Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sang Geon Kim is active.

Publication


Featured researches published by Sang Geon Kim.


Biochemical Pharmacology | 1997

Inhibition of cytochrome P450 2E1 expression by 2-(allylthio)Pyrazine, a Potential chemoprotective agent: hepatoprotective Effects

Nak Doo Kim; Mi Kyong Kwak; Sang Geon Kim

Cytochrome P450 2E1 (P450 2E1) is active in both the detoxification and activation of small organic molecules. The effects of 2-(allylthio)pyrazine (2-AP) on P450 2E1-catalytic activity and the expression of rat hepatic P450 2E1 were examined. 2-AP competitively inhibited 4-nitrophenol hydroxylase activity in vitro (Ki, 12 microM). 2-AP treatment of rats (200 mg/kg/day, p.o., 1-3 days old) resulted in 20-30% decreases in the rates of P450 2E1-specific metabolic activities. Immunoblot analysis also revealed that hepatic microsomes isolated from 2-AP-treated rats showed substantial decreases in P450 2E1 level. 2-AP-suppressed isoniazid (INH)-inducible hepatic P450 2E1 levels, as shown by both metabolic activities and immunoblot analyses. Thus, 2-AP was effective in suppressing both constitutive and inducible P450 2E1 expression. Northern blot analysis showed that 2-AP transiently suppressed the hepatic P450 2E1 mRNA level, suggesting that suppression in P450 2E1 expression by 2-AP may be mediated in part by transcriptional inactivation. Hepatoprotective effects of 2-AP against toxicants were monitored in mice. 2-AP pretreatment prior to the administration of lethal doses of acetaminophen (AAP) or INH substantially reduced toxicant-induced mortality. Whereas serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels were markedly elevated after AAP administration (i.e. 9-20-fold), 2-AP pretreatment of animals before AAP administration resulted in >95% decreases in elevated serum aminotransferase activities. 2-AP was also effective against CCl4-induced hepatotoxicity. Whereas CCl4 treatment caused 35-70-fold increases in aminotransferase activities, treatment of mice with 2-AP (>10 mg/kg) resulted in the blocking of CCl4-induced liver toxicity. The hepatoprotective effect of 2-AP was in part due to 2-AP-induced elevation of hepatic GSH levels. Whereas AAP or CCl4 treatment resulted in 70-80% reduction in hepatic GSH levels, pretreatment of mice with 2-AP caused a 40-210% elevation in hepatic GSH levels, as compared with either AAP or CCl4 alone. 2-AP pretreatment also reduced AAP- or CCl4-induced increases in lipid peroxidation in a dose-dependent manner. The results of these metabolic activities and of immunoblot and RNA blot analyses demonstrate that 2-AP is efficacious in suppressing constitutive and inducible P450 2E1 expression and effective in protecting against toxicant-induced liver toxicity.


Biochemical Pharmacology | 1994

Inhibition of cytochrome P4502E1 expression by organosulfur compounds allylsulfide, allylmercaptan and allylmethylsulfide in rats

Mi Kyong Kwak; Sang Geon Kim; Ji Yeon Kwak; Raymond F. Novak; Nak Doo Kim

Cytochrome P4502E1 (CYP2E1) is active in both detoxication and activation of small organic molecules. The effects of organosulfur compounds including allylsulfide (AS), allylmercaptan (AM) and allylmethylsulfide (AMS) on the expression of CYP2E1 were examined in rats. 4-Nitrophenol, aniline hydroxylase and N-nitrosodimethylamine demethylase activities, the rates of which represent the level of CYP2E1, decreased in hepatic microsomes isolated from rats treated with AS in a time-dependent manner by 45% to 90%, as compared to control. Pyrazine-induced hepatic microsomes exhibited approximately 5-fold increases in CYP2E1-catalysed metabolic activities, whereas the hepatic microsomes obtained after treatment of animals with both AS and pyrazine showed rates comparable to or less than those in control microsomes. AM or AMS suppressed constitutive and pyrazine-inducible levels of CYP2E1 similarly to AS. Immunoblot analyses of hepatic microsomes, using an anti-CYP2E1 antibody, showed that AS, AM and AMS significantly suppressed constitutive levels of CYP2E1 apoprotein after 24, 48 and 72 hr. Time-dependent induction of CYP2E1 by pyrazine was also completely blocked by treatment of animals with AS throughout the experimental period, as evidenced by immunoblot analysis. The levels of CYP2E1 apoprotein in the hepatic microsomes isolated from animals treated with both AM and pyrazine, or with both AMS and pyrazine were comparable to those in control hepatic microsomes at days 1-3 post-treatment. Treatment of rats with each of these organosulfur compounds caused no significant changes in the levels of CYP2E1 mRNA, as assessed by slot and northern blot analyses, suggesting that post-transcriptional regulation may be associated with the suppression of CYP2E1 apoprotein levels. The results of metabolic activities, immunoblot analyses and RNA blot analyses demonstrated that these organosulfur compounds are effective in suppressing constitutive and inducible expression of CYP2E1.


Biochemical Pharmacology | 1996

Expression of glutathione S-transferases Ya, Yb1, Yb2, Yc1 and Yc2 and microsomal epoxide hydrolase genes by thiazole, benzotbiazole and benzothiadiazole☆

Sang Geon Kim; Min Kyung Cho

The effects of thiazole (TH), benzothiazole (BT) and benzothiadiazole (BZ) on the expression of hepatic glutathione S-transferases (GSTs) Ya, Yb1, Yb2, Yc1 and Yc2 and microsomal epoxide hydrolase (mEH) genes were compared in rats. TH treatment resulted in 4- to 24-fold increases in GST Ya mRNA levels at 24 hr posttreatment; the ED50 value was 70 mg/kg. GST Ya mRNA levels were elevated 13-, 20-, 20- and 9-fold at 12, 24, 48 and 72 hr following 100 mg/kg of TH treatment, respectively, as compared with the control. BT was a moderate inducer of GST Ya with a maximal 18-fold increase observed, whereas BZ treatment caused a transient increase in GST Ya mRNA at 12 hr posttreatment, followed by a return to a 4-fold relative increase at 24 hr or afterward. Treatment of rats with TH at the dose of 100 mg/kg resulted in an approximately 10-fold increase in either Yb1 or Yb2 mRNA levels at 24 hr posttreatment. BT-treated rats showed 7- and 3-fold increases in the GST subunit Yb1 and Yb2 mRNA levels at 24 hr posttreatment. BZ was the least effective in modulating either GST Yb1 or Yb2 mRNA, resulting in < 2-fold changes. GST Yc1 and Yc2 mRNA levels were increased approximately 8-fold at the dose of 200 mg/kg of TH. BT minimally affected GST subunit Yc1 and Yc2 mRNA levels, with a maximal 4-fold relative increase observed. BZ was the least effective in enhancing Yc1 and Yc2 mRNA levels. Protein levels for GST subunit Ya, Yb1, Yb2 and Yc were also elevated in response to TH by 3-, 2-, 2-, and 2-fold, respectively. Thus, TH was effective in modulating both constitutive and inducible GST gene expression. BT or BZ was much less effective in increasing the expression of GST subunits. These RNA and Western blot analyses revealed that the levels of major GST were differentially increased after treatment with these thiazoles, exhibiting a rank order of GST expression of TH > BT > BZ. mEH expression by these compounds appeared to be consistent with that of GST Ya. The mRNA levels for GST Ya, Yb1, Yb2, Yc1 and Yc2 and mEH were also determined after treatment with triazole (TR), imidazole (IM), benzoxazole (BX), benzotriazole (BTR) or benzimidazole (BIM). TR, IM, BX or BTR caused increases in Ya, Yb1, Yc1 and Yc2 mRNA levels by 2- to 3-fold, whereas the agents failed to modulate the expression of GST Yb2. The fact that benzene, cyclohexane or n-hexane minimally affected the major GST or mEH mRNA levels provided evidence that certain heterocyclic compounds are more capable of modulating GST or mEH gene expression than hydrocarbons. These results corroborate evidence that the thiazoles differentially stimulate GST or mEH genes, with TH being the most efficacious; that thiazoles with carbocyclic ring are much less effective in increasing GST or mEH levels than is TH; and that the changes in these GST and mEH levels are primarily associated with increases in mRNA levels.


Biochemical Pharmacology | 1998

Lipopolysaccharide inhibition of rat hepatic microsomal epoxide hydrolase and glutathione S-transferase gene expression irrespective of nuclear factor-κb activation

Sung Hee Choi; Sang Geon Kim

Lipopolysaccharide (LPS) is an endotoxin involved in septic shock syndrome and potentiates toxicant-induced liver injury. The effects of LPS on the constitutive and inducible expression of hepatic microsomal epoxide hydrolase (mEH) and glutathione S-transferase (GST) genes were studied in rats. Northern blot analysis showed that treatment of rats with LPS caused suppression in mEH and GST gene expression. The mEH mRNA level was decreased in a time-dependent manner following a single dose of LPS (1 mg/kg, i.v.), resulting in levels of 52%, 22%, 17%, and 94% of those in untreated animals at 2, 6, 12, and 24 hr, respectively. The levels of rGSTA2 and rGSTA3 mRNA were suppressed in response to an LPS injection to the similar extents as observed in mEH mRNA, whereas rGSTM1 and rGSTM2 mRNA levels were less affected. LPS inhibited mEH gene expression at the doses of 1 microg or greater. Whereas treatment of rats with allyl disulfide (ADS), oltipraz (OZ) or pyrazine (PZ) at the dose of 50 mg/kg caused increases in the mEH mRNA level at 12 hr, a concomitant LPS injection (1 mg/kg) resulted in 80%-95% suppression of the inducible gene expression. The inducible rGSTA2, rGSTA3, rGSTM1, and rGSTM2 mRNA levels were also 50%-90% decreased at 12 hr after LPS treatment, with the relative change in rGSTA being greater than that in rGSTM. Three consecutive daily treatments with LPS (10 microg/kg/day) resulted in significant decreases of the constitutive and PZ (50 mg/kg/day, i.p. for 3 days)-inducible mEH and GST mRNA levels, which were consistent with those in the protein levels. Gel shift retardation analysis showed that LPS substantially activated the hepatic nuclear p65/p50 nuclear factor-kappaB (NF-kappaB) complex with the maximal effect observed at 1 hr at the doses of 1 microg/kg or greater. LPS-induced activation of nuclear NF-kappaB (1 microg/kg, i.v.) failed to be inhibited by concomitant treatment with the mEH and GST inducers, including ADS (300 mg/kg, p.o.), OZ (300 mg/kg, p.o.), and PZ (300 mg/kg, i.p.), indicating that NF-kappaB activation was not required for suppression of the gene expression by LPS. In contrast, GdCl3, an inhibitor of mEH and GST expression, inhibited LPS-induced activation of the p65/p50 NF-kappaB. These gel shift analyses provided evidence that LPS-induced activation of the NF-kappaB was not responsible for alterations in the gene expression. In summary, the results of this research demonstrate that LPS effectively inhibits constitutive and inducible mEH and GST expression with decreases in their mRNA levels, and that LPS suppression in the expression of the detoxifying enzymes is not mediated with its activation of NF-kappaB.


Xenobiotica | 1995

Effects of garlic oil on rat hepatic P4502E1 expression

Mi Kyong Kwak; Sang Geon Kim; Nak Doo Kim

1. The effects of garlic oil (GO) on the expression of P4502E1, glutathione S-transferase (GST) and microsomal epoxide hydrolase (mEH) were assessed by metabolic activities, immunoblot and RNA blot analyses in the rat. 2. p-Nitrophenol (PNP) hydroxylase activity decreased in the hepatic microsomes isolated from rats treated with GO at 200 mg/kg b.w. by 10-30% as compared with control. Pyrazine-inducible P4502E1 expression was decreased by approximately 40% following concomitant treatment of animals with GO at the dose of 200 mg/kg from day 1 to 3 post-treatment, as evidenced by PNP hydroxylase activity. The rates of aniline hydroxylase and NDMA demethylase activities in GO-treated animals were consistent with those of PNP hydroxylase activity. Treatment of animals with 500 mg/kg GO resulted in suppression of P4502E1-mediated catalytic activities, as monitored by both PNP and aniline hydroxylase activities, whereas the effects at the dose of 1000 mg/kg were identical with those at 500 mg/kg b.w. 3. Immunoblot analyses of hepatic microsomes, using an anti-P4502E1 antibody, showed that GO minimally suppressed constitutive P4502E1 expression at 24, 48 and 72 h post-treatment at the daily doses from 200 to 1000 mg/kg b.w., as compared with vehicle-treated animals. Time-dependent pyrazine induction of P4502E1, however, was substantially blocked by concomitant treatment of animals with 200 mg/kg GO to the levels of control. Treatment at the dose of 1000 mg/kg failed to further suppress P4502E1 levels. GO treatment caused no changes in the levels of P4502E1 mRNA, as assessed by slot blot analyses. 4. Cytosol produced from the GO-treated rat showed approximately 40% increases in GST conjugating activity toward 1-chloro-2,4-dinitrobenzene, whereas mEH protein levels were 1.5-2.0-fold greater than control with similar increases in the mRNA levels noted. 5. These results demonstrate that GO suppresses inducible P4502E1 expression more significantly than constitutive expression, and that GO induces GST and mEH expression to a certain extent.


Xenobiotica | 1994

Piperine effects on the expression of P4502E1, P4502B and P4501A in rat

Min Hee Kang; Sun Me Won; Sang Shin Park; Sang Geon Kim; Raymond F. Novak; Nak Doo Kim

1. Treatment of rat with piperine (PIP) (1.4 mmol/kg, 3 days ip injections) resulted in an approximate two-fold increase in total liver microsomal P450 content relative to that in uninduced animals. 2. 4-Nitrophenol and aniline hyroxylase activities in the hepatic microsomes prepared from rat treated with PIP decreased by 30 and 28% respectively as compared with control. Immunoblot analyses also revealed decreased P4502E1 levels in hepatic microsomes from PIP-treated animals. 3. In contrast with P4502E1 suppression, hepatic 2B1 and 2B2 levels were significantly increased in PIP-induced animals, as evidence by both metabolic activity and immunoblot analysis of the liver microsomal fractions. The rate of hexobarbital hydroxylase activity in microsomes from PIP-treated animals was markedly elevated and was inhibited by approximately 62% in the presence of monoclonal anti-P4502B IgG. Immunoblot analyses demonstrated that P4502B1 and 2B2 levels in hepatic microsomes from PIP-treated animals were comparable with those from phenobarbital-treated animals. 4. 7-Ethoxycoumarin deethylase activity was elevated approximately two-fold in PIP-induced animals and was 17% of that derived from 3-methylcholanthrene-induced animals. 7-ethoxycoumarin deethylase activity in PIP-induced hepatic microsomes was inhibited 63% in the presence of monoclonal anti-P4501A antibody. Immunoblot analysis confirmed the increase in P4501A levels by PIP, which was 15% of that in hepatic microsomes from 3-methylcholanthrene-induced animals. 5. PIP treatment failed to affect microsomal epoxide hydrolase (mEH) and glutathione S-transferases (GST) expression, as indicated by immunoblot analyses using polyclonal antibodies toward mEH and GST subunits Ya, Yb1, Yb2 and Yc. 6. These results demonstrate that PIP treatment suppressed P4502E1 expression and enhanced 2B and 1A expression, whereas this agent failed to affect hepatic mEH and GST expression.


Biochemical Pharmacology | 1994

Enhanced expression of rat microsomal epoxide hydrolase gene by organosulfur compounds

Nak Doo Kim; Sang Geon Kim; Mi Kyong Kwak

The effects of organosulfur compounds including allylsulfide (AS), allylmercaptan (AM) and allylmethylsulfide (AMS) on the expression of microsomal epoxide hydrolase (mEH) protein and its mRNA were examined in rats. The levels of mEH induction were examined with or without concomitant treatment of animals with pyrazine, a strong inducer of mEH, in order to establish whether a common molecular basis exists for mEH induction between these structurally different xenobiotics. Immunoblot analyses using anti-rat mEH antibody showed that treatment with AS caused an approximately 4-fold increase in hepatic mEH protein levels relative to controls whereas treatment with both AS and pyrazine resulted in only minimal additive increases in the elevation of mEH. Administration of AM to rats resulted in a comparable increase in mEH levels to that caused by AS, whereas an approximately 2-fold increase was noted after AMS treatment, as compared to control. mEH levels in the hepatic microsomes isolated from animals treated with both AMS and pyrazine were, however, approximately 50% less than those from pyrazine-treated rats. Thus, AS and AM appeared to be more effective than AMS in elevating mEH, as evidenced by immunoblot analyses. The levels of mEH mRNA were increased 10-16-fold following treatment with either AS or AM, while AMS caused a 3-7-fold increase relative to control, as assessed by slot blot analysis probed with a 1.3 kb mEH cDNA. Time-dependent increases in mRNA levels by each of these organosulfur compounds were consistent with those in mEH protein levels at 3 days. A marginal additive increase in mEH mRNA levels was noted following co-administration of either AS or AM with pyrazine, whereas treatment with both AMS and pyrazine decreased mEH mRNA levels by 55%. Significant mEH mRNA increases in poly(A)+ RNA fractions were confirmed by northern blot analysis. The results demonstrate that these organosulfur compounds are inducers of mEH and that the induction involves increases in its mRNA.


Biochemical Pharmacology | 1998

In Vivo Radioprotective Effects of Oltipraz in γ-Irradiated Mice

Sang Geon Kim; Seon Young Nam; Choon Won Kim

Abstract Previous studies in this laboratory have shown that oltipraz (Olt), a chemopreventive agent, enhances radiation(Rad)-inducible glutathione S-transferase (GST) and microsomal epoxide hydrolase (mEH) expression in the liver. The present study was designed to investigate the in vivo radioprotective effect of Olt in ICR mice exposed to a lethal dose of Rad. The 30-day survival rate of mice irradiated at the dose of 8 Gy was substantially increased to 91% by Olt pretreatment (100 mg/kg/day for 2 days), compared with 48% in animals irradiated alone. Light microscopic examinations revealed that exposure of mice to 8 Gy of γ-ray Rad resulted in hepatocyte degeneration in the surviving animals from Day 1 through Day 22 after irradiation with certain degrees of necrosis observed at early times, whereas Olt treatment provided protection of the liver against irradiation with no hepatic necrosis noted. Mice irradiated at the dose of 8 Gy exhibited time-related decreases in the white blood cell (WBC), red blood cell (RBC), and platelet counts with maximal reduction noted at Day 10. Animals irradiated with Olt treatment showed no difference in peripheral blood cell counts or in the ratio of myeloid to erythroid bone marrow cells, compared with those irradiated alone. Northern RNA blot analysis showed that treatment of mice with Olt at the dose of 100 mg/kg in combination with 8 Gy irradiation resulted in 12-fold increases in hepatic mEH and mGSTA3 mRNA levels at 24-hr post-treatment, whereas mGSTP1 mRNA levels were not altered. The mRNA levels for mEH and mGSTA3 were elevated after exposure of animals to both Olt and 8 Gy-γ ray to a greater extent than after irradiation alone. The enhanced survival rate (91%) in 8 Gy-irradiated animals after treatment with Olt (100 mg/kg/day for 2 days) was completely reversed by concomitant pretreatment with dexamethasone (Dexa) (0.1 and 1 mg/kg/day for 2 days), a known inhibitor of mEH and GST expression, resulting in a 42% and 28% survival rate, respectively. Mice irradiated after dexamethasone treatment at a dose of 1 mg/kg showed a reduced mean survival time compared with those treated with 0.1 mg/kg of dexamethasone (9 vs 14 days). The current study demonstrates that Olt is effective in increasing the survival rate of mice against ionizing Rad and that protective effects of Olt associated with enhanced expression of mEH and GST genes may represent its radioprotective efficacy.


Biochemical Pharmacology | 1998

Correlation of increased mortality with the suppression of radiation-inducible microsomal epoxide hydrolase and glutathione S-transferase gene expression by dexamethasone: effects on vitamin C and E-induced radioprotection

Seon Young Nam; Chul Koo Cho; Sang Geon Kim

Previous studies in this laboratory have shown that gamma-ray ionizing radiation in combination with oltipraz, a radioprotective agent, enhances hepatic microsomal epoxide hydrolase (mEH) and glutathione S-transferase (GST) expression. The present study was designed to investigate the effects of dexamethasone on the radiation-inducible expression of mEH and rGST genes and on the vitamin C and E-induced radioprotective effects in association with the expression of the genes. Treatment of rats with a single dose of dexamethasone (0.01-1 mg/kg, p.o.) caused a dose-dependent decrease in the constitutive mEH gene expression at 24 hr. The radiation-inducible mEH mRNA level (threefold increase after 3 Gy gamma-irradiation) was decreased by 21% and 88% by dexamethasone at the doses of 0.1 and 1 mg/kg, respectively. Although dexamethasone alone caused 2- to 5-fold increases in the hepatic rGSTA2 mRNA level, rats treated with dexamethasone prior to 3 Gy irradiation exhibited 80%-93% suppression in the radiation-inducible increases in the rGSTA2 mRNA level. The inducible rGSTA3 and rGSTA5 mRNA levels were also significantly decreased by dexamethasone, whereas the rGSTM1 mRNA level was reduced to a lesser extent. Vitamin C and/or E, however, failed to enhance the radiation-inducible increases in hepatic mEH and rGST mRNA levels. Whereas rats exposed to 3 Gy irradiation with or without vitamin C treatment (30 or 200 mg/kg/day, p.o., 2 days) exhibited approximately threefold increases in the mEH and rGSTA2/3/5 mRNA levels relative to untreated animals, dexamethasone treatment (1 mg/kg, p.o.) resulted in 64%-96% decreases in the mRNA levels at 24 hr. The inducible rGSTM1/2 mRNA levels in the vitamin C/E-treated rats were approximately 50% suppressed by dexamethasone. Although vitamin C and/or E treatment (200 mg/kg/day, p.o., 2 days) improved the 30-day survival rates of the 8 Gy gamma-irradiated mice from 39% up to 74%, the improved survival rate of gamma-irradiated animals was reduced to 30% by dexamethasone pretreatment (1 mg/kg/day, 2 days). The mean survival time of dexamethasone-treated animals was reduced to approximately 2 days from 14 days in the animals with total body irradiation alone. No significant hematologic changes were observed in mice at 10 days after dexamethasone plus gamma-irradiation, as compared with irradiation alone. These results demonstrate that: dexamethasone substantially suppresses radiation-inducible mEH, rGSTA and rGSTM expression in the liver; vitamins C/E exhibit radioprotective effects without enhancing radiation-inducible mEH and GST gene expression; and inhibition of radiation-inducible mEH and rGST gene expression in the vitamin C- and E-treated animals by dexamethasone was highly correlated with reduction in the survival rate and the mean survival time of gamma-irradiated animals.


Journal of Pharmacy and Pharmacology | 1995

Enhanced Effectiveness of Dimethyl‐4,4‘‐dimethoxy‐5,6,5’,6‘‐dimethylene dioxybiphenyl‐2,2’‐dicarboxylate in Combination with Garlic Oil against Experimental Hepatic Injury in Rats and Mice

Sang Geon Kim; Seon Young Nam; Hye Chin Chung; Su Yeun Hong; Ki Hwa Jung

The present study was designed to evaluate the effects of dimethyl‐4,4′‐dimethoxy‐5,6,5′,6′‐dimethylene dioxybiphenyl‐2,2′‐dicarboxylate (PMC) in combination with garlic oil against chemical‐induced hepatic injury in rats and mice.

Collaboration


Dive into the Sang Geon Kim's collaboration.

Top Co-Authors

Avatar

Nak Doo Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Seon Young Nam

Duksung Women's University

View shared research outputs
Top Co-Authors

Avatar

Mi Kyong Kwak

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Choon Won Kim

Duksung Women's University

View shared research outputs
Top Co-Authors

Avatar

Chul Koo Cho

Duksung Women's University

View shared research outputs
Top Co-Authors

Avatar

Jong-Wook Lee

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Joo Youn Cho

Duksung Women's University

View shared research outputs
Top Co-Authors

Avatar

Ki Hwa Jung

Duksung Women's University

View shared research outputs
Top Co-Authors

Avatar

Tae Gil Ha

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge