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.
Clin Liver Dis ; 11(3): 459-75, v, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17723915

ABSTRACT

Drug-induced liver injury depends initially on development of hepatocyte stress and cell death, which can be induced directly by parent drugs or by toxic metabolites. Hepatocyte stress can lead to activation of built-in death programs for apoptosis or necrosis. Subsequently, the innate immune system's participation is recruited. The interplay between proinflammatory and anti-inflammatory components of innate immune system determines the outcome of drug-induced liver injury. Both environmental factors and genetic differences in cellular responses to stress and the innate immune response may account for different susceptibilities between individuals to drug-induced liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury , Drug-Related Side Effects and Adverse Reactions , Acetaminophen/adverse effects , Apoptosis/drug effects , Humans , Immunity, Innate , Liver/drug effects , Liver/metabolism , Liver/pathology
2.
Gastroenterology ; 131(1): 165-78, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16831600

ABSTRACT

BACKGROUND & AIMS: In searching for effects of acetaminophen (APAP) on hepatocytes downstream of its metabolism that may participate in hepatotoxicity, we examined the role of stress kinases. METHODS: Mouse hepatocytes and C57BL/6 mice were administered a toxic dose of APAP with or without SP600125, a chemical c-jun N-terminal kinase (JNK) inhibitor. JNK activity as reflected in phospho-c-jun levels, serum alanine transaminase (ALT), and liver histology were assessed. Similar experiments were repeated in JNK1 and JNK2 knockout mice and by using antisense oligonucleotide (ASO) to knockdown JNK. RESULTS: Sustained activation of JNK was observed in cultured mouse hepatocytes and in vivo in the liver after APAP treatment. The importance of this pathway was identified by the marked protective effect of SP600125 against APAP toxicity in vitro and in vivo. The specificity of this protective effect was confirmed in vivo by the knockdown of JNK1 and 2 using ASO pretreatment. JNK2 knockout mice and mice treated with JNK2 ASO exhibited partial protection against APAP. One potential target of JNK is Bax translocation, which was enhanced by APAP and blocked by the JNK inhibitor. Protection by the JNK inhibitor persisted in Kupffer cell-depleted mice, whereas there was no protection against CCl(4) or concanavalin A toxicity. CONCLUSIONS: This work suggests that JNK acts downstream of APAP metabolism to promote hepatotoxicity. The results suggest that JNK2 plays a predominant role, although maximum protection was seen with decrease in both forms of JNK.


Subject(s)
Acetaminophen/toxicity , Chemical and Drug Induced Liver Injury/enzymology , JNK Mitogen-Activated Protein Kinases/metabolism , Alanine Transaminase/blood , Analgesics, Non-Narcotic/toxicity , Animals , Anthracenes/therapeutic use , Apoptosis , Blotting, Western , Cells, Cultured , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Disease Models, Animal , Hepatocytes/drug effects , Hepatocytes/enzymology , Hepatocytes/pathology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...