Animals. The BR.Cia21.D3-31 congenic founder mice were obtained from a partial advanced intercross (PAI) described elsewhere and they were subsequently back crossed for four additional generations 9. In order to ensure strain purity, BR.Cia21.D3-31 mice were screened with a custom designed 8k Illumina chip at genome wide level 54 and the mice were found to be devoid of any contaminating RIIIS/J alleles. No SNPs were present between the congenic and the B10.RIII background strain. Mice were kept under specific pathogen free (SPF) conditions in the animal house of the Section for Medical Inflammation Research, Karolinska Institute in Stockholm. Animals were housed in individually ventilated cages containing wood shavings in a climate-controlled environment with a 14 h light-dark cycle, fed with standard chow and water ad libitum. All the experiments were performed with age-, sex- and cage-matched mice and all the genetic experiments were performed with littermate controls. All the experimental procedures were approved by the ethical committees in Stockholm, Sweden with ethical permit numbers; 12923/18 and N134/13 (genotyping and serotyping), N35/16 (CIA) and N83/13 (EAE).
Preparation of mouse single cell suspensions. Briefly, spleen or lymph nodes were harvested and mechanically dissociated on a 40 µM cell strainer (Falcon) using a 1ml syringe plunger (Codan). Cells were counted on a Sysmex KX-21 cell counter. All centrifugation steps throughout the study were carried out a 350 x g for 5 min at RT. For spleen samples, red blood cells were lysed in RBC buffer (155 mM NH4Cl, 12 mM NaHCO3, 0.1 mM EDTA) before counting.
Preparation of human peripheral blood mononuclear cells (PBMC). Human PBMCs were prepared from 8 ml whole blood of healthy donors using SepMate (Stemcell Technologies) tubes and Ficoll density gradient medium (Sigma) according to the manufacturer. Ethical permit number: Dnr 2020–05001.
Cell culture. 106 splenocytes, 5×105 lymph node cells, or 105 PBMCs were cultured in 200 µl of complete RPMI per well in Nunclon U-shaped bottom 96-well plates (Thermo Scientific). Cells were incubated at 37°C and 5% CO2. Complete RPMI: RPMI 1640 with GlutaMAX™ (Thermo Scientific); 10% heat inactivated FBS (Thermo Scientific); 10 µM HEPES (Sigma); 50 µg/ml streptomycin sulfate (Sigma); 60 µg/ml penicillin C (Sigma); 50 µM β-Mercaptoethanol (Thermo Scientific). FBS was heat-inactivated for 30 min at 56°C. To assess the effect of 17-β-estradiol (Sigma) on CD2 expression, the medium was supplemented with charcoal-stripped FBS instead (Thermo Scientific). 17-β-estradiol was solved in ethanol.
ELISA. 106 lymph node cells from CIA mice were plated per well and stimulated with 100 µg/ml bovine collagen type II (bCII) in complete RPMI for 48 h as described in cell culture. Supernatants were used for cytokine analysis. Flat 96-well plates (Maxisorp, Nunc) were coated overnight at 4°C with the capture antibody (Ab, listed below) in PBS. After removing the coating solution, supernatant from cell cultures were added. Plates were incubated for 3 h at RT before washing (0.05% Tween PBS) and adding the biotinylated detection Ab (listed below) in PBS (1 h at RT). Plates were washed and incubated 30 min at RT with Eu-labelled streptavidin (PerkinElmer, 1:1000) in 50 mM Tris-HCl, 0.9% (w/v) NaCl, 0.5% (w/v) BSA and 0.1% Tween 20, 20 µM EDTA. After washing, DELFIA Enhancement Solution (PerkinElmer) was added and fluorescence read at 620 nm (Synergy 2, BioTek). Monoclonal antibodies (mAbs) to IL-2 (capture Ab 5 µg/ml JES6-IA12; detection Ab 2 µg/ml biotinylated-JES6-5H4, in-house produced), IL-17A (capture Ab 5 µg/ml TC11-18H10.1; detection Ab 2,5 µg/ml TC11-8H4, Biolgend), IFN-γ (capture Ab 5 µg/ml AN18; detection Ab 2,5 µg/ml biotinylated R46A2, in-house produced).
Analysis of mRNA expression. 106 lymph node cells per well were stimulated for 24 h using mAb LEAF hamster anti-mouse CD3 (1 µg/ml, 500A2, BD Pharmingen) and LEAF hamster anti-mouse CD28 (1 µg/ml, 37.51, BD Pharmingen) as described in cell culture. Cells were washed in PBS and RNA was extracted using Qiagen RNeasy columns according to the manufacturer without DNAse digestion. RNA concentration was determined using a NanoDrop 2000 (Thermo Scientific). Sample concentrations were normalized before proceeding with reverse transcription. Samples were stored at -20°C for short-term storage. cDNA synthesis was carried out using the iSrcipt cDNA synthesis kit (Bio-Rad) according to the manufacturer. qRT-PCR primers covered an exon-exon junction to minimize amplification of genomic DNA and were used at a final concentration of 300 nM. The qPCR reaction was carried out using the iQSYBR Green Mix (Bio-Rad) in white 96-well plates (Bio-Rad) using a CFX96 real-time PCR detection system (Bio-Rad). ACTB or GAPDH were used as an internal control. Primer sequences are listed in supplementary table 2. Data were analysed according to the ∆∆Ct method 55, assuming equal efficiency for all the primer pairs.
ChIP-qPCR. 10x106 spleen cells/ml were fixed for 10 min in 1% formaldehyde PBS at RT. The reaction was stopped by adding 125 mM glycine and cells were washed twice in ice-cold PBS. Complete protease inhibitor cocktail (Roche) was added in all the following steps. 2x106 cells were lysed in 1 ml cell lysis buffer 56 on ice for 15 min, and the extracted nuclei lysed in 1 ml nuclear lysis buffer 56 on ice for 15 min. Lysates were sonicated for 15 cycles (on high settings, 30’’ON-30’’OFF) using a Diagenode Bioruptor. The water bath was cooled to 4°C before beginning sonication. Average DNA length after sonication was 500 bp. 450 µl of the lysates were incubated with 10 µg/ml rabbit anti-mouse ERα Ab (clone E115, Abcam) or polyclonal rabbit IgG isotype control (Abcam) on a shaker at 4°C over night. Next day, DNA-Ab complexes were precipitated using protein G magnetic beads (Thermos Scientific). Beads were washed twice for 5 min at RT in buffers of increasing salt concentration according to 56. DNA was eluted by incubating beads in 100 µl elution buffer 56 at 65°C for 30 min with occasional vortex. Beads were pelleted and fixation was reversed by incubation of supernatants for 8 h at 65°C in the presence of 0.3 M NaCl in 96-well plates. On the third day, 10 µg/ml RNAse A (Thermo Scientific) was added for 30 min (37°C) before incubation with 10 µg/ml of Proteinase K (Thermo Scientific) at 55°C for 30 min. DNA was purified using GeneJET PCR purification kit (Thermo Scientific) and used for qPCR. Primers used for amplification of recovered DNA are listed in supplementary table 3. Data was analysed according to 56, but briefly, results are presented as fold change over their respective mock IP controls.
Flow cytometry. 106 cells were blocked in 20 µl of PBS containing 5 µg in-house produced 2.4G2 in 96-well plates for 10 min at RT. Samples were washed with 150 µl of PBS and subsequently stained with the indicated antibodies in 20µl of PBS diluted 1:100 or 1:200 at 4°C for 20 min in the dark (Ab list follows). Cells were washed once, fixed and permeabilized for intracellular staining using BD Cytofix/Cytoperm™ (BD) according to the manufacturer. Cells were stained intracellularly with 20 µl of permeabilization buffer (BD), using the antibodies at a 1:100 final dilution, for 20 min at RT. FOXP3 staining required nuclear permeabilization and was carried out using Bioscience™ FOXP3/Transcription Factor Staining Buffer. For intracellular cytokine staining, cells were stimulated in vitro with phorbol 12-myristate 13-acetate (PMA) 10 ng/ml, ionomycin 1 µg/ml, and BFA 10 µg/ml for 4–6 h at 37°C prior to fixation, permeabilization and staining.
Flow cytometry anti-mouse antibodies (BD Pharmingen): CD3 (clone: 145-2C11); TCRB (H57-597); CD4 (RM4-5); CD8 (53 − 6.7); CD19 (1D3, 6D5); CD11B (M1/70); CD11C (HL3, N418); FOXP3 (FJK-16s); CD25 (7D4); CD44 (IM7); CD62L (MEL-14); CD2 (RM2-5); LY6C (AL-21); LAG-3 (C9B7W); CD40L (MR1); IFN-γ (R46A2); IL-17A (TC11-18H10.1). CD16/CD32 (2.4G2, in house).
Flow cytometry anti-human antibodies (BD Pharmingen): CD45 (clone: HI30); CD2 (RPA-2,10); TCRB (IP26); CD4 (OKT4); CD45RA (Hl100); CD45RO (UCHL1).
Proliferation assay. 107 lymph node cells were labelled using CellTrace™ Violet Cell Proliferation Kit (ThermoFisher Scientific) according to the manufacturer. 5x105 naïve lymph node cells were cultured per well in U 96-well plates as described under cell culture in the presence of hamster anti-mouse CD3 (1 µg/ml, 500A2, BD Pharmingen) and hamster anti-mouse CD28 (1 µg/ml, 37.51, BD) for 72–96 h. Proliferation by dilution of CTV was assessed using flow cytometry. Complementary antibody staining was done as described under flow cytometry. Proliferation parameters were analysed and calculated using FlowJo 8.8.7.
Collagen-induced arthritis (CIA). 12-week-old mice were immunized with 100 µg of bovine collagen type II (bCII) in 100 µl of a 1:1 emulsion with CFA (BD ) and PBS intradermally at the base of the tail. Mice were challenged at day 35 with 50 µg of bCII in 50 µl of IFA (BD) emulsion. Mice were monitored for arthritis development as described in 57. In short, each visibly inflamed (i.e. swollen and red) ankle or wrist was given 5 points, whereas each inflamed knuckle and toe joint was given 1 point each, resulting in a total of 60 possible points per mouse and day.
Collagen antibody-induced arthritis (CAIA). CII-specific antibodies (M2139, CIIC1, CIIC2 and UL1) were generated and purified as previously described 15. The sterile cocktail of M2139, CIIC1, CIIC2 and UL1 mAbs (4 mg per mouse) was injected intravenously. On day 7, lipopolysaccharide (O55:B5 LPS from Merck; 25 µg in 200 µl per mouse) was injected intraperitoneally to all mice to increase severity of the disease. Mice were scored as described for CIA.
Experimental induced autoimmune encephalomyelitis (EAE). 12-week-old mice were immunized with a 100 µl emulsion of 250 µg myelin basic protein peptide (MBP) 89–101 peptide in PBS and 50 µl IFA (incomplete Freud’s adjuvant) containing 50 µg Mycobacterium tuberculosis H37RA (BD). Animals were boosted with 200 ng of Bordetella pertussis toxin (Sigma Aldrich, St. Louis, MO, USA) i.p. on day 0 and 48 h post initial immunization. EAE severity was evaluated as described in 58. Briefly, mice were scored as follows: 0, no clinical signs of disease; 1, tail weakness; 2, tail paralysis; 3, tail paralysis and mild waddle; 4, tail paralysis and severe waddle; 5, tail paralysis and paralysis of one limb; 6, tail paralysis and paralysis of two limbs; 7, tetraparesis; 8, moribund or deceased.
Delayed type hypersensitivity (DTH). Hypersensitivity reaction was elicited by initially immunizing mice with 100 µg bCII emulsified in 50 µl CFA (Difco, Detroit, MI, USA). Ten days later mice were challenged with an injection of 10 µg bCII in 10 mM acetic acid into the dorsal part of the right ear and vehicle control in the left one. Ear swelling was assessed 48 and 72 h later using a calliper.
Ovariectomy. In brief, ovaries of female mice were removed after a single incision through the back skin and bilateral flank incision through the peritoneum. Thereafter, mice were rested for a minimum of 14 days prior to immunization for EAE or CIA as described elsewhere.
Luciferase reporter assay. 2x104 MCF-7 cells were seeded into flat 96-well flat bottom plates (Thermo Scientific) and left to adhere overnight. Then cells were transfected with pGL4.17 (Promega) luciferase reporter construct containing the BR or R3 allele of the candidate ERBS (pGL4.17.BR and pGL4.17.R3, respectively). ERBS cloning primers 5’-3’, Fw: AGATCTCGAGGGGGAAAGCTCTGACTTGGG; Rv: GTCAAGCTTGAGAAAGAATTTTGCTTATTTAGTCC. Cells were transfected in OPTIMEM medium (Thermo Scientific) using lipofectamine 3000 (Thermo Scientific) according to the manufacturer. The transfection mix (per well) contained 400 ng plasmid, 0.3 µl lipofectamine, and 0.2 µl P3000 reagent. Respective stimuli (20 ng/ml PMA, 10–100 nM E2) were added after 24 h, and cells were further incubated overnight before lysis. Luciferase activity was measured using Pierce Firefly Luc One-Step Glow Assay Kit (Thermo Scientific) in a Synergy-2 plate reader (BioTek).
Genetic association study. Data for genetic variations within CD2-CD58 locus was extracted from previous Immunochip data published elsewhere (PMID: 23143596). After filtering these data correspond to 263 SNPs in 1940 healthy controls (M/F 524/1416) and 2762 RA patients (M/F 817/1945) from the Swedish EIRA study. Association was analysed by PLINK separately for female and male individuals.
Analysis of public microarray expression data. Microarray data was extracted from NCBI GEO Database 28 and analysed using Shiny GEO 59. GEO accession number is cited wherever NCBI GEO data has been used.
Statistical analysis. Statistical analysis was performed using GraphPad Prism v6.0 or higher. Statistical comparison of two unpaired groups was carried out using Mann-Whitney U non-parametric test. CIA and EAE disease curves were compared using two-way ANOVA multiple comparisons test. P-values under 0.05 were considered statistically significant and are denoted with the symbol *. P-values under 0.01 are denoted **.
Proteomic analysis of enriched CD4 + T cells. CD4+ T cells were enriched from naïve spleens using untouched CD4+ T cell mouse kit (Dynabeads, Life Technologies). 96-well U bottom plates were pre-coated with 1 µg/ml of anti-CD3 and 1 µg/ml of anti-CD2 in PBS for 3 h at 37°C. 2.5x105 CD4+ T cells were plated on the pre-coated plates and cultured for 48 h.
Cell pellets were lysed in a buffer consisting of 1% SDS, 8 M urea and 20 mM EPPS pH 8.5 and sonicated using a Branson probe sonicator (3 s on, 3 s off pulses, 45 s, 30% amplitude). Protein concentration was measured using BCA assay and subsequently 50 µg of protein from each sample were reduced with 5 mM DTT at RT for 45 min followed by alkylation with 15 mM IAA in the dark at RT for 45 min. The reaction was quenched by adding 10 mM DTT and the samples were precipitated using methanol-chloroform mixture. Dried protein pellets were dissolved into 8 M urea, 20 mM EPPS pH 8.5. EPPS (20 mM, pH 8.5) was added to lower the urea concentration to 4 M and LysC digestion was done at a 1:100 ratio (LysC/protein, w/w) overnight at RT. Then urea concentration was lowered to 1 M and trypsin digestion was conducted at a 1:100 ratio (Trypsin/protein, w/w) at RT for 5 h. TMTpro plex (Thermo Fischer Scientific) reagents were dissolved into dry acetonitrile (ACN) to a concentration of 20 µg/µl and 200 µg were added to each sample. The ACN concentration in the samples was adjusted to 20% and the labelling was conducted at RT for 2 h and quenched with 0.5% hydroxylamine (ThermoFischer Scientific) for 15 min at RT. The samples were then combined and dried using Speedvac to eliminate the ACN. Then samples were acidified to pH < 3 using TFA and desalted using SepPack (Waters). Lastly, peptide samples were dissolved into 20 mM NH4OH and 150 µg of each sample was used for off-line fractionation.
Samples were fractionated off-line in a high-pH reversed-phase manner using an UltimateTM 3000 RSLCnano System (Dionex) equipped with a XBridge Peptide BEH 25 cm column of 2.1 mm internal diameter, packed with 3.5 µm C18 beads having 300 Å pores (Waters). The mobile phase consisted of buffer A (20 mM NH4OH) and buffer B (100% ACN). The gradient started from 1% B to 23.5% in 42 min, then to 54% B in 9 min, 63% B in 2 min and stayed at 63% B for 5 min and finally back to 1% B and stayed at 1% B for 7 min. This resulted in 96 fractions that were concatenated into 24 fractions. Samples were then dried using Speedvac and re-suspended into 2% ACN and 0.1% FA prior to LC-MS/MS analysis.
Peptides were separated on a 50 cm EASY-spray column, with a 75 µm internal diameter, packed with 2 µm PepMap C18 beads, having 100 Å pores (Thermo Fischer Scientific). An UltiMate™ 3000 RSLCnano System (Thermo Fischer Scientific) was used that was programmed to a 91 min optimized LC gradient. The two mobile phases consisted of buffer A (98% milliQ water, 2% ACN and 0.1% FA) and buffer B (98% ACN, 2% milliQ water and 0.1% FA). The gradient was started with 4% B for 5 min and increased to 26% B in 91 min, 95% B in 9 min, stayed at 95% B for 4 min and finally decreased to 4% B in 3 min and stayed at 4% B for 8 more min. The injection was set to 5 µL corresponding to approximately 1 µg of peptides.
Mass spectra were acquired on a Q Exactive HF mass spectrometer (Thermo Fischer Scientific). The Q Exactive HF acquisition was performed in a data dependent manner with automatic switching between MS and MS/MS modes using a top-17 method. MS spectra were acquired at a resolution of 120,000 with a target value of 3.106 or maximum integration time of 100 ms. The m/z range was from 375 to 1500. Peptide fragmentation was performed using higher-energy collision dissociation (HCD), and the normalized collision energy was set at 33. The MS/MS spectra were acquired at a resolution of 60,000 with the target value of 2.105 ions and a maximum integration time of 120 ms. The isolation window and first fixed mass were set at 1.6 m/z units and m/z 100, respectively.
TMT-10 labelling quantification
Protein identification and quantification were performed with MaxQuant software (version 1.6.2.3). MS2 was selected as the quantification mode and masses of TMTpro labels were added manually and selected as peptide modification. Acetylation of N-terminal, oxidation of methionine and deamidation of asparagine and glutamine were selected as variable modifications while carbamidomethylation of the cysteine was selected as fixed modification. The Andromeda search engine was using the UP000000589_Mus musculus database (22129 entries) with the precursor mass tolerance for the first searches and the main search set to 20 and 4.5 ppm, respectively. Trypsin was selected as the enzyme, with up to two missed cleavages allowed; the peptide minimal length was set to seven amino acid. Default parameters were used for the instrument settings. The FDR was set to 0.01 for peptides and proteins. “Match between runs” option was selected with a time window of 0.7 min and an alignment time window of 20 min.