Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Neuroimage ; 297: 120716, 2024 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-38955254

RESUMEN

MAO-A catalyzes the oxidative degradation of monoamines and is thus implicated in sex-specific neuroplastic processes that influence gray matter (GM) density (GMD) and microstructure (GMM). Given the exact monitoring of plasma hormone levels and sex steroid intake, transgender individuals undergoing gender-affirming hormone therapy (GHT) represent a valuable cohort to potentially investigate sex steroid-induced changes of GM and concomitant MAO-A density. Here, we investigated the effects of GHT over a median time period of 4.5 months on GMD and GMM as well as MAO-A distribution volume. To this end, 20 cisgender women, 11 cisgender men, 20 transgender women and 10 transgender men underwent two MRI scans in a longitudinal design. PET scans using [11C]harmine were performed before each MRI session in a subset of 35 individuals. GM changes determined by diffusion weighted imaging (DWI) metrics for GMM and voxel based morphometry (VBM) for GMD were estimated using repeated measures ANOVA. Regions showing significant changes of both GMM and GMD were used for the subsequent analysis of MAO-A density. These involved the fusiform gyrus, rolandic operculum, inferior occipital cortex, middle and anterior cingulum, bilateral insula, cerebellum and the lingual gyrus (post-hoc tests: pFWE+Bonferroni < 0.025). In terms of MAO-A distribution volume, no significant effects were found. Additionally, the sexual desire inventory (SDI) was applied to assess GHT-induced changes in sexual desire, showing an increase of SDI scores among transgender men. Changes in the GMD of the bilateral insula showed a moderate correlation to SDI scores (rho = - 0.62, pBonferroni = 0.047). The present results are indicative of a reliable influence of gender-affirming hormone therapy on 1) GMD and GMM following an interregional pattern and 2) sexual desire specifically among transgender men.

2.
Hum Brain Mapp ; 44(6): 2654-2663, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36840505

RESUMEN

Seasonal changes in neurotransmitter systems have been demonstrated in imaging studies and are especially noticeable in diseased states such as seasonal affective disorder (SAD). These modulatory neurotransmitters, such as serotonin, are influencing glutamatergic and GABAergic neurotransmission. Furthermore, central components of the circadian pacemaker are regulated by GABA (the suprachiasmatic nucleus) or glutamate (e.g., the retinohypothalamic tract). Therefore, we explored seasonal differences in the GABAergic and glutamatergic system in 159 healthy individuals using magnetic resonance spectroscopy imaging with a GABA-edited 3D-MEGA-LASER sequence at 3T. We quantified GABA+/tCr, GABA+/Glx, and Glx/tCr ratios (GABA+, GABA+ macromolecules; Glx, glutamate + glutamine; tCr, total creatine) in five different subcortical brain regions. Differences between time periods throughout the year, seasonal patterns, and stationarity were tested using ANCOVA models, curve fitting approaches, and unit root and stationarity tests, respectively. Finally, Spearman correlation analyses between neurotransmitter ratios within each brain region and cumulated daylight and global radiation were performed. No seasonal or monthly differences, seasonal patterns, nor significant correlations could be shown in any region or ratio. Unit root and stationarity tests showed stable patterns of GABA+/tCr, GABA+/Glx, and Glx/tCr levels throughout the year, except for hippocampal Glx/tCr. Our results indicate that neurotransmitter levels of glutamate and GABA in healthy individuals are stable throughout the year. Hence, despite the important correction for age and gender in the analyses of MRS derived GABA and glutamate, a correction for seasonality in future studies does not seem necessary. Future investigations in SAD and other psychiatric patients will be of high interest.


Asunto(s)
Ácido Glutámico , Glutamina , Humanos , Espectroscopía de Resonancia Magnética/métodos , Estaciones del Año , Imagen por Resonancia Magnética/métodos , Encéfalo/diagnóstico por imagen , Ácido gamma-Aminobutírico/análisis , Neurotransmisores , Receptores de Antígenos de Linfocitos T
3.
Cereb Cortex ; 32(16): 3516-3524, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34952543

RESUMEN

The monoamine oxidase A (MAO-A) is integral to monoamine metabolism and is thus relevant to the pathophysiology of various neuropsychiatric disorders; however, associated gene-enzyme relations are not well understood. This study aimed to unveil genes coexpressed with MAO-A. Therefore, 18 179 mRNA expression maps (based on the Allen Human Brain Atlas) were correlated with the cerebral distribution volume (VT) of MAO-A assessed in 36 healthy subjects (mean age ± standard deviation: 32.9 ± 8.8 years, 18 female) using [11C]harmine positron emission tomography scans. Coexpression analysis was based on Spearman's ρ, over-representation tests on Fisher's exact test with false discovery rate (FDR) correction. The analysis revealed 35 genes in cortex (including B-cell translocation gene family, member 3, implicated in neuroinflammation) and 247 genes in subcortex (including kallikrein-related peptidase 10, implicated in Alzheimer's disease). Significantly over-represented Gene Ontology terms included "neuron development", "neuron differentiation", and "cell-cell signaling" as well as "axon" and "neuron projection". In vivo MAO-A enzyme distribution and MAOA expression did not correlate in cortical areas (ρ = 0.08) while correlation was found in subcortical areas (ρ = 0.52), suggesting influences of region-specific post-transcriptional and -translational modifications. The herein reported information could contribute to guide future genetic studies, deepen the understanding of associated pathomechanisms and assist in the pursuit of novel therapeutic targets.


Asunto(s)
Encéfalo , Monoaminooxidasa , Tomografía de Emisión de Positrones , Adulto , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Radioisótopos de Carbono , Femenino , Harmina/metabolismo , Humanos , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , Tomografía de Emisión de Positrones/métodos
4.
Mol Psychiatry ; 26(3): 1009-1018, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-31383926

RESUMEN

Attention deficit hyperactivity disorder (ADHD) is a common neurodevelopmental disorder with a robust genetic influence. The norepinephrine transporter (NET) is of particular interest as it is one of the main targets in treatment of the disorder. As ADHD is a complex and polygenetic condition, the possible regulation by epigenetic processes has received increased attention. We sought to determine possible differences in NET promoter DNA methylation between patients with ADHD and healthy controls. DNA methylation levels in the promoter region of the NET were determined in 23 adult patients with ADHD and 23 healthy controls. A subgroup of 18 patients with ADHD and 18 healthy controls underwent positron emission tomography (PET) with the radioligand (S,S)-[18F]FMeNER-D2 to quantify the NET in several brain areas in vivo. Analyses revealed significant differences in NET methylation levels at several cytosine-phosphate-guanine (CpG) sites between groups. A defined segment of the NET promoter ("region 1") was hypermethylated in patients in comparison with controls. In ADHD patients, a negative correlation between methylation of a CpG site in this region and NET distribution in the thalamus, locus coeruleus, and the raphe nuclei was detected. Furthermore, methylation of several sites in region 1 was negatively associated with the severity of hyperactivity-impulsivity symptoms. Our results point to an epigenetic dysregulation in ADHD, possibly due to a compensatory mechanisms or additional factors involved in transcriptional processing.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática , Adulto , Trastorno por Déficit de Atención con Hiperactividad/genética , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Humanos , Conducta Impulsiva , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Tomografía de Emisión de Positrones
5.
Neuroimage ; 236: 118039, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-33852940

RESUMEN

Animal studies using selective serotonin reuptake inhibitors (SSRIs) and learning paradigms have demonstrated that serotonin is important for flexibility in executive functions and learning. SSRIs might facilitate relearning through neuroplastic processes and thus exert their clinical effects in psychiatric diseases where cognitive functioning is affected. However, translation of these mechanisms to humans is missing. In this randomized placebo-controlled trial, we assessed functional brain activation during learning and memory retrieval in healthy volunteers performing associative learning tasks aiming to translate facilitated relearning by SSRIs. To this extent, seventy-six participants underwent three MRI scanning sessions: (1) at baseline, (2) after three weeks of daily associative learning and subsequent retrieval (face-matching or Chinese character-noun matching) and (3) after three weeks of relearning under escitalopram (10 mg/day) or placebo. Associative learning and retrieval tasks were performed during each functional MRI (fMRI) session. Statistical modeling was done using a repeated-measures ANOVA, to test for content-by-treatment-by-time interaction effects. During the learning task, a significant substance-by-time interaction was found in the right insula showing a greater deactivation in the SSRI cohort after 21 days of relearning compared to the learning phase. In the retrieval task, there was a significant content-by-time interaction in the left angular gyrus (AG) with an increased activation in face-matching compared to Chinese-character matching for both learning and relearning phases. A further substance-by-time interaction was found in task performance after 21 days of relearning, indicating a greater decrease of performance in the placebo group. Our findings that escitalopram modulate insula activation demonstrates successful translation of relearning as a mechanism of SSRIs in human. Furthermore, we show that the left AG is an active component of correct memory retrieval, which coincides with previous literature. We extend the function of this region by demonstrating its activation is not only stimulus dependent but also time constrained. Finally, we were able to show that escitalopram aids in relearning, irrespective of content.


Asunto(s)
Aprendizaje por Asociación/efectos de los fármacos , Corteza Cerebral , Citalopram/farmacología , Recuerdo Mental/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Adulto , Mapeo Encefálico , Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/fisiología , Citalopram/administración & dosificación , Método Doble Ciego , Femenino , Humanos , Estudios Longitudinales , Imagen por Resonancia Magnética , Masculino , Lóbulo Parietal/diagnóstico por imagen , Lóbulo Parietal/efectos de los fármacos , Lóbulo Parietal/fisiología , Reconocimiento Visual de Modelos/fisiología , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Adulto Joven
6.
Neuroimage ; 232: 117913, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33657450

RESUMEN

Impaired cognitive flexibility represents a widespread symptom in psychiatric disorders, including major depressive disorder (MDD), a disease, characterized by an imbalance of neurotransmitter concentrations. While memory formation is mostly associated with glutamate, also gamma-Aminobutyric acid (GABA) and serotonin show attributions in a complex interplay between neurotransmitter systems. Treatment with selective serotonin reuptake inhibitors (SSRIs) does not solely affect the serotonergic system but shows downstream effects on GABA- and glutamatergic neurotransmission, potentially helping to restore cognitive function via neuroplastic effects. Hence, this study aims to elaborate the effects of associative relearning and SSRI treatment on GABAergic and glutamatergic function within and between five brain regions using magnetic resonance spectroscopy imaging (MRSI). In this study, healthy subjects were randomized into four groups which underwent three weeks of an associative relearning paradigm, with or without emotional connotation, under SSRI (10mg escitalopram) or placebo administration. MRSI measurements, using a spiral-encoded, 3D-GABA-edited MEGA-LASER sequence at 3T, were performed on the first and last day of relearning. Mean GABA+/tCr (GABA+ = GABA + macromolecules; tCr = total creatine) and Glx/tCr (Glx = glutamate + glutamine) ratios were quantified in a ROI-based approach for the hippocampus, insula, putamen, pallidum and thalamus, using LCModel. A total of 66 subjects ((37 female, mean age ± SD = 25.4±4.7) for Glx/tCr and 58 subjects (32 female, mean age ± SD = 25.1±4.7) for GABA+/tCr were included in the final analysis. A significant measurement by region and treatment (SSRI vs placebo) interaction on Glx/tCr ratios was found (pcor=0.017), with post hoc tests confirming differential effects on hippocampus and thalamus (pcor=0.046). Moreover, treatment by time comparison, for each ROI independently, showed a reduction of hippocampal Glx/tCr ratios after SSRI treatment (puncor=0.033). No significant treatment effects on GABA+/tCr ratios or effects of relearning condition on any neurotransmitter ratio could be found. Here, we showed a significant SSRI- and relearning-driven interaction effect of hippocampal and thalamic Glx/tCr levels, suggesting differential behavior based on different serotonin transporter and receptor densities. Moreover, an indication for Glx/tCr adaptions in the hippocampus after three weeks of SSRI treatment could be revealed. Our findings are in line with animal studies reporting glutamate adaptions in the hippocampus following chronic SSRI intake. Due to the complex interplay of serotonin and hippocampal function, involving multiple serotonin receptor subtypes on glutamatergic cells and GABAergic interneurons, the interpretation of underlying neurobiological actions remains challenging.


Asunto(s)
Aprendizaje por Asociación/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Ácido Glutámico/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Ácido gamma-Aminobutírico/metabolismo , Adulto , Aprendizaje por Asociación/fisiología , Encéfalo/diagnóstico por imagen , Método Doble Ciego , Femenino , Humanos , Estudios Longitudinales , Imagen por Resonancia Magnética/métodos , Espectroscopía de Resonancia Magnética/métodos , Masculino , Estimulación Luminosa/métodos , Adulto Joven
7.
Int J Neuropsychopharmacol ; 24(7): 542-550, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-33667309

RESUMEN

BACKGROUND: Selective serotonin reuptake inhibitors (SSRIs) are predominantly prescribed for people suffering from major depressive disorder. These antidepressants exert their effects by blocking the serotonin transporter (SERT), leading to increased levels of serotonin in the synaptic cleft and subsequently to an attenuation of depressive symptoms and elevation in mood. Although long-term studies investigating white matter (WM) alterations after exposure to antidepressant treatment exist, results on the acute effects on the brain's WM microstructure are lacking. METHODS: In this interventional longitudinal study, 81 participants were included (33 patients and 48 healthy controls). All participants underwent diffusion weighted imaging on 2 separate days, receiving either citalopram or placebo using a randomized, double-blind, cross-over design. Fractional anisotropy, mean diffusivity, axial diffusivity, and radial diffusivity were calculated within the FMRIB software library and analyzed using tract-based spatial statistics. RESULTS: The repeated-measures ANOVA model revealed significant decreases after SSRI administration in mean diffusivity, axial diffusivity, and radial diffusivity regardless of the group (P < .05, family-wise error [FWE] corrected). Results were predominantly evident in frontal WM regions comprising the anterior corona radiata, corpus callosum, and external capsule and in distinct areas of the frontal blade. No increases in diffusivity were found, and no changes in fractional anisotropy were present. CONCLUSIONS: Our investigation provides the first evidence, to our knowledge, that fast WM microstructure adaptations within 1 hour after i.v. SSRI administration precede elevations in mood due to SSRI treatment. These results add a new facet to the complex mode of action of antidepressant therapy. This study was registered at clinicaltrials.gov with the identifier NCT02711215.


Asunto(s)
Trastorno Depresivo Mayor/tratamiento farmacológico , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Sustancia Blanca/efectos de los fármacos , Adulto , Trastorno Depresivo Mayor/diagnóstico por imagen , Imagen de Difusión Tensora , Femenino , Humanos , Estudios Longitudinales , Masculino , Sustancia Blanca/diagnóstico por imagen , Adulto Joven
8.
Neuroimage ; 209: 116497, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31899285

RESUMEN

Acute stress is often evoked during social interactions, by feelings of threat or negative evaluation by other people. We also constantly interact with others while under stress - in the workplace or in private alike. However, it is not clear how stress affects social interactions. For one, individuals could become more selfish and focused on their own goals. On the other hand, individuals might also become more focused on affiliating with potential social partners, in order to secure their support. There is, indeed, accumulating behavioral evidence that prosocial behaviors increase rather than decrease under stress. Here, we tested the underlying brain processes of such findings, by assessing the effects of stress on the neural representations of (monetary) value for self and other. Participants (N â€‹= â€‹30; male, 18-40 years) played a gambling task for themselves and for another participant while undergoing functional magnetic resonance imaging (fMRI). Each participant played the gambling task twice: once immediately following acute stress induction, and once in a control session. We compared neural patterns of value representation in the dorsomedial prefrontal cortex (dmPFC), ventromedial prefrontal cortex (vmPFC) and striatum using representational similarity analysis (RSA). We found that under stress, dmPFC and striatum showed higher dissimilarity between neural patterns underlying high and low value for the other. Dissimilarity of neural patterns underlying high and low value for the self was unaffected by stress. These findings suggest that participants track the magnitude of possible rewards for others more under stress, suggesting increased prosocial orientation.


Asunto(s)
Mapeo Encefálico , Conducta Cooperativa , Cuerpo Estriado/fisiología , Corteza Prefrontal/fisiología , Desempeño Psicomotor/fisiología , Recompensa , Estrés Psicológico/fisiopatología , Adolescente , Adulto , Cuerpo Estriado/diagnóstico por imagen , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Corteza Prefrontal/diagnóstico por imagen , Estrés Psicológico/diagnóstico por imagen , Adulto Joven
9.
Pharmazie ; 75(10): 456-462, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33305717

RESUMEN

Type 2 diabetes mellitus is characterized by insulin resistance and elevated blood glucose levels. Treatment protocols generally include dietary restriction of sugar, as well as drugs aiming at a reduction of blood glucose, mainly by activating the insulin system or supplementing insulin. This established approach does not take into account the outstanding physiological role of glucose as a key molecule in metabolism. Glucose is crucial to meet the high energy demand of the brain, which depends on it as an exclusive nutrient. Insulin independent glucose transporters GLUT1 import glucose into the brain. Reduction of blood glucose, as in current treatment concepts, may lead to energy deficiency in the brain and consecutively to worsening of - possibly already impaired - neurocognitive function. Reduced cell membrane fluidity of the vascular endothelium of the bloodbrain-barrier (BBB) - due to malnutrition and/or aging - is considered a major factor in pathogenesis of the cerebral metabolic syndrome, which is a key step in neurodegeneration. Under this aspect we suggest a novel approach to prophylaxis and treatment focusing on a sufficient supply of glucose to the brain.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Tipo 2/terapia , Azúcares de la Dieta/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Glucosa/metabolismo , Humanos , Hipoglucemiantes/farmacología , Insulina/metabolismo , Resistencia a la Insulina
10.
Acta Psychiatr Scand ; 140(5): 426-434, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31369144

RESUMEN

INTRODUCTION: Continuation electroconvulsive therapy (c-ECT) is highly effective for the prevention of depressive symptom relapse. There is a lack of understanding, about how c-ECT works in humans, particularly with regard to its effects on brain derived neurotrophic factor (BDNF) concentrations. Here, we aimed to close a gap in the literature by evaluating BDNF levels in patients receiving c-ECT. METHODS: We included 13 patients with either unipolar or bipolar depression (mean age ± SD: 55.5 ± 17.1; f/m: 10/3; unipolar/bipolar: 10/3) who received between one and four c-ECT (average per patient: 2.8). Serum BDNF (sBDNF) levels were assessed before and after each c-ECT sessions. Clinical assessments were also administered both before and after treatment. RESULTS: Our analysis revealed a significant increase in sBDNF after each treatment (c-ECT 1-3: P < 0.001, c-ECT 4: P = 0.018). The application of multiple c-ECT treatments was not, however, associated with further sBDNF enhancements. Psychometric scores were not significantly altered following c-ECT. DISCUSSION: An increase in sBDNF concentrations subsequent to c-ECT parallel data from the animal literature, which has linked regularly applied electrical stimulation to neuroplastic processes. This finding suggests a relationship between ECT-induced sBDNF concentrations and (sustained) remission status, considering a stable clinical condition across c-ECT.


Asunto(s)
Trastorno Bipolar/sangre , Trastorno Bipolar/terapia , Factor Neurotrófico Derivado del Encéfalo/sangre , Trastorno Depresivo/sangre , Trastorno Depresivo/terapia , Terapia Electroconvulsiva , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Prevención Secundaria , Adulto Joven
11.
Acta Psychiatr Scand ; 139(1): 78-88, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30291625

RESUMEN

OBJECTIVES: Clinical variables were investigated in the 'treatment resistant depression (TRD)- III' sample to replicate earlier findings by the European research consortium 'Group for the Study of Resistant Depression' (GSRD) and enable cross-sample prediction of treatment outcome in TRD. EXPERIMENTAL PROCEDURES: TRD was defined by a Montgomery and Åsberg Depression Rating Scale (MADRS) score ≥22 after at least two antidepressive trials. Response was defined by a decline in MADRS score by ≥50% and below a threshold of 22. Logistic regression was applied to replicate predictors for TRD among 16 clinical variables in 916 patients. Elastic net regression was applied for prediction of treatment outcome. RESULTS: Symptom severity (odds ratio (OR) = 3.31), psychotic symptoms (OR = 2.52), suicidal risk (OR = 1.74), generalized anxiety disorder (OR = 1.68), inpatient status (OR = 1.65), higher number of antidepressants administered previously (OR = 1.23), and lifetime depressive episodes (OR = 1.15) as well as longer duration of the current episode (OR = 1.022) increased the risk of TRD. Prediction of TRD reached an accuracy of 0.86 in the independent validation set, TRD-I. CONCLUSION: Symptom severity, suicidal risk, higher number of lifetime depressive episodes, and comorbid anxiety disorder were replicated as the most prominent risk factors for TRD. Significant predictors in TRD-III enabled robust prediction of treatment outcome in TRD-I.


Asunto(s)
Antidepresivos/farmacología , Trastorno Depresivo Resistente al Tratamiento/diagnóstico , Trastorno Depresivo Resistente al Tratamiento/psicología , Adulto , Trastornos Psicóticos Afectivos/diagnóstico , Trastornos Psicóticos Afectivos/psicología , Anciano , Antidepresivos/administración & dosificación , Antidepresivos/uso terapéutico , Trastornos de Ansiedad/diagnóstico , Trastornos de Ansiedad/psicología , Reglas de Decisión Clínica , Estudios Transversales , Trastorno Depresivo Resistente al Tratamiento/epidemiología , Episodio de Atención , Europa (Continente)/epidemiología , Femenino , Humanos , Pacientes Internos/psicología , Pacientes Internos/estadística & datos numéricos , Masculino , Persona de Mediana Edad , Escalas de Valoración Psiquiátrica , Estudios Retrospectivos , Riesgo , Índice de Severidad de la Enfermedad , Ideación Suicida , Resultado del Tratamiento
12.
Cereb Cortex ; 27(1): 117-130, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27909009

RESUMEN

Regional differences in posttranscriptional mechanisms may influence in vivo protein densities. The association of positron emission tomography (PET) imaging data from 112 healthy controls and gene expression values from the Allen Human Brain Atlas, based on post-mortem brains, was investigated for key serotonergic proteins. PET binding values and gene expression intensities were correlated for the main inhibitory (5-HT1A) and excitatory (5-HT2A) serotonin receptor, the serotonin transporter (SERT) as well as monoamine oxidase-A (MAO-A), using Spearman's correlation coefficients (rs) in a voxel-wise and region-wise analysis. Correlations indicated a strong linear relationship between gene and protein expression for both the 5-HT1A (voxel-wise rs = 0.71; region-wise rs = 0.93) and the 5-HT2A receptor (rs = 0.66; 0.75), but only a weak association for MAO-A (rs = 0.26; 0.66) and no clear correlation for SERT (rs = 0.17; 0.29). Additionally, region-wise correlations were performed using mRNA expression from the HBT, yielding comparable results (5-HT1Ars = 0.82; 5-HT2Ars = 0.88; MAO-A rs = 0.50; SERT rs = -0.01). The SERT and MAO-A appear to be regulated in a region-specific manner across the whole brain. In contrast, the serotonin-1A and -2A receptors are presumably targeted by common posttranscriptional processes similar in all brain areas suggesting the applicability of mRNA expression as surrogate parameter for density of these proteins.


Asunto(s)
Química Encefálica , Monoaminooxidasa/química , Proteínas del Tejido Nervioso/química , Tomografía de Emisión de Positrones/métodos , Receptores de Serotonina/química , Neuronas Serotoninérgicas/química , Proteínas de Transporte de Serotonina en la Membrana Plasmática/química , Adulto , Autopsia , Encéfalo/patología , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Masculino , Neuronas Serotoninérgicas/patología , Distribución Tisular
13.
Neuroimage ; 147: 198-203, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-27986606

RESUMEN

In the last years a plethora of studies have investigated morphological changes induced by behavioural or pharmacological interventions using structural T1-weighted MRI and voxel-based morphometry (VBM). Ketamine is thought to exert its antidepressant action by restoring neuroplasticity. In order to test for acute impact of a single ketamine infusion on grey matter volume we performed a placebo-controlled, double-blind investigation in healthy volunteers using VBM. 28 healthy individuals underwent two MRI sessions within a timeframe of 2 weeks, each consisting of two structural T1-weighted MRIs within a single session, one before and one 45min after infusion of S-ketamine (bolus of 0.11mg/kg, followed by an maintenance infusion of 0.12mg/kg) or placebo (0.9% NaCl infusion) using a crossover design. In the repeated-measures ANOVA with time (post-infusion/pre-infusion) and medication (placebo/ketamine) as factors, no significant effect of interaction and no effect of medication was found (FWE-corrected). Importantly, further post-hoc t-tests revealed a strong "decrease" of grey matter both in the placebo and the ketamine condition over time. This effect was evident mainly in frontal and temporal regions bilaterally with t-values ranging from 4.95 to 5.31 (FWE-corrected at p<0.05 voxel level). The vulnerabilities of VBM have been repeatedly demonstrated, with reports of influence of blood flow, tissue water and direct effects of pharmacological compounds on the MRI signal. Here again, we highlight that the relationship between intervention and VBM results is apparently subject to a number of physiological influences, which are partly unknown. Future studies focusing on the effects of ketamine on grey matter should try to integrate known influential factors such as blood flow into analysis. Furthermore, the results of this study highlight the importance of a carefully performed placebo condition in pharmacological fMRI studies.


Asunto(s)
Anestésicos Disociativos/farmacología , Procesamiento de Imagen Asistido por Computador/métodos , Ketamina/farmacología , Plasticidad Neuronal/efectos de los fármacos , Adulto , Circulación Cerebrovascular/fisiología , Estudios Cruzados , Método Doble Ciego , Femenino , Sustancia Gris/anatomía & histología , Sustancia Gris/fisiología , Humanos , Imagen por Resonancia Magnética , Masculino , Placebos , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Proyectos de Investigación , Adulto Joven
14.
Neuroimage ; 149: 23-32, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28119137

RESUMEN

INTRODUCTION: In-vivo quantification of serotonin transporters (SERT) in human brain has been a mainstay of molecular imaging in the field of neuropsychiatric disorders and helped to explore the underpinnings of several medical conditions, therapeutic and environmental influences. The emergence of PET/MR hybrid systems and the heterogeneity of SERT binding call for the development of efficient methods making the investigation of larger or vulnerable populations with limited scanner time and simultaneous changes in molecular and functional measures possible. We propose [11C]DASB bolus plus constant infusion for these applications and validate it against standard analyses of dynamic PET data. METHODS: [11C]DASB bolus/infusion optimization was performed on data acquired after [11C]DASB bolus in 8 healthy subjects. Subsequently, 16 subjects underwent one scan using [11C]DASB bolus plus constant infusion with Kbol 160-179min and one scan after [11C]DASB bolus for inter-method reliability analysis. Arterial blood sampling and metabolite analysis were performed for all scans. Distribution volumes (VT) were obtained using Logan plots for bolus scans and ratios between tissue and plasma parent activity for bolus plus infusion scans for different time spans of the scan (VT-70 for 60-70min after start of tracer infusion, VT-90 for 75-90min, VT-120 for 100-120min) in 9 subjects. Omitting blood data, binding potentials (BPND) obtained using multilinear reference tissue modeling (MRTM2) and cerebellar gray matter as reference region were compared in 11 subjects. RESULTS: A Kbol of 160min was observed to be optimal for rapid equilibration in thalamus and striatum. VT-70 showed good intraclass correlation coefficients (ICCs) of 0.61-0.70 for thalamus, striatal regions and olfactory cortex with bias ≤5.1% compared to bolus scans. ICCs increased to 0.72-0.78 for VT-90 and 0.77-0.93 for VT-120 in these regions. BPND-90 had negligible bias ≤2.5%, low variability ≤7.9% and ICCs of 0.74-0.87; BPND-120 had ICCs of 0.73-0.90. Low-binding cortical regions and cerebellar gray matter showed a positive bias of ~8% and ICCs 0.57-0.68 at VT-90. Cortical BPND suffered from high variability and bias, best results were obtained for olfactory cortex and anterior cingulate cortex with ICC=0.74-0.75 for BPND-90. High-density regions amygdala and midbrain had a negative bias of -5.5% and -22.5% at VT-90 with ICC 0.70 and 0.63, respectively. CONCLUSIONS: We have optimized the equilibrium method with [11C]DASB bolus plus constant infusion and demonstrated good inter-method reliability with accepted standard methods and for SERT quantification using both VT and BPND in a range of different brain regions. With as little as 10-15min of scanning valid estimates of SERT VT and BPND in thalamus, amygdala, striatal and high-binding cortical regions could be obtained. Blood sampling seems vital for valid quantification of SERT in low-binding cortical regions. These methods allow the investigation of up to three subjects with a single radiosynthesis.


Asunto(s)
Bencilaminas/administración & dosificación , Encéfalo/diagnóstico por imagen , Radioisótopos de Carbono/administración & dosificación , Tomografía de Emisión de Positrones/métodos , Radiofármacos/administración & dosificación , Proteínas de Transporte de Serotonina en la Membrana Plasmática/análisis , Adulto , Bencilaminas/farmacocinética , Radioisótopos de Carbono/farmacocinética , Método Doble Ciego , Femenino , Humanos , Infusiones Intravenosas , Inyecciones Intravenosas , Masculino , Radiofármacos/farmacocinética , Distribución Tisular
15.
Neuroimage ; 138: 257-265, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27236082

RESUMEN

Sex-steroid hormones have repeatedly been shown to influence empathy, which is in turn reflected in resting state functional connectivity (rsFC). Cross-sex hormone treatment in transgender individuals provides the opportunity to examine changes to rsFC over gender transition. We aimed to investigate whether sex-steroid hormones influence rsFC patterns related to unique aspects of empathy, namely emotion recognition and description as well as emotional contagion. RsFC data was acquired with 7Tesla magnetic resonance imaging in 24 male-to-female (MtF) and 33 female-to-male (FtM) transgender individuals before treatment, in addition to 33 male- and 44 female controls. Of the transgender participants, 15 MtF and 20 FtM were additionally assessed after 4 weeks and 4 months of treatment. Empathy scores were acquired at the same time-points. MtF differed at baseline from all other groups and assimilated over the course of gender transition in a rsFC network around the supramarginal gyrus, a region central to interpersonal emotion processing. While changes to sex-steroid hormones did not correlate with rsFC in this network, a sex hormone independent association between empathy scores and rsFC was found. Our results underline that 1) MtF transgender persons demonstrate unique rsFC patterns in a network related to empathy and 2) changes within this network over gender transition are likely related to changes in emotion recognition, -description, and -contagion, and are sex-steroid hormone independent.


Asunto(s)
Mapeo Encefálico/métodos , Empatía/fisiología , Hormonas Esteroides Gonadales/sangre , Red Nerviosa/fisiopatología , Lóbulo Parietal/fisiopatología , Transexualidad/tratamiento farmacológico , Transexualidad/fisiopatología , Adulto , Femenino , Hormonas Esteroides Gonadales/uso terapéutico , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Red Nerviosa/efectos de los fármacos , Lóbulo Parietal/efectos de los fármacos , Descanso , Caracteres Sexuales , Personas Transgénero , Resultado del Tratamiento
16.
Pharmacogenomics J ; 14(5): 463-72, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24709691

RESUMEN

Antidepressant pharmacogenetics represents a stimulating, but often discouraging field. The present study proposes a combination of several methodologies across three independent samples. Genes belonging to monoamine, neuroplasticity, circadian rhythm and transcription factor pathways were investigated in two samples (n=369 and 88) with diagnosis of major depression who were treated with antidepressants. Phenotypes were response, remission and treatment-resistant depression. Logistic regression including appropriate covariates was performed. Genes associated with outcomes were investigated in the STAR*D (Sequenced Treatment Alternatives to Relieve Depression) genome-wide study (n=1861). Top genes were further studied through a pathway analysis. In both original samples, markers associated with outcomes were concentrated in the PPP3CC gene. Other interesting findings were particularly in the HTR2A gene in one original sample and the STAR*D. The B-cell receptor signaling pathway proved to be the putative mediator of PPP3CC's effect on antidepressant response (P=0.03). Among innovative candidates, PPP3CC, involved in the regulation of immune system and synaptic plasticity, seems promising for further investigation.


Asunto(s)
Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Calcineurina/metabolismo , Depresión/tratamiento farmacológico , Depresión/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal/efectos de los fármacos , Calcineurina/genética , Depresión/genética , Depresión/inmunología , Humanos , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología
17.
Mol Psychiatry ; 18(1): 93-100, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22751491

RESUMEN

Electroconvulsive therapy (ECT) is a potent therapy in severe treatment-refractory depression. Although commonly applied in psychiatric clinical routine since decades, the exact neurobiological mechanism regarding its efficacy remains unclear. Results from preclinical and clinical studies emphasize a crucial involvement of the serotonin-1A receptor (5-HT(1A)) in the mode of action of antidepressant treatment. This includes associations between treatment response and changes in 5-HT(1A) function and density by antidepressants. Further, alterations of the 5-HT(1A) receptor are consistently reported in depression. To elucidate the effect of ECT on 5-HT(1A) receptor binding, 12 subjects with severe treatment-resistant major depression underwent three positron emission tomography (PET) measurements using the highly selective radioligand [carbonyl-(11)C]WAY100635, twice before (test-retest variability) and once after 10.08±2.35 ECT sessions. Ten patients (~83%) were responders to ECT. The voxel-wise comparison of the 5-HT(1A) receptor binding (BP(ND)) before and after ECT revealed a widespread reduction in cortical and subcortical regions (P<0.05 corrected), except for the occipital cortex and the cerebellum. Strongest reductions were found in regions consistently reported to be altered in major depression and involved in emotion regulation, such as the subgenual part of the anterior cingulate cortex (-27.5%), the orbitofrontal cortex (-30.1%), the amygdala (-31.8%), the hippocampus (-30.6%) and the insula (-28.9%). No significant change was found in the raphe nuclei. There was no significant difference in receptor binding in any region comparing the first two PET scans conducted before ECT. This PET study proposes a global involvement of the postsynaptic 5-HT(1A) receptor binding in the effect of ECT.


Asunto(s)
Trastorno Depresivo Mayor/diagnóstico por imagen , Trastorno Depresivo Mayor/terapia , Terapia Electroconvulsiva/métodos , Receptor de Serotonina 5-HT1A/metabolismo , Adulto , Antidepresivos/uso terapéutico , Antipsicóticos/uso terapéutico , Mapeo Encefálico , Isótopos de Carbono/farmacocinética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Piperazinas/farmacocinética , Tomografía de Emisión de Positrones/métodos , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Piridinas/farmacocinética , Antagonistas de la Serotonina/farmacocinética , Adulto Joven
18.
Nervenarzt ; 84(1): 14-9, 2013 Jan.
Artículo en Alemán | MEDLINE | ID: mdl-22318360

RESUMEN

Hormonal fluctuations during the perimenopausal transition lead to physical discomfort but are also frequently accompanied by mood swings, depressive symptoms, anxiety and sleeping disorders. The important role of the neurotransmitter serotonin in the pathogenesis of anxiety disorders and major depression is unquestioned, but only little is known about the influence of sex hormones on the serotonergic system. This review provides an overview of potential risk factors for the occurrence of affective disorders in the menopausal transition and discusses possible therapeutic options. Current research findings from longitudinal studies testing the efficacy of hormone replacement therapy and antidepressants with effects on the serotonergic neurotransmission on physical and mental discomforts during menopause are presented. Furthermore, studies using positron emission tomography and genetic methods that explore the effects of sex steroids on different components of the serotonergic system are shown. The interactions between estrogen, progesterone and the serotonergic system are described, and possible neurobiological and endocrinological mechanisms underlying depressive symptoms in the perimenopause are elucidated.


Asunto(s)
Trastornos de Ansiedad/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Climaterio/efectos de los fármacos , Climaterio/psicología , Trastorno Depresivo/tratamiento farmacológico , Terapia de Reemplazo de Estrógeno , Neuronas Serotoninérgicas/efectos de los fármacos , Serotonina/metabolismo , Antidepresivos/uso terapéutico , Trastornos de Ansiedad/psicología , Austria , Trastorno Depresivo/psicología , Quimioterapia Combinada , Femenino , Humanos , Estudios Longitudinales , Receptor de Serotonina 5-HT1A/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Resultado del Tratamiento
19.
J Affect Disord ; 301: 426-432, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35016914

RESUMEN

BACKGROUND: Neuroplastic processes are influenced by serotonergic agents, which reportedly alter white matter microstructure in humans in conjunction with learning. The goal of this double-blind, placebo-controlled imaging study was to investigate the neuroplastic properties of escitalopram and cognitive training on white matter plasticity during (re)learning as a model for antidepressant treatment and environmental factors. METHODS: Seventy-one healthy individuals (age=25.6 ± 5.0, 43 females) underwent three diffusion magnetic resonance imaging scans: at baseline, after 3 weeks of associative learning (emotional/non-emotional content), and after relearning shuffled associations for an additional 3 weeks. During the relearning phase, participants received a daily dose of 10 mg escitalopram or placebo orally. Fractional anisotropy (FA), and mean (MD), axial (AD), and radial diffusivity (RD) were calculated within the FMRIB software library and analyzed using tract-based spatial statistics. RESULTS: In a three-way repeated-measures marginal model with sandwich estimator standard errors, we found no significant effects of escitalopram and content on AD, FA, MD, and RD during both learning and relearning periods (pFDR>0.05). When testing for escitalopram or content effects separately, we also demonstrated no significant findings (pFDR>0.05) for any of the diffusion tensor imaging metrics. LIMITATIONS: The intensity of the study interventions might have been too brief to induce detectable white matter changes. DISCUSSION: Previous studies examining the effects of SSRIs on white matter tracts in humans have yielded inconclusive outcomes. Our results indicate that relearning under escitalopram does not affect the white matter microstructures in healthy individuals when administered for 3 weeks.


Asunto(s)
Imagen de Difusión Tensora , Sustancia Blanca , Anisotropía , Encéfalo , Imagen de Difusión por Resonancia Magnética , Imagen de Difusión Tensora/métodos , Método Doble Ciego , Escitalopram , Femenino , Humanos , Recuerdo Mental , Plasticidad Neuronal , Sustancia Blanca/diagnóstico por imagen
20.
Psychoneuroendocrinology ; 138: 105683, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35176535

RESUMEN

Sex hormones affect the GABAergic and glutamatergic neurotransmitter system as demonstrated in animal studies. However, human research has mostly been correlational in nature. Here, we aimed at substantiating causal interpretations of the interaction between sex hormones and neurotransmitter function by using magnetic resonance spectroscopy imaging (MRSI) to study the effect of gender-affirming hormone treatment (GHT) in transgender individuals. Fifteen trans men (TM) with a DSM-5 diagnosis of gender dysphoria, undergoing GHT, and 15 age-matched cisgender women (CW), receiving no therapy, underwent MRSI before and after at least 12 weeks. Additionally, sex differences in neurotransmitter levels were evaluated in an independent sample of 80 cisgender men and 79 cisgender women. Mean GABA+ (combination of GABA and macromolecules) and Glx (combination of glutamate and glutamine) ratios to total creatine (GABA+/tCr, Glx/tCr) were calculated in five predefined regions-of-interest (hippocampus, insula, pallidum, putamen and thalamus). Linear mixed models analysis revealed a significant measurement by gender identity effect (pcorr. = 0.048) for GABA+/tCr ratios in the hippocampus, with the TM cohort showing decreased GABA+/tCr levels after GHT compared to CW. Moreover, analysis of covariance showed a significant sex difference in insula GABA+/tCr ratios (pcorr. = 0.049), indicating elevated GABA levels in cisgender women compared to cisgender men. Our study demonstrates GHT treatment-induced GABA+/tCr reductions in the hippocampus, indicating hormone receptor activation on GABAergic cells and testosterone-induced neuroplastic processes within the hippocampus. Moreover, elevated GABA levels in the female compared to the male insula highlight the importance of including sex as factor in future MRS studies. DATA AVAILABILITY STATEMENT: Due to data protection laws processed data is available from the authors upon reasonable request. Please contact rupert.lanzenberger@meduniwien.ac.at with any questions or requests.


Asunto(s)
Ácido Glutámico , Personas Transgénero , Encéfalo/patología , Femenino , Identidad de Género , Hormonas Esteroides Gonadales , Humanos , Masculino , Neurotransmisores , Receptores de Antígenos de Linfocitos T , Testosterona , Ácido gamma-Aminobutírico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA