1
|
Kikuchi T, Okamura T, Zhang MR. Numerical simulation method for the assessment of the effect of molar activity on the pharmacokinetics of radioligands in small animals. EJNMMI Radiopharm Chem 2024; 9:78. [PMID: 39570519 PMCID: PMC11582259 DOI: 10.1186/s41181-024-00308-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND It is well recognized that the molar activity of a radioligand is an important pharmacokinetic parameter, especially in positron emission tomography (PET) of small animals. Occupation of a significant number of binding sites by radioligand molecules results in low radioligand accumulation in a target region (mass effect). Nevertheless, small-animal PET studies have often been performed without consideration of the molar activity or molar dose of radioligands. A simulation study would therefore help to assess the importance of the mass effect in small-animal PET. Here, we introduce a new compartmental model-based numerical method, which runs on commonly used spreadsheet software, to simulate the effect of molar activity or molar dose on the pharmacokinetics of radioligands. RESULTS Assuming a two-tissue compartmental model, time-concentration curves of a radioligand were generated using four simulation methods and the well-known Runge-Kutta numerical method. The values were compared with theoretical values obtained under an ultra-high molar activity condition (pseudo-first-order binding kinetics), a steady-state condition and an equilibrium condition (second-order binding kinetics). For all conditions, the simulation method using the simplest calculation yielded values closest to the theoretical values and comparable with those obtained using the Runge-Kutta method. To satisfy a maximum occupancy less than 5%, simulations showed that a molar activity greater than 150 GBq/μmol is required for a model radioligand when 20 MBq is administered to a 250 g rat and when the concentration of binding sites in target regions is greater than 1.25 nM. CONCLUSIONS The simulation method used in this study is based on a very simple calculation and runs on widely used spreadsheet software. Therefore, simulation of radioligand pharmacokinetics using this method can be performed on a personal computer and help to assess the importance of the mass effect in small-animal PET. This simulation method also enables the generation of a model time-activity curve for the evaluation of kinetic analysis methods.
Collapse
Affiliation(s)
- Tatsuya Kikuchi
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.
| | - Toshimitsu Okamura
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| |
Collapse
|
2
|
Maqbool M, Jakobsson JE, Alluri SR, Kramer V, Riss PJ. A protocol for controlled reactivity shift in the 2,2-difluorovinyl motif used for selective S- 18F and C- 18F bond formation. Commun Chem 2024; 7:97. [PMID: 38684771 PMCID: PMC11058245 DOI: 10.1038/s42004-024-01132-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/20/2024] [Indexed: 05/02/2024] Open
Abstract
Positron emission tomography (PET) is a powerful imaging technique for biomedical research, drug development and medical diagnosis. The power of PET lies in biochemically selective radiotracers, labelled with positron emitters like fluorine-18 image chemical processes in vivo. A rapid and remarkably efficient, unprecedented protocol to select between S-F and C-F bond formation based on activation of 1,1-difluoroethylene groups followed by selective oxidation or reduction is described. While transition metal mediated conditions can be employed, the reaction proceeds in high yield using unobjectionable chemical reagents amenable to routine radiotracer production. The latter bodes well for facile clinical translation of the method. The new technique affords radiotracers and the labelling reagent 2,2-difluoro-2-(fluoro-18F)ethyl 4-methylbenzenesulfonate ([18F]1b) in excellent yield. Following oxygenation of the reaction mixture with medical oxygen or air, sulfonyl fluorides are obtained as the primary product. The new protocol was employed in a proof of principle to develop a radiometric assay for quantitation of sulfonylation yield with sulfonyl fluoride reagents. With operational ease and mild conditions, the method bodes a high potential for radiolabelling of biomolecules, known enzyme inhibitors and other temperature-sensitive compounds.
Collapse
Affiliation(s)
- Mudasir Maqbool
- Department of Clinical Neurocience, OUS-Ullevål, Oslo, Norway
- Department of Chemistry, University of Oslo, Oslo, Norway
| | | | | | - Vasko Kramer
- Positronpharma SA, Rancangua, Santiago de Chile, Santiago, Chile
| | - Patrick Johannes Riss
- Department of Clinical Neurocience, OUS-Ullevål, Oslo, Norway.
- Department of Chemistry, University of Oslo, Oslo, Norway.
- Department of Chemistry, Johannes Gutenberg-University, Fritz-Strassmann-Weg 2, 55128, Mainz, Germany.
| |
Collapse
|
3
|
Curia G, Estrada-Camarena E, Manjarrez E, Mizuno H. Editorial: In vivo investigations on neurological disorders: From traditional approaches to forefront technologies. Front Neurosci 2022; 16:1052089. [PMID: 36330344 PMCID: PMC9623258 DOI: 10.3389/fnins.2022.1052089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 10/05/2022] [Indexed: 11/25/2022] Open
Affiliation(s)
- Giulia Curia
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- *Correspondence: Giulia Curia
| | - Erika Estrada-Camarena
- Laboratory of Neuropsychopharmacology, Neuroscience, National Institute of Psychiatry Ramon de la Fuente Muñiz (INPRFM), Mexico City, Mexico
| | - Elias Manjarrez
- Institute of Physiology, Benemerita Universidad Autonoma de Puebla, Puebla, Mexico
| | - Hidenobu Mizuno
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| |
Collapse
|
4
|
Accrombessi G, Galineau L, Tauber C, Serrière S, Moyer E, Brizard B, Le Guisquet AM, Surget A, Belzung C. An ecological animal model of subthreshold depression in adolescence: behavioral and resting state 18F-FDG PET imaging characterization. Transl Psychiatry 2022; 12:356. [PMID: 36050307 PMCID: PMC9436927 DOI: 10.1038/s41398-022-02119-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/09/2022] Open
Abstract
The different depressive disorders that exist can take root at adolescence. For instance, some functional and structural changes in several brain regions have been observed from adolescence in subjects that display either high vulnerability to depressive symptoms or subthreshold depression. For instance, adolescents with depressive disorder have been shown to exhibit hyperactivity in hippocampus, amygdala and prefrontal cortex as well as volume reductions in hippocampus and amygdala (prefrontal cortex showing more variable results). However, no animal model of adolescent subthreshold depression has been developed so far. Our objective was to design an animal model of adolescent subthreshold depression and to characterize the neural changes associated to this phenotype. For this purpose, we used adolescent Swiss mice that were evaluated on 4 tests assessing cognitive abilities (Morris water maze), anhedonia (sucrose preference), anxiety (open-field) and stress-coping strategies (forced swim test) at postnatal day (PND) 28-35. In order to identify neural alterations associated to behavioral profiles, we assessed brain resting state metabolic activity in vivo using 18F-FDG PET imaging at PND 37. We selected three profiles of mice distinguished in a composite Z-score computed from performances in the behavioral tests: High, Intermediate and Low Depressive Risk (HDR, IDR and LDR). Compared to both IDR and LDR, HDR mice were characterized by passive stress-coping behaviors, low cognition and high anhedonia and anxiety and were associated with significant changes of 18F-FDG uptakes in several cortical and subcortical areas including prelimbic cortex, infralimbic cortex, nucleus accumbens, amygdala, periaqueductal gray and superior colliculus, all displaying higher metabolic activity, while only the thalamus was associated with lower metabolic activity (compared to IDR). LDR displayed an opposing behavioral phenotype and were associated with significant changes of 18F-FDG uptakes in the dorsal striatum and thalamus that both exhibited markedly lower metabolic activity in LDR. In conclusion, our study revealed changes in metabolic activities that can represent neural signatures for behavioral profiles predicting subthreshold depression at adolescence in a mouse model.
Collapse
Affiliation(s)
- Georgine Accrombessi
- grid.411167.40000 0004 1765 1600UMR 1253, iBrain, Inserm, Université de Tours, CEDEX 1, 37032 Tours, France
| | - Laurent Galineau
- grid.411167.40000 0004 1765 1600UMR 1253, iBrain, Inserm, Université de Tours, CEDEX 1, 37032 Tours, France
| | - Clovis Tauber
- grid.411167.40000 0004 1765 1600UMR 1253, iBrain, Inserm, Université de Tours, CEDEX 1, 37032 Tours, France
| | - Sophie Serrière
- grid.411167.40000 0004 1765 1600UMR 1253, iBrain, Inserm, Université de Tours, CEDEX 1, 37032 Tours, France
| | - Esteban Moyer
- grid.411167.40000 0004 1765 1600UMR 1253, iBrain, Inserm, Université de Tours, CEDEX 1, 37032 Tours, France
| | - Bruno Brizard
- grid.411167.40000 0004 1765 1600UMR 1253, iBrain, Inserm, Université de Tours, CEDEX 1, 37032 Tours, France
| | - Anne-Marie Le Guisquet
- grid.411167.40000 0004 1765 1600UMR 1253, iBrain, Inserm, Université de Tours, CEDEX 1, 37032 Tours, France
| | - Alexandre Surget
- grid.411167.40000 0004 1765 1600UMR 1253, iBrain, Inserm, Université de Tours, CEDEX 1, 37032 Tours, France
| | - Catherine Belzung
- UMR 1253, iBrain, Inserm, Université de Tours, CEDEX 1, 37032, Tours, France.
| |
Collapse
|
5
|
Jaquins-Gerstl A, Nesbitt KM, Michael AC. In vivo evidence for the unique kinetics of evoked dopamine release in the patch and matrix compartments of the striatum. Anal Bioanal Chem 2021; 413:6703-6713. [PMID: 33843017 PMCID: PMC8551084 DOI: 10.1007/s00216-021-03300-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/11/2021] [Accepted: 03/16/2021] [Indexed: 11/29/2022]
Abstract
The neurochemical transmitter dopamine (DA) is implicated in a number of diseases states, including Parkinson's disease, schizophrenia, and drug abuse. DA terminal fields in the dorsal striatum and core region of the nucleus accumbens in the rat brain are organized as heterogeneous domains exhibiting fast and slow kinetic of DA release. The rates of dopamine release are significantly and substantially faster in the fast domains relative to the slow domains. The striatum is composed of a mosaic of spatial compartments known as the striosomes (patches) and the matrix. Extensive literature exists on the spatial organization of the patch and matrix compartments and their functions. However, little is known about these compartments as they relate to fast and slow kinetic DA domains observed by fast scan cyclic voltammetry (FSCV). Thus, we combined high spatial resolution of FSCV with detailed immunohistochemical analysis of these architectural compartments (patch and matrix) using fluorescence microscopy. Our findings demonstrated a direct correlation between patch compartments with fast domain DA kinetics and matrix compartments to slow domain DA kinetics. We also investigated the kinetic domains in two very distinct sub-regions in the striatum, the lateral dorsal striatum (LDS) and the medial dorsal striatum (MDS). The lateral dorsal striatum as opposed to the medial dorsal striatum is mainly governed by fast kinetic DA domains. These finding are highly relevant as they may hold key promise in unraveling the fast and slow kinetic DA domains and their physiological significance.
Collapse
Affiliation(s)
- Andrea Jaquins-Gerstl
- Department of Chemistry, Chevron Science Center, University of Pittsburgh, 219 Parkman Ave., Pittsburgh, PA, 15213, USA.
| | - Kathryn M Nesbitt
- Department of Chemistry, Chevron Science Center, University of Pittsburgh, 219 Parkman Ave., Pittsburgh, PA, 15213, USA
| | - Adrian C Michael
- Department of Chemistry, Chevron Science Center, University of Pittsburgh, 219 Parkman Ave., Pittsburgh, PA, 15213, USA
| |
Collapse
|
6
|
Chang CY, Luo DZ, Pei JC, Kuo MC, Hsieh YC, Lai WS. Not Just a Bystander: The Emerging Role of Astrocytes and Research Tools in Studying Cognitive Dysfunctions in Schizophrenia. Int J Mol Sci 2021; 22:ijms22105343. [PMID: 34069523 PMCID: PMC8160762 DOI: 10.3390/ijms22105343] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/14/2021] [Accepted: 05/14/2021] [Indexed: 12/16/2022] Open
Abstract
Cognitive dysfunction is one of the core symptoms in schizophrenia, and it is predictive of functional outcomes and therefore useful for treatment targets. Rather than improving cognitive deficits, currently available antipsychotics mainly focus on positive symptoms, targeting dopaminergic/serotoninergic neurons and receptors in the brain. Apart from investigating the neural mechanisms underlying schizophrenia, emerging evidence indicates the importance of glial cells in brain structure development and their involvement in cognitive functions. Although the etiopathology of astrocytes in schizophrenia remains unclear, accumulated evidence reveals that alterations in gene expression and astrocyte products have been reported in schizophrenic patients. To further investigate the role of astrocytes in schizophrenia, we highlighted recent progress in the investigation of the effect of astrocytes on abnormalities in glutamate transmission and impairments in the blood–brain barrier. Recent advances in animal models and behavioral methods were introduced to examine schizophrenia-related cognitive deficits and negative symptoms. We also highlighted several experimental tools that further elucidate the role of astrocytes. Instead of focusing on schizophrenia as a neuron-specific disorder, an additional astrocytic perspective provides novel and promising insight into its causal mechanisms and treatment. The involvement of astrocytes in the pathogenesis of schizophrenia and other brain disorders is worth further investigation.
Collapse
Affiliation(s)
- Chia-Yuan Chang
- Department of Psychology, National Taiwan University, Taipei 10617, Taiwan; (C.-Y.C.); (D.-Z.L.); (J.-C.P.); (Y.-C.H.)
- Neurobiology and Cognitive Science Center, National Taiwan University, Taipei 10617, Taiwan;
| | - Da-Zhong Luo
- Department of Psychology, National Taiwan University, Taipei 10617, Taiwan; (C.-Y.C.); (D.-Z.L.); (J.-C.P.); (Y.-C.H.)
| | - Ju-Chun Pei
- Department of Psychology, National Taiwan University, Taipei 10617, Taiwan; (C.-Y.C.); (D.-Z.L.); (J.-C.P.); (Y.-C.H.)
| | - Ming-Che Kuo
- Neurobiology and Cognitive Science Center, National Taiwan University, Taipei 10617, Taiwan;
- Department of Neurology, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Yi-Chen Hsieh
- Department of Psychology, National Taiwan University, Taipei 10617, Taiwan; (C.-Y.C.); (D.-Z.L.); (J.-C.P.); (Y.-C.H.)
| | - Wen-Sung Lai
- Department of Psychology, National Taiwan University, Taipei 10617, Taiwan; (C.-Y.C.); (D.-Z.L.); (J.-C.P.); (Y.-C.H.)
- Neurobiology and Cognitive Science Center, National Taiwan University, Taipei 10617, Taiwan;
- Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei 10617, Taiwan
- Correspondence: ; Tel.: +886-2-3366-3112; Fax: +886-2-3362-9909
| |
Collapse
|
7
|
Abstract
Positron emission tomography (PET) is a non-invasive imaging technology employed to describe metabolic, physiological, and biochemical processes in vivo. These include receptor availability, metabolic changes, neurotransmitter release, and alterations of gene expression in the brain. Since the introduction of dedicated small-animal PET systems along with the development of many novel PET imaging probes, the number of PET studies using rats and mice in basic biomedical research tremendously increased over the last decade. This article reviews challenges and advances of quantitative rodent brain imaging to make the readers aware of its physical limitations, as well as to inspire them for its potential applications in preclinical research. In the first section, we briefly discuss the limitations of small-animal PET systems in terms of spatial resolution and sensitivity and point to possible improvements in detector development. In addition, different acquisition and post-processing methods used in rodent PET studies are summarized. We further discuss factors influencing the test-retest variability in small-animal PET studies, e.g., different receptor quantification methodologies which have been mainly translated from human to rodent receptor studies to determine the binding potential and changes of receptor availability and radioligand affinity. We further review different kinetic modeling approaches to obtain quantitative binding data in rodents and PET studies focusing on the quantification of endogenous neurotransmitter release using pharmacological interventions. While several studies have focused on the dopamine system due to the availability of several PET tracers which are sensitive to dopamine release, other neurotransmitter systems have become more and more into focus and are described in this review, as well. We further provide an overview of latest genome engineering technologies, including the CRISPR/Cas9 and DREADD systems that may advance our understanding of brain disorders and function and how imaging has been successfully applied to animal models of human brain disorders. Finally, we review the strengths and opportunities of simultaneous PET/magnetic resonance imaging systems to study drug-receptor interactions and challenges for the translation of PET results from bench to bedside.
Collapse
|
8
|
Bekker RBW, Fjellaksel R, Hjornevik T, Nuruddin S, Rafique W, Hansen JH, Sundset R, Haraldsen IH, Riss PJ. Discovery of a Lead Brain-Penetrating Gonadotropin-Releasing Hormone Receptor Antagonist with Saturable Binding in Brain. ChemMedChem 2020; 15:1624-1628. [PMID: 32677155 PMCID: PMC7540054 DOI: 10.1002/cmdc.202000256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/12/2020] [Indexed: 11/30/2022]
Abstract
We report the synthesis, radiosynthesis and biological characterisation of two gonadotropin-releasing hormone receptor (GnRH-R) antagonists with nanomolar binding affinity. A small library of GnRH-R antagonists was synthesised in 20-67 % overall yield with the aim of identifying a high-affinity antagonist capable of crossing the blood-brain barrier. Binding affinity to rat GnRH-R was determined by autoradiography in competitive-binding studies against [125 I]buserelin, and inhibition constants were calculated by using the Cheng-Prusoff equation. The radioligands were obtained in 46-79 % radiochemical yield and >95 % purity and with a molar activity of 19-38 MBq/nmol by direct nucleophilic radiofluorination. Positron emission tomography imaging in rat under baseline conditions in comparison to pretreatment with a receptor-saturating dose of GnRH antagonist revealed saturable uptake (0.1 %ID/mL) into the brain.
Collapse
Affiliation(s)
| | - Richard Fjellaksel
- Department of Clinical MedicineUiT The Arctic University of NorwayHansine Hansens veg 189019TromsøNorway
- PET Imaging CenterUniversity Hospital of North NorwaySykehusvegen 389019TromsøNorway
- Department of ChemistryUiT – The Arctic University of NorwayHansine Hansens veg 189019TromsøNorway
| | - Trine Hjornevik
- Department of Diagnostic PhysicsOslo University HospitalSognsvannsveien 200372OsloNorway
| | - Syed Nuruddin
- Norwegian Medical Cyclotron AS, RikshospitaletSognsvannsveien 20OsloNorway
| | - Waqas Rafique
- Department of ChemistryUniversity of OsloSem Sælands vei, 260371OsloNorway
| | - Jørn H. Hansen
- Department of ChemistryUiT – The Arctic University of NorwayHansine Hansens veg 189019TromsøNorway
| | - Rune Sundset
- Department of Clinical MedicineUiT The Arctic University of NorwayHansine Hansens veg 189019TromsøNorway
- PET Imaging CenterUniversity Hospital of North NorwaySykehusvegen 389019TromsøNorway
| | - Ira H. Haraldsen
- Clinical NeurosciencesOslo University Hospital-UllevalKirkeveien 166, post code?OsloNorway
| | - Patrick J. Riss
- Department of ChemistryUniversity of OsloSem Sælands vei, 260371OsloNorway
- Norwegian Medical Cyclotron AS, RikshospitaletSognsvannsveien 20OsloNorway
- Clinical NeurosciencesOslo University Hospital-UllevalKirkeveien 166, post code?OsloNorway
| |
Collapse
|
9
|
Ott J, Spilhaug MM, Maschauer S, Rafique W, Jakobsson JE, Hartvig K, Hübner H, Gmeiner P, Prante O, Riss PJ. Pharmacological Characterization of Low-to-Moderate Affinity Opioid Receptor Agonists and Brain Imaging with 18F-Labeled Derivatives in Rats. J Med Chem 2020; 63:9484-9499. [PMID: 32787100 PMCID: PMC7497404 DOI: 10.1021/acs.jmedchem.0c00683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The 3,4-dichloro-N-(1-(dimethylamino)cyclohexyl)methyl benzamide scaffold was studied as a template for 18F-positron emission tomography (18F-PET) radiotracer development emphasizing sensitivity to changes in opioid receptor (OR) occupancy over high affinity. Agonist potency, binding affinity, and relevant pharmacological parameters of 15 candidates were investigated. Two promising compounds 3b and 3e with μ-OR (MOR) selective agonist activity in the moderate range (EC50 = 1-100 nM) were subjected to 18F-fluorination, autoradiography, and small-animal PET imaging. Radioligands [18F]3b and [18F]3e were obtained in activity yields of 21 ± 5 and 23 ± 4% and molar activities of 25-40 and 200-300 GBq/μmol, respectively. Displaceable binding matching MOR distribution in the brain was confirmed by imaging. Radioligands showed a rapid pharmacokinetic profile; however, metabolite-corrected, blood-based modeling was required for data analysis. Observed BPND was low, although treatment with naloxone leads to a marked decrease in specific binding, confirming the discovery of a new template for 18F-labeled OR-agonist PET ligands.
