Abstract

BackgroundProliferating tumor cells require continuous protein synthesis. De novo synthesis of most proteins is regulated through cap-dependent translation. Cellular stress such as ionizing radiation (IR) blocks cap-dependent translation resulting in shut-down of global protein translation which saves resources and energy needed for the stress response. At the same time, levels of proteins required for stress response are maintained or even increased. The study aimed to analyze the regulation of signaling pathways controlling protein translation in response to IR and the impact on Mcl-1, an anti-apoptotic and radioprotective protein, which levels rapidly decline upon IR.MethodsProtein levels and processing were analyzed by Western blot. The assembly of the translational pre-initiation complex was examined by Immunoprecipitation and pull-down experiments with 7-methyl GTP agarose. To analyze IR-induced cell death, dissipation of the mitochondrial membrane potential and DNA fragmentation were determined by flow cytometry. Protein levels of the different initiation factors were down-regulated using RNA interference approach.ResultsIR induced caspase-dependent cleavage of the translational initiation factors eIF4G1, eIF3A, and eIF4B resulting in disassembly of the cap-dependent initiation complex. In addition, DAP5-dependent initiation complex that regulates IRES-dependent translation was disassembled in response to IR. Moreover, IR resulted in dephosphorylation of 4EBP1, an inhibitor of cap-dependent translation upstream of caspase activation. However, knock-down of eIF4G1, eIF4B, DAP5, or 4EBP1 did not affect IR-induced decline of the anti-apoptotic protein Mcl-1.ConclusionOur data shows that cap-dependent translation is regulated at several levels in response to IR. However, the experiments indicate that IR-induced Mcl-1 decline is not a consequence of translational inhibition in Jurkat cells.

Highlights

  • Proliferating tumor cells require continuous protein synthesis

  • Using 7-methy-GTP agarose that imitates the cap structure in a pull-down assay, we could show that less eIF4G1, eIF4A, and eIF3A associated in the cap-binding initiation complex 24h after irradiation

  • Our results suggest that the ionizing radiation (IR)-induced Myeloid cell leukemia sequence 1 (Mcl-1) decline was not caused by inhibition of cap-dependent or internal ribosome entry site (IRES)/Death associated protein 5 (DAP5)-dependent translation Mcl-1 expression was translationally controlled by eIF4G1 in non-irradiated, healthy Jurkat cells

Read more

Summary

Introduction

Proliferating tumor cells require continuous protein synthesis. De novo synthesis of most proteins is regulated through cap-dependent translation. The rate limiting step during protein synthesis is the initiation of translation which is regulated by several initiation factors (IF). They allow the recruitment of the initiator tRNA and mRNA to the 40S ribosomal subunit, recognition of the start codon AUG, and joining of the 40S and 60S ribosomal subunits leading to the formation of peptid bonds during protein elongation. The eucaryotic initiation factors eIF3, eIF4A, eIF4B, eIF4E, eIF4G1, eIF4H, and the poly A-binding protein (PABP) regulate the recruitment of mRNA to the initiation complex. Binding directly to the initiation factors eIF3, eIF4A, eIF4E, and to PABP, the scaffolding protein eIF-4G1/p220 coordinates the recruitment of the different factors into the initiation complex [1]

Methods
Results
Discussion
Conclusion
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