Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Microbiol Biotechnol ; 23(12): 1791-801, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24100622

ABSTRACT

Diketopiperazine is produced by various organisms, including bacteria, fungi, and animals, and has been suggested as a novel signal molecule involved in the modulation of genes with various biological functions. Vibrio vulnificus, which causes septicemia in humans, produces cyclo(L-phenylalanine-L-proline) (cFP). To understand the biological roles of cFP, the effect of the compound on the expression of the total mRNA in V. vulnificus was assessed by nextgeneration sequencing. Based on the transcriptomic analysis, we classified the cFP-regulated genes into functional categories and clustered them according to the expression patterns resulted from treatment with cFP. From a total of 4,673 genes, excepting the genes encoding tRNA in V. vulnificus, 356 genes were up-regulated and 602 genes were down-regulated with an RPKM (reads per kilobase per million) value above 3. The genes most highly induced by cFP comprised those associated with the transport and metabolism of inorganic molecules, particularly iron. The genes negatively regulated by cFP included those associated with energy production and conversion, as well as carbohydrate metabolism. Noticeably, numerous genes related with biofilm formation were modulated by cFP. We demonstrated that cFP interferes significantly with the biofilm formation of V. vulnificus.


Subject(s)
Dipeptides/metabolism , Gene Expression Regulation, Bacterial/drug effects , Peptides, Cyclic/metabolism , Vibrio vulnificus/drug effects , Vibrio vulnificus/genetics , Gene Expression Profiling , High-Throughput Nucleotide Sequencing
2.
Comp Med ; 62(3): 203-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22776053

ABSTRACT

A 1-y-old male miniature pig housed in our laboratory facility was evaluated for weight loss and rough coat condition. CBC results revealed neutrophilia. Radiography of the thoracic area showed increased opacity throughout the thoracic cavity except for the right caudal lobe. ¹8F-labeled fluorodeoxyglucose positron emission tomography-computed tomography (FDG-PET-CT) revealed elevated standard uptake values in the area corresponding to the radiologic findings. Follow-up thoracic radiography taken 2 wk after FDG-PET-CT showed several interval changes, including markedly decreased opacity throughout the entire thoracic cavity. Necropsy revealed adhesions between the upper portion of the caudal lobe of the left lung and thoracic wall. ELISA for several closely related infectious species confirmed the presence of antibody to Actinobacillus pleuropneumoniae serovar V.


Subject(s)
Fluorodeoxyglucose F18 , Multimodal Imaging/methods , Pneumonia/diagnostic imaging , Positron-Emission Tomography , Tomography, X-Ray Computed , Animals , Swine , Swine, Miniature
3.
Oncol Rep ; 27(2): 499-503, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21971989

ABSTRACT

We cloned the ATP-binding cassette sub-family G member 2 (ABCG2) transporter, the most recently identified among several major human multidrug-resistance pumps, from A549 human lung adenocarcinoma cells in order to characterize its function and substrate specificity. In a previous report, we confirmed that a stem cell-like side population of A549 cells highly expressed the ABCG2 gene and had a unique ability to resist the anticancer drug methotrexate (MTX). In this study, ABCG2 cDNA was cloned by RT-PCR and converted into cRNA by an in vitro transcription system for expression in Xenopus laevis (X. laevis) oocytes. The transcribed cRNA of the ABCG2 gene was injected into the oocytes under the absence of cofactors or heterologous partner proteins or some lipids from the media. A high expression of ABCG2 was observed on the oocyte surface by immunofluorescence and confocal laser microscopy. We tested the functional effect of ABCG2 expression on drug efflux by directly injecting MTX into X. laevis oocytes. The drug concentration within the oocytes was quantified with LC-MS/MS; the analysis showed that the accumulation of MTX was significantly decreased in the X. laevis oocytes expressing ABCG2 compared with the control oocytes not expressing ABCG2. These findings show that the ABCG2 protein has an important role in the efflux of MTX through the cell membrane of X. laevis oocytes. Therefore, it might be that ABCG2, abundantly expressed in the stem cell population of A549 cells, can modulate resistance to MTX in lung cancer therapy.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adenocarcinoma/metabolism , Lung Neoplasms/metabolism , Neoplasm Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Amino Acid Sequence , Animals , Antimetabolites, Antineoplastic/metabolism , Base Sequence , Cell Line, Tumor , Cell Membrane/metabolism , DNA, Complementary/genetics , Female , Humans , Lung Neoplasms/genetics , Methotrexate/metabolism , Molecular Sequence Data , Neoplasm Proteins/genetics , Plasmids/genetics , Protein Transport , Sequence Alignment , Transcription, Genetic , Xenopus laevis
4.
Lab Anim Res ; 27(2): 109-16, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21826170

ABSTRACT

To clone the first anion channel from Xenopus laevis (X. laevis), we isolated a calcium-activated chloride channel (CLCA)-like membrane protein 6 gene (CMP6) in X. laevis. As a first step in gene isolation, an expressed sequence tags database was screened to find the partial cDNA fragment. A putative partial cDNA sequence was obtained by comparison with rat CLCAs identified in our laboratory. First stranded cDNA was synthesized by reverse transcription polymerase-chain reaction (RT-PCR) using a specific primer designed for the target cDNA. Repeating the 5' and 3' rapid amplification of cDNA ends, full-length cDNA was constructed from the cDNA pool. The full-length CMP6 cDNA completed via 5'- and 3'-RACE was 2,940 bp long and had an open reading frame (ORF) of 940 amino acids. The predicted 940 polypeptides have four major transmembrane domains and showed about 50% identity with that of rat brain CLCAs in our previously published data. Semi-quantification analysis revealed that CMP6 was most abundantly expressed in small intestine, colon and liver. However, all tissues except small intestine, colon and liver had undetectable levels. This result became more credible after we did real-time PCR quantification for the target gene. In view of all CLCA studies focused on human or murine channels, this finding suggests a hypothetical protein as an ion channel, an X. laevis CLCA.

5.
Biochem Biophys Res Commun ; 408(4): 559-65, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21521635

ABSTRACT

We have successfully isolated a novel anoctamin (xANO2), Ca(2+)-activated chloride channel (ANO1, TMEM16A), from Xenopus laevis. The cDNA sequence was determined to belong to the anoctamin family by comparison with the xTMEM16A sequence in a previous report. Full length cDNA synthesis was performed by repeating 5'- and 3'-rapid amplification of cDNA end (RACE). We successfully completed the entire cDNA sequence and transiently named this sequence xANO2. The xANO2 cDNA is 3884 base pair (bp) long and codes 980 amino acid (aa) proteins. According to an aa homology search using the Basic Local Alignment Search Tool (BLAST), xANO2 showed an overall identity of 92% to xTMEM16A (xANO1) independently sub-cloned in our laboratory. A primary sequence of xANO2 revealed typical characteristics of transmembrane proteins. In tissue distribution analysis, the gene products of anoctamins were ubiquitously detected by real-time PCR (RT-PCR). The expression profiles of each anoctamin were different among brain, oocytes, and digestive organs with relatively weak expression. To clarify the anoctamin activity, physiological studies were performed using the whole cell patch-clamp technique with HEK293T cells, enhanced green fluorescent protein (EGFP), and expression vectors carrying anoctamins. Characteristics typical of voltage-dependent chloride currents were detected in cells expressing both xANO2 and xTMEM16A but not with EGFP alone. Sensitive reactions to the anion channel blocker niflumic acid (NFA) were also revealed. Considering these results, xANO2 was regarded as a new TMEM16A belonging to the Xenopus anoctamin family.


Subject(s)
Chloride Channels/genetics , Xenopus laevis/genetics , Amino Acid Sequence , Animals , Chloride Channels/chemistry , Cloning, Molecular , Gene Expression , Molecular Sequence Data , Protein Conformation , Xenopus Proteins/chemistry , Xenopus Proteins/genetics
6.
J Biotechnol ; 131(3): 346-52, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17767971

ABSTRACT

In this study, the clostripain gene was modified and its signal sequence was replaced with that of penicillin G acylase (PGA). The core clostripain protein fused to the PGA signal peptide was also prepared. With regard to the expression of the clostripain precursors, the majority of clostripain activity was observed in the culture media, thereby indicating that both the clostripain signal peptide and the PGA signal peptide were recognized in the E. coli secretion pathway, and the precursors successfully matured into the active form. Otherwise, the activity was rather low when the core protein was expressed, which indicates that the clostripain pro-peptide is important in the formation of the active enzyme in E. coli. Enzyme activity reached a value of 3200U/L in CGY media for high expression. The recombinant clostripain and porcine carboxypeptidase B were used in the conversion of a proinsulin fusion protein into insulin. The leader peptide (LP) and the proinsulin C-peptide appeared to have been removed simultaneously, and the final cleavage product evidenced an HPLC retention time identical to that of the insulin standard, thereby implying that the clostripain specifically cleaved the arginine residues in the LP and in the C-peptide. We have also demonstrated the possibility that the recombinant clostripain might prove useful in the production of insulin from the proinsulin fusion protein.


Subject(s)
Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Escherichia coli/metabolism , Insulin/chemical synthesis , Proinsulin/chemistry , Protein Engineering/methods , Cloning, Molecular/methods , Cysteine Endopeptidases/genetics , Escherichia coli/genetics , Recombinant Fusion Proteins/chemistry
7.
Brain Res ; 1161: 1-10, 2007 Aug 03.
Article in English | MEDLINE | ID: mdl-17603028

ABSTRACT

Quercetin, one of the flavonoids, is a compound of low molecular weight found in fruits and vegetables. Besides its antioxidative effect, quercetin also shows a wide range of diverse neuropharmacological actions. However, the cellular mechanisms of quercetin's actions, especially on ligand-gated ion channels and synaptic transmissions, are not well studied. We investigated the effect of quercetin on the human glycine alpha1 receptor channel expressed in Xenopus oocytes using a two-electrode voltage clamp technique. Application of quercetin reversibly inhibited glycine-induced current (I(Gly)). Quercetin's inhibition depends on its dose, with an IC(50) of 21.5+/-.2 microM. The inhibition was sensitive to membrane voltages. Site-directed mutations of S267 to S267Y but not S267A, S267F, S267G, S267K, S267L and S267T at transmembrane domain 2 (TM2) nearly abolished quercetin-induced inhibition of I(Gly). In contrast, in site-directed mutant receptors such as S267 to S267I, S267R and S267V, quercetin enhanced I(Gly) compared to the wild-type receptor. The EC(50) was 22.6+/-1.4, 25.5+/-4.2, and 14.5+/-3.1 microM for S267I, S267R and S267V, respectively. These results indicate that quercetin might regulate the human glycine alpha(1) receptor via interaction with amino acid residue alpha267 and that alpha267 plays a key role in determining the regulatory consequences of the human glycine alpha1 receptor by quercetin.


Subject(s)
Antioxidants/pharmacology , Ion Channel Gating/drug effects , Mutation/physiology , Quercetin/pharmacology , Receptors, Glycine/physiology , Analysis of Variance , Animals , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Female , Glycine/pharmacology , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Microinjections/methods , Mutagenesis, Site-Directed/methods , Oocytes , Patch-Clamp Techniques , Receptors, Glycine/chemistry , Structure-Activity Relationship , Xenopus laevis
8.
Eur J Pharmacol ; 564(1-3): 7-17, 2007 Jun 14.
Article in English | MEDLINE | ID: mdl-17346698

ABSTRACT

We previously demonstrated that dextromethorphan (DM; 3-methoxy-17-methylmorphinan) analogs have neuroprotective effects. Here, we investigated the effects of DM, three of its analogs (DF, 3-methyl-17-methylmorphinan; AM, 3-allyloxy-17-methoxymorphian; and CM, 3-cyclopropyl-17-methoxymorphinan) and one of its metabolites (HM; 3-methoxymorphinan), on Na(+) channel activity. We used the two-microelectrode voltage-clamp technique to test the effects of DM, DF, AM, CM and HM on Na(+) currents (I(Na)) in Xenopus oocytes expressing cRNAs encoding rat brain Nav1.2 alpha and beta1 or beta2 subunits. In oocytes expressing Na(+) channels, DM, DF, AM and CM, but not HM, induced tonic and use-dependent inhibitions of peak I(Na) following low- and high-frequency stimulations. The order of potency for the inhibition of peak I(Na) was AM-CM > DM=DF. The DM, DF, AM and CM-induced tonic inhibitions of peak I(Na) were voltage-dependent, dose-dependent and reversible. The IC(50) values for DM, DF, AM and CM were 116.7+/-14.9, 175.8+/-16.9, 38.6+/-15.5, and 42.5+/-8.5 microM, respectively. DM and its analogs did not affect the steady-state activation and inactivation voltages. AM and CM, but not DM and DF, inhibited the plateau I(Na) more effectively than the peak I(Na) in oocytes expressing inactivation-deficient I1485Q-F1486Q-M1487Q (IFMQ3) mutant channels; the IC(50) values for AM and CM in this system were 8.4+/-1.3 and 8.7+/-1.3 microM, respectively, for the plateau I(Na) and 43.7+/-5.9 and 32.6+/-7.8 microM, respectively, for the peak I(Na). These results collectively indicate that DM and its analogs could be novel Na(+) channel blockers acting on the resting and open states of brain Na(+) channels.


Subject(s)
Brain/drug effects , Dextromethorphan/pharmacology , Neuroprotective Agents/pharmacology , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Animals , Brain/metabolism , Dextromethorphan/administration & dosage , Dextromethorphan/analogs & derivatives , Dose-Response Relationship, Drug , Electric Stimulation , Gene Expression , Membrane Potentials , Microelectrodes , Neuroprotective Agents/administration & dosage , Oocytes/drug effects , Protein Subunits , RNA, Complementary/metabolism , Rats , Sodium Channel Blockers/administration & dosage , Sodium Channels/metabolism , Structure-Activity Relationship , Xenopus laevis
9.
Neuropharmacology ; 52(4): 1139-50, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17257631

ABSTRACT

We previously demonstrated that 20(S)-ginsenoside Rg(3) (Rg(3)), one of the active components of Panax ginseng, non-competitively inhibits 5-HT(3A) receptor channel activity on extracellular side of the cell. Here, we sought to elucidate the molecular mechanisms underlying Rg(3)-induced 5-HT(3A) receptor regulation. We used the two-microelectrode voltage-clamp technique to investigate the effect of Rg(3) on 5-HT-mediated ion currents (I(5-HT)) in Xenopus oocytes expressing wild-type or 5-HT(3A) receptors harboring mutations in the gating pore region of transmembrane domain 2 (TM2). In oocytes expressing wild-type 5-HT(3A) receptors, Rg(3) dose-dependently inhibited peak I(5-HT) with an IC(50) of 27.6+/-4.3microM. Mutations V291A, F292A, and I295A in TM2 greatly attenuated or abolished the Rg(3)-induced inhibition of peak I(5-HT). Mutation V291A but not F292A and I295A induced constitutively active ion currents with decrease of current decay rate. Rg(3) accelerated the rate of current decay with dose-dependent manner in the presence of 5-HT. Rg(3) and TMB-8, an open channel blocker, dose-dependently inhibited constitutively active ion currents. The IC(50) values of constitutively active ion currents in V291A mutant receptor were 72.4+/-23.1 and 6.5+/-0.7microM for Rg(3) and TMB-8, respectively. Diltiazem did not prevent Rg(3)-induced inhibition of constitutively active ion currents in occlusion experiments. These results indicate that Rg(3) inhibits 5-HT(3A) receptor channel activity through interactions with residues V291, F292, and I295 in the channel gating region of TM2 and further demonstrate that Rg(3) regulates 5-HT(3A) receptor channel activity in the open state at different site(s) from those of TMB-8 and diltiazem.


Subject(s)
Ginsenosides/pharmacology , Receptors, Serotonin, 5-HT3/drug effects , Animals , Calcium Channel Blockers/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Gallic Acid/analogs & derivatives , Gallic Acid/pharmacology , Ginsenosides/chemistry , Membrane Potentials/drug effects , Membrane Potentials/genetics , Membrane Potentials/radiation effects , Models, Molecular , Mutation/physiology , Oocytes , Patch-Clamp Techniques/methods , Receptors, Serotonin, 5-HT3/chemistry , Receptors, Serotonin, 5-HT3/metabolism , Serotonin/pharmacology , Xenopus laevis
10.
Brain Res ; 1136(1): 190-9, 2007 Mar 09.
Article in English | MEDLINE | ID: mdl-17239831

ABSTRACT

We previously demonstrated that ginsenoside Rg(3) (Rg(3)), one of the active ingredients in Panax ginseng, attenuates NMDA receptor-mediated currents and NMDA-induced neurotoxicity (Kim, S., Kim, T., Ahn, K., Park, W.K., Nah, S.Y., Rhim, H., 2004. Ginsenoside Rg(3) antagonizes NMDA receptors through a glycine modulatory site in rat cultured hippocampal neurons. Biochem. Biophys. Res. Commun. 323, 416-424). Accumulating evidence suggests that homocysteine (HC), a metabolite of methionine, exerts its excitotoxicity through NMDA receptor activation. In the present study, we examined the neuroprotective effects of Rg(3) on HC-induced hippocampal excitotoxicity in vitro and in vivo. Our in vitro studies using rat cultured hippocampal neurons revealed that Rg(3) treatment significantly and dose-dependently inhibited HC-induced hippocampal cell death, with an EC(50) value of 28.7+/-7.5 muM. Rg(3) treatment not only significantly reduced HC-induced DNA damage, but also dose-dependently attenuated HC-induced caspase-3 activity in vitro. Our in vivo studies revealed that intracerebroventricular (i.c.v.) pre-administration of Rg(3) significantly and dose-dependently reduced i.c.v. HC-induced hippocampal damage in rats. To examine the mechanisms underlying the in vitro and in vivo neuroprotective effects of Rg(3) against HC-induced hippocampal excitotoxicity, we examined the effect of Rg(3) on HC-induced intracellular Ca(2+) elevations in cultured hippocampal cells and found that Rg(3) treatment dose-dependently inhibited HC-induced intracellular Ca(2+) elevation, with an IC(50) value of 41.5+/-17.5 muM. In addition, Rg(3) treatment dose-dependently inhibited HC-induced currents in Xenopus oocytes expressing the NMDA receptor, with an IC(50) of 47.3+/-14.2 muM. These results collectively indicate that Rg(3)-induced neuroprotection against HC in rat hippocampus might be achieved via inhibition of HC-mediated NMDA receptor activation.


Subject(s)
Ginsenosides/pharmacology , Hippocampus/cytology , Homocysteine/toxicity , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neurotoxins/toxicity , Animals , Calcium/metabolism , Caspase 3/metabolism , Cell Death/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Embryo, Mammalian , Embryo, Nonmammalian , In Situ Nick-End Labeling , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Oocytes , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Xenopus laevis
11.
Biol Pharm Bull ; 29(12): 2425-31, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17142976

ABSTRACT

Recent studies have shown that Panax ginseng has a variety of beneficial effects on the cardiovascular systems. Homocysteine (Hcy), which is derived from L-methionine (Met), has been closely associated with the increased risk of cardiovascular diseases. In the present study, we examined whether in vivo long-term administration of ginseng saponins (GS), active ingredients of Panax ginseng, attenuate adverse vascular effects associated with chronic Met-induced hyperhomocysteinemia (H-Hcy). We found that plasma Hcy level, which was measured after 30 and 60 d, in GS (100 mg/kg)+Met co-administration group was significantly reduced when it was compared with Met alone treatment group. We could also observe the alleviation of endothelial damages of aortic artery vessels in GS (100 mg/kg)+Met co-administration group compared with Met alone treatment group. We compared aortic vasocontractile and vasodilatory responses between Met alone and GS (100 mg/kg)+Met co-treatment groups. We found that norepinephrine-induced vasocontractile responses were greatly decreased in GS (100 mg/kg)+Met co-treatment group and that carbachol-induced dilatory responses were greatly enhanced in GS (100 mg/kg)+Met co-administration groups as compared with Met alone treatment group. The present results indicate that in vivo long-term administration of GS attenuates adverse vascular effects associated with chronic Met-induced H-Hcy in rats.


Subject(s)
Hyperhomocysteinemia/drug therapy , Methionine/adverse effects , Panax/chemistry , Saponins/therapeutic use , Animals , Blood Pressure , Body Weight , Heart Rate , Hyperhomocysteinemia/chemically induced , Hyperhomocysteinemia/physiopathology , Male , Rats , Rats, Wistar
12.
Biol Pharm Bull ; 29(11): 2168-73, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17077509

ABSTRACT

We previously reported the cloning of a calcium-activated chloride channel (CLCA) from rat brain (Biochem. Biophys. Res. Commun., 334, 569-576 (2005)), which we designated rbCLCA1. We further showed that rbCLCA1 is expressed in the central nervous system and peripheral organs, and may be functionally expressed in mammalian HEK293 cells. In the present study, we report the successful cloning of a second CLCA from rat cerebrum (designated rbCLCA2), using reverse transcription-PCR (RT-PCR) with primers specific for rbCLCA1. We have begun to clone this cDNA based on the rbCLCA1-like sequence. The full-length rbCLCA2 cDNA, obtained via 5' and 3' rapid amplification of cDNA ends (RACE), is 2900 bp long and encodes a putative polypeptide of 905 amino acids having at least two major transmembrane domains and showing 85.2% identity to rbCLCA1. RT-PCR analysis revealed that, similar to rbCLCA1, rbCLCA2 was predominantly expressed in the rat cerebrum, cerebellum, kidney, stomach, spinal cord, lung and small intestine, but not in the heart, large intestine, liver, orand spleen. Whole-cell patch clamp studies in HEK293 cells transiently co-transfected with expression vectors encoding rbCLCA2 and EGFP allowed us to identify the presence of niflumic acid (a CLCA channel blocker)-sensitive and voltage-dependent chloride currents in cells expressing rbCLCA2 but not EGFP alone. Treatment of these cells with ionomycin, a Ca2+ ionophore, significantly increased the novel currents in cells expressing rbCLCA2 and EGFP, but not those expressing EGFP alone, indicating that activation of the rbCLCA2 current is Ca2+-dependent. In sum, we herein report the cloning of a second member of the rbCLCA family from rat brain and its functional expression in vitro, thus adding to our knowledge of anion channels and facilitating future exploration of brain and other organ physiology.


Subject(s)
Brain/metabolism , Chloride Channels/genetics , Gene Expression/genetics , Amino Acid Sequence , Animals , Calcium Chloride/pharmacology , Cell Line , Chloride Channels/metabolism , Chloride Channels/physiology , Cloning, Molecular , DNA, Complementary/chemistry , DNA, Complementary/genetics , Egtazic Acid/pharmacology , Gene Expression/drug effects , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Kidney/cytology , Kidney/metabolism , Membrane Potentials/drug effects , Molecular Sequence Data , Niflumic Acid/pharmacology , Patch-Clamp Techniques , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Transfection/methods
13.
J Neurochem ; 99(3): 976-88, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16942594

ABSTRACT

Methamphetamine (MAP) is one of the most commonly abused drugs in Asia, and previous studies suggest that serotonin 3 receptors (5-HT(3)) are involved in MAP-induced locomotion and reward. However, little is known about the role of 5-HT(3) receptors in MAP-induced behavioral sensitization. Here, we measured the effects of MDL 72222, a 5-HT(3) antagonist, and SR 57227 A, a 5-HT(3) agonist, on the development and expression of MAP-induced behavioral sensitization, and alternations of 5-HT(3) receptor binding labeled with the 5-HT(3)-selective antagonist, [(3)H]GR65630, in mice. In addition, we investigated the effects of MAP on 5-HT(3A) receptor channel activity in Xenopus laevis oocytes expressing 5-HT(3A) receptors. We found that MDL 72222 attenuated both the development and expression of behavioral sensitization to MAP (1.0 mg/kg, i.p.), and that this attenuating effect of MDL 72222 was reversed by pre-treatment with SR 57227 A. In oocytes expressing 5-HT(3A) receptor, MAP exhibited a dual modulation of 5-HT(3A) receptor channel activity, i.e. pre-treatment with a low dose of MAP (0.1 microm) enhanced 5-HT-induced inward peak current (I(5-HT)) but a high dose of MAP (100 microm) inhibited I(5-HT). The acute administration of MDL 72222 with MAP decreased [(3)H]GR65630 binding versus MAP alone in the mouse striatum. Our results suggest that MDL 72222 attenuates MAP-induced behavioral sensitization via 5-HT(3) receptors in the caudate putamen, and that 5-HT(3) receptor antagonists like MDL 72222 have potential as novel anti-psychotic agents for the treatment of MAP dependence and psychosis.


Subject(s)
Behavior, Animal/drug effects , Dopamine Uptake Inhibitors/pharmacology , Methamphetamine/pharmacology , Receptors, Serotonin, 5-HT3/metabolism , Algorithms , Animals , Autoradiography , Dopamine Uptake Inhibitors/antagonists & inhibitors , Dopamine Uptake Inhibitors/metabolism , Imidazoles/pharmacology , Indoles/pharmacology , Male , Methamphetamine/antagonists & inhibitors , Methamphetamine/metabolism , Mice , Mice, Inbred ICR , Microinjections , Motor Activity/drug effects , Oocytes/metabolism , Piperidines/pharmacology , RNA, Complementary/biosynthesis , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Tropanes/pharmacology , Xenopus laevis
14.
Eur J Pharmacol ; 536(1-2): 85-92, 2006 Apr 24.
Article in English | MEDLINE | ID: mdl-16563374

ABSTRACT

We previously demonstrated that dextromethorphan (DM; 3-methoxy-17-methylmorphinan) analogs have neuroprotective effects, and a recent report showed that DM reduces the adverse effects of morphine and blocks alpha3beta4 nicotinic acetylcholine receptors, a major target of anti-addictive agents. Here, we investigated the effects of DM, three of its analogs (DF, 3-methyl-17-methylmorphinan; AM, 3-allyloxy-17-methoxymorphian; and CM, 3-cyclopropyl-17-methoxymorphinan) and one of its metabolites (HM; 3-methoxymorphinan), on neuronal alpha3beta4 nicotinic acetylcholine receptor channel activity expressed in Xenopus laevis oocytes, using the two-microelectrode voltage clamp technique. We found that intraoocyte injection of neuronal alpha3 and beta4 nicotinic acetylcholine receptor subunit cRNAs elicited an inward current (IACh) in the presence of acetylcholine. Co-treatment with DM, DF, AM, CM or HM inhibited IACh in a dose-dependent, voltage-independent and reversible manner. The IC50 values for DM, DF, AM, CM and HM were 19.5+/-5.2, 15.8+/-4.5, 16.3+/-1.7, 10.1+/-2.8, and 13.5+/-4.0 microM, respectively. The order of potency for the inhibition of IACh was CM>HM>DF=AM>DM in oocytes expressing alpha3beta4 nicotinic acetylcholine receptors. The inhibitions of (IACh) by DM, DF and HM, AM and CM were non-competitive. These results indicate that AM, CM and HM could be novel non-competitive agents regulating alpha3beta4 nicotinic acetylcholine receptor channel activity.


Subject(s)
Dextromethorphan/pharmacology , Oocytes/drug effects , Receptors, Nicotinic/physiology , Animals , Cattle , Dextromethorphan/chemistry , Dose-Response Relationship, Drug , Electric Stimulation , Female , Gene Expression , Membrane Potentials/drug effects , Molecular Structure , Oocytes/metabolism , Oocytes/physiology , RNA, Complementary/administration & dosage , RNA, Complementary/genetics , Receptors, Nicotinic/genetics , Structure-Activity Relationship , Time Factors , Xenopus laevis
15.
Mol Cells ; 21(1): 52-62, 2006 Feb 28.
Article in English | MEDLINE | ID: mdl-16511347

ABSTRACT

In previous reports we demonstrated that ginsenosides, active ingredients of Panax ginseng, affect some subsets of voltage-dependent Ca(2+) channels in neuronal cells expressed in Xenopus laevis oocytes. However, the major component(s) of ginseng that affect cloned Ca(2+) channel subtypes such as alpha(1C) (L)-, alpha(1B) (N)-, alpha(1A) (P/Q)-, a1E (R)- and a1G (T) have not been identified. Here, we used the two-microelectrode volt-age clamp technique to characterize the effects of ginsenosides and ginsenoside metabolites on Ba(2+) currents (IBa) in Xenopus oocytes expressing five different Ca(2+) channel subtypes. Exposure to ginseng total saponins (GTS) induced voltage-dependent, dose-dependent and reversible inhibition of the five channel subtypes, with particularly strong inhibition of the a1G-type. Of the various ginsenosides, Rb(1), Rc, Re, Rf, Rg(1), Rg(3), and Rh(2), ginsenoside Rg(3) also inhibited all five channel subtypes and ginsenoside Rh(2) had most effect on the a1C- and a1E-type Ca(2+) channels. Compound K (CK), a protopanaxadiol ginsenoside metabolite, strongly inhibited only the a(1G)-type of Ca(2+) channel, whereas M4, a protopanaxatriol ginsenoside metabolite, had almost no effect on any of the channels. Rg(3), Rh(2), and CK shifted the steady-state activation curves but not the inactivation curves in the depolarizing direction in the alpha(1B)- and alpha(1A)-types. These results reveal that Rg(3), Rh(2) and CK are the major inhibitors of Ca(2+) channels in Panax ginseng, and that they show some Ca(2+) channel selectivity.


Subject(s)
Calcium Channels/classification , Calcium Channels/metabolism , Ginsenosides/metabolism , Ginsenosides/pharmacology , Animals , Calcium Channels/genetics , Dose-Response Relationship, Drug , Ginsenosides/chemistry , Ion Channel Gating/drug effects , Oocytes/drug effects , Oocytes/metabolism , Xenopus
16.
Biol Pharm Bull ; 29(2): 365-70, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16462047

ABSTRACT

Previous reports have shown that ginseng saponins, the active ingredients of Panax ginseng, induce relaxation of hormone- or high K(+)-induced blood vessel contraction. We recently demonstrated that 20(R)- and 20(S)-ginsenoside Rg(3) epimers regulate ion channel activities in a stereospecific manner. Here, we examined whether ginsenoside Rg(3) epimers also exhibit differential effects on swine coronary artery contractions induced by high K(+) or 5-HT. We found that treatment with 20(S)- but not 20(R)-ginsenoside Rg(3) caused a potent concentration-dependent, endothelium-independent relaxation of coronary artery contraction induced by 25 mM KCl. However, treatment with both 20(S)- and 20(R)-ginsenoside Rg(3) induced a significant, concentration-dependent relaxation of 3 microM 5-HT-induced coronary artery contractions in intact samples, while only 20(S)-ginsenoside Rg(3) inhibited coronary artery contraction in endothelium-denuded arteries. 20(S)- but not 20(R)-ginsenoside Rg(3) inhibited L-type Ca(2+) channel currents in a dose- and voltage-dependent manner. These results indicate that 20(S)- and 20(R)-ginsenoside Rg(3) epimers might exhibit stereospecific relaxation effects on swine coronary artery contractions caused by high K(+) and 5-HT receptor activation.


Subject(s)
Coronary Vessels/drug effects , Ginsenosides/pharmacology , Vasoconstriction/drug effects , Vasodilator Agents/pharmacology , Animals , Calcium Channels, L-Type/metabolism , Coronary Vessels/metabolism , Dose-Response Relationship, Drug , Ginsenosides/chemistry , In Vitro Techniques , Molecular Structure , Potassium Chloride/pharmacology , Receptors, Serotonin/metabolism , Stereoisomerism , Swine , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/chemistry
17.
Biol Pharm Bull ; 28(11): 2120-4, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16272701

ABSTRACT

The 5-hydroxytryptamine3A (5-HT3) receptor is closely related with irritable bowel syndrome (IBS) in enteric nervous systems. We previously demonstrated that ginseng total saponins (GTS, also called ginsenosides), the active ingredients of Panax ginseng, inhibit the activity of 5-HT3A receptor channels expressed in Xenopus laevis oocytes. Here, we further investigated whether the in vitro inhibitory effect of ginsenosides on 5-HT3A receptor channel activity is coupled to in vivo attenuation of IBS. A rat model of IBS was induced by colorectal distention (CRD) and intracolonic infusion of 0.6% acetic acid (CRD-acetic acid), and visceral hypersensitivity was assessed by counting the contractions in the external oblique muscles of conscious rats during the 10 min distention period. We found that oral administration of GTS significantly and dose-dependently inhibited CRD-acetic acid-induced visceral hypersensitivity. The EC50 was 5.5+/-4.7 mg/kg (95% confidence intervals: 1.2-15.7) and the inhibitory effect of GTS against visceral hypersensitivity persisted for 4 h. When we compared the effects of protopanaxadiol (PD) ginsenosides and protopanaxatriol (PT) ginsenosides against CRD-acetic acid-induced visceral hypersensitivity, we found that PT but not PD ginsenosides significantly attenuated the CRD-acetic acid-induced visceral hypersensitivity. These results indicate that PT ginsenosides of Panax ginseng might be the main active components for the attenuation of experimentally CRD-acetic acid-induced visceral hypersensitivity, and may be clinically relevant for the future treatment of IBS.


Subject(s)
Irritable Bowel Syndrome/drug therapy , Panax/chemistry , Saponins/therapeutic use , Animals , Benzamides/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Catheterization , Colon/physiology , Dose-Response Relationship, Drug , Male , Peripheral Nervous System/drug effects , Peripheral Nervous System/physiology , Rats , Rats, Sprague-Dawley , Receptors, Serotonin, 5-HT3/drug effects , Receptors, Serotonin, 5-HT3/metabolism , Rectum/physiology , Serotonin Antagonists/pharmacology
18.
Mol Cells ; 20(1): 69-73, 2005 Aug 31.
Article in English | MEDLINE | ID: mdl-16258243

ABSTRACT

The flavonoid, quercetin, is a low molecular weight substance found in apple, tomato and other fruit. Besides its antioxidative effect, quercetin, like other flavonoids, has a wide range of neuropharmacological actions including analgesia, and motility, sleep, anticonvulsant, sedative and anxiolytic effects. In the present study, we investigated its effect on mouse 5-hydroxytryptamine type 3 (5-HT3A) receptor channel activity, which is involved in pain transmission, analgesia, vomiting, and mood disorders. The 5-HT3A receptor was expressed in Xenopus oocytes, and the current was measured with the two-electrode voltage clamp technique. In oocytes injected with 5-HT3A receptor cRNA, quercetin inhibited the 5-HT-induced inward peak current (I(5-HT)) with an IC50 of 64.7 +/- 2.2 microM. Inhibition was competitive and voltage-independent. Point mutations of pre-transmembrane domain 1 (pre-TM1) such as R222T and R222A, but not R222D, R222E and R222K, abolished inhibition, indicating that quercetin interacts with the pre-TM1 of the 5-HT3A receptor.


Subject(s)
Membrane Potentials/drug effects , Protein Structure, Tertiary/drug effects , Quercetin/pharmacology , Serotonin 5-HT3 Receptor Antagonists , Animals , Dose-Response Relationship, Drug , Female , Ion Channel Gating/drug effects , Ion Transport/drug effects , Mice , Mutagenesis, Site-Directed , Oocytes/metabolism , Oocytes/physiology , Protein Structure, Tertiary/physiology , Quercetin/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Xenopus laevis
19.
Arch Pharm Res ; 28(6): 680-4, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16042077

ABSTRACT

Ginseng, the root of Panax ginseng C.A. Meyer, is well known as a tonic medicine for restoring and enhancing human health. In traditional medicine, ginseng is utilized for the alleviation of emesis, which includes nausea and vomiting. However, it has not yet been demonstrated whether ginseng exhibits in vivo anti-nausea and anti-vomiting properties. In this study, we examined the anti-emetic effect of Korean red ginseng total extract (KRGE) on cisplatin-induced nausea and vomiting using ferrets. Intraperitoneal administration (i.p.) of cisplatin (7.5 mg/kg) induced both nausea and vomiting with one-hour latency. The episodes of nausea and vomiting reached a peak after 1.5 h and persisted for 3 h. Treatment with KRGE via oral route significantly reduced the cisplatin-induced nausea and vomiting in a dose-dependent manner. The anti-emetic effect was 12.7 +/- 8.6, 31.8 +/- 6.9, and 67.6 +/- 4.0% with doses of 0.3, 1.0, and 3.0 g/kg of KRGE, respectively. Pretreatment with KRGE via oral route 1 and 2 h before cisplatin administration also significantly attenuated the cisplatin-induced nausea and vomiting. However this did not occur with a pretreatment 4 h before cisplatin administration. These results are supportive of KRGE being utilized as an anti-emetic agent against nausea and vomiting caused by chemotherapy (i.e. cisplatin).


Subject(s)
Antiemetics/pharmacology , Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Nausea/prevention & control , Panax , Plant Extracts/pharmacology , Vomiting/prevention & control , Animals , Dose-Response Relationship, Drug , Ferrets , Male , Nausea/chemically induced , Orchiectomy , Phytotherapy , Plant Extracts/administration & dosage , Plant Extracts/therapeutic use , Plant Roots , Premedication , Time Factors , Vomiting/chemically induced
20.
Mol Pharmacol ; 68(4): 1114-26, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16014805

ABSTRACT

We demonstrated previously that ginsenoside Rg(3) (Rg(3)), an active ingredient of Panax ginseng, inhibits brain-type Na(+) channel activity. In this study, we sought to elucidate the molecular mechanisms underlying Rg(3)-induced Na(+) channel inhibition. We used the two-microelectrode voltage-clamp technique to investigate the effect of Rg(3) on Na(+) currents (I(Na)) in Xenopus laevis oocytes expressing wild-type rat brain Na(V)1.2 alpha and beta1 subunits, or mutants in the channel entrance, the pore region, the lidocaine/tetrodotoxin (TTX) binding sites, the S4 voltage sensor segments of domains I to IV, and the Ile-Phe-Met inactivation cluster. In oocytes expressing wild-type Na(+) channels, Rg(3) induced tonic and use-dependent inhibitions of peak I(Na). The Rg(3)-induced tonic inhibition of I(Na) was voltage-dependent, dose-dependent, and reversible, with an IC(50) value of 32 +/- 6 microM. Rg(3) treatment produced a 11.2 +/- 3.5 mV depolarizing shift in the activation voltage but did not alter the steady-state inactivation voltage. Mutations in the channel entrance, pore region, lidocaine/TTX binding sites, or voltage sensor segments did not affect Rg(3)-induced tonic blockade of peak I(Na). However, Rg(3) treatment inhibited the peak and plateau I(Na) in the IFMQ3 mutant, indicating that Rg(3) inhibits both the resting and open states of Na(+) channel. Neutralization of the positive charge at position 859 of voltage sensor segment domain II abolished the Rg(3)-induced activation voltage shift and use-dependent inhibition. These results reveal that Rg(3) is a novel Na(+) channel inhibitor capable of acting on the resting and open states of Na(+) channel via interactions with the S4 voltage-sensor segment of domain II.


Subject(s)
Brain/drug effects , Ginsenosides/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Animals , Brain/metabolism , DNA, Complementary , Lidocaine/pharmacology , Mutagenesis, Site-Directed , NAV1.2 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/genetics , Rats , Sodium Channels/genetics , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...