Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Ginseng Res ; 41(1): 52-59, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28123322

ABSTRACT

BACKGROUND: Korean Red Ginseng extract (KRG, Panax ginseng Meyer) and its constituents have been used for treating diabetes. However, in diet-induced obese mice, it is unclear whether KRG can enhance the glucose-lowering action of rosiglitazone (ROSI), a peroxisome proliferator-activated receptor gamma synthetic activator. METHODS: Oral glucose tolerance tests (oGTTs) were performed after 4 days of treatment with a vehicle (CON), KRG [500 mg/kg body weight (b.w.)], ROSI (3.75 mg/kg b.w, 7.5 mg/kg b.w, and 15 mg/kg b.w.), or ROSI and KRG (RK) in obese mice on a high-fat diet. Adipose tissue morphology, crown-like structures (CLSs), and inflammation were compared by hematoxylin-eosin staining or quantitative reverse transcription polymerase chain reaction. RESULTS: The area under the glucose curve (AUC) was significantly lower in the RK group (15 mg/kg b.w. and 500 mg/kg b.w. for ROSI and KRG, respectively) than in the CON group. There was no significant difference in the AUC between the CON and the other groups. Furthermore, the AUC was significantly lower in the RK group than in the ROSI group. The expression of the Ccl2 gene and the number of CLSs were significantly reduced in the RK group than in the CON group. CONCLUSION: Our results show a potential enhancement of ROSI-induced improvement of glucose regulation by the combined treatment with KRG.

2.
Korean J Physiol Pharmacol ; 17(5): 405-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24227940

ABSTRACT

We studied the sex different nicotine effect on evoked population spike amplitudes (ePSA) and connexin (Cx) expression in the hippocampus CA1 area of gerbils. Acute doses of nicotine bitartrate (0.5 mg/kg: NT-0.5) slightly reduced ePSA in males but markedly augmented that in females. Acute NT (5.0 mg/kg) markedly increased the ePSA in all gerbils. Unlike acute NT-0.5, repeated NT-0.5 injection (twice a day for 7 days) significantly increased the ePSA in males and slightly affected the NT-0.5 effect in females. The Cx36 and Cx43 expression levels as well as Cx expressing neuronal populations were significantly increased by repeated NT-0.5 in in both male and female gerbils, and particularly, Cx43 expression was somewhat prominent in females. These results demonstrated a sex difference with respect to the nicotine effect on hippocampal bisynaptic excitability, irrelevant to connexin expression.

3.
Am J Physiol Regul Integr Comp Physiol ; 305(5): R499-505, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23824960

ABSTRACT

Administration of metformin is known to reduce both body weight and food intake. Although the hypothalamus is recognized as a critical regulator of energy balance and body weight, there is currently no evidence for an effect of metformin in the hypothalamus. Therefore, we sought to determine the central action of metformin on energy balance and body weight, as well as its potential involvement with key hypothalamic energy sensors, including adenosine monophosphate-activated protein kinase (AMPK) and S6 kinase (S6K). We used meal pattern analysis and a conditioned taste aversion (CTA) test and measured energy expenditure in C56BL/6 mice administered metformin (0, 7.5, 15, or 30 µg) into the third ventricle (I3V). Furthermore, we I3V-administered either control or metformin (30 µg) and compared the phosphorylation of AMPK and S6K in the mouse mediobasal hypothalamus. Compared with the control, I3V administration of metformin decreased body weight and food intake in a dose-dependent manner and did not result in CTA. Furthermore, the reduction in food intake induced by I3V administration of metformin was accomplished by decreases in both nocturnal meal size and number. Compared with the control, I3V administration of metformin significantly increased phosphorylation of S6K at Thr(389) and AMPK at Ser(485/491) in the mediobasal hypothalamus, while AMPK phosphorylation at Thr(172) was not significantly altered. Moreover, I3V rapamycin pretreatment restored the metformin-induced anorexia and weight loss. These results suggest that the reduction in food intake induced by the central administration of metformin in the mice may be mediated by activation of S6K pathway.


Subject(s)
Appetite Regulation/drug effects , Appetite Regulation/physiology , Hypothalamus/enzymology , Meals/drug effects , Meals/physiology , Metformin/administration & dosage , Ribosomal Protein S6 Kinases/biosynthesis , Animals , Enzyme Activation , Hypoglycemic Agents/administration & dosage , Hypothalamus/drug effects , Infusions, Intraventricular , Male , Mice , Mice, Inbred C57BL , Third Ventricle/drug effects , Third Ventricle/physiology
4.
Physiol Behav ; 110-111: 213-20, 2013 Feb 17.
Article in English | MEDLINE | ID: mdl-23391573

ABSTRACT

Metformin is widely used to treat obese diabetics because of its beneficial effects on body weight, energy intake, and glucose regulation. However, it has not been investigated how oral metformin affects meal patterns, or whether the reduced food intake is associated with neuronal activation in the hindbrain. Accordingly, we investigated how orally administered metformin (150 or 300 mg/kg daily for 4 or 7 days) reduces body weight in obese mice on a high-fat diet by continuously measuring meal patterns, energy expenditure, and locomotor activity, and whether oral metformin (300 mg/kg daily for 3 days) increases c-Fos expression in the nucleus tractus solitarius (NTS) and area postrema. Furthermore, we determined whether oral metformin produces a conditioned taste aversion (CTA) in obese mice administered a single dose of metformin (75, 150, or 300 mg/kg, p.o.). Metformin (300 mg/kg daily for 7 days) reduced body weight and adiposity by decreasing nocturnal energy intake but did not significantly change energy expenditure or locomotor activity relative to vehicle, and it transiently decreased nocturnal meal size and reduced meal number throughout the experiments. Furthermore, metformin significantly increased c-Fos immunoreactivity within the NTS of obese mice compared to that in controls and pair-fed group, and induced a CTA at doses of 150 or 300 mg/kg. These results indicate that metformin-induced weight loss is associated with a sustained reduction in energy intake maintained by a reduction in meal size and number, and that oral administration of metformin causes visceral illness and neuronal activation in the NTS.


Subject(s)
Eating/drug effects , Genes, fos/drug effects , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Obesity/psychology , Solitary Nucleus/drug effects , Animals , Avoidance Learning/drug effects , Body Weight/drug effects , Circadian Rhythm/drug effects , Dietary Fats/pharmacology , Energy Intake/drug effects , Energy Metabolism/drug effects , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Obesity/metabolism , Rhombencephalon/drug effects , Rhombencephalon/metabolism , Solitary Nucleus/metabolism , Taste/drug effects
5.
Mol Cells ; 30(3): 219-26, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20803089

ABSTRACT

Nicotine treatment has known to produce an inverse relationship between body weight and food intake in rodents. Present study determined the effect of repeated treatment with nicotine and withdrawal in control and obese mice, on: (1) body weight, caloric intake and energy expenditure; (2) hypothalamic neuropeptides mRNA expression; and (3) serum leptin. 21-week-old C57BL/6 mice (n = 65) received nicotine (3.0 mg/kg/day; 2 weeks) and saline (1 ml/kg/day; 2 weeks) subcutaneously. Animals were given either a normal-fat (10% kcal from fat, NF) or a high-fat diet (45% kcal from fat, HF) from the 12th week to 25th week. While, nicotine treatment for 14 days induced an increase in hypothalamic agouti-related protein, cocaine- and amphetamine- regulated transcript, pro-opiomelanocortin mRNA expressions, nicotine also produced a reducing effect in body weight gain and leptin concentration in NF mice. High-fat diet induced obese mice showed a blunted hypothalamic and leptin response to nicotine. Remarkable weight loss in obese mice was mediated not just by decreasing caloric intake, but also by increasing total energy expenditure (EE). During nicotine withdrawal period, weight gain occurred in NF and HF groups, which was ascribed to a decrease in EE rather than changes in caloric intake. Hypothalamic AgRP might play a role for maintaining energy balance under the nicotine-induced negative energy status.


Subject(s)
Energy Metabolism , Hypothalamus/metabolism , Nicotine/administration & dosage , Obesity/metabolism , Pro-Opiomelanocortin/biosynthesis , Animals , Dietary Fats/administration & dosage , Energy Metabolism/drug effects , Hypothalamus/drug effects , Hypothalamus/pathology , Leptin/blood , Mice , Mice, Inbred C57BL , Nicotine/adverse effects , Obesity/blood , Obesity/chemically induced , Obesity/pathology , Pro-Opiomelanocortin/genetics
6.
Int J Neurosci ; 119(9): 1267-81, 2009.
Article in English | MEDLINE | ID: mdl-19922355

ABSTRACT

The hypothalamic proteomes were analyzed 1 and 6 hr after an intraperitoneal injection of lithium chloride or sodium chloride (0.15 M, 12 ml/kg). Results showed that expression of 14 and 32 proteomes was increased consistently by 1 hr and 6 hr of lithium treatment, respectively. Among them, tentative implications of glial fibrillary acidic protein, receptor-type protein tyrosine phosphatase, spectrin, and glutamate dehydrogenase in the lithium-induced activation of the hypothalamic-pituitary-adrenal axis, and conditioned taste aversion have been discussed. The proteomes listed in this study will provide, at least, a new insight to understand the molecular mechanism of lithium's action in the brain.


