<<

Supporting Information for

Real-time tracking of complex ubiquitination cascades using a fluorescent confocal on-bead assay

Joanna Koszela*, Nhan T Pham, David Evans, Stefan Mann, Irene Perez-Pi, Steven Shave,

Derek F J Ceccarelli, Frank Sicheri, Mike Tyers, Manfred Auer*

Supporting Methods

Inhibitors

BAY 11-7082, and heclin were purchased from Sigma Aldrich (cat. no B5556,

C7291 and SML1396, respectively). PYR-41 was purchased from Calbiochem (cat. no

662105) and CC0651 was provided by the Tyers group. The masses and purity of compounds were confirmed by analytical HPLC and LC/MS.

Proteins

The following were purchased from BostonBiochem: wild type (cat. no U-

100H); untagged human recombinant ubiquitin activating (UBE1) (cat. no E-304); N- terminally His6-tagged human recombinant ubiquitin activating enzyme (UBE1) (cat. no E-

305); human recombinant E6AP (UBE3A) with a C-terminal His6-tag (cat. no E3-230).

Protein expression and purification

His6-tagged Ube2L3 was expressed from pET28a-LIC (provided by the Sicheri group) in

BL21(DE3) E. coli. 1 L cultures were grown at 37°C in LB and induced with 0.5 mM isopropyl

β-D-1-thiogalactopyranoside (IPTG) at 20°C overnight. Cells were harvested by centrifugation at 4,000 rpm, 4°C and lysed in lysis buffer (50 mM sodium phosphate buffer pH 8, 400 mM NaCl, 10 mM , 10 mM β-mercaptoethanol, inhibitor cocktail

(Roche) with lysozyme (Sigma-Aldrich) and Benzonase Nuclease (Sigma-Aldrich, cat. no

E8263)) and sonicated on ice for 1 min in 10 second pulses at 50% of maximum power

1

(Sonic VibraCell). The lysate was cleared by centrifugation at 4,000 rpm, 4°C and incubated in rotation with pre-equilibrated 2 to 4 mL of 50% slurry Ni2+ NTA beads (Qiagen) for 2 hours at 4°C prior to standard -tag purification according to manufacturer’s instructions. Proteins were eluted in lysis buffer containing 300 mM imidazole. To obtain untagged Ube2L3, the His6-tag was removed with thrombin. Protein purity was analysed by

SDS-PAGE and by analytical HPLC. Protein concentration was measured by UV-Vis

(NanoDrop, ThermoScientific) based on absorption at 280 nm and stray light correction at

330 nm.

Expression, purification and labelling of Cys0-ubiquitin

Cys0-Ub (ubiquitin with an N-terminal insertion) was expressed as TEV-cleavable histidine6, -S-transferase (GST) fusion from pETM-30 plasmid (provided by the

Sicheri lab) in BL21(DE3) E. coli. Harvested cells were lysed and purified on Ni2+NTA resin as above, then the tag was cleaved overnight with TEV protease in 10 mL elution buffer with

2 mM DTT. Contaminants, TEV and cleaved tag were removed by perchloric acid precipitation (pH 4.5) and centrifugation at 4°C, 14,000 rpm. Cys0-Ub was then dialyzed into

50 mM NaOAc pH 4.5 and purified on a 5 mL MonoS column (Pharmacia Biotech) using

ÄKTA Explorer FPLC purification system (Amersham Pharmacia Biotech). Elution volumes of 0.5 mL were analyzed by SDS-PAGE and pure fractions were pooled and dialyzed into 20 mM HEPES, 100 mM NaCl, 0.5 mM TCEP pH 7.0. The concentration was measured by UV-

Vis (NanoDrop, ThermoScientific) and adjusted to 500 μM using Amicon-Ultra filter spin columns with 3 kDa cutoff (Merck Millipore).

For labelling, maleimide-Cy5 (GE Healthcare) was dissolved under argon in 100% dry

DMSO to obtain 50 mM stock solution, aliquoted and kept at -20°C. Labelling reactions were performed in 100 μL volumes. 300 μM of Cys0-Ub was incubated with 2.5 mM maleimide-

Cy5 on a shaking platform for 3 hours at 25°C, then purified by size exclusion chromatography over a PD10 column (GE Healthcare). Elution fractions were analysed by

HPLC and the fractions labelled Cy5-Ub with <1% free Cy5-maleimide were pooled together.

2

The purity of the sample was assessed by SDS-PAGE and the Cy5-ubiquitin concentration was measured by UV-Vis (NanoDrop, ThermoScientific) based on Cy5 absorption at 649 nm, prior to being aliquoted and stored at -80°C.

Western Blots

The proteins (E1, E2s, ubiquitin) were run on 12% Bolt Bis-Tris Plus SDS-PAGE gels

(ThermoFisher Scientific), transferred to a 0.2 μm nitrocellulose membrane (BioRad) and incubated with mouse anti-6His-tag antibody (1:500, Roche, cat. no 11 922 416 001), mouse anti-Ub antibody (1:1000, Enzo, cat. no ADI-SPA-203) or rabbit anti-Ube1 antibody (1:1000,

Enzo, cat. no BML-PW8395) and infrared dye-coupled goat anti-mouse or anti-rabbit secondary antibody (1:20,000, LI-COR, cat. no 925-32210 and 925-32211). Blots were scanned using an infrared imager (Odyssey, LI-COR Biosciences) at 800 nm.

Gel-based Ube1 activity assay

200 nM Ube1 in 10 μL volumes was pre-incubated with compounds in indicated concentrations or DMSO as control for 15 minutes in 50 mM Tris pH 7.5, 5 mM MgCl2 before adding 5 μM WT Ub and 1 μM ATP. The reactions were incubated at 30°C for 5 minutes and stopped by adding SDS loading buffer. The samples were resolved by SDS-PAGE and visualised by Coomassie staining (SimplyBlueTM, Invitrogen), or by Western blotting with anti-Ube1 and anti-Ub antibodies.

3

Supporting Information Figures

Figure S1: Ring fluorescence intensity is linearly proportional to the amount of bead- bound fluorescent substrate and is consistent across a test plate.

(a) His6-tagged emGFP-Ube2C fusion in different amounts (0-10 pmoles per well) was conjugated to Ni2+NTA agarose beads as described in the Methods. After washes, the emGFP-Ube2C beads were placed in a 384-well plate and imaged using the confocal scanning microscope OperaTM (Perkin Elmer). The ring emGFP fluorescence intensity was averaged for each well. Detected emGFP intensity increased linearly to the expected amount of the bead-bound emGFP-Ube2C. (b) A batch of His6-tagged emGFP beads was prepared at a concentration of 250 nM (5 pmoles per well) and distributed into 96 wells across a 384- well plate. Images were acquired on the OperaTM (Perkin Elmer). The detected emGFP intensity is consistent across the plate. (c) Frequency of wells with a given mean of emGFP intensity follows a normal distribution.

4

Figure S2: On-bead ubiquitination of a protein substrate, p53.

The tumour suppressor protein p53 tagged with His6 on the N-terminus (BostonBiochem, cat. no SP-40) was immobilized on Ni2+NTA agarose beads as described in the Methods. 150 pmoles of p53 was used per well. Washed p53 beads were placed in a 384-well plate and incubated with 500 nM FITC-Ub (LifeTechnologies, cat. no PV4378), 100 nM Ube1 (BPS Bioscience, cat. no 80301), 2.5 µM Ube2D2 (BostonBiochem, cat. no E2-622), 84 nM

(Millipore, cat. no 23-032) in energy-regeneration buffer (50 mM Tris-HCl, pH 7.5, 5 mM MgCl2, 10 mM phosphate, 3.5 U/mL creatine phosphokinase), in the absence or presence of ATP. After 6 h incubation at room temperature, the imaged were acquired on the OperaTM (Perkin Elmer) using FITC detection settings and the images were processed using ImageJ software. Bright rings appear upon addition of ATP, corresponding to FITC-Ub conjugation to the on-bead p53.

Figure S3: Ring fluorescence intensity is stable over time and remains linearly proportional to the amount of bead-bound fluorescent substrate.

(a) His6-tagged emGFP-Ube2C fusion in different amounts (0-20 pmoles per well) was bound to Ni2+NTA agarose beads as described in the Methods. After washes, the emGFP beads were placed in a 384-well plate and imaged using the OperaTM microscope (Perkin Elmer). The emGFP fluorescence intensity was averaged for each well. Detected emGFP intensity remained stable over 24 h of imaging for amounts up to 10 pmoles/well and was proportional to the amount of the bead-bound emGFP (b).

5

Figure S4: Fluorescence excitation and emission spectra for simultaneous detection of three different fluorescent-fusion proteins.

(a) Excitation spectra for mTurquise2 (mTurq2, blue), emGFP (green) and E2-Crimson (red) are represented, together with excitation wavelengths (black spikes) used on the confocal fluorescence scanning microscope OperaTM (Perkin Elmer) for excitation of these proteins: 445, 488 and 561 nm, respectively. (b) Emission spectra of mTurq2, emGFP and E2-Crimson were overlaid with corresponding emission filters on the OperaTM (Perkin Elmer) used for detection of these proteins: Camera 3: 475/34 nm, Camera 1: 520/35 nm and Camera 2: 660/150 nm.

6

Figure S6: Bead detection and analysis allows for differential detection of mixed small and large bead populations. emGFP was immobilised on small (40-70 µm) or large (100-120 µm) beads as described in the Methods and mixed with large or small (respectively) blank beads in the same well, resulting in a mixed bead populations: fluorescent large beads and blank small beads (a and c) or blank large beads and fluorescent small beads (b and d). Images were acquired in the brightfield and GFP detection channels using the OperaTM microscope (Perkin Elmer) and submitted to bead detection and analysis as described in Methods. When the parameters were set for detection of large beads (a and b), large beads were detected, either from the GFP detection image (GFP on large beads, red circles) or from the brightfield image (large blank beads, blue circles). Similarly, when the parameters were set for detection of small beads (a and b), small beads were detected from the GFP detection image (GFP on small beads, red circles) or from the brightfield image (small blank beads, blue circles). Distribution of detected bead intensity corresponding to each well and to detection settings as indicated is represented in the charts: high GFP intensity for GFP on large beads, large beads detection (a) and for GFP on small beads, small bead detection (d); null to low GFP intensity for GFP on small beads, large beads detection (b) and for GFP on large beads, small beads detection (c).

7

In solution (besides Reaction On bead (His6-tagged) Modifier type Ub/ULM) FITC-Ub, Cy5-Ub, E1 loading with ubiquitin Ube1 TMR-Ub Ube2C, Ube2D1, Ube2D2, Ube2D4, Ube2E2, Ube2G1, Ube2G2, Ube2H, Ube2K, Ube2L3, E2 charging with ubiquitin Ube1 FITC-Ub or Cy5-Ub Ube2N, Ube2R1, Ube2R2, Ube2S, Ube2U, Ube2W E2 charging with NEDD8 Ube2M Uba3/NAE1 FITC-NEDD8 emGFP or mTurq2 fusions: Ube2C, Ube2D1, Ube2D4, Ube2E2, Ube2G1, Ube2G2, Ube2H, Ratiometric E2 charging with ubiquitin Ube1 Cy5-Ub Ube2K, Ube2L3, Ube2L6, Ube2Q2, Ube2R1, Ube2R2, Ube2S, Ube2W HECT E3 charging/autoubiqutination E6AP, CHIP Ube1, Ube2L3 Cy5-Ub Substrate ubiquitination p53 Ube1, Ube2D3, Mdm2 FITC-Ub 2-step reaction: ubiquitin activation and Ube1 Ube2R1, Ube2L3 FITC-Ub, Cy5-Ub transfer to E2 3-step reaction: ubiquitin activation, Ube1 and E6AP Ube2L3 Cy5-Ub transfer to an E2 and transfer to a HECT E3

Ubiquitin binding Ube2R1 CC0651 Cy5-Ub

Cy5-Ub, FITC- Cross-pathway reactivity Ube2R1 or Ube2M Ube1, Uba3/NAE1 NEDD8 Ube1 PYR-41 FITC-Ub

8

Ube2M Uba3/NAE1, MLN4924 FITC-NEDD8 Ube2R1 CC0651 Cy5-Ub Chemical modulators of ubiquitin-like Ube1, BAY 11-7082, Various E2s Cy5-Ub reactions CC0651 Ube1, Ube2D3, Mdm2, p53 FITC-Ub nutlin-3 Screening for chemical modulators Ube2C Ube1, >2000 compounds Cy5-Ub SAR Ube2C Ube1, ~100 analogues Cy5-Ub Ube2C, other E2s; Ube1 Ube1, hit compounds Cy5-Ub Ube1, BAY 11-7082, Various E2s Cy5-Ub Evaluation of compound specificity CC0651 Ube2L3, PYR-41, BAY 11- Ube1 and E6AP Cy5-Ub 7082, clomipramine, heclin Evaluation of activity of a new chemical Ube1, Ube2L3 Ube1, Ube2L3, probe Cy5-Ub probe

Supporting Information Table 1: UPS-CONA is a broadly applicable assay. Listed are different ubiquitin-related reactions which have been tested using UPS-CONA. Various /substrates were immobilized on bead to monitor conjugation of Ub or ULM with fluorescent labels including Cy5, TMR and FITC. Reactions also included testing inhibitors against ubiquitination enzymes.

9