Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
EJNMMI Radiopharm Chem ; 9(1): 24, 2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38526746

RESUMEN

BACKGROUND: Production of [11C]CH4 from gas targets is notorious for weak performance with respect to yield, especially when using high beam currents. Post-target conversion of [11C]CO2 to [11C]CH4 is a widely used roundabout method in 11C-radiochemistry, but the added complexity increase the challenge to control carrier carbon. Thus in-target-produced [11C]CH4 is superior with respect to molar activity. We studied the in-target production of [11C]CO2 and [11C]CH4 from nitrogen gas targets as a function of beam current, irradiation time, and target temperature. RESULTS: [11C]CO2 production was practically unchanged across the range of varied parameters, but the [11C]CH4 yield, presented in terms of saturation yield YSAT(11CH4), had a negative correlation with beam current and a positive correlation with target chamber temperature. A formulated model equation indicates behavior where the [11C]CH4 formation follows a parabolic graph as a function of beam current. The negative square term, i.e., the yield loss, is postulated to arise from Haber-Bosch-like NH3 formation: N2 + 3H2 → 2NH3. The studied conditions suggest that the NH3 (liq.) would be condensed on the target chamber walls, thus depleting the hydrogen reserve needed for the conversion of nascent 11C to [11C]CH4. CONCLUSIONS: [11C]CH4 production can be improved by increasing the target chamber temperature, which is presented in a mathematical formula. Our observations have implications for targetry design (geometry, gas volume and composition, pressure) and irradiation conditions, providing specific knowledge to enhance [11C]CH4 production at high beam currents. Increased [11C]CH4 radioactivity is an obvious benefit in radiosynthesis in terms of product yield and molar radioactivity.

2.
J Behav Addict ; 12(3): 670-681, 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37561637

RESUMEN

Background: The neurobiological mechanisms of gambling disorder are not yet fully characterized, limiting the development of treatments. Defects in frontostriatal connections have been shown to play a major role in substance use disorders, but data on behavioral addictions, such as gambling disorder, are scarce. The aim of this study was to 1) investigate whether gambling disorder is associated with abnormal frontostriatal connectivity and 2) characterize the key neurotransmitter systems underlying the connectivity abnormalities. Methods: Fifteen individuals with gambling disorder and 17 matched healthy controls were studied with resting-state functional connectivity MRI and three brain positron emission tomography scans, investigating dopamine (18F-FDOPA), opioid (11C-carfentanil) and serotonin (11C-MADAM) function. Frontostriatal connectivity was investigated using striatal seed-to-voxel connectivity and compared between the groups. Neurotransmitter systems underlying the identified connectivity differences were investigated using region-of-interest and voxelwise approaches. Results: Individuals with gambling disorder showed loss of functional connectivity between the right nucleus accumbens (NAcc) and a region in the right dorsolateral prefrontal cortex (DLPFC) (PFWE <0.05). Similarly, there was a significant Group x right NAcc interaction in right DLPFC 11C-MADAM binding (p = 0.03) but not in 18F-FDOPA uptake or 11C-carfentanil binding. This was confirmed in voxelwise analyses showing a widespread Group x right NAcc interaction in the prefrontal cortex 11C-MADAM binding (PFWE <0.05). Right NAcc 11C-MADAM binding potential correlated with attentional impulsivity in individuals with gambling disorder (r = -0.73, p = 0.005). Discussion: Gambling disorder is associated with right hemisphere abnormal frontostriatal connectivity and serotonergic function. These findings will contribute to understanding the neurobiological mechanism and may help identify potential treatment targets for gambling disorder.


Asunto(s)
Juego de Azar , Humanos , Juego de Azar/diagnóstico por imagen , Juego de Azar/metabolismo , Serotonina , Imagen por Resonancia Magnética/métodos , Neurotransmisores
3.
Med Sci Sports Exerc ; 54(7): 1076-1084, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35195103

RESUMEN

INTRODUCTION: Central µ-opioid receptors (MORs) modulate affective responses to physical exercise. Individuals with higher aerobic fitness report greater exercise-induced mood improvements than those with lower fitness, but the link between cardiorespiratory fitness and the MOR system remains unresolved. Here we tested whether maximal oxygen uptake (V̇O2peak) and physical activity level are associated with cerebral MOR availability and whether these phenotypes predict endogenous opioid release after a session of exercise. METHODS: We studied 64 healthy lean men who performed a maximal incremental cycling test for V̇O2peak determination, completed a questionnaire assessing moderate-to-vigorous physical activity (MVPA; in minutes per week), and underwent positron emission tomography with [11C]carfentanil, a specific radioligand for MOR. A subset of 24 subjects underwent additional positron emission tomography scan also after a 1-h session of moderate-intensity exercise and 12 of them also after a bout of high-intensity interval training. RESULTS: Higher self-reported MVPA level predicted greater opioid release after high-intensity interval training, and both V̇O2peak and MVPA level were associated with a larger decrease in cerebral MOR binding after aerobic exercise in the ventral striatum, orbitofrontal cortex, and insula. That is, more trained individuals showed greater opioid release acutely after exercise in brain regions especially relevant for reward and cognitive processing. Fitness was not associated with MOR availability. CONCLUSIONS: We conclude that regular exercise training and higher aerobic fitness may induce neuroadaptation within the MOR system, which might contribute to improved emotional and behavioral responses associated with long-term exercise.


Asunto(s)
Analgésicos Opioides , Capacidad Cardiovascular , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Ejercicio Físico/fisiología , Humanos , Tomografía de Emisión de Positrones/métodos , Recompensa
4.
Psychol Med ; 52(14): 3280-3288, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-33568248

RESUMEN

BACKGROUND: Risk is an essential trait of most daily decisions. Our behaviour when faced with risks involves evaluation of many factors including the outcome probabilities, the valence (gains or losses) and past experiences. Several psychiatric disorders belonging to distinct diagnostic categories, including pathological gambling and addiction, show pathological risk-taking and implicate abnormal dopaminergic, opioidergic and serotonergic neurotransmission. In this study, we adopted a transdiagnostic approach to delineate the neurochemical substrates of decision making under risk. METHODS: We recruited 39 participants, including 17 healthy controls, 15 patients with pathological gambling and seven binge eating disorder patients, who completed an anticipatory risk-taking task. Separately, participants underwent positron emission tomography (PET) imaging with three ligands, [18F]fluorodopa (FDOPA), [11C]MADAM and [11C]carfentanil to assess presynaptic dopamine synthesis capacity and serotonin transporter and mu-opioid receptor binding respectively. RESULTS: Risk-taking behaviour when faced with gains positively correlated with dorsal cingulate [11C]carfentanil binding and risk-taking to losses positively correlated with [11C]MADAM binding in the caudate and putamen across all subjects. CONCLUSIONS: We show distinct neurochemical substrates underlying risk-taking with the dorsal cingulate cortex mu-opioid receptor binding associated with rewards and dorsal striatal serotonin transporter binding associated with losses. Risk-taking and goal-directed control appear to dissociate between dorsal and ventral fronto-striatal systems. Our findings thus highlight the potential role of pharmacological agents or neuromodulation on modifying valence-specific risk-taking biases.


Asunto(s)
Juego de Azar , Proteínas de Transporte de Serotonina en la Membrana Plasmática , Humanos , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Voluntarios Sanos , Tomografía de Emisión de Positrones/métodos , Recompensa , Receptores Opioides/metabolismo
5.
EJNMMI Res ; 10(1): 152, 2020 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-33296042

RESUMEN

BACKGROUND: Availability of the α2C-adrenoceptor (α2C-AR) positron emission tomography (PET) tracer, [11C]ORM-13070, and the α2C-AR antagonist ORM-12741 allows probing of the roles of this G-protein coupled receptor subtype in brain function, both in healthy humans and in patients with various brain disorders. This translational study employed [11C]ORM-13070 autoradiography and PET to determine α2C-AR occupancy by ORM-12741 in rat and human brain, respectively. RESULTS: ORM-12741 has high affinity (Ki: 0.08 nM) and potent antagonist activity (Kb: 0.04 nM) as well as selectivity (Ki estimates for the human α2A-AR and α2B-AR were 8.3 nM and 0.8 nM, respectively) for the human α2C-AR subtype. [11C]ORM-13070 had highest uptake in the basal ganglia of rat and human brain. Pretreatment with ORM-12741 inhibited [11C]ORM-13070 binding in rat striatum in a time- and dose-dependent manner at 10 and 50 µg/kg (s.c.) with an EC50 estimate of 1.42 ng/mL in rat plasma, corresponding to protein-free drug concentration of 0.23 nM. In the living human brain, time- and dose-related α2C-AR occupancy was detected with EC50 estimates of 24 ng/mL and 31 ng/mL for the caudate nucleus and putamen, respectively, corresponding to protein-free concentrations in plasma of 0.07 nM and 0.1 nM. Modelling-based maximum α2C-AR occupancy estimates were 63% and 52% in the caudate nucleus and the putamen, respectively. CONCLUSIONS: ORM-12741 is a selective α2C-AR antagonist which penetrates the rat and human brain to occupy α2C-ARs in a manner consistent with its receptor pharmacology. Trial registration number and date of registration: ClinicalTrial.cov NCT00829907. Registered 11 December 2008. https://clinicaltrials.gov/ .

6.
Transl Psychiatry ; 10(1): 84, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32127520

RESUMEN

Our daily decisions are governed by the arbitration between goal-directed and habitual strategies. However, the neurochemical basis of this arbitration is unclear. We assessed the contribution of dopaminergic, serotonergic, and opioidergic systems to this balance across reward and loss domains. Thirty-nine participants (17 healthy controls, 15 patients with pathological gambling, and 7 with binge eating disorder) underwent positron emission tomography (PET) imaging with [18F]FDOPA, [11C]MADAM and [11C]carfentanil to assess presynaptic dopamine, and serotonin transporter and mu-opioid receptor binding potential. Separately, participants completed a modified two-step task, which quantifies the degree to which decision-making is influenced by goal-directed or habitual strategies. All participants completed a version with reward outcomes; healthy controls additionally completed a version with loss outcomes. In the context of rewarding outcomes, we found that greater serotonin transporter binding potential in prefrontal regions was associated with habitual control, while greater serotonin transporter binding potential in the putamen was marginally associated with goal-directed control; however, the findings were no longer significant when controlling for the opposing valence (loss). In blocks with loss outcomes, we found that the opioidergic system, specifically greater [11C]carfentanil binding potential, was positively associated with goal-directed control and negatively associated with habit-directed control. Our findings illuminate the complex neurochemical basis of goal-directed and habitual behavior, implicating differential roles for prefrontal and subcortical serotonin in decision-making across healthy and pathological populations.


Asunto(s)
Juego de Azar , Objetivos , Humanos , Motivación , Tomografía de Emisión de Positrones , Recompensa
7.
Neuroimage ; 183: 1-6, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30077742

RESUMEN

Animal studies have suggested that dopamine and opioid neurotransmitter systems interact in brain regions that are relevant for reward functions, but data in humans are very limited. The interaction is potentially important in disorders affecting these neurotransmitter systems, such as addiction. Here, we investigated whether subcortical µ-opioid receptor (MOR) availability and presynaptic dopamine synthesis capacity are correlated in the healthy human brain or in pathological gamblers (PGs) using positron emission tomography with 6-[18F]fluoro-l-dopa and [11C]carfentanil. The specificity of the findings was further investigated by including a serotonin transporter ligand, [11C]MADAM, as a negative control. Thirteen PG patients and 15 age-, sex- and weight-matched controls underwent the scans. In both groups, presynaptic dopamine synthesis capacity was associated with MOR availability in the putamen, caudate nucleus and globus pallidus. No similar associations were observed between dopamine synthesis capacity and [11C]MADAM binding, supporting a specific interplay between presynaptic dopamine neurotransmission and opioid receptor function in the basal ganglia. Correlations were similar between the groups, suggesting that the dopamine-opioid link is general and unaffected by behavioral addiction. The results provide in vivo human evidence of a connection between endogenous opioid and dopamine signaling in the brain.


Asunto(s)
Ganglios Basales/metabolismo , Dopamina/metabolismo , Juego de Azar/metabolismo , Tomografía de Emisión de Positrones/métodos , Receptores Opioides mu/metabolismo , Adulto , Ganglios Basales/diagnóstico por imagen , Femenino , Juego de Azar/diagnóstico por imagen , Humanos , Masculino , Persona de Mediana Edad , Trazadores Radiactivos , Radiofármacos , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo
8.
Hum Brain Mapp ; 39(10): 3972-3983, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29885086

RESUMEN

Physical exercise modulates food reward and helps control body weight. The endogenous µ-opioid receptor (MOR) system is involved in rewarding aspects of both food and physical exercise, yet interaction between endogenous opioid release following exercise and anticipatory food reward remains unresolved. Here we tested whether exercise-induced opioid release correlates with increased anticipatory reward processing in humans. We scanned 24 healthy lean men after rest and after a 1 h session of aerobic exercise with positron emission tomography (PET) using MOR-selective radioligand [11 C]carfentanil. After both PET scans, the subjects underwent a functional magnetic resonance imaging (fMRI) experiment where they viewed pictures of palatable versus nonpalatable foods to trigger anticipatory food reward responses. Exercise-induced changes in MOR binding in key regions of reward circuit (amygdala, thalamus, ventral and dorsal striatum, and orbitofrontal and cingulate cortices) were used to predict the changes in anticipatory reward responses in fMRI. Exercise-induced changes in MOR binding correlated negatively with the exercise-induced changes in neural anticipatory food reward responses in orbitofrontal and cingulate cortices, insula, ventral striatum, amygdala, and thalamus: higher exercise-induced opioid release predicted higher brain responses to palatable versus nonpalatable foods. We conclude that MOR activation following exercise may contribute to the considerable interindividual variation in food craving and consumption after exercise, which might promote compensatory eating and compromise weight control.


Asunto(s)
Anticipación Psicológica/fisiología , Encéfalo/fisiología , Ejercicio Físico/fisiología , Alimentos , Neuroimagen/métodos , Reconocimiento Visual de Modelos/fisiología , Receptores Opioides mu/metabolismo , Recompensa , Adulto , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Tomografía de Emisión de Positrones/métodos
9.
J Head Trauma Rehabil ; 33(1): 25-32, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28060207

RESUMEN

OBJECTIVE: To investigate quantitative positron emission tomography (PET) findings and to study whether the cholinergic function differs between respondents to cholinergic medication versus nonrespondents. SETTING: Outpatient clinic and university PET imaging center. PARTICIPANTS: We studied 17 subjects for more than 1 year after at least moderate traumatic brain injury. Ten of the subjects were respondents and 7 nonrespondents to cholinergic medication. DESIGN: Cholinergic function was assessed with [methyl-C] N-methylpiperidyl-4-acetate-PET (C-MP4A-PET), which reflects the activity of the acetylcholinesterase (AChE) enzyme. The subjects were PET scanned twice: without medication and after a 4-week treatment with rivastigmine 1.5 mg twice a day. MEASURES: Regional cerebral AChE activity was measured with PET. RESULTS: At baseline Statistical Parametric Mapping analyses showed significantly lower AChE activity in respondents bilaterally in the frontal cortex as compared with nonrespondents. Region of interest (ROI) analysis revealed that the difference was most pronounced in the lateral frontal cortex (-9.4%, P = .034) and anterior cingulate (-6.0%, P = .049). After rivastigmine treatment, AChE activity was notably lower throughout the cortex in both respondents and nonrespondents, without significant differences between them. CONCLUSION: Our study suggests that frontal cholinergic dysfunction is associated with the clinical response to cholinergic stimulation in patients with traumatic brain injury.


Asunto(s)
Acetilcolinesterasa/metabolismo , Lesiones Traumáticas del Encéfalo/diagnóstico por imagen , Lesiones Traumáticas del Encéfalo/enzimología , Inhibidores de la Colinesterasa/uso terapéutico , Trastornos del Conocimiento/tratamiento farmacológico , Rivastigmina/uso terapéutico , Adulto , Lesiones Traumáticas del Encéfalo/psicología , Enfermedad Crónica , Trastornos del Conocimiento/diagnóstico por imagen , Trastornos del Conocimiento/enzimología , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Tomografía de Emisión de Positrones
10.
Neuropsychopharmacology ; 43(2): 246-254, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28722022

RESUMEN

Central opioidergic mechanisms may modulate the positive effects of physical exercise such as mood elevation and stress reduction. How exercise intensity and concomitant effective changes affect central opioidergic responses is unknown. We studied the effects of acute physical exercise on the cerebral µ-opioid receptors (MOR) of 22 healthy recreationally active males using positron emission tomography (PET) and the MOR-selective radioligand [11C]carfentanil. MOR binding was measured in three conditions on separate days: after a 60-min aerobic moderate-intensity exercise session, after a high-intensity interval training (HIIT) session, and after rest. Mood was measured repeatedly throughout the experiment. HIIT significantly decreased MOR binding selectively in the frontolimbic regions involved in pain, reward, and emotional processing (thalamus, insula, orbitofrontal cortex, hippocampus, and anterior cingulate cortex). Decreased binding correlated with increased negative emotionality. Moderate-intensity exercise did not change MOR binding, although increased euphoria correlated with decreased receptor binding. These observations, consistent with endogenous opioid release, highlight the role of the µ-opioid system in mediating affective responses to high-intensity training as opposed to recreational moderate physical exercise.


Asunto(s)
Afecto/fisiología , Corteza Cerebral/metabolismo , Ejercicio Físico/fisiología , Entrenamiento de Intervalos de Alta Intensidad , Tomografía de Emisión de Positrones/métodos , Receptores Opioides mu/metabolismo , Tálamo/metabolismo , Analgésicos Opioides/metabolismo , Corteza Cerebral/diagnóstico por imagen , Fentanilo/análogos & derivados , Fentanilo/metabolismo , Giro del Cíngulo/diagnóstico por imagen , Giro del Cíngulo/metabolismo , Voluntarios Sanos , Hipocampo/diagnóstico por imagen , Hipocampo/metabolismo , Humanos , Masculino , Corteza Prefrontal/diagnóstico por imagen , Corteza Prefrontal/metabolismo , Tálamo/diagnóstico por imagen , Adulto Joven
11.
Eur Neuropsychopharmacol ; 27(12): 1281-1288, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29032922

RESUMEN

Behavioral addictions, such as pathological gambling (PG) and binge eating disorder (BED), appear to be associated with specific changes in brain dopamine and opioid function, but the role of other neurotransmitter systems is less clear. Given the crucial role of serotonin in a number of psychiatric disorders, we aimed to compare brain serotonergic function among individuals with BED, PG and healthy controls. Seven BED patients, 13 PG patients and 16 healthy controls were scanned with high-resolution positron emission tomography (PET) using the serotonin transporter (SERT) tracer [11C]MADAM. Both region-of-interest and voxel-wise whole brain analyses were performed. Patients with BED showed increased SERT binding in the parieto-occipital cortical regions compared to both PG and healthy controls, with parallel decreases in binding in the nucleus accumbens, inferior temporal gyrus and lateral orbitofrontal cortex. No differences between PG patients and controls were observed. None of the subjects were on SSRI medications at the time of imaging, and there were no differences in the level of depression between PG and BED patients. The results highlight differences in brain SERT binding between individuals with BED and PG and provide further evidence of different neurobiological underpinnings in behavioral addictions that are unrelated to the co-existing mood disorder. The results aid in the conceptualization of behavioral addictions by characterizing the underlying serotonin changes and provide a framework for additional studies to examine syndrome-specific pharmaceutical treatments.


Asunto(s)
Bencilaminas/metabolismo , Trastorno por Atracón/diagnóstico por imagen , Encéfalo/diagnóstico por imagen , Juego de Azar/diagnóstico por imagen , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Adulto , Análisis de Varianza , Trastorno por Atracón/patología , Femenino , Juego de Azar/patología , Humanos , Masculino , Persona de Mediana Edad , Tomografía de Emisión de Positrones
12.
J Neurosci ; 37(25): 6125-6131, 2017 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-28536272

RESUMEN

The size of human social networks significantly exceeds the network that can be maintained by social grooming or touching in other primates. It has been proposed that endogenous opioid release after social laughter would provide a neurochemical pathway supporting long-term relationships in humans (Dunbar, 2012), yet this hypothesis currently lacks direct neurophysiological support. We used PET and the µ-opioid-receptor (MOR)-specific ligand [11C]carfentanil to quantify laughter-induced endogenous opioid release in 12 healthy males. Before the social laughter scan, the subjects watched laughter-inducing comedy clips with their close friends for 30 min. Before the baseline scan, subjects spent 30 min alone in the testing room. Social laughter increased pleasurable sensations and triggered endogenous opioid release in thalamus, caudate nucleus, and anterior insula. In addition, baseline MOR availability in the cingulate and orbitofrontal cortices was associated with the rate of social laughter. In a behavioral control experiment, pain threshold-a proxy of endogenous opioidergic activation-was elevated significantly more in both male and female volunteers after watching laughter-inducing comedy versus non-laughter-inducing drama in groups. Modulation of the opioidergic activity by social laughter may be an important neurochemical pathway that supports the formation, reinforcement, and maintenance of human social bonds.SIGNIFICANCE STATEMENT Social contacts are vital to humans. The size of human social networks significantly exceeds the network that can be maintained by social grooming in other primates. Here, we used PET to show that endogenous opioid release after social laughter may provide a neurochemical mechanism supporting long-term relationships in humans. Participants were scanned twice: after a 30 min social laughter session and after spending 30 min alone in the testing room (baseline). Endogenous opioid release was stronger after laughter versus the baseline scan. Opioid receptor density in the frontal cortex predicted social laughter rates. Modulation of the opioidergic activity by social laughter may be an important neurochemical mechanism reinforcing and maintaining social bonds between humans.


Asunto(s)
Química Encefálica/fisiología , Endorfinas/metabolismo , Risa/fisiología , Medio Social , Adulto , Mapeo Encefálico , Femenino , Humanos , Masculino , Apego a Objetos , Placer , Tomografía de Emisión de Positrones , Receptores Opioides mu/efectos de los fármacos , Receptores Opioides mu/metabolismo , Adulto Joven
13.
Neuropsychopharmacology ; 42(5): 1169-1177, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27882998

RESUMEN

Although behavioral addictions share many clinical features with drug addictions, they show strikingly large variation in their behavioral phenotypes (such as in uncontrollable gambling or eating). Neurotransmitter function in behavioral addictions is poorly understood, but has important implications in understanding its relationship with substance use disorders and underlying mechanisms of therapeutic efficacy. Here, we compare opioid and dopamine function between two behavioral addiction phenotypes: pathological gambling (PG) and binge eating disorder (BED). Thirty-nine participants (15 PG, 7 BED, and 17 controls) were scanned with [11C]carfentanil and [18F]fluorodopa positron emission tomography using a high-resolution scanner. Binding potentials relative to non-displaceable binding (BPND) for [11C]carfentanil and influx rate constant (Ki) values for [18F]fluorodopa were analyzed with region-of-interest and whole-brain voxel-by-voxel analyses. BED subjects showed widespread reductions in [11C]carfentanil BPND in multiple subcortical and cortical brain regions and in striatal [18F]fluorodopa Ki compared with controls. In PG patients, [11C]carfentanil BPND was reduced in the anterior cingulate with no differences in [18F]fluorodopa Ki compared with controls. In the nucleus accumbens, a key region involved in reward processing, [11C]Carfentanil BPND was 30-34% lower and [18F]fluorodopa Ki was 20% lower in BED compared with PG and controls (p<0.002). BED and PG are thus dissociable as a function of dopaminergic and opioidergic neurotransmission. Compared with PG, BED patients show widespread losses of mu-opioid receptor availability together with presynaptic dopaminergic defects. These findings highlight the heterogeneity underlying the subtypes of addiction and indicate differential mechanisms in the expression of pathological behaviors and responses to treatment.


Asunto(s)
Analgésicos Opioides/metabolismo , Conducta Adictiva/metabolismo , Encéfalo/metabolismo , Bulimia/metabolismo , Dopamina/metabolismo , Juego de Azar/metabolismo , Adulto , Analgésicos Opioides/administración & dosificación , Conducta Adictiva/diagnóstico por imagen , Encéfalo/diagnóstico por imagen , Bulimia/diagnóstico por imagen , Radioisótopos de Carbono , Dihidroxifenilalanina/administración & dosificación , Dihidroxifenilalanina/análogos & derivados , Femenino , Fentanilo/administración & dosificación , Fentanilo/análogos & derivados , Juego de Azar/diagnóstico por imagen , Humanos , Masculino , Persona de Mediana Edad , Tomografía de Emisión de Positrones , Radiofármacos
14.
Brain Res ; 1652: 97-102, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27693414

RESUMEN

Intravenous use of a psychostimulant drug containing methcathinone (ephedrone) and manganese causes an irreversible extrapyramidal syndrome in drug abusers. We aimed to reproduce the syndrome in mice to evaluate dopaminergic damage. C57/B6 mice were intraperitoneally injected once a day with the study drug or saline for a period of 27 weeks. Motor activity was recorded in an automated motility-box. After 13 and 27 weeks of treatment, ex vivo digital autoradiography was performed using [11C]dihydrotetrabenazine ([11C]DTBZ). After 27 weeks of treatment [11C]DTBZ autoradiography demonstrated a significant increase in the striatum-to-cerebellum binding ratio compared with saline treated controls. At the same time point, there was no evident change in motor activity. Increased [11C]DTBZ binding may indicate vesicular monoamine transporter type 2 (VMAT2) function is altered. The lack of extrapyramidal symptoms in animals could be attributed to low dosing regimen or high metabolic rate.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Manganeso/toxicidad , Propiofenonas/toxicidad , Psicotrópicos/toxicidad , Trastornos Relacionados con Sustancias/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/metabolismo , Animales , Autorradiografía , Fenómenos Biomecánicos , Radioisótopos de Carbono , Estimulantes del Sistema Nervioso Central/toxicidad , Cuerpo Estriado/diagnóstico por imagen , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Marcha/efectos de los fármacos , Marcha/fisiología , Drogas Ilícitas/toxicidad , Inyecciones Intraperitoneales , Masculino , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Tomografía de Emisión de Positrones , Radiofármacos , Distribución Aleatoria , Trastornos Relacionados con Sustancias/diagnóstico por imagen , Tetrabenazina/análogos & derivados , Factores de Tiempo
15.
Neuroimage ; 138: 242-247, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27238727

RESUMEN

In non-human primates, opioid-receptor blockade increases social grooming, and the endogenous opioid system has therefore been hypothesized to support maintenance of long-term relationships in humans as well. Here we tested whether social touch modulates opioidergic activation in humans using in vivo positron emission tomography (PET). Eighteen male participants underwent two PET scans with [11C]carfentanil, a ligand specific to µ-opioid receptors (MOR). During the social touch scan, the participants lay in the scanner while their partners caressed their bodies in a non-sexual fashion. In the baseline scan, participants lay alone in the scanner. Social touch triggered pleasurable sensations and increased MOR availability in the thalamus, striatum, and frontal, cingulate, and insular cortices. Modulation of activity of the opioid system by social touching might provide a neurochemical mechanism reinforcing social bonds between humans.


Asunto(s)
Encéfalo/metabolismo , Apego a Objetos , Placer/fisiología , Receptores Opioides mu/metabolismo , Conducta Social , Percepción del Tacto/fisiología , Tacto/fisiología , Adulto , Femenino , Humanos , Masculino , Imagen Molecular/métodos , Tomografía de Emisión de Positrones/métodos
16.
Synapse ; 70(2): 57-65, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26562363

RESUMEN

PET imaging can for some neurotransmitters be used to measure synaptic neurotransmitter concentrations. The objective of this study was to test whether the receptor binding of the α2C -AR antagonist PET tracer [(11)C]ORM-13070 would increase in response to reductions in synaptic noradrenaline, evoked by dexmedetomidine as a sympatholytic drug challenge. Six subjects underwent a control PET scan and two dexmedetomidine PET scans. Dexmedetomidine was infused with target plasma concentrations of 0.6 and 0.2 ng/ml. Tracer binding was measured by voxel-based analysis of bound per free (B/F) images. ROI-based analysis was performed in the dorsal striatum and in the thalamus. Vital signs and drug concentrations in plasma were measured and the sedative effect was estimated with the visual analog scale. In the voxel-based analysis, dexmedetomidine administration was associated with a tendency to increased B/F tracer in the right thalamus (mean, +17%, P = 0.14, and +19%, P = 0.05, with the low and high dose, respectively). Tracer binding in the dorsal striatum was unaffected by dexmedetomidine. A cluster with significantly increased B/F tracer (+42%, P = 0.01) was seen in the right superior temporal gyrus with low-dose dexmedetomidine, but not after the high dose. Brain uptake of [(11)C]ORM-13070 has previously been shown to be reduced in conditions of increased synaptic noradrenaline concentrations. In this study, tracer binding in the thalamus tended to increase in accordance with reduced activity of noradrenergic projections from the locus coeruleus, but statistical significance was not reached.


Asunto(s)
Analgésicos no Narcóticos/farmacología , Encéfalo/metabolismo , Dexmedetomidina/farmacología , Dioxanos/farmacocinética , Norepinefrina/metabolismo , Piperazinas/farmacocinética , Radiofármacos/farmacocinética , Analgésicos no Narcóticos/efectos adversos , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Dexmedetomidina/efectos adversos , Humanos , Masculino , Tomografía de Emisión de Positrones , Distribución Tisular
17.
Psychopharmacology (Berl) ; 232(21-22): 4169-78, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25918111

RESUMEN

RATIONALE: No validated methods have been available for studying brain noradrenergic neurotransmission in vivo in humans. Positron emission tomography (PET) radiotracers are widely used in clinical drug development targeted to brain receptors and can also in some cases be employed to monitor extracellular (synaptic) neurotransmitter concentrations. OBJECTIVES: The objective of this study is to test the sensitivity of [(11)C]ORM-13070 uptake to increased concentrations of extracellular (synaptic) noradrenaline in the human brain. METHODS: Eight subjects underwent a control PET scan with [(11)C]ORM-13070, a subtype-selective α2C-adrenoceptor antagonist radioligand, and two PET scans after two different noradrenaline challenges, i.e. during ketamine infusion and after a dose of atomoxetine combined with cold stimulation. Tracer uptake in the caudate nucleus and putamen was described with AUC values in scan time windows of 10-20 and 5-30 min post injection and quantified with the ratio method. Voxel-based analysis was performed with average bound per free (B/F) ratio images. RESULTS: Both noradrenaline challenges were consistently associated with 10-20 % (p < 0.05) reductions in tracer uptake in the dorsal striatum, as determined with region-of-interest-based analysis. Voxel-based analysis revealed significant reductions in B/F ratios in the dorsal striatum, in the brain stem and in several cortical areas. Reductions of 24 and 23 % were detected in the peak putamen clusters with ketamine and atomoxetine + cold, respectively. CONCLUSION: Direct experimental support was gained for the suitability of [(11)C]ORM-13070 for imaging of brain noradrenergic neurotransmission.


Asunto(s)
Núcleo Caudado/metabolismo , Norepinefrina/metabolismo , Putamen/metabolismo , Adulto , Núcleo Caudado/diagnóstico por imagen , Humanos , Masculino , Tomografía de Emisión de Positrones/métodos , Putamen/diagnóstico por imagen , Radiofármacos/metabolismo , Adulto Joven
19.
Eur J Nucl Med Mol Imaging ; 42(1): 120-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25201008

RESUMEN

PURPOSE: α2C-Adrenoceptors share inhibitory presynaptic functions with the more abundant α2A-adrenoceptor subtype, but they also have widespread postsynaptic modulatory functions in the brain. Research on the noradrenergic system of the human brain has been hampered by the lack of suitable PET tracers targeted to the α2-adrenoceptor subtypes. METHODS: PET imaging with the specific α2C-adrenoceptor antagonist tracer [(11)C]ORM-13070 was performed twice in six healthy male subjects to investigate the test-retest reliability of tracer binding. RESULTS: The bound/free ratio of tracer uptake relative to nonspecific uptake into the cerebellum during the time interval of 5 - 30 min was most prominent in the dorsal striatum: 0.77 in the putamen and 0.58 in the caudate nucleus. Absolute test-retest variability in bound/free ratios of tracer ranged from 4.3 % in the putamen to 29 % in the hippocampus. Variability was also <10 % in the caudate nucleus and thalamus. Intraclass correlation coefficients (ICC) ranged from 0.50 in the hippocampus to 0.89 in the thalamus (ICC >0.70 was also reached in the caudate nucleus, putamen, lateral frontal cortex and parietal cortex). The pattern of [(11)C]ORM-13070 binding, as determined by PET, was in good agreement with receptor density results previously derived from post-mortem autoradiography. PET data analysis results obtained with a compartmental model fit, the simplified reference tissue model and a graphical reference tissue analysis method were convergent with the tissue ratio method. CONCLUSION: The results of this study support the use of [(11)C]ORM-13070 PET in the quantitative assessment of α2C-adrenoceptors in the human brain in vivo. Reliable assessment of specific tracer binding in the dorsal striatum is possible with the help of reference tissue ratios.


Asunto(s)
Encéfalo/diagnóstico por imagen , Dioxanos/farmacocinética , Piperazinas/farmacocinética , Tomografía de Emisión de Positrones , Radiofármacos/farmacocinética , Receptores Adrenérgicos alfa 2/metabolismo , Adulto , Humanos , Masculino , Reproducibilidad de los Resultados , Distribución Tisular
20.
J Cereb Blood Flow Metab ; 35(1): 157-65, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25370856

RESUMEN

[(11)C]TMSX ([7-N-methyl-(11)C]-(E)-8-(3,4,5-trimethoxystyryl)-1,3,7-trimethylxanthine) is a selective adenosine A2A receptor (A2AR) radioligand. In the central nervous system (CNS), A2AR are linked to dopamine D2 receptor function in striatum, but they are also important modulators of inflammation. The golden standard for kinetic modeling of brain [(11)C]TMSX positron emission tomography (PET) is to obtain arterial input function via arterial blood sampling. However, this method is laborious, prone to errors and unpleasant for study subjects. The aim of this work was to evaluate alternative input function acquisition methods for brain [(11)C]TMSX PET imaging. First, a noninvasive, automated method for the extraction of gray matter reference region using supervised clustering (SCgm) was developed. Second, a method for obtaining a population-based arterial input function (PBIF) was implemented. These methods were created using data from 28 study subjects (7 healthy controls, 12 multiple sclerosis patients, and 9 patients with Parkinson's disease). The results with PBIF correlated well with original plasma input, and the SCgm yielded similar results compared with cerebellum as a reference region. The clustering method for extracting reference region and the population-based approach for acquiring input for dynamic [(11)C]TMSX brain PET image analyses appear to be feasible and robust methods, that can be applied in patients with CNS pathology.


Asunto(s)
Encéfalo/metabolismo , Esclerosis Múltiple/metabolismo , Enfermedad de Parkinson/metabolismo , Tomografía de Emisión de Positrones/métodos , Receptor de Adenosina A2A/metabolismo , Xantinas , Adulto , Encéfalo/diagnóstico por imagen , Mapeo Encefálico , Radioisótopos de Carbono , Estudios de Casos y Controles , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Ligandos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/diagnóstico por imagen , Enfermedad de Parkinson/diagnóstico por imagen , Unión Proteica , Ensayo de Unión Radioligante , Valores de Referencia , Reproducibilidad de los Resultados , Distribución Tisular , Xantinas/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...