Subject(s)
Antimanic Agents/pharmacology , Gene Expression/drug effects , Hypothalamus/metabolism , Lithium Chloride/pharmacology , Nerve Tissue Proteins/biosynthesis , Proteomics , Animals , Databases, Genetic , Echocardiography , Hypothalamus/drug effects , Injections, Intraperitoneal , Male , Nerve Tissue Proteins/genetics , Proteins/chemistry , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
7.
Eur J Pharmacol ; 589(1-3): 58-65, 2008 Jul 28.
Article in English | MEDLINE | ID: mdl-18550051

ABSTRACT

MK801 (dizocilpine) induces selective neurotoxic effects in the retrosplenial cortex, ranging from neuronal vacuolization to irreversible neurodegeneration depending on the dose administered. Although lamotrigine prevents MK801-induced neuronal vacuolization in the retrosplenial cortex 4 h after injection, it is not clear whether lamotrigine attenuates the subsequent neurodegeneration that occurs 3-4 days later. Because early growth response factor-1 (egr-1) plays a key role in neurodegeneration and its expression is induced in the retrosplenial cortex following MK801 treatment, it is possible that lamotrigine may attenuate MK801-induced neurodegeneration via inhibition of egr-1 expression in the retrosplenial cortex. To address this issue, we treated rats with lamotrigine (10 or 20 mg/kg) followed by MK801 (2 mg/kg) and measured changes in the levels of egr-1 mRNA and immunoreactivity in the retrosplenial cortex and other brain regions 3 h later. We also evaluated the effects of these treatments on neurodegeneration 4 days following treatment using Fluoro-Jade B staining. MK801 treatment increased egr-1 mRNA and immunoreactivity in the restrosplenial, cingulate, entorhinal and piriform cortices, but decreased levels in hippocampal subfields. These MK801-induced changes in egr-1 expression were significantly inhibited by lamotrigine pretreatment. In addition, MK801-induced neurodegeneration in the retrosplenial cortex was partially blocked by lamotrigine pretreatment in a dose dependent manner. These results demonstrate that lamotrigine pretreatment prevents the MK801-induced upregulation of egr-1 expression in a region-selective manner, and suggest that this effect may contribute, in part, to the attenuation of MK801-induced neurodegeneration in the retrosplenial cortex.


Subject(s)
Cerebral Cortex/drug effects , Early Growth Response Protein 1/metabolism , Hippocampus/drug effects , Nerve Degeneration/prevention & control , Neuroprotective Agents/pharmacology , RNA, Messenger/metabolism , Triazines/pharmacology , Animals , Cerebral Cortex/metabolism , Disease Models, Animal , Dizocilpine Maleate , Dose-Response Relationship, Drug , Early Growth Response Protein 1/genetics , Hippocampus/metabolism , Immunohistochemistry , Lamotrigine , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
8.
Eur J Pharmacol ; 571(2-3): 106-15, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17643412

ABSTRACT

The lysosomal destabilization that precedes mitochondrial apoptotic changes is an important step in cell death, particularly in oxidative cell death. This study describes the novel pharmacological effects of zaprinast, a cGMP-elevating phosphodiesterase inhibitor, on the inhibition of oxidative cell death in astrocyte cultures. H2O2-induced oxidative cytotoxicity was measured grossly by monitoring lactate dehydrogenase (LDH) release, and was found to be associated with lysosomal acridine orange relocation, lysosomal cathepsin D release into cytosol, and reduced mitochondrial potentials. Moreover, zaprinast (100 microM) inhibited all of these cytotoxic phenomena. In addition, H2O2-induced LDH release was not inhibited by 8-pCPT-cGMP, and the inhibition of this release by zaprinast was unaffected by Rp-8-pCPT-cGMP, a protein kinase G inhibitor. Zaprinast was found to inhibit sphingosine-induced lysosomal acridine orange relocation and the induced decrease in mitochondrial potential, but zaprinast had no effect on rotenone-induced mitochondrial collapse, which was not associated with lysosomal destabilization. However, zaprinast did not inhibit the cellular increase of reactive oxygen species induced by H2O2, which suggests that its protective mechanism differs from that of desferrioxamine, which does inhibit such cellular increase of oxygen free radicals. We suggest that the novel protective effect of zaprinast on H2O2-induced oxidative cell death is primarily associated with its inhibition of lysosomal destabilization.


Subject(s)
Astrocytes/drug effects , Cerebral Cortex/drug effects , Hydrogen Peroxide/toxicity , Lysosomes/drug effects , Oxidative Stress/drug effects , Phosphodiesterase Inhibitors/pharmacology , Protective Agents/pharmacology , Purinones/pharmacology , Animals , Animals, Newborn , Astrocytes/enzymology , Astrocytes/metabolism , Astrocytes/pathology , Cathepsin D/metabolism , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/metabolism , Cytoprotection , Cytosol/enzymology , Cytosol/metabolism , Deferoxamine/pharmacology , Dose-Response Relationship, Drug , Intracellular Membranes/drug effects , L-Lactate Dehydrogenase/metabolism , Lysosomes/enzymology , Lysosomes/metabolism , Lysosomes/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Rotenone/toxicity , Sphingosine/toxicity , Thionucleotides/pharmacology , Uncoupling Agents/toxicity
9.
Int J Neurosci ; 117(3): 387-400, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17365122

