General cell culture
NHEM (normal adult epidermal melanocytes) were grown in Lonza MGM-4 growth media; prior to harvest for IB analysis, the cells were switched to the same media as the other cells overnight. WM793,1205Lu, A375, WM1366, WM164, and SW1 melanoma cells were obtained from the Ronai laboratory (Sanford-Burnham Prebys Medical Discovery Institute (La Jolla, CA), WM983A/B cells were purchased from Rockland Immunochemicals (Limerick, PA). WM115 and WM266-4 cells were purchased from ATCC (Manassas, VA). SM1 (Gift from the Smalley Laboratory at Moffitt), were cultured in Dulbecco's Modified Eagle Medium containing 10% fetal bovine serum (FBS), 1 g/mL glucose, 4 mM L-glutamine in 37oC in 5% CO2. Cell lines were transfected using Lipofectamine 2000 (Invitrogen, Waltham, MA). Primary CD4 + T cells were harvested using the EasySep (StemCell Technologies) Human CD4+ negative selection isolation kit (#17952) according to manufacturer's protocols.
Antibodies
The following antibodies were used as indicated: mouse anti-V5 (0.2 µg/mL Millipore Sigma (St. Louis, MO)), mouse anti-V5 gel (V5-10, Millipore Sigma (St. Louis, MO)), mouse anti-human HLA-DRB1 (0.2 µg/mL, IF, ab215835, Abcam (Cambridge, UK)), rabbit anti-human HLA-DRB1 (0.2 µg/mL WB, ab92371, Abcam (Cambridge, UK)), β-tubulin (0.3 µg/mL, E7, developed by M. McCutcheon and S. Carroll and obtained from Developmental Studies Hybridoma Bank (University of Iowa, Iowa City, IA)), goat anti-biotin (0.1 µg/mL Vector Labs (Burlingame, CA)), biotinylated AAL (0.4 µg/mL Vector Labs, Burlingame, CA), fluorescein-conjugated AAL (0.4 µg/mL Vector Laboratories, Burlingame, CA), Agarose UEA1 and AAL (Vector Laboratories, (Burlingame, CA)), anti-mouse CD4 (20 mg/kg, for immunodepletion, GK1.5, Bioxcell (West Lebanon, NH)), anti-mouse CD8 (20 mg/kg, for immunodepletion, 2.43, Bioxcell (West Lebanon, NH)), goat anti-mouse IgGκ horseradish peroxidase (HRP) (0.04 µg/mL, Santa Cruz Biotechnology (Dallas, TX)), mouse anti-rabbit HRP (0.04 µg/mL, Santa Cruz Biotechnology (Dallas, TX)), goat anti-rabbit AlexaFluor 488 (0.04 µg/mL, ThermoFisher Scientific (Waltham, MA)),), donkey anti-mouse AlexaFluor 594 (0.05 µg/mL, ThermoFisher Scientific (Waltham, MA)), AlexaFluor 594 donkey anti-rabbit (0.05 µg/mL, ThermoFisher Scientific (Waltham, MA)), rabbit anti-Mart1 (0.2 µg/mL, Millipore Sigma (St. Louis, MO), rabbit anti-S100 (0.2 µg/mL, Agilent Technologies (Santa Clara, CA)), APC anti-mouse CD3 (0.5 µg/mL, Biolegend (San Jose, CA)), Pacific Blue anti-mouse CD4 (0.5 µg/mL, BD Biosciences (San Jose, CA)), BV785 anti-mouse CD8 (0.5 µg/mL, BD Biosciences (San Jose, CA)), FITC anti-mouse F4/80 (0.5 µg/mL, BD Biosciences (San Jose, CA)), APC anti-mouse GR-1 (0.5 µg/mL, BD Biosciences (San Jose, CA)), PeCy7 anti-mouse CD11c (0.5 µg/mL, BD Biosciences (San Jose, CA)), PE anti-mouse NK1.1(0.5 µg/mL, BD Biosciences (San Jose, CA)), PE anti-mouse DX5 (0.5 µg/mL, BD Biosciences (San Jose, CA)), PerCP-Cy5.5 anti-mouse CD11b (0.5 µg/mL, BD Biosciences (San Jose, CA)), rabbit anti-human PD-L1 (clone #NBP1-76769; Noveus Biologicals, Centennial, CO), PE rat anti-mouse PD-L1 (clone #10F.9G2; (Biolegend, (San Diego, CA)), and phalloidin Alexafluor 488 (0.2µg/mL, ThermoFisher Scientific (Waltham, MA)), mouse anti-FLAG (0.2 µg/mL, clone M2, Millipore Sigma (St. Louis, MO)), rabbit anti-HLA-A (0.2 µg/mL, Proteintech (Rosemont, IL)), normal mouse IgG (Santa Cruz Biotechnology (Dallas, TX)), rabbit anti-KDEL (0.1 µg/mL, ThermoFisher Scientific (Waltham, MA)), mouse anti-PD1 (for in vivo studies, 20 mg/kg, clone# RMP1-14 Bioxcell (West Lebanon, NH)), donkey anti-goat plus PLA secondary antibody (Millipore Sigma (St. Louis, MO)), donkey anti-mouse plus PLA secondary antibody (Millipore Sigma (St. Louis, MO)), rat anti-mouse CD8 antibody (0.2 µg/mL, ThermoFisher Scientific (Waltham, MA)), AlexaFluor 594 goat anti-rat secondary antibody (0.05 µg/mL, ThermoFisher Scientific (Waltham, MA)), anti-CD3 (0.2 µg/mL, Clone PS1, Santa Cruz Biotechnology (Dallas, TX), PE anti-pan-MHC-I (HLA-A,B,C) (BD Pharmingen (San Jose, CA), FITC anti-pan-MHC-II (HLA-DP, DQ, DQ)(BD Pharmingen (San Jose, CA), PerPCy5.5 anti-CD45 (Invitrogen (Waltham, MA)), APC anti-CD90 (Biolegend (San Diego, CA)), and BV421 anti EpCAM (Biolegend (San Diego, CA)).
Cloning and mutagenesis
Mouse fucokinase (mFuk) was cloned using cDNA from SW1 cells into pLenti-C-Myc-DDK-IRES-Puro expression vector (Origene Technologies (Rockville, MD)) into BAMHI and NHEI restriction sites. Mouse EB1 constructs was cloned using cDNA from SW1 cells into pLenti-C-Myc-DDK-IRES-Puro expression vector (Origene Technologies (Rockville, MD)) into ASCI and XHOI restriction sites. pLKO Non-targeting shRNA (shNT), pLKO shK1-1, pLKO shK1-2, pLKO shEB1-1, and pLKO shEB1-2 were obtained from Millapore Sigma (St. Louis). pLX304::EV was obtained from Origene Technologies (Rockville, MD). pLX304::HLA-A and pLX304::HLA-DRB1 constructs were obtained from DNAasu (PMID:21706014). HLA-DRB1 N48G and T129A as well as EB1 N46G mutants were generated using QuikChange II XL site-directed mutagenesis kit according to the manufacturer’s protocol (Agilent Technologies (Santa Clara, CA)).
Proteomic mass spectrometric profiling of fucosylated proteins
WM793 cells stably transduced with pLenti-GFP empty vector (EV), pLenti-FUK-GFP, or shFUK were grown in triplicate to ~ 30–40% confluence in (3 x 15 cm3 plates each). The cells were further cultured in the presence of 50µM L-fucose-alkyne for ~ 72 h to ~ 80% confluence. The cells were lysed in 1.5% N-dodecyl-beta-D-maltoside/20mM HEPES pH 7.4/protease and phosphatase inhibitors. Lysates were sonicated and cleared by centrifugation at full speed for 5 min at 4C. Lysates were acetone precipitated overnight. The pelleted proteins were resuspended and subjected to click-chemistry labeling with biotin-azide using the Click-It kit per manufacturer's protocol (Invitrogen). For negative control, pLenti-GFP-EV cells were not labeled with L-fucose-alkyne but were lysed, pelleted, and click-reacted with biotin-azide. All biotin-azide (biotinylated-fucosylated) samples were pulled down using neutravidin beads that were pre-blocked with 2% IgG-free BSA. Samples were submitted to the Sanford-Burnham Prebys proteomics core facility for on-bead digest; supernatants from on-bead digest were analyzed by LC/MS/MS. Hits that were increased by > 1.5 fold in pLenti-FUK-GFP-expressing cells and unchanged or decreased in pLenti-EV-GFP-expressing cells or decreased in pLenti-shFUK-expressing cells. Hits were subjected to Ingenuity Pathway Analysis (Qiagen).
Lectin pulldown
Control beads and AAL or UEA1 lectin-conjugated agarose beads were pre-blocked for 2 h in blocking buffer (2% IgG-Free BSA (Jackson ImmunoResearch Laboratories (Westgrove, PA)) on a rotator at 4oC. Cells were lysed on ice in 1% Triton-X100 lysis buffer (1% Triton-X100, 20mM Tris-HCl, pH 7.4, 150mM NaCl in ddH2O + protease and phosphatase inhibitors (ThermoFisher Scientific (Waltham, MA)), briefly sonicated, pelleted, and the resulting lysates were normalized in protein concentration to the sample with the lowest concentration and diluted to a final 0.25% Triton-X-100 with dilution buffer (0% Triton X-100, 20mM Tris-HCl, pH 7.4, 150mM NaCl in ddH2O + protease and phosphatase inhibitors (ThermoFisher Scientific (Waltham, MA)), and incubated with 15µl of pre-blocked beads (beads were spun out of block and resuspended in dilution buffer) and rotated overnight at 4oC. Next, the beads were washed twice with dilution buffer and subjected to (12%) SDS-PAGE and IB analysis using the indicated antibodies.
Mass spectrometric analysis of glycosylation on HLA-DRB1
Stained bands of approximately 1ug of exogenously expressed V5-HLA-DRB1 purified from WM793 cells were cut into 1-mm3 pieces and reduced and alkylated using 20mM TCEP (tris(2-carboxyehtyl)phosphine) and iodoacetamide in 50mM Tris-HCl. The gel pieces were washed in a 20mM ammonium phosphate solution with 50% methanol overnight at 4°C. The following day, the gel pieces were dehydrated for 30 minutes with 100% acetonitrile. After gel pieces were completely dry, trypsin protease solution was added to the samples (300ng trypsin). Samples were digested for 4 hours at 37°C. The digests were applied to a C-18 Zip-Tip and eluted with 50% methanol and 0.1% formic acid. Five microliters of the elution were diluted in 0.1% formic acid and then injected into a Q-Exactive Orbitrap mass spectrometer (ThermoFisher Scientific, (Waltham, MA)) equipped with an Easy nano-LC HPLC system with reverse-phase column (ThermoFisher Scientific, (Waltham, MA)). A binary gradient solvent system consisting of 0.1% formic acid in water (solvent A) an 90% acetonitrile and 0.1% formic acid in water (solvent B) was used to separate peptides. Raw data files were analyzed using both Proteome Discoverer v2.1 (ThermoFisher Scientific, (Waltham, MA)) with Byonic (Protein Metrics) as a module and Byonic standalone v2.10.5. All extracted ion chromatograms (EICs) were generated using Xcalibur Qual Browser v4.0 (ThermoFisher Scientific, (Waltham, MA)). UniProt sequence Q5Y7D1_Human was used as the reference sequence for peptide analysis.
Phosphoproteomics mass spectrometric profiling of CD4 + T cells
CD4+T cells cultured and treated as indicated in the main text were harvested and lysed in standard RIPA buffer + protease and phosphatase inhibitors. Protein concentration was estimated by BCA assay (Bio-Rad) and 1 mg lysates were subjected to trypsin digestion. Briefly, lysates were reduced with 4.5 mM dithiothreitol (DTT) for 30 min at 600C, alkylated with 10mM iodoacetamide (IAA) at room temperature in the dark for 20 minutes, and digested overnight at 370C with 1:20 enzyme-to-protein ratio of trypsin (Worthington). The resulting peptide solution was de-salted using reversed-phase Sep-Pak C18 cartridge (Waters) and lyophilized for 48 hours.
Fuco-proteomic mass spectrometric profiling of CD4 + T cells
CD4+T cells cultured and treated as indicated in the main text were harvested, lysed in standard RIPA buffer + protease and phosphatase inhibitors, and subjected to lectin pulldown using control or AAL beads as described above. The beads were washed with PBS and subjected to on-bead trypsin digestion. Proteins bound to beads were denatured with 30mM ammonium bicarbonate at 950C for 5 minutes. Samples were reduced with 4.5 mM dithiothreitol (DTT) for 30 min at 600C, alkylated with 10mM iodoacetamide (IAA) at room temperature in the dark for 20 minutes, and digested overnight at 370C with 1:20 enzyme-to-protein ratio of trypsin (Worthington). The resulting peptide solution was acidified with a final concentration of 1% TFA. Samples were centrifuged at high speed and the supernatants were subjected to Ziptip purification (Millipore Ziptips, #Z720070). The eluted peptides were concentrated in a SpeedVac and suspended in 15 µL loading buffer (5% ACN and 0.1% TFA) prior to auto sampling. Samples were then subjected to LC-MS/MS as described below
Mass spectrometric identification of WT vs. glycofucomutant HLA-DRB1 interactors
V5-tagged WT or N48G glycofucomutant HLA-DRB1-expressing WM793 cells were lysed and subjected to V5 bead pulldown. Five percent of pulled down protein was immunblotted to ensure for equal sample submission for LC-MS/MS (Extended Data FIg. 5a). Samples were then subjected to LC-MS/MS as described below.
Liquid chromatography-MS/MS
On-bead digestion was performed with trypsin and tryptic peptides were then analyzed using a nanoflow ultra-high-performance liquid chromatograph (RSLC, Dionex, Sunnyvale, CA) coupled to an electrospray orbitrap mass spectrometer (Q-Exactive Plus, Thermo, San Jose, CA) for tandem mass spectrometry peptide sequencing. The peptide mixtures were loaded onto a pre-column (2 cm x 100 µm ID packed with C18 reversed-phase resin, 5µm, 100Å) and washed for 5 minutes with aqueous 2% acetonitrile and 0.1% formic acid. The trapped peptides were eluted and separated on a 75 µm ID × 50 cm, 2 µm, 100Å, C18 analytical column (Dionex, Sunnyvale, CA) using a 90-minute program at a flow rate of 300 nL/min of 2–3% solvent B over 5 minutes, 3 to 30% solvent B over 27 minutes, then 30–38.5% solvent B over 5 minutes, 38.5–90% solvent B over 3 minutes, then held at 90% for 3 minutes, followed by 90–2% solvent B in 1 minute and re-equilibrated for 18 minutes. Solvent A was composed of 98% ddH2O and 2% acetonitrile containing 0.1% FA. Solvent B was 90% acetonitrile and 10% ddH2O containing 0.1% FA. MS resolution was set at 70,000 and MS/MS resolution was set at 17,500 with max IT of 50 ms. The top sixteen tandem mass spectra were collected using data-dependent acquisition (DDA) following each survey scan. MS and MS/MS scans were performed in an Orbitrap for accurate mass measurement using 60 second exclusion for previously sampled peptide peaks. MaxQuant 66 software (version 1.6.2.10) was used to identify and quantify the proteins for the DDA runs.
PyMOL structural modeling
In Fig. 4a, structural modeling was performed using PyMOL (Molecular Graphics System, Version 2.0 Schrödinger, LLC) of the HLA-DRB1:HLA-DM complex (PDB ID, 4FQX); HLA-DRB1 (yellow) and DM (gray). For the CD4:HLA-DRB1:TCR complex, the model was reconstituted by superimposing the DRB1 beta chains from CD4:HLA-DR1 complex (PDB ID, 3S5L) and TCR:HLA-DR1 complex (PDB ID, 6CQR) using PyMOL. RMSD between the 163 backbone atoms is 0.497. The potential glycosylation sites, N48 and T129, of HLA-DR1 beta chain are shown as sticks. CD4 (cyan), HLA-DRB1 (yellow), antigen peptide (magenta), and TCR (green)(lower right).
TIL isolation protocol
Tumors of SW1 or SM1 melanoma cells from C3H/HeJ or C57BL/6 mice, respectively) were digested using 1X tumor digest buffer (0.5 mg/mL Collagenase I, 0.5 mg/mL Collagenase IV, 0.25 mg/mL Hyalyronidase V, 0.1 mg/ mL DNAse I in HBSS (Millipore Sigma (St. Louis, MO)). Tumors were homogenized using the Miltenyi MACs dissociator. Red blood cells were lysed using ACK lysis buffer (Life Technologies, (Grand Island, NY)). Tumor homogenate cells were counted using a standard hemocytometer.
Human donor peripheral CD4 + T cell isolation protocol
Human CD4+ T cells were isolated from fresh peripheral blood monocyte cells (PBMC) using a CD4+ T cell negative selection isolation kit (Stem Cell Technologies, (Vancouver CA)) according to manufacturer’s protocols. CD4+ T cells were cultured in the presence of vehicle or 250µM L-fucose and were activated using anti-CD3/CD28 Dynabeads (ThermoFisher Scientific (Waltham, MA)) in a 1:1 bead:CD4+ T cell ratio. After 48 h, cell pellets were collected and lysed for either lectin-based fucoproteomics or phosphoproteomics.
Flow Cytometry
Gating schemes can be found in the Supplementary Information
• itIC and splenic profiling:
Total TILs were gated first to single cells (based on forward scatter height vs width, followed by side scatter height vs. width). Live cells were gated from the Zombie negative population from the population above. TILs were gated based on splenocyte size from a control spleen. Individual immune subpopulations were sub-gated from the total TIL population using the following staining criteria: CD3+ for CD3+ T cells; CD3+/CD4+/CD8− for CD4 + T cells; CD3+/CD4−/CD8+ for CD8+ T cells, CD11c+/CD11b+ for DCs; either NK1.1 (for C57/BL6 mice) or DX5 (for C3H/HeJ) for NK cells; CD11b+/GR1+ for MDSC-like cells; and F4/80+ for macrophages. Single-cell suspensions from tumor and spleen tissue were stained with Live/Dead Zombie NIR (Biolegend, (San Diego, CA)) at 1:1,000 in PBS for 20 min. Cell suspensions were spun down and stained with the following with antibodies at 0.5 µg/ml per antibody: APC anti-mouse CD3, Pacific Blue anti-mouse CD4, BV785 anti-mouse CD8, PerCP anti-mouse CD25, FITC anti-mouse F4/80, PeCy7 anti-mouse CD11c, PE anti-mouse NK1.1 or PE anti-mouse DX5, and PerCP-Cy5.5 anti-mouse CD11b. After staining, the cells were washed and fixed (2% formaldehyde), followed by another wash and flow cytometric analysis. The compensation controls were prepared using 0.5 µg/mL of each antibody with UltraComp eBeads, (ThermoFisher Scientific (Waltham, MA)). All samples were subject to flow cytometric profiling using a LSR Flow Cytometer (BD Biosciences (San Jose, CA)) and analysis as indicated using FlowJo software (BD Biosciences (San Jose, CA)).
• Assessment of cell surface fucosylation, HLA-DRB1, and PD-L1: Indicated cells were treated for 72 h with DMSO, 250 µM fucosyltransferase inhibitor (FUTi) (Millipore Sigma (St. Louis, MO)), or 250 µM of L-fucose (Biosynth (Oak Terrace, IL)). After 72 h, cells were stained with 0.1 µM PKH26 (Millipore Sigma (St. Louis, MO)) prior to fixation in 4% formaldehyde solution. The cells were stained with anti-HLA-DRB1 and fluorescein AAL, or anti-human or anti-mouse PD-L1 overnight. The following day the cells were washed 3 times prior to adding AlexaFluor 594 donkey anti-rabbit. Cells were washed 3 times and then subject to flow cytometric analyses using a FACSCalibur (BD Biosciences (San Jose, CA)). Samples were analyzed using FlowJo analysis software (BD Biosciences (San Jose, CA)). Median values of DRB1 and AAL were normalized to PKH26 values and statistical analysis was performed using GraphPad Prism.
• Assessment of cell surface pan-MHC-I and pan-MHC-II: Surgically resected patient tumors were minced to less than 1-mm fragments. Minced tumor sample was enzymatically digested in enzyme media comprised of RPMI with collagenase type IV (1 mg/mL), DNase type IV (30 U/mL), and hyaluronidase type V (100 µg/mL) (Sigma). Single cell suspensions were strained through 40-micron nylon mesh and counted for viability via trypan blue exclusion, followed by cryopreservation for future analysis. Tumor homogenates were thawed and stained using Live / Dead Zombie NIR, PE anti-pan-MHC-I (HLA-A,B,C), FITC anti-pan-MHC-II, PerPCy5.5 anti-CD45, APC anti-CD90, and BV421 anti EpCAM. Flow cytometric data was analyzed using FlowJo analysis software (BD Biosciences (San Jose, CA)). MHC-I and MHC-II expression was dichotomized as positive or negative based on FMO samples for each marker. Statistical analysis was performed using GraphPad Prism.
Immunoprecipitation and immunoblot analyses
Cells were lysed on ice in RIPA lysis buffer (25 mM Tris-HCl pH 7.6, 150 mM NaCl, 5 mM EDTA, 1% NP-40 or 1% Triton X-100, 1% sodium deoxycholate, 0.1% SDS in diH20 + protease and phosphatase inhibitors (ThermoFisher Scientific (Waltham, MA)), briefly sonicated, pelleted, and the resulting lysates were normalized by protein concentration using DC assay (BioRad Laboratories, (Hercules, CA)). The indicated samples were subjected to (12%) SDS-PAGE and immunoblot analysis using the indicated antibodies. Immunoblot imaging and analysis was performed using either an Odyssey FC scanner and ImageStudio (LiCor Biosciences, Lincoln, NE) or film.
qRT-PCR
RNA from cells subjected to the indicated treatments was extracted using Gene Elute Mammalian Total RNA Extraction System (Millipore Sigma (St. Louis, MO)). RNA was reversed transcribed to cDNA using High-Capacity cDNA Reverse Transcription Kit (ThermoFisher Scientific (Waltham, MA)). qRT-PCR analysis was performed using FastStart Universal SYBR Green Master Mix (Rox) (Roche Diagnostics, (Indianapolis, IN)) using a BioRad CFX96 Real-time system (BioRad Laboratories, (Hercules, CA)). The qRT-PCR cycles used were as follows: 95°C for 10 min, 35 cycles of 95°C for 15 seconds, 55°C for 60 seconds, and 72°C for 30 seconds. Expression of the indicated genes was normalized to histone H3A expression. Primers for qRT-PCR were generated using NCBI primer blast software (National Center for Biotechnology Information (Washington, D.C.)) as detailed the table below.
Primer
|
Sequence (5’ -> 3’)
|
Cloning Primers
|
|
Fuk (mouse)
|
F: cgcgcgcgGGATCCatggagcagtcagagggagtcaattggactg
R: cgcgcgcgGCTAGCggtggtgcccacttcagagggcc
|
HLA-DRB1 N48G
|
F: GTCTTTGAAGGATACACAGCCACCTTAGGATGGACTCG
R: tgagtgtcatttcttcggtgggacggagcggg
|
HLA-DRB1 T129A
|
F: CGAGTCCATCCTAAGGTGGCTGTGTATCCTTCAAAGAC
R: GTCTTTGAAGGATACACAGCCACCTTAGGATGGACTCG
|
EB1
|
F: cgcgcccgGGCGCGCCatggtggtgtggc
R: cgcgcccgCTCGAGgctcaggagtcc
|
EB1 N46G
|
F: GTGTCATTTCTACGGCGGGACGCAGCGC
R: GCGCTGCGTCCCGCCGTAGAAATGACAC
|
qRT-PCR
|
|
H3A (human and mouse)
|
F: AAGCAGACTGCCGCAAAT
R: GGCCTGTAACGATGAGGTTTC
|
Fuk (mouse)
|
F: ACTTCCGCCGAGATCTGTTC
R: GGATCAGTGGACGTAGGCAG
|
EB1
|
F: GAACACGCTTCTTCCTTGGG
R: CAGGCTCCTTACCTTTCTGGT
|
H2-K1
|
F: CCGCGGACGCTGGATA
R: GGCGATTCGCGACTTCTG
|
HLA-DRB1
|
F: CCATAGTAGCTCAGCACCCG
R: GTCCTGTCCTGTTCTCCAGC
|
Fluorescent immunocytochemical and immunohistological staining and analysis
• General fluorescent immunocytochemical staining protocol: Melanoma cells were grown on German glass coverslips (Electron Microscopy Services (Hatfield, PA)) and fixed in fixation buffer (4% formaldehyde, 2% sucrose in phosphate buffered saline (PBS) for 20 min at room temperature (RT). The coverslip-grown cells were subject to two 5-min standing washes in PBS prior to permeabilization in permeabilization buffer (0.4% Triton-X-100 and 0.4% IgG-free bovine serum albumin (BSA, Jackson ImmunoResearch Laboratories (Westgrove, PA) in PBS) for 20 min at RT. The coverslip-grown cells were next subject to 2 PBS washes and incubated with the indicated primary antibodies.
• General fluorescent immunohistochemical tissue staining protocol: In general, paraffin-embedded FFPE tumor tissue sections (or the TMA slide) were melted at 70°C for 30 min. The slides were further de-paraffinizeded using xylene and rehydrated in serial alcohol washes. The slides were pressure cooked at 15 PSI for 15 min in a 1X DAKO antigen retrieval buffer (Agilent Technologies (Santa Clara, CA)). The tumor sections were subject to two 5-min standing washes in PBS prior to blocking in 1X Carb-Free Blocking Solution (Vector Labs (Burlingame, CA)) for 2-3h. The slides were next washed twice and incubated with indicated lectin and/or antibodies.
General fluorescent analysis of mouse tumor tissue fucosylation (Extended Data Figs. 1a,d,k):
For assessment of mouse tumor fucosylation, FFPE tumor sections were immunostained with FITC-conjugated AAL lectin (0.4 µg/mL, Vector Laboratories (Burlingame, CA)) and rabbit anti-Mart1 + rabbit anti-S100 (melanoma marker cocktail). The slides were mounted with Vectashield + DAPI (Vector Laboratories (Burlingame, CA)). Four representative microscopy images per tumor were acquired using a Keyence BZ-X710, and images were process and analyzed using FIJI (NIH) as follows: melanoma marker-positive regions were assigned as regions of interest (ROI) in which we measured Integrated density of AAL signal. Integrated densities of control tumors were assigned as 1, and Integrated AAL density values of experimental tumors were divided by control to produce relative fold changes and plotted as column charts.
• Immunofluorescent staining and analysis of melanoma tissues and TMA (Fig. 1N):
Immunostaining and image acquisition: Melanoma TMA (Serial #ME1002b; US BioMax, Inc. (Derwood, MD)) was immunostained with FITC-conjugated AAL lectin (0.4 µg/mL, Vector Laboratories (Burlingame, CA)), rabbit anti-Mart1, rabbit anti-S100, and anti-CD3 followed by AlexaFluor 568 (Cy3) donkey anti-rabbit and AlexaFluor 647 (Cy5) donkey anti-mouse secondary antibodies. The slides were mounted with Vectashield + DAPI (Vector Laboratories (Burlingame, CA)). An Aperio Scanscope FL (Leica Biosystems) was used to scan the TMA slide at 20X magnification and the digital slide saved into the Spectrum e-slide database.
Analysis: The multiplex fluorescence TMA image file was imported into Definiens Tissue Studio version 4.7 (Definiens AG, Munich, Germany), where individual cores were identified using the software’s automated TMA segmentation tool. First, nucleus segmentation (DAPI channel) and cell growth algorithms were used to segment individual cells within each core. A minimum size threshold was used to refine the cell segmentation. Next, mean fluorescence intensity (MFI) values for the FITC (fucosylation), Cy3 (melanoma markers Mart1 + S100) and Cy5 (CD3 marker) channels were extracted from each segmented cell and minimum thresholds for MFI was set to enumerate positive Cy3 and Cy5 cells. Identical thresholds were used for each core. Finally average MFI values for each core were reported for the FITC and Cy3 channels.
Melanoma-specific fucosylation (FITC in CY3-positive cells) MFI and CD3+ cell numbers were subject to statistical analyses and correlation with clinical parameters as follows: We used the nonparametric Wilcoxon rank sum test to compare melanoma-specific fucosylation levels between CD3+ T cells high vs low groups. The density values of CD3+ T cells were all log2 transformed in the statistical analysis. Multivariable linear regression was used to assess the association between fucosylation and T cells while adjusting for confounding factors including sex, age and stage. The Spearman correlation coefficient was used to measure the correlation between melanoma-specific fucosylation and T cells in different sex groups.
Lectin-mediated proximity ligation assay (L-PLA)
Coverslip-grown cells subjected to L-PLA were processed upfront as described in the fluorescent immunocytochemistry protocol detailed above, whereas FFPE tumor tissue sections were processed according to the fluorescent immunohistochemistry protocol detailed above. Both approaches used mouse-anti-HLA-DRB1 (applied at 0.2 µg/mL, ab215835, Abcam, Cambridge, UK), biotinylated AAL lectin (applied at 0.2 µg/mL, Vector Laboratories (Burlingame, CA)), on coverslips overnight in 4oC. The coverslip-grown cells were again washed twice with PBS followed and then incubated with phalloidin Alexafluor 488 (applied at 0.05 µg/mL, ThermoFisher Scientific (Waltham, MA) with goat anti-biotin (applied at 0.1 µg/mL, Vector Laboratories (Burlingame, CA)) for 2h in 4oC. Subsequent steps of the protocol were adapted from the DUOlink In Situ Green PLA kit's manufacturer’s protocol (Millipore Sigma (St. Louis, MO)). PLA anti-goat MINUS and PLA anti-mouse PLUS probes were applied at 1:5 for 1 h at 37oC. The coverslips were washed twice with Wash Buffer A prior to ligation with 1:5 ligation buffer and 1:40 ligase in ddH2O for 30 min at 37oC. The coverslips were washed twice with wash buffer A prior to incubation in amplification mix (1:5 amplification buffer and 1:80 polymerase in ddH2O for 1.5 h at 37oC). Coverslips were washed twice with Wash Buffer B prior to mounting to slide with DAPI with VectaShield (Vector Labs, Burlingame, CA). Microscopy images were acquired using a Keyence BZ-X710, and images were process and analyzed using FIJI (NIH).
• Immunofluorescent staining, image acquisition, and analysis of anti-PD1-treated melanoma patients (FIG. 5c):
The indicated FFPE sections were immunostained with anti-DRB1 antibody or L-PLA stained as detailed above with the addition of anti-CD4+ antibody. WTS imaging was performed using the Vectra3 Automated Quantitative Pathology Imaging System (PerkinElmer, Waltham, MA). 20X ROI tiles were sequentially scanned across the slide and spectrally unmixed using InForm (PerkinElmer, Waltham, MA) and the multilayer Tiff files were exported. HALO (indica labs, Albuquerque, NM) was used to fuse the tile images together prior to WTS image analysis. For each whole tumor image, (i) every individual melanoma marker (MART1 + S100)-positive cell was segmented and quantitatively measured for total fucosylation, total HLA-DRB1, and fucosylated HLA-DRB1, and (ii) every CD4+T cell within the melanoma marker-positive tissue region was counted. Per patient (Pt.), these marker values were box plotted to visualize the staining distribution of individual tumor cells. The total numbers of melanoma cells per patient section measured and analyzed were as follows: Pt. 1: 557,146 cells; Pt. 2: 743,172 cells; Pt. 3: 95,628 cells; and Pt. 4: 13,423 cells.
Anti-PD1-treated patient specimens (FIGs. 5d & 5e, and EXTENDED DATA FIGs. 4D & 4E)
Moffitt Cancer Center patient specimens: Patients with advanced stage melanoma being treated at Moffitt Cancer Center were identified, and specimens collected and analyzed following patient consent under Moffitt Cancer Center Institutional Review Board approved protocols.
• For FIGs. 5d & 5e: De-identified Moffitt “Responder” patients exhibited greater than 20 months of progression-free survival, whereas “Non-Responder” patients progressed in less than 6 months after receiving anti-PD1.
• For Extended Data FIGs 4D & 4E: Non-response status to PD1 checkpoint blockade therapy (nivolumab or pembrolizumab) was defined as progression of disease by RECIST 1.1 while on PD-1 checkpoint blockade therapy or within 3 months of last dose.
MD Anderson Cancer Center patient specimens: Biospecimens were retrieved, collected and analyzed after patient consent under UT MD Anderson Cancer Center Institutional Review Board-approved protocols. Patients with advanced (stage III/IV) melanoma treated at The University of Texas MD Anderson Cancer Center between 07/01/2015 and 05/01/2020 who received at least one dose of PD-1 checkpoint blockade agent (either nivolumab or pembrolizumab) were identified from detailed retrospective and prospective review of clinic records. Responder status was defined as a complete or partial response and non-responder was defined as stable or progressive disease by RECIST 1.1. Pathologic response was defined by the presence or absence of viable tumor on pathologic review when available.
Massachusetts General Hospital patient specimens: Patients initiating anti-PD1 (Pembrolizumab) as front-line treatment for metastatic melanoma at MGH provided written informed consent for the collection of tissue and blood samples for research and genomic profiling (DF/HCC IRB approved Protocol 11–181). Patients classified as responders (R) showed clear radiographic decrease in disease at initial staging through a minimum of 12 weeks. Patients classified as non-responders (NR) did not respond to treatment radiographically and/or had clear and rapid progression. Progression free survival (PFS) is given in days from treatment start to radiographic scan when progression was first noted (uncensored) or last progression free scan (censured). Overall survival (OS) is given in days from treatment start to date of death (uncensored) or last follow-up (censored).
Animal models
All animals were housed at the Vincent A. Stabile Research building animal facility at H. Lee Moffitt Cancer Center & Research Institute, which is fully accredited by the Association for Assessment and Accreditation of Laboratory Animal Care International (AAALAC, #434), and are managed in accordance with the Guide for the Care and Use of Laboratory Animals (“The Guide”), the Animal Welfare Regulations Title 9 Code of Federal Regulations Subchapter A, “Animal Welfare”, Parts 1–3 (AWR), the Public Health Service Policy on Humane Care and Use of Laboratory Animals (PHS Policy), and by the USF Institutional Animal Care and Use Committee’s Principles and Procedures of Animal Care and Use (IACUC Principles). The experiments and protocols detailed in this study received institutional approval by the Moffitt IACUC (RIS00001625). Four-to-six-week-old female C3H/HeN and male C57BL6 mice were purchased from Charles Rivers Laboratories for the indicated experiments. Four-to-six-week-old male NSG mice from the Lau laboratory breeding colony were used for the indicated experiments. Power calculations were used to determine mouse cohort sizes to detect significant changes in tumor sizes. In general, 10 mice per indicated cohort to accommodate for incidental loss of mice due to issues beyond our control (e.g., incidental tumor ulceration that required exclusion from the study). Mouse tumor volumes were measured using length, width and depth divided by 2. At each experimental endpoint, mice were humanely euthanized using CO2 inhalation in accordance to the American Veterinary Medical Association guidelines. Mice were observed daily and humanely euthanized if the tumor reached 2,000 mm3 or mice showed signs of metastatic disease.
For all mouse models, 1 x 106 melanoma cells were injected subcutaneously in the right hind flanks of each mouse. Between 7–14 days, when the tumor volumes reached ~ 150 mm3, the mice were either supplemented with or without 100 mM L-fucose (Biosynth (Oak Terrace, IL)) via drinking water, which was provided ad libitum and which we previously demonstrated to increase tumor fucosylation and to suppress melanomas 8. This dosage is within previously reported ranges for dietary supplementation with L-fucose and other similar dietary sugars (e.g., D-mannose) in other rodent studies 67–71. When the tumors reached ~ 2 cm3, the animals were sacrificed, and the tumors either processed for flow cytometric profiling or for histological analysis as indicated.
• Control vs. mFuk ± L-fucose models (Fig. 1 & Extended Data Fig. 1): SW1 or SM1 mouse melanoma cells were injected into syngeneic C3H/HeN (or NSG) female or C57BL/6 male mice, respectively, as follows: parental SW1 cells for Fig. 1A; parental SM1 cells for Fig. 1E; SW1 cells stably expressing either empty vector (EV) or mouse fucose kinase (mFuk) for Fig. 1L; and parental SW1 cells for Fig. 1M.
• Control vs. L-fucose ± FTY720 models (Fig. 2): SW1 or SM1 mouse melanoma cells were injected into syngeneic C3H/HeN (or NSG) female or C57BL/6 male mice, respectively. Cells were injected as follows: parental SW1 cells for Fig. 1A; parental SM1 cells for Fig. 1E; SW1 cells stably expressing either empty vector (EV) or mouse fucose kinase (mFuk) for Fig. 1L; and parental SW1 cells for Fig. 1M. FTY720 was administered at 20 µg every 2 days starting on Day 12, just prior to the initiation of LF, until endpoint.
• Immunodepletion mouse models (Fig. 1 & Extended Data Fig. 1): Three days prior to tumor engraftment, PBS (control) or ~ 300 µg α-CD4 (20 mg/kg, for immunodepletion, GK1.5, Bioxcell (West Lebanon, NH)) or α-CD8 (20 mg/kg, for immunodepletion, 2.43, Bioxcell (West Lebanon, NH)) was administered by intraperitoneal injection into the indicated cohorts of mice. Injections of immunodepletion antibody or PBS were continued every 3–4 days until endpoint. Syngeneic recipient C3H/HeN female or C57BL/6 male mice were injected with SW1 or SM1 cells, respectively.
• HLA-A/HLA-DRB1 knockdown and glyco-fucomutant H2-EB1 reconstitution mouse model (Figs. 2 & 3): SW1 mouse melanoma cells expressing either shNT (non-targeting shRNA), shH2K1, shEB1, shNT + EV, shEB1 + EV, shEB1 + EB1 WT, or shEB1 + EB1 N46G were injected into syngeneic C3H/HeN female mice.
• anti-PD-1 mouse model (Figs. 4): SW1 or SM1 mouse melanoma cells were injected into syngeneic C3H/HeN female or C57BL/6 male mice, respectively. After approximately 7 days, when the mice tumors reached ~ 150 mm3, the mice were either supplemented with or without 100 mM L-fucose (Biosynth (Oak Terrace, IL)) via drinking water, which was provided ad libitum. Simultaneously, PBS (control) or anti-PD1 (20 mg/kg, clone RMP1-14, Bioxcell (West Lebanon, NH)) were administered via intraperitoneal injection every 3–4 days until endpoint. Mice were sacrificed, and tumors and indicated organs were harvested for analysis at indicated timepoints.
• NSG melanoma model (Fig. 1 model): SW1 murine mouse melanoma cells were subcutaneously injected into the right rear flanks of NSG mice.
Quantification and statistical analysis
GraphPad Prism was used for statistical calculations unless otherwise indicated. For all comparisons between 2 independent conditions, t tests were performed to obtain p values and standard error of the mean (SEM). For comparisons between ≥ 2 groups, one way or two-way ANOVAs were performed, and p values and SEMs were obtained. For the TMA data, Wilcoxon signed-rank test was used to determine significance.