Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Schizophr Res ; 155(1-3): 1-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24680031

ABSTRACT

Neurexin 1 (NRXN1) is a presynaptic neuronal adhesion molecule that interacts with postsynaptic neuroligins in both glutamatergic and GABAergic synapses and is important in synaptic formation and function. NRXN1 deletions increase the risk of schizophrenia, so our aims were to explore this in our family sample, to distinguish de novo from inherited mutations, to examine transmission to affected and unaffected siblings and to estimate penetrance. We performed copy number analyses in NRXN1 using data from Illumina BeadArrays from 635 subjects with schizophrenia (276 in genotyped families), 487 of their unaffected parents and 309 unaffected siblings as well as 635 normal controls, all from the CBDB/NIMH Genetic Study of Schizophrenia. Deletions called by software were confirmed by quantitative PCR and comparative genome hybridization. There were deletions in 15 individuals in 11 families, including de novo exonic deletions in one case and one unaffected sibling. We observed no deletions in controls, 7 deletions in cases (1.10%), and an unexpectedly high deletion frequency in parents (n=5, 1.02%) and siblings (n=3, 0.97%). Three families showed inheritance from an unaffected parent, and in two families an unaffected parent did not transmit to the affected offspring. Thus we have added to the evidence that NRXN1 deletions are more frequent in patients with schizophrenia than in healthy individuals. However, the presence of de novo deletions in unaffected relatives and transmission from and to unaffected family members demonstrated that while the deletions may well have been necessary for some carriers to develop schizophrenia, they were not always sufficient.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Family Health , Genetic Predisposition to Disease/genetics , Nerve Tissue Proteins/genetics , Penetrance , Schizophrenia/genetics , Sequence Deletion/genetics , Adolescent , Adult , Calcium-Binding Proteins , Female , Genotype , Humans , Male , Middle Aged , Neural Cell Adhesion Molecules , Young Adult
2.
Biol Psychiatry ; 75(9): 693-700, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24290728

ABSTRACT

BACKGROUND: Episodic memory (EM) declines with age and the rate of decline is variable across individuals. A single nucleotide polymorphism (rs17070145) in the WWC1 gene that encodes the KIBRA protein critical for long-term potentiation and memory consolidation has previously been associated with EM performance, as well as differences in hippocampal engagement during EM tasks using functional magnetic resonance imaging (fMRI). In the current study, we explore the effect of this polymorphism on EM-related activity and cognitive performance across the adult life span using fMRI. METHODS: Two hundred thirty-two healthy, Caucasian subjects (18-89 years) completed a battery of cognitive tests, as well as an EM task during an fMRI scan. RESULTS: WWC1 T carriers had significantly better delayed recall performance than CC individuals (p = .006). The relationship between increasing age and recall scores (immediate and delayed) was also significantly different between WWC1 genotype groups (p = .01). In addition to the age-related decline in hippocampal formation (HF) activation (p < .05; false discovery ratesmall volume correction-HF-region of interest), we observed an age by WWC1 genotype interaction on HF activation during encoding and retrieval. The CC group showed a significant negative association between HF activity and increasing age, while no such association was observed in the T carrier group (left HF p = .04; r-z correlation difference during encoding and retrieval; right HF p = .0008; r-z correlation difference during retrieval). CONCLUSIONS: Our results show a dynamic relationship between rs17070145 polymorphism and increasing age on neuronal activity in the hippocampal region.


Subject(s)
Aging/genetics , Aging/physiology , Hippocampus/physiology , Intracellular Signaling Peptides and Proteins/genetics , Memory, Episodic , Phosphoproteins/genetics , Polymorphism, Single Nucleotide , Adolescent , Adult , Aged , Aged, 80 and over , Brain/physiology , Brain Mapping , Genotype , Humans , Learning/physiology , Magnetic Resonance Imaging , Mental Recall , Middle Aged , Neuropsychological Tests , White People/genetics , Young Adult
3.
J Neurosci ; 32(20): 7074-81, 2012 May 16.
Article in English | MEDLINE | ID: mdl-22593075

ABSTRACT

The human Val66Met single nucleotide polymorphism in the brain-derived neurotrophic factor (BDNF) gene impacts BDNF signaling at the cellular level. At the neural-systems level, it is associated with differences in prefrontal cortex (PFC) and hippocampal function during performance of cognitive and affective tasks. Because the impact of this variant on basal prefrontal and hippocampal activity is not known but may be relevant to understanding the function of this gene in health and disease, we studied 94 healthy individuals with H2 ¹5O PET to assess regional cerebral blood flow (rCBF) during rest and tested for between-genotype differences. Because BDNF and gonadal steroid hormones conjointly influence neuronal growth, survival, and plasticity in hippocampus and PFC, we also tested for sex × genotype interactions. Finally, in light of the known impact of BDNF on plasticity and dendritic arborization, we complimented direct rCBF comparisons with connectivity analyses to determine how activity in hippocampal and prefrontal regions showing between-genotype group differences covaries with rCBF in other nodes throughout the brain in a genotype- or sex-dependent manner. Compared with Val homozygotes, Met carriers had higher rCBF in prefrontal (BA25 extending into BA10) and hippocampal/parahippocampal regions. Moreover, there were significant sex × genotype interactions in regions (including frontal, parahippocampal, and lateral temporal cortex) in which Val homozygotes showed higher rCBF in females than males, but Met carriers showed the opposite relationship. Functional connectivity analysis demonstrated that correlations of BA25, hippocampus, and parahippocampus with frontal and temporal networks were positive for Val homozygotes and negative for Met carriers. In addition, sex × genotype analysis of functional connectivity revealed that genotype affected directionality of the inter-regional correlations differentially in men versus women. Our data indicate that BDNF allelic variation and sex interactively affect basal prefrontal and hippocampal function.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Cerebrovascular Circulation/physiology , Hippocampus/physiology , Prefrontal Cortex/physiology , Adolescent , Adult , Alleles , Brain-Derived Neurotrophic Factor/genetics , Cerebrovascular Circulation/genetics , Female , Functional Neuroimaging/methods , Genotype , Hippocampus/diagnostic imaging , Humans , Male , Middle Aged , Neural Pathways/diagnostic imaging , Neural Pathways/physiology , Oxygen Radioisotopes , Polymorphism, Single Nucleotide , Positron-Emission Tomography/methods , Prefrontal Cortex/diagnostic imaging , Sex Characteristics , Temporal Lobe/diagnostic imaging , Temporal Lobe/physiology
4.
PLoS One ; 5(5): e10789, 2010 May 24.
Article in English | MEDLINE | ID: mdl-20520724

ABSTRACT

BACKGROUND: Neuregulin1 (NRG1)-ErbB signaling has been implicated in the pathogenesis of cancer and schizophrenia. We have previously reported that NRG1-stimulated migration of B lymphoblasts is PI3K-AKT1dependent and impaired in patients with schizophrenia and significantly linked to the catechol-o-methyltransferase (COMT) Val108/158Met functional polymorphism. METHODOLOGY/PRINCIPAL FINDINGS: We have now examined AKT1 activation in NRG1-stimulated B lymphoblasts and other cell models and explored a functional relationship between COMT and AKT1. NRG1-induced AKT1 phosphorylation was significantly diminished in Val carriers compared to Met carriers in both normal subjects and in patients. Further, there was a significant epistatic interaction between a putatively functional coding SNP in AKT1 (rs1130233) and COMT Val108/158Met genotype on AKT1 phosphorylation. NRG1 induced translocation of AKT1 to the plasma membrane also was impaired in Val carriers, while PIP(3) levels were not decreased. Interestingly, the level of COMT enzyme activity was inversely correlated with the cells' ability to synthesize phosphatidylserine (PS), a factor that attracts the pleckstrin homology domain (PHD) of AKT1 to the cell membrane. Transfection of SH-SY5Y cells with a COMT Val construct increased COMT activity and significantly decreased PS levels as well as NRG1-induced AKT1 phosphorylation and migration. Administration of S-adenosylmethionine (SAM) rescued all of these deficits. These data suggest that AKT1 function is influenced by COMT enzyme activity through competition with PS synthesis for SAM, which in turn dictates AKT1-dependent cellular responses to NRG1-mediated signaling. CONCLUSION/SIGNIFICANCE: Our findings implicate genetic and functional interactions between COMT and AKT1 and may provide novel insights into pathogenesis of schizophrenia and other ErbB-associated human diseases such as cancer.


Subject(s)
Catechol O-Methyltransferase/metabolism , Epistasis, Genetic , ErbB Receptors/metabolism , Models, Biological , Neuregulin-1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Amino Acid Substitution/genetics , B-Lymphocytes/enzymology , Catechol O-Methyltransferase/genetics , Cell Line, Tumor , Cell Movement , Enzyme Activation , Humans , Phosphatidylinositol Phosphates/metabolism , Phosphatidylserines/metabolism , Phosphorylation , Polymorphism, Single Nucleotide/genetics , Protein Transport , Proto-Oncogene Proteins c-akt/genetics , Transfection
5.
Brain ; 132(Pt 11): 2958-69, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19812213

ABSTRACT

Cognitive abnormalities are common in Parkinson's disease, with important social and economic implications. Factors influencing their evolution remain unclear but are crucial to the development of targeted therapeutic strategies. We have investigated the development of cognitive impairment and dementia in Parkinson's disease using a longitudinal approach in a population-representative incident cohort (CamPaIGN study, n = 126) and here present the 5-year follow-up data from this study. Our previous work has implicated two genetic factors in the development of cognitive dysfunction in Parkinson's disease, namely the genes for catechol-O-methyltransferase (COMT Val(158)Met) and microtubule-associated protein tau (MAPT) H1/H2. Here, we have explored the influence of these genes in our incident cohort and an additional cross-sectional prevalent cohort (n = 386), and investigated the effect of MAPT H1/H2 haplotypes on tau transcription in post-mortem brain samples from patients with Lewy body disease and controls. Seventeen percent of incident patients developed dementia over 5 years [incidence 38.7 (23.9-59.3) per 1000 person-years]. We have demonstrated that three baseline measures, namely, age >or=72 years, semantic fluency less than 20 words in 90 s and inability to copy an intersecting pentagons figure, are significant predictors of dementia risk, thus validating our previous findings. In combination, these factors had an odds ratio of 88 for dementia within the first 5 years from diagnosis and may reflect the syndrome of mild cognitive impairment of Parkinson's disease. Phonemic fluency and other frontally based tasks were not associated with dementia risk. MAPT H1/H1 genotype was an independent predictor of dementia risk (odds ratio = 12.1) and the H1 versus H2 haplotype was associated with a 20% increase in transcription of 4-repeat tau in Lewy body disease brains. In contrast, COMT genotype had no effect on dementia, but a significant impact on Tower of London performance, a frontostriatally based executive task, which was dynamic, such that the ability to solve this task changed with disease progression. Hence, we have identified three highly informative predictors of dementia in Parkinson's disease, which can be easily translated into the clinic, and established that MAPT H1/H1 genotype is an important risk factor with functional effects on tau transcription. Our work suggests that the dementing process in Parkinson's disease is predictable and related to tau while frontal-executive dysfunction evolves independently with a more dopaminergic basis and better prognosis.


Subject(s)
Cognition Disorders , Dementia , Parkinson Disease , Aged , Catechol O-Methyltransferase/genetics , Cognition/physiology , Cognition Disorders/etiology , Cognition Disorders/genetics , Cognition Disorders/physiopathology , Cohort Studies , Dementia/etiology , Dementia/genetics , Dementia/physiopathology , Disease Progression , Dopamine/metabolism , Female , Follow-Up Studies , Genetic Predisposition to Disease , Haplotypes , Humans , Male , Middle Aged , Neuropsychological Tests , Parkinson Disease/complications , Parkinson Disease/genetics , Parkinson Disease/physiopathology , Regression Analysis , Risk Factors , tau Proteins/genetics
6.
Neuroimage ; 45(1): 44-51, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19071221

ABSTRACT

BACKGROUND: Functional variants in the catechol-O-methyltransferase (COMT) gene have been shown to impact cognitive function, cortical physiology and risk for schizophrenia. A recent study showed that previously reported effects of the functional val158met SNP (rs4680) on brain function are modified by other functional SNPs and haplotypes in the gene, though it was unknown if these effects are also seen in brain structure. METHODS: We used voxel-based morphometry to investigate the impact of multiple functional variants in COMT on gray matter volume in a large group of 151 healthy volunteers from the CBDB/NIMH Genetic Study of Schizophrenia. RESULTS: We found that the previously described rs4680 val risk variant affects hippocampal and dorsolateral prefrontal (DLPFC) gray matter volume. In addition, we found that this SNP interacts with a variant in the P2 promoter region (rs2097603) in predicting changes in hippocampal gray matter volume consistent with a nonlinear effect of extracellular dopamine. CONCLUSIONS: We report evidence that interacting functional variants in COMT affect gray matter regional volume in hippocampus and DLPFC, providing further in vivo validation of the biological impact of complex genetic variation in COMT on neural systems relevant for the pathophysiology of schizophrenia and extending observations of nonlinear dependence of prefrontal neurons on extracellular dopamine to the domain of human brain structure.


Subject(s)
Catechol O-Methyltransferase/genetics , Hippocampus/cytology , Hippocampus/physiology , Neurons/cytology , Neurons/physiology , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology , Adult , Female , Genetic Variation/genetics , Humans , Imaging, Three-Dimensional/methods , Magnetic Resonance Imaging/methods , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Statistics as Topic
7.
Am J Psychiatry ; 166(2): 216-25, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19074979

ABSTRACT

OBJECTIVE: Although amygdala dysfunction is reported in schizophrenia, it is unknown whether this deficit represents a heritable phenotype that is related to risk for schizophrenia or whether it is related to disease state. The purpose of the present study was to examine amygdala response to threatening faces among healthy siblings of schizophrenia patients in whom a subtler heritable deficit might be observed. METHOD: Participants were 34 schizophrenia patients, 29 unaffected siblings, and 20 healthy comparison subjects. Blood-oxygen-level-dependent (BOLD) functional magnetic resonance imaging (fMRI) was conducted during an implicit facial information processing task. The N-back working memory task, which has been shown to elicit prefrontal cortex abnormalities in unaffected siblings of schizophrenia patients, was employed as a positive experimental control. RESULTS: Schizophrenia patients demonstrated a deficit in amygdala reactivity to negative face stimuli and an alteration, correlated with neuroleptic drug dosage, in the functional coupling between the amygdala and subgenual cingulate. In contrast, unaffected siblings showed a pattern that was not statistically different from that of healthy comparison subjects. During the N-back working memory task, both schizophrenia patients and their unaffected siblings demonstrated a pattern of inefficient prefrontal cortex engagement, which is consistent with earlier evidence that this pattern is related to genetic risk for schizophrenia. CONCLUSIONS: These data suggest that the pathophysiological mechanism underlying the inability of individuals with schizophrenia to normally engage the amygdala in processing fearful and angry facial representations is more likely a phenomenon related to the disease state, specifically to treatment.


Subject(s)
Amygdala/physiopathology , Cognition Disorders/genetics , Cognition Disorders/physiopathology , Genetic Predisposition to Disease/genetics , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Neuropsychological Tests , Oxygen/blood , Schizophrenia/genetics , Schizophrenia/physiopathology , Adult , Anger , Cognition Disorders/diagnosis , Dominance, Cerebral/physiology , Facial Expression , Fear/physiology , Female , Gyrus Cinguli/physiopathology , Humans , Imaging, Three-Dimensional , Male , Memory, Short-Term , Nerve Net/physiopathology , Pattern Recognition, Visual/physiology , Phenotype , Schizophrenia/diagnosis
8.
J Clin Invest ; 118(6): 2200-8, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18497887

ABSTRACT

AKT1-dependent molecular pathways control diverse aspects of cellular development and adaptation, including interactions with neuronal dopaminergic signaling. If AKT1 has an impact on dopaminergic signaling, then genetic variation in AKT1 would be associated with brain phenotypes related to cortical dopaminergic function. Here, we provide evidence that a coding variation in AKT1 that affects protein expression in human B lymphoblasts influenced several brain measures related to dopaminergic function. Cognitive performance linked to frontostriatal circuitry, prefrontal physiology during executive function, and frontostriatal gray-matter volume on MRI were altered in subjects with the AKT1 variation. Moreover, on neuroimaging measures with a main effect of the AKT1 genotype, there was significant epistasis with a functional polymorphism (Val158Met) in catechol-O-methyltransferase [COMT], a gene that indexes cortical synaptic dopamine. This genetic interaction was consistent with the putative role of AKT1 in dopaminergic signaling. Supportive of an earlier tentative association of AKT1 with schizophrenia, we also found that this AKT1 variant was associated with risk for schizophrenia. These data implicate AKT1 in modulating human prefrontal-striatal structure and function and suggest that the mechanism of this effect may be coupled to dopaminergic signaling and relevant to the expression of psychosis.


Subject(s)
Dopamine/metabolism , Gene Expression Regulation, Enzymologic , Proto-Oncogene Proteins c-akt/metabolism , Schizophrenia/genetics , Adolescent , Adult , Alleles , Brain/metabolism , Brain/pathology , Case-Control Studies , Cognition , Genotype , Humans , Middle Aged , Neurons/metabolism , Phenotype , Schizophrenia/diagnosis , Signal Transduction
9.
Biol Psychiatry ; 63(1): 72-9, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17707347

ABSTRACT

BACKGROUND: Catechol-O-methyltransferase (COMT) val(108/158)met (rs4680) is thought to affect dopamine regulated prefrontal cortical activity during working memory (WM) tasks, and to weakly increase risk for developing schizophrenia. Recently, other single nucleotide polymorphisms (SNPs) across the gene have emerged as additional risk factors for schizophrenia: namely rs737865, rs165599, and rs2097603. In a large sample, we examined whether these SNPs affect WM. METHODS: Schizophrenic probands (n = 325), their nonpsychotic siblings (n = 359), and normal control subjects (n = 330) completed tests of WM function. Data were analyzed with a series of mixed model analyses of variance (ANOVAs). RESULTS: Val homozygotes performed most poorly on all conditions of the n-back, irrespective of diagnosis. Additionally, there was a trend towards a disease-only val(108/158)met effect on a test of attentional set-shifting; val homozygote probands performed most poorly. Significant or near-significant effects of rs737865 were found on all conditions of the n-back, with G homozygotes performing worst. There also was a disease-only COMT rs737865 effect on the 0-back. None of the other SNPs showed main effects by themselves. A haplotype constructed from promoter and val(108/158)met SNPs showed main effects on WM parameters, consistent with inverted U models of dopamine signaling. CONCLUSIONS: We extended earlier findings of a val(108/158)met effect on WM function, and suggest that combinations of alleles within COMT may modulate the val(108/158)met effect in a nonlinear manner.


Subject(s)
Catechol O-Methyltransferase/genetics , Family Health , Memory, Short-Term/physiology , Polymorphism, Single Nucleotide , Schizophrenia/genetics , Schizophrenia/physiopathology , Adult , Case-Control Studies , Female , Genotype , Humans , Male , Methionine/genetics , Middle Aged , Neuropsychological Tests , Promoter Regions, Genetic/genetics , Valine/genetics
10.
Hum Genet ; 120(6): 889-906, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17006672

ABSTRACT

Catechol-O-methyltransferase (COMT) regulates dopamine degradation and is located in a genomic region that is deleted in a syndrome associated with psychosis, making it a promising candidate gene for schizophrenia. COMT also has been shown to influence prefrontal cortex processing efficiency. Prefrontal processing dysfunction is a common finding in schizophrenia, and a background of inefficient processing may modulate the effect of other candidate genes. Using the NIMH sibling study (SS), a non-independent case-control set, and an independent German (G) case-control set, we performed conditional/unconditional logistic regression to test for epistasis between SNPs in COMT (rs2097603, Val158Met (rs4680), rs165599) and polymorphisms in other schizophrenia susceptibility genes. Evidence for interaction was evaluated using a likelihood ratio test (LRT) between nested models. SNPs in RGS4, G72, GRM3, and DISC1 showed evidence for significant statistical epistasis with COMT. A striking result was found in RGS4: three of five SNPs showed a significant increase in risk [LRT P-values: 90387 = 0.05 (SS); SNP4 = 0.02 (SS), 0.02 (G); SNP18 = 0.04 (SS), 0.008 (G)] in interaction with COMT; main effects for RGS4 SNPs were null. Significant results for SNP4 and SNP18 were also found in the German study. We were able to detect statistical interaction between COMT and polymorphisms in candidate genes for schizophrenia, many of which had no significant main effect. In addition, we were able to replicate other studies, including allelic directionality. The use of epistatic models may improve replication of psychiatric candidate gene studies.


Subject(s)
Carrier Proteins/genetics , Catechol O-Methyltransferase/genetics , Epistasis, Genetic , Nerve Tissue Proteins/genetics , Polymorphism, Single Nucleotide , RGS Proteins/genetics , Receptors, Metabotropic Glutamate/genetics , Schizophrenia/enzymology , Schizophrenia/genetics , Adolescent , Adult , Alleles , Case-Control Studies , Female , Genetic Predisposition to Disease , Genotype , Haplotypes , Humans , Intracellular Signaling Peptides and Proteins , Male , Middle Aged , Models, Genetic , Risk Factors , Siblings
11.
Neuropsychopharmacology ; 32(5): 1011-20, 2007 May.
Article in English | MEDLINE | ID: mdl-17063156

ABSTRACT

Prefrontal cortical dopamine (DA) regulates various executive cognitive functions, including attention and working memory. Efforts to enhance prefrontal-related cognition, which have focused on catecholaminergic stimulant drugs, have been unsatisfactory. Recently, the demonstration that a functional polymorphism in the catecholamine-O-methyltransferase (COMT) gene impacts prefrontal cognition raises the possibility of a novel pharmacological approach for the treatment of prefrontal lobe executive dysfunction. To explore in a proof of concept study the effects of tolcapone, a CNS penetrant specific COMT inhibitor, we performed a randomized, double blind, placebo controlled, and crossover design of this drug in normal subjects stratified by COMT (val158met) genotype. COMT enzyme activity was determined in peripheral blood. Forty-seven normal volunteers with no family history of psychiatric disorders underwent neuropsychological testing and 34 of those subjects underwent physiological measurement of prefrontal information processing assessed by blood oxygen level-dependent functional magnetic resonance imaging (fMRI). We found significant drug effects on measures of executive function and verbal episodic memory and a significant drug by genotype interaction on the latter, such that individuals with val/val genotypes improved, whereas individuals with met/met genotypes worsened on tolcapone. fMRI revealed a significant tolcapone-induced improvement in the efficiency of information processing in prefrontal cortex during a working memory test. This study demonstrates enhancement of prefrontal cortical function in normal human subjects with a nonstimulant drug having COMT inhibitory activity. Our results are consistent with data from animal studies and from computational models of the effects of selective enhancement of DA signaling in the prefrontal cortex.


Subject(s)
Benzophenones/pharmacology , Catechol O-Methyltransferase Inhibitors , Cognition/drug effects , Memory/drug effects , Nitrophenols/pharmacology , Prefrontal Cortex/drug effects , Adult , Amino Acid Substitution/genetics , Benzophenones/adverse effects , Brain Mapping , Catechol O-Methyltransferase/genetics , Catechol O-Methyltransferase/metabolism , Cognition/physiology , Cross-Over Studies , Double-Blind Method , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacology , Female , Genotype , Humans , Magnetic Resonance Imaging , Male , Memory/physiology , Middle Aged , Neuropsychological Tests , Nitrophenols/adverse effects , Placebos , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiology , Reference Values , Tolcapone , Treatment Outcome
12.
Arch Gen Psychiatry ; 63(12): 1396-406, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17146014

ABSTRACT

CONTEXT: Catechol O-methyltransferase (COMT), the major enzyme determining cortical dopamine flux, has a common functional polymorphism (val(158)met) that affects prefrontal function and working memory capacity and has also been associated with anxiety and emotional dysregulation. OBJECTIVES: To examine COMT val(158)met effects on corticolimbic circuitry reactivity and functional connectivity during processing of biologically salient stimuli, as well as the relationship to the temperamental trait of novelty seeking. DESIGN: Within-subject functional magnetic resonance imaging study. SETTING: National Institute of Mental Health, Genes, Cognition, and Psychosis Program, Bethesda, Md. Patients One hundred one healthy subjects of both sexes. RESULTS: We found that the met allele was associated with a dose-dependent increase in hippocampal formation and ventrolateral prefrontal cortex activation during viewing of faces displaying negative emotion. In met/met homozygotes, limbic and prefrontal regions showed increased functional coupling. Moreover, in these same subjects, the magnitude of amygdala-orbitofrontal coupling was inversely correlated with novelty seeking, an index of temperamental inflexibility. CONCLUSIONS: Our results indicate that heritable variation in dopamine neurotransmission associated with the met allele of the COMT polymorphism results in heightened reactivity and connectivity in corticolimbic circuits. This may reflect a genetic predisposition for inflexible processing of affective stimuli, a mechanism possibly accounting for aspects of arousal and behavioral control that contribute to emotional dysregulation previously reported in met/met individuals.


Subject(s)
Affect/physiology , Affect/radiation effects , Affective Symptoms/genetics , Arousal/physiology , Catechol O-Methyltransferase/genetics , Limbic System/physiology , Polymorphism, Genetic/physiology , Prefrontal Cortex/physiology , Adult , Affective Symptoms/diagnosis , Amygdala/physiology , Arousal/genetics , Brain Mapping/methods , Exploratory Behavior/physiology , Facial Expression , Female , Genotype , Humans , Magnetic Resonance Imaging/statistics & numerical data , Male , Methionine/genetics , Neural Pathways/physiology , Oxygen/blood , Synaptic Transmission/physiology , Temperament/physiology , Valine/genetics , Visual Perception/physiology
13.
Biol Psychiatry ; 60(6): 578-84, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16730334

ABSTRACT

BACKGROUND: Increased variability of stimulus-induced prefrontal electromagnetic activity ("noise") has been associated with genetic risk for schizophrenia. On the basis of animal experiments and computational models, we have predicted that this prefrontal "noise" phenotype would be related to variation in prefrontal dopamine (DA) signaling, which itself might be abnormal in schizophrenia. In the present study, the effect of a functional single nucleotide polymorphism (val(108/158)met) within the catechol-O-methyltransferase (COMT) gene on prefrontal "noise" was examined, because the COMT enzyme is involved in cortical synaptic dopamine metabolism and weakly predictive of risk for schizophrenia. METHODS: A Caucasian sample comprising 112 unrelated normal subjects, 83 schizophrenic probands, and 87 of their unaffected siblings was investigated, all of whom had measures of prefrontal "noise" estimated from event-related electroencephalogram during an auditory oddball task. RESULTS: The val(108/158)met genotype was significantly associated with prefrontal "noise"; homozygous Val-carriers had greatest prefrontal "noise" values; odds ratio (OR) = 2.37 (95% confidence interval [CI] 1.37-4.10), p = 003. The genotype-phenotype association was stronger when only considering male subjects with an OR = 3.37 (95% CI: 1.63-6.98), p = 002. CONCLUSIONS: The results suggest that COMT genotype impacts the level of prefrontal physiologic "noise."


Subject(s)
Catechol O-Methyltransferase/physiology , Genetic Predisposition to Disease , Noise , Prefrontal Cortex/physiopathology , Schizophrenia , Acoustic Stimulation/methods , Adolescent , Adult , Analysis of Variance , Confidence Intervals , DNA Mutational Analysis , Female , Gene Frequency , Genotype , Humans , Male , Methionine/genetics , Middle Aged , Odds Ratio , Polymorphism, Single Nucleotide/genetics , Retrospective Studies , Schizophrenia/genetics , Schizophrenia/pathology , Schizophrenia/physiopathology , Sex Factors , Valine/genetics
14.
Nat Neurosci ; 8(6): 828-34, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15880108

ABSTRACT

Carriers of the short allele of a functional 5' promoter polymorphism of the serotonin transporter gene have increased anxiety-related temperamental traits, increased amygdala reactivity and elevated risk of depression. Here, we used multimodal neuroimaging in a large sample of healthy human subjects to elucidate neural mechanisms underlying this complex genetic association. Morphometrical analyses showed reduced gray matter volume in short-allele carriers in limbic regions critical for processing of negative emotion, particularly perigenual cingulate and amygdala. Functional analysis of those regions during perceptual processing of fearful stimuli demonstrated tight coupling as a feedback circuit implicated in the extinction of negative affect. Short-allele carriers showed relative uncoupling of this circuit. Furthermore, the magnitude of coupling inversely predicted almost 30% of variation in temperamental anxiety. These genotype-related alterations in anatomy and function of an amygdala-cingulate feedback circuit critical for emotion regulation implicate a developmental, systems-level mechanism underlying normal emotional reactivity and genetic susceptibility for depression.


Subject(s)
Amygdala/metabolism , Anxiety Disorders/genetics , Depressive Disorder/genetics , Genetic Predisposition to Disease/genetics , Gyrus Cinguli/metabolism , Membrane Glycoproteins/genetics , Membrane Transport Proteins/genetics , Nerve Tissue Proteins/genetics , Amygdala/pathology , Amygdala/physiopathology , Anthropometry , Anxiety Disorders/metabolism , Anxiety Disorders/pathology , Atrophy/genetics , Atrophy/metabolism , Atrophy/pathology , Brain Chemistry/genetics , Brain Mapping , Depressive Disorder/metabolism , Depressive Disorder/pathology , Fear/physiology , Fear/psychology , Gyrus Cinguli/pathology , Gyrus Cinguli/physiopathology , Humans , Magnetic Resonance Imaging , Mutation/genetics , Neural Pathways/metabolism , Neural Pathways/pathology , Neural Pathways/physiopathology , Neuropsychological Tests , Polymorphism, Genetic/genetics , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins , Surveys and Questionnaires
15.
J Neurosci ; 25(20): 5038-45, 2005 May 18.
Article in English | MEDLINE | ID: mdl-15901785

ABSTRACT

The cingulate cortex is richly innervated by dopaminergic projections and plays a critical role in attentional control (AC). Evidence indicates that dopamine enhances the neurophysiological signal-to-noise ratio and that dopaminergic tone in the frontal cortex is critically dependent on catechol-O-methyltransferase (COMT). A functional polymorphism (val158met) in the COMT gene accounts for some of the individual variability in executive function mediated by the dorsolateral prefrontal cortex. We explored the effect of this genetic polymorphism on cingulate engagement during a novel AC task. We found that the COMT val158met polymorphism also affects the function of the cingulate during AC. Individuals homozygous for the high-activity valine ("val") allele show greater activity and poorer performance than val/methionine ("met") heterozygotes, who in turn show greater activity and poorer performance than individuals homozygous for the low-activity met allele, and these effects are most evident at the highest demand for AC. These results indicate that met allele load and presumably enhanced dopaminergic tone improve the "efficiency" of local circuit processing within the cingulate cortex and thereby its function during AC.


Subject(s)
Attention/physiology , Catechol O-Methyltransferase/genetics , Methionine/genetics , Polymorphism, Genetic , Prefrontal Cortex/physiology , Valine/genetics , Adult , Analysis of Variance , Brain Mapping , Case-Control Studies , Catechol O-Methyltransferase/physiology , Female , Genotype , Humans , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Male , Neuropsychological Tests/statistics & numerical data , Oxygen/blood , Photic Stimulation/methods , Prefrontal Cortex/blood supply , Reaction Time/genetics , Regression Analysis
16.
J Neurol ; 252(7): 833-8, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15772739

ABSTRACT

Parkinson's disease (PD) patients show a range of cognitive deficits,which may relate to abnormalities in dopaminergic transmission in fronto-striatal circuitry. In this study, we have investigated the impact of brainderived neurotrophic factor (BDNF) val66met polymorphisms on performance of the Tower of London (TOL) test of planning by PD patients. This polymorphism significantly influences BDNF secretion in the CNS, and BDNF is known to influence dopaminergic neurons and cognitive processes. Patients with PD totalling 291 who had undergone detailed motor and cognitive assessments as part of a population-based study of PD were genotyped for the BDNF val66met polymorphism. The impact of this polymorphism on cognitive ability was determined using multivariate analysis to adjust for possible confounding variables. Patients with low rates of BDNF secretion (met alleles) performed significantly better at the TOL task than those with high rates of secretion (val alleles). Furthermore, subgroup analyses revealed that the effect is most apparent in women and among patients with prior dopaminergic exposure. We speculate that BDNF may interact with dopaminergic transmission and dopamine receptor stimulation in the frontostriatal circuitry, with subsequent consequences on cognition in Parkinson's disease.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Mental Processes/physiology , Methionine/genetics , Parkinson Disease/physiopathology , Polymorphism, Genetic , Sex Characteristics , Valine/genetics , Aged , Analysis of Variance , Female , Gene Frequency , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Neuropsychological Tests/statistics & numerical data , Parkinson Disease/genetics
17.
Arch Gen Psychiatry ; 62(2): 146-52, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15699291

ABSTRACT

BACKGROUND: A common regulatory variant (5-HTTLPR) in the human serotonin transporter gene (SLC6A4), resulting in altered transcription and transporter availability, has been associated with vulnerability for affective disorders, including anxiety and depression. A recent functional magnetic resonance imaging study suggested that this association may be mediated by 5-HTTLPR effects on the response bias of the human amygdala-a brain region critical for emotional and social behavior-to environmental threat. OBJECTIVES AND DESIGN: To examine the effects of 5-HTTLPR genotype on the reactivity of the human amygdala to salient environmental cues with functional magnetic resonance imaging in a large (N = 92) cohort of volunteers carefully screened for past and present medical or psychiatric illness, and to explore the effects of 5-HTTLPR genotype as well as amygdala reactivity on harm avoidance, a putative personality measure related to trait anxiety. RESULTS: We now confirm the finding of 5-HTTLPR short allele-driven amygdala hyperreactivity in a large independent cohort of healthy subjects with no history of psychiatric illness or treatment. Furthermore, we demonstrate that these genotype effects on amygdala function are consistent with a dominant short allele effect and are equally prominent in men and women. However, neither 5-HTTLPR genotype, amygdala reactivity, nor genotype-driven variability in this reactivity was reflected in harm avoidance scores. CONCLUSIONS: Our results reveal a potent modulatory effect of the 5-HTTLPR on amygdala reactivity to environmental threat. Since this genetically driven effect exists in healthy subjects, it does not, in and of itself, predict dimensions of mood or temperament. As such, the 5-HTTLPR may represent a classic susceptibility factor for affective disorders by biasing the functional reactivity of the human amygdala in the context of stressful life experiences and/or deficient cortical regulatory input.


Subject(s)
Amygdala/physiology , Emotions/physiology , Membrane Glycoproteins/genetics , Membrane Transport Proteins/genetics , Mood Disorders/genetics , Nerve Tissue Proteins/genetics , Adult , Anger/physiology , Facial Expression , Fear/physiology , Female , Genetic Predisposition to Disease/genetics , Genetic Variation/physiology , Genotype , Humans , Magnetic Resonance Imaging , Male , Mood Disorders/diagnosis , Mood Disorders/epidemiology , Oxygen/blood , Serotonin Plasma Membrane Transport Proteins , Sex Distribution , Stress, Psychological/diagnosis , Stress, Psychological/genetics , Transcription, Genetic/physiology , Visual Perception/physiology
18.
J Neurosci ; 24(45): 10099-102, 2004 Nov 10.
Article in English | MEDLINE | ID: mdl-15537879

ABSTRACT

A variation in the BDNF gene (val66met) affects the function of BDNF in neurons, predicts variation in human memory, and is associated with several neurological and psychiatric disorders. Here, we show that, in magnetic resonance imaging scans of a large sample of normal individuals, this polymorphism affects the anatomy of the hippocampus and prefrontal cortex, identifying a genetic mechanism of variation in brain morphology related to learning and memory.


Subject(s)
Amino Acid Substitution , Brain-Derived Neurotrophic Factor/genetics , Genetic Variation , Hippocampus/ultrastructure , Neuronal Plasticity , Prefrontal Cortex/ultrastructure , Adolescent , Adult , Alleles , Brain-Derived Neurotrophic Factor/physiology , Female , Humans , Imaging, Three-Dimensional , Learning/physiology , Magnetic Resonance Imaging , Male , Memory/physiology , Middle Aged , Organ Size , Polymorphism, Genetic
19.
Biol Psychiatry ; 56(9): 677-82, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15522252

ABSTRACT

BACKGROUND: The gene encoding catechol-O-methyltransferase (COMT), an enzyme that regulates prefrontal cortex dopamine, contains a common functional polymorphism (val(108/158)met) that influences prefrontal cortex function in an allelic dose-dependent manner. A recent study reported that the COMT val(108/158)met polymorphism influences cognitive- and physiologic-related prefrontal cortex responses to antipsychotic treatment. The present study tested the effects of several COMT polymorphisms on the cognitive response to antipsychotic medication in patients with schizophrenia. METHODS: Twenty inpatients with schizophrenia or schizoaffective disorder (5 with the val-val genotype, 11 with val-met, and 4 with met-met) were administered cognitive tests at two time points: once after 4 weeks of treatment with antipsychotic medication and once after 4 weeks of placebo administration, according to a counterbalanced, double-blind, within-subject study design. RESULTS: Patients homozygous for the COMT met allele displayed significant improvement on the working memory task after treatment. Patients homozygous for the COMT val allele did not show working memory improvement with treatment. Other COMT polymorphisms were not associated with significant differences between treatment and placebo conditions. CONCLUSIONS: These results support other data suggesting that the COMT val(108/158)met polymorphism might be an important factor in the cognitive response to antipsychotic medication.


Subject(s)
Antipsychotic Agents/therapeutic use , Catechol O-Methyltransferase/genetics , Memory, Short-Term/drug effects , Schizophrenia/drug therapy , Schizophrenia/genetics , Adult , Analysis of Variance , Case-Control Studies , Double-Blind Method , Female , Genotype , Humans , Male , Mental Status Schedule , Methionine/genetics , Neuropsychological Tests/statistics & numerical data , Polymorphism, Genetic , Retrospective Studies , Valine/genetics
20.
Am J Hum Genet ; 75(5): 807-21, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15457404

ABSTRACT

Catechol-O-methyltransferase (COMT) is a key enzyme in the elimination of dopamine in the prefrontal cortex of the human brain. Genetic variation in the COMT gene (MIM 116790) has been associated with altered prefrontal cortex function and higher risk for schizophrenia, but the specific alleles and their functional implications have been controversial. We analyzed the effects of several single-nucleotide polymorphisms (SNPs) within COMT on mRNA expression levels (using reverse-transcriptase polymerase chain reaction analysis), protein levels (using Western blot analysis), and enzyme activity (using catechol methylation) in a large sample (n = 108) of postmortem human prefrontal cortex tissue, which predominantly expresses the -membrane-bound isoform. A common coding SNP, Val158Met (rs4680), significantly affected protein abundance and enzyme activity but not mRNA expression levels, suggesting that differences in protein integrity account for the difference in enzyme activity between alleles. A SNP in intron 1 (rs737865) and a SNP in the 3' flanking region (rs165599)--both of which have been reported to contribute to allelic expression differences and to be associated with schizophrenia as part of a haplotype with Val--had no effect on mRNA expression levels, protein immunoreactivity, or enzyme activity. In lymphocytes from 47 subjects, we confirmed a similar effect on enzyme activity in samples with the Val/Met genotype but no effect in samples with the intron 1 or 3' SNPs. Separate analyses revealed that the subject's sex, as well as the presence of a SNP in the P2 promoter region (rs2097603), had small effects on COMT enzyme activity. Using site-directed mutagenesis of mouse COMT cDNA, followed by in vitro translation, we found that the conversion of Leu at the homologous position into Met or Val progressively and significantly diminished enzyme activity. Thus, although we cannot exclude a more complex genetic basis for functional effects of COMT, Val is a predominant factor that determines higher COMT activity in the prefrontal cortex, which presumably leads to lower synaptic dopamine levels and relatively deleterious prefrontal function.


Subject(s)
Catechol O-Methyltransferase/genetics , Gene Expression Regulation, Enzymologic/physiology , Genetic Variation , Prefrontal Cortex/enzymology , Proteins/metabolism , RNA, Messenger/metabolism , Analysis of Variance , Animals , Blotting, Western , DNA Primers , Genotype , Humans , Lymphocytes/enzymology , Methylation , Mice , Mutagenesis, Site-Directed , Polymorphism, Single Nucleotide , Prefrontal Cortex/pathology , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...