Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Hepatol ; 50(6): 1184-91, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19398231

ABSTRACT

BACKGROUND/AIMS: Oxidative stress via generation of reactive oxygen species is suggested to be the major mechanism of alcohol-induced liver injury. We investigated the effects of glutathione peroxidase-1 and catalase double deficiency (Gpx-1(-/-)/Cat(-/-)) on liver injury and changes in the sulfur amino acid metabolism induced by binge ethanol administration. METHODS: Ethanol (5 g/kg) was administered orally to the wild-type and the Gpx-1(-/-)/Cat(-/-) mice every 12 h for a total of three doses. Mice were sacrificed 6 h after the final dose. RESULTS: The Gpx-1/Cat deficiency alone increased malondialdehyde levels in liver significantly. Hepatic methionine adenosyltransferase (MAT) activity and S-adenosylmethionine levels were decreased, however, glutathione contents were not changed. Ethanol administration to the Gpx-1(-/-)/Cat(-/-) mice increased the elevation of serum alanine aminotransferase activity, plasma homocysteine levels, hepatic fat accumulation and lipid peroxidation compared with the wild-type animals challenged with ethanol. Also the reduction of MAT activity and S-adenosylmethionine levels was enhanced, but MATI/III expression was increased significantly. CONCLUSIONS: The results indicate that Gpx-1 and Cat have critical roles in the protection of liver against binge ethanol exposure. Augmentation of ethanol-induced oxidative stress may be responsible for the impairment of the transsulfuration reactions and the aggravation of acute liver injury in the Gpx-1(-/-)/Cat(-/-) mice.


Subject(s)
Acatalasia/metabolism , Amino Acids, Sulfur/metabolism , Ethanol/toxicity , Glutathione Peroxidase/deficiency , Liver/drug effects , Liver/metabolism , Acatalasia/genetics , Animals , Catalase/genetics , Catalase/metabolism , Cytochrome P-450 CYP2E1/metabolism , Glutathione Peroxidase/genetics , Liver/injuries , Liver/pathology , Male , Metabolomics , Methionine Adenosyltransferase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Reactive Oxygen Species/metabolism , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism , Glutathione Peroxidase GPX1
2.
Chem Biol Interact ; 177(3): 204-11, 2009 Feb 12.
Article in English | MEDLINE | ID: mdl-18930038

ABSTRACT

Previous studies suggested that betaine intake might antagonize the induction of oxidative stress-mediated acute liver injury through regulation of the sulfur-amino acid metabolism. In this study we examined the protective effects of betaine on chronic liver injury and fibrosis induced by dimethylnitrosamine (DMN). Male rats were supplemented with betaine (1%, w/v) in drinking water from 2 weeks prior to the initiation of DMN treatment (10mg/(kg day), i.p., 3 days/week, for 1, 2, or 4 weeks) until sacrifice. Induction of liver injury was determined by quantifying serum alanine aminotransferase, aspartate aminotransferase activities, bilirubin levels, hepatic xenobiotic-metabolizing capacity, histopathological changes and 4-hydroxyproline levels. Development of oxidative injury was estimated by malondialdehyde (MDA) levels and total oxyradical scavenging capacity (TOSC) of liver and serum toward hydroxyl, peroxyl radicals, and peroxynitrite. Progressive changes in the parameters of liver injury and fibrosis were evident in the rats challenged with DMN. Elevation of MDA levels in liver was significant before the onset of a change in any parameters determined in this study. Betaine supplementation markedly attenuated the induction of hepatotoxicity and fibrosis by DMN. Elevation of MDA and the reduction of TOSC were also depressed significantly. Development of liver injury corresponded well with the induction of oxidative stress in rats treated with DMN, both of which are inhibited effectively by betaine supplementation. It is suggested that betaine may protect liver from fibrogenesis by maintaining the cellular antioxidant capacity.


Subject(s)
Betaine/administration & dosage , Dimethylnitrosamine/toxicity , Liver Cirrhosis/prevention & control , Liver/drug effects , Alanine Transaminase/metabolism , Animals , Aspartate Aminotransferases/metabolism , Liver/enzymology , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/enzymology , Liver Cirrhosis/pathology , Male , Malondialdehyde/metabolism , Oxidative Stress , Rats , Rats, Sprague-Dawley
3.
Int J Urol ; 15(2): 178-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18269460

ABSTRACT

A 56-year-old male who had renal transplants at the age of 34 and 49 years, presented with painless gross hematuria six years after the second renal transplantation. An abdominal computed tomography scan revealed diffuse wall thickening of the distal ureter of the failed first allograft and a bulging lesion about 1.2 cm in size on the lower pole of the right native kidney. Both lesions were suspected for tumors, but they showed a ureteral nephrogenic adenoma and a renal hemorrhagic simple cyst.


