1
|
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
|
2
|
Bucher ML, Dunn AR, Bradner JM, Stout Egerton K, Burkett JP, Johnson MA, Miller GW. Synaptic vesicle glycoprotein 2C enhances vesicular storage of dopamine and counters dopaminergic toxicity. Eur J Neurosci 2024; 59:2483-2501. [PMID: 38532289 PMCID: PMC11647951 DOI: 10.1111/ejn.16311] [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: 06/20/2023] [Revised: 02/14/2024] [Accepted: 02/22/2024] [Indexed: 03/28/2024]
Abstract
Dopaminergic neurons of the substantia nigra exist in a persistent state of vulnerability resulting from high baseline oxidative stress, high-energy demand, and broad unmyelinated axonal arborisations. Impairments in the storage of dopamine compound this stress because of cytosolic reactions that transform the vital neurotransmitter into an endogenous neurotoxicant, and this toxicity is thought to contribute to the dopamine neuron degeneration that occurs Parkinson's disease. We have previously identified synaptic vesicle glycoprotein 2C (SV2C) as a modifier of vesicular dopamine function, demonstrating that genetic ablation of SV2C in mice results in decreased dopamine content and evoked dopamine release in the striatum. Here, we adapted a previously published in vitro assay utilising false fluorescent neurotransmitter 206 (FFN206) to visualise how SV2C regulates vesicular dopamine dynamics and determined that SV2C promotes the uptake and retention of FFN206 within vesicles. In addition, we present data indicating that SV2C enhances the retention of dopamine in the vesicular compartment with radiolabelled dopamine in vesicles isolated from immortalised cells and from mouse brain. Further, we demonstrate that SV2C enhances the ability of vesicles to store the neurotoxicant 1-methyl-4-phenylpyridinium (MPP+) and that genetic ablation of SV2C results in enhanced 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced vulnerability in mice. Together, these findings suggest that SV2C functions to enhance vesicular storage of dopamine and neurotoxicants and helps maintain the integrity of dopaminergic neurons.
Collapse
Affiliation(s)
- Meghan L Bucher
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| | - Amy R Dunn
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Joshua M Bradner
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Kristen Stout Egerton
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - James P Burkett
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Michelle A Johnson
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York NY 10031, USA
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
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: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [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
|
6
|
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
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
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: 166] [Impact Index Per Article: 33.2] [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
|
11
|
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: 54] [Impact Index Per Article: 9.0] [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
|
12
|
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
|
13
|
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
|
14
|
Ciruelas K, Marcotulli D, Bajjalieh SM. Synaptic vesicle protein 2: A multi-faceted regulator of secretion. Semin Cell Dev Biol 2019; 95:130-141. [PMID: 30826548 DOI: 10.1016/j.semcdb.2019.02.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/11/2019] [Accepted: 02/21/2019] [Indexed: 01/01/2023]
Abstract
Synaptic Vesicle Protein 2 (SV2) comprises a recently evolved family of proteins unique to secretory vesicles that undergo calcium-regulated exocytosis. In this review we consider SV2s' structural features, evolution, and function and discuss its therapeutic potential as the receptors for an expanding class of drugs used to treat epilepsy and cognitive decline.
Collapse
Affiliation(s)
- Kristine Ciruelas
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Daniele Marcotulli
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Sandra M Bajjalieh
- Department of Pharmacology, University of Washington, Seattle, WA, United States.
| |
Collapse
|
15
|
Antipova V, Wree A, Holzmann C, Mann T, Palomero-Gallagher N, Zilles K, Schmitt O, Hawlitschka A. Unilateral Botulinum Neurotoxin-A Injection into the Striatum of C57BL/6 Mice Leads to a Different Motor Behavior Compared with Rats. Toxins (Basel) 2018; 10:E295. [PMID: 30018211 PMCID: PMC6070800 DOI: 10.3390/toxins10070295] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 07/13/2018] [Accepted: 07/15/2018] [Indexed: 12/26/2022] Open
Abstract
Different morphological changes in the caudate-putamen (CPu) of naïve rats and mice were observed after intrastriatal botulinum neurotoxin-A (BoNT-A) injection. For this purpose we here studied various motor behaviors in mice (n = 46) longitudinally up to 9 months after intrastriatal BoNT-A administration as previously reported for rats, and compared both outcomes. Apomorphine- and amphetamine-induced rotational behavior, spontaneous motor behavior, as well as lateralized neglect were studied in mice after the injection of single doses of BoNT-A into the right CPu, comparing them with sham-injected animals. Unilateral intrastriatal injection of BoNT-A in mice induced significantly increased contralateral apomorphine-induced rotations for 1 to 3 months, as well as significantly increased contralateral amphetamine-induced rotations 1 to 9 months after injection. In rats (n = 28), unilateral BoNT-A injection also induced significantly increased contralateral apomorphine-induced rotations 3 months after injection, but did not provoke amphetamine-induced rotations at all. Lateralized sensorimotor integration, forelimb preference, and forelimb stepping were significantly impaired on the left side. The differences in motor behaviors between rats and mice may be caused by different BoNT-A effects on cholinergic and catecholaminergic fibers in rat and mouse striata, interspecies differences in striatal receptor densities, and different connectomes of the basal ganglia.
Collapse
Affiliation(s)
- Veronica Antipova
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057 Rostock, Germany.
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Macroscopic and Clinical Anatomy, Medical University of Graz, Harrachgasse 21/1, A-8010 Graz, Austria.
| | - Andreas Wree
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057 Rostock, Germany.
| | - Carsten Holzmann
- Institute of Medical Genetics, Rostock University Medical Center, Ernst-Heydemann-Strasse 8, D-18057 Rostock, Germany.
| | - Teresa Mann
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057 Rostock, Germany.
| | - Nicola Palomero-Gallagher
- Institute of Neuroscience and Medicine INM-1, Research Center Jülich, D-52425 Jülich, Germany.
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen, D-52062 Aachen, Germany.
| | - Karl Zilles
- Institute of Neuroscience and Medicine INM-1, Research Center Jülich, D-52425 Jülich, Germany.
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen, D-52062 Aachen, Germany.
- JARA-Translational Brain Medicine, D-52062 Aachen, Germany.
| | - Oliver Schmitt
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057 Rostock, Germany.
| | - Alexander Hawlitschka
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057 Rostock, Germany.
| |
Collapse
|
16
|
Mutant Huntingtin Causes a Selective Decrease in the Expression of Synaptic Vesicle Protein 2C. Neurosci Bull 2018; 34:747-758. [PMID: 29713895 DOI: 10.1007/s12264-018-0230-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 03/24/2018] [Indexed: 12/11/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disease caused by a polyglutamine expansion in the huntingtin (Htt) protein. Mutant Htt causes synaptic transmission dysfunctions by interfering in the expression of synaptic proteins, leading to early HD symptoms. Synaptic vesicle proteins 2 (SV2s), a family of synaptic vesicle proteins including 3 members, SV2A, SV2B, and SV2C, plays important roles in synaptic physiology. Here, we investigated whether the expression of SV2s is affected by mutant Htt in the brains of HD transgenic (TG) mice and Neuro2a mouse neuroblastoma cells (N2a cells) expressing mutant Htt. Western blot analysis showed that the protein levels of SV2A and SV2B were not significantly changed in the brains of HD TG mice expressing mutant Htt with 82 glutamine repeats. However, in the TG mouse brain there was a dramatic decrease in the protein level of SV2C, which has a restricted distribution pattern in regions particularly vulnerable in HD. Immunostaining revealed that the immunoreactivity of SV2C was progressively weakened in the basal ganglia and hippocampus of TG mice. RT-PCR demonstrated that the mRNA level of SV2C progressively declined in the TG mouse brain without detectable changes in the mRNA levels of SV2A and SV2B, indicating that mutant Htt selectively inhibits the transcriptional expression of SV2C. Furthermore, we found that only SV2C expression was progressively inhibited in N2a cells expressing a mutant Htt containing 120 glutamine repeats. These findings suggest that the synaptic dysfunction in HD results from the mutant Htt-mediated inhibition of SV2C transcriptional expression. These data also imply that the restricted distribution and decreased expression of SV2C contribute to the brain region-selective pathology of HD.
Collapse
|
17
|
Yang M, Li Y, Hu L, Luo D, Zhang Y, Xiao X, Li G, Zhang L, Zhu G. Lead exposure inhibits expression of SV2C through NRSF. Toxicology 2018; 398-399:23-30. [DOI: 10.1016/j.tox.2018.02.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/11/2018] [Accepted: 02/27/2018] [Indexed: 12/31/2022]
|
18
|
Dunn AR, Hoffman CA, Stout KA, Ozawa M, Dhamsania RK, Miller GW. Immunochemical analysis of the expression of SV2C in mouse, macaque and human brain. Brain Res 2017; 1702:85-95. [PMID: 29274878 DOI: 10.1016/j.brainres.2017.12.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 11/30/2017] [Accepted: 12/19/2017] [Indexed: 11/17/2022]
Abstract
The synaptic vesicle glycoprotein 2C (SV2C) is an undercharacterized protein with enriched expression in phylogenetically old brain regions. Its precise role within the brain is unclear, though various lines of evidence suggest that SV2C is involved in the function of synaptic vesicles through the regulation of vesicular trafficking, calcium-induced exocytosis, or synaptotagmin function. SV2C has been linked to multiple neurological disorders, including Parkinson's disease and psychiatric conditions. SV2C is expressed in various cell types-primarily dopaminergic, GABAergic, and cholinergic cells. In mice, it is most highly expressed in nuclei within the basal ganglia, though it is unknown if this pattern of expression is consistent across species. Here, we use a custom SV2C-specific antiserum to describe localization within the brain of mouse, nonhuman primate, and human, including cell-type localization. We found that the immunoreactivity with this antiserum is consistent with previously-published antibodies, and confirmed localization of SV2C in the basal ganglia of rodent, rhesus macaque, and human. We observed strongest expression of SV2C in the substantia nigra, ventral tegmental area, dorsal striatum, pallidum, and nucleus accumbens of each species. Further, we demonstrate colocalization between SV2C and markers of dopaminergic, GABAergic, and cholinergic neurons within these brain regions. SV2C has been increasingly linked to dopamine and basal ganglia function. These antisera will be an important resource moving forward in our understanding of the role of SV2C in vesicle dynamics and neurological disease.
Collapse
Affiliation(s)
- Amy R Dunn
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Carlie A Hoffman
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Kristen A Stout
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Minagi Ozawa
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Rohan K Dhamsania
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Gary W Miller
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States; Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, United States.
| |
Collapse
|
19
|
Hawlitschka A, Holzmann C, Witt S, Spiewok J, Neumann AM, Schmitt O, Wree A, Antipova V. Intrastriatally injected botulinum neurotoxin-A differently effects cholinergic and dopaminergic fibers in C57BL/6 mice. Brain Res 2017; 1676:46-56. [DOI: 10.1016/j.brainres.2017.09.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 08/10/2017] [Accepted: 09/12/2017] [Indexed: 11/29/2022]
|
20
|
Synaptic vesicle 2C and its synaptic-related function. Clin Chim Acta 2017; 472:112-117. [PMID: 28774501 DOI: 10.1016/j.cca.2017.07.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 07/27/2017] [Accepted: 07/30/2017] [Indexed: 11/22/2022]
Abstract
Synaptic vesicle 2 C (SV2C) is a novel isoform belonging to the synaptic vesicle 2 (SV2) protein superfamily; a family of proteins known to have roles in vesicle trafficking, exocytosis and neurotransmission. In humans, SV2C is expressed in evolutionarily older brain regions, and is a known receptor for botulinum neurotoxin/A (BoNT/A), controlling glucose-evoked granule recruitment and regulating dopamine release, thus serving as a potential target molecule in the treatment of psychosis. In addition, recent researches have shown that SV2C regulates hypertension and accelerates venous thromboembolism (VTE) and coagulation pathways and may play roles in several non-nervous system diseases. In terms of regulation, SV2C is positively regulated by both alendronate and statins. As SV2C may provide a potential novel therapeutic target for psychosis and other diseases, this article reviews the progress made thus far in understanding the structure, distribution, function and regulation of SV2C.
Collapse
|
21
|
Schmitt M, Dehay B, Bezard E, Garcia-Ladona FJ. U18666A, an activator of sterol regulatory element binding protein pathway, modulates presynaptic dopaminergic phenotype of SH-SY5Y neuroblastoma cells. Synapse 2017; 71. [DOI: 10.1002/syn.21980] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 03/22/2017] [Accepted: 04/10/2017] [Indexed: 11/06/2022]
Affiliation(s)
- Mathieu Schmitt
- Neuroscience Therapeutic Area, New Medicines, UCB Biopharma SPRL; 1420 Braine l'Alleud Belgium
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293; Bordeaux 33000 France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293; Bordeaux 33000 France
| | - Benjamin Dehay
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293; Bordeaux 33000 France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293; Bordeaux 33000 France
| | - Erwan Bezard
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293; Bordeaux 33000 France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293; Bordeaux 33000 France
| | - F. Javier Garcia-Ladona
- Neuroscience Therapeutic Area, New Medicines, UCB Biopharma SPRL; 1420 Braine l'Alleud Belgium
| |
Collapse
|
22
|
Bartholome O, Van den Ackerveken P, Sánchez Gil J, de la Brassinne Bonardeaux O, Leprince P, Franzen R, Rogister B. Puzzling Out Synaptic Vesicle 2 Family Members Functions. Front Mol Neurosci 2017; 10:148. [PMID: 28588450 PMCID: PMC5438990 DOI: 10.3389/fnmol.2017.00148] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 05/02/2017] [Indexed: 01/18/2023] Open
Abstract
Synaptic vesicle proteins 2 (SV2) were discovered in the early 80s, but the clear demonstration that SV2A is the target of efficacious anti-epileptic drugs from the racetam family stimulated efforts to improve understanding of its role in the brain. Many functions have been suggested for SV2 proteins including ions or neurotransmitters transport or priming of SVs. Moreover, several recent studies highlighted the link between SV2 and different neuronal disorders such as epilepsy, Schizophrenia (SCZ), Alzheimer's or Parkinson's disease. In this review article, we will summarize our present knowledge on SV2A function(s) and its potential role(s) in the pathophysiology of various brain disorders.
Collapse
Affiliation(s)
- Odile Bartholome
- Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences, University of LiègeLiège, Belgium
| | | | - Judit Sánchez Gil
- Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences, University of LiègeLiège, Belgium
| | | | - Pierre Leprince
- Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences, University of LiègeLiège, Belgium
| | - Rachelle Franzen
- Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences, University of LiègeLiège, Belgium
| | - Bernard Rogister
- Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences, University of LiègeLiège, Belgium.,Department of Neurology, Centre Hospitalier Universitaire de Liège (CHU), University of LiègeLiège, Belgium
| |
Collapse
|
23
|
Synaptic vesicle glycoprotein 2C (SV2C) modulates dopamine release and is disrupted in Parkinson disease. Proc Natl Acad Sci U S A 2017; 114:E2253-E2262. [PMID: 28246328 PMCID: PMC5358362 DOI: 10.1073/pnas.1616892114] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Here we describe a role for the synaptic vesicle glycoprotein 2C (SV2C) in dopamine neurotransmission and Parkinson disease (PD). SV2C is expressed on the vesicles of dopamine-producing neurons, and genetic deletion of SV2C causes a reduction in synaptic release of dopamine. The reduced dopamine release is associated with a decrease in motor activity. SV2C is suspected of mediating the neuroprotective effects of nicotine, and we show an ablated neurochemical response to nicotine in SV2C-knockout mice. Last, we demonstrate that SV2C expression is specifically disrupted in mice that express mutated α-synuclein and in humans with PD. Together, these data establish SV2C as an important mediator of dopamine homeostasis and a potential contributor to PD pathogenesis. Members of the synaptic vesicle glycoprotein 2 (SV2) family of proteins are involved in synaptic function throughout the brain. The ubiquitously expressed SV2A has been widely implicated in epilepsy, although SV2C with its restricted basal ganglia distribution is poorly characterized. SV2C is emerging as a potentially relevant protein in Parkinson disease (PD), because it is a genetic modifier of sensitivity to l-DOPA and of nicotine neuroprotection in PD. Here we identify SV2C as a mediator of dopamine homeostasis and report that disrupted expression of SV2C within the basal ganglia is a pathological feature of PD. Genetic deletion of SV2C leads to reduced dopamine release in the dorsal striatum as measured by fast-scan cyclic voltammetry, reduced striatal dopamine content, disrupted α-synuclein expression, deficits in motor function, and alterations in neurochemical effects of nicotine. Furthermore, SV2C expression is dramatically altered in postmortem brain tissue from PD cases but not in Alzheimer disease, progressive supranuclear palsy, or multiple system atrophy. This disruption was paralleled in mice overexpressing mutated α-synuclein. These data establish SV2C as a mediator of dopamine neuron function and suggest that SV2C disruption is a unique feature of PD that likely contributes to dopaminergic dysfunction.
Collapse
|
24
|
Löscher W, Gillard M, Sands ZA, Kaminski RM, Klitgaard H. Synaptic Vesicle Glycoprotein 2A Ligands in the Treatment of Epilepsy and Beyond. CNS Drugs 2016; 30:1055-1077. [PMID: 27752944 PMCID: PMC5078162 DOI: 10.1007/s40263-016-0384-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The synaptic vesicle glycoprotein SV2A belongs to the major facilitator superfamily (MFS) of transporters and is an integral constituent of synaptic vesicle membranes. SV2A has been demonstrated to be involved in vesicle trafficking and exocytosis, processes crucial for neurotransmission. The anti-seizure drug levetiracetam was the first ligand to target SV2A and displays a broad spectrum of anti-seizure activity in various preclinical models. Several lines of preclinical and clinical evidence, including genetics and protein expression changes, support an important role of SV2A in epilepsy pathophysiology. While the functional consequences of SV2A ligand binding are not fully elucidated, studies suggest that subsequent SV2A conformational changes may contribute to seizure protection. Conversely, the recently discovered negative SV2A modulators, such as UCB0255, counteract the anti-seizure effect of levetiracetam and display procognitive properties in preclinical models. More broadly, dysfunction of SV2A may also be involved in Alzheimer's disease and other types of cognitive impairment, suggesting potential novel therapies for levetiracetam and its congeners. Furthermore, emerging data indicate that there may be important roles for two other SV2 isoforms (SV2B and SV2C) in the pathogenesis of epilepsy, as well as other neurodegenerative diseases. Utilization of recently developed SV2A positron emission tomography ligands will strengthen and reinforce the pharmacological evidence that SV2A is a druggable target, and will provide a better understanding of its role in epilepsy and other neurological diseases, aiding in further defining the full therapeutic potential of SV2A modulation.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany.
- Center for Systems Neuroscience, Hannover, Germany.
| | | | | | | | | |
Collapse
|
25
|
Stout KA, Dunn AR, Lohr KM, Alter SP, Cliburn RA, Guillot TS, Miller GW. Selective Enhancement of Dopamine Release in the Ventral Pallidum of Methamphetamine-Sensitized Mice. ACS Chem Neurosci 2016; 7:1364-1373. [PMID: 27501345 PMCID: PMC5073372 DOI: 10.1021/acschemneuro.6b00131] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
![]()
Drugs of abuse induce
sensitization, which is defined as enhanced
response to additional drug following a period of withdrawal. Sensitization
occurs in both humans and animal models of drug reinforcement and
contributes substantially to the addictive nature of drugs of abuse,
because it is thought to represent enhanced motivational wanting for
drug. The ventral pallidum, a key member of the reward pathway, contributes
to behaviors associated with reward, such as sensitization. Dopamine
inputs to the ventral pallidum have not been directly characterized.
Here we provide anatomical, neurochemical, and behavioral evidence
demonstrating that dopamine terminals in the ventral pallidum contribute
to reward in mice. We report subregional differences in dopamine release,
measured by ex vivo fast-scan cyclic voltammetry:
rostral ventral pallidum exhibits increased dopamine release and uptake
compared with caudal ventral pallidum, which is correlated with tissue
expression of dopaminergic proteins. We then subjected mice to a methamphetamine-sensitization
protocol to investigate the contribution of dopaminergic projections
to the region in reward related behavior. Methamphetamine-sensitized
animals displayed a 508% and 307% increase in baseline dopamine release
in the rostral and caudal ventral pallidum, respectively. Augmented
dopamine release in the rostral ventral pallidum was significantly
correlated with sensitized locomotor activity. Moreover, this presynaptic
dopaminergic plasticity occurred only in the ventral pallidum and
not in the ventral or dorsal striatum, suggesting that dopamine release
in the ventral pallidum may be integrally important to drug-induced
sensitization.
Collapse
Affiliation(s)
- Kristen A. Stout
- Department
of Environmental Health, Rollins School of Public Health, ‡Center for Neurodegenerative
Diseases, §Department of Pharmacology, and ∥Department of Neurology, Emory University, Atlanta, Georgia 30322, United States
| | - Amy R. Dunn
- Department
of Environmental Health, Rollins School of Public Health, ‡Center for Neurodegenerative
Diseases, §Department of Pharmacology, and ∥Department of Neurology, Emory University, Atlanta, Georgia 30322, United States
| | - Kelly M. Lohr
- Department
of Environmental Health, Rollins School of Public Health, ‡Center for Neurodegenerative
Diseases, §Department of Pharmacology, and ∥Department of Neurology, Emory University, Atlanta, Georgia 30322, United States
| | - Shawn P. Alter
- Department
of Environmental Health, Rollins School of Public Health, ‡Center for Neurodegenerative
Diseases, §Department of Pharmacology, and ∥Department of Neurology, Emory University, Atlanta, Georgia 30322, United States
| | - Rachel A. Cliburn
- Department
of Environmental Health, Rollins School of Public Health, ‡Center for Neurodegenerative
Diseases, §Department of Pharmacology, and ∥Department of Neurology, Emory University, Atlanta, Georgia 30322, United States
| | - Thomas S. Guillot
- Department
of Environmental Health, Rollins School of Public Health, ‡Center for Neurodegenerative
Diseases, §Department of Pharmacology, and ∥Department of Neurology, Emory University, Atlanta, Georgia 30322, United States
| | - Gary W. Miller
- Department
of Environmental Health, Rollins School of Public Health, ‡Center for Neurodegenerative
Diseases, §Department of Pharmacology, and ∥Department of Neurology, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
26
|
Altmann V, Schumacher-Schuh AF, Rieck M, Callegari-Jacques SM, Rieder CRM, Hutz MH. Influence of genetic, biological and pharmacological factors on levodopa dose in Parkinson's disease. Pharmacogenomics 2016; 17:481-8. [PMID: 27019953 DOI: 10.2217/pgs.15.183] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
AIM Levodopa is first-line treatment of Parkinson's disease motor symptoms but, dose response is highly variable. Therefore, the aim of this study was to determine how much levodopa dose could be explained by biological, pharmacological and genetic factors. PATIENTS & METHODS A total of 224 Parkinson's disease patients were genotyped for SV2C and SLC6A3 polymorphisms by allelic discrimination assays. Comedication, demographic and clinical data were also assessed. RESULTS All variables with p < 0.20 were included in a multiple regression analysis for dose prediction. The final model explained 23% of dose variation (F = 11.54; p < 0.000001). CONCLUSION Although a good prediction model was obtained, it still needs to be tested in an independent sample to be validated.
Collapse
Affiliation(s)
- Vivian Altmann
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Mariana Rieck
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Carlos R M Rieder
- Serviço de Neurologia, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Mara H Hutz
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
27
|
Schmitt M, Dehay B, Bezard E, Garcia-Ladona FJ. Harnessing the trophic and modulatory potential of statins in a dopaminergic cell line. Synapse 2016; 70:71-86. [DOI: 10.1002/syn.21881] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 11/12/2015] [Accepted: 12/15/2015] [Indexed: 01/15/2023]
Affiliation(s)
- Mathieu Schmitt
- Neuroscience Therapeutic Area, New Medicines, UCB Biopharma SPRL; 1420 Braine L'alleud Belgium
- University De Bordeaux, Institut Des Maladies Neurodégénératives; UMR 5293 Bordeaux 33000 France
- CNRS, Institut Des Maladies Neurodégénératives; UMR 5293 Bordeaux 33000 France
| | - Benjamin Dehay
- University De Bordeaux, Institut Des Maladies Neurodégénératives; UMR 5293 Bordeaux 33000 France
- CNRS, Institut Des Maladies Neurodégénératives; UMR 5293 Bordeaux 33000 France
| | - Erwan Bezard
- University De Bordeaux, Institut Des Maladies Neurodégénératives; UMR 5293 Bordeaux 33000 France
- CNRS, Institut Des Maladies Neurodégénératives; UMR 5293 Bordeaux 33000 France
| | - F. Javier Garcia-Ladona
- Neuroscience Therapeutic Area, New Medicines, UCB Biopharma SPRL; 1420 Braine L'alleud Belgium
| |
Collapse
|
28
|
Abstract
This paper is the thirty-sixth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2013 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia; stress and social status; tolerance and dependence; learning and memory; eating and drinking; alcohol and drugs of abuse; sexual activity and hormones, pregnancy, development and endocrinology; mental illness and mood; seizures and neurologic disorders; electrical-related activity and neurophysiology; general activity and locomotion; gastrointestinal, renal and hepatic functions; cardiovascular responses; respiration and thermoregulation; and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
29
|
Ramsey TL, Liu Q, Massey BW, Brennan MD. Genotypic variation in the SV2C gene impacts response to atypical antipsychotics the CATIE study. Schizophr Res 2013; 149:21-5. [PMID: 23886675 PMCID: PMC3845218 DOI: 10.1016/j.schres.2013.07.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/27/2013] [Accepted: 07/02/2013] [Indexed: 11/27/2022]
Abstract
Pharmacogenetic (PGx) predictors of response would improve outcomes in antipsychotic treatment. Based on both biological rationale and prior evidence of an impact on Parkinson's disease, we conducted an association study for 106 SNPs in the synaptic vesicle protein 2C (SV2C) gene using genetic and treatment response data from the Clinical Trial of Antipsychotic Intervention Effectiveness (CATIE). We examined response to the atypical antipsychotics for Caucasian subjects in the blinded phases, Phases 1A, 1B, and 2, of CATIE with sample sizes as follows: olanzapine (N=134), quetiapine (N=124), risperidone (N=134), and ziprasidone (N=74). Response was defined as change in the Positive and Negative Syndrome Scale (PANSS) score using a mixed model repeat measures approach. Subjects homozygous for the T allele of rs11960832 displayed significantly worse response to olanzapine treatment, the only finding with study-wide significance (p=2.94×10(-5); false discovery rate=2.18×10(-2)). These subjects also displayed worse response to quetiapine with nominal significance (p=4.56×10(-2)). While no other SNP achieved study-wide significance, one SNP (rs10214163) influencing Parkinson's disease displayed nominally significant association with olanzapine and quetiapine response, while the second such SNP (rs30196) showed a statistical trend toward correlating with olanzapine and quetiapine response. Furthermore, both coding SNPs examined (rs31244 and rs2270927) displayed nominally significant correlations with treatment response: one for olanzapine (rs227092), and one for quetiapine (rs31244). The fact that multiple SNPs in SV2C may impact response to atypical antipsychotics suggests that further evaluation of SNPs in this gene as PGx predictors of antipsychotic response is warranted.
Collapse
Affiliation(s)
- Timothy L. Ramsey
- SureGene, LLC, 600 Envoy Circle, suite 601, Louisville, KY 40299 United States
| | - Qian Liu
- SureGene, LLC, 600 Envoy Circle, suite 601, Louisville, KY 40299 United States
| | - Bill W. Massey
- SureGene, LLC, 600 Envoy Circle, suite 601, Louisville, KY 40299 United States
| | - Mark D. Brennan
- SureGene, LLC, 600 Envoy Circle, suite 601, Louisville, KY 40299 United States,Communicating author, Mark D. Brennan, SureGene, LLC, 600 Envoy Circle, suite 601, Louisville, KY 40299 United States, , Phone: 502-287-0899, Fax: 859-663-2984
| |
Collapse
|