Collapse
Affiliation(s)
- Julian Ott
- Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Translational Research Center, Friedrich Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 12, D-91054 Erlangen, Germany
| | - Mona M Spilhaug
- Realomics SRI, Kjemisk Institutt, Universitetet i Oslo, N-0376 Oslo, Norway
| | - Simone Maschauer
- Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Translational Research Center, Friedrich Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 12, D-91054 Erlangen, Germany
| | - Waqas Rafique
- Realomics SRI, Kjemisk Institutt, Universitetet i Oslo, N-0376 Oslo, Norway
| | - Jimmy E Jakobsson
- Realomics SRI, Kjemisk Institutt, Universitetet i Oslo, N-0376 Oslo, Norway
| | - Karoline Hartvig
- Realomics SRI, Kjemisk Institutt, Universitetet i Oslo, N-0376 Oslo, Norway
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, Germany
| | - Olaf Prante
- Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Translational Research Center, Friedrich Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 12, D-91054 Erlangen, Germany
| | - Patrick J Riss
- Realomics SRI, Kjemisk Institutt, Universitetet i Oslo, N-0376 Oslo, Norway.,Division of Clinical Neuroscience, Neuroscience Research Unit, OUS-UllevÅl, N-0450 Oslo, Norway
| |
Collapse
|
10
|
Neves LT, Neves PFR, Paz LV, Zancan M, Milanesi BB, Lazzari GZ, da Silva RB, de Oliveira MMBP, Venturin GT, Greggio S, da Costa JC, Rasia-Filho AA, Mestriner RG, Xavier LL. Increases in dendritic spine density in BLA without metabolic changes in a rodent model of PTSD. Brain Struct Funct 2019; 224:2857-2870. [PMID: 31440907 DOI: 10.1007/s00429-019-01943-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 08/13/2019] [Indexed: 01/07/2023]
Abstract
Imaging studies have shown abnormal amygdala function in patients with posttraumatic stress disorder (PTSD). In addition, alterations in synaptic plasticity have been associated with psychiatric disorders and previous reports have indicated alterations in the amygdala morphology, especially in basolateral (BLA) neurons, are associated with stress-related disorders. Since, some individuals exposed to a traumatic event develop PTSD, the goals of this study were to evaluate the early effects of PTSD on amygdala glucose metabolism and analyze the possible BLA dendritic spine plasticity in animals with different levels of behavioral response. We employed the inescapable footshock protocol as an experimental model of PTSD and the animals were classified according to the duration of their freezing behavior into distinct groups: "extreme behavioral response" (EBR) and "minimal behavioral response". We evaluated the amygdala glucose metabolism at baseline (before the stress protocol) and immediately after the situational reminder using the microPET and the radiopharmaceutical 18F-FDG. The BLA dendritic spines were analyzed according to their number, density, shape and morphometric parameters. Our results show the EBR animals exhibited longer freezing behavior and increased proximal dendritic spines density in the BLA neurons. Neither the amygdaloid glucose metabolism, the types of dendritic spines nor their morphometric parameters showed statistically significant differences. The extreme behavior response induced by this PTSD protocol produces an early increase in BLA spine density, which is unassociated with either additional changes in the shape of spines or metabolic changes in the whole amygdala of Wistar rats.
Collapse
Affiliation(s)
- Laura Tartari Neves
- Laboratório de Biologia Celular e Tecidual, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Av. Ipiranga 6681, Prédio 12C, Sala 104, Porto Alegre, Rio Grande do Sul, CEP 90619-900, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Paula Fernanda Ribas Neves
- Laboratório de Biologia Celular e Tecidual, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Av. Ipiranga 6681, Prédio 12C, Sala 104, Porto Alegre, Rio Grande do Sul, CEP 90619-900, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Lisiê Valéria Paz
- Laboratório de Biologia Celular e Tecidual, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Av. Ipiranga 6681, Prédio 12C, Sala 104, Porto Alegre, Rio Grande do Sul, CEP 90619-900, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Mariana Zancan
- Departamento de Ciências Básicas/Fisiologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Bruna Bueno Milanesi
- Laboratório de Biologia Celular e Tecidual, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Av. Ipiranga 6681, Prédio 12C, Sala 104, Porto Alegre, Rio Grande do Sul, CEP 90619-900, Brazil
| | - Gabriele Zenato Lazzari
- Laboratório de Biologia Celular e Tecidual, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Av. Ipiranga 6681, Prédio 12C, Sala 104, Porto Alegre, Rio Grande do Sul, CEP 90619-900, Brazil
| | - Rafaela Barboza da Silva
- Laboratório de Biologia Celular e Tecidual, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Av. Ipiranga 6681, Prédio 12C, Sala 104, Porto Alegre, Rio Grande do Sul, CEP 90619-900, Brazil
| | - Marina Mena Barreto Peres de Oliveira
- Laboratório de Biologia Celular e Tecidual, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Av. Ipiranga 6681, Prédio 12C, Sala 104, Porto Alegre, Rio Grande do Sul, CEP 90619-900, Brazil
| | - Gianina Teribele Venturin
- Instituto do Cérebro do Rio Grande do Sul (InsCer), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Samuel Greggio
- Instituto do Cérebro do Rio Grande do Sul (InsCer), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Jaderson Costa da Costa
- Instituto do Cérebro do Rio Grande do Sul (InsCer), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Alberto A Rasia-Filho
- Departamento de Ciências Básicas/Fisiologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Régis Gemerasca Mestriner
- Laboratório de Biologia Celular e Tecidual, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Av. Ipiranga 6681, Prédio 12C, Sala 104, Porto Alegre, Rio Grande do Sul, CEP 90619-900, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Léder Leal Xavier
- Laboratório de Biologia Celular e Tecidual, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Av. Ipiranga 6681, Prédio 12C, Sala 104, Porto Alegre, Rio Grande do Sul, CEP 90619-900, Brazil. .,Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
11
|
Abstract
To aid in the analysis of rhesus macaque brain images, we aligned digitized anatomical regions from the widely used atlas of Paxinos et al. to a published magnetic resonance imaging (MRI) template based on a large number of subjects. Digitally labelled atlas images were aligned to the template in 2D and then in 3D. The resulting grey matter regions appear qualitatively to be well registered to the template. To quantitatively validate the procedure, MR brain images of 20 rhesus macaques were aligned to the template along with regions drawn by hand in striatal and cortical areas in each subject's MRI. There was good geometric overlap between the hand drawn regions and the template regions. Positron emission tomography (PET) images of the same subjects showing uptake of a dopamine D2 receptor ligand were aligned to the template space, and good agreement was found between tracer binding measures calculated using the hand drawn and template regions. In conclusion, an anatomically defined set of rhesus macaque brain regions has been aligned to an MRI template and has been validated for analysis of PET imaging in a subset of striatal and cortical areas. The entire set of over 200 regions is publicly available at https://www.nitrc.org/ . Graphical Abstract ᅟ.
Collapse
|
12
|
Yasmin A, Pitkänen A, Jokivarsi K, Poutiainen P, Gröhn O, Immonen R. MRS Reveals Chronic Inflammation in T2w MRI-Negative Perilesional Cortex - A 6-Months Multimodal Imaging Follow-Up Study. Front Neurosci 2019; 13:863. [PMID: 31474824 PMCID: PMC6707062 DOI: 10.3389/fnins.2019.00863] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/31/2019] [Indexed: 12/14/2022] Open
Abstract
Sustained inflammation in the injured cortex is a promising therapeutic target for disease-modification after traumatic brain injury (TBI). However, its extent and dynamics of expansion are incompletely understood which challenges the timing and placement of therapeutics to lesioned area. Our aim was to characterize the evolution of chronic inflammation during lesion expansion in lateral fluid-percussion injury (FPI) rat model with focus on the MRI-negative perilesional cortex. T2-weighted MR imaging (T2w MRI) and localized magnetic resonance spectroscopy (MRS) were performed at 1, 3, and 6 months post-injury. End-point histology, including Nissl for neuronal death, GFAP for astrogliosis, and Prussian Blue for iron were used to assess perilesional histopathology. An additional animal cohort was imaged with a positron emission tomography (PET) using translocator protein 18 kDa (TSPO) radiotracer [18F]-FEPPA. T2w MRI assessed lesion growth and detected chronic inflammation along the lesion border while rest of the ipsilateral cortex was MRI-negative (MRI-). Instead, myo-inositol that is an inflammatory MRS marker for gliosis, glutathione for oxidative stress, and choline for membrane turnover were elevated throughout the 6-months follow-up in the MRI- perilesional cortex (all p < 0.05). MRS markers revealed chronically sustained inflammation across the ipsilateral cortex but did not indicate the upcoming lesion expansion. Instead, the rostral expansion of the cortical lesion was systematically preceded by a hyperintense band in T2w images months earlier. Histologic analysis of the hyperintensity indicated scattered astrocytes, incomplete glial scar, and intracellularly packed and free iron. Yet, the band was negative in [18F]-FEPPA-PET. [18F]-FEPPA also showed no cortical TSPO expression within the MRS voxel in MRI- perilesional cortex or anywhere along glial scar when assessed at 2 months post-injury. However, [18F]-FEPPA showed a robust signal increase, indicating reactive microgliosis in the ipsilateral thalamus at 2 months post-TBI. We present evidence that MRS reveals chronic posttraumatic inflammation in MRI-negative perilesional cortex. The mismatch in MRS, MRI, and PET measures may allow non-invasive endophenotyping of beneficial and detrimental inflammatory processes to aid targeting and timing of anti-inflammatory therapeutics.
Collapse
Affiliation(s)
- Amna Yasmin
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Asla Pitkänen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Kimmo Jokivarsi
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Pekka Poutiainen
- Center of Diagnostic Imaging, Department of Cyclotron and Radiopharmacy, Kuopio University Hospital, Kuopio, Finland
| | - Olli Gröhn
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Riikka Immonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
13
|
Chao PH, Lazari M, Hanet S, Narayanam MK, Murphy JM, van Dam RM. Automated concentration of [ 18F]fluoride into microliter volumes. Appl Radiat Isot 2018; 141:138-148. [PMID: 30243135 PMCID: PMC6502507 DOI: 10.1016/j.apradiso.2018.06.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/07/2018] [Accepted: 06/20/2018] [Indexed: 11/23/2022]
Abstract
Concentration of [18F]fluoride has been mentioned in literature, however, reports have lacked details about system designs, operation, and performance. Here, we describe in detail a compact, fast, fully-automated concentration system based on a micro-sized strong anion exchange cartridge. The concentration of radionuclides enables scaled-up microfluidic synthesis. Our system can also be used to provide highly concentrated [18F]fluoride with minimal water content. We demonstrate how the concentrator can produce varying concentrations of [18F]fluoride for the macroscale synthesis of N-boc-5-[18F]fluoroindole without an azeotropic drying process, while enabling high starting radioactivity. By appropriate choice of solid-phase resin, flow conditions, and eluent solution, we believe this approach can be extended beyond [18F]fluoride to other radionuclides.
Collapse
Affiliation(s)
- Philip H Chao
- Department of Bioengineering, Henry Samueli School of Engineering, UCLA, Los Angeles, CA 90095, USA; Crump Institute for Molecular Imaging, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Mark Lazari
- Department of Bioengineering, Henry Samueli School of Engineering, UCLA, Los Angeles, CA 90095, USA; Crump Institute for Molecular Imaging, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Sebastian Hanet
- Crump Institute for Molecular Imaging, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Maruthi Kumar Narayanam
- Crump Institute for Molecular Imaging, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Jennifer M Murphy
- Crump Institute for Molecular Imaging, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - R Michael van Dam
- Department of Bioengineering, Henry Samueli School of Engineering, UCLA, Los Angeles, CA 90095, USA; Crump Institute for Molecular Imaging, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
14
|
Estimation of and correction for finite motion sampling errors in small animal PET rigid motion correction. Med Biol Eng Comput 2018; 57:505-518. [PMID: 30242596 PMCID: PMC6347657 DOI: 10.1007/s11517-018-1899-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 09/14/2018] [Indexed: 11/23/2022]
Abstract
Motion tracking with finite time sampling causing an associated unknown residual motion between two motion measurements is one of the factors contributing to resolution loss in small animal PET motion correction. The aim of this work is (i) to provide a means to estimate the effect of the finite motion sampling on the spatial resolution of the motion correction reconstructions and (ii) to correct for this residual motion thereby minimizing resolution loss. We calculate a tailored spatially variant deconvolution kernel from the measured motion data which is then used to deconvolve the motion corrected image using a 3D Richardson-Lucy algorithm. A simulation experiment of numerical phantoms as well as a microDerenzo phantom experiment wherein the phantom was manually moved at different speeds was performed to assess the performance of our proposed method. In the motion corrected images of the microDerenzo phantom there was an average rod FWHM differences between the slow and fast motion cases of 9.7%. This difference was reduced to 5.8% after applying the residual motion deconvolution. In awake animal experiments, the proposed method can serve to mitigate the finite sampling factor degrading the spatial resolution as well as the resolution differences between fast-moving and slow-moving animals. Motion correction of positron emission tomography (PET) scans of moving subjects can be performed by measuring the motion of the subject during the PET scan with an optical tracking camera. The motion tracking data obtained from the tracking camera is then used to correct the PET image reconstructions for motion. Due to finite time sampling of the motion data, the motion corrected reconstructions suffer from loss of spatial resolution. In the proposed method, a spatially variant deconvolution kernel is calculated from the motion tracking data, which is then used to correct the motion-corrected PET reconstructions for the blurring effect of the finite motion sampling through a Richardson-Lucy deconvolution. ![]()
Collapse
|
15
|
Barbone GE, Bravin A, Romanelli P, Mittone A, Bucci D, Gaaβ T, Le Duc G, Auweter S, Reiser MF, Kraiger MJ, Hrabě de Angelis M, Battaglia G, Coan P. Micro-imaging of Brain Cancer Radiation Therapy Using Phase-contrast Computed Tomography. Int J Radiat Oncol Biol Phys 2018; 101:965-984. [PMID: 29976510 DOI: 10.1016/j.ijrobp.2018.03.063] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 03/23/2018] [Accepted: 03/29/2018] [Indexed: 01/20/2023]
Abstract
PURPOSE Experimental neuroimaging provides a wide range of methods for the visualization of brain anatomic morphology down to subcellular detail. Still, each technique-specific detection mechanism presents compromises among the achievable field-of-view size, spatial resolution, and nervous tissue sensitivity, leading to partial sample coverage, unresolved morphologic structures, or sparse labeling of neuronal populations and often also to obligatory sample dissection or other sample invasive manipulations. X-ray phase-contrast imaging computed tomography (PCI-CT) is an experimental imaging method that simultaneously provides micrometric spatial resolution, high soft-tissue sensitivity, and ex vivo full organ rodent brain coverage without any need for sample dissection, staining or labeling, or contrast agent injection. In the present study, we explored the benefits and limitations of PCI-CT use for in vitro imaging of normal and cancerous brain neuromorphology after in vivo treatment with synchrotron-generated x-ray microbeam radiation therapy (MRT), a spatially fractionated experimental high-dose radiosurgery. The goals were visualization of the MRT effects on nervous tissue and a qualitative comparison of the results to the histologic and high-field magnetic resonance imaging findings. METHODS AND MATERIALS MRT was administered in vivo to the brain of both healthy and cancer-bearing rats. At 45 days after treatment, the brain was dissected out and imaged ex vivo using propagation-based PCI-CT. RESULTS PCI-CT visualizes the brain anatomy and microvasculature in 3 dimensions and distinguishes cancerous tissue morphology, necrosis, and intratumor accumulation of iron and calcium deposits. Moreover, PCI-CT detects the effects of MRT throughout the treatment target areas (eg, the formation of micrometer-thick radiation-induced tissue ablation). The observed neurostructures were confirmed by histologic and immunohistochemistry examination and related to the micro-magnetic resonance imaging data. CONCLUSIONS PCI-CT enabled a unique 3D neuroimaging approach for ex vivo studies on small animal models in that it concurrently delivers high-resolution insight of local brain tissue morphology in both normal and cancerous micro-milieu, localizes radiosurgical damage, and highlights the deep microvasculature. This method could assist experimental small animal neurology studies in the postmortem evaluation of neuropathology or treatment effects.
Collapse
Affiliation(s)
- Giacomo E Barbone
- Department of Physics, Ludwig Maximilians University, Garching, Germany
| | - Alberto Bravin
- European Synchrotron Radiation Facility, Grenoble, France
| | | | | | - Domenico Bucci
- Department of Molecular Pathology, Neuropharmacology Section, I.R.C.C.S. Neuromed, Pozzilli, Italy
| | - Thomas Gaaβ
- Department of Clinical Radiology, Ludwig Maximilians University, Munich, Germany
| | | | - Sigrid Auweter
- Department of Clinical Radiology, Ludwig Maximilians University, Munich, Germany
| | - Maximilian F Reiser
- Department of Clinical Radiology, Ludwig Maximilians University, Munich, Germany
| | - Markus J Kraiger
- Institute of Experimental Genetics and German Mouse Clinic, German Research Center for Environmental Health, Neuherberg, Germany
| | - Martin Hrabě de Angelis
- Institute of Experimental Genetics and German Mouse Clinic, German Research Center for Environmental Health, Neuherberg, Germany; Department of Experimental Genetics, School of Life Science Weihenstephan, Technical University of Munich, Freising, Germany; German Center for Diabetes Research, Neuherberg, Germany
| | - Giuseppe Battaglia
- Department of Molecular Pathology, Neuropharmacology Section, I.R.C.C.S. Neuromed, Pozzilli, Italy
| | - Paola Coan
- Department of Physics, Ludwig Maximilians University, Garching, Germany; Department of Clinical Radiology, Ludwig Maximilians University, Munich, Germany.
| |
Collapse
|
16
|
Omidvari N, Cabello J, Topping G, Schneider FR, Paul S, Schwaiger M, Ziegler SI. PET performance evaluation of MADPET4: a small animal PET insert for a 7 T MRI scanner. Phys Med Biol 2017; 62:8671-8692. [PMID: 28976912 DOI: 10.1088/1361-6560/aa910d] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
MADPET4 is the first small animal PET insert with two layers of individually read out crystals in combination with silicon photomultiplier technology. It has a novel detector arrangement, in which all crystals face the center of field of view transaxially. In this work, the PET performance of MADPET4 was evaluated and compared to other preclinical PET scanners using the NEMA NU 4 measurements, followed by imaging a mouse-size hot-rod resolution phantom and two in vivo simultaneous PET/MRI scans in a 7 T MRI scanner. The insert had a peak sensitivity of 0.49%, using an energy threshold of 350 keV. A uniform transaxial resolution was obtained up to 15 mm radial offset from the axial center, using filtered back-projection with single-slice rebinning. The measured average radial and tangential resolutions (FWHM) were 1.38 mm and 1.39 mm, respectively. The 1.2 mm rods were separable in the hot-rod phantom using an iterative image reconstruction algorithm. The scatter fraction was 7.3% and peak noise equivalent count rate was 15.5 kcps at 65.1 MBq of activity. The FDG uptake in a mouse heart and brain were visible in the two in vivo simultaneous PET/MRI scans without applying image corrections. In conclusion, the insert demonstrated a good overall performance and can be used for small animal multi-modal research applications.
Collapse
Affiliation(s)
- Negar Omidvari
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
17
|
Altomonte S, Telu S, Lu S, Pike VW. Pd(0)-Mediated 11C-Carbonylation of Aryl(mesityl)iodonium Salts as a Route to [ 11C]Arylcarboxylic Acids and Derivatives. J Org Chem 2017; 82:11925-11932. [PMID: 28972758 DOI: 10.1021/acs.joc.7b01704] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Pd(0)-mediated 11C-carbonylation of aryl(mesityl)iodonium salts followed by suitable quench provides a rapid room-temperature two-pot procedure for labeling arylcarboxylic acids and amide derivatives with the short-lived positron emitter carbon-11 (t1/2 = 20.4 min) in generally good to high yields (up to 71%). High product ring selectivity (≥13) was achieved when using mesityl as a spectator group in the diaryliodonium salt precursors. This process has potential for preparing new radiotracers for molecular imaging with positron emission tomography.
Collapse
Affiliation(s)
- Stefano Altomonte
- Molecular Imaging Branch, NIMH, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Sanjay Telu
- Molecular Imaging Branch, NIMH, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Shuiyu Lu
- Molecular Imaging Branch, NIMH, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Victor W Pike
- Molecular Imaging Branch, NIMH, National Institutes of Health , Bethesda, Maryland 20892, United States
| |
Collapse
|
18
|
Rafique W, Khanapur S, Spilhaug MM, Riss PJ. Reaching out for Sensitive Evaluation of the Mu Opioid Receptor in Vivo: Positron Emission Tomography Imaging of the Agonist [ 11C]AH7921. ACS Chem Neurosci 2017; 8:1847-1852. [PMID: 28590714 DOI: 10.1021/acschemneuro.7b00075] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Imaging of the mu opioid receptor (MOR) availability with positron emission tomography (PET) is a pertinent challenge in Neuroscience. Both, regulation of receptor expression and occupancy by endogeneous opioids play into cognitive and behavioral phenotypes of healthy function and disease. Receptor expression in the active and inactive states can be measured using high affinity radioagonist and radioantagonist PET tracers, respectively. Occupancy assessment requires radioligands showing competitive and reversible binding with moderate affinity to the MOR, which may lead to physical extinction of the receptor specific signal in vivo. We investigated a moderately potent, selective MOR agonist in rat to test if a radiotracer design paradigm tailored to competition with endogeneous opioids leads to viable imaging results. The benzamide 3,4-dichlorobenzenecarboxylic acid (dimethylamino)cyclohexyl)methyl amide (AH-7921, 1) was synthesized and characterized in rat brain using autoradiography and positron emission tomography. Compound 1 was found to activate with low nanomolar potency the MOR and to a lesser extent KOR as a full agonist. Concentration dependent binding studies with agonist and antagonist radioligands were conducted to assess competition behavior and obtain inhibition constants. Kinetic analysis of 3,4-dichlorobenzene[11C]carboxylic acid (dimethylamino)cyclohexyl)methyl amide binding in rat brain resulted in low but reproducible binding potential in the thalamus (0.8 ± 0.1). A radioactive metabolite was detected in brain (17%, after 15 min). Nonetheless, we conclude that quantitative imaging of MOR availability is possible when using a moderate affinity radiotracer.
Collapse
Affiliation(s)
- Waqas Rafique
- realomics
SFI, Kjemisk Institutt, Universitetet i Oslo, Sem Sælands vei 26, Kjemibygningen, 0371 Oslo, Norway
| | - Shivashankar Khanapur
- realomics
SFI, Kjemisk Institutt, Universitetet i Oslo, Sem Sælands vei 26, Kjemibygningen, 0371 Oslo, Norway
- Radboud Translational Medicine BV, Geert Grooteplein
21, Postbus 9101, 6500HB Nijmegen, Netherland
| | - Mona M. Spilhaug
- realomics
SFI, Kjemisk Institutt, Universitetet i Oslo, Sem Sælands vei 26, Kjemibygningen, 0371 Oslo, Norway
| | - Patrick J. Riss
- realomics
SFI, Kjemisk Institutt, Universitetet i Oslo, Sem Sælands vei 26, Kjemibygningen, 0371 Oslo, Norway
- Klinik
for Kirurgi og Nevrofag, Oslo Universitets Sykehus HF−Rikshospitalet, Postboks
4950 Nydalen, 0424 Oslo, Norway
- Norsk Medisinsk Syklotronsenter AS, Gaustad, Postboks 4950 Nydalen, 0424 Oslo, Norway
- Radboud Translational Medicine BV, Geert Grooteplein
21, Postbus 9101, 6500HB Nijmegen, Netherland
| |
Collapse
|
19
|
The brain during free movement - What can we learn from the animal model. Brain Res 2017; 1716:3-15. [PMID: 28893579 DOI: 10.1016/j.brainres.2017.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/11/2017] [Accepted: 09/04/2017] [Indexed: 11/21/2022]
Abstract
Animals, just like humans, can freely move. They do so for various important reasons, such as finding food and escaping predators. Observing these behaviors can inform us about the underlying cognitive processes. In addition, while humans can convey complicated information easily through speaking, animals need to move their bodies to communicate. This has prompted many creative solutions by animal neuroscientists to enable studying the brain during movement. In this review, we first summarize how animal researchers record from the brain while an animal is moving, by describing the most common neural recording techniques in animals and how they were adapted to record during movement. We further discuss the challenge of controlling or monitoring sensory input during free movement. However, not only is free movement a necessity to reflect the outcome of certain internal cognitive processes in animals, it is also a fascinating field of research since certain crucial behavioral patterns can only be observed and studied during free movement. Therefore, in a second part of the review, we focus on some key findings in animal research that specifically address the interaction between free movement and brain activity. First, focusing on walking as a fundamental form of free movement, we discuss how important such intentional movements are for understanding processes as diverse as spatial navigation, active sensing, and complex motor planning. Second, we propose the idea of regarding free movement as the expression of a behavioral state. This view can help to understand the general influence of movement on brain function. Together, the technological advancements towards recording from the brain during movement, and the scientific questions asked about the brain engaged in movement, make animal research highly valuable to research into the human "moving brain".
Collapse
|
20
|
McArthur RA. Aligning physiology with psychology: Translational neuroscience in neuropsychiatric drug discovery. Neurosci Biobehav Rev 2017; 76:4-21. [DOI: 10.1016/j.neubiorev.2017.02.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 02/03/2017] [Indexed: 12/12/2022]
|
21
|
Herde A, Ioanas HI, Boss S, Seifritz E, Ametamey S, Saab B. Using Tandem Behaviour-PET to Examine Dopaminergic Signalling Underlying Exploration. ACTA ACUST UNITED AC 2017. [DOI: 10.19185/matters.201702000008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
22
|
Brendel M, Kleinberger G, Probst F, Jaworska A, Overhoff F, Blume T, Albert NL, Carlsen J, Lindner S, Gildehaus FJ, Ozmen L, Suárez-Calvet M, Bartenstein P, Baumann K, Ewers M, Herms J, Haass C, Rominger A. Increase of TREM2 during Aging of an Alzheimer's Disease Mouse Model Is Paralleled by Microglial Activation and Amyloidosis. Front Aging Neurosci 2017; 9:8. [PMID: 28197095 PMCID: PMC5282474 DOI: 10.3389/fnagi.2017.00008] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/12/2017] [Indexed: 12/23/2022] Open
Abstract
Heterozygous missense mutations in the triggering receptor expressed on myeloid cells 2 (TREM2) have been reported to significantly increase the risk of developing Alzheimer’s disease (AD). Since TREM2 is specifically expressed by microglia in the brain, we hypothesized that soluble TREM2 (sTREM2) levels may increase together with in vivo biomarkers of microglial activity and amyloidosis in an AD mouse model as assessed by small animal positron-emission-tomography (μPET). In this cross-sectional study, we examined a strong amyloid mouse model (PS2APP) of four age groups by μPET with [18F]-GE180 (glial activation) and [18F]-florbetaben (amyloidosis), followed by measurement of sTREM2 levels and amyloid levels in the brain. Pathology affected brain regions were compared between tracers (dice similarity coefficients) and pseudo-longitudinally. μPET results of both tracers were correlated with terminal TREM2 levels. The brain sTREM2 levels strongly increased with age of PS2APP mice (5 vs. 16 months: +211%, p < 0.001), and correlated highly with μPET signals of microglial activity (R = 0.89, p < 0.001) and amyloidosis (R = 0.92, p < 0.001). Dual μPET enabled regional mapping of glial activation and amyloidosis in the mouse brain, which progressed concertedly leading to a high overlap in aged PS2APP mice (dice similarity 67%). Together, these results substantiate the use of in vivo μPET measurements in conjunction with post mortem sTREM2 in future anti-inflammatory treatment trials. Taking human data into account sTREM2 may increase during active amyloid deposition.
Collapse
Affiliation(s)
- Matthias Brendel
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München Munich, Germany
| | - Gernot Kleinberger
- Department of Biochemistry, Biomedical Center (BMC), Ludwig-Maximilians-Universität MünchenMunich, Germany; Munich Cluster for Systems Neurology (SyNergy), Ludwig-Maximilians-Universität MünchenMunich, Germany
| | - Federico Probst
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München Munich, Germany
| | - Anna Jaworska
- DZNE-German Center for Neurodegenerative DiseasesMunich, Germany; Laboratory of Neurodegeneration, International Institute of Molecular and Cell BiologyWarsaw, Poland
| | - Felix Overhoff
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München Munich, Germany
| | - Tanja Blume
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München Munich, Germany
| | - Janette Carlsen
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München Munich, Germany
| | - Franz Josef Gildehaus
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München Munich, Germany
| | - Laurence Ozmen
- Roche Pharma Research and Early Development, Neuroscience Discovery, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd. Basel, Switzerland
| | - Marc Suárez-Calvet
- Department of Biochemistry, Biomedical Center (BMC), Ludwig-Maximilians-Universität MünchenMunich, Germany; DZNE-German Center for Neurodegenerative DiseasesMunich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität MünchenMunich, Germany; Munich Cluster for Systems Neurology (SyNergy), Ludwig-Maximilians-Universität MünchenMunich, Germany
| | - Karlheinz Baumann
- Roche Pharma Research and Early Development, Neuroscience Discovery, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd. Basel, Switzerland
| | - Michael Ewers
- DZNE-German Center for Neurodegenerative Diseases Munich, Germany
| | - Jochen Herms
- Munich Cluster for Systems Neurology (SyNergy), Ludwig-Maximilians-Universität MünchenMunich, Germany; DZNE-German Center for Neurodegenerative DiseasesMunich, Germany
| | - Christian Haass
- Department of Biochemistry, Biomedical Center (BMC), Ludwig-Maximilians-Universität MünchenMunich, Germany; Munich Cluster for Systems Neurology (SyNergy), Ludwig-Maximilians-Universität MünchenMunich, Germany; DZNE-German Center for Neurodegenerative DiseasesMunich, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität MünchenMunich, Germany; Munich Cluster for Systems Neurology (SyNergy), Ludwig-Maximilians-Universität MünchenMunich, Germany
| |
Collapse
|
23
|
Mandeville ET, Ayata C, Zheng Y, Mandeville JB. Translational MR Neuroimaging of Stroke and Recovery. Transl Stroke Res 2016; 8:22-32. [PMID: 27578048 DOI: 10.1007/s12975-016-0497-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/16/2016] [Accepted: 08/18/2016] [Indexed: 12/26/2022]
Abstract
Multiparametric magnetic resonance imaging (MRI) has become a critical clinical tool for diagnosing focal ischemic stroke severity, staging treatment, and predicting outcome. Imaging during the acute phase focuses on tissue viability in the stroke vicinity, while imaging during recovery requires the evaluation of distributed structural and functional connectivity. Preclinical MRI of experimental stroke models provides validation of non-invasive biomarkers in terms of cellular and molecular mechanisms, while also providing a translational platform for evaluation of prospective therapies. This brief review of translational stroke imaging discusses the acute to chronic imaging transition, the principles underlying common MRI methods employed in stroke research, and the experimental results obtained by clinical and preclinical imaging to determine tissue viability, vascular remodeling, structural connectivity of major white matter tracts, and functional connectivity using task-based and resting-state fMRI during the stroke recovery process.
Collapse
Affiliation(s)
- Emiri T Mandeville
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Charlestown, MA, USA. .,Department of Radiology, Massachusetts General Hospital, Bldg 149 13th Street, Charlestown, MA, 02129, USA.
| | - Cenk Ayata
- Neurovascular Research Laboratory, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Radiology, Massachusetts General Hospital, Bldg 149 13th Street, Charlestown, MA, 02129, USA
| | - Yi Zheng
- Neurovascular Research Laboratory, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Radiology, Massachusetts General Hospital, Bldg 149 13th Street, Charlestown, MA, 02129, USA
| | - Joseph B Mandeville
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Radiology, Massachusetts General Hospital, Bldg 149 13th Street, Charlestown, MA, 02129, USA
| |
Collapse
|
24
|
Bascuñana P, Javela J, Delgado M, Fernández de la Rosa R, Shiha AA, García-García L, Pozo MÁ. [18F]FDG PET Neuroimaging Predicts Pentylenetetrazole (PTZ) Kindling Outcome in Rats. Mol Imaging Biol 2016; 18:733-40. [DOI: 10.1007/s11307-016-0950-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
25
|
Richard MA, Fouquet JP, Lebel R, Lepage M. MRI-Guided Derivation of the Input Function for PET Kinetic Modeling. PET Clin 2016; 11:193-202. [DOI: 10.1016/j.cpet.2015.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
26
|
Gunn RN, Slifstein M, Searle GE, Price JC. Quantitative imaging of protein targets in the human brain with PET. Phys Med Biol 2015; 60:R363-411. [DOI: 10.1088/0031-9155/60/22/r363] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
27
|
Abstract
The mass transport or flux of neurochemicals in the brain and how this flux affects chemical measurements and their interpretation is reviewed. For all endogenous neurochemicals found in the brain, the flux of each of these neurochemicals exists between sources that produce them and the sites that consume them all within μm distances. Principles of convective-diffusion are reviewed with a significant emphasis on the tortuous paths and discrete point sources and sinks. The fundamentals of the primary methods of detection, microelectrodes and microdialysis sampling of brain neurochemicals are included in the review. Special attention is paid to the change in the natural flux of the neurochemicals caused by implantation and consumption at microelectrodes and uptake by microdialysis. The detection of oxygen, nitric oxide, glucose, lactate, and glutamate, and catecholamines by both methods are examined and where possible the two techniques (electrochemical vs. microdialysis) are compared. Non-invasive imaging methods: magnetic resonance, isotopic fluorine MRI, electron paramagnetic resonance, and positron emission tomography are also used for different measurements of the above-mentioned solutes and these are briefly reviewed. Although more sophisticated, the imaging techniques are unable to track neurochemical flux on short time scales, and lack spatial resolution. Where possible, determinations of flux using imaging are compared to the more classical techniques of microdialysis and microelectrodes.
Collapse
Affiliation(s)
- David W Paul
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA.
| | | |
Collapse
|
28
|
Durham TB, Blanco MJ. Target Engagement in Lead Generation. Bioorg Med Chem Lett 2015; 25:998-1008. [DOI: 10.1016/j.bmcl.2014.12.076] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 12/15/2014] [Accepted: 12/23/2014] [Indexed: 12/15/2022]
|
29
|
Brendel M, Jaworska A, Grießinger E, Rötzer C, Burgold S, Gildehaus FJ, Carlsen J, Cumming P, Baumann K, Haass C, Steiner H, Bartenstein P, Herms J, Rominger A. Cross-sectional comparison of small animal [18F]-florbetaben amyloid-PET between transgenic AD mouse models. PLoS One 2015; 10:e0116678. [PMID: 25706990 PMCID: PMC4338066 DOI: 10.1371/journal.pone.0116678] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 12/11/2014] [Indexed: 12/13/2022] Open
Abstract
We aimed to compare [18F]-florbetaben PET imaging in four transgenic mouse strains modelling Alzheimer’s disease (AD), with the main focus on APPswe/PS2 mice and C57Bl/6 mice serving as controls (WT). A consistent PET protocol (N = 82 PET scans) was used, with cortical standardized uptake value ratio (SUVR) relative to cerebellum as the endpoint. We correlated methoxy-X04 staining of β-amyloid with PET results, and undertook ex vivo autoradiography for further validation of a partial volume effect correction (PVEC) of PET data. The SUVR in APPswe/PS2 increased from 0.95±0.04 at five months (N = 5) and 1.04±0.03 (p<0.05) at eight months (N = 7) to 1.07±0.04 (p<0.005) at ten months (N = 6), 1.28±0.06 (p<0.001) at 16 months (N = 6) and 1.39±0.09 (p<0.001) at 19 months (N = 6). SUVR was 0.95±0.03 in WT mice of all ages (N = 22). In APPswe/PS1G384A mice, the SUVR was 0.93/0.98 at five months (N = 2) and 1.11 at 16 months (N = 1). In APPswe/PS1dE9 mice, the SUVR declined from 0.96/0.96 at 12 months (N = 2) to 0.91/0.92 at 24 months (N = 2), due to β-amyloid plaques in cerebellum. PVEC reduced the discrepancy between SUVR-PET and autoradiography from −22% to +2% and increased the differences between young and aged transgenic animals. SUVR and plaque load correlated highly between strains for uncorrected (R = 0.94, p<0.001) and PVE-corrected (R = 0.95, p<0.001) data. We find that APPswe/PS2 mice may be optimal for longitudinal amyloid-PET monitoring in planned interventions studies.
Collapse
Affiliation(s)
- Matthias Brendel
- Dept. of Nuclear Medicine, University of Munich, Munich, Germany
| | - Anna Jaworska
- Dept. of Translational Research I, German Center for Neurodegenerative Diseases (DZNE)—site Munich, University of Munich, Munich, Germany
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Eric Grießinger
- Dept. of Translational Research I, German Center for Neurodegenerative Diseases (DZNE)—site Munich, University of Munich, Munich, Germany
| | - Christina Rötzer
- Dept. of Nuclear Medicine, University of Munich, Munich, Germany
| | - Steffen Burgold
- Dept. of Translational Research I, German Center for Neurodegenerative Diseases (DZNE)—site Munich, University of Munich, Munich, Germany
| | | | - Janette Carlsen
- Dept. of Nuclear Medicine, University of Munich, Munich, Germany
| | - Paul Cumming
- Department of Nuclear Medicine, University of Erlangen, Erlangen, Germany
- Department of Neuroscience and Pharmacology, Copenhagen University, Copenhagen, Denmark
| | | | - Christian Haass
- Adolf-Butenandt-Institute, Biochemistry, University of Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Harald Steiner
- Adolf-Butenandt-Institute, Biochemistry, University of Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
| | - Peter Bartenstein
- Dept. of Nuclear Medicine, University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jochen Herms
- Dept. of Translational Research I, German Center for Neurodegenerative Diseases (DZNE)—site Munich, University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Axel Rominger
- Dept. of Nuclear Medicine, University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- * E-mail:
| |
Collapse
|
30
|
Shiha AA, de Cristóbal J, Delgado M, Fernández de la Rosa R, Bascuñana P, Pozo MA, García-García L. Subacute administration of fluoxetine prevents short-term brain hypometabolism and reduces brain damage markers induced by the lithium-pilocarpine model of epilepsy in rats. Brain Res Bull 2015; 111:36-47. [DOI: 10.1016/j.brainresbull.2014.12.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/12/2014] [Accepted: 12/16/2014] [Indexed: 12/30/2022]
|
31
|
Stewart AM, Ullmann JF, Norton WH, Brennan CH, Parker MO, Gerlai R, Kalueff AV. Molecular psychiatry of zebrafish. Mol Psychiatry 2015; 20:2-17. [PMID: 25349164 PMCID: PMC4318706 DOI: 10.1038/mp.2014.128] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 08/27/2014] [Accepted: 08/28/2014] [Indexed: 12/31/2022]
Abstract
Due to their well-characterized neural development and high genetic homology to mammals, zebrafish (Danio rerio) have emerged as a powerful model organism in the field of biological psychiatry. Here, we discuss the molecular psychiatry of zebrafish, and its implications for translational neuroscience research and modeling central nervous system (CNS) disorders. In particular, we outline recent genetic and technological developments allowing for in vivo examinations, high-throughput screening and whole-brain analyses in larval and adult zebrafish. We also summarize the application of these molecular techniques to the understanding of neuropsychiatric disease, outlining the potential of zebrafish for modeling complex brain disorders, including attention-deficit/hyperactivity disorder (ADHD), aggression, post-traumatic stress and substance abuse. Critically evaluating the advantages and limitations of larval and adult fish tests, we suggest that zebrafish models become a rapidly emerging new field in modern molecular psychiatry research.
Collapse
Affiliation(s)
- Adam Michael Stewart
- ZENEREI Institute, 309 Palmer Court, Slidell, LA 70458, USA
- International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA 70458, USA
| | - Jeremy F.P. Ullmann
- International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA 70458, USA
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland 4072, Australia
| | - William H.J. Norton
- International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA 70458, USA
- Department of Biology, College of Medicine, Biological Sciences and Psychiatry, University of Leicester, University Road, Leicester LE1 7RH, UK
| | - Caroline H. Brennan
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London, E1-4NS, UK
| | - Matthew O. Parker
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London, E1-4NS, UK
| | - Robert Gerlai
- Department of Psychology, University of Toronto at Mississauga, 3359 Mississauga Rd N Mississauga, Ontario L5L1C6, Canada
| | - Allan V. Kalueff
- ZENEREI Institute, 309 Palmer Court, Slidell, LA 70458, USA
- International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA 70458, USA
- Research Institute for Marine Drugs and Nutrition, Guangdong Ocean University, Zhanjiang, Guangdong 524025, China
| |
Collapse
|
32
|
Piel M, Vernaleken I, Rösch F. Positron Emission Tomography in CNS Drug Discovery and Drug Monitoring. J Med Chem 2014; 57:9232-58. [DOI: 10.1021/jm5001858] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Markus Piel
- Institute
of Nuclear Chemistry, Johannes Gutenberg-University, Fritz-Strassmann-Weg 2, D-55128 Mainz, Germany
| | - Ingo Vernaleken
- Department
of Psychiatry, Psychotherapy, and Psychosomatics, RWTH Aachen University, Pauwelsstraße 30, D-52074 Aachen, Germany
| | - Frank Rösch
- Institute
of Nuclear Chemistry, Johannes Gutenberg-University, Fritz-Strassmann-Weg 2, D-55128 Mainz, Germany
| |
Collapse
|
33
|
Development of (18)F-labeled radiotracers for neuroreceptor imaging with positron emission tomography. Neurosci Bull 2014; 30:777-811. [PMID: 25172118 DOI: 10.1007/s12264-014-1460-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Accepted: 06/02/2014] [Indexed: 12/14/2022] Open
Abstract
Positron emission tomography (PET) is an in vivo molecular imaging tool which is widely used in nuclear medicine for early diagnosis and treatment follow-up of many brain diseases. PET uses biomolecules as probes which are labeled with radionuclides of short half-lives, synthesized prior to the imaging studies. These probes are called radiotracers. Fluorine-18 is a radionuclide routinely used in the radiolabeling of neuroreceptor ligands for PET because of its favorable half-life of 109.8 min. The delivery of such radiotracers into the brain provides images of transport, metabolic, and neurotransmission processes on the molecular level. After a short introduction into the principles of PET, this review mainly focuses on the strategy of radiotracer development bridging from basic science to biomedical application. Successful radiotracer design as described here provides molecular probes which not only are useful for imaging of human brain diseases, but also allow molecular neuroreceptor imaging studies in various small-animal models of disease, including genetically-engineered animals. Furthermore, they provide a powerful tool for in vivo pharmacology during the process of pre-clinical drug development to identify new drug targets, to investigate pathophysiology, to discover potential drug candidates, and to evaluate the pharmacokinetics and pharmacodynamics of drugs in vivo.
Collapse
|
34
|
Riss PJ, Stockhofe K, Roesch F. Tropane-derived (11) C-labelled and (18) F-labelled DAT ligands. J Labelled Comp Radiopharm 2014; 56:149-58. [PMID: 24285320 DOI: 10.1002/jlcr.3018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 11/30/2012] [Accepted: 12/04/2012] [Indexed: 01/02/2023]
Abstract
Radiolabelling of cocaine-derived 3-phenyltropanes for dopamine transporter positron emission tomography with (18) F and (11) C is reviewed.
Collapse
Affiliation(s)
- P J Riss
- Wolfson Brain Imaging Centre, University of Cambridge, Box 65 Addenbrooke's Hospital, CB2 0QQ, Cambridge, UK
| | | | | |
Collapse
|
35
|
Radiosynthesis of [18)F]trifluoroalkyl groups: scope and limitations. BIOMED RESEARCH INTERNATIONAL 2014; 2014:380124. [PMID: 25110676 PMCID: PMC4119740 DOI: 10.1155/2014/380124] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 04/21/2014] [Accepted: 05/06/2014] [Indexed: 12/04/2022]
Abstract
The present paper is concerned with radiochemical methodology to furnish the trifluoromethyl motif labelled with 18F. Literature spanning the last four decades is comprehensively reviewed and radiochemical yields and specific activities are discussed.
Collapse
|
36
|
Dedeurwaerdere S, Shultz SR, Federico P, Engel J. Workshop on Neurobiology of Epilepsy appraisal: new systemic imaging technologies to study the brain in experimental models of epilepsy. Epilepsia 2014; 55:819-28. [PMID: 24836499 DOI: 10.1111/epi.12642] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2014] [Indexed: 12/14/2022]
Abstract
Modern functional neuroimaging provides opportunities to visualize activity of the entire brain, making it an indispensable diagnostic tool for epilepsy. Various forms of noninvasive functional neuroimaging are now also being performed as research tools in animal models of epilepsy and provide opportunities for parallel animal/human investigations into fundamental mechanisms of epilepsy and identification of epilepsy biomarkers. Recent animal studies of epilepsy using positron emission tomography, tractography, and functional magnetic resonance imaging were reviewed. Epilepsy is an abnormal emergent property of disturbances in neuronal networks which, even for epilepsies characterized by focal seizures, involve widely distributed systems, often in both hemispheres. Functional neuroimaging in animal models now provides opportunities to examine neuronal disturbances in the whole brain that underlie generalized and focal seizure generation as well as various types of epileptogenesis. Tremendous advances in understanding the contribution of specific properties of widely distributed neuronal networks to both normal and abnormal human behavior have been provided by current functional neuroimaging methodologies. Successful application of functional neuroimaging of the whole brain in the animal laboratory now permits investigations during epileptogenesis and correlation with deep brain electroencephalography (EEG) activity. With the continuing development of these techniques and analytical methods, the potential for future translational research on epilepsy is enormous. A PowerPoint slide summarizing this article is available for download in the Supporting Information section here.
Collapse
|
37
|
Abstract
Epilepsy is one of the most common chronic neurological conditions worldwide. Anti-epileptic drugs (AEDs) can suppress seizures, but do not affect the underlying epileptic state, and many epilepsy patients are unable to attain seizure control with AEDs. To cure or prevent epilepsy, disease-modifying interventions that inhibit or reverse the disease process of epileptogenesis must be developed. A major limitation in the development and implementation of such an intervention is the current poor understanding, and the lack of reliable biomarkers, of the epileptogenic process. Neuroimaging represents a non-invasive medical and research tool with the ability to identify early pathophysiological changes involved in epileptogenesis, monitor disease progression, and assess the effectiveness of possible therapies. Here we will provide an overview of studies conducted in animal models and in patients with epilepsy that have utilized various neuroimaging modalities to investigate epileptogenesis.
Collapse
Affiliation(s)
- Sandy R Shultz
- Department of Medicine, The Melbourne Brain Centre, The Royal Melbourne Hospital, The University of Melbourne, Building 144, Royal Parade, Parkville, VIC, 3010, Australia,
| | | | | | | |
Collapse
|
38
|
Present status and future challenges of electroencephalography- and magnetic resonance imaging-based monitoring in preclinical models of focal cerebral ischemia. Brain Res Bull 2014; 102:22-36. [PMID: 24462642 DOI: 10.1016/j.brainresbull.2014.01.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 01/07/2014] [Accepted: 01/14/2014] [Indexed: 12/16/2022]
Abstract
Animal models are useful tools for better understanding the mechanisms underlying neurological deterioration after an ischemic insult as well as subsequent evolution of changes and recovery of functions. In response to the updated requirements for preclinical investigations of stroke to include relevant functional measurement techniques and biomarker endpoints, we here review the state of knowledge on application of some translational electrophysiological and neuroimaging methods, and in particular, electroencephalography monitoring and magnetic resonance imaging in rodent models of ischemic stroke. This may lead to improvement of diagnostic methods and identification of new therapeutic targets, which would considerably advance the translational value of preclinical stroke research.
Collapse
|
39
|
Rühl T, Rafique W, Lien VT, Riss PJ. Cu(i)-mediated 18F-trifluoromethylation of arenes: Rapid synthesis of 18F-labeled trifluoromethyl arenes. Chem Commun (Camb) 2014; 50:6056-9. [DOI: 10.1039/c4cc01641f] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An efficient, CuI-mediated method for the radiosynthesis of [18F]trifluoromethyl arenes, abundant motifs in small molecule drug candidates and potential radiotracers for positron emission tomography starting from [18F]fluoride ions is described.
Collapse
Affiliation(s)
- T. Rühl
- Kjemisk Institutt
- Universitetet I Oslo
- Oslo, Norway
| | - W. Rafique
- Kjemisk Institutt
- Universitetet I Oslo
- Oslo, Norway
| | - V. T. Lien
- Norsk Medisinsk Syklotronsenter AS
- Oslo, Norway
| | - P. J. Riss
- Kjemisk Institutt
- Universitetet I Oslo
- Oslo, Norway
- Norsk Medisinsk Syklotronsenter AS
- Oslo, Norway
| |
Collapse
|
40
|
Abstract
Drug addiction is a progressive, relapsing disease comprised of interlocking stages of disordered motivation. Numerous animal models describing various stages of the addiction process have been developed over the past few decades, providing considerable advantages for the modeling of drug addiction compared with other complex psychiatric disease states. Escalation of drug self-administration has emerged as a widely accepted operant conditioning model of excessive drug intake. We further argue here that drug-escalated animals represent a comprehensive model of addiction according to the manifestations of behavioral neuroadaptations resulting directly or indirectly from excessive drug consumption. In particular, drug-escalated animals exhibit a host of symptoms in line with multiple Diagnostic and Statistical Manual of Mental Disorders criteria for substance dependence, which can be summarized as an emergence of uncontrollable drug-taking and drug-seeking behaviors as a consequence of within-circuit and between-circuit neuroadaptations. Such a transition from impulsive drug sampling to compulsive intake represents a highly valid conceptualization of the addiction timeline in humans, and further investigation of persistent or near-permanent (e.g. epigenetic) neuroadaptations generated by operant drug intake escalation models will continue to provide mechanisms and therapeutic interventions for reversing the aberrant neuroplasticity underlying addiction.
Collapse
|
41
|
Brendel M, Delker A, Rötzer C, Böning G, Carlsen J, Cyran C, Mille E, Gildehaus FJ, Cumming P, Baumann K, Steiner H, Haass C, Herms J, Bartenstein P, Rominger A. Impact of partial volume effect correction on cerebral β-amyloid imaging in APP-Swe mice using [(18)F]-florbetaben PET. Neuroimage 2013; 84:843-53. [PMID: 24055703 DOI: 10.1016/j.neuroimage.2013.09.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 09/03/2013] [Accepted: 09/10/2013] [Indexed: 11/17/2022] Open
Abstract
We previously investigated the progression of β-amyloid deposition in brain of mice over-expressing amyloid-precursor protein (APP-Swe), a model of Alzheimer's disease (AD), in a longitudinal PET study with the novel β-amyloid tracer [(18)F]-florbetaben. There were certain discrepancies between PET and autoradiographic findings, which seemed to arise from partial volume effects (PVE). Since this phenomenon can lead to bias, most especially in the quantitation of brain microPET studies of mice, we aimed in the present study to investigate the magnitude of PVE on [(18)F]-florbetaben quantitation in murine brain, and to establish and validate a useful correction method (PVEC). Phantom studies with solutions of known radioactivity concentration were performed to measure the full-width-at-half-maximum (FWHM) resolution of the Siemens Inveon DPET and to validate a volume-of-interest (VOI)-based PVEC algorithm. Several VOI-brain-masks were applied to perform in vivo PVEC on [(18)F]-florbetaben data from C57BL/6(N=6) mice, while uncorrected and PVE-corrected data were cross-validated with gamma counting and autoradiography. Next, PVEC was performed on longitudinal PET data set consisting of 43 PET scans in APP-Swe (13-20months) and age-matched wild-type (WT) mice using the previously defined masks. VOI-based cortex-to-cerebellum ratios (SUVR) were compared for uncorrected and PVE-corrected results. Brains from a subset of transgenic mice were ultimately examined by autoradiography ex vivo and histochemistry in vitro as gold standard assessments, and compared to VOI-based PET results. The phantom study indicated a FWHM of 1.72mm. Applying a VOI-brain-mask including extracerebral regions gave robust PVEC, with increased precision of the SUVR results. Cortical SUVR increased with age in APP-Swe mice compared to baseline measurements (16months: +5.5%, p<0.005; 20months: +15.5%, p<0.05) with uncorrected data, and to a substantially greater extent with PVEC (16months: +12.2% p<0.005; 20months: +36.4% p<0.05). WT animals showed no binding changes, irrespective of PVEC. Relative to autoradiographic results, the error [%] for uncorrected cortical SUVR was 18.9% for native PET data, and declined to 4.8% upon PVEC, in high correlation with histochemistry results. We calculate that PVEC increases by 10% statistical power for detecting altered [(18)F]-florbetaben uptake in aging APP-Swe mice in planned studies of disease modifying treatments on amyloidogenesis.
Collapse
Affiliation(s)
- Matthias Brendel
- Department of Nuclear Medicine, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kumazawa-Manita N, Katayama M, Hashikawa T, Iriki A. Three-dimensional reconstruction of brain structures of the rodent Octodon degus: a brain atlas constructed by combining histological and magnetic resonance images. Exp Brain Res 2013; 231:65-74. [PMID: 23995563 PMCID: PMC3824219 DOI: 10.1007/s00221-013-3667-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 07/30/2013] [Indexed: 12/18/2022]
Abstract
Degus (Octodon degus) are rodents that are becoming more widely used in the neuroscience field. Degus display several more complex behaviors than rats and mice, including complicated social behaviors, vocal communications, and tool usage with superb manual dexterity. However, relatively little information is known about the anatomy of degu brains. Therefore, for these complex behaviors to be correlated with specific brain regions, a contemporary atlas of the degu brain is required. This manuscript describes the construction of a three-dimensional (3D) volume rendered model of the degu brain that combines histological and magnetic resonance images. This atlas provides several advantages, including the ability to visualize the surface of the brain from any angle. The atlas also permits virtual cutting of brain sections in any plane and provides stereotaxic coordinates for all sections, to be beneficial for both experimental surgeries and radiological studies. The reconstructed 3D atlas is freely available online at: http://brainatlas.brain.riken.jp/degu/modules/xoonips/listitem.php?index_id=24 .
Collapse
Affiliation(s)
- Noriko Kumazawa-Manita
- Laboratory for Symbolic Cognitive Development, RIKEN, Brain Science Institute, Wako, Saitama, 351-0198, Japan,
| | | | | | | |
Collapse
|
43
|
Nagy K, Tóth M, Major P, Patay G, Egri G, Häggkvist J, Varrone A, Farde L, Halldin C, Gulyás B. Performance evaluation of the small-animal nanoScan PET/MRI system. J Nucl Med 2013; 54:1825-32. [PMID: 23990683 DOI: 10.2967/jnumed.112.119065] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED nanoScan is a high-resolution integrated system for consecutive PET and MR imaging of small laboratory animals. We evaluated the performance of the system, using the NEMA NU 4-2008 protocol for the PET component and the NEMA MS 1-2007, MS 2-2008, and MS 3-2007 standards for the MR imaging component. METHODS The imaging system uses magnetically shielded position-sensitive photomultiplier tubes and a compact 1-T permanent-magnet MR imaging platform. Spatial resolution, sensitivity, counting rate capabilities, and image quality parameters were evaluated in accordance with the aforementioned NEMA standards. Further in vivo evaluation experiments complement the physical validation results. RESULTS The spatial resolution of the PET system enabled the 0.8-mm rods of a Derenzo phantom to be resolved. With point source and 2-dimensional filtered backprojection reconstruction, the resolution varied from 1.50 to 2.01 mm in full width at half maximum in the radial direction and from 1.32 to 1.65 mm in the tangential direction within the radius of 25 mm. Peak absolute sensitivity was 8.41%. Scatter fraction was 17.3% and 34.0%, and maximum noise-equivalent counting rate was 406 and 119 kcps in the mouselike and ratlike phantom, respectively. The image quality test found a nonuniformity of 3.52% and a spillover ratio of 6.2% and 5.8% in water and air, respectively. In testing of the MR imaging component, artifact-free images with high signal-to-noise ratio were recorded. Geometric distortion was below 5%, and image uniformity was at least 94.5% and 96.6% for the 60- and 35-mm radiofrequency coils, respectively. CONCLUSION The nanoScan integrated small-animal PET/MR imaging system has excellent spatial resolution and sensitivity. The performance characteristics of the PET and the MR imaging components are not compromised as a result of their integration onto a single platform. Because of its combination of features and performance parameters, the system provides crucial advantages for preclinical imaging studies over existing PET/CT systems, especially in neurologic and oncologic research.
Collapse
Affiliation(s)
- Kálmán Nagy
- Psychiatry Section, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; and
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Simplified programming and control of automated radiosynthesizers through unit operations. EJNMMI Res 2013; 3:53. [PMID: 23855995 PMCID: PMC3717018 DOI: 10.1186/2191-219x-3-53] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 07/02/2013] [Indexed: 11/29/2022] Open
Abstract
Background Many automated radiosynthesizers for producing positron emission tomography (PET) probes provide a means for the operator to create custom synthesis programs. The programming interfaces are typically designed with the engineer rather than the radiochemist in mind, requiring lengthy programs to be created from sequences of low-level, non-intuitive hardware operations. In some cases, the user is even responsible for adding steps to update the graphical representation of the system. In light of these unnecessarily complex approaches, we have created software to perform radiochemistry on the ELIXYS radiosynthesizer with the goal of being intuitive and easy to use. Methods Radiochemists were consulted, and a wide range of radiosyntheses were analyzed to determine a comprehensive set of basic chemistry unit operations. Based around these operations, we created a software control system with a client–server architecture. In an attempt to maximize flexibility, the client software was designed to run on a variety of portable multi-touch devices. The software was used to create programs for the synthesis of several 18F-labeled probes on the ELIXYS radiosynthesizer, with [18F]FDG detailed here. To gauge the user-friendliness of the software, program lengths were compared to those from other systems. A small sample group with no prior radiosynthesizer experience was tasked with creating and running a simple protocol. Results The software was successfully used to synthesize several 18F-labeled PET probes, including [18F]FDG, with synthesis times and yields comparable to literature reports. The resulting programs were significantly shorter and easier to debug than programs from other systems. The sample group of naive users created and ran a simple protocol within a couple of hours, revealing a very short learning curve. The client–server architecture provided reliability, enabling continuity of the synthesis run even if the computer running the client software failed. The architecture enabled a single user to control the hardware while others observed the run in progress or created programs for other probes. Conclusions We developed a novel unit operation-based software interface to control automated radiosynthesizers that reduced the program length and complexity and also exhibited a short learning curve. The client–server architecture provided robustness and flexibility.
Collapse
|
45
|
Kim M, Woo SK, Yu JW, Lee YJ, Kim KM, Kang JH, Eom K, Nahm SS. Effect of Harderian adenectomy on the statistical analyses of mouse brain imaging using positron emission tomography. J Vet Sci 2013; 15:157-61. [PMID: 23820224 PMCID: PMC3973759 DOI: 10.4142/jvs.2014.15.1.157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 06/28/2013] [Indexed: 11/30/2022] Open
Abstract
Positron emission tomography (PET) using 2-deoxy-2-[18F] fluoro-D-glucose (FDG) as a radioactive tracer is a useful technique for in vivo brain imaging. However, the anatomical and physiological features of the Harderian gland limit the use of FDG-PET imaging in the mouse brain. The gland shows strong FDG uptake, which in turn results in distorted PET images of the frontal brain region. The purpose of this study was to determine if a simple surgical procedure to remove the Harderian gland prior to PET imaging of mouse brains could reduce or eliminate FDG uptake. Measurement of FDG uptake in unilaterally adenectomized mice showed that the radioactive signal emitted from the intact Harderian gland distorts frontal brain region images. Spatial parametric measurement analysis demonstrated that the presence of the Harderian gland could prevent accurate assessment of brain PET imaging. Bilateral Harderian adenectomy efficiently eliminated unwanted radioactive signal spillover into the frontal brain region beginning on postoperative Day 10. Harderian adenectomy did not cause any post-operative complications during the experimental period. These findings demonstrate the benefits of performing a Harderian adenectomy prior to PET imaging of mouse brains.
Collapse
Affiliation(s)
- Minsoo Kim
- Department of Veterinary Medicine, College of Veterinary Medicine, Konkuk University, Seoul 143-701, Korea
| | | | | | | | | | | | | | | |
Collapse
|
46
|
In vitro, in vivo and in silico models of drug distribution into the brain. J Pharmacokinet Pharmacodyn 2013; 40:301-14. [DOI: 10.1007/s10928-013-9303-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Accepted: 01/31/2013] [Indexed: 10/27/2022]
|
47
|
Riss PJ, Hong YT, Marton J, Caprioli D, Williamson DJ, Ferrari V, Saigal N, Roth BL, Henriksen G, Fryer TD, Dalley JW, Aigbirhio FI. Synthesis and Evaluation of 18F-FE-PEO in Rodents: An 18F-Labeled Full Agonist for Opioid Receptor Imaging. J Nucl Med 2013; 54:299-305. [DOI: 10.2967/jnumed.112.108688] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|