Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Front Neurosci ; 15: 766176, 2021.
Article in English | MEDLINE | ID: mdl-34924935

ABSTRACT

Mutations in the huntingtin gene (HTT) triggers aggregation of huntingtin protein (mHTT), which is the hallmark pathology of neurodegenerative Huntington's disease (HD). Development of a high affinity 18F radiotracer would enable the study of Huntington's disease pathology using a non-invasive imaging modality, positron emission tomography (PET) imaging. Herein, we report the first synthesis of fluorine-18 imaging agent, 6-(5-((5-(2,2-difluoro-2-(fluoro-18F)ethoxy)pyridin-2-yl)methoxy)benzo[d]oxazol-2-yl)-2-methylpyridazin-3(2H)-one ([18F]1), a radioligand for HD and its preclinical evaluation in vitro (autoradiography of post-mortem HD brains) and in vivo (rodent and non-human primate brain PET). [18F]1 was synthesized in a 4.1% RCY (decay corrected) and in an average molar activity of 16.5 ± 12.5 GBq/µmol (445 ± 339 Ci/mmol). [18F]1 penetrated the blood-brain barrier of both rodents and primates, and specific saturable binding in post-mortem brain slices was observed that correlated to mHTT aggregates identified by immunohistochemistry.

2.
ACS Chem Neurosci ; 10(8): 3839-3846, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31339297

ABSTRACT

[18F]AV-1451 is one of the most widely used radiotracers for positron emission tomography (PET) imaging of tau protein aggregates in neurodegenerative disorders. While the radiotracer binds with high affinity to tau neurofibrillary tangles, extensive clinical studies have simultaneously revealed off-target tracer accumulation in areas of low tau burden such as the basal ganglia and choroid plexus. Though there are a number of possible reasons for this accumulation, it is often attributed to off-target binding to monoamine oxidase (MAO). In this paper, we investigate the association between [18F]AV-1451 and MAO through (i) enzyme inhibition assays, (ii) autoradiography with postmortem tissue samples, and (iii) nonhuman primate PET imaging. We confirm that [18F]AV-1451 is a weak inhibitor of MAO-A and -B and that MAO inhibitors can alter binding of [18F]AV-1451 in autoradiography and in vivo PET imaging.


Subject(s)
Basal Ganglia/drug effects , Carbolines/pharmacology , Choroid Plexus/drug effects , Monoamine Oxidase Inhibitors/pharmacology , Substantia Nigra/drug effects , Aged , Animals , Autoradiography , Basal Ganglia/diagnostic imaging , Basal Ganglia/metabolism , Choroid Plexus/diagnostic imaging , Choroid Plexus/metabolism , Humans , Monoamine Oxidase/metabolism , Positron-Emission Tomography , Primates , Radiopharmaceuticals , Substantia Nigra/diagnostic imaging , Substantia Nigra/metabolism
3.
Mol Imaging ; 18: 1536012119848927, 2019.
Article in English | MEDLINE | ID: mdl-31099304

ABSTRACT

Noise-induced hearing loss leads to anatomic and physiologic changes in primary auditory cortex (A1) and the adjacent dorsal rostral belt (RB). Since acetylcholine is known to modulate plasticity in other cortical areas, changes in A1 and RB following noise damage may be due to changes in cholinergic receptor expression. We used [3H]scopolamine and [18F]flubatine binding to measure muscarinic acetylcholine receptor (mAChR) and nicotinic acetylcholine receptor (nAChR) expression, respectively, in guinea pig A1 and RB 3 weeks following unilateral, left ear noise exposure, and a temporary threshold shift in hearing. [3H]Scopolamine binding decreased in right A1 and RB (contralateral to noise) compared to sham controls across all cortical layers. [18F]Flubatine binding showed a nonsignificant upward trend in right A1 following noise but only significantly increased in right RB and 2 layers of left RB (ipsilateral to noise). This selective response may ultimately influence cortical plasticity and function. The mechanism(s) by which cholinergic receptors are altered following noise exposure remain unknown. However, these data demonstrate noise exposure may differentially influence mAChRs that typically populate interneurons in A1 and RB more than nAChRs that are traditionally located on thalamocortical projections and provide motivation for cholinergic imaging in clinical patient populations of temporary or permanent hearing loss.


Subject(s)
Autoradiography/methods , Benzamides/chemistry , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Receptors, Muscarinic/analysis , Receptors, Nicotinic/analysis , Scopolamine/chemistry , Animals , Female , Guinea Pigs , Hearing Loss/metabolism
4.
EJNMMI Radiopharm Chem ; 3: 12, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30363401

ABSTRACT

BACKGROUND: We recently upgraded our [18F]fludeoxyglucose (FDG) production capabilities with the goal of futureproofing our FDG clinical supply, expanding the number of batches of FDG we can manufacture each day, and improving patient throughput in our nuclear medicine clinic. In this paper we report upgrade of the synthesis modules to the GE FASTLab 2 platform (Phase 1) and cyclotron updates (Phase 2) from both practical and regulatory perspectives. We summarize our experience manufacturing FDG on the FASTLab 2 module with a high-yielding self-shielded niobium (Nb) fluorine-18 target. RESULTS: Following installation of Nb targets for production of fluorine-18, a 55 µA beam for 22 min generated 1330 ± 153 mCi of [18F]fluoride. Using these cyclotron beam parameters in combination with the FASTLab 2, activity yields (AY) of FDG were 957 ± 102 mCi at EOS, corresponding to 72% non-corrected AY (n = 235). Our workflow, inventory management and regulatory compliance have been greatly simplified following the synthesis module and cyclotron upgrades, and patient wait times for FDG PET have been cut in half at our nuclear medicine clinic. CONCLUSIONS: The combination of FASTlab 2 and self-shielded Nb fluorine-18 targets have improved our yield of FDG, and enabled reliable and repeatable manufacture of the radiotracer for clinical use.

5.
Medchemcomm ; 9(3): 454-459, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-30108935

ABSTRACT

Radiolabeled erythrocytes have multiple applications in nuclear medicine, including blood pool imaging. Historically they have been labeled with SPECT radionuclides. A PET blood pool imaging agent is highly desirable as it would improve clinical applications with better image quality and resolution, higher sensitivity, and dynamic scanning capabilities. With the coming of age of modern 68Ge/68Ga generator systems, gallium-68 is now widely accessible. In this paper we describe an updated method for the preparation of 68Ga-labeled erythrocytes and their preliminary use in rodent blood pool imaging. A novel automated synthesis of [68Ga]oxine using a 68Ga/68Ge generator and automated synthesis module is reported. [68Ga]Oxine was synthesized in 50 ± 5% (n = 3) non-decay corrected radiochemical yield and >99% radiochemical purity. Rat and human erythrocytes were successfully labeled with the complex in 42% RCY, and the 68Ga-labeled erythrocytes have been shown to clearly image the blood pool in a healthy rat. Human erythrocytes labelled with [68Ga]oxine were shown to be viable up to 2 hours post-labelling, and washout of the radiolabel was minimal up to 1 hour post-labelling. Further optimization of the labeling method to translate for use in human cardiac and oncologic blood pool PET imaging studies, is underway.

6.
Medchemcomm ; 9(8): 1315-1322, 2018 Aug 01.
Article in English | MEDLINE | ID: mdl-30151086

ABSTRACT

There is considerable interest in using positron emission tomography (PET) imaging to understand the function of dopamine D3 receptors. Due to high sequence homology with D2 receptors, development of D3-selective PET radiotracers has been challenging. In an effort to overcome this issue, we report the radiosynthesis of a new selective D3 ligand with carbon-11 ([11C]1 ), and its initial preclincial evaluation as a potential PET radiotracer for in vivo imaging of D3 receptors. [11C]1 was prepared via [11C]CO2 fixation in 0.1% non-corrected radiochemical yield, good radiochemical purity (>95%) and high specific activity (>2000 Ci mmol-1). [11C]1 exhibited specific binding to D3 receptors using ex vivo autoradiography experiments with rat brain, but only 14-fold selectivity over D2 receptors which is lower than the 1400-fold value reported previously for cell studies. Rodent PET imaging revealed reasonable uptake of the radiotracer in areas of the brain known to be rich in D3 receptors.