ABSTRACT

One major pathogenesis in degenerative disorders of the central nervous system (CNS), including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and ischemia, is the oxidative stress induced by reactive oxygen species (ROS). The present study investigated the protective effect of colloidal silver, which is widely marketed as a dietary supplement for diseases like diabetes, AIDS, cancer, and various infections, upon the oxidative brain damage induced by H(2)O(2) or naphthazarin treatment. LDH release from primary cultured astrocytes was enhanced by naphthazarin treatment, and this elevation of the LDH concentration in medium was blocked by colloidal silver treatment. However, hydrogen peroxide was little affected by the colloidal silver. Fluorescence of DCF (peroxides) increased in astrocytes incubated with hydrogen peroxide or naphthazarin compared to the control. When exposed to naphthazarin-induced cells, ROS formation appeared to be reduced by colloidal silver. However, intracellular ROS formation in hydrogen peroxide-treated cells slightly reduced by colloidal silver. These results suggest that colloidal silver has a protective activity against the oxidative stress induced by naphthazarin, but not by hydrogen peroxide.


Subject(s)
Astrocytes/drug effects , Cerebral Cortex/cytology , Hydrogen Peroxide/toxicity , Naphthoquinones/toxicity , Oxidants/toxicity , Silver Compounds/pharmacology , Animals , Animals, Newborn , Astrocytes/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , L-Lactate Dehydrogenase/metabolism , Rats , Rats, Sprague-Dawley , Reactive Nitrogen Species/metabolism , Tetrazolium Salts , Thiazoles
10.
Clin Pharmacol Ther ; 80(6): 646-56, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17178265

ABSTRACT

BACKGROUND AND OBJECTIVE: 1,4-Dihydropyridine calcium channel blockers, including amlodipine, are mainly metabolized by cytochrome P450 (CYP) 3A. We investigated the effect of CYP3A5*3 genotype on the pharmacokinetics and pharmacodynamics of amlodipine in healthy Korean male subjects. METHODS: Forty healthy male participants were enrolled and genotyped for the CYP3A5*3 gene. Each subject ingested a 5-mg dose of amlodipine, and plasma amlodipine concentrations were measured for 144 hours after dosing. Blood pressure and pulse rate were also measured for pharmacodynamic analysis. RESULTS: Among the 40 volunteers, 24 were CYP3A5*3/*3 carriers and 16 were CYP3A5*1 carriers (CYP3A5*1/*1 in 2 and CYP3A5*1/*3 in 14). The difference in the oral clearance of amlodipine approached statistical significance between the 2 major genotype groups, with CYP3A5*1 carriers (27.0 +/- 8.2 L/h) showing 20% lower clearance than CYP3A5*3/*3 carriers (32.4 +/- 10.2 L/h) (P = .063). However, the mean area under the plasma concentration-time curve of amlodipine was 200.9 +/- 61.9 ng . h/mL for CYP3A5*1 carriers and 167.6 +/- 45.0 ng . h/mL for CYP3A5*3/*3 carriers (P = .029). Moreover, the peak plasma concentration was significantly higher in CYP3A5*1 carriers (3.8 +/- 1.1 ng/mL) than in CYP3A5*3/*3 carriers (3.1 +/- 0.8 ng/mL) (P = .037). Pharmacodynamically, blood pressure and pulse rate were not significantly different between the 2 groups. CONCLUSIONS: CYP3A5*3/*3 carriers exhibited lower plasma amlodipine concentrations than CYP3A5*1 carriers. These findings suggest that the polymorphic CYP3A5 gene affects the disposition of amlodipine and provides a plausible explanation for interindividual variability in amlodipine disposition.


Subject(s)
Amlodipine/pharmacology , Cytochrome P-450 Enzyme System/genetics , Polymorphism, Genetic/genetics , Adult , Amlodipine/blood , Amlodipine/pharmacokinetics , Area Under Curve , Blood Pressure/drug effects , Cytochrome P-450 CYP3A , Genotype , Half-Life , Humans , Korea , Male , Metabolic Clearance Rate , Pharmacogenetics
11.
Eur J Pharmacol ; 545(2-3): 142-6, 2006 Sep 18.
Article in English | MEDLINE | ID: mdl-16884712

ABSTRACT

To investigate whether the anticonvulsant valproate influences the changes in brain-derived neurotrophic factor (BDNF) mRNA expression induced by MK801 in rat brain, we injected valproate prior to MK801 and observed the changes in the BDNF expression 3 h later. MK801 significantly increased BDNF expression in the retrosplenial and entorhinal cortex, and these increases were prevented by valproate pretreatment. Valproate pretreatment significantly blocked the MK801-induced increase of BDNF expression in retrosplenial cortex at 3 h, 6 h, and 9 h after MK801 injection, suggesting that valproate pretreatment did not delay the MK801-induced increase of BDNF expression. However, MK801 significantly decreased BDNF expression in the granule cell layer of hippocampus, and valproate pretreatment before MK801 potentiated the MK801-induced decrease in BDNF expression in granule cell layer. These results indicate that valproate pretreatment differentially affects the MK801-induced changes in BDNF expression in a region-selective manner.


Subject(s)
Anticonvulsants/pharmacology , Brain-Derived Neurotrophic Factor/genetics , Brain/drug effects , Dizocilpine Maleate/pharmacology , RNA, Messenger/analysis , Valproic Acid/pharmacology , Animals , Brain/metabolism , Male , Rats , Rats, Sprague-Dawley
12.
Neuropharmacology ; 50(7): 824-33, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16504218

ABSTRACT

Accumulating evidence suggests that dysregulation of corticotropin-releasing factor (CRF) may play a role in depression and that this dysregulation may be corrected by antidepressant drug treatment. Here, we examined whether chronic mild stress (CMS) alters CRF mRNA levels in stress-related brain areas including the bed nucleus of the stria terminalis (BNST) and the central nucleus of amygdala (CeA), and whether repeated tianeptine treatment can attenuate CMS-induced changes in CRF mRNA levels. Male rats were exposed to CMS for 19 days, and control animals were subjected to brief handling. Both groups were injected daily with tianeptine or saline. CMS significantly increased CRF mRNA levels in the dorsal BNST (dBNST), but not in other areas. Repeated tianeptine treatment prevented the CMS-induced increase in CRF mRNA levels in the dBNST, and reduced CRF mRNA levels in dBNST in non-stressed controls. Moreover, repeated tianeptine treatment significantly decreased CRF mRNA levels in the ventral BNST and CeA of non-stressed controls as well as CMS-exposed rats. These results show that CMS induces a rather selective increase of CRF mRNA in the dBNST. In addition, these results suggest that repeated tianeptine treatment diminishes the basal activity of CRF neurons and reduces their sensitivity to stress.


Subject(s)
Corticotropin-Releasing Hormone/biosynthesis , Gene Expression Regulation/drug effects , RNA, Messenger/biosynthesis , Stress, Physiological/metabolism , Thiazepines/administration & dosage , Animals , Corticotropin-Releasing Hormone/genetics , Gene Expression Regulation/physiology , Male , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Stress, Physiological/drug therapy , Stress, Physiological/genetics
13.
Exp Mol Med ; 38(1): 44-54, 2006 Feb 28.
Article in English | MEDLINE | ID: mdl-16520552

ABSTRACT

New-born cells continue to proliferate and survive to become mature granule cells in adult rat hippocampus. Although this process, known as neurogenesis, is inhibited by acute stress, it is not clear whether chronic stress affects neurogenesis. To determine whether chronic mild stress (CMS) influences neurogenesis in the adult rat hippocampus, male Sprague-Dawley rats were exposed to CMS and administered bromodeoxyuridine (BrdU) before or after CMS to observe the survival/differentiation or proliferation of new-born cells, respectively. In addition, we measured brain-derived neurotrophic factor (BDNF) mRNA in the granule cell layer (GCL) of the hippocampus, because BDNF is known to play an important role in the survival of new-born cells. CMS significantly decreased the survival of new-born cells in the GCL, but did not influence the proliferation or differentiation of new-born cells. CMS did not affect the proliferation and survival of new-born cells in the hilus. In addition, CMS did not change BDNF mRNA levels in the GCL. These results demonstrate that CMS reduces the survival of new-born cells but not of their proliferation, suggesting that repeated mild stress could influence a part of neurogenesis, but not the whole part of neurogenesis. These results raise the possibility that the survival of new-born cells may be suppressed in the presence of normal BDNF mRNA levels in GCL.


Subject(s)
Bromodeoxyuridine/administration & dosage , Hippocampus/pathology , Stress, Physiological/physiopathology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Calbindins , Cell Proliferation , Cell Survival , Fluorescein-5-isothiocyanate , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/cytology , Hippocampus/growth & development , Immunohistochemistry , In Situ Hybridization , Male , Microscopy, Confocal , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Restraint, Physical , Rhodamines , S100 Calcium Binding Protein G/metabolism , Stress, Physiological/pathology
14.
Peptides ; 26(11): 2246-51, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16269353

ABSTRACT

Doxapram causes panic anxiety in humans. To determine whether doxapram alters corticotropin-releasing factor (CRF) expression in the central nucleus of the amygdala (CeA), paraventricular nucleus of hypothalamus (PVN), or bed nucleus of the stria terminalis (BNST), we used immunohistochemistry to measure CRF peptide in these brain areas after doxapram injection. Doxapram injection significantly increased CRF-like immunoreactivity (CRF-IR) within the CeA, but not in the BNST or PVN, and this increase was significant 2h after injection. In addition, doxapram significantly increased CRF mRNA expression within the CeA, and this was most prominent 30min after injection. These results suggest that doxapram selectively increases CRF expression within the CeA, and that this is mediated by increased CRF gene transcription. This increase in CRF-IR within the CeA might explain the doxapram-induced anxiety reaction.


