Abstract

Autophagy is a conserved cellular process that functions as a first-line defense to restrict the growth of invading parasitic bacteria. As an intracellular pathogen, Salmonella (S) Typhimurium invades host cells through two Type III secretion systems (T3SS) and resides in the Salmonella-containing vacuole (SCV). When the SCV membrane is perforated and ruptured by T3SS-1, a small portion of the Salmonella egresses from the SCV and replicates rapidly in the nutrient-rich cytosol. Cytosolic Salmonella and those residing in the membrane-damaged SCV are tagged by ubiquitination and marked for autophagy through the ubiquitin-binding adaptor proteins such as p62, NDP52, and optineurin. Prior studies suggest that transient intracellular amino-acid starvation and subsequent inactivation of the mechanistic target of rapamycin (mTOR), a key molecule that phosphorylates Unc-51 like autophagy activating kinase (ULK1) and inhibits its activity, can trigger autophagy in S. Typhimurium-infected cells. Other studies suggest that energy stress in S. Typhimurium-infected cells leads to AMP-activated protein kinase (AMPK) activation and autophagy. In the present study, we report that autophagy was rapidly induced in S. Typhimurium-infected cells, as evidenced by increased LC3 lipidation and decreased p62 levels. However, S. Typhimurium infection drastically increased AKT phosphorylation but decreased S6K1T389, 4E-BPT37/46, and ULK1S757 phosphorylation, suggesting that mTOR activation by AKT is subverted. Further studies showed that AMPK was activated in S. Typhimurium-infected cells, as evidenced by increased ULK1S317 and ACCS79 phosphorylation. AMPK activation was mediated by Toll-like receptor-activated TAK1. Functional studies revealed that AMPK and TAK1 inhibitors accelerated S. Typhimurium growth in HeLa cells. Our results strongly suggest that TAK1 activation leads to AMPK activation, which activates ULK1 by phosphorylating ULK1S317 and suppressing mTOR activity and ULK1S757 phosphorylation. Our study has unveiled a previously unrecognized pathway for S. Typhimurium-induced autophagy.

Highlights

  • Autophagy is a highly conserved self-digestion process that plays a crucial role in maintaining cellular homeostasis in response to nutrient depletion or other cellular stresses such as accumulation of damaged organelles, unneeded protein aggregates, and invading microbes[1,2,3,4]

  • Our results strongly suggest that TAK1 activation leads to AMPK activation, which activates ULK1 by phosphorylating ULK1S317 and suppressing mechanistic target of rapamycin (mTOR) activity and ULK1S757 phosphorylation

  • Typhimurium to induce autophagy, we analyzed the formation of autophagosomes in RFP-GFP-LC3-transfected HeLa cells

Read more

Summary

Introduction

Autophagy is a highly conserved self-digestion process that plays a crucial role in maintaining cellular homeostasis in response to nutrient depletion or other cellular stresses such as accumulation of damaged organelles, unneeded protein aggregates, and invading microbes[1,2,3,4]. Autophagy is controlled by mTOR and AMP-activated protein kinase (AMPK), two nutrient- and energy-sensitive kinases[5]. Liu et al Cell Death and Disease (2018)9:570. These two kinases phosphorylate ULK1/2 at different serine residues and have the opposite effect on ULK activity: mTOR phosphorylates ULK1 at serine 757 (ULK1S757) and inhibits its activity[5,6,7], whereas AMPK phosphorylates. ULK1 at multiple sites, including the serine residues 317, 555, and 777, and activates its activity[6,7,8,9,10]. ATG13 and FIP200 proteins to form a preinitiation complex, which controls the activation of the initiation complex that comprises Beclin 1, ATG14L, VPS34, and VSP1511–13. Microbial proteins and the components of host cell signaling pathways can regulate TAK1 activity[16]

Methods
Results
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