Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Psychiatry ; 23(9): 1920-1928, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-28948973

RESUMO

Maternal inflammation and diabetes increase the risk for psychiatric disorders in offspring. We hypothesized that these co-occurring risk factors may potentiate each other. To test this, we maternally exposed developing mice in utero to gestational diabetes mellitus (GDM) and/or maternal immune activation (MIA). Fetal mouse brains were exposed to either vehicle, GDM, MIA or GDM+MIA. At gestational day (GD) 12.5, GDM produced a hyperglycemic, hyperleptinemic maternal state, whereas MIA produced significant increases in proinflammatory cytokines and chemokines. Each condition alone resulted in an altered, inflammatory and neurodevelopmental transcriptome profile. In addition, GDM+MIA heightened the maternal inflammatory state and gave rise to a new, specific transcriptional response. This exacerbated response was associated with pathways implicated in psychiatric disorders, including dopamine neuron differentiation and innate immune response. Based on these data, we hypothesize that children born to GDM mothers and exposed to midgestation infections have an increased vulnerability to psychiatric disorder later in life, and this should be tested in follow-up epidemiological studies.


Assuntos
Diabetes Gestacional/imunologia , Diabetes Gestacional/fisiopatologia , Imunidade Ativa/imunologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/embriologia , Quimiocinas/metabolismo , Citocinas/metabolismo , Feminino , Imunidade Ativa/fisiologia , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mães , Gravidez , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Fatores de Risco
2.
Neuroscience ; 320: 105-21, 2016 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-26855193

RESUMO

In order to decipher the disease etiology, progression and treatment of multifactorial human brain diseases we utilize a host of different experimental models. Recently, patient-derived human dermal fibroblast (HDF) cultures have re-emerged as promising in vitro functional system for examining various cellular, molecular, metabolic and (patho)physiological states and traits of psychiatric disorders. HDF studies serve as a powerful complement to postmortem and animal studies, and often appear to be informative about the altered homeostasis in neural tissue. Studies of HDFs from patients with schizophrenia (SZ), depression, bipolar disorder (BD), autism, attention deficit and hyperactivity disorder and other psychiatric disorders have significantly advanced our understanding of these devastating diseases. These reports unequivocally prove that signal transduction, redox homeostasis, circadian rhythms and gene*environment (G*E) interactions are all amenable for assessment by the HDF model. Furthermore, the reported findings suggest that this underutilized patient biomaterial, combined with modern molecular biology techniques, may have both diagnostic and prognostic value, including prediction of response to therapeutic agents.


Assuntos
Fibroblastos , Transtornos Mentais , Modelos Biológicos , Psiquiatria/métodos , Pesquisa Biomédica/métodos , Células Cultivadas , Humanos , Pele
3.
Transl Psychiatry ; 5: e656, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26460480

RESUMO

Antagonists of the N-methyl-D-aspartate (NMDA)-type glutamate receptor induce psychosis in healthy individuals and exacerbate schizophrenia symptoms in patients. In this study we have produced an animal model of NMDA receptor hypofunction by chronically treating rats with low doses of the NMDA receptor antagonist MK-801. Subsequently, we performed an expression study and identified 20 genes showing altered expression in the brain of these rats compared with untreated animals. We then explored whether the human orthologs of these genes are associated with schizophrenia in the largest schizophrenia genome-wide association study published to date, and found evidence for association for 4 out of the 20 genes: SF3B1, FOXP1, DLG2 and VGLL4. Interestingly, three of these genes, FOXP1, SF3B1 and DLG2, have previously been implicated in neurodevelopmental disorders.


Assuntos
Maleato de Dizocilpina/farmacologia , Fatores de Transcrição Forkhead/genética , Guanilato Quinases/genética , Hipocampo , Fosfoproteínas/genética , Proteínas Repressoras/genética , Ribonucleoproteína Nuclear Pequena U2/genética , Esquizofrenia , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Adulto , Idoso , Animais , Estudos de Casos e Controles , Feminino , Estudo de Associação Genômica Ampla , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Psicotrópicos/farmacologia , Fatores de Processamento de RNA , Ratos , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Esquizofrenia/genética , Esquizofrenia/patologia
4.
Transl Psychiatry ; 5: e523, 2015 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-25756806

RESUMO

Major depressive disorder (MDD) is increasingly viewed as interplay of environmental stressors and genetic predisposition, and recent data suggest that the disease affects not only the brain, but the entire body. As a result, we aimed at determining whether patients with major depression have aberrant molecular responses to stress in peripheral tissues. We examined the effects of two metabolic stressors, galactose (GAL) or reduced lipids (RL), on the transcriptome and miRNome of human fibroblasts from 16 pairs of patients with MDD and matched healthy controls (CNTR). Our results demonstrate that both MDD and CNTR fibroblasts had a robust molecular response to GAL and RL challenges. Most importantly, a significant part (messenger RNAs (mRNAs): 26-33%; microRNAs (miRNAs): 81-90%) of the molecular response was only observed in MDD, but not in CNTR fibroblasts. The applied metabolic challenges uncovered mRNA and miRNA signatures, identifying responses to each stressor characteristic for the MDD fibroblasts. The distinct responses of MDD fibroblasts to GAL and RL revealed an aberrant engagement of molecular pathways, such as apoptosis, regulation of cell cycle, cell migration, metabolic control and energy production. In conclusion, the metabolic challenges evoked by GAL or RL in dermal fibroblasts exposed adaptive dysfunctions on mRNA and miRNA levels that are characteristic for MDD. This finding underscores the need to challenge biological systems to bring out disease-specific deficits, which otherwise might remain hidden under resting conditions.


Assuntos
Transtorno Depressivo Maior/genética , Transtorno Depressivo Maior/metabolismo , Fibroblastos/metabolismo , Estresse Fisiológico/genética , Transcriptoma/genética , Adulto , Feminino , Humanos , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , RNA Mensageiro/genética , Pele/metabolismo , Adulto Jovem
5.
Mol Psychiatry ; 19(5): 580-7, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24322205

RESUMO

Gamma-aminobutyric acid (GABA)-ergic disturbances are hallmark features of schizophrenia and other neuropsychiatric disorders and encompass multiple interneuronal cell types. Using bacterial artificial chromosome-driven, miRNA silencing technology we generated transgenic mouse lines that suppress glutamic acid decarboxylase 1 (GAD1) in either cholecystokinin (CCK)- or neuropeptide Y (NPY)-expressing interneurons. In situ lipidomic and proteomic analyses on brain tissue sections revealed distinct, brain region-specific profiles in each transgenic line. Behavioral analyses revealed that suppression of GAD1 in CCK+ interneurons resulted in locomotor and olfactory sensory changes, whereas suppression in NPY+ interneurons affected anxiety-related behaviors and social interaction. Both transgenic mouse lines had altered sensitivity to amphetamine albeit in opposite directions. Together, these data argue that reduced GAD1 expression leads to altered molecular and behavioral profiles in a cell type-dependent manner, and that these subpopulations of interneurons are strong and opposing modulators of dopamine system function. Furthermore, our findings also support the hypothesis that neuronal networks are differentially controlled by diverse inhibitory subnetworks.


Assuntos
Comportamento/fisiologia , Colecistocinina/metabolismo , Glutamato Descarboxilase/metabolismo , Interneurônios/fisiologia , Neuropeptídeo Y/metabolismo , Ácido gama-Aminobutírico/metabolismo , Anfetamina/farmacologia , Animais , Ansiedade/fisiopatologia , Encéfalo/fisiologia , Estimulantes do Sistema Nervoso Central/farmacologia , Colecistocinina/genética , Glutamato Descarboxilase/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Neuropeptídeo Y/genética , Percepção Olfatória/fisiologia , Proteômica/métodos , Comportamento Social
7.
Transl Psychiatry ; 2: e98, 2012 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-22832908

RESUMO

We are exploring the mechanisms underlying how maternal infection increases the risk for schizophrenia and autism in the offspring. Several mouse models of maternal immune activation (MIA) were used to examine the immediate effects of MIA induced by influenza virus, poly(I:C) and interleukin IL-6 on the fetal brain transcriptome. Our results indicate that all three MIA treatments lead to strong and common gene expression changes in the embryonic brain. Most notably, there is an acute and transient upregulation of the α, ß and γ crystallin gene family. Furthermore, levels of crystallin gene expression are correlated with the severity of MIA as assessed by placental weight. The overall gene expression changes suggest that the response to MIA is a neuroprotective attempt by the developing brain to counteract environmental stress, but at a cost of disrupting typical neuronal differentiation and axonal growth. We propose that this cascade of events might parallel the mechanisms by which environmental insults contribute to the risk of neurodevelopmental disorders such as schizophrenia and autism.


Assuntos
Encéfalo/embriologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/genética , Influenza Humana/imunologia , Interleucina-6/imunologia , Poli I-C/imunologia , Complicações Infecciosas na Gravidez/imunologia , Transcriptoma/genética , Animais , Criança , Transtornos Globais do Desenvolvimento Infantil/genética , Transtornos Globais do Desenvolvimento Infantil/imunologia , Cristalinas/genética , Feminino , Humanos , Influenza Humana/genética , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sequência com Séries de Oligonucleotídeos , Tamanho do Órgão , Placenta/imunologia , Gravidez , Complicações Infecciosas na Gravidez/genética , Efeitos Tardios da Exposição Pré-Natal , RNA/genética , Proteínas Recombinantes/imunologia , Fatores de Risco , Esquizofrenia/genética , Esquizofrenia/imunologia , Transcriptoma/imunologia , Regulação para Cima/genética
8.
Mol Psychiatry ; 16(7): 751-62, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20479761

RESUMO

The etiology of major depression (MDD), a common and complex disorder, remains obscure. Gene expression profiling was conducted on post-mortem brain tissue samples from Brodmann Area 10 (BA10) in the prefrontal cortex from psychotropic drug-free persons with a history of MDD and age, gender, and post-mortem interval-matched normal controls (n=14 pairs of subjects). Microarray analysis was conducted using the Affymetrix Exon 1.0 ST arrays. A set of differential expression changes was determined by dual-fold change-probability criteria (∣average log ratios∣>0.585 [equivalent to a 1.5-fold difference in either direction], P<0.01), whereas molecular pathways of interest were evaluated using Gene Set Enrichment Analysis software. The results strongly implicate increased apoptotic stress in the samples from the MDD group. Three anti-apoptotic factors, Y-box-binding protein 1, caspase-1 dominant-negative inhibitor pseudo-ICE, and the putative apoptosis inhibitor FKGS2, were over-expressed. Gene set analysis suggested up-regulation of a variety of pro- and anti-inflammatory cytokines, including interleukin 1α (IL-1α), IL-2, IL-3, IL-5, IL-8, IL-9, IL-10, IL-12A, IL-13, IL-15, IL-18, interferon gamma (IFNγ), and lymphotoxin α (TNF superfamily member 1). The genes showing reduced expression included metallothionein 1M (MT1M), a zinc-binding protein with a significant function in the modulation of oxidative stress. The results of this study indicate that post-mortem brain tissue samples from BA10, a region that is involved in reward-related behavior, show evidence of local inflammatory, apoptotic, and oxidative stress in MDD.


Assuntos
Apoptose/fisiologia , Citocinas/metabolismo , Transtorno Depressivo Maior/patologia , Lobo Frontal/metabolismo , Lobo Frontal/fisiopatologia , Adulto , Idoso , Análise por Conglomerados , Citocinas/genética , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Estatística como Assunto
9.
Mol Psychiatry ; 15(10): 987-95, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20125089

RESUMO

In schizophrenia, glutamic acid decarboxylase 1 (GAD1) disturbances are robust, consistently observed, cell-type specific and represent a core feature of the disease. In addition, neuropeptide Y (NPY), which is a phenotypic marker of a sub-population of GAD1-containing interneurons, has shown reduced expression in the prefrontal cortex in subjects with schizophrenia, suggesting that dysfunction of the NPY+ cortical interneuronal sub-population might be a core feature of this devastating disorder. However, modeling gene expression disturbances in schizophrenia in a cell type-specific manner has been extremely challenging. To more closely mimic these molecular and cellular human post-mortem findings, we generated a transgenic mouse in which we downregulated GAD1 mRNA expression specifically in NPY+ neurons. This novel, cell type-specific in vivo system for reducing gene expression uses a bacterial artificial chromosome (BAC) containing the NPY promoter-enhancer elements, the reporter molecule (eGFP) and a modified intron containing a synthetic microRNA (miRNA) targeted to GAD1. The animals of isogenic strains are generated rapidly, providing a new tool for better understanding the molecular disturbances in the GABAergic system observed in complex neuropsychiatric disorders such as schizophrenia. In the future, because of the small size of the silencing miRNAs combined with our BAC strategy, this method may be modified to allow generation of mice with simultaneous silencing of multiple genes in the same cells with a single construct, and production of splice-variant-specific knockdown animals.


Assuntos
Cromossomos Artificiais Bacterianos , Modelos Animais de Doenças , Inativação Gênica , Camundongos Transgênicos , MicroRNAs/genética , Esquizofrenia/genética , Processamento Alternativo , Animais , Encefalopatias/genética , Encefalopatias/fisiopatologia , Regulação da Expressão Gênica/fisiologia , Glutamato Descarboxilase/genética , Células HEK293 , Humanos , Camundongos , Neuropeptídeo Y/genética , Esquizofrenia/fisiopatologia
10.
Mol Psychiatry ; 14(7): 705-18, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18317465

RESUMO

Protein kinase C enzymes play an important role in signal transduction, regulation of gene expression and control of cell division and differentiation. The fsI and betaII isoenzymes result from the alternative splicing of the PKCbeta gene (PRKCB1), previously found to be associated with autism. We performed a family-based association study in 229 simplex and 5 multiplex families, and a postmortem study of PRKCB1 gene expression in temporocortical gray matter (BA41/42) of 11 autistic patients and controls. PRKCB1 gene haplotypes are significantly associated with autism (P<0.05) and have the autistic endophenotype of enhanced oligopeptiduria (P<0.05). Temporocortical PRKCB1 gene expression was reduced on average by 35 and 31% for the PRKCB1-1 and PRKCB1-2 isoforms (P<0.01 and <0.05, respectively) according to qPCR. Protein amounts measured for the PKCbetaII isoform were similarly decreased by 35% (P=0.05). Decreased gene expression characterized patients carrying the 'normal' PRKCB1 alleles, whereas patients homozygous for the autism-associated alleles displayed mRNA levels comparable to those of controls. Whole genome expression analysis unveiled a partial disruption in the coordinated expression of PKCbeta-driven genes, including several cytokines. These results confirm the association between autism and PRKCB1 gene variants, point toward PKCbeta roles in altered epithelial permeability, demonstrate a significant downregulation of brain PRKCB1 gene expression in autism and suggest that it could represent a compensatory adjustment aimed at limiting an ongoing dysreactive immune process. Altogether, these data underscore potential PKCbeta roles in autism pathogenesis and spur interest in the identification and functional characterization of PRKCB1 gene variants conferring autism vulnerability.


Assuntos
Transtorno Autístico/genética , Transtorno Autístico/patologia , Expressão Gênica/genética , Predisposição Genética para Doença , Neocórtex/metabolismo , Proteína Quinase C/genética , Adolescente , Adulto , Distribuição de Qui-Quadrado , Criança , Pré-Escolar , Saúde da Família , Feminino , Frequência do Gene , Estudo de Associação Genômica Ampla/métodos , Genótipo , Humanos , Masculino , Polimorfismo de Nucleotídeo Único/genética , Proteína Quinase C beta , Adulto Jovem
11.
Mol Psychiatry ; 13(2): 147-61, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17471287

RESUMO

In subjects with schizophrenia, impairments in working memory are associated with dysfunction of the dorsolateral prefrontal cortex (DLPFC). This dysfunction appears to be due, at least in part, to abnormalities in gamma-aminobutyric acid (GABA)-mediated inhibitory circuitry. To test the hypothesis that altered GABA-mediated circuitry in the DLPFC of subjects with schizophrenia reflects expression changes of genes that encode selective presynaptic and postsynaptic components of GABA neurotransmission, we conducted a systematic expression analysis of GABA-related transcripts in the DLPFC of 14 pairs of schizophrenia and age-, sex- and post-mortem interval-matched control subjects using a customized DNA microarray with enhanced sensitivity and specificity. Subjects with schizophrenia exhibited expression deficits in GABA-related transcripts encoding (1) presynaptic regulators of GABA neurotransmission (67 kDa isoform of glutamic acid decarboxylase (GAD(67)) and GABA transporter 1), (2) neuropeptides (somatostatin (SST), neuropeptide Y (NPY) and cholecystokinin (CCK)) and (3) GABA(A) receptor subunits (alpha1, alpha4, beta3, gamma2 and delta). Real-time qPCR and/or in situ hybridization confirmed the deficits for six representative transcripts tested in the same pairs and in an extended cohort, respectively. In contrast, GAD(67), SST and alpha1 subunit mRNA levels, as assessed by in situ hybridization, were not altered in the DLPFC of monkeys chronically exposed to antipsychotic medications. These findings suggest that schizophrenia is associated with alterations in inhibitory inputs from SST/NPY-containing and CCK-containing subpopulations of GABA neurons and in the signaling via certain GABA(A) receptors that mediate synaptic (phasic) or extrasynaptic (tonic) inhibition. In concert with previous findings, these data suggest that working memory dysfunction in schizophrenia is mediated by altered GABA neurotransmission in certain DLPFC microcircuits.


Assuntos
Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Regulação da Expressão Gênica/fisiologia , Glutamato Descarboxilase/metabolismo , Córtex Pré-Frontal/metabolismo , Receptores de GABA-A/metabolismo , Esquizofrenia/patologia , Adulto , Idoso , Animais , Antipsicóticos/farmacologia , Benzodiazepinas/farmacologia , Estudos de Casos e Controles , Cloroquinolinóis/farmacologia , Feminino , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Glutamato Descarboxilase/genética , Humanos , Macaca fascicularis , Masculino , Pessoa de Meia-Idade , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Olanzapina , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Receptores de GABA-A/genética
12.
Genes Brain Behav ; 6(3): 229-39, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16827919

RESUMO

Many candidate gene association studies have evaluated incomplete, unrepresentative sets of single nucleotide polymorphisms (SNPs), producing non-significant results that are difficult to interpret. Using a rapid, efficient strategy designed to investigate all common SNPs, we tested associations between schizophrenia and two positional candidate genes: ACSL6 (Acyl-Coenzyme A synthetase long-chain family member 6) and SIRT5 (silent mating type information regulation 2 homologue 5). We initially evaluated the utility of DNA sequencing traces to estimate SNP allele frequencies in pooled DNA samples. The mean variances for the DNA sequencing estimates were acceptable and were comparable to other published methods (mean variance: 0.0008, range 0-0.0119). Using pooled DNA samples from cases with schizophrenia/schizoaffective disorder (Diagnostic and Statistical Manual of Mental Disorders edition IV criteria) and controls (n=200, each group), we next sequenced all exons, introns and flanking upstream/downstream sequences for ACSL6 and SIRT5. Among 69 identified SNPs, case-control allele frequency comparisons revealed nine suggestive associations (P<0.2). Each of these SNPs was next genotyped in the individual samples composing the pools. A suggestive association with rs 11743803 at ACSL6 remained (allele-wise P=0.02), with diminished evidence in an extended sample (448 cases, 554 controls, P=0.062). In conclusion, we propose a multi-stage method for comprehensive, rapid, efficient and economical genetic association analysis that enables simultaneous SNP detection and allele frequency estimation in large samples. This strategy may be particularly useful for research groups lacking access to high throughput genotyping facilities. Our analyses did not yield convincing evidence for associations of schizophrenia with ACSL6 or SIRT5.


Assuntos
Coenzima A Ligases/genética , DNA/genética , Frequência do Gene , Polimorfismo de Nucleotídeo Único/genética , Esquizofrenia/genética , Sirtuínas/genética , Estudos de Casos e Controles , Análise Mutacional de DNA/métodos , Pool Gênico , Predisposição Genética para Doença , Testes Genéticos/métodos , Humanos , Valores de Referência
13.
Mol Psychiatry ; 11(7): 633-48, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16702976

RESUMO

Brain-derived neurotrophic factor (BDNF) has been reported to be critical for the development of cortical inhibitory neurons. However, the effect of BDNF on the expression of transcripts whose protein products are involved in gamma amino butric acid (GABA) neurotransmission has not been assessed. In this study, gene expression profiling using oligonucleotide microarrays was performed in prefrontal cortical tissue from mice with inducible deletions of BDNF. Both embryonic and adulthood ablation of BDNF gave rise to many shared transcriptome changes. BDNF appeared to be required to maintain gene expression in the SST-NPY-TAC1 subclass of GABA neurons, although the absence of BDNF did not alter their general phenotype as inhibitory neurons. Furthermore, we observed expression alterations in genes encoding early-immediate genes (ARC, EGR1, EGR2, FOS, DUSP1, DUSP6) and critical cellular signaling systems (CDKN1c, CCND2, CAMK1g, RGS4). These BDNF-dependent gene expression changes may illuminate the biological basis for transcriptome changes observed in certain human brain disorders.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas do Tecido Nervoso/biossíntese , Córtex Pré-Frontal/metabolismo , Transcrição Gênica , Animais , Encefalopatias/genética , Fator Neurotrófico Derivado do Encéfalo/deficiência , Fator Neurotrófico Derivado do Encéfalo/genética , Cruzamentos Genéticos , Doxiciclina/farmacologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genes Precoces , Humanos , Proteínas Imediatamente Precoces/biossíntese , Interneurônios/química , Interneurônios/ultraestrutura , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Neurônios/classificação , Neurônios/metabolismo , Neuropeptídeo Y/biossíntese , Neuropeptídeo Y/genética , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos , Fenótipo , Córtex Pré-Frontal/embriologia , Córtex Pré-Frontal/crescimento & desenvolvimento , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/fisiologia , Deleção de Sequência , Somatostatina/biossíntese , Somatostatina/genética , Fatores de Tempo , Ácido gama-Aminobutírico/análise
14.
Mol Psychiatry ; 8(10): 863-78, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14515137

RESUMO

Presenilin 1 (PS1) plays a critical role in the nervous system development and PS1 mutations have been associated with familial Alzheimer's disease. PS1-deficient mice exhibit alterations in neural and vascular development and die in late embryogenesis. The present study was aimed at uncovering transcript networks that depend on intact PS1 function in the developing brain. To achieve this, we analyzed the brains of PS1-deficient and control animals at embryonic ages E12.5 and E14.5 using MG_U74Av2 oligonucleotide microarrays by Affymetrix. Based on the microarray data, overall molecular brain development appeared to be comparable between the E12.5 and E14.5 PS1-deficient and control embryos. However, in brains of PS1-deficient mice, we observed significant differences in the expression of genes encoding molecules that are associated with neural differentiation, extracellular matrix, vascular development, Notch-related signaling and lipid metabolism. Many of the expression differences between wild-type and PS1-deficient animals were present at both E12.5 and E14.5, whereas other transcript alterations were characteristic of only one developmental stage. The results suggest that the role of PS1 in development includes influences on a highly co-regulated transcript network; some of the genes participating in this expression network may contribute to the pathophysiology of Alzheimer's disease.


Assuntos
Doença de Alzheimer/genética , Química Encefálica/genética , Encéfalo/embriologia , Encéfalo/fisiologia , Proteínas de Membrana/genética , Análise de Sequência com Séries de Oligonucleotídeos , Doença de Alzheimer/fisiopatologia , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Masculino , Camundongos , Camundongos Mutantes , Gravidez , Presenilina-1 , Transcrição Gênica
16.
Trends Neurosci ; 24(8): 479-86, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11476888

RESUMO

The level of cellular and molecular complexity of the nervous system creates unique problems for the neuroscientist in the design and implementation of functional genomic studies. Microarray technologies can be powerful, with limitations, when applied to the analysis of human brain disorders. Recently, using cDNA microarrays, altered gene expression patterns between subjects with schizophrenia and controls were shown. Functional data mining led to two novel discoveries: a consistent decrease in the group of transcripts encoding proteins that regulate presynaptic function; and the most changed gene, which has never been previously associated with schizophrenia, regulator of G-protein signaling 4. From these and other findings, a hypothesis has been formulated to suggest that schizophrenia is a disease of the synapse. In the context of a neurodevelopmental model, it is proposed that impaired mechanics of synaptic transmission in specific neural circuits during childhood and adolescence ultimately results in altered synapse formation or pruning, or both, which manifest in the clinical onset of the disease.


Assuntos
Química Encefálica/genética , Análise de Sequência com Séries de Oligonucleotídeos , Esquizofrenia/genética , Sinapses/fisiologia , Humanos
18.
Nat Rev Neurosci ; 2(6): 444-7, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11389480

RESUMO

Making sense of microarray data is a complex process, in which the interpretation of findings will depend on the overall experimental design and judgement of the investigator performing the analysis. As a result, differences in tissue harvesting, microarray types, sample labelling and data analysis procedures make post hoc sharing of microarray data a great challenge. To ensure rapid and meaningful data exchange, we need to create some order out of the existing chaos. In these ground-breaking microarray standardization and data sharing efforts, NIH agencies should take a leading role


Assuntos
Bases de Dados Factuais/normas , Bases de Dados Factuais/tendências , Expressão Gênica/fisiologia , Neurociências/métodos , Neurociências/tendências , Análise de Sequência com Séries de Oligonucleotídeos/normas , Análise de Sequência com Séries de Oligonucleotídeos/tendências , Animais , Processamento Eletrônico de Dados/normas , Processamento Eletrônico de Dados/tendências , Humanos , National Institutes of Health (U.S.)/normas , National Institutes of Health (U.S.)/tendências , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Reprodutibilidade dos Testes , Projetos de Pesquisa/normas , Projetos de Pesquisa/tendências , Estados Unidos
19.
Mol Psychiatry ; 6(3): 293-301, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11326297

RESUMO

Complex defects in neuronal signaling may underlie the dysfunctions that characterize schizophrenia. Using cDNA microarrays, we discovered that the transcript encoding regulator of G-protein signaling 4 (RGS4) was the most consistently and significantly decreased in the prefrontal cortex of all schizophrenic subjects examined. The expression levels of ten other RGS family members represented on the microarrays were unchanged and hierarchical data analysis revealed that as a group, 274 genes associated with G-protein signaling were unchanged. Quantitative in situ hybridization verified the microarray RGS4 data, and demonstrated highly correlated decreases in RGS4 expression across three cortical areas of ten subjects with schizophrenia. RGS4 expression was not altered in the prefrontal cortex of subjects with major depressive disorder or in monkeys treated chronically with haloperidol. Interestingly, targets for 70 genes mapped to the major schizophrenia susceptibility locus 1q21--22 were present on the microarrays, of which only RGS4 gene expression was consistently altered. The combined data indicate that a decrease in RGS4 expression may be a common and specific feature of schizophrenia, which could be due either to genetic factors or a disease- specific adaptation, both of which could affect neuronal signaling.


Assuntos
Cromossomos Humanos Par 1 , Proteínas de Ligação ao GTP/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo , Esquizofrenia/genética , Adulto , Animais , Antipsicóticos/uso terapêutico , Transtorno Depressivo Maior/genética , Transtorno Depressivo Maior/metabolismo , Saúde da Família , Feminino , Expressão Gênica/fisiologia , Predisposição Genética para Doença , Haloperidol/uso terapêutico , Humanos , Macaca fascicularis , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Córtex Pré-Frontal/fisiologia , Esquizofrenia/tratamento farmacológico , Esquizofrenia/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...