Cargando…

Assessment of synaptic loss in mouse models of β-amyloid and tau pathology using [(18)F]UCB-H PET imaging

OBJECTIVE: In preclinical research, the use of [(18)F]Fluorodesoxyglucose (FDG) as a biomarker for neurodegeneration may induce bias due to enhanced glucose uptake by immune cells. In this study, we sought to investigate synaptic vesicle glycoprotein 2A (SV2A) PET with [(18)F]UCB-H as an alternative...

Descripción completa

Detalles Bibliográficos
Autores principales: Vogler, Letizia, Ballweg, Anna, Bohr, Bernd, Briel, Nils, Wind, Karin, Antons, Melissa, Kunze, Lea H., Gnörich, Johannes, Lindner, Simon, Gildehaus, Franz-Josef, Baumann, Karlheinz, Bartenstein, Peter, Boening, Guido, Ziegler, Sibylle I., Levin, Johannes, Zwergal, Andreas, Höglinger, Günter U., Herms, Jochen, Brendel, Matthias
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Elsevier 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10407951/
https://www.ncbi.nlm.nih.gov/pubmed/37541098
http://dx.doi.org/10.1016/j.nicl.2023.103484
_version_ 1785086078069243904
author Vogler, Letizia
Ballweg, Anna
Bohr, Bernd
Briel, Nils
Wind, Karin
Antons, Melissa
Kunze, Lea H.
Gnörich, Johannes
Lindner, Simon
Gildehaus, Franz-Josef
Baumann, Karlheinz
Bartenstein, Peter
Boening, Guido
Ziegler, Sibylle I.
Levin, Johannes
Zwergal, Andreas
Höglinger, Günter U.
Herms, Jochen
Brendel, Matthias
author_facet Vogler, Letizia
Ballweg, Anna
Bohr, Bernd
Briel, Nils
Wind, Karin
Antons, Melissa
Kunze, Lea H.
Gnörich, Johannes
Lindner, Simon
Gildehaus, Franz-Josef
Baumann, Karlheinz
Bartenstein, Peter
Boening, Guido
Ziegler, Sibylle I.
Levin, Johannes
Zwergal, Andreas
Höglinger, Günter U.
Herms, Jochen
Brendel, Matthias
author_sort Vogler, Letizia
collection PubMed
description OBJECTIVE: In preclinical research, the use of [(18)F]Fluorodesoxyglucose (FDG) as a biomarker for neurodegeneration may induce bias due to enhanced glucose uptake by immune cells. In this study, we sought to investigate synaptic vesicle glycoprotein 2A (SV2A) PET with [(18)F]UCB-H as an alternative preclinical biomarker for neurodegenerative processes in two mouse models representing the pathological hallmarks of Alzheimer’s disease (AD). METHODS: A total of 29 PS2APP, 20 P301S and 12 wild-type mice aged 4.4 to 19.8 months received a dynamic [(18)F]UCB-H SV2A-PET scan (14.7 ± 1.5 MBq) 0–60 min post injection. Quantification of tracer uptake in cortical, cerebellar and brainstem target regions was implemented by calculating relative volumes of distribution (V(T)) from an image-derived-input-function (IDIF). [(18)F]UCB-H binding was compared across all target regions between transgenic and wild-type mice. Additional static scans were performed in a subset of mice to compare [(18)F]FDG and [(18)F]GE180 (18 kDa translocator protein tracer as a surrogate for microglial activation) standardized uptake values (SUV) with [(18)F]UCB-H binding at different ages. Following the final scan, a subset of mouse brains was immunohistochemically stained with synaptic markers for gold standard validation of the PET results. RESULTS: [(18)F]UCB-H binding in all target regions was significantly reduced in 8-months old P301S transgenic mice when compared to wild-type controls (temporal lobe: p = 0.014; cerebellum: p = 0.0018; brainstem: p = 0.0014). Significantly lower SV2A tracer uptake was also observed in 13-months (temporal lobe: p = 0.0080; cerebellum: p = 0.006) and 19-months old (temporal lobe: p = 0.0042; cerebellum: p = 0.011) PS2APP transgenic versus wild-type mice, whereas the brainstem revealed no significantly altered [(18)F]UCB-H binding. Immunohistochemical analyses of post-mortem mouse brain tissue confirmed the SV2A PET findings. Correlational analyses of [(18)F]UCB-H and [(18)F]FDG using Pearson’s correlation coefficient revealed a significant negative association in the PS2APP mouse model (R = -0.26, p = 0.018). Exploratory analyses further stressed microglial activation as a potential reason for this inverse relationship, since [(18)F]FDG and [(18)F]GE180 quantification were positively correlated in this cohort (R = 0.36, p = 0.0076). CONCLUSION: [(18)F]UCB-H reliably depicts progressive synaptic loss in PS2APP and P301S transgenic mice, potentially qualifying as a more reliable alternative to [(18)F]FDG as a biomarker for assessment of neurodegeneration in preclinical research.
format Online
Article
Text
id pubmed-10407951
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Elsevier
record_format MEDLINE/PubMed
