Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biol Pharm Bull ; 47(2): 361-365, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38311396

RESUMEN

Immune-related adverse events (irAEs) affect all organs and are associated with various symptoms. The identification of biomarkers that can predict irAEs may be particularly clinically useful. This study aimed to investigate whether the prognostic nutritional index (PNI) before the initiation of immune checkpoint inhibitor (ICI) treatment can predict the occurrence of irAEs. We conducted a survey of 111 patients with cancer who were receiving ICI fixed-dose monotherapy at Saga University Hospital from the time each ICI became available until January 2020. We compared the PNI between the patients with and without irAE expression, established a cutoff value for PNI associated with the development of irAEs, and investigated the incidence of irAEs and progression-free survival (PFS) in groups divided by the cutoff value. Patients with irAEs had significantly higher PNI than did those without, and there was a significant association between PNI and irAEs after adjusting for potential factors (odds ratio, 1.12; 95% confidence interval, 1.03-1.21). In addition, PNI ≥44.2 was associated with a significantly higher incidence of irAEs (75.0% vs. 35.2%, p = 0.0001) and significantly longer PFS than PNI <44.2 (p = 0.025). In conclusion, pretreatment PNI may be associated with the risk of developing irAEs in patients with advanced recurrent solid tumors. When the PNI is ≥44.2, patient management is important for avoiding serious AEs because while the treatment may be effective, the occurrence of irAEs is a concern.


Asunto(s)
Enfermedades del Sistema Inmune , Neoplasias , Humanos , Evaluación Nutricional , Pronóstico , Neoplasias/tratamiento farmacológico , Biomarcadores , Inmunoterapia/efectos adversos , Estudios Retrospectivos
2.
Neurobiol Dis ; 155: 105382, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33940180

RESUMEN

The unique fast spiking (FS) phenotype of cortical parvalbumin-positive (PV) neurons depends on the expression of multiple subtypes of voltage-gated potassium channels (Kv). PV neurons selectively express Kcns3, the gene encoding Kv9.3 subunits, suggesting that Kcns3 expression is critical for the FS phenotype. KCNS3 expression is lower in PV neurons in the neocortex of subjects with schizophrenia, but the effects of this alteration are unclear, because Kv9.3 subunit function is poorly understood. Therefore, to assess the role of Kv9.3 subunits in PV neuron function, we combined gene expression analyses, computational modeling, and electrophysiology in acute slices from the cortex of Kcns3-deficient mice. Kcns3 mRNA levels were ~ 50% lower in cortical PV neurons from Kcns3-deficient relative to wildtype mice. While silent per se, Kv9.3 subunits are believed to amplify the Kv2.1 current in Kv2.1-Kv9.3 channel complexes. Hence, to assess the consequences of reducing Kv9.3 levels, we simulated the effects of decreasing the Kv2.1-mediated current in a computational model. The FS cell model with reduced Kv2.1 produced spike trains with irregular inter-spike intervals, or stuttering, and greater Na+ channel inactivation. As in the computational model, PV basket cells (PVBCs) from Kcns3-deficient mice displayed spike trains with strong stuttering, which depressed PVBC firing. Moreover, Kcns3 deficiency impaired the recruitment of PVBC firing at gamma frequency by stimuli mimicking synaptic input observed during cortical UP states. Our data indicate that Kv9.3 subunits are critical for PVBC physiology and suggest that KCNS3 deficiency in schizophrenia could impair PV neuron firing, possibly contributing to deficits in cortical gamma oscillations in the illness.


Asunto(s)
Potenciales de Acción/fisiología , Neuronas/fisiología , Parvalbúminas/fisiología , Canales de Potasio con Entrada de Voltaje/deficiencia , Corteza Prefrontal/fisiopatología , Esquizofrenia/fisiopatología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Canales de Potasio con Entrada de Voltaje/genética , Esquizofrenia/genética
3.
Cereb Cortex ; 29(8): 3540-3550, 2019 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-30247542

RESUMEN

Visuospatial working memory (WM), which is impaired in schizophrenia, depends on a distributed network including visual, posterior parietal, and dorsolateral prefrontal cortical regions. Within each region, information processing is differentially regulated by subsets of γ-aminobutyric acid (GABA) neurons that express parvalbumin (PV), somatostatin (SST), or vasoactive intestinal peptide (VIP). In schizophrenia, WM impairments have been associated with alterations of PV and SST neurons in the dorsolateral prefrontal cortex. Here, we quantified transcripts selectively expressed in GABA neuron subsets across four cortical regions in the WM network from comparison and schizophrenia subjects. In comparison subjects, PV mRNA levels declined and SST mRNA levels increased from posterior to anterior regions, whereas VIP mRNA levels were comparable across regions except for the primary visual cortex (V1). In schizophrenia subjects, each transcript in PV and SST neurons exhibited similar alterations across all regions, whereas transcripts in VIP neurons were unaltered in any region except for V1. These findings suggest that the contribution of each GABA neuron subset to inhibitory regulation of local circuitry normally differs across cortical regions of the visuospatial WM network and that in schizophrenia alterations of PV and SST neurons are a shared feature across these regions, whereas VIP neurons are affected only in V1.


Asunto(s)
Encéfalo/metabolismo , Neuronas GABAérgicas/metabolismo , Memoria a Corto Plazo/fisiología , Parvalbúminas/genética , Esquizofrenia/genética , Somatostatina/genética , Péptido Intestinal Vasoactivo/genética , Adulto , Estudios de Casos y Controles , Femenino , Perfilación de la Expresión Génica , Glutamato Descarboxilasa/genética , Humanos , Proteínas con Homeodominio LIM/genética , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Lóbulo Parietal/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , Corteza Prefrontal/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Opioides mu/genética , Esquizofrenia/fisiopatología , Procesamiento Espacial , Factores de Transcripción/genética , Corteza Visual/metabolismo
4.
Biochim Biophys Acta Biomembr ; 1859(10): 2076-2085, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28754537

RESUMEN

Tryptophan is an essential amino acid in humans and an important serotonin and melatonin precursor. Monocarboxylate transporter MCT10 is a member of the SLC16A family proteins that mediates low-affinity tryptophan transport across basolateral membranes of kidney, small intestine, and liver epithelial cells, although the precise transport mechanism remains unclear. Here we developed a simple functional assay to analyze tryptophan transport by human MCT10 using a deletion mutant for the high-affinity tryptophan permease Tat2 in Saccharomyces cerevisiae. tat2Δtrp1 cells are defective in growth in YPD medium because tyrosine present in the medium competes for the low-affinity tryptophan permease Tat1 with tryptophan. MCT10 appeared to allow growth of tat2Δtrp1 cells in YPD medium, and accumulate in cells deficient for Rsp5 ubiquitin ligase. These results suggest that MCT10 is functional in yeast, and is subject to ubiquitin-dependent quality control. Whereas growth of Tat2-expressing cells was significantly impaired by neutral pH, that of MCT10-expressing cells was nearly unaffected. This property is consistent with the transport mechanism of MCT10 via facilitated diffusion without a need for pH gradient across the plasma membrane. Single-nucleotide polymorphisms (SNPs) are known to occur in the human MCT10 coding region. Among eight SNP amino acid changes in MCT10, the N81K mutation completely abrogated tryptophan import without any abnormalities in the expression or localization. In the MCT10 modeled structure, N81 appeared to protrude into the putative trajectory of tryptophan. Plasma membrane localization of MCT10 and the variant proteins was also verified in human embryonic kidney 293T cells.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Sistemas de Transporte de Aminoácidos/metabolismo , Aminoácidos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/genética , Línea Celular , Membrana Celular/metabolismo , Células HEK293 , Humanos , Polimorfismo de Nucleótido Simple/genética , Triptófano/metabolismo , Tirosina/metabolismo , Ubiquitina/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo
5.
Genes Cells ; 21(2): 185-99, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26791143

RESUMEN

Ultraviolet (UV) B is a major factor in melanomagenesis. This fact is linked to the resistance of melanocytes to UVB-induced apoptosis. In this study, we characterized the involvement of Mcl-1L in the regulation of UVB-induced apoptosis in melanocytes and in melanoma cells. In melanocytes, apoptosis was not evident at 24 h after UVB irradiation. The Mcl-1L expression increased after UVB irradiation, and the high Mcl-1L expression continued for at least 24 h. This UVB-dependent increase in Mcl-1L was mediated by the MEK-ERK-pS-STAT3 (STAT3 phosphorylated at Ser727) pathway. The Ser727 phosphorylation facilitated nuclear localization of STAT3. In melanoma cells, the expression levels of Mcl-1L varied depending on the cell line. WM39 melanoma cells expressed high levels of Mcl-1L via the MEK-ERK-pS-STAT3 pathway and were resistant to UVB-induced apoptosis without up-regulation of Mcl-1L. In melanocytes and in WM39 cells, transfection with Mcl-1 siRNA promoted UVB-induced apoptosis. Immunohistochemical studies showed that melanoma cells in in situ lesions expressed high amounts of Mcl-1L. These results indicate that the high expression of Mcl-1L mediated by the MEK-ERK-pS-STAT3 pathway protects melanocytes and melanoma cells from UVB-induced apoptosis.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Melanocitos/citología , Melanoma/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Rayos Ultravioleta/efectos adversos , Apoptosis , Línea Celular Tumoral , Núcleo Celular/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Melanocitos/metabolismo , Melanoma/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Metástasis de la Neoplasia , Fosforilación/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Regulación hacia Arriba
6.
Psychiatry Res ; 339: 116084, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39033685