7.
ACS Med Chem Lett ; 7(8): 746-50, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27563397

ABSTRACT

Clarithromycin is a potential treatment for hypersomnia acting through proposed negative allosteric modulation of GABAA receptors. We were interested whether this therapeutic benefit might extend to Parkinson's disease (PD) patients because GABAergic neurotransmission is implicated in postural control. Prior to initiating clinical studies in PD patients, we wished to better understand clarithromycin's mechanism of action. In this work we investigated whether the proposed activity of clarithromycin at the GABAA receptor is associated with the benzodiazepine binding site using in vivo [(11)C]flumazenil positron emission tomography (PET) in primates and ex vivo [(3)H]flumazenil autoradiography in rat brain. While the studies demonstrate that clarithromycin does not change the K d of FMZ, nor does it competitively displace FMZ, there is preliminary evidence from the primate PET imaging studies that clarithromycin delays dissociation and washout of flumazenil from the primate brain in a dose-dependent fashion. These findings would be consistent with the proposed GABAA allosteric modulator function of clarithromycin. While the results are only preliminary, further investigation of the interaction of clarithromycin with GABA receptors and/or GABAergic medications is warranted, and therapeutic applications of clarithromycin alone or in combination with flumazenil, to treat hyper-GABAergic status in PD at minimally effective doses, should also be pursued.

8.
ACS Chem Neurosci ; 7(3): 391-8, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26771209

ABSTRACT

The receptor for advanced glycation endproducts (RAGE) is a 35 kDa transmembrane receptor that belongs to the immunoglobulin superfamily of cell surface molecules. Its role in Alzheimer's disease (AD) is complex, but it is thought to mediate influx of circulating amyloid-ß into the brain as well as amplify Aß-induced pathogenic responses. RAGE is therefore of considerable interest as both a diagnostic and a therapeutic target in AD. Herein we report the synthesis and preliminary preclinical evaluation of [(18)F]RAGER, the first small molecule PET radiotracer for RAGE (Kd = 15 nM). Docking studies proposed a likely binding interaction between RAGE and RAGER, [(18)F]RAGER autoradiography showed colocalization with RAGE identified by immunohistochemistry in AD brain samples, and [(18)F]RAGER microPET confirmed CNS penetration and increased uptake in areas of the brain known to express RAGE. This first generation radiotracer represents initial proof-of-concept and a promising first step toward quantifying CNS RAGE activity using PET. However, there were high levels of nonspecific [(18)F]RAGER binding in vitro, likely due to its high log P (experimental log P = 3.5), and rapid metabolism of [(18)F]RAGER in rat liver microsome studies. Therefore, development of second generation ligands with improved imaging properties would be advantageous prior to anticipated translation into clinical PET imaging studies.


Subject(s)
Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Receptor for Advanced Glycation End Products/analysis , Alzheimer Disease/metabolism , Animals , Autoradiography , Brain/metabolism , Chromatography, High Pressure Liquid , Fluorine Radioisotopes/pharmacokinetics , Humans , Immunohistochemistry , Macaca mulatta , Molecular Docking Simulation , Positron-Emission Tomography , Rats , Rats, Sprague-Dawley
9.
ACS Med Chem Lett ; 6(2): 112-116, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25705326

