Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Mol Psychiatry ; 23(4): 1066-1075, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28416813

RESUMO

Preclinical evidence suggests that the actions of ovarian steroid hormones and brain-derived neurotrophic factor (BDNF) are highly convergent on brain function. Studies in humanized mice document an interaction between estrus cycle-related changes in estradiol secretion and BDNF Val66Met genotype on measures of hippocampal function and anxiety-like behavior. We believe our multimodal imaging data provide the first demonstration in women that the effects of the BDNF Val/Met polymorphism on hippocampal function are selectively modulated by estradiol. In a 6-month pharmacological hormone manipulation protocol, healthy, regularly menstruating, asymptomatic women completed positron emission tomography (PET) and functional magnetic resonance imaging (fMRI) scans while performing the n-back working memory task during three hormone conditions: ovarian suppression induced by the gonadotropin-releasing hormone agonist, leuprolide acetate; leuprolide plus estradiol; and leuprolide plus progesterone. For each of the three hormone conditions, a discovery data set was obtained with oxygen-15 water regional cerebral blood flow PET in 39 healthy women genotyped for BDNF Val66Met, and a confirmatory data set was obtained with fMRI in 27 women. Our results, in close agreement across the two imaging platforms, demonstrate an ovarian hormone-by-BDNF interaction on working memory-related hippocampal function (PET: F2,37=9.11, P=0.00026 uncorrected, P=0.05, familywise error corrected with small volume correction; fMRI: F2,25=5.43, P=0.01, uncorrected) that reflects differential hippocampal recruitment in Met carriers but only in the presence of estradiol. These findings have clinical relevance for understanding the neurobiological basis of individual differences in the cognitive and behavioral effects of ovarian steroids in women, and may provide a neurogenetic framework for understanding neuropsychiatric disorders related to reproductive hormones as well as illnesses with sex differences in disease expression and course.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Hormônios Esteroides Gonadais/metabolismo , Hipocampo/metabolismo , Memória de Curto Prazo/fisiologia , Adulto , Circulação Cerebrovascular , Método Duplo-Cego , Estradiol/administração & dosagem , Estradiol/sangue , Feminino , Hipocampo/diagnóstico por imagem , Humanos , Leuprolida/farmacologia , Imageamento por Ressonância Magnética , Metionina/genética , Pessoa de Meia-Idade , Imagem Multimodal/métodos , Neuroimagem/métodos , Testes Neuropsicológicos , Ovário/metabolismo , Polimorfismo de Nucleotídeo Único , Tomografia por Emissão de Pósitrons , Progesterona/administração & dosagem , Progesterona/sangue , Distribuição Aleatória , Supositórios , Valina/genética
2.
Transl Psychiatry ; 5: e622, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-26285132

RESUMO

Characterizing the molecular mechanisms underlying the heritability of complex behavioral traits such as human anxiety remains a challenging endeavor for behavioral neuroscience. Copy-number variation (CNV) in the general transcription factor gene, GTF2I, located in the 7q11.23 chromosomal region that is hemideleted in Williams syndrome and duplicated in the 7q11.23 duplication syndrome (Dup7), is associated with gene-dose-dependent anxiety in mouse models and in both Williams syndrome and Dup7. Because of this recent preclinical and clinical identification of a genetic influence on anxiety, we examined whether sequence variation in GTF2I, specifically the single-nucleotide polymorphism rs2527367, interacts with trait and state anxiety to collectively impact neural response to anxiety-laden social stimuli. Two hundred and sixty healthy adults completed the Tridimensional Personality Questionnaire Harm Avoidance (HA) subscale, a trait measure of anxiety proneness, and underwent functional magnetic resonance imaging (fMRI) while matching aversive (fearful or angry) facial identity. We found an interaction between GTF2I allelic variations and HA that affects brain response: in individuals homozygous for the major allele, there was no correlation between HA and whole-brain response to aversive cues, whereas in heterozygotes and individuals homozygous for the minor allele, there was a positive correlation between HA sub-scores and a selective dorsolateral prefrontal cortex (DLPFC) responsivity during the processing of aversive stimuli. These results demonstrate that sequence variation in the GTF2I gene influences the relationship between trait anxiety and brain response to aversive social cues in healthy individuals, supporting a role for this neurogenetic mechanism in anxiety.


Assuntos
Ansiedade/genética , Variações do Número de Cópias de DNA/genética , Córtex Pré-Frontal/fisiopatologia , Fatores de Transcrição TFII/genética , Síndrome de Williams/genética , Adolescente , Adulto , Ira/fisiologia , Ansiedade/complicações , Ansiedade/fisiopatologia , Medo/fisiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Síndrome de Williams/complicações , Síndrome de Williams/fisiopatologia , Adulto Jovem
3.
Mol Psychiatry ; 19(2): 192-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23295814

RESUMO

Dopamine- and cAMP-regulated phosphoprotein of molecular weight 32 kDa (DARPP-32 or PPP1R1B) has been of interest in schizophrenia owing to its critical function in integrating dopaminergic and glutaminergic signaling. In a previous study, we identified single-nucleotide polymorphisms (SNPs) and a frequent haplotype associated with cognitive and imaging phenotypes that have been linked with schizophrenia, as well as with expression of prefrontal cortical DARPP-32 messenger RNA (mRNA) in a relatively small sample of postmortem brains. In this study, we examined the association of expression of two major DARPP-32 transcripts, full-length (FL-DARPP-32) and truncated (t-DARPP-32), with genetic variants of DARPP-32 in three brain regions receiving dopaminergic input and implicated in schizophrenia (the dorsolateral prefrontal cortex (DLPFC), hippocampus and caudate) in a much larger set of postmortem samples from patients with schizophrenia, bipolar disorder, major depression and normal controls (>700 subjects). We found that the expression of t-DARPP-32 was increased in the DLPFC of patients with schizophrenia and bipolar disorder, and was strongly associated with genotypes at SNPs (rs879606, rs90974 and rs3764352), as well as the previously identified 7-SNP haplotype related to cognitive functioning. The genetic variants that predicted worse cognitive performance were associated with higher t-DARPP-32 expression. Our results suggest that variation in PPP1R1B affects the abundance of the splice variant t-DARPP-32 mRNA and may reflect potential molecular mechanisms implicated in schizophrenia and affective disorders.


Assuntos
Transtorno Bipolar/metabolismo , Encéfalo/metabolismo , Fosfoproteína 32 Regulada por cAMP e Dopamina/genética , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Esquizofrenia/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antipsicóticos/farmacologia , Transtorno Bipolar/genética , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Criança , Pré-Escolar , Transtorno Depressivo Maior/genética , Transtorno Depressivo Maior/metabolismo , Feminino , Feto , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Ratos , Ratos Sprague-Dawley , Esquizofrenia/tratamento farmacológico , Esquizofrenia/genética
5.
Mol Psychiatry ; 18(6): 713-20, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23319002

RESUMO

A Val(66)Met single-nucleotide polymorphism (SNP) in the brain-derived neurotrophic factor (BDNF) gene impairs activity-dependent BDNF release in cultured hippocampal neurons and predicts impaired memory and exaggerated basal hippocampal activity in healthy humans. Several clinical genetic association studies along with multi-modal evidence for hippocampal dysfunction in schizophrenia indirectly suggest a relationship between schizophrenia and genetically determined BDNF function in the hippocampus. To directly test this hypothesized relationship, we studied 47 medication-free patients with schizophrenia or schizoaffective disorder and 74 healthy comparison individuals with genotyping for the Val(66)Met SNP and [(15)O]H(2)O positron emission tomography (PET) to measure resting and working memory-related hippocampal regional cerebral blood flow (rCBF). In patients, harboring a Met allele was associated with significantly less hippocampal rCBF. This finding was opposite to the genotype effect seen in healthy participants, resulting in a significant diagnosis-by-genotype interaction. Exploratory analyses of interregional resting rCBF covariation revealed a specific and significant diagnosis-by-genotype interaction effect on hippocampal-prefrontal coupling. A diagnosis-by-genotype interaction was also found for working memory-related hippocampal rCBF change, which was uniquely attenuated in Met allele-carrying patients. Thus, both task-independent and task-dependent hippocampal neurophysiology accommodates a Met allelic background differently in patients with schizophrenia than in control subjects. Potentially consistent with the hypothesis that cellular sequelae of the BDNF Val(66)Met SNP interface with aspects of schizophrenic hippocampal and frontotemporal dysfunction, these results warrant future investigation to understand the contributions of unique patient trait or state variables to these robust interactions.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Hipocampo/fisiopatologia , Polimorfismo de Nucleotídeo Único/genética , Esquizofrenia/genética , Esquizofrenia/patologia , Adulto , Técnicas de Apoio para a Decisão , Óxido de Deutério , Feminino , Genótipo , Hipocampo/irrigação sanguínea , Hipocampo/diagnóstico por imagem , Humanos , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Masculino , Memória de Curto Prazo/fisiologia , Metionina/genética , Testes Neuropsicológicos , Oxigênio/sangue , Tomografia por Emissão de Pósitrons , Descanso/fisiologia , Valina/genética , Adulto Jovem
6.
Mol Psychiatry ; 17(10): 1007-16, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21788944

RESUMO

AKT1 controls important processes in medial temporal lobe (MTL) development and plasticity, but the impact of human genetic variation in AKT1 on these processes is not known in healthy or disease states. Here, we report that an AKT1 variant (rs1130233) previously associated with AKT1 protein expression, prefrontal function and schizophrenia, affects human MTL structure and memory function. Further, supporting AKT1's role in transducing hippocampal neuroplasticity and dopaminergic processes, we found epistasis with functional polymorphisms in BDNF and COMT--genes also implicated in MTL biology related to AKT1. Consistent with prior predictions that these biologic processes relate to schizophrenia, we found epistasis between the same AKT1, BDNF and COMT functional variants on schizophrenia risk, and pharmacogenetic interactions of AKT1 with the effects on cognition and brain volume measures by AKT1 activators in common clinical use--lithium and sodium valproate. Our findings suggest that AKT1 affects risk for schizophrenia and accompanying cognitive deficits, at least in part through specific genetic interactions related to brain neuroplasticity and development, and that these AKT1 effects may be pharmacologically modulated in patients.


Assuntos
Polimorfismo de Nucleotídeo Único/genética , Proteínas Proto-Oncogênicas c-akt/genética , Esquizofrenia/genética , Esquizofrenia/patologia , Lobo Temporal/patologia , Antipsicóticos/uso terapêutico , Mapeamento Encefálico , Fator Neurotrófico Derivado do Encéfalo/genética , Catecol O-Metiltransferase/genética , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Farmacoeconomia , Epistasia Genética/efeitos dos fármacos , Epistasia Genética/genética , Feminino , Estudos de Associação Genética , Humanos , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Masculino , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/etiologia , Transtornos da Memória/genética , Testes Neuropsicológicos , Oxigênio/sangue , Estimulação Luminosa , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/genética , Esquizofrenia/complicações , Esquizofrenia/tratamento farmacológico , Lobo Temporal/irrigação sanguínea , Lobo Temporal/efeitos dos fármacos
7.
Mol Psychiatry ; 14(10): 968-75, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18490926

RESUMO

In mammals, the neuropeptide vasopressin is a key molecule for complex emotional and social behaviours. Two microsatellite polymorphisms, RS1 and RS3, near the promoter of AVPR1A, encoding the receptor subtype most heavily implicated in behaviour regulation, have been linked to autism and behavioural traits. However, the impact of these variants on human brain function is unknown. Here we show that human amygdala function is strongly associated with genetic variation in AVPR1A. Using an imaging genetics approach in a sample of 121 volunteers studied with an emotional face-matching paradigm, we found that differential activation of amygdala is observed in carriers of risk alleles for RS3 and RS1. Alleles in RS1 previously reported to be significantly over- and undertransmitted to autistic probands showed opposing effects on amygdala activation. Furthermore, we show functional difference in human brain between short and long repeat lengths that mirror findings recently obtained in a corresponding variant in voles. Our results indicate a neural mechanism mediating genetic risk for autism through an impact on amygdala signalling and provide a rationale for exploring therapeutic strategies aimed at abnormal amygdala function in this disorder.


Assuntos
Tonsila do Cerebelo/fisiologia , Arginina Vasopressina/genética , Transtorno Autístico/genética , Variação Genética , Personalidade/genética , Receptores de Vasopressinas/genética , Adulto , Expressão Facial , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Repetições de Microssatélites
8.
Mol Psychiatry ; 13(7): 709-16, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18347599

RESUMO

Complex genetic disorders such as depression likely exhibit epistasis, but neural mechanisms of such gene-gene interactions are incompletely understood. 5-HTTLPR and BDNF VAL66MET, functional polymorphisms of the serotonin (5-HT) transporter (SLC6A4) and brain-derived neurotrophic factor (BDNF) gene, impact on two distinct, but interacting signaling systems, which have been related to depression and to the modulation of neurogenesis and plasticity of circuitries of emotion processing. Recent clinical studies suggest that the BDNF MET allele, which shows abnormal intracellular trafficking and regulated secretion, has a protective effect regarding the development of depression and in mice of social defeat stress. Here we show, using anatomical neuroimaging techniques in a sample of healthy subjects (n=111), that the BDNF MET allele, which is predicted to have reduced responsivity to 5-HT signaling, protects against 5-HTTLPR S allele-induced effects on a brain circuitry encompassing the amygdala and the subgenual portion of the anterior cingulate (rAC). Our analyses revealed no effect of the 5-HTTLPR S allele on rAC volume in the presence of BDNF MET alleles, whereas a significant volume reduction (P<0.001) was seen on BDNF VAL/VAL background. Interacting genotype effects were also found in structural connectivity between amygdala and rAC (P=0.002). These data provide in vivo evidence of biologic epistasis between SLC6A4 and BDNF in the human brain by identifying a neural mechanism linking serotonergic and neurotrophic signaling on the neural systems level, and have implications for personalized treatment planning in depression.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Depressão/genética , Transtorno Depressivo/genética , Epistasia Genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Substituição de Aminoácidos , Encéfalo/patologia , Depressão/patologia , Transtorno Depressivo/patologia , Giro do Cíngulo/patologia , Humanos , Imageamento por Ressonância Magnética , Polimorfismo Genético , Valores de Referência , População Branca/genética
9.
Mol Psychiatry ; 13(8): 821-7, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18317466

RESUMO

A common polymorphism (val158met) in the gene encoding catechol-O-methyltransferase (COMT) has been shown to affect dopamine (DA) tone in cortex and cortical functioning. D1 receptors are the main DA receptors in the cortex, and studies have shown that decreased levels of cortical DA are associated with upregulation of D1 receptor availability, as measured with the positron-emission tomography (PET) radiotracer [11C]NNC112. We compared [11C]NNC 112 binding in healthy volunteers homozygous for the Val allele compared with Met carriers. Subjects were otherwise matched for parameters known to affect [11C]NNC 112 binding. Subjects with Val/Val alleles had significantly higher cortical [11C]NNC 112 binding compared with Met carriers, but did not differ in striatal binding. These results confirm the prominent role of COMT in regulating DA transmission in cortex but not striatum, and the reliability of [11C]NNC 112 as a marker for low DA tone as previously suggested by studies in patients with schizophrenia.


Assuntos
Catecol O-Metiltransferase/genética , Córtex Cerebral/fisiologia , Corpo Estriado/fisiologia , Sistema Límbico/fisiologia , Polimorfismo de Nucleotídeo Único , Receptores de Dopamina D1/fisiologia , Adulto , Substituição de Aminoácidos , Benzazepinas , Benzofuranos , Mapeamento Encefálico/métodos , Feminino , Genótipo , Humanos , Masculino , Anamnese , Tomografia por Emissão de Pósitrons
10.
Mol Psychiatry ; 13(3): 313-24, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17519928

RESUMO

Little is known about neural mechanisms underlying human personality and temperament, despite their considerable importance as highly heritable risk mediators for somatic and psychiatric disorders. To identify these circuits, we used a combined genetic and imaging approach focused on Monoamine Oxidase A (MAOA), encoding a key enzyme for monoamine metabolism previously associated with temperament and antisocial behavior. Male carriers of a low-expressing genetic variant exhibited dysregulated amygdala activation and increased functional coupling with ventromedial prefrontal cortex (vmPFC). Stronger coupling predicted increased harm avoidance and decreased reward dependence scores, suggesting that this circuitry mediates a part of the association of MAOA with these traits. We utilized path analysis to parse the effective connectivity within this system, and provide evidence that vmPFC regulates amygdala indirectly by influencing rostral cingulate cortex function. Our data implicate a neural circuit for variation in human personality under genetic control, provide an anatomically consistent mechanism for vmPFC-amygdala interactions underlying this variation, and suggest a role for vmPFC as a superordinate regulatory area for emotional arousal and social behavior.


Assuntos
Variação Genética , Individualidade , Monoaminoxidase/genética , Personalidade/genética , Córtex Pré-Frontal/fisiologia , Adulto , Mapeamento Encefálico , Expressão Facial , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Masculino , Modelos Biológicos , Vias Neurais/irrigação sanguínea , Vias Neurais/fisiologia , Testes Neuropsicológicos , Oxigênio/sangue , Estimulação Luminosa/métodos , Córtex Pré-Frontal/irrigação sanguínea
11.
Mol Psychiatry ; 12(9): 854-69, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17767149

RESUMO

Cortical GABAergic dysfunction has been implicated as a key component of the pathophysiology of schizophrenia and decreased expression of the gamma-aminobutyric acid (GABA) synthetic enzyme glutamic acid decarboxylase 67 (GAD(67)), encoded by GAD1, is found in schizophrenic post-mortem brain. We report evidence of distorted transmission of single-nucleotide polymorphism (SNP) alleles in two independent schizophrenia family-based samples. In both samples, allelic association was dependent on the gender of the affected offspring, and in the Clinical Brain Disorders Branch/National Institute of Mental Health (CBDB/NIMH) sample it was also dependent on catechol-O-methyltransferase (COMT) Val158Met genotype. Quantitative transmission disequilibrium test analyses revealed that variation in GAD1 influenced multiple domains of cognition, including declarative memory, attention and working memory. A 5' flanking SNP affecting cognition in the families was also associated in unrelated healthy individuals with inefficient BOLD functional magnetic resonance imaging activation of dorsal prefrontal cortex (PFC) during a working memory task, a physiologic phenotype associated with schizophrenia and altered cortical inhibition. In addition, a SNP in the 5' untranslated (and predicted promoter) region that also influenced cognition was associated with decreased expression of GAD1 mRNA in the PFC of schizophrenic brain. Finally, we observed evidence of statistical epistasis between two SNPs in COMT and SNPs in GAD1, suggesting a potential biological synergism leading to increased risk. These coincident results implicate GAD1 in the etiology of schizophrenia and suggest that the mechanism involves altered cortical GABA inhibitory activity, perhaps modulated by dopaminergic function.


Assuntos
Córtex Cerebral/fisiopatologia , Expressão Gênica/fisiologia , Predisposição Genética para Doença , Glutamato Descarboxilase/genética , Polimorfismo de Nucleotídeo Único , Esquizofrenia , Adolescente , Adulto , Alelos , Catecol O-Metiltransferase/genética , Córtex Cerebral/irrigação sanguínea , Saúde da Família , Feminino , Frequência do Gene , Humanos , Processamento de Imagem Assistida por Computador/métodos , Desequilíbrio de Ligação , Imageamento por Ressonância Magnética/métodos , Masculino , Pessoa de Meia-Idade , Testes Neuropsicológicos , Oxigênio/sangue , Esquizofrenia/genética , Esquizofrenia/patologia , Esquizofrenia/fisiopatologia , Fatores Sexuais
12.
Mol Psychiatry ; 12(10): 946-57, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17440436

RESUMO

Neuregulin1 (NRG1), a candidate susceptibility gene for schizophrenia, plays a critical role in neuronal migration and central nervous system development. However, its relation to schizophrenia pathogenesis is unknown. Here we show that B lymphoblasts migrate to NRG1 through the ErbB-signaling system as observed in neuronal cells. We assessed NRG1-induced cell migration in B lymphoblasts from patients with schizophrenia and found that NRG1-induced migration is significantly decreased compared with control individuals in two independent cohorts. This impaired migration is related at least in part to reduced AKT phosphorylation in the patients. Moreover, the magnitude of NRG1-induced migration is associated with polymorphisms of the NRG1 and catechol-o-methyltransferase genes and with an epistatic interaction of these genes. This study demonstrates that the migratory response of schizophrenia-derived cells to NRG1 is impaired and is associated with genetic variations in more than one schizophrenia susceptibility gene, providing a novel insight into potential neurodevelopmental mechanisms of schizophrenia.


Assuntos
Linfócitos B/efeitos dos fármacos , Catecol O-Metiltransferase/genética , Movimento Celular/efeitos dos fármacos , Neuregulina-1/genética , Neuregulina-1/farmacologia , Polimorfismo Genético/efeitos dos fármacos , Esquizofrenia/patologia , Adulto , Análise de Variância , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Esquizofrenia/genética , Transdução de Sinais/efeitos dos fármacos , Estatísticas não Paramétricas
13.
Cereb Cortex ; 17(5): 1206-12, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-16835293

RESUMO

The prefrontal cortex (PFC) dopamine system, which is critical for modulating PFC function, undergoes remodeling until at least young adulthood in primates. Catechol-o-methyltransferase (COMT) alters extracellular dopamine levels in PFC, and its gene contains a functional polymorphism (Val(158)Met) that has been associated with variation in PFC function. We examined COMT enzyme activity and protein immunoreactivity in the PFC during human postnatal development. Protein was extracted from PFC of normal individuals from 6 age groups: neonates (1-4 months), infants (5-11 months), teens (14-18 years), young adults (20-24 years), adults (31-43 years), and aged individuals (68-86 years; n = 5-8 per group). There was a significant 2-fold increase in COMT enzyme activity from neonate to adulthood, paralleled by increases in COMT protein immunoreactivity. Furthermore, COMT protein immunoreactivity was related to Val(158)Met genotype, as has been previously demonstrated. The significant increase in COMT activity from neonate to adulthood complements previous findings of protracted postnatal changes in the PFC dopamine system and may reflect an increasing importance of COMT for PFC dopamine regulation during maturation.


Assuntos
Envelhecimento/metabolismo , Catecol O-Metiltransferase/genética , Catecol O-Metiltransferase/metabolismo , Dopamina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Córtex Pré-Frontal/metabolismo , Adolescente , Adulto , Idoso , Ativação Enzimática , Feminino , Humanos , Lactente , Recém-Nascido , Masculino
14.
Mol Psychiatry ; 11(9): 867-77, 797, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16786032

RESUMO

Catechol-O-methyltransferase (COMT) has been shown to be critical for prefrontal dopamine flux, prefrontal cortex-dependent cognition and activation. Several potentially functional variants in the gene have been identified, but considerable controversy exists regarding the contribution of individual alleles and haplotypes to risk for schizophrenia, partly because clinical phenotypes are ill-defined and preclinical studies are limited by lack of adequate models. Here, we propose a neuroimaging approach to overcome these limitations by characterizing the functional impact of ambiguous haplotypes on a neural system-level intermediate phenotype in humans. Studying 126 healthy control subjects during a working-memory paradigm, we find that a previously described risk variant in a functional Val158Met (rs4680) polymorphism interacts with a P2 promoter region SNP (rs2097603) and an SNP in the 3' region (rs165599) in predicting inefficient prefrontal working memory response. We report evidence that the nonlinear response of prefrontal neurons to dopaminergic stimulation is a neural mechanism underlying these nonadditive genetic effects. This work provides an in vivo approach to functional validation in brain of the biological impact of complex genetic variations within a gene that may be critical for its clinical association.


Assuntos
Encéfalo/fisiologia , Catecol O-Metiltransferase/genética , Variação Genética , Memória/fisiologia , Polimorfismo de Nucleotídeo Único , Adulto , Substituição de Aminoácidos , Sequência de Bases , Encéfalo/enzimologia , Dopamina/metabolismo , Feminino , Humanos , Íntrons , Masculino , Polimorfismo Genético , Córtex Pré-Frontal/fisiologia , Tempo de Reação/genética
15.
Pharmacogenomics J ; 3(1): 27-40, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12629581

RESUMO

CNS-focused cDNA microarrays were used to examine gene expression profiles in dorsolateral prefrontal cortex (dlPFC, Area 46) from seven individual sets of age- and post-mortem interval-matched male cocaine abusers and controls. The presence of cocaine and related metabolites was confirmed by gas chromatography-mass spectrometry. Sixty-five transcripts were differentially expressed, indicating alterations in energy metabolism, mitochondria and oligodendrocyte function, cytoskeleton and related signaling, and neuronal plasticity. There was evidence for two distinct states of transcriptional regulation, with increases in gene expression predominating in subjects testing positive for a metabolite indicative of recent 'crack' cocaine abuse and decreased expression profiles in the remaining cocaine subjects. This pattern was confirmed by quantitative polymerase chain reaction for select transcripts. These data suggest that cocaine abuse targets a distinct subset of genes in the dlPFC, resulting in either a state of acute activation in which increased gene expression predominates, or a relatively destimulated, refractory phase.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/genética , Perfilação da Expressão Gênica/métodos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Córtex Pré-Frontal/fisiologia , Ativação Transcricional/efeitos dos fármacos , Adulto , Análise por Conglomerados , Cocaína Crack/efeitos adversos , Perfilação da Expressão Gênica/estatística & dados numéricos , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos/estatística & dados numéricos , Reação em Cadeia da Polimerase/métodos , Transcrição Gênica/efeitos dos fármacos
16.
Proc Natl Acad Sci U S A ; 98(12): 6917-22, 2001 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-11381111

RESUMO

Abnormalities of prefrontal cortical function are prominent features of schizophrenia and have been associated with genetic risk, suggesting that susceptibility genes for schizophrenia may impact on the molecular mechanisms of prefrontal function. A potential susceptibility mechanism involves regulation of prefrontal dopamine, which modulates the response of prefrontal neurons during working memory. We examined the relationship of a common functional polymorphism (Val(108/158) Met) in the catechol-O-methyltransferase (COMT) gene, which accounts for a 4-fold variation in enzyme activity and dopamine catabolism, with both prefrontally mediated cognition and prefrontal cortical physiology. In 175 patients with schizophrenia, 219 unaffected siblings, and 55 controls, COMT genotype was related in allele dosage fashion to performance on the Wisconsin Card Sorting Test of executive cognition and explained 4% of variance (P = 0.001) in frequency of perseverative errors. Consistent with other evidence that dopamine enhances prefrontal neuronal function, the load of the low-activity Met allele predicted enhanced cognitive performance. We then examined the effect of COMT genotype on prefrontal physiology during a working memory task in three separate subgroups (n = 11-16) assayed with functional MRI. Met allele load consistently predicted a more efficient physiological response in prefrontal cortex. Finally, in a family-based association analysis of 104 trios, we found a significant increase in transmission of the Val allele to the schizophrenic offspring. These data suggest that the COMT Val allele, because it increases prefrontal dopamine catabolism, impairs prefrontal cognition and physiology, and by this mechanism slightly increases risk for schizophrenia.


Assuntos
Catecol O-Metiltransferase/genética , Lobo Frontal/fisiologia , Esquizofrenia/etiologia , Adulto , Alelos , Feminino , Genótipo , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Fatores de Risco
17.
Curr Protoc Neurosci ; Chapter 7: Unit7.3, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-18428531

RESUMO

This unit presents a technique that allows for routine repeated microdialysis experiments in the monkey using a sedated preparation or, with further minor modification, in the awake behaving animal. Protocols are provided for construction and in vitro calibration of dialysis probes, obtaining magnetic resonance imaging (MRI) scans of the monkey brain, surgically attaching a guide holder to the skull, and performing postoperative MRI scanning, in vivo dialysis sample collection, anatomical verification of the probes, and neurochemical analysis. An alternate protocol describes microdialysis in awake, behaving monkeys; this requires substantial preparatory work in training the monkeys to sit quietly in a restraining chair and/or to perform a series of behavioral tasks.


Assuntos
Macaca mulatta/metabolismo , Microdiálise/métodos , Animais , Comportamento Animal , Química Encefálica , Desenho de Equipamento , Feminino , Implantes Experimentais , Imageamento por Ressonância Magnética , Masculino , Microdiálise/instrumentação , Técnicas Estereotáxicas , Vigília
18.
J Neural Transm (Vienna) ; 106(2): 183-96, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10226938

RESUMO

In vivo microdialysis was used to study the effects of restraint stress (30 min) and amphetamine (AMPH) (5 mg/kg, i.p.) in awake adult male rats with neonatal ventral hippocampal (VH) damage. Extracellular levels of dopamine (DA), dihydrophenylacetate (DOPAC), homovanillate (HVA) and 5-hydroxyindolacetate (5-HIAA) were measured in the nucleus accumbens (NA). There were no differences in the baseline levels of DA, DOPAC, HVA or 5-HIAA in the lesioned as compared to the sham rats. Release from restraint resulted in increased extracellular levels of DA in the sham but not in the lesioned animals. AMPH increased DA release in both sham operated and lesioned animals, but this increase was significantly attenuated in the lesioned rats. Our data suggest that this developmental lesion alters function of the dopaminergic system in response to environmental and pharmacological challenge.


Assuntos
Anfetamina/farmacologia , Dopamina/metabolismo , Hipocampo/fisiologia , Núcleo Accumbens/metabolismo , Estresse Fisiológico/metabolismo , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Animais , Animais Recém-Nascidos , Hipocampo/efeitos dos fármacos , Ácido Homovanílico/metabolismo , Ácido Hidroxi-Indolacético/metabolismo , Ácido Ibotênico/toxicidade , Masculino , Núcleo Accumbens/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Restrição Física , Fatores de Tempo
19.
Synapse ; 32(2): 71-9, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10231127

RESUMO

Developmental cortical damage has been implicated in the basic neurobiology of schizophrenia. Adult rhesus monkeys with neonatal temporal limbic damage show a stimulus-dependent disinhibition of subcortical dopamine (DA) release. We measured dopamine D2 receptors and transporters in vivo in rhesus monkeys with neonatal and adult mesial temporal limbic lesions and control monkeys to explore further the effects of this developmental lesion on striatal DA function. All monkeys were studied with [I-123]IBZM SPECT to assess the availability of striatal dopamine D2 receptors and with [I-123]beta-CIT SPECT to measure the availability of dopamine transporters in the striatum. IBZM binding was significantly reduced in monkeys with neonatal limbic lesions. No group difference in beta-CIT binding was found. The reduction in IBZM binding was significantly correlated with subcortical dopamine release after monoaminergic prefrontal stimulation as determined with in vivo microdialysis. Our findings imply specific interactions between age at lesion and the availability of DA transporter and receptors in non-human primates, and suggest that stimulus-dependent DA activity affects the expression of DA receptors.


Assuntos
Proteínas de Transporte/análise , Corpo Estriado/química , Sistema Límbico/lesões , Glicoproteínas de Membrana , Proteínas de Membrana Transportadoras , Proteínas do Tecido Nervoso , Receptores de Dopamina D1/análise , Receptores de Dopamina D2/análise , Lobo Temporal/lesões , Fatores Etários , Animais , Animais Recém-Nascidos , Benzamidas/metabolismo , Benzamidas/farmacologia , Ligação Competitiva/fisiologia , Proteínas de Transporte/metabolismo , Cocaína/análogos & derivados , Cocaína/metabolismo , Cocaína/farmacologia , Meios de Contraste , Corpo Estriado/diagnóstico por imagem , Dopamina/análise , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina , Radioisótopos do Iodo , Macaca mulatta , Masculino , Microdiálise , Córtex Pré-Frontal/metabolismo , Pirrolidinas/metabolismo , Pirrolidinas/farmacologia , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Tomografia Computadorizada de Emissão de Fóton Único
20.
Biol Psychiatry ; 45(6): 660-7, 1999 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10187995

RESUMO

BACKGROUND: Pathology of dorsolateral prefrontal cortex and dysregulation of dopaminergic neurons have been associated with the pathophysiology of schizophrenia, but how these phenomena relate to each other in patients has not been known. It has been hypothesized that prefrontal cortical pathology might induce both diminished steady-state and exaggerated responses of dopaminergic neurons to certain stimuli (e.g., stress). We examined the relationship between a measure of prefrontal neuronal pathology and striatal dopamine activity in patients with schizophrenia and in a nonhuman primate model of abnormal prefrontal cortical development. METHODS: In the patients, we studied in vivo markers of cortical neuronal pathology with NMR spectroscopic imaging and of steady-state striatal dopamine activity with radioreceptor imaging. In the monkeys, we used the same NMR technique and in vivo microdialysis. RESULTS: Measures of N-acetyl-aspartate concentrations (NAA) in dorsolateral prefrontal cortex strongly and selectively predicted D2 receptor availability in the striatum (n = 14, rho = -.64, p < .01), suggesting that the greater the apparent dorsolateral prefrontal cortex pathology, the less the steady-state dopamine activity in these patients. A similar relationship between NAA measures in dorsolateral prefrontal cortex and steady-state dopamine concentrations in the striatum was found in the monkeys (n = 5, rho = .70, p < .05). We then tested in the same monkeys the relationship of prefrontal NAA and striatal dopamine overflow following amphetamine infusion into dorsolateral prefrontal cortex. Under these conditions, the relationship was inverted, i.e., the greater the apparent dorsolateral prefrontal cortex pathology, the greater the dopamine release. CONCLUSIONS: These data demonstrate direct relationships between putative neuronal pathology in dorsolateral prefrontal cortex and striatal dopamine activity in human and nonhuman primates and implicate a mechanism for dopamine dysregulation in schizophrenia.


Assuntos
Ácido Aspártico/metabolismo , Corpo Estriado/metabolismo , Dopamina/metabolismo , Córtex Pré-Frontal/metabolismo , Esquizofrenia/metabolismo , Adulto , Animais , Colina/metabolismo , Doença Crônica , Corpo Estriado/diagnóstico por imagem , Creatinina/metabolismo , Feminino , Humanos , Macaca mulatta , Espectroscopia de Ressonância Magnética/métodos , Masculino , Neurônios/fisiologia , Escalas de Graduação Psiquiátrica , Receptores de Dopamina D2/metabolismo , Esquizofrenia/diagnóstico , Tomografia Computadorizada de Emissão de Fóton Único
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA