4,545
Views
15
CrossRef citations to date
0
Altmetric
Research Paper

RAB33B recruits the ATG16L1 complex to the phagophore via a noncanonical RAB binding protein

ORCID Icon, ORCID Icon, , , , ORCID Icon & ORCID Icon show all
Pages 2290-2304 | Received 13 May 2020, Accepted 08 Sep 2020, Published online: 22 Sep 2020

References

  • Mizushima N, Yoshimori T, Ohsumi Y. The role of Atg proteins in autophagosome formation. Annu Rev Cell Dev Biol. 2011;27:107–132.
  • Rubinsztein DC, Codogno P, Levine B. Autophagy modulation as a potential therapeutic target for diverse diseases. Nat Rev Drug Discov. 2012;11(9):709–730.
  • Yang Z, Klionsky DJ. Eaten alive: a history of macroautophagy. Nat Cell Biol. 2010;12(9):814–822.
  • Noda NN, Ohsumi Y, Inagaki F. ATG systems from the protein structural point of view. Chem Rev. 2009;109(4):1587–1598.
  • Xie Z, Klionsky DJ. Autophagosome formation: core machinery and adaptations. Nat Cell Biol. 2007;9(10):1102–1109.
  • Nakatogawa H, Ichimura Y, Ohsumi Y. Atg8, a ubiquitin-like protein required for autophagosome formation, mediates membrane tethering and hemifusion. Cell. 2007;130(1):165–178.
  • Weidberg H, Shpilka T, Shvets E, et al. LC3 and GATE-16 N termini mediate membrane fusion processes required for autophagosome biogenesis. Dev Cell. 2011;20(4):444–454.
  • Yang A, Li Y, Pantoom S, et al. Semisynthetic lipidated LC3 protein mediates membrane fusion. Chembiochem. 2013;14(11):1296–1300.
  • Dooley HC, Razi M, Polson HE, et al. WIPI2 links LC3 conjugation with PI3P, autophagosome formation, and pathogen clearance by recruiting Atg12-5-16L1. Mol Cell. 2014;55(2):238–252.
  • Gammoh N, Florey O, Overholtzer M, et al. Interaction between FIP200 and ATG16L1 distinguishes ULK1 complex-dependent and -independent autophagy. Nat Struct Mol Biol. 2013;20(2):144–149.
  • Nishimura T, Kaizuka T, Cadwell K, et al. FIP200 regulates targeting of Atg16L1 to the isolation membrane. EMBO Rep. 2013;14(3):284–291.
  • Fujita N, Morita E, Itoh T, et al. Recruitment of the autophagic machinery to endosomes during infection is mediated by ubiquitin. J Cell Biol. 2013;203(1):115–128.
  • Dudley LJ, Cabodevilla AG, Makar AN, et al. Intrinsic lipid binding activity of ATG16L1 supports efficient membrane anchoring and autophagy. Embo J. 2019;38(9):e100554.
  • Fletcher K, Ulferts R, Jacquin E, et al. The WD40 domain of ATG16L1 is required for its non-canonical role in lipidation of LC3 at single membranes. Embo J. 2018;37(4):e97840.
  • Lystad AH, Carlsson SR, de la Ballina LR, et al. Distinct functions of ATG16L1 isoforms in membrane binding and LC3B lipidation in autophagy-related processes. Nat Cell Biol. 2019;21(3):372–383.
  • Hutagalung AH, Novick PJ. Role of Rab GTPases in membrane traffic and cell physiology. Physiol Rev. 2011;91(1):119–149.
  • Stenmark H. Rab GTPases as coordinators of vesicle traffic. Nat Rev Mol Cell Biol. 2009;10(8):513–525.
  • Longatti A, Tooze SA. Vesicular trafficking and autophagosome formation. Cell Death Differ. 2009;16(7):956–965.
  • Szatmari Z, Sass M. The autophagic roles of Rab small GTPases and their upstream regulators: a review. Autophagy. 2014;10(7):1154–1166.
  • Gutierrez MG, Munafo DB, Beron W, et al. Rab7 is required for the normal progression of the autophagic pathway in mammalian cells. J Cell Sci. 2004;117(Pt 13):2687–2697.
  • Jager S, Bucci C, Tanida I, et al. Role for Rab7 in maturation of late autophagic vacuoles. J Cell Sci. 2004;117(Pt 20):4837–4848.
  • Ravikumar B, Imarisio S, Sarkar S, et al. Rab5 modulates aggregation and toxicity of mutant huntingtin through macroautophagy in cell and fly models of Huntington disease. J Cell Sci. 2008;121(Pt 10):1649–1660.
  • Dou Z, Pan JA, Dbouk HA, et al. Class IA PI3K p110beta subunit promotes autophagy through Rab5 small GTPase in response to growth factor limitation. Mol Cell. 2013;50(1):29–42.
  • Zhou F, Wu Z, Zhao M, et al. Autophagosome closure by ESCRT: vps21/RAB5-regulated ESCRT recruitment via an Atg17-Snf7 interaction. Autophagy. 2019;15(9):1653–1654.
  • Longatti A, Lamb CA, Razi M, et al. TBC1D14 regulates autophagosome formation via Rab11- and ULK1-positive recycling endosomes. J Cell Biol. 2012;197(5):659–675.
  • Puri C, Vicinanza M, Ashkenazi A, et al. The RAB11A-positive compartment is a primary platform for autophagosome assembly mediated by WIPI2 recognition of PI3P-RAB11A. Dev Cell. 2018;45(1):114–131. e118
  • Lipatova Z, Belogortseva N, Zhang XQ, et al. Regulation of selective autophagy onset by a Ypt/Rab GTPase module. Proc Natl Acad Sci U S A. 2012;109(18):6981–6986.
