Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Biofactors ; 38(1): 34-43, 2012.
Article in English | MEDLINE | ID: mdl-22253071

ABSTRACT

Typically, chemopreventive agents involve either induction of phase II detoxifying enzymes and/or inhibition of cytochrome P450 enzymes (CYPs) that are required for the activation of procarcinogens. In this study, we investigated the protective effects of phloretin against aflatoxin B1 (AFB1) activation to the ultimate carcinogenic intermediate, AFB(1)-8, 9-epoxide (AFBO), and its subsequent detoxification. Phloretin markedly inhibited formation of the epoxide with human liver microsomes in a dose-dependent manner. Phloretin also inhibited the activities of nifedipine oxidation and ethoxyresorufin O-deethylase (EROD) in human liver microsomes. These data show that phloretin strongly inhibits CYP1A2 and CYP3A4 activities, which are involved in the activation of AFB1. Phloretin increased glutathione S-transferase (GST) activity of alpha mouse liver 12 (AML 12) cells in a dose-dependent manner. GST activity toward AFBO in cell lysates treated with 20 µM phloretin was 23-fold that of untreated control cell lysates. The expression of GSTA3, GSTA4, GSTM1, GSTP1 and GSTT1 was induced by phloretin in a dose-dependent manner in AML 12 cells. GSTP1, GSTM1, and GSTT1 were able to significantly increase the conjugation of AFBO with glutathione. Concurrently, induction of the GST isozyme genes was partially associated with the Nrf2/ARE pathway. Taken together, the results demonstrate that phloretin has a strong chemopreventive effect against AFB1 through its inhibitory effect on CYP1A2, CYP3A4, and its inductive effect on GST activity.


Subject(s)
Aflatoxin B1/pharmacokinetics , Phloretin/pharmacokinetics , Animals , Cell Line , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP1A2 Inhibitors , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors , Enzyme Induction/drug effects , Genes, Reporter , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Inactivation, Metabolic , Isoenzymes/genetics , Isoenzymes/metabolism , Luciferases/biosynthesis , Luciferases/genetics , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Phloretin/pharmacology , Response Elements
2.
Pharmacol Rep ; 63(3): 708-16, 2011.
Article in English | MEDLINE | ID: mdl-21857081

ABSTRACT

Cisplatin is a highly effective chemotherapeutic agent, but it has significant ototoxic side effects. Apoptosis is an important mechanism of cochlear hair cell loss following exposure to cisplatin. The present study examined the effects of phloretin, a natural polyphenolic compound found in apples and pears, on cisplatin-induced apoptosis. We found that phloretin induced the expression of heme oxygenase-1 (HO-1) protein in a concentration- and time-dependent manner. Phloretin induced nuclear factor-E2-related factor 2 (Nrf2) nuclear translocation, and dominant-negative Nrf2 attenuated phloretin-induced expression of HO-1. Phloretin activated the JNK, ERK and p38 mitogen-activated protein kinase (MAPK) pathways, and the JNK pathway played an important role in phloretin-induced HO-1 expression. Phloretin protected the cells against cisplatin-induced apoptosis. The protective effect of phloretin was abrogated by zinc protoporphyrin IX (ZnPP IX), a HO inhibitor. Furthermore, phloretin pretreatment inhibited mitochondrial dysfunction and the activation of caspases. These results demonstrate that the expression of HO-1 induced by phloretin is mediated by both the JNK pathway and Nrf2; the expression inhibits cisplatin-induced apoptosis in HEI-OC1 cells.


Subject(s)
Apoptosis/drug effects , Cisplatin/toxicity , Organ of Corti/drug effects , Phloretin/pharmacology , Animals , Antineoplastic Agents/toxicity , Cell Line , Dose-Response Relationship, Drug , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase-1/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , NF-E2-Related Factor 2/metabolism , Phloretin/administration & dosage , Time Factors
3.
Biofactors ; 36(4): 289-96, 2010.
Article in English | MEDLINE | ID: mdl-20818711

ABSTRACT

The aflatoxin B(1)-8,9-epoxide (AFBO) is hepatocarcinogenic intermediate of aflatoxin B(1) (AFB(1)) and is detoxified by glutathione S-transferases (GSTs). In this study, we investigated whether sulforaphane (SFN) could increase the rate of conjugation between AFBO and glutathione (GSH) as well as which of the GST isozymes were involved in the conjugation reaction. The conjugation potential was inhibited dose dependently with curcumin, an inhibitor of GSTs. SFN induced the expression of GST A3, GST A4, GST M1, GST P1, and GST T1 in alpha mouse line (AML) 12 cells. The cells treated with SFN (10 microM) for 12 h showed a 35-fold increase in conjugation potential of AFBO with GSH compared with the vehicle-treated cell. The conjugation potential was blocked partially by transfection of cells with siRNAs against each of the GST isozymes. The activity of GST A3 had the strongest effect on the conjugation potential. SFN treatment also increased total GST activity detected with 1-chloro-2,4-dinitrobenzene (CDNB) up to 4.3-fold. The induction fold was much lower than that detected with AFBO. These results suggest that the chemopreventive effect of SFN on the decomposition of AFBO is related to the upregulation of several GST isozymes genes. The increase of GST activity by SFN was extremely specific toward the conjugation reaction of AFBO compared with CDNB. Therefore, this system for detecting GST activity seems to be an excellent method for screening chemopreventive compounds toward AFB(1) toxicity.


Subject(s)
Aflatoxin B1/analogs & derivatives , Anticarcinogenic Agents/pharmacology , Glutathione Transferase/metabolism , Thiocyanates/pharmacology , Aflatoxin B1/metabolism , Animals , Cells, Cultured , Chemoprevention , Dinitrochlorobenzene/chemistry , Inactivation, Metabolic , Isoenzymes/metabolism , Isothiocyanates , Mice , Sulfoxides , Transfection
4.
Pharm Res ; 27(2): 235-45, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19937094

ABSTRACT

PURPOSE: The present study was undertaken to elucidate the chemoprotective mechanism of kaempferol, which possesses anti-oxidative and anti-apoptotic properties. METHODS: House Ear Institute-Organ of Corti 1 (HEI-OC1) cells were treated with kaempferol in the presence or absence of cisplatin. Cisplatin-induced oxidative stress was assessed by analysis of Comet assay, DNA-laddering assay and activation of caspases. Heme oxygenase-1 (HO-1), mitogen-activated protein kinase (MAPK) pathway and nuclear factor-E2-related factor 2 (Nrf2) were measured by Western blot analysis. Transfection of small interfering RNAs (siRNA), glutathione (GSH) assay and RT-PCR were performed in this study. RESULTS: Kaempferol protected cells against cisplatin-induced apoptosis in a dose-dependent manner in HEI-OC1 cells. Kaempferol-induced HO-1 expression protected against cell death though the c-Jun N-terminal kinase (JNK) pathway and by the aid of Nrf2 translocation. Kaempferol increased the cellular level of GSH and the expression of GCLC time-dependently. siRNA GCLC blocked the increase of GSH level by kaempferol and the protective effect of kaempferol against cisplatin-induced cell death. CONCLUSION: The expression of HO-1 by kaempferol inhibits cisplatin-induced apoptosis in HEI-OC1 cells, and the mechanism of protective effect is also associated with its inductive effect of GCLC expression.


Subject(s)
Apoptosis/drug effects , Catalytic Domain , Cisplatin/toxicity , Glutamate-Cysteine Ligase/biosynthesis , Heme Oxygenase-1/biosynthesis , Kaempferols/physiology , Organ of Corti/enzymology , Animals , Apoptosis/genetics , Catalytic Domain/drug effects , Catalytic Domain/genetics , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Cisplatin/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Induction/drug effects , Enzyme Induction/genetics , Gene Expression Regulation/drug effects , Glutamate-Cysteine Ligase/genetics , Heme Oxygenase-1/genetics , Mice , Organ of Corti/cytology , Organ of Corti/drug effects
5.
J Med Food ; 11(2): 230-6, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18598163

ABSTRACT

Luteolin has been shown to possess antitumorigenic, antioxidant, and anti-inflammatory properties. In the present study, we investigated the protective mechanism of luteolin against cisplatin-induced apoptosis in auditory (House Ear Institute-Organ of Corti 1 [HEI-OC1]) cells. Luteolin was found to induce the expression of heme oxygenase-1 (HO-1) in a dose- and time-dependent manner. Luteolin also activated the extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase pathway, which plays an important role in the expression of HO-1. Luteolin protected the cells against cisplatin-induced apoptotic cell death. The protective effect of luteolin was abrogated by zinc protoporphyrin IX (ZnPP IX), an HO inhibitor, and antisense oligodeoxynucleotides against the HO-1 gene. Furthermore, pretreatment with luteolin inhibited the activation of caspase-3 and the mitochondrial dysfunction, and the effect of luteolin on the activation of caspase-3 disappeared in the presence of ZnPP IX or PD098059. These results demonstrate that the expression of HO-1 by luteolin is mediated by the ERK pathway, and also that the activating of HO-1 inhibits cisplatin-induced apoptosis in HEI-OC1 1 cells.


Subject(s)
Apoptosis/drug effects , Cisplatin/pharmacology , Heme Oxygenase-1/genetics , Luteolin/pharmacology , Organ of Corti/cytology , Animals , Caspase 3/metabolism , Cell Line, Transformed , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/physiology , Gene Expression/drug effects , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/biosynthesis , Mice , Oligonucleotides, Antisense/pharmacology , Organ of Corti/enzymology , Proto-Oncogene Proteins c-bcl-2/genetics
6.
Nucleic Acids Res ; 36(10): 3226-34, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18417535

ABSTRACT

Rad51 plays a key role in the repair of DNA double-strand breaks through homologous recombination, which is the central process in the maintenance of genomic integrity. Five paralogs of the human Rad51 gene (hRad51) have been identified to date, including hRad51B, hRad51C, hRad51D, Xrcc2 and Xrcc3. In searches of additional hRad51 paralogs, we identified a novel hRad51 variant that lacked the sequence corresponding to exon 9 (hRad51-Deltaex9). The expected amino acid sequence of hRad51-Deltaex9 showed a frame-shift at codon 259, which resulted in a truncated C-terminus. RT-PCR analysis revealed that both hRad51 and hRad51-Deltaex9 were prominently expressed in the testis, but that there were subtle differences in tissue specificity. The hRad51-Deltaex9 protein was detected as a 31-kDa protein in the testis and localized at the nucleus. In addition, the hRad51-Deltaex9 protein showed a DNA-strand exchange activity comparable to that of hRad51. Taken together, these results indicate that hRad51-Deltaex9 promotes homologous pairing and DNA strand exchange in the nucleus, suggesting that alternative pathways in hRad51- or hRad51-Deltaex9-dependent manners exist for DNA recombination and repair.


Subject(s)
Rad51 Recombinase/genetics , Amino Acid Sequence , Blotting, Western , Cell Nucleus/enzymology , Humans , Male , Molecular Sequence Data , Polymerase Chain Reaction , RNA Splicing , Rad51 Recombinase/chemistry , Rad51 Recombinase/metabolism , Sequence Homology, Amino Acid , Testis/metabolism , Tissue Distribution
7.
Eur J Pharmacol ; 580(1-2): 12-8, 2008 Feb 02.
Article in English | MEDLINE | ID: mdl-18021765

ABSTRACT

Heme oxygenase (HO)-1 is a cytoprotective enzyme that is activated by various phytochemicals. We examined the ability of brazilin to upregulate HO-1 expression in auditory cells. We found that brazilin induced the expressions of HO-1 mRNA and protein in concentration- and time-dependent manners. Brazilin induced nuclear factor-E2-related factor 2 (Nrf2) nuclear translocation, and dominant-negative Nrf2 attenuated brazilin-induced expression of HO-1. Brazilin induced a temporary increase in the phosphorylation of Akt. While LY294002, a non-selective phosphotidylinositol 3-kinase (PI3K) inhibitor, was able to reduce brazilin-induced phosphorylation of Akt and the subsequent induction of HO-1. Brazilin activated the extracellular signal-regulated kinase (ERK) and p38 pathways, and the ERK pathway played an important role in HO-1 expression. Brazilin protected the cells against t-butyl hydroperoxide (t-BHP)-induced cell death. The protective effect of brazilin was abrogated by anti-sense oligodeoxynucleotides (ODN) against the HO-1 gene. These results demonstrate that the expression of HO-1 by brazilin is mediated via the PI3K/Akt and ERK pathways, and this expression inhibits t-BHP-induced cell death in House Ear Institute-Organ of Corti 1 (HEI-OC1) cells.


Subject(s)
Benzopyrans/pharmacology , Heme Oxygenase-1/drug effects , Up-Regulation/drug effects , Animals , Benzopyrans/administration & dosage , Benzopyrans/isolation & purification , Caesalpinia/chemistry , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Heme Oxygenase-1/metabolism , Mice , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
8.
J Nutr Biochem ; 18(9): 615-22, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17418561

ABSTRACT

Piperine is a major component of black pepper, Piper nigrum Linn, used widely in traditional medicine. In this study, we examined whether piperine could protect House Ear Institute-Organ of Corti 1 (HEI-OC1) cells against cisplatin-induced apoptosis through the induction of heme oxygenase (HO)-1 expression. Piperine (10-100 microM) induced the expression of HO-1 in dose- and time-dependent manners. Piperine also induced antioxidant response element-luciferase and translocated nuclear factor-E2-related factor-2 (Nrf2) to nucleus. Piperine activated the c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase and p38 mitogen-activated protein kinase (MAPK) pathways, and the JNK pathway played an important role in piperine-induced HO-1 expression. Piperine protected the cells against cisplatin-induced apoptosis. The protective effect of piperine was abrogated by zinc protoporphyrin IX, an HO inhibitor, and antisense oligodeoxynucleotides against HO-1 gene. These results demonstrate that the expression of HO-1 by piperine is mediated by both JNK pathway and Nrf2, and the expression inhibits cisplatin-induced apoptosis in HEI-OC1 cells.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Benzodioxoles/pharmacology , Cisplatin/toxicity , Heme Oxygenase-1/metabolism , Organ of Corti/drug effects , Piperidines/pharmacology , Polyunsaturated Alkamides/pharmacology , Animals , Cell Nucleus/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Hair Cells, Auditory, Outer/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , NF-E2-Related Factor 2/metabolism , Organ of Corti/enzymology , Signal Transduction , Transfection
9.
Biofactors ; 30(3): 149-57, 2007.
Article in English | MEDLINE | ID: mdl-18525109

ABSTRACT

In this study, we examined the protective effects of Caesalpinia sappan L. and its major component, brazilin, against tert-butylhydroperoxide (t-BHP)-induced cell death in House Ear Institute-Organ of Corti 1 (HEI-OC1) cells. We found that the extract of C. sappan L. and brazilin induced antioxidant response element (ARE)-luciferase activity and heme oxygenase-1 (HO-1) expression in a concentration-dependent manner. The inductive effect of brazilin was more potent than the extract of C. sappan L. and the expression of HO-1 reached a peak at 12 h after brazilin treatment. The extract and brazilin protected the cells against t-BHP-induced cell death. Their protective effects were abrogated by zinc protoporphyrin IX (ZnPP IX), a HO inhibitor. These results demonstrate that the extract of C. sappan L. and brazilin induce the expression of HO-1 and the enzyme diminishes t-BHP-induced cell death in HEI-OC1 cells.


Subject(s)
Benzopyrans/pharmacology , Caesalpinia/chemistry , Heme Oxygenase-1/metabolism , Plant Extracts/pharmacology , Animals , Antioxidants/pharmacology , Benzopyrans/chemistry , Blotting, Western , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Heme Oxygenase-1/antagonists & inhibitors , Luciferases/genetics , Luciferases/metabolism , Mice , Molecular Structure , Organ of Corti/cytology , Organ of Corti/drug effects , Organ of Corti/metabolism , Oxidative Stress/drug effects , Plant Extracts/chemistry , Protoporphyrins/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Response Elements/genetics , Transfection , tert-Butylhydroperoxide/pharmacology
10.
Arch Biochem Biophys ; 444(2): 121-9, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16297369

ABSTRACT

Polycyclic aromatic hydrocarbons, such as benzo(a)pyrene (BaP), are widespread in the environment and cause untoward effects, including carcinogenesis, in mammalian cells. However, the molecular mechanism of apoptosis by BaP is remained to be elusive. Pharmacological inhibition of p38 kinase markedly inhibited the BaP-induced cytotoxicity, which was proven as apoptosis characterized by an increase in sub-G(0)/G(1) fraction of DNA content, ladder-pattern fragmentation of genomic DNA, and catalytic activation of caspase-3 with PARP cleavage. Our data also demonstrated that activation of caspase-3 was accompanied with activation of caspase-9 and mitochondrial dysfunction, which was also apparently suppressed by pretreatment with p38 kinase inhibitors. Also, pharmacological inhibition of p38 markedly inhibited the phosphorylation, accumulated expression, and transactivation activity of p53 in BaP-treated cells. Adenoviral overexpression of human p53 (wild-type) further augmented in increase of PARP cleavage and the sub-G(0)/G(1) fraction of DNA content. Furthermore, p53 mediated apoptotic activity in BaP-treated cells was inhibited by p38 kinase inhibitor. The current data collectively indicate that BaP induces apoptosis of Hepa1c1c7 cells via activation of p53-related signaling, which was, in part, regulated by p38 kinase.


Subject(s)
Apoptosis/drug effects , Benzo(a)pyrene/administration & dosage , Carcinoma, Hepatocellular/enzymology , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Activation , Mice
11.
Life Sci ; 76(20): 2299-314, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15748624

ABSTRACT

Resveratrol, a polyphenolic compound found in grape skin and peanuts has been shown to prevent many diseases including cardiovascular diseases and cancer. To better understand resveratrol's potential in vivo toxicity, we studied the dose response using cDNA stress arrays coupled with drug metabolizing enzymatic (DME) assays to investigate the expression of stress-responsive genes and Phase I and II detoxifying enzymes in rat livers. Male and female CD rats were treated with high doses of resveratrol (0.3, 1.0 and 3.0 gm/kg/day) for a period of 28 days. Total RNA from rat liver was reverse-transcribed using gene-specific primers and hybridized to stress-related cDNA arrays. Among female rats, Phase I DME genes were repressed at 0.3 and 1.0 gm/kg/day doses, while genes such as manganese superoxide dismutase, cytochrome P450 reductase, quinone oxidoreductase and thiosulfate sulfurtransferase demonstrated a dose-dependent increase in gene expression. The modulation of these liver genes may implicate the potential toxicity as observed among the rats at the highest dose level of resveratrol. Real-Time PCR was conducted on some of the Phase II DME genes and anti-oxidant genes to validate the cDNA array data. The gene expression from real-time PCR demonstrated good correlation with the cDNA array data. UGT1A genes were amongst the most robustly induced especially at the high doses of resveratrol. We next performed Phase I and Phase II enzymatic assays on cytochrome P450 2E1 (CYP2E1), cytochrome P450 1A1 (CYP1A1), NAD(P)H:quinone oxidoreductase (NQO1), glutathione S-transferase (GST) and UDP-glucuronosyl transferase (UGT). Induction of Phase II detoxifying enzymes was most pronounced at the highest dose of resveratrol. CYP1A1 activity demonstrated a decreasing trend among the 3 dose groups and CYP2E1 activity increased marginally among female rats over controls. In summary, at lower doses of resveratrol there are few significant changes in gene expression whereas the modulation of liver genes at the high dose of resveratrol may implicate the potential toxicity observed.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Gene Expression Regulation/drug effects , Liver/metabolism , Stilbenes/toxicity , Animals , Dose-Response Relationship, Drug , Female , Gene Expression Profiling , Genomics , Male , Oligonucleotide Array Sequence Analysis , Rats , Resveratrol , Sex Factors
12.
Toxicology ; 199(1): 35-46, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15125997

ABSTRACT

Benzo(a)pyrene (BaP), a potent carcinogen, has been shown to induce apoptosis via activation of caspase-3. However, the upstream of caspase-3 and other apoptosis signaling remain to be elusive. Herein, we demonstrated that treatment of Hepa1c1c7 cells with BaP increased the transcriptional expression of aryl hydrocarbon nuclear transporter and cytochrome p450 1A1 in a time and dose-dependent manner but did not aromatic hydrocarbon receptor. Also, the catalytic activation of caspase-3 and caspase-9 was induced whereas that of caspase-3 and caspase-9 was not by the addition of BaP. BaP also induced the mitochondrial dysfunction, including transition of mitochondria membrane potential and cytosolic release of cytochrome c. Furthermore, a decrease in the expression of Bcl-2 to Bax ratio and phosphorylation of p53(Ser 15) were observed in BaP-treated cells. Taken together, these results demonstrated that BaP-induced apoptosis of Hepa1c1c7 cells via activation of intrinsic caspase pathway including caspase-3, caspase-9, with mitochondrial dysfunction and p53 activation.


Subject(s)
Apoptosis/drug effects , Benzo(a)pyrene/toxicity , Carcinogens/toxicity , Caspases/metabolism , Mitochondria/drug effects , Animals , Carcinoma, Hepatocellular , Cell Line, Tumor , Enzyme Activation/drug effects , Hepatocytes/drug effects , Hepatocytes/pathology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mitochondria/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein
13.
Free Radic Biol Med ; 36(7): 858-71, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15019971

ABSTRACT

We recently demonstrated that heme oxygenase (HO)-1 is constitutively expressed in human CD4+CD25+ regulatory T cells and induced by anti-CD28 or anti-CD28/anti-CD3 stimulation, even in CD4+CD25- responder T cells. To study the effects of HO-1 expression on lymphocyte survival, we transfected the HO-1 gene or induced the gene to express HO-1 protein with cobalt protoporphyrin (CoPP) in Jurkat T cells. Consistently, anti-Fas antibody triggered apoptotic cell death in wild-type Jurkat T cells. Surprisingly, however, HO-1-overexpressing Jurkat T cells showed strong resistance to Fas-mediated apoptosis. In contrast, abrogation of HO-1 expression by antisense oligomer against HO-1 gene from CoPP-treated cells or depletion of iron by desferrioxamine from HO-1-transfected cells abolished the resistance. In addition, exogenously added iron rendered wild-type Jurkat T cells resistant. The resistance involved IkappaB kinase (IKK) activation via iron-induced reactive oxygen species formation, NF-kappaB activation by activated IKK, and c-FLIP expression by activated NF-kappaB. Primary CD4+ T cells induced by CoPP to express HO-1 also showed more resistance to Fas-mediated apoptosis than untreated cells. Our findings suggest that HO-1 plays a critical and nonredundant role in Fas-mediated activation-induced cell death of T lymphocytes.


Subject(s)
Apoptosis , CD4-Positive T-Lymphocytes/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Iron/metabolism , fas Receptor/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/enzymology , Caspases/metabolism , DNA, Complementary/genetics , Flow Cytometry , Genes, bcl-2/physiology , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1 , Humans , Immunochemistry , Intracellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , Membrane Proteins , NF-kappa B/metabolism , Oligonucleotides, Antisense/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , Protoporphyrins/pharmacology , Reactive Oxygen Species/metabolism , Transfection , NF-kappaB-Inducing Kinase
14.
J Pharmacol Exp Ther ; 310(1): 263-71, 2004 Jul.
Article in English | MEDLINE | ID: mdl-14988420

ABSTRACT

Sulforaphane (SUL) is one member of the isothiocyanate class of cancer chemopreventive compounds that has been shown to be effective in blocking initiation and progression of carcinogenesis. Previously, many studies have shown that SUL can potently induce phase II detoxifying enzymes, which contributes to its chemopreventive functions. In this study, we used 4967 oligonucleotides microarray to assess the genes that are modulated by SUL in in vivo rat livers, as well as time course of expression of these genes. The pharmacokinetics of SUL was assessed after oral dose of 50 micromol of SUL. The plasma concentration occurred at 1 h and peaked around 20 microM at 4 h after dosing and declined with a half-life of about 2.2 h. Analysis of the gene expression data found various clusters of genes that are important in cellular defense mechanisms and cell cycle regulation. The most robust cluster of genes is the metallothionein-like genes (MT-1/2 and MT-1a), which are increased up to 10-fold by 2 to 4 h after SUL dosing. The second cluster of genes is the glutathione S-transferase-A3-like genes, which include aflatoxin B1 aldehyde reductase and aldehyde oxidase. These genes are increased slightly by 4 h and peaked at 12 h. Real-time polymerase chain reaction was performed to authenticate the mRNA expression of some of these genes. In summary, this in vivo study of SUL provides the first clue as to the plasma concentrations of SUL, in vivo mitogen-activated protein kinase activations in rat livers, as well as what other genes are modulated in addition to phase II detoxifying genes. The results from this study may yield better insights for its chemopreventive functions.


Subject(s)
Gene Expression/drug effects , Isothiocyanates/blood , Thiocyanates/blood , Animals , Gene Expression Profiling , Isothiocyanates/chemistry , Isothiocyanates/pharmacology , Liver/drug effects , Liver/metabolism , Male , Oligonucleotide Array Sequence Analysis , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Sulfoxides , Thiocyanates/chemistry , Thiocyanates/pharmacology
15.
Cancer Res ; 63(21): 7520-5, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14612554

ABSTRACT

Sulforaphane (SFN) and its N-acetyl-L-cysteine (NAC) conjugate are effective inhibitors of tumorigenesis in animal models. These compounds induce the expression of the antioxidant response element (ARE)-related genes and cause apoptosis. We studied the role of reduced glutathione (GSH) in the activations of ARE-mediated gene expression, apoptosis, and the activation of c-Jun NH(2)-terminal kinase (JNK) in HepG2-C8 cells. The cellular level of GSH decreased transiently when cells were exposed to SFN and then increased from 4 h, reaching 2.2-fold over control at 24 h. In contrast, SFN-NAC did not change the GSH level substantially during the time of incubation. ARE expression was increased in a dose-dependent manner up to 35 micro M SFN and 75 micro M SFN-NAC, respectively. The induction of ARE by SFN was 8.6-fold higher than that by SFN-NAC. Pretreatment with L-buthionine sulfoximine increased SFN-induced ARE expression significantly. The decrease in ARE expression at higher concentrations of SFN and SFN-NAC was correlated with accelerated apoptotic cell death, with a dose-dependent activation of caspase 3 activity by SFN. On addition of extracellular GSH within 6 h of treatment with SFN, the effect on ARE expression was blocked almost completely. SFN was able to activate JNK1/2, and that activation was blocked by treatment with exogenous GSH. Taken together, these results suggest that the biological effects of SFN and SFN-NAC on the induction of ARE-related gene expression and apoptosis could be different from each other; however, the different effects on ARE-related gene expression and apoptosis elicited by SFN can be blocked by the addition of GSH.


Subject(s)
Antioxidants/metabolism , Apoptosis/drug effects , Glutathione/physiology , Response Elements/drug effects , Thiocyanates/pharmacology , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Apoptosis/physiology , Buthionine Sulfoximine/pharmacology , Caspase 3 , Caspases/metabolism , Cell Line, Tumor , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glutathione/metabolism , Glutathione/pharmacology , Humans , Isothiocyanates , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Sulfoxides , Thiocyanates/chemistry
16.
Pharm Res ; 20(9): 1351-6, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14567627

ABSTRACT

PURPOSE: Phenethyl isothiocyanate (PEITC) has been of great interest as a promising cancer chemopreventive agent. To better understand its chemopreventive activity, we examined the effect of PEITC on the antioxidant responsive element (ARE), which is an important gene regulatory element of many phase II drug-metabolizing/detoxification enzymes as well as cellular defensive enzymes. METHODS: HeLa cells were transiently transfected with different cDNA plasmids using calcium phosphate precipitation. Subsequently, the cells were maintained in fresh media, and various concentrations of PEITC were added to the transfected cells. After harvesting and lysing of the cells, ARE-luciferase reporter gene activity was measured and normalized against beta-galactosidase activity. RESULTS: Treatments of HeLa cells with PEITC transiently stimulated ARE-reporter gene expressions in a dose-dependent manner. Overexpression of wild-type NF-E2 related factor-2 (Nrf2) dramatically increased ARE-reporter gene expression in a dose-dependent manner. Similar effects were seen when wild-type c-Jun N-terminal kinase 1 (JNK1) was transfected, although the transactivating potential of JNK1 was much less than that of Nrf2. Cotransfection of Nrf2 and JNK1 showed additional enhancement of ARE reporter gene expression, implying that JNK1 might be an upstream activator of Nrf2. To support this, overexpression of dominant-negative JNK1 suppressed Nrf2-induced ARE reporter gene expression in a dose-dependent manner. When PEITC was added, slight enhancement of ARE reporter gene expression was observed in either Nrf2- or JNK1-transfected cells. Finally, ARE reporter activity induced by PEITC was substantially attenuated by transfection of either dominant-negative mutant of Nrf2 or dominant-negative mutant of JNK1. CONCLUSION: Taken together, these data suggest that JNK1 acts as an upstream activator of Nrf2 and that PEITC activates ARE-mediated phase II drug metabolism gene expressions via the JNK1- and Nrf2-dependent pathways.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antioxidants/metabolism , DNA-Binding Proteins/metabolism , Isothiocyanates/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Response Elements , Trans-Activators/metabolism , Anticarcinogenic Agents/therapeutic use , Chemoprevention , Enzyme Induction , Genes, Reporter , HeLa Cells , Humans , Isothiocyanates/therapeutic use , Luciferases/metabolism , Mitogen-Activated Protein Kinase 8 , NF-E2-Related Factor 2 , Signal Transduction , Transfection
17.
Carcinogenesis ; 24(8): 1361-7, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12819185

ABSTRACT

Phenethyl isothiocyanate (PEITC) is a potential chemopreventive agent that is present naturally in widely consumed vegetables, especially in watercress. It has been extensively investigated for its anticancer activities against lung, forestomach and esophageal tumorigenesis. Here we investigated the pro-apoptotic effect of PEITC in HT-29 human colorectal carcinoma cell line, and the mechanism of apoptosis induced by PEITC. PEITC-induced apoptosis was determined by DNA fragmentation assay and diamidino-2-phenylindole (DAPI) staining technique. To understand the mechanisms of apoptosis induced by PEITC, we studied the role of caspases, mitochondria-cytochrome c release, and mitogen-activated protein kinase (MAPK) signaling pathways involved in PEITC-induced apoptosis in HT-29 cells. Both the caspase-3 and -9 activities were stimulated by PEITC. The release of cytochrome c from the mitochondrial inter-space was time- and dose-dependent, with a maximal release at 50 micro M after 10 h treatment. Three MAPKs [JNK (c-Jun N-terminal kinase), extracellular signal-regulated protein kinase (ERK) and p38 kinase] were activated shortly after PEITC treatment in HT-29 cells. Importantly, the SP600125 compound, an anthrapyrazolone inhibitor of JNK, but not the ERK and p38 inhibitor, suppressed apoptosis induced by PEITC. Similarly, this JNK inhibitor attenuated both cytochrome c release and caspase-3 activation induced by PEITC. In summary, this study shows that PEITC can induce apoptosis in HT-29 cells in a time- and dose-dependent manner via the mitochondria caspase cascade, and the activation of JNK is critical for the initiation of the apoptotic processes. This mechanism of PEITC may play an important role in the killing of cancerous cells and offer a potential mechanism for its anticancer action in vivo.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Isothiocyanates/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , Aldehyde Dehydrogenase/antagonists & inhibitors , Anthracenes/pharmacology , Blotting, Western , Caspases/metabolism , Cell Nucleus/metabolism , Cytochrome c Group/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , HT29 Cells/drug effects , Humans , JNK Mitogen-Activated Protein Kinases , MAP Kinase Signaling System , Mitochondria/drug effects , Mitochondria/metabolism
18.
J Toxicol Environ Health A ; 65(5-6): 373-81, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11936218

ABSTRACT

A series of flavonoids isolated from Scutellariae radix were evaluated for their effects on cytochrome P-450 (CYP) activities in human liver microsomes. All flavonoids did not substantially inhibit pentoxyresorufin O-deethylation (CYP2B 1), mephenytoin 4-hydroxylation (CYP2C19), dextromethorphan O-demethylation (CYP2D6), and chlorzoxazone 6-hydroxylation (CYP2E1) activities (IC50: >50 microM). Baicalein and 2',5,6',7-tetrahydroxyflavone inhibited hepatic testosterone 6beta-hydroxylation (CYP3A4) activity with a IC50 of 17.4 and 7.8 microM, respectively. Oroxylin A inhibited diclofenac 4-hydroxylation (CYP2C9) activity with a IC50 of 6.7 microM. In contrast, all flavonoids tested inhibited hepatic caffeine N'-demethylation (CYP1A2) with IC50 values ranging from 0.7 to 51.3 microM. Kinetic analysis revealed that the mechanism of inhibition varied according to the flavonoids. These results suggest that flavonoids tested are inhibitors of hepatic CYP1A2 and that the extracts of Scutellariae radix, widely used as a hepatoprotective agent, may protect the liver through the prevention of CYPIA2-induced metabolic activation of protoxicants.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Lamiaceae/chemistry , Cell Culture Techniques , Humans , Microsomes, Liver/enzymology , Plant Extracts
SELECTION OF CITATIONS
SEARCH DETAIL
...