Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 450
Filtrar
1.
Mol Psychiatry ; 23(5): 1251-1260, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-28485405

RESUMO

The role of the immune system in schizophrenia remains controversial despite numerous studies to date. Most studies have profiled expression of select genes or proteins in peripheral blood, but none have focused on the expression of canonical pathways that mediate overall immune response. The current study used a systematic genetic approach to investigate the role of the immune system in a large sample of post-mortem brain of patients with schizophrenia: RNA sequencing was performed to assess the differential expression of 561 immune genes and 20 immune pathways in dorsolateral prefrontal cortex (DLPFC) (144 schizophrenia and 196 control subjects) and hippocampus (83 schizophrenia and 187 control subjects). The effect of RNA quality on gene expression was found to be highly correlated with the effect of diagnosis even after adjustment for observable RNA quality parameters (i.e. RNA integrity), thus this confounding relationship was statistically controlled using principal components derived from the gene expression matrix. In DLPFC, 23 immune genes were found to be differentially expressed (false discovery rate <0.05), of which seven genes replicated in both directionality and at nominal significance (P<0.05) in an independent post-mortem DLPFC data set (182 schizophrenia and 212 control subjects), although notably at least five of these genes have prominent roles in pathways other than immune function and overall the effect sizes were minimal (fold change <1.1). In the hippocampus, no individual immune genes were identified to be differentially expressed, and in both DLPFC and hippocampus none of the individual immune pathways were relatively differentially expressed. Further, genomic schizophrenia risk profiles scores were not correlated with the expression of individual immune pathways or differentially expressed genes. Overall, past reports claiming a primary pathogenic role of the immune system intrinsic to the brain in schizophrenia could not be confirmed.


Assuntos
Esquizofrenia/imunologia , Esquizofrenia/patologia , Adulto , Encéfalo/patologia , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Neuroimunomodulação , Análise de Sequência com Séries de Oligonucleotídeos , Esquizofrenia/genética , Análise de Sequência de RNA
2.
Mol Psychiatry ; 23(6): 1496-1505, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-28485403

RESUMO

Genetic variations and adverse environmental events in utero or shortly after birth can lead to abnormal brain development and increased risk of schizophrenia. γ-Aminobutyric acid (GABA), the major inhibitory neurotransmitter in the mammalian brain, plays a vital role in normal brain development. GABA synthesis is controlled by enzymes derived from two glutamic acid decarboxylase (GAD) genes, GAD1 and GAD2, both of which produce transcript isoforms. While the full-length GAD1 transcript (GAD67) has been implicated in the neuropathology of schizophrenia, the transcript structure of GAD1 in the human brain has not been fully characterized. In this study, with the use of RNA sequencing and PCR technologies, we report the discovery of 10 novel transcripts of GAD1 in the human brain. Expression levels of four novel GAD1 transcripts (8A, 8B, I80 and I86) showed a lifespan trajectory expression pattern that is anticorrelated with the expression of the full-length GAD1 transcript. In addition, methylation levels of two CpG loci within the putative GAD1 promoter were significantly associated with the schizophrenia-risk SNP rs3749034 and with the expression of GAD25 in dorsolateral prefrontal cortex (DLPFC). Moreover, schizophrenia patients who had completed suicide and/or were positive for nicotine exposure had significantly higher full-length GAD1 expression in the DLPFC. Alternative splicing of GAD1 and epigenetic state appear to play roles in the developmental profile of GAD1 expression and may contribute to GABA dysfunction in the PFC and hippocampus of patients with schizophrenia.


Assuntos
Glutamato Descarboxilase/genética , Esquizofrenia/genética , Adolescente , Adulto , Processamento Alternativo/genética , Autopsia , Encéfalo/metabolismo , Criança , Pré-Escolar , Metilação de DNA/genética , Feminino , Expressão Gênica/genética , Variação Genética/genética , Glutamato Descarboxilase/metabolismo , Hipocampo/metabolismo , Humanos , Recém-Nascido , Masculino , Córtex Pré-Frontal/metabolismo , Regiões Promotoras Genéticas/genética , Isoformas de RNA/genética , RNA Mensageiro/metabolismo , Esquizofrenia/metabolismo
3.
Mol Psychiatry ; 23(5): 1145-1156, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-28630453

RESUMO

In order to determine the impact of the epigenetic response to traumatic stress on post-traumatic stress disorder (PTSD), this study examined longitudinal changes of genome-wide blood DNA methylation profiles in relation to the development of PTSD symptoms in two prospective military cohorts (one discovery and one replication data set). In the first cohort consisting of male Dutch military servicemen (n=93), the emergence of PTSD symptoms over a deployment period to a combat zone was significantly associated with alterations in DNA methylation levels at 17 genomic positions and 12 genomic regions. Evidence for mediation of the relation between combat trauma and PTSD symptoms by longitudinal changes in DNA methylation was observed at several positions and regions. Bioinformatic analyses of the reported associations identified significant enrichment in several pathways relevant for symptoms of PTSD. Targeted analyses of the significant findings from the discovery sample in an independent prospective cohort of male US marines (n=98) replicated the observed relation between decreases in DNA methylation levels and PTSD symptoms at genomic regions in ZFP57, RNF39 and HIST1H2APS2. Together, our study pinpoints three novel genomic regions where longitudinal decreases in DNA methylation across the period of exposure to combat trauma marks susceptibility for PTSD.


Assuntos
Epigênese Genética , Transtornos de Estresse Pós-Traumáticos/genética , Adulto , Estudos de Coortes , Metilação de DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Predisposição Genética para Doença , Testes Genéticos/métodos , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Estudos Longitudinais , Masculino , Militares/psicologia , Estudos Prospectivos , Proteínas Repressoras , Transtornos de Estresse Pós-Traumáticos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
Mol Psychiatry ; 19(3): 311-6, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24145376

RESUMO

Cognitive functions are highly heritable and the impact of complex genetic interactions, though undoubtedly important, has received little investigation. Here we show in an animal model and in a human neuroimaging experiment a consistent non-linear interaction between two genes--catechol-O-methyl transferase (COMT) and dysbindin (dys; dystrobrevin-binding protein 1 (DTNBP1))--implicated through different mechanisms in cortical dopamine signaling and prefrontal cognitive function. In mice, we found that a single genetic mutation reducing expression of either COMT or DTNBP1 alone produced working memory advantages, while, in dramatic contrast, genetic reduction of both in the same mouse produced working memory deficits. We found evidence of the same non-linear genetic interaction in prefrontal cortical function in humans. In healthy volunteers (N=176) studied with functional magnetic resonance imaging during a working memory paradigm, individuals homozygous for the COMT rs4680 Met allele that reduces COMT enzyme activity showed a relatively more efficient prefrontal engagement. In contrast, we found that the same genotype was less efficient on the background of a dys haplotype associated with decreased DTNBP1 expression. These results illustrate that epistasis can be functionally multi-directional and non-linear and that a putatively beneficial allele in one epistastic context is a relatively deleterious one in another. These data also have important implications for single-locus association analyses of complex traits.


Assuntos
Proteínas de Transporte/fisiologia , Catecol O-Metiltransferase/genética , Catecol O-Metiltransferase/fisiologia , Epistasia Genética , Memória de Curto Prazo/fisiologia , Córtex Pré-Frontal/fisiologia , Alelos , Animais , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Catecol O-Metiltransferase/biossíntese , Disbindina , Proteínas Associadas à Distrofina , Neuroimagem Funcional , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Homozigoto , Humanos , Imageamento por Ressonância Magnética , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Knockout , Mutação
5.
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
6.
Mol Psychiatry ; 19(12): 1258-66, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24322206

RESUMO

Dopamine 2 receptor (DRD2) is of major interest to the pathophysiology of schizophrenia (SCZ) both as a target for antipsychotic drug action as well as a SCZ-associated risk gene. The dopamine 1 receptor (DRD1) is thought to mediate some of the cognitive deficits in SCZ, including impairment of working memory that relies on normal dorsolateral prefrontal cortex (DLPFC) function. To better understand the association of dopamine receptors with SCZ, we studied the expression of three DRD2 splice variants and the DRD1 transcript in DLPFC, hippocampus and caudate nucleus in a large cohort of subjects (~700), including patients with SCZ, affective disorders and nonpsychiatric controls (from 14th gestational week to 85 years of age), and examined genotype-expression associations of 278 single-nucleotide polymorphisms (SNPs) located in or near DRD2 and DRD1 genes. Expression of D2S mRNA and D2S/D2-long (D2L) ratio were significantly increased in DLPFC of patients with SCZ relative to controls (P<0.0001 and P<0.0001, respectively), whereas D2L, D2Longer and DRD1 were decreased (P<0.0001). Patients with affective disorders showed an opposite pattern: reduced expression of D2S (major depressive disorder, P<0.0001) and increased expression of D2L and DRD1 (bipolar disorder, P<0.0001). Moreover, SCZ-associated risk alleles at rs1079727, rs1076560 and rs2283265 predicted increased D2S/D2L expression ratio (P<0.05) in control individuals. Our data suggest that altered splicing of DRD2 and expression of DRD1 may constitute a pathophysiological mechanism in risk for SCZ and affective disorders. The association between SCZ risk-associated polymorphism and the ratio of D2S/D2L is consistent with this possibility.


Assuntos
Transtorno Bipolar/genética , Encéfalo/metabolismo , Transtorno Depressivo Maior/genética , Receptores de Dopamina D1/genética , Receptores de Dopamina D2/genética , Esquizofrenia/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Transtorno Bipolar/metabolismo , Encéfalo/crescimento & desenvolvimento , Criança , Pré-Escolar , Estudos de Coortes , Transtorno Depressivo Maior/metabolismo , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Splicing de RNA , RNA Mensageiro/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Esquizofrenia/metabolismo , Adulto Jovem
7.
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
8.
Mol Psychiatry ; 17(1): 85-98, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20956979

RESUMO

Dysbindin-1 regulates D2-receptor trafficking and is implicated in schizophrenia and related cognitive abnormalities, but whether this molecular effect mediates the clinical manifestations of the disorder is unknown. We explored in dysbindin-1-deficient mice (dys-/-) (1) schizophrenia-related behaviors, (2) molecular and electrophysiological changes in medial prefrontal cortex (mPFC) and (3) the dependence of these on D2-receptor stimulation. Dysbindin-1 disruption altered dopamine-related behaviors and impaired working memory under challenging/stressful conditions. Dys-/- pyramidal neurons in mPFC layers II/III were hyperexcitable at baseline but hypoexcitable following D2 stimulation. Dys-/- were also respectively more and less sensitive to D2 agonist- and antagonist-induced behavioral effects. Dys-/- had reduced expression of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and CaMKKß in mPFC. Chronic D2 agonist treatment reproduced these changes in protein expression, and some of the dys-/- behavioral effects. These results elucidate dysbindin's modulation of D2-related behavior, cortical activity and mPFC CaMK components, implicating cellular and molecular mechanisms of the association of dysbindin with psychosis.


Assuntos
Proteínas de Transporte/genética , Dopamina/metabolismo , Córtex Pré-Frontal/fisiopatologia , Receptores de Dopamina D2/metabolismo , Esquizofrenia/patologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Anfetamina/efeitos adversos , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Modelos Animais de Doenças , Dopaminérgicos/farmacologia , Disbindina , Proteínas Associadas à Distrofina , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Hipercinese/tratamento farmacológico , Hipercinese/etiologia , Hipercinese/genética , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/etiologia , Memória de Curto Prazo/efeitos dos fármacos , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Endogâmicos DBA , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/patologia , Células Piramidais/efeitos dos fármacos , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/genética , Esquizofrenia/complicações , Filtro Sensorial/efeitos dos fármacos , Filtro Sensorial/genética , Estresse Psicológico/fisiopatologia
9.
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
10.
Mol Psychiatry ; 16(6): 620-5, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21519338

RESUMO

The antipsychotic drug, olanzapine, one of the most widely used drugs in clinical medicine, has a high rate of discontinuation due to inefficacy and/or adverse effects. We identified a single nucleotide polymorphism in the drug metabolizing enzyme, cytochrome P450 3A43 (CYP3A43; rs472660), that highly significantly predicted olanzapine clearance in the Clinical Antipsychotic Trials of Intervention Effectiveness trial (P=5.9e(-7)). Moreover, at standard antipsychotic doses, 50% of individuals with the high clearance genotype (AA) have trough blood levels below the therapeutic range. Interestingly, a much higher proportion of African Americans carry the A allele compared with Caucasians (allele frequency 67 vs 14%). After accounting for CYP3A43 genotype, race is no longer a significant predictor of olanzapine clearance. Olanzapine clearance was associated with measures of clinical response. Patients with greater clearance had higher symptom ratings and were more likely to discontinue treatment due to an inadequate response. Our data identify a genetic mechanism for variation in olanzapine response and demonstrate that blood level monitoring of olanzapine treatment is advisable.


Assuntos
Antipsicóticos/farmacocinética , Benzodiazepinas/farmacocinética , Negro ou Afro-Americano/genética , Citocromo P-450 CYP3A/genética , Polimorfismo de Nucleotídeo Único/genética , População Branca/genética , Antipsicóticos/uso terapêutico , Benzodiazepinas/uso terapêutico , Feminino , Frequência do Gene , Genótipo , Glucose/metabolismo , Humanos , Modelos Lineares , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Modelos Químicos , Olanzapina , Escalas de Graduação Psiquiátrica , Esquizofrenia/tratamento farmacológico , Esquizofrenia/metabolismo , Fatores Sexuais , Fumar/genética , Aumento de Peso/efeitos dos fármacos
11.
Mol Psychiatry ; 16(11): 1117-29, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20838396

RESUMO

We conducted data-mining analyses using the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) and molecular genetics of schizophrenia genome-wide association study supported by the genetic association information network (MGS-GAIN) schizophrenia data sets and performed bioinformatic prioritization for all the markers with P-values ≤0.05 in both data sets. In this process, we found that in the CMYA5 gene, there were two non-synonymous markers, rs3828611 and rs10043986, showing nominal significance in both the CATIE and MGS-GAIN samples. In a combined analysis of both the CATIE and MGS-GAIN samples, rs4704591 was identified as the most significant marker in the gene. Linkage disequilibrium analyses indicated that these markers were in low LD (3 828 611-rs10043986, r(2)=0.008; rs10043986-rs4704591, r(2)=0.204). In addition, CMYA5 was reported to be physically interacting with the DTNBP1 gene, a promising candidate for schizophrenia, suggesting that CMYA5 may be involved in the same biological pathway and process. On the basis of this information, we performed replication studies for these three single-nucleotide polymorphisms. The rs3828611 was found to have conflicting results in our Irish samples and was dropped out without further investigation. The other two markers were verified in 23 other independent data sets. In a meta-analysis of all 23 replication samples (family samples, 912 families with 4160 subjects; case-control samples, 11 380 cases and 15 021 controls), we found that both markers are significantly associated with schizophrenia (rs10043986, odds ratio (OR)=1.11, 95% confidence interval (CI)=1.04-1.18, P=8.2 × 10(-4) and rs4704591, OR=1.07, 95% CI=1.03-1.11, P=3.0 × 10(-4)). The results were also significant for the 22 Caucasian replication samples (rs10043986, OR=1.11, 95% CI=1.03-1.17, P=0.0026 and rs4704591, OR=1.07, 95% CI=1.02-1.11, P=0.0015). Furthermore, haplotype conditioned analyses indicated that the association signals observed at these two markers are independent. On the basis of these results, we concluded that CMYA5 is associated with schizophrenia and further investigation of the gene is warranted.


Assuntos
Estudo de Associação Genômica Ampla , Proteínas Musculares/genética , Polimorfismo de Nucleotídeo Único , Esquizofrenia/genética , Negro ou Afro-Americano/genética , Proteínas de Transporte/genética , Estudos de Casos e Controles , Mineração de Dados , Disbindina , Proteínas Associadas à Distrofina , Alemanha/epidemiologia , Alemanha/etnologia , Humanos , Irlanda/epidemiologia , Judeus/genética , Desequilíbrio de Ligação , Pennsylvania/epidemiologia , Risco , Esquizofrenia/epidemiologia , Esquizofrenia/etnologia , População Branca/genética
12.
Environ Pollut ; 278: 116777, 2021 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-33689951

RESUMO

A recent surge in the use and abuse of diverse prescribed psychotic and illicit drugs necessitates the surveillance of drug residues in source water and the associated ecological impacts of chronic exposure to the aquatic organism. Thirty-six psychotic and illicit drug residues were determined in discharged wastewater from two centralized municipal wastewater treatment facilities and two wastewater receiving creeks for seven consecutive days in Kentucky. Zebrafish (Danio rerio) larvae were exposed to the environmental relevant mixtures of all drug residues, all illicit drugs, and all prescribed psychotic drugs. The extracted RNA from fish homogenates was sequenced, and differentially expressed sequences were analyzed for known or predicted nervous system expression, and screened annotated protein-coding genes to the true environmental cocktail mixture. Illicit stimulant (cocaine and one metabolite), opioids (methadone, methadone metabolite, and oxycodone), hallucinogen (MDA), benzodiazepine (oxazepam and temazepam), carbamazepine, and all target selective serotonin reuptake inhibitors including sertraline, fluoxetine, venlafaxine, and citalopram were quantified in 100% of collected samples from both creeks. The high dose cocktail mixture exposure group revealed the largest group of differentially expressed genes: 100 upregulated and 77 downregulated (p ≤ 0.05; q ≤ 0.05). The top 20 differentially expressed sequences in each exposure group comprise 82 unique transcripts corresponding to 74% annotated genes, 7% non-coding sequences, and 19% uncharacterized sequences. Among 61 differentially expressed sequences that corresponded to annotated protein-coding genes, 23 (38%) genes or their homologs are known to be expressed in the nervous system of fish or other organisms. Several of the differentially expressed sequences are associated primarily with the immune system, including several major histocompatibility complex class I and interferon-induced proteins. Interleukin-1 beta (downregulated in this study) abnormalities are considered a risk factor for psychosis. This is the first study to assess the contributions of multiple classes of psychotic and illicit drugs in combination with developmental gene expression.


Assuntos
Poluentes Químicos da Água , Peixe-Zebra , Animais , Fluoxetina , Larva , Inibidores Seletivos de Recaptação de Serotonina , Poluentes Químicos da Água/toxicidade
15.
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
18.
Mol Psychiatry ; 13(9): 873-7, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18195713

RESUMO

The etiology of schizophrenia is thought to include both epistasis and gene-environment interactions. We sought to test whether a set of schizophrenia candidate genes regulated by hypoxia or involved in vascular function in the brain (AKT1, BDNF, CAPON, CHRNA7, COMT, DTNBP1, GAD1, GRM3, NOTCH4, NRG1, PRODH, RGS4, TNF-alpha) interacted with serious obstetric complications to influence risk for schizophrenia. A family-based study of transmission disequilibrium was conducted in 116 trios. Twenty-nine probands had at least one serious obstetric complication (OC) using the McNeil-Sjostrom Scale, and many of the OCs reported were associated with the potential for fetal hypoxia. Analyses were conducted using conditional logistic regression and a likelihood ratio test (LRT) between nested models was performed to assess significance. Of the 13 genes examined, four (AKT1 (three SNPs), BDNF (two SNPs), DTNBP1 (one SNP) and GRM3 (one SNP)) showed significant evidence for gene-by-environment interaction (LRT P-values ranged from 0.011 to 0.037). Although our sample size was modest and the power to detect interactions was limited, we report significant evidence for genes involved in neurovascular function or regulated by hypoxia interacting with the presence of serious obstetric complications to increase risk for schizophrenia.


Assuntos
Regulação da Expressão Gênica/fisiologia , Complicações do Trabalho de Parto , Esquizofrenia/etiologia , Esquizofrenia/genética , Saúde da Família , Feminino , Humanos , Hipóxia-Isquemia Encefálica/embriologia , Hipóxia-Isquemia Encefálica/genética , Hipóxia-Isquemia Encefálica/fisiopatologia , Desequilíbrio de Ligação , Modelos Logísticos , Masculino , Razão de Chances , Polimorfismo de Nucleotídeo Único , Gravidez , Proteínas Proto-Oncogênicas c-akt/genética , Fatores de Risco , Inquéritos e Questionários
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA