Abstract

Activated Ras has been shown to provide powerful antiapoptotic signals to cells through well defined transcriptional and post- translational pathways, whereas translational control as a mechanism of Ras survival signaling remains unexplored. Here we show a direct relationship between assembly of the cap-dependent translation initiation apparatus and suppression of apoptosis by oncogenic Ras in vitro and in vivo. Decreasing protein synthesis with rapamycin, which is known to inhibit cap-dependent translation, increases the susceptibility of Ras-transformed fibroblasts to cytostatic drug-induced apoptosis. In contrast, suppressing global protein synthesis with equipotent concentrations of cycloheximide actually prevents apoptosis. Enforced expression of the cap-dependent translational repressor, the eukaryotic translation initiation factor (eIF) 4E-binding protein (4E-BPI), sensitizes fibroblasts to apoptosis in a manner strictly dependent on its ability to sequester eIF4E from a translationally active complex with eIF4GI and the co-expression of oncogenic Ras. Ectopic expression of 4E-BP1 also promotes apoptosis of Ras-transformed cells injected into immunodeficient mice and markedly diminishes their tumorigenicity. These results establish that eIF4E-dependent protein synthesis is essential for survival of fibroblasts bearing oncogenic Ras and support the concept that activation of cap-dependent translation by extracellular ligands or intrinsic survival signaling molecules suppresses apoptosis, whereas synthesis of proteins mediating apoptosis can occur independently of the cap.

Highlights

  • When rapamycin was added to Ras-transformed cells, it caused a dose-dependent decline in protein synthesis, which paralleled its ability to sensitize cells to lovastatin-induced apoptosis (Fig. 1c)

  • Equipotent doses of the peptide elongation inhibitor cycloheximide blocked apoptosis (Fig. 1d), demonstrating that the execution of lovastatin-induced cell death requires global protein synthesis. These results suggest that a generalized inhibition of mRNA translation is not the means by which rapamycin exerts its proapoptotic effect, rather they point toward a selective inhibition of antiapoptotic mRNA translation or a mechanism independent of its ability to repress translation

  • Examples of selective control have emerged involving regulation at the translation initiation step, in the integration of pleiotropic responses leading to differentiation, FIG. 3. 4E-BP1-promoted apoptosis in Ras-transformed cells is associated with displacement of translation factor eIF4GI from eIF4E. a, immunoblot analysis of 4E-BP1 and eIF4GI associated with cap-bound eIF4E in clones of Cloned rat embryo fibroblasts (CREF)/RasV12 ectopically expressing wild type 4E-BP1. b and c, apoptosis is shown as a function of the 4EBP1/eIF4E (b) or eIF4G/eIF4E (c) ratio in the indicated CREF/Ras/BP1-wt clones incubated in growth medium for 24 h in the presence or absence of 5 ␮M lovastatin. d, 4EBP1 lacking an eIF4E binding domain does not promote apoptosis

Read more

Summary

Translational Control of the Antiapoptotic Function of Ras*

Enforced expression of the cap-dependent translational repressor, the eukaryotic translation initiation factor (eIF) 4Ebinding protein (4E-BPI), sensitizes fibroblasts to apoptosis in a manner strictly dependent on its ability to sequester eIF4E from a translationally active complex with eIF4GI and the co-expression of oncogenic Ras. Ectopic expression of 4E-BP1 promotes apoptosis of Ras-transformed cells injected into immunodeficient mice and markedly diminishes their tumorigenicity. To elucidate the role of translational control in Ras survival signaling, here we focus directly on the 5Ј-mRNA cap binding complex and examine the induction of apoptosis in cells transformed with oncogenic Ras after a generalized reduction in protein synthesis or after specific repression of cap-dependent translation initiation. Extracellular survival factors suppress the intrinsic apoptotic apparatus through cognate receptor kinases at the cell surface, which activate the proto-oncogene ras and a number of pleiotropic transcriptional and post-translational effector pathways [1]. Akt-mediated phosphorylation of the Bcl-2 family member Bad [6] and the cell death protease caspase-9 [7] is implicated in post-translational suppression of

EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.