  • Lynch-Day MA, Bhandari D, Menon S, et al. Trs85 directs a Ypt1 GEF, TRAPPIII, to the phagophore to promote autophagy. Proc Natl Acad Sci U S A. 2010;107(17):7811–7816.
  • Zoppino FC, Militello RD, Slavin I, et al. Autophagosome formation depends on the small GTPase Rab1 and functional ER exit sites. Traffic. 2010;11(9):1246–1261.
  • Wang J, Menon S, Yamasaki A, et al. Ypt1 recruits the Atg1 kinase to the preautophagosomal structure. Proc Natl Acad Sci U S A. 2013;110(24):9800–9805.
  • Itoh T, Fujita N, Kanno E, et al. Golgi-resident small GTPase Rab33B interacts with Atg16L and modulates autophagosome formation. Mol Biol Cell. 2008;19(7):2916–2925.
  • Itoh T, Kanno E, Uemura T, et al. OATL1, a novel autophagosome-resident Rab33B-GAP, regulates autophagosomal maturation. J Cell Biol. 2011;192(5):839–853.
  • Lee MT, Mishra A, Lambright DG. Structural mechanisms for regulation of membrane traffic by rab GTPases. Traffic. 2009;10(10):1377–1389.
  • Fujioka Y, Noda NN, Nakatogawa H, et al. Dimeric coiled-coil structure of Saccharomyces cerevisiae Atg16 and its functional significance in autophagy. J Biol Chem. 2010;285(2):1508–1515.
  • Merithew E, Hatherly S, Dumas JJ, et al. Structural plasticity of an invariant hydrophobic triad in the switch regions of Rab GTPases is a determinant of effector recognition. J Biol Chem. 2001;276(17):13982–13988.
  • Chandra M, Saran R, Datta S. Deciphering the role of Atg5 in nucleotide dependent interaction of Rab33B with the dimeric complex, Atg5-Atg16L1. Biochem Biophys Res Commun. 2016;473(1):8–16.
  • Eathiraj S, Pan X, Ritacco C, et al. Structural basis of family-wide Rab GTPase recognition by rabenosyn-5. Nature. 2005;436(7049):415–419.
  • Li F, Yi L, Zhao L, et al. The role of the hypervariable C-terminal domain in Rab GTPases membrane targeting. Proc Natl Acad Sci U S A. 2014;111(7):2572–2577.
  • Wu YW, Goody RS, Abagyan R, et al. Structure of the disordered C terminus of Rab7 GTPase induced by binding to the Rab geranylgeranyl transferase catalytic complex reveals the mechanism of Rab prenylation. J Biol Chem. 2009;284(19):13185–13192.
  • Aivazian D, Serrano RL, Pfeffer S. TIP47 is a key effector for Rab9 localization. J Cell Biol. 2006;173(6):917–926.
  • Mizushima N, Yamamoto A, Hatano M, et al. Dissection of autophagosome formation using Apg5-deficient mouse embryonic stem cells. J Cell Biol. 2001;152(4):657–668.
  • Ravikumar B, Moreau K, Jahreiss L, et al. Plasma membrane contributes to the formation of pre-autophagosomal structures. Nat Cell Biol. 2010;12(8):747–757.
  • Li J, Chen Z, Stang MT, et al. Transiently expressed ATG16L1 inhibits autophagosome biogenesis and aberrantly targets RAB11-positive recycling endosomes. Autophagy. 2017;13(2):345–358.
  • Saitoh T, Fujita N, Jang MH, et al. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production. Nature. 2008;456(7219):264–268.
  • Fujita N, Itoh T, Omori H, et al. The Atg16L complex specifies the site of LC3 lipidation for membrane biogenesis in autophagy. Mol Biol Cell. 2008;19(5):2092–2100.
  • Schoebel S, Cichy AL, Goody RS, et al. Protein LidA from Legionella is a Rab GTPase supereffector. Proc Natl Acad Sci U S A. 2011;108(44):17945–17950.
  • Otomo C, Metlagel Z, Takaesu G, et al. Structure of the human ATG12~ATG5 conjugate required for LC3 lipidation in autophagy. Nat Struct Mol Biol. 2013;20(1):59–66.
  • McCoy AJ, Grosse-Kunstleve RW, Adams PD, et al. Phaser crystallographic software. J Appl Crystallogr. 2007;40(Pt 4):658–674.
  • Li X, He L, Che KH, et al. Imperfect interface of Beclin1 coiled-coil domain regulates homodimer and heterodimer formation with Atg14L and UVRAG. Nat Commun. 2012;3:662.
  • Adams PD, Afonine PV, Bunkoczi G, et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr D Biol Crystallogr. 2010;66(Pt 2):213–221.
  • Emsley P, Cowtan K. Coot: model-building tools for molecular graphics. Acta Crystallogr D Biol Crystallogr. 2004;60(Pt 12 Pt 1):2126–2132.
  • Chen VB, Arendall WB 3rd, Headd JJ, et al. MolProbity: all-atom structure validation for macromolecular crystallography. Acta Crystallogr D Biol Crystallogr. 2010;66(Pt 1):12–21.
  • Wu YW, Waldmann H, Reents R, et al. A protein fluorescence amplifier: continuous fluorometric assay for Rab geranylgeranyltransferase. Chembiochem. 2006;7(12):1859–1861.
  • Grecco HE, Roda-Navarro P, Verveer PJ. Global analysis of time correlated single photon counting FRET-FLIM data. Opt Express. 2009;17(8):6493–6508.
  • Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9(7):671–675.