Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurochem ; 101(5): 1205-13, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17403030

ABSTRACT

When activated by proinflammatory stimuli, microglia release substantial levels of glutamate, and mounting evidence suggests this contributes to neuronal damage during neuroinflammation. Prior studies indicated a role for the Xc exchange system, an amino acid transporter that antiports glutamate for cystine. Because cystine is used for synthesis of glutathione (GSH) synthesis, we hypothesized that glutamate release is an indirect consequence of GSH depletion by the respiratory burst, which produces superoxide from NADPH oxidase. Microglial glutamate release triggered by lipopolysaccharide was blocked by diphenylene iodonium chloride and apocynin, inhibitors of NADPH oxidase. This glutamate release was also blocked by vitamin E and elicited by lipid peroxidation products 4-hydroxynonenal and acrolein, suggesting that lipid peroxidation makes crucial demands on GSH. Although NADPH oxidase inhibitors also suppressed nitrite accumulation, vitamin E did not; moreover, glutamate release was largely unaffected by nitric oxide donors, inhibitors of nitric oxide synthase, or changes in gene expression. These findings indicate that a considerable degree of the neurodegenerative consequences of neuroinflammation may result from conversion of oxidative stress to excitotoxic stress. This phenomenon entails a biochemical chain of events initiated by a programmed oxidative stress and resultant mass-action amino acid transport. Indeed, some of the neuroprotective effects of antioxidants may be due to interference with these events rather than direct protection against neuronal oxidation.


Subject(s)
Glutamic Acid/metabolism , Lipid Peroxidation/physiology , Microglia/metabolism , Oxidation-Reduction , Acetophenones/pharmacology , Animals , Animals, Newborn , Antioxidants/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Lipid Peroxidation/drug effects , Lipopolysaccharides/pharmacology , Models, Biological , Onium Compounds/pharmacology , Oxidation-Reduction/drug effects , Rats , Vitamin E/pharmacology
2.
Biochem Pharmacol ; 66(3): 459-69, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12907245

ABSTRACT

Assessment of specific apoptosis and survival pathways implicated in anticancer drug action is important for understanding drug mechanisms and modes of resistance in order to improve the benefits of chemotherapy. In order to better examine the role of mitogen-activated protein kinases, including JNK and ERK, as well as the tumor suppressor p53, in the response of tumor cells to chemotherapy, we compared the effects on these pathways of three structurally and functionally distinct antitumor agents. Drug concentrations equal to 50 times the concentration required to reduce cell proliferation by 50% were used. Vinblastine, doxorubicin, or etoposide (VP-16) induced apoptotic cell death in KB-3 carcinoma cells, with similar kinetic profiles of PARP cleavage, caspase 3 activation, and mitochondrial cytochrome c release. All three drugs strongly activated JNK, but only vinblastine induced c-Jun phosphorylation and AP-1 activation. Inhibition of JNK by SP600125 protected cells from drug-induced cytotoxicity. Vinblastine caused inactivation of ERK whereas ERK was unaffected in cells exposed to doxorubicin or VP-16. Inhibition of ERK signaling by the MEK inhibitor, U0126, potentiated the cytotoxic effects of vinblastine and doxorubicin, but not that of VP-16. Vinblastine induced p53 downregulation, and chemical inhibition of p53 potentiated vinblastine-induced cell death, suggesting a protective effect of p53. In contrast, doxorubicin and VP-16 induced p53, and inhibition of p53 decreased drug-induced cell death, suggesting a pro-apoptotic role for p53. These results highlight the differential roles played by several key signal transduction pathways in the mechanisms of action of key antitumor agents, and suggest ways to specifically potentiate their effects in a context-dependent manner. In addition, the novel finding that JNK activation can occur without c-Jun phosphorylation or AP-1 activation has important implications for our understanding of JNK function.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Mitogen-Activated Protein Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Doxorubicin/pharmacology , Enzyme Activation , Etoposide/pharmacology , Gene Expression/drug effects , Humans , JNK Mitogen-Activated Protein Kinases , Kinetics , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Phosphorylation/drug effects , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured , Vinblastine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...