Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
1.
Mol Psychiatry ; 23(5): 1251-1260, 2018 05.
Article in English | MEDLINE | ID: mdl-28485405

ABSTRACT

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.


Subject(s)
Schizophrenia/immunology , Schizophrenia/pathology , Adult , Brain/pathology , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Neuroimmunomodulation , Oligonucleotide Array Sequence Analysis , Schizophrenia/genetics , Sequence Analysis, RNA
2.
Mol Psychiatry ; 23(6): 1496-1505, 2018 06.
Article in English | MEDLINE | ID: mdl-28485403

ABSTRACT

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.


Subject(s)
Glutamate Decarboxylase/genetics , Schizophrenia/genetics , Adolescent , Adult , Alternative Splicing/genetics , Autopsy , Brain/metabolism , Child , Child, Preschool , DNA Methylation/genetics , Female , Gene Expression/genetics , Genetic Variation/genetics , Glutamate Decarboxylase/metabolism , Hippocampus/metabolism , Humans , Infant, Newborn , Male , Prefrontal Cortex/metabolism , Promoter Regions, Genetic/genetics , RNA Isoforms/genetics , RNA, Messenger/metabolism , Schizophrenia/metabolism
3.
Mol Psychiatry ; 23(5): 1145-1156, 2018 05.
Article in English | MEDLINE | ID: mdl-28630453

ABSTRACT

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.


Subject(s)
Epigenesis, Genetic , Stress Disorders, Post-Traumatic/genetics , Adult , Cohort Studies , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genetic Predisposition to Disease , Genetic Testing/methods , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Longitudinal Studies , Male , Military Personnel/psychology , Prospective Studies , Repressor Proteins , Stress Disorders, Post-Traumatic/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Pharmacogenomics J ; 18(1): 121-126, 2018 01.
Article in English | MEDLINE | ID: mdl-27897267

ABSTRACT

The efflux pump, p-glycoprotein, controls bioavailability and excretion of pharmaceutical compounds. In the blood-brain barrier, p-glycoprotein regulates the delivery of pharmaceutical substances to the brain, influencing efficacy and side effects for some drugs notably antipsychotics. Common side effects to antipsychotics include obesity and metabolic disease. Polymorphisms in the ABCB1 gene coding for p-glycoprotein are associated with more severe side effects to neuro-pharmaceuticals as well as weight gain, indicating a potential link between p-glycoprotein function and metabolic regulation. Using microarray data analysis from 145 neurologically sound adults, this study investigated the association between body mass index (BMI) and ABCB1 expression in the frontal cortex. Increasing BMI values were associated with a statistically significantly reduced expression of ABCB1. Investigation of DNA methylation patterns in a subgroup of 52 individuals found that the methylation/expression ratios of ABCB1 were unaffected by increasing BMI values. Interestingly, the effect of BMI on ABCB1 expression appeared stronger in African Americans than in Caucasians.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Black or African American/genetics , Brain/metabolism , White People/genetics , ATP Binding Cassette Transporter, Subfamily B/genetics , Adolescent , Adult , Antipsychotic Agents/therapeutic use , Blood-Brain Barrier/metabolism , Body Mass Index , Brain/drug effects , DNA Methylation/drug effects , DNA Methylation/genetics , Female , Humans , Male , Middle Aged , Obesity/genetics , Polymorphism, Genetic/genetics , Weight Gain/drug effects , Weight Gain/genetics , Young Adult
5.
Mol Psychiatry ; 21(5): 701-6, 2016 May.
Article in English | MEDLINE | ID: mdl-26216298

ABSTRACT

Neurexin 1 (NRXN1), a presynaptic cell adhesion molecule, is implicated in several neurodevelopmental disorders characterized by synaptic dysfunction including autism, intellectual disability and schizophrenia. To gain insight into NRXN1's involvement in human cortical development we used quantitative real-time PCR to examine the expression trajectories of NRXN1, and its predominant isoforms, NRXN1-α and NRXN1-ß, in prefrontal cortex from fetal stages to aging. In addition, we investigated whether prefrontal cortical expression levels of NRXN1 transcripts are altered in schizophrenia or bipolar disorder in comparison with non-psychiatric control subjects. We observed that all three NRXN1 transcripts were highly expressed during human fetal cortical development, markedly increasing with gestational age. In the postnatal dorsolateral prefrontal cortex, expression levels were negatively correlated with age, peaking at birth until ~3 years of age, after which levels declined markedly to be stable across the lifespan. NRXN1-ß expression was modestly but significantly elevated in the brains of patients with schizophrenia compared with non-psychiatric controls, whereas NRXN1-α expression was increased in bipolar disorder. These data provide novel evidence that NRXN1 expression is highest in human dorsolateral prefrontal cortex during critical developmental windows relevant to the onset and diagnosis of a range of neurodevelopmental disorders, and that NRXN1 expression may be differentially altered in neuropsychiatric disorders.


Subject(s)
Aging/metabolism , Bipolar Disorder/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Neocortex/growth & development , Neocortex/metabolism , Nerve Tissue Proteins/metabolism , Schizophrenia/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Calcium-Binding Proteins , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Neural Cell Adhesion Molecules , Protein Isoforms , Young Adult
6.
Mol Psychiatry ; 20(9): 1057-68, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26169973

ABSTRACT

Accumulating data indicate that the glutamate system is disrupted in major depressive disorder (MDD), and recent clinical research suggests that ketamine, an antagonist of the N-methyl-d-aspartate (NMDA) glutamate receptor (GluR), has rapid antidepressant efficacy. Here we report findings from gene expression studies of a large cohort of postmortem subjects, including subjects with MDD and controls. Our data reveal higher expression levels of the majority of glutamatergic genes tested in the dorsolateral prefrontal cortex (DLPFC) in MDD (F21,59=2.32, P=0.006). Posthoc data indicate that these gene expression differences occurred mostly in the female subjects. Higher expression levels of GRIN1, GRIN2A-D, GRIA2-4, GRIK1-2, GRM1, GRM4, GRM5 and GRM7 were detected in the female patients with MDD. In contrast, GRM5 expression was lower in male MDD patients relative to male controls. When MDD suicides were compared with MDD non-suicides, GRIN2B, GRIK3 and GRM2 were expressed at higher levels in the suicides. Higher expression levels were detected for several additional genes, but these were not statistically significant after correction for multiple comparisons. In summary, our analyses indicate a generalized disruption of the regulation of the GluRs in the DLPFC of females with MDD, with more specific GluR alterations in the suicides and in the male groups. These data reveal further evidence that, in addition to the NMDA receptor, the AMPA, kainate and the metabotropic GluRs may be targets for the development of rapidly acting antidepressant drugs.


Subject(s)
Depressive Disorder, Major/metabolism , Depressive Disorder, Major/psychology , Prefrontal Cortex/metabolism , Receptors, Glutamate/biosynthesis , Receptors, N-Methyl-D-Aspartate/metabolism , Suicide/psychology , Adult , Case-Control Studies , Depressive Disorder, Major/genetics , Female , Gene Expression , Glutamic Acid/metabolism , Humans , Ketamine/therapeutic use , Male , Receptors, Glutamate/genetics , Sex Factors , Transcriptome
7.
Mol Psychiatry ; 19(2): 192-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23295814

ABSTRACT

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.


Subject(s)
Bipolar Disorder/metabolism , Brain/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/genetics , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Schizophrenia/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antipsychotic Agents/pharmacology , Bipolar Disorder/genetics , Brain/drug effects , Brain/growth & development , Child , Child, Preschool , Depressive Disorder, Major/genetics , Depressive Disorder, Major/metabolism , Female , Fetus , Humans , Infant , Male , Middle Aged , Rats , Rats, Sprague-Dawley , Schizophrenia/drug therapy , Schizophrenia/genetics
8.
Mol Psychiatry ; 19(11): 1243-50, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24492349

ABSTRACT

The common APOE2 gene variant is neuroprotective against Alzheimer's disease (AD) and reduces risk by nearly 50%. However, the mechanisms by which APOE2 confers neuroprotection are largely unknown. Here we showed that ApoE protein abundance in human postmortem cortex follows an isoform-dependent pattern (E2>E3>E4). We also identified a unique downstream transcriptional profile determined by microarray and characterized by downregulation of long-term potentiation (LTP) related transcripts and upregulation of extracellular matrix (ECM)/integrin-related transcripts in E2 cases and corroborated this finding at the protein level by demonstrating increases in ECM collagens and laminins. In vivo studies of healthy older individuals demonstrated a unique and advantageous biomarker signature in E2 carriers. APOE2 also reduced the risk of mild cognitive impairment to AD conversion by half. Our findings suggest that ApoE2 protein abundance, coupled with its inability to bind to LDLRs, may act to increase amyloid-beta (Ab) clearance. In addition, increased ECM and reduced LTP-related expression results in diminished activity-dependent Ab secretion and/or excitotoxicity, and thus also promotes neuroprotection.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Apolipoprotein E2/genetics , Apolipoprotein E2/metabolism , Adult , Aged , Alzheimer Disease/diagnosis , Biomarkers/metabolism , Cerebral Cortex/physiopathology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/physiopathology , Collagen/metabolism , Disease Progression , Extracellular Matrix/metabolism , Female , Humans , Integrins/metabolism , Laminin/metabolism , Long-Term Potentiation/physiology , Male , Middle Aged , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Risk
9.
Mol Psychiatry ; 19(12): 1258-66, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24322206

ABSTRACT

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.


Subject(s)
Bipolar Disorder/genetics , Brain/metabolism , Depressive Disorder, Major/genetics , Receptors, Dopamine D1/genetics , Receptors, Dopamine D2/genetics , Schizophrenia/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Bipolar Disorder/metabolism , Brain/growth & development , Child , Child, Preschool , Cohort Studies , Depressive Disorder, Major/metabolism , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Infant , Infant, Newborn , Male , Middle Aged , Polymorphism, Single Nucleotide , RNA Splicing , RNA, Messenger/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Schizophrenia/metabolism , Young Adult
10.
Mol Psychiatry ; 19(4): 478-85, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23528911

ABSTRACT

The underlying pathology of schizophrenia (SZ) is likely as heterogeneous as its symptomatology. A variety of cortical and subcortical regions, including the prefrontal cortex, have been implicated in its pathology, and a number of genes have been identified as risk factors for disease development. We used in situ hybridization (ISH) to examine the expression of 58 genes in the dorsolateral prefrontal cortex (DLPFC, comprised of Brodmann areas 9 and 46) from 19 individuals with a premorbid diagnosis of SZ and 33 control individuals. Genes were selected based on: (1) previous identification as risk factors for SZ; (2) cell type markers or (3) laminar markers. Cell density and staining intensity were compared in the DLPFC, as well as separately in Brodmann areas 9 and 46. The expression patterns of a variety of genes, many of which are associated with the GABAergic system, were altered in SZ when compared with controls. Additional genes, including C8orf79 and NR4A2, showed alterations in cell density or staining intensity between the groups, highlighting the need for additional studies. Alterations were, with only a few exceptions, limited to Brodmann area 9, suggesting regional specificity of pathology in the DLPFC. Our results agree with previous studies on the GABAergic involvement in SZ, and suggest that areas 9 and 46 may be differentially affected in the disease. This study also highlights additional genes that may be altered in SZ, and indicates that these potentially interesting genes can be identified by ISH and high-throughput image analysis techniques.


Subject(s)
Gene Expression Regulation/physiology , Prefrontal Cortex/physiopathology , Schizophrenia/pathology , Adult , Cell Count , Female , Humans , Male , Middle Aged , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroglia/metabolism , Neuroimaging , Neurons/metabolism , Prefrontal Cortex/pathology , Schizophrenia/genetics , Young Adult
11.
Mol Psychiatry ; 16(8): 836-47, 2011 Aug.
Article in English | MEDLINE | ID: mdl-20479757

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative condition characterized histopathologically by neuritic plaques and neurofibrillary tangles. The objective of this transcriptional profiling study was to identify both neurosusceptibility and intrinsic neuroprotective factors at the molecular level, not confounded by the downstream consequences of pathology. We thus studied post-mortem cortical tissue in 28 cases that were non-APOE4 carriers (called the APOE3 group) and 13 cases that were APOE4 carriers. As APOE genotype is the major genetic risk factor for late-onset AD, the former group was at low risk for development of the disease and the latter group was at high risk for the disease. Mean age at death was 42 years and none of the brains had histopathology diagnostic of AD at the time of death. We first derived interregional difference scores in expression between cortical tissue from a region relatively invulnerable to AD (primary somatosensory cortex, BA 1/2/3) and an area known to be susceptible to AD pathology (middle temporal gyrus, BA 21). We then contrasted the magnitude of these interregional differences in between-group comparisons of the APOE3 (low risk) and APOE4 (high risk) genotype groups. We identified 70 transcripts that differed significantly between the groups. These included EGFR, CNTFR, CASP6, GRIA2, CTNNB1, FKBPL, LGALS1 and PSMC5. Using real-time quantitative PCR, we validated these findings. In addition, we found regional differences in the expression of APOE itself. We also identified multiple Kyoto pathways that were disrupted in the APOE4 group, including those involved in mitochondrial function, calcium regulation and cell-cycle reentry. To determine the functional significance of our transcriptional findings, we used bioinformatics pathway analyses to demonstrate that the molecules listed above comprised a network of connections with each other, APOE, and APP and MAPT. Overall, our results indicated that the abnormalities that we observed in single transcripts and in signaling pathways were not the consequences of diagnostic plaque and tangle pathology, but preceded it and thus may be a causative link in the long molecular prodrome that results in clinical AD.


Subject(s)
Alzheimer Disease/genetics , Apolipoprotein E4/genetics , Gene Expression/genetics , Genetic Predisposition to Disease/genetics , Signal Transduction/genetics , Adult , Apolipoprotein E3/genetics , Apolipoprotein E4/metabolism , Apolipoproteins E/biosynthesis , Apolipoproteins E/genetics , Cerebral Cortex/metabolism , Databases, Genetic , Female , Genotype , Heterozygote , Humans , Male , Middle Aged
14.
Science ; 273(5279): 1225-7, 1996 Aug 30.
Article in English | MEDLINE | ID: mdl-8703056

ABSTRACT

Tourette syndrome, a chronic tic disorder with autosomal dominant inheritance, exhibits considerable phenotypic variability even within monozygotic twin pairs. The origins of this variability remain unclear. Recent findings have implicated the caudate nucleus as a locus of pathology, and pharmacological evidence supports dopaminergic involvement. Within monozygotic twins discordant for Tourette syndrome severity, differences in D2 dopamine receptor binding in the head of the caudate nucleus predicted differences in phenotypic severity (r = 0.99); this relation was not observed in putamen. These data may link Tourette syndrome with a spectrum of neuropsychiatric disorders that involve associative striatal circuitry.


Subject(s)
Caudate Nucleus/metabolism , Diseases in Twins , Receptors, Dopamine D2/metabolism , Tourette Syndrome/metabolism , Twins, Monozygotic , Adult , Benzamides/metabolism , Dopamine Antagonists/metabolism , Female , Humans , Male , Phenotype , Putamen/metabolism , Pyrrolidines/metabolism , Tomography, Emission-Computed, Single-Photon , Tourette Syndrome/genetics
15.
Transl Psychiatry ; 7(2): e1044, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28244985

ABSTRACT

Several studies link increasing body mass index (BMI) to cognitive decline both as a consequence of obesity per se and as a sequela of obesity-induced type 2 diabetes. Obese individuals are prone to a chronic low-grade inflammation as the metabolically active visceral fat produces proinflammatory cytokines. Animal studies indicate that these cytokines can cross the blood-brain barrier. Such crossover could potentially affect the immune system in the brain by inducing gene expression of proinflammatory genes. The relationship between obesity and neuroinflammation in the human brain is currently unknown. Therefore we aim to examine the relationship between BMI and gene expression of central inflammatory markers in the human frontal cortex. Microarray data of 141 neurologically and psychiatrically healthy individuals were obtained through the BrainCloud database. A simple linear regression analysis was performed with BMI as variable on data on IL10, IL1ß, IL6, PTGS2 (COX2) and NOS2 (iNOS). Increasing BMI is associated with a decrease in the mRNA expression of IL10 (P=0.014) and an increase in the expression of NOS2 (iNOS; P=0.040). Expressions of IL10 and NOS2 (iNOS) were negatively correlated (P<0.001). The expression of IL10 was mostly affected by individuals with BMI ⩾40. Multiple linear regression analyses with BMI, age, sex and race as variables were performed in order to identify potential confounders. In conclusion, increasing BMI could affect the IL10-mediated anti-inflammatory defense in the brain and induce iNOS-mediated inflammatory activity.


Subject(s)
Frontal Lobe/metabolism , Interleukin-10/genetics , Nitric Oxide Synthase Type II/genetics , Obesity/metabolism , RNA, Messenger/metabolism , Thinness/metabolism , Adolescent , Adult , Aged , Body Mass Index , Child , Child, Preschool , Cyclooxygenase 2/genetics , Female , Humans , Infant , Infant, Newborn , Interleukin-1beta/genetics , Interleukin-6/genetics , Linear Models , Male , Middle Aged , Overweight/metabolism , Young Adult
16.
Transl Psychiatry ; 7(5): e1126, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28485729

ABSTRACT

The histaminergic system (HS) has a critical role in cognition, sleep and other behaviors. Although not well studied in autism spectrum disorder (ASD), the HS is implicated in many neurological disorders, some of which share comorbidity with ASD, including Tourette syndrome (TS). Preliminary studies suggest that antagonism of histamine receptors 1-3 reduces symptoms and specific behaviors in ASD patients and relevant animal models. In addition, the HS mediates neuroinflammation, which may be heightened in ASD. Together, this suggests that the HS may also be altered in ASD. Using RNA sequencing (RNA-seq), we investigated genome-wide expression, as well as a focused gene set analysis of key HS genes (HDC, HNMT, HRH1, HRH2, HRH3 and HRH4) in postmortem dorsolateral prefrontal cortex (DLPFC) initially in 13 subjects with ASD and 39 matched controls. At the genome level, eight transcripts were differentially expressed (false discovery rate <0.05), six of which were small nucleolar RNAs (snoRNAs). There was no significant diagnosis effect on any of the individual HS genes but expression of the gene set of HNMT, HRH1, HRH2 and HRH3 was significantly altered. Curated HS gene sets were also significantly differentially expressed. Differential expression analysis of these gene sets in an independent RNA-seq ASD data set from DLPFC of 47 additional subjects confirmed these findings. Understanding the physiological relevance of an altered HS may suggest new therapeutic options for the treatment of ASD.


Subject(s)
Autism Spectrum Disorder/genetics , Histamine/genetics , Receptors, Histamine/drug effects , Sequence Analysis, RNA/methods , Tourette Syndrome/genetics , Adolescent , Adult , Aged , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/physiopathology , Child , Child, Preschool , Cognition/physiology , Diagnosis , Female , Genome-Wide Association Study/methods , Histamine/metabolism , Humans , Male , Middle Aged , Neurogenic Inflammation/genetics , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Sleep/physiology , Tourette Syndrome/metabolism , Tourette Syndrome/physiopathology , Transcriptome/genetics , Young Adult
17.
Transl Psychiatry ; 6(6): e838, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27300264

ABSTRACT

The incidence of Alzheimer's disease (AD) is increasing. Major risk factors for AD are advancing age and diabetes. Lately, obesity has been associated with an increased risk of dementia. Obese and diabetic individuals are prone to decreased circulating levels of zinc, reducing the amount of zinc available for crucial intracellular processes. In the brain, zinc co-localizes with glutamate in synaptic vesicles, and modulates NMDA receptor activity. Intracellular zinc is involved in apoptosis and fluctuations in cytoplasmic Zn(2+) affect modulation of intracellular signaling. The ZNT and ZIP proteins participate in intracellular zinc homeostasis. Altered expression of zinc-regulatory proteins has been described in AD patients. Using microarray data from human frontal cortex (BrainCloud), this study investigates expression of the SCLA30A (ZNT) and SCLA39A (ZIP) families of genes in a Caucasian and African-American sample of 145 neurologically and psychiatrically normal individuals. Expression of ZNT3 and ZNT4 were significantly reduced with increasing age, whereas expression of ZIP1, ZIP9 and ZIP13 were significantly increased. Increasing body mass index (BMI) correlated with a significant reduction in ZNT1 expression similar to what is seen in the early stages of AD. Increasing BMI also correlated with reduced expression of ZNT6. In conclusion, we found that the expression of genes that regulate intracellular zinc homeostasis in the human frontal cortex is altered with increasing age and affected by increasing BMI. With the increasing rates of obesity throughout the world, these findings warrant continuous scrutiny of the long-term consequences of obesity on brain function and the development of neurodegenerative diseases.


Subject(s)
Alzheimer Disease/genetics , Black or African American/genetics , Carrier Proteins/genetics , DNA-Binding Proteins/genetics , Frontal Lobe/metabolism , Gene Expression/genetics , Obesity/complications , Obesity/genetics , Transcription Factors/genetics , White People/genetics , Adult , Age Factors , Aged , Aged, 80 and over , Alzheimer Disease/physiopathology , Body Mass Index , Cation Transport Proteins/genetics , Databases, Genetic , Female , Homeostasis/genetics , Humans , Intracellular Signaling Peptides and Proteins/physiology , Middle Aged , Obesity/physiopathology , RNA, Messenger/genetics , Risk Factors , Statistics as Topic , Zinc/metabolism , Transcription Factor MTF-1
18.
J Neurosci ; 20(20): 7798-806, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11027244

ABSTRACT

Chronic treatment of humans or experimental animals with classical neuroleptic drugs can lead to abnormal, tardive movements that persist long after the drugs are withdrawn. A role in these neuroleptic-induced dyskinesias may be played by a structural change in the shell of the nucleus accumbens where the opioid peptide dynorphin is upregulated in treated rats that show vacuous chewing movements (VCMs). The shell of the nucleus accumbens normally contains a dense plexus of dynorphinergic fibers especially in its caudomedial part. After 27 weeks of haloperidol administration and 18 weeks of withdrawal, the immunoreactive labeling of this plexus is intensified when compared with that after vehicle treatment. In addition, medium spiny neurons here show a significant increase in spine density, dendritic branching, and numbers of terminal segments. In the VCM-positive animals, the dendritic surface area is reduced, and dynorphin-positive terminals contact more spines and form more asymmetrical specializations than do those in animals without the syndrome (VCM-negative and vehicle-treated groups). Persistent, neuroleptic-induced oral dyskinesias could therefore be caused by incontrovertible alterations, involving terminal remodeling or sprouting, to the synaptic connectivity of the accumbal shell.


Subject(s)
Dendrites/metabolism , Dynorphins/metabolism , Dyskinesia, Drug-Induced/metabolism , Nucleus Accumbens/metabolism , Synapses/metabolism , Animals , Antipsychotic Agents/toxicity , Behavior, Animal/drug effects , Dendrites/drug effects , Dendrites/ultrastructure , Disease Models, Animal , Dyskinesia, Drug-Induced/pathology , Haloperidol/toxicity , Male , Mastication/drug effects , Microscopy, Electron , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nucleus Accumbens/drug effects , Nucleus Accumbens/pathology , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , Rats, Wistar , Synapses/drug effects , Synapses/ultrastructure
19.
Diabetes ; 45(7): 992-4, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8666155

ABSTRACT

Leptin-receptor gene expression in hypothalamic tissue from lean and obese humans was examined. The full-length leptin receptor, that is believed to transmit the leptin signal, is expressed in human hypothalamus. There was no difference in the amount of leptin-receptor mRNA In seven lean (BMI 23.3 +/- 0.9 kg/m2) and eight obese (BMI 36.9 +/- 1.5) subjects as determined by reverse transcription-polymerase chain reaction. A sequence polymorphism (A-->G) was detected at position 668 of the leptin receptor cDNA. This second base substitution changed a glutamine to an arginine at position 223 of the leptin receptor protein. Of 15 subjects analyzed, 11 were heterozygous for this base change and 3 were homozygous. The occurrence [correction of occurance] of the polymorphic allele(s) did not correlate with BMI in the population studied. The mutation responsible for the defect in the leptin receptor in db/db mice was not detected in any obese human, nor was the fa/fa rat mutation. These results provide evidence that the leptin resistance observed in obese humans is not due to a defect in the leptin receptor.


Subject(s)
Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Hypothalamus/metabolism , Polymorphism, Genetic , Receptors, Cell Surface , Animals , Arginine , Body Weight , DNA, Complementary , Gene Expression , Glutamine , Humans , Mice , Mice, Mutant Strains , Obesity , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Rats , Rats, Mutant Strains , Receptors, Cytokine/biosynthesis , Receptors, Cytokine/genetics , Receptors, Leptin , Thinness
20.
Neuroscience ; 131(1): 219-33, 2005.
Article in English | MEDLINE | ID: mdl-15680705

ABSTRACT

Basic fibroblast growth factor (bFGF) is an important mitogen and neurotrophic factor that binds and signals through the high-affinity receptor, fibroblast growth factor receptor 1 (FGFR1). However, only a limited amount of information is available concerning the molecular forms and anatomical distribution of fibroblast growth factors (FGFs) in the normal human brain. We found multiple bFGF and FGFR1 mRNA transcripts which vary in expression pattern across human brain regions. Using in situ hybridization and immunohistochemistry, we localized bFGF and FGFR1 mRNA and protein to cells in the normal adult human hippocampus and caudal entorhinal cortex (ERC). The majority of pyramidal neurons contained FGFR1 mRNA and protein in the mesial temporal lobe, with neurons in the CA2/CA3 region demonstrating the highest levels of FGFR1 mRNA. In contrast to FGFR1, bFGF mRNA expression was detected at very low levels in a small fraction of the neurons in the human hippocampus and caudal ERC. While bFGF mRNA may be expressed at low levels in neurons, bFGF-immunopositive cells with astrocytic features were detected throughout the mesial temporal lobe in rats, monkeys and humans. bFGF immunoreactive processes are found traversing the dentate gyrus, and bFGF immunoreactive cells are found in the neurogenic subgranular zone in all three mammalian species studied. The anatomical distribution of these two FGF family members suggests that bFGF is endogenously positioned to be involved in ongoing neurogenesis in the adult hippocampus, and that FGF trophic signaling to differentiated neurons could involve the release of astrocytic bFGF acting on neuronal FGFR1 in the normal adult human hippocampus.


Subject(s)
Fibroblast Growth Factor 2/genetics , Gene Expression Regulation , Hippocampus/physiology , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Fibroblast Growth Factor/genetics , Adult , Autopsy , Black People , Female , Humans , In Situ Hybridization , Male , Mesothelin , Middle Aged , RNA, Messenger/genetics , Receptor, Fibroblast Growth Factor, Type 1 , Reference Values , United States , White People
SELECTION OF CITATIONS
SEARCH DETAIL