Subject(s)
Adenoma/diagnosis , Cysts/diagnosis , Hematuria/etiology , Kidney Diseases/diagnosis , Ureteral Neoplasms/diagnosis , Cysts/complications , Humans , Kidney Transplantation , Male , Middle Aged
4.
Pharmacol Ther ; 113(1): 88-120, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17097148

ABSTRACT

Endogenous factors, including hormones, growth factors and cytokines, play an important role in the regulation of hepatic drug metabolizing enzyme expression in both physiological and pathophysiological conditions. Diabetes, fasting, obesity, protein-calorie malnutrition and long-term alcohol consumption produce changes in hepatic drug metabolizing enzyme gene and protein expression. This difference in expression alters the metabolism of xenobiotics, including procarcinogens, carcinogens, toxicants and therapeutic agents, potentially impacting the efficacy and safety of therapeutic agents, and/or resulting in drug-drug interactions. Although the mechanisms by which xenobiotics regulate drug metabolizing enzymes have been studied intensively, less is known regarding the cellular signaling pathways and components which regulate drug metabolizing enzyme gene and protein expression in response to hormones and cytokines. Recent findings, however, have revealed that several cellular signaling pathways are involved in hormone- and growth factor-mediated regulation of drug metabolizing enzymes. Our laboratory has reported that insulin and growth factors regulate drug metabolizing enzyme gene and protein expression, including cytochromes P450 (CYP), glutathione S-transferases (GST) and microsomal epoxide hydrolase (mEH), through receptors which are members of the large receptor tyrosine kinase (RTK) family, and by downstream effectors such as phosphatidylinositol 3-kinase, mitogen activated protein kinase (MAPK), Akt/protein kinase B (PKB), mammalian target of rapamycin (mTOR), and the p70 ribosomal protein S6 kinase (p70S6 kinase). Here, we review current knowledge of the signaling pathways implicated in regulation of drug metabolizing enzyme gene and protein expression in response to insulin and growth factors, with the goal of increasing our understanding of how disease affects these signaling pathways, components, and ultimately gene expression and translational control.


Subject(s)
Gene Expression Regulation, Enzymologic , Hypoglycemic Agents/metabolism , Insulin/metabolism , Liver/enzymology , Signal Transduction , Animals , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/genetics , Epoxide Hydrolases/biosynthesis , Epoxide Hydrolases/genetics , Gene Expression Regulation, Enzymologic/drug effects , Glutamate-Cysteine Ligase/biosynthesis , Glutamate-Cysteine Ligase/genetics , Glutathione Transferase/biosynthesis , Glutathione Transferase/genetics , Humans , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Isoenzymes , Liver/drug effects , MAP Kinase Signaling System/drug effects , Metabolic Detoxication, Phase II/genetics , Phosphoric Monoester Hydrolases/metabolism , Receptor, Insulin/metabolism , Receptors, Growth Factor/metabolism , Signal Transduction/drug effects
5.
Eur J Appl Physiol ; 96(5): 581-6, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16408233

ABSTRACT

Proenkephalin peptide F [107-140] is an enkephalin-containing peptide found predominantly within the adrenal medulla and is co-packaged with epinephrine within adrenal medullary chromaffin granules. Peptide F has been shown to have the classic opioid analgesia effects along with immune cell interactions. This is only the second peptide F study in women, and in it we compare the responses of peptide F to a maximal cycle exercise test and recovery values over the follicular and luteal phases of the menstrual cycle. Eight untrained (directly documented in this study) women who were eumenorrheic performed a progressive maximal exercise test to volitional exhaustion on a cycle ergometer, once during the follicular phase, and once during the luteal phases of the menstrual cycle. Blood was obtained pre-exercise, immediately post-exercise and at 0, 15, and 30 min into recovery. Typical exercise changes in response to the cycle tests were observed with blood lactate increases that remained elevated 30 min into recovery. No significant exercise-induced elevations were observed for peptide F concentrations with exercise nor were any differences observed between the two menstrual phases. Thus, the effects of the menstrual cycle on peptide F concentrations appear to be minimal under the conditions of this investigation. With high concentrations of peptide F observed at rest (approx. 0.2-0.3 pmol ml(-1)) pre-exercise arousal mechanisms may have obviated any exercise-induced response. In addition, inhibition via elevated epinephrine may have inhibited any post-exercise increases and finally adrenal medullary capacity for circulatory concentrations of peptide F may have been reached in such untrained women. Pre-exercise arousal mechanisms potentially related to analgesia may also be involved to prepare untrained women for the stress of maximal exercise.


Subject(s)
Adrenal Medulla/physiology , Enkephalin, Methionine/analogs & derivatives , Exercise/physiology , Follicular Phase/physiology , Luteal Phase/physiology , Protein Precursors/blood , Adolescent , Adult , Enkephalin, Methionine/blood , Estradiol/blood , Exercise Test , Female , Heart Rate/physiology , Humans , Lactic Acid/blood , Plasma Volume/physiology , Progesterone/blood
6.
Drug Metab Dispos ; 34(4): 683-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16443668

ABSTRACT

The antioxidant activity of flavonoids, directly through scavenging oxidizing species and indirectly through modulating drug-metabolizing enzyme activities, is associated with chemopreventive and chemotherapeutic effects. However, little published information is available concerning the effect of flavonoids on glutathione (GSH) homeostasis. We previously demonstrated that PD98059 (2'-amino-3'-methoxyflavone), a flavone derivative and selective mitogen-activated protein kinase kinase (MEK) 1 inhibitor, enhanced the insulin-mediated increase in GSH levels. To determine whether the PD98059-mediated increase in GSH levels was associated with MEK inhibition, primary cultured rat hepatocytes were treated with PD98059, the MEK inhibitor U0126, which is not a flavone derivative, or flavone. PD98059 increased GSH levels in a concentration-dependent manner in hepatocytes cultured in the presence or absence of insulin. In contrast, GSH levels were not affected by U0126 at concentrations sufficient to inhibit insulin-mediated extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation. Flavone, however, markedly increased GSH levels without inhibition of ERK1/2 phosphorylation. The concentration of GSH in the culture medium was also elevated by PD98059 or flavone, suggesting that the cellular GSH elevation could not be accounted for by the inhibition of GSH efflux into medium. Interestingly, PD98059 and flavone increased cellular cysteine levels, which may be responsible for the PD98059- and flavone-mediated elevation of GSH levels. These results provide evidence that PD98059 and flavone produce dramatic changes in GSH homeostasis in hepatocytes, through a mechanism(s) unrelated to MEK inhibition. Moreover, the current study implies that flavonoid-induced chemopreventive and chemotherapeutic effects may be mediated by regulation of redox state through the stimulation of GSH synthesis.


Subject(s)
Flavonoids/pharmacology , Glutathione/metabolism , Hepatocytes/drug effects , Protein Kinase Inhibitors/pharmacology , Animals , Cell Culture Techniques , Cells, Cultured , Cysteine/metabolism , Dose-Response Relationship, Drug , Flavones , Hepatocytes/enzymology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/metabolism , Male , Rats , Rats, Sprague-Dawley
7.
J Pharmacol Exp Ther ; 316(3): 1255-61, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16293713

ABSTRACT

We reported previously that insulin elevated alpha-class glutathione S-transferase (GSTs) protein levels in primary cultured rat hepatocytes (Kim et al., 2003b). In contrast, glucagon down-regulated alpha- and pi-class GST expression, and mechanistic research implicated cAMP and protein kinase A in this process (Kim et al., 2003b). The present study examines the signaling pathways involved in the regulation of alpha-class GST in response to insulin in primary cultured rat hepatocytes. Protein levels of GSTA1/2 and GSTA3/5 and activity of GST toward 7-chloro-4-nitrobenzo-2-oxa-1,3-diazole (NBD) were increased in an insulin concentration-dependent manner. Treatment of cells with the phosphatidylinositol 3-kinase (PI3K) inhibitors wortmannin and LY294002 [2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one] or rapamycin, an inhibitor of mammalian target of rapamycin and ribosomal p70 S6 kinase (p70S6K) phosphorylation, or with an adenovirus containing green fluorescent protein and a dominant-negative and kinase-dead Akt, effectively inhibited the insulin-mediated increase in alpha-class GST expression and GST activity toward NBD. In contrast, PD98059 (2'-amino-3'-methoxyflavone), an inhibitor of mitogen-activated protein kinase kinase, SP600125 (1,9-pyrazoloanthrone), an inhibitor of c-Jun N-terminal kinase, SB203580 [4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imadazole], an inhibitor of p38 mitogen-activated protein kinase, or bisindolylmaleimide, a broad spectrum inhibitor of protein kinase C, did not inhibit the insulin-mediated increase in alpha-class GST protein levels in hepatocytes. These results show that PI3K/Akt/p70S6K signaling is active in the insulin-mediated up-regulation of the antioxidant defense system and that low insulin levels, as encountered in diabetes, potentially increase the susceptibility of hepatocytes to xenobiotic-mediated and/or oxidative stress-mediated damage.


Subject(s)
Glutathione Transferase/biosynthesis , Hepatocytes/enzymology , Insulin/pharmacology , Signal Transduction/physiology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Epoxide Hydrolases/biosynthesis , Male , Phosphatidylinositol 3-Kinases/physiology , Protein Kinase C/physiology , Proto-Oncogene Proteins c-akt/physiology , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 70-kDa/physiology
8.
Biochem Pharmacol ; 70(12): 1785-95, 2005 Dec 05.
Article in English | MEDLINE | ID: mdl-16242670

ABSTRACT

Mechanical stress is known to activate signaling cascades, including mitogen-activated protein kinase (MAPK) pathways. Although mechanical stress has been implicated in hepatic cirrhosis and liver regeneration following hepatectomy, the signaling pathway(s) that may be activated in hepatocytes in response to mechanical stress have not been determined. Using primary cultured rat hepatocytes to examine cellular signaling in response to mechanical stress associated with medium change, we observed that the phosphorylation status of extracellular signal-regulated kinase 1/2 (ERK1/2), Jun N-terminal kinase and p38 MAPK, but not Akt, was altered. MAPK activation, especially ERK1/2, was rapidly increased within 5 min, followed by a subsequent decrease to below basal levels between 30 min and 1 h following medium change. MAPK/ERK kinase (MEK1/2) phosphorylation followed the same pattern. The phosphorylation of Raf-1 in response to medium change was also consistent with Raf-1 serving as an upstream regulator of MEK1/2-ERK1/2 signaling. Phosphorylation of ERK1/2 was increased by mechanical stress alone, suggesting that mechanical stress may be primarily responsible for ERK1/2 activation in response to medium change. Medium change produced a marked decline in oxidized glutathione and malondialdehyde levels, and the antioxidant N-acetyl-L-cysteine decreased basal ERK1/2 phosphorylation, suggesting a role for oxidative stress in maintaining basal ERK1/2 phosphorylation in cultured hepatocytes. These data suggest that medium change results in immediate activation of the MAPK signaling pathway due to mechanical stress, followed by a subsequent inactivation of MAPK signaling due to a reduction in oxidative stress levels. These processes may be associated with alteration of hepatic hemodynamic circulation observed in hepatic diseases and in liver transplantation.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Hepatocytes/enzymology , Oxidative Stress , Animals , Cells, Cultured , Culture Media , Enzyme Activation , Epidermal Growth Factor/pharmacology , MAP Kinase Signaling System , Male , Phosphorylation , Proto-Oncogene Proteins c-raf/metabolism , Rats , Rats, Sprague-Dawley , Stress, Mechanical
9.
J Pharmacol Exp Ther ; 315(1): 203-13, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15980059

ABSTRACT

The ketone body acetoacetate (AA) in the absence of insulin or in the presence of diabetic insulin levels decreases CYP2E1 mRNA expression in a concentration- and time-dependent manner in primary cultured rat hepatocytes. AA activates p70 ribosomal S6 kinase (p70S6K) and protein kinase C (PKC) by approximately 2- to 2.5-fold, respectively, following 6-h treatment. The AA-mediated activation of p70S6K, but not PKC, was abolished by inhibition of PI 3-K with LY294002 [2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one] or wortmannin, in agreement with p70S6K being downstream of phosphatidylinositol 3-kinase (PI 3-K). Inhibition of PI 3-K, mTOR with rapamycin, or PKC with bisindolylmaleimide ameliorated the AA-mediated down-regulation of CYP2E1 mRNA expression. Neither the mitogen-activated protein kinase kinase inhibitor PD98059 (2'-amino-3'-methoxyflavone) nor the p38 mitogen-activated protein kinase inhibitor SB203580 [4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imidazole] ameliorated the AA-mediated suppression of CYP2E1 mRNA expression. Heterogeneous nuclear RNA analysis revealed that AA suppressed CYP2E1 gene transcription by approximately 50% and that inhibition of PI 3-K and PKC diminished this AA-mediated effect on transcription. CYP2E1 mRNA half-life slightly increased from approximately 24 h in untreated hepatocytes to approximately 32 h in AA-treated cells. Interestingly, AA increased CYP2E1 protein levels by approximately 2- and 2.5-fold at 24 and 48 h, respectively. DL-beta-hydroxybutyrate was without effect. Polysomal distribution studies revealed that AA increased the proportion of RNA associated with the actively translated polysomal fractions versus the 40S to 60S untranslated fractions by approximately 40%. CYP2E1 protein half-life increased from approximately 8 h in untreated hepatocytes to approximately 24 in AA-treated cells. These data show that AA decreases CYP2E1 mRNA expression through inhibition of gene transcription while simultaneously elevating CYP2E1 protein levels through increased translation and decreased protein degradation.


Subject(s)
Acetoacetates/pharmacology , Cytochrome P-450 CYP2E1/genetics , Gene Expression Regulation/drug effects , Hepatocytes/metabolism , Animals , Cells, Cultured , Cytochrome P-450 CYP2E1/analysis , Dose-Response Relationship, Drug , Insulin/pharmacology , Male , Phosphatidylinositol 3-Kinases/physiology , Protein Kinases/physiology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 70-kDa/physiology , Signal Transduction , TOR Serine-Threonine Kinases
10.
J Hepatol ; 42(6): 907-13, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15885362

ABSTRACT

BACKGROUND/AIMS: We previously reported that acute betaine treatment induced significant changes in the hepatic glutathione and cysteine levels in mice and rats. The present study was aimed to determine the effects of dietary betaine on the metabolism of sulfur-containing amino acids. METHODS/RESULTS: Male mice were supplemented with betaine (1%) in drinking water for up to 3 weeks. Changes in hepatic levels of major sulfur amino acid metabolites and products were stabilized after 2 weeks of betaine supplementation. Betaine intake increased methionine, S-adenosylmethionine, and S-adenosylhomocysteine levels significantly, but homocysteine and cystathionine were reduced. Methionine adenosyltransferase activity was elevated to three-fold of control. Cysteine catabolism to taurine was inhibited as evidenced by a decrease in cysteine dioxygenase activity and taurine levels in liver and plasma. Despite the significant changes in the transsulfuration reactions, neither hepatic cysteine nor glutathione was altered. Betaine supplementation decreased the hepatotoxicity induced by chloroform (0.5 ml/kg, ip) significantly. CONCLUSIONS: Betaine supplementation enhances recycling of homocysteine for the generation of methionine and S-adenosylmethionine while reducing its utilization for the synthesis of cystathionine and cysteine. However, the hepatic levels of cysteine or glutathione are not affected, most probably due to the depression of taurine generation from cysteine.


Subject(s)
Amino Acids, Sulfur/metabolism , Betaine/pharmacology , Lipotropic Agents/pharmacology , Liver/drug effects , Liver/metabolism , Amino Acids, Sulfur/blood , Animals , Chemical and Drug Induced Liver Injury/metabolism , Chloroform/toxicity , Cysteine/blood , Cysteine/metabolism , Glutathione/metabolism , Kidney/drug effects , Kidney/metabolism , Male , Methionine/blood , Methionine/metabolism , Methionine Adenosyltransferase/metabolism , Mice , Mice, Inbred ICR , S-Adenosylmethionine/metabolism , Solvents/toxicity , Taurine/blood , Taurine/metabolism
11.
Food Chem Toxicol ; 43(5): 663-70, 2005 May.
Article in English | MEDLINE | ID: mdl-15778005

ABSTRACT

Alterations in the hepatic metabolism of sulfur amino acids in experimental cholestasis induced by alpha-naphthylisothiocyanate (ANIT) (100 mg/kg, po) were monitored in male mice for 1 week. We also examined the effects of betaine supplementation (1% in drinking water) for 2 weeks on the hepatotoxicity and changes in the sulfur amino acid metabolism induced by ANIT treatment. Acute ANIT challenge elevated the serum alkaline phosphatase (ALP), alanine aminotransferase (ALT), aspartate aminotransferase (AST) activities, and total bilirubin contents from 5 h after the treatment, reaching a peak at t = 48-72 h. Hepatic S-adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH) levels were decreased significantly in a manner almost inversely proportional to the changes in serum parameters measured to determine the ANIT-induced toxicity. Hepatic glutathione and cysteine levels were elevated at t = 120 h after the treatment. Betaine supplementation blocked or significantly attenuated induction of the hepatotoxicity by ANIT. The decrease in SAM and SAH levels was also inhibited by betaine intake. The results indicate that betaine supplementation may antagonize the induction of experimental cholestasis and changes in the metabolism of sulfur amino acids associated with ANIT treatment. The underlying mechanism and pharmacological significance of its action are discussed.


Subject(s)
Amino Acids, Sulfur/metabolism , Betaine/pharmacology , Cholestasis/metabolism , Liver/metabolism , 1-Naphthylisothiocyanate/toxicity , Alanine Transaminase/metabolism , Alkaline Phosphatase/metabolism , Analysis of Variance , Animals , Aspartate Aminotransferases/metabolism , Betaine/administration & dosage , Bilirubin/metabolism , Cholestasis/chemically induced , Cholestasis/enzymology , Cysteine/metabolism , Dietary Supplements , Glutathione/metabolism , Kinetics , Liver/drug effects , Liver/enzymology , Male , Mice , Mice, Inbred ICR , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism
12.
J Pharmacol Exp Ther ; 311(1): 99-108, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15169830

ABSTRACT

Decreased glutathione (GSH) levels and gamma-glutamylcysteine ligase (GCL) activity have been observed in diabetic patients, and insulin reportedly increases GSH synthesis via increased GCL catalytic subunit (GCLC) gene expression. The signaling pathways responsible for mediating insulin effects on GCLC expression and GSH levels, however, are unknown. The signaling pathways involved in the regulation of GSH synthesis in response to insulin were examined in primary cultured rat hepatocytes. GSH levels, GCL activity, GCLC protein, and mRNA levels were increased to 140, 160, 600, and 340% of that monitored in untreated cells, respectively, in hepatocytes cultured with 100 nM insulin. The phosphatidylinositol 3-kinase (PI3K) inhibitors, wortmannin and LY294002 [2-(4-morpholinyl)-9-phenyl-4H-1-benzopyran-4-one], dominant-negative Akt, or rapamycin, an inhibitor of mTOR (mammalian target of rapamycin) and ribosomal p70 S6 kinase (p70S6K) phosphorylation, inhibited the insulin-mediated increase in GCLC protein and GSH levels. Although the mitogen-activated protein kinases (MAPKs) extracellular signal-regulated kinase, p38 MAPK, and JNK (c-Jun N-terminal kinase) were activated in response to insulin, PD98059 (2'-amino-3'-methoxyflavone), an inhibitor of mitogen-activated protein kinase kinase, SP600125 (1,9-pyrazoloanthrone), an inhibitor of JNK, and SB203580 [4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imidazole], an inhibitor of p38 MAPK, failed to inhibit the insulin-mediated increase in GCLC protein levels. In conclusion, these data show that insulin signaling pathways involving PI3K/Akt/p70S6K, but not MAPKs, are active in the insulin-mediated regulation of GSH synthesis via increased GCLC expression.


Subject(s)
Dipeptides/metabolism , Hepatocytes/drug effects , Insulin/pharmacology , Ligases/metabolism , Signal Transduction/physiology , Animals , Catalytic Domain/drug effects , Catalytic Domain/physiology , Glutathione/metabolism , Hepatocytes/enzymology , Hepatocytes/metabolism , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Ligases/genetics , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-akt , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
13.
J Pharmacol Exp Ther ; 310(2): 728-36, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15051799

ABSTRACT

Diabetes is characterized by elevated levels of ketone bodies acetoacetate (AA) and 3-hydroxybutyrate (3HB). High levels of ketone bodies have been implicated in generation of cellular oxidative stress. Ketone body activation of cellular signaling pathways associated with oxidative stress, however, has not been established. Thus, ketone body effects on kinase activation in primary cultured rat hepatocytes have been examined. Treatment with AA increased the phosphorylation of extracellular signal-regulated kinase 1/2 (Erk1/2) and p38 mitogen-activated protein kinase (MAPK), maximally by approximately 2.5- and 4-fold, respectively. AA failed to activate c-Jun NH(2)-terminal kinase. AA-mediated Erk1/2 and p38 MAPK activation was detectable at 3 h post-treatment with maximal activation occurring at 12 h. In contrast, 3HB failed to activate any of these kinases. Elevated phosphorylation of Raf and MKK3/6 also occurred in response to AA. Bisindolylmaleimide, a generalized protein kinase C (PKC) inhibitor, and B581, a Ras farnesylation inhibitor, inhibited AA-mediated activation of Erk1/2 and p38 MAPK, suggesting a role for PKC and Ras in mediating such activation. Interestingly, the tyrosine kinase inhibitor genistein prevented the AA-mediated phosphorylation of Erk1/2, but not p38 MAPK. AA treatment resulted in the generation of reactive oxygen species (ROS) and the depletion of cellular glutathione levels, which was ameliorated by the antioxidants N-Acetyl-l-cysteine (NAC) and Trolox (6-hydroxy-2,5,7,8-tetramethyl-chroman-2-carboxylic acid). NAC and Trolox also ameliorated AA-mediated Erk1/2 and p38 MAPK activation, suggesting that this activation is associated with ROS and oxidative stress.


Subject(s)
Acetoacetates/pharmacology , Hepatocytes/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oxidative Stress/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/physiology , Hepatocytes/drug effects , Male , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley
14.
Drug Metab Dispos ; 31(10): 1260-8, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12975336

ABSTRACT

Microsomal epoxide hydrolase (mEH) plays an important role in the detoxification of a broad range of epoxide intermediates and has been reported to be decreased during diabetes and fasting. The signaling pathways involved in the regulation of mEH expression in response to insulin and glucagon were examined in primary cultured rat hepatocytes. mEH protein levels were increased 2- to 6-fold in hepatocytes cultured for 1 to 4 days, respectively, in the presence of insulin. Concentration-response studies revealed that insulin concentrations >or=1 nM resulted in increased mEH protein levels. The phosphatidylinositol 3-kinase (PI3K) inhibitors wortmannin or LY294002 [2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one], and rapamycin, an inhibitor of p70 S6 kinase phosphorylation, ameliorated the insulin-mediated increase in mEH protein levels. The p38 mitogen-activated protein (MAP) kinase inhibitors SB203580 and SB202190 also abrogated the insulin-mediated increase in mEH protein. Treatment of cells with glucagon, 8-bromo-cAMP, or dibutyryl-cAMP for 3 days resulted in decreased mEH protein levels. Pretreatment with the protein kinase A (PKA) inhibitor H89 (N-[2-(4-bromocinnamylamino)ethyl]-5-isoquinoline) prior to glucagon addition markedly attenuated the glucagon effect, implicating PKA signaling in the regulation of mEH expression. These data demonstrate that insulin and glucagon regulate, in an opposing manner, the expression of mEH in primary cultured rat hepatocytes. Furthermore, these data suggest that PI3K and p70 S6 kinase are active in the regulation of insulin-mediated mEH expression. We also provide data implicating p38 MAP kinase in the insulin-mediated increase in mEH levels. Moreover, cAMP and PKA are implicated in mediating the inhibitory effect of glucagon on mEH expression.


Subject(s)
Epoxide Hydrolases/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Glucagon/physiology , Hepatocytes/enzymology , Insulin/physiology , Microsomes, Liver/enzymology , Signal Transduction/physiology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/genetics , Gene Expression Regulation, Enzymologic/physiology , Hepatocytes/drug effects , Hepatocytes/physiology , Male , Microsomes, Liver/drug effects , Microsomes, Liver/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
15.
Biochem Pharmacol ; 65(9): 1565-74, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12732369

ABSTRACT

Alterations of hepatic glutathione level by betaine were observed previously. In this study effects of betaine administration (1000 mg/kg, i.p.) on S-amino acid metabolism in rats and mice were investigated. Hepatic glutathione level decreased rapidly followed by marked elevation in 24 hr. Concentrations of S-adenosylmethionine, S-adenosylhomocysteine, and methionine were increased whereas cystathionine decreased significantly, suggesting that homocysteine generated in the methionine cycle is preferentially remethylated to methionine rather than being utilized for synthesis of cysteine. Hepatic cysteine concentration declined immediately, but plasma cysteine increased. Effect of betaine on hepatic cysteine uptake was estimated from the difference in cysteine concentration in major blood vessels connected to liver. Cysteine concentration either in the portal vein or abdominal aorta was not altered, however, a significant increase was noted in the hepatic vein, indicating that hepatic uptake of cysteine was decreased by betaine treatment. Activities of glutamate cysteine ligase, cystathionine beta-synthase, and cystathionine gamma-lyase were elevated in 24 hr. Pretreatment with propargylglycine, an irreversible inhibitor of cystathionine gamma-lyase, did not abolish the betaine-induced reduction of hepatic glutathione in 4 hr, however, the elevation at t=24 hr was blocked completely. In conclusion the present results indicate that betaine administration induces time-dependent changes on hepatic metabolism of S-amino acids. Betaine enhances metabolic reactions in the methionine cycle, but inhibits cystathionine synthesis and cysteine uptake, leading to a decrease in supply of cysteine for glutathione synthesis. Reduction in glutathione is subsequently reversed due to induction of cysteine synthesis and glutamate cysteine ligase activity.


Subject(s)
Betaine/administration & dosage , Dioxygenases , Glycine/analogs & derivatives , Liver/drug effects , S-Adenosylmethionine/metabolism , Alkynes/pharmacology , Amino Acids/metabolism , Animals , Betaine/pharmacology , Biological Transport/drug effects , Cystathionine/metabolism , Cystathionine gamma-Lyase/metabolism , Cysteine Dioxygenase , Enzyme Inhibitors/pharmacology , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Glycine/pharmacology , Liver/metabolism , Methionine/metabolism , Mice , Mice, Inbred ICR , Oxygenases/metabolism , Rats , Rats, Sprague-Dawley , S-Adenosylhomocysteine/metabolism , Taurine/metabolism , gamma-Glutamyltransferase/metabolism
16.
J Pharmacol Exp Ther ; 305(1): 353-61, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12649389

ABSTRACT

Diabetes is a major cause of morbidity and mortality, and complications resulting from diabetes have been attributed in part to increased oxidative stress. Glutathione S-transferases (GSTs) constitute a major protective mechanism against oxidative stress. Studies of the expression and activity of GSTs during diabetes are inconclusive, with both increased and decreased GST expression being reported in vivo. Insulin and glucagon effects on GST expression and the signaling pathway involved in the glucagon regulation of GST expression were examined in primary cultured rat hepatocytes. The addition of insulin resulted in the elevation of alpha-class GST protein levels, whereas alpha- and pi-class GST protein levels were markedly decreased in hepatocytes cultured with glucagon. In contrast, mu-class GST protein expression was unaffected by insulin or glucagon treatment. Insulin concentrations >/=1 nM resulted in increased GST activities and alpha-class GST protein levels, whereas glucagon concentrations >/=20 nM decreased alpha- and pi-class protein levels and activity. Treatment of cells with 8-bromo-cAMP or dibutyryl-cAMP also resulted in decreased alpha- and pi-class GST protein levels. Pretreatment with N-[2-(4-bromocinnamylamino)ethyl]-5-isoquinoline sulfonamide (H89), a selective inhibitor of protein kinase A, before glucagon addition markedly attenuated the glucagon effect. This study demonstrates that insulin and glucagon regulate, in an opposing manner, the expression of alpha-class GSTs and that glucagon completely inhibits pi-class GST expression in vitro, suggesting that hepatic GST expression may be decreased during diabetes. Furthermore, the present study implicates cAMP and protein kinase A in mediating the inhibitory effect of glucagon on GST expression.


