Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
2.
Nat Commun ; 12(1): 4913, 2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-34389722

RESUMEN

Epitranscriptomic mechanisms linking tRNA function and the brain proteome to cognition and complex behaviors are not well described. Here, we report bi-directional changes in depression-related behaviors after genetic disruption of neuronal tRNA cytosine methylation, including conditional ablation and transgene-derived overexpression of Nsun2 in the mouse prefrontal cortex (PFC). Neuronal Nsun2-deficiency was associated with a decrease in tRNA m5C levels, resulting in deficits in expression of 70% of tRNAGly isodecoders. Altogether, 1488/5820 proteins changed upon neuronal Nsun2-deficiency, in conjunction with glycine codon-specific defects in translational efficiencies. Loss of Gly-rich proteins critical for glutamatergic neurotransmission was associated with impaired synaptic signaling at PFC pyramidal neurons and defective contextual fear memory. Changes in the neuronal translatome were also associated with a 146% increase in glycine biosynthesis. These findings highlight the methylation sensitivity of glycinergic tRNAs in the adult PFC. Furthermore, they link synaptic plasticity and complex behaviors to epitranscriptomic modifications of cognate tRNAs and the proteomic homeostasis associated with specific amino acids.


Asunto(s)
Trastorno Depresivo/fisiopatología , Epigénesis Genética/genética , Metiltransferasas/genética , Proteoma/metabolismo , ARN de Transferencia/genética , Transmisión Sináptica/genética , Animales , Trastorno Depresivo/genética , Trastorno Depresivo/metabolismo , Perfilación de la Expresión Génica/métodos , Metiltransferasas/deficiencia , Metiltransferasas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neuronas/metabolismo , Corteza Prefrontal/citología , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiología , Proteómica/métodos , ARN de Transferencia/metabolismo , Transducción de Señal/genética
3.
NPJ Schizophr ; 4(1): 16, 2018 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-30131491

RESUMEN

Epigenetic changes may account for the doubled risk to develop schizophrenia in individuals exposed to famine in utero. We therefore investigated DNA methylation in a unique sample of patients and healthy individuals conceived during the great famine in China. Subsequently, we examined two case-control samples without famine exposure in whole blood and brain tissue. To shed light on the causality of the relation between famine exposure and DNA methylation, we exposed human fibroblasts to nutritional deprivation. In the famine-exposed schizophrenia patients, we found significant hypermethylation of the dual specificity phosphatase 22 (DUSP22) gene promoter (Chr6:291687-293285) (N = 153, p = 0.01). In this sample, DUSP22 methylation was also significantly higher in patients independent of famine exposure (p = 0.025), suggesting that hypermethylation of DUSP22 is also more generally involved in schizophrenia risk. Similarly, DUSP22 methylation was also higher in two separate case-control samples not exposed to famine using DNA from whole blood (N = 64, p = 0.03) and postmortem brains (N = 214, p = 0.007). DUSP22 methylation showed strong genetic regulation across chromosomes by a region on chromosome 16 which was consistent with new 3D genome interaction data. The presence of a direct link between famine and DUSP22 transcription was supported by data from cultured human fibroblasts that showed increased methylation (p = 0.048) and expression (p = 0.019) in response to nutritional deprivation (N = 10). These results highlight an epigenetic locus that is genetically regulated across chromosomes and that is involved in the response to early-life exposure to famine and that is relevant for a major psychiatric disorder.

4.
Mol Psychiatry ; 23(1): 123-132, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28115742

RESUMEN

Large-scale consortia mapping the genomic risk architectures of schizophrenia provide vast amounts of molecular information, with largely unexplored therapeutic potential. We harnessed publically available information from the Psychiatric Genomics Consortium, and report myocyte enhancer factor 2C (MEF2C) motif enrichment in sequences surrounding the top scoring single-nucleotide polymorphisms within risk loci contributing by individual small effect to disease heritability. Chromatin profiling at base-pair resolution in neuronal nucleosomes extracted from prefrontal cortex of 34 subjects, including 17 cases diagnosed with schizophrenia, revealed MEF2C motif enrichment within cis-regulatory sequences, including neuron-specific promoters and superenhancers, affected by histone H3K4 hypermethylation in disease cases. Vector-induced short- and long-term Mef2c upregulation in mouse prefrontal projection neurons consistently resulted in enhanced cognitive performance in working memory and object recognition paradigms at baseline and after psychotogenic drug challenge, in conjunction with remodeling of local connectivity. Neuronal genome tagging in vivo by Mef2c-Dam adenine methyltransferase fusion protein confirmed the link between cognitive enhancement and MEF2C occupancy at promoters harboring canonical and variant MEF2C motifs. The multilayered integrative approaches presented here provide a roadmap to uncover the therapeutic potential of transcriptional regulators for schizophrenia and related disorders.


Asunto(s)
Trastornos del Conocimiento , Regulación de la Expresión Génica/genética , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Polimorfismo de Nucleótido Simple/genética , Esquizofrenia/complicaciones , Animales , Encéfalo/metabolismo , Encéfalo/patología , Inmunoprecipitación de Cromatina , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/terapia , Biología Computacional , Modelos Animales de Enfermedad , Epigenómica/métodos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Histonas/genética , Histonas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Esquizofrenia/genética , Esquizofrenia/patología , Transducción Genética
5.
Mol Cell Neurosci ; 82: 157-166, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28549865

RESUMEN

CRISPR/Cas9 technology has transformed our ability to manipulate the genome and epigenome, from efficient genomic editing to targeted localization of effectors to specific loci. Through the manipulation of DNA- and histone-modifying enzyme activities, activation or repression of gene expression, and targeting of transcriptional regulators, the role of gene-regulatory and epigenetic pathways in basic biology and disease processes can be directly queried. Here, we discuss emerging CRISPR-based methodologies, with specific consideration of neurobiological applications of human induced pluripotent stem cell (hiPSC)-based models.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Sistemas CRISPR-Cas/genética , Edición Génica , Expresión Génica/genética , Células Madre Pluripotentes Inducidas/citología , Encefalopatías/terapia , Edición Génica/métodos , Humanos
6.
Transl Psychiatry ; 5: e679, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26575220

RESUMEN

Regulators of the histone H3-trimethyl lysine-4 (H3K4me3) mark are significantly associated with the genetic risk architecture of common neurodevelopmental disease, including schizophrenia and autism. Typical H3K4me3 is primarily localized in the form of sharp peaks, extending in neuronal chromatin on average only across 500-1500 base pairs mostly in close proximity to annotated transcription start sites. Here, through integrative computational analysis of epigenomic and transcriptomic data based on next-generation sequencing, we investigated H3K4me3 landscapes of sorted neuronal and non-neuronal nuclei in human postmortem, non-human primate and mouse prefrontal cortex (PFC), and blood. To explore whether H3K4me3 peak signals could also extend across much broader domains, we examined broadest domain cell-type-specific H3K4me3 peaks in an unbiased manner with an innovative approach on 41+12 ChIP-seq and RNA-seq data sets. In PFC neurons, broadest H3K4me3 distribution ranged from 3.9 to 12 kb, with extremely broad peaks (~10 kb or broader) related to synaptic function and GABAergic signaling (DLX1, ELFN1, GAD1, IGSF9B and LINC00966). Broadest neuronal peaks showed distinct motif signatures and were centrally positioned in prefrontal gene-regulatory Bayesian networks and sensitive to defective neurodevelopment. Approximately 120 of the broadest H3K4me3 peaks in human PFC neurons, including many genes related to glutamatergic and dopaminergic signaling, were fully conserved in chimpanzee, macaque and mouse cortical neurons. Exploration of spread and breadth of lysine methylation markings could provide novel insights into epigenetic mechanism involved in neuropsychiatric disease and neuronal genome evolution.


Asunto(s)
Encéfalo/metabolismo , Epigénesis Genética/genética , Redes Reguladoras de Genes/genética , Histonas/genética , Histonas/metabolismo , Adulto , Animales , Femenino , Humanos , Macaca , Masculino , Ratones , Pan troglodytes
7.
Neuroscience ; 286: 79-86, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25446346

RESUMEN

Prenatal protein malnutrition (PPM) in rats causes enduring changes in brain and behavior including increased cognitive rigidity and decreased inhibitory control. A preliminary gene microarray screen of PPM rat prefrontal cortex (PFC) identified alterations in KCNJ3 (GIRK1/Kir3.1), a gene important for regulating neuronal excitability. Follow-up with polymerase chain reaction and Western blot showed decreased KCNJ3 expression in the PFC, but not hippocampus or brainstem. To verify localization of the effect to the PFC, baseline regional brain activity was assessed with (14)C-2-deoxyglucose. Results showed decreased activation in the PFC but not hippocampus. Together these findings point to the unique vulnerability of the PFC to the nutritional insult during early brain development, with enduring effects in adulthood on KCNJ3 expression and baseline metabolic activity.


Asunto(s)
Desoxiglucosa/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Desnutrición/genética , Desnutrición/metabolismo , Corteza Prefrontal/metabolismo , Fenómenos Fisiologicos de la Nutrición Prenatal , Animales , Regulación hacia Abajo , Femenino , Expresión Génica , Masculino , Embarazo , Ratas , Ratas Long-Evans
8.
Neuroscience ; 264: 131-41, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23831425

RESUMEN

A large, and still rapidly expanding literature on epigenetic regulation in the nervous system has provided fundamental insights into the dynamic regulation of DNA methylation and post-translational histone modifications in the context of neuronal plasticity in health and disease. Remarkably, however, very little is known about the potential role of chromatin-bound RNAs, including many long non-coding transcripts and various types of small RNAs. Here, we provide an overview on RNA-mediated regulation of chromatin structure and function, with focus on histone lysine methylation and psychiatric disease. Examples of recently discovered chromatin-bound long non-coding RNAs important for neuronal health and function include the brain-derived neurotrophic factor antisense transcript (Bdnf-AS) which regulates expression of the corresponding sense transcript, and LOC389023 which is associated with human-specific histone methylation signatures at the chromosome 2q14.1 neurodevelopmental risk locus by regulating expression of DPP10, an auxillary subunit for voltage-gated K(+) channels. We predict that the exploration of chromatin-bound RNA will significantly advance our current knowledge base in neuroepigenetics and biological psychiatry.


Asunto(s)
Ensamble y Desensamble de Cromatina , Epigénesis Genética , Histonas/metabolismo , Trastornos Mentales/genética , ARN no Traducido/metabolismo , Animales , Cromatina/metabolismo , Humanos , Trastornos Mentales/metabolismo
9.
Oper Dent ; 39(1): E22-30, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23865583

RESUMEN

OBJECTIVES: Microleakage of composite restorations at the cervical margin placed apically to the cementoenamel junction (CEJ) is still a concern. This study evaluated the effect of simultaneous bonding application on cervical sealing of nano-ionomer/silorane- or methacrylate-based composite open sandwich Class II restorations in the modified technique compared with that of conventional bonding. METHODS AND MATERIALS: In 60 sound maxillary premolars, two standardized Class II cavities were prepared with cervical margins 1 mm below the CEJ. The teeth were randomly divided into six groups of 10 teeth each. In the first three groups (groups 1-3), Clearfil SE Bond and Clearfil APX (Kuraray) were used for restoration in the total bonding technique (group 1), conventional open sandwich technique associated with a nano-ionomer (Ketac N100, 3M ESPE) (group 2), and modified open sandwich technique with simultaneous bonding application for both nano-ionomer and composite (group 3). In the second three groups (groups 4-6), Silorane Adhesive and Filtek Silorane composite (3M ESPE) were used in the same manner as in the first three groups, respectively. RESULTS: The simultaneous bonding application in the modified sandwich restorations (with SE Bond or Silorane Adhesive) resulted in a significant reduction of the cervical microleakage compared with that of the conventional bonding (p<0.05). However, microleakage of the modified technique was similar to that of the total bonding (with SE Bond or Silorane Adhesive) (p>0.05), both showing good marginal seal.


Asunto(s)
Resinas Compuestas/uso terapéutico , Filtración Dental/etiología , Restauración Dental Permanente/métodos , Cementos de Ionómero Vítreo/uso terapéutico , Resinas Compuestas/efectos adversos , Recubrimiento Dental Adhesivo/efectos adversos , Recubrimiento Dental Adhesivo/métodos , Filtración Dental/prevención & control , Cementos de Ionómero Vítreo/efectos adversos , Humanos , Metacrilatos/efectos adversos , Metacrilatos/uso terapéutico , Nanoestructuras/uso terapéutico , Cementos de Resina/efectos adversos , Cementos de Resina/uso terapéutico , Resinas de Silorano/efectos adversos , Resinas de Silorano/uso terapéutico , Cuello del Diente/efectos de los fármacos
10.
Neurobiol Dis ; 8(5): 784-91, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11592848

RESUMEN

Dysfunction of the prefrontal cortex may contribute to the autistic features and mental retardation of Rett syndrome, a neuropsychiatric condition caused by mutations of the gene encoding methyl-CpG-binding protein 2 (MeCP2). Because nothing is known about the expression of MeCP2 and other chromatin-associated factors in primate brain, we studied in monkey prefrontal cortex and murine cerebral cortex expression patterns of MeCP2 and of macrohistone H2A (MacroH2A), which like MeCP2 is associated with transcriptionally silent chromatin. In both species, MeCP2 and MacroH2A appeared to be ubiquitously expressed by cortical neurons, including projection neurons and GABAergic interneurons. In the adult monkey, MeCP2 expression was robust throughout all layers of the prefrontal cortex but it was limited in fetal monkeys at embryonic day 110 to the deeper cortical layers and the subplate. These results suggest that MeCP2 may be important for neuronal maintenance in the developing and in the mature primate prefrontal cortex, consistent with the previously reported phenotype of MeCP2-null mutant mice.


Asunto(s)
Proteínas Cromosómicas no Histona , Proteínas de Unión al ADN/biosíntesis , Regulación de la Expresión Génica , Proteínas del Tejido Nervioso/biosíntesis , Corteza Prefrontal/metabolismo , Proteínas Represoras , Síndrome de Rett/genética , Factores de Edad , Animales , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/genética , Silenciador del Gen , Histonas/biosíntesis , Histonas/genética , Humanos , Macaca mulatta , Proteína 2 de Unión a Metil-CpG , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Especificidad de Órganos , Corteza Prefrontal/crecimiento & desarrollo
11.
Mol Psychiatry ; 6(5): 593-604, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11526474

RESUMEN

Somatic symptoms and aversion of opiate withdrawal, regulated by noradrenergic signaling, were attenuated in mice with a CNS-wide conditional ablation of neurotrophin-3. This occurred in conjunction with altered cAMP-mediated excitation and reduced upregulation of tyrosine hydroxylase in A6 (locus coeruleus) without loss of neurons. Transgene-derived NT-3 expressed by noradrenergic neurons of conditional mutants restored opiate withdrawal symptoms. Endogenous NT-3 expression, strikingly absent in noradrenergic neurons of postnatal and adult brain, is present in afferent sources of the dorsal medulla and is upregulated after chronic morphine exposure in noradrenergic projection areas of the ventral forebrain. NT-3 expressed by non-catecholaminergic neurons may modulate opiate withdrawal and noradrenergic signalling.


Asunto(s)
Encéfalo/fisiología , Dependencia de Morfina/genética , Proteínas del Tejido Nervioso , Neuronas/fisiología , Neurotrofina 3/fisiología , Síndrome de Abstinencia a Sustancias/genética , Tirosina 3-Monooxigenasa/genética , Envejecimiento , Animales , Reacción de Prevención/fisiología , Encéfalo/crecimiento & desarrollo , Colforsina/farmacología , AMP Cíclico/fisiología , Estimulación Eléctrica , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Regulación Enzimológica de la Expresión Génica , Técnicas In Vitro , Proteínas de Filamentos Intermediarios/genética , Locus Coeruleus/enzimología , Locus Coeruleus/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Morfina/farmacología , Dependencia de Morfina/fisiopatología , Nestina , Neuronas/efectos de los fármacos , Neurotrofina 3/deficiencia , Neurotrofina 3/genética , Transducción de Señal/fisiología , Síndrome de Abstinencia a Sustancias/fisiopatología , Tirosina 3-Monooxigenasa/metabolismo
12.
Nat Genet ; 27(3): 327-31, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11242118

RESUMEN

Mecp2 is an X-linked gene encoding a nuclear protein that binds specifically to methylated DNA (ref. 1) and functions as a general transcriptional repressor by associating with chromatin-remodeling complexes. Mecp2 is expressed at high levels in the postnatal brain, indicating that methylation-dependent regulation of gene expression may have a crucial role in the mammalian central nervous system. Consistent with this notion is the recent demonstration that MECP2 mutations cause Rett syndrome (RTT, MIM 312750), a childhood neurological disorder that represents one of the most common causes of mental retardation in females. Here we show that Mecp2-deficient mice exhibit phenotypes that resemble some of the symptoms of RTT patients. Mecp2-null mice were normal until 5 weeks of age, when they began to develop disease, leading to death between 6 and 12 weeks. Mutant brains showed substantial reduction in both weight and neuronal cell size, but no obvious structural defects or signs of neurodegeneration. Brain-specific deletion of Mecp2 at embryonic day (E) 12 resulted in a phenotype identical to that of the null mutation, indicating that the phenotype is caused by Mecp2 deficiency in the CNS rather than in peripheral tissues. Deletion of Mecp2 in postnatal CNS neurons led to a similar neuronal phenotype, although at a later age. Our results indicate that the role of Mecp2 is not restricted to the immature brain, but becomes critical in mature neurons. Mecp2 deficiency in these neurons is sufficient to cause neuronal dysfunction with symptomatic manifestation similar to Rett syndrome.


Asunto(s)
Proteínas Cromosómicas no Histona , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas Represoras , Síndrome de Rett/genética , Animales , Diferenciación Celular , Tamaño de la Célula , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Islas de CpG , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Proteína 2 de Unión a Metil-CpG , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/patología , Fenotipo , Síndrome de Rett/metabolismo , Síndrome de Rett/patología
13.
Arch Gen Psychiatry ; 53(5): 425-36, 1996 May.
Artículo en Inglés | MEDLINE | ID: mdl-8624186

RESUMEN

BACKGROUND: The cortical subplate is a transitory structure involved in the formation of connections in developing cerebral cortex. Interstitial neurons, normally present in subcortical white matter (WM) of the adult brain, have escaped the programmed cell death that eliminates most subplate neurons. Previous investigations indicated a maldistribution of one population of interstitial neurons in the WM of brains of schizophrenic patients, suggesting a defect of the subplate during brain development. METHODS: Three histochemically or immunocytochemically defined neuronal populations were studied in WM beneath the middle frontal gyrus of 20 schizophrenic patients and 20 matched control subjects. RESULTS: Brains of schizophrenic patients showed significant changes in the distribution of the three neuronal populations: microtubule-associated protein 2 and nonphosphorylated neurofilament-immunoreactive neurons showed a decreased density in superficial WM and an increased density in deeper WM. Nicotinamide adenine dinucleotide phosphate-diaphorase neurons were reduced in superficial WM and showed variable densities in deeper WM. Thirty-five percent of the brains of schizophrenic patients but no brains of the control subjects showed a maldistribution of neurons toward deeper WM with at least two of the three markers. Changes in neuronal distribution were not linked to age, gender, autolysis time, or subtype of schizophrenia. CONCLUSIONS: Selective displacement of interstitial WM neurons in the frontal lobe of brains of schizophrenic patients may indicate alteration in the migration of subplate neurons or in the pattern of programmed cell death. Both could lead to defective cortical circuitry in the brains of schizophrenic patients.


Asunto(s)
Corteza Prefrontal/citología , Esquizofrenia/diagnóstico , Adulto , Edad de Inicio , Anciano , Anticuerpos Monoclonales , Autólisis , Recuento de Células , Muerte Celular , Femenino , Humanos , Inmunohistoquímica , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Persona de Mediana Edad , NADPH Deshidrogenasa/metabolismo , Neuronas/citología , Neuronas/enzimología , Neuronas/patología , Corteza Prefrontal/enzimología , Corteza Prefrontal/patología , Esquizofrenia/enzimología , Esquizofrenia/patología , Factores de Tiempo
14.
J Neurosci ; 16(1): 19-30, 1996 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8613785

RESUMEN

NMDA receptor antagonists can induce a schizophrenia-like psychosis, but the role of NMDA receptors in the pathophysiology of schizophrenia remains unclear. Expression patterns of mRNAs for five NMDA receptor subunits (NR1/NR2A-D) were determined by in situ hybridization in prefrontal, parieto-temporal, and cerebellar cortex of brains from schizophrenics and from neuroleptic-treated and nonmedicated controls. In the cerebral cortex of both schizophrenics and controls, mRNAs for NR1, NR2A, NR2B, and NR2D subunits were preferentially expressed in layers II/III, Va, and VIa, with much higher levels in the prefrontal than in the parieto-temporal cortex. Levels of mRNA for the NR2C subunit were very low overall. By contrast, the cerebellar cortex of both schizophrenics and controls contained very high levels of NR2C subunit mRNA, whereas levels for the other subunit mRNAs were very low, except NR1, for which levels were moderate. Significant alterations in the schizophrenic cohort were confined to the prefrontal cortex. Here there was a shift in the relative proportions of mRNAs for the NR2 subunit family, with a 53% relative increase in expression of the NR2D subunit mRNA. No comparable changes were found in neuroleptic-treated or untreated controls. These findings indicate regional heterogeneity of NMDA receptor subunit expression in human cerebral and cerebellar cortex. In schizophrenics, the alterations in expression of NR2 subunit mRNA in prefrontal cortex are potential indicators of deficits in NMDA receptor-mediated neurotransmission accompanying functional hypoactivity of the frontal lobes.


Asunto(s)
Corteza Prefrontal/fisiopatología , Receptores de N-Metil-D-Aspartato/genética , Esquizofrenia/genética , Autorradiografía , Cerebelo/fisiopatología , Cerebelo/ultraestructura , Femenino , Expresión Génica/fisiología , Humanos , Hibridación in Situ , Masculino , Persona de Mediana Edad , Lóbulo Parietal/fisiopatología , Lóbulo Parietal/ultraestructura , Corteza Prefrontal/ultraestructura , ARN Mensajero/análisis , Receptores de N-Metil-D-Aspartato/ultraestructura , Esquizofrenia/fisiopatología , Lóbulo Temporal/fisiopatología , Lóbulo Temporal/ultraestructura
15.
Brain Res ; 699(2): 297-304, 1995 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-8616634

RESUMEN

Animal studies and cell culture experiments demonstrated that posttranscriptional editing of the transcript of the GluR-2 gene, resulting in substitution of an arginine for glutamine in the second transmembrane region (TM II) of the expressed protein, is associated with a reduction in Ca2+ permeability of the receptor channel. Thus, disturbances in GluR-2 RNA editing with alteration of intracellular Ca2+ homeostasis could lead to neuronal dysfunction and even neuronal degeneration. The present study determined the proportions of edited and unedited GluR-2 RNA in the prefrontal cortex of brains from patients with Alzheimer's disease, in the striatum of brains from patients with Huntington's disease, and in the same areas of brains from age-matched schizophrenics and controls, by using reverse transcriptase-polymerase chain reaction, restriction endonuclease digestion, gel electrophoresis and scintillation radiometry. In the prefrontal cortex of controls, < 0.1% of all GluR-2 RNA molecules were unedited and > 99.9% were edited; in the prefrontal cortex both of schizophrenics and of Alzheimer's patients approximately 1.0% of all GluR-2 RNA molecules were unedited and 99% were edited. In the striatum of controls and of schizophrenics, approximately 0.5% of GluR-2 RNA molecules were unedited and 99.5% were edited; in the striatum of Huntington's patients nearly 5.0% of GluR-2 RNA was unedited. In the prefrontal white matter of controls, approximately 7.0% of GluR-2 RNA was unedited. In the normal human prefrontal cortex and striatum, the large majority of GluR-2 RNA molecules contains a CGG codon for arginine in the TMII coding region; this implies that the corresponding AMPA receptors have a low Ca2+ permeability, as previously demonstrated for the rat brain. The process of GluR-2 RNA editing is compromised in a region-specific manner in schizophrenia, in Alzheimer's disease and Huntington's Chorea although in each of these disorders there is still a large excess of edited GluR-2 RNA molecules. Disturbances of GluR-2 RNA editing leading to excessive Ca2+ permeability, may contribute to neuronal dysfunction in schizophrenia and to neuronal death in Alzheimer's disease and Huntington's disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Cuerpo Estriado/metabolismo , Corteza Prefrontal/metabolismo , ARN/metabolismo , Receptores AMPA/metabolismo , Anciano , Autorradiografía , Humanos , Enfermedad de Huntington/metabolismo , Persona de Mediana Edad , Degeneración Nerviosa , Esquizofrenia/metabolismo
16.
Cereb Cortex ; 5(6): 550-60, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-8590827

RESUMEN

The prefrontal cortex of schizophrenics is hypoactive and displays changes related to inhibitory, GABAergic neurons, and GABAergic synapses. These changes include decreased levels of glutamic acid decarboxylase (GAD), the enzyme for GABA synthesis, upregulation of muscimol binding, and downregulation of benzodiazepine binding to GABAA receptors. Studies in the visual cortex of nonhuman primates have demonstrated that gene expression for GAD and for several GABAA receptor subunit polypeptides is under control of neuronal activity, raising the possibility that similar mechanisms in the hypoactive prefrontal cortex of schizophrenics may explain the abnormalities in GAD and in GABAA receptor regulation. In the present study, which is the first of its type on human cerebral cortex, levels of mRNAs for six GABAA receptor subunits (alpha 1, alpha 2, alpha 5, beta 1, beta 2, gamma 2) and their laminar expression patterns were analyzed in the prefrontal cortex of schizophrenics and matched controls, using in situ hybridization histochemistry and densitometry. Three types of laminar expression pattern were observed: mRNAs for the alpha 1, beta 2, and gamma 2 subunits, which are the predominant receptor subunits expressed in the mature cortex, were expressed at comparatively high levels by cells of all six cortical layers, but most intensely by cells in lower layer III and layer IV. mRNAs for the alpha 2, alpha 5, and beta 1 subunits were expressed at lower levels; alpha 2 and beta 1 were expressed predominantly by cells in layers II, III, and IV; alpha 5 was expressed predominantly in layers IV, V, and VI. There were no significant changes in overall mRNA levels for any of the receptor subunits in the prefrontal cortex of schizophrenics, and the laminar expression pattern of all six receptor subunit mRNAs did not differ between schizophrenics and controls. Because gene expression for GABAA receptor subunits is not consistently altered in the prefrontal cortex of schizophrenics, the previously reported upregulation of muscimol binding sites and downregulation of benzodiazepine binding sites in the prefrontal and adjacent cingulate cortex of schizophrenics are possibly due to posttranscriptional modifications of mRNAs and their translated polypeptides.


Asunto(s)
Expresión Génica , Corteza Prefrontal/metabolismo , Receptores de GABA-A/genética , Esquizofrenia/metabolismo , Autorradiografía , Femenino , Humanos , Hibridación in Situ , Masculino , Sondas ARN , ARN Mensajero/biosíntesis , Receptores de GABA-A/biosíntesis , Esquizofrenia/genética
17.
J Neurosci ; 15(10): 6509-20, 1995 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7472413

RESUMEN

A novel NMDA receptor-like (NMDAR-L) cDNA was isolated that contained an open reading frame coding for a predicted polypeptide of 1115 amino acids that shares approximately 27% identity with NMDA receptor subunits. In situ hybridization experiments indicated that NMDAR-L mRNA was expressed in the developing rodent CNS. On postnatal day 1 (P1), NMDAR-L mRNA expression was pronounced in the entorhinal cortex, the subiculum and the thalamus, in layer V of the developing neocortex, in the superior and inferior colliculi, and various regions of the hindbrain, excluding the cerebellum. On P5, NMDAR-L mRNA was expressed in layer V of the neocortex, in the entorhinal cortex, in the subiculum, and in the thalamus. On P14, NMDAR-L mRNA was expressed in layers II-VI of the neocortex, in the entorhinal and piriform cortex, in the subiculum and CA1 field, and in the nucleus of the lateral olfactory tract. In the adult brain, NMDAR-L mRNA was detected predominately in the nucleus of the lateral olfactory tract. Injection of NMDAR-L cRNA into Xenopus oocytes did not lead to the expression of homomeric glutamate-activated channels. However, coinjection of the triple combination of NMDAR-L with NMDAR1 and NMDAR2B cRNAs led to a striking decrease in the current magnitude compared to currents obtained after coexpression of the double combination of NMDAR1 with NMDAR2B. While the function of NMDAR-L remains to be established, its developmental and regional expression pattern suggests that NMDAR-L may influence axonal outgrowth and synaptogenesis during brain development.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Roedores/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Ratones , Sondas Moleculares/genética , Datos de Secuencia Molecular , Oocitos/metabolismo , Reacción en Cadena de la Polimerasa , ARN Complementario/metabolismo , ARN Mensajero/metabolismo , Ratas , Distribución Tisular , Xenopus
18.
Arch Gen Psychiatry ; 52(4): 258-66, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7702443

RESUMEN

BACKGROUND: Up-regulation of gamma-aminobutyric acidA (GABAA) receptors and decreased GABA uptake in the cerebral cortex of schizophrenics suggest altered GABAergic transmission, which could be caused by primary disturbance of GABA synapses or by decreased production of the transmitter. Decreased production could be due to a shutdown in GABA production or to loss of GABA neurons caused by cell death or their failure to migrate to the cortex during brain development. METHODS: To discriminate between these possibilities, we quantified levels of messenger RNA (mRNA) for the 67-kd isoform of glutamic acid decarboxylase (GAD), the key enzyme in GABA synthesis, and the number and laminar distribution of GAD mRNA--expressing neurons in the dorsolateral prefrontal cortex (DLPFC) of schizophrenics and matched controls, using in situ hybridization-histochemistry, densitometry, and cell-counting methods. These data were compared with the total number of neurons, the number of small, round or ovoid neurons 8 to 15 microns in diameter, and overall frontal lobe volume. As a control, mRNA levels for type II calcium-calmodulin-dependent protein kinase (CamIIK) were quantified. RESULTS: Schizophrenics showed a pronounced decrease in GAD mRNA levels in neurons of layer I (40%) and layer II (48%) and an overall 30% decrease in layers III to VI. There were also strong overall reductions in GAD mRNA levels. The CamIIK mRNA levels showed no significant differences between samples. No differences were found in the total number of neurons nor in small, round or ovoid neurons, which should include a majority of the GABA cells. Prefrontal gray and white matter volume did not differ significantly between controls and schizophrenics. CONCLUSIONS: The prefrontal cortex of schizophrenics shows reduced expression for GAD in the absence of significant cell loss. This may be brought about by an activity-dependent down-regulation associated with the functional hypoactivity of the DLPFC. The lack of significant alterations in cell numbers in the DLPFC and frontal lobe volume in schizophrenics also implies that overall cortical neuronal migration had not been compromised in development. Previous reports of altered neuronal distribution in the subcortical white matter of schizophrenic brains in comparison with that of controls may indicate disturbances of migration or programmed cell death in the cortical subplate, leading to altered connection formation in the overlying cortex of schizophrenics and activity-dependent down-regulation of neurotransmitter-related gene expression.


Asunto(s)
Glutamato Descarboxilasa/análisis , Corteza Prefrontal/enzimología , Esquizofrenia/enzimología , Ácido gamma-Aminobutírico/biosíntesis , Adulto , Edad de Inicio , Anciano , Autorradiografía , Recuento de Células , Muerte Celular , Densitometría , Regulación hacia Abajo , Femenino , Expresión Génica , Glutamato Descarboxilasa/genética , Humanos , Hibridación in Situ , Masculino , Persona de Mediana Edad , Neuronas/enzimología , Neuronas/metabolismo , Corteza Prefrontal/anatomía & histología , Corteza Prefrontal/citología , ARN Mensajero/análisis , Receptores de GABA/química , Receptores de GABA/metabolismo , Esquizofrenia/genética , Esquizofrenia/metabolismo , Ácido gamma-Aminobutírico/genética
19.
J Comp Neurol ; 339(3): 421-37, 1994 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-7510732

RESUMEN

The distribution of cortical efferent connections to brainstem vestibular nuclei was quantitatively analysed by means of retrograde tracer substances injected into different electrophysiologically identified parts of the brainstem vestibular nuclear complex of five Java monkeys (Macaca fascicularis). Three polysensory vestibular areas were found to have a substantial projection to the vestibular nuclei: area 2v located at the tip of the intraparietal sulcus, the parietoinsular vestibular cortex (PIVC) covering the most occipital part of the granular insula (Ig) and the retroinsular area (Ri or reipt), and the dorsolateral part of the somatosensory area 3a ("area 3aV" neck/trunk region). From physiological recording experiments, these three cortical fields were known to contain many neurons responding to stimulation of semicircular canals as well as to optokinetic (area 2v, PIVC) and somatosensory stimuli (PIVC, area 3a). These three regions form the inner cortical vestibular circuit. Besides these polysensory vestibular cortical fields, three other circumscribed cortical regions of the macaque brain were also found to project directly to the brainstem vestibular nuclei: a circumscribed part of the postarcuate premotor cortex (area 6pa), part of the agranular and the adjacent dysgranular cortex located around the cingulate sulcus (area 6c/23c), and a predominantly visual (optokinetic) association field located at the fundus of the lateral sulcus (area T3). These areas are known to have connections with the structures of the inner cortical vestibular circuit. Only a few efferent connections to the brainstem vestibular nuclei were found for the different parts of cytoarchitectonic area 7. Significant differences were found between the efferent innervation patterns of the axons originating in the six cortical areas mentioned and ending in the various compartments of the vestibular nuclear complex. Vestibular nuclei with a dominant output to the gaze motor system of the brainstem receive efferent connections preferably from the parietoinsular vestibular cortex. Vestibular structures with their primary output to skeletomotor centers, however, receive stronger efferent connections from areas 6pa and 3a. The ventrolateral nucleus, which sends efferent axons to both the oculomotor and skeletomotor systems of the brainstem and the spinal cord, also receives its main cortical efferents from the somatomotor area 6 and from area 3aV. Through these connections the cortical somatomotor system may directly influence vestibuloocular and vestibulocollic reflexes. It is speculated that the corticofugal connections to the vestibular brainstem nuclei are predominantly inhibitory, suppressing vestibular reflexes during cortically controlled goal-directed movements.


Asunto(s)
Corteza Cerebral/fisiología , Núcleos Vestibulares/fisiología , Animales , Cerebelo/citología , Cerebelo/fisiología , Corteza Cerebral/citología , Vías Eferentes/citología , Vías Eferentes/fisiología , Histocitoquímica , Peroxidasa de Rábano Silvestre , Macaca fascicularis , Microelectrodos , Núcleos Vestibulares/citología , Aglutinina del Germen de Trigo-Peroxidasa de Rábano Silvestre Conjugada , Aglutininas del Germen de Trigo
20.
J Comp Neurol ; 332(1): 89-104, 1993 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-7685782

RESUMEN

Single- and multiple-unit recordings were made from nerve cells located in the different nuclei of the brainstem vestibular nuclear complex (VNC) of anaesthetized squirrel monkeys (Saimiri sciureus) by conventional stereotaxic techniques. After neurons responding to semicircular canal stimulation in a yaw, roll, or pitch direction or to otholith stimulation were identified, small amounts of retrograde tracer substances were deposited at the recording sites. Up to three different tracers were administered to different parts of the VNC in the same animal (Fast Blue, HRP-WGA, and Rhodamine-dextranes). After adequate survival times, the animals were sacrificed. Following histological processing, the cortical grey matter was screened systematically for cells labelled with the retrograde tracers (fluorescence microscopy or light microscopy for HRP processing). Labelled nerve cells which clearly project to the VNC directly were found predominantly in the cytoarchitectonic layer 5 of seven different cortical areas: 1) The parieto-insular vestibular cortex PIVC, which in squirrel monkeys consists mainly of the medial area Ri and parts of the anterior area Ig; 2) area 7ant, which presumably corresponds to the macaque area 2v; 3) area 3aV, a vestibular field of area 3a; 4) the temporal area T3 bordering on area Ri; 5) the premotor area 6a; and 6, 7) the areas 6c and 23c of the anterior cingulate cortex. The PIVC, area 7ant, and area 3aV form the "inner cortical vestibular circuit" (Guldin et al.: J. Comp. Neurol. 326:375-401, '92), while the other cortical areas mentioned also have direct projections to the structures of the inner cortical vestibular circuit. It is speculated that the direct projections of the cortical vestibular structures to the brainstem vestibular nuclei regulate the vestibulo-ocular, the vestibulo-spinal, and the optokinetic reflexes mediated through the VNC, thus preventing counteractions of these reflexes during voluntary, goal-directed head movements or locomotion.


Asunto(s)
Corteza Cerebral/anatomía & histología , Giro del Cíngulo/anatomía & histología , Propiocepción/fisiología , Saimiri/anatomía & histología , Corteza Somatosensorial/anatomía & histología , Núcleos Vestibulares/anatomía & histología , Animales , Transporte Axonal , Mapeo Encefálico , Corteza Cerebral/fisiología , Vías Eferentes/anatomía & histología , Giro del Cíngulo/fisiología , Peroxidasa de Rábano Silvestre , Membrana Otolítica/inervación , Postura , Saimiri/fisiología , Canales Semicirculares/inervación , Corteza Somatosensorial/fisiología , Técnicas Estereotáxicas , Núcleos Vestibulares/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA