Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 50(4): e19, 2022 02 28.
Article in English | MEDLINE | ID: mdl-34850115

ABSTRACT

Accurately quantifying gene and isoform expression changes is essential to understanding cell functions, differentiation and disease. Sequencing full-length native RNAs using long-read direct RNA sequencing (DRS) has the potential to overcome many limitations of short and long-read sequencing methods that require RNA fragmentation, cDNA synthesis or PCR. However, there are a lack of tools specifically designed for DRS and its ability to identify differential expression in complex organisms is poorly characterised. We developed NanoCount for fast, accurate transcript isoform quantification in DRS and demonstrate it outperforms similar methods. Using synthetic controls and human SH-SY5Y cell differentiation into neuron-like cells, we show that DRS accurately quantifies RNA expression and identifies differential expression of genes and isoforms. Differential expression of 231 genes, 333 isoforms, plus 27 isoform switches were detected between undifferentiated and differentiated SH-SY5Y cells and samples clustered by differentiation state at the gene and isoform level. Genes upregulated in neuron-like cells were associated with neurogenesis. NanoCount quantification of thousands of novel isoforms discovered with DRS likewise enabled identification of their differential expression. Our results demonstrate enhanced DRS isoform quantification with NanoCount and establish the ability of DRS to identify biologically relevant differential expression of genes and isoforms.


Subject(s)
Nanopore Sequencing , Nanopores , Gene Expression Profiling/methods , Humans , Protein Isoforms/genetics , RNA/genetics , Sequence Analysis, RNA/methods , Transcriptome
2.
J Psychopharmacol ; 31(12): 1519-1526, 2017 12.
Article in English | MEDLINE | ID: mdl-28655286

ABSTRACT

As well as being expressed as a full-length transcript, the group II metabotropic glutamate receptor 3 (GRM3, mGlu3) gene is expressed as an mRNA isoform which lacks exon 4 (GRM3Δ4) and which is predicted to encode a protein with a novel C terminus (called mGlu3Δ4). This variant may contribute to the mechanism by which GRM3 acts as a schizophrenia risk gene. However, little is known about the properties or function of mGlu3Δ4. Here, using transiently transfected HEK293T/17 cells, we confirm that GRM3Δ4 cDNA is translated, with mGlu3Δ4 existing as a homodimer as well as a monomer, and localizing primarily to cell membranes including the plasma membrane. Co-immunoprecipitation shows that mGlu3Δ4 interacts with canonical mGlu3. mGlu3Δ4 does not bind the mGlu2/3 antagonist [3H]LY341495, but the presence of mGlu3Δ4 reduces binding of [3H]LY341495 to mGlu3, paralleled by a decrease in the abundance of membrane-associated mGlu3. These experiments indicate that mGlu3Δ4 may negatively modulate mGlu3, and thereby impact on the roles of GRM3/mGlu3 in schizophrenia and as a therapeutic target.


Subject(s)
Receptors, Metabotropic Glutamate/classification , Receptors, Metabotropic Glutamate/metabolism , Schizophrenia/metabolism , Amino Acids/pharmacology , Cell Membrane/metabolism , HEK293 Cells , Humans , Ligands , Protein Isoforms/metabolism , Radioligand Assay , Transfection , Tritium/metabolism , Xanthenes/pharmacology
3.
Schizophr Res ; 177(1-3): 18-27, 2016 11.
Article in English | MEDLINE | ID: mdl-27130562

ABSTRACT

BACKGROUND: Metabotropic glutamate receptor 3 (mGlu3, mGluR3), encoded by GRM3, is a risk gene for schizophrenia and a therapeutic target. It is unclear whether expression of the receptor is altered in the disorder or related to GRM3 risk genotype. Antibodies used to date to assess mGlu3 in schizophrenia have not been well validated. OBJECTIVE: To characterise six commercially available anti-mGlu3 antibodies for use in human brain, and then conduct a semi-quantitative study of mGlu3 immunoreactivity in schizophrenia. METHODS: Antibodies tested using Grm3-/- and Grm2-/-/3-/- mice and transfected HEK293T/17 cells. Western blotting on membrane protein isolated from superior temporal cortex of 70 patients with schizophrenia and 87 healthy comparison subjects, genotyped for GRM3 SNP rs10234440. RESULTS: One (out of six) anti-mGlu3 antibodies was fully validated, a C-terminal antibody which detected monomeric (~100kDa) and dimeric (~200kDa) mGlu3. A second, N-terminal, antibody detected the 200kDa band but also produced non-specific bands. Using the C-terminal antibody for western blotting in human brain, mGlu3 immunoreactivity was found to decline with age, and was affected by pH and post mortem interval. There were no differences in monomeric or dimeric mGlu3 immunoreactivity in schizophrenia or in relation to GRM3 genotype. The antibody was not suitable for immunohistochemistry. INTERPRETATION: These data highlight the value of knockout mouse tissue for antibody validation, and the need for careful antibody characterisation. The schizophrenia data show that involvement of GRM3 in the disorder and its genetic risk architecture is not reflected in total membrane mGlu3 immunoreactivity in superior temporal cortex.


Subject(s)
Antibodies , Receptors, Metabotropic Glutamate/immunology , Receptors, Metabotropic Glutamate/metabolism , Schizophrenia/metabolism , Adult , Aged , Aged, 80 and over , Animals , Blotting, Western , Brain Chemistry , Cell Membrane/metabolism , Cell Membrane/pathology , Female , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Immunohistochemistry , Male , Mice, Knockout , Middle Aged , Polymorphism, Single Nucleotide , Receptors, Metabotropic Glutamate/deficiency , Receptors, Metabotropic Glutamate/genetics , Schizophrenia/genetics , Schizophrenia/pathology , Temporal Lobe/metabolism , Temporal Lobe/pathology , Time Factors , Young Adult
4.
Psychoneuroendocrinology ; 60: 163-72, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26150358

ABSTRACT

PURPOSE: Prenatal depression increases risk for a number of adverse offspring outcomes, however the biological mechanisms underlying this association remain unclear. It has been suggested that maternal glucocorticoids may mediate this link, though supporting evidence has been mixed. An alternative mechanism of effect may be via depression-induced changes in maternal sympathetic nervous system (SNS) function. We examined this hypothesis by determining the relationship between symptoms of maternal prenatal depression and diurnal salivary alpha-amylase (sAA) levels. METHODS: 76 pregnant women were recruited during either the second or third trimester of pregnancy. Participants self-reported depressive symptoms using the Edinburgh postnatal depression scale. Saliva samples, to be assayed for alpha-amylase activity, were collected at home over two working days. RESULTS: Participants with depressive symptoms in later pregnancy had elevated awakening sAA levels compared with non-depressed controls (t(73) = -2.737, p = 0.008), and continued to have raised sAA throughout the day (F(1) = 10.924, p = 0.002). CONCLUSIONS: Our findings highlight that symptoms of depression during late pregnancy are associated with increased maternal SNS activity. Thus, changes in maternal SNS function, which may include increased vasoconstriction and reduced foetal blood flow, could, in part, mediate associations between prenatal depression and adverse offspring outcomes.


Subject(s)
Depression/enzymology , Depression/psychology , Pregnancy Complications/enzymology , Pregnancy Complications/psychology , Salivary alpha-Amylases/metabolism , Adult , Circadian Rhythm , Female , Fetus/blood supply , Humans , Life Style , Longitudinal Studies , Pregnancy , Pregnancy Trimester, Second , Pregnancy Trimester, Third , Psychiatric Status Rating Scales , Salivary alpha-Amylases/analysis , Socioeconomic Factors , Young Adult
5.
Neuropharmacology ; 89: 19-32, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25158312

ABSTRACT

Group II metabotropic glutamate receptors (mGlu2 and mGlu3, encoded by GRM2 and GRM3) have been implicated in both cognitive and emotional processes, although their precise role remains to be established. Studies with knockout (KO) mice provide an important approach for investigating the role of specific receptor genes in behaviour. In the present series of experiments we extended our prior characterisation of GRM2/3(-/-) double KO mice and, in complementary experiments, investigated the behavioural phenotype of single GRM2(-/-) and GRM3(-/-) mice. We found no consistent effect on anxiety in either the double or single KO mice. The lack of an anxiety phenotype in any of the lines contrasts with the clear anxiolytic effects of mGlu2/3 ligands. Motor co-ordination was impaired in GRM2/3(-/-) mice, but spared in single GRM2(-/-) and GRM3(-/-) mice. Spatial working memory (rewarded alternation) testing on the elevated T-maze revealed a deficit in GRM2(-/-) mice throughout testing, whereas GRM3(-/-) mice exhibited a biphasic effect (initially impaired, but performing better than controls by the end of training). A biphasic effect on activity levels was seen for the GRM2(-/-) mice. Overall, the phenotype in both GRM2(-/-) and GRM3(-/-) mice was less pronounced - if present at all - compared to GRM2/3(-/-) mice, across the range of task domains. This is consistent with possible redundancy of function and/or compensation in the single KO lines. Results are discussed with reference to a possible role for group II metabotropic glutamate receptors at the interface between arousal and behavioural performance, according to an inverted U-shaped function.


Subject(s)
Anxiety/metabolism , Cognition/physiology , Receptors, Metabotropic Glutamate/physiology , Animals , Anxiety/psychology , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
JAMA Psychiatry ; 71(10): 1112-20, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25162540

ABSTRACT

IMPORTANCE: The single-nucleotide polymorphism rs1344706 in the zinc finger protein 804A gene (ZNF804A) shows genome-wide association with schizophrenia and bipolar disorder. Little is known regarding the expression of ZNF804A and the functionality of rs1344706. OBJECTIVES: To characterize ZNF804A expression in human brain and to investigate how it changes across the life span and how it is affected by rs1344706, schizophrenia, bipolar disorder, and major depressive disorder. DESIGN, SETTING, AND PARTICIPANTS: Molecular and immunochemical methods were used to study ZNF804A messenger RNA (mRNA) and ZNF804A protein, respectively. ZNF804A transcripts were investigated using next-generation sequencing and polymerase chain reaction-based methods, and ZNF804A protein was investigated using Western blots and immunohistochemistry. Samples of dorsolateral prefrontal cortex and inferior parietal lobe tissue were interrogated from 697 participants between 14 weeks' gestational age and age 85 years, including patients with schizophrenia, bipolar disorder, or major depressive disorder. MAIN OUTCOMES AND MEASURES: Quantitative measurements of ZNF804A mRNA and immunoreactivity, and the effect of diagnosis and rs1344706 genotype. RESULTS: ZNF804A was expressed across the life span, with highest expression prenatally. An abundant and developmentally regulated truncated ZNF804A transcript was identified, missing exons 1 and 2 (ZNF804AE3E4) and predicted to encode a protein lacking the zinc finger domain. rs1344706 influenced expression of ZNF804AE3E4 mRNA in fetal brain (P = .02). In contrast, full-length ZNF804A showed no association with genotype (P > .05). ZNF804AE3E4 mRNA expression was decreased in patients with schizophrenia (P = .006) and increased in those with major depressive disorder (P < .001), and there was a genotype-by-diagnosis interaction in bipolar disorder (P = .002). ZNF804A immunoreactivity was detected in fetal and adult human cerebral cortex. It was localized primarily to pyramidal neurons, with cytoplasmic as well as dendritic and nuclear staining. No differences in ZNF804A-immunoreactive neurons were seen in schizophrenia or related to rs1344706 (P > .05). CONCLUSIONS AND RELEVANCE: rs1344706 influences the expression of ZNF804AE3E4, a novel splice variant. The effect is limited to fetal brain and to this isoform. It may be part of the mechanism by which allelic variation in ZNF804A affects risk of psychosis. ZNF804A is translated in human brain, where its functions may extend beyond its predicted role as a transcription factor.


Subject(s)
Bipolar Disorder/genetics , Brain Chemistry , Depressive Disorder, Major/genetics , Kruppel-Like Transcription Factors/genetics , Polymorphism, Single Nucleotide/genetics , Schizophrenia/genetics , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Bipolar Disorder/metabolism , Blotting, Western , Brain/embryology , Child , Child, Preschool , Depressive Disorder, Major/metabolism , Female , Gene Expression Regulation/genetics , Genetic Predisposition to Disease/genetics , Genotype , Humans , Infant , Infant, Newborn , Kruppel-Like Transcription Factors/analysis , Kruppel-Like Transcription Factors/physiology , Male , Middle Aged , Parietal Lobe/chemistry , Prefrontal Cortex/chemistry , Protein Isoforms/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Schizophrenia/metabolism , Sex Factors , Young Adult
7.
BMC Neurosci ; 14: 102, 2013 Sep 22.
Article in English | MEDLINE | ID: mdl-24053122

ABSTRACT

BACKGROUND: Group II metabotropic glutamate receptors (mGlu2 and mGlu3, encoded by Grm2 and Grm3) have been the focus of attention as treatment targets for a number of psychiatric conditions. Double knockout mice lacking mGlu2 and mGlu3 (mGlu2/3-/-) show a subtle behavioural phenotype, being hypoactive under basal conditions and in response to amphetamine, and with a spatial memory deficit that depends on the arousal properties of the task. The neurochemical correlates of this profile are unknown. Here, we measured tissue levels of dopamine, 5-HT, noradrenaline and their metabolites in the striatum and frontal cortex of mGlu2/3-/- double knockout mice, using high performance liquid chromatography. We also measured the same parameters in mGlu2-/- and mGlu3-/- single knockout mice. RESULTS: mGlu2/3-/-mice had reduced dopamine levels in the striatum but not in frontal cortex, compared to wild-types. In a separate cohort we replicated this deficit and, using tissue punches, found it was more prominent in the nucleus accumbens than in dorsolateral striatum. Noradrenaline, 5-HT and their metabolites were not altered in the striatum of mGlu2/3-/- mice, although the noradrenaline metabolite MHPG was increased in the cortex. In mGlu2-/- and mGlu3-/- single knockout mice we found no difference in any monoamine or metabolite, in either brain region, compared to their wild-type littermates. CONCLUSIONS: Group II metabotropic glutamate receptors impact upon striatal dopamine. The effect may contribute to the behavioural phenotype of mGlu2/3-/- mice. The lack of dopaminergic alterations in mGlu2-/- and mGlu3-/- single knockout mice reveals a degree of redundancy between the two receptors. The findings support the possibility that interactions between mGlu2/3 and dopamine may be relevant to the pathophysiology and therapy of schizophrenia and other disorders.


Subject(s)
Corpus Striatum/metabolism , Dopamine/metabolism , Receptors, Metabotropic Glutamate/deficiency , Animals , Brain Chemistry , Chromatography, High Pressure Liquid , Corpus Striatum/chemistry , Dopamine/analysis , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
Neuropharmacology ; 62(3): 1164-7, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21864547

ABSTRACT

Genetic mouse models relevant to schizophrenia complement, and have to a large extent supplanted, pharmacological and lesion-based rat models. The main attraction is that they potentially have greater construct validity; however, they share the fundamental limitations of all animal models of psychiatric disorder, and must also be viewed in the context of the uncertain and complex genetic architecture of psychosis. Some of the key issues, including the choice of gene to target, the manner of its manipulation, gene-gene and gene-environment interactions, and phenotypic characterization, are briefly considered in this commentary, illustrated by the relevant papers reported in this special issue.


Subject(s)
Disease Models, Animal , Gene-Environment Interaction , Genetic Predisposition to Disease , Schizophrenia/genetics , Animals , Humans , Mice , Schizophrenia/etiology
9.
Cereb Cortex ; 22(7): 1520-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21878485

ABSTRACT

Neuregulin 1 (NRG1) is a growth factor involved in neurodevelopment and plasticity. It is a schizophrenia candidate gene, and hippocampal expression of the NRG1 type I isoform is increased in the disorder. We have studied transgenic mice overexpressing NRG1 type I (NRG1(tg-type I)) and their wild-type littermates and measured hippocampal electrophysiological and behavioral phenotypes. Young NRG1(tg-type I) mice showed normal memory performance, but in older NRG1(tg-type I) mice, hippocampus-dependent spatial working memory was selectively impaired. Hippocampal slice preparations from NRG1(tg-type I) mice exhibited a reduced frequency of carbachol-induced gamma oscillations and an increased tendency to epileptiform activity. Long-term potentiation in NRG1(tg-type I) mice was normal. The results provide evidence that NRG1 type I impacts on hippocampal function and circuitry. The effects are likely mediated via inhibitory interneurons and may be relevant to the involvement of NRG1 in schizophrenia. However, the findings, in concert with those from other genetic and pharmacological manipulations of NRG1, emphasize the complex and pleiotropic nature of the gene, even with regard to a single isoform.


Subject(s)
Action Potentials/physiology , Biological Clocks/physiology , Hippocampus/physiology , Long-Term Potentiation/physiology , Memory, Short-Term/physiology , Neuregulin-1/metabolism , Animals , Cells, Cultured , Hippocampus/cytology , Mice , Mice, Transgenic , Protein Isoforms/genetics , Protein Isoforms/metabolism , Up-Regulation/physiology
10.
Neuropsychopharmacology ; 36(13): 2616-28, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21832989

ABSTRACT

Group II metabotropic glutamate receptors (mGluR2 and mGluR3, encoded by GRM2 and GRM3) are implicated in hippocampal function and cognition, and in the pathophysiology and treatment of schizophrenia and other psychiatric disorders. However, pharmacological and behavioral studies with group II mGluR agonists and antagonists have produced complex results. Here, we studied hippocampus-dependent memory in GRM2/3 double knockout (GRM2/3(-/-)) mice in an iterative sequence of experiments. We found that they were impaired on appetitively motivated spatial reference and working memory tasks, and on a spatial novelty preference task that relies on animals' exploratory drive, but were unimpaired on aversively motivated spatial memory paradigms. GRM2/3(-/-) mice also performed normally on an appetitively motivated, non-spatial, visual discrimination task. These results likely reflect an interaction between GRM2/3 genotype and the arousal-inducing properties of the experimental paradigm. The deficit seen on appetitive and exploratory spatial memory tasks may be absent in aversive tasks because the latter induce higher levels of arousal, which rescue spatial learning. Consistent with an altered arousal-cognition relationship in GRM2/3(-/-) mice, injection stress worsened appetitively motivated, spatial working memory in wild-types, but enhanced performance in GRM2/3(-/-) mice. GRM2/3(-/-) mice were also hypoactive in response to amphetamine. This fractionation of hippocampus-dependent memory depending on the appetitive-aversive context is to our knowledge unique, and suggests a role for group II mGluRs at the interface of arousal and cognition. These arousal-dependent effects may explain apparently conflicting data from previous studies, and have translational relevance for the involvement of these receptors in schizophrenia and other disorders.


Subject(s)
Arousal/physiology , Cognition/physiology , Hippocampus/metabolism , Memory Disorders/metabolism , Receptors, Metabotropic Glutamate/deficiency , Receptors, Metabotropic Glutamate/physiology , Animals , Hippocampus/physiopathology , Male , Memory Disorders/genetics , Memory Disorders/physiopathology , Mice , Mice, Knockout , Receptors, Metabotropic Glutamate/genetics
11.
Brain Res ; 1301: 197-206, 2009 Dec 08.
Article in English | MEDLINE | ID: mdl-19747464

ABSTRACT

Single nucleotide polymorphisms (SNPs) within the gene encoding the serine/threonine kinase KIS (Kinase Interacting with Stathmin, also known as UHMK1) have recently been associated with schizophrenia. As none of the disease associated SNPs are coding, they may confer susceptibility by altering some facet of KIS expression. Here we have characterised the cellular distribution of KIS in human brain using in situ hybridisation and immunohistochemistry, and quantified KIS protein and mRNA in two large brain series to determine if KIS expression is altered in schizophrenia or bipolar disorder or in relation to a schizophrenia-associated SNP (rs7513662). Post-mortem tissue from the superior temporal gyrus of schizophrenia and control subjects, and also dorsolateral prefrontal cortex, anterior cingulate cortex, and cerebellum from schizophrenia, bipolar disorder, and control subjects were used. KIS expression was measured by quantitative PCR (mRNA) and immunoautoradiography (protein), and was also quantified by immunoblot in lymphoblast cell lines derived from schizophrenia and control subjects. Our results demonstrate that KIS is expressed in neurons, and its encoded protein is localised to the nucleus and cytoplasm. No difference in KIS expression was found between diagnostic groups, or in the lymphoblast cell lines, and no effect of rs7513662 genotype on KIS expression was found. Hence, these data do not provide support for the hypothesis that altered expression is the mechanism by which genetic variation of KIS may increase susceptibility to schizophrenia, nor evidence that KIS expression is altered in the disease itself, at least in terms of the parameters studied here.


Subject(s)
Brain/metabolism , Gene Expression/genetics , Genotype , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Schizophrenia/genetics , Adult , Aged , Analysis of Variance , Autoradiography , Blotting, Western , Cell Line, Tumor , Female , Humans , Immunohistochemistry , In Situ Hybridization , Intracellular Signaling Peptides and Proteins/metabolism , Male , Middle Aged , Neurons/metabolism , Polymorphism, Single Nucleotide/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Schizophrenia/metabolism
13.
Am J Med Genet B Neuropsychiatr Genet ; 144B(6): 834-9, 2007 Sep 05.
Article in English | MEDLINE | ID: mdl-17477346

ABSTRACT

Catechol-o-methyltransferase (COMT) is important for modulating dopamine levels, prefrontal cortex (PFC) function, and several psychiatric phenotypes. A single COMT mRNA has been described in human brain, which gives rise to membrane-bound (MB)- and soluble (S)-COMT proteins. In addition, we have recently described a novel COMT protein isoform in the human PFC, suggesting that there are more COMT gene products expressed than are currently appreciated. Therefore, we have investigated whether variant COMT mRNAs are present in human brain. We used reverse transcription-PCR (RT-PCR) to screen systematically for variant COMT mRNAs in human frontal cortex. Intron-spanning primers were used for exon-to-exon PCR reactions; additionally, specific primers were designed to sequences in the NCBI Aceview database. The identity of amplicons was confirmed by sequencing, and their regional distributions and 3' untranslated regions (UTRs) were characterised using RT-PCR. We detected 7 COMT variant mRNAs, resulting from both insertions and deletions within the known COMT brain transcript. Several of the variants alter the predicted coding sequence. Three of these variants correspond to sequences within the Aceview database and could be reliably amplified, while the remaining four do not correspond to any expressed sequence tags and were amplified only once. The regional distributions of these transcripts are described. The results demonstrate multiple COMT mRNAs in human brain, revealing an additional complexity to the biology of COMT. The alternate gene products may be of significant functional importance, and differentially impacted by polymorphisms within the COMT gene.


Subject(s)
Brain/enzymology , Catechol O-Methyltransferase/genetics , RNA, Messenger/genetics , Adult , Base Sequence , DNA Primers/genetics , Fetus/enzymology , Gene Expression , Genetic Variation , Humans , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...