1
|
Lee SY, Oh HR, Kim YH, Bae SH, Lee Y, Lee YS, Lee BC, Cheon GJ, Kang KW, Youn H. Cerenkov luminescence imaging of interscapular brown adipose tissue using a TSPO-targeting PET probe in the UCP1 ThermoMouse. Am J Cancer Res 2022; 12:6380-6394. [PMID: 36168637 PMCID: PMC9475450 DOI: 10.7150/thno.74828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/20/2022] [Indexed: 11/21/2022] Open
Abstract
Rationale: [18F]fluorodeoxyglucose-positron emission tomography ([18F]FDG-PET) has been widely used as an imaging technique to measure interscapular brown adipose tissue (iBAT) activity. However, it is challenging to obtain iBAT-specific images using [18F]FDG-PET because increased uptake of [18F]FDG is observed in tumors, muscle, and inflamed tissues. Uncoupling protein 1 (UCP1) in the mitochondrial membrane, a well-known molecular marker of BAT, has been proposed as a useful BAT imaging marker. Recently, the UCP1 ThermoMouse was developed as a reporter mouse for monitoring UCP1 expression and investigating BAT activation. In addition, Translocator protein-18 kDa (TSPO) located in the outer mitochondrial membrane is also overexpressed in BAT, suggesting that TSPO-targeting PET has potential for iBAT imaging. However, there are no studies monitoring BAT using TSPO-targeting PET probes in the UCP1 ThermoMouse. Moreover, the non-invasive Cerenkov luminescence imaging (CLI) using Cerenkov radiation from the PET probe has been proposed as an alternative option for PET as it is less expensive and user-friendly. Therefore, we selected [18F]fm-PBR28-d2 as a TSPO-targeting PET probe for iBAT imaging to evaluate the usefulness of CLI in the UCP1 ThermoMouse. Methods: UCP1 ThermoMouse was used to monitor UCP1 expression. Western blotting and immunohistochemistry were performed to measure the level of protein expression. [18F]fm-PBR28-d2 and [18F]FDG were used as radioactive probes for iBAT imaging. PET images were acquired with SimPET, and optical images were acquired with IVIS 100. Results: UCP1 ThermoMouse showed that UCP1 and TSPO expressions were correlated in iBAT. In both PET and CLI, the TSPO-targeting probe [18F]fm-PBR28-d2 was superior to [18F]FDG for acquiring iBAT images. The high molar activity of the probe was essential for CLI and PET imaging. We tested the feasibility of TSPO-targeting probe under cold exposure by imaging with TSPO-PET/CLI. Both signals of iBAT were clearly increased after cold stimulation. Under prolonged isoflurane anesthesia, TSPO-targeting images showed higher signals from iBAT in the short-term than in long-term groups. Conclusion: We demonstrated that TSPO-PET/CLI reflected UCP1 expression in iBAT imaging better than [18F]FDG-PET/CLI under the various conditions. Considering convenience and cost, TSPO-CLI could be used as an alternative TSPO-PET technique for iBAT imaging.
Collapse
Affiliation(s)
- Seok-Yong Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ho Rim Oh
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Young-Hwa Kim
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sung-Hwan Bae
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yongseok Lee
- Cancer Imaging Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Imaging Center, Seoul National University Hospital, Seoul, Republic of Korea.,Radiation Medicine Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Byung Chul Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.,Center for Nanomolecular Imaging and Innovative Drug Development, Advanced Institutes of Convergence Technology, Seoul National University, Suwon, Republic of Korea
| | - Gi Jeong Cheon
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyewon Youn
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Imaging Center, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
2
|
Landau AM, Noer O, Alstrup AKO, Audrain H, Wegener G, Gjedde A, Doudet DJ, Winterdahl M. Type of Anaesthetic Influences [ 11C]MDL100,907 Binding to 5HT 2A Receptors in Porcine Brain. Mol Imaging Biol 2020; 22:797-804. [PMID: 31993926 DOI: 10.1007/s11307-020-01476-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
PURPOSE Anaesthesia routinely is used in animal neuroimaging in order to reduce head motion artefacts and minimize the influence of stress. However, anaesthetics can modify radioligand binding profiles at receptor targets studied by positron emission tomography (PET). Here, we determined the effects of two routine anaesthetics on the binding of a tracer of the serotonin 5HT2A receptors. PROCEDURES Isoflurane- and propofol-anesthetised Göttingen minipigs were imaged with [11C]MDL100,907 PET and analysed using regions of interest and statistical non-parametric mapping. RESULTS The binding potentials of the tracer in striatum under isoflurane anaesthesia significantly exceeded those obtained under propofol anaesthesia, an effect we attribute to the higher blood flow in brain induced by the former. CONCLUSIONS Interactions between radioligands and anaesthesia must be carefully evaluated in the design of in vivo neuroimaging and interpretation of data.
Collapse
Affiliation(s)
- Anne M Landau
- Department of Nuclear Medicine and PET, Aarhus University, Aarhus, Denmark. .,Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark.
| | - Ove Noer
- Department of Nuclear Medicine and PET, Aarhus University, Aarhus, Denmark
| | | | - Hélène Audrain
- Department of Nuclear Medicine and PET, Aarhus University, Aarhus, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
| | - Albert Gjedde
- Department of Nuclear Medicine and PET, Aarhus University, Aarhus, Denmark.,Department of Nuclear Medicine, University of Southern Denmark, Odense University Hospital, Odense, Denmark.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
| | - Doris J Doudet
- Department of Medicine/Neurology, University of British Columbia, Vancouver, Canada
| | - Michael Winterdahl
- Department of Nuclear Medicine and PET, Aarhus University, Aarhus, Denmark
| |
Collapse
|
3
|
Yang KC, Stepanov V, Martinsson S, Ettrup A, Takano A, Knudsen GM, Halldin C, Farde L, Finnema SJ. Fenfluramine Reduces [11C]Cimbi-36 Binding to the 5-HT2A Receptor in the Nonhuman Primate Brain. Int J Neuropsychopharmacol 2017; 20:683-691. [PMID: 28911007 PMCID: PMC5581490 DOI: 10.1093/ijnp/pyx051] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/18/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND [11C]Cimbi-36 is a serotonin 2A receptor agonist positron emission tomography radioligand that has recently been examined in humans. The binding of agonist radioligand is expected to be more sensitive to endogenous neurotransmitter concentrations than antagonist radioligands. In the current study, we compared the effect of serotonin releaser fenfluramine on the binding of [11C]Cimbi-36, [11C]MDL 100907 (a serotonin 2A receptor antagonist radioligand), and [11C]AZ10419369 (a serotonin 1B receptor partial agonist radioligand with established serotonin sensitivity) in the monkey brain. METHODS Eighteen positron emission tomography measurements, 6 for each radioligand, were performed in 3 rhesus monkeys before or after administration of 5.0 mg/kg fenfluramine. Binding potential values were determined with the simplified reference tissue model using cerebellum as the reference region. RESULTS Fenfluramine significantly decreased [11C]Cimbi-36 (26-62%) and [11C]AZ10419369 (35-58%) binding potential values in most regions (P < 0.05). Fenfluramine-induced decreases in [11C]MDL 100907 binding potential were 8% to 30% and statistically significant in 3 regions. Decreases in [11C]Cimbi-36 binding potential were larger than for [11C]AZ10419369 in neocortical and limbic regions (~35%) but smaller in striatum and thalamus (~40%). Decreases in [11C]Cimbi-36 binding potential were 0.9 to 2.8 times larger than for [11C]MDL 100907, and the fraction of serotonin 2A receptor in the high-affinity state was estimated as 54% in the neocortex. CONCLUSIONS The serotonin sensitivity of serotonin 2A receptor agonist radioligand [11C]Cimbi-36 was higher than for antagonist radioligand [11C]MDL 100907. The serotonin sensitivity of [11C]Cimbi-36 was similar to [11C]AZ10419369, which is one of the most sensitive radioligands. [11C]Cimbi-36 is a promising radioligand to examine serotonin release in the primate brain.
