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










Base de datos
Intervalo de año de publicación
1.
Neurobiol Aging ; 127: 23-32, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37030016

RESUMEN

We aimed to assess the utility of AT(N) classification in clinical practice. We measured the cerebrospinal fluid levels of amyloid-ß (Aß) 42, Aß40, phosphorylated tau, total tau, and neurofilament light chain (NfL) in samples from 230 patients with Alzheimer's clinical syndrome (ACS) and 328 patients with non-ACS. The concordance of two A-markers (i.e., Aß42 alone and the Aß42/Aß40 ratio) was not significantly different between the ACS (87.4%) and non-ACS (74.1%) groups. However, the frequency of discordant cases with AAß42-alone+/AAß-ratio- was significantly higher in the non-ACS (23.8%) than in the ACS group (7.4%). The concordance of two N-markers (i.e., total tau and NfL) was 40.4% in the ACS group and 24.4% in the non-ACS group. In the ACS samples, the frequency of biological Alzheimer's disease (i.e., A+T+) in Ntau+ cases was 95% while that in NNfL+ cases was 65%. Reflecting Aß deposition and neurodegeneration more accurately, we recommend the use of AT(N) classification defined by cerebrospinal fluid AAß-ratioTNNfL in clinical practice.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/líquido cefalorraquídeo , Proteínas tau/líquido cefalorraquídeo , Biomarcadores/líquido cefalorraquídeo , Péptidos beta-Amiloides/líquido cefalorraquídeo , Síndrome , Fragmentos de Péptidos/líquido cefalorraquídeo
2.
PLoS One ; 6(12): e28208, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22164246

RESUMEN

Various antidepressants are commonly used for the treatment of depression and several other neuropsychiatric disorders. In addition to their primary effects on serotonergic or noradrenergic neurotransmitter systems, antidepressants have been shown to interact with several receptors and ion channels. However, the molecular mechanisms that underlie the effects of antidepressants have not yet been sufficiently clarified. G protein-activated inwardly rectifying K(+) (GIRK, Kir3) channels play an important role in regulating neuronal excitability and heart rate, and GIRK channel modulation has been suggested to have therapeutic potential for several neuropsychiatric disorders and cardiac arrhythmias. In the present study, we investigated the effects of various classes of antidepressants on GIRK channels using the Xenopus oocyte expression assay. In oocytes injected with mRNA for GIRK1/GIRK2 or GIRK1/GIRK4 subunits, extracellular application of sertraline, duloxetine, and amoxapine effectively reduced GIRK currents, whereas nefazodone, venlafaxine, mianserin, and mirtazapine weakly inhibited GIRK currents even at toxic levels. The inhibitory effects were concentration-dependent, with various degrees of potency and effectiveness. Furthermore, the effects of sertraline were voltage-independent and time-independent during each voltage pulse, whereas the effects of duloxetine were voltage-dependent with weaker inhibition with negative membrane potentials and time-dependent with a gradual decrease in each voltage pulse. However, Kir2.1 channels were insensitive to all of the drugs. Moreover, the GIRK currents induced by ethanol were inhibited by sertraline but not by intracellularly applied sertraline. The present results suggest that GIRK channel inhibition may reveal a novel characteristic of the commonly used antidepressants, particularly sertraline, and contributes to some of the therapeutic effects and adverse effects.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Canales de Potasio/química , Amoxapina/farmacología , Animales , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Depresión/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Clorhidrato de Duloxetina , Etanol/farmacología , Femenino , Humanos , Ratones , Oocitos/metabolismo , ARN Mensajero/metabolismo , Sertralina/farmacología , Tiofenos/farmacología , Xenopus laevis
3.
Neuropsychopharmacology ; 35(7): 1560-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20393461

RESUMEN

Atomoxetine and reboxetine are commonly used as selective norepinephrine reuptake inhibitors (NRIs) for the treatment of attention-deficit/hyperactivity disorder and depression, respectively. Furthermore, recent studies have suggested that NRIs may be useful for the treatment of several other psychiatric disorders. However, the molecular mechanisms underlying the various effects of NRIs have not yet been sufficiently clarified. G-protein-activated inwardly rectifying K(+) (GIRK or Kir3) channels have an important function in regulating neuronal excitability and heart rate, and GIRK channel modulation has been suggested to be a potential treatment for several neuropsychiatric disorders and cardiac arrhythmias. In this study, we investigated the effects of atomoxetine and reboxetine on GIRK channels using the Xenopus oocyte expression assay. In oocytes injected with mRNA for GIRK1/GIRK2, GIRK2, or GIRK1/GIRK4 subunits, extracellular application of atomoxetine or reboxetine reversibly reduced GIRK currents. The inhibitory effects were concentration-dependent, but voltage-independent, and time-independent during each voltage pulse. However, Kir1.1 and Kir2.1 channels were insensitive to atomoxetine and reboxetine. Atomoxetine and reboxetine also inhibited GIRK currents induced by activation of cloned A(1) adenosine receptors or by intracellularly applied GTPgammaS, a nonhydrolyzable GTP analogue. Furthermore, the GIRK currents induced by ethanol were concentration-dependently inhibited by extracellularly applied atomoxetine but not by intracellularly applied atomoxetine. The present results suggest that atomoxetine and reboxetine inhibit brain- and cardiac-type GIRK channels, revealing a novel characteristic of clinically used NRIs. GIRK channel inhibition may contribute to some of the therapeutic effects of NRIs and adverse side effects related to nervous system and heart function.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Morfolinas/farmacología , Propilaminas/farmacología , Inhibidores de Captación Adrenérgica , Animales , Clorhidrato de Atomoxetina , Biofisica , Depresores del Sistema Nervioso Central/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Estimulación Eléctrica/métodos , Etanol/farmacología , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Guanosina 5'-O-(3-Tiotrifosfato)/farmacología , Potenciales de la Membrana/genética , Microinyecciones/métodos , Oocitos , Técnicas de Placa-Clamp/métodos , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , Reboxetina , Receptor de Adenosina A1/metabolismo , Xenopus laevis
4.
PLoS One ; 4(7): e6311, 2009 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-19621089

RESUMEN

BACKGROUND: Neurosteroids have various physiological and neuropsychopharmacological effects. In addition to the genomic effects of steroids, some neurosteroids modulate several neurotransmitter receptors and channels, such as N-methyl-D-aspartate receptors, gamma-aminobutyric acid type A (GABA(A)) receptors, and sigma(1) receptors, and voltage-gated Ca(2+) and K(+) channels. However, the molecular mechanisms underlying the various effects of neurosteroids have not yet been sufficiently clarified. In the nervous system, inwardly rectifying K(+) (Kir) channels also play important roles in the control of resting membrane potential, cellular excitability and K(+) homeostasis. Among constitutively active Kir2 channels in a major Kir subfamily, Kir2.3 channels are expressed predominantly in the forebrain, a brain area related to cognition, memory, emotion, and neuropsychiatric disorders. METHODOLOGY/PRINCIPAL FINDINGS: The present study examined the effects of various neurosteroids on Kir2.3 channels using the Xenopus oocyte expression assay. In oocytes injected with Kir2.3 mRNA, only pregnenolone sulfate (PREGS), among nine neurosteroids tested, reversibly potentiated Kir2.3 currents. The potentiation effect was concentration-dependent in the micromolar range, and the current-voltage relationship showed inward rectification. However, the potentiation effect of PREGS was not observed when PREGS was applied intracellularly and was not affected by extracellular pH conditions. Furthermore, although Kir1.1, Kir2.1, Kir2.2, and Kir3 channels were insensitive to PREGS, in oocytes injected with Kir2.1/Kir2.3 or Kir2.2/Kir2.3 mRNA, but not Kir2.1/Kir2.2 mRNA, PREGS potentiated Kir currents. These potentiation properties in the concentration-response relationships were less potent than for Kir2.3 channels, suggesting action of PREGS on Kir2.3-containing Kir2 heteromeric channels. CONCLUSIONS/SIGNIFICANCE: The present results suggest that PREGS acts as a positive modulator of Kir2.3 channels. Kir2.3 channel potentiation may provide novel insights into the various effects of PREGS.


Asunto(s)
Canales de Potasio/efectos de los fármacos , Pregnenolona/farmacología , Animales , Canales de Potasio/genética , ARN Mensajero/genética , Xenopus laevis
5.
Neuropharmacology ; 56(2): 499-506, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18977371

RESUMEN

Antiepileptic drugs protect against seizures by modulating neuronal excitability. Ethosuximide is selectively used for the treatment of absence epilepsy, and has also been shown to have the potential for treating several other neuropsychiatric disorders in addition to several antiepileptic drugs. Although ethosuximide inhibits T-type Ca(2+), noninactivating Na(+), and Ca(2+)-activated K(+) channels, the molecular mechanisms underlying the effects of ethosuximide have not yet been sufficiently clarified. G protein-activated inwardly rectifying K(+) channels (GIRK, or Kir3) play an important role in regulating neuronal excitability, heart rate and platelet aggregation. In the present study, the effects of various antiepileptic drugs on GIRK channels were examined first by using the Xenopus oocyte expression assay. Ethosuximide at clinically relevant concentrations inhibited GIRK channels expressed in Xenopus oocytes. The inhibition was concentration-dependent, but voltage-independent, and time-independent during each voltage pulse. However, the other antiepileptic drugs tested: phenytoin, valproic acid, carbamazepine, phenobarbital, gabapentin, topiramate and zonisamide, had no significant effects on GIRK channels even at toxic concentrations. In contrast, Kir1.1 and Kir2.1 channels were insensitive to all of the drugs tested. Ethosuximide also attenuated ethanol-induced GIRK currents. These inhibitory effects of ethosuximide were not observed when ethosuximide was applied intracellularly. In granule cells of cerebellar slices, ethosuximide inhibited GTPgammaS-activated GIRK currents. Moreover, ADP- and epinephrine-induced platelet aggregation was inhibited by ethosuximide, but not by charybdotoxin, a platelet Ca(2+)-activated K(+) channel blocker. These results suggest that the inhibitory effects of ethosuximide on GIRK channels may affect some of brain, heart and platelet functions.