Subject(s)
Gene Expression Regulation, Enzymologic/drug effects , Glucagon/pharmacology , Glutathione Transferase/metabolism , Hepatocytes/drug effects , Insulin/pharmacology , Sulfonamides , Animals , Cells, Cultured , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors , Glutathione Transferase/genetics , Hepatocytes/enzymology , Isoquinolines/pharmacology , Male , Rats , Rats, Sprague-Dawley
17.
Life Sci ; 72(10): 1171-81, 2003 Jan 24.
Article in English | MEDLINE | ID: mdl-12505547

ABSTRACT

The changes in amino acid concentrations and transsulfuration enzyme activities in liver were investigated after 4-week fed on 23% casein diet (control group) and 5% casein diet without (protein-calorie malnutrition, PCM group) or with (PCMC group) oral administration of cysteine, 250 mg/kg (twice daily, starting from the fourth week) using rats as an animal model. By supplementation with cysteine in PCM rats (PCMC group), cysteine level was elevated almost close to the control level, and glutathione (GSH), aspartic acid and serine levels were restored greater than the control levels. The measurement of transsulfuration enzyme activities exhibited that gamma-glutamylcysteine ligase (gamma-GCL) activity was up-regulated in rats with protein restriction (PCM group), and cysteine supplementation (PCMC group) down-regulated to the control level. One-week supplementation of cysteine (PCMC group) significantly down-regulated the cysteine sulfinate decarboxylase activity. These results indicate that the availability of sulfur amino acid(s) especially cysteine appears to play a role in determining the flux of cysteine between cysteine catabolism and GSH synthesis.


Subject(s)
Amino Acids/metabolism , Cysteine/pharmacology , Dioxygenases , Liver/enzymology , Protein-Energy Malnutrition/enzymology , Sulfur/metabolism , Animals , Carboxy-Lyases/metabolism , Cystathionine beta-Synthase/metabolism , Cysteine Dioxygenase , Cytosol/drug effects , Cytosol/enzymology , Diet , Eating/drug effects , Glutamate-Cysteine Ligase/metabolism , Male , Organ Size/drug effects , Oxygenases/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Weight Gain/drug effects
18.
Food Chem Toxicol ; 40(4): 545-9, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11893413

ABSTRACT

The effects of betaine or taurine on hepatotoxicity induced by lipopolysaccharide (LPS) were examined in adult male SD rats. Rats were provided with drinking water containing either 1% betaine or taurine for 2 weeks prior to challenge with LPS (5 mg/kg, iv). Supplementation with betaine or taurine protected the animals from induction of LPS hepatotoxicity as measured by changes in aspartate aminotransferase (AST), alanine aminotransferase (ALT) activities and total bilirubin levels in serum, and hepatic glutathione contents. LPS challenge increased serum TNF-alpha and nitrate/nitrite in rats, which were reduced by betaine or taurine intake. Taurine depletion induced by supply of drinking water containing 3% beta-alanine for 7 days did not enhance the LPS-induced hepatic damage or the decrease in hepatic glutathione level. The results indicate that intake of betaine or taurine attenuates the LPS-induced hepatotoxicity resulting from activation of Kupffer cells.


Subject(s)
Betaine/pharmacology , Gastrointestinal Agents/pharmacology , Lipopolysaccharides/toxicity , Liver/pathology , Taurine/pharmacology , Administration, Oral , Alanine Transaminase/pharmacology , Animals , Aspartate Aminotransferases/pharmacology , Bilirubin/blood , Kupffer Cells , Liver/drug effects , Liver/enzymology , Male , Rats , Rats, Sprague-Dawley
19.
Toxicol Lett ; 129(1-2): 107-14, 2002 Mar 24.
Article in English | MEDLINE | ID: mdl-11879980

ABSTRACT

Dichloromethane (DCM) elimination and carboxyhemoglobin (COHb) generation were examined in adult female SD rats pretreated with a glutathione (GSH) depletor(s). Rats were treated with either buthionine sulfoximine (BSO; 2 mmol/kg, i.p.), diethylmaleate (DEM; 3 mmol/kg, i.p.), phorone (PHO; 1 mmol/kg, i.p.) or BSO plus PHO (BSO; 2 mmol/kg +PHO; 0.5 mmol/kg, i.p.). The hepatic GSH concentration was significantly reduced by each treatment. Decrease in hepatic GSH was maintained at least for 10 h after BSO treatment but recovered rapidly in rats treated with DEM or PHO. The hepatic p-nitrophenol hydroxylase activity was not affected by the GSH depletors at the dose used in this study. Rats were treated with an i.p. injection of DCM (3 mmol/kg) and the concentrations of DCM and the COHb levels in blood were monitored. In rats pretreated with a GSH depletor, the peak COHb level was significantly greater than that of rats treated with DCM only. The peak COHb level attained in each group of rats appeared to be inversely related to the magnitude of reduction in hepatic GSH levels. The half-life of DCM in blood was also increased in rats pretreated with the GSH depletor(s). The results indicate that the GSH-dependent metabolic reaction has an important role in the overall elimination of DCM as well as in the metabolic generation of carbon monoxide (CO) from this solvent.


Subject(s)
Glutathione/physiology , Methylene Chloride/metabolism , Animals , Buthionine Sulfoximine/pharmacology , Carbon Monoxide/metabolism , Carboxyhemoglobin/metabolism , Cytochrome P-450 CYP2E1/physiology , Female , Glutathione/analysis , Half-Life , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...