Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Apoptosis ; 10(1): 193-200, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15711935

ABSTRACT

Our previous studies demonstrated that antiallergic effects of herbs such as clove and Magnoliae Flos (MF) resulted from the induction of apoptosis in mast cells. We here examined whether the antiallergic activity was caused by eugenol (4-allyl-2-methoxyphenol) which was one of major ingredients in the essential oils or extracts of numerous plants including clove and Magnoliae Flos. RBL-2H3 cells were treated with eugenol, and DNA electrophoresis, Western blotting, immunocytochemistry, confocal microscopy and immunoprecipitation were conducted. Effect of eugenol was tested using a rat anaphylaxis model. RBL-2H3 cells treated with eugenol showed typical apoptotic manifestations and translocation of p53 into mitochondria. Antisense p53 partially prevented the induction of apoptosis. Noticeably, we observed that p53 translocated into mitochondria was phosphorylated on ser 15. Phospho-ser 15-p53 physically interacted with Bcl-2 and Bcl-xL in mitochondria and its translocation into mitochondria preceded cytochrome c release and mitochondrial membrane potential (MMP) reduction. We also depicted that the survival of animals even after administration of the fatal dose of compound 48/80 might result from the decreased number of mast cells by eugenol pretreatment. In conclusion, eugenol induces apoptosis in mast cells via translocation of phospho-ser 15-p53 into mitochondria.


Subject(s)
Apoptosis/drug effects , Eugenol/pharmacology , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Rats , bcl-X Protein
2.
Apoptosis ; 9(5): 583-9, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15314286

ABSTRACT

Although histone deacetylase (HDAC) inhibitors are emerging as a promising new treatment strategy in malignancy, how they exert their effect on osteosarcoama cells is as yet unclear. This study was undertaken to investigate the underlying mechanism of a HDAC inhibitor Trichostatin A (TSA)-induced apoptosis in a osteosarcoma cell line HOS. We observed that TSA treatment decreased the viability of the cells and prominently increased acetylation of histone H3. Evidence was obtained indicating that TSA induced apoptosis of HOS cells as follows: (1) Generation of DNA fragmentation; (2) activation of procaspase-3; (3) cleavage of PARP; and (4) increase of DNA hypoploidy. The reduction of MMP and the release of cytochrome c to cytosol were also shown, indicating that TSA induces apoptosis in HOS cells in a histone acetylation- and mitochondria-dependent fashions. We also examined whether TSA can sensitize HOS cells to the action of an antitumor agent genistein. The combination therapy of TSA and genistein showed synergistic anticancer effect indicating that TSA can be considered as a novel therapeutic strategy for osteosarcoma not only from its direct apoptosis-inducing activity but also from the possibility of sensitization to other antitumor agents.


Subject(s)
Apoptosis/drug effects , Bone Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Osteosarcoma/pathology , Caspase Inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Humans , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/physiology
3.
Toxicol Appl Pharmacol ; 166(3): 173-85, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10906281

ABSTRACT

Individuals are commonly exposed to bacterial endotoxin (lipopolysaccharide [LPS]) through gram-negative bacterial infection and from its translocation from the gastrointestinal lumen into the circulation. Inasmuch as noninjurious doses of LPS augment the hepatotoxicity of certain xenobiotic agents, exposure to small amounts of LPS may be an important determinant of susceptibility to chemical intoxication. Monocrotaline (MCT) is a pyrrolizidine alkaloid phytotoxin that at large doses produces centrilobular liver lesions in rats. In the present study, MCT was coadministered with LPS to determine whether LPS would enhance its hepatotoxicity. Doses of MCT (100 mg/kg, ip) and LPS (7.4 x 10(6) EU/kg, iv), which were nonhepatotoxic when administered separately, produced significant liver injury in male, Sprague-Dawley rats when given in combination. Within 18 h after MCT administration, this cotreatment resulted in enhanced plasma alanine aminotransferase and aspartate aminotransferase activities, two markers of liver injury. Histologically, overt hemorrhage and necrosis appeared between 12 and 18 h. The lesions were centrilobular and midzonal and exhibited characteristics similar to lesions associated with larger doses of MCT and LPS, respectively. In the presence of LPS, the threshold for MCT toxicity was reduced to 13-33% of the dose required for toxicity with MCT alone. A study in isolated, hepatic parenchymal cells revealed no interaction between MCT and LPS in producing cytotoxicity. In summary, coexposure of rats to noninjurious doses of MCT and LPS resulted in pronounced liver injury. Results in vitro suggest that the enhanced toxicity does not result from a direct interaction of MCT and LPS with hepatic parenchymal cells. These results provide additional evidence that exposure to small amounts of LPS may be a determinant of susceptibility to food-borne hepatotoxins.


