Your browser doesn't support javascript.
loading
Protein kinase C (PKC) participates in acetaminophen hepatotoxicity through c-jun-N-terminal kinase (JNK)-dependent and -independent signaling pathways.
Saberi, Behnam; Ybanez, Maria D; Johnson, Heather S; Gaarde, William A; Han, Derick; Kaplowitz, Neil.
Afiliação
  • Saberi B; University of Southern California Research Center for Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089-9121, USA.
  • Ybanez MD; University of Southern California Research Center for Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089-9121, USA.
  • Johnson HS; University of Southern California Research Center for Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089-9121, USA.
  • Gaarde WA; Isis Pharmaceuticals, Carlsbad, CA 92008-7326.
  • Han D; KGI School of Biopharmacy, Keck Graduate Institute, 535 Watson Drive Claremont, CA 91711.
  • Kaplowitz N; University of Southern California Research Center for Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089-9121, USA.
Hepatology ; 59(4): 1543-1554, 2014 Apr.
Article em En | MEDLINE | ID: mdl-23873604
ABSTRACT
UNLABELLED This study examines the role of protein kinase C (PKC) and AMP-activated kinase (AMPK) in acetaminophen (APAP) hepatotoxicity. Treatment of primary mouse hepatocytes with broad-spectrum PKC inhibitors (Ro-31-8245, Go6983), protected against APAP cytotoxicity despite sustained c-jun-N-terminal kinase (JNK) activation. Broad-spectrum PKC inhibitor treatment enhanced p-AMPK levels and AMPK regulated survival-energy pathways including autophagy. AMPK inhibition by compound C or activation using an AMPK activator oppositely modulated APAP cytotoxicity, suggesting that p-AMPK and AMPK regulated energy survival pathways, particularly autophagy, play a critical role in APAP cytotoxicity. Ro-31-8245 treatment in mice up-regulated p-AMPK levels, increased autophagy (i.e., increased LC3-II formation, p62 degradation), and protected against APAP-induced liver injury, even in the presence of sustained JNK activation and translocation to mitochondria. In contrast, treatment of hepatocytes with a classical PKC inhibitor (Go6976) protected against APAP by inhibiting JNK activation. Knockdown of PKC-α using antisense (ASO) in mice also protected against APAP-induced liver injury by inhibiting JNK activation. APAP treatment resulted in PKC-α translocation to mitochondria and phosphorylation of mitochondrial PKC substrates. JNK 1 and 2 silencing in vivo decreased APAP-induced PKC-α translocation to mitochondria, suggesting PKC-α and JNK interplay in a feed-forward mechanism to mediate APAP-induced liver injury.

CONCLUSION:

PKC-α and other PKC(s) regulate death (JNK) and survival (AMPK) proteins, to modulate APAP-induced liver injury.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Proteína Quinase C / Transdução de Sinais / Proteínas Quinases JNK Ativadas por Mitógeno / Proteínas Quinases Ativadas por AMP / Doença Hepática Induzida por Substâncias e Drogas / Acetaminofen Limite: Animals Idioma: En Revista: Hepatology Ano de publicação: 2014 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Proteína Quinase C / Transdução de Sinais / Proteínas Quinases JNK Ativadas por Mitógeno / Proteínas Quinases Ativadas por AMP / Doença Hepática Induzida por Substâncias e Drogas / Acetaminofen Limite: Animals Idioma: En Revista: Hepatology Ano de publicação: 2014 Tipo de documento: Article País de afiliação: Estados Unidos