#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Activation of Focal Adhesion Kinase by Suppresses Autophagy via an Akt/mTOR Signaling Pathway and Promotes Bacterial Survival in Macrophages


Salmonella enterica is a food- and water-borne pathogen that has evolved closely with vertebrate hosts. Two medically relevant serovars include S. typhimurium, which causes gastroenteritis and S. typhi, which is the causative agent of typhoid fever. Host cells can utilize a process called autophagy, normally involved in the elimination of defective proteins and organelles, to capture and degrade intracellular pathogens. Enteric Salmonella express numerous virulence factors that enable the bacterium to subvert host defense mechanisms. Here we report that Salmonella specifically activates the host molecule focal adhesion kinase (FAK) in macrophages, triggering a signaling cascade that suppresses the autophagic elimination of intracellular bacteria. A key regulator of autophagy in mammalian cells is the target of rapamycin, mTOR, which transmits inhibitory signals that downregulate the autophagic response. We show that Salmonella-induced FAK activation leads to the Akt-dependent activation of mTOR, thereby repressing autophagic signaling. Inhibition of autophagy results in increased bacterial survival, while in FAK-deficient cells, autophagy is enhanced and intracellular Salmonella are eliminated. We also show that in mice lacking macrophage-specific FAK, animals were less susceptible to oral Salmonella infection. Together, these data identify FAK as a novel regulator of autophagy in macrophages with broad implications for host survival.


Vyšlo v časopise: Activation of Focal Adhesion Kinase by Suppresses Autophagy via an Akt/mTOR Signaling Pathway and Promotes Bacterial Survival in Macrophages. PLoS Pathog 10(6): e32767. doi:10.1371/journal.ppat.1004159
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004159

Souhrn

Salmonella enterica is a food- and water-borne pathogen that has evolved closely with vertebrate hosts. Two medically relevant serovars include S. typhimurium, which causes gastroenteritis and S. typhi, which is the causative agent of typhoid fever. Host cells can utilize a process called autophagy, normally involved in the elimination of defective proteins and organelles, to capture and degrade intracellular pathogens. Enteric Salmonella express numerous virulence factors that enable the bacterium to subvert host defense mechanisms. Here we report that Salmonella specifically activates the host molecule focal adhesion kinase (FAK) in macrophages, triggering a signaling cascade that suppresses the autophagic elimination of intracellular bacteria. A key regulator of autophagy in mammalian cells is the target of rapamycin, mTOR, which transmits inhibitory signals that downregulate the autophagic response. We show that Salmonella-induced FAK activation leads to the Akt-dependent activation of mTOR, thereby repressing autophagic signaling. Inhibition of autophagy results in increased bacterial survival, while in FAK-deficient cells, autophagy is enhanced and intracellular Salmonella are eliminated. We also show that in mice lacking macrophage-specific FAK, animals were less susceptible to oral Salmonella infection. Together, these data identify FAK as a novel regulator of autophagy in macrophages with broad implications for host survival.


Zdroje

1. FigueiraR, HoldenDW (2012) Functions of the Salmonella pathogenicity island 2 (SPI-2) type III secretion system effectors. Microbiology 158: 1147–1161.

2. MoestTP, MeresseS (2013) Salmonella T3SSs: successful mission of the secret(ion) agents. Curr Opin Microbiol 16: 38–44.

3. LevineB (2005) Eating oneself and uninvited guests: autophagy-related pathways in cellular defense. Cell 120: 159–162.

4. CemmaM, BrumellJH (2012) Interactions of pathogenic bacteria with autophagy systems. Curr Biol 22: R540–545.

5. LevineB, MizushimaN, VirginHW (2011) Autophagy in immunity and inflammation. Nature 469: 323–335.

6. DereticV, SaitohT, AkiraS (2013) Autophagy in infection, inflammation and immunity. Nat Rev Immunol 13: 722–737.

7. IntoT, InomataM, TakayamaE, TakigawaT (2012) Autophagy in regulation of Toll-like receptor signaling. Cell Signal 24: 1150–1162.

8. DelgadoMA, DereticV (2009) Toll-like receptors in control of immunological autophagy. Cell Death Differ 16: 976–983.

9. ShiCS, KehrlJH (2008) MyD88 and Trif target Beclin 1 to trigger autophagy in macrophages. J Biol Chem 283: 33175–33182.

10. SanjuanMA, DillonCP, TaitSW, MoshiachS, DorseyF, et al. (2007) Toll-like receptor signalling in macrophages links the autophagy pathway to phagocytosis. Nature 450: 1253–1257.

11. HuangJ, CanadienV, LamGY, SteinbergBE, DinauerMC, et al. (2009) Activation of antibacterial autophagy by NADPH oxidases. Proc Natl Acad Sci U S A 106: 6226–6231.

12. GongL, CullinaneM, TreeratP, RammG, PrescottM, et al. (2011) The Burkholderia pseudomallei type III secretion system and BopA are required for evasion of LC3-associated phagocytosis. PLoS One 6: e17852.

13. MehtaP, HenaultJ, KolbeckR, SanjuanMA (2014) Noncanonical autophagy: one small step for LC3, one giant leap for immunity. Curr Opin Immunol 26C: 69–75.

14. FloreyO, KimSE, SandovalCP, HaynesCM, OverholtzerM (2011) Autophagy machinery mediates macroendocytic processing and entotic cell death by targeting single membranes. Nat Cell Biol 13: 1335–1343.

15. HenaultJ, MartinezJ, RiggsJM, TianJ, MehtaP, et al. (2012) Noncanonical autophagy is required for type I interferon secretion in response to DNA-immune complexes. Immunity 37: 986–997.

16. KimM, ParkHL, ParkHW, RoSH, NamSG, et al. (2013) Drosophila Fip200 is an essential regulator of autophagy that attenuates both growth and aging. Autophagy 9: 1201–1213.

17. BirminghamCL, BrumellJH (2006) Autophagy recognizes intracellular Salmonella enterica serovar Typhimurium in damaged vacuoles. Autophagy 2: 156–158.

18. BirminghamCL, SmithAC, BakowskiMA, YoshimoriT, BrumellJH (2006) Autophagy controls Salmonella infection in response to damage to the Salmonella-containing vacuole. J Biol Chem 281: 11374–11383.

19. ThurstonTL, RyzhakovG, BloorS, von MuhlinenN, RandowF (2009) The TBK1 adaptor and autophagy receptor NDP52 restricts the proliferation of ubiquitin-coated bacteria. Nat Immunol 10: 1215–1221.

20. ZhengYT, ShahnazariS, BrechA, LamarkT, JohansenT, et al. (2009) The adaptor protein p62/SQSTM1 targets invading bacteria to the autophagy pathway. J Immunol 183: 5909–5916.

21. CemmaM, KimPK, BrumellJH (2011) The ubiquitin-binding adaptor proteins p62/SQSTM1 and NDP52 are recruited independently to bacteria-associated microdomains to target Salmonella to the autophagy pathway. Autophagy 7: 341–345.

22. WildP, FarhanH, McEwanDG, WagnerS, RogovVV, et al. (2011) Phosphorylation of the autophagy receptor optineurin restricts Salmonella growth. Science 333: 228–233.

23. TattoliI, PhilpottDJ, GirardinSE (2012) The bacterial and cellular determinants controlling the recruitment of mTOR to the Salmonella-containing vacuole. Biol Open 1: 1215–1225.

24. GutierrezMG, MasterSS, SinghSB, TaylorGA, ColomboMI, et al. (2004) Autophagy is a defense mechanism inhibiting BCG and Mycobacterium tuberculosis survival in infected macrophages. Cell 119: 753–766.

25. WangYH, WuJJ, LeiHY (2009) The autophagic induction in Helicobacter pylori-infected macrophage. Exp Biol Med (Maywood) 234: 171–180.

26. OwenKA, PixleyFJ, ThomasKS, Vicente-ManzanaresM, RayBJ, et al. (2007) Regulation of lamellipodial persistence, adhesion turnover, and motility in macrophages by focal adhesion kinase. J Cell Biol 179: 1275–1287.

27. HaraT, TakamuraA, KishiC, IemuraS, NatsumeT, et al. (2008) FIP200, a ULK-interacting protein, is required for autophagosome formation in mammalian cells. J Cell Biol 181: 497–510.

28. LimST, ChenXL, LimY, HansonDA, VoTT, et al. (2008) Nuclear FAK promotes cell proliferation and survival through FERM-enhanced p53 degradation. Mol Cell 29: 9–22.

29. SandilandsE, SerrelsB, WilkinsonS, FrameMC (2012) Src-dependent autophagic degradation of Ret in FAK-signalling-defective cancer cells. EMBO Rep 13: 733–740.

30. ChenLM, KanigaK, GalanJE (1996) Salmonella spp. are cytotoxic for cultured macrophages. Mol Microbiol 21: 1101–1115.

31. BrownJ, WangH, HajishengallisGN, MartinM (2011) TLR-signaling networks: an integration of adaptor molecules, kinases, and cross-talk. J Dent Res 90: 417–427.

32. MonackDM, DetweilerCS, FalkowS (2001) Salmonella pathogenicity island 2-dependent macrophage death is mediated in part by the host cysteine protease caspase-1. Cell Microbiol 3: 825–837.

33. Slack-DavisJK, MartinKH, TilghmanRW, IwanickiM, UngEJ, et al. (2007) Cellular characterization of a novel focal adhesion kinase inhibitor. J Biol Chem 282: 14845–14852.

34. WangS, BassonMD (2011) Akt directly regulates focal adhesion kinase through association and serine phosphorylation: implication for pressure-induced colon cancer metastasis. Am J Physiol Cell Physiol 300: C657–670.

35. ThurstonTL, WandelMP, von MuhlinenN, FoegleinA, RandowF (2012) Galectin 8 targets damaged vesicles for autophagy to defend cells against bacterial invasion. Nature 482: 414–418.

36. KageyamaS, OmoriH, SaitohT, SoneT, GuanJL, et al. (2011) The LC3 recruitment mechanism is separate from Atg9L1-dependent membrane formation in the autophagic response against Salmonella. Mol Biol Cell 22: 2290–2300.

37. TattoliI, SorbaraMT, VuckovicD, LingA, SoaresF, et al. (2012) Amino acid starvation induced by invasive bacterial pathogens triggers an innate host defense program. Cell Host Microbe 11: 563–575.

38. MartinezJ, AlmendingerJ, OberstA, NessR, DillonCP, et al. (2011) Microtubule-associated protein 1 light chain 3 alpha (LC3)-associated phagocytosis is required for the efficient clearance of dead cells. Proc Natl Acad Sci U S A 108: 17396–17401.

39. LamGY, CemmaM, MuiseAM, HigginsDE, BrumellJH (2013) Host and bacterial factors that regulate LC3 recruitment to Listeria monocytogenes during the early stages of macrophage infection. Autophagy 9: 985–995.

40. FujitaN, YoshimoriT (2011) Ubiquitination-mediated autophagy against invading bacteria. Curr Opin Cell Biol 23: 492–497.

41. SrikanthCV, CherayilBJ (2007) Intestinal innate immunity and the pathogenesis of Salmonella enteritis. Immunol Res 37: 61–78.

42. SrikanthCV, WallDM, Maldonado-ContrerasA, ShiHN, ZhouD, et al. (2010) Salmonella pathogenesis and processing of secreted effectors by caspase-3. Science 330: 390–393.

43. KnodlerLA, FinlayBB, Steele-MortimerO (2005) The Salmonella effector protein SopB protects epithelial cells from apoptosis by sustained activation of Akt. J Biol Chem 280: 9058–9064.

44. CooperKG, WinfreeS, Malik-KaleP, JollyC, IrelandR, et al. (2011) Activation of Akt by the bacterial inositol phosphatase, SopB, is wortmannin insensitive. PLoS One 6: e22260.

45. KnodlerLA, WinfreeS, DrecktrahD, IrelandR, Steele-MortimerO (2009) Ubiquitination of the bacterial inositol phosphatase, SopB, regulates its biological activity at the plasma membrane. Cell Microbiol 11: 1652–1670.

46. KuijlC, SavageND, MarsmanM, TuinAW, JanssenL, et al. (2007) Intracellular bacterial growth is controlled by a kinase network around PKB/AKT1. Nature 450: 725–730.

47. CollinsCA, De MaziereA, van DijkS, CarlssonF, KlumpermanJ, et al. (2009) Atg5-independent sequestration of ubiquitinated mycobacteria. PLoS Pathog 5: e1000430.

48. CodognoP, MehrpourM, Proikas-CezanneT (2012) Canonical and non-canonical autophagy: variations on a common theme of self-eating? Nat Rev Mol Cell Biol 13: 7–12.

49. ShahnazariS, YenWL, BirminghamCL, ShiuJ, NamolovanA, et al. (2010) A diacylglycerol-dependent signaling pathway contributes to regulation of antibacterial autophagy. Cell Host Microbe 8: 137–146.

50. HueckCJ, HantmanMJ, BajajV, JohnstonC, LeeCA, et al. (1995) Salmonella typhimurium secreted invasion determinants are homologous to Shigella Ipa proteins. Mol Microbiol 18: 479–490.

51. GuyRL, GoniasLA, SteinMA (2000) Aggregation of host endosomes by Salmonella requires SPI2 translocation of SseFG and involves SpvR and the fms-aroE intragenic region. Mol Microbiol 37: 1417–1435.

52. LeeCA, FalkowS (1990) The ability of Salmonella to enter mammalian cells is affected by bacterial growth state. Proc Natl Acad Sci U S A 87: 4304–4308.

53. CrissAK, AhlgrenDM, JouTS, McCormickBA, CasanovaJE (2001) The GTPase Rac1 selectively regulates Salmonella invasion at the apical plasma membrane of polarized epithelial cells. J Cell Sci 114: 1331–1341.

54. OwenKA, AbshireMY, TilghmanRW, CasanovaJE, BoutonAH (2011) FAK regulates intestinal epithelial cell survival and proliferation during mucosal wound healing. PLoS One 6: e23123.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 6
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#