Subject(s)
Amygdala/metabolism , Central Nervous System Stimulants/adverse effects , Corticotropin-Releasing Hormone/biosynthesis , Doxapram/adverse effects , Gene Expression Regulation/drug effects , Amygdala/pathology , Animals , Central Nervous System Stimulants/administration & dosage , Doxapram/administration & dosage , Immunohistochemistry , In Situ Hybridization , Male , Panic Disorder/chemically induced , Panic Disorder/metabolism , Panic Disorder/pathology , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/pathology , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Septal Nuclei/metabolism , Septal Nuclei/pathology
15.
Mol Cells ; 19(1): 74-80, 2005 Feb 28.
Article in English | MEDLINE | ID: mdl-15750343

ABSTRACT

Stress is known to inhibit granule cell proliferation in the hippocampus. However, recent studies suggest that the commonly used dose of bromodeoxyuridine (BrdU) is insufficient to label all fractions of granule cells. Furthermore, stress-induced changes in BrdU availability may influence the labeling of newly born cells. To investigate whether changes in BrdU availability affect measurements of stress-induced granule cell proliferation, granule cell proliferation was assessed using injection of high doses of BrdU before and after restraint stress lasting 1 h. In addition, to determine whether stress-induced changes in plasma corticosterone levels were influenced by the BrdU, time-dependent changes in plasma corticosterone levels over 2 h after BrdU injection were compared with total accumulated plasma corticosterone levels [as determined by areas under the curve (AUC)]. Restraint stress significantly reduced the numbers of BrdU-labeled cells and clusters in the granule cell layer (GCL) of rats that received BrdU after stress, and decreases of similar magnitude were observed when the rats were given BrdU before stress. BrdU injection enhanced the stress-induced plasma corticosterone response, but there was no difference between the mean AUCs of plasma corticosterone levels of animals injected with BrdU before or after stress. These observations suggest that restraint stress decreases granule cell proliferation, and that this may be influenced by the extent and duration of plasma corticosterone increases rather than by changes in the availability of BrdU.


Subject(s)
Bromodeoxyuridine/administration & dosage , Hippocampus/pathology , Stress, Physiological/physiopathology , Animals , Cell Proliferation , Corticosterone/blood , Hippocampus/cytology , Hippocampus/growth & development , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley , Restraint, Physical , Stress, Physiological/pathology
16.
Yonsei Med J ; 45(4): 649-60, 2004 Aug 31.
Article in English | MEDLINE | ID: mdl-15344206

ABSTRACT

Hepatic stellate cells (HSCs) are known to play a role in the pathogenesis of the increased intrahepatic vascular resistance found in chronic liver diseases. The aim of this study was to evaluate the K+ and Ca2+ currents in cultured HSCs from rat liver, through the patch-clamp technique. Most cells were positive for desmin immunostain after isolation and in alpha-smooth muscle actin immunostain after 10 - 14 days of culturing. Outward and inward rectifying K+ currents were confirmed. Two different types of K+ currents were distinguished: one with the inward rectifying current and the other without. The outward K+ currents consisted of at least four components: tetraethylammonium (TEA)-sensitive current, 4-aminopyridine (4-AP)-sensitive current, pimozide-sensitive current and three blocker-resistant current. The peaks of the outward K+ currents evoked by a depolarizing pulse were decreased to 32.0 +/- 3.0, 62.8 +/- 3.7 and 32.8 +/- 3.5% by 5 mM TEA, 2 mM 4-AP and 15 micro M pimozide, respectively. Moreover, the combined application of three blockers caused 86.6 +/- 4.8% suppression. The inward currents evoked hyperpolarizing pulses were inwardly rectifying and almost blocked by Ba2+. Elevation of external K+ increased the inward current amplitude and positively shifted its reversal potential. Voltage- dependent Ca2+ currents which were completely abolished by Cd2+ and nimodipine were detected in 14 day cultured HSCs. In this study, the cultured HSCs were found to express outward K+ currents composed of multiple pharmacological components, Ba2+-sensitive inward rectifying K+ current and L-type Ca2+ current.


Subject(s)
Calcium Channels, L-Type/physiology , Calcium/metabolism , Hepatocytes/physiology , Potassium Channels, Voltage-Gated/physiology , Potassium/metabolism , Animals , Calcium Channel Blockers/pharmacology , Cells, Cultured , Hepatocytes/cytology , Immunohistochemistry , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley
17.
Free Radic Biol Med ; 33(10): 1321-33, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12419464

ABSTRACT

Oxidative stress and mitochondrial dysfunction are important aspects of pathogenesis, particularly in the brain, which is highly dependent on oxygen, and the protection of astrocytes is essential for neuroprotection. In this context, imidazoline drugs have been reported to be neuroprotective. Our recent study showed that imidazoline drugs, including guanabenz, inhibit the naphthazarin-induced oxidative cytotoxicity associated with lysosomal destabilization. We now report on a study into the protective effects of rilmenidine and AGN 192403, which have affinity for imidazoline-1 receptors, on the cytotoxicity induced by naphthazarin and inhibitors of mitochondrial respiration in astrocytes. Cytotoxicity was measured grossly by LDH release and by measuring changes in lysosomal membrane stability and features of mitochondrial membrane permeabilization. Naphthazarin-induced cytotoxicity was evidenced by the ordered development of lysosomal acridine orange relocation, decrease in mitochondrial potential, cytochrome c release, and caspase-9 activation, and was inhibited by guanabenz, rilmenidine, and AGN 192403. Antimycin A and rotenone induced mitochondrial dysfunction primarily, and their cytotoxicities were inhibited only by AGN 192403. Rilmenidine and guanabenz may have a lysosomal stabilizing effect, which underlies their protective effects. AGN 192403 might affect the mitochondrial cell death cascades, and had a novel protective effect on the cytotoxicity associated with mitochondrial dysfunction.


Subject(s)
Astrocytes/drug effects , Bridged Bicyclo Compounds/pharmacology , Heptanes/pharmacology , Mitochondria/metabolism , Oxazoles/pharmacology , Oxygen/metabolism , Animals , Animals, Newborn , Antihypertensive Agents/pharmacology , Antimycin A/pharmacology , Astrocytes/metabolism , Caspase 9 , Caspases/metabolism , Cell Death , Cytochrome c Group/metabolism , Dose-Response Relationship, Drug , Free Radicals , Lysosomes/metabolism , Membrane Potentials , Models, Chemical , Naphthoquinones/pharmacology , Neuroglia/cytology , Oxidative Stress , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , Rilmenidine , Time Factors
18.
Eur J Pharmacol ; 443(1-3): 119-24, 2002 May 17.
Article in English | MEDLINE | ID: mdl-12044801

ABSTRACT

Repeated injections of nicotine are well known to produce progressively larger increases in locomotor activity, an effect defined as behavioral sensitization. This study was carried out to investigate the role of nitric oxide (NO) and N-methyl-D-aspartate (NMDA) receptors in nicotine-induced behavioral sensitization. Rats were given repeated injections of nicotine (0.4 mg/kg, s.c., twice daily for 7 days) followed by one challenge injection on the fourth day after the last daily injection. Systemic challenge with nicotine produced a much larger increase in locomotor activity in nicotine-pretreated rats. Rats were pretreated with the nonselective nitric oxide synthase (NOS) inhibitor, N(G)-nitro-arginine-methyl-ester (L-NAME; 75 mg/kg, i.p.), the selective constitutive NOS inhibitor, N-nitro-L-arginine (L-NNA; 15 mg/kg, i.p.), the prototypical selective inducible NOS inhibitor, aminoguanidine (100 mg/kg, i.p.) or NMDA receptor antagonist, MK-801 ((5R,10S)-(+)-5-Methyl-10,11-dihydro-5H-dibenzo[a,d] cyclohepten-5,10-imine; 0.3 mg/kg, i.p.), 30 min before injections of nicotine during a 7-day development or a 3-day withdrawal phase after which challenged with nicotine on day 11. Pretreatment with L-NAME, L-NNA and MK-801, but not aminoguanidine, blocked the development of nicotine-induced sensitization to subsequent nicotine challenge. Injections of MK-801 twice daily during 3-day withdrawal periods after a 7-day induction period of nicotine attenuated nicotine-induced behavioral sensitization, whereas injections of L-NAME, L-NNA or aminoguanidine had no effects on the expression of sensitization produced by repeated nicotine. This study demonstrates that NMDA receptors can play a major role in the expression as well as development of nicotine-induced behavioral sensitization, and that NO is also involved in the development, but not critically involved in the expression of behavioral sensitization to nicotine.


Subject(s)
Dizocilpine Maleate/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Motor Activity/drug effects , Nicotine/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Behavior, Animal/drug effects , Male , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/physiology , Nitric Oxide Synthase Type II , Rats , Rats, Sprague-Dawley
19.
Mol Cells ; 13(1): 137-43, 2002 Feb 28.
Article in English | MEDLINE | ID: mdl-11911465

ABSTRACT

We previously reported that a cytostatic protein that is found in ASC-17D Sertoli cell-conditioned media was Mycoplasma arginine deiminase (ADI), which hydrolyzes L-arginine into L-citrulline and ammonia. Here, we report the over-expression of recombinant ADI (rADI) in E. coli and the down-regulation of lipopolysaccharide (LPS) induced-nitric oxide (NO) production by rADI treatment. We cloned the ADI gene from Mycoplasma arginini genomic DNA by a polymerase chain reaction, and changed five TGA tryptophan codons (stop codon in E. coli) to TGG codons in the coding region by site-directed mutagenesis in order to express in E. coli. The rADI was purified to apparent homogeneity by DEAE-Sepharose and arginine-affinity chromatography. The rADI expressed in E. coli was identified as 45 kDa on SDS-PAGE and 90 kDa on native PAGE, implying that it exists as a dimer like ADI of M. arginini. The Km for arginine of rADI was approximately 370+/-50 microM. Its optimal temperature and pH were 41 degrees C and pH 6.4, respectively, and enzyme activity remained > or = 50% for 5 d at physiological temperature and pH. Treatment of purified rADI suppressed NO production in macrophage-like RAW 264.7 and primary glial cells that were exposed to LPS. Furthermore, an intraperitoneal injection of rADI significantly suppressed the rise of blood nitrite/nitrate levels that were induced by the systemic administration of bacterial endotoxin LPS to mice, resulting in an improvement in their survival rate. These results suggest that the depletion of blood arginine with an arginine-metabolizing enzyme, such as ADI, could suppress excessive production of NO that is caused by inducible NOS (iNOS) during the endotoxemia. Also, rADI may be used as a new approach to control NO-related diseases, such as sepsis.


Subject(s)
Hydrolases/genetics , Hydrolases/metabolism , Mycoplasma/enzymology , Mycoplasma/genetics , Nitric Oxide/biosynthesis , Animals , Arginine/blood , Base Sequence , DNA, Bacterial/genetics , Endotoxemia/blood , Endotoxemia/drug therapy , Escherichia coli/genetics , Gene Expression , Genes, Bacterial , Hydrolases/chemistry , Hydrolases/therapeutic use , Kinetics , Lipopolysaccharides/toxicity , Mice , Mice, Inbred ICR , Molecular Sequence Data , Molecular Weight , Mutagenesis, Site-Directed , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use
20.
Free Radic Biol Med ; 32(5): 394-405, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11864779

ABSTRACT

Oxidative stress is a primary pathogenesis in the brain, which is particularly vulnerable to oxidative stress. Maintenance of astrocyte functions under oxidative stress is essential to prevent neuronal injuries and to recover neuronal functions in various pathologic conditions. Imidazoline drugs have affinities for imidazoline receptors, which are highly distributed in the brain, and have been shown to be neuroprotective. This study presented the protective effects of several imidazoline drugs against oxidative cytotoxicity, in primary cultures of astrocytes. Imidazoline drugs, such as idazoxan, guanabenz, guanfacine, BU224, and RS-45041-190, showed protective effects against naphthazarin-induced oxidative cytotoxicity, as evidenced by LDH release and Hoechst 33342/propidium iodide staining. The imidazoline drugs stabilized lysosomes and inhibited naphthazarin-induced lysosomal destabilization, as evidenced by acridine orange relocation. Guanabenz inhibited, the leakage of lysosomal cathepsin D to cytosol, the decreased mitochondrial potential, and the release of mitochondrial cytochrome c, which were induced by naphthazarin. The lysosomal destabilization by oxidative stress and other apoptotic signals and subsequent cathepsin D leakage to the cytosol can induce apoptotic changes of mitochondria and eventually cell death. Therefore, lysosomal stabilization by imidazoline drugs may be ascribed to their protective effects against oxidative cytotoxicity.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Astrocytes/metabolism , Cell Division/drug effects , Imidazoles/therapeutic use , Lysosomes/enzymology , Naphthoquinones/pharmacology , Acridine Orange , Adrenergic alpha-Antagonists/therapeutic use , Animals , Animals, Newborn , Antineoplastic Agents/adverse effects , Astrocytes/drug effects , Cathepsin D/metabolism , Cell Line , Cerebral Cortex/metabolism , Cytochrome c Group/metabolism , Fish Venoms , Guanabenz/therapeutic use , Guanfacine/therapeutic use , HIV Protease Inhibitors/pharmacology , Idazoxan/therapeutic use , Indoles/therapeutic use , Isoindoles , L-Lactate Dehydrogenase/metabolism , Ligands , Lysosomes/drug effects , Membrane Potentials/drug effects , Mitochondria/physiology , Naphthoquinones/adverse effects , Neuroglia/metabolism , Oxidation-Reduction , Pepstatins/pharmacology , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptors, Drug
SELECTION OF CITATIONS
SEARCH DETAIL
...