Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Schizophr Res ; 251: 22-29, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36527956

RESUMEN

BACKGROUND: Cognitive disturbances in schizophrenia have been linked to a lower density of dendritic spines on pyramidal neurons in the prefrontal cortex (PFC). Complement component C4, which has previously been found at higher levels in schizophrenia, marks synapses for phagocytosis by microglia. Thus, elevated consumption of dendritic spines by microglia mediated through excessive complement activity may play a role in lower spine density in schizophrenia. However, it is unclear if microglia themselves have the molecular capacity for enhanced phagocytosis of spines in schizophrenia. METHODS: Transcript levels for complement components and microglia-specific phagocytic markers were quantified using quantitative PCR in the PFC of 62 matched pairs of schizophrenia and unaffected comparison subjects and in antipsychotic-exposed monkeys. RESULTS: Relative to comparison subjects, schizophrenia subjects had higher mRNA levels for C4 (+154 %); C1q (+69 %), which initiates the classical complement pathway that includes C4; and for microglia-specific markers that enable phagocytic activity including TAM receptor tyrosine kinases Axl (+27 %) and MerTK (+27 %) and lysosome-associated glycoprotein CD68 (+27 %) (all p ≤ .042). Transcript levels for microglial phagocytic markers were correlated with C4 mRNA levels in schizophrenia subjects (all r ≥ 0.31, p ≤ .015). We also found further evidence consistent with microglial activation in schizophrenia, including higher mRNA levels for THIK1 (TWIK-related halothane-inhibited potassium channel: +30 %) and lower mRNA levels for the purinergic receptor P2Y12 (-27 %) (all p ≤ .016). Transcript levels were unchanged in antipsychotic-exposed monkeys. CONCLUSIONS: These results are consistent with the presence of increased complement activity and an elevated molecular capacity of microglia for phagocytosis in the same schizophrenia subjects.


Asunto(s)
Antipsicóticos , Esquizofrenia , Animales , Esquizofrenia/tratamiento farmacológico , Antipsicóticos/farmacología , Antipsicóticos/uso terapéutico , Microglía , Corteza Prefrontal/metabolismo , Fagocitosis , ARN Mensajero/metabolismo , Haplorrinos
2.
Transl Psychiatry ; 11(1): 40, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436571

RESUMEN

Bipolar disorder and schizophrenia have multiple clinical and genetic features in common, including shared risk associated with overlapping susceptibility loci in immune-related genes. Higher activity of the nuclear factor-κB (NF-κB) transcription factor complex, which regulates the transcription of multiple immune markers, has been reported to contribute to immune activation in the prefrontal cortex in schizophrenia. These findings suggest the hypothesis that elevated NF-κB activity is present in the prefrontal cortex in bipolar disorder in a manner similar to that seen in schizophrenia. Therefore, we quantified levels of NF-κB-related mRNAs in the prefrontal cortex of 35 matched pairs of bipolar disorder and unaffected comparison subjects using quantitative PCR. We found that transcript levels were higher in the prefrontal cortex of bipolar disorder subjects for several NF-κB family members, NF-κB activation receptors, and NF-κB-regulated mRNAs, and were lower for an NF-κB inhibitor. Transcript levels for NF-κB family members, NF-κB activation receptors, and NF-κB-regulated mRNAs levels were also highly correlated with each other. This pattern of elevated transcript levels for NF-κB-related markers in bipolar disorder is similar to that previously reported in schizophrenia, suggesting that cortical immune activation is a shared pathophysiological feature between the two disorders.


Asunto(s)
Trastorno Bipolar , Esquizofrenia , Trastorno Bipolar/genética , Humanos , FN-kappa B/metabolismo , Corteza Prefrontal/metabolismo , ARN Mensajero , Esquizofrenia/genética
4.
Biol Psychiatry ; 85(1): 25-34, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30082065

RESUMEN

BACKGROUND: Transcript levels for cytokines and the viral restriction factor interferon-induced transmembrane protein are markedly higher in the prefrontal cortex in schizophrenia. These gene products are regulated by the nuclear factor-κB (NF-κB) transcriptional complex. NF-κB activity, which requires the formation of NF-κB family member heterodimers, is regulated by activation receptors, kinases, and inhibitors. Whether any of these factors are altered in schizophrenia is not known. It is also unclear whether NF-κB-related disturbances reflect ongoing cortical immune activation or a long-lasting response to a prenatal immune-related insult. METHODS: Transcript levels for NF-κB pathway markers were assessed using quantitative polymerase chain reaction in the prefrontal cortex from 1) 62 matched pairs of schizophrenia and unaffected comparison subjects, 2) antipsychotic-exposed monkeys, and 3) adult mice exposed prenatally to maternal immune activation or in adulthood to the immune stimulant polyinosinic-polycytidylic acid. RESULTS: In schizophrenia subjects, but not antipsychotic-exposed monkeys, we found higher messenger RNA levels for 1) most NF-κB family members, 2) all NF-κB activation receptors, 3) several kinases, and 4) one inhibitor (IκBα) whose transcript level is itself regulated by NF-κB activity. A similar pattern of elevated NF-κB-related messenger RNA levels was seen in adult mice that received daily polyinosinic-polycytidylic acid injections, but not in adult mice subjected to maternal immune activation in utero. CONCLUSIONS: Higher NF-κB activity, evidenced by elevated transcript levels for NF-κB family members, activation receptors, and kinases, may contribute to increased markers of cortical immune activation in schizophrenia.


Asunto(s)
Citocinas/sangre , FN-kappa B/sangre , Esquizofrenia/metabolismo , Adulto , Animales , Antipsicóticos/uso terapéutico , Estudios de Casos y Controles , Femenino , Humanos , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Corteza Prefrontal/inmunología , Embarazo , ARN Mensajero/sangre , Esquizofrenia/tratamiento farmacológico , Transducción de Señal
5.
Schizophr Res ; 201: 254-260, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29705007

RESUMEN

Altered cannabinoid 1 receptor (CB1R) expression has been reported in the brain of subjects with schizophrenia, a developmental mental illness that usually emerges in late adolescence/early adulthood. However, the developmental period at which changes in the CB1R expression appear in schizophrenia is unknown. To gain insight into this factor, we assessed the postnatal developmental trajectory of CB1R expression in the methylazoxymethanol (MAM) model of schizophrenia. Using in situ hybridization with film and grain analyses, CB1R messenger RNA (mRNA) levels were quantified in multiple brain regions, including the medial prefrontal cortex (mPFC), secondary motor cortex, dorsomedial and dorsolateral striatum, dorsal subregions and ventral subiculum of the hippocampus, of MAM-treated rats and normal controls at three developmental periods [juvenile - postnatal day (PD) 30; adolescence - PD45; and adulthood - PD85]. In all brain regions studied, CB1R mRNA levels were highest in juveniles and then decreased progressively toward adolescent and adult levels in control and MAM-treated rats. However, in MAM-treated rats, CB1R mRNA levels were lower in the mPFC at PD85 and higher in the dorsolateral striatum at PD45 and PD85 relative to controls. Cellular analyses confirmed the changes in CB1R mRNA expression in MAM-treated rats. These findings are in accordance with previous studies showing a decrease in the CB1R mRNA expression from juvenile period to adolescence to adulthood in cortical, striatal, and hippocampal regions. Additionally, similar to most of the schizophrenia-like signs observed in the MAM model, embryonic exposure to MAM leads to schizophrenia-related changes in CB1R mRNA expression that only emerge later in development.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , ARN Mensajero/metabolismo , Receptor Cannabinoide CB1/metabolismo , Esquizofrenia/metabolismo , Animales , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Masculino , Acetato de Metilazoximetanol/análogos & derivados , Distribución Aleatoria , Ratas Sprague-Dawley
6.
Neurobiol Dis ; 99: 58-65, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28007586

RESUMEN

Studies of genetics, serum cytokines, and autoimmune illnesses suggest that immune-related abnormalities are involved in the disease process of schizophrenia. Furthermore, direct evidence of cortical immune activation, including markedly elevated levels of many immune-related markers, have been reported in the prefrontal cortex in multiple cohorts of schizophrenia subjects. Within the prefrontal cortex in schizophrenia, deficits in the basilar dendritic spines of layer 3 pyramidal neurons and disturbances in inhibitory inputs to pyramidal neurons have also been commonly reported. Interestingly, microglia, the resident immune-related cells of the brain, also regulate excitatory and inhibitory input to pyramidal neurons. Consequently, in this review, we describe the cytological and molecular evidence of immune activation that has been reported in the brains of individuals with schizophrenia and the potential links between these immune-related disturbances with previously reported disturbances in pyramidal and inhibitory neurons in the disorder. Finally, we discuss the role that activated microglia may play in connecting these observations and as potential therapeutic treatment targets in schizophrenia.


Asunto(s)
Microglía/metabolismo , Corteza Prefrontal/metabolismo , Esquizofrenia/metabolismo , Animales , Humanos , Microglía/patología , Vías Nerviosas/metabolismo , Vías Nerviosas/patología , Corteza Prefrontal/patología , Esquizofrenia/epidemiología , Esquizofrenia/patología
7.
Am J Psychiatry ; 174(2): 163-171, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27523502

RESUMEN

OBJECTIVE: Lower dendritic spine density on layer 3 pyramidal cells in the dorsolateral prefrontal cortex (DLPFC) appears to contribute to cognitive dysfunction in schizophrenia, whereas psychosis is associated with excessive dopamine release in the striatum. These findings may be related via excitatory projections from the DLPFC to the ventral mesencephalon, the location of dopamine cells projecting to the striatum. Consistent with this hypothesis, deletion of the actin-related protein-2/3 (ARP2/3) complex, which regulates the actin cytoskeleton supporting dendritic spines, produced spine loss in cortical pyramidal cells and striatal hyperdopaminergia in mice. The authors sought to determine whether the ARP2/3 complex is altered in schizophrenia. METHOD: In matched pairs of schizophrenia and comparison subjects, transcript levels of ARP2/3 complex signaling pathway were assessed in laser-microdissected DLPFC layer 3 and 5 pyramidal cells and layer 3 parvalbumin interneurons, and in total DLPFC gray matter. RESULTS: Transcript levels of ARP2/3 complex subunits and of nucleation promotion factors that regulate the ARP2/3 complex were significantly lower in DLPFC layer 3 and 5 pyramidal cells in schizophrenia. In contrast, these transcripts were unaltered, or only modestly changed, in parvalbumin interneurons and DLPFC gray matter. CONCLUSIONS: Down-regulation of the ARP2/3 complex signaling pathway, a common final pathway for multiple signaling cascades that regulate the actin cytoskeleton, would compromise the structural stability of spines, leading to their loss. In concert with findings from deletion of the ARP2/3 complex in mice, these findings support the idea that spine deficits in the DLPFC may contribute to subcortical hyperdopaminergia in schizophrenia.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina/genética , Regulación hacia Abajo/genética , Expresión Génica/genética , Corteza Prefrontal/metabolismo , Células Piramidales/metabolismo , Esquizofrenia/genética , Transducción de Señal/genética , Adulto , Animales , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Esquizofrenia/diagnóstico
8.
Trends Neurosci ; 39(12): 797-798, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27832914

RESUMEN

Altered inhibition from parvalbumin-containing GABA neurons is thought to contribute to impaired gamma frequency oscillations and cognitive deficits in schizophrenia. Crabtree and colleagues report that proline dehydrogenase deficits produce excessive cytosolic levels of the GABA-mimetic l-proline which impairs GABA synthesis and gamma oscillations in a manner that mimics schizophrenia.


Asunto(s)
Prolina , Esquizofrenia , Neuronas GABAérgicas , Parvalbúminas , Corteza Prefrontal , Ácido gamma-Aminobutírico
9.
Schizophr Res ; 177(1-3): 3-9, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-26972474

RESUMEN

Dysfunction of prefrontal cortex (PFC) inhibitory neurons that express the calcium-binding protein parvalbumin or the neuropeptide somatostatin in schizophrenia may be related to disturbances in the migration, phenotypic specification, and/or maturation of these neurons. These pre- and postnatal developmental stages are regulated in a cell type-specific manner by various transcription factors and co-activators, fibroblast growth factor receptors (FgfR), and other molecular markers. Consequently, we used quantitative PCR to quantify mRNA levels for these developmental regulators in the PFC of 62 schizophrenia subjects in whom parvalbumin and somatostatin neuron disturbances were previously reported, and in antipsychotic-exposed monkeys. Relative to unaffected comparison subjects, subjects with schizophrenia exhibited elevated mRNA levels for 1) the transcription factor MafB, which is expressed by parvalbumin and somatostatin neurons as they migrate from the medial ganglionic eminence to the cortex, 2) the transcriptional coactivator PGC-1α, which is expressed postnatally by parvalbumin neurons to maintain parvalbumin levels and inhibitory function, and 3) FgfR1, which is required for the migration and phenotypic specification of parvalbumin and somatostatin neurons. Elevations in these markers were most prominent in younger schizophrenia subjects and were not present in antipsychotic-exposed monkeys. Finally, expression levels of other important developmental regulators (i.e. Dlx1, Dlx5, Dlx6, SATB1, Sip1/Zeb2, ST8SIA4, cMaf, Nkx6.2, and Arx) were not altered in schizophrenia. The over-expression of a subset of molecular markers with distinct roles in the pre- and postnatal development of parvalbumin and somatostatin neurons might reflect compensatory mechanisms to sustain the development of these neurons in the face of other insults.


Asunto(s)
Neuronas/metabolismo , Parvalbúminas/metabolismo , Corteza Prefrontal/metabolismo , Esquizofrenia/metabolismo , Somatostatina/metabolismo , Animales , Antipsicóticos/farmacología , Antipsicóticos/uso terapéutico , Benzodiazepinas/farmacología , Benzodiazepinas/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Haloperidol/farmacología , Haloperidol/uso terapéutico , Humanos , Macaca fascicularis , Factor de Transcripción MafB/metabolismo , Masculino , Persona de Mediana Edad , Neuronas/efectos de los fármacos , Olanzapina , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Corteza Prefrontal/efectos de los fármacos , ARN Mensajero/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Esquizofrenia/tratamiento farmacológico
10.
Schizophr Bull ; 42(2): 396-405, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26424323

RESUMEN

N-methyl-d-aspartate receptor (NMDAR) hypofunction in the dorsolateral prefrontal cortex (DLPFC) has been implicated in the pathology of schizophrenia. NMDAR activity is negatively regulated by some G protein-coupled receptors (GPCRs). Signaling through these GPCRs is reduced by Regulator of G protein Signaling 4 (RGS4). Thus, lower levels of RGS4 would enhance GPCR-mediated reductions in NMDAR activity and could contribute to NMDAR hypofunction in schizophrenia. In this study, we quantified RGS4 mRNA and protein levels at several levels of resolution in the DLPFC from subjects with schizophrenia and matched healthy comparison subjects. To investigate molecular mechanisms that could contribute to altered RGS4 levels, we quantified levels of small noncoding RNAs, known as microRNAs (miRs), which regulate RGS4 mRNA integrity after transcription. RGS4 mRNA and protein levels were significantly lower in schizophrenia subjects and were positively correlated across all subjects. The RGS4 mRNA deficit was present in pyramidal neurons of DLPFC layers 3 and 5 of the schizophrenia subjects. In contrast, levels of miR16 were significantly higher in the DLPFC of schizophrenia subjects, and higher miR16 levels predicted lower RGS4 mRNA levels. These findings provide convergent evidence of lower RGS4 mRNA and protein levels in schizophrenia that may result from increased expression of miR16. Given the role of RGS4 in regulating GPCRs, and consequently the strength of NMDAR signaling, these findings could contribute to the molecular substrate for NMDAR hypofunction in DLPFC pyramidal cells in schizophrenia.


Asunto(s)
MicroARNs/metabolismo , Corteza Prefrontal/metabolismo , Trastornos Psicóticos/metabolismo , Proteínas RGS/metabolismo , Esquizofrenia/metabolismo , Transducción de Señal/fisiología , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/metabolismo
11.
Am J Psychiatry ; 173(1): 60-8, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26337038

RESUMEN

OBJECTIVE: Alternative splicing of ErbB4 transcripts is dysregulated in the dorsolateral prefrontal cortex in schizophrenia. ErbB4 regulates the activity of parvalbumin interneurons, and therefore dysregulated ErbB4 splicing could contribute to lower parvalbumin interneuron activity and consequently lower parvalbumin levels in schizophrenia. However, ErbB4 is also present in calretinin interneurons, which are not affected in schizophrenia. Therefore, the authors hypothesized that dysregulated ErbB4 splicing occurs selectively in parvalbumin interneurons and is associated with lower parvalbumin levels in schizophrenia. METHOD: Tissue samples enriched in calretinin and parvalbumin interneurons were laser microdissected from dorsolateral prefrontal cortex layers 2 and 4, respectively, from matched pairs of schizophrenia and comparison subjects. Transcript levels for pan-ErbB4, four ErbB4 splicing variants (JM-a, JM-b, CYT-1, CYT-2), parvalbumin, and calretinin were quantified by quantitative polymerase chain reaction (qPCR) in each layer. Transcript levels for myocardial infarction associated transcript (MIAT), which regulates ErbB4 splicing, were quantified in gray matter by qPCR and in parvalbumin interneurons by microarray. RESULTS: Calretinin and parvalbumin mRNAs were preferentially expressed in layers 2 and 4, respectively. In schizophrenia subjects, lower parvalbumin levels, higher CYT-1 and JM-a levels, and lower CYT-2 and JM-b levels were detected selectively in layer 4. In layer 4, the JM-a/JM-b ratio was inversely correlated with parvalbumin levels in schizophrenia subjects. MIAT levels were preferentially higher in parvalbumin interneurons in schizophrenia subjects. CONCLUSIONS: These findings suggest that elevated MIAT expression alters ErbB4 splicing selectively in parvalbumin interneurons in schizophrenia. Dysregulated ErbB4 splicing in schizophrenia may contribute to lower activity of parvalbumin interneurons and an activity-dependent down-regulation of parvalbumin expression.


Asunto(s)
Parvalbúminas/metabolismo , Corteza Prefrontal/metabolismo , Receptor ErbB-4/genética , Esquizofrenia , Adulto , Empalme Alternativo , Regulación hacia Abajo , Femenino , Humanos , Interneuronas/metabolismo , Masculino , Esquizofrenia/genética , Esquizofrenia/metabolismo
12.
Biol Psychiatry ; 79(7): 595-603, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26210060

RESUMEN

Cannabis use has been reported to increase the risk of developing schizophrenia and to worsen symptoms of the illness. Both of these outcomes might be attributable to the disruption by cannabis of the endogenous cannabinoid system's spatiotemporal regulation of the inhibitory circuitry in the prefrontal cortex that is essential for core cognitive processes, such as working memory, which are impaired in schizophrenia. In the healthy brain, the endocannabinoid 2-arachidonylglycerol 1) is synthesized by diacylglycerol lipase in pyramidal neurons; 2) travels retrogradely to nearby inhibitory axon terminals that express the primary type 1 cannabinoid receptor (CB1R); 3) binds to CB1R, which inhibits gamma-aminobutyric acid release from the cholecystokinin-containing population of interneurons; and 4) is metabolized by either monoglyceride lipase, which is located in the inhibitory axon terminal, or by α-ß-hydrolase domain 6, which is co-localized presynaptically with diacylglycerol lipase. Investigations of the endogenous cannabinoid system in the prefrontal cortex of subjects with schizophrenia have found evidence of higher metabolism of 2-arachidonylglycerol, as well as both greater CB1R receptor binding and lower levels of CB1R messenger RNA and protein. Current views on the potential pathogenesis of these alterations, including disturbances in the development of the endogenous cannabinoid system, are discussed. In addition, how interactions between these alterations in the endocannabinoid system and those in other inhibitory neurons in the prefrontal cortex in subjects with schizophrenia might increase the liability to adverse outcomes with cannabis use is considered.


Asunto(s)
Ácidos Araquidónicos/efectos adversos , Endocannabinoides/efectos adversos , Glicéridos/efectos adversos , Interneuronas/efectos de los fármacos , Corteza Prefrontal/fisiopatología , Células Piramidales/efectos de los fármacos , Esquizofrenia/fisiopatología , Ácidos Araquidónicos/farmacología , Cognición , Endocannabinoides/farmacología , Glicéridos/farmacología , Humanos , ARN Mensajero/genética , Receptor Cannabinoide CB1/genética , Transducción de Señal , Ácido gamma-Aminobutírico/metabolismo
13.
Am J Psychiatry ; 172(11): 1112-21, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26133963

RESUMEN

OBJECTIVE: Immune-related abnormalities are commonly reported in schizophrenia, including higher mRNA levels for the viral restriction factor interferon-induced transmembrane protein (IFITM) in the prefrontal cortex. The authors sought to clarify whether higher IFITM mRNA levels and other immune-related disturbances in the prefrontal cortex are the consequence of an ongoing molecular cascade contributing to immune activation or the reflection of a long-lasting maladaptive response to an in utero immune-related insult. METHOD: Quantitative polymerase chain reaction was employed to measure mRNA levels for immune-related cytokines and transcriptional regulators, including those reported to regulate IFITM expression, in the prefrontal cortex from 62 schizophrenia and 62 healthy subjects and from adult mice exposed prenatally to maternal immune activation or in adulthood to the immune stimulant poly(I:C). RESULTS: Schizophrenia subjects had markedly higher mRNA levels for interleukin 6 (IL-6) (+379%) and interferon-ß (+29%), which induce IFITM expression; lower mRNA levels for Schnurri-2 (-10%), a transcriptional inhibitor that lowers IFITM expression; and higher mRNA levels for nuclear factor-κB (+86%), a critical transcription factor that mediates cytokine regulation of immune-related gene expression. In adult mice that received daily poly(I:C) injections, but not in offspring with prenatal exposure to maternal immune activation, frontal cortex mRNA levels were also markedly elevated for IFITM (+304%), multiple cytokines including IL-6 (+493%), and nuclear factor-κB (+151%). CONCLUSIONS: These data suggest that higher prefrontal cortex IFITM mRNA levels in schizophrenia may be attributable to adult, but not prenatal, activation of multiple immune markers and encourage further investigation into the potential role of these and other immune markers as therapeutic targets in schizophrenia.


Asunto(s)
Corteza Prefrontal/inmunología , ARN Mensajero/inmunología , Esquizofrenia/inmunología , Adulto , Animales , Antígenos de Diferenciación/efectos de los fármacos , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Estudios de Casos y Controles , Corteza Cerebral/inmunología , Corteza Cerebral/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Humanos , Inductores de Interferón/farmacología , Interferón beta/efectos de los fármacos , Interferón beta/genética , Interferón beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Masculino , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Persona de Mediana Edad , FN-kappa B/efectos de los fármacos , FN-kappa B/genética , FN-kappa B/inmunología , Poli I-C/farmacología , Corteza Prefrontal/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/inmunología , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/efectos de los fármacos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Esquizofrenia/genética , Factores de Transcripción/genética , Factores de Transcripción/inmunología
14.
Schizophr Res ; 167(1-3): 12-7, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25464914

RESUMEN

Alterations in inhibitory (GABA) neurons, including deficiencies in the GABA synthesizing enzyme GAD67, in the prefrontal cortex in schizophrenia are pronounced in the subpopulations of neurons that contain the calcium-binding protein parvalbumin or the neuropeptide somatostatin. The presence of similar illness-related deficits in the transcription factor Lhx6, which regulates prenatal development of parvalbumin and somatostatin neurons, suggests that cortical GABA neuron dysfunction may be related to disturbances in utero. Since the chemokine receptors CXCR4 and CXCR7 guide the migration of cortical parvalbumin and somatostatin neurons from their birthplace in the medial ganglionic eminence to their final destination in the neocortex, we sought to determine whether altered CXCR4 and/or CXCR7 mRNA levels were associated with disturbances in GABA-related markers in schizophrenia. Quantitative PCR was used to quantify CXCR4 and CXCR7 mRNA levels in the prefrontal cortex of 62 schizophrenia and 62 healthy comparison subjects that were previously characterized for markers of parvalbumin and somatostatin neurons and in antipsychotic-exposed monkeys. We found elevated mRNA levels for CXCR7 (+29%; p<.0001) and CXCR4 (+14%, p=.052) in schizophrenia subjects but not in antipsychotic-exposed monkeys. CXCR7 mRNA levels were inversely correlated with mRNA levels for GAD67, parvalbumin, somatostatin, and Lhx6 in schizophrenia but not in healthy subjects. These findings suggest that higher mRNA levels for CXCR7, and possibly CXCR4, may represent a compensatory mechanism to sustain the migration and correct positioning of cortical parvalbumin and somatostatin neurons in the face of other insults that disrupt the prenatal development of cortical GABA neurons in schizophrenia.


Asunto(s)
Interneuronas/patología , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Esquizofrenia/patología , Adulto , Análisis de Varianza , Femenino , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Humanos , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Parvalbúminas/genética , Parvalbúminas/metabolismo , ARN Mensajero/metabolismo , Receptores CXCR/genética , Receptores CXCR/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Somatostatina/genética , Somatostatina/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ácido gamma-Aminobutírico/metabolismo
15.
Schizophr Bull ; 41(1): 180-91, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24361861

RESUMEN

BACKGROUND: Schizophrenia is a neurodevelopmental disorder with altered expression of GABA-related genes in the prefrontal cortex (PFC). However, whether these gene expression abnormalities reflect disturbances in postnatal developmental processes before clinical onset or arise as a consequence of clinical illness remains unclear. METHODS: Expression levels for 7 GABA-related transcripts (vesicular GABA transporter [vGAT], GABA membrane transporter [GAT1], GABAA receptor subunit α1 [GABRA1] [novel in human and monkey cohorts], glutamic acid decarboxylase 67 [GAD67], parvalbumin, calretinin, and somatostatin [previously reported in human cohort, but not in monkey cohort]) were quantified in the PFC from 42 matched pairs of schizophrenia and comparison subjects and from 49 rhesus monkeys ranging in age from 1 week postnatal to adulthood. RESULTS: Levels of vGAT and GABRA1, but not of GAT1, messenger RNAs (mRNAs) were lower in the PFC of the schizophrenia subjects. As previously reported, levels of GAD67, parvalbumin, and somatostatin, but not of calretinin, mRNAs were also lower in these subjects. Neither illness duration nor age accounted for the levels of the transcripts with altered expression in schizophrenia. In monkey PFC, developmental changes in expression levels of many of these transcripts were in the opposite direction of the changes observed in schizophrenia. For example, mRNA levels for vGAT, GABRA1, GAD67, and parvalbumin all increased with age. CONCLUSIONS: Together with published reports, these findings support the interpretation that the altered expression of GABA-related transcripts in schizophrenia reflects a blunting of normal postnatal development changes, but they cannot exclude a decline during the early stages of clinical illness.


Asunto(s)
Corteza Prefrontal/metabolismo , Trastornos Psicóticos/genética , ARN Mensajero/metabolismo , Esquizofrenia/genética , Ácido gamma-Aminobutírico/metabolismo , Adulto , Animales , Calbindina 2/genética , Estudios de Casos y Controles , Progresión de la Enfermedad , Femenino , Proteínas Transportadoras de GABA en la Membrana Plasmática/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Glutamato Descarboxilasa/genética , Humanos , Macaca mulatta , Masculino , Persona de Mediana Edad , Parvalbúminas/genética , Corteza Prefrontal/crecimiento & desarrollo , Trastornos Psicóticos/metabolismo , Receptores de GABA-A/genética , Esquizofrenia/metabolismo , Somatostatina/genética , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/genética
16.
Schizophr Res ; 159(1): 124-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25107849

RESUMEN

The deleterious effects of cannabis use in schizophrenia have been linked, in part, to underlying disturbances in endogenous cannabinoid signaling in the prefrontal cortex. However, while receptor autoradiography studies of the primary cannabinoid receptor (CB1R) have consistently found higher CB1R binding in the prefrontal cortex in schizophrenia, deficits in CB1R mRNA levels and protein immunoreactivity have also been reported in the illness. To investigate this apparent discrepancy, we quantified CB1R binding using receptor autoradiography with the selective CB1R ligand [(3)H]-OMAR in the prefrontal cortex of 21 subjects with schizophrenia who were previously found to have lower levels of both CB1R mRNA using in situ hybridization and CB1R protein using radioimmunocytochemistry relative to matched healthy comparison subjects. We observed higher levels of [(3)H]-OMAR binding in the prefrontal cortex of schizophrenia subjects that did not appear to be attributable to psychotropic medications or substance abuse. The combination of lower levels of CB1R mRNA and immunoreactivity with higher CB1R receptor binding may reflect 1) altered trafficking of the receptor resulting in higher levels of membrane-bound CB1R or 2) higher CB1R affinity. In either case, greater CB1R receptor availability may contribute to the increased susceptibility of schizophrenia subjects to the deleterious effects of cannabis use.


Asunto(s)
Corteza Prefrontal/metabolismo , ARN Mensajero/metabolismo , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB1/metabolismo , Esquizofrenia/patología , Adulto , Análisis de Varianza , Autorradiografía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Piperidinas/farmacocinética , Tomografía de Emisión de Positrones , Corteza Prefrontal/diagnóstico por imagen , Corteza Prefrontal/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Pirazoles/farmacocinética , Tritio/farmacocinética
17.
Schizophr Bull ; 40(5): 952-7, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25053651

RESUMEN

Cognitive dysfunction is a disabling and core feature of schizophrenia. Cognitive impairments have been linked to disturbances in inhibitory (gamma-aminobutyric acid [GABA]) neurons in the prefrontal cortex. Cognitive deficits are present well before the onset of psychotic symptoms and have been detected in early childhood with developmental delays reported during the first year of life. These data suggest that the pathogenetic process that produces dysfunction of prefrontal GABA neurons in schizophrenia may be related to altered prenatal development. Interestingly, adult postmortem schizophrenia brain tissue studies have provided evidence consistent with a disease process that affects different stages of prenatal development of specific subpopulations of prefrontal GABA neurons. Prenatal ontogeny (ie, birth, proliferation, migration, and phenotypic specification) of distinct subpopulations of cortical GABA neurons is differentially regulated by a host of transcription factors, chemokine receptors, and other molecular markers. In this review article, we propose a strategy to investigate how alterations in the expression of these developmental regulators of subpopulations of cortical GABA neurons may contribute to the pathogenesis of cortical GABA neuron dysfunction and consequently cognitive impairments in schizophrenia.


Asunto(s)
Trastornos del Conocimiento/fisiopatología , Desarrollo Fetal/fisiología , Neuronas GABAérgicas/fisiología , Corteza Prefrontal/fisiopatología , Esquizofrenia/fisiopatología , Animales , Trastornos del Conocimiento/etiología , Humanos , Corteza Prefrontal/crecimiento & desarrollo , Esquizofrenia/etiología
18.
Schizophr Bull ; 40(5): 1053-61, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24837792

RESUMEN

Disturbances in parvalbumin- and somatostatin-containing neurons, including deficits in the gamma-aminobutyric acid (GABA)-synthesizing enzyme GAD67 in the prefrontal cortex (PFC) in schizophrenia, may be related to disrupted pre- and/or postnatal development. Deficits in the transcription factor Lhx6, which regulates parvalbumin and somatostatin neuron development, are associated with GAD67 deficits in schizophrenia. Therefore, we investigated the potential pre- and postnatal roles of Lhx6 in GABA-related disturbances using qPCR and/or in situ hybridization to quantify PFC levels of (1) Lhx6 mRNA in a new cohort of schizophrenia subjects; (2) Lhx6 mRNA in monkeys across postnatal development; (3) GABA-related mRNAs in Lhx6 heterozygous (Lhx6+/−) mice, which model Lhx6 deficits in schizophrenia; and (4) Lhx6 mRNA in GAD67+/− mice, which model GAD67 deficits in schizophrenia. Lhx6 mRNA levels were lower (−15%) in schizophrenia and correlated with lower GAD67 mRNA levels. In addition, Lhx6 mRNA levels declined 24% from the perinatal to prepubertal periods then stabilized in monkeys. Finally, GAD67, parvalbumin, and somatostatin mRNAs were not altered in Lhx6+/− mice, and Lhx6 mRNA was not altered in GAD67+/− mice. These data suggest that PFC Lhx6 and GAD67 mRNA deficits are common components of GABA neuron pathology in schizophrenia. An excessive early postnatal decline in Lhx6 mRNA might contribute to Lhx6 mRNA deficits in schizophrenia. However, a partial loss of Lhx6 is not sufficient in isolation to produce deficits in GAD67 mRNA and vice versa, suggesting that the concurrence of Lhx6 and GAD67 mRNA deficits in schizophrenia may instead be the consequence of a common upstream factor.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Glutamato Descarboxilasa/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Parvalbúminas/metabolismo , Corteza Prefrontal/metabolismo , Esquizofrenia/metabolismo , Somatostatina/metabolismo , Factores de Transcripción/metabolismo , Adulto , Factores de Edad , Anciano , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Macaca mulatta , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , ARN Mensajero/metabolismo
19.
Biol Psychiatry ; 76(2): 160-7, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24209773

RESUMEN

BACKGROUND: Higher tissue transcript levels of immune-related markers-including the recently discovered viral restriction factor interferon-induced transmembrane protein (IFITM), which inhibits viral entry and replication-have been reported in the prefrontal cortex in schizophrenia. Interestingly, mouse models of neuroinflammation have higher IFITM levels and deficits in γ-aminobutyric acid (GABA)-related markers that are similar to findings in schizophrenia, suggesting that a shared pathogenetic process might underlie diverse cortical pathology in the disorder. However, the cell types that overexpress IFITM messenger RNA (mRNA) in schizophrenia are unknown, and it is unclear whether higher IFITM mRNA levels are associated with lower GABA-related marker levels in the same schizophrenia subjects. METHODS: We used quantitative polymerase chain reaction and in situ hybridization with film and grain counting analyses to quantify IFITM mRNA levels in prefrontal cortex area 9 of 57 schizophrenia and 57 healthy comparison subjects and in antipsychotic-exposed monkeys. RESULTS: Quantitative polymerase chain reaction and in situ hybridization film analysis revealed markedly elevated IFITM mRNA levels (+114% and +117%, respectively) in prefrontal gray matter in schizophrenia. Interestingly, emulsion-dipped, Nissl-stained sections from schizophrenia and comparison subjects revealed IFITM mRNA expression in pia mater and blood vessels. The IFITM grain density over blood vessels was 71% higher in schizophrenia. The IFITM mRNA levels were negatively correlated with GABA-related mRNAs in the same schizophrenia subjects. CONCLUSIONS: The finding that schizophrenia subjects with higher IFITM mRNA levels in cortical blood vessels have greater disturbances in cortical GABA neurons suggests that these cell-type distinct pathological disturbances might be influenced by a shared upstream insult that involves immune activation.


Asunto(s)
Antígenos de Diferenciación/genética , Corteza Prefrontal/irrigación sanguínea , Corteza Prefrontal/metabolismo , Esquizofrenia/genética , Adulto , Animales , Antígenos de Diferenciación/metabolismo , Antipsicóticos/farmacología , Benzodiazepinas/farmacología , Femenino , Neuronas GABAérgicas/metabolismo , Haloperidol/farmacología , Humanos , Macaca fascicularis , Masculino , Persona de Mediana Edad , Olanzapina , Corteza Prefrontal/efectos de los fármacos , ARN Mensajero/metabolismo , Esquizofrenia/metabolismo
20.
Schizophr Res ; 147(1): 53-57, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23561296

RESUMEN

Adolescent cannabis use is associated with greater relative risk, increased symptom severity, and earlier age of onset of schizophrenia. We investigated whether this interaction may be partly attributable to disease-related disturbances in metabolism of the major cortical endocannabinoid 2-arachidonoylglycerol (2-AG). Transcript levels for the recently discovered 2-AG metabolizing enzyme, α-ß-hydrolase domain 6 (ABHD6), were assessed using quantitative PCR in the prefrontal cortex of schizophrenia and healthy subjects (n=84) and antipsychotic- or tetrahydrocannabinol-exposed monkeys. ABHD6 mRNA levels were elevated in schizophrenia subjects who were younger and had a shorter illness duration but not in antipsychotic- or tetrahydrocannabinol-exposed monkeys. Higher ABHD6 mRNA levels may increase 2-AG metabolism which may influence susceptibility to cannabis in the earlier stages of schizophrenia.


Asunto(s)
Endocannabinoides/metabolismo , Corteza Prefrontal/metabolismo , Esquizofrenia/patología , Adolescente , Adulto , Factores de Edad , Animales , Antipsicóticos/farmacología , Benzodiazepinas/farmacología , Berberina/análogos & derivados , Berberina/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Haloperidol/farmacología , Humanos , Macaca fascicularis , Masculino , Persona de Mediana Edad , Monoacilglicerol Lipasas/metabolismo , Olanzapina , Corteza Prefrontal/efectos de los fármacos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...