Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Psychopharmacology (Berl) ; 236(8): 2437-2449, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31254091

ABSTRACT

BACKGROUND: Aversive stimuli in the environment influence human actions. This includes valence-dependent influences on action selection, e.g., increased avoidance but decreased approach behavior. However, it is yet unclear how aversive stimuli interact with complex learning and decision-making in the reward and avoidance domain. Moreover, the underlying computational mechanisms of these decision-making biases are unknown. METHODS: To elucidate these mechanisms, 54 healthy young male subjects performed a two-step sequential decision-making task, which allows to computationally model different aspects of learning, e.g., model-free, habitual, and model-based, goal-directed learning. We used a within-subject design, crossing task valence (reward vs. punishment learning) with emotional context (aversive vs. neutral background stimuli). We analyzed choice data, applied a computational model, and performed simulations. RESULTS: Whereas model-based learning was not affected, aversive stimuli interacted with model-free learning in a way that depended on task valence. Thus, aversive stimuli increased model-free avoidance learning but decreased model-free reward learning. The computational model confirmed this effect: the parameter lambda that indicates the influence of reward prediction errors on decision values was increased in the punishment condition but decreased in the reward condition when aversive stimuli were present. Further, by using the inferred computational parameters to simulate choice data, our effects were captured. Exploratory analyses revealed that the observed biases were associated with subclinical depressive symptoms. CONCLUSION: Our data show that aversive environmental stimuli affect complex learning and decision-making, which depends on task valence. Further, we provide a model of the underlying computations of this affective modulation. Finally, our finding of increased decision-making biases in subjects reporting subclinical depressive symptoms matches recent reports of amplified Pavlovian influences on action selection in depression and suggests a potential vulnerability factor for mood disorders. We discuss our findings in the light of the involvement of the neuromodulators serotonin and dopamine.


Subject(s)
Avoidance Learning/physiology , Computer Simulation , Decision Making/physiology , Depression/psychology , Models, Psychological , Reward , Affect/physiology , Choice Behavior/physiology , Humans , Male , Photic Stimulation/methods , Punishment/psychology , Young Adult
2.
Neuropsychopharmacology ; 44(3): 590-597, 2019 02.
Article in English | MEDLINE | ID: mdl-30375508

ABSTRACT

Cognitive control represents an essential neuropsychological characteristic that allows for the rapid adaption of a changing environment by constant re-allocation of cognitive resources. This finely tuned mechanism is impaired in psychiatric disorders such as schizophrenia and contributes to cognitive deficits. Neuroimaging has highlighted the contribution of the anterior cingulate cortex (ACC) and prefrontal regions (PFC) on cognitive control and demonstrated the impact of genetic variation, as well as genetic liability for schizophrenia. In this study, we aimed to examine the influence of the functional single-nucleotide polymorphism (SNP) rs6265 of a plasticity-related neurotrophic factor gene, BDNF (Val66Met), on cognitive control. Strong evidence implicates BDNF Val66Met in neural plasticity in humans. Furthermore, several studies suggest that although the variant is not convincingly associated with schizophrenia risk, it seems to be a modifier of the clinical presentation and course of the disease. In order to clarify the underlying mechanisms using functional magnetic resonance imaging (fMRI), we studied the effects of this SNP on ACC and PFC activation, and the connectivity between these regions in a discovery sample of 85 healthy individuals and sought to replicate this effect in an independent sample of 253 individuals. Additionally, we tested the identified imaging phenotype in relation to schizophrenia familial risk in a sample of 58 unaffected first-degree relatives of schizophrenia patients. We found a significant increase in interregional connectivity between ACC and PFC in the risk-associated BDNF 66Met allele carriers. Furthermore, we replicated this effect in an independent sample and demonstrated its independence of structural confounds, as well as task specificity. A similar coupling increase was detectable in individuals with increased familial risk for schizophrenia. Our results show that a key neural circuit for cognitive control is influenced by a plasticity-related genetic variant, which may render this circuit particular susceptible to genetic and environmental risk factors for schizophrenia.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Connectome , Executive Function/physiology , Gyrus Cinguli/physiopathology , Nerve Net/physiopathology , Neuronal Plasticity/genetics , Prefrontal Cortex/physiopathology , Schizophrenia/genetics , Adult , Female , Genetic Predisposition to Disease , Gyrus Cinguli/diagnostic imaging , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Polymorphism, Single Nucleotide , Prefrontal Cortex/diagnostic imaging , Young Adult
3.
Eur Neuropsychopharmacol ; 28(3): 415-427, 2018 03.
Article in English | MEDLINE | ID: mdl-29358097

ABSTRACT

Major depressive disorder (MDD) is characterized by low mood for at least two weeks. Impaired emotion regulation has been suggested to be the consequence of dysfunctional serotonergic regulation of limbic and prefrontal regions, especially the amygdala, the anterior cingulate cortex (ACC) and the prefrontal cortex (PFC). The impact of genetic variation on brain function can be investigated with intermediate phenotypes. A suggested intermediate phenotype of MDD is emotion recognition: The 5-HTTLPR polymorphism of SLC6A4 as well as other serotonergic genes have been associated with amygdala and prefrontal function during emotion recognition. Previously, it has been suggested that habituation is a more reliable index of emotion recognition than functional activation. We examined the relationship of genes involved in serotonergic signaling with amygdala as well as prefrontal functional activation and habituation during an emotion recognition task in 171 healthy subjects. While effects of 5-HTTLPR and of a serotonergic multi-marker score (5-HTTLPR, TPH1(rs1800532), TPH2(rs4570625), HTR1A(rs6295) and HTR2A(rs6311)) on amygdala activation did not withstand correction for multiple regions of interest, we observed a strong correlation of the multi-marker score and habituation in the amygdala, DLPFC, and ACC. We replicated a well-studied intermediate phenotype for association with 5-HTTLPR and provided additional evidence for polygenic involvement. Furthermore, we showed that task habituation may be influenced by genetic variation in serotonergic signaling, particularly by a serotonergic multi-marker score. We provided preliminary evidence that PFC activation is an important intermediate phenotype of MDD. Future studies are needed to corroborate the results in larger samples.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Habituation, Psychophysiologic/physiology , Magnetic Resonance Imaging , Serotonin Plasma Membrane Transport Proteins/genetics , Signal Transduction/physiology , Adolescent , Adult , Amygdala/diagnostic imaging , Cohort Studies , Female , Genotype , Gyrus Cinguli/diagnostic imaging , Healthy Volunteers , Humans , Image Processing, Computer-Assisted , Male , Middle Aged , Oxygen/blood , Phosphorylation , Prefrontal Cortex/diagnostic imaging , PubMed/statistics & numerical data , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism , Surveys and Questionnaires , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism , Young Adult
4.
Transl Psychiatry ; 7(1): e997, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28072415

ABSTRACT

Recently, 125 loci with genome-wide support for association with schizophrenia were identified. We investigated the impact of these variants and their accumulated genetic risk on brain activation in five neurocognitive domains of the Research Domain Criteria (working memory, reward processing, episodic memory, social cognition and emotion processing). In 578 healthy subjects we tested for association (i) of a polygenic risk profile score (RPS) including all single-nucleotide polymorphisms (SNPs) reaching genome-wide significance in the recent genome-wide association studies (GWAS) meta-analysis and (ii) of all independent genome-wide significant loci separately that showed sufficient distribution of all allelic groups in our sample (105 SNPs). The RPS was nominally associated with perigenual anterior cingulate and posterior cingulate/precuneus activation during episodic memory (PFWE(ROI)=0.047) and social cognition (PFWE(ROI)=0.025), respectively. Single SNP analyses revealed that rs9607782, located near EP300, was significantly associated with amygdala recruitment during emotion processing (PFWE(ROI)=1.63 × 10-4, surpassing Bonferroni correction for the number of SNPs). Importantly, this association was replicable in an independent sample (N=150; PFWE(ROI)<0.025). Other SNP effects previously associated with imaging phenotypes were nominally significant, but did not withstand correction for the number of SNPs tested. To assess whether there was true signal within our data, we repeated single SNP analyses with 105 randomly chosen non-schizophrenia-associated variants, observing fewer significant results and lower association probabilities. Applying stringent methodological procedures, we found preliminary evidence for the notion that genetic risk for schizophrenia conferred by rs9607782 may be mediated by amygdala function. We critically evaluate the potential caveats of the methodological approaches employed and offer suggestions for future studies.


Subject(s)
Brain/physiopathology , Emotions , Memory, Episodic , Memory, Short-Term , Reward , Schizophrenia/genetics , Social Perception , Amygdala/diagnostic imaging , Amygdala/physiopathology , Brain/diagnostic imaging , Functional Neuroimaging , Genetic Predisposition to Disease , Genotype , Gyrus Cinguli/diagnostic imaging , Gyrus Cinguli/physiopathology , Healthy Volunteers , Humans , Magnetic Resonance Imaging , Multifactorial Inheritance , Parietal Lobe/diagnostic imaging , Parietal Lobe/physiopathology , Phenotype , Polymorphism, Single Nucleotide , Schizophrenia/diagnostic imaging , Schizophrenia/physiopathology , Schizophrenic Psychology
5.
Psychol Med ; 46(15): 3187-3198, 2016 11.
Article in English | MEDLINE | ID: mdl-27573157

ABSTRACT

BACKGROUND: Bipolar disorder (BD), with the hallmark symptoms of elevated and depressed mood, is thought to be characterized by underlying alterations in reward-processing networks. However, to date the neural circuitry underlying abnormal responses during reward processing in BD remains largely unexplored. The aim of this study was to investigate whether euthymic BD is characterized by aberrant ventral striatal (VS) activation patterns and altered connectivity with the prefrontal cortex in response to monetary gains and losses. METHOD: During functional magnetic resonance imaging 20 euthymic BD patients and 20 age-, gender- and intelligence quotient-matched healthy controls completed a monetary incentive delay paradigm, to examine neural processing of reward and loss anticipation. A priori defined regions of interest (ROIs) included the VS and the anterior prefrontal cortex (aPFC). Psychophysiological interactions (PPIs) between these ROIs were estimated and tested for group differences for reward and loss anticipation separately. RESULTS: BD participants, relative to healthy controls, displayed decreased activation selectively in the left and right VS during anticipation of reward, but not during loss anticipation. PPI analyses showed decreased functional connectivity between the left VS and aPFC in BD patients compared with healthy controls during reward anticipation. CONCLUSIONS: This is the first study showing decreased VS activity and aberrant connectivity in the reward-processing circuitry in euthymic, medicated BD patients during reward anticipation. Our findings contrast with research supporting a reward hypersensitivity model of BD, and add to the body of literature suggesting that blunted activation of reward processing circuits may be a vulnerability factor for mood disorders.


Subject(s)
Anticipation, Psychological , Bipolar Disorder/physiopathology , Prefrontal Cortex/physiopathology , Reward , Ventral Striatum/physiopathology , Adult , Bipolar Disorder/diagnostic imaging , Brain/physiopathology , Female , Functional Neuroimaging , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Motivation , Neural Pathways/physiopathology , Prefrontal Cortex/diagnostic imaging , Ventral Striatum/diagnostic imaging
6.
Brain Struct Funct ; 220(4): 2373-85, 2015 Jul.
Article in English | MEDLINE | ID: mdl-24874919

ABSTRACT

The s/s-genotype of the 5-HTTLPR polymorphism and the personality trait of neuroticism have both been associated with experiences of negative affect, anxiety and mood disorders, as well as an emotional processing bias towards negative facial emotions. On a neural level, this bias can be characterized by altered amygdala and fusiform gyrus (FFG) activity during perception of negative facial expressions. Using resting-state functional magnetic resonance imaging in a multi-center-sample of 178 healthy subjects of European descent, this study investigated the association of 5-HTTLPR (short s- and long l-allele) including the genotype of the single nucleotide polymorphism (SNP) rs25531 (A/G) within this region polymorphism, and trait neuroticism on resting-state functional connectivity (rs-FC) between amygdala and the FFG. Moreover, we aimed to identify additional brain regions with associations of 5-HTTLPR/rs25531 (combined according to its expression; low: s/s; high: l(A)/l(A); intermediate: s/l(A), s/l(G), l(G)/l(G), l(A)/l(G)) and trait neuroticism to amygdala rs-FC. Separate analyses for 5-HTTLPR/rs25531 and neuroticism (controlling for age, gender, handedness, and research site) revealed that s/s-homozygotes and individuals high in neuroticism obtained altered amygdala rs-FC in the right occipital face area, which is considered to be a "core component" of the face processing system. Importantly, effects of neuroticism were replicated across three independent research sites. Additionally, associations of 5-HTTLPR/rs25531 genotype and amygdala rs-FC were observed in the anterior and posterior cingulate cortex, whereas neuroticism was not related to rs-FC in these areas. The presented data implies that 5-HTTLPR/rs25531 variants and neuroticism are linked by resting state functional connectivity of amygdala and fusiform gyrus and suggests that variants of 5-HTTLPR/rs25531 genotype and different levels of neuroticism may partly account for altered processing of negative facial emotions.


Subject(s)
Amygdala/blood supply , Anxiety Disorders/genetics , Anxiety Disorders/pathology , Polymorphism, Single Nucleotide/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Temporal Lobe/blood supply , Adult , Female , Genotype , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Neural Pathways/blood supply , Neuroticism , Oxygen/blood , Rest , Young Adult
7.
Transl Psychiatry ; 4: e372, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24643163

ABSTRACT

Recent genome-wide association studies have pointed to single-nucleotide polymorphisms (SNPs) in genes encoding the neuronal calcium channel CaV1.2 (CACNA1C; rs1006737) and the presynaptic active zone protein Piccolo (PCLO; rs2522833) as risk factors for affective disorders, particularly major depression. Previous neuroimaging studies of depression-related endophenotypes have highlighted the role of the subgenual cingulate cortex (CG25) in negative mood and depressive psychopathology. Here, we aimed to assess how recently associated PCLO and CACNA1C depression risk alleles jointly affect memory-related CG25 activity as an intermediate phenotype in clinically healthy humans. To investigate the combined effects of rs1006737 and rs2522833 on the CG25 response, we conducted three functional magnetic resonance imaging studies of episodic memory formation in three independent cohorts (N=79, 300, 113). An epistatic interaction of PCLO and CACNA1C risk alleles in CG25 during memory encoding was observed in all groups, with carriers of no risk allele and of both risk alleles showing higher CG25 activation during encoding when compared with carriers of only one risk allele. Moreover, PCLO risk allele carriers showed lower memory performance and reduced encoding-related hippocampal activation. In summary, our results point to region-specific epistatic effects of PCLO and CACNA1C risk variants in CG25, potentially related to episodic memory. Our data further suggest that genetic risk factors on the SNP level do not necessarily have additive effects but may show complex interactions. Such epistatic interactions might contribute to the 'missing heritability' of complex phenotypes.


Subject(s)
Calcium Channels, L-Type/genetics , Cytoskeletal Proteins/genetics , Depressive Disorder, Major/genetics , Epistasis, Genetic/genetics , Gyrus Cinguli/physiopathology , Memory, Episodic , Neuropeptides/genetics , Adult , Functional Neuroimaging , Hippocampus/physiopathology , Humans , Magnetic Resonance Imaging , Phenotype , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL
...