Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Transl Psychiatry ; 6(11): e964, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27898074

ABSTRACT

The choroid plexus (CP) has a key role in maintaining brain homeostasis by producing cerebrospinal fluid (CSF), by mediating transport of nutrients and removing metabolic products from the central nervous system and by responding to peripheral inflammatory signals. Although abnormal markers of immune response and inflammation are apparent in individuals with schizophrenia, the CP of these individuals has not been characterized. We therefore sequenced mRNA from the CP from two independent collections of individuals with schizophrenia and unaffected controls. Genes related to immune function and inflammation were upregulated in both collections. In addition, a co-expression module related to immune/inflammation response that was generated by combining mRNA-Seq data from both collections was significantly associated with disease status. The immune/inflammation-related co-expression module was positively correlated with levels of C-reactive protein (CRP), cortisol and several immune modulator proteins in the serum of the same individuals and was also positively correlated with CRP, cortisol and pro-inflammatory cytokines in the frontal cortex of the same individuals. In addition, we found a substantial number of nodes (genes) that were common to our schizophrenia-associated immune/inflammation module from the pooled data and a module we generated from lippopolysaccharides-treated mouse model data. These results suggest that the CP of individuals with schizophrenia are responding to signals from the periphery by upregulating immune/inflammation-related genes to protect the brain and maintain the homeostasis but nevertheless fails to completely prevent immune/inflammation related changes in the brain.


Subject(s)
Choroid Plexus/metabolism , Exome Sequencing , Schizophrenia/genetics , Adult , Animals , C-Reactive Protein/genetics , Cytokines/blood , Disease Models, Animal , Frontal Lobe/metabolism , Humans , Hydrocortisone/blood , Inflammation/genetics , Inflammation Mediators/blood , Mice , RNA, Messenger/genetics , Reference Values , Statistics as Topic , Up-Regulation/genetics
2.
Mol Psychiatry ; 21(3): 376-85, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26077692

ABSTRACT

The Stanley Neuropathology Consortium Integrative Database (SNCID, http://sncid.stanleyresearch.org) is a data-mining tool that includes 379 neuropathology data sets from hippocampus, as well as RNA-Seq data measured in 15 well-matched cases in each of four groups: schizophrenia, bipolar disorder (BPD), major depression (MD) and unaffected controls. We analyzed the neuropathology data from the hippocampus to identify those abnormalities that are shared between psychiatric disorders and those that are specific to each disorder. Of the 379 data sets, 20 of them showed a significant abnormality in at least one disorder as compared with unaffected controls. GABAergic markers and synaptic proteins were mainly abnormal in schizophrenia and the two mood disorders, respectively. Two immune/inflammation-related co-expression modules built from RNA-seq data from both schizophrenia and controls combined were associated with disease status, as well as negatively correlated with the GABAergic markers. The correlation between immune-related modules and schizophrenia was replicated using microarray data from an independent tissue collection. Immune/inflammation-related co-expression modules were also built from RNA-seq data from BPD cases or from MD cases but were not preserved when using data from control cases. Moreover, there was no overlap in the genes that comprise the immune/inflammation response-related modules across the different disorders. Thus, there appears to be differential activation of the immune/inflammatory response, as determined by co-expression of genes, which is associated with the major psychiatric disorders and which is also associated with the abnormal neuropathology in the disorders.


Subject(s)
Gene Regulatory Networks/genetics , Hippocampus/metabolism , Mental Disorders/immunology , Mental Disorders/pathology , Mood Disorders/pathology , Analysis of Variance , Case-Control Studies , Databases, Factual , Female , Gene Expression Regulation/physiology , Glutamate Decarboxylase/metabolism , Humans , Male , Mood Disorders/immunology , Parvalbumins/metabolism , RNA, Messenger/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Retrospective Studies , gamma-Aminobutyric Acid/genetics , gamma-Aminobutyric Acid/metabolism
3.
Transl Psychiatry ; 4: e389, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24802307

ABSTRACT

Abnormalities in brain-derived neurotrophic factor (BDNF)/trkB signaling have been implicated in the etiology of schizophrenia and mood disorders. Patients with schizophrenia, bipolar disorder (BPD) and major depression (MDD) have reduced levels of neurotrophins in their brains when compared with normal unaffected individuals; however, only a few brain areas have been examined to date. Owing to the broad range of symptoms manifested in these disorders, we hypothesized that multiple associative areas of the neocortex may be implicated and that the degree of change in BDNF and trkB-TK+ mRNA expression and the cortical region or layers involved may vary according to Diagnostic and Statistical Manual of Mental Disorders (DSM) diagnosis. We compared BDNF and trkB-TK+ mRNA levels across all layers of the prefrontal cortex (dorsolateral prefrontal cortex, DLPFC), orbital frontal cortex (OFC), anterior cingulate cortex (ACC), inferior temporal gyrus (ITG) and superior temporal gyrus (STG) in four groups: schizophrenia, BPD, MDD and unaffected controls (n=60). BDNF mRNA levels were significantly decreased in layers IV and V of DLPFC in schizophrenia patients, in layer VI of ACC in schizophrenia and MDD and in layer VI of ITG in schizophrenia, BPD and MDD. BDNF mRNA levels were also significantly decreased in layer V and/or VI of STG in schizophrenia, BPD and MDD. TrkB-TK+ mRNA levels were only significantly decreased in the cortical layer VI of OFC in BPD. The shared and distinct patterns of neurotrophin transcript reductions, with some specific to each group, may compromise the function and plasticity of distinct cortical areas to various degrees in the different groups and contribute to the range and overlap of symptoms manifested across the diagnoses.


Subject(s)
Bipolar Disorder/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Depressive Disorder, Major/metabolism , Frontal Lobe/metabolism , Gyrus Cinguli/metabolism , Membrane Glycoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Schizophrenia/metabolism , Temporal Lobe/metabolism , Adult , Female , Frontal Lobe/pathology , Gyrus Cinguli/pathology , Humans , Male , Middle Aged , RNA, Messenger/metabolism , Receptor, trkB , Temporal Lobe/pathology
4.
Transl Psychiatry ; 4: e365, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24569695

ABSTRACT

Schizophrenia and bipolar disorder share a number of common features, both symptomatically and biologically. Abnormalities in the neuroimmune and the stress-signaling pathways have been previously identified in brains of individuals with both diseases. However, the possible relationship between abnormalities in stress and neuroimmune signaling within the cortex of people with psychotic illness has not been defined. To test the hypothesis that combined alterations in brain stress responsiveness and neuroimmune/inflammatory status are characteristic of some individuals suffering from major mental illness, we examined gene expression in the Stanley Array Cohort of 35 controls, 35 individuals with schizophrenia and 34 individuals with bipolar disorder. We used levels of 8 inflammatory-related transcripts, of which SERPINA3 was significantly elevated in individuals with schizophrenia (F(2,88)=4.137, P<0.05), and 12 glucocorticoid receptor signaling (stress) pathway transcripts previously examined, to identify two clusters of individuals: a high inflammation/stress group (n=32) and a low (n=68) inflammation/stress group. The high inflammation/stress group has a significantly greater number of individuals with schizophrenia (n=15), and a trend toward having more bipolar disorder individuals (n=11), when compared with controls (n=6). Using these subgroups, we tested which microarray-assessed transcriptional changes may be associated with high inflammatory/stress groups using ingenuity analysis and found that an extended network of gene expression changes involving immune, growth factors, inhibitory signaling and cell death factors also distinguished these groups. Our work demonstrates that some of the heterogeneity in schizophrenia and bipolar disorder may be partially explained by inflammation/stress interactions, and that this biological subtype cuts across Diagnostic and Statistical Manual of Mental Disorders (DSM)-defined categories.


Subject(s)
Bipolar Disorder/immunology , Cerebral Cortex/metabolism , Gene Expression , Inflammation/immunology , Schizophrenia/immunology , Stress, Psychological/immunology , Adult , Biomarkers/metabolism , Bipolar Disorder/classification , Cohort Studies , Female , Humans , Male , Middle Aged , Schizophrenia/classification , Young Adult
5.
Transl Psychiatry ; 3: e321, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24169640

ABSTRACT

Whole-genome expression profiling in postmortem brain tissue has recently provided insight into the pathophysiology of schizophrenia. Previous microarray and RNA-Seq studies identified several biological processes including synaptic function, mitochondrial function and immune/inflammation response as altered in the cortex of subjects with schizophrenia. Now using RNA-Seq data from the hippocampus, we have identified 144 differentially expressed genes in schizophrenia cases as compared with unaffected controls. Immune/inflammation response was the main biological process over-represented in these genes. The upregulation of several of these genes, IFITM1, IFITM2, IFITM3, APOL1 (Apolipoprotein L1), ADORA2A (adenosine receptor 2A), IGFBP4 and CD163 were validated in the schizophrenia subjects using data from the SNCID database and with quantitative RT-PCR. We identified a co-expression module associated with schizophrenia that includes the majority of differentially expressed genes related to immune/inflammation response as well as with the density of parvalbumin-containing neurons in the hippocampus. The results indicate that abnormal immune/inflammation response in the hippocampus may underlie the pathophysiology of schizophrenia and may be associated with abnormalities in the parvalbumin-containing neurons that lead to the cognitive deficits of the disease.


Subject(s)
Hippocampus/immunology , RNA, Messenger/analysis , Schizophrenia/immunology , Adult , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/immunology , Apolipoprotein L1 , Apolipoproteins/genetics , Apolipoproteins/immunology , Case-Control Studies , Female , Gene Expression Profiling , Hippocampus/metabolism , Humans , Inflammation/genetics , Insulin-Like Growth Factor Binding Protein 4/genetics , Insulin-Like Growth Factor Binding Protein 4/immunology , Lipoproteins, HDL/genetics , Lipoproteins, HDL/immunology , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Middle Aged , Oligonucleotide Array Sequence Analysis , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Reverse Transcriptase Polymerase Chain Reaction , Schizophrenia/genetics , Up-Regulation
6.
Transl Psychiatry ; 2: e87, 2012 Mar 13.
Article in English | MEDLINE | ID: mdl-22832852

ABSTRACT

Major depressive disorder (MDD) is a leading cause of disability worldwide and results tragically in the loss of almost one million lives in Western societies every year. This is due to poor understanding of the disease pathophysiology and lack of empirical medical tests for accurate diagnosis or for guiding antidepressant treatment strategies. Here, we have used shotgun proteomics in the analysis of post-mortem dorsolateral prefrontal cortex brain tissue from 24 MDD patients and 12 matched controls. Brain proteomes were pre-fractionated by gel electrophoresis and further analyzed by shotgun data-independent label-free liquid chromatography-mass spectrometry. This led to identification of distinct proteome fingerprints between MDD and control subjects. Some of these differences were validated by Western blot or selected reaction monitoring mass spectrometry. This included proteins associated with energy metabolism and synaptic function and we also found changes in the histidine triad nucleotide-binding protein 1 (HINT1), which has been implicated recently in regulation of mood and behavior. We also found differential proteome profiles in MDD with (n=11) and without (n=12) psychosis. Interestingly, the psychosis fingerprint showed a marked overlap to changes seen in the brain proteome of schizophrenia patients. These findings suggest that it may be possible to contribute to the disease understanding by distinguishing different subtypes of MDD based on distinct brain proteomic profiles.


Subject(s)
Affective Disorders, Psychotic/genetics , Depressive Disorder, Major/genetics , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Proteomics , Transcriptome/genetics , Adult , Affective Disorders, Psychotic/pathology , Depressive Disorder, Major/pathology , Female , Humans , Male , Mass Spectrometry , Middle Aged , Nerve Tissue Proteins/genetics , Peptide Mapping , Schizophrenia/genetics , Schizophrenia/pathology
7.
Transl Psychiatry ; 2: e113, 2012 May 08.
Article in English | MEDLINE | ID: mdl-22832957

ABSTRACT

Identifying the genetic cis associations between DNA variants (single-nucleotide polymorphisms (SNPs)) and gene expression in brain tissue may be a promising approach to find functionally relevant pathways that contribute to the etiology of psychiatric disorders. In this study, we examined the association between genetic variations and gene expression in prefrontal cortex, hippocampus, temporal cortex, thalamus and cerebellum in subjects with psychiatric disorders and in normal controls. We identified cis associations between 648 transcripts and 6725 SNPs in the various brain regions. Several SNPs showed brain regional-specific associations. The expression level of only one gene, PDE4DIP, was associated with a SNP, rs12124527, in all the brain regions tested here. From our data, we generated a list of brain cis expression quantitative trait loci (eQTL) genes that we compared with a list of schizophrenia candidate genes downloaded from the Schizophrenia Forum (SZgene) database (http://www.szgene.org/). Of the SZgene candidate genes, we found that the expression levels of four genes, HTR2A, PLXNA2, SRR and TCF4, were significantly associated with cis SNPs in at least one brain region tested. One gene, SRR, was also involved in a coexpression module that we found to be associated with disease status. In addition, a substantial number of cis eQTL genes were also involved in the module, suggesting eQTL analysis of brain tissue may identify more reliable susceptibility genes for schizophrenia than case-control genetic association analyses. In an attempt to facilitate the identification of genetic variations that may underlie the etiology of major psychiatric disorders, we have integrated the brain eQTL results into a public and online database, Stanley Neuropathology Consortium Integrative Database (SNCID; http://sncid.stanleyresearch.org).


Subject(s)
Alleles , Bipolar Disorder/genetics , Brain/metabolism , Depressive Disorder, Major/genetics , Gene Expression/genetics , Genetic Association Studies , Genetic Predisposition to Disease/genetics , Muscle Proteins/genetics , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics , Schizophrenia/genetics , Adaptor Proteins, Signal Transducing , Bipolar Disorder/pathology , Brain/pathology , Cytoskeletal Proteins , Depressive Disorder, Major/pathology , Humans , Oligonucleotide Array Sequence Analysis , Schizophrenia/pathology
8.
Med Phys ; 39(6Part14): 3777, 2012 Jun.
Article in English | MEDLINE | ID: mdl-28517269

ABSTRACT

PURPOSE: We introduced the concept of Dynamic Modulated Brachytherapy (DMBT) for rectal cancer, last year. To continue our work, we studied different shield designs and investigated the system's tolerance against systematic setup errors. METHODS: As previously presented, our system uses a cylindrical tungsten shield to create a directional radiation profile, which is modulated through translation and rotation using a specialized robotic arm. We used Monte Carlo simulations and an in-house gradient projection optimization algorithm to look at key design parameters. First, we used ideal phantoms to study treatment quality from shield radii ranging 0.5-1.5 cm in 0.25 cm increments. Second, using 36 patient plans, the dependence on radial source position within the shield was studied. We also analyzed the tolerance of the system to systematic setup errors by simulating dose distributions from possible inaccuracies. These included translational and rotational errors as well as possible Ir-192 source misplacements by the afterloading system. RESULTS: Changes in shield radius followed steady patterns. Increasing the radius showed a consistent increase in dose conformality to the tumor volume and better sparing to surrounding tissues. However, there was also a linear increase in total dwell time. There was a trade off to changing the radial position of the source. As the source is brought away from the center, there is a decrease in conformality to the tumor volume, but sparing to healthy tissues was increased and there is a decrease in total dwell time. We found that any potential setup errors for our system, within anticipated margins, had negligible effects on the dose distributions (< 3% deviation). CONCLUSION: Various parameters for shield designs must be balanced for an effective DMBT application. It was found that the system is highly robust against systematic setup uncertainties.

9.
Mol Psychiatry ; 16(5): 504-15, 2011 May.
Article in English | MEDLINE | ID: mdl-20308989

ABSTRACT

The glucocorticoid receptor (GR) has a critical role in determining the brain's capacity to respond to stress, and has been implicated in the pathogenesis of psychiatric illness. We hypothesized that key changes in cortical GR occur during adolescence and young adulthood, at a time when individuals are at increased risk of developing schizophrenia, bipolar disorder and major depression. We investigated the mRNA and protein expression of GR in the dorsolateral prefrontal cortex across seven developmental time points from infancy to adulthood. GR mRNA expression, determined by microarray and quantitative real-time PCR, was lowest in neonates and peaked around young adulthood. Western blotting revealed two dynamic patterns of GRα protein expression across the lifespan, with N-terminal variants displaying differing unique patterns of abundance. GRα-A and a 67-kDa GRα isoform mirrored mRNA trends and peaked in toddlers and late in adolescence, whereas a 40-kDa isoform, very likely a GRα-D variant, peaked in neonates and decreased across the lifespan. GRα protein was localized to pyramidal neurons throughout life and most strikingly in young adulthood, but to white matter astrocytes only in neonates and infants (<130 days). These results suggest that the neonatal and late adolescent periods represent critical windows of stress pathway development, and highlight the importance of white matter astrocytes and pyramidal neurons, respectively, at these stages of cortical development. Evidence of dynamic patterns of GR isoform expression and cellular localization across development strengthens the hypothesis that windows of vulnerability to stress exist across human cortical development.


Subject(s)
Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Gene Expression Regulation, Developmental/physiology , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Adolescent , Adult , Child , Child, Preschool , Female , Gene Expression Profiling , Glial Fibrillary Acidic Protein/metabolism , Humans , Infant , Male , Middle Aged , Molecular Weight , Nonlinear Dynamics , Oligonucleotide Array Sequence Analysis , Phosphopyruvate Hydratase/metabolism , RNA, Messenger/metabolism , Young Adult
10.
Mol Psychiatry ; 16(4): 452-61, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20308991

ABSTRACT

Cytoarchitectural abnormalities have been described in the prefrontal cortex (PFC) of subjects with psychiatric disorders. We explored the possible genetic causalities that may underlie the cytoarchitectural abnormalities of calbindin-containing γ-aminobutyric acid (GABA)ergic neurons and perineuronal oligodendrocytes in the PFC of subjects with psychiatric disorders by converging results from genome-wide single-nucleotide polymorphism (SNP) scans for the traits and expression SNP (eSNP) associations. In the initial genome-wide scans, we identified several development- and apoptosis-related genes associated with the cytoarchitectural traits. Moreover, the susceptibility gene for bipolar disorder, PPP2R2C, was found to be associated with the number of perineuronal oligodendrocytes. Further eSNP analyses indicated that two novel candidate genes, RAB2A and SLC38A1, were associated with the density of calbindin-positive neurons and the number of perineuronal oligodendrocytes, respectively. Our findings may provide novel insights into the genetic causalities associated with cytoarchitectural abnormalities in the PFC of subjects with major psychiatric disorders as well as into the etiology of such disorders.


Subject(s)
Amino Acid Transport System A/genetics , Genetic Predisposition to Disease , Mental Disorders/genetics , Mental Disorders/pathology , Polymorphism, Single Nucleotide/genetics , Prefrontal Cortex/pathology , S100 Calcium Binding Protein G/genetics , Adult , Calbindins , Databases, Genetic , Female , Gene Expression Profiling , Genome-Wide Association Study , Genotype , Humans , Male , Middle Aged , Oligodendroglia/pathology , Oligonucleotide Array Sequence Analysis , Protein Phosphatase 2/genetics , gamma-Aminobutyric Acid/genetics , rab2 GTP-Binding Protein/genetics
11.
Mol Psychiatry ; 15(3): 326-36, 2010 Mar.
Article in English | MEDLINE | ID: mdl-18762803

ABSTRACT

Cytoarchitectural abnormalities have been described in the prefrontal cortex of subjects with schizophrenia, bipolar disorder and depression. However, little is known about the gene expression profiles associated with these abnormalities. Genome-wide expression profiling technology provides an unbiased approach to identifying candidate genes and biological processes that may be associated with complex biological traits such as cytoarchitecture. In this study, we explored expression profiles associated with the abnormalities by using publicly available microarray metadata and cytoarchitectural data from post-mortem samples of the frontal cortex from 54 subjects (schizophrenia, n=14; bipolar disorder, n=13; depression, n=12 and controls n=15). Correlation analysis between genome-wide expression levels and cytoarchitectural traits revealed that 818 genes were significantly correlated with a decrease in the number of perineuronal oligodendrocytes across all subjects. A total of 600 genes were significantly correlated with a decrease in density of calbindin-positive interneurons across all subjects. Multiple biological processes including cellular metabolism, central nervous system development, cell motility and programmed cell death were significantly overrepresented in both correlated gene lists. These findings may provide novel insights into the molecular mechanisms that underlie the cytoarchitectural abnormalities of perineuronal oligodendrocytes and calbindin-containing GABAergic interneurons in the prefrontal cortex of the major psychiatric disorders.


Subject(s)
Gene Expression Profiling/methods , Genome-Wide Association Study/methods , Mental Disorders/genetics , Mental Disorders/pathology , Biological Phenomena/genetics , Bipolar Disorder/genetics , Bipolar Disorder/pathology , Depression/genetics , Depression/pathology , Humans , Interneurons/metabolism , Interneurons/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Oligonucleotide Array Sequence Analysis/methods , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Schizophrenia/genetics , Schizophrenia/pathology
13.
Neuroscience ; 153(4): 1126-34, 2008 Jun 02.
Article in English | MEDLINE | ID: mdl-18440154

ABSTRACT

Posttraumatic stress disorder (PTSD) is one of the most common psychiatric disorders. Despite the extensive study of the neurobiological correlates of this disorder, the underlying mechanisms of PTSD are still poorly understood. Recently, a study demonstrated that dexamethasone (Dex), a synthetic glucocorticoid, can up-regulate p11, known as S100A10-protein which is down-regulated in patients with depression, (Yao et al., 1999; Huang et al., 2003) a common comorbid disorder in PTSD. These observations led to our hypothesis that traumatic stress may alter expression of p11 mediated through a glucocorticoid receptor. Here, we demonstrate that inescapable tail shock increased both prefrontal cortical p11 mRNA levels and plasma corticosterone levels in rats. We also found that Dex up-regulated p11 expression in SH-SY5Y cells through glucocorticoid response elements (GREs) within the p11 promoter. This response was attenuated by either RU486, a glucocorticoid receptor (GR) antagonist or mutating two of three glucocorticoid response elements (GRE2 and GRE3) in the p11 promoter. Finally, we showed that p11 mRNA levels were increased in postmortem prefrontal cortical tissue (area 46) of patients with PTSD. The data obtained from our work in a rat model of inescapable tail shock, a p11-transfected cell line and postmortem brain tissue from PTSD patients outline a possible mechanism by which p11 is regulated by glucocorticoids elevated by traumatic stress.


Subject(s)
Annexin A2/metabolism , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Nuclear Proteins/metabolism , Promoter Regions, Genetic/drug effects , Prosencephalon/metabolism , S100 Proteins/metabolism , Stress, Psychological/pathology , Up-Regulation/drug effects , Animals , Animals, Newborn , Annexin A2/genetics , Cells, Cultured , Chromatin Immunoprecipitation/methods , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Electroshock/adverse effects , Hormone Antagonists/pharmacology , Humans , Male , Mifepristone/pharmacology , Nuclear Proteins/genetics , Prosencephalon/cytology , Prosencephalon/drug effects , Rats , Rats, Sprague-Dawley , S100 Proteins/genetics , Stress, Psychological/etiology , Time Factors , Up-Regulation/physiology
14.
Neuroscience ; 144(3): 1109-19, 2007 Feb 09.
Article in English | MEDLINE | ID: mdl-17123740

ABSTRACT

Dopamine in the prefrontal cortex plays a critical role in normal cognition throughout the lifespan and has been implicated in the pathophysiology of neuropsychiatric disorders such as schizophrenia and attention deficit disorder. Little is known, however, about the postnatal development of the dopaminergic system in the human prefrontal cortex. In this study, we examined pre- and post-synaptic markers of the dopaminergic system in postmortem tissue specimens from 37 individuals ranging in age from 2 months to 86 years. We measured the levels of tyrosine hydroxylase, the rate limiting enzyme in dopamine biosynthesis, using Western immunoblotting. We also examined the gene expression of the three most abundant dopamine receptors (DARs) in the human prefrontal cortex: DAR1, DAR2 and DAR4, by in situ hybridization. We found that tyrosine hydroxylase concentrations and DAR2 mRNA levels were highest in the cortex of neonates. In contrast, the gene expression of DAR1 was highest in adolescents and young adults. No significant changes across age groups were detected in mRNA levels of DAR4. Both DAR1 and DAR2 mRNA were significantly lower in the aged cortex. Taken together, our data suggest dynamic changes in markers of the dopamine system in the human frontal cortex during postnatal development at both pre-and post-synaptic sites. The peak in DAR1 mRNA levels around adolescence/early adulthood may be of particular relevance to neuropsychiatric disorders such as schizophrenia in which symptoms manifest during the same developmental period.


Subject(s)
Aging/physiology , Dopamine/metabolism , Neurons/metabolism , Prefrontal Cortex/growth & development , Prefrontal Cortex/metabolism , Receptors, Dopamine/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , Cell Division/physiology , Down-Regulation/physiology , Female , Gene Expression Regulation, Developmental/genetics , Humans , Infant , Infant, Newborn , Male , Neurons/cytology , Prefrontal Cortex/cytology , RNA, Messenger/metabolism , Schizophrenia/metabolism , Schizophrenia/physiopathology , Synaptic Transmission/physiology , Tyrosine 3-Monooxygenase/metabolism
15.
Mol Psychiatry ; 11(10): 965-78, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16894394

ABSTRACT

Bipolar affective disorder is a severe psychiatric disorder with a strong genetic component but unknown pathophysiology. We used microarray technology to determine the expression of approximately 22,000 mRNA transcripts in post-mortem tissue from two brain regions in patients with bipolar disorder and matched healthy controls. Dorsolateral prefrontal cortex tissue from a cohort of 70 subjects and orbitofrontal cortex tissue from a separate cohort of 30 subjects was investigated. The final analysis included 30 bipolar and 31 control subjects for the dorsolateral prefrontal cortex and 10 bipolar and 11 control subjects for the orbitofrontal cortex. Differences between disease and control groups were identified using a rigorous statistical analysis with correction for confounding variables and multiple testing. In the orbitofrontal cortex, 393 differentially expressed transcripts were identified by microarray analysis and a representative subset was validated by quantitative real-time PCR. Pathway analysis revealed significant upregulation of genes involved in G-protein coupled receptor signalling and response to stimulus (in particular the immune response), while genes relating to the ubiquitin cycle and intracellular transport showed coordinated downregulation in bipolar disorder. Additionally, several genes involved in synaptic function were significantly downregulated in bipolar disorder. No significant changes in gene expression were observed in the dorsolateral prefrontal cortex using microarray analysis or quantitative real-time PCR. Our findings implicate the orbitofrontal cortex as a region prominently involved in bipolar disorder and indicate that diverse processes are affected. Overall, our results suggest that dysregulation of the ubiquitin pathway and synaptic function may be central to the disease process.


Subject(s)
Bipolar Disorder/genetics , Cerebral Cortex/metabolism , Protein Transport/physiology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Ubiquitin/metabolism , Adult , Bipolar Disorder/metabolism , Female , Frontal Lobe/metabolism , Gene Expression Profiling , Gene Expression Regulation , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Prefrontal Cortex/metabolism , Protein Transport/genetics , RNA, Messenger/analysis , Receptors, G-Protein-Coupled/genetics , Reference Values , Signal Transduction/genetics , Synaptic Transmission , Ubiquitin/genetics
16.
Gene Expr Patterns ; 6(8): 941-51, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16713371

ABSTRACT

Brain-derived neurotrophic factor (BDNF) and its receptor tyrosine kinase B (trkB) influence neuronal survival, differentiation, synaptogenesis, and maintenance. Using in situ hybridization we examined the spatial and temporal expression of mRNAs encoding these proteins during diverse stages of life in the human hippocampus and inferior temporal cortex. We examined six postnatal time points: neonatal (1-3 months), infant (4-12 months), adolescent (14-18 years), young adult (20-24 years), adult (34-43 years), and aged (68-86 years). Within the hippocampus, levels of BDNF mRNA did not change significantly with age. However, levels of both the full-length form of trkB (trkB TK+) mRNA and the truncated form of trkB (trkB TK-) decreased over the life span (p < 0.05). In the temporal cortex, BDNF and trkB TK+ mRNA levels were highest in neonates and decreased with age (r = -0.4 and r = -0.7, respectively, both p < 0.05). In contrast, TrkB TK- mRNA levels remained constant across the life span in the temporal cortex. The peak in both BDNF and trkB TK+ mRNA expression in the neonate temporal cortex differs from that previously described for the frontal cortex where both mRNAs peak in expression during young adulthood. The increase in BDNF and trkB TK+ mRNA in the temporal cortex of the neonate suggests that neurotrophin signaling is important in the early development of the temporal cortex. In addition, since BDNF and both forms of its high affinity receptor are expressed throughout the development, maturation, and aging of the human hippocampus and surrounding neocortex they are likely to play roles not only in early growth but also in maintenance of neurons throughout life.


Subject(s)
Aging/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/metabolism , Hippocampus/metabolism , Receptor, trkB/metabolism , Temporal Lobe/metabolism , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Autopsy , Cerebral Cortex/anatomy & histology , Cerebral Cortex/growth & development , Female , Gene Expression Profiling , Hippocampus/anatomy & histology , Hippocampus/growth & development , Humans , Infant , Infant, Newborn , Male , RNA, Messenger/metabolism , Temporal Lobe/anatomy & histology , Temporal Lobe/growth & development , Tissue Distribution
17.
Neuroscience ; 134(1): 81-95, 2005.
Article in English | MEDLINE | ID: mdl-15964702

ABSTRACT

Although estrogen receptor alpha (ERalpha) mRNA has been detected in the primate frontal cortex, the types of ERalpha transcripts expressed, including exon-deleted variants (Delta), have not been determined in the monkey or human frontal cortex. Because the types of ERalpha mRNA expressed in brain could define neuronal responses to estrogens, we examined the transcript pool of ERalpha mRNAs expressed in normal adult and developing human and macaque frontal cortex. We reverse transcribed total RNA from the postmortem frontal cortex of 29 normal adult humans, 12 rhesus macaques, and 19 people ranging from infants to adults and employed two rounds of nested polymerase chain reaction (PCR) to generate ERalpha products spanning the coding domain. In a third nested PCR, we used primers specific for novel sequences of exon-exon junctions created when whole exons are missing. By sequencing PCR products, we detected 60 instances of 12 distinct DeltaERalpha mRNAs in adult humans and 94 instances of 13 distinct DeltaERalpha mRNAs in monkeys in differing patterns from one individual to another. In adult humans, 83% of individuals expressed at least 1 DeltaERalpha mRNA variant, and 100% of the monkeys expressed at least 1 DeltaERalpha mRNA variant. The single Delta2, Delta5, and Delta7 variants were frequently expressed in both human and monkey frontal cortex, Delta3 variants were rare in both species, and Delta6 variants were more frequently expressed in monkeys. In both species, we detected double, triple and quadruple Deltas, but these were less common than single Deltas. The pattern of human variant expression did not appear to change dramatically as a function of age. These findings imply the potential to produce different ERalpha proteins in frontal cortex, possibly with altered structure and function which may have physiological relevance for gene transcription by virtue of altered functional interactions with each other, other steroid hormone receptors, and genomic DNA.


Subject(s)
Estrogen Receptor alpha/genetics , Exons/genetics , Frontal Lobe/metabolism , Gene Deletion , Gene Expression Regulation, Developmental/physiology , Adolescent , Adult , Age Factors , Animals , Blotting, Northern , Child , Child, Preschool , Chromosomes, Human, Pair 6 , Estrogen Receptor alpha/metabolism , Female , Gene Expression/physiology , Humans , Infant , Macaca mulatta , Male , Middle Aged , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Sex Factors
18.
Neuroscience ; 133(2): 453-61, 2005.
Article in English | MEDLINE | ID: mdl-15885920

ABSTRACT

Recent studies have shown a decrease in glial number and glial fibrillary acidic protein (GFAP) levels in the frontal and cingulate cortices of individuals with mood disorders and schizophrenia. In an attempt to verify and expand these findings we examined GFAP messenger ribonucleic acid (mRNA) levels in postmortem sections of the anterior cingulate cortex (ACC) from the Stanley Neuropathology Consortium (SNC). The consortium consists of 15 cases in each of four groups (schizophrenia, bipolar disorder, non-psychotic depression and unaffected controls). By in situ hybridization, we found higher levels of GFAP mRNA in white matter and at the pial surface as compared with gray matter levels in all cases. In the white matter of ACC we detected a significant effect of diagnosis (P<0.04) with GFAP mRNA levels decreased in individuals with schizophrenia and bipolar disorder as compared with normal controls. In the gray matter there was a significant effect of layer (P<0.01) with the highest levels of GFAP mRNA in layer VI in all groups. As in the white matter, the mean GFAP mRNA levels were decreased in individuals with schizophrenia and bipolar disorder as compared with the unaffected controls, however the difference failed to reach statistical significance. Thus, astrocytes positive for GFAP may contribute to the decrease in glial density previously described in subjects with major mental illness, however the relative contribution of astrocytes may vary with diagnosis.


Subject(s)
Bipolar Disorder/metabolism , Depression/metabolism , Glial Fibrillary Acidic Protein/metabolism , Gyrus Cinguli/metabolism , Schizophrenia/metabolism , Adult , Analysis of Variance , Area Under Curve , Autoradiography/methods , Bipolar Disorder/complications , Bipolar Disorder/genetics , Depression/complications , Depression/genetics , Diagnostic Imaging , Female , Glial Fibrillary Acidic Protein/genetics , Humans , In Situ Hybridization/methods , Male , Middle Aged , Postmortem Changes , RNA, Messenger/metabolism , Schizophrenia/complications , Schizophrenia/genetics
19.
Mol Psychiatry ; 9(7): 684-97, 643, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15098003

ABSTRACT

The etiology and pathophysiology of schizophrenia remain unknown. A parallel transcriptomics, proteomics and metabolomics approach was employed on human brain tissue to explore the molecular disease signatures. Almost half the altered proteins identified by proteomics were associated with mitochondrial function and oxidative stress responses. This was mirrored by transcriptional and metabolite perturbations. Cluster analysis of transcriptional alterations showed that genes related to energy metabolism and oxidative stress differentiated almost 90% of schizophrenia patients from controls, while confounding drug effects could be ruled out. We propose that oxidative stress and the ensuing cellular adaptations are linked to the schizophrenia disease process and hope that this new disease concept may advance the approach to treatment, diagnosis and disease prevention of schizophrenia and related syndromes.


Subject(s)
Brain/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Schizophrenia/genetics , Schizophrenia/metabolism , Fatty Acids/metabolism , Genome, Human , Glucose/metabolism , Humans , Hypoxia, Brain/etiology , Hypoxia, Brain/genetics , Hypoxia, Brain/metabolism , Mitochondrial Diseases/complications , Oligonucleotide Array Sequence Analysis , Oxidative Phosphorylation , Oxidative Stress , Proteomics , Schizophrenia/etiology , Signal Transduction
20.
J Anim Sci ; 82(1): 250-61, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14753369

ABSTRACT

Four experiments with 1,040 weanling pigs (17 +/- 2 d of age at weaning) were conducted to evaluate the effects of spray-dried animal plasma source, drying technique, and methods of bacterial reduction on nursery pig performance. In Exp. 1, 180 barrows and gilts (initial BW 5.9 +/- 1.8 kg) were used to compare effects of animal plasma, animal plasma source, drying technique (spray-dried or freeze-dried), and plasma irradiation in nursery pig diets. From d 0 to 10, pigs fed diets containing irradiated spray-dried animal plasma had increased ADG and ADFI (P < 0.05) compared with pigs fed diets containing nonirradiated spray-dried animal plasma. Pigs fed irradiated animal plasma Sources 1 and 2 were similar in ADG and ADFI, but pigs fed animal plasma Source 1 had greater ADG (P < 0.05) than pigs fed animal plasma Source 2 and pigs not fed plasma. Pigs fed freeze-dried animal plasma had growth performance similar (P > 0.36) to pigs fed spray-dried animal plasma. Overall (d 0 to 24), pigs fed irradiated spray-dried animal plasma were heavier (P < 0.05) than pigs fed no animal plasma, whereas pigs fed nonirradiated spray-dried plasma were intermediate. In Exp. 2, 325 barrows and gilts (initial BW 5.8 +/- 1.7 kg) were used to compare the effects of irradiation or formaldehyde treatment of animal plasma and formaldehyde treatment of the whole diet. Pigs fed diets containing irradiated animal plasma had greater ADG (P < 0.05) than pigs fed nonirradiated plasma. Pigs fed formaldehyde-treated plasma had greater ADG and ADFI (P < 0.05) than pigs fed diets with either nonirradiated plasma or whole diet treated with formaldehyde. In Exp. 3 (360 barrows and gilts; initial BW 6.3 +/- 2.7 kg) and Exp. 4 (175 barrows and gilts; initial BW 6.1 +/- 1.7 kg), the irradiation of feed (high bacteria) and food-grade (low bacteria) animal plasma in nursery pig diets was examined. Pigs fed irradiated feed-grade plasma Product 2 had increased ADG (P < 0.05) compared with pigs fed nonirradiated plasma Product 2 and pigs fed the control diet without plasma. In Exp. 3 and 4, pigs fed irradiated food-grade plasma had growth performance similar to pigs fed nonirradiated food-grade plasma (P > 0.12). These studies indicate that bacterial reduction of feed-grade, but not food-grade animal plasma, improves nursery pig performance.


Subject(s)
Animal Feed/radiation effects , Food Irradiation , Formaldehyde/pharmacology , Plasma/microbiology , Swine/growth & development , Animal Feed/microbiology , Animal Nutritional Physiological Phenomena , Animals , Female , Gamma Rays , Male , Random Allocation , Weaning , Weight Gain
SELECTION OF CITATIONS
SEARCH DETAIL
...