Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
EJNMMI Phys ; 10(1): 65, 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37861929

ABSTRACT

BACKGROUND: Q.Clear, a Bayesian penalized likelihood reconstruction algorithm, has shown high potential in improving quantitation accuracy in PET systems. The Q.Clear algorithm controls noise during the iterative reconstruction through a ß penalization factor. This study aimed to determine the optimal ß-factor for accurate quantitation of dynamic PET scans. METHODS: A Flangeless Esser PET Phantom with eight hollow spheres (4-25 mm) was scanned on a GE Discovery MI PET/CT system. Data were reconstructed into five sets of variable acquisition times using Q.Clear with 18 different ß-factors ranging from 100 to 3500. The recovery coefficient (RC), coefficient of variation (CVRC) and root-mean-square error (RMSERC) were evaluated for the phantom data. Two male patients with recurrent glioblastoma were scanned on the same scanner using 18F-PSMA-1007. Using an irreversible two-tissue compartment model, the area under curve (AUC) and the net influx rate Ki were calculated to assess the impact of different ß-factors on the pharmacokinetic analysis of clinical PET brain data. RESULTS: In general, RC and CVRC decreased with increasing ß-factor in the phantom data. For small spheres (< 10 mm), and in particular for short acquisition times, low ß-factors resulted in high variability and an overestimation of measured activity. Increasing the ß-factor improves the variability, however at a cost of underestimating the measured activity. For the clinical data, AUC decreased and Ki increased with increased ß-factor; a change in ß-factor from 300 to 1000 resulted in a 25.5% increase in the Ki. CONCLUSION: In a complex dynamic dataset with variable acquisition times, the optimal ß-factor provides a balance between accuracy and precision. Based on our results, we suggest a ß-factor of 300-500 for quantitation of small structures with dynamic PET imaging, while large structures may benefit from higher ß-factors. TRIAL REGISTRATION: Clinicaltrials.gov, NCT03951142. Registered 5 October 2019, https://clinicaltrials.gov/ct2/show/NCT03951142 . EudraCT no 2018-003229-27. Registered 26 February 2019, https://www.clinicaltrialsregister.eu/ctr-search/trial/2018-003229-27/NO .

2.
Neurooncol Adv ; 5(1): vdad021, 2023.
Article in English | MEDLINE | ID: mdl-37066109

ABSTRACT

Background: Biomechanical tissue properties of glioblastoma tumors are heterogeneous, but the molecular mechanisms involved and the biological implications are poorly understood. Here, we combine magnetic resonance elastography (MRE) measurement of tissue stiffness with RNA sequencing of tissue biopsies to explore the molecular characteristics of the stiffness signal. Methods: MRE was performed preoperatively in 13 patients with glioblastoma. Navigated biopsies were harvested during surgery and classified as "stiff" or "soft" according to MRE stiffness measurements (|G*|norm). Twenty-two biopsies from eight patients were analyzed by RNA sequencing. Results: The mean whole-tumor stiffness was lower than normal-appearing white matter. The surgeon's stiffness evaluation did not correlate with the MRE measurements, which suggests that these measures assess different physiological properties. Pathway analysis of the differentially expressed genes between "stiff" and "soft" biopsies showed that genes involved in extracellular matrix reorganization and cellular adhesion were overexpressed in "stiff" biopsies. Supervised dimensionality reduction identified a gene expression signal separating "stiff" and "soft" biopsies. Using the NIH Genomic Data Portal, 265 glioblastoma patients were divided into those with (n = 63) and without (n = 202) this gene expression signal. The median survival time of patients with tumors expressing the gene signal associated with "stiff" biopsies was 100 days shorter than that of patients not expressing it (360 versus 460 days, hazard ratio: 1.45, P < .05). Conclusion: MRE imaging of glioblastoma can provide noninvasive information on intratumoral heterogeneity. Regions of increased stiffness were associated with extracellular matrix reorganization. An expression signal associated with "stiff" biopsies correlated with shorter survival of glioblastoma patients.

3.
ChemMedChem ; 15(17): 1624-1628, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32677155

ABSTRACT

We report the synthesis, radiosynthesis and biological characterisation of two gonadotropin-releasing hormone receptor (GnRH-R) antagonists with nanomolar binding affinity. A small library of GnRH-R antagonists was synthesised in 20-67 % overall yield with the aim of identifying a high-affinity antagonist capable of crossing the blood-brain barrier. Binding affinity to rat GnRH-R was determined by autoradiography in competitive-binding studies against [125 I]buserelin, and inhibition constants were calculated by using the Cheng-Prusoff equation. The radioligands were obtained in 46-79 % radiochemical yield and >95 % purity and with a molar activity of 19-38 MBq/nmol by direct nucleophilic radiofluorination. Positron emission tomography imaging in rat under baseline conditions in comparison to pretreatment with a receptor-saturating dose of GnRH antagonist revealed saturable uptake (0.1 %ID/mL) into the brain.


Subject(s)
Brain/drug effects , Drug Discovery , Hydrocarbons, Fluorinated/pharmacology , Pyrimidines/pharmacology , Radiopharmaceuticals/pharmacology , Receptors, LHRH/antagonists & inhibitors , Animals , Binding Sites/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Fluorine Radioisotopes , Hydrocarbons, Fluorinated/chemical synthesis , Hydrocarbons, Fluorinated/chemistry , Molecular Structure , Positron-Emission Tomography , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Rats , Receptors, LHRH/metabolism , Structure-Activity Relationship
4.
Transl Psychiatry ; 8(1): 264, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504860

ABSTRACT

Major depressive disorder (MDD) is characterized by the altered integration of reward histories and reduced responding of the striatum. We have posited that this reduced striatal activation in MDD is due to tonically decreased stimulation of striatal dopamine synapses which results in decremented propagation of information along the cortico-striatal-pallido-thalamic (CSPT) spiral. In the present investigation, we tested predictions of this formulation by conducting concurrent functional magnetic resonance imaging (fMRI) and 11C-raclopride positron emission tomography (PET) in depressed and control (CTL) participants. We scanned 16 depressed and 14 CTL participants with simultaneous fMRI and 11C-raclopride PET. We estimated raclopride binding potential (BPND), voxel-wise, and compared MDD and CTL samples with respect to BPND in the striatum. Using striatal regions that showed significant between-group BPND differences as seeds, we conducted whole-brain functional connectivity analysis using the fMRI data and identified brain regions in each group in which connectivity with striatal seed regions scaled linearly with BPND from these regions. We observed increased BPND in the ventral striatum, bilaterally, and in the right dorsal striatum in the depressed participants. Further, we found that as BPND increased in both the left ventral striatum and right dorsal striatum in MDD, connectivity with the cortical targets of these regions (default-mode network and salience network, respectively) decreased. Deficits in stimulation of striatal dopamine receptors in MDD could account in part for the failure of transfer of information up the CSPT circuit in the pathophysiology of this disorder.


Subject(s)
Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/physiopathology , Dopamine/metabolism , Adult , Brain Mapping , Corpus Striatum/diagnostic imaging , Depressive Disorder, Major/diagnostic imaging , Female , Humans , Magnetic Resonance Imaging , Male , Neural Pathways/diagnostic imaging , Neural Pathways/metabolism , Neural Pathways/physiopathology , Positron-Emission Tomography , Raclopride
5.
Psychoneuroendocrinology ; 91: 123-131, 2018 05.
Article in English | MEDLINE | ID: mdl-29550675

ABSTRACT

The µ-opioid system modulates responses to pain and psychosocial stress and mediates non-social and social reward. In humans, the µ-opioid agonist morphine can increase overt attention to the eye-region and visual exploration of faces with neutral expressions. However, little is known about how the human µ-opioid system influences sensitivity to and appraisal of subtle and explicit cues of social threats and reward. Here, we examined the effects of selective µ-opioid stimulation on perception of anger and happiness in faces with explicit, neutral or implicit emotion expressions. Sixty-three healthy adults (32 females) attended two sessions where they received either placebo or 10 mg per oral morphine in randomised order under double-blind conditions. Based on the known µ-opioid reduction of pain and discomfort, as well as reports suggesting that the non-specific partial agonist buprenorphine or the non-specific antagonist naltrexone affect appraisal of social emotional stimuli, we hypothesised that morphine would reduce threat sensitivity and enhance perception of happy facial expressions. While overall perception of others' happiness was unaffected by morphine treatment, morphine reduced perception of anger in stimuli with neutral and implicit expressions without affecting perception of explicit anger. This effect was statistically unrelated to gender, subjective drug effects, mood and autism trait measures. The finding that a low dose of µ-agonist reduced the propensity to perceive anger in photos with subtle facial expressions is consistent with the notion that µ-opioids mediate social confidence and reduce sensitivity to threat cues.


Subject(s)
Facial Recognition/drug effects , Morphine/pharmacology , Perception/drug effects , Adult , Affect/drug effects , Analgesics, Opioid/pharmacology , Anger/drug effects , Attention/drug effects , Cues , Double-Blind Method , Emotions/drug effects , Expressed Emotion/drug effects , Facial Expression , Female , Happiness , Humans , Male , Middle Aged , Morphine/metabolism
6.
J Nucl Med ; 58(12): 2004-2009, 2017 12.
Article in English | MEDLINE | ID: mdl-28572487

ABSTRACT

The purpose of this study was to assess safety, biodistribution, and radiation dosimetry in humans for the highly selective σ-1 receptor PET agent 18F-6-(3-fluoropropyl)-3-(2-(azepan-1-yl)ethyl)benzo[d]thiazol-2(3H)-one (18F-FTC-146). Methods: Ten healthy volunteers (5 women, 5 men; age ± SD, 34.3 ± 6.5 y) were recruited, and written informed consent was obtained from all participants. Series of whole-body PET/MRI examinations were acquired for up to 3 h after injection (357.2 ± 48.8 MBq). Blood samples were collected, and standard vital signs (heart rate, pulse oximetry, and body temperature) were monitored at regular intervals. Regions of interest were delineated, time-activity curves were calculated, and organ uptake and dosimetry were estimated. Results: All subjects tolerated the PET/MRI examination well, and no adverse reactions to 18F-FTC-146 were reported. High accumulation of 18F-FTC-146 was observed in σ-1 receptor-dense organs such as the pancreas and spleen, moderate uptake in the brain and myocardium, and low uptake in bone and muscle. High uptake was also observed in the kidneys and bladder, indicating renal tracer clearance. The effective dose of 18F-FTC-146 was 0.0259 ± 0.0034 mSv/MBq (range, 0.0215-0.0301 mSv/MBq). Conclusion: First-in-human studies with clinical-grade 18F-FTC-146 were successful. Injection of 18F-FTC-146 is safe, and absorbed doses are acceptable. The potential of 18F-FTC-146 as an imaging agent for a variety of neuroinflammatory diseases is currently under investigation.


Subject(s)
Azepines/pharmacokinetics , Benzothiazoles/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Adult , Azepines/adverse effects , Azepines/chemical synthesis , Benzothiazoles/adverse effects , Benzothiazoles/chemical synthesis , Female , Healthy Volunteers , Humans , Isotope Labeling , Magnetic Resonance Imaging , Male , Multimodal Imaging , Radiometry , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/chemical synthesis , Receptors, sigma/drug effects , Receptors, sigma/metabolism , Tissue Distribution , Whole Body Imaging , Sigma-1 Receptor
7.
Mol Imaging Biol ; 19(5): 779-786, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28280965

ABSTRACT

PURPOSE: Sigma-1 receptors (S1Rs) play an important role in many neurological disorders. Simultaneous positron emission tomography (PET)/magnetic resonance imaging (MRI) with S1R radioligands may provide valuable information for diagnosing and guiding treatment for these diseases. Our previously reported S1R radioligand, [18F]FTC-146, demonstrated high affinity for the S1R (K i = 0.0025 nM) and excellent selectivity for the S1R over the sigma-2 receptor (S2Rs; K i = 364 nM) across several species (from mouse to non-human primate). Herein, we report the clinical-grade radiochemistry filed with exploratory Investigational New Drug (eIND) and first-in-human PET/MRI evaluation of [18F]FTC-146. PROCEDURES: [18F]FTC-146 is prepared via a direct [18F] fluoride nucleophilic radiolabeling reaction and formulated in 0.9 % NaCl containing no more than 10 % ethanol through sterile filtration. Quality control (QC) was performed based on USP 823 before doses were released for clinical use. The safety and whole body biodistribution of [18F]FTC-146 were evaluated using a simultaneous PET/MR scanner in two representative healthy human subjects. RESULTS: [18F]FTC-146 was synthesized with a radiochemical yield of 3.3 ± 0.7 % and specific radioactivity of 8.3 ± 3.3 Ci/µmol (n = 10, decay corrected to EOB). Both radiochemical and chemical purities were >95 %; the prepared doses were stable for 4 h at ambient temperature. All QC test results met specified clinical criteria. The in vivo PET/MRI investigations showed that [18F]FTC-146 rapidly crossed the blood brain barrier and accumulated in S1R-rich regions of the brain. There were also radioactivity distributed in the peripheral organs, i.e., the lungs, spleen, pancreas, and thyroid. Furthermore, insignificant uptake of [18F]FTC-146 was observed in cortical bone and muscle. CONCLUSION: A reliable and automated radiosynthesis for providing routine clinical-grade [18F]FTC-146 for human studies was established in a modified GE TRACERlab FXFN. PET/MRI demonstrated the initial tracer biodistribution in humans, and clinical studies investigating different S1R-related diseases are in progress.


Subject(s)
Azepines/chemistry , Azepines/chemical synthesis , Benzothiazoles/chemistry , Benzothiazoles/chemical synthesis , Magnetic Resonance Imaging , Positron-Emission Tomography , Adult , Azepines/pharmacokinetics , Benzothiazoles/pharmacokinetics , Female , Humans , Male , Tissue Distribution
8.
Nucl Med Biol ; 43(8): 478-89, 2016 08.
Article in English | MEDLINE | ID: mdl-27236283

ABSTRACT

INTRODUCTION: The gonadotropin releasing hormone receptor (GnRH-R) has a well-described neuroendocrine function in the anterior pituitary. However, little is known about its function in the central nervous system (CNS), where it is most abundantly expressed in hippocampus and amygdala. Since peptide ligands based upon the endogenous decapetide GnRH do not pass the blood-brain-barrier, we are seeking a high-affinity small molecule GnRH-R ligand suitable for brain imaging by positron emission tomography. We have previously reported the radiosynthesis and in vitro evaluation of two novel [(18)F]fluorinated GnRH-R ligands belonging to the furamide class of antagonists, with molecular weight less than 500 Da. We now extend this work using palladium coupling for the synthesis of four novel radioligands, with putatively reduced polar surface area and hydrophilicity relative to the two previously described compounds, and report the uptake of these (18)F-labeled compounds in brain of living rats. METHODS: We synthesized reference standards of the small molecule GnRH-R antagonists as well as mesylate precursors for (18)F-labeling. The antagonists were tested for binding affinity for both human and rat GnRH-R. Serum and blood stability in vitro and in vivo were studied. Biodistribution and PET imaging studies were performed in male rats in order to assess brain penetration in vivo. RESULTS: A palladium coupling methodology served for the synthesis of four novel fluorinated furamide GnRH receptor antagonists with reduced heteroatomic count. Radioligand binding assays in vitro revealed subnanomolar affinity of the new fluorinated compounds for both human and rat GnRH-R. The (18)F-GnRH antagonists were synthesized from the corresponding mesylate precursors in 5-15% overall radiochemical yield. The radiolabeled compounds demonstrated good in vivo stability. PET imaging with the (18)F-radiotracers in naive rats showed good permeability into brain and rapid washout, but absence of discernible specific binding in vivo. CONCLUSIONS: The novel small molecule (18)F-fluorinated GnRH-R antagonist compounds show high receptor affinity in vitro, and may prove useful for quantitative autoradiographic studies in vitro. The compounds were permeable to the blood-brain barrier, but nonetheless failed to reveal significant specific binding in brain of living rats. Nonetheless, our approach may serve as a foundation for designing PET ligands suitable to image the GnRH-R distribution in brain.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Fluorine Radioisotopes , Furans/chemical synthesis , Furans/metabolism , Receptors, LHRH/antagonists & inhibitors , Animals , Chemistry Techniques, Synthetic , Furans/pharmacokinetics , Furans/pharmacology , Male , Permeability , Positron-Emission Tomography , Radiochemistry , Rats , Rats, Sprague-Dawley , Substrate Specificity , Tissue Distribution
9.
J Med Chem ; 57(12): 5464-9, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24933507

ABSTRACT

We report the synthesis and biological evaluation of a triplet of 6-O-(18)F-fluoroethylated derivatives of structurally related orvinols that span across the full range of intrinsic activities, the antagonist diprenorphine, the partial agonist buprenorphine, and the full agonist phenethyl-orvinol. [(18)F]fluoroethyl-diprenorphine, [(18)F]fluoroethyl-buprenorphine, and [(18)F]fluoroethyl-phenethyl-orvinol were prepared in high yields and quality from their 6-O-desmethyl-precursors. The results indicate suitable properties of the three 6-O-(18)F-fluoroethylated derivatives as functional analogues to the native carbon-11 labeled versions with similar pharmacological properties.


Subject(s)
Buprenorphine/analogs & derivatives , Diprenorphine/analogs & derivatives , Morphinans/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Animals , Brain/diagnostic imaging , Brain/metabolism , Buprenorphine/chemical synthesis , Buprenorphine/chemistry , Buprenorphine/pharmacokinetics , CHO Cells , Carbon Radioisotopes , Cricetulus , Diprenorphine/chemical synthesis , Diprenorphine/chemistry , Diprenorphine/pharmacokinetics , Fluorine Radioisotopes , Humans , Morphinans/chemistry , Morphinans/pharmacokinetics , Narcotic Antagonists , Positron-Emission Tomography , Radioligand Assay , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Rats , Receptors, Opioid/agonists , Receptors, Opioid/metabolism , Structure-Activity Relationship
10.
J Neurosci Methods ; 199(1): 166-72, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21550366

ABSTRACT

Anatomical standardization (also called spatial normalization) of positron emission tomography (PET) small animal brain images is required to make statistical comparisons across individuals. Frequently, PET images are co-registered to an individual MR or CT image of the same subject in order to transform the functional images to an anatomical space. In the present work, we evaluate the normalization of synthetic PET (synPET) images to a synthetic PET template. To provide absolute error in terms of pixel misregistration, we created a synthetic PET image from the individual MR image through segmentation of the brain into gray and white matter which produced functional and anatomical images in the same space. When comparing spatial normalization of synPET images to a synPET template with the gold standard (MR images to an MR template), a mean translation error of 0.24mm (±0.20) and a maximal mean rotational error of 0.85° (±0.91) were found. Significant decrease in misregistration error was measured when achieving spatial normalization of functional images to a functional template instead of an anatomical template. This accuracy strengthens the use of standardization methods where individual PET images are registered to a customized PET template in order to statistically assess physiological changes in rat brains.


Subject(s)
Brain/diagnostic imaging , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/veterinary , Positron-Emission Tomography/veterinary , Rats/anatomy & histology , Animals , Brain/metabolism , Female , Fluorine Radioisotopes/pharmacokinetics , Fluorodeoxyglucose F18/pharmacokinetics , Positron-Emission Tomography/standards , Radiopharmaceuticals/pharmacokinetics , Rats, Sprague-Dawley , Reference Values
11.
Clin Physiol Funct Imaging ; 30(4): 285-93, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20662879

ABSTRACT

INTRODUCTION: Neuronal events leading to development of long-term potentiation (LTP) in the nociceptive pathways may be a cellular mechanism underlying hyperalgesia. In the present study, we examine if induction of spinal LTP may be associated with functional changes in the supraspinal opioidergic system. The opioid receptors (ORs) play a key role in nociceptive processing and controlling the descending modulatory system to the spinal cord. METHODS: Spinal LTP was induced by electrical high-frequency stimulation (HFS) conditioning applied to the sciatic nerve, and the excitability at spinal level was verified by spinal field potential recordings. To study supraspinal changes in opioid neurotransmission following the same HFS conditioning, we used small animal positron emission tomography (PET) and [(11)C]Phenethyl-Orvinol ([(11)C]PEO). All rats included in the PET study were scanned at baseline and 150 min after HFS, and specific binding was calculated with a reference tissue model. RESULTS: A clear C-fibre LTP, i.e. increased C-fibre response and reduced C-fibre threshold, was observed 150 min after HFS conditioning (t-test, P<0.05, n = 6). Moreover, increased OR binding, relative to baseline, was observed after the same type of HFS conditioning ipsilaterally in the amygdala, hippocampus, somatosensory cortex and superior colliculus, and bilaterally in the nucleus accumbens, caudate putamen and hypothalamus (paired t-test, HFS>baseline, P<0.05, n = 8). CONCLUSIONS: HFS conditioning of the sciatic nerve resulted in both spinal LTP and functional changes in supraspinal opioidergic signalling. Our findings suggest that induction of spinal LTP may be associated with reduced opioid neurotransmission in brain regions involved in pain modulation and affective-emotional responses.


Subject(s)
Brain/metabolism , Long-Term Potentiation , Pain/physiopathology , Receptors, Opioid/metabolism , Sciatic Nerve/physiopathology , Synaptic Transmission , Animals , Brain/diagnostic imaging , Electric Stimulation , Evoked Potentials , Female , Morphinans/metabolism , Nerve Fibers, Unmyelinated , Neural Pathways/physiopathology , Pain/metabolism , Positron-Emission Tomography , Radiopharmaceuticals/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
12.
Eur J Nucl Med Mol Imaging ; 37(6): 1174-80, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20157708

ABSTRACT

PURPOSE: The recent development in radiosynthesis of the (11)C-carbamate function increases the potential of [(11)C]GR103545, which for the last decade has been regarded as promising for imaging the kappa-opioid receptor (kappa-OR) with PET. In the present study, [(11)C]GR103545 was evaluated in awake rhesus macaques. Separate investigations were performed to clarify the OR subtype selectivity of this compound. METHODS: Regional brain uptake kinetics of [(11)C]GR103545 was studied 0-120 min after injection. The binding affinity and opioid subtype selectivity of [(11)C]GR103545 was determined in cells transfected with cloned human opioid receptors. RESULTS: In vitro binding assays demonstrated a high affinity of GR103545 for kappa-OR (K(i) = 0.02 +/- 0.01 nM) with excellent selectivity over mu-OR (6 x 10(2)-fold) and) delta-OR (2 x 10(4)-fold). PET imaging revealed a volume of distribution (V(T)) pattern consistent with the known distribution of kappa-OR, with striatum = temporal cortex > cingulate cortex > frontal cortex > parietal cortex > thalamus > cerebellum. CONCLUSION: [(11)C]GR103545 is selective for kappa-OR and holds promise for use to selectively depict and quantify this receptor in humans by means of PET.


Subject(s)
Macaca mulatta , Piperazines/metabolism , Pyrrolidines/metabolism , Receptors, Opioid, kappa/metabolism , Wakefulness , Animals , Carbon Radioisotopes , Positron-Emission Tomography , Radioactive Tracers , Substrate Specificity
13.
J Med Chem ; 52(18): 5586-9, 2009 Sep 24.
Article in English | MEDLINE | ID: mdl-19694469

ABSTRACT

Antagonist radiotracers have shown only a low sensitivity for detecting competition from high-efficacy agonists at opioid receptors (ORs) in vivo. We report that [(11)C]PEO binds with high affinity to mu and kappa-opioid receptors, is a full agonist, and concentrates in brain regions of rats with a high density of the mu-OR after intravenous injection. Blocking studies with mu and kappa-OR selective compounds demonstrated that the binding of [(11)C]PEO is saturable and selective to the mu-OR in rat brain.


Subject(s)
Morphinans/chemical synthesis , Morphinans/pharmacology , Positron-Emission Tomography , Receptors, Opioid/agonists , Animals , Brain/diagnostic imaging , Brain/metabolism , CHO Cells , Carbon Radioisotopes/chemistry , Cricetinae , Cricetulus , Humans , Kinetics , Male , Morphinans/metabolism , Rats , Rats, Wistar , Receptors, Opioid/metabolism , Substrate Specificity
14.
Pain ; 140(3): 456-464, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19004552

ABSTRACT

It has been suggested that spinal cord long-term potentiation (LTP) may contribute to hypersensitivity and hyperalgesia. We have investigated if noxious stimulus-induced spinal cord LTP might have a long lasting effect on supraspinal neuronal activity. First, we verified that spinal LTP was induced by electrical high frequency stimuli (HFS) conditioning applied to the sciatic nerve. The C-fibre response in the dorsal horn reached a twofold increase 150 min after HFS (t-test, p<0.01, n=6). Then, to study the metabolic supraspinal activity following the same stimulation protocol, we used small animal positron emission tomography (PET) and the glucose analog [(18)F]-fluorodeoxyglucose (FDG). With this combined approach we measured changes in regional supraspinal activity at two time points in HFS conditioned and in sham animals; acute (immediately after HFS/sham, n=4) and late phase (150 min after HFS/sham, n=10). Comparisons between HFS and sham groups revealed that induction of spinal LTP was followed by an acute metabolic response in the primary somatosensory cortex (S1), but also various slower metabolic adaptations in brain regions involved in modulation of nociceptive signaling and descending inhibition, i.e., amygdala, periaqueductal gray (PAG), rostral ventromedial medulla (RVM), and the dorsolateral pontomesencephalic tegmentum (DLPT) (t-test, p<0.05). The study demonstrates that PET may be used as an in vivo method to study regional brain metabolic activity between different conditions. It is concluded that noxious sciatic stimuli which induce spinal cord LTP also affect supraspinal metabolic activity. We suggest that these changes might illustrate a supraspinal maladaptive dysfunction involved in pain hypersensitivity and hyperalgesia.


Subject(s)
Brain/physiopathology , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Pain/physiopathology , Spinal Cord/physiopathology , Animals , Brain/diagnostic imaging , Brain/metabolism , Brain Stem/metabolism , Brain Stem/physiopathology , Electric Stimulation , Energy Metabolism/physiology , Female , Fluorodeoxyglucose F18 , Hyperalgesia/diagnostic imaging , Neural Pathways/metabolism , Neural Pathways/physiopathology , Pain/diagnostic imaging , Pain/metabolism , Pain Measurement/methods , Pain Threshold/physiology , Positron-Emission Tomography , Rats , Rats, Sprague-Dawley , Sciatic Nerve/physiopathology , Somatosensory Cortex/metabolism , Somatosensory Cortex/physiopathology , Time
15.
Front Neuroinform ; 1: 4, 2007.
Article in English | MEDLINE | ID: mdl-18974799

ABSTRACT

Tomographic neuroimaging techniques allow visualization of functionally and structurally specific signals in the mouse and rat brain. The interpretation of the image data relies on accurate determination of anatomical location, which is frequently obstructed by the lack of structural information in the data sets. Positron emission tomography (PET) generally yields images with low spatial resolution and little structural contrast, and many experimental magnetic resonance imaging (MRI) paradigms give specific signal enhancements but often limited anatomical information. Side-by-side comparison of image data with conventional atlas diagram is hampered by the 2-D format of the atlases, and by the lack of an analytical environment for accumulation of data and integrative analyses. We here present a method for reconstructing 3-D atlases from digital 2-D atlas diagrams, and exemplify 3-D atlas-based analysis of PET and MRI data. The reconstruction procedure is based on two seminal mouse and brain atlases, but is applicable to any stereotaxic atlas. Currently, 30 mouse brain structures and 60 rat brain structures have been reconstructed. To exploit the 3-D atlas models, we have developed a multi-platform atlas tool (available via The Rodent Workbench, http://rbwb.org) which allows combined visualization of experimental image data within the 3-D atlas space together with 3-D viewing and user-defined slicing of selected atlas structures. The tool presented facilitates assignment of location and comparative analysis of signal location in tomographic images with low structural contrast.

SELECTION OF CITATIONS
SEARCH DETAIL
...