Skip to main content
Top
Published in: EJNMMI Research 1/2020

Open Access 01-12-2020 | Obsessive-Compulsive Disorder | Original research

Sapap3 deletion causes dynamic synaptic density abnormalities: a longitudinal [11C]UCB-J PET study in a model of obsessive–compulsive disorder-like behaviour

Authors: Dorien Glorie, Jeroen Verhaeghe, Alan Miranda, Stef De Lombaerde, Sigrid Stroobants, Steven Staelens

Published in: EJNMMI Research | Issue 1/2020

Login to get access

Abstract

Background

Currently, the evidence on synaptic abnormalities in neuropsychiatric disorders—including obsessive–compulsive disorder (OCD)—is emerging. The newly established positron emission tomography (PET) ligand ((R)-1-((3-((11)C-methyl-(11)C)pyridin-4-yl)methyl)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one) ([11C]UCB-J) provides the opportunity to visualize synaptic density changes in vivo, by targeting the synaptic vesicle protein 2A (SV2A). Here, we aim to evaluate such alterations in the brain of the SAP90/PSD-95-associated protein 3 (Sapap3) knockout (ko) mouse model, showing an abnormal corticostriatal neurotransmission resulting in OCD-like behaviour.

Methods

Longitudinal [11C]UCB-J µPET/CT scans were acquired in Sapap3 ko and wildtype (wt) control mice (n = 9/group) to study SV2A availability. Based on the Logan reference method, we calculated the volume of distribution (VT(IDIF)) for [11C]UCB-J. Both cross-sectional (wt vs. ko) and longitudinal (3 vs. 9 months) volume-of-interest-based statistical analysis and voxel-based statistical parametric mapping were performed. Both [11C]UCB-J ex vivo autoradiography and [3H]UCB-J in vitro autoradiography were used for the validation of the µPET data.

Results

At the age of 3 months, Sapap3 ko mice are already characterized by a significantly lower SV2A availability compared to wt littermates (i.a. cortex − 12.69%, p < 0.01; striatum − 14.12%, p < 0.001, thalamus − 13.11%, p < 0.001, and hippocampus − 12.99%, p < 0.001). Healthy ageing in control mice was associated with a diffuse and significant (p < 0.001) decline throughout the brain, whereas in Sapap3 ko mice this decline was more confined to the corticostriatal level. A strong linear relationship (p < 0.0001) was established between the outcome parameters of [11C]UCB-J µPET and [11C]UCB-J ex vivo autoradiography, while such relationship was absent for [3H]UCB-J in vitro autoradiography.

Conclusions

[11C]UCB-J PET is a potential marker for synaptic density deficits in the Sapap3 ko mouse model for OCD, parallel to disease progression. Our data suggest that [11C]UCB-J ex vivo autoradiography is a suitable proxy for [11C]UCB-J PET data in mice.
Appendix
Available only for authorised users
Literature
2.
go back to reference Abramowitz JS, Taylor S, McKay D. Obsessive-compulsive disorder. Lancet. 2009;374:491–9. CrossRef Abramowitz JS, Taylor S, McKay D. Obsessive-compulsive disorder. Lancet. 2009;374:491–9. CrossRef
4.
go back to reference Graybiel AM, Rauch SL. Toward a neurobiology of obsessive-compulsive disorder. Neuron. 2000;28:343–7. CrossRef Graybiel AM, Rauch SL. Toward a neurobiology of obsessive-compulsive disorder. Neuron. 2000;28:343–7. CrossRef
5.
go back to reference Pauls DL, Abramovitch A, Rauch SL, Geller DA. Obsessive-compulsive disorder: an integrative genetic and neurobiological perspective. Nat Rev Neurosci. 2014;15:410–24. CrossRef Pauls DL, Abramovitch A, Rauch SL, Geller DA. Obsessive-compulsive disorder: an integrative genetic and neurobiological perspective. Nat Rev Neurosci. 2014;15:410–24. CrossRef
6.
go back to reference Ting JT, Feng G. Neurobiology of obsessive-compulsive disorder: insights into neural circuitry dysfunction through mouse genetics. Curr Opin Neurobiol. 2011;21:842–8. CrossRef Ting JT, Feng G. Neurobiology of obsessive-compulsive disorder: insights into neural circuitry dysfunction through mouse genetics. Curr Opin Neurobiol. 2011;21:842–8. CrossRef
17.
go back to reference Burguière E, Monteiro P, Feng G, Graybiel AM. Optogenetic stimulation of lateral orbitofronto-striatal pathway suppresses compulsive behaviors. Science. 2013;340:1243–6. CrossRef Burguière E, Monteiro P, Feng G, Graybiel AM. Optogenetic stimulation of lateral orbitofronto-striatal pathway suppresses compulsive behaviors. Science. 2013;340:1243–6. CrossRef
22.
go back to reference Ade KK, Wan Y, Hamann HC, et al. Increased metabotropic glutamate receptor 5 signaling underlies obsessive-compulsive disorder-like behavioral and striatal circuit abnormalities in mice. Biol Psychiatry. 2016;80:522–33. CrossRef Ade KK, Wan Y, Hamann HC, et al. Increased metabotropic glutamate receptor 5 signaling underlies obsessive-compulsive disorder-like behavioral and striatal circuit abnormalities in mice. Biol Psychiatry. 2016;80:522–33. CrossRef
24.
go back to reference Wan Y, Ade KK, Caffall Z, Ilcim Ozlu M, Eroglu C, Feng G, Calakos N. Circuit-selective striatal synaptic dysfunction in the Sapap3 knockout mouse model of obsessive-compulsive disorder. Biol Psychiatry. 2014;75:623–30. CrossRef Wan Y, Ade KK, Caffall Z, Ilcim Ozlu M, Eroglu C, Feng G, Calakos N. Circuit-selective striatal synaptic dysfunction in the Sapap3 knockout mouse model of obsessive-compulsive disorder. Biol Psychiatry. 2014;75:623–30. CrossRef
43.
go back to reference Sherer M, Padilla S. A case of obsessiveness induced by levetiracetam in a patient with epilepsy, intellectual disability and pervasive developmental disorder. Ment Heal Asp Dev Disabil. 2008;11:22–6. Sherer M, Padilla S. A case of obsessiveness induced by levetiracetam in a patient with epilepsy, intellectual disability and pervasive developmental disorder. Ment Heal Asp Dev Disabil. 2008;11:22–6.
46.
go back to reference Khouzam HR. Levetiracetam treatment of refractory obsessive-compulsive disorder. Ann Psychiatry Ment Health. 2015;3(6):1045. Khouzam HR. Levetiracetam treatment of refractory obsessive-compulsive disorder. Ann Psychiatry Ment Health. 2015;3(6):1045.
47.
go back to reference Hume SP, Gunn RN, Jones T. Pharmacological constraints associated with positron emission tomographic scanning of small laboratory animals. Eur J Nucl Med. 1998;25:173–6. CrossRef Hume SP, Gunn RN, Jones T. Pharmacological constraints associated with positron emission tomographic scanning of small laboratory animals. Eur J Nucl Med. 1998;25:173–6. CrossRef
66.
go back to reference Milad MR, Rauch SL. Obsessive-compulsive disorder: beyond segregated cortico-striatal pathways. Trends Cogn Sci. 2012;16:43–51. CrossRef Milad MR, Rauch SL. Obsessive-compulsive disorder: beyond segregated cortico-striatal pathways. Trends Cogn Sci. 2012;16:43–51. CrossRef
70.
go back to reference Carson R, Naganawa M, Matuskey D, et al. Age and sex effects on synaptic density in healthy humans as assessed with SV2A PET. J Nucl Med. 2018;59:541. Carson R, Naganawa M, Matuskey D, et al. Age and sex effects on synaptic density in healthy humans as assessed with SV2A PET. J Nucl Med. 2018;59:541.
Metadata
Title
Sapap3 deletion causes dynamic synaptic density abnormalities: a longitudinal [11C]UCB-J PET study in a model of obsessive–compulsive disorder-like behaviour
Authors
Dorien Glorie
Jeroen Verhaeghe
Alan Miranda
Stef De Lombaerde
Sigrid Stroobants
Steven Staelens
Publication date
01-12-2020
Publisher
Springer Berlin Heidelberg
Published in
EJNMMI Research / Issue 1/2020
Electronic ISSN: 2191-219X
DOI
https://doi.org/10.1186/s13550-020-00721-2

Other articles of this Issue 1/2020

EJNMMI Research 1/2020 Go to the issue