spelling pubmed-104079512023-08-09 Assessment of synaptic loss in mouse models of β-amyloid and tau pathology using [(18)F]UCB-H PET imaging Vogler, Letizia Ballweg, Anna Bohr, Bernd Briel, Nils Wind, Karin Antons, Melissa Kunze, Lea H. Gnörich, Johannes Lindner, Simon Gildehaus, Franz-Josef Baumann, Karlheinz Bartenstein, Peter Boening, Guido Ziegler, Sibylle I. Levin, Johannes Zwergal, Andreas Höglinger, Günter U. Herms, Jochen Brendel, Matthias Neuroimage Clin Regular Article OBJECTIVE: In preclinical research, the use of [(18)F]Fluorodesoxyglucose (FDG) as a biomarker for neurodegeneration may induce bias due to enhanced glucose uptake by immune cells. In this study, we sought to investigate synaptic vesicle glycoprotein 2A (SV2A) PET with [(18)F]UCB-H as an alternative preclinical biomarker for neurodegenerative processes in two mouse models representing the pathological hallmarks of Alzheimer’s disease (AD). METHODS: A total of 29 PS2APP, 20 P301S and 12 wild-type mice aged 4.4 to 19.8 months received a dynamic [(18)F]UCB-H SV2A-PET scan (14.7 ± 1.5 MBq) 0–60 min post injection. Quantification of tracer uptake in cortical, cerebellar and brainstem target regions was implemented by calculating relative volumes of distribution (V(T)) from an image-derived-input-function (IDIF). [(18)F]UCB-H binding was compared across all target regions between transgenic and wild-type mice. Additional static scans were performed in a subset of mice to compare [(18)F]FDG and [(18)F]GE180 (18 kDa translocator protein tracer as a surrogate for microglial activation) standardized uptake values (SUV) with [(18)F]UCB-H binding at different ages. Following the final scan, a subset of mouse brains was immunohistochemically stained with synaptic markers for gold standard validation of the PET results. RESULTS: [(18)F]UCB-H binding in all target regions was significantly reduced in 8-months old P301S transgenic mice when compared to wild-type controls (temporal lobe: p = 0.014; cerebellum: p = 0.0018; brainstem: p = 0.0014). Significantly lower SV2A tracer uptake was also observed in 13-months (temporal lobe: p = 0.0080; cerebellum: p = 0.006) and 19-months old (temporal lobe: p = 0.0042; cerebellum: p = 0.011) PS2APP transgenic versus wild-type mice, whereas the brainstem revealed no significantly altered [(18)F]UCB-H binding. Immunohistochemical analyses of post-mortem mouse brain tissue confirmed the SV2A PET findings. Correlational analyses of [(18)F]UCB-H and [(18)F]FDG using Pearson’s correlation coefficient revealed a significant negative association in the PS2APP mouse model (R = -0.26, p = 0.018). Exploratory analyses further stressed microglial activation as a potential reason for this inverse relationship, since [(18)F]FDG and [(18)F]GE180 quantification were positively correlated in this cohort (R = 0.36, p = 0.0076). CONCLUSION: [(18)F]UCB-H reliably depicts progressive synaptic loss in PS2APP and P301S transgenic mice, potentially qualifying as a more reliable alternative to [(18)F]FDG as a biomarker for assessment of neurodegeneration in preclinical research. Elsevier 2023-07-26 /pmc/articles/PMC10407951/ /pubmed/37541098 http://dx.doi.org/10.1016/j.nicl.2023.103484 Text en © 2023 The Author(s) https://creativecommons.org/licenses/by/4.0/This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Regular Article
Vogler, Letizia
Ballweg, Anna
Bohr, Bernd
Briel, Nils
Wind, Karin
Antons, Melissa
Kunze, Lea H.
Gnörich, Johannes
Lindner, Simon
Gildehaus, Franz-Josef
Baumann, Karlheinz
Bartenstein, Peter
Boening, Guido
Ziegler, Sibylle I.
Levin, Johannes
Zwergal, Andreas
Höglinger, Günter U.
Herms, Jochen
Brendel, Matthias
Assessment of synaptic loss in mouse models of β-amyloid and tau pathology using [(18)F]UCB-H PET imaging
title Assessment of synaptic loss in mouse models of β-amyloid and tau pathology using [(18)F]UCB-H PET imaging
title_full Assessment of synaptic loss in mouse models of β-amyloid and tau pathology using [(18)F]UCB-H PET imaging
title_fullStr Assessment of synaptic loss in mouse models of β-amyloid and tau pathology using [(18)F]UCB-H PET imaging
title_full_unstemmed Assessment of synaptic loss in mouse models of β-amyloid and tau pathology using [(18)F]UCB-H PET imaging
title_short Assessment of synaptic loss in mouse models of β-amyloid and tau pathology using [(18)F]UCB-H PET imaging
title_sort assessment of synaptic loss in mouse models of β-amyloid and tau pathology using [(18)f]ucb-h pet imaging
topic Regular Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10407951/
https://www.ncbi.nlm.nih.gov/pubmed/37541098
http://dx.doi.org/10.1016/j.nicl.2023.103484
work_keys_str_mv AT voglerletizia assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT ballweganna assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT bohrbernd assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT brielnils assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT windkarin assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT antonsmelissa assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT kunzeleah assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT gnorichjohannes assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT lindnersimon assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT gildehausfranzjosef assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT baumannkarlheinz assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT bartensteinpeter assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT boeningguido assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT zieglersibyllei assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT levinjohannes assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT zwergalandreas assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT hoglingergunteru assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT hermsjochen assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging
AT brendelmatthias assessmentofsynapticlossinmousemodelsofbamyloidandtaupathologyusing18fucbhpetimaging