Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Brain ; 138(Pt 10): 3016-29, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26198591

ABSTRACT

There is an urgent need for early biomarkers and novel disease-modifying therapies in Huntington's disease. Huntington's disease pathology involves the toxic effect of mutant huntingtin primarily in striatal medium spiny neurons, which highly express phosphodiesterase 10A (PDE10A). PDE10A hydrolyses cAMP/cGMP signalling cascades, thus having a key role in the regulation of striatal output, and in promoting neuronal survival. PDE10A could be a key therapeutic target in Huntington's disease. Here, we used combined positron emission tomography (PET) and multimodal magnetic resonance imaging to assess PDE10A expression in vivo in a unique cohort of 12 early premanifest Huntington's disease gene carriers with a mean estimated 90% probability of 25 years before the predicted onset of clinical symptoms. We show bidirectional changes in PDE10A expression in premanifest Huntington's disease gene carriers, which are associated with the probability of symptomatic onset. PDE10A expression in early premanifest Huntington's disease was decreased in striatum and pallidum and increased in motor thalamic nuclei, compared to a group of matched healthy controls. Connectivity-based analysis revealed prominent PDE10A decreases confined in the sensorimotor-striatum and in striatonigral and striatopallidal projecting segments. The ratio between higher PDE10A expression in motor thalamic nuclei and lower PDE10A expression in striatopallidal projecting striatum was the strongest correlate with higher probability of symptomatic conversion in early premanifest Huntington's disease gene carriers. Our findings demonstrate in vivo, a novel and earliest pathophysiological mechanism underlying Huntington's disease with direct implications for the development of new pharmacological treatments, which can promote neuronal survival and improve outcome in Huntington's disease gene carriers.


Subject(s)
Gene Expression Regulation, Enzymologic , Huntington Disease/enzymology , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Signal Transduction/physiology , Adolescent , Adult , Brain Mapping , Female , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/physiopathology , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Nerve Tissue Proteins/genetics , Positron-Emission Tomography , Quinoxalines/pharmacokinetics , Severity of Illness Index , Signal Transduction/genetics , Terminal Repeat Sequences/genetics , Young Adult
2.
Brain ; 138(Pt 10): 3003-15, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26210536

ABSTRACT

The mechanisms underlying neurodegeneration and loss of dopaminergic signalling in Parkinson's disease are still only partially understood. Phosphodiesterase 10A (PDE10A) is a basal ganglia expressed dual substrate enzyme, which regulates cAMP and cGMP signalling cascades, thus having a key role in the regulation of dopaminergic signalling in striatal pathways, and in promoting neuronal survival. This study aimed to assess in vivo the availability of PDE10A in patients with Parkinson's disease using positron emission tomography molecular imaging with (11)C-IMA107, a highly selective PDE10A radioligand. We studied 24 patients with levodopa-treated, moderate to advanced Parkinson's disease. Their positron emission tomography imaging data were compared to those from a group of 12 healthy controls. Parametric images of (11)C-IMA107 binding potential relative to non-displaceable binding (BPND) were generated from the dynamic (11)C-IMA107 scans using the simplified reference tissue model with the cerebellum as the reference tissue. Corresponding region of interest analysis showed lower mean (11)C-IMA107 BPND in the caudate (P < 0.001), putamen (P < 0.001) and globus pallidus (P = 0.025) in patients with Parkinson's disease compared to healthy controls, which was confirmed with voxel-based analysis. Longer Parkinson's duration correlated with lower (11)C-IMA107 BPND in the caudate (r = -0.65; P = 0.005), putamen (r = -0.51; P = 0.025), and globus pallidus (r = -0.47; P = 0.030). Higher Unified Parkinson's Disease Rating Scale part-III motor scores correlated with lower (11)C-IMA107 BPND in the caudate (r = -0.54; P = 0.011), putamen (r = -0.48; P = 0.022), and globus pallidus (r = -0.70; P < 0.001). Higher Unified Dyskinesia Rating Scale scores in those Parkinson's disease with levodopa-induced dyskinesias (n = 12), correlated with lower (11)C-IMA107 BPND in the caudate (r = -0.73; P = 0.031) and putamen (r = -0.74; P = 0.031). Our findings demonstrate striatal and pallidal loss of PDE10A expression, which is associated with Parkinson's duration and severity of motor symptoms and complications. PDE10A is an enzyme that could be targeted with novel pharmacotherapy, and this may help improve dopaminergic signalling and striatal output, and therefore alleviate symptoms and complications of Parkinson's disease.


Subject(s)
Brain/pathology , Gene Expression Regulation, Enzymologic , Parkinson Disease/diagnosis , Parkinson Disease/enzymology , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Aged , Brain/diagnostic imaging , Brain Mapping , Disease Progression , Female , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Middle Aged , Motor Activity , Multivariate Analysis , Parkinson Disease/diagnostic imaging , Parkinson Disease/genetics , Positron-Emission Tomography , Quinoxalines/pharmacokinetics , Severity of Illness Index , Statistics as Topic
3.
Neuropsychopharmacology ; 39(7): 1703-12, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24469594

ABSTRACT

Animal studies support the role of the dopamine D3 receptor (DRD3) in alcohol reinforcement or liking. Sustained voluntary alcohol drinking in rats has been associated with an upregulation of striatal DRD3 gene expression and selective blockade of DRD3 reduces ethanol preference, consumption, and cue-induced reinstatement. In vivo measurement of DRD3 in the living human brain has not been possible until recently owing to a lack of suitable tools. In this study, DRD3 status was assessed for the first time in human alcohol addiction. Brain DRD3 availability was compared between 16 male abstinent alcohol-dependent patients and 13 healthy non-dependent age-matched males using the DRD3-preferring agonist positron emission tomography (PET) radioligand [(11)C]PHNO with and without blockade with a selective DRD3 antagonist (GSK598809 60 mg p.o.). In striatal regions of interest, where the [(11)C]PHNO PET signal represents primarily DRD2 binding, no differences were seen in [(11)C]PHNO binding between the groups at baseline. However, baseline [(11)C]PHNO binding was higher in alcohol-dependent patients in hypothalamus (VT: 16.5 ± 4 vs 13.7 ± 2.9, p = 0.040), a region in which the [(11)C]PHNO signal almost entirely reflects DRD3 availability. The reductions in regional receptor binding (VT) following a single oral dose of GSK598809 (60 mg) were consistent with those observed in previous studies across all regions. There were no differences in regional changes in VT following DRD3 blockade between the two groups, indicating that the regional fractions of DRD3 are similar in the two groups, and the increased [(11)C]PHNO binding in the hypothalamus in alcohol-dependent patients is explained by elevated DRD3 in this group. Although we found no difference between alcohol-dependent patients and controls in striatal DRD3 levels, increased DRD3 binding in the hypothalamus of alcohol-dependent patients was observed. This may be relevant to the development of future therapeutic strategies to treat alcohol abuse.


Subject(s)
Alcoholism/pathology , Cerebral Cortex/metabolism , Oxazines , Receptors, Dopamine D3/metabolism , Adult , Age Factors , Alcoholism/blood , Alcoholism/diagnostic imaging , Azabicyclo Compounds/pharmacology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/drug effects , Dopamine Antagonists/blood , Dopamine Antagonists/pharmacology , Female , Humans , Male , Middle Aged , Oxazoles/pharmacology , Positron-Emission Tomography , Protein Binding/drug effects , Psychiatric Status Rating Scales , Receptors, Dopamine D3/antagonists & inhibitors , Smoking , Young Adult
4.
Neuroimage ; 84: 225-35, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23994455

ABSTRACT

In dynamic positron emission tomography (PET) neuroimaging studies, where scan durations often exceed 1h, registration of motion-corrupted dynamic PET images is necessary in order to maintain the integrity of the physiological, pharmacological, or biochemical information derived from the tracer kinetic analysis of the scan. In this work, we incorporate a pharmacokinetic model, which is traditionally used to analyse PET data following any registration, into the registration process itself in order to allow for a groupwise registration of the temporal time frames. The new method is shown to achieve smaller registration errors and improved kinetic parameter estimates on validation data sets when compared with image similarity based registration approaches. When applied to measured clinical data from 10 healthy subjects scanned with [(11)C]-(+)-PHNO (a dopamine D3/D2 receptor tracer), it reduces the intra-class variability on the receptor binding outcome measure, further supporting the improvements in registration accuracy. Our method incorporates a generic tracer kinetic model which makes it applicable to different PET radiotracers to remove motion artefacts and increase the integrity of dynamic PET studies.


Subject(s)
Brain/metabolism , Imaging, Three-Dimensional/methods , Models, Neurological , Oxazines/pharmacokinetics , Positron-Emission Tomography/methods , Receptors, Dopamine D3/metabolism , Subtraction Technique , Algorithms , Brain/diagnostic imaging , Carbon Isotopes/pharmacokinetics , Computer Simulation , Female , Humans , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Male , Neuroimaging/methods , Radiopharmaceuticals/pharmacokinetics , Receptors, Dopamine D3/antagonists & inhibitors , Reproducibility of Results , Sensitivity and Specificity , Spatio-Temporal Analysis , Time Factors , Young Adult
5.
Cereb Cortex ; 24(5): 1165-77, 2014 May.
Article in English | MEDLINE | ID: mdl-23283687

ABSTRACT

The striatum acts in conjunction with the cortex to control and execute functions that are impaired by abnormal dopamine neurotransmission in disorders such as Parkinson's and schizophrenia. To date, in vivo quantification of striatal dopamine has been restricted to structure-based striatal subdivisions. Here, we present a multimodal imaging approach that quantifies the endogenous dopamine release following the administration of d-amphetamine in the functional subdivisions of the striatum of healthy humans with [(11)C]PHNO and [(11)C]Raclopride positron emission tomography ligands. Using connectivity-based (CB) parcellation, we subdivided the striatum into functional subregions based on striato-cortical anatomical connectivity information derived from diffusion magnetic resonance imaging (MRI) and probabilistic tractography. Our parcellation showed that the functional organization of the striatum was spatially coherent across individuals, congruent with primate data and previous diffusion MRI studies, with distinctive and overlapping networks. d-amphetamine induced the highest dopamine release in the limbic followed by the sensory, motor, and executive areas. The data suggest that the relative regional proportions of D2-like receptors are unlikely to be responsible for this regional dopamine release pattern. Notably, the homogeneity of dopamine release was significantly higher within the CB functional subdivisions in comparison with the structural subdivisions. These results support an association between local levels of dopamine release and cortical connectivity fingerprints.


Subject(s)
Corpus Striatum/diagnostic imaging , Corpus Striatum/metabolism , Diffusion Tensor Imaging , Dopamine/metabolism , Nerve Net/diagnostic imaging , Positron-Emission Tomography , Adult , Brain Mapping , Corpus Striatum/drug effects , Dopamine Antagonists/pharmacokinetics , Dopamine Antagonists/pharmacology , Executive Function/physiology , Humans , Male , Middle Aged , Nerve Net/drug effects , Nerve Net/metabolism , Probability , Raclopride/pharmacokinetics , Raclopride/pharmacology
6.
Neuroimage ; 68: 119-32, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23207573

ABSTRACT

The D(2)/D(3) agonist radioligand [(11)C]-(+)-PHNO is currently the most suitable D(3) imaging agent available, despite its limited selectivity for the D(3) over the D(2). Given the collocation of D(2) and D(3) receptors, and generally higher densities of D(2), the separation of D(2) and D(3) information from [(11)C]-(+)-PHNO PET data are somewhat complex. This complexity is compounded by recent data suggesting that [(11)C]-(+)-PHNO PET scans might be routinely performed in non-tracer conditions (with respect to D(3) receptors), and that the cerebellum (used as a reference region) might manifest some displaceable binding signal. Here we present the modelling and analysis of data from two human studies which employed an adequate dose range of selective D(3) antagonists (GSK598809 and GSK618334) to interrogate the [(11)C]-(+)-PHNO PET signal. Models describing the changes observed in the PET volume of distribution (V(T)) and binding potential (BP(ND)) were used to identify and quantify a [(11)C]-(+)-PHNO mass dose effect at the D(3), and displaceable signal in the cerebellum, as well as providing refined estimates of regional D(3) fractions of [(11)C]-(+)-PHNO BP(ND). The dose of (+)-PHNO required to occupy half of the available D(3) receptors (ED(50)(PHNO,D3)) was estimated as 40ng/kg, and the cerebellum BP(ND) was estimated as 0.40. These findings confirm that [(11)C]-(+)-PHNO human PET studies are in fact routinely performed under non-tracer conditions. This suggests that (+)-PHNO injection masses should be minimised and tightly controlled in order to mitigate the mass dose effect. The specific binding detected in the cerebellum was modest but could have a significant effect, for example on estimates of D(3) potency in drug occupancy studies. A range of methods for the analysis of future [(11)C]-(+)-PHNO data, incorporating models for the effects quantified here, were developed and evaluated. The comparisons and conclusions drawn from these can inform the design and analysis of future PET studies with [(11)C]-(+)-PHNO.


Subject(s)
Brain/diagnostic imaging , Models, Theoretical , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Receptors, Dopamine D3/antagonists & inhibitors , Adult , Binding, Competitive , Carbon Radioisotopes/pharmacokinetics , Dopamine D2 Receptor Antagonists , Humans , Image Interpretation, Computer-Assisted , Male , Middle Aged
7.
Biol Psychiatry ; 72(5): 371-7, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22386378

ABSTRACT

BACKGROUND: We aimed to demonstrate a pharmacologically stimulated endogenous opioid release in the living human brain by evaluating the effects of amphetamine administration on [(11)C]carfentanil binding with positron emission tomography (PET). METHODS: Twelve healthy male volunteers underwent [(11)C]carfentanil PET before and 3 hours after a single oral dose of d-amphetamine (either a "high" dose, .5 mg/kg, or a sub-pharmacological "ultra-low" dose, 1.25 mg total dose or approximately .017 mg/kg). Reductions in [(11)C]carfentanil binding from baseline to post-amphetamine scans (ΔBP(ND)) after the "high" and "ultra-low" amphetamine doses were assessed in 10 regions of interest. RESULTS: [(11)C]carfentanil binding was reduced after the "high" but not the "ultra-low" amphetamine dose in the frontal cortex, putamen, caudate, thalamus, anterior cingulate, and insula. CONCLUSIONS: Our findings indicate that oral amphetamine administration induces endogenous opioid release in different areas of human brain, including basal ganglia, frontal cortex areas, and thalamus. The combination of an amphetamine challenge and [(11)C]carfentanil PET is a practical and robust method to probe the opioid system in the living human brain.


Subject(s)
Amphetamine/pharmacology , Brain/drug effects , Opioid Peptides/metabolism , Reward , Adult , Amphetamine/metabolism , Brain/anatomy & histology , Brain/diagnostic imaging , Brain/metabolism , Brain Mapping , Carbon Radioisotopes/metabolism , Fentanyl/analogs & derivatives , Fentanyl/metabolism , Humans , Male , Positron-Emission Tomography/methods , Statistics, Nonparametric
8.
J Cereb Blood Flow Metab ; 32(1): 127-36, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21878947

ABSTRACT

[(11)C]PHNO is a D(2)/D(3) agonist positron emission tomography radiotracer, with higher in vivo affinity for D(3) than for D(2) receptors. As [(11)C]-(+)-PHNO is an agonist, its in vivo binding is expected to be more affected by acute fluctuations in synaptic dopamine than that of antagonist radiotracers such as [(11)C]raclopride. In this study, the authors compared the effects of an oral dose of the dopamine releaser amphetamine (0.3 mg/kg) on in vivo binding of [(11)C]-(+)-PHNO and [(11)C]raclopride in healthy subjects, using a within-subjects, counterbalanced, open-label design. In the dorsal striatum, where the density of D(3) receptors is negligible and both tracers predominantly bind to D(2) receptors, the reduction of [(11)C]-(+)-PHNO binding potential (BP(ND)) was 1.5 times larger than that of [(11)C]raclopride. The gain in sensitivity associated with the agonist [(11)C]-(+)-PHNO implies that ∼65% of D(2) receptors are in the high-affinity state in vivo. In extrastriatal regions, where [(11)C]-(+)-PHNO predominantly binds to D(3) receptors, the amphetamine effect on [(11)C]-(+)-PHNO BP(ND) was even larger, consistent with the higher affinity of dopamine for D(3). This study indicates that [(11)C]-(+)-PHNO is superior to [(11)C]raclopride for studying acute fluctuations in synaptic dopamine in the human striatum. [(11)C]-(+)-PHNO also enables measurement of synaptic dopamine in D(3) regions.


Subject(s)
Amphetamine/pharmacology , Dopamine Agonists/pharmacokinetics , Dopamine Antagonists/pharmacokinetics , Oxazines/pharmacokinetics , Positron-Emission Tomography/methods , Raclopride/pharmacokinetics , Adult , Amphetamine/blood , Binding, Competitive , Brain/drug effects , Brain/metabolism , Dopamine/metabolism , Dopamine D2 Receptor Antagonists , Humans , Ligands , Male , Middle Aged , Protein Binding , Radioligand Assay , Radiopharmaceuticals/pharmacokinetics , Receptors, Dopamine D2/agonists , Receptors, Dopamine D3/agonists , Receptors, Dopamine D3/antagonists & inhibitors , Sensitivity and Specificity
9.
Cereb Cortex ; 22(12): 2784-93, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22186675

ABSTRACT

Animal research and human postmortem evidence highlight the importance of brain dopamine D3 receptor (D3R) function in multiple neuropsychiatric disorders, including addiction. Separate anatomical and functional neuroimaging findings implicate disrupted frontal cortical connectivity with distributed brain networks in processes relevant for these diseases. This potential conjunction between molecular and functional markers has not, however, been tested directly. Here, we used a novel combination of [(11)C]-(+)-PHNO positron emission tomography and resting-state functional magnetic resonance imaging in the same healthy individuals to investigate whether differences in midbrain D3R availability are associated with functional interactions between large-scale networks and regions involved in reward processing and cognition. High midbrain D3R availability was associated with reduced functional connectivity between orbitofrontal cortex (OFC) and networks implicated in cognitive control and salience processing. The opposite pattern was observed in subcortical reward circuitry and the "default mode" network, which showed greater connectivity with OFC in individuals with high D3R availability. These findings demonstrate that differential interactions between OFC and networks implicated in cognitive control and reward are associated with midbrain D3R availability, consistent with the hypothesis that dopamine D3R signaling is an important molecular pathway underlying goal-directed behavior.


Subject(s)
Frontal Lobe/physiology , Mesencephalon/anatomy & histology , Mesencephalon/physiology , Nerve Net/physiology , Neural Pathways/physiology , Receptors, Dopamine D3/metabolism , Adult , Brain Mapping , Connectome/methods , Female , Humans , Male , Middle Aged , Rest/physiology
10.
Comput Med Imaging Graph ; 35(5): 407-16, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21295443

ABSTRACT

In dynamic positron emission tomography data many researchers have attempted to exploit kinetic models within reconstruction such that parametric images are estimated directly from measurements. This work studies a direct parametric maximum likelihood expectation maximization algorithm applied to [(18)F]DOPA data using reference-tissue input function. We use a modified version for direct reconstruction with a gradually descending scheme of subsets (i.e. 18-6-1) initialized with the FBP parametric image for faster convergence and higher accuracy. The results compared with analytic reconstructions show quantitative robustness (i.e. minimal bias) and clinical reproducibility within six human acquisitions in the region of clinical interest. Bland-Altman plots for all the studies showed sufficient quantitative agreement between the direct reconstructed parametric maps and the indirect FBP (--0.035x+0.48E--5).


Subject(s)
Algorithms , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Positron-Emission Tomography/methods , Humans , Reproducibility of Results , Sensitivity and Specificity
11.
Drug Discov Today Technol ; 8(2-4): e45-51, 2011.
Article in English | MEDLINE | ID: mdl-24990262

ABSTRACT

The quantitative application of PET neuroreceptor imaging to study pathophysiology, diagnostics and drug development has continued to benefit from associated advances in biomathematical imaging methodology. We review some of these advances with particular focus on multi-modal image processing, tracer kinetic modeling, occupancy studies and discovery and development of novel radioligands.:

12.
Neuroimage ; 54(1): 264-77, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20600980

ABSTRACT

[(11)C]-(+)-PHNO is a D3 preferring PET radioligand which has recently opened the possibility of imaging D3 receptors in the human brain in vivo. This imaging tool allows characterisation of the distribution of D3 receptors in vivo and further investigation of their functional role. The specific [(11)C]-(+)-PHNO signal is a mixture of D3 and D2 components with the relative magnitude of each component determined by the regional receptor densities. An accurate and reproducible delineation of regions of interest (ROI) is therefore important for optimal analysis of human PET data. We present a set of anatomical guidelines for the delineation of D3 relevant ROIs including substantia nigra, hypothalamus, ventral pallidum/substantia innominata, ventral striatum, globus pallidus and thalamus. Delineation of these structures using this approach allowed for high intra- and inter-operator reproducibility. Subsequently we used a selective D3 antagonist to dissect the total [(11)C]-(+)-PHNO signal in each region into its D3 and D2 components and estimated the regional fraction of the D3 signal (f(PHNO)(D3)). In descending order of magnitude the following results for the f(PHNO)(D3) were obtained: hypothalamus=100%, substantia nigra=100%, ventral pallidum/substantia innominata=75%, globus pallidus=65%, thalamus=43%, ventral striatum=26% and precommissural-ventral putamen=6%. An automated approach for the delineation of these anatomical regions of interest was also developed and investigated in terms of its reproducibility and accuracy.


Subject(s)
Brain/diagnostic imaging , Carbon Radioisotopes , Corpus Striatum/diagnostic imaging , Oxazines , Receptors, Dopamine/analysis , Brain/physiology , Brain Mapping/methods , Corpus Striatum/physiology , Dopamine Agonists , Humans , Magnetic Resonance Imaging/methods , Male , Positron-Emission Tomography/methods , Reference Values , Reproducibility of Results , Supraoptic Nucleus/diagnostic imaging , Supraoptic Nucleus/physiology , Thalamus/diagnostic imaging , Thalamus/physiology
13.
Biol Psychiatry ; 68(4): 392-9, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20599188

ABSTRACT

BACKGROUND: Dopamine D(3) receptors are involved in the pathophysiology of several neuropsychiatric conditions. [(11)C]-(+)-PHNO is a radiolabeled D(2) and D(3) agonist, suitable for imaging the agonist binding sites (denoted D(2HIGH) and D(3)) of these receptors with positron emission tomography (PET). PET studies in nonhuman primates documented that, in vivo, [(11)C]-(+)-PHNO displays a relative selectivity for D(3) compared with D(2HIGH) receptor sites and that the [(11)C]-(+)-PHNO signal is enriched in D(3) contribution compared with conventional ligands such as [(11)C] raclopride. METHODS: To define the D(3) contribution (f(PHNO)(D3)) to [(11)C]-(+)-PHNO binding potential (BP(ND)) in healthy humans, 52 PET scans were obtained in 19 healthy volunteers at baseline and following oral administration of various doses of the selective D(3) receptor antagonist, GSK598809. RESULTS: The impact of GSK598809 on [(11)C]-(+)-PHNO was regionally selective. In dorsal regions of the striatum, GSK598809 did not significantly affect [(11)C]-(+)-PHNO BP(ND) (f(PHNO)(D3) approximately 0%). Conversely, in the substantia nigra, GSK598809 dose-dependently reduced [(11)C]-(+)-PHNO binding to nonspecific level (f(PHNO)(D3) approximately 100%). In ventral striatum (VST), globus pallidus and thalamus (THA), [(11)C]-(+)-PHNO BP(ND) was attributable to a combination of D(2HIGH) and D(3) receptor sites, with f(PHNO)(D3) of 26%, 67% and 46%, respectively. D(3) receptor binding potential (BP(ND)(D3)) was highest in globus pallidus (1.90) and substantial nigra (1.39), with lower levels in VST (.77) and THA (.18) and negligible levels in dorsal striatum. CONCLUSIONS: This study elucidated the pharmacologic nature of the [(11)C]-(+)-PHNO signal in healthy subjects and provided the first quantification of D(3) receptor availability with PET in the living human brain.


Subject(s)
Brain Mapping , Brain/diagnostic imaging , Dopamine Agonists/pharmacokinetics , Oxazines/pharmacokinetics , Receptors, Dopamine D3/metabolism , Adult , Azabicyclo Compounds/administration & dosage , Binding, Competitive/drug effects , Binding, Competitive/physiology , Brain/metabolism , Carbon Radioisotopes/pharmacokinetics , Dopamine Antagonists/administration & dosage , Dose-Response Relationship, Drug , Humans , Oxadiazoles/administration & dosage , Oxazoles/administration & dosage , Positron-Emission Tomography , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...