RESUMEN

Visuospatial working memory (vsWM), which is impaired in schizophrenia (SZ), is mediated by multiple cortical regions including the primary (V1) and association (V2) visual, posterior parietal (PPC) and dorsolateral prefrontal (DLPFC) cortices. In these regions, parvalbumin (PV) or somatostatin (SST) GABA neurons are altered in SZ as reflected in lower levels of activity-regulated transcripts. As PV and SST neurons receive excitatory inputs from neighboring pyramidal neurons, we hypothesized that levels of activity-regulated transcripts are also lower in pyramidal neurons in these regions. Thus, we quantified levels of four activity-regulated, pyramidal neuron-selective transcripts, namely adenylate cyclase-activating polypeptide-1 (ADCYAP1), brain-derived neurotrophic factor (BDNF), neuronal pentraxin-2 (NPTX2) and neuritin-1 (NRN1) mRNAs, in V1, V2, PPC and DLPFC from unaffected comparison and SZ individuals. In SZ, BDNF and NPTX2 mRNA levels were lower across all four regions, whereas ADCYAP1 and NRN1 mRNA levels were lower in V1 and V2. The regional pattern of deficits in BDNF and NPTX2 mRNAs was similar to that in transcripts in PV and SST neurons in SZ. These findings suggest that lower activity of pyramidal neurons expressing BDNF and/or NPTX2 mRNAs might contribute to alterations in PV and SST neurons across the vsWM network in SZ.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Memoria a Corto Plazo , Proteínas del Tejido Nervioso , Células Piramidales , ARN Mensajero , Esquizofrenia , Humanos , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología , Esquizofrenia/genética , Masculino , Células Piramidales/metabolismo , Adulto , Femenino , Memoria a Corto Plazo/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , ARN Mensajero/metabolismo , ARN Mensajero/genética , Persona de Mediana Edad , Proteína C-Reactiva/metabolismo , Proteína C-Reactiva/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Corteza Cerebral/metabolismo , Adulto Joven , Lóbulo Parietal/metabolismo
7.
Cereb Cortex ; 21(5): 999-1011, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20843900

RESUMEN

Dysfunction of the dorsolateral prefrontal cortex (DLPFC) in schizophrenia is associated with lamina-specific alterations in particular subpopulations of interneurons. In pyramidal cells, postsynaptic γ-aminobutyric acid (GABA(A)) receptors containing different α subunits are inserted preferentially in distinct subcellular locations targeted by inputs from specific interneuron subpopulations. We used in situ hybridization to quantify the laminar expression of α1, α2, α3, and α5 subunit, and of ß1-3 subunit, mRNAs in the DLFPC of schizophrenia, and matched normal comparison subjects. In subjects with schizophrenia, mean GABA(A) α1 mRNA expression was 17% lower in layers 3 and 4, α2 expression was 14% higher in layer 2, α5 expression was 15% lower in layer 4, and α3 expression did not differ relative to comparison subjects. The mRNA expression of ß2, which preferentially assembles with α1 subunits, was also 20% lower in layers 3 and 4, whereas ß1 and ß3 mRNA levels were not altered in schizophrenia. These expression differences were not attributable to medication effects or other potential confounds. These findings suggest that GABA neurotransmission in the DLPFC is altered at the postsynaptic level in a receptor subunit- and layer-specific manner in subjects with schizophrenia and support the hypothesis that GABA neurotransmission in this illness is predominantly impaired in certain cortical microcircuits.


Asunto(s)
Corteza Prefrontal/metabolismo , Receptores de GABA-A/genética , Esquizofrenia/genética , Esquizofrenia/metabolismo , Adulto , Anciano , Femenino , Regulación de la Expresión Génica/genética , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/biosíntesis , Receptores de GABA-A/biosíntesis , Esquizofrenia/fisiopatología , Transmisión Sináptica/genética
8.
Neuropsychopharmacology ; 47(12): 2061-2070, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35034100

RESUMEN

Visuospatial working memory (vsWM), which is impaired in schizophrenia (SZ), is mediated by a distributed cortical network. In one node of this network, the dorsolateral prefrontal cortex (DLPFC), altered expression of transcripts for actin assembly and mitochondrial oxidative phosphorylation (OXPHOS) have been reported in SZ. To understand the relationship between these processes, and the extent to which similar alterations are present in other regions of vsWM network in SZ, a subset of actin- (CDC42, BAIAP2, ARPC3, and ARPC4) and OXPHOS-related (ATP5H, COX4I1, COX7B, and NDUFB3) transcripts were quantified in DLPFC by RNA sequencing in 139 SZ and unaffected comparison (UC) subjects, and in DLPFC and three other regions of the cortical vsWM network by qPCR in 20 pairs of SZ and UC subjects. By RNA sequencing, levels of actin- and OXPHOS-related transcripts were significantly altered in SZ, and robustly correlated in both UC and SZ subject groups. By qPCR, cross-regional expression patterns of these transcripts in UC subjects were consistent with greater actin assembly in DLPFC and higher OXPHOS activity in primary visual cortex (V1). In SZ, CDC42 and ARPC4 levels were lower in all regions, BAIAP2 levels higher only in V1, and ARPC3 levels unaltered across regions. All OXPHOS-related transcript levels were lower in SZ, with the disease effect decreasing from posterior to anterior regions. The differential alterations in markers of actin assembly and energy production across regions of the cortical vsWM network in SZ suggest that each region may make specific contributions to vsWM impairments in the illness.


Asunto(s)
Esquizofrenia , Actinas/genética , Actinas/metabolismo , Humanos , Memoria a Corto Plazo , Fosforilación Oxidativa , Corteza Prefrontal/metabolismo , Esquizofrenia/genética , Esquizofrenia/metabolismo
9.
Curr Psychiatry Rep ; 12(4): 335-44, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20556669

RESUMEN

The hypothesis that alterations of cortical inhibitory gamma-aminobutyric acid (GABA) neurons are a central element in the pathology of schizophrenia has emerged from a series of postmortem studies. How such abnormalities may contribute to the clinical features of schizophrenia has been substantially informed by a convergence with basic neuroscience studies revealing complex details of GABA neuron function in the healthy brain. Importantly, activity of the parvalbumin-containing class of GABA neurons has been linked to the production of cortical network oscillations. Furthermore, growing knowledge supports the concept that gamma band oscillations (30-80 Hz) are an essential mechanism for cortical information transmission and processing. Herein we review recent studies further indicating that inhibition from parvalbumin-positive GABA neurons is necessary to produce gamma oscillations in cortical circuits; provide an update on postmortem studies documenting that deficits in the expression of glutamic acid decarboxylase67, which accounts for most GABA synthesis in the cortex, are widely observed in schizophrenia; and describe studies using novel, noninvasive approaches directly assessing potential relations between alterations in GABA, oscillations, and cognitive function in schizophrenia.


Asunto(s)
Corteza Cerebral/fisiopatología , Red Nerviosa/fisiopatología , Neuronas/metabolismo , Esquizofrenia/fisiopatología , Ácido gamma-Aminobutírico/metabolismo , Relojes Biológicos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Humanos , Red Nerviosa/metabolismo , Red Nerviosa/patología , Neuronas/patología , Esquizofrenia/metabolismo , Esquizofrenia/patología , Transmisión Sináptica/fisiología
10.
Seishin Shinkeigaku Zasshi ; 112(5): 439-52, 2010.
Artículo en Japonés | MEDLINE | ID: mdl-20560363

RESUMEN

Individuals with schizophrenia show disturbances in a number of brain functions that regulate cognitive, affective, motor, and sensory processing. The cognitive deficits associated with dysfunction of the dorsolateral prefrontal cortex result, at least in part, from abnormalities in GABA neurotransmission, as reflected in a specific pattern of altered expression of GABA-related molecules. First, mRNA levels for the 67-kilodalton isoform of glutamic acid decarboxylase (GAD67), an enzyme principally responsible for GABA synthesis, and the GABA membrane transporter GAT1, which regulates the reuptake of synaptically released GABA, are decreased in a subset of GABA neurons. Second, affected GABA neurons include those that express the calcium-binding protein parvalbumin (PV), because PV mRNA levels are decreased in the prefrontal cortex of subjects with schizophrenia and GAD67 mRNA is undetectable in almost half of PV-containing neurons. These changes are accompanied by decreased GAT1 expression in the presynaptic terminals of PV-containing neurons and by increased postsynaptic GABA-A receptor alpha2 subunit expression at the axon initial segments of pyramidal neurons. These findings indicate decreased GABA synthesis/release by PV-containing GABA neurons and compensatory changes at synapses formed by these neurons. Third, another subset of GABA neurons that express the neuropeptide somatostatin (SST) also appear to be affected because their specific markers, SST and neuropeptide Y mRNAs, are decreased in a manner highly correlated with the decreases in GAD67 mRNA. Finally, mRNA levels for GABA-A receptor subunits for synaptic (alpha1 and gamma2) and extra-synaptic (delta) receptors are decreased, indicating alterations in both synaptic and extra-synaptic GABA neurotransmission. Together, this pattern of changes indicates that the altered GABA neurotransmission is specific to PV-containing and SST-containing GABA neuron subsets and involves both synaptic and extra-synaptic GABA-A receptors. Our recent analyses demonstrated that this pattern exists across diverse cortical areas including the prefrontal, anterior cingulate, primary motor, and primary visual cortices. GABA neurotransmission by PV-containing and SST-containing neurons is important for the generation of cortical oscillatory activities in the gamma (30-100 Hz) and theta (4-7 Hz) bands, respectively. These oscillatory activities have been proposed to play critical roles in regulating the efficiency of information transfer between neurons and neuronal networks in the cortex. Altered cortical GABA neurotransmission appears to contribute to disturbances in diverse functions through affecting the generation of cortical oscillations in schizophrenia.


Asunto(s)
Corteza Cerebral/fisiopatología , Esquizofrenia/fisiopatología , Transmisión Sináptica/fisiología , Ácido gamma-Aminobutírico/fisiología , Humanos , Esquizofrenia/tratamiento farmacológico
11.
J Neurosci ; 28(48): 12691-9, 2008 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-19036962

RESUMEN

Animals with a neonatal ventral hippocampal lesion (NVHL) develop abnormal behaviors during or after adolescence, suggesting that early insults can have delayed consequences. Many of these behaviors depend on the prefrontal cortex (PFC), and we have reported that PFC pyramidal neurons of adult rats with an NVHL respond to stimulation of the ventral tegmental area with an increase in firing instead of the characteristic decrease. As the dopamine modulation of cortical interneurons matures during adolescence, these findings raise the possibility that maturation of local inhibitory circuits within the PFC may have been altered in NVHL rats. Here, we assessed the state of PFC interneurons in NVHL rats with in situ hybridization measures of the mRNAs for the calcium binding protein parvalbumin (PV) and the GABA synthesizing enzyme GAD(67), as well as with electrophysiological measures of interneuron function. Although no differences were observed with PV or GAD(67), whole-cell recordings in slices revealed abnormal responses to the D(2) agonist quinpirole in interneurons from NVHL rats. The loss of D(2) modulation of local inhibition in slices from NVHL rats was also evident in the absence of a lasting component in the D(2) attenuation of excitatory synaptic responses in pyramidal neurons, which in sham treated rats was picrotoxin sensitive. The results suggest that the neonatal lesion causes improper maturation, but not loss, of PFC interneurons during adolescence, a finding consistent with current interpretations of imaging data in schizophrenia that suggest a hyperactive, "noisy" cortex underlying dysfunction in the PFC and other cortical areas.


Asunto(s)
Envejecimiento/fisiología , Hipocampo/anomalías , Hipocampo/lesiones , Interneuronas/metabolismo , Neurogénesis/fisiología , Corteza Prefrontal/fisiopatología , Animales , Animales Recién Nacidos , Biomarcadores/análisis , Biomarcadores/metabolismo , Desnervación , Agonistas de Dopamina/farmacología , Glutamato Descarboxilasa/genética , Hipocampo/fisiopatología , Masculino , Inhibición Neural/fisiología , Vías Nerviosas/anomalías , Vías Nerviosas/lesiones , Vías Nerviosas/fisiopatología , Técnicas de Cultivo de Órganos , Parvalbúminas/genética , Técnicas de Placa-Clamp , Corteza Prefrontal/citología , Corteza Prefrontal/crecimiento & desarrollo , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Esquizofrenia/etiología , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología , Ácido gamma-Aminobutírico/biosíntesis
12.
Cereb Cortex ; 18(7): 1575-87, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18203698

RESUMEN

Alterations in the inhibitory circuitry of the dorsolateral prefrontal cortex (DLPFC) in schizophrenia include reduced expression of the messenger RNA (mRNA) for somatostatin (SST), a neuropeptide present in a subpopulation of gamma-aminobutyric acid (GABA) neurons. However, neither the cellular substrate nor the causal mechanisms for decreased SST mRNA levels in schizophrenia are known. We used in situ hybridization to quantify the compartmental, laminar, and cellular levels of SST mRNA expression in the DLPFC of 23 pairs of schizophrenia or schizoaffective disorder and control subjects. We also explored potential causal mechanisms by utilizing similar methods to analyze SST mRNA expression in 2 animal models. The expression of SST mRNA was significantly decreased in layers 2-superficial 6 of subjects with schizophrenia, but not in layer 1, deep 6 or the white matter. At the cellular level, both the density of cortical SST mRNA-positive neurons and the expression of SST mRNA per neuron were reduced in the subjects with schizophrenia. These alterations were not due to potential confounds and appeared to be a downstream consequence of impaired neurotrophin signaling through the trkB receptor. These findings support the hypothesis that a marked reduction in SST mRNA expression in a subset of GABA neurons contributes to DLPFC dysfunction in schizophrenia.


Asunto(s)
Corteza Prefrontal/metabolismo , Trastornos Psicóticos/metabolismo , ARN Mensajero/metabolismo , Esquizofrenia/metabolismo , Somatostatina/metabolismo , Adaptación Fisiológica/genética , Adulto , Anciano , Femenino , Regulación de la Expresión Génica/genética , Humanos , Masculino , Persona de Mediana Edad , Trastornos Psicóticos/genética , Esquizofrenia/genética , Somatostatina/genética
13.
Am J Psychiatry ; 165(4): 479-89, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18281411

RESUMEN

OBJECTIVE: Individuals with schizophrenia exhibit disturbances in a number of cognitive, affective, sensory, and motor functions that depend on the circuitry of different cortical areas. The cognitive deficits associated with dysfunction of the dorsolateral prefrontal cortex result, at least in part, from abnormalities in GABA neurotransmission, as reflected in a specific pattern of altered expression of GABA-related genes. Consequently, the authors sought to determine whether this pattern of altered gene expression is restricted to the dorsolateral prefrontal cortex or could also contribute to the dysfunction of other cortical areas in subjects with schizophrenia. METHOD: Real-time quantitative polymerase chain reaction was used to assess the levels of eight GABA-related transcripts in four cortical areas (dorsolateral prefrontal cortex, anterior cingulate cortex, and primary motor and primary visual cortices) of subjects (N=12) with schizophrenia and matched normal comparison subjects. RESULTS: Expression levels of seven transcripts were lower in subjects with schizophrenia, with the magnitude of reduction for each transcript comparable across the four areas. The largest reductions were detected for mRNA encoding somatostatin and parvalbumin, followed by moderate decreases in mRNA expression for the 67-kilodalton isoform of glutamic acid decarboxylase, the GABA membrane transporter GAT-1, and the alpha 1 and delta subunits of GABA(A) receptors. In contrast, the expression of calretinin mRNA did not differ between the subject groups in any of the four areas. CONCLUSIONS: Because the areas examined represent the major functional domains (e.g., association, limbic, motor, and sensory) of the cerebral cortex, our findings suggest that a conserved set of molecular alterations affecting GABA neurotransmission contribute to the pathophysiology of different clinical features of schizophrenia.


Asunto(s)
Perfilación de la Expresión Génica , Neocórtex/metabolismo , Esquizofrenia/genética , Ácido gamma-Aminobutírico/genética , Adulto , Anciano , Causas de Muerte , Femenino , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Giro del Cíngulo/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Corteza Motora/metabolismo , Neocórtex/química , Parvalbúminas/genética , Parvalbúminas/metabolismo , Reacción en Cadena de la Polimerasa , Corteza Prefrontal/química , Corteza Prefrontal/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Esquizofrenia/metabolismo , Esquizofrenia/mortalidad , Somatostatina/genética , Somatostatina/metabolismo , Transmisión Sináptica/genética , Transmisión Sináptica/fisiología , Distribución Tisular/genética , Ácido gamma-Aminobutírico/metabolismo
14.
Neurotox Res ; 14(2-3): 237-48, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19073429

RESUMEN

Impairments in cognitive control, such as those involved in working memory, are associated with dysfunction of the dorsolateral prefrontal cortex (DLPFC) in individuals with schizophrenia. This dysfunction appears to result, at least in part, from abnormalities in GABA-mediated neurotransmission. In this paper, we review recent findings indicating that the altered DLPFC circuitry in subjects with schizophrenia reflects changes in the expression of genes that encode selective presynaptic and postsynaptic components of GABA neurotransmission. Specifically, using a combination of methods, we found that subjects with schizophrenia exhibited expression deficits in GABA-related transcripts encoding presynaptic regulators of GABA neurotransmission, neuropeptide markers of specific subpopulations of GABA neurons, and certain subunits of the GABA(A) receptor. In particular, alterations in the expression of the neuropeptide somatostatin suggested that GABA neurotransmission is impaired in the Martinotti subset of GABA neurons that target the dendrites of pyramidal cells. In contrast, none of the GABA-related transcripts assessed to date were altered in the DLPFC of monkeys chronically exposed to antipsychotic medications, suggesting that the effects observed in the human studies reflect the disease process and not its treatment. In concert with previous findings, these data suggest that working memory dysfunction in schizophrenia may be attributable to altered GABA neurotransmission in specific DLPFC microcircuits.


Asunto(s)
Memoria/fisiología , Corteza Prefrontal/fisiopatología , Receptores de Neurotransmisores/fisiología , Esquizofrenia/fisiopatología , Transmisión Sináptica , Ácido gamma-Aminobutírico/fisiología , Animales , Biomarcadores/metabolismo , Expresión Génica , Humanos , Vías Nerviosas , Neuronas/metabolismo , Neuronas/fisiología , Corteza Prefrontal/metabolismo , Terminales Presinápticos/fisiología , Células Piramidales/metabolismo , Células Piramidales/fisiopatología , Esquizofrenia/genética , Esquizofrenia/metabolismo , Ácido gamma-Aminobutírico/genética , Ácido gamma-Aminobutírico/metabolismo
15.
Neuropeptides ; 72: 1-11, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30287150

RESUMEN

Social recognition is the sensitive domains of complex behavior critical for identification, interpretation and storage of socially meaningful information. Social recognition develops throughout childhood and adolescent, and is affected in a wide variety of psychiatric disorders. Recently, new data appeared on the molecular mechanisms of these processes, particularly, the excitatory-inhibitory (E/I) ratio which is modified during development, and then E/I balance is established in the adult brain. While E/I imbalance has been proposed as a mechanism for schizophrenia, it also seems to be the common mechanism in autism spectrum disorder (ASD). In addition, there is a strong suggestion that the oxytocinergic system is related to GABA-mediated E/I control in the context of brain socialization. In this review, we attempt to summarize the underpinning molecular mechanisms of E/I balance and its imbalance, and related biomarkers in the brain in healthiness and pathology. In addition, because there are increasing interest on oxytocin in the social neuroscience field, we will pay intensive attention to the role of oxytocin in maintaining E/I balance from the viewpoint of its effects on improving social impairment in psychiatric diseases, especially in ASD.


Asunto(s)
Encéfalo/fisiología , Oxitocina/fisiología , Reconocimiento en Psicología/fisiología , Conducta Social , Animales , Humanos
16.
Schizophr Res ; 185: 33-40, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28073605

RESUMEN

Involvement of the gamma-aminobutyric acid (GABA)-ergic system in schizophrenia pathogenesis through disrupted neurodevelopment has been highlighted in numerous studies. However, the function of common genetic variants of this system in determining schizophrenia risk is unknown. We therefore tested the association of 375 tagged SNPs in genes derived from the GABAergic system, such as GABAA receptor subunit genes, and GABA related genes (glutamate decarboxylase genes, GABAergic-marker gene, genes involved in GABA receptor trafficking and scaffolding) in Japanese schizophrenia case-control samples (n=2926; 1415 cases and 1511 controls). We observed nominal association of SNPs in nine GABAA receptor subunit genes and the GPHN gene with schizophrenia, although none survived correction for study-wide multiple testing. Two SNPs located in the GABRA1 gene, rs4263535 (Pallele=0.002; uncorrected) and rs1157122 (Pallele=0.006; uncorrected) showed top hits, followed by rs723432 (Pallele=0.007; uncorrected) in the GPHN gene. All three were significantly associated with schizophrenia and survived gene-wide multiple testing. Haplotypes containing associated variants in GABRA1 but not GPHN were significantly associated with schizophrenia. To conclude, we provided substantiating genetic evidence for the involvement of the GABAergic system in schizophrenia susceptibility. These results warrant further investigations to replicate the association of GABRA1 and GPHN with schizophrenia and to discern the precise mechanisms of disease pathophysiology.


Asunto(s)
Proteínas Portadoras/genética , Predisposición Genética a la Enfermedad , Proteínas de la Membrana/genética , Polimorfismo de Nucleótido Simple/genética , Esquizofrenia/genética , Transducción de Señal/genética , Adulto , Anciano , Pueblo Asiatico , Femenino , Frecuencia de los Genes , Pruebas Genéticas , Glutamato Descarboxilasa/genética , Humanos , Desequilibrio de Ligamiento , Masculino , Persona de Mediana Edad , Receptores de GABA/genética , Esquizofrenia/metabolismo , Ácido gamma-Aminobutírico/genética , Ácido gamma-Aminobutírico/metabolismo
17.
J Neurosci ; 25(2): 372-83, 2005 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-15647480

RESUMEN

Dysfunction of inhibitory neurons in the prefrontal cortex (PFC), represented by decreased expression of GABA-related genes such as the 67 kDa isoform of glutamate decarboxylase (GAD67) and parvalbumin (PV), appears to contribute to cognitive deficits in subjects with schizophrenia. We investigated the involvement of signaling mediated by brain-derived neurotrophic factor (BDNF) and its receptor tyrosine kinase TrkB in producing the altered GABA-related gene expression in schizophrenia. In 15 pairs of subjects with schizophrenia and matched control subjects, both BDNF and TrkB mRNA levels, as assessed by in situ hybridization, were significantly decreased in the PFC of the subjects with schizophrenia, whereas the levels of mRNA encoding the receptor tyrosine kinase for neurotrophin-3, TrkC, were unchanged. In this cohort, within-pair changes in TrkB mRNA levels were significantly correlated with those in both GAD67 and PV mRNA levels. Decreased BDNF, TrkB, and GAD67 mRNA levels were replicated in a second cohort of 12 subject pairs. In the combined cohorts, the correlation between within-pair changes in TrkB and GAD67 mRNA levels was significantly stronger than the correlation between the changes in BDNF and GAD67 mRNA levels. Neither BDNF nor TrkB mRNA levels were changed in the PFC of monkeys after a long-term exposure to haloperidol. Genetically introduced decreases in TrkB expression, but not in BDNF expression, also resulted in decreased GAD67 and PV mRNA levels in the PFC of adult mice; in addition, the cellular pattern of altered GAD67 mRNA expression paralleled that present in schizophrenia. Decreased TrkB signaling appears to underlie the dysfunction of inhibitory neurons in the PFC of subjects with schizophrenia.


Asunto(s)
Benzotropina/análogos & derivados , Factor Neurotrófico Derivado del Encéfalo/fisiología , Corteza Prefrontal/fisiopatología , Receptor trkB/fisiología , Esquizofrenia/fisiopatología , Adulto , Anciano , Animales , Antidiscinéticos/farmacología , Antipsicóticos/farmacología , Benzotropina/farmacología , Factor Neurotrófico Derivado del Encéfalo/genética , Estudios de Casos y Controles , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Haloperidol/farmacología , Humanos , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Persona de Mediana Edad , Inhibición Neural/fisiología , ARN Mensajero/metabolismo , Receptor trkB/genética , Esquizofrenia/tratamiento farmacológico , Transducción de Señal/fisiología , Ácido gamma-Aminobutírico/genética
18.
Am J Psychiatry ; 163(3): 534-7, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16513879

RESUMEN

OBJECTIVE: In the prefrontal cortex of subjects with schizophrenia, decreased signaling mediated by brain-derived neurotrophic factor (BDNF) and its receptor tyrosine kinase (TrkB) appears to contribute to the reduced expression of mRNA encoding the 67-kilodalton isoform of glutamate decarboxylase (GAD(67)), an enzyme for GABA synthesis. The authors examined in subjects with schizophrenia the effect in the human BDNF gene of a single nucleotide polymorphism (Val66Met), which reduces the trafficking and secretion of BDNF protein, on the expression of GAD(67) mRNA. METHOD: BDNF Val66Met genotyping was performed in 27 matched pairs of schizophrenia and comparison subjects. The impact of this polymorphism on prefrontal cortex GAD(67) mRNA expression in schizophrenia subjects was assessed by comparing within-pair differences in GAD(67) mRNA expression between schizophrenia subjects with versus without the Met66 allele after the level of BDNF mRNA expression was controlled. RESULTS: In contrast to expectations, the within-pair reduction in GAD(67) mRNA expression was not greater in schizophrenia subjects who were hetero- or homozygous for the Met66 allele. These subjects did tend to exhibit less marked within-pair reductions in both GAD(67) and BDNF mRNA expression compared with schizophrenia subjects homozygous for the Val allele. CONCLUSIONS: The presence of the BDNF Met66 allele does not contribute to the decreased level of GAD(67) mRNA expression in the prefrontal cortex of subjects with schizophrenia.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Polimorfismo de Nucleótido Simple , Corteza Prefrontal/metabolismo , Esquizofrenia/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Estudios de Cohortes , Diazepam/metabolismo , Femenino , Expresión Génica , Frecuencia de los Genes , Genotipo , Humanos , Masculino , Metionina , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor trkB/genética , Receptor trkB/metabolismo , Esquizofrenia/metabolismo , Ácido gamma-Aminobutírico/biosíntesis
19.
Schizophr Bull ; 42(4): 992-1002, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26980143

RESUMEN

In the cortex of subjects with schizophrenia, expression of glutamic acid decarboxylase 67 (GAD67), the enzyme primarily responsible for cortical GABA synthesis, is reduced in the subset of GABA neurons that express parvalbumin (PV). This GAD67 deficit is accompanied by lower cortical levels of other GABA-associated transcripts, including GABA transporter-1, PV, brain-derived neurotrophic factor (BDNF), tropomyosin receptor kinase B, somatostatin, GABAA receptor α1 subunit, and KCNS3 potassium channel subunit mRNAs. In contrast, messenger RNA (mRNA) levels for glutamic acid decarboxylase 65 (GAD65), another enzyme for GABA synthesis, are not altered. We tested the hypothesis that this pattern of GABA-associated transcript levels is secondary to the GAD67 deficit in PV neurons by analyzing cortical levels of these GABA-associated mRNAs in mice with a PV neuron-specific GAD67 knockout. Using in situ hybridization, we found that none of the examined GABA-associated transcripts had lower cortical expression in the knockout mice. In contrast, PV, BDNF, KCNS3, and GAD65 mRNA levels were higher in the homozygous mice. In addition, our behavioral test battery failed to detect a change in sensorimotor gating or working memory, although the homozygous mice exhibited increased spontaneous activities. These findings suggest that reduced GAD67 expression in PV neurons is not an upstream cause of the lower levels of GABA-associated transcripts, or of the characteristic behaviors, in schizophrenia. In PV neuron-specific GAD67 knockout mice, increased levels of PV, BDNF, and KCNS3 mRNAs might be the consequence of increased neuronal activity secondary to lower GABA synthesis, whereas increased GAD65 mRNA might represent a compensatory response to increase GABA synthesis.


Asunto(s)
Corteza Cerebral/metabolismo , Neuronas GABAérgicas/metabolismo , Expresión Génica/fisiología , Glutamato Descarboxilasa/metabolismo , Parvalbúminas/metabolismo , Esquizofrenia/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Conducta Animal , Modelos Animales de Enfermedad , Femenino , Glutamato Descarboxilasa/genética , Masculino , Memoria a Corto Plazo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibición Prepulso
20.
J Neurosci ; 23(15): 6315-26, 2003 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-12867516

RESUMEN

Markers of inhibitory neurotransmission are altered in the prefrontal cortex (PFC) of subjects with schizophrenia, and several lines of evidence suggest that these alterations may be most prominent in the subset of GABA-containing neurons that express the calcium-binding protein, parvalbumin (PV). To test this hypothesis, we evaluated the expression of mRNAs for PV, another calcium-binding protein, calretinin (CR), and glutamic acid decarboxylase (GAD67) in postmortem brain specimens from 15 pairs of subjects with schizophrenia and matched control subjects using single- and dual-label in situ hybridization. Signal intensity for PV mRNA expression in PFC area 9 was significantly decreased in the subjects with schizophrenia, predominantly in layers III and IV. Analysis at the cellular level revealed that this decrease was attributable principally to a reduction in PV mRNA expression per neuron rather than by a decreased density of PV mRNA-positive neurons. In contrast, the same measures of CR mRNA expression were not altered in schizophrenia. These findings were confirmed by findings from cDNA microarray studies using different probes. Across the subjects with schizophrenia, the decrease in neuronal PV mRNA expression was highly associated (r = 0.84) with the decrease in the density of neurons containing detectable levels of GAD67 mRNA. Furthermore, simultaneous detection of PV and GAD67 mRNAs revealed that in subjects with schizophrenia only 55% of PV mRNA-positive neurons had detectable levels of GAD67 mRNA. Given the critical role that PV-containing GABA neurons appear to play in regulating the cognitive functions mediated by the PFC, the selective alterations in gene expression in these neurons may contribute to the cognitive deficits characteristic of schizophrenia.


Asunto(s)
Benzotropina/análogos & derivados , Perfilación de la Expresión Génica , Haloperidol/análogos & derivados , Neuronas/metabolismo , Corteza Prefrontal/metabolismo , Esquizofrenia/genética , Ácido gamma-Aminobutírico/biosíntesis , Adulto , Anciano , Animales , Autorradiografía , Benzotropina/farmacología , Calbindina 2 , Recuento de Células , Femenino , Glutamato Descarboxilasa/genética , Haloperidol/farmacología , Humanos , Hibridación in Situ , Isoenzimas/genética , Macaca fascicularis , Masculino , Persona de Mediana Edad , Inhibición Neural , Neuronas/clasificación , Neuronas/efectos de los fármacos , Neuronas/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Parvalbúminas/genética , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/patología , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Proteína G de Unión al Calcio S100/genética , Esquizofrenia/metabolismo , Esquizofrenia/patología , Especificidad por Sustrato , Transmisión Sináptica , Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA