Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Neurobiol Aging ; 127: 23-32, 2023 07.
Article in English | MEDLINE | ID: mdl-37030016

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/diagnosis , Alzheimer Disease/cerebrospinal fluid , tau Proteins/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Syndrome , Peptide Fragments/cerebrospinal fluid
2.
PLoS One ; 6(12): e28208, 2011.
Article in English | MEDLINE | ID: mdl-22164246

ABSTRACT

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.


Subject(s)
GTP-Binding Proteins/metabolism , Potassium Channels/chemistry , Amoxapine/pharmacology , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Depression/drug therapy , Dose-Response Relationship, Drug , Duloxetine Hydrochloride , Ethanol/pharmacology , Female , Humans , Mice , Oocytes/metabolism , RNA, Messenger/metabolism , Sertraline/pharmacology , Thiophenes/pharmacology , Xenopus laevis
3.
Neuropsychopharmacology ; 35(7): 1560-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20393461

ABSTRACT

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.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Membrane Potentials/drug effects , Morpholines/pharmacology , Propylamines/pharmacology , Adrenergic Uptake Inhibitors , Animals , Atomoxetine Hydrochloride , Biophysics , Central Nervous System Depressants/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation/methods , Ethanol/pharmacology , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Membrane Potentials/genetics , Microinjections/methods , Oocytes , Patch-Clamp Techniques/methods , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Reboxetine , Receptor, Adenosine A1/metabolism , Xenopus laevis
4.
PLoS One ; 4(7): e6311, 2009 Jul 21.
Article in English | MEDLINE | ID: mdl-19621089

ABSTRACT

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.


Subject(s)
Potassium Channels/drug effects , Pregnenolone/pharmacology , Animals , Potassium Channels/genetics , RNA, Messenger/genetics , Xenopus laevis
5.
Neuropharmacology ; 56(2): 499-506, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18977371

ABSTRACT

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.


Subject(s)
Anticonvulsants/pharmacology , Ethosuximide/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Membrane Potentials/drug effects , Neural Inhibition/drug effects , Adenosine Diphosphate/pharmacology , Animals , Animals, Newborn , Barium Compounds/pharmacology , Central Nervous System Depressants/pharmacology , Cerebellum/cytology , Chlorides/pharmacology , Dose-Response Relationship, Drug , Electric Stimulation , Ethanol/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , In Vitro Techniques , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Microinjections/methods , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques/methods , Xenopus
6.
Brain Tumor Pathol ; 25(1): 9-17, 2008.
Article in English | MEDLINE | ID: mdl-18415661

ABSTRACT

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.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Tumor Suppressor Protein p14ARF/genetics , Adult , Aged , Base Sequence , Blotting, Northern , Blotting, Southern , Blotting, Western , Cell Line, Tumor , Female , Gene Expression , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Middle Aged , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
7.
Ann N Y Acad Sci ; 1074: 42-51, 2006 Aug.
Article in English | MEDLINE | ID: mdl-17105902

ABSTRACT

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.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Brain/drug effects , Brain/metabolism , Fluoxetine/administration & dosage , Oligonucleotide Array Sequence Analysis/methods , Animals , Down-Regulation , Injections, Intraperitoneal , Male , Mice , Up-Regulation
8.
J Pharmacol Sci ; 102(3): 278-87, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17072103

ABSTRACT

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.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/antagonists & inhibitors , Paroxetine/pharmacology , Potassium Channel Blockers , Selective Serotonin Reuptake Inhibitors/pharmacology , Adrenergic Uptake Inhibitors/pharmacology , Animals , Barium/pharmacology , Cyclopropanes/pharmacology , Electrophysiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Hydrogen-Ion Concentration , Milnacipran , Oocytes/metabolism , Plasmids/genetics , Potassium Channels, Inwardly Rectifying/drug effects , Potassium Channels, Inwardly Rectifying/metabolism , Receptor, Adenosine A1/drug effects , Receptor, Adenosine A1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trazodone/pharmacology , Xenopus laevis
9.
Neuropsychopharmacology ; 31(3): 516-24, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16123769

ABSTRACT

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.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/antagonists & inhibitors , Piperidines/pharmacology , Animals , Brain Chemistry/genetics , Brain Chemistry/physiology , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Female , Humans , Hydrogen-Ion Concentration , Myocardium/metabolism , Oocytes/physiology , Plasmids , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, G-Protein-Coupled/drug effects , Xenopus
11.
J Neurooncol ; 74(2): 105-11, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16193380

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Astrocytoma/drug therapy , Cell Proliferation/drug effects , Curcumin/therapeutic use , NF-kappa B/drug effects , Astrocytoma/metabolism , Astrocytoma/pathology , Electrophoretic Mobility Shift Assay , Humans , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Thymidine/metabolism , Tumor Cells, Cultured
12.
J Neuropathol Exp Neurol ; 64(6): 537-44, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15977646

ABSTRACT

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.


Subject(s)
Cerebral Cortex , Chromosomal Proteins, Non-Histone/deficiency , DNA-Binding Proteins/deficiency , Gene Expression Regulation, Developmental/physiology , Neurons/pathology , Synapses/pathology , Age Factors , Animals , Animals, Newborn , Cell Count , Cell Size , Cerebral Cortex/growth & development , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry/methods , Male , Methyl-CpG-Binding Protein 2 , Mice , Mice, Knockout , Microscopy, Electron, Transmission/methods , Neurons/metabolism , Neurons/ultrastructure , Parvalbumins/metabolism , Repressor Proteins , Silver Staining/methods , Synapses/ultrastructure
13.
Cancer Sci ; 96(1): 38-41, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15649253

ABSTRACT

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.


Subject(s)
Brain Neoplasms/genetics , Genes, Tumor Suppressor , Lymphoma/genetics , Tumor Suppressor Protein p14ARF/genetics , Adult , Aged , Base Sequence , Blotting, Southern , DNA Methylation , Female , Humans , Male , Middle Aged , Mutation
14.
Life Sci ; 76(4): 407-15, 2004 Dec 10.
Article in English | MEDLINE | ID: mdl-15530503

ABSTRACT

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.


Subject(s)
Interferon-alpha/pharmacology , Oocytes/drug effects , Receptors, Opioid/drug effects , Xenopus laevis , Animals , Cloning, Molecular , Dose-Response Relationship, Drug , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Oocytes/metabolism , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , RNA, Messenger/metabolism , Receptors, Opioid/genetics , Receptors, Opioid/metabolism , Receptors, Opioid, delta/drug effects , Receptors, Opioid, delta/genetics , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Recombinant Proteins
15.
Ann N Y Acad Sci ; 1025: 590-4, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15542767

ABSTRACT

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.


Subject(s)
Behavior, Addictive/metabolism , GTP-Binding Proteins/physiology , Potassium Channels, Inwardly Rectifying/metabolism , Substance-Related Disorders/metabolism , Animals , Behavior, Addictive/drug therapy , Female , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/therapeutic use , Substance-Related Disorders/drug therapy , Xenopus laevis
16.
Neuropsychopharmacology ; 29(10): 1841-51, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15150531

ABSTRACT

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.


Subject(s)
Antidepressive Agents/pharmacology , GTP-Binding Proteins/physiology , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/metabolism , Animals , Antidepressive Agents, Tricyclic/pharmacology , Electrophysiology , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Heart/drug effects , Humans , Myocardium/metabolism , Oocytes/metabolism , Potassium Channels, Inwardly Rectifying/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Xenopus laevis
17.
Br J Pharmacol ; 138(6): 1119-28, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12684268

ABSTRACT

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.


Subject(s)
Fluoxetine/pharmacology , Fluvoxamine/pharmacology , Potassium Channels, Inwardly Rectifying , Potassium Channels/drug effects , Zimeldine/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Ethanol/antagonists & inhibitors , Ethanol/pharmacology , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Heart/drug effects , Mice , Potassium Channels/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Xenopus laevis
18.
Jpn J Cancer Res ; 93(12): 1308-16, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12495470

ABSTRACT

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.


Subject(s)
Brain Neoplasms/genetics , Gene Rearrangement , Genes, Immunoglobulin , Immunoglobulin Heavy Chains/genetics , Lymphoma, B-Cell/genetics , Mutation , Aged , Base Sequence , Brain Neoplasms/immunology , Female , Humans , Immunoglobulin Variable Region/genetics , In Situ Hybridization , Lymphoma, B-Cell/immunology , Male , Middle Aged , Molecular Sequence Data , Polymerase Chain Reaction
19.
Brain Res Mol Brain Res ; 100(1-2): 13-20, 2002 Apr 30.
Article in English | MEDLINE | ID: mdl-12008017

ABSTRACT

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.


Subject(s)
Brain Chemistry/genetics , Brain/metabolism , Gene Expression Regulation/physiology , Isoenzymes/metabolism , Neurons/metabolism , Protein Kinase C/metabolism , RNA, Messenger/metabolism , Tyrosine 3-Monooxygenase/chemistry , Tyrosine 3-Monooxygenase/isolation & purification , 14-3-3 Proteins , Amino Acid Sequence/genetics , Animals , Base Sequence/genetics , Brain/cytology , Exons/genetics , Genes, Regulator/genetics , Immunohistochemistry , Mice , Molecular Sequence Data , Polyadenylation/genetics , Promoter Regions, Genetic/genetics , TATA Box/genetics , Tyrosine 3-Monooxygenase/genetics
20.
J Neurosurg ; 96(5): 909-17, 2002 May.
Article in English | MEDLINE | ID: mdl-12005399

ABSTRACT

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.


Subject(s)
Astrocytoma , Brain Neoplasms , I-kappa B Proteins , NF-kappa B/genetics , NF-kappa B/metabolism , Age Factors , Astrocytes/cytology , Astrocytes/metabolism , Brain/cytology , Cell Division/physiology , DNA/biosynthesis , DNA-Binding Proteins/genetics , Fetus/cytology , Humans , NF-KappaB Inhibitor alpha , NF-kappa B p50 Subunit , Oligodeoxyribonucleotides, Antisense/pharmacology , RNA, Messenger/analysis , Transcription Factor RelA , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...