Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 43
1.
Wien Klin Wochenschr ; 2024 Apr 17.
Article En | MEDLINE | ID: mdl-38662240

Modern electroconvulsive therapy (ECT) and the approval of nasal esketamine for clinical use have significantly improved the approach to treatment-resistant depression (TRD), which is defined as non-response to at least two different courses of antidepressants with verified adherence to treatment, adequate dosage, and duration of treatment. The goal of this literature review is to present the newest evidence regarding efficacy and safety. Furthermore, we aim to provide an overview of future perspectives in this field of research, for example, regarding structural and molecular effects. Both treatment methods will be critically evaluated for their individual advantages, disadvantages, and response rates. Firstly, we will discuss the well-established method of ECT and its different treatment modalities. Secondly, we will discuss the properties of ketamine, the discovery of its antidepressive effects and the route to clinical approval of the esketamine nasal spray. We will comment on research settings which have evaluated intravenous ketamine against ECT. The decision-making process between esketamine nasal spray or ECT should include the assessment of contraindications, age, severity of disease, presence of psychotic symptoms, patient preference and treatment accessibility. We conclude that both treatment options are highly effective in TRD. If both are indicated, pragmatically esketamine will be chosen before ECT; however, ECT studies in ketamine non-responders are missing.

2.
Arch Sex Behav ; 53(5): 1859-1871, 2024 May.
Article En | MEDLINE | ID: mdl-38216784

Self-reported sexual orientation of transgender individuals occasionally changes over transition. Using functional magnetic resonance imaging, we tested the hypothesis that neural and behavioral patterns of sexual arousal in transgender individuals would shift from the assigned to the experienced gender (e.g., trans women's responses becoming more dissimilar to those of cis men and more similar to those of cis women). To this aim, trans women (N = 12) and trans men (N = 20) as well as cisgender women (N = 24) and cisgender men (N = 14) rated visual stimuli showing male-female, female-female or male-male intercourse for sexual arousal before and after four months of gender-affirming hormone therapy. A Bayesian framework allowed us to incorporate previous behavioral findings. The hypothesized changes could indeed be observed in the behavioral responses with the strongest results for trans men and female-female scenes. Activation of the ventral striatum supported our hypothesis only for female-female scenes in trans women. The respective application or depletion of androgens in trans men and trans women might partly explain this observation. The prominent role of female-female stimuli might be based on the differential responses they elicit in cis women and men or, in theory, the controversial concept of autogynephilia. We show that correlates of sexual arousal in transgender individuals might change in the direction of the experienced gender. Future investigations should elucidate the mechanistic role of sex hormones and the cause of the differential neural and behavioral findings.The study was registered at ClinicalTrials.gov (NCT02715232), March 22, 2016.


Bayes Theorem , Gender Dysphoria , Magnetic Resonance Imaging , Sexual Arousal , Transgender Persons , Humans , Male , Female , Adult , Gender Dysphoria/psychology , Gender Dysphoria/drug therapy , Transgender Persons/psychology , Sexual Behavior/drug effects , Sexual Behavior/psychology , Young Adult , Ventral Striatum/drug effects , Ventral Striatum/diagnostic imaging
4.
Sci Rep ; 13(1): 16426, 2023 09 30.
Article En | MEDLINE | ID: mdl-37777558

The serotonin transporter (5-HTT) critically shapes serotonin neurotransmission by regulating extracellular brain serotonin levels; it remains unclear to what extent 5-HTT levels in the human brain are genetically determined. Here we applied [11C]DASB positron emission tomography to image brain 5-HTT levels and evaluated associations with five common serotonin-related genetic variants that might indirectly regulate 5-HTT levels (BDNF rs6265, SLC6A4 5-HTTLPR, HTR1A rs6295, HTR2A rs7333412, and MAOA rs1137070) in 140 healthy volunteers. In addition, we explored whether these variants could predict in vivo 5-HTT levels using a five-fold cross-validation random forest framework. MAOA rs1137070 T-carriers showed significantly higher brain 5-HTT levels compared to C-homozygotes (2-11% across caudate, putamen, midbrain, thalamus, hippocampus, amygdala and neocortex). We did not observe significant associations for the HTR1A rs6295 and HTR2A rs7333412 genotypes. Our previously observed lower subcortical 5-HTT availability for rs6265 met-carriers remained in the presence of these additional variants. Despite this significant association, our prediction models showed that genotype moderately improved prediction of 5-HTT in caudate, but effects were not statistically significant after adjustment for multiple comparisons. Our observations provide additional evidence that serotonin-related genetic variants modulate adult human brain serotonin neurotransmission.


Neocortex , Serotonin Plasma Membrane Transport Proteins , Humans , Adult , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin , Tomography, X-Ray Computed , Brain/diagnostic imaging , Brain/metabolism , Mesencephalon/metabolism , Positron-Emission Tomography , Neocortex/metabolism
5.
J Psychiatry Neurosci ; 48(5): E369-E375, 2023.
Article En | MEDLINE | ID: mdl-37751919

BACKGROUND: Among its pleiotropic properties, gender-affirming hormone therapy (GHT) affects regional brain volumes. The hypothalamus, which regulates neuroendocrine function and associated emotional and cognitive processes, is an intuitive target for probing GHT effects. We sought to assess changes to hypothalamus and hypothalamic subunit volumes after GHT, thereby honouring the region's anatomical and functional heterogeneity. METHODS: Individuals with gender dysphoria and cisgender controls underwent 2 MRI measurements, with a median interval of 145 days (interquartile range [IQR] 128.25-169.75 d, mean 164.94 d) between the first and second MRI. Transgender women (TW) and transgender men (TM) underwent the first MRI before GHT and the second MRI after approximately 4.5 months of GHT, which comprised estrogen and anti-androgen therapy in TW or testosterone therapy in TM. Hypothalamic volumes were segmented using FreeSurfer, and effects of GHT were tested using repeated-measures analysis of covariance. RESULTS: The final sample included 106 participants: 38 TM, 15 TW, 32 cisgender women (CW) and 21 cisgender men (CM). Our analyses revealed group × time interaction effects for total, left and right hypothalamus volume, and for several subunits (left and right inferior tubular, left superior tubular, right anterior inferior, right anterior superior, all p corr < 0.01). In TW, volumes decreased between the first and second MRI in these regions (all p corr ≤ 0.01), and the change from the first to second MRI in TW differed significantly from that in CM and CW in several subunits (p corr < 0.05). LIMITATIONS: We did not address the influence of transition-related psychological and behavioural changes. CONCLUSION: Our results suggest a subunit-specific effect of GHT on hypothalamus volumes in TW. This finding is in accordance with previous reports of positive and negative effects of androgens and estrogens, respectively, on cerebral volumes.


Emotions , Gender Dysphoria , Male , Female , Humans , Gender Dysphoria/diagnostic imaging , Gender Dysphoria/drug therapy , Hypothalamus/diagnostic imaging , Testosterone
6.
Psychoneuroendocrinology ; 155: 106336, 2023 09.
Article En | MEDLINE | ID: mdl-37499299

BACKGROUND: Sex-specific differences in brain connectivity were found in various neuroimaging studies, though little is known about sex steroid effects on insular functioning. Based on well-characterized sex differences in emotion regulation, interoception and higher-level cognition, gender-dysphoric individuals receiving gender-affirming hormone therapy represent an interesting cohort to investigate how sex hormones might influence insular connectivity and related brain functions. METHODS: To analyze the potential effect of sex steroids on insular connectivity at rest, 11 transgender women, 14 transgender men, 20 cisgender women, and 11 cisgender men were recruited. All participants underwent two magnetic resonance imaging sessions involving resting-state acquisitions separated by a median time period of 4.5 months and also completed the Bermond-Vorst alexithymia questionnaire at the initial and final examination. Between scans, transgender subjects received gender-affirming hormone therapy. RESULTS: A seed based functional connectivity analysis revealed a significant 2-way interaction effect of group-by-time between right insula, cingulum, left middle frontal gyrus and left angular gyrus. Post-hoc tests demonstrated an increase in connectivity for transgender women when compared to cisgender men. Furthermore, spectral dynamic causal modelling showed reduced effective connectivity from the posterior cingulum and left angular gyrus to the left middle frontal gyrus as well as from the right insula to the left middle frontal gyrus. Alexithymia changes were found after gender-affirming hormone therapy for transgender women in both fantasizing and identifying. CONCLUSION: These findings suggest a considerable influence of estrogen administration and androgen suppression on brain networks implicated in interoception, own-body perception and higher-level cognition.


Gender Dysphoria , Transsexualism , Humans , Male , Female , Gender Dysphoria/drug therapy , Gender Identity , Transsexualism/drug therapy , Brain , Magnetic Resonance Imaging/methods , Gonadal Steroid Hormones/pharmacology , Steroids
7.
Transl Psychiatry ; 13(1): 208, 2023 Jun 15.
Article En | MEDLINE | ID: mdl-37322010

Variants within the monoamine oxidase A (MAO-A, MAOA) and tryptophan hydroxylase 2 (TPH2) genes, the main enzymes in cerebral serotonin (5-HT) turnover, affect risk for depression. Depressed cohorts show increased cerebral MAO-A in positron emission tomography (PET) studies. TPH2 polymorphisms might also influence brain MAO-A because availability of substrates (i.e. monoamine concentrations) were shown to affect MAO-A levels. We assessed the effect of MAOA (rs1137070, rs2064070, rs6323) and TPH2 (rs1386494, rs4570625) variants associated with risk for depression and related clinical phenomena on global MAO-A distribution volume (VT) using [11C]harmine PET in 51 participants (21 individuals with seasonal affective disorder (SAD) and 30 healthy individuals (HI)). Statistical analyses comprised general linear models with global MAO-A VT as dependent variable, genotype as independent variable and age, sex, group (individuals with SAD, HI) and season as covariates. rs1386494 genotype significantly affected global MAO-A VT after correction for age, group and sex (p < 0.05, corr.), with CC homozygotes showing 26% higher MAO-A levels. The role of rs1386494 on TPH2 function or expression is poorly understood. Our results suggest rs1386494 might have an effect on either, assuming that TPH2 and MAO-A levels are linked by their common product/substrate, 5-HT. Alternatively, rs1386494 might influence MAO-A levels via another mechanism, such as co-inheritance of other genetic variants. Our results provide insight into how genetic variants within serotonin turnover translate to the cerebral serotonin system. Clinicaltrials.gov Identifier: NCT02582398. EUDAMED Number: CIV-AT-13-01-009583.


Seasonal Affective Disorder , Serotonin , Humans , Brain/diagnostic imaging , Brain/metabolism , Harmine/metabolism , Monoamine Oxidase/genetics , Monoamine Oxidase/metabolism , Seasonal Affective Disorder/metabolism , Serotonin/metabolism
8.
Int J Neuropsychopharmacol ; 26(2): 116-124, 2023 02 14.
Article En | MEDLINE | ID: mdl-36573644

BACKGROUND: Epigenetic modifications like DNA methylation are understood as an intermediary between environmental factors and neurobiology. Cerebral monoamine oxidase A (MAO-A) levels are altered in depression, as are DNA methylation levels within the MAOA gene, particularly in the promoter/exon I/intron I region. An effect of MAOA methylation on peripheral protein expression was shown, but the extent to which methylation affects brain MAO-A levels is not fully understood. METHODS: Here, the influence of MAOA promoter/exon I/intron I region DNA methylation on global MAO-A distribution volume (VT), an index of MAO-A density, was assessed via [11C]harmine positron emission tomography in 22 patients (14 females) suffering from seasonal affective disorder and 30 healthy controls (17 females). RESULTS: No significant influence of MAOA DNA methylation on global MAO-A VT was found, despite correction for health status, sex, season, and MAOA variable number of tandem repeat genotype. However, season affected average methylation in women, with higher levels in spring and summer (Puncorr = .03). We thus did not find evidence for an effect of MAOA DNA methylation on brain MAO-A VT. CONCLUSIONS: In contrast to a previous study demonstrating an effect of methylation of a MAOA promoter region located further 5' on brain MAO-A, MAOA methylation of the region assessed here appears to affect brain protein levels to a limited extent at most. The observed effect of season on methylation levels is in accordance with extensive evidence for seasonal effects within the serotonergic system. CLINICALTRIALS.GOV IDENTIFIER: NCT02582398 (https://clinicaltrials.gov/ct2/show/NCT02582398).


DNA Methylation , Harmine , Humans , Female , Monoamine Oxidase/genetics , Monoamine Oxidase/metabolism , Carbon Radioisotopes , Positron-Emission Tomography/methods
9.
Psychoneuroendocrinology ; 133: 105381, 2021 11.
Article En | MEDLINE | ID: mdl-34416504

The sex hormones testosterone and estradiol influence brain structure and function and are implicated in the pathogenesis, prevalence and disease course of major depression. Recent research employing gender-affirming hormone treatment (GHT) of gender dysphoric individuals and utilizing positron emission tomography (PET) indicates increased serotonin transporter binding upon high-dosages of testosterone treatment. Here, we investigated the effects of GHT on levels of monoamine oxidase A (MAO-A), another key target of antidepressant treatment. Participants underwent PET with the radioligand [11C]harmine to assess cerebral MAO-A distribution volumes (VT) before and four months after initiation of GHT. By the time this study was terminated for technical reasons, 18 transgender individuals undergoing GHT (11 transmen, TM and 7 transwomen, TW) and 17 cis-gender subjects had been assessed. Preliminary analysis of available data revealed statistically significant MAO-A VT reductions in TM under testosterone treatment in six of twelve a priori defined regions of interest (middle frontal cortex (-10%), anterior cingulate cortex (-9%), medial cingulate cortex (-10.5%), insula (-8%), amygdala (-9%) and hippocampus (-8.5%, all p<0.05)). MAO-A VT did not change in TW receiving estrogen treatment. Despite the limited sample size, pronounced MAO-A VT reduction could be observed, pointing towards a potential effect of testosterone. Considering MAO-A's central role in regulation of serotonergic neurotransmission, changes to MAO-A VT should be further investigated as a possible mechanism by which testosterone mediates risk for, symptomatology of, and treatment response in affective disorders.


Brain , Monoamine Oxidase , Testosterone , Brain/diagnostic imaging , Brain/metabolism , Dose-Response Relationship, Drug , Female , Humans , Male , Monoamine Oxidase/drug effects , Monoamine Oxidase/metabolism , Positron-Emission Tomography , Testosterone/administration & dosage , Testosterone/pharmacology
10.
Front Neurosci ; 15: 609485, 2021.
Article En | MEDLINE | ID: mdl-33841073

Ketamine is a powerful glutamatergic long-lasting antidepressant, efficient in intractable major depression. Whereas ketamine's immediate psychomimetic side-effects were linked to glutamate changes, proton MRS (1H-MRS) showed an association between the ratio of glutamate and glutamine and delayed antidepressant effect emerging ∼2 h after ketamine administration. While most 1H-MRS studies focused on anterior cingulate, recent functional MRI connectivity studies revealed an association between ketamine's antidepressant effect and disturbed connectivity patterns to the posterior cingulate cortex (PCC), and related PCC dysfunction to rumination and memory impairment involved in depressive pathophysiology. The current study utilized the state-of-the-art single-voxel 3T sLASER 1H-MRS methodology optimized for reproducible measurements. Ketamine's effects on neurochemicals were assessed before and ∼3 h after intravenous ketamine challenge in PCC. Concentrations of 11 neurochemicals, including glutamate (CRLB ∼ 4%) and glutamine (CRLB ∼ 13%), were reliably quantified with the LCModel in 12 healthy young men with between-session coefficients of variation (SD/mean) <8%. Also, ratios of glutamate/glutamine and glutamate/aspartate were assessed as markers of synaptic function and activated glucose metabolism, respectively. Pairwise comparison of metabolite profiles at baseline and 193 ± 4 min after ketamine challenge yielded no differences. Minimal detectable concentration differences estimated with post hoc power analysis (power = 80%, alpha = 0.05) were below 0.5 µmol/g, namely 0.39 µmol/g (∼4%) for glutamate, 0.28 µmol/g (∼10%) for Gln, ∼14% for glutamate/glutamine and ∼8% for glutamate/aspartate. Despite the high sensitivity to detect between-session differences in glutamate and glutamine concentrations, our study did not detect delayed glutamatergic responses to subanesthetic ketamine doses in PCC.

11.
Handb Clin Neurol ; 175: 117-140, 2020.
Article En | MEDLINE | ID: mdl-33008520

Most psychiatric disorders demonstrate sex differences in their prevalence and symptomatology, and in their response to treatment. These differences are particularly pronounced in mood disorders. Differences in sex hormone levels are among the most overt distinctions between males and females and are thus an intuitive underpinning for these clinical observations. In fact, treatment with estrogen and testosterone was shown to exert antidepressant effects, which underscores this link. Changes to monoaminergic signaling in general, and serotonergic transmission in particular, are understood as central components of depressive pathophysiology. Thus, modulation of the serotonin system may serve as a mechanism via which sex hormones exert their clinical effects in mental health disorders. Over the past 20 years, various experimental approaches have been applied to identify modes of influence of sex and sex hormones on the serotonin system. This chapter provides an overview of different molecular components of the serotonin system, followed by a review of studies performed in animals and in humans with the purpose of elucidating sex hormone effects. Particular emphasis will be placed on studies performed with positron emission tomography, a method that allows for human in vivo molecular imaging and, therefore, assessment of effects in a clinically representative context. The studies addressed in this chapter provide a wealth of information on the interaction between sex, sex hormones, and serotonin in the brain. In general, they offer evidence for the concept that the influence of sex hormones on various components of the serotonin system may serve as an underpinning for the clinical effects these hormones demonstrate.


Depression , Migraine Disorders , Animals , Antidepressive Agents , Brain/diagnostic imaging , Female , Humans , Male , Serotonin
12.
Front Psychiatry ; 11: 549903, 2020.
Article En | MEDLINE | ID: mdl-33101078

INTRODUCTION: Converging evidence suggests that ketamine elicits antidepressant effects via enhanced neuroplasticity precipitated by a surge of glutamate and modulation of GABA. Magnetic resonance spectroscopic imaging (MRSI) illustrates changes to cerebral glutamate and GABA immediately following ketamine administration during dissociation. However, few studies assess subacute changes in the first hours following application, when ketamine's antidepressant effects emerge. Moreover, ketamine metabolites implicated in its antidepressant effects develop during this timeframe. Thus, this study aimed to investigate subacute changes in cerebral Glx (glutamate + glutamine), GABA and their ratio in seven brain regions central to depressive pathophysiology and treatment. METHODS: Twenty-five healthy subjects underwent two multivoxel MRS scans using a spiral encoded, MEGA-edited LASER-localized 3D-MRSI sequence, at baseline and 2 h following intravenous administration of racemic ketamine (0.8 mg/kg bodyweight over 50 min). Ketamine, norketamine and dehydronorketamine plasma levels were determined at routine intervals during and after infusion. Automated region-of-interest (ROI)-based quantification of mean metabolite concentration was used to assess changes in GABA+/total creatine (tCr), Glx/tCr, and GABA+/Glx ratios in the thalamus, hippocampus, insula, putamen, rostral anterior cingulate cortex (ACC), caudal ACC, and posterior cingulate cortex. Effects of ketamine on neurotransmitter levels and association with ketamine- and metabolite plasma levels were tested with repeated measures analyses of variance (rmANOVA) and correlation analyses, respectively. RESULTS: For GABA+/tCr rmANOVA revealed a measurement by region interaction effect (puncorr < 0.001) and post hoc pairwise comparisons showed a reduction in hippocampal GABA+/tCr after ketamine (pcorr = 0.02). For Glx/tCr and GABA+/Glx neither main effects of measurement nor measurement by region interactions were observed (all puncorr > 0.05). Furthermore, no statistically significant associations between changes in any of the neurotransmitter ratios and plasma levels of ketamine, norketamine, or dehydronorketamine were observed (pcorr > 0.05). CONCLUSION: This study provides evidence for decreased hippocampal GABA+/tCr ratio 2 h following ketamine administration. As MRS methodology measures total levels of intra- and extracellular GABA, results might indicate drug induced alterations in GABA turnover. Our study in healthy humans suggests that changes in GABA levels, particularly in the hippocampus, should be further assessed for their relevance to ketamine´s antidepressant effects.

13.
Hum Brain Mapp ; 41(16): 4518-4528, 2020 11.
Article En | MEDLINE | ID: mdl-32697408

The serotonin 2A receptor (5-HT2AR) is implicated in the pathophysiology and treatment of various psychiatric disorders. [18 F]altanserin and [11 C]Cimbi-36 positron emission tomography (PET) allow for high-resolution imaging of 5-HT2AR in the living human brain. Cerebral 5-HT2AR binding is strongly genetically determined, though the impact of specific variants is poorly understood. Candidate gene studies suggest that HTR2A single nucleotide polymorphisms including rs6311/rs6313, rs6314, and rs7997012 may influence risk for psychiatric disorders and mediate treatment response. Although known to impact in vitro expression of 5-HT2AR or other serotonin (5-HT) proteins, their effect on human in vivo brain 5-HT2AR binding has as of yet been insufficiently studied. We thus assessed the extent to which these variants and the commonly studied 5-HTTLPR predict neocortex in vivo 5-HT2AR binding in healthy adult humans. We used linear regression analyses and likelihood ratio tests in 197 subjects scanned with [18 F]altanserin or [11 C]Cimbi-36 PET. Although we observed genotype group differences in 5-HT2AR binding of up to ~10%, no genetic variants were statistically significantly predictive of 5-HT2AR binding in what is the largest human in vivo 5-HT2AR imaging genetics study to date. Thus, in vitro and post mortem results suggesting effects on 5-HT2AR expression did not carry over to the in vivo setting. To any extent these variants might affect clinical risk, our findings do not support that 5-HT2AR binding mediates such effects. Our observations indicate that these individual variants do not significantly contribute to genetic load on human in vivo 5-HT2AR binding.


Neocortex/metabolism , Receptor, Serotonin, 5-HT2A/genetics , Receptor, Serotonin, 5-HT2A/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Benzylamines/pharmacokinetics , Female , Fluorine Radioisotopes/pharmacokinetics , Humans , Ketanserin/analogs & derivatives , Ketanserin/pharmacokinetics , Male , Middle Aged , Neocortex/diagnostic imaging , Phenethylamines/pharmacokinetics , Positron-Emission Tomography , Serotonin 5-HT2 Receptor Agonists/pharmacokinetics , Serotonin Antagonists/pharmacokinetics , Young Adult
14.
Front Hum Neurosci ; 13: 289, 2019.
Article En | MEDLINE | ID: mdl-31507394

Reward anticipation is essential for directing behavior toward positively valenced stimuli, creating motivational salience. Task-related activation of the ventral striatum (VS) has long been used as a target for understanding reward function. However, some subjects may not be able to perform the respective tasks because of their complexity or subjects' physical or mental disabilities. Moreover, task implementations may differ, which results in limited comparability. Hence, developing a task-free method for evaluating neural gain circuits is essential. Research has shown that fluctuations in neuronal activity at rest denoted individual differences in the brain functional networks. Here, we proposed novel models to predict the activation of the VS during gain anticipation, using the functional magnetic resonance imaging data of 45 healthy subjects acquired during a monetary incentive delay task and under rest. In-sample validation and held-out data were used to estimate the generalizability of the models. It was possible to predict three measures of reward activation (sensitivity, average, maximum) from resting-state functional connectivity (Pearson's r = 0.38-0.54 in validation data). Especially high contributions to the models were observed from the default mode network. These findings highlight the potential of using functional connectivity at rest as a task-free alternative for predicting activation in the VS, offering a possibility to estimate reward response in the broader sampling of subject populations.

15.
Sci Rep ; 9(1): 11484, 2019 08 07.
Article En | MEDLINE | ID: mdl-31391479

Numerous studies demonstrate ketamine's influence on resting-state functional connectivity (rsFC). Seed-based and static rsFC estimation methods may oversimplify FC. These limitations can be addressed with whole-brain, dynamic rsFC estimation methods. We assessed data from 27 healthy subjects who underwent two 3 T resting-state fMRI scans, once under subanesthetic, intravenous esketamine and once under placebo, in a randomized, cross-over manner. We aimed to isolate only highly robust effects of esketamine on dynamic rsFC by using eight complementary methodologies derived from two dynamic rsFC estimation methods, two functionally defined atlases and two statistical measures. All combinations revealed a negative influence of esketamine on dynamic rsFC within the left visual network and inter-hemispherically between visual networks (p < 0.05, corrected), hereby suggesting that esketamine's influence on dynamic rsFC is highly stable in visual processing networks. Our findings may be reflective of ketamine's role as a model for psychosis, a disorder associated with alterations to visual processing and impaired inter-hemispheric connectivity. Ketamine is a highly effective antidepressant and studies have shown changes to sensory processing in depression. Dynamic rsFC in sensory processing networks might be a promising target for future investigations of ketamine's antidepressant properties. Mechanistically, sensitivity of visual networks for esketamine's effects may result from their high expression of NMDA-receptors.


Antidepressive Agents/administration & dosage , Ketamine/administration & dosage , Nerve Net/drug effects , Visual Cortex/drug effects , Adult , Antidepressive Agents/pharmacokinetics , Connectome , Cross-Over Studies , Female , Healthy Volunteers , Humans , Ketamine/pharmacokinetics , Magnetic Resonance Imaging , Male , Nerve Net/physiology , Placebos/administration & dosage , Receptors, N-Methyl-D-Aspartate/metabolism , Rest/physiology , Visual Cortex/physiology , Young Adult
16.
World J Biol Psychiatry ; 20(6): 427-448, 2019 07.
Article En | MEDLINE | ID: mdl-31340696

Objectives: The overview outlines two decades of research from the European Group for the Study of Resistant Depression (GSRD) that fundamentally impacted evidence-based algorithms for diagnostics and psychopharmacotherapy of treatment-resistant depression (TRD). Methods: The GSRD staging model characterising response, non-response and resistance to antidepressant (AD) treatment was applied to 2762 patients in eight European countries. Results: In case of non-response, dose escalation and switching between different AD classes did not show superiority over continuation of original AD treatment. Predictors for TRD were symptom severity, duration of the current major depressive episode (MDE), suicidality, psychotic and melancholic features, comorbid anxiety and personality disorders, add-on treatment, non-response to the first AD, adverse effects, high occupational level, recurrent disease course, previous hospitalisations, positive family history of MDD, early age of onset and novel associations of single nucleoid polymorphisms (SNPs) within the PPP3CC, ST8SIA2, CHL1, GAP43 and ITGB3 genes and gene pathways associated with neuroplasticity, intracellular signalling and chromatin silencing. A prediction model reaching accuracy of above 0.7 highlighted symptom severity, suicidality, comorbid anxiety and lifetime MDEs as the most informative predictors for TRD. Applying machine-learning algorithms, a signature of three SNPs of the BDNF, PPP3CC and HTR2A genes and lacking melancholia predicted treatment response. Conclusions: The GSRD findings offer a unique and balanced perspective on TRD representing foundation for further research elaborating on specific clinical and genetic hypotheses and treatment strategies within appropriate study-designs, especially interaction-based models and randomized controlled trials.


Depressive Disorder, Treatment-Resistant/genetics , Depressive Disorder, Treatment-Resistant/therapy , Algorithms , Antidepressive Agents/therapeutic use , Comorbidity , Depressive Disorder, Treatment-Resistant/diagnosis , Europe , Humans , Machine Learning , Pharmacogenetics , Polymorphism, Single Nucleotide , Psychiatric Status Rating Scales , Treatment Outcome
17.
Front Mol Neurosci ; 12: 28, 2019.
Article En | MEDLINE | ID: mdl-30837839

Purpose: Advanced analysis methods for multi-voxel magnetic resonance spectroscopy (MRS) are crucial for neurotransmitter quantification, especially for neurotransmitters showing different distributions across tissue types. So far, only a handful of studies have used region of interest (ROI)-based labeling approaches for multi-voxel MRS data. Hence, this study aims to provide an automated ROI-based labeling tool for 3D-multi-voxel MRS data. Methods: MRS data, for automated ROI-based labeling, was acquired in two different spatial resolutions using a spiral-encoded, LASER-localized 3D-MRS imaging sequence with and without MEGA-editing. To calculate the mean metabolite distribution within selected ROIs, masks of individual brain regions were extracted from structural T1-weighted images using FreeSurfer. For reliability testing of automated labeling a comparison to manual labeling and single voxel selection approaches was performed for six different subcortical regions. Results: Automated ROI-based labeling showed high consistency [intra-class correlation coefficient (ICC) > 0.8] for all regions compared to manual labeling. Higher variation was shown when selected voxels, chosen from a multi-voxel grid, uncorrected for voxel composition, were compared to labeling methods using spatial averaging based on anatomical features within gray matter (GM) volumes. Conclusion: We provide an automated ROI-based analysis approach for various types of 3D-multi-voxel MRS data, which dramatically reduces hands-on time compared to manual labeling without any possible inter-rater bias.

18.
Behav Sleep Med ; 17(6): 713-720, 2019.
Article En | MEDLINE | ID: mdl-29775085

Objective: Therapeutic sleep deprivation (SD) is a nonpharmacological treatment that is used most often for depression. The aim of this study was to examine the pattern of use of SD in psychiatric hospitals in Austria, Germany, and Switzerland. Methods: A questionnaire about perceived usage of SD was sent by mail to all 511 psychiatric hospitals in the three countries. Nonresponders were asked to answer the questionnaire by phone. We achieved a response rate of 75.3%. Results: SD was recommended by 61.3% of all hospitals. Despite this degree of recommendation, nearly two thirds of the psychiatric hospitals had not treated a patient with SD during the last 12 months. Of the respondents, 59.5% considered SD to be indicated for major depressive disorder, 17.7% for bipolar depression, and 7.8% for other indications. SD was administered most frequently in inpatient settings and in combination with other therapies. Total SD (patients kept awake entire night) and partial late SD (patients kept awake in second half of night) were judged equally effective. Of the hospitals, 53.0% reported having seen hypomania and 13.2% manic episodes as side effects (rates do not represent actual incident rates). Conclusion: The lack of large controlled studies for SD with its different forms of treatment probably still hinders a broader use of the therapy. Therefore, further efforts should be undertaken to provide high-quality scientific evidence for the usage of SD.


Hospitals, Psychiatric/trends , Sleep Deprivation/psychology , Adult , Austria , Female , Germany , Humans , Male , Surveys and Questionnaires , Switzerland
19.
Cereb Cortex ; 29(1): 372-382, 2019 01 01.
Article En | MEDLINE | ID: mdl-30357321

Parcellation of distinct areas in the cerebral cortex has a long history in neuroscience and is of great value for the study of brain function, specialization, and alterations in neuropsychiatric disorders. Analysis of cytoarchitectonical features has revealed their close association with molecular profiles based on protein density. This provides a rationale for the use of in vivo molecular imaging data for parcellation of the cortex with the advantage of whole-brain coverage. In the current work, parcellation was based on expression of key players of the serotonin neurotransmitter system. Positron emission tomography was carried out for the quantification of serotonin 1A (5-HT1A, n = 30) and 5-HT2A receptors (n = 22), the serotonin-degrading enzyme monoamine oxidase A (MAO-A, n = 32) and the serotonin transporter (5-HTT, n = 24) in healthy participants. Cortical protein distribution maps were obtained using surface-based quantification. Based on k-means clustering, silhouette criterion and bootstrapping, five distinct clusters were identified as the optimal solution. The defined clusters proved of high explanatory value for the effects of psychotropic drugs acting on the serotonin system, such as antidepressants and psychedelics. Therefore, the proposed method constitutes a sensible approach towards integration of multimodal imaging data for research and development in neuropharmacology and psychiatry.


Cerebral Cortex/metabolism , Monoamine Oxidase/metabolism , Positron-Emission Tomography/methods , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Adult , Cerebral Cortex/diagnostic imaging , Female , Humans , Male , Middle Aged , Molecular Imaging/methods , Serotonin/metabolism , Young Adult
20.
Transl Psychiatry ; 8(1): 198, 2018 09 21.
Article En | MEDLINE | ID: mdl-30242221

Increased cerebral monoamine oxidase A (MAO-A) levels have been shown in non-seasonal depression using positron emission tomography (PET). Seasonal affective disorder (SAD) is a sub-form of major depressive disorder and is typically treated with bright light therapy (BLT). The serotonergic system is affected by season and light. Hence, this study aims to assess the relevance of brain MAO-A levels to the pathophysiology and treatment of SAD. Changes to cerebral MAO-A distribution (1) in SAD in comparison to healthy controls (HC), (2) after treatment with BLT and (3) between the seasons, were investigated in 24 patients with SAD and 27 HC using [11C]harmine PET. PET scans were performed in fall/winter before and after 3 weeks of placebo-controlled BLT, as well as in spring/summer. Cerebral MAO-A distribution volume (VT, an index of MAO-A density) did not differ between patients and HC at any of the three time-points. However, MAO-A VT decreased from fall/winter to spring/summer in the HC group (F1, 187.84 = 4.79, p < 0.050), while SAD showed no change. In addition, BLT, but not placebo, resulted in a significant reduction in MAO-A VT (F1, 208.92 = 25.96, p < 0.001). This is the first study to demonstrate an influence of BLT on human cerebral MAO-A levels in vivo. Furthermore, we show that SAD may lack seasonal dynamics in brain MAO-A levels. The lack of a cross-sectional difference between patients and HC, in contrast to studies in non-seasonal depression, may be due to the milder symptoms typically shown by patients with SAD.


Brain/metabolism , Monoamine Oxidase/metabolism , Phototherapy , Seasonal Affective Disorder/metabolism , Seasonal Affective Disorder/therapy , Adult , Brain/physiopathology , Carbon Radioisotopes , Female , Harmine , Humans , Male , Positron-Emission Tomography , Treatment Outcome
...