ABSTRACT

Interest in quantifying metal-Aß species in vivo led to the synthesis and evaluation of [11C]L2-b and [18F]FL2-b as radiopharmaceuticals for studying the metallobiology of Alzheimer's disease (AD) using positron emission tomography (PET) imaging. [11C]L2-b was synthesized in 3.6% radiochemical yield (nondecay corrected, n = 3), >95% radiochemical purity, from the corresponding desmethyl precursor. [18F]FL2-b was synthesized in 1.0% radiochemical yield (nondecay corrected, n = 3), >99% radiochemical purity, from a 6-chloro pyridine precursor. Autoradiography experiments with AD positive and healthy control brain samples were used to determine the specificity of binding for the radioligands compared to [11C]PiB, a known imaging agent for ß-amyloid (Aß) aggregates. The Kd for [11C]L2-b and [18F]FL2-b were found to be 3.5 and 9.4 nM, respectively, from those tissue studies. Displacement studies of [11C]L2-b and [18F]FL2-b with PiB and AV-45 determined that L2-b binds to Aß aggregates differently from known radiopharmaceuticals. Finally, brain uptake of [11C]L2-b was examined through microPET imaging in healthy rhesus macaque, which revealed a maximum uptake at 2.5 min (peak SUV = 2.0) followed by rapid egress (n = 2).

10.
ACS Chem Neurosci ; 5(8): 718-30, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-24896980

ABSTRACT

Abnormally aggregated tau is the hallmark pathology of tauopathy neurodegenerative disorders and is a target for development of both diagnostic tools and therapeutic strategies across the tauopathy disease spectrum. Development of carbon-11- or fluorine-18-labeled radiotracers with appropriate affinity and specificity for tau would allow noninvasive quantification of tau burden using positron emission tomography (PET) imaging. We have synthesized [(18)F]lansoprazole, [(11)C]N-methyl lansoprazole, and [(18)F]N-methyl lansoprazole and identified them as high affinity radiotracers for tau with low to subnanomolar binding affinities. Herein, we report radiosyntheses and extensive preclinical evaluation with the aim of selecting a lead radiotracer for translation into human PET imaging trials. We demonstrate that [(18)F]N-methyl lansoprazole, on account of the favorable half-life of fluorine-18 and its rapid brain entry in nonhuman primates, favorable kinetics, low white matter binding, and selectivity for binding to tau over amyloid, is the lead compound for progression into clinical trials.


Subject(s)
Alzheimer Disease/diagnostic imaging , Lansoprazole , Positron-Emission Tomography , Radiopharmaceuticals , Supranuclear Palsy, Progressive/diagnostic imaging , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Autoradiography , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes/chemistry , Carbon Radioisotopes/pharmacokinetics , Drug Evaluation, Preclinical , Fluorine Radioisotopes/chemistry , Fluorine Radioisotopes/pharmacokinetics , Humans , Lansoprazole/chemistry , Lansoprazole/pharmacokinetics , Mice , Peptide Fragments/metabolism , Positron-Emission Tomography/methods , Primates , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Rats , Supranuclear Palsy, Progressive/metabolism , tau Proteins/metabolism
11.
Nucl Med Biol ; 41(6): 507-12, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24768148

ABSTRACT

INTRODUCTION: The dysfunction of glycogen synthase kinase-3ß (GSK-3ß) has been implicated in a number of diseases, including Alzheimer's disease. The ability to non-invasively quantify GSK-3ß activity in vivo is therefore of critical importance, and this work is focused upon development of inhibitors of GSK-3ß radiolabeled with carbon-11 to examine quantification of the enzyme using positron emission tomography (PET) imaging. METHODS: (11)C PyrATP-1 was prepared from the corresponding desmethyl-piperazine precursor in an automated synthesis module. In vivo rodent and primate imaging studies were conducted on a Concorde MicroPET P4 scanner to evaluate imaging properties and in vitro autoradiography studies with rat brain samples were carried out to examine specific binding. RESULTS: 2035±518MBq (55±14mCi) of [(11)C]PyrATP-1 was obtained (1%-2% non-corrected radiochemical yield at end-of-synthesis based upon [(11)C]CO2) with high chemical (>95%) and radiochemical (>99%) purities, and good specific activities (143±52GBq/µmol (3874±1424Ci/mmol)), n=5. In vivo microPET imaging studies revealed poor brain uptake in rodents and non-human primates. Pretreatment of rodents with cyclosporin A resulted in moderately increased brain uptake suggesting Pgp transporter involvement. Autoradiography demonstrated high levels of specific binding in areas of the rodent brain known to be rich in GSK-3ß. CONCLUSION: (11)C PyrATP-1 is readily synthesized using standard carbon-11 radiochemistry. However the poor brain uptake in rodents and non-human primates indicates that the radiotracer is not suitable for the purposes of quantifying GSK-3ß in neurological and psychiatric disorders.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Positron-Emission Tomography/methods , Protein Kinase Inhibitors/chemical synthesis , Pyrazinamide/analogs & derivatives , Pyrazines/chemical synthesis , Sulfonamides/chemical synthesis , Animals , Brain/diagnostic imaging , Carbon Radioisotopes , Chemistry Techniques, Synthetic , Female , Glycogen Synthase Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazinamide/chemical synthesis , Pyrazinamide/chemistry , Pyrazinamide/pharmacology , Pyrazines/chemistry , Pyrazines/pharmacology , Radioactive Tracers , Radiochemistry , Rats , Sulfonamides/chemistry , Sulfonamides/pharmacology
12.
ACS Med Chem Lett ; 3(11): 936-41, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-24900410

ABSTRACT

[(11)C]N-Methyl lansoprazole ([(11)C]NML, 3) was synthesized and evaluated as a radiopharmaceutical for quantifying tau neurofibrillary tangle (NFT) burden using positron emission tomography (PET) imaging. [(11)C]NML was synthesized from commercially available lansoprazole in 4.6% radiochemical yield (noncorrected RCY, based upon [(11)C]MeI), 99% radiochemical purity, and 16095 Ci/mmol specific activity (n = 5). Log P was determined to be 2.18. A lack of brain uptake in rodent microPET imaging revealed [(11)C]NML to be a substrate for the rodent permeability-glycoprotein 1 (PGP) transporter, but this could be overcome by pretreating with cyclosporin A to block the PGP. Contrastingly, [(11)C]NML was not found to be a substrate for the primate PGP, and microPET imaging in rhesus revealed [(11)C]NML uptake in the healthy primate brain of ∼1600 nCi/cc maximum at 3 min followed by rapid egress to 500 nCi/cc. Comparative autoradiography between wild-type rats and transgenic rats expressing human tau (hTau +/+) revealed 12% higher uptake of [(11)C]NML in the cortex of brains expressing human tau. Further autoradiography with tau positive brain samples from progressive supranuclear palsy (PSP) patients revealed colocalization of [(11)C]NML with tau NFTs identified using modified Bielschowsky staining. Finally, saturation binding experiments with heparin-induced tau confirmed K d and Bmax values of [(11)C]NML as 700 pM and 0.214 fmol/µg, respectively.

13.
J Neurosci ; 25(39): 8978-87, 2005 Sep 28.
Article in English | MEDLINE | ID: mdl-16192388

ABSTRACT

In recent studies, inactivation of the dorsal hippocampus before the retrieval of extinguished fear memories disrupted the context-dependent expression of these memories. In the present experiments, we examined the role of the dorsal hippocampus in the acquisition of extinction. After pairing an auditory conditional stimulus (CS) with an aversive footshock [unconditional stimulus (US)], rats received an extinction session in which the CS was presented without the US. In experiment 1, infusion of muscimol, a GABAA receptor agonist, into the dorsal hippocampus before the extinction training session decreased the rate of extinction. Moreover, when later tested for fear to the extinguished CS, all rats that had received hippocampal inactivation before extinction training demonstrated renewed fear regardless of the context in which testing took place. This suggests a role for the dorsal hippocampus in both acquiring the extinction memory and encoding the CS-context relationship that yields the context dependence of extinction. In experiment 2, inactivation of the dorsal hippocampus before testing also disrupted the context dependence of fear to the extinguished CS. In experiment 3, quantitative autoradiography revealed the boundaries of muscimol diffusion after infusion into the dorsal hippocampus. Together, these results reveal that the dorsal hippocampus is involved in the acquisition, contextual encoding, and context-dependent retrieval of fear extinction. Learning and remembering when and where aversive events occur is essential for adaptive emotional regulation.


Subject(s)
Extinction, Psychological/physiology , Fear , Hippocampus/physiology , Acoustic Stimulation , Animals , Autoradiography , Behavior, Animal/drug effects , Conditioning, Psychological , Electroshock , Extinction, Psychological/drug effects , Fear/drug effects , GABA Agonists/administration & dosage , GABA Agonists/pharmacokinetics , GABA Agonists/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Infusion Pumps , Male , Muscimol/administration & dosage , Muscimol/pharmacokinetics , Muscimol/pharmacology , Rats , Rats, Long-Evans
14.
J Neurooncol ; 73(3): 225-38, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15980973

ABSTRACT

Solvent facilitated perfusion (SFP) has been proposed as a technique to increase the delivery of chemotherapeutic agents to tumors. SFP entails direct injection of the agent into the tumor in a water-miscible organic solvent, and because the solvent moves easily through both aqueous solutions and cellular membranes it drives the penetration of the solubilized anticancer agent throughout the tumor. To test this hypothesis, we compared the pharmacokinetics (PK) of 14C-labeled 1,3-bis-chlorethyl-1-nitrosourea (BCNU) in intra-cerebral 9L rat gliomas after intravenous (IV) infusion in 90% saline--10% ethanol or direct intratumoral (IT) injection of 14C-BCNU in 100% ethanol (DTI-015). Treatment with DTI-015 yielded a peak radioactive count (Cmax) for the 14C label that was 100-1000 fold higher in the tumor than in all other tissues in addition to a concentration in the tumor that was 100-fold higher than that achieved following IV infusion of 14C-BCNU. Pathologic and auto-radiographic analysis of tissue sections following IT injection of 14C-BCNU in ethanol into either tumor or normal rat brain revealed both an enhanced local volume of distribution and an increased concentration of BCNU delivered to tumor compared to non-tumor bearing brain. To investigate the mechanism behind the SFP of BCNU to the tumor both dynamic contrast and perfusion MRI were performed on 9L tumors before and after treatment and demonstrated a decrease in tumor perfusion following IT injection of DTI-015. Finally, initial PK of patient blood samples following administration of DTI-015 into relapsed high-grade glioma indicated a 20-fold decrease in systemic exposure to BCNU compared to IV infusion of BCNU providing further evidence for the enhanced therapeutic ratio observed for DTI-015.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/pharmacokinetics , Brain Neoplasms/drug therapy , Carmustine/administration & dosage , Carmustine/pharmacokinetics , Glioma/drug therapy , Animals , Antineoplastic Agents, Alkylating/blood , Area Under Curve , Brain Neoplasms/pathology , Carmustine/blood , Ethanol , Glioma/pathology , Half-Life , Humans , Injections, Intralesional , Injections, Intravenous , Injections, Intraventricular , Magnetic Resonance Imaging , Male , Rats , Tissue Distribution
15.
Exp Neurol ; 188(2): 387-90, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15246838

ABSTRACT

The in vivo equilibrium specific binding of d-threo-[3H]methylphenidate, a radioligand for the dopamine transporter (DAT), and +-alpha-[3H]dihydrotetrabenazine, a radioligand for the vesicular monoamine transporter (VMAT2), were examined in rat brain with and without prior administration of 5 mg/kg scopolamine. Drug-treated animals exhibited a 30% increase in d-threo-[3H]methylphenidate binding to the DAT in the striatum relative to controls. No changes in specific binding of +-alpha-[3H]dihydrotetrabenazine were observed in any brain region following scopolamine pretreatment. Cholinergic drugs thus differentially affect in vivo specific binding of DAT and VMAT2 radioligands, suggesting this should be a consideration in selection of in vivo markers for imaging studies of dopaminergic terminals in the brain of animals and humans.


Subject(s)
Brain/metabolism , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuropeptides , Scopolamine/pharmacology , Tetrabenazine/analogs & derivatives , Animals , Binding, Competitive/drug effects , Brain Chemistry , Dopamine Plasma Membrane Transport Proteins , Ligands , Male , Methylphenidate/pharmacokinetics , Muscarinic Antagonists/pharmacology , Radioligand Assay , Rats , Tetrabenazine/pharmacokinetics , Tritium , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
16.
Eur J Neurosci ; 18(11): 3080-8, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14656303

ABSTRACT

The amygdala is an essential neural substrate for Pavlovian fear conditioning. Nevertheless, long-term synaptic plasticity in amygdaloid afferents, such as the auditory thalamus, may contribute to the formation of fear memories. We therefore compared the influence of protein synthesis inhibition in the amygdala and the auditory thalamus on the consolidation of Pavlovian fear conditioning in Long-Evans rats. Rats received three tone-footshock trials in a novel conditioning chamber. Immediately after fear conditioning, rats were infused intra-cranially with the protein synthesis inhibitor, anisomycin. Conditional fear to the tone and conditioning context was assessed by measuring freezing behaviour in separate retention tests conducted at least 24 h following conditioning. Post-training infusion of anisomycin into the amygdala impaired conditional freezing to both the auditory and contextual stimuli associated with footshock. In contrast, intra-thalamic infusions of anisomycin or a broad-spectrum protein kinase inhibitor [1-(5'-isoquinolinesulphonyl)-2-methylpiperazine, H7] did not affect conditional freezing during the retention tests. Pre-training intra-thalamic infusion of the NMDA receptor antagonist 2-amino-5-phosphonopentanoic acid (APV), which blocks synaptic transmission in the auditory thalamus, produced a selective deficit in the acquisition of auditory fear conditioning. Autoradiographic assays of cerebral [14C]-leucine incorporation revealed similar levels of protein synthesis inhibition in the amygdala and thalamus following intra-cranial anisomycin infusions. These results reveal that the establishment of long-term fear memories requires protein synthesis in the amygdala, but not the thalamus, after auditory fear conditioning. Forms of synaptic plasticity that depend on protein synthesis, such as long-term potentiation, are likely candidates for the encoding and long-term storage of fear memories in the amygdala.


Subject(s)
Amygdala/chemistry , Amygdala/metabolism , Conditioning, Classical/physiology , Fear/physiology , Protein Biosynthesis , Amygdala/drug effects , Amygdala/physiology , Animals , Anisomycin/pharmacology , Autoradiography , Behavior, Animal , Benzoxazines , Carbon Isotopes/metabolism , Functional Laterality , Immobilization , Leucine/metabolism , Male , Oxazines/metabolism , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Long-Evans , Thalamus/chemistry , Thalamus/drug effects , Thalamus/physiology , Time Factors
17.
Exp Neurol ; 183(1): 81-6, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12957491

ABSTRACT

There is controversy over potential effects of dopaminergic replacement therapies on the partially lesioned nigrostriatal dopaminergic projection. We evaluated indirect (levodopa, L-DOPA) versus direct (pramipexole, PRA) dopaminergic treatment effects on nigrostriatal lesion severity as measured with vesicular monoamine transporter type-2 (VMAT2) binding. Prior studies have shown that striatal VMAT2 density provides an objective estimate of dopaminergic neuronal integrity, without confounding effects of compensatory regulation. Partial unilateral median forebrain bundle lesions were made by injection of 6-hydroxydopamine in adult male Sprague-Dawley rats. Lesion severity was estimated using rotational behavior after injections of apomorphine and amphetamine. Rats were ranked and matched in pairs by rotation and assigned to receive either PRA (1 mg/kg/day) or L-DOPA/benserazide (100/25 mg/kg/day) ip via osmotic pump. After 4 weeks of drug treatment, in vitro autoradiography was performed with [(3)H]methoxytetrabenazine to measure striatal VMAT2 binding density. Lesion-to-intact VMAT2 density correlated with rotation in both treatment groups. There was no treatment effect on VMAT2 expression in the partially lesioned striatum and thus no differential effect of indirect versus direct dopamimetic treatment on nigrostriatal integrity.


Subject(s)
Corpus Striatum/drug effects , Levodopa/therapeutic use , Membrane Transport Proteins , Neuropeptides , Parkinsonian Disorders/drug therapy , Substantia Nigra/drug effects , Thiazoles/therapeutic use , Amphetamine/pharmacology , Animals , Antiparkinson Agents/therapeutic use , Apomorphine/pharmacology , Autoradiography , Behavior, Animal/drug effects , Benzothiazoles , Binding, Competitive/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Male , Membrane Glycoproteins/biosynthesis , Motor Activity/drug effects , Oxidopamine , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/pathology , Pramipexole , Rats , Rats, Sprague-Dawley , Substantia Nigra/metabolism , Substantia Nigra/pathology , Treatment Outcome , Tritium , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
18.
Ann Neurol ; 51(6): 767-71, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12112084

ABSTRACT

Vesicular monoamine transporter type 2 and benzodiazepine binding site expressions were examined with quantitative autoradiography in postmortem striata from 19 patients with dementia with Lewy bodies, seven patients with dementia with Lewy bodies and Alzheimer's disease, 12 patients with Alzheimer's disease, and eight neurologically normal subjects. Striatal vesicular monoamine transporter type 2 expression in dementia with Lewy bodies and in dementia with Lewy bodies plus Alzheimer's disease was reduced significantly, indicating degeneration of nigrostriatal projections. Striatal benzodiazepine binding site expression was unchanged, indicating preserved intrinsic striatal neuropil. Vesicular monoamine transporter type 2 and benzodiazepine binding site expressions were each preserved in Alzheimer's disease striatum. We conclude that dementia with Lewy bodies may be distinguished from Alzheimer's disease by postmortem examination or by future in vivo measurements of the striatal vesicular monoamine transporter type 2.


Subject(s)
Alzheimer Disease/pathology , Corpus Striatum/pathology , Lewy Body Disease/pathology , Membrane Glycoproteins/analysis , Membrane Transport Proteins , Neurons/chemistry , Neuropeptides , Aged , Aged, 80 and over , Female , Humans , Male , Neurotransmitter Agents/analysis , Receptors, GABA-A/analysis , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
19.
Neurology ; 58(7): 1013-8, 2002 Apr 09.
Article in English | MEDLINE | ID: mdl-11940684

ABSTRACT

OBJECTIVE: To determine the status of cholinergic and monoaminergic vesicular transporter binding sites in progressive supranuclear palsy (PSP). METHODS: The authors determined autoradiographically the regional expression of acetylcholine vesicular transporter (VAChT) and monoamine vesicular transporter type 2 (VMAT2) binding sites in postmortem basal ganglia samples from subjects with PSP. Comparison neurochemical measures included choline acetyltransferase (ChAT) enzyme activity and benzodiazepine (BZ) binding sites. RESULTS: VAChT expressions and ChAT activities in caudate nucleus and putamen were markedly decreased in PSP, whereas BZ binding was unaffected, consistent with selective losses of striatal cholinergic interneurons. VMAT2 density was reduced significantly in the caudate nucleus, putamen, and substantia nigra pars compacta, consistent with degeneration of dopaminergic nigrostriatal projection neurons in PSP. In the globus pallidus, BZ receptor binding sites were reduced, whereas VMAT2 and VAChT binding sites were unchanged, indicating losses of intrinsic pallidal neurons and synapses. CONCLUSIONS: These results confirm selective and marked degenerations of basal ganglia cholinergic and dopaminergic terminals in PSP. Striatal VAChT reduction may provide a unique neurochemical imaging marker for distinction of PSP from other types of basal ganglia neurodegeneration.


Subject(s)
Acetylcholine/metabolism , Carrier Proteins/metabolism , Membrane Transport Proteins , Neuropeptides , Supranuclear Palsy, Progressive/metabolism , Synaptic Vesicles/metabolism , Vesicular Transport Proteins , Aged , Aged, 80 and over , Basal Ganglia/metabolism , Biomarkers/chemistry , Brain/metabolism , Choline O-Acetyltransferase/metabolism , Female , Humans , Male , Membrane Glycoproteins/metabolism , Middle Aged , Receptors, GABA-A/metabolism , Vesicular Acetylcholine Transport Proteins , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...