Materials.
HEK293T and HeLa cell lines were kindly provided by Prof. Junying Yuan from Interdisciplinary Research Center on Biology and Chemistry, CAS, Shanghai, China. The full length human WIPI2b, ATG8s, ATG16L1 and FIP200 genes were obtained from Prof. Jiahuai Han from School of Life Sciences, Xiamen University, Xiamen, China. The synthetic peptide "EQDDDIEVIVDET" (ATG16L1 FIR) was purchased from the China Peptides company, and the purity of the commercially synthesized peptides was > 98%.
Protein expression and purification.
The DNA fragments encoding human FIP200 (residues 1490–1594) and WIPI2b (residues 13–362 without 265–297) were cloned into pET-SUMO-3C vector or pET-32M-3C vector (modified versions of pET-32a vector containing an N-terminal SUMO or Trx tag). The DNA fragments encoding ATG16L1 (residues 207–247, 78–235, 235–247 and 78–247) were all cloned into pACYC-Trx1-3C vector (a modified version of pACYC vector containing an N-terminal Trx tag). Meanwhile, the DNA fragments encoding six human ATG8s were all cloned into pET-32M-3C vector or pET-GST-3C vector (modified versions of pET-32a vector containing an N-terminal GST tag). Of note, 6xHis was placed on either the N-terminal or the C-terminal of target proteins. For Co-immunoprecipitation assays, the DNA fragments encoding human GABARAPL1(1-115) and full-length FIP200 were separately cloned into pEGFP-C1 vector and pmEGFP-C1 vector (a modified version of pEGFP-C1 with an A206K mutation that monomerizes EGFP) as well as full-length ATG16L1 into pFlag-CMV-2 vector. All point mutations of WIPI2b, FIP200, GABARAPL1 and ATG16L1 used in this study were generated through standard PCR-based mutagenesis method and further confirmed by DNA sequencing.
Recombinant proteins were all expressed in BL21 (DE3) E. coli cells induced by 200 µM IPTG overnight at 16°C. The bacterial cell pellets were re-suspended in the binding buffer (50 mM Tris, 500 mM NaCl, and 5 mM imidazole at pH 7.9), and then lysed by the ultrahigh pressure homogenizer FB-110XNANO homogenizer machine (Shanghai Litu Machinery Equipment Engineering Co., Ltd.). Then, the lysate was spun down by centrifuge at 17000 rpm (35000 g) for 35 minutes to remove the pellets fractions. All proteins were purified by Ni2+-NTA agarose (GE Healthcare) affinity chromatography and further purified by size-exclusion chromatography (Superdex 75 or 200 26/60 column; GE Healthcare) equilibrated with the column buffer containing 20 mM Tris, 100 mM NaCl, 1 mM DTT and 1 mM EDTA at pH 7.5. To obtain the FIP200 Claw/ATG16L1 FIR complex used for crystallization, the FIP200/ATG16L1 complex was obtained through the co-expression of Trx-FIP200(1490–1594) and Trx-ATG16L1(235–247). The N-terminal Trx tags of relevant FIP200 and ATG16L1 proteins were cleaved by 3C protease and removed by HisTrap excel column (GE Healthcare). Finally, the FIP200 Claw/ATG16L1 FIR complex and the GABARAPL1/ATG16L1 FIR complex were further purified through Superdex 75 column equilibrated with the aforementioned column buffer. Special for six human ATG8 family proteins, their Trx tags were removed by MonoQ 10/10 ion-exchange column (GE Healthcare). Meanwhile, uniformly 15N-labelled GABARAPL1 proteins were prepared by growing bacteria in M9 minimal medium using 15NH4Cl (Cambridge Isotope Laboratories Inc.) as the sole nitrogen source.
ITC assay.
ITC measurements were all carried out on a MicroCal PEAQ-ITC (Malvern) calorimeter at 25°C. All protein samples were prepared in the same buffer containing 20 mM Tris, 100 mM NaCl, 1 mM DTT at pH 7.5. For each ITC experiment in this study, the concentrated (~ 50 µM) proteins were loaded into the cell, and the other titrated proteins (~ 500 µM) were loaded into the syringe. The titration processes were performed by injecting proteins from syringe into the cell at time intervals of 2 minutes to ensure that the titration peak returned to the baseline. The titration data were analyzed using the Malvern MicroCal PEAQ-ITC analysis program and fitted using the one-site binding model.
Size exclusion chromatography.
Size exclusion chromatography was carried out on an AKTA FPLC system (GE Healthcare). Purified proteins were loaded on to a Superdex 200 or 75 increase 10/300 GL column (GE Healthcare) equilibrated with the same column buffer. The fitting results were further output to the Origin 9 software and aligned with each other.
NMR spectroscopy.
The 15N-labelled protein samples for NMR titration experiments were concentrated to ~ 0.1 mM. All the protein samples for NMR studies were prepared in the 50 mM potassium phosphate buffer containing 100 mM NaCl and 1 mM DTT at pH 6.5, and NMR spectra were acquired at 25°C on an Agilent 800 MHz spectrometer equipped with an actively z gradient shielded triple resonance cryogenic probe at the Shanghai Institute of Organic Chemistry.
Multi-angle light scattering.
For multi-angle light-scattering measurement, the FIP200(1490–1594)/ATG16L1(78–247) complex samples were injected into an AKTA FPLC system (GE Healthcare) with a Superdex 200 increase 10/300 GL column (GE Healthcare) with the same column buffer mentioned before. The chromatography system was coupled to a static light scattering detector (miniDawn, Wyatt Technology) and a differential refractive index detector (Optilab, Wyatt Technology). Data were collected every 0.5 s with a flow rate of 0.5 mL/min. Data were analyzed using the ASTRA 6 software (Wyatt Technology) and drawn using the Origin 9 software.
Fluorescence polarization assay.
Fluorescence anisotropy binding assays were performed on the SpectraMax i3x Multi-Mode Detection Platform from Molecular Devices, using a 485 nm excitation filter and a 535 nm emission filter. Peptides were labeled with fluorescein isothiocyanate isomer I (Sigma-Aldrich) at their N-terminal NH2. In this assay, the FITC-labeled peptide (~ 0.3 µM) was titrated with increasing amount of testing proteins in the column buffer at 25 °C. The Kd values were obtained by fitting the titration curves with the classical one-site binding model using GraphPad Prism 9 software.
Protein crystallization and structural elucidation.
Crystals of the FIP200(1490–1594)/ATG16L1(235–247) complex and the GABARAPL1/ATG16L1(235–247) complex were all obtained using the sitting-drop vapor-diffusion method at 16°C. The crystal-growing condition of the FIP200(1490–1594)/ATG16L1(235–247) complex (38 mg/mL) contains 10% v/v 2-Propanol, 0.1 M BICINE (pH 8.5), and 30% w/v Polyethylene glycol 1500. As for the GABARAPL1/ATG16L1(235–247) complex, the purified GABARAPL1 protein (27 mg/mL) was saturated with ATG16L1 FIR peptide with a molar ratio up to 1:10. Crystals were observed in the condition containing 0.1 M Sodium cacodylate (pH 6.5), 40% v/v MPD and 5% w/v PEG 8000. Before diffraction experiments, relevant amount of glycerol was added as the cryo-protectant. A 1.76 Å resolution X-ray data set for the FIP200(1490–1594)/ATG16L1(235–247) complex and a 1.54 Å resolution X-ray data set for the GABARAPL1/ATG16L1(235–247) complex were collected at the beamline BL19U1, BL02U1 and BL10U2 of the Shanghai Synchrotron Radiation Facility48. The diffraction data were processed upon autoPROC49. The phase problems of the FIP200/ATG16L1 complex and the GABARAPL1/ATG16L1 complex were all solved by molecular replacement method by using the FIP200 Claw structure (PDB ID: 6DCE) and the GABARAPL1 structure (PDB ID: 5LXI) respectively as the search model with PHASER50. The initial structural models were rebuilt manually using COOT51, and then refined through PHENIX52. Further manual model building and adjustments were completed via COOT 51. The qualities of the final models were validated by MolProbity53. The final refinement statistics of solved structures in this study were listed in Supplementary Table 1. All the structural diagrams were prepared using the program PyMOL (http://www.pymol.org/).
Co-immunoprecipitation assay.
Flag-tagged ATG16L1 plasmids (wild-type or mutants) were co-transfected into HEK293T cells using Lipofectamine 2000 transfection reagent (Thermo Fisher Scientific) or Lipofectamine 6000 transfection reagent (Beyotime) with mEGFP-tagged FIP200 or EGFP-tagged GABARAPL1(1-115) plasmids. Cells were collected 24 hours after transfection and lysed in ice-cold cell lysis buffer (50 mM Tris, 150 mM NaCl, 0.5% NP-40, 1 mM PMSF, 1% protease inhibitor cocktail at pH 7.5) for 20 to 40 minutes at 4°C. Lysates were centrifuged at 14500 g for 15 minutes at 4°C to separate soluble fractions and cell debris. Supernatants were applied to anti-GFP mAb-Agarose (Medical & Biological Laboratories) and incubated for 40 to 60 minutes at 4°C. The beads and non-bound proteins were separated by centrifugation at 800 g for 3 minutes at 4°C. After washing several times with the cold wash buffer (50 mM Tris, 150 mM NaCl and 0.1% or 0.5% NP-40 at pH 7.5), the beads were re-suspended with the 1X SDS-PAGE sample buffer and boiled at 65°C for 10 minutes. The prepared samples were analyzed by SDS-PAGE. The EGFP-tagged GABARAPL1(1-115), mEGFP-tagged FIP200 and Flag-tagged ATG16L1 were detected by western blot using the anti-GFP (Proteintech, 50430-2-AP, 1:1000 dilution), anti-GFP (Proteintech, 66002-1-Ig, 1:2000 dilution), anti-Flag (Proteintech, 20543-1-AP, 1:1000 dilution) and anti-Flag (Proteintech, 66008-4-Ig, 1:2000 dilution) primary antibodies.
Generation of relevant ATG16L1 stable cell lines.
The AcGFP1-tagged mutant ATG16L1 was cloned into the pMSCV-blasticidin vector and was co-transfected into HEK293T cells with VSV-G and gag/pol using Lipofectamine 6000 transfection reagent (Beyotime). Notably, the sgRNA-targeting region of ATG16L1 at pMSCV-blasticidin vector was synonymously mutated to avoid being targeted again by Cas9 enzyme. ATG16L1-knockout cells were incubated with polybrene (Sigma-Aldrich) and concentrated virus-containing medium filtered through a 0.45-µm-pore syringe filter. Transfected cells were treated with blasticidin (5 µg/mL; InvivoGen) to generate stable polyclonal cell lines.
Autophagy induction
The ATG16L1-knockout HeLa cells, which were generated in our previous study12, were rescued by lentiviral transduction with AcGFP1-tagged WT ATG16L1, ATG16L1 D239R/I240F mutant (DRIF), or ATG16L1 I240Q/I243Q mutant (IQIQ). Rescued HeLa cells were separately seeded on a six-well plate. The following day, cells were incubated for 4 hours with DMEM (Thermo Fisher Scientific) supplemented with 10% FBS (Thermo Fisher Scientific) and 1% penicillin-streptomycin (Thermo Fisher Scientific), amino acid starvation medium (BOSTER), and amino acid starvation medium with bafilomycin A1 (Selleck) at 400 nM. After starvation treatment, cells were resuspended with the 1X SDS-PAGE sample buffer and boiled for 7 minutes at 100°C. The samples were detected by Western blot using specific ATG16L1 antibody (1:1000; Abcam, catalog no. ab187671), LC3B antibody (1:1000; Abcam, catalog no. ab192890), β-actin antibody (1:5000; Proteintech, catalog no. 66009-1-lg), and p62 antibody (1:1000; Cell Signaling Technology, catalog no. #39749). The data are presented as means ± SEM from three independent experiments. Statistical analyses were performed in GraphPad Prism 9 by two-way analysis of variance (ANOVA) followed by Bonferroni multiple comparisons test, and P value style is P = 0.1234 (not significant (ns)), *P = 0.0332, **P = 0.0021, ***P = 0.0002, and ****P < 0.0001.