Collapse
Affiliation(s)
- Kai-Chun Yang
- Karolinska Institutet and Stockholm County Council, Department of Clinical Neuroscience, Center for Psychiatric Research, Stockholm, Sweden (Drs Yang and Stepanov, Mr Martinsson, and Drs Takano, Halldin, Farde, and Finnema); Rigshospitalet, Center for Integrated Molecular Brain Imaging, Copenhagen, Denmark and University of Copenhagen, Faculty of Health and Medicine Sciences, Copenhagen, Denmark (Drs Ettrup and Knudsen); AstraZeneca, PET Science Center at Karolinska Institutet, Personalized Health Care and Biomarkers, Stockholm, Sweden (Dr Farde).,Correspondence: Kai-Chun Yang, MD, Karolinska Institutet and Stockholm County Council, Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska University Hospital, Building R5:02, SE-171 76 Stockholm, Sweden ()
| | - Vladimir Stepanov
- Karolinska Institutet and Stockholm County Council, Department of Clinical Neuroscience, Center for Psychiatric Research, Stockholm, Sweden (Drs Yang and Stepanov, Mr Martinsson, and Drs Takano, Halldin, Farde, and Finnema); Rigshospitalet, Center for Integrated Molecular Brain Imaging, Copenhagen, Denmark and University of Copenhagen, Faculty of Health and Medicine Sciences, Copenhagen, Denmark (Drs Ettrup and Knudsen); AstraZeneca, PET Science Center at Karolinska Institutet, Personalized Health Care and Biomarkers, Stockholm, Sweden (Dr Farde)
| | - Stefan Martinsson
- Karolinska Institutet and Stockholm County Council, Department of Clinical Neuroscience, Center for Psychiatric Research, Stockholm, Sweden (Drs Yang and Stepanov, Mr Martinsson, and Drs Takano, Halldin, Farde, and Finnema); Rigshospitalet, Center for Integrated Molecular Brain Imaging, Copenhagen, Denmark and University of Copenhagen, Faculty of Health and Medicine Sciences, Copenhagen, Denmark (Drs Ettrup and Knudsen); AstraZeneca, PET Science Center at Karolinska Institutet, Personalized Health Care and Biomarkers, Stockholm, Sweden (Dr Farde)
| | - Anders Ettrup
- Karolinska Institutet and Stockholm County Council, Department of Clinical Neuroscience, Center for Psychiatric Research, Stockholm, Sweden (Drs Yang and Stepanov, Mr Martinsson, and Drs Takano, Halldin, Farde, and Finnema); Rigshospitalet, Center for Integrated Molecular Brain Imaging, Copenhagen, Denmark and University of Copenhagen, Faculty of Health and Medicine Sciences, Copenhagen, Denmark (Drs Ettrup and Knudsen); AstraZeneca, PET Science Center at Karolinska Institutet, Personalized Health Care and Biomarkers, Stockholm, Sweden (Dr Farde)
| | - Akihiro Takano
- Karolinska Institutet and Stockholm County Council, Department of Clinical Neuroscience, Center for Psychiatric Research, Stockholm, Sweden (Drs Yang and Stepanov, Mr Martinsson, and Drs Takano, Halldin, Farde, and Finnema); Rigshospitalet, Center for Integrated Molecular Brain Imaging, Copenhagen, Denmark and University of Copenhagen, Faculty of Health and Medicine Sciences, Copenhagen, Denmark (Drs Ettrup and Knudsen); AstraZeneca, PET Science Center at Karolinska Institutet, Personalized Health Care and Biomarkers, Stockholm, Sweden (Dr Farde)
| | - Gitte M Knudsen
- Karolinska Institutet and Stockholm County Council, Department of Clinical Neuroscience, Center for Psychiatric Research, Stockholm, Sweden (Drs Yang and Stepanov, Mr Martinsson, and Drs Takano, Halldin, Farde, and Finnema); Rigshospitalet, Center for Integrated Molecular Brain Imaging, Copenhagen, Denmark and University of Copenhagen, Faculty of Health and Medicine Sciences, Copenhagen, Denmark (Drs Ettrup and Knudsen); AstraZeneca, PET Science Center at Karolinska Institutet, Personalized Health Care and Biomarkers, Stockholm, Sweden (Dr Farde)
| | - Christer Halldin
- Karolinska Institutet and Stockholm County Council, Department of Clinical Neuroscience, Center for Psychiatric Research, Stockholm, Sweden (Drs Yang and Stepanov, Mr Martinsson, and Drs Takano, Halldin, Farde, and Finnema); Rigshospitalet, Center for Integrated Molecular Brain Imaging, Copenhagen, Denmark and University of Copenhagen, Faculty of Health and Medicine Sciences, Copenhagen, Denmark (Drs Ettrup and Knudsen); AstraZeneca, PET Science Center at Karolinska Institutet, Personalized Health Care and Biomarkers, Stockholm, Sweden (Dr Farde)
| | - Lars Farde
- Karolinska Institutet and Stockholm County Council, Department of Clinical Neuroscience, Center for Psychiatric Research, Stockholm, Sweden (Drs Yang and Stepanov, Mr Martinsson, and Drs Takano, Halldin, Farde, and Finnema); Rigshospitalet, Center for Integrated Molecular Brain Imaging, Copenhagen, Denmark and University of Copenhagen, Faculty of Health and Medicine Sciences, Copenhagen, Denmark (Drs Ettrup and Knudsen); AstraZeneca, PET Science Center at Karolinska Institutet, Personalized Health Care and Biomarkers, Stockholm, Sweden (Dr Farde)
| | - Sjoerd J Finnema
- Karolinska Institutet and Stockholm County Council, Department of Clinical Neuroscience, Center for Psychiatric Research, Stockholm, Sweden (Drs Yang and Stepanov, Mr Martinsson, and Drs Takano, Halldin, Farde, and Finnema); Rigshospitalet, Center for Integrated Molecular Brain Imaging, Copenhagen, Denmark and University of Copenhagen, Faculty of Health and Medicine Sciences, Copenhagen, Denmark (Drs Ettrup and Knudsen); AstraZeneca, PET Science Center at Karolinska Institutet, Personalized Health Care and Biomarkers, Stockholm, Sweden (Dr Farde)
| |
Collapse
|
4
|
Oxytocin and Serotonin Brain Mechanisms in the Nonhuman Primate. J Neurosci 2017; 37:6741-6750. [PMID: 28607170 DOI: 10.1523/jneurosci.0659-17.2017] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/31/2017] [Accepted: 06/01/2017] [Indexed: 01/29/2023] Open
Abstract
Oxytocin (OT) is increasingly studied for its therapeutic potential in psychiatric disorders, which are associated with the deregulation of several neurotransmission systems. Studies in rodents demonstrated that the interaction between OT and serotonin (5-HT) is critical for several aspects of social behavior. Using PET scan in humans, we have recently found that 5-HT 1A receptor (5-HT1AR) function is modified after intranasal oxytocin intake. However, the underlying mechanism between OT and 5-HT remains unclear. To understand this interaction, we tested 3 male macaque monkeys using both [11C]DASB and [18F]MPPF, two PET radiotracers, marking the serotonin transporter and the 5-HT1AR, respectively. Oxytocin (1 IU in 20 μl of ACSF) or placebo was injected into the brain lateral ventricle 45 min before scans. Additionally, we performed postmortem autoradiography. Compared with placebo, OT significantly reduced [11C]DASB binding potential in right amygdala, insula, and hippocampus, whereas [18F]MPPF binding potential increased in right amygdala and insula. Autoradiography revealed that [11C]DASB was sensitive to physiological levels of 5-HT modification, and that OT does not act directly on the 5-HT1AR. Our results show that oxytocin administration in nonhuman primates influences serotoninergic neurotransmission via at least two ways: (1) by provoking a release of serotonin in key limbic regions; and (2) by increasing the availability of 5-HT1AR receptors in the same limbic areas. Because these two molecules are important for social behavior, our study sheds light on the specific nature of their interaction, therefore helping to develop new mechanisms-based therapies for psychiatric disorders.SIGNIFICANCE STATEMENT Social behavior is largely controlled by brain neuromodulators, such as oxytocin and serotonin. While these are currently targeted in the context of psychiatric disorders such as autism and schizophrenia, a new promising pharmaceutical strategy is to study the interaction between these systems. Here we depict the interplay between oxytocin and serotonin in the nonhuman primate brain. We found that oxytocin provokes the release of serotonin, which in turn impacts on the serotonin 1A receptor system, by modulating its availability. This happens in several key brain regions for social behavior, such as the amygdala and insula. This novel finding can open ways to advance treatments where drugs are combined to influence several neurotransmission networks.
Collapse
|
5
|
Kolesnikova TO, Khatsko SL, Shevyrin VA, Morzherin YY, Kalueff AV. Effects of a non-competitive N-methyl-d-aspartate (NMDA) antagonist, tiletamine, in adult zebrafish. Neurotoxicol Teratol 2017; 59:62-67. [DOI: 10.1016/j.ntt.2016.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 11/25/2016] [Accepted: 11/29/2016] [Indexed: 10/20/2022]
|
6
|
Effects of common anesthetic agents on [ 18F]flumazenil binding to the GABA A receptor. EJNMMI Res 2016; 6:80. [PMID: 27826950 PMCID: PMC5101239 DOI: 10.1186/s13550-016-0235-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 10/29/2016] [Indexed: 12/25/2022] Open
Abstract
Background The availability of GABAA receptor binding sites in the brain can be assessed by positron emission tomography (PET) using the radioligand, [18F]flumazenil. However, the brain uptake and binding of this PET radioligand are influenced by anesthetic drugs, which are typically needed in preclinical imaging studies and clinical imaging studies involving patient populations that do not tolerate relatively longer scan times. The objective of this study was to examine the effects of anesthesia on the binding of [18F]flumazenil to GABAA receptors in mice. Methods Brain and whole blood radioactivity concentrations were measured ex vivo by scintillation counting or in vivo by PET in four groups of mice following administration of [18F]flumazenil: awake mice and mice anesthetized with isoflurane, dexmedetomidine, or ketamine/dexmedetomidine. Dynamic PET recordings were obtained for 60 min in mice anesthetized by either isoflurane or ketamine/dexmedetomidine. Static PET recordings were obtained at 25 or 55 min after [18F]flumazenil injection in awake or dexmedetomidine-treated mice acutely anesthetized with isoflurane. The apparent distribution volume (VT*) was calculated for the hippocampus and frontal cortex from either the full dynamic PET scans using an image-derived input function or from a series of ex vivo experiments using whole blood as the input function. Results PET images showed persistence of high [18F]flumazenil uptake (up to 20 % ID/g) in the brains of mice scanned under isoflurane or ketamine/dexmedetomidine anesthesia, whereas uptake was almost indiscernible in late samples or static scans from awake or dexmedetomidine-treated animals. The steady-state VT* was twofold higher in hippocampus of isoflurane-treated mice and dexmedetomidine-treated mice than in awake mice. Conclusions Anesthesia has pronounced effects on the binding and blood-brain distribution of [18F]flumazenil. Consequently, considerable caution must be exercised in the interpretation of preclinical and clinical PET studies of GABAA receptors involving the use of anesthesia.
Collapse
|
7
|
Yokoyama C, Mawatari A, Kawasaki A, Takeda C, Onoe K, Doi H, Newman-Tancredi A, Zimmer L, Onoe H. Marmoset Serotonin 5-HT1A Receptor Mapping with a Biased Agonist PET Probe 18F-F13714: Comparison with an Antagonist Tracer 18F-MPPF in Awake and Anesthetized States. Int J Neuropsychopharmacol 2016; 19:pyw079. [PMID: 27608810 PMCID: PMC5203761 DOI: 10.1093/ijnp/pyw079] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 09/05/2016] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND In vivo mapping by positron emission tomography of the serotonin 1A receptors has been hindered by the lack of suitable agonist positron emission tomography probes. 18F-labeled F13714 is a recently developed biased agonist positron emission tomography probe that preferentially targets subpopulations of serotonin 1A receptors in their "active state," but its brain labeling pattern in nonhuman primate has not been described. In addition, a potential confound in the translatability of PET data between nonhuman animal and human arise from the use of anesthetics that may modify the binding profiles of target receptors. METHODS Positron emission tomography scans were conducted in a cohort of common marmosets (n=4) using the serotonin 1A receptor biased agonist radiotracer, 18F-F13714, compared with a well-characterized 18F-labeled antagonist radiotracer, 18F-MPPF. Experiments on each animal were performed under both consciousness and isoflurane-anesthesia conditions. RESULTS 18F-F13714 binding distribution in marmosets by positron emission tomography differs markedly from that of the 18F-MPPF. Whereas 18F-MPPF showed highest binding in hippocampus and amygdala, 18F-F13714 showed highest labeling in other regions, including insular and cingulate cortex, thalamus, raphe, caudate nucleus, and putamen. The binding potential values of 18F-F13714 were about one-third of those observed with 18F-MPPF, with marked individual- and region-specific differences under isoflurane-anesthetized vs conscious conditions. CONCLUSIONS These findings highlight the importance of investigating the brain imaging of serotonin 1A receptors using agonist probes such as 18F-F13714, which may preferentially target subpopulations of serotonin 1A receptors in specific brain regions of nonhuman primate as a biased agonist.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hirotaka Onoe
- RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan (Dr Yokoyama, Ms Mawatari, Mr Kawasaki, Ms Takeda, Ms K. Onoe, Dr Doi, Dr H. Onoe); Neurolixis Inc, Dana Point, CA (Dr Newman-Tancredi); Université Claude Bernard Lyon 1, Hospices Civils de Lyon, INSERM, CNRS, Lyon Neuroscience Research Center, Lyon, France (Dr Zimmer).
| |
Collapse
|
8
|
Imaging Dopamine and Serotonin Systems on MPTP Monkeys: A Longitudinal PET Investigation of Compensatory Mechanisms. J Neurosci 2016; 36:1577-89. [PMID: 26843639 DOI: 10.1523/jneurosci.2010-15.2016] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED It is now widely accepted that compensatory mechanisms are involved during the early phase of Parkinson's disease (PD) to delay the expression of motor symptoms. However, the neurochemical mechanisms underlying this presymptomatic period are still unclear. Here, we measured in vivo longitudinal changes of both the dopaminergic and serotonergic systems in seven asymptomatic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated monkeys (when motor symptoms are less apparent) using PET. We used the progressively MPTP-intoxicated monkey model that expresses recovery from motor symptoms to study the changes in dopamine synthesis ([(18)F]DOPA), dopamine D2/D3 receptors ([(11)C]raclopride), and serotonin transporter (11)C-N,N-dimethyl-2-(-2-amino-4-cyanophenylthio) benzylamine ([(11)C]DASB) and serotonin 1A receptor ([(18)F]MPPF) levels between four different states (baseline, early symptomatic, full symptomatic and recovered). During the early symptomatic state, we observed increases of [(18)F]DOPA uptake in the anterior putamen, [(11)C]raclopride binding in the posterior striatum, and 2'-methoxyphenyl-(N-2'-pyridinyl)-p-[(18)F]fluoro-benzamidoethylpiperazine [(18)F]MPPF uptake in the orbitofrontal cortex and dorsal ACC. After recovery from motor symptoms, the results mainly showed decreased [(11)C]raclopride binding in the anterior striatum and limbic ACC. In addition, our findings supported the importance of pallidal dopaminergic neurotransmission in both the early compensatory mechanisms and the functional recovery mechanisms, with reduced aromatic L-amino acid decarboxylase (AAAD) activity closely related to the appearance or perseveration of motor symptoms. In parallel, this study provides preliminary evidence of the role of the serotonergic system in compensatory mechanisms. Nonetheless, future studies are needed to determine whether there are changes in SERT availability in the early symptomatic state and if [(18)F]MPPF PET imaging might be a promising biomarker of early degenerative changes in PD. SIGNIFICANCE STATEMENT The present research provides evidence of the potential of combining a multitracer PET imaging technique and a longitudinal protocol applied on a progressively 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-intoxicated monkey model to further elucidate the nature of the compensatory mechanisms involved in the preclinical period of Parkinson's disease (PD). In particular, by investigating the dopaminergic and serotonergic changes both presynaptically and postsynaptically at four different motor states (baseline, early symptomatic, full symptomatic, and recovered), this study has allowed us to identify putative biomarkers for future therapeutic interventions to prevent and/or delay disease expression. For example, our findings suggest that the external pallidum could be a new target for cell-based therapies to reduce PD symptoms.
Collapse
|
9
|
Stehouwer JS, Goodman MM. Fluorine-18 Radiolabeled PET Tracers for Imaging Monoamine Transporters: Dopamine, Serotonin, and Norepinephrine. PET Clin 2016; 4:101-28. [PMID: 20216936 DOI: 10.1016/j.cpet.2009.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This review focuses on the development of fluorine-18 radiolabeled PET tracers for imaging the dopamine transporter (DAT), serotonin transporter (SERT), and norepinephrine transporter (NET). All successful DAT PET tracers reported to date are members of the 3β-phenyl tropane class and are synthesized from cocaine. Currently available carbon-11 SERT PET tracers come from both the diphenylsulfide and 3β-phenyl nortropane class, but so far only the nortropanes have found success with fluorine-18 derivatives. NET imaging has so far employed carbon-11 and fluorine-18 derivatives of reboxetine but due to defluorination of the fluorine-18 derivatives further research is still necessary.
Collapse
|
10
|
Vanhove C, Bankstahl JP, Krämer SD, Visser E, Belcari N, Vandenberghe S. Accurate molecular imaging of small animals taking into account animal models, handling, anaesthesia, quality control and imaging system performance. EJNMMI Phys 2015; 2:31. [PMID: 26560138 PMCID: PMC4642455 DOI: 10.1186/s40658-015-0135-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/05/2015] [Indexed: 11/22/2022] Open
Abstract
Small-animal imaging has become an important technique for the development of new radiotracers, drugs and therapies. Many laboratories have now a combination of different small-animal imaging systems, which are being used by biologists, pharmacists, medical doctors and physicists. The aim of this paper is to give an overview of the important factors in the design of a small animal, nuclear medicine and imaging experiment. Different experts summarize one specific aspect important for a good design of a small-animal experiment.
Collapse
Affiliation(s)
- Christian Vanhove
- Department of Electronics and Information Systems, MEDISIP, Ghent University-iMinds Medical IT-IBiTech, De Pintelaan 185 block B, B-9000, Ghent, Belgium.
| | - Jens P Bankstahl
- Department of Nuclear Medicine, Preclinical Molecular Imaging, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Stefanie D Krämer
- Radiopharmaceutical Sciences/Biopharmacy, ETH Zurich, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Vladimir-Prelog-Weg 4, CH-8093, Zurich, Switzerland
| | - Eric Visser
- Department of Radiology and Nuclear Medicine, Radboudumc, 6525 GA Nijmegen, the Netherlands
| | - Nicola Belcari
- Department of Physics, University of Pisa and INFN sezione di Pisa, 56127 Pisa, Italy
| | - Stefaan Vandenberghe
- Department of Electronics and Information Systems, MEDISIP, Ghent University-iMinds Medical IT-IBiTech, De Pintelaan 185 block B, B-9000, Ghent, Belgium
| |
Collapse
|
11
|
Finnema SJ, Halldin C, Bang-Andersen B, Bundgaard C, Farde L. Serotonin transporter occupancy by escitalopram and citalopram in the non-human primate brain: a [(11)C]MADAM PET study. Psychopharmacology (Berl) 2015; 232:4159-67. [PMID: 25980484 DOI: 10.1007/s00213-015-3961-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/05/2015] [Indexed: 11/29/2022]
Abstract
RATIONALE A number of serotonin receptor positron emission tomography (PET) radioligands have been shown to be sensitive to changes in extracellular serotonin concentration, in a generalization of the well-known dopamine competition model. High doses of selective serotonin reuptake inhibitors (SSRIs) decrease serotonin receptor availability in monkey brain, consistent with increased serotonin concentrations. However, two recent studies on healthy human subjects, using a single, lower and clinically relevant SSRI dose, showed increased cortical serotonin receptor radioligand binding, suggesting potential decreases in serotonin concentration in projection regions when initiating treatment. OBJECTIVES The cross-species differential SSRI effect may be partly explained by serotonin transporter (SERT) occupancy in monkey brain being higher than is clinically relevant. We here determine SERT occupancy after single doses of escitalopram or citalopram by conducting PET measurements with [(11)C]MADAM in monkeys. Relationships between dose, plasma concentration and SERT occupancy were estimated by one-site binding analyses. Binding affinity was expressed as dose (ID50) or plasma concentration (K i) where 50 % SERT occupancy was achieved. RESULTS Estimated ID50 and K i values were 0.020 mg/kg and 9.6 nmol/L for escitalopram and 0.059 mg/kg and 9.7 nmol/L for citalopram, respectively. Obtained K i values are comparable to values reported in humans. CONCLUSIONS Escitalopram or citalopram doses nearly saturated SERT in previous monkey studies which examined serotonin sensitivity of receptor radioligands. PET-measured cross-species differential effects of SSRI on cortical serotonin concentration may thus be related to SSRI dose. Future monkey studies using SSRI doses inducing clinically relevant SERT occupancy may further illuminate the delayed onset of SSRI therapeutic effects.
Collapse
Affiliation(s)
- Sjoerd J Finnema
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Karolinska University Hospital, Building R5:02, SE-17176, Stockholm, Sweden. .,Department of Diagnostic Radiology, Yale University, New Haven, CT, USA.
| | - Christer Halldin
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Karolinska University Hospital, Building R5:02, SE-17176, Stockholm, Sweden
| | | | | | - Lars Farde
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Karolinska University Hospital, Building R5:02, SE-17176, Stockholm, Sweden.,AstraZeneca, Translational Science Center at Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
12
|
Chen Z, Tang J, Liu C, Li X, Huang H, Xu X, Yu H. Effects of anesthetics on vesicular monoamine transporter type 2 binding to ¹⁸F-FP-(+)-DTBZ: a biodistribution study in rat brain. Nucl Med Biol 2015; 43:124-129. [PMID: 26526872 DOI: 10.1016/j.nucmedbio.2015.09.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/14/2015] [Accepted: 09/28/2015] [Indexed: 11/30/2022]
Abstract
OBJECTIVES The in vivo binding analysis of vesicular monoamine transporter type 2 (VMAT2) to radioligand has provided a means of investigating related disorders. Anesthesia is often inevitable when the investigations are performed in animals. In the present study, we tested effects of four commonly-used anesthetics: isoflurane, pentobarbital, chloral hydrate and ketamine, on in vivo VMAT2 binding to (18)F-FP-(+)-DTBZ, a specific VMAT2 radioligand, in rat brain. METHODS The transient equilibrium time window for in vivo binding of (18)F-FP-(+)-DTBZ after a bolus injection was firstly determined. The brain biodistribution studies under anesthetized and awake rats were then performed at the equilibrium time. Standard uptake values (SUVs) of the interest brain regions: the striatum (ST), hippocampus (HP), cortex (CX) and cerebellum (CB) were obtained; and ratios of tissue to cerebellum were calculated. RESULTS Isoflurane and pentobarbital did not alter distribution of (18)F-FP-(+)-DTBZ in the brain relative to the awake group; neither SUVs nor ratios of ST/CB and HP/CB were altered significantly. Chloral hydrate significantly increased SUVs of all the brain regions, but did not significantly alter ratios of ST/CB and HP/CB. Ketamine significantly increased SUVs of the striatum, hippocampus and cortex, and insignificantly increased the SUV of the cerebellum; consequently, ketamine significantly increased ratios of ST/CB and HP/CB. CONCLUSIONS It is concluded that in vivo VMAT2 binding to (18)F-FP-(+)-DTBZ are not altered by isoflurane and pentobarbital, but altered by chloral hydrate and ketamine. Isoflurane and pentobarbital may be promising anesthetic compounds for investigating in vivo VMAT2 binding. Further studies are warranted to investigate the interactions of anesthetics with VMAT2 binding potential with in vivo PET studies.
Collapse
Affiliation(s)
- Zhengping Chen
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China, 214063.
| | - Jie Tang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China, 214063
| | - Chunyi Liu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China, 214063
| | - Xiaomin Li
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China, 214063
| | - Hongbo Huang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China, 214063
| | - Xijie Xu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China, 214063
| | - Huixin Yu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China, 214063
| |
Collapse
|
13
|
Kroll T, Elmenhorst D, Weisshaupt A, Beer S, Bauer A. Reproducibility of non-invasive a1 adenosine receptor quantification in the rat brain using [(18)F]CPFPX and positron emission tomography. Mol Imaging Biol 2015; 16:699-709. [PMID: 24595700 DOI: 10.1007/s11307-014-0729-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
PURPOSE The A1AR antagonist 8-cyclopentyl-3-(3-fluoropropyl)-1-propylxanthine ([(18)F]CPFPX) has recently been shown to be a suitable radiotracer for quantitative in vivo imaging of the A1 adenosine receptor (A1AR) in rats. The present study evaluates the reproducibility of non-invasive longitudinal A1AR studies with [(18)F]CPFPX and a dedicated small animal positron emission tomography (PET) scanner. PROCEDURES Twelve male Sprague Dawley rats underwent four repeated dynamic PET scans with a bolus injection of [(18)F]CPFPX. A1AR availability was determined by different non-invasive approaches including simplified and multilinear reference tissue (olfactory bulb)-based models and graphical methods. The outcome parameter binding potential (BP) was evaluated in terms of variability and reproducibility. RESULTS Repeated estimations of [(18)F]CPFPX BP ND gave reliable results with acceptable variability (mean 12 %) and reproducibility (intraclass correlation coefficients raging from 0.57 to 0.68) in cortical and subcortical regions of the rat brain. With regard to kinetic models, test-retest stability of the simplified reference-tissue model (SRTM) was superior to multilinear and graphical approaches. CONCLUSIONS Non-invasive quantification of A1AR density in the rat brain is reproducible and reliable with [(18)F]CPFPX PET and allows longitudinal designs of in vivo imaging studies in rodents.
Collapse
Affiliation(s)
- Tina Kroll
- Institute of Neuroscience and Medicine (INM-2), Forschungszentrum Jülich GmbH, 52425, Jülich, Germany,
| | | | | | | | | |
Collapse
|
14
|
Waelbers T, Polis I, Vermeire S, Dobbeleir A, Eersels J, De Spiegeleer B, Audenaert K, Peremans K. Effect of ketamine on the regional cerebral blood flow and binding index of the 5-HT2A receptor radioligand 123I-R91150 in the canine brain. J Vet Behav 2015. [DOI: 10.1016/j.jveb.2015.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
15
|
Saba W, Goutal S, Kuhnast B, Dollé F, Auvity S, Fontyn Y, Cayla J, Peyronneau MA, Valette H, Tournier N. Differential influence of propofol and isoflurane anesthesia in a non-human primate on the brain kinetics and binding of [(18)F]DPA-714, a positron emission tomography imaging marker of glial activation. Eur J Neurosci 2015; 42:1738-45. [PMID: 25962575 DOI: 10.1111/ejn.12946] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 05/05/2015] [Accepted: 05/07/2015] [Indexed: 01/19/2023]
Abstract
Translocator protein 18 kDa (TSPO) expression at the mitochondrial membrane of glial cells is related to glial activation. TSPO radioligands such as [(18)F]DPA-714 are useful for the non-invasive study of neuroimmune processes using positron emission tomography (PET). Anesthetic agents were shown to impact mitochondrial function and may influence [(18)F]DPA-714 binding parameters and PET kinetics. [(18) F]DPA-714 PET imaging was performed in Papio anubis baboons anesthetized using either intravenous propofol (n = 3) or inhaled isoflurane (n = 3). Brain kinetics and metabolite-corrected input function were measured to estimate [(18) F]DPA-714 brain distribution (VT). Displacement experiments were performed using PK11195 (1.5 mg/kg). In vitro [(18)F]DPA-714 binding experiments were performed using baboon brain tissue in the absence and presence of tested anesthetics. Brain radioactivity peaked higher in isoflurane-anesthetized animals compared with propofol (SUVmax = 2.7 ± 0.5 vs. 1.3 ± 0.2, respectively) but was not different after 30 min. Brain VT was not different under propofol and isoflurane. Displacement resulted in a 35.8 ± 8.4% decrease of brain radioactivity under propofol but not under isoflurane (0.1 ± 7.0%). In vitro, the presence of propofol increased TSPO density and dramatically reduced its affinity for [(18)F]DPA-714 compared with control. This in vitro effect was not significant with isoflurane. Exposure to propofol and isoflurane differentially influences TSPO interaction with its specific radioligand [(18)F]DPA-714 with subsequent impact on its tissue kinetics and specific binding estimated in vivo using PET. Therefore, the choice of anesthetics and their potential influence on PET data should be considered for the design of imaging studies using TSPO radioligands, especially in a translational research context.
Collapse
Affiliation(s)
- Wadad Saba
- Inserm / CEA / Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, F-91406, France
| | - Sébastien Goutal
- Inserm / CEA / Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, F-91406, France
| | - Bertrand Kuhnast
- Inserm / CEA / Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, F-91406, France
| | - Frédéric Dollé
- Inserm / CEA / Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, F-91406, France
| | - Sylvain Auvity
- Inserm / CEA / Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, F-91406, France
| | - Yoan Fontyn
- Inserm / CEA / Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, F-91406, France
| | - Jérôme Cayla
- Inserm / CEA / Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, F-91406, France
| | - Marie-Anne Peyronneau
- Inserm / CEA / Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, F-91406, France
| | - Héric Valette
- Inserm / CEA / Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, F-91406, France
| | - Nicolas Tournier
- Inserm / CEA / Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, F-91406, France
| |
Collapse
|
16
|
Stehouwer JS, Birnbaum MS, Voll RJ, Owens MJ, Plott SJ, Bourke CH, Wassef MA, Kilts CD, Goodman MM. Synthesis, F-18 radiolabeling, and microPET evaluation of 3-(2,4-dichlorophenyl)-N-alkyl-N-fluoroalkyl-2,5-dimethylpyrazolo[1,5-a]pyrimidin-7-amines as ligands of the corticotropin-releasing factor type-1 (CRF1) receptor. Bioorg Med Chem 2015; 23:4286-4302. [PMID: 26145817 DOI: 10.1016/j.bmc.2015.06.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/04/2015] [Accepted: 06/12/2015] [Indexed: 12/28/2022]
Abstract
A series of 3-(2,4-dichlorophenyl)-N-alkyl-N-fluoroalkyl-2,5-dimethylpyrazolo[1,5-a]pyrimidin-7-amines were synthesized and evaluated as potential positron emission tomography (PET) tracers for the corticotropin-releasing factor type-1 (CRF1) receptor. Compounds 27, 28, 29, and 30 all displayed high binding affinity (⩽1.2 nM) to the CRF1 receptor when assessed by in vitro competition binding assays at 23 °C, whereas a decrease in affinity (⩾10-fold) was observed with compound 26. The logP7.4 values of [(18)F]26-[(18)F]29 were in the range of ∼2.2-2.8 and microPET evaluation of [(18)F]26-[(18)F]29 in an anesthetized male cynomolgus monkey demonstrated brain penetrance, but specific binding was not sufficient enough to differentiate regions of high CRF1 receptor density from regions of low CRF1 receptor density. Radioactivity uptake in the skull, and sphenoid bone and/or sphenoid sinus during studies with [(18)F]28, [(18)F]28-d8, and [(18)F]29 was attributed to a combination of [(18)F]fluoride generated by metabolic defluorination of the radiotracer and binding of intact radiotracer to CRF1 receptors expressed on mast cells in the bone marrow. Uptake of [(18)F]26 and [(18)F]27 in the skull and sphenoid region was rapid but then steadily washed out which suggests that this behavior was the result of binding to CRF1 receptors expressed on mast cells in the bone marrow with no contribution from [(18)F]fluoride.
Collapse
Affiliation(s)
- Jeffrey S Stehouwer
- Center for Systems Imaging, Department of Radiology and Imaging Sciences, Emory University, WWHC 209, 1841 Clifton Rd NE, Atlanta, GA 30329, USA.
| | - Matthew S Birnbaum
- Center for Systems Imaging, Department of Radiology and Imaging Sciences, Emory University, WWHC 209, 1841 Clifton Rd NE, Atlanta, GA 30329, USA
| | - Ronald J Voll
- Center for Systems Imaging, Department of Radiology and Imaging Sciences, Emory University, WWHC 209, 1841 Clifton Rd NE, Atlanta, GA 30329, USA
| | - Michael J Owens
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Susan J Plott
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Chase H Bourke
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Michael A Wassef
- Center for Systems Imaging, Department of Radiology and Imaging Sciences, Emory University, WWHC 209, 1841 Clifton Rd NE, Atlanta, GA 30329, USA
| | - Clinton D Kilts
- Department of Psychiatry and Behavioral Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Mark M Goodman
- Center for Systems Imaging, Department of Radiology and Imaging Sciences, Emory University, WWHC 209, 1841 Clifton Rd NE, Atlanta, GA 30329, USA; Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| |
Collapse
|
17
|
Lewis DY, Champion S, Wyper D, Dewar D, Pimlott S. Assessment of [125I]WYE-230949 as a novel histamine H3 receptor radiopharmaceutical. PLoS One 2014; 9:e115876. [PMID: 25542008 PMCID: PMC4277420 DOI: 10.1371/journal.pone.0115876] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 12/02/2014] [Indexed: 12/15/2022] Open
Abstract
Histamine H3 receptor therapeutics have been proposed for several diseases such as schizophrenia, attention deficit hyperactivity disorder, Alzheimer's disease and obesity. We set out to evaluate the novel compound, [125I]WYE-230949, as a potential radionuclide imaging agent for the histamine H3 receptor in brain. [125I]WYE-230949 had a high in vitro affinity for the rat histamine H3 receptor (Kd of 6.9 nM). The regional distribution of [125I]WYE-230949 binding sites in rat brain, demonstrated by in vitro autoradiography, was consistent with the known distribution of the histamine H3 receptor. Rat brain uptake of intravenously injected [125I]WYE-230949 was low (0.11 %ID/g) and the ratio of specific: non-specific binding was less than 1.4, as determined by ex vivo autoradiography. In plasma, metabolism of [125I]WYE-230949 into a less lipophilic species occurred, such that less than 38% of the parent compound remained 30 minutes after injection. Brain uptake and metabolism of [125I]WYE-230949 were increased and specific binding was reduced in anaesthetised compared to conscious rats. [125I]WYE230949 is not a potential radiotracer for imaging rat histamine H3 receptors in vivo due to low brain uptake, in vivo metabolism of the parent compound and low specific binding.
Collapse
Affiliation(s)
- David Y. Lewis
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- * E-mail:
| | - Sue Champion
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - David Wyper
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Deborah Dewar
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sally Pimlott
- Department of Clinical Physics, Greater Glasgow NHS Trust and University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
18
|
Li Y, Wang X, Zhang J, Deuther-Conrad W, Xie F, Zhang X, Liu J, Qiao J, Cui M, Steinbach J, Brust P, Liu B, Jia H. Synthesis and Evaluation of Novel 18F-Labeled Spirocyclic Piperidine Derivatives as σ1 Receptor Ligands for Positron Emission Tomography Imaging. J Med Chem 2013; 56:3478-91. [DOI: 10.1021/jm301734g] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Yan Li
- Key Laboratory
of Radiopharmaceuticals
(Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, People’s
Republic of China
| | - Xia Wang
- Key Laboratory
of Radiopharmaceuticals
(Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, People’s
Republic of China
| | - Jinming Zhang
- Nuclear
Medicine Department, Chinese PLA General Hospital, Beijing 100853, People’s
Republic of China
| | - Winnie Deuther-Conrad
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Research
Site Leipzig, Department of Neuroradiopharmaceuticals, 04318 Leipzig,
Germany
| | - Fang Xie
- Key Laboratory
of Radiopharmaceuticals
(Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, People’s
Republic of China
| | - Xiaojun Zhang
- Nuclear
Medicine Department, Chinese PLA General Hospital, Beijing 100853, People’s
Republic of China
| | - Jian Liu
- Nuclear
Medicine Department, Chinese PLA General Hospital, Beijing 100853, People’s
Republic of China
| | - Jinping Qiao
- Key Laboratory
of Radiopharmaceuticals
(Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, People’s
Republic of China
| | - Mengchao Cui
- Key Laboratory
of Radiopharmaceuticals
(Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, People’s
Republic of China
| | - Jörg Steinbach
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Research
Site Leipzig, Department of Neuroradiopharmaceuticals, 04318 Leipzig,
Germany
| | - Peter Brust
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Research
Site Leipzig, Department of Neuroradiopharmaceuticals, 04318 Leipzig,
Germany
| | - Boli Liu
- Key Laboratory
of Radiopharmaceuticals
(Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, People’s
Republic of China
| | - Hongmei Jia
- Key Laboratory
of Radiopharmaceuticals
(Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, People’s
Republic of China
| |
Collapse
|
19
|
Alstrup AKO, Smith DF. Anaesthesia for positron emission tomography scanning of animal brains. Lab Anim 2013; 47:12-8. [PMID: 23349451 DOI: 10.1258/la.2012.011173] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Positron emission tomography (PET) provides a means of studying physiological and pharmacological processes as they occur in the living brain. Mice, rats, dogs, cats, pigs and non-human primates are often used in studies using PET. They are commonly anaesthetized with ketamine, propofol or isoflurane in order to prevent them from moving during the imaging procedure. The use of anaesthesia in PET studies suffers, however, from the drawback of possibly altering central neuromolecular mechanisms. As a result, PET findings obtained in anaesthetized animals may fail to correctly represent normal properties of the awake brain. Here, we review findings of PET studies carried out either in both awake and anaesthetized animals or in animals given at least two different anaesthetics. Such studies provide a means of estimating the extent to which anaesthesia affects the outcome of PET neuroimaging in animals. While no final conclusion can be drawn concerning the 'best' general anaesthetic for PET neuroimaging in laboratory animals, such studies provide findings that can enhance an understanding of neurobiological mechanisms in the living brain.
Collapse
Affiliation(s)
- Aage Kristian Olsen Alstrup
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospitals, Nørrebrogade 44, 10G, DK-8000 Aarhus C, Denmark.
| | | |
Collapse
|
20
|
Hines CS, Fujita M, Zoghbi SS, Kim JS, Quezado Z, Herscovitch P, Miao N, Ferraris Araneta MD, Morse C, Pike VW, Labovsky J, Innis RB. Propofol decreases in vivo binding of 11C-PBR28 to translocator protein (18 kDa) in the human brain. J Nucl Med 2012; 54:64-9. [PMID: 23148296 DOI: 10.2967/jnumed.112.106872] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED The PET radioligand (11)C-PBR28 targets translocator protein (18 kDa) (TSPO) and is a potential marker of neuroimmune activation in vivo. Although several patient populations have been studied using (11)C-PBR28, no investigators have studied cognitively impaired patients who would require anesthesia for the PET procedure, nor have any reports investigated the effects that anesthesia may have on radioligand uptake. The purpose of this study was to determine whether the anesthetic propofol alters brain uptake of (11)C-PBR28 in healthy subjects. METHODS Ten healthy subjects (5 men; 5 women) each underwent 2 dynamic brain PET scans on the same day, first at baseline and then with intravenous propofol anesthesia. The subjects were injected with 680 ± 14 MBq (mean ± SD) of (11)C-PBR28 for each PET scan. Brain uptake was measured as total distribution volume (V(T)) using the Logan plot and metabolite-corrected arterial input function. RESULTS Propofol decreased V(T), which corrects for any alteration of metabolism of the radioligand, by about 26% (P = 0.011). In line with the decrease in V(T), brain time-activity curves showed decreases of about 20% despite a 13% increase in plasma area under the curve with propofol. Reduction of V(T) with propofol was observed across all brain regions, with no significant region X condition interaction (P = 0.40). CONCLUSION Propofol anesthesia reduces the V(T) of (11)C-PBR28 by about 26% in the brains of healthy human subjects. Given this finding, future studies will measure neuroimmune activation in the brains of autistic volunteers and their age and sex-matched healthy controls using propofol anesthesia. We recommend that future PET studies using (11)C-PBR28 and concomitant propofol anesthesia, as would be required in impaired populations, include a control arm to account for the effects of propofol on brain measurements of TSPO.
Collapse
Affiliation(s)
- Christina S Hines
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland 20892-1026, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Tantawy MN, Peterson TE, Jones CK, Johnson K, Rook JM, Conn PJ, Baldwin RM, Ansari MS, Kessler RM. Impact of isoflurane anesthesia on D2 receptor occupancy by [18F]fallypride measured by microPET with a modified Logan plot. Synapse 2011; 65:1173-80. [PMID: 21584868 DOI: 10.1002/syn.20955] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2011] [Accepted: 05/11/2011] [Indexed: 11/11/2022]
Abstract
UNLABELLED In the previous work, we reported a method that utilized imaging data collected from 60 to 120 min following [(18) F]fallypride administration to estimate the distribution volume ratio DVR' (DVR' ∝ DVR; DVR = 1 + BP(ND) , where BP(ND) is a measure of receptor density, DA D2 in this case). In this work, we use this method to assess the effects of isoflurane anesthesia on [(18) F]fallypride DVR'. METHODS Rats were injected with [(18) F]fallypride either unconsciously under ∼1.5% isoflurane via the tail vein (Group 1) or consciously via a catheter inserted either in the jugular vein (Group 2) or the tail vein (Group 3). After about 1 h of free access to food and water the rats were anesthetized with 1.5% isoflurane and imaged in a microPET for 60 min. The rats that were injected consciously (Groups 2 and 3) were placed in a rat restrainer during [(18) F]fallypride injection. They were habituated in that restrainer for 3 days prior to the experiment day to minimize restraint-related stress. For comparison, a control group of rats was imaged for 120 min simultaneously with the administration of [(18) F]fallypride i.v. while under 1.5% isoflurane. The DVR' estimates from the 60 min acquisitions were compared with the DVR' from the last 60 min of the 120 min acquisitions (after neglecting the first 60 min). In addition, the striatal time-activity curves were fit with a 2-tissue + plasma compartment model using an arbitrary simulated plasma input function to obtain k(3) /k(4) (≈ BP(ND) ) for the 60 and 120 min acquisitions. RESULTS Isoflurane anesthesia caused a significant reduction, up to 22%, in the DVR' estimates, which were 15.7 ± 0.3 (mean ± SE) for the controls, 17.7 ± 0.3 for Group 1, 19.2 ± 0.4 for Group 2, and 18.8 ± 0.7 for Group 3. The compartmental model fit produced similar results, ∼30% reduction in k(3) /k(4) for the 120-min acquisitions compared with the 60-min acquisitions (initial conscious uptake of the radiotracer). CONCLUSION The results of this study demonstrate that isoflurane anesthesia significantly decreases striatal [(18) F]fallypride BP(ND) in rats. Of similar importance, this work demonstrates the effectiveness of delayed scans following radiotracer injection and the implication that different types of studies can be conducted simultaneously with this method, including studies of behavioral and environmental impact on brain receptors.
Collapse
Affiliation(s)
- Mohammed N Tantawy
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Palner M, Underwood MD, Kumar DJS, Arango V, Knudsen GM, John Mann J, Parsey RV. Ex vivo evaluation of the serotonin 1A receptor partial agonist [³H]CUMI-101 in awake rats. Synapse 2011; 65:715-23. [PMID: 21108237 DOI: 10.1002/syn.20888] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Accepted: 11/11/2010] [Indexed: 11/11/2022]
Abstract
[³H]CUMI-101 is a 5-HT(1A) partial agonist, which has been evaluated for use as a positron emission tracer in baboon and humans. We sought to evaluate the properties of [³H]CUMI-101 ex vivo in awake rats and determine if [³H]CUMI-101 can measure changes in synaptic levels of serotonin after different challenge paradigms. [³H]CUMI-101 shows good uptake and good specific binding ratio (SBR) in frontal cortex 5.18 and in hippocampus 3.18. Binding was inhibited in a one-binding-site fashion by WAY100635 and unlabeled CUMI-101. The ex vivo B(max) of [³H]CUMI-101 in frontal cortex (98.7 fmol/mg) and hippocampus (131 fmol/kg) agree with the ex vivo B(max) of [³H]MPPF in frontal cortex (147.1 fmol/mg) and hippocampus (72.1 fmol/mg) and with in vitro values reported with 8-OH-DPAT. Challenges with citalopram, a selective serotonin reuptake inhibitor, fenfluramine, a serotonin releaser, and 4-chloro-DL-phenylalanine, a serotonin synthesis inhibitor, did not show any effect on the standardized uptake values (SUVs) in any region. Citalopram did alter SBR, but this was due to changes in cerebellar SUVs. Our results indicate that [³H]CUMI-101 is a good radioligand for imaging 5-HT(1A) high-density regions in rats; however, the results from pharmacological challenges remain inconclusive.
Collapse
Affiliation(s)
- Mikael Palner
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York, USA.
| | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Molecular in vivo neuroimaging techniques can be used to measure regional changes in endogenous neurotransmitters, evoked by challenges that alter synaptic neurotransmitter concentration. This technique has most successfully been applied to the study of endogenous dopamine release using positron emission tomography, but has not yet been adequately extended to other neurotransmitter systems. This review focuses on how the technique has been applied to the study of the 5-hydroxytryptamine (5-HT) system. The principles behind visualising fluctuations in neurotransmitters are introduced, with reference to the dopaminergic system. Studies that aim to image acute, endogenous 5-HT release or depletion at 5-HT receptor targets are summarised, with particular attention to studies in humans. Radiotracers targeting the 5-HT(1A), 5-HT(2A), and 5-HT(4) receptors and the serotonin reuptake transporter have been explored for their sensitivity to 5-HT fluctuations, but with mixed outcomes; tracers for these targets cannot reliably image endogenous 5-HT in humans. Shortcomings in our basic knowledge of the mechanisms underlying changes in binding potential are addressed, and suggestions are made as to how the selection of targets, radiotracers, challenge paradigms, and experimental design might be optimised to improve our chances of successfully imaging endogenous neurotransmitters in the future.
Collapse
|
24
|
Stehouwer JS, Daniel LM, Chen P, Voll RJ, Williams L, Plott SJ, Votaw JR, Owens MJ, Howell L, Goodman MM. Synthesis, fluorine-18 radiolabeling, and biological evaluation of N-((E)-4-fluorobut-2-en-1-yl)-2beta-carbomethoxy-3beta-(4'-halophenyl)nortropanes: candidate radioligands for in vivo imaging of the brain dopamine transporter with positron emission tomography. J Med Chem 2010; 53:5549-57. [PMID: 20597489 DOI: 10.1021/jm100269c] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The N-(E)-fluorobutenyl-3beta-(para-halo-phenyl)nortropanes 9-12 were synthesized as ligands of the dopamine transporter (DAT) for use as (18)F-labeled positron emission tomography (PET) imaging agents. In vitro competition binding assays demonstrated that compounds 9-12 have a high affinity for the DAT and are selective for the DAT compared to the serotonin and norepinephrine transporters. MicroPET imaging with [(18)F]9-[(18)F]11 in anesthetized cynomolgus monkeys showed high uptake in the putamen with lesser uptake in the caudate, but significant washout of the radiotracer was only observed for [(18)F]9. PET imaging with [(18)F]9 in an awake rhesus monkey showed high and nearly equal uptake in both the putamen and caudate with peak uptake achieved after 20 min followed by a leveling-off for about 10 min and then a steady washout and attainment of a quasi-equilibrium. During the time period 40-80 min postinjection of [(18)F]9, the ratio of uptake in the putamen and caudate vs cerebellum uptake was > or = 4.
Collapse
|
25
|
Yatham LN, Liddle PF, Lam RW, Zis AP, Stoessl AJ, Sossi V, Adam MJ, Ruth TJ. Effect of electroconvulsive therapy on brain 5-HT(2) receptors in major depression. Br J Psychiatry 2010; 196:474-9. [PMID: 20513859 DOI: 10.1192/bjp.bp.109.069567] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Brain serotonin(2) (5-hydroxytryptamine(2); 5-HT(2)) receptors were considered potential targets for therapeutic efficacy of electroconvulsive therapy (ECT), but pre-clinical studies showed that electroconvulsive shock up-regulates 5-HT(2) receptors in contrast to antidepressant medications, which down-regulate brain 5-HT(2) receptors. Positron emission tomography (PET) studies in individuals with depression confirmed that antidepressant medications reduce brain 5-HT(2) receptors, but the effects of ECT on these receptors in individuals with depression are unknown. AIMS To determine if a course of ECT alters brain 5-HT(2) receptors in individuals with depression and whether such changes correlate with improvement in symptoms. METHOD Fifteen people with major depression, refractory to antidepressant therapy and referred for a course of ECT, had an [18F]setoperone scan during baseline drug-free washout period and another after a course of ECT. We assessed changes in brain 5-HT(2) receptors with ECT and their relationship to therapeutic outcome. RESULTS Widespread reduction in brain 5-HT(2) receptors was observed in all cortical areas with changes slightly more prominent in the right hemisphere. There was a trend for correlation between reduction in brain 5-HT(2) receptors in right parahippocampal gyrus, right lingual gyrus and right medial frontal gyrus, and improvement in depressive symptoms. CONCLUSIONS Unlike in rodents, and similar to antidepressants, ECT reduces brain 5-HT(2) receptors in individuals with depression. The ability of ECT to further down-regulate brain 5-HT(2) receptors in antidepressant non-responsive individuals may explain its efficacy in those people with antidepressant refractory depression.
Collapse
Affiliation(s)
- Lakshmi N Yatham
- UBC Department of Psychiatry, The University of British Columbia, UBC Hospital, 2255 Wesbrook Mall, Vancouver, BC V6T2A1, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Tantawy MN, Jones CK, Baldwin RM, Ansari MS, Conn PJ, Kessler RM, Peterson TE. [(18)F]Fallypride dopamine D2 receptor studies using delayed microPET scans and a modified Logan plot. Nucl Med Biol 2009; 36:931-40. [PMID: 19875049 DOI: 10.1016/j.nucmedbio.2009.06.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Revised: 06/02/2009] [Accepted: 06/22/2009] [Indexed: 10/20/2022]
Abstract
UNLABELLED [(18)F]Fallypride PET studies can be used to estimate the nondisplaceable binding potential (BP(ND)) in vivo of dopamine D2/D3 receptor-rich regions of the brain. These studies often take considerable time, up to >or=2 h, limiting the throughput. In this work, we investigated whether limited-duration scans performed subsequent to tracer administration yielded stable BP(ND) estimates. In particular, we applied a modified version of the Logan plot method on the last 60 min of 120-min data and compared the results to those from analysis of the full data set. METHODS Fourteen male Sprague-Dawley rats were injected with [(18)F]fallypride intravenously while under isoflurane anesthesia, and dynamic data were acquired on the microPET Focus 220 scanner for 120 min. The distribution volume ratio (DVR=BP(ND)+1) was calculated from a Logan plot using 120 min of data and from a modified version using only the last 60 min. Three of these rats were imaged again on a second day to test the reproducibility. A two-tissue compartment model also was used to fit the time-activity curves (TACs) of the 120-min scans to estimate the parameters K(1), k(2), k(on), k(4) and B(max). These parameters were then used to simulate similar TACs while changing k(on) to reflect changes in the dopaminergic system. The simulated TACs were used as a means for exploring the differences in DVR estimates between the last 60 min only and the full 120 min of simulated data. RESULTS The average DVR from the full 120-min scans was 13.8+/-0.9, whereas the average DVR estimated from only the last 60 min of data (DVR') was 16.3+/-1.0. The DVR estimates showed good reproducibility in the three rats (mean DVR=13.8+/-1.5 on Day 1 and DVR=13.8+/-0.9 on Day 2). The simulations showed that the relationship between DVR' and DVR estimates follows a semilinear form with varying k(on). CONCLUSION Although the BP(ND) estimates are slightly overestimated in a delayed scan mode (i.e., no initial radiotracer uptake measurements) compared to a full scan, this overestimation depends primarily on k(3) (approximately k(on) x B(max)) and has been evaluated in this work for a wide range of k(on) values using simulated TACs. In particular, the sensitivity of DVR' to changes in k(on) is similar to that of DVR. This method of delayed scans eliminates the necessity of imaging during the initial uptake of the radiotracer and, thus, can be used to increase the throughput of studies.
Collapse
Affiliation(s)
- Mohammed N Tantawy
- Department of Radiology and Radiological Sciences, Vanderbilt University Institute of Imaging Science, Nashville, TN 37232, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Christian BT, Fox AS, Oler JA, Vandehey NT, Murali D, Rogers J, Oakes TR, Shelton SE, Davidson RJ, Kalin NH. Serotonin transporter binding and genotype in the nonhuman primate brain using [C-11]DASB PET. Neuroimage 2009; 47:1230-6. [PMID: 19505582 DOI: 10.1016/j.neuroimage.2009.05.090] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Revised: 05/21/2009] [Accepted: 05/27/2009] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED The length polymorphism of the serotonin (5-HT) transporter gene promoter region has been implicated in altered 5-HT function and, in turn, neuropsychiatric illnesses, such as anxiety and depression. The nonhuman primate has been used as a model to study anxiety-related mechanisms in humans based upon similarities in behavior and the presence of a similar 5-HT transporter gene polymorphism. Stressful and threatening contexts in the nonhuman primate model have revealed 5-HT transporter genotype dependent differences in regional glucose metabolism. Using the rhesus monkey, we examined the extent to which serotonin transporter genotype is associated with 5-HT transporter binding in brain regions implicated in emotion-related pathology. METHODS Genotype data and high resolution PET scans were acquired in 29 rhesus (Macaca mulatta) monkeys. [C-11]DASB dynamic PET scans were acquired for 90 min in the anesthetized animals and images of distribution volume ratio (DVR) were created to serve as a metric of 5-HT transporter binding for group comparison based on a reference region method of analysis. Regional and voxelwise statistical analysis were performed with corrections for anatomical differences in gray matter probability, sex, age and radioligand mass. RESULTS There were no significant differences when comparing l/l homozygotes with s-carriers in the regions of the brain implicated in anxiety and mood related illnesses (amygdala, striatum, thalamus, raphe nuclei, temporal and prefrontal cortex). There was a significant sex difference in 5-HT transporter binding in all regions with females having 18%-28% higher DVR than males. CONCLUSIONS Because these findings are consistent with similar genotype findings in humans, this further strengthens the use of the rhesus model for studying anxiety-related neuropathologies.
Collapse
Affiliation(s)
- B T Christian
- Department of Psychiatry, Harlow Primate Center, University of Wisconsin-Madison, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Tokunaga M, Seneca N, Shin RM, Maeda J, Obayashi S, Okauchi T, Nagai Y, Zhang MR, Nakao R, Ito H, Innis RB, Halldin C, Suzuki K, Higuchi M, Suhara T. Neuroimaging and physiological evidence for involvement of glutamatergic transmission in regulation of the striatal dopaminergic system. J Neurosci 2009; 29:1887-96. [PMID: 19211895 PMCID: PMC2746446 DOI: 10.1523/jneurosci.2559-08.2009] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2008] [Revised: 11/30/2008] [Accepted: 12/27/2008] [Indexed: 11/21/2022] Open
Abstract
Aberrant neurotransmissions via glutamate and dopamine receptors have been the focus of biomedical research on the molecular basis of psychiatric disorders, but the mode of their interaction is yet to be uncovered. In this study, we demonstrated the pharmacological reversal of methamphetamine-stimulated dopaminergic overflow by suppression of group I metabotropic glutamate (mGlu) receptor in living primates and rodents. In vivo positron emission tomography (PET) was conducted on cynomolgus monkeys and rats using a full agonistic tracer for dopamine D(2/3) receptor, [(11)C]MNPA [(R)-2-(11)CH(3)O-N-n-propylnorapomorphine], and fluctuation of kinetic data resulting from anesthesia was avoided by scanning awake subjects. Excessive release of dopamine induced by methamphetamine and abolishment of this alteration by treatment with an antagonist of group I mGlu receptors, 2-methyl-6-(phenylethynyl)pyridine (MPEP), were measured in both species as decreased binding potential because of increased dopamine and its recovery to baseline levels, respectively. Counteraction of MPEP to the methamphetamine-induced dopamine spillover was also supported neurochemically by microdialysis of unanesthetized rat striatum. Moreover, patch-clamp electrophysiological assays using acute brain slices prepared from rats indicated that direct targets of MPEP mechanistically involved in the effects of methamphetamine are present locally within the striatum. Because MPEP alone did not markedly alter the baseline dopaminergic neurotransmission according to our PET and electrophysiological data, the present findings collectively extend the insights on dopamine-glutamate cross talk from extrastriatal localization of responsible mGlu receptors to intrastriatal synergy and support therapeutic interventions in case of disordered striatal dopaminergic status using group I mGlu receptor antagonists assessable by in vivo imaging techniques.
Collapse
Affiliation(s)
- Masaki Tokunaga
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Nicholas Seneca
- Molecular Imaging Branch, National Institute of Mental Health–National Institutes of Health, Bethesda, Maryland 20892, and
| | - Ryong-Moon Shin
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Jun Maeda
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Shigeru Obayashi
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Takashi Okauchi
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Yuji Nagai
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Ming-Rong Zhang
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Ryuji Nakao
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Hiroshi Ito
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Robert B. Innis
- Molecular Imaging Branch, National Institute of Mental Health–National Institutes of Health, Bethesda, Maryland 20892, and
| | - Christer Halldin
- Department of Clinical Neuroscience, Psychiatry Section, Kalorinska Institute, S-17176 Stockholm, Sweden
| | - Kazutoshi Suzuki
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Makoto Higuchi
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Tetsuya Suhara
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| |
Collapse
|
29
|
Stehouwer JS, Jarkas N, Zeng F, Voll RJ, Williams L, Camp VM, Malveaux EJ, Votaw JR, Howell L, J.Owens M, Goodman MM. Synthesis, radiosynthesis, and biological evaluation of fluorine-18-labeled 2beta-carbo(fluoroalkoxy)-3beta-(3'-((Z)-2-haloethenyl)phenyl)nortropanes: candidate radioligands for in vivo imaging of the serotonin transporter with positron emission tomography. J Med Chem 2008; 51:7788-99. [PMID: 19053782 PMCID: PMC2668213 DOI: 10.1021/jm800781a] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The meta-vinylhalide fluoroalkyl ester nortropanes 1-4 were synthesized as ligands of the serotonin transporter (SERT) for use as positron emission tomography (PET) imaging agents. In vitro competition binding assays demonstrated that 1-4 have a high affinity for the SERT (K(i) values = 0.3-0.4 nM) and are selective for the SERT over the dopamine and norepinephrine transporters (DAT and NET). MicroPET imaging in anesthetized cynomolgus monkeys with [(18)F]1-[(18)F]4 demonstrated that all four tracers behave similarly with peak uptake in the SERT-rich brain regions achieved after 45-55 min, followed by a steady washout. An awake monkey study was performed with [(18)F]1, which demonstrated that the uptake of [(18)F]1 was not influenced by anesthesia. Chase studies with the SERT ligand 15 displaced [(18)F]1-[(18)F]4, but chase studies with the DAT ligand 16 did not displace [(18)F]1-[(18)F]4 thus indicating that the tracers were binding specifically to the SERT.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Mark M. Goodman
- To whom correspondence should be addressed. Department of Radiology, Emory University, 1364 Clifton Road NE, Atlanta, GA 30322 Phone: (404) 727-9366. Fax: (404) 727-3488. E-mail:
| |
Collapse
|
30
|
Syvänen S, Lindhe O, Palner M, Kornum BR, Rahman O, Långström B, Knudsen GM, Hammarlund-Udenaes M. Species differences in blood-brain barrier transport of three positron emission tomography radioligands with emphasis on P-glycoprotein transport. Drug Metab Dispos 2008; 37:635-43. [PMID: 19047468 DOI: 10.1124/dmd.108.024745] [Citation(s) in RCA: 251] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Species differences occur in the brain concentrations of drugs, but the reasons for these differences are not yet apparent. This study was designed to compare brain uptake of three radiolabeled P-glycoprotein (P-gp) substrates across species using positron emission tomography. Brain concentrations and brain-to-plasma ratios were compared; [(11)C]verapamil in rats, guinea pigs, and monkeys; [(11)C](S)-(2-methoxy-5-(5-trifluoromethyltetrazol-1-yl)-phenylmethylamino)-2(S)-phenylpiperidine (GR205171) in rats, guinea pigs, monkeys, and humans; and [(18)F]altanserin in rats, minipigs, and humans. The fraction of the unbound radioligand in plasma was studied along with its metabolism. The effect of P-gp inhibition was investigated by administering cyclosporin A (CsA). Pronounced species differences were found in the brain and brain-to-plasma concentrations of [(11)C]verapamil, [(11)C]GR205171, and [(18)F]altanserin with higher brain distribution in humans, monkeys, and minipigs than in rats and guinea pigs. For example, the brain-to-plasma ratio of [(11)C]GR205171 was almost 9-fold higher in humans compared with rats. The species differences were still present after P-gp inhibition, although the increase in brain concentrations after P-gp inhibition was somewhat greater in rats than in the other species. Differences in plasma protein binding and metabolism did not explain the species-related differences. The findings are important for interpretation of brain drug delivery when extrapolating preclinical data to humans. Compounds found to be P-gp substrates in rodents are likely to also be substrates in higher species, but sufficient blood-brain barrier permeability may be retained in humans to allow the compound to act at intracerebral targets.
Collapse
Affiliation(s)
- Stina Syvänen
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Evaluation of serotonin-2A receptor occupancy with 123I-5-I-R91150 and single-photon emission tomography before and after low-dose pipamperone administration in the canine brain. Nucl Med Commun 2008; 29:724-9. [DOI: 10.1097/mnm.0b013e3282fdc989] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
Emond P, Guilloteau D, Chalon S. PE2I: a radiopharmaceutical for in vivo exploration of the dopamine transporter. CNS Neurosci Ther 2008; 14:47-64. [PMID: 18482099 DOI: 10.1111/j.1527-3458.2007.00033.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The membrane dopamine transporter (DAT) has a pivotal role in the regulation of dopamine (DA) neurotransmission involved in a number of physiological functions and brain disorders. Molecular imaging techniques, such as positron emission tomography (PET) and single photon emission computerized tomography (SPECT), are relevant tools to explore the DAT, and we developed the cocaine derivative N-(3-iodopro-2E-enyl)-2beta-carbomethoxy-3beta-(4'-methylphenyl) nortropane (PE2I) that has proved to be a very potent radiopharmaceutical to image the DAT by these techniques. Several methods are available to obtain PE2I labeled with iodine-123 or -125, carbon-11 and tritium. The pharmacological properties of PE2I have demonstrated that it has good affinity for the DAT (4 nM) and is one of the most selective DAT ligands. [(125)I]PE2I characterized postmortem in human brains has revealed very intense and selective binding in the basal ganglia. Ex vivo autoradiography in rats has shown that high level of [(125)I]PE2I accumulates in the striatum and also in the substantia nigra and ventral tegmental area. [(125)I]PE2I accumulation in the rat striatum is rapid, high, and selective, providing a maximum striatum/cerebellum ratio of 10 during the first 30 min post injection. Using SPECT or PET, rapid, high, and selective accumulation of PE2I was found in the caudate nucleus and putamen in monkeys, whereas rapid wash out from the cerebellum was observed. In vivo investigations in healthy humans have demonstrated that PE2I has high striatal uptake, low nonspecific binding, low radiation exposure, and a fairly short scanning time. A number of findings in various animal models of Parkinson's disease in rats and monkeys have demonstrated the high efficacy of PE2I for detection of reduction in the density of DAT, thus showing the potential value of PE2I for early diagnosis and evaluation of treatment of this disease. The excellent properties of PE2I are basis for the development of new DAT tracers for use in future PET explorations using fluor-18.
Collapse
|
33
|
Imaging dopamine release with Positron Emission Tomography (PET) and 11C-raclopride in freely moving animals. Neuroimage 2008; 41:1051-66. [DOI: 10.1016/j.neuroimage.2008.02.065] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Revised: 02/16/2008] [Accepted: 02/29/2008] [Indexed: 11/19/2022] Open
|
34
|
Emond P, Guilloteau D, Chalon S. PE2I: A Radiopharmaceutical for In vivo Exploration of the Dopamine Transporter. CNS Neurosci Ther 2008. [DOI: 10.1111/j.1755-5949.2007.00033.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
35
|
Matusch A, Hurlemann R, Rota Kops E, Winz OH, Elmenhorst D, Herzog H, Zilles K, Bauer A. Acute S-ketamine application does not alter cerebral [18F]altanserin binding: a pilot PET study in humans. J Neural Transm (Vienna) 2007; 114:1433-42. [PMID: 17541696 DOI: 10.1007/s00702-007-0751-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2007] [Accepted: 04/19/2007] [Indexed: 10/23/2022]
Abstract
Modeling short-term psychotic states with subanaesthetic doses of ketamine provides substantial experimental evidence in support of the glutamate hypothesis of schizophrenia. Ketamine exerts its pharmacological effects both directly via interactions with glutamate receptors and indirectly by stimulating presynaptic release of endogenous serotonin (5-HT). The aim of this feasibility study was to examine whether acute ketamine-induced 5-HT release interferes with the binding of the 5-HT(2A) receptor (5-HT(2A)R) radioligand [(18)F]altanserin and positron emission tomography (PET). Two subjects treated with ketamine and one subject treated with placebo underwent [(18)F]altanserin PET at distribution equilibrium conditions. Robust physiological, psychopathological and cognitive effects were present at ketamine plasma concentrations exceeding 100 microg/l during >70 min. Notwithstanding, we observed stable radioligand binding (changes +/-95% CI of -1.0 +/- 1.6% and +4.1 +/- 1.8% versus -1.2 +/- 2.6%) in large cortical regions presenting high basal uptake of both, [(18)F]altanserin and ketamine. Marginal decreases of 4% of radioligand binding were observed in the frontal lobe, and 8% in a posteriorily specified frontomesial subregion. This finding is not compatible with a specific radioligand displacement from 5-HT(2A)R which should occur proportionally throughout the whole brain. Instead, the spatial pattern of these minor reductions was congruent with ketamine-induced increases in cerebral blood flow observed in a previous study using [(15)O]butanol PET. This may caused by accelerated clearance of unspecifically bound [(18)F]altanserin from cerebral tissue with increased perfusion. In conclusion, this study suggests that [(18)F]altanserin PET is not sensitive to acute neurotransmitter fluctuations under ketamine. Advantageously, the stability of [(18)F]altanserin PET towards acute influences is a prerequisite for its future use to detect sub-acute and chronic effects of ketamine.
Collapse
Affiliation(s)
- A Matusch
- Institute of Medicine and Brain Imaging Center West, Research Center Juelich, Juelich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Peremans K, Goethals I, De Vos F, Dobbeleir A, Ham H, Van Bree H, Van Heeringen C, Audenaert K. Serotonin transporter and dopamine transporter imaging in the canine brain. Nucl Med Biol 2007; 33:907-13. [PMID: 17045171 DOI: 10.1016/j.nucmedbio.2006.07.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2006] [Revised: 07/17/2006] [Accepted: 07/24/2006] [Indexed: 11/16/2022]
Abstract
The serotonergic and dopaminergic systems are involved in a wide range of emotional and behavioral aspects of animals and humans and are involved in many neuropsychiatric disorders. Selective serotonin (5-HT) reuptake inhibitors (SSRIs) are designed to block the 5-HT transporter (SERT), thereby increasing the available 5-HT in the brain. Functional imaging with specific SERT and dopamine transporter (DAT) ligands contributes to the study of the SSRI-transporter interaction. First, we evaluated the feasibility of a canine model in the study of the SERT and DAT with the radioligands [123I]-beta-CIT and [123I]-FP-CIT as well as single-photon emission computed tomography imaging. Second, we studied the effect of SSRIs (sertraline, citalopram and escitalopram) on the SERT and DAT in two dogs. The position of the canine model in the study of the SERT and DAT is discussed and compared with other animal models.
Collapse
Affiliation(s)
- Kathelijne Peremans
- Department of Medical Imaging, Faculty of Veterinary Sciences, Ghent University, B-9000 Ghent, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Honer M, Hengerer B, Blagoev M, Hintermann S, Waldmeier P, Schubiger PA, Ametamey SM. Comparison of [18F]FDOPA, [18F]FMT and [18F]FECNT for imaging dopaminergic neurotransmission in mice. Nucl Med Biol 2006; 33:607-14. [PMID: 16843835 DOI: 10.1016/j.nucmedbio.2006.04.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2005] [Revised: 04/03/2006] [Accepted: 04/11/2006] [Indexed: 11/23/2022]
Abstract
INTRODUCTION The clinically established positron emission tomography (PET) tracers 6-[(18)F]-fluoro-l-DOPA ([(18)F]FDOPA), 6-[(18)F]-fluoro-l-m-tyrosine ([(18)F]FMT) and 2beta-carbomethoxy-3beta-(4-chlorophenyl)-8-(2-[(18)F]-fluoroethyl)-nortropane ([(18)F]FECNT) serve as markers of presynaptic integrity of dopaminergic nerve terminals in humans. This study describes our efforts to adopt the methodology of human Parkinson's disease (PD) PET studies to mice. METHODS The PET imaging characteristics of [(18)F]FDOPA, [(18)F]FMT and [(18)F]FECNT were analyzed in healthy C57BL/6 mice using the dedicated small-animal PET tomograph quad-HIDAC. Furthermore, [(18)F]FECNT was tested in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. RESULTS [(18)F]FDOPA and [(18)F]FMT failed to clearly visualize the mouse striatum, whereas PET experiments using [(18)F]FECNT proved that the employed methodology is capable of delineating the striatum in mice with exquisite resolution. Moreover, [(18)F]FECNT PET imaging of healthy and MPTP-lesioned mice demonstrated that the detection and quantification of striatal degeneration in lesioned mice can be accomplished. CONCLUSIONS This study shows the feasibility of using [(18)F]FECNT PET to analyze noninvasively the striatal degeneration in the MPTP mouse model of PD. This methodology can be therefore considered as a viable complement to established in vivo microdialysis and postmortem techniques.
Collapse
Affiliation(s)
- Michael Honer
- Center for Radiopharmaceutical Science of ETH, PSI and USZ, CH-8093 Zurich, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
38
|
Booij J, de Bruin K, Gunning WB. Repeated administration of d-amphetamine induces loss of [123I]FP-CIT binding to striatal dopamine transporters in rat brain: a validation study. Nucl Med Biol 2006; 33:409-11. [PMID: 16631090 DOI: 10.1016/j.nucmedbio.2005.12.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2005] [Revised: 11/15/2005] [Accepted: 12/09/2005] [Indexed: 10/24/2022]
Abstract
UNLABELLED In recent years, several PET and SPECT studies have shown loss of striatal dopamine transporter (DAT) binding in amphetamine (AMPH) users. However, the use of DAT SPECT tracers to detect AMPH-induced changes in DAT binding has not been validated. We therefore examined if repeated administration of D-AMPH or methamphetamine (METH) may induce loss of binding to striatal DATs in rats by using an experimental biodistribution study design and a SPECT tracer for the DAT ([123I]FP-CIT). METHODS Groups of male rats (n = 10 per group) were treated with D-AMPH (10 mg/kg body weight), METH (10 mg/kg body weight), or saline, twice a day for 5 consecutive days. Five days later, [123I]FP-CIT was injected intravenously, and 2 h later, the rats were sacrificed and radioactivity was assayed. RESULTS In d-AMPH but not METH-treated rats, striatal [123I]FP-CIT uptake was significantly lower (approximately 17%) than in the control group. CONCLUSION These data show that [123I]FP-CIT can be used to detect AMPH-induced changes in DAT binding and may validate the use of DAT radiotracers to study AMPH-induced changes in striatal DAT binding in vivo.
Collapse
Affiliation(s)
- Jan Booij
- Department of Nuclear Medicine, Academic Medical Center, 1105 AZ Amsterdam, The Netherlands.
| | | | | |
Collapse
|
39
|
Hosoi R, Matsumura A, Mizokawa S, Tanaka M, Nakamura F, Kobayashi K, Watanabe Y, Inoue O. MicroPET detection of enhanced 18F-FDG utilization by PKA inhibitor in awake rat brain. Brain Res 2005; 1039:199-202. [PMID: 15781062 DOI: 10.1016/j.brainres.2005.01.064] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2004] [Revised: 01/12/2005] [Accepted: 01/12/2005] [Indexed: 11/25/2022]
Abstract
To obtain PET imaging of glucose metabolism in the brains of conscious rats, a method of rat head fixation was developed. PET measurement with microPET was performed for 60 min after 18F-FDG injection. Significant enhancement of glucose utilization in the right striatum was observed with infusion of Rp-adenosine-3,5-cyclic phosphorothioate triethylamine (Rp-cAMPS). FDG uptake increments were also seen in the ipsilateral frontal cortex and thalamus. As initial FDG uptake in the brain was not significantly altered by Rp-cAMPS, increased glucose metabolism might be due to an increase in the phosphorylation rate by hexokinase rather than the delivery process from plasma to the brain. In contrast to awake rats, the effect of Rp-cAMPS was abolished by anesthesia using chloral hydrate, indicating that neuronal activity has an important role in short term regulation of hexokinase activity through the cAMP/PKA system in the brain. These results strongly demonstrated the value of measuring glucose utilization in the brains of conscious rats.
Collapse
Affiliation(s)
- Rie Hosoi
- Course of Allied Health Sciences, Graduate School of Medicine, Osaka University, 1-7 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Datla KP, Zbarsky V, Dexter DT. Effects of anaesthetics on the loss of nigrostriatal dopaminergic neurons by 6-hydroxydopamine in rats. J Neural Transm (Vienna) 2005; 113:583-91. [PMID: 16082506 DOI: 10.1007/s00702-005-0353-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2005] [Accepted: 06/25/2005] [Indexed: 12/21/2022]
Abstract
Various studies use ketamine/xylazine, fentanyl/medetomidine, etorphine/methotrimeprazine, and isoflurane anaesthesia for creating the 6-hydroxydopamine (6-OHDA)-lesion rat model of Parkinson's disease. As these anaesthetics are known to modulate uptake and turnover of dopamine and that 6-OHDA-induced neurotoxicity is also dependents on uptake/turnover, we studied the effects of these anaesthetics on the extent of nigrostriatal dopaminergic damage caused by 6-OHDA. Infusion of 8 microg of 6-OHDA into the medial forebrain bundle significantly reduced the numbers of dopaminergic cells in nigra and striatal concentrations of dopamine in animals anaesthetized with fentanyl/medetomidine, etorphine/methotrimeprazine and isoflurane but not with ketamine/xylazine. In the latter group, however, increasing the dose of 6-OHDA to 10 and 12 microg resulted in a moderate (15 and 29%), but significant loss of dopaminergic cells. A severe loss of dopaminergic cells (59% and 81%) was seen with these doses in isoflurane-anaesthetized animals, but with only 8 microg in etorphine/methotrimeprazine-anaesthetized animals. Thus, these results suggest that the extent of nigrostriatal dopaminergic neuronal loss with 6-OHDA seems to be influenced by anaesthetic used during the surgery.
Collapse
Affiliation(s)
- K P Datla
- Department of Cellular and Molecular Neuroscience, Division of Neuroscience and Mental Health, Faculty of Medicine, Imperial College London, Charing Cross Campus, London, United Kingdom
| | | | | |
Collapse
|
41
|
Momosaki S, Hatano K, Kawasumi Y, Kato T, Hosoi R, Kobayashi K, Inoue O, Ito K. Rat-PET study without anesthesia: Anesthetics modify the dopamine D1 receptor binding in rat brain. Synapse 2004; 54:207-13. [PMID: 15476291 DOI: 10.1002/syn.20083] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Positron emission tomography (PET) measurements in 6-month-old F344/N rats were performed in the conscious state and the influence of chloral hydrate, ketamine, and pentobarbital anesthesia on dopamine D(1) (DA-D(1)) receptor binding was evaluated using [(11)C]SCH23390, a selective DA-D(1) receptor ligand. To perform the PET study in conscious rats, an original fixation apparatus was developed and the animals were trained to acclimate to the scanning atmosphere for 3 h. This training was carried out twice a day for 2 weeks. PET measurements in conscious rats were successful, since the trained rats scarcely moved during the scanning (as monitored by video camera) and since highly reproducible measurements of binding potential (BP) were derived from their scanning. Chloral hydrate and ketamine anesthesia significantly increased the striatal BP of DA-D(1) receptors by 36% and 46%, respectively, compared to that observed in the conscious state. In contrast, pentobarbital markedly decreased the BP by 41%. These BP values of DA-D(1) receptors were calculated using a curve-fitting method. Our results indicate that PET studies in rats should be performed in the conscious state since the anesthetics dramatically modified ligand-receptor bindings, such as DA-D(1) receptor binding, in rat brain.
Collapse
Affiliation(s)
- Sotaro Momosaki
- Department of Brain Science and Molecular Imaging, National Institute for Longevity Sciences, Aichi 474-8522, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Nikolaus S, Beu M, Wirrwar A, Vosberg H, Müller HW, Larisch R. The Contribution of Small Animal Positron Emission Tomography to the Neurosciences - A Critical Evaluation. Rev Neurosci 2004; 15:131-56. [PMID: 15202685 DOI: 10.1515/revneuro.2004.15.2.131] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
This article presents an overview of those animal studies which so far have been performed with dedicated small animal positron emission tomographs in the field of the neurosciences. In vivo investigations focus on energy metabolism, perfusion and receptor/transporter binding in rat models of reinforcement, learning and memory, traumatic brain injury, epilepsy, depression, cardiovascular diseases--such as ischemia and focal stroke--and neurodegenerative disorders such as Alzheimer's, Parkinson's and Huntington's disease. In the majority of studies, important novel aspects arise from the fact that the investigators made use of an option inherent to in vivo studies, namely to conduct longitudinal investigations on the same animals. Relevant findings pertain to the relationship of brain metabolism/perfusion and the cholinergic system, the regulation state of dopamine receptors upon cocaine administration and withdrawal, the regulation state of dopamine receptors and transporters in animal models of Parkinson's and Huntington's disease, and potential treatments of progressive dopaminergic depletion with adenoviral vectors, embryonic grafts, stem cells and nerve growth factors.
Collapse
Affiliation(s)
- Susanne Nikolaus
- Clinic of Nuclear Medicine, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
Molecular imaging of small animals in vivo is vital in the study of mouse and rat models of human diseases, and will provide important clues to the pathogenesis, progression and treatment of many disorders. Functional imaging of small animals using ultra-high resolution single photon emission tomography (SPECT) should be a valuable tool in the molecular imaging armamentarium. SPECT has been used to study cerebral binding sites, to image the expression of reporter genes, and in applications in cardiology and oncology. In this review, we summarize the most recent developments in SPECT imaging of small animals, with particular reference to the types of systems available, their application, and some of the potential limitations.
Collapse
Affiliation(s)
- Paul D Acton
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA.
| | | |
Collapse
|