160 results on '"Maria Karayiorgou"'
Search Results
2. Family history identifies sporadic schizoaffective disorder as a subtype for genetic studies
- Author
-
Nicolaas J. van der Merwe, Maria Karayiorgou, René Ehlers, and Johannes L. Roos
- Subjects
schizophrenia ,family history ,genetic studies ,novo genetic events ,familial-sporadic distinction ,Psychiatry ,RC435-571 - Abstract
Background: Schizophrenia is a heterogeneous disorder with strong genetic vulnerability. Family history of schizophrenia has been considered in genetic studies under several models. De novo genetic events seem to play a larger role in sporadic cases. Aim: This study used the familial–sporadic distinction with the aim of identifying a more homogeneous phenotype to delineate the genetic and clinical complexity of schizophrenia. Setting: The study was conducted at Weskoppies Hospital, Pretoria, South Africa. Methods: The study included 384 participants with schizophrenia or schizoaffective disorder from the Afrikaner founder population in South Africa who are considered comparable to Caucasian patients from the United States. A comprehensive data capturing sheet was completed. Results: When schizophrenia and schizoaffective disorder diagnoses were considered jointly, we found no significant differences between the sporadic and the familial groups for age at disease onset, season of birth, comorbid diagnoses, clinical symptomatology, history of suicide or marital status. When the diagnoses were examined separately, however, the sporadic schizoaffective disorder, bipolar type, was found to have a significantly lower age at onset (mean 20.6 vs. 25.3 years). Conclusion: The sporadic schizoaffective disorder, bipolar type, forms a more homogeneous subgroup for genetic studies.
- Published
- 2020
- Full Text
- View/download PDF
3. The BDNF Val66Met variant affects gene expression through miR-146b
- Author
-
Pei-Ken Hsu, Bin Xu, Jun Mukai, Maria Karayiorgou, and Joseph A. Gogos
- Subjects
BDNF ,Val66Met ,MicroRNA ,Knock-in mice ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Variation in gene expression is an important mechanism underlying susceptibility to complex disease and traits. Single nucleotide polymorphisms (SNPs) account for a substantial portion of the total detected genetic variation in gene expression but how exactly variants acting in trans modulate gene expression and disease susceptibility remains largely unknown. The BDNF Val66Met SNP has been associated with a number of psychiatric disorders such as depression, anxiety disorders, schizophrenia and related traits. Using global microRNA expression profiling in hippocampus of humanized BDNF Val66Met knock-in mice we showed that this variant results in dysregulation of at least one microRNA, which in turn affects downstream target genes. Specifically, we show that reduced levels of miR-146b (mir146b), lead to increased Per1 and Npas4 mRNA levels and increased Irak1 protein levels in vitro and are associated with similar changes in the hippocampus of hBDNFMet/Met mice. Our findings highlight trans effects of common variants on microRNA-mediated gene expression as an integral part of the genetic architecture of complex disorders and traits.
- Published
- 2015
- Full Text
- View/download PDF
4. MicroRNA dysregulation in neuropsychiatric disorders and cognitive dysfunction
- Author
-
Bin Xu, Pei-Ken Hsu, Maria Karayiorgou, and Joseph A. Gogos
- Subjects
MicroRNA ,Psychiatric disorder ,Schizophrenia ,Risk gene ,22q11.2 ,Intellectual disability ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
MicroRNAs (miRNA), a class of non-coding RNAs, are emerging as important modulators of neuronal development, structure and function. A connection has been established between abnormalities in miRNA expression and miRNA-mediated gene regulation and psychiatric and neurodevelopmental disorders as well as cognitive dysfunction. Establishment of this connection has been driven by progress in elucidating the genetic etiology of these phenotypes and has provided a context to interpret additional supporting evidence accumulating from parallel expression profiling studies in brains and peripheral blood of patients. Here we review relevant evidence that supports this connection and explore possible mechanisms that underlie the contribution of individual miRNAs and miRNA-related pathways to the pathogenesis and pathophysiology of these complex clinical phenotypes. The existing evidence provides useful hypotheses for further investigation as well as important clues for identifying novel therapeutic targets.
- Published
- 2012
- Full Text
- View/download PDF
5. Nogo Receptor 1 (RTN4R) as a candidate gene for schizophrenia: analysis using human and mouse genetic approaches.
- Author
-
Ruby Hsu, Abigail Woodroffe, Wen-Sung Lai, Melloni N Cook, Jun Mukai, Jonathan P Dunning, Douglas J Swanson, J Louw Roos, Gonçalo R Abecasis, Maria Karayiorgou, and Joseph A Gogos
- Subjects
Medicine ,Science - Abstract
NOGO Receptor 1 (RTN4R) regulates axonal growth, as well as axon regeneration after injury. The gene maps to the 22q11.2 schizophrenia susceptibility locus and is thus a strong functional and positional candidate gene.We evaluate evidence for genetic association between common RTN4R polymorphisms and schizophrenia in a large family sample of Afrikaner origin and screen the exonic sequence of RTN4R for rare variants in an independent sample from the U.S. We also employ animal model studies to assay a panel of schizophrenia-related behavioral tasks in an Rtn4r-deficient mouse model. We found weak sex-specific evidence for association between common RTN4R polymorphisms and schizophrenia in the Afrikaner patients. In the U.S. sample, we identified two novel non-conservative RTN4R coding variants in two patients with schizophrenia that were absent in 600 control chromosomes. In our complementary mouse model studies, we identified a haploinsufficient effect of Rtn4r on locomotor activity, but normal performance in schizophrenia-related behavioral tasks. We also provide evidence that Rtn4r deficiency can modulate the long-term behavioral effects of transient postnatal N-methyl-D-aspartate (NMDA) receptor hypofunction.Our results do not support a major role of RTN4R in susceptibility to schizophrenia or the cognitive and behavioral deficits observed in individuals with 22q11 microdeletions. However, they suggest that RTN4R may modulate the genetic risk or clinical expression of schizophrenia in a subset of patients and identify additional studies that will be necessary to clarify the role of RTN4R in psychiatric phenotypes. In addition, our results raise interesting issues about evaluating the significance of rare genetic variants in disease and their role in causation.
- Published
- 2007
- Full Text
- View/download PDF
6. AUTS2 gene dosage affects synaptic AMPA receptors via a local dendritic spine AUTS2-TTC3-AKT-mTORC1 signaling dysfunction
- Author
-
Aude-Marie Lepagnol-Bestel, Arnaud Duchon, Julia Viard, Mirna Kvajo, Rachel Daudin, Malik Khelfaoui, Simon Haziza, Yann Loe-Mie, Mattia Aime, Futoshi Suizu, Marie-Christine Birling, Mounia Bensaid, Sylvie Jacquot, Pascale Koebel, Céline Reverdy, Jean-Christophe Rain, Masayuki Noguchi, Xavier Marquez, Antoine Triller, Yann Humeau, Yann Hérault, Maria Karayiorgou, Joseph A. Gogos, and Michel Simonneau
- Abstract
The Human 1.2-MbAUTS2locus on chromosome 7q11.22 encodes a 1259-aa full-length protein, and a 711-aa C-terminal isoform. Functions of these AUTS2 proteins are only partly known. The major traits found in patients displayingAUTS2locus mutations are Intellectual Disabilities, microcephaly attention deficit hyperactivity disorder (ADHD) (54%), and autistic traits. Furthermore,AUTS2common variants were recently found associated to alcohol consumption and dyslexia using GWAS approaches. Auts2 localizes mainly in cell nuclei. We evidenced by super-resolution that Auts2 is present in dendritic spines. Auts2 interacts with Ttc3, the Akt2 E3 ligase, and negatively regulates Akt2 ubiquitination. Auts2 haploinsufficiency affects Akt/mTorc1 pathway with a decrease in AMPA and NMDA receptor subunits and in synaptic currents. Akt2 injection in postsynaptic neurons is sufficient to reverse changes in synaptic currents generated by Auts2 haploinsufficiency. Using chromosome engineering based on targeted meiotic recombination, we generated two mouse models withAuts2locus deletion and duplication. DeletedAuts2locus mice display stereotypies (rearing), perseveration and abnormal recognition memory. DuplicatedAuts2locus mice display similar perseveration and abnormal recognition memory but also a decrease in cued and contextual fear memory. Gene dosage induce changes in brain sub-region neuronal networks. In the thalamo-lateral amygdala pathway linked to cued fear memory, we found synaptic impairments linked to AMPA receptors, with a specific decrease in pAKT/total AKT ratio in duplicated Auts2 mice. Altogether, our study thereby provides a novel mechanistic and potentially therapeutic understanding of synaptic AKT/mTORC1 deregulated signaling and its related behavioral and cognitive phenotypes.
- Published
- 2022
7. Family history identifies sporadic schizoaffective disorder as a subtype for genetic studies
- Author
-
Rene Ehlers, Nicolaas J van der Merwe, Johannes L. Roos, and Maria Karayiorgou
- Subjects
genetic studies ,medicine.medical_specialty ,Season of birth ,lcsh:RC435-571 ,Schizoaffective disorder ,lcsh:RZ400-408 ,03 medical and health sciences ,0302 clinical medicine ,lcsh:Psychiatry ,mental disorders ,Medicine ,Family history ,Psychiatry ,Heterogeneous disorder ,Original Research ,family history ,familial-sporadic distinction ,schizophrenia ,novo genetic events ,familialsporadic distinction ,business.industry ,lcsh:Mental healing ,medicine.disease ,3. Good health ,030227 psychiatry ,Psychiatry and Mental health ,Homogeneous ,Schizophrenia ,Marital status ,business ,030217 neurology & neurosurgery ,Founder effect - Abstract
Background: Schizophrenia is a heterogeneous disorder with strong genetic vulnerability. Family history of schizophrenia has been considered in genetic studies under several models. De novo genetic events seem to play a larger role in sporadic cases. Aim: This study used the familial–sporadic distinction with the aim of identifying a more homogeneous phenotype to delineate the genetic and clinical complexity of schizophrenia. Setting: The study was conducted at Weskoppies Hospital, Pretoria, South Africa. Methods: The study included 384 participants with schizophrenia or schizoaffective disorder from the Afrikaner founder population in South Africa who are considered comparable to Caucasian patients from the United States. A comprehensive data capturing sheet was completed. Results: When schizophrenia and schizoaffective disorder diagnoses were considered jointly, we found no significant differences between the sporadic and the familial groups for age at disease onset, season of birth, comorbid diagnoses, clinical symptomatology, history of suicide or marital status. When the diagnoses were examined separately, however, the sporadic schizoaffective disorder, bipolar type, was found to have a significantly lower age at onset (mean 20.6 vs. 25.3 years). Conclusion: The sporadic schizoaffective disorder, bipolar type, forms a more homogeneous subgroup for genetic studies.
- Published
- 2020
8. Combined small-molecule inhibition accelerates the derivation of functional cortical neurons from human pluripotent stem cells
- Author
-
M. Zeeshan Ozair, Song-Hai Shi, Yosif Ganat, Nicolas Renier, Talia Atkin, Jason Tchieu, Faranak Fattahi, Joseph A. Gogos, Lorenz Studer, Mark J. Tomishima, Xin-Jun Zhang, Ziyi Sun, Ricardo Azevedo, Bastian Zimmer, Maria Karayiorgou, Zhuhao Wu, Marc Tessier-Lavigne, Ali H. Brivanlou, Yuchen Qi, and Nadja Zeltner
- Subjects
Pluripotent Stem Cells ,0301 basic medicine ,Neurogenesis ,Cell ,Central nervous system ,Drug Evaluation, Preclinical ,Biomedical Engineering ,Bioengineering ,Sensory system ,Biology ,Applied Microbiology and Biotechnology ,Regenerative medicine ,Article ,Cell therapy ,03 medical and health sciences ,medicine ,Humans ,Induced pluripotent stem cell ,Cells, Cultured ,Neurons ,Dose-Response Relationship, Drug ,Cell Differentiation ,Anatomy ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Batch Cell Culture Techniques ,Molecular Medicine ,Neuron ,Neuroscience ,Central Nervous System Agents ,Biotechnology - Abstract
Considerable progress has been made in converting human pluripotent stem cells (hPSCs) into functional neurons. However, the protracted timing of human neuron specification and functional maturation remains a key challenge that hampers the routine application of hPSC-derived lineages in disease modeling and regenerative medicine. Using a combinatorial small-molecule screen, we previously identified conditions for the rapid differentiation of hPSCs into peripheral sensory neurons. Here we generalize the approach to central nervous system (CNS) fates by developing a small-molecule approach for accelerated induction of early-born cortical neurons. Combinatorial application of 6 pathway inhibitors induces post-mitotic cortical neurons with functional electrophysiological properties by day 16 of differentiation, in the absence of glial cell co-culture. The resulting neurons, transplanted at 8 days of differentiation into the postnatal mouse cortex, are functional and establish long-distance projections, as shown using iDISCO whole brain imaging. Accelerated differentiation into cortical neuron fates should facilitate hPSC-based strategies for disease modeling and cell therapy in CNS disorders.
- Published
- 2017
9. Aberrant function of the C-terminal tail of HIST1H1E Aacelerates cellular senescence and causes premature aging
- Author
-
Giuseppe Matullo, Brett H. Graham, Elisa Coluzzi, Karit Reinson, Antonella Sgura, Monica H. Wojcik, Luca Pannone, Melissa P. Wasserstein, Lucia Pedace, Seema R. Lalani, Elena Carcarino, Daniela Q.C.M. Barge-Schaapveld, Anke Van Dijck, Austin Larson, Giovanna Carpentieri, Alessandro Bruselles, Simona Petrucci, Simone Pizzi, Elisabetta Flex, Cornelia Di Gaetano, Francesca Clementina Radio, Bruno Dallapiccola, Serena Cecchetti, Clara Viberti, Enrico Bertini, Chieko Chijiwa, Emilia K. Bijlsma, Elisabetta Ferretti, William J. Craigen, Cristina Andreoli, Brian G. Skotko, Daan J. Kamphuis, Alessandro De Luca, J. Louw Roos, Giuseppina Catanzaro, Sandra Kenis, Mariëtte J.V. Hoffer, Katrin Õunap, Maria Karayiorgou, Gijs W. E. Santen, Annette P.M. van den Elzen, Kathleen Brown, Haley Streff, M. E. Suzanne Lewis, Claudia A. L. Ruivenkamp, Xiaoyan Ge, Andrea Ciolfi, Nathalie Van der Aa, Marco Tartaglia, Rossella Rota, Amber Begtrup, Richard E. Person, Simone Martinelli, Koen L.I. van Gassen, R. Frank Kooy, Marije Meuwissen, Magdalena Walkiewicz, Evelina Miele, Marije Koopmans, Sander Pajusalu, Flex, E., Martinelli, S., Van Dijck, A., Ciolfi, A., Cecchetti, S., Coluzzi, E., Pannone, L., Andreoli, C., Radio, F. C., Pizzi, S., Carpentieri, G., Bruselles, A., Catanzaro, G., Pedace, L., Miele, E., Carcarino, E., Ge, X., Chijiwa, C., Lewis, M. E. S., Meuwissen, M., Kenis, S., Van der Aa, N., Larson, A., Brown, K., Wasserstein, M. P., Skotko, B. G., Begtrup, A., Person, R., Karayiorgou, M., Roos, J. L., Van Gassen, K. L., Koopmans, M., Bijlsma, E. K., Santen, G. W. E., Barge-Schaapveld, D. Q. C. M., Ruivenkamp, C. A. L., Hoffer, M. J. V., Lalani, S. R., Streff, H., Craigen, W. J., Graham, B. H., van den Elzen, A. P. M., Kamphuis, D. J., Ounap, K., Reinson, K., Pajusalu, S., Wojcik, M. H., Viberti, C., Di Gaetano, C., Bertini, E., Petrucci, S., De Luca, A., Rota, R., Ferretti, E., Matullo, G., Dallapiccola, B., Sgura, A., Walkiewicz, M., Kooy, R. F., and Tartaglia, M.
- Subjects
0301 basic medicine ,Premature aging ,Senescence ,Male ,Cell division ,methylation profiling ,Article ,Chromatin remodeling ,chromatin remodeling ,Histones ,03 medical and health sciences ,chemistry.chemical_compound ,replicative senescence ,0302 clinical medicine ,HIST1H1E ,chromatin dynamic ,Genetics ,accelerated aging ,cellular senescence ,Humans ,Genetics(clinical) ,Child ,Biology ,Genetics (clinical) ,chromatin compaction ,chromatin dynamics ,linker histone ,linker histone H1.4 ,Aneuploidy ,Cell Nucleolus ,Cellular Senescence ,Chromatin ,DNA Methylation ,Female ,Infant ,Middle Aged ,biology ,DNA replication ,Cell biology ,030104 developmental biology ,Histone ,chemistry ,biology.protein ,Human medicine ,030217 neurology & neurosurgery ,DNA - Abstract
Histones mediate dynamic packaging of nuclear DNA in chromatin, a process that is precisely controlled to guarantee efficient compaction of the genome and proper chromosomal segregation during cell division and to accomplish DNA replication, transcription, and repair. Due to the important structural and regulatory roles played by histones, it is not surprising that histone functional dysregulation or aberrant levels of histones can have severe consequences for multiple cellular processes and ultimately might affect development or contribute to cell transformation. Recently, germline frameshift mutations involving the C-terminal tail of HIST1H1E, which is a widely expressed member of the linker histone family and facilitates higher-order chromatin folding, have been causally linked to an as-yet poorly defined syndrome that includes intellectual disability. We report that these mutations result in stable proteins that reside in the nucleus, bind to chromatin, disrupt proper compaction of DNA, and are associated with a specific methylation pattern. Cells expressing these mutant proteins have a dramatically reduced proliferation rate and competence, hardly enter into the S phase, and undergo accelerated senescence. Remarkably, clinical assessment of a relatively large cohort of subjects sharing these mutations revealed a premature aging phenotype as a previously unrecognized feature of the disorder. Our findings identify a direct link between aberrant chromatin remodeling, cellular senescence, and accelerated aging.
- Published
- 2019
10. System-based proteomic and metabonomic analysis of the Df(16)A+/− mouse identifies potential miR-185 targets and molecular pathway alterations
- Author
-
Elaine Holmes, Elizabeth J. Want, Hendrik Wesseling, Joseph A. Gogos, Bin Xu, Paul M. Guest, Maria Karayiorgou, Sabine Bahn, and Neurosciences
- Subjects
0301 basic medicine ,Gene isoform ,Biochemistry & Molecular Biology ,HIPPOCAMPAL VOLUME REDUCTION ,MATRIX METALLOPROTEINASES ,Transgene ,Biology ,Proteomics ,MITOCHONDRIAL-FUNCTION ,17 Psychology And Cognitive Sciences ,Transcriptome ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Metabolomics ,Lipid biosynthesis ,microRNA ,22Q11.2 DELETION SYNDROME ,Epigenetics ,Molecular Biology ,Psychiatry ,INSULIN-RESISTANCE ,Science & Technology ,PSYCHIATRIC-DISORDERS ,Neurosciences ,LONG-TERM DEPRESSION ,11 Medical And Health Sciences ,06 Biological Sciences ,LINKED N-ACETYLGLUCOSAMINE ,Cell biology ,Psychiatry and Mental health ,030104 developmental biology ,O-GLCNAC TRANSFERASE ,Biochemistry ,SCHIZOPHRENIA-RELATED MICRODELETION ,Neurosciences & Neurology ,Life Sciences & Biomedicine - Abstract
Deletions on chromosome 22q11.2 are a strong genetic risk factor for development of schizophrenia and cognitive dysfunction. We employed shotgun liquid chromatography-mass spectrometry (LC-MS) proteomic and metabonomic profiling approaches on prefrontal cortex (PFC) and hippocampal (HPC) tissue from Df(16)A(+/-) mice, a model of the 22q11.2 deletion syndrome. Proteomic results were compared with previous transcriptomic profiling studies of the same brain regions. The aim was to investigate how the combined effect of the 22q11.2 deletion and the corresponding miRNA dysregulation affects the cell biology at the systems level. The proteomic brain profiling analysis revealed PFC and HPC changes in various molecular pathways associated with chromatin remodelling and RNA transcription, indicative of an epigenetic component of the 22q11.2DS. Further, alterations in glycolysis/gluconeogenesis, mitochondrial function and lipid biosynthesis were identified. Metabonomic profiling substantiated the proteomic findings by identifying changes in 22q11.2 deletion syndrome (22q11.2DS)-related pathways, such as changes in ceramide phosphoethanolamines, sphingomyelin, carnitines, tyrosine derivates and panthothenic acid. The proteomic findings were confirmed using selected reaction monitoring mass spectrometry, validating decreased levels of several proteins encoded on 22q11.2, increased levels of the computationally predicted putative miR-185 targets UDP-N-acetylglucosamine-peptide N-acetylglucosaminyltransferase 110 kDa subunit (OGT1) and kinesin heavy chain isoform 5A and alterations in the non-miR-185 targets serine/threonine-protein phosphatase 2B catalytic subunit gamma isoform, neurofilament light chain and vesicular glutamate transporter 1. Furthermore, alterations in the proteins associated with mammalian target of rapamycin signalling were detected in the PFC and with glutamatergic signalling in the hippocampus. Based on the proteomic and metabonomic findings, we were able to develop a schematic model summarizing the most prominent molecular network findings in the Df(16)A(+/-) mouse. Interestingly, the implicated pathways can be linked to one of the most consistent and strongest proteomic candidates, (OGT1), which is a predicted miR-185 target. Our results provide novel insights into system-biological mechanisms associated with the 22q11DS, which may be linked to cognitive dysfunction and an increased risk to develop schizophrenia. Further investigation of these pathways could help to identify novel drug targets for the treatment of schizophrenia.Molecular Psychiatry advance online publication, 22 March 2016; doi:10.1038/mp.2016.27.
- Published
- 2016
11. De Novo Synonymous Mutations in Regulatory Elements Contribute to the Genetic Etiology of Autism and Schizophrenia
- Author
-
Iuliana Ionita-Laza, Bin Xu, Maria Karayiorgou, Atsushi Takata, and Joseph A. Gogos
- Subjects
0301 basic medicine ,Male ,Databases, Factual ,Neuroscience(all) ,Nerve Tissue Proteins ,Disease ,Biology ,medicine.disease_cause ,Article ,Machine Learning ,03 medical and health sciences ,mental disorders ,medicine ,Humans ,Exome ,Genetic Predisposition to Disease ,Regulatory Elements, Transcriptional ,Autistic Disorder ,Gene ,Genetic Association Studies ,Genetics ,Mutation ,General Neuroscience ,Rab2 GTP-Binding Protein ,Histone-Lysine N-Methyltransferase ,Sequence Analysis, DNA ,medicine.disease ,MicroRNAs ,rab2 GTP-Binding Protein ,030104 developmental biology ,Schizophrenia ,Autism ,Female ,Synonymous substitution - Abstract
We analyze de novo synonymous mutations identified in autism spectrum disorders (ASDs) and schizophrenia (SCZ) with potential impact on regulatory elements using data from whole-exome sequencing (WESs) studies. Focusing on five types of genetic regulatory functions, we found that de novo near-splice site synonymous mutations changing exonic splicing regulators and those within frontal cortex-derived DNase I hypersensitivity sites are significantly enriched in ASD and SCZ, respectively. These results remained significant, albeit less so, after incorporating two additional ASD datasets. Among the genes identified, several are hit by multiple functional de novo mutations, with RAB2A and SETD1A showing the highest statistical significance in ASD and SCZ, respectively. The estimated contribution of these synonymous mutations to disease liability is comparable to de novo protein-truncating mutations. These findings expand the repertoire of functional de novo mutations to include "functional" synonymous ones and strengthen the role of rare variants in neuropsychiatric disease risk.
- Published
- 2016
- Full Text
- View/download PDF
12. Loss-of-function mutation in
- Author
-
Anastasia, Diamantopoulou, Ziyi, Sun, Jun, Mukai, Bin, Xu, Karine, Fenelon, Maria, Karayiorgou, and Joseph A, Gogos
- Subjects
Male ,Mice, Inbred C57BL ,Mice ,Neuronal Plasticity ,Phenotype ,PNAS Plus ,Loss of Function Mutation ,Schizophrenia ,Animals ,Membrane Proteins ,Female ,Genetic Predisposition to Disease - Abstract
Despite substantial progress in the field of schizophrenia (SCZ) genetics, the heterogeneity of genetic etiology and neural complexity has rendered the task of developing improved treatments inauspicious. Thus, there is need to identify convergent neural substrates and underlying molecular mechanisms that can serve the prevention or reversal of disease progression. Our extensive characterization of an animal model of the 22q11.2 deletion, one of the strongest genetic risk factors for SCZ, when combined with a loss-of-function (LoF) mutation for a microRNA-dependent upregulated target, offers a proof of principle for such approaches. Hence, identification of variants that confer protection against disease by disabling protein function via LoF effects holds great promise for devising therapeutic schemes to restore or prevent disease symptoms.
- Published
- 2017
13. Loss-of-function mutation in Mirta22/Emc10 rescues specific schizophrenia-related phenotypes in a mouse model of the 22q11.2 deletion
- Author
-
Joseph A. Gogos, Jun Mukai, Ziyi Sun, Maria Karayiorgou, Anastasia Diamantopoulou, Bin Xu, and Karine Fénelon
- Subjects
0301 basic medicine ,Genetics ,Multidisciplinary ,Working memory ,Biology ,medicine.disease ,Phenotype ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Schizophrenia ,microRNA ,Knockout mouse ,medicine ,Allele ,Prefrontal cortex ,030217 neurology & neurosurgery ,Prepulse inhibition - Abstract
Identification of protective loss-of-function (LoF) mutations holds great promise for devising novel therapeutic interventions, although it faces challenges due to the scarcity of protective LoF alleles in the human genome. Exploiting the detailed mechanistic characterization of animal models of validated disease mutations offers an alternative. Here, we provide insights into protective-variant biology based on our characterization of a model of the 22q11.2 deletion, a strong genetic risk factor for schizophrenia (SCZ). Postnatal brain up-regulation of Mirta22/Emc10, an inhibitor of neuronal maturation, represents the major transcriptional effect of the 22q11.2-associated microRNA dysregulation. Here, we demonstrate that mice in which the Df(16)A deficiency is combined with a LoF Mirta22 allele show rescue of key SCZ-related deficits, namely prepulse inhibition decrease, working memory impairment, and social memory deficits, as well as synaptic and structural plasticity abnormalities in the prefrontal cortex. Additional analysis of homozygous Mirta22 knockout mice, in which no alteration is observed in the above-mentioned SCZ-related phenotypes, highlights the deleterious effects of Mirta22 up-regulation. Our results support a causal link between dysregulation of a miRNA target and SCZ-related deficits and provide key insights into beneficial LoF mutations and potential new treatments.
- Published
- 2017
14. Scan statistic-based analysis of exome sequencing data identifies FAN1 at 15q13.3 as a susceptibility gene for schizophrenia and autism
- Author
-
Iuliana Ionita-Laza, Joseph D. Buxbaum, Vlad Makarov, Bin Xu, Joseph A. Gogos, J. Louw Roos, and Maria Karayiorgou
- Subjects
Genetic Markers ,Male ,Risk ,Nonsynonymous substitution ,DNA Repair ,Molecular Sequence Data ,Locus (genetics) ,Biology ,South Africa ,mental disorders ,medicine ,Cluster Analysis ,Humans ,Computer Simulation ,Exome ,Genetic Predisposition to Disease ,Amino Acid Sequence ,Copy-number variation ,Autistic Disorder ,Gene ,Exome sequencing ,Genetics ,Chromosomes, Human, Pair 15 ,Endodeoxyribonucleases ,Multidisciplinary ,Sequence Homology, Amino Acid ,Nucleotides ,Genetic Variation ,Biological Sciences ,medicine.disease ,Multifunctional Enzymes ,United States ,Exodeoxyribonucleases ,Phenotype ,Autism spectrum disorder ,Schizophrenia ,Autism ,Female - Abstract
We used a family-based cluster detection approach designed to localize significant rare disease-risk variants clusters within a region of interest to systematically search for schizophrenia (SCZ) susceptibility genes within 49 genomic loci previously implicated by de novo copy number variants. Using two independent whole-exome sequencing family datasets and a follow-up autism spectrum disorder (ASD) case/control whole-exome sequencing dataset, we identified variants in one gene, Fanconi-associated nuclease 1 (FAN1), as being associated with both SCZ and ASD. FAN1 is located in a region on chromosome 15q13.3 implicated by a recurrent copy number variant, which predisposes to an array of psychiatric and neurodevelopmental phenotypes. In both SCZ and ASD datasets, rare nonsynonymous risk variants cluster significantly in affected individuals within a 20-kb window that spans several key functional domains of the gene. Our finding suggests that FAN1 is a key driver in the 15q13.3 locus for the associated psychiatric and neurodevelopmental phenotypes. FAN1 encodes a DNA repair enzyme, thus implicating abnormalities in DNA repair in the susceptibility to SCZ or ASD.
- Published
- 2013
15. Fine Mapping on Chromosome 13q32–34 and Brain Expression Analysis Implicates MYO16 in Schizophrenia
- Author
-
Gonçalo R. Abecasis, Laura Rodriguez-Murillo, Maria Karayiorgou, Bin Xu, Joseph A. Gogos, and J. Louw Roos
- Subjects
Male ,Genotype ,Genetic Linkage ,Population ,Single-nucleotide polymorphism ,Biology ,White People ,South Africa ,Genetic linkage ,Genetic variation ,medicine ,Humans ,Genetic Predisposition to Disease ,International HapMap Project ,education ,Gene ,Pharmacology ,Linkage (software) ,Genetics ,education.field_of_study ,Chromosomes, Human, Pair 13 ,Myosin Heavy Chains ,Brain ,Chromosome Mapping ,Genetic Variation ,medicine.disease ,Psychiatry and Mental health ,Schizophrenia ,Original Article ,Female - Abstract
We previously reported linkage of schizophrenia and schizoaffective disorder to 13q32-34 in the European descent Afrikaner population from South Africa. The nature of genetic variation underlying linkage peaks in psychiatric disorders remains largely unknown and both rare and common variants may be contributing. Here, we examine the contribution of common variants located under the 13q32-34 linkage region. We used densely spaced SNPs to fine map the linkage peak region using both a discovery sample of 415 families and a meta-analysis incorporating two additional replication family samples. In a second phase of the study, we use one family-based data set with 237 families and independent case-control data sets for fine mapping of the common variant association signal using HapMap SNPs. We report a significant association with a genetic variant (rs9583277) within the gene encoding for the myosin heavy-chain Myr 8 (MYO16), which has been implicated in neuronal phosphoinositide 3-kinase signaling. Follow-up analysis of HapMap variation within MYO16 in a second set of Afrikaner families and additional case-control data sets of European descent highlighted a region across introns 2-6 as the most likely region to harbor common MYO16 risk variants. Expression analysis revealed a significant increase in the level of MYO16 expression in the brains of schizophrenia patients. Our results suggest that common variation within MYO16 may contribute to the genetic liability to schizophrenia.
- Published
- 2013
16. A Disc1 mutation differentially affects neurites and spines in hippocampal and cortical neurons
- Author
-
Michel Simonneau, Mirna Kvajo, Maria Karayiorgou, Aude-Marie Lepagnol-Bestel, and Joseph A. Gogos
- Subjects
Dendritic spine ,Neurite ,Dendritic Spines ,Neurogenesis ,Golgi Apparatus ,Hippocampus ,Nerve Tissue Proteins ,Hippocampal formation ,medicine.disease_cause ,Article ,Mice ,Cellular and Molecular Neuroscience ,DISC1 ,symbols.namesake ,Neurites ,medicine ,Animals ,Molecular Biology ,Cells, Cultured ,Cerebral Cortex ,Mutation ,biology ,Cytoplasmic Vesicles ,Cell Biology ,Golgi apparatus ,Axons ,Cell biology ,Mice, Inbred C57BL ,Protein Transport ,Organ Specificity ,biology.protein ,symbols ,Neuroscience - Abstract
A balanced chromosomal translocation segregating with schizophrenia and affective disorders in a large Scottish family disrupting DISC1 implicated this gene as a susceptibility gene for major mental illness. Here we study neurons derived from a genetically engineered mouse strain with a truncating lesion disrupting the endogenous Disc1 ortholog. We provide a detailed account of the consequences of this mutation on axonal and dendritic morphogenesis as well as dendritic spine development in cultured hippocampal and cortical neurons. We show that the mutation has distinct effects on these two types of neurons, supporting a cell-type specific role of Disc1 in establishing structural connections among neurons. Moreover, using a validated antibody we provide evidence indicating that Disc1 localizes primarily to Golgi apparatus-related vesicles. Our results support the notion that in vitro cultures derived from Disc1(Tm1Kara) mice provide a valuable model for future mechanistic analysis of the cellular and biochemical effects of this mutation, and can thus serve as a platform for drug discovery efforts.
- Published
- 2013
17. Derepression of a Neuronal Inhibitor due to miRNA Dysregulation in a Schizophrenia-Related Microdeletion
- Author
-
Pei Ken Hsu, Joseph A. Gogos, Maria Karayiorgou, Kimberly L. Stark, and Bin Xu
- Subjects
Genetics ,Biochemistry, Genetics and Molecular Biology(all) ,Locus (genetics) ,RNA-binding protein ,Golgi apparatus ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,symbols.namesake ,Membrane protein ,microRNA ,Neuroplasticity ,symbols ,Gene ,Derepression - Abstract
Summary22q11.2 microdeletions result in specific cognitive deficits and schizophrenia. Analysis of Df(16)A+/− mice, which model this microdeletion, revealed abnormalities in the formation of neuronal dendrites and spines, as well as altered brain microRNAs. Here, we show a drastic reduction of miR-185, which resides within the 22q11.2 locus, to levels more than expected by a hemizygous deletion, and we demonstrate that this reduction alters dendritic and spine development. miR-185 represses, through an evolutionarily conserved target site, a previously unknown inhibitor of these processes that resides in the Golgi apparatus and shows higher prenatal brain expression. Sustained derepression of this inhibitor after birth represents the most robust transcriptional disturbance in the brains of Df(16)A+/− mice and results in structural alterations in the hippocampus. Reduction of miR-185 also has milder age- and region-specific effects on the expression of some Golgi-related genes. Our findings illuminate the contribution of microRNAs in psychiatric disorders and cognitive dysfunction.
- Published
- 2013
- Full Text
- View/download PDF
18. De novo gene mutations highlight patterns of genetic and neural complexity in schizophrenia
- Author
-
Yan-Yan Sun, Braden E. Boone, Joseph A. Gogos, J. Louw Roos, Scarlet Woodrick, Bin Xu, Maria Karayiorgou, Shawn Levy, and Iuliana Ionita-Laza
- Subjects
Nonsynonymous substitution ,Adult ,Male ,Sequence analysis ,Molecular Sequence Data ,Vesicular Transport Proteins ,Autophagy-Related Proteins ,Biology ,Gene mutation ,medicine.disease_cause ,Polymorphism, Single Nucleotide ,Article ,Cohort Studies ,03 medical and health sciences ,South Africa ,Young Adult ,0302 clinical medicine ,Genetics ,medicine ,Humans ,Exome ,Genetic Predisposition to Disease ,Gene ,Exome sequencing ,Dihydrouracil Dehydrogenase (NADP) ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,Neurons ,0303 health sciences ,Mutation ,Base Sequence ,Intracellular Signaling Peptides and Proteins ,Brain ,Gene Expression Regulation, Developmental ,Nuclear Proteins ,Sequence Analysis, DNA ,United States ,Schizophrenia ,DPYD ,Female ,Laminin ,030217 neurology & neurosurgery - Abstract
To evaluate evidence for de novo etiologies in schizophrenia, we sequenced at high coverage the exomes of families recruited from two populations with distinct demographic structures and history. We sequenced a total of 795 exomes from 231 parent-proband trios enriched for sporadic schizophrenia cases, as well as 34 unaffected trios. We observed in cases an excess of de novo nonsynonymous single-nucleotide variants as well as a higher prevalence of gene-disruptive de novo mutations relative to controls. We found four genes (LAMA2, DPYD, TRRAP and VPS39) affected by recurrent de novo events within or across the two populations, which is unlikely to have occurred by chance. We show that de novo mutations affect genes with diverse functions and developmental profiles, but we also find a substantial contribution of mutations in genes with higher expression in early fetal life. Our results help define the genomic and neural architecture of schizophrenia.
- Published
- 2012
19. Physiological and behavioural responsivity to stress and anxiogenic stimuli in COMT-deficient mice
- Author
-
Maria Karayiorgou, Lieve Desbonnet, Orna Tighe, Colm M. P. O’Tuathaigh, John L. Waddington, and Joseph A. Gogos
- Subjects
Male ,Restraint, Physical ,medicine.medical_specialty ,Time Factors ,medicine.drug_class ,Midazolam ,Statistics as Topic ,Dark Adaptation ,Catechol O-Methyltransferase ,COMT inhibitor ,behavioral disciplines and activities ,Chlordiazepoxide ,Nitrophenols ,Benzophenones ,Mice ,Behavioral Neuroscience ,chemistry.chemical_compound ,Corticosterone ,Internal medicine ,mental disorders ,medicine ,Animals ,Chronic stress ,Enzyme Inhibitors ,Psychiatry ,Mice, Knockout ,Analysis of Variance ,Benzodiazepine ,Catechol-O-methyl transferase ,Behavior, Animal ,Dose-Response Relationship, Drug ,Tolcapone ,fungi ,Organ Size ,Mice, Inbred C57BL ,Disease Models, Animal ,Endocrinology ,Anti-Anxiety Agents ,nervous system ,Anxiogenic ,chemistry ,Cytokines ,Female ,Psychology ,Stress, Psychological ,medicine.drug - Abstract
Catechol-O-methyltransferase, an enzyme involved in regulating brain catecholamine levels, has been implicated in anxiety, pain and/or stress responsivity. Elements of this putative association remain unclarified, notably whether: (a) COMT variation modulates responses to acute and/or chronic stress equally; (b) acute pharmacological inhibition of COMT produces comparable effects on anxiety to that observed after deletion of the COMT gene; (c) COMT genotype modulates action of anxiolytic drugs. We aimed to further investigate the relationship between reduced COMT function, anxiety and stress responsivity in mice. To compare the effect of acute vs . chronic restraint stress in female COMT KO vs . WT mice, serum corticosterone and cytokine concentrations were measured [Experiment 1]. Sensitivity to the benzodiazepines midazolam and chlordiazepoxide in the light–dark test was assessed in female COMT KO vs . WT mice [Experiment 2]. Effects of acute administration of the COMT inhibitor tolcapone, and of these same benzodiazepines thereon, in the light–dark test were assessed in female C57BL/6 mice [Experiment 3]. COMT KO mice demonstrated an increased corticosterone response to acute but not chronic stress, and a modified cytokine profile after chronic, but not acute stress. COMT KO mice showed increased anxiety, but benzodiazepine sensitivity was affected by COMT genotype. Whilst tolcapone had no effect on light/dark performance in C57BL6/J mice it decreased benzodiazepine sensitivity. These data elaborate earlier findings of increased anxiety in female COMT KO mice and also clarify a role for COMT in modulating stress-related hormonal and immune parameters in a manner that depends on chronicity of the stressor.
- Published
- 2012
20. The Genetic Architecture of Schizophrenia: New Mutations and Emerging Paradigms
- Author
-
Maria Karayiorgou, Joseph A. Gogos, and Laura Rodriguez-Murillo
- Subjects
Genetics ,Point mutation ,Locus (genetics) ,Genome-wide association study ,General Medicine ,Biology ,Genome ,General Biochemistry, Genetics and Molecular Biology ,Genetic architecture ,Risk Factors ,Evolutionary biology ,Genetic marker ,Schizophrenia ,Humans ,Genetic Predisposition to Disease ,Rare disease ,Genetic association - Abstract
Although a genetic component of schizophrenia has been acknowledged for a long time, the underlying architecture of the genetic risk remains a contentious issue. Early linkage and candidate association studies led to largely inconclusive results. More recently, the availability of powerful technologies, samples of sufficient sizes, and genome-wide panels of genetic markers facilitated systematic and agnostic scans throughout the genome for either common or rare disease risk variants of small or large effect size, respectively. Although the former had limited success, the role of rare genetic events, such as copy-number variants (CNVs) or rare point mutations, has become increasingly important in gene discovery for schizophrenia. Importantly, recent research building upon earlier findings of de novo recurrent CNVs at the 22q11.2 locus, has highlighted a de novo mutational paradigm as a major component of the genetic architecture of schizophrenia. Recent progress is bringing us closer to earlier intervention and new therapeutic targets.
- Published
- 2012
21. Genetic vs. pharmacological inactivation of COMT influences cannabinoid-induced expression of schizophrenia-related phenotypes
- Author
-
John F. Cryan, Ted Dinan, Emilie Petit, John L. Waddington, Niamh Clarke, Orna Tighe, Lieve Desbonnet, Jeremy Walsh, Colm M. P. O’Tuathaigh, Gerard Clarke, Joseph A. Gogos, Claire O'Leary, and Maria Karayiorgou
- Subjects
Reflex, Startle ,medicine.medical_treatment ,Schizophrenia/enzymology ,Swimming/psychology ,Anxiety ,Pharmacology ,COMT inhibitor ,Nitrophenols ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Nitrophenols/pharmacology ,Reflex, Startle/drug effects ,Pharmacology (medical) ,Enzyme Inhibitors ,Benzophenones/pharmacology ,Chromatography, High Pressure Liquid ,Prepulse inhibition ,Pain Measurement ,Enzyme Inhibitors/pharmacology ,Mice, Knockout ,Biogenic Monoamines/metabolism ,3. Good health ,Cannabinoids/pharmacology ,Psychiatry and Mental health ,Phenotype ,Schizophrenia ,Schizophrenic Psychology ,Morpholines/pharmacology ,Psychology ,Naphthalenes/pharmacology ,medicine.drug ,Psychosis ,Benzoxazines/pharmacology ,Morpholines ,Anxiety/psychology ,Catechol O-Methyltransferase/genetics ,Naphthalenes ,Catechol O-Methyltransferase ,behavioral disciplines and activities ,Benzophenones ,03 medical and health sciences ,Dopamine ,mental disorders ,medicine ,Animals ,Biogenic Monoamines ,Social Behavior ,Alleles ,Swimming ,Cannabinoid Receptor Agonists ,Catechol-O-methyl transferase ,Cannabinoids ,fungi ,Catechol O-Methyltransferase Inhibitors ,Cyclohexanols ,medicine.disease ,Benzoxazines ,030227 psychiatry ,Mice, Inbred C57BL ,chemistry ,Endophenotype ,Cannabinoid Receptor Agonists/pharmacology ,Tolcapone ,Cannabinoid ,Cyclohexanols/pharmacology ,Pain Measurement/drug effects ,030217 neurology & neurosurgery - Abstract
Catechol-O-methyltransferase (COMT) is an important enzyme in the metabolism of dopamine and disturbance in dopamine function is proposed to be central to the pathogenesis of schizophrenia. Clinical epidemiological studies have indicated cannabis use to confer a 2-fold increase in risk for subsequent onset of psychosis, with adolescent-onset use conveying even higher risk. There is evidence that a high activity COMT polymorphism moderates the effects of adolescent exposure to cannabis on risk for adult psychosis. In this paper we compared the effect of chronic adolescent exposure to the cannabinoid WIN 55212 on sensorimotor gating, behaviours related to the negative symptoms of schizophrenia, anxiety- and stress-related behaviours, as well as ex-vivo brain dopamine and serotonin levels, in COMT KO vs. wild-type (WT) mice. Additionally, we examined the effect of pretreatment with the COMT inhibitor tolcapone on acute effects of this cannabinoid on sensorimotor gating in C57BL/6 mice. COMT KO mice were shown to be more vulnerable than WT to the disruptive effects of adolescent cannabinoid treatment on prepulse inhibition (PPI). Acute pharmacological inhibition of COMT in C57BL/6 mice also modified acute cannabinoid effects on startle reactivity, as well as PPI, indicating that chronic and acute loss of COMT can produce dissociable effects on the behavioural effects of cannabinoids. COMT KO mice also demonstrated differential effects of adolescent cannabinoid administration on sociability and anxiety-related behaviour, both confirming and extending earlier reports of COMT×cannabinoid effects on the expression of schizophrenia-related endophenotypes.
- Published
- 2012
- Full Text
- View/download PDF
22. High Frequencies of De Novo CNVs in Bipolar Disorder and Schizophrenia
- Author
-
Verena Krause, Vladimir Vacic, Francis J. McMahon, Ellen Leibenluft, Vladimir Makarov, Sydney Gary, Marcella Rietschel, Shane McCarthy, Sven Cichon, Deborah L. Levy, Jonathan Sebat, Markus M. Nöthen, Katherine E. Burdick, Seungtai Yoon, Elliot S. Gershon, Thomas G. Schulze, Maria Karayiorgou, James B. Potash, Michael Gill, Anil K. Malhotra, Aiden Corvin, Olga Krastoshevsky, Jacob J. Michaelson, John R. Kelsoe, Dheeraj Malhotra, and Abhishek Bhandari
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Bipolar Disorder ,endocrine system diseases ,Adolescent ,DNA Copy Number Variations ,genetics [DNA Copy Number Variations] ,Offspring ,Neuroscience(all) ,Genome-wide association study ,Biology ,methods [Genome-Wide Association Study] ,diagnosis [Schizophrenia] ,Young Adult ,Internal medicine ,mental disorders ,medicine ,Humans ,genetics [Schizophrenia] ,ddc:610 ,Copy-number variation ,Bipolar disorder ,Young adult ,diagnosis [Bipolar Disorder] ,Child ,Genetics ,General Neuroscience ,Case-control study ,Genetic Variation ,medicine.disease ,genetics [Genetic Variation] ,Mood disorders ,Schizophrenia ,Case-Control Studies ,Female ,genetics [Bipolar Disorder] ,Genome-Wide Association Study - Abstract
SummaryWhile it is known that rare copy-number variants (CNVs) contribute to risk for some neuropsychiatric disorders, the role of CNVs in bipolar disorder is unclear. Here, we reasoned that a contribution of CNVs to mood disorders might be most evident for de novo mutations. We performed a genome-wide analysis of de novo CNVs in a cohort of 788 trios. Diagnoses of offspring included bipolar disorder (n = 185), schizophrenia (n = 177), and healthy controls (n = 426). Frequencies of de novo CNVs were significantly higher in bipolar disorder as compared with controls (OR = 4.8 [1.4,16.0], p = 0.009). De novo CNVs were particularly enriched among cases with an age at onset younger than 18 (OR = 6.3 [1.7,22.6], p = 0.006). We also confirmed a significant enrichment of de novo CNVs in schizophrenia (OR = 5.0 [1.5,16.8], p = 0.007). Our results suggest that rare spontaneous mutations are an important contributor to risk for bipolar disorder and other major neuropsychiatric diseases.
- Published
- 2011
- Full Text
- View/download PDF
23. Advancing drug discovery for schizophrenia
- Author
-
John A. Allen, Patrick F. Sullivan, Jay A. Gingrich, John H. Krystal, Alessandro Guidotti, Jeffrey A. Lieberman, Akira Saw, Eric J. Nestler, Marc G. Caron, Bryan L. Roth, P. Jeffrey Conn, Susan R. George, Stephen J. Haggarty, Stephen R. Marder, Mark A. Geyer, Amanda J. Law, Bita Moghaddam, Edward M. Scolnick, Schahram Akbarian, Robert W. Buchanan, Brian M. Campbell, Michelle Solis, Daniel R. Welnberger, and Maria Karayiorgou
- Subjects
medicine.medical_specialty ,Medical education ,business.industry ,Drug discovery ,General Neuroscience ,Schizophrenia (object-oriented programming) ,Alternative medicine ,Translational research ,behavioral disciplines and activities ,Mental health ,General Biochemistry, Genetics and Molecular Biology ,History and Philosophy of Science ,Vanguard ,medicine ,Schizophrenia research ,business ,Neuroscience - Abstract
Sponsored by the New York Academy of Sciences and with support from the National Institute of Mental Health, the Life Technologies Foundation, and the Josiah Macy Jr. Foundation, “Advancing Drug Discovery for Schizophrenia” was held March 9–11 at the New York Academy of Sciences in New York City. The meeting, comprising individual talks and panel discussions, highlighted basic, clinical, and translational research approaches, all of which contribute to the overarching goal of enhancing the pharmaceutical armamentarium for treating schizophrenia. This report surveys work by the vanguard of schizophrenia research in such topics as genetic and epigenetic approaches; small molecule therapeutics; and the relationships between target genes, neuronal function, and symptoms of schizophrenia.
- Published
- 2011
24. A Transient Inhibition and Permanent Lack of Catechol-O-Methyltransferase have Minor Effects on Feeding Pattern of Female Rodents
- Author
-
Nadia Schendzielorz, Joseph A. Gogos, Maria Karayiorgou, Atso Raasmaja, and Pekka T. Männistö
- Subjects
medicine.medical_specialty ,Biology ,Toxicology ,COMT inhibitor ,behavioral disciplines and activities ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Dopamine ,Internal medicine ,mental disorders ,medicine ,Dopamine metabolism ,Prefrontal cortex ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Meal ,Catechol-O-methyl transferase ,fungi ,Dopaminergic ,General Medicine ,Single injection ,Endocrinology ,nervous system ,chemistry ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Abnormal feeding behaviours have long been linked to disruptions in brain dopaminergic activity. Dopamine is metabolized, amongst others, by catechol-O-methyltransferase (COMT). Normally, COMT only plays a subordinate role in dopamine metabolism. However, changes in COMT activity, especially in the prefrontal cortex, become more important during events that evoke dopamine release. The current study investigated the effect of acute COMT inhibition on feeding in Wistar rats and C57BL/6 mice using a selective, brain penetrating COMT inhibitor (OR-1139). Furthermore, the effect of a long-term lack of COMT on feeding behaviour was studied in COMT-deficient (COMT −/−) mice. Apart from following the gross feeding behaviour of fasted rats and mice, the first 4 hr of re-feeding were recorded with a video camera to allow a more detailed analysis of feeding microstructure. In the acute study, rats and mice received a single injection of OR-1139 (3, 10 or 30 mg/kg), just before the experiment. We found that rats and mice receiving OR-1139 had fewer very short meals but more long meals than the controls. Treated mice even ate more frequently than the controls, but other feeding parameters remained unchanged. Conversely, COMT −/− mice displayed an increased latency to initiate the first meal and spent less total time eating than wild-type mice. In conclusion, although decreased/lack of COMT activity did not robustly alter feeding behaviour of female rodents, we observed some alterations in the microstructure of feeding. However, these minor changes were highly dependent on the extent and fashion in which COMT was manipulated.
- Published
- 2011
25. Exome sequencing supports a de novo mutational paradigm for schizophrenia
- Author
-
Shawn Levy, Brooks Plummer, Maria Karayiorgou, Phillip J. Dexheimer, Braden E. Boone, Joseph A. Gogos, J. Louw Roos, and Bin Xu
- Subjects
Chromosomes, Human, Pair 22 ,DNA Mutational Analysis ,Disease ,Biology ,medicine.disease_cause ,Article ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Family history ,Gene ,Exome sequencing ,Sequence Deletion ,030304 developmental biology ,0303 health sciences ,Mutation ,Membrane Glycoproteins ,Exons ,medicine.disease ,Platelet Glycoprotein GPIb-IX Complex ,Schizophrenia ,030217 neurology & neurosurgery - Abstract
Despite its high heritability, a large fraction of individuals with schizophrenia do not have a family history of the disease (sporadic cases). Here we examined the possibility that rare de novo protein-altering mutations contribute to the genetic component of schizophrenia by sequencing the exomes of 53 sporadic cases, 22 unaffected controls and their parents. We identified 40 de novo mutations in 27 cases affecting 40 genes, including a potentially disruptive mutation in DGCR2, a gene located in the schizophrenia-predisposing 22q11.2 microdeletion region. A comparison to rare inherited variants indicated that the identified de novo mutations show a large excess of non-synonymous changes in schizophrenia cases, as well as a greater potential to affect protein structure and function. Our analyses suggest a major role for de novo mutations in schizophrenia as well as a large mutational target, which together provide a plausible explanation for the high global incidence and persistence of the disease.
- Published
- 2011
26. Deficiency of Dgcr8 , a gene disrupted by the 22q11.2 microdeletion, results in altered short-term plasticity in the prefrontal cortex
- Author
-
Jun Mukai, Karine Fénelon, Maria Karayiorgou, Liam Drew, Gerald D. Fischbach, Amy B. MacDermott, Bin Xu, Pei Ken Hsu, and Joseph A. Gogos
- Subjects
Time Factors ,Dendritic spine ,Neural substrate ,Chromosomes, Human, Pair 22 ,Dendritic Spines ,Prefrontal Cortex ,Biology ,Mice ,Laminar organization ,Neuroplasticity ,Animals ,Prefrontal cortex ,CA1 Region, Hippocampal ,Genetics ,Neuronal Plasticity ,Multidisciplinary ,Working memory ,Excitatory Postsynaptic Potentials ,Proteins ,RNA-Binding Proteins ,Long-term potentiation ,Biological Sciences ,CA3 Region, Hippocampal ,Synapses ,Synaptic plasticity ,Chromosome Deletion ,Neuroscience ,Gene Deletion - Abstract
Individuals with 22q11.2 microdeletions have cognitive and behavioral impairments and the highest known genetic risk for developing schizophrenia. One gene disrupted by the 22q11.2 microdeletion is DGCR8 , a component of the “microprocessor” complex that is essential for microRNA production, resulting in abnormal processing of specific brain miRNAs and working memory deficits. Here, we determine the effect of Dgcr8 deficiency on the structure and function of cortical circuits by assessing their laminar organization, as well as the neuronal morphology, and intrinsic and synaptic properties of layer 5 pyramidal neurons in the prefrontal cortex of Dgcr8 +/− mutant mice. We found that heterozygous Dgcr8 mutant mice have slightly fewer cortical layer 2/4 neurons and that the basal dendrites of layer 5 pyramidal neurons have slightly smaller spines. In addition to the modest structural changes, field potential and whole-cell electrophysiological recordings performed in layer 5 of the prefrontal cortex revealed greater short-term synaptic depression during brief stimulation trains applied at 50 Hz to superficial cortical layers. This finding was accompanied by a decrease in the initial phase of synaptic potentiation. Our results identify altered short-term plasticity as a neural substrate underlying the cognitive dysfunction and the increased risk for schizophrenia associated with the 22q11.2 microdeletions.
- Published
- 2011
27. The 22q11.2 microdeletion: Fifteen years of insights into the genetic and neural complexity of psychiatric disorders
- Author
-
Liam Drew, Maria Karayiorgou, Florence Chaverneff, Jun Mukai, Kimberly L. Stark, Karine Fénelon, Gregg W. Crabtree, Joseph A. Gogos, Pei Ken Hsu, Sander Markx, and Bin Xu
- Subjects
Chromosomes, Human, Pair 22 ,Schizophrenia (object-oriented programming) ,Induced Pluripotent Stem Cells ,Population ,Disease ,Biology ,Catechol O-Methyltransferase ,Bioinformatics ,Article ,Developmental Neuroscience ,Chromosome (genetic algorithm) ,Proline Oxidase ,Animals ,Humans ,Genetic Predisposition to Disease ,education ,Genetics ,education.field_of_study ,Models, Genetic ,Mental Disorders ,Brain ,Epistasis, Genetic ,22q11 2 microdeletion ,Syndrome ,Disease Models, Animal ,MicroRNAs ,Homogeneous ,Mutation (genetic algorithm) ,Schizophrenia ,Etiology ,Chromosome Deletion ,Developmental Biology - Abstract
Over the last fifteen years it has become established that 22q11.2 deletion syndrome (22q11DS) is a true genetic risk factor for schizophrenia. Carriers of deletions in chromosome 22q11.2 develop schizophrenia at rate of 25–30% and such deletions account for as many as 1–2% of cases of sporadic schizophrenia in the general population. Access to a relatively homogeneous population of individuals that suffer from schizophrenia as the result of a shared etiological factor and the potential to generate etiologically valid mouse models provides an immense opportunity to better understand the pathobiology of this disease. In this review we survey the clinical literature associated with the 22q11.2 microdeletions with a focus on neuroanatomical changes. Then, we highlight results from work modeling this structural mutation in animals. The key biological pathways disrupted by the mutation are discussed and how these changes impact the structure and function of neural circuits is described.
- Published
- 2010
28. Quantitative role of COMT in dopamine clearance in the prefrontal cortex of freely moving mice
- Author
-
M. Käenmäki, Maria Karayiorgou, Anne Tammimäki, Carolina Louise Amberg, Kaisa Pakarinen, Pekka T. Männistö, Joseph A. Gogos, and Timo T. Myöhänen
- Subjects
0303 health sciences ,medicine.medical_specialty ,Microdialysis ,Catechol-O-methyl transferase ,biology ,Chemistry ,Striatum ,Nucleus accumbens ,Biochemistry ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Monoamine neurotransmitter ,Endocrinology ,nervous system ,Norepinephrine transporter ,Dopamine ,Internal medicine ,mental disorders ,biology.protein ,medicine ,030217 neurology & neurosurgery ,030304 developmental biology ,Dopamine transporter ,medicine.drug - Abstract
Catechol-O-methyltransferase (COMT) plays an active role in the metabolism of dopamine (DA) in the prefrontal cortex (PFC). Because of low levels of dopamine transporter (DAT), it is proposed that the majority of released DA is taken up by either norepinephrine transporter (NET) and subsequently metabolized by monoamine oxidize (MAO) or by uptake(2) (to glial cells and post-synaptic neurons) and metabolized by COMT. However, a comprehensive in vivo study of rating the mechanisms involved in DA clearance in the PFC has not been done. Here, we employ two types of microdialysis to study these pathways using DAT, NET and MAO blockers in conscious mice, with or without Comt gene disruption. In quantitative no-net-flux microdialysis, DA levels were increased by 60% in the PFC of COMT-knockout (ko) mice, but not in the striatum and nucleus accumbens. In conventional microdialysis studies, we showed that selective NET and MAO inhibition increased DA levels in the PFC of wild-type mice by two- to fourfold, an effect that was still doubled in COMT-ko mice. Inhibition of DAT had no effect on DA levels in either genotype. Therefore, we conclude that in the mouse, PFC COMT contributes about one half of the total DA clearance.
- Published
- 2010
29. S189. CARVING A MORE SPECIFIC SUBTYPE OF SCHIZOPHRENIA FOR GENETIC STUDIES: SPORADIC SCHIZOAFFECTIVE BIPOLAR TYPE
- Author
-
Rene Ehlers, Johannes L. Roos, Maria Karayiorgou, and Nicolaas Albertus Van der Merwe
- Subjects
Abstracts ,Poster Session III ,Psychiatry and Mental health ,medicine.medical_specialty ,Carving ,business.industry ,Schizophrenia (object-oriented programming) ,mental disorders ,Medicine ,business ,Psychiatry - Abstract
Background Schizophrenia is a heterogeneous group of disorders. The familial-sporadic distinction has been considered under a range of genetic models. Research supports a strong association of de novo copy mutations with sporadic schizophrenia. The aim of the study was to determine a more homogenous phenotype for genetic research via comparison of various clinical and socio-demographic variables in familial and sporadic schizophrenia. Methods A cross-sectional observational design was used. This study included 384 participants with schizophrenia/schizoaffective disorder from an Afrikaner founder population in South Africa that previously participated in genetic research. A comprehensive data capturing sheet was completed from a pre-existing database that contains information obtained from the Diagnostic Interview for Genetic Studies, chronological clinical summary reports and additional sources of information. The study protocol was approved by the Research Ethics Committee from the Faculty of Health Sciences at the University of Pretoria. Logit models were fitted using the backward elimination procedure to investigate relationships where the dependent variable was binary. Odds ratio estimates were calculated for independent variables that were significant in the model. The Kruskal-Wallis test was conducted to compare the means of groups. For cases where there were significant differences a post-hoc test with a Bonferroni correction was done to determine which groups differ significantly. Results There were 214 familial and 170 sporadic subjects. 279 had a diagnosis of schizophrenia, 66 schizoaffective, bipolar type and 39 schizoaffective disorder, depressive type. 242 were male and 142 female. The age at onset of the primary psychiatric diagnosis, season of birth, co-morbid diagnoses, symptomatology, suicidality history and marital status weren’t significantly different when considering the combined schizophrenia/schizoaffective disorder group and its relationship to familiality. Early deviant behaviour was however decreased in the sporadic group. These findings were replicated when analysing schizophrenia independently from schizoaffective disorder. The sporadic schizoaffective disorder, bipolar type did however have a significantly lower age at onset (mean 20.18 versus 25.07 years), 8.8 times more hallucinations, 6.6 times more odd behaviour before the age of 10 and were 2.8 times more likely to be single. The bipolar type also had 2.9 times more suicide attempts as opposed to ideation. This finding wasn’t statistically significant. The sporadic schizoaffective group overall was 2.2 times more likely to abuse substances. The depressive type didn’t differ significantly with regards to age at onset, season of birth, co-morbidities, early deviant behaviour, symptomatology, suicidality or marital status. Discussion The combined schizophrenia/schizoaffective group didn’t differ significantly, nor did schizophrenia and schizoaffective disorder depressive type when analysed independently. The sporadic schizoaffective bipolar type differed significantly on multiple important variables that suggest a poorer prognosis and increased disease severity. The sporadic schizoaffective bipolar type forms a more homogenous group, with genetic studies hinting at relatively specific genetic risk factors. Studies elucidating the genetic architecture of this group could prove invaluable in clarifying the aetiology of schizophrenia.
- Published
- 2018
30. Impaired hippocampal–prefrontal synchrony in a genetic mouse model of schizophrenia
- Author
-
Torfi Sigurdsson, Maria Karayiorgou, Joshua A. Gordon, Kimberly L. Stark, and Joseph A. Gogos
- Subjects
Male ,Psychosis ,Brain activity and meditation ,Chromosomes, Human, Pair 22 ,Models, Neurological ,Action Potentials ,Prefrontal Cortex ,Hippocampus ,Hippocampal formation ,Biology ,Bioinformatics ,Article ,Mice ,Memory ,medicine ,Biological neural network ,Animals ,Humans ,Genetic Predisposition to Disease ,Prefrontal cortex ,Alleles ,Multidisciplinary ,Behavior, Animal ,Models, Genetic ,Working memory ,medicine.disease ,Chromosomes, Mammalian ,Mice, Inbred C57BL ,Disease Models, Animal ,Schizophrenia ,Female - Abstract
Abnormalities in functional connectivity between brain areas have been postulated as an important pathophysiological mechanism underlying schizophrenia1,2. In particular, macroscopic measurements of brain activity in patients suggest that functional connectivity between the frontal and temporal lobes may be altered3,4. However, it remains unclear whether such dysconnectivity relates to the aetiology of the illness, and how it is manifested in the activity of neural circuits. Because schizophrenia has a strong genetic component5, animal models of genetic risk factors are likely to aid our understanding of the pathogenesis and pathophysiology of the disease. Here we study Df(16)A+/− mice, which model a microdeletion on human chromosome 22 (22q11.2) that constitutes one of the largest known genetic risk factors for schizophrenia6. To examine functional connectivity in these mice, we measured the synchronization of neural activity between the hippocampus and the prefrontal cortex during the performance of a task requiring working memory, which is one of the cognitive functions disrupted in the disease. In wild-type mice, hippocampal–prefrontal synchrony increased during working memory performance, consistent with previous reports in rats7. Df(16)A+/− mice, which are impaired in the acquisition of the task, showed drastically reduced synchrony, measured both by phase-locking of prefrontal cells to hippocampal theta oscillations and by coherence of prefrontal and hippocampal local field potentials. Furthermore, the magnitude of hippocampal–prefrontal coherence at the onset of training could be used to predict the time it took the Df(16)A+/− mice to learn the task and increased more slowly during task acquisition. These data suggest how the deficits in functional connectivity observed in patients with schizophrenia may be realized at the single-neuron level. Our findings further suggest that impaired long-range synchrony of neural activity is one consequence of the 22q11.2 deletion and may be a fundamental component of the pathophysiology underlying schizophrenia.
- Published
- 2010
31. Development of animal models for schizophrenia
- Author
-
Sander Markx, Maria Karayiorgou, P. Alexander Arguello, and Joseph A. Gogos
- Subjects
Genetics ,education.field_of_study ,Population ,Neuroscience (miscellaneous) ,Medicine (miscellaneous) ,Locus (genetics) ,Disease ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Disease Models, Animal ,Mice ,Phenotype ,Chromosome 16 ,Immunology and Microbiology (miscellaneous) ,Clinical Puzzle ,Schizophrenia ,Animals ,Human genome ,Copy-number variation ,Allele ,education ,Gene - Abstract
Schizophrenia is a devastating psychiatric disorder that affects around 1% of the population worldwide. The disease is characterized by ‘positive symptoms’, ‘negative symptoms’ and cognitive deficits. Over the last 60 years, a large number of family, twin and adoption studies have clearly demonstrated a strong genetic component for schizophrenia, but the mode of inheritance of the disease is complex and, in all likelihood, involves contribution from multiple genes in conjunction with environmental and stochastic factors. Recently, several genome-wide scans have demonstrated that rare alleles contribute significantly to schizophrenia risk. Assessments of rare variants have identified specific and probably causative, disease-associated structural mutations or copy number variants (CNVs, which result from genomic gains or losses). The fact that the effects of such lesions are transparent allows the generation of etiologically valid animal models and the opportunity to explore the molecular, cellular and circuit-level abnormalities underlying the expression of psychopathology. To date, the most common genomic structural rearrangements that are unequivocally associated with the development of schizophrenia, are de novo microdeletions of the 22q11.2 locus. Fortunately, the human 22q11.2 locus is conserved within the syntenic region of mouse chromosome 16, which harbors nearly all orthologues of the human genes. This has made it possible to engineer genetically faithful, and thus etiologically valid, animal models of this schizophrenia susceptibility locus.
- Published
- 2010
32. Importance of membrane-bound catechol-O-methyltransferase in L-DOPA metabolism: a pharmacokinetic study in two types of Comt gene modified mice
- Author
-
Timo T. Myöhänen, Maria Karayiorgou, Pekka T. Männistö, J.A. Garcia-Horsman, Nadia Schendzielorz, M. Käenmäki, Joseph A. Gogos, and Anne Tammimäki
- Subjects
Pharmacology ,0303 health sciences ,medicine.medical_specialty ,Catechol-O-methyl transferase ,Methyltransferase ,Ratón ,fungi ,Biology ,behavioral disciplines and activities ,Isozyme ,3. Good health ,Blot ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,nervous system ,Pharmacokinetics ,Dopamine ,Internal medicine ,Carbidopa ,mental disorders ,medicine ,030217 neurology & neurosurgery ,030304 developmental biology ,medicine.drug - Abstract
Background and purpose: Catechol-O-methyltransferase (COMT) metabolizes compounds containing catechol structures and has two forms: soluble (S-COMT) and membrane-bound (MB-COMT). Here we report the generation of a mouse line that expresses MB-COMT but not S-COMT. We compared the effects of deleting S-COMT only or both COMT forms on the pharmacokinetics of oral L-DOPA. Experimental approach: L-DOPA (10 mg·kg−1) and carbidopa (30 mg·kg−1) were given to mice by gastric tube, and samples were taken at various times. HPLC was used to measure L-DOPA in plasma and tissue samples, and dopamine and its metabolites in brain. Immunohistochemistry and Western blotting were used to characterize the distribution of COMT protein isoforms. Key results: Lack of S-COMT did not affect the levels of L-DOPA in plasma or peripheral tissues, whereas in the full COMT-knock-out mice, these levels were increased. The levels of 3-O-methyldopa were significantly decreased in the S-COMT-deficient mice. In the brain, L-DOPA levels were not significantly increased, and dopamine was increased only in females. The total COMT activity in the S-COMT-deficient mice was 22–47% of that in the wild-type mice. In peripheral tissues, female mice had lower COMT activity than the males. Conclusions and implications: In S-COMT-deficient mice, MB-COMT in the liver and the duodenum is able to O-methylate about one-half of exogenous L-DOPA. Sexual dimorphism and activity of the two COMT isoforms seems to be tissue specific and more prominent in peripheral tissues than in the brain.
- Published
- 2009
33. Elucidating the genetic architecture of familial schizophrenia using rare copy number variant and linkage scans
- Author
-
Gonçalo R. Abecasis, Laura Rodriguez-Murillo, Elizabeth J. van Rensburg, Abigail Woodroffe, Bin Xu, Joseph A. Gogos, J. Louw Roos, and Maria Karayiorgou
- Subjects
Genetics ,education.field_of_study ,Multidisciplinary ,Genetic Linkage ,Genome, Human ,Population ,Gene Dosage ,Genetic Variation ,Chromosome ,Locus (genetics) ,Biological Sciences ,Biology ,Genetic architecture ,Cohort Studies ,South Africa ,Genetic linkage ,Genetic variation ,Schizophrenia ,Humans ,Human genome ,Copy-number variation ,education - Abstract
To elucidate the genetic architecture of familial schizophrenia we combine linkage analysis with studies of fine-level chromosomal variation in families recruited from the Afrikaner population in South Africa. We demonstrate that individually rare inherited copy number variants (CNVs) are more frequent in cases with familial schizophrenia as compared to unaffected controls and affect almost exclusively genic regions. Interestingly, we find that while the prevalence of rare structural variants is similar in familial and sporadic cases, the type of variants is markedly different. In addition, using a high-density linkage scan with a panel of nearly 2,000 markers, we identify a region on chromosome 13q34 that shows genome-wide significant linkage to schizophrenia and show that in the families not linked to this locus, there is evidence for linkage to chromosome 1p36. No causative CNVs were identified in either locus. Overall, our results from approaches designed to detect risk variants with relatively low frequency and high penetrance in a well-defined and relatively homogeneous population, provide strong empirical evidence supporting the notion that multiple genetic variants, including individually rare ones, that affect many different genes contribute to the genetic risk of familial schizophrenia. They also highlight differences in the genetic architecture of the familial and sporadic forms of the disease.
- Published
- 2009
34. Strong association of de novo copy number mutations with sporadic schizophrenia
- Author
-
Joseph A. Gogos, J. Louw Roos, Bin Xu, E. J. Van Rensburg, Maria Karayiorgou, and Shawn Levy
- Subjects
Male ,Psychosis ,Genotype ,Genetic Linkage ,Population ,Gene Dosage ,Inheritance Patterns ,Biology ,medicine.disease_cause ,Germline mutation ,Gene Frequency ,Genetic linkage ,mental disorders ,Genetics ,medicine ,Chromosomes, Human ,Humans ,Family ,Genetic Testing ,education ,Germ-Line Mutation ,Oligonucleotide Array Sequence Analysis ,Genetic testing ,education.field_of_study ,Mutation ,medicine.diagnostic_test ,Genetic heterogeneity ,Chromosome Mapping ,medicine.disease ,Genetics, Population ,Schizophrenia ,Female ,Algorithms - Abstract
Schizophrenia is an etiologically heterogeneous psychiatric disease, which exists in familial and nonfamilial (sporadic) forms. Here, we examine the possibility that rare de novo copy number (CN) mutations with relatively high penetrance contribute to the genetic component of schizophrenia. We carried out a whole-genome scan and implemented a number of steps for finding and confirming CN mutations. Confirmed de novo mutations were significantly associated with schizophrenia (P = 0.00078) and were collectively approximately 8 times more frequent in sporadic (but not familial) cases with schizophrenia than in unaffected controls. In comparison, rare inherited CN mutations were only modestly enriched in sporadic cases. Our results suggest that rare de novo germline mutations contribute to schizophrenia vulnerability in sporadic cases and that rare genetic lesions at many different loci can account, at least in part, for the genetic heterogeneity of this disease.
- Published
- 2008
35. Altered brain microRNA biogenesis contributes to phenotypic deficits in a 22q11-deletion mouse model
- Author
-
Xiang Wan, Kimberly L. Stark, Wen-Sung Lai, Maria Karayiorgou, Alea A. Mills, Anindya Bagchi, Joseph A. Gogos, Bin Xu, Ruby Hsu, Paul Pavlidis, and Hui Liu
- Subjects
Male ,Heterozygote ,Chromosomes, Human, Pair 22 ,ved/biology.organism_classification_rank.species ,Locus (genetics) ,Biology ,Mice ,DGCR8 Gene ,RNA interference ,microRNA ,Genetics ,Animals ,Humans ,Gene silencing ,Habituation, Psychophysiologic ,Model organism ,Oligonucleotide Array Sequence Analysis ,Mice, Knockout ,Behavior, Animal ,Learning Disabilities ,ved/biology ,Gene Expression Profiling ,Brain ,Proteins ,RNA-Binding Proteins ,Spine ,Mice, Inbred C57BL ,Disease Models, Animal ,MicroRNAs ,Phenotype ,Sensation Disorders ,Female ,Chromosome Deletion ,Cognition Disorders ,Haploinsufficiency ,Biogenesis - Abstract
Individuals with 22q11.2 microdeletions show behavioral and cognitive deficits and are at high risk of developing schizophrenia. We analyzed an engineered mouse strain carrying a chromosomal deficiency spanning a segment syntenic to the human 22q11.2 locus. We uncovered a previously unknown alteration in the biogenesis of microRNAs (miRNAs) and identified a subset of brain miRNAs affected by the microdeletion. We provide evidence that the abnormal miRNA biogenesis emerges because of haploinsufficiency of the Dgcr8 gene, which encodes an RNA-binding moiety of the 'microprocessor' complex and contributes to the behavioral and neuronal deficits associated with the 22q11.2 microdeletion.
- Published
- 2008
36. Exploratory and habituation phenotype of heterozygous and homozygous COMT knockout mice
- Author
-
Jeremiah J Clifford, Maria Karayiorgou, David T. Croke, Joseph A. Gogos, Orna Tighe, Colm M. P. O’Tuathaigh, Gerard J. O'Sullivan, Daniela Babovic, David Cotter, and John L. Waddington
- Subjects
Male ,Heterozygote ,Psychosis ,Motor Activity ,Catechol O-Methyltransferase ,behavioral disciplines and activities ,Mice ,Behavioral Neuroscience ,Dopamine ,mental disorders ,medicine ,Animals ,Habituation, Psychophysiologic ,Prefrontal cortex ,Mice, Knockout ,Genetics ,Analysis of Variance ,Catechol-O-methyl transferase ,Behavior, Animal ,Homozygote ,fungi ,Heterozygote advantage ,medicine.disease ,Phenotype ,nervous system ,Endophenotype ,Knockout mouse ,Exploratory Behavior ,Female ,Psychology ,medicine.drug - Abstract
Catechol-O-methyltransferase (COMT) inactivates dopamine in prefrontal cortex and is associated clinically with a schizophrenia endophenotype. Using an ethologically based approach, the phenotype of mice with heterozygous COMT deletion was characterised by decreased rearing with increased sifting and chewing. Heterozygous COMT deletion is associated with a distinctive phenotype. This differs from that which we have reported previously for heterozygous deletion of the schizophrenia risk gene neuregulin-1.
- Published
- 2007
37. Site-Specific Role of Catechol-O-Methyltransferase in Dopamine Overflow within Prefrontal Cortex and Dorsal Striatum
- Author
-
Maria Karayiorgou, Markus M. Forsberg, Pekka T. Männistö, Leonid Yavich, and Joseph A. Gogos
- Subjects
Male ,Dopamine ,Caudate nucleus ,Prefrontal Cortex ,Striatum ,In Vitro Techniques ,Nucleus accumbens ,Catechol O-Methyltransferase ,behavioral disciplines and activities ,Reuptake ,Mice ,mental disorders ,medicine ,Animals ,Prefrontal cortex ,Mice, Knockout ,Binding Sites ,Catechol-O-methyl transferase ,Chemistry ,General Neuroscience ,fungi ,Extracellular Fluid ,Articles ,Corpus Striatum ,Mice, Inbred C57BL ,nervous system ,Knockout mouse ,Neuroscience ,medicine.drug - Abstract
Accumulating evidence from clinical and preclinical studies shows that catechol-O-methyltransferase (COMT) plays a significant role in dopamine metabolism in the prefrontal cortex, but not in the striatum. However, to what extent dopamine overflow in the prefrontal cortex and striatum is controlled by enzymatic degradation versus reuptake is unknown. We used COMT deficient mice to investigate the role of COMT in these two brain regions within vivovoltammetry. A real-time analysis of evoked dopamine overflow showed that removal of dopamine was twofold slower in the prefrontal cortex of mice lacking COMT than in wild-type mice, indicating that half of the dopamine decline in this brain region results from COMT-mediated enzymatic degradation. Lack of COMT did not influence dopamine overflow/decline in the dorsal striatum. COMT-deficient mice demonstrated a small (20–25%) but consistent increase in evoked dopamine release in the prefrontal cortex, but not in the dorsal striatum. Cocaine affected equally dopaminergic neurotransmission in the prefrontal cortex in both genotypes by prolonging 3–4 times dopamine elimination from extracellular space. Paradoxically, this happened without increase of the peak levels of evoked dopamine release. The present findings represent the first demonstration of the significant contribution of COMT in modulating the dynamics of dopamine overflow in the prefrontal cortex and underscore the therapeutic potential of manipulating COMT activity to alter dopaminergic neurotransmission in the prefrontal cortex.
- Published
- 2007
38. Cobalt(III) Protoporphyrin Activates the DGCR8 Protein and Can Compensate microRNA Processing Deficiency
- Author
-
Joseph A. Gogos, Sara H. Weitz, Talia Atkin, Feng Guo, Pei Ken Hsu, Maria Karayiorgou, Ian G. Barr, and Shimon Weiss
- Subjects
Clinical Biochemistry ,Post-Transcriptional ,Protoporphyrins ,Inbred C57BL ,Biochemistry ,Microprocessor complex ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Coordination Complexes ,Drug Discovery ,RNA Processing, Post-Transcriptional ,Heme ,Cells, Cultured ,Mice, Knockout ,Neurons ,0303 health sciences ,Cultured ,biology ,Protoporphyrin IX ,RNA-Binding Proteins ,General Medicine ,Cobalt ,Recombinant Proteins ,Molecular Medicine ,Biotechnology ,Protein Binding ,RNA Processing ,RNase P ,DGCR8 ,Cells ,Knockout ,1.1 Normal biological development and functioning ,Article ,Medicinal and Biomolecular Chemistry ,03 medical and health sciences ,Underpinning research ,microRNA ,Genetics ,Animals ,Humans ,Molecular Biology ,Drosha ,030304 developmental biology ,Pharmacology ,Organic Chemistry ,Neurosciences ,Molecular biology ,Mice, Inbred C57BL ,MicroRNAs ,chemistry ,Hela Cells ,biology.protein ,Protoporphyrin ,Biochemistry and Cell Biology ,030217 neurology & neurosurgery ,HeLa Cells - Abstract
© 2015 Elsevier Ltd All rights reserved. Summary Processing of microRNA primary transcripts (pri-miRNAs) is highly regulated and defects in the processing machinery play a key role in many human diseases. In 22q11.2 deletion syndrome (22q11.2DS), heterozygous deletion of DiGeorge critical region gene 8 (DGCR8) causes a processing deficiency, which contributes to abnormal brain development. The DGCR8 protein is the RNA-binding partner of Drosha RNase, both essential for processing canonical pri-miRNAs. To identify an agent that can compensate reduced DGCR8 expression, we screened for metalloporphyrins that can mimic the natural DGCR8 heme cofactor. We found that Co(III) protoporphyrin IX (PPIX) stably binds DGCR8 and activates it for pri-miRNA processing in vitro and in HeLa cells. Importantly, treating cultured Dgcr8+/-mouse neurons with Co(III)PPIX can compensate the pri-miRNA processing defects. Co(III)PPIX is effective at concentrations as low as 0.2 μM and is not degraded by heme degradation enzymes, making it useful as a research tool and a potential therapeutic.
- Published
- 2015
39. The BDNF Val66Met Variant Affects Gene Expression through miR-146b
- Author
-
Jun Mukai, Joseph A. Gogos, Bin Xu, Pei Ken Hsu, and Maria Karayiorgou
- Subjects
Molecular Sequence Data ,Single-nucleotide polymorphism ,Mice, Transgenic ,Biology ,In Vitro Techniques ,Hippocampus ,Article ,lcsh:RC321-571 ,Mice ,Methionine ,Val66Met ,Gene knockin ,Genetic variation ,Gene expression ,microRNA ,Basic Helix-Loop-Helix Transcription Factors ,Knock-in mice ,Animals ,Humans ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Gene ,Genetics ,Brain-Derived Neurotrophic Factor ,MicroRNA ,IRAK1 ,Valine ,Period Circadian Proteins ,Gene expression profiling ,MicroRNAs ,BDNF ,Interleukin-1 Receptor-Associated Kinases ,Neurology ,Gene Expression Regulation - Abstract
Variation in gene expression is an important mechanism underlying susceptibility to complex disease and traits. Single nucleotide polymorphisms (SNPs) account for a substantial portion of the total detected genetic variation in gene expression but how exactly variants acting in trans modulate gene expression and disease susceptibility remains largely unknown. The BDNF Val66Met SNP has been associated with a number of psychiatric disorders such as depression, anxiety disorders, schizophrenia and related traits. Using global microRNA expression profiling in hippocampus of humanized BDNF Val66Met knock-in mice we showed that this variant results in dysregulation of at least one microRNA, which in turn affects downstream target genes. Specifically, we show that reduced levels of miR-146b (mir146b), lead to increased Per1 and Npas4 mRNA levels and increased Irak1 protein levels in vitro and are associated with similar changes in the hippocampus of hBDNF(Met/Met) mice. Our findings highlight trans effects of common variants on microRNA-mediated gene expression as an integral part of the genetic architecture of complex disorders and traits.
- Published
- 2015
40. Deletion of Rapgef6, a candidate schizophrenia susceptibility gene, disrupts amygdala function in mice
- Author
-
Yan Li, Joseph A. Gogos, Yoko Yoshikawa, Rebecca J. Levy, Maria Karayiorgou, W. Dong, Vadim Y. Bolshakov, Evgeny Tsvetkov, Mirna Kvajo, and Tohru Kataoka
- Subjects
Dendritic spine ,DNA Copy Number Variations ,Dendritic Spines ,Long-Term Potentiation ,Hippocampus ,Prefrontal Cortex ,Hippocampal formation ,Anxiety ,Amygdala ,Cellular and Molecular Neuroscience ,Mice ,Conditioning, Psychological ,medicine ,Animals ,Guanine Nucleotide Exchange Factors ,Fear conditioning ,Phosphorylation ,Prefrontal cortex ,Biological Psychiatry ,Spatial Memory ,Mice, Knockout ,Pyramidal Cells ,Long-term potentiation ,Fear ,CA3 Region, Hippocampal ,Psychiatry and Mental health ,medicine.anatomical_structure ,Memory, Short-Term ,Knockout mouse ,Schizophrenia ,Original Article ,Psychology ,Neuroscience ,Proto-Oncogene Proteins c-fos - Abstract
In human genetic studies of schizophrenia, we uncovered copy-number variants in RAPGEF6 and RAPGEF2 genes. To discern the effects of RAPGEF6 deletion in humans, we investigated the behavior and neural functions of a mouse lacking Rapgef6. Rapgef6 deletion resulted in impaired amygdala function measured as reduced fear conditioning and anxiolysis. Hippocampal-dependent spatial memory and prefrontal cortex-dependent working memory tasks were intact. Neural activation measured by cFOS phosphorylation demonstrated a reduction in hippocampal and amygdala activation after fear conditioning, while neural morphology assessment uncovered reduced spine density and primary dendrite number in pyramidal neurons of the CA3 hippocampal region of knockout mice. Electrophysiological analysis showed enhanced long-term potentiation at cortico–amygdala synapses. Rapgef6 deletion mice were most impaired in hippocampal and amygdalar function, brain regions implicated in schizophrenia pathophysiology. The results provide a deeper understanding of the role of the amygdala in schizophrenia and suggest that RAPGEF6 may be a novel therapeutic target in schizophrenia.
- Published
- 2015
41. Akt1 deficiency affects neuronal morphology and predisposes to abnormalities in prefrontal cortex functioning
- Author
-
Wen-Sung Lai, Marta Paterlini, Paul Pavlidis, Maria Karayiorgou, Berend Olivier, Koen G.C. Westphal, Bin Xu, Joseph A. Gogos, Dierexperimenteel, and Dep Farmaceutische wetenschappen
- Subjects
Male ,Prefrontal Cortex ,AKT1 ,Mice, Transgenic ,Biology ,Pathogenesis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Neurochemical ,Memory ,Animals ,Permissive ,Maze Learning ,Prefrontal cortex ,Protein kinase B ,030304 developmental biology ,Mice, Knockout ,Neurons ,0303 health sciences ,Multidisciplinary ,Working memory ,Gene Expression Profiling ,Biological Sciences ,Mice, Inbred C57BL ,Gene expression profiling ,nervous system ,Proto-Oncogene Proteins c-akt ,Neuroscience ,030217 neurology & neurosurgery - Abstract
There is accumulating evidence that AKT signaling plays a role in the pathogenesis of schizophrenia. We asked whether Akt1 deficiency in mice results in structural and functional abnormalities in prefrontal cortex (PFC). Exploratory transcriptional profiling revealed concerted alterations in the expression of PFC genes controlling synaptic function, neuronal development, myelination, and actin polymerization, and follow-up ultrastructural analysis identified consistent changes in the dendritic architecture of pyramidal neurons. Behavioral analysis indicated that Akt1 -mutant mice have normal acquisition of a PFC-dependent cognitive task but abnormal working memory retention under neurochemical challenge of three distinct neurotransmitter systems. Thus, Akt1 deficiency creates a context permissive for gene–gene and gene–environment interactions that modulate PFC functioning and contribute to the disease risk associated with this locus, the severity of the clinical syndrome, or both.
- Published
- 2006
42. Olfactory Disorder in Children With 22q11 Deletion Syndrome
- Author
-
Christina Sobin, Samantha Hadley Monk, Karen Kiley-Brabeck, Maria Karayiorgou, Jananne Khuri, and Kathryn A. Dale
- Subjects
Male ,medicine.medical_specialty ,Velopharyngeal Insufficiency ,Adolescent ,Chromosomes, Human, Pair 22 ,Olfaction ,Article ,Olfaction Disorders ,22q11 Deletion Syndrome ,Hyposmia ,Dopamine ,DiGeorge syndrome ,Internal medicine ,DiGeorge Syndrome ,medicine ,Humans ,Child ,business.industry ,Age Factors ,Case-control study ,Syndrome ,medicine.disease ,Phenotype ,Endocrinology ,Odor ,Case-Control Studies ,Child, Preschool ,Odorants ,Pediatrics, Perinatology and Child Health ,Immunology ,Female ,Chromosome Deletion ,medicine.symptom ,business ,medicine.drug - Abstract
OBJECTIVE. 22q11 deletion syndrome, a common human interstitial deletion syndrome (1:5000), is associated with a heterogeneous physical phenotype, including several factors that markedly increase the risk for olfactory disorder. Despite its potential consequences, pediatric studies of impaired olfaction are rare, and odor detection in children with 22q11 deletion syndrome has not yet been examined.METHODS. The University of Pennsylvania Smell Identification Test was administered to 62 children, including 39 with 22q11 deletion syndrome and 23 neurotypical control siblings who ranged in age from 5.3 to 14.8 years. Lowered smell detection accuracy among affected children was predicted.RESULTS. Substantially more children with 22q11 deletion syndrome (68%) as compared with neurotypical control subjects (13%) had University of Pennsylvania Smell Identification Test scores ≥2 SDs below the standardization sample mean. Frequency of impairment in younger versus older children did not differ. The score distributions of children with and without velopharyngeal insufficiency did not differ; however, markedly lower score variance among children with velopharyngeal insufficiency suggested its negative impact on olfaction. Posthoc error analyses revealed that affected children had special difficulty detecting smells that are associated with fumes and smoke.CONCLUSIONS. Odor detection failures are ubiquitous among children with 22q11 deletion syndrome and are not associated with developmental delay or performance characteristics of younger affected children. Additional studies are needed to examine further the impact on olfaction of velopharyngeal insufficiency and compromised nasal airway patency. Children with 22q11 deletion syndrome should be evaluated routinely for olfactory disorder. When deficits are identified, caregivers should be warned of potential dangers that are associated with this type of sensory impairment.
- Published
- 2006
43. Schizophrenia genetics: uncovering positional candidate genes
- Author
-
Joseph A. Gogos and Maria Karayiorgou
- Subjects
Genetics ,Psychosis ,Candidate gene ,Genetic Linkage ,Mechanism (biology) ,Schizophrenia (object-oriented programming) ,Chromosome Mapping ,Neuropathology ,Biology ,medicine.disease ,Genetic determinism ,Gene mapping ,mental disorders ,Schizophrenia ,medicine ,Humans ,Genetic Predisposition to Disease ,Cloning, Molecular ,Causes of schizophrenia ,Alleles ,Genetics (clinical) - Abstract
The efforts to decipher the genetic causes of schizophrenia, one of the most devastating mental illnesses, have reached a turning point. Several linkage findings in schizophrenia have been replicated and, in the last few years, have been followed by systematic fine-mapping efforts to identify positional susceptibility genes. Here, we outline the evidence supporting each of the proposed positional candidate genes and identify some general areas of caution in their interpretation. Several of these findings hold considerable promise both for understanding the neuropathology of this brain disorder, the causes of which remain a mystery, but also for development of novel, mechanism-based treatments for the patients.
- Published
- 2006
44. Neuromotor deficits in children with the 22q11 deletion syndrome
- Author
-
Jananne Khuri, Christina Sobin, Karen Kiley-Brabeck, Maria Karayiorgou, and Samantha Hadley Monk
- Subjects
Male ,medicine.medical_specialty ,Movement disorders ,Adolescent ,Chromosomes, Human, Pair 22 ,Developmental Disabilities ,Intelligence ,Chromosome Disorders ,Audiology ,Article ,Sex Factors ,22q11 Deletion Syndrome ,medicine ,Humans ,Longitudinal Studies ,Age of Onset ,Child ,Psychiatry ,Chi-Square Distribution ,Movement Disorders ,Intelligence quotient ,Siblings ,Age Factors ,Neurology ,Chromosomal deletion syndrome ,El Niño ,Child, Preschool ,Disease Progression ,Etiology ,Female ,Neurology (clinical) ,Chromosome Deletion ,medicine.symptom ,Age of onset ,Psychology ,Neurocognitive - Abstract
The 22q11 chromosomal deletion syndrome (22q11DS) is associated with a heterogeneous physical phenotype, neurocognitive deficits, and increased risk of later psychiatric illness. Sporadic clinical reports suggested motor differences, but quantitative studies of movement in children with 22q11DS are rare. If present in a majority of affected school-age children, characterization of neuromotor deficits may prove to be critical for intervention, neurocognitive test interpretation, and understanding etiology. We administered the Movement Assessment Battery for Children to 72 children ages 4.3 to 16.1, including 49 children confirmed positive for the 22q11 deletion and 23 control siblings. We predicted a higher frequency of global and domain impairment in manual dexterity, eye-hand coordination, and balance among affected children. Ninety-four percent of affected children had marked neuromotor deficits, and group scores differed broadly for both global and subarea measures. Secondary analyses showed no impairment differences between younger and older children with 22q11DS, and longitudinal trajectories for 12 affected children suggested stability of deficits over 3-year intervals. Neuromotor deficits in children with 22q11DS occur early in development, continue throughout the school-age years, should be considered in the interpretation of motor-based achievement and IQ tests, and require targeted and ongoing remediation throughout childhood and adolescence. Further studies examining the specificity of motor impairment to 22q11DS are needed.
- Published
- 2006
45. Transcriptional and behavioral interaction between 22q11.2 orthologs modulates schizophrenia-related phenotypes in mice
- Author
-
Stanislav S. Zakharenko, Maria Karayiorgou, Jun Mukai, Koen G.C. Westphal, Marta Paterlini, Steven A. Siegelbaum, Berend Olivier, Joseph A. Gogos, Hui Zhang, Wen-Sung Lai, David Sulzer, Paul Pavlidis, and Jie Qin
- Subjects
Transcriptional Activation ,Chromosomes, Human, Pair 22 ,Locus (genetics) ,Mice ,chemistry.chemical_compound ,Dopamine ,Proline Oxidase ,medicine ,Animals ,Humans ,Maze Learning ,Neurotransmitter ,Gene ,Behavior, Animal ,General Neuroscience ,Neurodegeneration ,Dopaminergic ,Excitatory Postsynaptic Potentials ,medicine.disease ,Chromosomes, Mammalian ,Phenotype ,Frontal Lobe ,Associative learning ,Disease Models, Animal ,Animals, Newborn ,chemistry ,Dopamine Agonists ,Schizophrenia ,Central Nervous System Stimulants ,Psychology ,Neuroscience ,medicine.drug - Abstract
Microdeletions of 22q11.2 represent one of the highest known genetic risk factors for schizophrenia. It is likely that more than one gene contributes to the marked risk associated with this locus. Two of the candidate risk genes encode the enzymes proline dehydrogenase (PRODH) and catechol-O-methyltransferase (COMT), which modulate the levels of a putative neuromodulator (L-proline) and the neurotransmitter dopamine, respectively. Mice that model the state of PRODH deficiency observed in humans with schizophrenia show increased neurotransmitter release at glutamatergic synapses as well as deficits in associative learning and response to psychomimetic drugs. Transcriptional profiling and pharmacological manipulations identified a transcriptional and behavioral interaction between the Prodh and Comt genes that is likely to represent a homeostatic response to enhanced dopaminergic signaling in the frontal cortex. This interaction modulates a number of schizophrenia-related phenotypes, providing a framework for understanding the high disease risk associated with this locus, the expression of the phenotype, or both.
- Published
- 2005
46. Lower Prepulse Inhibition in Children With the 22q11 Deletion Syndrome
- Author
-
Christina Sobin, Maria Karayiorgou, and Karen Kiley-Brabeck
- Subjects
Adult ,Male ,Reflex, Startle ,Adolescent ,Chromosomes, Human, Pair 22 ,Developmental Disabilities ,Population ,Article ,Craniofacial Abnormalities ,22q11 Deletion Syndrome ,DiGeorge syndrome ,DiGeorge Syndrome ,Reaction Time ,medicine ,Humans ,Abnormalities, Multiple ,Child ,education ,Prepulse inhibition ,Psychiatric Status Rating Scales ,education.field_of_study ,Blinking ,Learning Disabilities ,Working memory ,Siblings ,Neuropsychology ,Syndrome ,medicine.disease ,Startle reaction ,Psychiatry and Mental health ,Acoustic Stimulation ,Schizophrenia ,Female ,Psychology ,Neuroscience ,Gene Deletion - Abstract
The 22q11 deletion syndrome is associated with a range of possible physical anomalies, probable ongoing learning disabilities, and a specific constellation of neuropsychological deficits, including impairments in selective and executive visual attention, working memory, and sensorimotor functioning. It has been estimated that 25% of the children with 22q11 deletion syndrome go on to develop schizophrenia in late adolescence or adulthood. This is of urgent concern. Specification of early brain network vulnerabilities may provide a basis for early intervention while indicating critical links between genes and severe psychiatric illness. Neuropsychological studies of children with 22q11 deletion syndrome have implicated an array of potentially aberrant brain pathways. This study was conducted to determine whether preattentive processing ("sensorimotor gating") deficits are present in this population.The authors administered a test of prepulse inhibition to 25 children with 22q11 deletion syndrome and their 23 sibling comparison subjects, ages 6-13. It was predicted that the children with 22q11 deletion syndrome would have lower prepulse inhibition than the comparison subjects.Prepulse inhibition in the children with 22q11 deletion syndrome (26.06%) was significantly less than that of the sibling comparison subjects (46.41%). Secondary analyses suggested that this decrement did not reflect developmental delay, and lower prepulse inhibition was associated with particular subsyndromal symptoms in some children.Sensorimotor gating is lower in children with 22q11 deletion syndrome. These findings may indicate specific brain circuits that are anomalous in 22q11 deletion syndrome.
- Published
- 2005
47. NEUROPSYCHOLOGICAL CHARACTERISTICS OF CHILDREN WITH THE 22Q11 DELETION SYNDROME: A DESCRIPTIVE ANALYSIS
- Author
-
Maria Karayiorgou, Christina Sobin, Kwame Anyane-Yeboa, Karen Kiley-Brabeck, Lisa Taylor, Maude L. Blundell, Sarah Daniels, and Jananne Khuri
- Subjects
Male ,medicine.medical_specialty ,Neuropsychological function ,Chromosomes, Human, Pair 22 ,Stanford-Binet Test ,Audiology ,Severity of Illness Index ,Article ,Developmental psychology ,22q11 Deletion Syndrome ,DiGeorge Syndrome ,Developmental and Educational Psychology ,medicine ,Humans ,Cognitive skill ,Child ,Memory Disorders ,Descriptive statistics ,Working memory ,Neuropsychology ,Brain ,Neuropsychology and Physiological Psychology ,Child, Preschool ,Pediatrics, Perinatology and Child Health ,Female ,Verbal memory ,Cognition Disorders ,Psychology ,Neurocognitive ,Gene Deletion ,Follow-Up Studies - Abstract
Previous reports of cognitive functioning in children with the 22q11 Deletion Syndrome have reported marked variability in IQ and achievement subtest scores. Studies have begun to explore neuropsychological function in 22q11 DS however results are inconsistent and the profile incomplete. We assessed 40 children ages 5–12 with 22q11 DS. Consistent with past results, visual-spatial memory was significantly lower than verbal memory. Differentially lowered scores were found only in visual attention, working memory and motor function. Contrary with some past results quantitative, verbal ability, and visual spatial memory scores were within 1 SD from the standardization sample mean. Motor behavior, not typically discussed with regard to 22q11 DS school-age children, may be critical to incorporate in neurocognitive studies of children with 22q11 DS. Implications of these findings are considered with regard to past results.
- Published
- 2005
48. The molecular genetics of the 22q11-associated schizophrenia
- Author
-
Maria Karayiorgou and Joseph A. Gogos
- Subjects
Genetics ,Psychosis ,medicine.medical_specialty ,Candidate gene ,Genetic Linkage ,Chromosomes, Human, Pair 22 ,Epigenetics of schizophrenia ,Locus (genetics) ,Biology ,medicine.disease ,Contiguous gene syndrome ,Cellular and Molecular Neuroscience ,Genetic linkage ,Molecular genetics ,mental disorders ,Schizophrenia ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Protein palmitoylation ,Molecular Biology - Abstract
Schizophrenia has a strong genetic component but the mode of inheritance of the disease is complex and in all likelihood involves interaction among multiple genes and also possibly environmental or stochastic factors. A number of studies have shown that the 22q11 deletion syndrome (22q11DS) is a true genetic subtype of schizophrenia and as such may play an extremely important role in deciphering the genetic basis of schizophrenia. Microdeletions of the 22q11 locus are associated with a staggering increased risk to develop schizophrenia. The same locus has also been implicated by some linkage studies. Systematic examination of individual genes from the 1.5 Mb critical region has identified so far the PRODH and ZDHHC8 as strong candidate schizophrenia susceptibility genes from this locus. Discovery of these genes implicates neuromodulatory aminoacids and protein palmitoylation as important for disease development. Other genes, including the gene encoding for COMT, have been implicated by candidate gene approaches. It therefore appears that the 22q11-associated schizophrenia may have the characteristics of a contiguous gene syndrome, where deficiency in more than one gene contributes to the strikingly increased disease risk. Mouse models for individual candidate genes will provide the investigators with the opportunity to start understanding the function of these genes and how they may impact on schizophrenia. Mouse models that carry long-range deletions will likely capture the interactions among the culprit genes and help explain the genetic contribution of this locus to the high risk for schizophrenia. In-depth human and animal model studies of 22q11DS promise to answer critical questions relating to the devastating illness of schizophrenia, whose causes remain largely unknown.
- Published
- 2004
49. Networks of Attention in Children With the 22q11 Deletion Syndrome
- Author
-
Maude L. Blundell, Karen Kiley-Brabeck, Kwame Anyane-Yeboa, Maria Karayiorgou, Christina Sobin, and Sarah Daniels
- Subjects
Male ,medicine.medical_specialty ,Chromosomes, Human, Pair 22 ,Population ,Neuropsychological Tests ,Audiology ,Article ,Developmental psychology ,22q11 Deletion Syndrome ,Reaction Time ,Developmental and Educational Psychology ,medicine ,Humans ,Attention ,Child ,education ,Analysis of Variance ,education.field_of_study ,Learning Disabilities ,Siblings ,Cognitive disorder ,Neuropsychology ,medicine.disease ,Cross-Sectional Studies ,Neuropsychology and Physiological Psychology ,Chromosomal deletion syndrome ,El Niño ,Child, Preschool ,Female ,Analysis of variance ,Chromosome Deletion ,Cues ,Psychology ,Neurocognitive - Abstract
The 22q11 chromosomal deletion syndrome (22q11 DS) is associated with learning disabilities and a complex neuropsychological profile. Previous findings have suggested that executive attention deficits might underlie other neurocognitive anomalies. We administered the child Attention Network Test (ANT) to 52 children ages 5.0 to 11.5, 32 22q11 DS children (19 girls) and 20 controls (13 girls) and assessed the efficiency of segregated executive, orienting, and alerting networks. We hypothesized that 22q11 DS children have impaired executive network efficiency as compared to control siblings. The internal validity of the child ANT was confirmed for this population. Analysis of variance results showed significant main effects for flanker and cue types and no interaction effect in either 22q11 DS children or control siblings. Compared to control siblings, 22q11 DS children had significantly larger (less efficient) executive network scores, significantly increased errors on only incongruent trials, and a significant correlation between executive network scores and accuracy. The implications of these findings for future neurocognitive studies of 22q11 DS children are considered.
- Published
- 2004
50. The contribution of three strong candidate schizophrenia susceptibility genes in demographically distinct populations
- Author
-
Diana Hall, Maria Karayiorgou, and Joseph A. Gogos
- Subjects
Candidate gene ,Neuregulin-1 ,Locus (genetics) ,South Africa ,Behavioral Neuroscience ,mental disorders ,Genetic variation ,Genetics ,Humans ,Genetic Predisposition to Disease ,Neuregulin 1 ,Gene ,Genetic association ,Chromosome 13 ,biology ,Dysbindin ,Haplotype ,Intracellular Signaling Peptides and Proteins ,Genetic Variation ,United States ,Pedigree ,Neurology ,Dystrophin-Associated Proteins ,Schizophrenia ,biology.protein ,Carrier Proteins - Abstract
Here we characterize and compare the contribution of three recently identified strong candidate schizophrenia susceptibility genes; G72, neuregulin 1 (NRG1) and dystrobrevin-binding protein 1 (DTNBP1) in two independent datasets of patients with distinct genetic backgrounds. On the basis of corrected P-values from single- and multilocus transmission distortion tests our analysis provides no support for a contribution of G72, NRG1 or DTNBP1 in the tested samples. When transmission of individual haplotypes was considered, a picture more consistent with the original studies emerged, where transmission distortions in the same direction as the original samples and involving the same core haplotypes were observed for G72 and NRG1. Interestingly, whereas the NRG1 gene analysis was dominated by the presence of over-transmitted haplotypes, the G72 gene analysis was consistently dominated in both datasets by under-transmissions. Negative transmissions involved a core haplotype complementary to the originally detected over-transmitted haplotype, suggesting the presence of a protective variant within the G72 locus.
- Published
- 2004
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.