying neurophysiology of RSNs. The aim of the current study was to investigate associations between RSN connectivity and synaptic density assessed using the synaptic vesicle glycoprotein 2A radioligand 11C-UCB-J PET. Methods: Independent component analyses (ICA) were performed on resting-state fMRI and PET data from 34 healthy adult participants (16F, mean age: 46 ± 15 years) to identify a priori RSNs of interest (default-mode, right frontoparietal executive-control, salience, and sensorimotor networks) and select sources of 11C-UCB-J variability (medial prefrontal, striatal, and medial parietal). Pairwise correlations were performed to examine potential intermodal associations between the fractional amplitude of low-frequency fluctuations (fALFF) of RSNs and subject loadings of 11C-UCB-J source networks both locally and along known anatomical and functional pathways. Results: Greater medial prefrontal synaptic density was associated with greater fALFF of the anterior default-mode, posterior default-mode, and executive-control networks. Greater striatal synaptic density was associated with greater fALFF of the anterior default-mode and salience networks. Post-hoc mediation analyses exploring relationships between aging, synaptic density, and RSN activity revealed a significant indirect effect of greater age on fALFF of the anterior default-mode network mediated by the medial prefrontal 11C-UCB-J source. Discussion: RSN functional connectivity may be linked to synaptic architecture through multiple local and circuit-based associations. Findings regarding healthy aging, lower prefrontal synaptic density, and lower default-mode activity provide initial evidence of a neurophysiological link between RSN activity and local synaptic density, which may have relevance in neurodegenerative and psychiatric disorders.
Collapse
Affiliation(s)
- Xiaotian T. Fang
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Tommaso Volpi
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Sophie E. Holmes
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States
| | - Irina Esterlis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States
- Department of Psychology, Yale University, New Haven, CT, United States
| | - Richard E. Carson
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | | |
Collapse
|
15
|
Ng XY, Wu Y, Lin Y, Yaqoob SM, Greene LE, De Camilli P, Cao M. Mutations in Parkinsonism-linked endocytic proteins synaptojanin1 and auxilin have synergistic effects on dopaminergic axonal pathology. NPJ Parkinsons Dis 2023; 9:26. [PMID: 36792618 PMCID: PMC9932162 DOI: 10.1038/s41531-023-00465-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 01/20/2023] [Indexed: 02/17/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by defective dopaminergic (DAergic) input to the striatum. Mutations in two genes encoding synaptically enriched clathrin-uncoating factors, synaptojanin 1 (SJ1) and auxilin, have been implicated in atypical Parkinsonism. SJ1 knock-in (SJ1-KIRQ) mice carrying a disease-linked mutation display neurological manifestations reminiscent of Parkinsonism. Here we report that auxilin knockout (Aux-KO) mice display dystrophic changes of a subset of nigrostriatal DAergic terminals similar to those of SJ1-KIRQ mice. Furthermore, Aux-KO/SJ1-KIRQ double mutant mice have shorter lifespan and more severe synaptic defects than single mutant mice. These include increase in dystrophic striatal nerve terminals positive for DAergic markers and for the PD risk protein SV2C, as well as adaptive changes in striatal interneurons. The synergistic effect of the two mutations demonstrates a special lability of DAergic neurons to defects in clathrin uncoating, with implications for PD pathogenesis in at least some forms of this condition.
Collapse
Affiliation(s)
- Xin Yi Ng
- Programme in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Yumei Wu
- Departments of Neuroscience and Cell Biology, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Youneng Lin
- Programme in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Sidra Mohamed Yaqoob
- Programme in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Lois E Greene
- Laboratory of Cell Biology, NHLBI, National Institutes of Health, Bethesda, MD, USA
| | - Pietro De Camilli
- Departments of Neuroscience and Cell Biology, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Mian Cao
- Programme in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore, Singapore.
- Department of Physiology, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
16
|
Xiao L, Zhou M, Tang Y, Hu S. 18 F-SynVesT-1 positron emission tomography in a hypothalamic hamartoma with abnormal uptake. Epilepsia 2023; 64:e43-e47. [PMID: 36745038 DOI: 10.1111/epi.17533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/07/2023]
Abstract
Hypothalamic hamartomas (HHs) are uncommon benign lesions of neuronal and glial cells in the inferior hypothalamus. They have been linked to epilepsy, premature puberty, and cognitive and behavioral impairment. We report a 13-year-old patient who was referred to a multidisciplinary treatment team for epilepsy with 6 months of convulsive seizures. Sustained seizure control was not achieved despite the use of multiple antiepileptic agents. He had been plagued by unexplained paroxysmal bursts of laughter for >11 years. Video-electroencephalography showed diffuse epileptic discharges prominent in the right hemisphere in both interictal and ictal phases. Magnetic resonance imaging demonstrated an isointense gray matter mass on the right lateral walls of the third ventricle, with focal hypometabolism on 18 F-fluorodeoxyglucose positron emission tomography (PET). The patient subsequently was enrolled in a clinical trial of 18 F-SynVesT-1 PET in epilepsy, and an increased 18 F-SynVesT-1 uptake was noted in the mass. After excluding hormonal abnormalities, the patient underwent open resection targeting HHs. We used 18 F-SynVesT-1 as a specific PET tracer for synaptic vesicle glycoprotein 2A, which is ubiquitously expressed in brain synapses. 18 F-SynVesT-1 PET may hold promise as a supplementary tool in the presurgical localization and evaluation of HHs.
Collapse
Affiliation(s)
- Ling Xiao
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Ming Zhou
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yongxiang Tang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Biological Nanotechnology of the National Health Commission, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
17
|
Dickson E, Dwijesha AS, Andersson N, Lundh S, Björkqvist M, Petersén Å, Soylu-Kucharz R. Microarray profiling of hypothalamic gene expression changes in Huntington's disease mouse models. Front Neurosci 2022; 16:1027269. [PMID: 36408416 PMCID: PMC9671106 DOI: 10.3389/fnins.2022.1027269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/10/2022] [Indexed: 09/11/2024] Open
Abstract
Structural changes and neuropathology in the hypothalamus have been suggested to contribute to the non-motor manifestations of Huntington's disease (HD), a neurodegenerative disorder caused by an expanded cytosine-adenine-guanine (CAG) repeat in the huntingtin (HTT) gene. In this study, we investigated whether hypothalamic HTT expression causes transcriptional changes. Hypothalamic RNA was isolated from two different HD mouse models and their littermate controls; BACHD mice with ubiquitous expression of full-length mutant HTT (mHTT) and wild-type mice with targeted hypothalamic overexpression of either wild-type HTT (wtHTT) or mHTT fragments. The mHTT and wtHTT groups showed the highest number of differentially expressed genes compared to the BACHD mouse model. Gene Set Enrichment Analysis (GSEA) with leading-edge analysis showed that suppressed sterol- and cholesterol metabolism were shared between hypothalamic wtHTT and mHTT overexpression. Most distinctive for mHTT overexpression was the suppression of neuroendocrine networks, in which qRT-PCR validation confirmed significant downregulation of neuropeptides with roles in feeding behavior; hypocretin neuropeptide precursor (Hcrt), tachykinin receptor 3 (Tacr3), cocaine and amphetamine-regulated transcript (Cart) and catecholamine-related biological processes; dopa decarboxylase (Ddc), histidine decarboxylase (Hdc), tyrosine hydroxylase (Th), and vasoactive intestinal peptide (Vip). In BACHD mice, few hypothalamic genes were differentially expressed compared to age-matched WT controls. However, GSEA indicated an enrichment of inflammatory- and gonadotropin-related processes at 10 months. In conclusion, we show that both wtHTT and mHTT overexpression change hypothalamic transcriptome profile, specifically mHTT, altering neuroendocrine circuits. In contrast, the ubiquitous expression of full-length mHTT in the BACHD hypothalamus moderately affects the transcriptomic profile.
Collapse
Affiliation(s)
- Elna Dickson
- Biomarkers in Brain Disease, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Amoolya Sai Dwijesha
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Natalie Andersson
- Pathways of Cancer Cell Evolution, Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Sofia Lundh
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Maria Björkqvist
- Biomarkers in Brain Disease, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Åsa Petersén
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Rana Soylu-Kucharz
- Biomarkers in Brain Disease, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
18
|
Castro PA, Pinto-Borguero I, Yévenes GE, Moraga-Cid G, Fuentealba J. Antiseizure medication in early nervous system development. Ion channels and synaptic proteins as principal targets. Front Pharmacol 2022; 13:948412. [PMID: 36313347 PMCID: PMC9614143 DOI: 10.3389/fphar.2022.948412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/05/2022] [Indexed: 12/04/2022] Open
Abstract
The main strategy for the treatment of epilepsy is the use of pharmacological agents known as antiseizure medication (ASM). These drugs control the seizure onset and improves the life expectancy and quality of life of patients. Several ASMs are contraindicated during pregnancy, due to a potential teratogen risk. For this reason, the pharmacological treatments of the pregnant Women with Epilepsy (WWE) need comprehensive analyses to reduce fetal risk during the first trimester of pregnancy. The mechanisms by which ASM are teratogens are still under study and scientists in the field, propose different hypotheses. One of them, which will be addressed in this review, corresponds to the potential alteration of ASM on ion channels and proteins involved in relevant signaling and cellular responses (i.e., migration, differentiation) during embryonic development. The actual information related to the action of ASM and its possible targets it is poorly understood. In this review, we will focus on describing the eventual presence of some ion channels and synaptic proteins of the neurotransmitter signaling pathways present during early neural development, which could potentially interacting as targets of ASM. This information leads to elucidate whether these drugs would have the ability to affect critical signaling during periods of neural development that in turn could explain the fetal malformations observed by the use of ASM during pregnancy.
Collapse
Affiliation(s)
- Patricio A. Castro
- Laboratory of Physiology and Pharmacology for Neural Development, LAND, Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- *Correspondence: Patricio A. Castro,
| | - Ingrid Pinto-Borguero
- Laboratory of Physiology and Pharmacology for Neural Development, LAND, Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Gonzalo E. Yévenes
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Gustavo Moraga-Cid
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Jorge Fuentealba
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| |
Collapse
|
19
|
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
|
20
|
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: 54] [Impact Index Per Article: 18.0] [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
|
21
|
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
|
22
|
Pazarlar BA, Aripaka SS, Petukhov V, Pinborg L, Khodosevich K, Mikkelsen JD. Expression profile of synaptic vesicle glycoprotein 2A, B, and C paralogues in temporal neocortex tissue from patients with temporal lobe epilepsy (TLE). Mol Brain 2022; 15:45. [PMID: 35578248 PMCID: PMC9109314 DOI: 10.1186/s13041-022-00931-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 05/05/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractSynaptic vesicle glycoprotein-2 (SV2) is a family of proteins consisting of SV2A, SV2B, and SV2C. This protein family has attracted attention in recent years after SV2A was shown to be an epileptic drug target and a perhaps a biomarker of synaptic density. So far, the anatomical localization of these proteins in the rodent and human brain have been reported, but co-expression of SV2 genes on a cellular level, their expressions in the human brain, comparison to radioligand binding, any possible regulation in epilepsy are not known. We have here analyzed the expression of SV2 genes in neuronal subtypes in the temporal neocortex in selected specimens by using single nucleus-RNA sequencing, and performed quantitative PCR in populations of temporal lobe epilepsy (TLE) patients and healthy controls. [3H]-UCB-J autoradiography was performed to analyze the correlation between the mRNA transcript and binding capacity to SV2A. Our data showed that the SV2A transcript is expressed in all glutamatergic and GABAergic cortical subtypes, while SV2B expression is restricted to only the glutamatergic neurons and SV2C has very limited expression in a small subgroup of GABAergic interneurons. The level of [3H]-UCB-J binding and the concentration of SV2A mRNA is strongly correlated in each patient, and the expression is lower in the TLE patients. There is no relationship between SV2A expression and age, sex, seizure frequency, duration of epilepsy, or whether patients were recently treated with levetiracetam or not. Collectively, these findings point out a neuronal subtype-specific distribution of the expression of the three SV2 genes, and the lower levels of both radioligand binding and expression further emphasize the significance of these proteins in this disease.
Collapse
|
23
|
Kim HR, Jung Y, Shin J, Park M, Kweon DH, Ban C. Neuron-recognizable characteristics of peptides recombined using a neuronal binding domain of botulinum neurotoxin. Sci Rep 2022; 12:4980. [PMID: 35322139 PMCID: PMC8943039 DOI: 10.1038/s41598-022-09145-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/14/2022] [Indexed: 11/09/2022] Open
Abstract
Recombinant peptides were designed using the C-terminal domain (receptor binding domain, RBD) and its subdomain (peptide A2) of a heavy chain of botulinum neurotoxin A-type 1 (BoNT/A1), which can bind to the luminal domain of synaptic vesicle glycoprotein 2C (SV2C-LD). Peptide A2- or RBD-containing recombinant peptides linked to an enhanced green fluorescence protein (EGFP) were prepared by expression in Escherichia coli. A pull-down assay using SV2C-LD-covered resins showed that the recombinant peptides for CDC297 BoNT/A1, referred to EGFP-A2' and EGFP-RBD', exhibited ≥ 2.0-times stronger binding affinity to SV2C-LD than those for the wild-type BoNT/A1. Using bio-layer interferometry, an equilibrium dissociation rate constant (KD) of EGFP-RBD' to SV2C-LD was determined to be 5.45 μM, which is 33.87- and 15.67-times smaller than the KD values for EGFP and EGFP-A2', respectively. Based on confocal laser fluorescence micrometric analysis, the adsorption/absorption of EGFP-RBD' to/in differentiated PC-12 cells was 2.49- and 1.29-times faster than those of EGFP and EGFP-A2', respectively. Consequently, the recombinant peptides acquired reasonable neuron-specific binding/internalizing ability through the recruitment of RBD'. In conclusion, RBDs of BoNTs are versatile protein domains that can be used to mark neural systems and treat a range of disorders in neural systems.
Collapse
Affiliation(s)
- Hye Rin Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Younghun Jung
- Department of Integrative Biotechnology, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi, 16419, Republic of Korea.,Institute of Biomolecule Control, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Jonghyeok Shin
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Myungseo Park
- Environmental Health Sciences, School of Public Health, University of Minnesota, Saint Paul, MN, 55108, USA
| | - Dae-Hyuk Kweon
- Department of Integrative Biotechnology, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi, 16419, Republic of Korea. .,Institute of Biomolecule Control, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi, 16419, Republic of Korea. .,Biologics Research Center, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi, 16419, Republic of Korea. .,Interdisciplinary Program in BioCosmetics, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi, 16419, Republic of Korea.
| | - Choongjin Ban
- Department of Environmental Horticulture, University of Seoul, 163 Seoulsiripdaero, Dongdaemun-gu, Seoul, 02504, Republic of Korea.
| |
Collapse
|
24
|
Angarita GA, Worhunsky PD, Naganawa M, Toyonaga T, Nabulsi NB, Li CSR, Esterlis I, Skosnik PD, Radhakrishnan R, Pittman B, Gueorguieva R, Potenza MN, Finnema SJ, Huang Y, Carson RE, Malison RT. Lower prefrontal cortical synaptic vesicle binding in cocaine use disorder: An exploratory 11 C-UCB-J positron emission tomography study in humans. Addict Biol 2022; 27:e13123. [PMID: 34852401 PMCID: PMC8891080 DOI: 10.1111/adb.13123] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/22/2021] [Accepted: 11/12/2021] [Indexed: 12/22/2022]
Abstract
Preclinical studies have revealed robust and long-lasting alterations in dendritic spines in the brain following cocaine exposure. Such alterations are hypothesized to underlie enduring maladaptive behaviours observed in cocaine use disorder (CUD). The current study explored whether synaptic density is altered in CUD. Fifteen individuals with DSM-5 CUD and 15 demographically matched healthy control (HC) subjects participated in a single 11 C-UCB-J positron emission tomography scan to assess density of synaptic vesicle glycoprotein 2A (SV2A). The volume of distribution (VT ) and the plasma-free fraction-corrected form of the total volume of distribution (VT /fP ) were analysed in the anterior cingulate cortex (ACC), dorsomedial and ventromedial prefrontal cortex (PFC), lateral and medial orbitofrontal cortex (OFC) and ventral striatum. A significant diagnostic-group-by-region interaction was observed for VT and VT /fP . Post hoc analyses revealed no differences on VT , while for VT /fP showed lower values in CUD as compared with HC subjects in the ACC (-10.9%, p = 0.02), ventromedial PFC (-9.9%, p = 0.02) and medial OFC (-9.9%, p = 0.04). Regional VT /fP values in CUD, though unrelated to measures of lifetime cocaine use, were positively correlated with the frequency of recent cocaine use (p = 0.02-0.03) and negatively correlated with cocaine abstinence (p = 0.008-0.03). These findings provide initial preliminary in vivo evidence of altered (lower) synaptic density in the PFC of humans with CUD. Cross-sectional variation in SV2A availability as a function of recent cocaine use and abstinence suggests that synaptic density may be dynamically and plastically regulated by acute cocaine, an observation that merits direct testing by studies using more definitive longitudinal designs.
Collapse
Affiliation(s)
- Gustavo A. Angarita
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
- Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519
| | - Patrick D. Worhunsky
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
| | - Mika Naganawa
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, New Haven, CT 06519
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, New Haven, CT 06519
| | - Nabeel B. Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, New Haven, CT 06519
| | - Chiang-Shan R. Li
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
- Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519
| | - Irina Esterlis
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
| | - Patrick D. Skosnik
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
| | - Rajiv Radhakrishnan
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
| | - Brian Pittman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
| | - Ralitza Gueorguieva
- Department of Biostatistics, Yale School of Public Health, 60 College Street, New Haven CT 06520
| | - Marc N. Potenza
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
- Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519
- Connecticut Council on Problem Gambling, 100 Great Meadow Road, Wethersfield, CT 06109
- Child Study Center, Yale University School of Medicine, 234 South Frontage Road, New Haven, CT 06510
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510
| | - Sjoerd J. Finnema
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, New Haven, CT 06519
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, New Haven, CT 06519
| | - Richard E. Carson
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, New Haven, CT 06519
| | - Robert T. Malison
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
- Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519
| |
Collapse
|
25
|
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
|
26
|
In vivo evidence of lower synaptic vesicle density in schizophrenia. Mol Psychiatry 2021; 26:7690-7698. [PMID: 34135473 DOI: 10.1038/s41380-021-01184-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 05/20/2021] [Accepted: 05/28/2021] [Indexed: 02/05/2023]
Abstract
Decreased synaptic spine density has been the most consistently reported postmortem finding in schizophrenia (SCZ). A recently developed in vivo measure of synaptic vesicle density estimated using the novel positron emission tomography (PET) ligand [11C]UCB-J is a proxy measure of synaptic density. In this study we determined whether [11C]UCB-J binding, an in vivo measure of synaptic vesicle density, is altered in SCZ. SCZ patients (n = 13, 3 F) and age-, gender-matched healthy controls (HCs) (n = 15, 3 F) underwent PET imaging using [11C]UCB-J and high-resolution research tomography (HRRT). [11C]UCB-J distribution volume (VT) and binding potential (BPND) were estimated using a 1T model with centrum-semiovale as the reference region. Relative to HCs, SCZ patients, showed significantly lower [11C]UCB-J BPND with significant differences in the frontal cortex (-10%, Cohen's d = 1.01), anterior cingulate (-11%, Cohen's d = 1.24), hippocampus (-15%, Cohen's d = 1.29), occipital cortex (-14%, Cohen's d = 1.34), parietal cortex (-10%, p = 0.03, Cohen's d = 0.85) and temporal cortex (-11%, Cohen's d = 1.23). These differences remained significant after partial volume correction. [11C]UCB-J BPND did not correlate with cumulative antipsychotic exposure or gray-matter volume. Consistent with the postmortem and in vivo findings, synaptic vesicle density is lower across several brain regions in SCZ. Frontal synaptic vesicle density correlated with psychosis symptom severity and cognitive performance on social cognition and processing speed. These findings indicate that [11C]UCB-J PET is a sensitive tool to detect lower synaptic density in SCZ and holds promise for future studies of early detection and disease progression.
Collapse
|
27
|
Knockin mouse models demonstrate differential contributions of synaptotagmin-1 and -2 as receptors for botulinum neurotoxins. PLoS Pathog 2021; 17:e1009994. [PMID: 34662366 PMCID: PMC8553082 DOI: 10.1371/journal.ppat.1009994] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 10/28/2021] [Accepted: 09/30/2021] [Indexed: 12/11/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) are the most potent toxins known and are also utilized to treat a wide range of disorders including muscle spasm, overactive bladder, and pain. BoNTs' ability to target neurons determines their specificity, potency, and therapeutic efficacy. Homologous synaptic vesicle membrane proteins synaptotagmin-1 (Syt1) and synaptotagmin-2 (Syt2) have been identified as receptors for BoNT family members including BoNT/B, DC, and G, but their contributions at physiologically relevant toxin concentrations in vivo have yet to be validated and established. Here we generated two knockin mutant mouse models containing three designed point-mutations that specifically disrupt BoNT binding in endogenous Syt1 or Syt2, respectively. Utilizing digit abduction score assay by injecting toxins into the leg muscle, we found that Syt1 mutant mice showed similar sensitivity as the wild type mice, whereas Syt2 mutant mice showed reduced sensitivity to BoNT/B, DC, and G, demonstrating that Syt2 is the dominant receptor at skeletal neuromuscular junctions. We further developed an in vivo bladder injection assay for analyzing BoNT action on bladder tissues and demonstrated that Syt1 is the dominant toxin receptor in autonomic nerves controlling bladder tissues. These findings establish the critical role of protein receptors for the potency and specificity of BoNTs in vivo and demonstrate the differential contributions of Syt1 and Syt2 in two sets of clinically relevant target tissues.
Collapse
|
28
|
Bertoglio D, Verhaeghe J, Wyffels L, Miranda A, Stroobants S, Mrzljak L, Dominguez C, Skinbjerg M, Bard J, Liu L, Munoz-Sanjuan I, Staelens S. Synaptic vesicle glycoprotein 2A is affected in the CNS of Huntington's Disease mice and post-mortem human HD brain. J Nucl Med 2021; 63:942-947. [PMID: 34531262 DOI: 10.2967/jnumed.121.262709] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/26/2021] [Indexed: 11/16/2022] Open
Abstract
Synaptic dysfunction is a primary mechanism underlying Huntington's Disease (HD) progression. This study investigated changes in synaptic vesicle glycoprotein 2A (SV2A) density by means of 11C-UCB-J microPET imaging in the central nervous system (CNS) of HD mice. METHODS: Dynamic 11C-UCB-J microPET imaging was performed at clinically relevant disease stages (at 3, 7, 10, and 16 months, M) in the heterozygous knock-in Q175DN mouse model of HD and WT littermates (n = 16-18/genotype and time point). Cerebral 11C-UCB-J analyses were performed to assess genotypic differences during pre-symptomatic (3M) and symptomatic (7-16M) disease stages. 11C-UCB-J binding in the spinal cord was quantified at 16M. 3H-UCB-J autoradiography and SV2A immunofluorescence were performed post-mortem in mouse and human brain tissue. RESULTS: 11C-UCB-J binding was declined in symptomatic heterozygous mice compared to WT littermates in parallel with disease progression (7M: p<0.01, 16M: p<0.0001). Specific 11C-UCB-J binding was detectable in the spinal cord, with symptomatic heterozygous mice displaying a significant reduction (p<0.0001). 3H-UCB-J autoradiography and SV2A immunofluorescence corroborated the in vivo measurements demonstrating lowered SV2A in heterozygous mice (p<0.05). Finally, preliminary analysis of SV2A in post-mortem human brain suggested lower SV2A in HD gene carrier compared to nondemented control. CONCLUSION: 11C-UCB-J PET detects SV2A deficits during symptomatic disease in heterozygous mice in both brain and spinal cord, offering a novel marker of synaptic integrity widely distributed in CNS. Upon clinical application, 11C-UCB-J PET imaging yields promise for SV2A measurement in patients with HD during disease progression and following disease-modifying therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Longbin Liu
- CHDI Management/CHDI Foundation, United States
| | | | | |
Collapse
|
29
|
Fang XT, Toyonaga T, Hillmer AT, Matuskey D, Holmes SE, Radhakrishnan R, Mecca AP, van Dyck CH, D’Souza DC, Esterlis I, Worhunsky PD, Carson RE. Identifying brain networks in synaptic density PET ( 11C-UCB-J) with independent component analysis. Neuroimage 2021; 237:118167. [PMID: 34000404 PMCID: PMC8452380 DOI: 10.1016/j.neuroimage.2021.118167] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/27/2021] [Accepted: 05/11/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The human brain is inherently organized into distinct networks, as reported widely by resting-state functional magnetic resonance imaging (rs-fMRI), which are based on blood-oxygen-level-dependent (BOLD) signal fluctuations. 11C-UCB-J PET maps synaptic density via synaptic vesicle protein 2A, which is a more direct structural measure underlying brain networks than BOLD rs-fMRI. METHODS The aim of this study was to identify maximally independent brain source networks, i.e., "spatial patterns with common covariance across subjects", in 11C-UCB-J data using independent component analysis (ICA), a data-driven analysis method. Using a population of 80 healthy controls, we applied ICA to two 40-sample subsets and compared source network replication across samples. We examined the identified source networks at multiple model orders, as the ideal number of maximally independent components (IC) is unknown. In addition, we investigated the relationship between the strength of the loading weights for each source network and age and sex. RESULTS Thirteen source networks replicated across both samples. We determined that a model order of 18 components provided stable, replicable components, whereas estimations above 18 were not stable. Effects of sex were found in two ICs. Nine ICs showed age-related change, with 4 remaining significant after correction for multiple comparison. CONCLUSION This study provides the first evidence that human brain synaptic density can be characterized into organized covariance patterns. Furthermore, we demonstrated that multiple synaptic density source networks are associated with age, which supports the potential utility of ICA to identify biologically relevant synaptic density source networks.
Collapse
Affiliation(s)
- Xiaotian T. Fang
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University, 801 Howard Avenue, New Haven, CT 06520, USA,Corresponding author. (X.T. Fang)
| | - Takuya Toyonaga
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University, 801 Howard Avenue, New Haven, CT 06520, USA
| | - Ansel T. Hillmer
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University, 801 Howard Avenue, New Haven, CT 06520, USA,Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - David Matuskey
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University, 801 Howard Avenue, New Haven, CT 06520, USA,Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA,Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Sophie E. Holmes
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | | | - Adam P. Mecca
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Christopher H. van Dyck
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA,Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | | | - Irina Esterlis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | | | - Richard E. Carson
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University, 801 Howard Avenue, New Haven, CT 06520, USA
| |
Collapse
|
30
|
Shemesh N, Jubran J, Dror S, Simonovsky E, Basha O, Argov C, Hekselman I, Abu-Qarn M, Vinogradov E, Mauer O, Tiago T, Carra S, Ben-Zvi A, Yeger-Lotem E. The landscape of molecular chaperones across human tissues reveals a layered architecture of core and variable chaperones. Nat Commun 2021; 12:2180. [PMID: 33846299 PMCID: PMC8042005 DOI: 10.1038/s41467-021-22369-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
The sensitivity of the protein-folding environment to chaperone disruption can be highly tissue-specific. Yet, the organization of the chaperone system across physiological human tissues has received little attention. Through computational analyses of large-scale tissue transcriptomes, we unveil that the chaperone system is composed of core elements that are uniformly expressed across tissues, and variable elements that are differentially expressed to fit with tissue-specific requirements. We demonstrate via a proteomic analysis that the muscle-specific signature is functional and conserved. Core chaperones are significantly more abundant across tissues and more important for cell survival than variable chaperones. Together with variable chaperones, they form tissue-specific functional networks. Analysis of human organ development and aging brain transcriptomes reveals that these functional networks are established in development and decline with age. In this work, we expand the known functional organization of de novo versus stress-inducible eukaryotic chaperones into a layered core-variable architecture in multi-cellular organisms.
Collapse
Affiliation(s)
- Netta Shemesh
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Juman Jubran
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Shiran Dror
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Eyal Simonovsky
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Omer Basha
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Chanan Argov
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Idan Hekselman
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Mehtap Abu-Qarn
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ekaterina Vinogradov
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Omry Mauer
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Tatiana Tiago
- Centre for Neuroscience and Nanotechnology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Serena Carra
- Centre for Neuroscience and Nanotechnology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Anat Ben-Zvi
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| | - Esti Yeger-Lotem
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| |
Collapse
|
31
|
Goutal S, Guillermier M, Becker G, Gaudin M, Bramoullé Y, Luxen A, Lemaire C, Plenevaux A, Salmon E, Hantraye P, Barret O, Van Camp N. The pharmacokinetics of [ 18F]UCB-H revisited in the healthy non-human primate brain. EJNMMI Res 2021; 11:36. [PMID: 33826008 PMCID: PMC8026785 DOI: 10.1186/s13550-021-00777-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/22/2021] [Indexed: 12/29/2022] Open
Abstract
Background Positron Emission Tomography (PET) imaging of the Synaptic Vesicle glycoprotein (SV) 2A is a new tool to quantify synaptic density. [18F]UCB-H was one of the first promising SV2A-ligands to be labelled and used in vivo in rodent and human, while limited information on its pharmacokinetic properties is available in the non-human primate. Here, we evaluate the reliability of the three most commonly used modelling approaches for [18F]UCB-H in the non-human cynomolgus primate, adding the coupled fit of the non-displaceable distribution volume (VND) as an alternative approach to improve unstable fit. The results are discussed in the light of the current state of SV2A PET ligands. Results [18F]UCB-H pharmacokinetic data was optimally fitted with a two-compartment model (2TCM), although the model did not always converge (large total volume of distribution (VT) or large uncertainty of the estimate). 2TCM with coupled fit K1/k2 across brain regions stabilized the quantification, and confirmed a lower specific signal of [18F]UCB-H compared to the newest SV2A-ligands. However, the measures of VND and the influx parameter (K1) are similar to what has been reported for other SV2A ligands. These data were reinforced by displacement studies using [19F]UCB-H, demonstrating only 50% displacement of the total [18F]UCB-H signal at maximal occupancy of SV2A. As previously demonstrated in clinical studies, the graphical method of Logan provided a more robust estimate of VT with only a small bias compared to 2TCM. Conclusions Modeling issues with a 2TCM due to a slow component have previously been reported for other SV2A ligands with low specific binding, or after blocking of specific binding. As all SV2A ligands share chemical structural similarities, we hypothesize that this slow binding component is common for all SV2A ligands, but only hampers quantification when specific binding is low. Supplementary Information The online version contains supplementary material available at 10.1186/s13550-021-00777-8.
Collapse
Affiliation(s)
- Sébastien Goutal
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire Des Maladies Neurodégénératives, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
| | - Martine Guillermier
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire Des Maladies Neurodégénératives, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
| | - Guillaume Becker
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liege, Allee du 6 Aout, 8, Sart Tilman B30, 4000, Liege, Belgium
| | - Mylène Gaudin
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire Des Maladies Neurodégénératives, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
| | - Yann Bramoullé
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire Des Maladies Neurodégénératives, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
| | - André Luxen
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liege, Allee du 6 Aout, 8, Sart Tilman B30, 4000, Liege, Belgium
| | - Christian Lemaire
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liege, Allee du 6 Aout, 8, Sart Tilman B30, 4000, Liege, Belgium
| | - Alain Plenevaux
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liege, Allee du 6 Aout, 8, Sart Tilman B30, 4000, Liege, Belgium
| | - Eric Salmon
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liege, Allee du 6 Aout, 8, Sart Tilman B30, 4000, Liege, Belgium
| | - Philippe Hantraye
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire Des Maladies Neurodégénératives, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
| | - Olivier Barret
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire Des Maladies Neurodégénératives, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
| | - Nadja Van Camp
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire Des Maladies Neurodégénératives, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France.
| |
Collapse
|
32
|
Sun L, Bai D, Lin M, Eerdenidalai, Zhang L, Wang F, Jin S. miR-96 Inhibits SV2C to Promote Depression-Like Behavior and Memory Disorders in Mice. Front Behav Neurosci 2021; 14:575345. [PMID: 33815074 PMCID: PMC8017146 DOI: 10.3389/fnbeh.2020.575345] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 12/29/2020] [Indexed: 11/17/2022] Open
Abstract
Accumulating evidence continues to emphasize the role of microRNAs as significant contributors to depression-like behavior and memory disorders. The current study aimed to investigate the mechanism by which miR-96 influences depression-like behavior and memory deficit in mice. A depression-like behavior and memory disorder mouse model was initially established by means of intraperitoneal injection with lipopolysaccharide. Memory deficits in the mice were evaluated using the Novel Object Recognition Test and Morris water maze experiments, whereas the Sucrose Preference Experiment and forced swimming experiments were performed to identify depression-like behavior in mice. The levels of tumor necrosis factor-α, malondialdehyde, superoxide dismutase, glutathione, and the monoamine transmitters 5-hydroxytryptamine and dopamine were subsequently detected in the serum. Reverse transcription-quantitative polymerase chain reaction and Western blot analysis evaluated the expression of miR-96 and SV2C expression in the CA1 hippocampal region of the mice. Finally, the relationship of miR-96 and SV2C was verified by dual-luciferase reporter gene assay. Our data indicated that the expression of miR-96 was increased, whereas that of SV2C was decreased in the CA1 region of mice exhibiting depression-like behavior and memory impairment. When miR-96 was downregulated or SV2C was overexpressed via intra-cerebroventricular injection with a miR-96 antagonist (miR-96 antagomir) or overexpression of SV2C vector, the Novel Object Recognition Test and sucrose preference index were increased, whereas the escape latency, the number of water maze platform crossings, and the immobility time of the mice were decreased. The serum levels of tumor necrosis factor-α, interleukin-1β, and malondialdehyde in the mouse CA1 region of mice were reduced, whereas the levels of superoxide dismutase and glutathione were elevated after the downregulation of miR-96 or overexpression of SV2C. Collectively, our study demonstrates that miR-96 negatively regulates the expression of SV2C, which consequently leads to depression-like behavior and memory impairment in mice. Our findings highlight the potential of miR-96-targeted therapeutics.
Collapse
Affiliation(s)
- Lidong Sun
- Outpatient Department, Ordos Fourth People's Hospital, Ordos, China
| | - Donghao Bai
- Outpatient Department, Ordos Fourth People's Hospital, Ordos, China
| | - Maoguang Lin
- Outpatient Department, Ordos Fourth People's Hospital, Ordos, China
| | - Eerdenidalai
- Outpatient Department, Ordos Fourth People's Hospital, Ordos, China
| | - Li Zhang
- Outpatient Department, Ordos Fourth People's Hospital, Ordos, China
| | - Fengzhen Wang
- Outpatient Department, Ordos Fourth People's Hospital, Ordos, China
| | - Shangwu Jin
- Clinical Laboratory, Ordos Fourth People's Hospital, Ordos, China
| |
Collapse
|
33
|
Gcwensa NZ, Russell DL, Cowell RM, Volpicelli-Daley LA. Molecular Mechanisms Underlying Synaptic and Axon Degeneration in Parkinson's Disease. Front Cell Neurosci 2021; 15:626128. [PMID: 33737866 PMCID: PMC7960781 DOI: 10.3389/fncel.2021.626128] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/05/2021] [Indexed: 01/13/2023] Open
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disease that impairs movement as well as causing multiple other symptoms such as autonomic dysfunction, rapid eye movement (REM) sleep behavior disorder, hyposmia, and cognitive changes. Loss of dopamine neurons in the substantia nigra pars compacta (SNc) and loss of dopamine terminals in the striatum contribute to characteristic motor features. Although therapies ease the symptoms of PD, there are no treatments to slow its progression. Accumulating evidence suggests that synaptic impairments and axonal degeneration precede neuronal cell body loss. Early synaptic changes may be a target to prevent disease onset and slow progression. Imaging of PD patients with radioligands, post-mortem pathologic studies in sporadic PD patients, and animal models of PD demonstrate abnormalities in presynaptic terminals as well as postsynaptic dendritic spines. Dopaminergic and excitatory synapses are substantially reduced in PD, and whether other neuronal subtypes show synaptic defects remains relatively unexplored. Genetic studies implicate several genes that play a role at the synapse, providing additional support for synaptic dysfunction in PD. In this review article we: (1) provide evidence for synaptic defects occurring in PD before neuron death; (2) describe the main genes implicated in PD that could contribute to synapse dysfunction; and (3) show correlations between the expression of Snca mRNA and mouse homologs of PD GWAS genes demonstrating selective enrichment of Snca and synaptic genes in dopaminergic, excitatory and cholinergic neurons. Altogether, these findings highlight the need for novel therapeutics targeting the synapse and suggest that future studies should explore the roles for PD-implicated genes across multiple neuron types and circuits.
Collapse
Affiliation(s)
- Nolwazi Z Gcwensa
- Department of Neurobiology, Center for Neurodegeneration and Experimental Therapeutics, Civitan International Research Center, Birmingham, AL, United States
| | - Drèson L Russell
- Department of Neurobiology, Center for Neurodegeneration and Experimental Therapeutics, Civitan International Research Center, Birmingham, AL, United States
| | - Rita M Cowell
- Department of Neuroscience, Southern Research, Birmingham, AL, United States
| | - Laura A Volpicelli-Daley
- Department of Neurobiology, Center for Neurodegeneration and Experimental Therapeutics, Civitan International Research Center, Birmingham, AL, United States
| |
Collapse
|
34
|
Lekholm E, Ceder MM, Forsberg EC, Schiöth HB, Fredriksson R. Differentiation of two human neuroblastoma cell lines alters SV2 expression patterns. Cell Mol Biol Lett 2021; 26:5. [PMID: 33588752 PMCID: PMC7885392 DOI: 10.1186/s11658-020-00243-8] [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: 07/08/2020] [Accepted: 12/07/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The synaptic vesicle glycoprotein 2 (SV2) family is essential to the synaptic machinery involved in neurotransmission and vesicle recycling. The isoforms SV2A, SV2B and SV2C are implicated in neurological diseases such as epilepsy, Alzheimer's and Parkinson's disease. Suitable cell systems for studying regulation of these proteins are essential. Here we present gene expression data of SV2A, SV2B and SV2C in two human neuroblastoma cell lines after differentiation. METHODS Human neuroblastoma cell lines SiMa and IMR-32 were treated for seven days with growth supplements (B-27 and N-2), all-trans-retinoic acid (ATRA) or vasoactive intestinal peptide (VIP) and gene expression levels of SV2 and neuronal targets were analyzed. RESULTS The two cell lines reacted differently to the treatments, and only one of the three SV2 isoforms was affected at a time. SV2B and choline O-acetyltransferase (CHAT) expression was changed in concert after growth supplement treatment, decreasing in SiMa cells while increasing in IMR-32. ATRA treatment resulted in no detected changes in SV2 expression in either cell line while VIP increased both SV2C and dopamine transporter (DAT) in IMR-32 cells. CONCLUSION The synergistic expression patterns between SV2B and CHAT as well as between SV2C and DAT mirror the connectivity between these targets found in disease models and knock-out animals, although here no genetic alteration was made. These cell lines and differentiation treatments could possibly be used to study SV2 regulation and function.
Collapse
Affiliation(s)
- Emilia Lekholm
- Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden. .,Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.
| | - Mikaela M Ceder
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Erica C Forsberg
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Helgi B Schiöth
- Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden.,Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Robert Fredriksson
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
35
|
Goud NS, Bhattacharya A, Joshi RK, Nagaraj C, Bharath RD, Kumar P. Carbon-11: Radiochemistry and Target-Based PET Molecular Imaging Applications in Oncology, Cardiology, and Neurology. J Med Chem 2021; 64:1223-1259. [PMID: 33499603 DOI: 10.1021/acs.jmedchem.0c01053] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The positron emission tomography (PET) molecular imaging technique has gained its universal value as a remarkable tool for medical diagnosis and biomedical research. Carbon-11 is one of the promising radiotracers that can report target-specific information related to its pharmacology and physiology to understand the disease status. Currently, many of the available carbon-11 (t1/2 = 20.4 min) PET radiotracers are heterocyclic derivatives that have been synthesized using carbon-11 inserted different functional groups obtained from primary and secondary carbon-11 precursors. A spectrum of carbon-11 PET radiotracers has been developed against many of the upregulated and emerging targets for the diagnosis, prognosis, prediction, and therapy in the fields of oncology, cardiology, and neurology. This review focuses on the carbon-11 radiochemistry and various target-specific PET molecular imaging agents used in tumor, heart, brain, and neuroinflammatory disease imaging along with its associated pathology.
Collapse
Affiliation(s)
- Nerella Sridhar Goud
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Ahana Bhattacharya
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Raman Kumar Joshi
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Chandana Nagaraj
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Rose Dawn Bharath
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Pardeep Kumar
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| |
Collapse
|
36
|
Hirano AA, Vuong HE, Kornmann HL, Schietroma C, Stella SL, Barnes S, Brecha NC. Vesicular Release of GABA by Mammalian Horizontal Cells Mediates Inhibitory Output to Photoreceptors. Front Cell Neurosci 2020; 14:600777. [PMID: 33335476 PMCID: PMC7735995 DOI: 10.3389/fncel.2020.600777] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Abstract
Feedback inhibition by horizontal cells regulates rod and cone photoreceptor calcium channels that control their release of the neurotransmitter glutamate. This inhibition contributes to synaptic gain control and the formation of the center-surround antagonistic receptive fields passed on to all downstream neurons, which is important for contrast sensitivity and color opponency in vision. In contrast to the plasmalemmal GABA transporter found in non-mammalian horizontal cells, there is evidence that the mechanism by which mammalian horizontal cells inhibit photoreceptors involves the vesicular release of the inhibitory neurotransmitter GABA. Historically, inconsistent findings of GABA and its biosynthetic enzyme, L-glutamate decarboxylase (GAD) in horizontal cells, and the apparent lack of surround response block by GABAergic agents diminished support for GABA's role in feedback inhibition. However, the immunolocalization of the vesicular GABA transporter (VGAT) in the dendritic and axonal endings of horizontal cells that innervate photoreceptor terminals suggested GABA was released via vesicular exocytosis. To test the idea that GABA is released from vesicles, we localized GABA and GAD, multiple SNARE complex proteins, synaptic vesicle proteins, and Cav channels that mediate exocytosis to horizontal cell dendritic tips and axonal terminals. To address the perceived relative paucity of synaptic vesicles in horizontal cell endings, we used conical electron tomography on mouse and guinea pig retinas that revealed small, clear-core vesicles, along with a few clathrin-coated vesicles and endosomes in horizontal cell processes within photoreceptor terminals. Some small-diameter vesicles were adjacent to the plasma membrane and plasma membrane specializations. To assess vesicular release, a functional assay involving incubation of retinal slices in luminal VGAT-C antibodies demonstrated vesicles fused with the membrane in a depolarization- and calcium-dependent manner, and these labeled vesicles can fuse multiple times. Finally, targeted elimination of VGAT in horizontal cells resulted in a loss of tonic, autaptic GABA currents, and of inhibitory feedback modulation of the cone photoreceptor Cai, consistent with the elimination of GABA release from horizontal cell endings. These results in mammalian retina identify the central role of vesicular release of GABA from horizontal cells in the feedback inhibition of photoreceptors.
Collapse
Affiliation(s)
- Arlene A. Hirano
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Helen E. Vuong
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Helen L. Kornmann
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Cataldo Schietroma
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Salvatore L. Stella
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Steven Barnes
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Doheny Eye Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nicholas C. Brecha
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
37
|
Oliver EE, Hughes EK, Puckett MK, Chen R, Lowther WT, Howlett AC. Cannabinoid Receptor Interacting Protein 1a (CRIP1a) in Health and Disease. Biomolecules 2020; 10:biom10121609. [PMID: 33261012 PMCID: PMC7761089 DOI: 10.3390/biom10121609] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/15/2022] Open
Abstract
Endocannabinoid signaling depends upon the CB1 and CB2 cannabinoid receptors, their endogenous ligands anandamide and 2-arachidonoylglycerol, and intracellular proteins that mediate responses via the C-terminal and other intracellular receptor domains. The CB1 receptor regulates and is regulated by associated G proteins predominantly of the Gi/o subtypes, β-arrestins 1 and 2, and the cannabinoid receptor-interacting protein 1a (CRIP1a). Evidence for a physiological role for CRIP1a is emerging as data regarding the cellular localization and function of CRIP1a are generated. Here we summarize the neuronal distribution and role of CRIP1a in endocannabinoid signaling, as well as discuss investigations linking CRIP1a to development, vision and hearing sensory systems, hippocampus and seizure regulation, and psychiatric disorders including schizophrenia. We also examine the genetic and epigenetic association of CRIP1a within a variety of cancer subtypes. This review provides evidence upon which to base future investigations on the function of CRIP1a in health and disease.
Collapse
Affiliation(s)
- Emily E. Oliver
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC 20157, USA; (E.E.O.); (E.K.H.); (M.K.P.); (R.C.)
- Department of Biochemistry and Center for Structural Biology, Wake Forest School of Medicine, Winston-Salem, NC 20157, USA;
| | - Erin K. Hughes
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC 20157, USA; (E.E.O.); (E.K.H.); (M.K.P.); (R.C.)
- Department of Biochemistry and Center for Structural Biology, Wake Forest School of Medicine, Winston-Salem, NC 20157, USA;
| | - Meaghan K. Puckett
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC 20157, USA; (E.E.O.); (E.K.H.); (M.K.P.); (R.C.)
| | - Rong Chen
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC 20157, USA; (E.E.O.); (E.K.H.); (M.K.P.); (R.C.)
| | - W. Todd Lowther
- Department of Biochemistry and Center for Structural Biology, Wake Forest School of Medicine, Winston-Salem, NC 20157, USA;
| | - Allyn C. Howlett
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC 20157, USA; (E.E.O.); (E.K.H.); (M.K.P.); (R.C.)
- Correspondence: ; Tel.: +1-336-716-8545
| |
Collapse
|
38
|
Cheng J, Liu HP, Lin WY, Tsai FJ. Identification of contributing genes of Huntington's disease by machine learning. BMC Med Genomics 2020; 13:176. [PMID: 33228685 PMCID: PMC7684976 DOI: 10.1186/s12920-020-00822-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Background Huntington’s disease (HD) is an inherited disorder caused by the polyglutamine (poly-Q) mutations of the HTT gene results in neurodegeneration characterized by chorea, loss of coordination, cognitive decline. However, HD pathogenesis is still elusive. Despite the availability of a wide range of biological data, a comprehensive understanding of HD’s mechanism from machine learning is so far unrealized, majorly due to the lack of needed data density.
Methods To harness the knowledge of the HD pathogenesis from the expression profiles of postmortem prefrontal cortex samples of 157 HD and 157 controls, we used gene profiling ranking as the criteria to reduce the dimension to the order of magnitude of the sample size, followed by machine learning using the decision tree, rule induction, random forest, and generalized linear model. Results These four Machine learning models identified 66 potential HD-contributing genes, with the cross-validated accuracy of 90.79 ± 4.57%, 89.49 ± 5.20%, 90.45 ± 4.24%, and 97.46 ± 3.26%, respectively. The identified genes enriched the gene ontology of transcriptional regulation, inflammatory response, neuron projection, and the cytoskeleton. Moreover, three genes in the cognitive, sensory, and perceptual systems were also identified. Conclusions The mutant HTT may interfere with both the expression and transport of these identified genes to promote the HD pathogenesis.
Collapse
Affiliation(s)
- Jack Cheng
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan.,Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Hsin-Ping Liu
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Wei-Yong Lin
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan. .,Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan. .,Brain Diseases Research Center, China Medical University, Taichung, 40402, Taiwan.
| | - Fuu-Jen Tsai
- Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan. .,School of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan. .,Department of Biotechnology, Asia University, Taichung, 41354, Taiwan. .,Children's Medical Center, China Medical University Hospital, Taichung, 40447, Taiwan.
| |
Collapse
|
39
|
Delva A, Van Weehaeghe D, Koole M, Van Laere K, Vandenberghe W. Loss of Presynaptic Terminal Integrity in the Substantia Nigra in Early Parkinson's Disease. Mov Disord 2020; 35:1977-1986. [PMID: 32767618 DOI: 10.1002/mds.28216] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/26/2020] [Accepted: 06/22/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND It has been hypothesized that the pathology of Parkinson's disease (PD) primarily affects presynaptic terminals and spreads trans-synaptically. OBJECTIVES The main objective of this study was to assess the magnitude and anatomical extent of presynaptic terminal loss across the brain in early PD. A second objective was to compare loss of presynaptic terminals and cell bodies within the nigrostriatal tract. METHODS A total of 30 patients with early PD and 20 age- and gender-matched healthy controls underwent positron emission tomography with 11 C-UCB-J, a ligand for the universal presynaptic terminal marker synaptic vesicle protein 2A (SV2A), and with the dopamine transporter ligand 18 F-FE-PE2I, as well as a detailed clinical assessment. Volumes of interest were delineated based on individual 3-dimensional T1 magnetic resonance imaging. BPND images were calculated. RESULTS Patients with PD showed significant loss of SV2A binding in the substantia nigra only. Loss of dopamine transporter binding in the PD group was much greater in the putamen than in the substantia nigra. We found no correlations between SV2A or dopamine transporter binding and any of the clinical motor or nonmotor scores. Homologous voxel-based analysis in the PD group showed significant correlations between SV2A and dopamine transporter binding in the caudate and substantia nigra. CONCLUSIONS Presynaptic terminals appear to be the most heavily affected subcellular compartment of nigrostriatal neurons in early PD. Moreover, early PD causes loss of presynaptic terminals that innervate the nigrostriatal neurons. This loss of presynaptic boutons in the substantia nigra may reflect an axonal response to target deprivation or could possibly point to a trans-synaptic mode of propagation of the disease process. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Aline Delva
- Department of Neurosciences, KU Leuven, Flanders, Belgium.,Department of Neurology, University Hospitals Leuven, Flanders, Belgium
| | - Donatienne Van Weehaeghe
- Division of Nuclear Medicine, University Hospitals Leuven, Flanders, Belgium.,Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Flanders, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Flanders, Belgium
| | - Koen Van Laere
- Division of Nuclear Medicine, University Hospitals Leuven, Flanders, Belgium.,Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Flanders, Belgium
| | - Wim Vandenberghe
- Department of Neurosciences, KU Leuven, Flanders, Belgium.,Department of Neurology, University Hospitals Leuven, Flanders, Belgium
| |
Collapse
|
40
|
Bertoglio D, Verhaeghe J, Miranda A, Kertesz I, Cybulska K, Korat Š, Wyffels L, Stroobants S, Mrzljak L, Dominguez C, Liu L, Skinbjerg M, Munoz-Sanjuan I, Staelens S. Validation and noninvasive kinetic modeling of [ 11C]UCB-J PET imaging in mice. J Cereb Blood Flow Metab 2020; 40:1351-1362. [PMID: 31307287 PMCID: PMC7232782 DOI: 10.1177/0271678x19864081] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Synaptic pathology is associated with several brain disorders, thus positron emission tomography (PET) imaging of synaptic vesicle glycoprotein 2A (SV2A) using the radioligand [11C]UCB-J may provide a tool to measure synaptic alterations. Given the pivotal role of mouse models in understanding neuropsychiatric and neurodegenerative disorders, this study aims to validate and characterize [11C]UCB-J in mice. We performed a blocking study to verify the specificity of the radiotracer to SV2A, examined kinetic models using an image-derived input function (IDIF) for quantification of the radiotracer, and investigated the in vivo metabolism. Regional TACs during baseline showed rapid uptake of [11C]UCB-J into the brain. Pretreatment with levetiracetam confirmed target engagement in a dose-dependent manner. VT (IDIF) values estimated with one- and two-tissue compartmental models (1TCM and 2TCM) were highly comparable (r=0.999, p < 0.0001), with 1TCM performing better than 2TCM for K1 (IDIF). A scan duration of 60 min was sufficient for reliable VT (IDIF) and K1 (IDIF) estimations. In vivo metabolism of [11C]UCB-J was relatively rapid, with a parent fraction of 22.5 ± 4.2% at 15 min p.i. In conclusion, our findings show that [11C]UCB-J selectively binds to SV2A with optimal kinetics in the mouse representing a promising tool to noninvasively quantify synaptic density in comparative or therapeutic studies in neuropsychiatric and neurodegenerative disorder models.
Collapse
Affiliation(s)
- Daniele Bertoglio
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
| | - Jeroen Verhaeghe
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
| | - Alan Miranda
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
| | - Istvan Kertesz
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium.,Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Klaudia Cybulska
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium.,Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Špela Korat
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium.,Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Leonie Wyffels
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium.,Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium.,Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | | | | | - Longbin Liu
- CHDI Management/CHDI Foundation, Los Angeles, CA, USA
| | | | | | - Steven Staelens
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
41
|
Foo JN, Chew EGY, Chung SJ, Peng R, Blauwendraat C, Nalls MA, Mok KY, Satake W, Toda T, Chao Y, Tan LCS, Tandiono M, Lian MM, Ng EY, Prakash KM, Au WL, Meah WY, Mok SQ, Annuar AA, Chan AYY, Chen L, Chen Y, Jeon BS, Jiang L, Lim JL, Lin JJ, Liu C, Mao C, Mok V, Pei Z, Shang HF, Shi CH, Song K, Tan AH, Wu YR, Xu YM, Xu R, Yan Y, Yang J, Zhang B, Koh WP, Lim SY, Khor CC, Liu J, Tan EK. Identification of Risk Loci for Parkinson Disease in Asians and Comparison of Risk Between Asians and Europeans: A Genome-Wide Association Study. JAMA Neurol 2020; 77:746-754. [PMID: 32310270 PMCID: PMC7171584 DOI: 10.1001/jamaneurol.2020.0428] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/03/2020] [Indexed: 02/05/2023]
Abstract
Importance Large-scale genome-wide association studies in the European population have identified 90 risk variants associated with Parkinson disease (PD); however, there are limited studies in the largest population worldwide (ie, Asian). Objectives To identify novel genome-wide significant loci for PD in Asian individuals and to compare genetic risk between Asian and European cohorts. Design Setting, and Participants Genome-wide association data generated from PD cases and controls in an Asian population (ie, Singapore/Malaysia, Hong Kong, Taiwan, mainland China, and South Korea) were collected from January 1, 2016, to December 31, 2018, as part of an ongoing study. Results were combined with inverse variance meta-analysis, and replication of top loci in European and Japanese samples was performed. Discovery samples of 31 575 individuals passing quality control of 35 994 recruited were used, with a greater than 90% participation rate. A replication cohort of 1 926 361 European-ancestry and 3509 Japanese samples was analyzed. Parkinson disease was diagnosed using UK Parkinson's Disease Society Brain Bank Criteria. Main Outcomes and Measures Genotypes of common variants, association with disease status, and polygenic risk scores. Results Of 31 575 samples identified, 6724 PD cases (mean [SD] age, 64.3 [10] years; age at onset, 58.8 [10.6] years; 3472 [53.2%] men) and 24 851 controls (age, 59.4 [11.4] years; 11 030 [45.0%] men) were analyzed in the discovery study. Eleven genome-wide significant loci were identified; 2 of these loci were novel (SV2C and WBSCR17) and 9 were previously found in Europeans. Replication in European-ancestry and Japanese samples showed robust association for SV2C (rs246814; odds ratio, 1.16; 95% CI, 1.11-1.21; P = 1.17 × 10-10 in meta-analysis of discovery and replication samples) but showed potential genetic heterogeneity at WBSCR17 (rs9638616; I2=67.1%; P = 3.40 × 10-3 for hetereogeneity). Polygenic risk score models including variants at these 11 loci were associated with a significant improvement in area under the curve over the model based on 78 European loci alone (63.1% vs 60.2%; P = 6.81 × 10-12). Conclusions and Relevance This study identified 2 apparently novel gene loci and found 9 previously identified European loci to be associated with PD in this large, meta-genome-wide association study in a worldwide population of Asian individuals and reports similarities and differences in genetic risk factors between Asian and European individuals in the risk for PD. These findings may lead to improved stratification of Asian patients and controls based on polygenic risk scores. Our findings have potential academic and clinical importance for risk stratification and precision medicine in Asia.
Collapse
Affiliation(s)
- Jia Nee Foo
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- Human Genetics, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Elaine Guo Yan Chew
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- Human Genetics, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Sun Ju Chung
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Rong Peng
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Mike A. Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
- Data Tecnica International LLC, Glen Echo, Maryland
| | - Kin Y. Mok
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Wataru Satake
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- Department of Neurology, The University of Tokyo Graduate School of Medicine, Bunkyo, Tokyo, Japan
| | - Tatsushi Toda
- Department of Neurology, The University of Tokyo Graduate School of Medicine, Bunkyo, Tokyo, Japan
| | - Yinxia Chao
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore, Singapore
- Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Louis C. S. Tan
- Duke-National University of Singapore Medical School, Singapore, Singapore
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
| | - Moses Tandiono
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- Human Genetics, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Michelle M. Lian
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- Human Genetics, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Ebonne Y. Ng
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore, Singapore
| | - Kumar-M. Prakash
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore, Singapore
| | - Wing-Lok Au
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
| | - Wee-Yang Meah
- Human Genetics, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Shi Qi Mok
- Human Genetics, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Azlina Ahmad Annuar
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Anne Y. Y. Chan
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Gerald Choa Neuroscience Centre, Lui Che Woo Institute of Innovative Medicine, Prince of Wales Hospital, Division of Neurology, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, PR China
| | - Ling Chen
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Yongping Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Beom S. Jeon
- Department of Neurology, Seoul National University Hospital, Jongno-gu, Seoul, South Korea
| | - Lulu Jiang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Jia Lun Lim
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Mah Pooi Soo and Tan Chin Nam Centre for Parkinson's and Related Disorders, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Juei-Jueng Lin
- Department of Neurology, Chushang Show-Chwan Hospital, Zhushan District, Nantou, Taiwan
| | - Chunfeng Liu
- Department of Neurology, Second Affiliated Hospital of Soochow University, Suzhou, PR China
| | - Chengjie Mao
- Department of Neurology, Second Affiliated Hospital of Soochow University, Suzhou, PR China
| | - Vincent Mok
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Gerald Choa Neuroscience Centre, Lui Che Woo Institute of Innovative Medicine, Prince of Wales Hospital, Division of Neurology, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, PR China
| | - Zhong Pei
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Hui-Fang Shang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Chang-He Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Kyuyoung Song
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ai Huey Tan
- Mah Pooi Soo and Tan Chin Nam Centre for Parkinson's and Related Disorders, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yih-Ru Wu
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University, Taipei, Taiwan
| | - Yu-ming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Renshi Xu
- Department of Neurology, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, Nanchang, Jiangxi, PR China
| | - Yaping Yan
- Second Affiliated Hospital, Department of Neurology, Zhejiang University College of Medicine, Hangzhou, Zhejiang Province, PR China
| | - Jing Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - BaoRong Zhang
- Second Affiliated Hospital, Department of Neurology, Zhejiang University College of Medicine, Hangzhou, Zhejiang Province, PR China
| | - Woon-Puay Koh
- Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Shen-Yang Lim
- Mah Pooi Soo and Tan Chin Nam Centre for Parkinson's and Related Disorders, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Chiea Chuen Khor
- Human Genetics, Genome Institute of Singapore, A*STAR, Singapore, Singapore
- Singapore Eye Research Institute, Singapore, Singapore
| | - Jianjun Liu
- Human Genetics, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore, Singapore
- Duke-National University of Singapore Medical School, Singapore, Singapore
| |
Collapse
|
42
|
Tsymbalyuk S, Smith M, Gore C, Tsymbalyuk O, Ivanova S, Sansur C, Gerzanich V, Simard JM. Brivaracetam attenuates pain behaviors in a murine model of neuropathic pain. Mol Pain 2020; 15:1744806919886503. [PMID: 31615323 PMCID: PMC6880061 DOI: 10.1177/1744806919886503] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Background The antiseizure racetams may provide novel molecular insights into
neuropathic pain due to their unique mechanism involving synaptic vesicle
glycoprotein 2A. Anti-allodynic effects of levetiracetam have been shown in
animal models of neuropathic pain. Here, we studied the effect of
brivaracetam, which binds to synaptic vesicle glycoprotein 2A with 20-fold
greater affinity, and has fewer off-target effects. Methods Mice underwent unilateral sciatic nerve cuffing and were evaluated for
mechanical sensitivity using von Frey filaments. Pain behaviors were
assessed with prophylactic treatment using levetiracetam (100 or 10 mg/kg)
or brivaracetam (10 or 1 mg/kg) beginning after surgery and continuing for
21 days, or with therapeutic treatment using brivaracetam (10 or 1 mg/kg)
beginning on day 14, after allodynia was established, and continuing for 28
or 63 days. Spinal cord tissues from the prophylaxis experiment with10 mg/kg
brivaracetam were examined for neuroinflammation (Iba1 and tumor necrosis
factor), T-lymphocyte (CD3) infiltration, and synaptic vesicle glycoprotein
2A expression. Results When used prophylactically, levetiracetam, 100 mg/kg, and brivaracetam,
10 mg/kg, prevented the development of allodynia, with lower doses of each
being less effective. When used therapeutically, brivaracetam extinguished
allodynia, requiring 10 days with 10 mg/kg, and six weeks with 1 mg/kg.
