Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cereb Cortex ; 33(5): 1581-1594, 2023 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-35441221

RESUMEN

Reciprocal connections between primate dorsolateral prefrontal (DLPFC) and posterior parietal (PPC) cortices, furnished by subsets of layer 3 pyramidal neurons (PNs), contribute to cognitive processes including working memory (WM). A different subset of layer 3 PNs in each region projects to the homotopic region of the contralateral hemisphere. These ipsilateral (IP) and callosal (CP) projections, respectively, appear to be essential for the maintenance and transfer of information during WM. To determine if IP and CP layer 3 PNs in each region differ in their transcriptomes, fluorescent retrograde tracers were used to label IP and CP layer 3 PNs in the DLPFC and PPC from macaque monkeys. Retrogradely-labeled PNs were captured by laser microdissection and analyzed by RNAseq. Numerous differentially expressed genes (DEGs) were detected between IP and CP neurons in each region and the functional pathways containing many of these DEGs were shared across regions. However, DLPFC and PPC displayed opposite patterns of DEG enrichment between IP and CP neurons. Cross-region analyses indicated that the cortical area targeted by IP or CP layer 3 PNs was a strong correlate of their transcriptome profile. These findings suggest that the transcriptomes of layer 3 PNs reflect regional, projection type and target region specificity.


Asunto(s)
Cuerpo Calloso , Lóbulo Parietal , Animales , Haplorrinos , Neuronas , Expresión Génica
2.
Cereb Cortex ; 32(22): 5216-5229, 2022 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-35106549

RESUMEN

Visual spatial working memory (vsWM) is mediated by a distributed cortical network composed of multiple nodes, including primary visual (V1), posterior parietal (PPC), and dorsolateral prefrontal (DLPFC) cortices. Feedforward and feedback information is transferred among these nodes via projections furnished by pyramidal neurons (PNs) located primarily in cortical layer 3. Morphological and electrophysiological differences among layer 3 PNs across these nodes have been reported; however, the transcriptional signatures underlying these differences have not been examined in the human brain. Here we interrogated the transcriptomes of layer 3 PNs from 39 neurotypical human subjects across 3 critical nodes of the vsWM network. Over 8,000 differentially expressed genes were detected, with more than 6,000 transcriptional differences present between layer 3 PNs in V1 and those in PPC and DLPFC. Additionally, over 600 other genes differed in expression along the rostral-to-caudal hierarchy formed by these 3 nodes. Moreover, pathway analysis revealed enrichment of genes in V1 related to circadian rhythms and in DLPFC of genes involved in synaptic plasticity. Overall, these results show robust regional differences in the transcriptome of layer 3 PNs, which likely contribute to regional specialization in their morphological and physiological features and thus in their functional contributions to vsWM.


Asunto(s)
Memoria a Corto Plazo , Corteza Visual , Humanos , Memoria a Corto Plazo/fisiología , Corteza Visual/fisiología , Corteza Prefrontal/fisiología , Células Piramidales/fisiología , Expresión Génica
3.
Front Neuroanat ; 14: 578483, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33328902

RESUMEN

cAMP signaling has powerful, negative effects on cognitive functions of the primate dorsolateral prefrontal cortex (dlPFC), opening potassium channels to reduce firing and impair working memory, and increasing tau phosphorylation in aging neurons. This contrasts with cAMP actions in classic circuits, where it enhances plasticity and transmitter release. PDE4 isozymes regulate cAMP actions, and thus have been a focus of research and drug discovery. Previous work has focused on the localization of PDE4A and PDE4B in dlPFC, but PDE4D is also of great interest, as it is the predominant PDE4 isoform in primate association cortex, and PDE4D expression decreases with aging in human dlPFC. Here we used laser-capture microdissection transcriptomics and found that PDE4D message is enriched in pyramidal cells compared to GABAergic PV-interneurons in layer III of the human dlPFC. A parallel study in rhesus macaques using high-spatial resolution immunoelectron microscopy revealed the ultrastructural locations of PDE4D in primate dlPFC with clarity not possible in human post-mortem tissue. PDE4D was especially prominent in dendrites associated with microtubules, mitochondria, and likely smooth endoplasmic reticulum (SER). There was substantial postsynaptic labeling in dendritic spines, associated with the SER spine-apparatus near glutamatergic-like axospinous synapses, but sparse labeling in axon terminals. We also observed dense PDE4D labeling perisynaptically in astroglial leaflets ensheathing glutamatergic connections. These data suggest that PDE4D is strategically positioned to regulate cAMP signaling in dlPFC glutamatergic synapses and circuits, especially in postsynaptic compartments where it is localized to influence cAMP actions on intracellular trafficking, mitochondrial physiology, and internal calcium release.

4.
PLoS One ; 15(6): e0233895, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32497062

RESUMEN

Deficits in fast-spiking inhibitory interneurons (FSINs) within the dorsolateral prefrontal cortex (dlPFC) are hypothesized to underlie cognitive impairment associated with schizophrenia. Though representing a minority of interneurons, this key cell type coordinates broad neural network gamma-frequency oscillations, associated with cognition and cognitive flexibility. Here we report expression of GluN2D mRNA selectively in parvalbumin positive cells of human postmortem dlPFC tissue, but not pyramidal neurons, with little to no GluN2C expression in either cell type. In acute murine mPFC slices the GluN2C/D selective positive allosteric modulator (PAM), CIQ(+), increased the intrinsic excitability as well as enhanced NMDAR-mediated EPSCs onto FSINs. This increase in intrinsic excitability with GluN2C/D PAM was also observed in the Dlx 5/6+/- FSIN developmental deficit model with reported FSIN hypoexcitability. Together these data speak to selective modulation of FSINs by a GluN2D PAM, providing a potential mechanism to counter the FSIN-deficit seen in schizophrenia.


Asunto(s)
Interneuronas/metabolismo , Parvalbúminas/metabolismo , Corteza Prefrontal/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Potenciales de Acción , Adulto , Animales , Femenino , Expresión Génica , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Inhibición Neural , Células Piramidales/metabolismo , ARN Mensajero/genética , Receptores de N-Metil-D-Aspartato/genética
5.
J Neurosci ; 39(37): 7277-7290, 2019 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-31341029

RESUMEN

In primates, working memory function depends on activity in a distributed network of cortical areas that display different patterns of delay task-related activity. These differences are correlated with, and might depend on, distinctive properties of the neurons located in each area. For example, layer 3 pyramidal neurons (L3PNs) differ significantly between primary visual and dorsolateral prefrontal (DLPFC) cortices. However, to what extent L3PNs differ between DLPFC and other association cortical areas is less clear. Hence, we compared the properties of L3PNs in monkey DLPFC versus posterior parietal cortex (PPC), a key node in the cortical working memory network. Using patch-clamp recordings and biocytin cell filling in acute brain slices, we assessed the physiology and morphology of L3PNs from monkey DLPFC and PPC. The L3PN transcriptome was studied using laser microdissection combined with DNA microarray or quantitative PCR. We found that in both DLPFC and PPC, L3PNs were divided into regular spiking (RS-L3PNs) and bursting (B-L3PNs) physiological subtypes. Whereas regional differences in single-cell excitability were modest, B-L3PNs were rare in PPC (RS-L3PN:B-L3PN, 94:6), but were abundant in DLPFC (50:50), showing greater physiological diversity. Moreover, DLPFC L3PNs display larger and more complex basal dendrites with higher dendritic spine density. Additionally, we found differential expression of hundreds of genes, suggesting a transcriptional basis for the differences in L3PN phenotype between DLPFC and PPC. These data show that the previously observed differences between DLPFC and PPC neuron activity during working memory tasks are associated with diversity in the cellular/molecular properties of L3PNs.SIGNIFICANCE STATEMENT In the human and nonhuman primate neocortex, layer 3 pyramidal neurons (L3PNs) differ significantly between dorsolateral prefrontal (DLPFC) and sensory areas. Hence, L3PN properties reflect, and may contribute to, a greater complexity of computations performed in DLPFC. However, across association cortical areas, L3PN properties are largely unexplored. We studied the physiology, dendrite morphology and transcriptome of L3PNs from macaque monkey DLPFC and posterior parietal cortex (PPC), two key nodes in the cortical working memory network. L3PNs from DLPFC had greater diversity of physiological properties and larger basal dendrites with higher spine density. Moreover, transcriptome analysis suggested a molecular basis for the differences in the physiological and morphological phenotypes of L3PNs from DLPFC and PPC.


Asunto(s)
Neocórtex/fisiología , Lóbulo Parietal/fisiología , Corteza Prefrontal/fisiología , Células Piramidales/fisiología , Potenciales de Acción/fisiología , Animales , Femenino , Captura por Microdisección con Láser/métodos , Macaca mulatta , Masculino , Neocórtex/citología , Técnicas de Cultivo de Órganos , Lóbulo Parietal/citología , Corteza Prefrontal/citología
6.
Neuropsychopharmacology ; 44(4): 743-748, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30390066

RESUMEN

The mammalian neocortex is organized into layers distinguished by the size, packing density, and connectivity of their constituent neurons. Many neuropsychiatric illnesses are complex trait disorders with etiologic factors converging on neuronal protein networks. Cortical pathology of neuropsychiatric diseases, such as schizophrenia, is often restricted to, or more pronounced in, certain cortical layers, suggesting that genetic vulnerabilities manifest with laminar specificity. Thus, the ability to investigate cortical layer-specific protein levels in human postmortem brain is highly desirable. Here, we developed and validated a laser capture microdissection-mass spectrometry (LCM-MS) approach to quantify over 200 proteins in cortical layers 3 and 5 of two cohorts of human subjects as well as a monkey model of postmortem interval. LCM-MS was readily implementable and reliably identified protein patterns that differed between cortical layers 3 and 5. Our findings suggest that LCM-MS facilitates the precise quantification of proteins within individual cortical layers from human postmortem brain tissue, providing a powerful tool in the study of neuropsychiatric disease.


Asunto(s)
Captura por Microdisección con Láser/normas , Espectrometría de Masas/normas , Corteza Prefrontal/química , Corteza Prefrontal/patología , Adulto , Anciano , Animales , Autopsia , Estudios de Cohortes , Humanos , Captura por Microdisección con Láser/métodos , Macaca fascicularis , Masculino , Espectrometría de Masas/métodos , Persona de Mediana Edad , Reproducibilidad de los Resultados
7.
Biol Psychiatry ; 82(8): 594-600, 2017 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-28476208

RESUMEN

BACKGROUND: Impairments in certain cognitive processes (e.g., working memory) are typically most pronounced in schizophrenia (SZ), intermediate in bipolar disorder, and least in major depressive disorder. Given that working memory depends, in part, on neural circuitry that includes pyramidal cells in layer 3 (L3) and layer 5 (L5) of the dorsolateral prefrontal cortex (DLPFC), we sought to determine if transcriptome alterations in these neurons were shared or distinctive for each diagnosis. METHODS: Pools of L3 and L5 pyramidal cells in the DLPFC were individually captured by laser microdissection from 19 matched tetrads of unaffected comparison subjects and subjects with SZ, bipolar disorder, and major depressive disorder, and the messenger RNA was subjected to transcriptome profiling by microarray. RESULTS: In DLPFC L3 and L5 pyramidal cells, transcriptome alterations were numerous in subjects with SZ, but rare in subjects with bipolar disorder and major depressive disorder. The leading molecular pathways altered in subjects with SZ involved mitochondrial energy production and the regulation of protein translation. In addition, we did not find any significant transcriptome signatures related to psychosis or suicide. CONCLUSIONS: In concert, these findings suggest that molecular alterations in DLPFC L3 and L5 pyramidal cells might be characteristic of the disease processes operative in individuals diagnosed with SZ and thus might contribute to the circuitry alterations underlying cognitive dysfunction in individuals with SZ.


Asunto(s)
Trastorno Bipolar/diagnóstico , Trastorno Depresivo Mayor/diagnóstico , Corteza Prefrontal/patología , Células Piramidales/metabolismo , Esquizofrenia/diagnóstico , Transcriptoma/fisiología , Adulto , Femenino , Perfilación de la Expresión Génica , Humanos , Captura por Microdisección con Láser , Masculino , Análisis por Micromatrices , Persona de Mediana Edad , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Esquizofrenia/patología , Índice de Severidad de la Enfermedad , Transducción de Señal/fisiología
8.
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
9.
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
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.
Biol Psychiatry ; 78(11): 775-85, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25981171

RESUMEN

BACKGROUND: Cognitive dysfunction in schizophrenia is associated with a lower density of dendritic spines on deep layer 3 pyramidal cells in the dorsolateral prefrontal cortex (DLPFC). These alterations appear to reflect dysregulation of the actin cytoskeleton required for spine formation and maintenance. Consistent with this idea, altered expression of genes in the cell division cycle 42 (CDC42)-CDC42 effector protein (CDC42EP) signaling pathway, a key organizer of the actin cytoskeleton, was previously reported in DLPFC gray matter from subjects with schizophrenia. We examined the integrity of the CDC42-p21-activated serine/threonine protein kinases (PAK)-LIM domain-containing serine/threonine protein kinases (LIMK) signaling pathway in schizophrenia in a layer-specific and cell type-specific fashion in DLPFC deep layer 3. METHODS: Using laser microdissection, samples of DLPFC deep layer 3 were collected from 56 matched pairs of subjects with schizophrenia and comparison subjects, and levels of CDC42-PAK-LIMK pathway messenger RNAs were measured by quantitative polymerase chain reaction. These same transcripts also were quantified by microarray in samples of individually microdissected deep layer 3 pyramidal cells from a subset of the same subjects and from monkeys exposed to antipsychotics. RESULTS: Relative to comparison subjects, CDC42EP4, LIMK1, LIMK2, ARHGDIA, and PAK3 messenger RNA levels were significantly upregulated in subjects with schizophrenia in laminar and cellular samples. In contrast, CDC42 and PAK1 messenger RNA levels were significantly downregulated specifically in deep layer 3 pyramidal cells. These differences were not attributable to psychotropic medications or other comorbid factors. CONCLUSIONS: Findings from the present and prior studies converge on synergistic alterations in CDC42 signaling pathway that could destabilize actin dynamics and produce spine deficits preferentially in deep layer 3 pyramidal cells in schizophrenia.


Asunto(s)
Corteza Prefrontal/metabolismo , Esquizofrenia/patología , Transducción de Señal/fisiología , Proteína de Unión al GTP cdc42/metabolismo , Adulto , Animales , Antipsicóticos/farmacología , Benzodiazepinas/farmacología , Proteínas del Citoesqueleto , Femenino , Reguladores de Proteínas de Unión al GTP/genética , Reguladores de Proteínas de Unión al GTP/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Haloperidol/farmacología , Humanos , Captura por Microdisección con Láser , Quinasas Lim/genética , Quinasas Lim/metabolismo , Macaca fascicularis , Masculino , Persona de Mediana Edad , Olanzapina , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/patología , Proteínas de Unión al ARN , Transducción de Señal/efectos de los fármacos , Proteína de Unión al GTP cdc42/genética , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo , Proteínas de Unión al GTP rho , Inhibidor alfa de Disociación del Nucleótido Guanina rho/genética , Inhibidor alfa de Disociación del Nucleótido Guanina rho/metabolismo
12.
Cereb Cortex ; 25(8): 2295-305, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24610118

RESUMEN

Cortical pyramidal neuron activity is regulated in part through inhibitory inputs mediated by GABAA receptors. The subunit composition of these receptors confers distinct functional properties. Thus, developmental shifts in subunit expression will likely influence the characteristics of pyramidal cell firing and the functional maturation of processes that depend on these neurons. We used laser microdissection and PCR to quantify postnatal developmental changes in the expression of GABAA receptor subunits (α1, α2, α5, ß2, γ2, and δ) in layer 3 pyramidal cells of monkey prefrontal cortex, which are critical for working memory. To determine the specificity of these changes, we examined glutamate receptor subunits (AMPA Glur1 and NMDA Grin1) and conducted the same analyses in layer 5 pyramidal cells. Expression of GABAA receptor subunit mRNAs changed substantially, whereas glutamate receptor subunit changes were modest over postnatal development. Some transcripts (e.g., GABAA α1) progressively increased from birth until adulthood, whereas others (e.g., GABAA α2) declined with age. Changes in some transcripts were present in only one layer (e.g., GABAA δ). The development of GABAA receptor subunit expression in primate prefrontal pyramidal neurons is protracted and subunit- and layer-specific. These trajectories might contribute to the molecular basis for the maturation of working memory.


Asunto(s)
Corteza Prefrontal/crecimiento & desarrollo , Corteza Prefrontal/metabolismo , Células Piramidales/metabolismo , Receptores de GABA-A/metabolismo , Envejecimiento/metabolismo , Animales , Femenino , Captura por Microdisección con Láser , Macaca mulatta , Masculino , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
13.
Am J Psychiatry ; 171(1): 62-71, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24170294

RESUMEN

OBJECTIVE: In schizophrenia, alterations in markers of cortical GABA neurotransmission are prominent in parvalbumin-containing neurons. Parvalbumin neurons selectively express KCNS3, the gene encoding the Kv9.3 potassium channel α-subunit. Kv9.3 subunits are present in voltage-gated potassium channels that contribute to the precise detection of coincident excitatory synaptic inputs to parvalbumin neurons. This distinctive feature of parvalbumin neurons appears important for the synchronization of cortical neural networks in γ-oscillations. Because impaired prefrontal cortical γ-oscillations are thought to underlie the cognitive impairments in schizophrenia, the authors investigated whether KCNS3 mRNA levels are altered in the prefrontal cortex of schizophrenia subjects. METHOD: KCNS3 mRNA expression was evaluated by in situ hybridization in 22 matched pairs of schizophrenia and comparison subjects and by microarray analyses of pooled samples of individually dissected neurons that were labeled with Vicia villosa agglutinin (VVA), a parvalbumin neuron-selective marker, in a separate cohort of 14 pairs. Effects of chronic antipsychotic treatments on KCNS3 expression were tested in the prefrontal cortex of antipsychotic-exposed monkeys. RESULTS: By in situ hybridization, KCNS3 mRNA levels were 23% lower in schizophrenia subjects. At the cellular level, both KCNS3 mRNA-expressing neuron density and KCNS3 mRNA level per neuron were significantly lower. By microarray, KCNS3 mRNA levels were lower by 40% in VVA-labeled neurons from schizophrenia subjects. KCNS3 mRNA levels were not altered in antipsychotic-exposed monkeys. CONCLUSIONS: These findings reveal lower KCNS3 expression in prefrontal cortical parvalbumin neurons in schizophrenia, providing a molecular basis for compromised detection of coincident synaptic inputs to parvalbumin neurons that could contribute to altered γ-oscillations and impaired cognition in schizophrenia.


Asunto(s)
Neuronas/metabolismo , Parvalbúminas/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , Corteza Prefrontal/metabolismo , Esquizofrenia/genética , Adulto , Anciano , Animales , Antipsicóticos/farmacología , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Macaca fascicularis , Masculino , Persona de Mediana Edad , Neuronas/efectos de los fármacos , Canales de Potasio con Entrada de Voltaje/metabolismo , Corteza Prefrontal/efectos de los fármacos , Esquizofrenia/metabolismo
14.
Curr Opin Neurobiol ; 21(6): 866-72, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21680173

RESUMEN

Deficits of cognitive control in schizophrenia are associated with altered gamma oscillations in the prefrontal cortex. Paralbumin basket interneurons, which innervate the perisomatic region of pyramidal neurons, appear to play a key role in generating cortical gamma oscillations. In the prefrontal cortex of subjects with schizophrenia, alterations are present in both pre- and post-synaptic markers of the strength of GABA inputs from parvalbumin basket neurons to pyramidal neurons. These alterations may contribute to the neural substrate for impaired gamma oscillations in schizophrenia.


Asunto(s)
Vías Nerviosas/fisiopatología , Neuronas/fisiología , Corteza Prefrontal/fisiopatología , Esquizofrenia/fisiopatología , Transmisión Sináptica/fisiología , Humanos , Neuronas/patología
15.
Am J Psychiatry ; 168(9): 921-9, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21632647

RESUMEN

OBJECTIVE: Cognitive deficits in schizophrenia are associated with altered activity of the dorsolateral prefrontal cortex, which has been attributed to lower expression of the 67 kDa isoform of glutamic acid decarboxylase (GAD67), the major γ-aminobutyric acid (GABA)-synthesizing enzyme. However, little is known about the relationship of prefrontal GAD67 mRNA levels and illness severity, translation of the transcript into protein, and protein levels in axon terminals, the key site of GABA production and function. METHOD: Quantitative polymerase chain reaction was used to measure GAD67 mRNA levels in postmortem specimens of dorsolateral prefrontal cortex from subjects with schizophrenia and matched comparison subjects with no known history of psychiatric or neurological disorders (N=42 pairs). In a subset of this cohort in which potential confounds of protein measures were controlled (N=19 pairs), Western blotting was used to quantify tissue levels of GAD67 protein in tissue. In five of these pairs, multilabel confocal immunofluorescence was used to quantify GAD67 protein levels in the axon terminals of parvalbumin-containing GABA neurons, which are known to have low levels of GAD67 mRNA in schizophrenia. RESULTS: GAD67 mRNA levels were significantly lower in schizophrenia subjects (by 15%), but transcript levels were not associated with predictors or measures of illness severity or chronicity. In schizophrenia subjects, GAD67 protein levels were significantly lower in total gray matter (by 10%) and in parvalbumin axon terminals (by 49%). CONCLUSIONS: The findings that lower GAD67 mRNA expression is common in schizophrenia, that it is not a consequence of having the illness, and that it leads to less translation of the protein, especially in the axon terminals of parvalbumin-containing neurons, support the hypothesis that lower GABA synthesis in parvalbumin neurons contributes to dorsolateral prefrontal cortex dysfunction and impaired cognition in schizophrenia.


Asunto(s)
Trastornos del Conocimiento/genética , Glutamato Descarboxilasa/genética , Neuronas/metabolismo , Parvalbúminas/metabolismo , Corteza Prefrontal/enzimología , Esquizofrenia/genética , Ácido gamma-Aminobutírico/metabolismo , Adulto , Western Blotting , Trastornos del Conocimiento/patología , Estudios de Cohortes , Femenino , Humanos , Masculino , Microscopía Fluorescente , Persona de Mediana Edad , Neuronas/patología , Corteza Prefrontal/patología , Terminales Presinápticos , ARN Mensajero/genética , Valores de Referencia , Esquizofrenia/patología
16.
Arch Gen Psychiatry ; 68(1): 21-31, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20819979

RESUMEN

CONTEXT: Disturbances in markers of cortical γ-aminobutyric acid neurotransmission are a common finding in schizophrenia. The nature of γ-aminobutyric acid neurotransmission (hyperpolarizing or depolarizing) depends on the local intracellular chloride concentration. In the central nervous system, the intracellular chloride level is determined by the activity of 2 cation-chloride transporters, NKCC1 and KCC2. The activities of these transporters are in turn regulated by a network of serine-threonine kinases that includes OXSR1, STK39, and the WNK kinases WNK1, WNK3, and WNK4. OBJECTIVE: To compare the levels of NKCC1, KCC2, OXSR1, STK39, WNK1, WNK3, and WNK4 transcripts in prefrontal cortex area 9 between subjects with schizophrenia and healthy comparison subjects. DESIGN: Real-time quantitative polymerase chain reaction technique was used to measure transcript levels in the prefrontal cortex. SETTING: Human brain specimens were obtained from autopsies conducted at the Allegheny County Medical Examiner's Office, Pittsburgh, Pennsylvania. PARTICIPANTS: Postmortem brain specimens from 42 subjects with schizophrenia and 42 matched healthy comparison subjects. Brain specimens from 18 macaque monkeys exposed to haloperidol, olanzapine, or sham long-term. MAIN OUTCOME MEASURES: Relative expression levels for NKCC1, KCC2, OXSR1, STK39, WNK1, WNK3, and WNK4 transcripts compared with the mean expression level of 3 housekeeping transcripts. RESULTS: OXSR1 and WNK3 transcripts were substantially overexpressed in subjects with schizophrenia relative to comparison subjects. In contrast, NKCC1, KCC2, STK39, WNK1, and WNK4 transcript levels did not differ between subject groups. OXSR1 and WNK3 transcript expression levels were not changed in antipsychotic-exposed monkeys and were not affected by potential confounding factors in the subjects with schizophrenia. CONCLUSION: In schizophrenia, increased expression levels, and possibly increased kinase activities, of OXSR1 and WNK3 may shift the balance of chloride transport by NKCC1 and KCC2 and alter the nature of γ-aminobutyric acid neurotransmission in the prefrontal cortex.


Asunto(s)
Corteza Prefrontal/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Esquizofrenia/metabolismo , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Adulto , Autopsia , Estudios de Casos y Controles , Cloruros/metabolismo , Manual Diagnóstico y Estadístico de los Trastornos Mentales , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular , Transporte Iónico/genética , Masculino , Persona de Mediana Edad , Antígenos de Histocompatibilidad Menor , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esquizofrenia/genética , Simportadores de Cloruro de Sodio-Potasio/genética , Miembro 1 de la Familia de Transportadores de Soluto 12 , Miembro 2 de la Familia de Transportadores de Soluto 12 , Regulación hacia Arriba/genética , Proteína Quinasa Deficiente en Lisina WNK 1
17.
Neurobiol Dis ; 37(3): 738-46, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20034564

RESUMEN

The development of the human neocortex gives rise to a complex cytoarchitecture, grouping together cells with similar structure, connectivity and function. As a result, the six neocortical laminae show distinct molecular content. In schizophrenia, many anatomical and neurochemical changes appear to be restricted to a subset of lamina and/or cell types. In this study, we hypothesized that supragranular (SG; laminae II-III) and infragranular layers (IG; laminae V-VI) of area 46 in the human prefrontal cortex will show distinct and specific transcriptome alterations between subjects with schizophrenia and matched controls. To enhance sample homogeneity, we compared the gene expression patterns of the SG and IG layers of 8 matched middle-aged male subjects with schizophrenia to 8 pairwise matched controls using two replicate DNA microarrays for each sample. The study revealed strong disease-related laminar expression differences between the SG and IG layers. Expression changes were dominated by an overall underexpression of the IG-enriched genes in the schizophrenia subjects compared to normal control subjects. Furthermore, using a diagnosis-blind, unsupervised clustering of the control-derived SG or IG-enriched transcripts, the IG-enriched markers segregated the subjects with schizophrenia from the matched controls with a high degree of confidence. Importantly, multiple members of the semaphorin gene family reported altered gene expression, suggesting that the IG gene expression disturbances in subjects with schizophrenia may be a result of altered cortical development and disrupted brain connectivity.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Malformaciones del Sistema Nervioso/genética , Corteza Prefrontal/anomalías , Esquizofrenia/genética , Esquizofrenia/patología , Tipificación del Cuerpo/genética , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Persona de Mediana Edad , Familia de Multigenes/genética , Red Nerviosa/anomalías , Red Nerviosa/metabolismo , Red Nerviosa/fisiopatología , Malformaciones del Sistema Nervioso/metabolismo , Malformaciones del Sistema Nervioso/fisiopatología , Vías Nerviosas/anomalías , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiopatología , Neuronas/metabolismo , Neuronas/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Esquizofrenia/fisiopatología , Semaforinas/genética , Transducción de Señal/genética
18.
Biol Psychiatry ; 65(12): 1015-23, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19249749

RESUMEN

BACKGROUND: In schizophrenia, working memory dysfunction is associated with altered expression of gamma-aminobutyric acid (GABA)(A) receptor alpha1 and alpha2 subunits in the dorsolateral prefrontal cortex (DLPFC). In rodents, cortical alpha subunit expression shifts from low alpha1 and high alpha2 to high alpha1 and low alpha2 during early postnatal development. Because these two alpha subunits confer different functional properties to the GABA(A) receptors containing them, we determined whether this shift in alpha1 and alpha2 subunit expression continues through adolescence in the primate DLPFC, potentially contributing to the maturation of working memory during this developmental period. METHODS: Levels of GABA(A) receptor alpha1 and alpha2 subunit mRNAs were determined in the DLPFC of monkeys aged 1 week, 4 weeks, 3 months, 15-17 months (prepubertal), and 43-47 months (postpubertal) and in adult monkeys using in situ hybridization, followed by the quantification of alpha1 subunit protein by western blotting. We also performed whole-cell patch clamp recording of miniature inhibitory postsynaptic potentials (mIPSPs) in DLPFC slices prepared from pre- and postpubertal monkeys. RESULTS: The mRNA and protein levels of alpha1 and alpha2 subunits progressively increased and decreased, respectively, throughout postnatal development including adolescence. Furthermore, as predicted by the different functional properties of alpha1-containing versus alpha2-containing GABA(A) receptors, the mIPSP duration was significantly shorter in postpubertal than in prepubertal animals. CONCLUSIONS: In contrast to rodents, the developmental shift in GABA(A) receptor alpha subunit expression continues through adolescence in primate DLPFC, inducing a marked change in the kinetics of GABA neurotransmission. Disturbances in this shift might underlie impaired working memory in schizophrenia.


Asunto(s)
Corteza Prefrontal/crecimiento & desarrollo , Corteza Prefrontal/metabolismo , Receptores de GABA-A/biosíntesis , Animales , Western Blotting , Electrofisiología , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Hibridación in Situ , Cinética , Macaca mulatta , Masculino , Memoria a Corto Plazo/fisiología , Técnicas de Placa-Clamp , Sondas ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de GABA-A/genética , Maduración Sexual
19.
Biol Psychiatry ; 62(7): 711-21, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17568569

RESUMEN

BACKGROUND: Schizophrenia is characterized by complex gene expression changes. The transcriptome alterations in the prefrontal cortex have been the subject of several recent postmortem studies that yielded both convergent and divergent findings. METHODS: To increase measurement precision, we used a custom-designed DNA microarray platform with long oligonucleotides and multiple probes with replicates. The platform was designed to assess the expression of > 1800 genes specifically chosen because of their hypothesized roles in the pathophysiology of schizophrenia. The gene expression differences in dorsolateral prefrontal cortex samples from 14 matched pairs of schizophrenia and control subjects were analyzed with two technical replicates and four data mining approaches. RESULTS: In addition to replicating many expression changes in synaptic, oligodendrocyte, and signal transduction genes, we uncovered and validated a robust immune/chaperone transcript upregulation in the schizophrenia samples. CONCLUSIONS: We speculate that the overexpression of SERPINA3, IFITM1, IFITM2, IFITM3, CHI3L1, MT2A, CD14, HSPB1, HSPA1B, and HSPA1A in schizophrenia subjects represents a long-lasting and correlated signature of an early environmental insult during development that actively contributes to the pathophysiology of prefrontal dysfunction.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Inmunidad/genética , Chaperonas Moleculares/genética , Corteza Prefrontal/metabolismo , Esquizofrenia/genética , Adulto , Anciano , Alcoholismo/genética , Alcoholismo/psicología , Antipsicóticos/farmacología , Antipsicóticos/uso terapéutico , Análisis por Conglomerados , ADN/genética , Interpretación Estadística de Datos , Bases de Datos Genéticas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Oligodendroglía/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Oligonucleótidos/química , Oligonucleótidos/genética , Escalas de Valoración Psiquiátrica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología , Transducción de Señal/genética , Transducción de Señal/fisiología , Sinapsis/fisiología , Transcripción Genética/fisiología , Regulación hacia Arriba/fisiología
20.
Eur J Neurosci ; 25(6): 1843-54, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17432970

RESUMEN

The human neocortex is organized into six layers that are differentiated by the size and packing density of their constituent neurons. The gene products that guide the establishment of this lamination have been studied extensively, but the gene expression gradients present across the layers of the adult human neocortex are mostly unknown. As the supragranular (SG) and infragranular (IG) layers of the human prefrontal cortex (PFC) differ in their connectivity and developmental time course, we hypothesized that the SG and IG layers will show distinct differences in their transcriptomes. To test this prediction, we used laser capture microdissection coupled with DNA microarray transcriptome profiling. Sixty-nine genes exhibited robust and highly consistent expression differences between the SG and IG layers. For six selected markers, in addition to validating the microarray findings, in situ hybridization revealed a complex, subpopulation-specific neuronal distribution. The markers we identified are likely to be related to the functional differences between the SG and IG layers of the human PFC and can be used for assessing alterations in structure and function of this cortical region in human brain disorders.


Asunto(s)
Mapeo Encefálico , Neuronas/clasificación , Neuronas/metabolismo , Corteza Prefrontal/citología , Adulto , Perfilación de la Expresión Génica/métodos , Marcadores Genéticos , Humanos , Hibridación in Situ/métodos , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Corteza Prefrontal/anatomía & histología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...