Subject(s)
Lipopolysaccharides/toxicity , Liver/drug effects , Monocrotaline/toxicity , Animals , Dose-Response Relationship, Drug , Liver/pathology , Male , Rats , Rats, Sprague-Dawley
4.
Toxicol Sci ; 55(2): 444-52, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10828277

ABSTRACT

Bacterial endotoxin (lipopolysaccharide; LPS) given to animals in large doses results in pronounced, midzonal liver injury. Exposure to smaller, non-injurious doses of LPS augments the toxicity of certain hepatotoxicants. This study was conducted to delineate the development of injury in a rat model of augmentation of aflatoxin B(1) (AFB(1)) hepatotoxicity by LPS. At large doses (i.e., > 1 mg/kg, ip), AFB(1) administration resulted in pronounced injury to the periportal regions of the liver. Male, Sprague-Dawley rats (250-350 g) were treated with 1 mg AFB(1)/kg, ip or its vehicle (0.5% DMSO/saline) and 4 h later with either E. coli LPS (7.4 x 106 EU/kg, iv) or its saline vehicle. Liver injury was assessed 6, 12, 24, 48, 72, or 96 h after AFB(1) administration. Hepatic parenchymal cell injury was evaluated as increased alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities in serum and from histologic examination of liver sections. Biliary tract alterations were evaluated as increased concentration of serum bile acids and activities of gamma-glutamyltransferase (GGT), alkaline phosphatase (ALP), and 5'-nucleotidase (5'-ND) in serum. At all times and for all markers, injury in rats treated with either AFB(1) or LPS alone was absent or modest. In the AFB(1)/LPS cotreated group, hepatic parenchymal cell injury was pronounced by 24 h and had returned to control values by 72 h. The injury began in the periportal region and spread midzonally with time. Furthermore, changes in serum markers indicative of biliary tract alterations were evident by 12 h and had returned to control values by 72 h. Thus, the nature of the hepatic lesions suggested that LPS potentiated the effects of AFB(1) on both parenchymal and bile duct epithelial cells.


Subject(s)
Aflatoxin B1/pharmacology , Chemical and Drug Induced Liver Injury/etiology , Lipopolysaccharides/pharmacology , Liver/drug effects , 5'-Nucleotidase/blood , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Animals , Apoptosis/drug effects , Aspartate Aminotransferases/blood , Bile Acids and Salts/blood , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/pathology , Cholestasis/blood , Cholestasis/chemically induced , Cholestasis/pathology , Drug Synergism , Escherichia coli , In Situ Nick-End Labeling , Liver/pathology , Male , Rats , Rats, Sprague-Dawley , gamma-Glutamyltransferase/blood
5.
Arch Biochem Biophys ; 347(2): 235-41, 1997 Nov 15.
Article in English | MEDLINE | ID: mdl-9367530

ABSTRACT

Investigations into the enzymes responsible for the reductive activation of antineoplastic agents are of particular interest with regard to the use of these agents in the treatment of solid tumors. Xanthine oxidase (EC 1.1.3.22; XO) and xanthine dehydrogenase (EC 1. 1.1.204; XDH) are two enzymes capable of the reductive activation of antineoplastic agents. Previously, XDH, the enzymatic precursor of XO, was not extensively studied because of difficulties in its isolation. Research in the reductive activation of antineoplastic agents by XDH has increased with the discovery of a rapid and high-yield purification procedure for XDH. In the present investigation, the potential for drug activation of doxorubicin (DOX), streptonigrin (STN), and menadione (MD) by XO and XDH was assessed through oxygen consumption studies. These studies were conducted at pH 7.4 and pH 6.0 to reflect physiological and the acidic pH of solid tumors, respectively. Apparent kinetic constants were determined for DOX, STN, and MD activation by XO and XDH at both pH levels. Higher oxygen consumption was observed for XDH drug activation in comparison to XO drug activation at equivalent enzyme activity for both pH levels. Drug-induced oxygen consumption was affected by pH. Hence, drug activation for DOX, STN, and MD was dependent upon the form of the xanthine-converting enzyme and the pH.


Subject(s)
Doxorubicin/metabolism , Streptonigrin/metabolism , Vitamin K/metabolism , Xanthine Dehydrogenase/metabolism , Xanthine Oxidase/metabolism , Animals , Antineoplastic Agents/metabolism , Female , Hydrogen-Ion Concentration , Mammary Neoplasms, Experimental/enzymology , Mice , Mice, Inbred BALB C , Mice, Nude , Oxidation-Reduction , Oxygen Consumption , Prodrugs/metabolism , Reactive Oxygen Species
6.
Neurosci Res ; 28(4): 373-7, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9274833

ABSTRACT

Three cDNA clones for the Machado-Joseph disease gene (MJD1) were isolated, two of which have a new exon sequence and a distinct 3' terminal nucleotide sequence resulting in a new carboxyl terminal domain in the translated product. The nucleotide sequence of the other one is similar to the previously published one except for five polymorphisms, one of which is a single nucleotide substitution resulting in a change from the stop codon (TAA; allele A) to a tyrosine residue (TAC; allele C). Genetic analysis results suggest that Japanese MJD mutations are associated with allele A.


Subject(s)
Machado-Joseph Disease/metabolism , Nerve Tissue Proteins/genetics , Amino Acid Sequence , Ataxin-3 , Base Sequence , Codon/genetics , Humans , Machado-Joseph Disease/genetics , Molecular Sequence Data , Nuclear Proteins , Polymorphism, Genetic , Repressor Proteins
7.
Chem Biol Interact ; 104(2-3): 87-101, 1997 May 02.
Article in English | MEDLINE | ID: mdl-9212777

ABSTRACT

The role of enzymes in the reductive activation of various chemotherapeutic agents is an area of considerable interest in studies to better understand the selective toxicities of these agents. Xanthine dehydrogenase (XDH) is an enzyme capable of reductive activation of chemotherapeutic agents. Previously, this enzyme has not been extensively studied because of difficulties in its isolation. We recently isolated this enzyme from EMT6 mouse mammary carcinoma cells and showed that this enzyme is capable of activating mitomycin C. In this study, we examined whether XDH could activate the clinically important antineoplastic agent, doxorubicin. Drug activation was determined under aerobic and hypoxic conditions and at various pHs in order to simulate the different environments found in solid tumors. The results of these studies show that XDH reacts with doxorubicin via a two-electron reduction. This reduction is different from the modified and more extensively studied form of the enzyme, xanthine oxidase (XO), which reacts with doxorubicin via a one-electron reduction. Under hypoxic conditions, the formation of large quantities of 7-deoxydoxorubicin aglycone, a deactivation product of doxorubicin metabolism, may serve to moderate doxorubicin's antineoplastic activity. Under aerobic conditions, however, XDH activation led to a greater rate of formation of oxygen radicals than XO thereby possibly potentiating doxorubicin's cytotoxicity to aerobic tumor cells. Kinetic constants were determined for doxorubicin activation by XDH.


Subject(s)
Antibiotics, Antineoplastic/metabolism , Carcinoma/enzymology , Doxorubicin/metabolism , Mammary Neoplasms, Experimental/enzymology , Xanthine Dehydrogenase/metabolism , Animals , Antibiotics, Antineoplastic/chemistry , Carcinoma/pathology , Chromatography, High Pressure Liquid , Doxorubicin/chemistry , Electrons , Female , Hydrogen-Ion Concentration , Kinetics , Mammary Neoplasms, Experimental/pathology , Mice , NAD/chemistry , NAD/metabolism , Naphthacenes/metabolism , Oxidation-Reduction , Oxygen Consumption/physiology , Reactive Oxygen Species , Xanthine Dehydrogenase/isolation & purification , Xanthine Oxidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...