Brivaracetam was associated with reduced neuroinflammation and reduced
T-lymphocyte infiltration in the dorsal horn. After sciatic nerve cuffing,
synaptic vesicle glycoprotein 2A expression was identified in neurons,
activated astrocytes, microglia/macrophages, and T lymphocytes in the dorsal
horn. Conclusion Synaptic vesicle glycoprotein 2A may represent a novel target for neuropathic
pain. Brivaracetam may warrant study in humans with neuropathic pain due to
peripheral nerve injury.
Collapse
Affiliation(s)
- Solomiya Tsymbalyuk
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Madeleine Smith
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Charles Gore
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Orest Tsymbalyuk
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Svetlana Ivanova
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Charles Sansur
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
43
|
Poulain B, Lemichez E, Popoff MR. Neuronal selectivity of botulinum neurotoxins. Toxicon 2020; 178:20-32. [PMID: 32094099 DOI: 10.1016/j.toxicon.2020.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022]
Abstract
Botulinum neurotoxins (BoNTs) are highly potent toxins responsible for a severe disease, called botulism. They are also efficient therapeutic tools with an increasing number of indications ranging from neuromuscular dysfunction to hypersecretion syndrome, pain release, depression as well as cosmetic application. BoNTs are known to mainly target the motor-neurons terminals and to induce flaccid paralysis. BoNTs recognize a specific double receptor on neuronal cells consisting of gangliosides and synaptic vesicle protein, SV2 or synaptotagmin. Using cultured neuronal cells, BoNTs have been established blocking the release of a wide variety of neurotransmitters. However, BoNTs are more potent in motor-neurons than in the other neuronal cell types. In in vivo models, BoNT/A impairs the cholinergic neuronal transmission at the motor-neurons but also at neurons controlling secretions and smooth muscle neurons, and blocks several neuronal pathways including excitatory, inhibitory, and sensitive neurons. However, only a few reports investigated the neuronal selectivity of BoNTs in vivo. In the intestinal wall, BoNT/A and BoNT/B target mainly the cholinergic neurons and to a lower extent the other non-cholinergic neurons including serotonergic, glutamatergic, GABAergic, and VIP-neurons. The in vivo effects induced by BoNTs on the non-cholinergic neurons remain to be precisely investigated. We report here a literature review of the neuronal selectivity of BoNTs.
Collapse
Affiliation(s)
- Bernard Poulain
- Université de Strasbourg, CNRS, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | | | | |
Collapse
|
44
|
Wood M, Daniels V, Provins L, Wolff C, Kaminski RM, Gillard M. Pharmacological Profile of the Novel Antiepileptic Drug Candidate Padsevonil: Interactions with Synaptic Vesicle 2 Proteins and the GABA A Receptor. J Pharmacol Exp Ther 2020; 372:1-10. [PMID: 31619465 DOI: 10.1124/jpet.119.261149] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/10/2019] [Indexed: 03/08/2025] Open
Abstract
Padsevonil is an antiepileptic drug (AED) candidate synthesized in a medicinal chemistry program initiated to rationally design compounds with high affinity for synaptic vesicle 2 (SV2) proteins and low-to-moderate affinity for the benzodiazepine binding site on GABAA receptors. The pharmacological profile of padsevonil was characterized in binding and electrophysiological experiments. At recombinant SV2 proteins, padsevonil's affinity for SV2A was greater than that of levetiracetam and brivaracetam (pKi 8.5, 5.2, and 6.6, respectively). Unlike the latter AEDs, both selective SV2A ligands, padsevonil also displayed high affinity for the SV2B and SV2C isoforms (pKi 7.9 and 8.5, respectively). Padsevonil's interaction with SV2A differed from that of levetiracetam and brivaracetam; it exhibited slower binding kinetics: dissociation t 1/2 30 minutes from the human protein at 37°C compared with <0.5 minute for levetiracetam and brivaracetam. In addition, its binding was not potentiated by the allosteric modulator UCB1244283. At recombinant GABAA receptors, padsevonil displayed low to moderate affinity (pIC50≤6.1) for the benzodiazepine site, and in electrophysiological studies, its relative efficacy compared with zolpidem (full-agonist reference drug) was 40%, indicating partial agonist properties. In in vivo (mice) receptor occupancy studies, padsevonil exhibited SV2A occupancy at low ED50 (0.2 mg/kg) and benzodiazepine site occupancy at higher doses (ED50 36 mg/kg), supporting in vitro results. Padsevonil's selectivity for its intended targets was confirmed in profiling studies, where it lacked significant effects on a wide variety of ion channels, receptors, transporters, and enzymes. Padsevonil is a first-in-class AED candidate with a unique target profile allowing for presynaptic and postsynaptic activity. SIGNIFICANCE STATEMENT: Padsevonil is an antiepileptic drug candidate developed as a single molecular entity interacting with both presynaptic and postsynaptic targets. Results of in vitro and in vivo radioligand binding assays confirmed this target profile: padsevonil displayed nanomolar affinity for the three synaptic vesicle 2 protein isoforms (SV2A, B, and C) and micromolar affinity for the benzodiazepine binding site on GABAA receptors. Furthermore, padsevonil showed greater affinity for and slower binding kinetics at SV2A than the selective SV2A ligands, levetiracetam, and brivaracetam.
Collapse
Affiliation(s)
- Martyn Wood
- UCB Pharma, Neurosciences Therapeutic Area, Braine l'Alleud, Belgium
| | - Veronique Daniels
- UCB Pharma, Neurosciences Therapeutic Area, Braine l'Alleud, Belgium
| | - Laurent Provins
- UCB Pharma, Neurosciences Therapeutic Area, Braine l'Alleud, Belgium
| | - Christian Wolff
- UCB Pharma, Neurosciences Therapeutic Area, Braine l'Alleud, Belgium
| | - Rafal M Kaminski
- UCB Pharma, Neurosciences Therapeutic Area, Braine l'Alleud, Belgium
| | - Michel Gillard
- UCB Pharma, Neurosciences Therapeutic Area, Braine l'Alleud, Belgium
| |
Collapse
|
45
|
Chen MK, Mecca AP, Naganawa M, Finnema SJ, Toyonaga T, Lin SF, Najafzadeh S, Ropchan J, Lu Y, McDonald JW, Michalak HR, Nabulsi NB, Arnsten AFT, Huang Y, Carson RE, van Dyck CH. Assessing Synaptic Density in Alzheimer Disease With Synaptic Vesicle Glycoprotein 2A Positron Emission Tomographic Imaging. JAMA Neurol 2019; 75:1215-1224. [PMID: 30014145 DOI: 10.1001/jamaneurol.2018.1836] [Citation(s) in RCA: 310] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Importance Synaptic loss is well established as the major structural correlate of cognitive impairment in Alzheimer disease (AD). The ability to measure synaptic density in vivo could accelerate the development of disease-modifying treatments for AD. Synaptic vesicle glycoprotein 2A is an essential vesicle membrane protein expressed in virtually all synapses and could serve as a suitable target for synaptic density. Objective To compare hippocampal synaptic vesicle glycoprotein 2A (SV2A) binding in participants with AD and cognitively normal participants using positron emission tomographic (PET) imaging. Design, Setting, and Participants This cross-sectional study recruited 10 participants with AD and 11 participants who were cognitively normal between November 2015 and June 2017. We hypothesized a reduction in hippocampal SV2A binding in AD, based on the early degeneration of entorhinal cortical cell projections to the hippocampus (via the perforant path) and hippocampal SV2A reductions that had been observed in postmortem studies. Participants underwent high-resolution PET scanning with ((R)-1-((3-(11C-methyl-11C)pyridin-4-yl)methyl)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one), a compound more commonly known as 11C-UCB-J, for SV2A. They also underwent high-resolution PET scanning with carbon 11-labeled Pittsburgh Compound B (11C-PiB) for β-amyloid, magnetic resonance imaging, and cognitive and neurologic evaluation. Main Outcomes and Measures Outcomes were 11C-UCB-J-specific binding (binding potential [BPND]) via PET imaging in brain regions of interest in participants with AD and participants who were cognitively normal. Results Ten participants with AD (5 male and 5 female; mean [SD] age, 72.7 [6.3] years; 10 [100%] β-amyloid positive) were compared with 11 participants who were cognitively normal (5 male and 6 female; mean [SD] age, 72.9 [8.7] years; 11 [100%] β-amyloid negative). Participants with AD spanned the disease stages from amnestic mild cognitive impairment (n = 5) to mild dementia (n = 5). Participants with AD had significant reduction in hippocampal SV2A specific binding (41%) compared with cognitively normal participants, as assessed by 11C-UCB-J-PET BPND (cognitively normal participants: mean [SD] BPND, 1.47 [0.37]; participants with AD: 0.87 [0.50]; P = .005). These reductions remained significant after correction for atrophy (ie, partial volume correction; participants who were cognitively normal: mean [SD], 2.71 [0.46]; participants with AD: 2.15 [0.55]; P = .02). Hippocampal SV2A-specific binding BPND was correlated with a composite episodic memory score in the overall sample (R = 0.56; P = .01). Conclusions and Relevance To our knowledge, this is the first study to investigate synaptic density in vivo in AD using 11C-UCB-J-PET imaging. This approach may provide a direct measure of synaptic density, and it therefore holds promise as an in vivo biomarker for AD and as an outcome measure for trials of disease-modifying therapies, particularly those targeted at the preservation and restoration of synapses.
Collapse
Affiliation(s)
- Ming-Kai Chen
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Adam P Mecca
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Mika Naganawa
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Sjoerd J Finnema
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Shu-Fei Lin
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Soheila Najafzadeh
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Jim Ropchan
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Yihuan Lu
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Julia W McDonald
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Hannah R Michalak
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Nabeel B Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Amy F T Arnsten
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | | |
Collapse
|
46
|
Stout K, Dunn A, Hoffman C, Miller GW. The Synaptic Vesicle Glycoprotein 2: Structure, Function, and Disease Relevance. ACS Chem Neurosci 2019; 10:3927-3938. [PMID: 31394034 PMCID: PMC11562936 DOI: 10.1021/acschemneuro.9b00351] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The synaptic vesicle glycoprotein 2 (SV2) family is comprised of three paralogues: SV2A, SV2B, and SV2C. In vertebrates, SV2s are 12-transmembrane proteins present on every secretory vesicle, including synaptic vesicles, and are critical to neurotransmission. Structural and functional studies suggest that SV2 proteins may play several roles to promote proper vesicular function. Among these roles are their potential to stabilize the transmitter content of vesicles, to maintain and orient the releasable pool of vesicles, and to regulate vesicular calcium sensitivity to ensure efficient, coordinated release of the transmitter. The SV2 family is highly relevant to human health in a number of ways. First, SV2A plays a role in neuronal excitability and as such is the specific target for the antiepileptic drug levetiracetam. SV2 proteins also act as the target by which potent neurotoxins, particularly botulinum, gain access to neurons and exert their toxicity. Both SV2B and SV2C are increasingly implicated in diseases such as Alzheimer's disease and Parkinson's disease. Interestingly, despite decades of intensive research, their exact function remains elusive. Thus, SV2 proteins are intriguing in their potentially diverse roles within the presynaptic terminal, and several recent developments have enhanced our understanding and appreciation of the protein family. Here, we review the structure and function of SV2 proteins as well as their relevance to disease and therapeutic development.
Collapse
Affiliation(s)
- Kristen Stout
- Department of Physiology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, United States
| | - Amy Dunn
- The Jackson Laboratory, Bar Harbor, Maine, United States
| | - Carlie Hoffman
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States
| | - Gary W. Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, United States
| |
Collapse
|
47
|
AD molecular: Molecular imaging of Alzheimer's disease: PET imaging of neurotransmitter systems. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019. [PMID: 31481161 DOI: 10.1016/bs.pmbts.2019.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Current understanding of Alzheimer's disease (AD) pathogenesis relies on the observed accumulations of amyloid β and phosphorylated tau aggregates that are thought to play key roles in initiating or propagating disease. However, other processes including changes in synaptic proteins and neurotransmitter loss have been suggested as important etiologies or contributors. Positron emission tomography (PET) imaging allows in vivo investigations of molecular changes associated with AD. PET imaging with multiple radiotracers can be used in combination with other modalities such as magnetic resonance imaging (MRI), and with assessments of cognition and neuropsychiatric symptoms to investigate the molecular underpinnings of AD. Studies of synaptic protein changes may improve the understanding of disease mechanisms and provide valuable markers of disease progression and therapeutic efficacy. This chapter will illustrate the importance of in vivo molecular imaging in the study of AD with a specific emphasis on PET and radioligands for several non-amyloid targets.
Collapse
|
48
|
Pellett S, Tepp WH, Johnson EA. Botulinum neurotoxins A, B, C, E, and F preferentially enter cultured human motor neurons compared to other cultured human neuronal populations. FEBS Lett 2019; 593:2675-2685. [PMID: 31240706 PMCID: PMC7751886 DOI: 10.1002/1873-3468.13508] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 12/11/2022]
Abstract
Human-induced pluripotent stem cell (hiPSC)-derived neurons can be exquisitely sensitive to botulinum neurotoxins (BoNTs), exceeding sensitivity of the traditionally used mouse bioassay. In this report, four defined hiPSC-derived neuronal populations including primarily GABAergic, glutamatergic, dopaminergic, and motor neurons were examined for BoNT/A, B, C, D, E, and F sensitivity. The data indicate that sensitivity varies markedly for the BoNTs tested. Motor neurons are significantly more sensitive than other neuron types for all BoNTs except BoNT/D. Examination of SNARE protein levels and BoNT-specific cell surface protein receptors reveals few differences between the cell types except greater expression levels of the receptor protein SV2C and synapsin-IIa in motor neurons. This indicates that differential toxicity of BoNTs for motor neurons compared to other neuronal cell types involves multiple mechanisms.
Collapse
Affiliation(s)
- Sabine Pellett
- Department of Bacteriology, University of Wisconsin-Madison, WI, USA
| | - William H Tepp
- Department of Bacteriology, University of Wisconsin-Madison, WI, USA
| | - Eric A Johnson
- Department of Bacteriology, University of Wisconsin-Madison, WI, USA
| |
Collapse
|
49
|
Anxiety-like features and spatial memory problems as a consequence of hippocampal SV2A expression. PLoS One 2019; 14:e0217882. [PMID: 31166988 PMCID: PMC6550411 DOI: 10.1371/journal.pone.0217882] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/20/2019] [Indexed: 02/08/2023] Open
Abstract
The Synaptic Vesicle Protein 2A (SV2A) is a transmembrane protein whose presence is reduced both in animal models and in patients with chronic epilepsy. Besides its implication in the epileptic process, the behavioural consequences of the changes in its expression remain unclear. The purpose of our research is to better understand the possible role(s) of this protein through the phenotype of cKO (Grik4 Cre+/-, SV2A lox/lox) mice, male and female, which present a specific decrease of SV2A expression levels in the hippocampal glutamatergic neurons but without any epileptic seizures. In this study, we compare the cKO mice with cHZ (Grik4 Cre+/-, SV2A lox/+) and WT (Grik4 Cre+/+, SV2A lox/lox) mice through a battery of tests, used to evaluate different features: the anxiety-related features (Elevated Plus Maze), the locomotor activity (Activity Chambers), the contextual fear-related memory (Contextual Fear Conditioning), and the spatial memory (Barnes Maze). Our results showed statistically significant differences in the habituation to a new environment, an increase in the anxiety levels and spatial memory deficit in the cHZ and cKO groups, compared to the WT group. No statistically significant differences due to the genotype appeared in the spontaneous locomotor activity or the fear-linked memory. However, sexual differences were observed in this last feature. These results highlight not only an important role of the SV2A protein in the cognitive and anxiety problems typically encountered in epileptic patients, but also a possible role in the symptomatology of other neurodegenerative diseases, such as the Alzheimer’s disease.
Collapse
|
50
|
Evaluating the In Vivo Specificity of [ 18F]UCB-H for the SV2A Protein, Compared with SV2B and SV2C in Rats Using microPET. Molecules 2019; 24:molecules24091705. [PMID: 31052478 PMCID: PMC6538996 DOI: 10.3390/molecules24091705] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/28/2019] [Accepted: 04/29/2019] [Indexed: 11/25/2022] Open
Abstract
The synaptic vesicle protein 2 (SV2) is involved in synaptic vesicle trafficking. The SV2A isoform is the most studied and its implication in epilepsy therapy led to the development of the first SV2A PET radiotracer [18F]UCB-H. The objective of this study was to evaluate in vivo, using microPET in rats, the specificity of [18F]UCB-H for SV2 isoform A in comparison with the other two isoforms (B and C) through a blocking assay. Twenty Sprague Dawley rats were pre-treated either with the vehicle, or with specific competitors against SV2A (levetiracetam), SV2B (UCB5203) and SV2C (UCB0949). The distribution volume (Vt, Logan plot, t* 15 min) was obtained with a population-based input function. The Vt analysis for the entire brain showed statistically significant differences between the levetiracetam group and the other groups (p < 0.001), but also between the vehicle and the SV2B group (p < 0.05). An in-depth Vt analysis conducted for eight relevant brain structures confirmed the statistically significant differences between the levetiracetam group and the other groups (p < 0.001) and highlighted the superior and the inferior colliculi along with the cortex as regions also displaying statistically significant differences between the vehicle and SV2B groups (p < 0.05). These results emphasize the in vivo specificity of [18F]UCB-H for SV2A against SV2B and SV2C, confirming that [18F]UCB-H is a suitable radiotracer for in vivo imaging of the SV2A proteins with PET.
Collapse
|