Asunto(s)
Anticonvulsivantes/farmacología , Etosuximida/farmacología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/fisiología , Potenciales de la Membrana/efectos de los fármacos , Inhibición Neural/efectos de los fármacos , Adenosina Difosfato/farmacología , Animales , Animales Recién Nacidos , Compuestos de Bario/farmacología , Depresores del Sistema Nervioso Central/farmacología , Cerebelo/citología , Cloruros/farmacología , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Etanol/farmacología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Técnicas In Vitro , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Microinyecciones/métodos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Técnicas de Placa-Clamp/métodos , Xenopus
6.
Brain Tumor Pathol ; 25(1): 9-17, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18415661

RESUMEN

Ten glioma cell lines were examined for abnormalities of exon 1beta of the p14 gene and then for abnormalities of the entire p14 gene with the use of previous findings of other exons. Abnormalities of exon 1beta and the entire p14 gene were detected in eight of ten cases: homozygous deletion of the entire gene in six cases, hemizygous deletion of exon 1beta with homozygous deletion of downstream exons in one case, and hemizygous deletion of the entire coding region with a missense mutation (A97V) at the C-terminal nucleolar localization domain in one case. The remaining two cases revealed no such abnormalities. p14 gene expression was observed in the latter two cases and one case with A97V mutation in the hemizygously deleted coding region, but not in the others, including one case with only exon 1beta. In the three cases with p14 gene expression, immunocytochemistry revealed p14 nucleolar staining, suggesting the retention of the functional activity of p14 protein and, in the case with the A97V mutation, an insufficient mutational effect as well. The present findings of the frequent and variable p14 gene abnormalities, including rare-type ones with or without sufficient mutational effect in glioma cell lines, might be of value for better understanding of the p14 gene and its related pathways in glioma carcinogenesis.


Asunto(s)
Neoplasias Encefálicas/genética , Glioma/genética , Proteína p14ARF Supresora de Tumor/genética , Adulto , Anciano , Secuencia de Bases , Northern Blotting , Southern Blotting , Western Blotting , Línea Celular Tumoral , Femenino , Expresión Génica , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-mdm2/genética , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Ann N Y Acad Sci ; 1074: 42-51, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17105902

RESUMEN

Previously we have examined the effects of phencyclidine and clozapine upon the gene expression in the mouse brain. Recently, fluoxetine (Prozac) has been introduced for the therapeutic purpose as an antidepressant drug. Miledi et al. reported blockage of mouse muscle and neuronal nicotinic acetylcholine receptor by various concentrations of fluoxetine. Furthermore, Kobayashi et al. discovered that fluoxetine inhibits G protein activated inwardly rectifying G protein activated K(+) (GIRK) channels using Xenopus oocyte expression assay. From these experiments, we considered that it might be interesting to study the effects of fluoxetine on the gene expression in the mouse brain. After we have injected fluoxetine once a day into mouse for 20 days, we sacrificed mouse by decapitation and extracted RNA from mouse cerebral cortex. We used DNA microarray method for examining the gene expression in the brain. We found the downregulation of many spot signals in the fluoxetine-treated mouse, for example cholecystockinin and prostaglandin D2 synthase.


Asunto(s)
Antidepresivos de Segunda Generación/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Fluoxetina/administración & dosificación , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Animales , Regulación hacia Abajo , Inyecciones Intraperitoneales , Masculino , Ratones , Regulación hacia Arriba
8.
J Pharmacol Sci ; 102(3): 278-87, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17072103

RESUMEN

Paroxetine is commonly used as a selective serotonin reuptake inhibitor for the treatment of depression and other psychiatric disorders. However, the molecular mechanisms of the paroxetine effects have not yet been sufficiently clarified. Using Xenopus oocyte expression assays, we investigated the effects of paroxetine on G protein-activated inwardly rectifying K+ (GIRK) channels, which play an important role in reducing neuronal excitability in most brain regions and the heart rate. In oocytes injected with mRNAs for GIRK1/GIRK2, GIRK2, or GIRK1/GIRK4 subunits, paroxetine reversibly reduced inward currents through the expressed GIRK channels. The inhibition was concentration-dependent, but voltage-independent and time-independent during each voltage pulse. However, two structurally different antidepressants: milnacipran and trazodone, caused only a small inhibition of basal GIRK currents. Additionally, Kir1.1 and Kir2.1 channels were insensitive to all of the antidepressants. Furthermore, the GIRK currents induced by activation of A1 adenosine receptors or by ethanol were inhibited by extracellularly applied paroxetine in a concentration-dependent manner, but not affected by intracellularly applied paroxetine. Our results suggest that inhibition of GIRK channels by paroxetine may contribute partly to some of its therapeutic effects and adverse side effects.


Asunto(s)
Antidepresivos de Segunda Generación/farmacología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/antagonistas & inhibidores , Paroxetina/farmacología , Bloqueadores de los Canales de Potasio , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Inhibidores de Captación Adrenérgica/farmacología , Animales , Bario/farmacología , Ciclopropanos/farmacología , Electrofisiología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Concentración de Iones de Hidrógeno , Milnaciprán , Oocitos/metabolismo , Plásmidos/genética , Canales de Potasio de Rectificación Interna/efectos de los fármacos , Canales de Potasio de Rectificación Interna/metabolismo , Receptor de Adenosina A1/efectos de los fármacos , Receptor de Adenosina A1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trazodona/farmacología , Xenopus laevis
9.
Neuropsychopharmacology ; 31(3): 516-24, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16123769

RESUMEN

G protein-activated inwardly rectifying K+ channels (GIRK, also known as Kir3) are regulated by various G-protein-coupled receptors. Activation of GIRK channels plays an important role in reducing neuronal excitability in most brain regions and the heart rate. Ifenprodil, which is a clinically used cerebral vasodilator, interacts with several receptors, such as alpha1 adrenergic, N-methyl-D-aspartate, serotonin and sigma receptors. However, the molecular mechanisms underlying the various clinically related effects of ifenprodil remain to be clarified. Here, we examined the effects of ifenprodil on GIRK channels by using Xenopus oocyte expression assays. In oocytes injected with mRNAs for GIRK1/GIRK2, GIRK2 or GIRK1/GIRK4 subunits, ifenprodil reversibly reduced inward currents through the basal GIRK activity. The inhibition was concentration-dependent, but voltage- and time-independent, suggesting that ifenprodil may not act as an open channel blocker of the channels. In contrast, Kir1.1 and Kir2.1 channels in other Kir channel subfamilies were insensitive to ifenprodil. Furthermore, GIRK current responses activated by the cloned kappa-opioid receptor were similarly inhibited by ifenprodil. The inhibitory effects of ifenprodil were not observed when ifenprodil was applied intracellularly, and were not affected by extracellular pH, which changed the proportion of the uncharged to protonated ifenprodil, suggesting its action from the extracellular side. The GIRK currents induced by ethanol were also attenuated in the presence of ifenprodil. Our results suggest that direct inhibition of GIRK channels by ifenprodil, at submicromolar concentrations or more, may contribute to some of its therapeutic effects and adverse side effects.


Asunto(s)
Antagonistas Adrenérgicos alfa/farmacología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/antagonistas & inhibidores , Piperidinas/farmacología , Animales , Química Encefálica/genética , Química Encefálica/fisiología , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Femenino , Humanos , Concentración de Iones de Hidrógeno , Miocardio/metabolismo , Oocitos/fisiología , Plásmidos , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores Acoplados a Proteínas G/efectos de los fármacos , Xenopus
11.
J Neurooncol ; 74(2): 105-11, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16193380

RESUMEN

Nuclear factor (NF)-kappaB is known to control cellular proliferation and apoptosis. In malignant astrocytoma cells, it was reported that NF-kappaB was activated aberrantly and promoted their proliferation. Thus, inhibition of NF-kappaB activity is considered to be a promising therapeutic strategy for malignant astrocytoma. Recently, curcumin, the major constituent of turmeric, was reported to inhibit NF-kappaB activity. In this study, we investigated inhibitory effects of curcumin on NF-kappaB activity and cellular proliferation, and induction of apoptosis by curcumin in human malignant astrocytoma cell lines. Alteration of NF-kappaB activity in NP-2 human malignant astrocytoma cell line after treatment with curcumin was examined using electrophoretic mobility shift assay. Alterations of DNA synthesis and cellular growth in five human malignant astrocytoma cell lines after treatment with curcumin were examined using [(3)H]thymidine incorporation assay and the trypan blue dye exclusion method, respectively. Induction of apoptosis by curcumin in NP-2 and NP-3 human malignant astrocytoma cell lines was examined by DNA-fragmentation analysis and morphological observation. We found that the NF-kappaB activity in NP-2 was significantly reduced by curcumin. The DNA synthesis and the cellular growth were inhibited by curcumin in dose-dependent manner in all the five malignant astrocytoma cell lines. Nuclear condensation and fragmentation, and DNA fragmentation were observed in both NP-2 and NP-3 after the treatment with curcumin. These results indicate that curcumin inhibits the cellular proliferation and induces apoptosis in human malignant astrocytoma cell lines. These results are considered to be resulted from the inhibition of NF-kappaB activity by curcumin.


Asunto(s)
Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Astrocitoma/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Curcumina/uso terapéutico , FN-kappa B/efectos de los fármacos , Astrocitoma/metabolismo , Astrocitoma/patología , Ensayo de Cambio de Movilidad Electroforética , Humanos , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Timidina/metabolismo , Células Tumorales Cultivadas
12.
J Neuropathol Exp Neurol ; 64(6): 537-44, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15977646

RESUMEN

We detected morphologic abnormalities in the cerebral cortex of Mecp2-hemizygous (Mecp2(-/y)) mice. The cortical thickness of both somatosensory and motor cortices in mutants did not increase after 4 weeks of age, as compared with that in wild-type male mice. The density of neurons in those areas was significantly higher in layers II/III and V of Mecp2(-/y) mice than in wild-type mice, particularly in layers II/ III after 4 weeks of age. In layer II/III of the somatosensory cortex of Mecp2(-/y) mice, the diameter of the apical dendrite was thin and the number of dendritic spines was small. Electron microscopy revealed that two-week-old mutants already had numerous premature postsynaptic densities. These results indicate that Mecp2(-/y) mice suffered delayed neuronal maturation of the cerebral cortex and that the initial neuronal changes were caused by premature synaptogenesis. Rett syndrome patients with a heterozygous mutation of Mecp2 display developmental disorders including cortical malfunctions such as mental retardation, autism, and epilepsy. Our results provide evidence of the similarity with Rett syndrome brains in some respects and suggest that MeCP2/Mecp2 plays some role in synaptogenesis.


Asunto(s)
Corteza Cerebral , Proteínas Cromosómicas no Histona/deficiencia , Proteínas de Unión al ADN/deficiencia , Regulación del Desarrollo de la Expresión Génica/fisiología , Neuronas/patología , Sinapsis/patología , Factores de Edad , Animales , Animales Recién Nacidos , Recuento de Células , Tamaño de la Célula , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunohistoquímica/métodos , Masculino , Proteína 2 de Unión a Metil-CpG , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión/métodos , Neuronas/metabolismo , Neuronas/ultraestructura , Parvalbúminas/metabolismo , Proteínas Represoras , Tinción con Nitrato de Plata/métodos , Sinapsis/ultraestructura
13.
Cancer Sci ; 96(1): 38-41, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15649253

RESUMEN

Ten primary central nervous system lymphomas (PCNSL, brain lymphomas) were examined for p14 gene exon 1beta deletion, mutation and methylation by Southern blot analysis, nucleotide analysis of polymerase chain reaction clones and Southern blot-based methylation assay. In Southern blot analysis, from the signal densities of the hybridized bands and their similarities to those of exons 2 and 3 in our previous quantitative study, we found that exon 1beta was homozygously deleted in four cases, hemizygously deleted in five cases and not deleted in one case. Thus, the same deletion patterns covered the entire p14 gene for all cases except for one case, which suggested the hemizygous deletion of exons 1beta and 2 and homozygous deletion of exon 3. In addition, although exon 1beta mutation is rare in various tumors, we detected a missense mutation (L50R) in one case with a hemizygous deletion. Methylation of the 5'CpG island of the p14 gene was not suggested for any case without homozygous deletion. Our observation of frequent p14 gene abnormalities (90%) and inactivation (40-60%) was in striking contrast to the same pathological subtype of systemic lymphoma in which p14 gene abnormalities and inactivation were infrequent, suggesting a difference in carcinogenesis between PCNSL and systemic lymphoma.


Asunto(s)
Neoplasias Encefálicas/genética , Genes Supresores de Tumor , Linfoma/genética , Proteína p14ARF Supresora de Tumor/genética , Adulto , Anciano , Secuencia de Bases , Southern Blotting , Metilación de ADN , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación
14.
Life Sci ; 76(4): 407-15, 2004 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-15530503

RESUMEN

Interferon-alpha (IFNalpha) affects the opioid system. However, the direct action of IFNalpha on cloned opioid receptors remains unknown. Taking advantage of the functional coupling of cloned opioid receptors to G protein-activated inwardly rectifying K+ (GIRK) channels in a Xenopus oocyte expression system, we investigated the effects of recombinant IFNalpha on cloned mu-, delta- and kappa-opioid receptors. In oocytes co-injected with mRNAs for either the delta- or kappa-opioid receptor and for GIRK channel subunits, IFNalpha at high concentrations induced small GIRK currents that were abolished by naloxone, an opioid-receptor antagonist, compared with the control responses to each selective opioid agonist. Additionally, IFNalpha induced no significant current response in oocytes injected with mRNA(s) for either opioid receptor alone or GIRK channels. In oocytes expressing the mu-opioid receptor and GIRK channels, IFNalpha had little or no effect. Moreover, in oocytes expressing each opioid receptor and GIRK channels, GIRK current responses to each selective opioid agonist were not affected by the presence of IFNalpha, indicating no significant antagonism of IFNalpha toward the opioid receptors. Furthermore, IFNalpha had little or no effect on the mu/delta-, delta/kappa- or mu/kappa-opioid receptors expressed together with GIRK channels in oocytes. Our results suggest that IFNalpha weakly activates the delta and kappa-opioid receptors. The direct activation of the delta- and kappa-opioid receptors by IFNalpha may partly contribute to some of the IFNalpha effects under its high-dose medication.


Asunto(s)
Interferón-alfa/farmacología , Oocitos/efectos de los fármacos , Receptores Opioides/efectos de los fármacos , Xenopus laevis , Animales , Clonación Molecular , Relación Dosis-Respuesta a Droga , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G , Oocitos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , ARN Mensajero/metabolismo , Receptores Opioides/genética , Receptores Opioides/metabolismo , Receptores Opioides delta/efectos de los fármacos , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/efectos de los fármacos , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/efectos de los fármacos , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Proteínas Recombinantes
15.
Ann N Y Acad Sci ; 1025: 590-4, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15542767

RESUMEN

G protein-activated inwardly rectifying K+ (GIRK, Kir3) channels play an important role in the inhibitory regulation of neuronal excitability in most brain regions and heart rate through activation of various G protein-coupled receptors, such as opioid, cannabinoid, and D2 dopamine receptors. Therefore, modulators of GIRK channels may affect many brain functions. We have shown using Xenopus oocyte expression assays that ethanol directly activates GIRK channels, whereas various antipsychotics (thioridazine, clozapine, pimozide, and haloperidol) inhibit the channels. Here we investigated not only the effects of various selective serotonin reuptake inhibitor (SSRI) antidepressants (fluoxetine, citalopram, fluvoxamine, and zimelidine) and risperidone, an atypical antipsychotic, on GIRK channels, but also those of the various drugs tested on other Kir channels using the Xenopus oocyte system. Fluoxetine inhibited GIRK channels, whereas the other SSRIs and risperidone had a small or no effect on the channels. In contrast, Kir1.1 and Kir2.1 channels were insensitive to ethanol and various SSRIs and antipsychotics, although thioridazine weakly inhibited Kir1.1 channels. It has been shown that the function of GIRK channels is involved in seizure susceptibility, antinociception by opioids, cannabinoids, or ethanol, and cocaine reinforcement in studies using GIRK knockout mice and weaver mutant mice that have mutant GIRK2 channels insensitive to G proteins and ethanol. Activation of GIRK channels by opioids, cannabinoids, or ethanol may be one of these key effects. Therefore, GIRK channel modulators might be potential agents for the treatment of users of addictive drugs, such as cocaine, opioids, cannabinoids, and ethanol, as well as for the treatment of epilepsy and pain.


Asunto(s)
Conducta Adictiva/metabolismo , Proteínas de Unión al GTP/fisiología , Canales de Potasio de Rectificación Interna/metabolismo , Trastornos Relacionados con Sustancias/metabolismo , Animales , Conducta Adictiva/tratamiento farmacológico , Femenino , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores de los Canales de Potasio/uso terapéutico , Trastornos Relacionados con Sustancias/tratamiento farmacológico , Xenopus laevis
16.
Neuropsychopharmacology ; 29(10): 1841-51, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15150531

RESUMEN

G protein-activated inwardly rectifying K+ channels (GIRK, also known as Kir3) are activated by various G protein-coupled receptors. GIRK channels play an important role in the inhibitory regulation of neuronal excitability in most brain regions and the heart rate. Modulation of GIRK channel activity may affect many brain functions. Here, we report the inhibitory effects of various antidepressants: imipramine, desipramine, amitriptyline, nortriptyline, clomipramine, maprotiline, and citalopram, on GIRK channels. In Xenopus oocytes injected with mRNAs for GIRK1/GIRK2, GIRK2 or GIRK1/GIRK4 subunits, the various antidepressants tested, except fluvoxamine, zimelidine, and bupropion, reversibly reduced inward currents through the basal GIRK activity at micromolar concentrations. The inhibitions were concentration-dependent with various degrees of potency and effectiveness, but voltage- and time-independent. In contrast, Kir1.1 and Kir2.1 channels in other Kir channel subfamilies were insensitive to all of the drugs. Furthermore, GIRK current responses activated by the cloned A1 adenosine receptor were similarly inhibited by the tricyclic antidepressant desipramine. The inhibitory effects of desipramine were not observed when desipramine was applied intracellularly, and were not affected by extracellular pH, which changed the proportion of the uncharged to protonated desipramine, suggesting its action from the extracellular side. The GIRK currents induced by ethanol were also attenuated in the presence of desipramine. Our results suggest that inhibition of GIRK channels by the tricyclic antidepressants and maprotiline may contribute to some of the therapeutic effects and adverse side effects, especially seizures and atrial arrhythmias in overdose, observed in clinical practice.


Asunto(s)
Antidepresivos/farmacología , Proteínas de Unión al GTP/fisiología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Rectificación Interna/metabolismo , Animales , Antidepresivos Tricíclicos/farmacología , Electrofisiología , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G , Corazón/efectos de los fármacos , Humanos , Miocardio/metabolismo , Oocitos/metabolismo , Canales de Potasio de Rectificación Interna/efectos de los fármacos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Xenopus laevis
17.
Br J Pharmacol ; 138(6): 1119-28, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12684268

RESUMEN

1. The effects of fluoxetine, a commonly used antidepressant drug, on G protein-activated inwardly rectifying K(+) channels (GIRK, Kir3) were investigated using Xenopus oocyte expression assays. 2. In oocytes injected with mRNAs for GIRK1/GIRK2, GIRK2 or GIRK1/GIRK4 subunits, fluoxetine reversibly reduced inward currents through the basal GIRK activity. The inhibition by fluoxetine showed a concentration-dependence, a weak voltage-dependence and a slight time-dependence with a predominant effect on the instantaneous current elicited by voltage pulses and followed by slight further inhibition. Furthermore, in oocytes expressing GIRK1/2 channels and the cloned Xenopus A(1) adenosine receptor, GIRK current responses activated by the receptor were inhibited by fluoxetine. In contrast, ROMK1 and IRK1 channels in other Kir channel subfamilies were insensitive to fluoxetine. 3. The inhibitory effect on GIRK channels was not obtained by intracellularly applied fluoxetine, and not affected by extracellular pH, which changed the proportion of the uncharged to protonated fluoxetine, suggesting that fluoxetine inhibits GIRK channels from the extracellular side. 4. The GIRK currents induced by ethanol were also attenuated in the presence of fluoxetine. 5. We demonstrate that fluoxetine, at low micromolar concentrations, inhibits GIRK channels that play an important role in the inhibitory regulation of neuronal excitability in most brain regions and the heart rate through activation of various G-protein-coupled receptors. The present results suggest that inhibition of GIRK channels by fluoxetine may contribute to some of its therapeutic effects and adverse side effects, particularly seizures in overdose, observed in clinical practice.


Asunto(s)
Fluoxetina/farmacología , Fluvoxamina/farmacología , Canales de Potasio de Rectificación Interna , Canales de Potasio/efectos de los fármacos , Zimeldina/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Relación Dosis-Respuesta a Droga , Etanol/antagonistas & inhibidores , Etanol/farmacología , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G , Corazón/efectos de los fármacos , Ratones , Canales de Potasio/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Xenopus laevis
18.
Jpn J Cancer Res ; 93(12): 1308-16, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12495470

RESUMEN

Using reverse transcription-polymerase chain reaction (RT-PCR), six primary brain lymphomas, pathologically diagnosed as diffuse large B-cell lymphoma, were examined for rearranged VH-D-JH sequences of the immunoglobulin heavy chain gene, focusing on somatic mutations and intraclonal heterogeneity. The reliability of the isolated PCR clones was confirmed by in situ hybridization (ISH) with complementarity-determining region (CDR) 3 oligonucleotide probes. Sequence analysis of the PCR clones revealed a high frequency of somatic mutation, ranging from 8.8 to 27.3% (mean 18.2%) in the VH gene segments in all the lymphomas. A significantly lower frequency of replacement (R) mutations than expected was also seen in their frameworks (FRs) in all cases. These findings suggested that the precursor cells were germinal center (GC)-related cells in these lymphomas. However, despite extensive cloning experiments, intraclonal heterogeneity was not detected in any case except for one in which it could not be ruled out. Thus, it seemed likely that all of our brain lymphomas were derived from GC-related cells and that at least most of them were from post-GC cells.


Asunto(s)
Neoplasias Encefálicas/genética , Reordenamiento Génico , Genes de Inmunoglobulinas , Cadenas Pesadas de Inmunoglobulina/genética , Linfoma de Células B/genética , Mutación , Anciano , Secuencia de Bases , Neoplasias Encefálicas/inmunología , Femenino , Humanos , Región Variable de Inmunoglobulina/genética , Hibridación in Situ , Linfoma de Células B/inmunología , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa
19.
Brain Res Mol Brain Res ; 100(1-2): 13-20, 2002 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-12008017

RESUMEN

14-3-3 protein is a brain-specific protein discovered by Moore and Perez, but at present is thought to be a multifunctional protein. To clarify the brain-specific function of the protein, we intend constructing a 14-3-3 eta gene knock-out mouse. As the first step of this process, we isolated the mouse 14-3-3 eta chain gene and determined its structure. The mouse gene is about 10 kb long and composed of two exons separated by a long intron. The transcription start site was identified and the polyadenylation signals (AATAAA) were found in exon 2 of the mouse gene. In the 5'-upstream sequence, we found several cis elements including a CRE sequence, a TATA box-like sequence, and a C/EBP element. Furthermore, the distribution of 14-3-3 eta mRNA in the mouse brain was examined by in situ hybridization histochemistry. The highest signals were found in the Purkinje cells of the cerebellum, the pyramidal cells of the hippocampus and the olfactory bulb neurons of the adult mouse. Neuronal expression of 14-3-3 eta in these regions mRNA may generally increase during postnatal brain development. The distribution of protein kinase C gamma in the mouse brain was also examined by immunohistochemistry. From the distribution of 14-3-3 eta mRNA and protein kinase C gamma in the mouse brain, the involvement of these compounds in the induction and maintenance of LTP was discussed.


Asunto(s)
Química Encefálica/genética , Encéfalo/metabolismo , Regulación de la Expresión Génica/fisiología , Isoenzimas/metabolismo , Neuronas/metabolismo , Proteína Quinasa C/metabolismo , ARN Mensajero/metabolismo , Tirosina 3-Monooxigenasa/química , Tirosina 3-Monooxigenasa/aislamiento & purificación , Proteínas 14-3-3 , Secuencia de Aminoácidos/genética , Animales , Secuencia de Bases/genética , Encéfalo/citología , Exones/genética , Genes Reguladores/genética , Inmunohistoquímica , Ratones , Datos de Secuencia Molecular , Poliadenilación/genética , Regiones Promotoras Genéticas/genética , TATA Box/genética , Tirosina 3-Monooxigenasa/genética
20.
J Neurosurg ; 96(5): 909-17, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12005399

RESUMEN

OBJECT: It has been suggested that nuclear factor (NF)-kappaB, a pleiotropic transcription factor, controls cell proliferation. The authors examined NF-kappaB activity and its participation in the growth of human malignant astrocytoma. METHODS: The authors examined NF-kappaB activity in human malignant astrocytoma cell lines and high-grade astrocytoma tissues by using electrophoretic mobility shift assays and immunohistochemical studies, respectively. In addition, messenger (m)RNA expression of p50 and RelA, which are representative subunits of NF-kappaB, and IkappaBalpha, which is a representative inhibitory protein of NF-KB, were analyzed using Northern blot hybridization in the astrocytoma cell lines. Furthermore, alterations in DNA synthesis and cell growth in the astrocytoma cell lines were examined after inhibition of NF-kappaB activity by RelA antisense oligodeoxynucleotide. The authors found NF-kappaB activity in all astrocytoma cell lines and high-grade astrocytoma tissues that were examined, but not in the fetal astrocyte strain or in normal cerebral tissue. Expression of p50, RelA, and IkappaBalpha mRNA was found in the fetal astrocyte strain and normal adult brain tissue, in addition to the astrocytoma cell lines. The relative levels of expression of these mRNAs were similar among these cell lines, the cell strain, and normal tissue. The RelA antisense oligodeoxynucleotide specifically reduced the levels of RelA mRNA expression and NF-kappaB activity in the astrocytoma cell lines, thus significantly inhibiting their DNA synthesis and cell growth. CONCLUSIONS: Human malignant astrocytoma cells have aberrant NF-KB activity, which promotes their growth. This activity is not associated with aberrant expression of p50 and RelA.


Asunto(s)
Astrocitoma , Neoplasias Encefálicas , Proteínas I-kappa B , FN-kappa B/genética , FN-kappa B/metabolismo , Factores de Edad , Astrocitos/citología , Astrocitos/metabolismo , Encéfalo/citología , División Celular/fisiología , ADN/biosíntesis , Proteínas de Unión al ADN/genética , Feto/citología , Humanos , Inhibidor NF-kappaB alfa , Subunidad p50 de NF-kappa B , Oligodesoxirribonucleótidos Antisentido/farmacología , ARN Mensajero/análisis , Factor de Transcripción ReIA , Células Tumorales Cultivadas/citología , Células Tumorales Cultivadas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA