Abstract

Fas-associated death domain (FADD) is a crucial adaptor protein mediating death signals triggered by death receptors. It was reported that FADD protein level are upregulated 20 fold after TNFa/Actinomycin D (ActD) treatment in cultured hepatocytes. In contrast, FADD mRNA level are reduced under the same conditions. This suggests a post-translational mechanism may be responsible for upregulation of FADD during hepatocyte apoptosis. We addressed whether phosphorylation of FADD plays a role in mouse hepatocytes (MHC) apoptosis induced by TNFa/ActD treatment. TNFa and ActD, but not TNFa alone, strongly upregulated a 28kDa FADD protein in MHCs 6h after treatment. Surprisingly, an extra band at ~66 kDa was consistently detected in MHCs even when using two different anti-FADD monoclonal antibodies. We show that the ~66kDa band was specific for phosphorylated FADD (pFADD). We confirmed this finding by ectopically expressing mouse FADD fused with a myc tagged protein in 293T cells and immunoblotting with both anti-myc and anti-pFADD antibodies. Mutation of Serine 191, a known FADD phosphorylation site, completely abolished the ~66 kDa band. We used immunofluorescence to show that pFADD is exclusively located in the nucleus of mouse hepatocytes. Our results indicate that this novel ~66 kDa protein is part of a complex that contains pFADD. This suggests that phosphorylation plays an important role in FADD nuclear localization and may prove to be important in FADD protein upregulation in TNFa stimulated MHCs.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call