Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 511(7508): 236-40, 2014 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-24870235

RESUMEN

Although considerable evidence suggests that the chemical synapse is a lynchpin underlying affective disorders, how molecular insults differentially affect specific synaptic connections remains poorly understood. For instance, Neurexin 1a and 2 (NRXN1 and NRXN2) and CNTNAP2 (also known as CASPR2), all members of the neurexin superfamily of transmembrane molecules, have been implicated in neuropsychiatric disorders. However, their loss leads to deficits that have been best characterized with regard to their effect on excitatory cells. Notably, other disease-associated genes such as BDNF and ERBB4 implicate specific interneuron synapses in psychiatric disorders. Consistent with this, cortical interneuron dysfunction has been linked to epilepsy, schizophrenia and autism. Using a microarray screen that focused upon synapse-associated molecules, we identified Cntnap4 (contactin associated protein-like 4, also known as Caspr4) as highly enriched in developing murine interneurons. In this study we show that Cntnap4 is localized presynaptically and its loss leads to a reduction in the output of cortical parvalbumin (PV)-positive GABAergic (γ-aminobutyric acid producing) basket cells. Paradoxically, the loss of Cntnap4 augments midbrain dopaminergic release in the nucleus accumbens. In Cntnap4 mutant mice, synaptic defects in these disease-relevant neuronal populations are mirrored by sensory-motor gating and grooming endophenotypes; these symptoms could be pharmacologically reversed, providing promise for therapeutic intervention in psychiatric disorders.


Asunto(s)
Dopamina/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal , Transmisión Sináptica/genética , Ácido gamma-Aminobutírico/metabolismo , Animales , Antipsicóticos/farmacología , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Sinapsis Eléctricas/genética , Sinapsis Eléctricas/ultraestructura , Femenino , Genotipo , Humanos , Masculino , Ratones , Polimorfismo de Nucleótido Simple
2.
Mol Psychiatry ; 22(3): 384-395, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27001617

RESUMEN

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.


Asunto(s)
Síndrome de DiGeorge/genética , MicroARNs/genética , MicroARNs/metabolismo , Proteómica/métodos , Animales , Encéfalo/metabolismo , Cromatografía Liquida , Deleción Cromosómica , Síndrome de DiGeorge/metabolismo , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Humanos , Masculino , Espectrometría de Masas , Metabolómica/métodos , Ratones , Ratones Transgénicos , Corteza Prefrontal/metabolismo , Esquizofrenia/genética
3.
Mol Psychiatry ; 19(1): 99-107, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23999526

RESUMEN

Recurrent deletions at the 22q11.2 locus have been established as a strong genetic risk factor for the development of schizophrenia and cognitive dysfunction. Individuals with 22q11.2 deletions have a range of well-defined volumetric abnormalities in a number of critical brain structures. A mouse model of the 22q11.2 deletion (Df(16)A(+/-)) has previously been utilized to characterize disease-associated abnormalities on synaptic, cellular, neurocircuitry, and behavioral levels. We performed a high-resolution MRI analysis of mutant mice compared with wild-type littermates. Our analysis revealed a striking similarity in the specific volumetric changes of Df(16)A(+/-) mice compared with human 22q11.2 deletion carriers, including in cortico-cerebellar, cortico-striatal and cortico-limbic circuits. In addition, higher resolution magnetic resonance imaging compared with neuroimaging in human subjects allowed the detection of previously unknown subtle local differences. The cerebellar findings in Df(16)A(+/-) mice are particularly instructive as they are localized to specific areas within both the deep cerebellar nuclei and the cerebellar cortex. Our study indicates that the Df(16)A(+/-)mouse model recapitulates most of the hallmark neuroanatomical changes observed in 22q11.2 deletion carriers. Our findings will help guide the design and interpretation of additional complementary studies and thereby advance our understanding of the abnormal brain development underlying the emergence of 22q11.2 deletion-associated psychiatric and cognitive symptoms.


Asunto(s)
Encéfalo/patología , Deleción Cromosómica , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/patología , Modelos Animales de Enfermedad , Animales , Humanos , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Tercer Ventrículo/patología
4.
Mol Cell Neurosci ; 54: 84-92, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23396153

RESUMEN

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.


Asunto(s)
Corteza Cerebral/citología , Espinas Dendríticas/metabolismo , Hipocampo/citología , Mutación , Proteínas del Tejido Nervioso/genética , Neuritas/metabolismo , Animales , Axones/metabolismo , Células Cultivadas , Corteza Cerebral/metabolismo , Vesículas Citoplasmáticas/metabolismo , Aparato de Golgi/metabolismo , Hipocampo/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis , Especificidad de Órganos , Transporte de Proteínas
5.
Nat Genet ; 21(4): 434-9, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10192398

RESUMEN

Hemizygous cryptic deletions of the q11 band of human chromosome 22 have been associated with a number of psychiatric and behavioural phenotypes, including schizophrenia. Here we report the isolation and characterization of PRODH, a human homologue of Drosophila melanogaster sluggish-A (slgA), which encodes proline dehydrogenase responsible for the behavioural phenotype of the slgA mutant. PRODH is localized at chromosome 22q11 in a region deleted in some psychiatric patients. We also isolated the mouse homologue of slgA (Prodh), identified a mutation in this gene in the Pro/Re hyperprolinaemic mouse strain and found that these mice have a deficit in sensorimotor gating accompanied by regional neurochemical alterations in the brain. Sensorimotor gating is a neural filtering process that allows attention to be focused on a given stimulus, and is affected in patients with neuropsychiatric disorders. Furthermore, several lines of evidence suggest that proline may serve as a modulator of synaptic transmission in the mammalian brain. Our observations, in conjunction with the chromosomal location of PRODH, suggest a potential involvement of this gene in the 22q11-associated psychiatric and behavioural phenotypes.


Asunto(s)
Prolina Oxidasa/genética , Prolina Oxidasa/metabolismo , Reflejo de Sobresalto/fisiología , Estimulación Acústica , Errores Innatos del Metabolismo de los Aminoácidos/genética , Secuencia de Aminoácidos , Animales , Conducta Animal/fisiología , Northern Blotting , Encéfalo/anatomía & histología , Encéfalo/metabolismo , Cromosomas Humanos Par 22 , Femenino , Humanos , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Masculino , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Mutación , Neurotransmisores/análisis , Neurotransmisores/metabolismo , Prolina/análisis , Prolina/sangre , Prolina/metabolismo , Homología de Secuencia de Aminoácido
8.
J Cell Biol ; 134(4): 837-47, 1996 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8769410

RESUMEN

To identify genes regulated during skeletal muscle differentiation, we have infected mouse C2C12 myoblasts with retroviral gene trap vectors, containing a promoterless marker gene with a 5' splice acceptor signal. Integration of the vector adjacent to an actively transcribed gene places the marker under the transcriptional control of the endogenous gene, while the adjacent vector sequences facilitate cloning. The vector insertionally mutates the trapped locus and may also form fusion proteins with the endogenous gene product. We have screened several hundred clones, each containing a trapping vector integrated into a different endogenous gene. In agreement with previous estimates based on hybridization kinetics, we find that a large proportion of all genes expressed in myoblasts are regulated during differentiation. Many of these genes undergo unique temporal patterns of activation or repression during cell growth and myotube formation, and some show specific patterns of subcellular localization. The first gene we have identified with this strategy is the lysosomal cysteine protease cathepsin B. Expression from the trapped allele is upregulated during early myoblast fusion and downregulated in myotubes. A direct role for cathepsin B in myoblast growth and fusion is suggested by the observation that the trapped cells deficient in cathepsin B activity have an unusual morphology and reduced survival in low-serum media and undergo differentiation with impaired cellular fusion. The phenotype is reproduced by antisense cathepsin B expression in parental C2C12 myoblasts. The cellular phenotype is similar to that observed in cultured myoblasts from patients with I cell disease, in which there is diminished accumulation of lysosomal enzymes. This suggests that a specific deficiency of cathepsin B could contribute to the myopathic component of this illness.


Asunto(s)
Catepsina B/genética , Regulación del Desarrollo de la Expresión Génica/genética , Desarrollo de Músculos , Músculo Esquelético/citología , Músculo Esquelético/crecimiento & desarrollo , Animales , Secuencia de Bases , Catepsina B/biosíntesis , Diferenciación Celular , Fusión Celular , Línea Celular , Clonación Molecular/métodos , Regulación hacia Abajo , Marcación de Gen , Genes/fisiología , Prueba de Complementación Genética , Vectores Genéticos/genética , Lisosomas/enzimología , Ratones , Datos de Secuencia Molecular , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/fisiología , Mutagénesis Insercional , Fenotipo , Transcripción Genética , Regulación hacia Arriba
9.
Mol Psychiatry ; 13(7): 685-96, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17684499

RESUMEN

G72 is a strong candidate susceptibility gene for schizophrenia and bipolar disorder, whose function remains enigmatic. Here we show that one splicing isoform of the gene (LG72) encodes for a mitochondrial protein. We also provide convergent lines of evidence that increase of endogenous or exogenous G72 levels promotes robust mitochondrial fragmentation in mammalian cell lines and primary neurons, which proceeds in a manner that does not depend on induction of apoptosis or alteration in mitochondrial transmembrane potential. Finally, we show that increase in G72 levels in immature primary neurons is accompanied by a marked increase in dendritic arborization. By contrast, we failed to confirm the originally proposed functional interaction between G72 and D-amino acid oxidase (DAO) in two tested cell lines. Our results suggest an alternative role for G72 in modulating mitochondrial function.


Asunto(s)
Trastorno Bipolar/genética , Proteínas Portadoras/genética , Predisposición Genética a la Enfermedad , Mitocondrias/fisiología , Esquizofrenia/genética , Empalme Alternativo , Animales , Apoptosis , Línea Celular , ADN Complementario/genética , Dendritas/fisiología , Dendritas/ultraestructura , Amplificación de Genes , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular , Potenciales de la Membrana/fisiología , Mitocondrias/patología , Membranas Mitocondriales/fisiología , Neuronas/patología , Primates/genética , Mapeo Restrictivo
10.
Science ; 257(5078): 1951-5, 1992 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-1290524

RESUMEN

Two major developmentally regulated isoforms of the Drosophila chorion transcription factor CF2 differ by an extra zinc finger within the DNA binding domain. The preferred DNA binding sites were determined and are distinguished by an internal duplication of TAT in the site recognized by the isoform with the extra finger. The results are consistent with modular interactions between zinc fingers and trinucleotides and also suggest rules for recognition of AT-rich DNA sites by zinc finger proteins. The results show how modular finger interactions with trinucleotides can be used, in conjunction with alternative splicing, to alter the binding specificity and increase the spectrum of sites recognized by a DNA binding domain. Thus, CF2 may potentially regulate distinct sets of target genes during development.


Asunto(s)
Empalme Alternativo , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción/metabolismo , Dedos de Zinc , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión , Drosophila melanogaster/genética , Enlace de Hidrógeno , Datos de Secuencia Molecular , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/metabolismo , Unión Proteica , Relación Estructura-Actividad
11.
Science ; 257(5078): 1946-50, 1992 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-1411512

RESUMEN

Transcripts encoding the Drosophila putative transcription factor CF2 are subject to developmentally regulated alternative splicing, and they encode protein isoforms that differ in the number of zinc fingers. One testis-specific RNA encodes an isoform that includes three zinc fingers and a frame-shifted segment. Two other transcripts encode isoforms with six and seven zinc fingers which bind to distinct promoters and DNA target sequences. Thus, because of alternative splicing, a single gene appears to encode distinct DNA-binding proteins, each capable of regulating different gene sets in different tissues and developmental periods.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Drosophila , Regulación de la Expresión Génica , Factores de Transcripción/genética , Dedos de Zinc , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Corion , Drosophila melanogaster/genética , Genes , Datos de Secuencia Molecular , Oligodesoxirribonucleótidos/química , Reacción en Cadena de la Polimerasa , Empalme del ARN , ARN Mensajero/genética , Alineación de Secuencia
12.
Neuroscience ; 155(4): 1021-9, 2008 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-18674597

RESUMEN

Catechol-O-methyltransferase is an important enzyme in the metabolism of dopamine and an important regulator of aspects of dopamine-dependent working memory in prefrontal cortex that are disturbed in schizophrenia. This study investigated the phenotype of mice with heterozygous deletion vs. homozygous knockout of the catechol-O-methyltransferase gene across paradigms that access processes relevant for psychotic illness. Homozygotes evidenced improved performance in spontaneous alternation, an index of immediate spatial working memory; this effect appeared more substantive in males and was reflected in performance in aspects of the Barnes maze, an index of spatial learning/memory. Heterozygotes evidenced impaired performance in object recognition, an index of recognition memory; this effect was evident for both sexes at a retention interval of 5 min but appeared more enduring in males. There were no material effects for either genotype in relation to sociability or social novelty preference. While homozygous catechol-O-methyltransferase deletion results in improvement in spatial learning/working memory with little effect on social behavior, heterozygous deletion results in impairment of recognition memory. We have reported recently, using similar methods, that mice with deletion of the schizophrenia risk gene neuregulin-1 evidence disruption to social behavior, with little effect on spatial learning/working memory. The data suggest that catechol-O-methyltransferase and neuregulin-1 may influence, respectively, primarily cognitive and social endophenotypes of the overall schizophrenia syndrome.


Asunto(s)
Catecol O-Metiltransferasa/deficiencia , Cognición/fisiología , Heterocigoto , Homocigoto , Fenotipo , Conducta Social , Análisis de Varianza , Animales , Conducta Animal/fisiología , Conducta Exploratoria/fisiología , Femenino , Masculino , Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/genética , Pruebas Neuropsicológicas , Factores Sexuales
13.
Transl Psychiatry ; 5: e577, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-26057047

RESUMEN

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.


Asunto(s)
Amígdala del Cerebelo/fisiopatología , Ansiedad/genética , Condicionamiento Psicológico , Espinas Dendríticas/patología , Miedo , Factores de Intercambio de Guanina Nucleótido/genética , Hipocampo/fisiopatología , Células Piramidales/patología , Esquizofrenia/genética , Animales , Región CA3 Hipocampal/patología , Variaciones en el Número de Copia de ADN , Hipocampo/patología , Potenciación a Largo Plazo/genética , Memoria a Corto Plazo , Ratones , Ratones Noqueados , Fosforilación , Corteza Prefrontal/fisiopatología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Memoria Espacial
14.
Genes Brain Behav ; 3(4): 240-8, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15248869

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/genética , Predisposición Genética a la Enfermedad , Neurregulina-1/genética , Esquizofrenia/genética , Disbindina , Proteínas Asociadas a la Distrofina , Variación Genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Linaje , Esquizofrenia/epidemiología , Sudáfrica/epidemiología , Estados Unidos/epidemiología
15.
Biol Psychiatry ; 45(9): 1178-89, 1999 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-10331110

RESUMEN

BACKGROUND: Obsessive-compulsive disorder (OCD) is a common and severe psychiatric illness that affects 1-3% of the population and presents a well-established co-morbidity with major depressive disorder (MDD). Twin and family studies have suggested a genetic component in the etiology of OCD, although the mode of inheritance is unknown. Pharmacotherapy of the disease implicates both serotonergic and dopaminergic pathways. Previously, guided by the 22q11 microdeletion-related psychiatric phenotype, we provided evidence for a sexually dimorphic association between OCD and the gene for catechol-O-methyltransferase (COMT). In this report, we use 110 nuclear OCD families to analyze the inheritance of variants of COMT and monoamine oxidase-A (MAOA), another gene modulating monoamine metabolism. METHODS: A sample of 110 nuclear OCD families was collected, and lifetime diagnoses were ascertained using the Diagnostic Interview for Genetic Studies (DIGS). DNA was genotyped for functional variants of the COMT and MAO genes, and allele inheritance was examined using the Transmission Disequilibrium Test (TDT) and Haplotype-based Haplotype Relative Risk (HHRR) test. RESULTS: We provide evidence supporting the previously reported sexually dimorphic association between low COMT enzymatic activity and OCD. We also provide evidence for a similar sexually dimorphic association between OCD and an allele of the MAOA gene, previously linked to high MAO-A enzymatic activity. In agreement with the well-established action of MAO-A inhibitors as antidepressants, this association is particularly marked among male OCD probands with co-morbid MDD, who represent more than 50% of our male OCD sample. CONCLUSIONS: Our analysis indicates that variants of two genes modulating monoamine metabolism contribute significantly to OCD susceptibility. Most importantly, an unexpected sexually dimorphic pattern of genetic susceptibility to OCD is revealed and suggests the possibility that profound gender differences in genetic predisposition may exist not only for other OCD susceptibility genes, but for an array of other psychiatric disorders as well.


Asunto(s)
Catecol O-Metiltransferasa/genética , Monoaminooxidasa/genética , Trastorno Obsesivo Compulsivo/genética , Cromosoma X/genética , Adolescente , Adulto , Femenino , Genotipo , Humanos , Masculino , Trastorno Obsesivo Compulsivo/psicología , Escalas de Valoración Psiquiátrica , Factores Sexuales
16.
Biol Psychiatry ; 43(6): 425-31, 1998 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-9532347

RESUMEN

BACKGROUND: Deletions of 1.5-2 MB of chromosome 22q11 have been previously associated with schizophrenia. The deleted region includes proximally the region harboring genes involved in DiGeorge and velocardiofacial syndromes. Distally, it includes the gene for catechol-O-methyl-transferase (COMT), an enzyme that catalyzes the O-methylation of catecholamine neurotransmitters, including dopamine, and which therefore is considered a candidate gene for schizophrenia. METHODS: We address the issue of a direct involvement of the COMT gene in the development of schizophrenia by employing the first extensive mutational analysis of this gene in a sample of 157 schizophrenia patients and 129 healthy controls, using single-strand conformation polymorphism and chemical cleavage methodologies. RESULTS: No mutations were found, but several sequence variants were identified, including the genetic polymorphism that underlies the high/low activity of the enzyme (a Val158-->Met change, which results in the creation of an NlaIII restriction site in the low-activity allele). The distribution of the NlaIII genotypes among subsets of schizophrenia patients was analyzed. CONCLUSIONS: The results presented here argue against a major role of COMT in schizophrenia in general (although a minor effect could not be excluded) and represent a first step toward a more refined delineation of the phenotype/genotype relationship between 22q11 microdeletions and schizophrenia susceptibility.


Asunto(s)
Catecol O-Metiltransferasa/genética , Esquizofrenia/genética , Adulto , Secuencia de Bases , Exones/genética , Femenino , Variación Genética , Humanos , Masculino , Datos de Secuencia Molecular , Mutación , Oligonucleótidos/análisis , Reacción en Cadena de la Polimerasa , Polimorfismo Conformacional Retorcido-Simple , Esquizofrenia/enzimología
17.
Am J Med Genet ; 105(1): 50-2, 2001 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-11424997

RESUMEN

The greatest challenge of schizophrenia research remains the identification of the multiple, common, interacting, and moderately penetrant mutations that interfere with the highly complex function of human brain and result to this devastating disease. The inaccessibility of the human central nervous system to experimental manipulations and the paramount difficulties in identifying genes for schizophrenia has led researchers to generate mouse models for candidate genes using gene-targeting approaches. Although such mouse models have proven very useful in deciphering the causes of several diseases of the central nervous system (such as neurodegenerative diseases), their use in dissecting the biology of schizophrenia is still in its infancy. We argue that progress in this direction depends highly on progress in human genetic studies and requires careful and critical interpretation of the accumulating data.


Asunto(s)
Modelos Animales , Esquizofrenia/genética , Animales , Humanos , Ratones , Mutación
19.
Neuroscience ; 211: 136-64, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21821099

RESUMEN

Schizophrenia is one of the most common psychiatric disorders, but despite progress in identifying the genetic factors implicated in its development, the mechanisms underlying its etiology and pathogenesis remain poorly understood. Development of mouse models is critical for expanding our understanding of the causes of schizophrenia. However, translation of disease pathology into mouse models has proven to be challenging, primarily due to the complex genetic architecture of schizophrenia and the difficulties in the re-creation of susceptibility alleles in the mouse genome. In this review we highlight current research on models of major susceptibility loci and the information accrued from their analysis. We describe and compare the different approaches that are necessitated by diverse susceptibility alleles, and discuss their advantages and drawbacks. Finally, we discuss emerging mouse models, such as second-generation pathophysiology models based on innovative approaches that are facilitated by the information gathered from the current genetic mouse models.


Asunto(s)
Conducta Animal/fisiología , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Ratones , Esquizofrenia/genética , Esquizofrenia/fisiopatología , Animales , Estudios de Asociación Genética/métodos , Humanos
20.
Brain Res ; 1348: 114-9, 2010 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-20561508

RESUMEN

Abnormalities in pain perception, especially altered warmth and heat pain sensitivity, have been reported in schizophrenia. Therefore, genes associated with schizophrenia, including neuregulin-1 (NRG1), catechol-O-methyltranferase (COMT) and disrupted-in-schizophrenia-1 (DISC1), may play a role in modulating the physiological and psychological effects of pain stimuli in such patients. Thermal pain sensitivity was assessed in NRG1, COMT and DISC1 mutant mice, and the anti-nociceptive effects of acute Delta(9)-tetrahydrocannabinol (THC) were compared in NRG1 and COMT mutants. At baseline, deletion of NRG1 and DISC1 each reduced thermal pain sensitivity, while deletion of COMT increased pain sensitivity. Neither NRG1 nor COMT deletion altered the anti-nociceptive effects of acute systemic THC (8.0mg/kg). These results indicate a differential contribution of NRG1 and DISC1 vis-à-vis COMT to the processing of thermal nociceptive stimuli and extend their phenotypic relationship to psychotic illness.


Asunto(s)
Catecol O-Metiltransferasa/genética , Hiperalgesia/genética , Proteínas del Tejido Nervioso/genética , Neurregulina-1/genética , Umbral del Dolor/efectos de los fármacos , Analgésicos no Narcóticos/uso terapéutico , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Catecol O-Metiltransferasa/deficiencia , Dronabinol/uso terapéutico , Femenino , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/fisiopatología , Masculino , Ratones , Ratones Noqueados , Mutagénesis/genética , Proteínas del Tejido Nervioso/deficiencia , Neurregulina-1/deficiencia , Dimensión del Dolor , Tiempo de Reacción/efectos de los fármacos , Factores Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA