Specifc-pathogen-free (SPF) experiments
All SPF animal experiment protocols were approved by the Animal Welfare Service at the Luxembourg Institute of Health, and further approved by the Veterinary Services Administration within the Ministry of Agriculture (national authorization no. LUPA2018/18, LUPA2019/29). Six-week-old, female BALB/c mice were purchased from Charles River Laboratories (Saint Germain Nuelles, France). Mice were housed in groups of five and assigned randomly to a diet and treatment group. The fiber-rich (FR) mice were fed the Standard CRM (P) Rat and Mouse Breeder and Grower Diet (Special Diets Services, catalogue no. 801722). The fiber-free (FF) diet was custom-manufactured by SAFE diets (Augy, France) according to the TD.140343 diet formulation (Envigo, Indianapolis, IN, USA), as previously described14. Each cage was provided their respective diets ad libitum with autoclaved water. For baseline readouts (before sensitization protocol), mice were fed for 40 days and then sacrificed for tissue/sample processing.
Germ-free and gnotobiotic experiments
All germ-free and gnotobiotic animal experiment protocols were approved by the Animal Experimentation Ethics Committee (AEEC) at the University of Luxembourg (national authorization no. LUPA2019/50). The germ-free status of the mice was verified by culture-based (both aerobic and anaerobic) methods. For all gnotobiotic experiments, 6–8-week-old BALB/c mice were housed in maintained in isocages and were fed the standard diet SAFE R04, irradiated at 40 kGy (SAFE, Augy, France). Mice were colonized with two consecutive daily intragastric gavages of the synthetic microbiota, as described in Steimle et al.47. One week after the gavage, fecal samples were collected, DNA was extracted and purified, and the colonization of individual strains in the synthetic microbiota was confirmed using phylotype-specifc qPCR primers47. Diet was switched for half of the cages to the FF diet 10 days after colonization.
Allergen sensitization and challenge
The ovalbumin sensitization protocol was adapted from Ganeshan et al.25 and was conducted as per Fig. 3a. All animals started with a feeding period on either the FR diet or the FF diet prior to sensitization. The feeding period lasted for 40 days for SPF mice and at least 7 days for gnotobiotic and germ-free mice. Mice were sensitized by intragastric gavage of 100 μL of the solution related to their treatment group using a reusable, 20G feeding needle (FST, Heildelberg, Germany), once per week for 8 weeks. All solutions were prepared in autoclaved Dulbecco's phosphate-buffered saline (PBS, Catalogue no. LOBE17-515F, Westburg, Leudsen, Netherlands). Treatment groups were as follows: OVA+CTX (100 µg ovalbumin (Catalogue no. A5503, Merck, Overijse, Belgium), 10 µg cholera toxin (Catalogue no. C8052, Merck, Overijse, Belgium), OVA (100 µg ovalbumin), CTX (10 µg cholera toxin) and PBS (PBS alone). One week after the 8th gavage, the mice were challenged with 5 mg of OVA in 200 ml of PBS. During the one-hour challenge period, two researchers took core body temperature readouts using a rectal probe (BiosebLab, Vitrolles, France), and recorded the clinical symptom score25; the researchers were blinded to the identities of the mouse treatment groups while recording the symptoms scores. Briefly, “0” was assigned if no symptoms were evident; “1” represents mild scratching, rubbing, or both of the nose, head, or feet; “2” and “3” represent intermediate symptoms (e.g. edema around the eyes or mouth, piloerection, and/or labored breathing); “4” represents significantly reduced motility, tremors, and/or significant respiratory distress; and “5” represents death. Mice were sacrificed 24 h after the allergen challenge (end of the experiment) and serum, tissues and fecal samples were collected for analysis.
The peanut sensitization protocol was adapted from Stefka et al.7 and was conducted as per Extended Data Fig. 3a. As with the OVA sensitization protocol, all SPF mice started with a 40-day feeding period on either a fiber-rich (FR) diet or a fiber-free (FF) diet. All solutions were made up in filter-sterilized Tris 20mM, pH 7.2. Peanut protein was extracted from defatted peanut flour (Bell Plantation, Tifton, GA, USA)7. Mice were sensitized by intragastric gavage of 400 μL of the solution related to their treatment group: PN+CTX (6 mg PN and 10 µg cholera toxin on days 40 and 42, then 6 mg PN and 15 µg cholera toxin), PN (6 mg PN), CTX (10 µg cholera toxin on days 40 and 42, then 15 µg cholera toxin). On day 75, the mice were challenged twice by gavage with 20 mg of PN, thirty minutes apart. During the one-hour challenge period, two researchers took core body temperature readouts using a rectal probe, and recorded clinical symptom score25; the researchers were blinded to the identities of the mouse treatment groups while recording the symptoms scores. Mice were sacrificed 3 h after challenge, and serum and tissues were retreived for analysis.
Sample processing
All animals were euthanized by cervical dislocation. Blood was extracted immediately by cardiac puncture, and collected into 1.5 ml eppendorf tubes. Serum was separated by incubating blood samples at 37°C for 30 min, followed by centrifugation at 3000 rpm for 30 min. Serum was stored in 1.5 ml eppendorf tubes at –20°C for short term storage, or –80°C for long term storage.
Colons were removed and placed in either Methacarn for mucus layer measurements, or collected in HBSS (w/o) (HBSS without Ca2+ and Mg2+ (Catalogue no. LOBE10-543F, Westburg, Leudsen, Netherlands) supplemented with 10 mM HEPES (Catalogue no. LOBE17-737E, Westburg, Leudsen, Netherlands) for lamina propria (LP) cell isolation. Ileal tissues were collected in HBSS (w/o) for LP cell isolation. Both ilea and colons were processed using the Lamina Propria Dissociation Kit (Catalogue no. 130-097-410, Miltenyi Biotec, Leiden, Netherlands) and gentleMACS™ Dissociator (Miltenyi Biotec, Leiden, Netherlands), according to the manufacturer’s instructions.
Cecal, fecal and ileal contents were collected in 2 ml screw cap tubes, and flash frozen in liquid nitrogen. All contents were stored at -80°C. Cecal, ileal and proximal colonic tissues were cleaned in PBS and stored in 1 ml RNAprotect Tissue Reagent (Catalogue no. 76106, QIAGEN, Venlo, Netherlands) for up to 1 week, followed by long term storage in -80°C.
Mucus layer measurements
After incubation in Methacarnoys fixative14, colons were stored in methanol until processing. For processing, samples underwent paraffin embedding and thin cross sections were set on glass slides. Slides were stained with Alcian blue or anti-MUC2, and quantification was determined in a blinded fashion, as previously described14.
For MUC2 staining, the slides were deparafinized in xylene (Catalogue no. 28973.328, VWR International, Leuven, Belgium) 3 times for 5 min at room temperature. The same 3 consecutive 5 min washes were repeated with pure ethanol (Catalogue no. 10437341, Fisher Scientific, Erembodegem-Aalst, Belgium) to dehydrate the sections and then with fresh Milli-Q water to wash the slides. To retrieve the antigens from the tissue sections, Retrievagen A, pH 6.0 (Catalogue no. 550524, BD Biosciences, Erembodegem-Aalst, Belgium) was heated and immediately poured onto the slides and covered to retain the heat as much as possible. After a 10 min incubation, the slides were left to cool in the solution for 20 min. Then the slides were washed in fresh Milli-Q water 3 times for 5 min, incubated in blocking buffer (Tris-buffered saline (TBS), 10% goat serum (Gibco, Catalogue no. 11540526, Fisher Scientific, Erembodegem-Aalst, Belgium) for 1 h at room temperature, and gently blotted dry. For the staining with the primary antibody, the tissue sections on the slides were covered with the Rabbit monoclonal anti-MUC2 antibody (Catalogue no. ab272692, Abcam, Amsterdam, Netherlands) 1/200 in the blocking buffer, in the dark for 2 hours at RT. The excess liquid was blotted away and the slides were washed by dipping them into fresh TBS for 5 min, 3 consecutive times. The tissue sections were covered with the secondary antibody Alexa Fluor-488-conjugated Goat anti-Rabbit IgG (H+L) (10 mg/mL; Catalogue no. 10729174, Fisher Scientific, Erembodegem-Aalst, Belgium) in blocking buffer, in the dark for 1 h at room temperature. The excess liquid was blotted away and the slides were washed by dipping them into TBS, 3 consecutive times for 5 min. The samples were finally mounted by applying 2–3 drops of ProLong Gold Antifade Mountant with DAPI (Invitrogen, Catalogue no. 11549306, Fisher Scientific, Erembodegem-Aalst, Belgium) and a cover slip to the colon section. After incubating for 24 h at room temperature, the slides were sealed using nail varnish and stored in the dark at 4 °C until they were visualized on the Axio Observer Z1 (Zeiss Carl, Zaventem, Belgium). Images were analysed using the MATLAB software BacSpace15 to determine mucus layer thickness.
Mucus penetrability assay
Penetrability of the colonic mucus was assessed as described by Gustafsson et al21 . Briefly, colons were flushed with ice-cold oxygenated KREBS Buffer “Transport” (116 mM NaCl, 1.3 mM CaCl2, 3.6 mM KCl, 1.4 mM KH2PO4, 23 mM NaHCO3, and 1.2 mM MgSO4 – Carl Roth) and open along the mesenteric axis. The longitudinal muscle layer was removed by blunt dissection and the distal mucosa was inserted in a perfusion chamber. The basolateral chamber was filled with 0.6µg/ml SYTO9 (Fisher Scientific, 10237582) in oxygenated KREBS “Base” Glucose Buffer (KREBS “Base” Buffer : 136 mM NaCl, 1.5 mM CaCl2, 4.3 mM KCl, 1.6 mM KH2PO4, 27.1 mM NaHCO3, and 1.4 mM MgSO4; KREBS Glucose Buffer containing 10mM Glucose, 5.7 mM sodium pyruvate and 5.1 mM sodium-l-glutamate in KREBS “Base” Oxygenated Buffer – Carl Roth), and the apical chamber was filled with oxygenated KREBS Mannitol Buffer (containing 10mM Mannitol, 5.7 mM sodium pyruvate and 5.1 mM sodium-l-glutamate in KREBS “Base” Oxygenated Buffer). After 10 min incubation in the dark at room temperature, the excess of KREBS Mannitol Buffer is removed and FluoSphere™ carboxylate beads (1µm, red 580/605, Invitrogen, F882) were applied on top and let to sediment on the tissue for 5 min in the dark at room temperature. The apical chamber was then gently washed several times with KREBS Mannitol Buffer to remove excess of beads. The chamber was incubated for 10 min in the dark before being visualized with a microscope Axio Examiner KMAT (Carl Zeiss Microscopy GmbH) using the software Zen3.0 (Blue Edition, Carl Zeiss Microscopy GmbH). For each tissue, 4-7 confocal images were taken in XY stacks from the epithelium at the bottom to the beads on top, with 5 µm-intervals between sections. Images were then analyzed with Imaris software (Oxford Instruments Imaris). The penetrability was computed by comparing the distance between the outer border of the beads and the epithelium with the distance between the most inner beads and the epithelium (Extended data Fig. 1b). The same method was used to compute the mucus thickness as the distance between the median position of the beads and the median position of the epithelium (Extended data Fig. 1b).
Intestinal permeability assay
Mice were fasted for 4 h prior to the assay. They were orally gavaged with FITC-dextran (Catalogue no. FD4-250MG, Merck, Overijse, Belgium) solution (100 mg/mL in PBS) so that they received 44 mg FITC-dextran per 100g of body weight. The mice were anaesthetized 4 hours later by intraperitoneal injection of ketamine (12.5mg/100g body weight) and medetomidine (0.025mg/100g body weight) and blood was collected via cardiac puncture in a 1.5 mL tube protected from light. Serum was separated by incubating blood samples at 37°C for 30 min, followed by centrifugation at 3000 rpm for 30 min. 100 ml of serum diluted with an equal volume of PBS was added to a 96-well black/clear flat bottom polystyrene microplate (Catalogue no. 655906, Greiner, Vilvoorde, Belgium). The concentration of FITC in serum was determined by spectrophotofluorometry with an excitation at 485 nm (20 nm band width) and an emission wavelength of 528 nm (20 nm band width), using serially diluted FITC-dextran (0, 125, 250, 500, 1000, 2000, 4000, 6000, 8000 ng/ml) as a standard. Serum from mice not administered with FITC-dextran was used to determine the background.
Bacterial genomic DNA extraction and qPCR analysis
Bacterial DNA was extracted from fecal pellets using phenol-chloroform extraction. Fecal pellets were disrupted in a mix of 500 µl Buffer A (200 mM NaCl, 200 mM Tris, 20 mM EDTA), 210 µl 20% SDS, 500 µl equal parts phenol:chloroform and approximately 250 µl acid-washed glass beads (Catalogue no. G1277, Merck, Overijse, Belgium) per sample. Mechanical lysis was performed on a Mixer Mill MM 400 (Retsch, Fisher Scientific, Erembodegem-Aalst, Belgium) for 5 min at 30 Hz, then samples were centrifuged at 18000 × g for 3 min at 4°C. The aqueous phase was recovered and 500 µl phenol:chloroform was added, mixed by inversion, and then centrifuged at 18000 × g for 3 min at 4°C. The aqueous phase was recovered and 500 µl pure chloroform was added. Samples were mixed by inversion, and then centrifuged at 18000 × g for 3 min at 4°C. The aqueous phase was recovered into a new tube and 1 volume of isopropanol and 1/10 volume 3 M sodium acetate (pH 5.2) were added. Samples were stored at –20°C for 1 h, followed by centrifugation at max speed for 20 min at 4°C. The supernatants were discarded and the DNA pellets were further cleaned with 1 mL 70% ethanol. Pellets were air-dried for approximately 1 h, and then resuspended in 100 µl of ultrapure water (Invitrogen, Catalogue no. 12060346, Fisher Scientific, Erembodegem-Aalst, Belgium). The DNA was purified using QIAGEN DNeasy Blood & Tissue Kit (Catalogue no. 69506, QIAGEN, Venlo, Netherlands) according to the manufacturer’s instructions. Final quantification was determined with a NanoPhotometer® N60 (Implen, Fisher Scientific, Erembodegem-Aalst, Belgium).
20 ng DNA was amplified within 12.5 µl of master mix: buffer (Catalogue no. 10966034, Invitrogen,), 1.5 mM MgCl2 (Catalogue no. 10966034, Invitrogen), 200 µM dNTP (Catalogue no. 10297117, Invitrogen), GelStar Nucleic Acid Gel Stain (Lonza, Catalogue no. LONZ50535, VWR International, Leuven, Belgium), 0.5 U Platinum™ Taq DNA Polymerase (Catalogue no. 10966034, Invitrogen) and 0.2 µM each of forward and reverse primers14. The qPCR cycle consisted of pre-denaturation at 95°C for 3 min, followed by 40 cycles of 3 sec denaturation at 95°C, 20 sec annealing at 55°C, and 20 sec extension at 68°C. Samples were held at 95°C for 15 sec post-extension and then a melting curve was generated by heating from 60°C to 95°C with 0.3°C interval increases over 15 sec.
16S rRNA gene sequencing and analysis
DNA concentration was measured using a Qubit® dsDNA HS assay kit (Catalogue no. Q32854, Invitrogen) on a Qubit4 fluorometer (Catalogue no. Q33238, Invitrogen). The V4 region of the 16S rRNA gene was amplified using the dual-index primers described by Kozich et al.48 and sequenced on an Illumina MiSeq system. Raw sequences were processed using QIIME 2 (version 2020.6)49 with DADA2 for sequence quality control. Sequences were aligned and classified using SILVA 138 reference database50. PCoA plots were generated in RStudio version 1.3.1093 (R version 4.0.2), using the package ‘vegan’ version 2.5-751, with clustering significance testing using the betadisper() function in vegan and pairwise.adonis() package ‘pairwiseAdonis’ version 0.452 to test for heterogeneity of dispersion and difference in centroids, respectively. Relative abundance plots were generated using the package ‘phyloseq’ version 1.34.053 and ‘ggplot2’ version 3.3.554. The package ‘DESeq2’ version 1.30.155 was used to perform differential abundance analyses.
RNA extraction
Tissues were defrosted on ice in 1 ml of TRIzol Reagent (Catalogue no. 15596026, Life Technologies, Merelbeke, Belgium). A 0.5 mm metal bead (Catalogue no. 69989, QIAGEN, Venlo, Netherlands) was added prior to mechanical lysis on a Mixer Mill MM 400 (Retsch, Fisher Scientific, Erembodegem-Aalst, Belgium) for 8–10 min at 30 Hz. The extraction was performed according to the TRIzol Reagent protocol with a few adaptations. After bead-beating, samples were centrifuged at 12000 × g for 5 min at 4°C. The supernatant was transferred into a fresh 1.5 ml tube and 200 µl of pure chloroform was added. Samples were mixed by shaking for 15 sec and left to incubate for 3 min at room temperature, then centrifuged at 12000 × g for 15 min at 4°C. The aqueous phase was added to 500 µl isopropanol in a new tube, mixed by shaking for 10 sec and left to incubate for 10 min at room temperature. The samples were then centrifuged for 10 min at 12000 × g at 4°C for the precipitation of total RNA. Supernatant was removed and samples were washed with 1 ml cold 75% EtOH. The pellets were left to dry at 37°C for 5–10 min. Pellets were resuspended in 50 µl nuclease-free water and incubated at 56°C for 15 min. A DNase treatment was performed following the Thermo Scientific DNase I, RNase-free (Catalogue no. EN0521, Thermo Scientific™,) protocol, followed by a purification with Qiagen RNeasy Mini Kit (Catalogue no. 74106, QIAGEN, Venlo, Netherlands) according to the manufacturer’s instructions. Final RNA concentrations were quantifed using a NanoPhotometer® N60 (Implen). Reverse transcriptase was performed to obtain cDNA according to Invitrogen Superscript™ IV Reverse Transcriptase protocol (Catalogue no. 18090010, Invitrogen) in combination with RNaseOUT (Catalogue no. 10777019, Invitrogen).
Cytokine gene expression
qPCR reactions were performed using SYBR Green detection method and Platinum™ Taq DNA Polymerase Kit (Invitrogen) on a C1000 Touch Thermal Cycler (Biorad). Prior to qPCR, cDNA samples were diluted 1/5 in 80µl of nuclease-free water. 1 µl of cDNA was amplified with 11,5 µl of master mix containing buffer (Catalogue no. 10966034, Invitrogen), 2.5 mM MgCl2 (Catalogue no. 10966034, Invitrogen,), 400 µM dNTP (Catalogue no. 1029711, Invitrogen), GelStar Nucleic Acid Gel Stain (Lonza, Catalogue no. LONZ50535, VWR International, Leuven, Belgium), 0.5 U Platinum™ Taq DNA Polymerase (Catalogue no. 10966034, Invitrogen) and 0.2 µM each of forward and reverse primers. The qPCR cycle consisted of pre-denaturation at 94°C for 5 min, followed by 40 cycles of 20 sec denaturation at 94°C, 50 sec annealing at 60°C, and 45 sec extension at 72°C. Samples were held at 72°C for 5 min post-extension and then a melting curve was generated by heating from 65°C to 95°C with 0.3°C interval increases over 15 sec. The mRNA cytokine expression cytokines was normalized to expression of GAPDH. The following primers (Kaneka Eurogentec,Ougrée, Belgium) were used: Ifng forward (5’-ATGAACGCTACACACTGCATC-3’), Ifng reverse (5’-CCATCCTTTTGCCAGTTCCTC-3’), Il22 forward (5’-GCAGCCGTACATCGTCAACC-3’), Il22 reverse (5’-TCCCCGATGAGCCGGACA-3’), Il25 forward (5’-ACAGGGACTTGAATCGGGTC-3’), Il25 reverse (5’-TGGTAAAGTGGGACGGAGTTG-3’), Gapdh forward (5’-AATTCAACGGCACAGTCAAGGC-3’), Gapdh reverse (5’-GTGGTTCACACCCATCACAAA-3’).
Mass cytometry time-of-flight (CyTOF)
Single cell suspension of colonic lamina propria cells were stained with a 28-marker panel, as previously described18 . Briefly, 3 x 106 cells were stained with 5 µM cisplatin solution (Catalogue no. 201064, Fluidigm, Amsterdam Zuidoost, Netherlands) for 5 min. Cells were washed in FACS buffer (PBS; 2% FBS; 2 mM EDTA) and extracellular cell surface staining mix was added for 30 min at room temperature. Cells were washed twice, fixed using the Foxp3 Transcription Factor Staining Buffer Set (eBioscience, catalogue no. 00-5523-00, Life Technologies, Merelbeke, Belgium) for 45 min at 4°C, and washed in the permeabilization buffer. The intracellular cell staining mix was added to the cells for 30 min at room temperature. Samples were washed twice in FACS buffer, resuspended in Cell-ID™ iIntercalator-Ir (Catalogue no. 201192 A, Fluidigm, Amsterdam Zuidoost, Netherlands) in MaxPar fixation solution (Catalogue no. 201192 A, Fluidigm, Amsterdam Zuidoost, Netherlands) and refrigerated overnight, or for up to five days. Prior to acquisition on the Helios™ mass cytometer (Fluidigm, Amsterdam Zuidoost, Netherlands), samples were washed twice in PBS and then twice in deionized water. Samples were resuspended in deionized water at 0.5 x 106 cells per mL, with 10% calibration beads (EQ Four Element Calibration Beads, catalogue no. 201078, Fluidigm, Amsterdam Zuidoost, Netherlands). Normalized FCS files were imported in FlowJo™ (BD Life Sciences). The files were cleaned to exclude calibration beads and doublets, and CD45+ cells were exported as new files, which were later imported into RStudio (version 1.0.143, R version 3.4.4) using the R package flowcore (version 1.44.2) for unsupervised analysis, following a pipeline previously described56,57.
Cell stimulation and flow cytometry
Prior to staining for cytokines, single-cell suspensions from ileal and colonic lamina propria were incubated for 4 h at 37°C, 5% CO2, with Lymphocyte Activation Cocktail (Catalogue no. 423304, Biolegend Europe BV, Amsterdam, Netherlands) in DMEM (Catalogue no. LOBE12-604F, Westburg, Leudsen, Netherlands) complemented with 10% FBS, 100 U/ml penicillin/streptomycin (Catalogue no. LOBE17-602E, Westburg, Leudsen, Netherlands), glutamine (Catalogue no. LOBE17-605E, Westburg, Leudsen, Netherlands), 10 mM HEPES (Catalogue no. LOBE17-737E, Westburg, Leudsen, Netherlands) and 0.1% β-mercaptoethanol (Catalogue no. M6250, Merck, Overijse, Belgium). For eosinophils and cytokine expression analysis, cells were washed with PBS and incubated with Zombie NIR (Catalogue no. 423105, Biolegend Europe BV, Amsterdam, Netherlands), for 15 min at 4°C, prior to fixation with the Cytofix/Cytoperm solution kit (BD Biosciences, Erembodegem-Aalst, Belgium). Cells were then washed with FACS buffer, incubated with Fc block (1 mg/106 cells, catalogue no. 553142, BD Biosciences, Erembodegem-Aalst, Belgium) for 15 min and stained with the antibody mix (Extended data Table 1) for 30 min at 4°C. Samples were finally washed and resuspended in PBS for acquisition on a NovoCyte Quanteon Flow Cytometer (ACEA Biosciences). FCS files were analyzed in FlowJo™ (BD Biosciences). For each reported population, counts were normalized to the LD-CD45+/Single Cells/Width, SSC-H subset and results are presented as a percentage of CD45+ cells.
Lipocalin-2 ELISA
Fecal samples were resuspended in 500 μl ice-cold PBS with 1% Tween 20, followed by agitation at 2000 rpm for 20 min at 4°C on a thermomixer (Eppendorf, Aarschot, Belgium). Samples were then centrifuged for 10 min at 21000 × g at 4°C. Supernatants were stored at –20°C until further analysis. Lipocalin-2 detection was performed using the Mouse Lipocalin-2/NGAL DuoSet Elisa, R&D Systems (Catalogue no. DY1857, Bio-Techne, Abingdon, UK), according to the manufacturer’s instructions.
MCPT1 ELISA
Mouse mast cell protease 1 (MCPT1) was detected in serum samples using the MCPT-1 Mouse Uncoated ELISA Kit (Catalogue no. 88-7503-88, Life Technologies, Merelbeke, Belgium), according to the manufacturer’s instructions, but adapted for 384-well microplates.
Allergen-specific ELISA
Allergen-specific IgE and IgG1 antibodies were quantified from serum samples by sandwich ELISA. For OVA-specific assays, 20 ml of ovalbumin (Catalogue no. A5503, Merck, Overijse, Belgium) at 100 ng/ml in PBS were added to each well of a 384-well microplate (Catalogue no. 781061, Greiner Bio-One, Vilvoorde, Belgium), and incubated overnight at 4°C. Wells were washed four times with 100 μL of wash buffer (1% Tween-20, 154 mM sodium chloride and 10 mM Trisma-base), and blocked with 75 μl of blocking buffer (15 mM Trizma-acetate, 136 mM sodium chloride, 2 mM potassium chloride and 1% (w/v) BSA (bovine serum albumin) for 2 h at room temperature. Undiluted mouse sera were used to determine the OVA-specific IgE antibody concentrations, using the Mouse Anti-Ovalbumin IgE Monoclonal Antibody (Clone E-C1, catalogue no. 7091, Ams Biotechnology, Abingdon, UK) as serially diluted standard (range 0–1000 pg/ml) in dilution buffer (DB; 15 mM Trizma-acetate, 136 mM sodium chloride, 2 mM potassium chloride, 0.1% (w/v) Tween-20, and 1% BSA). To determine the OVA-specific IgG1 antibody concentrations, mouse sera were serially diluted from 1/400 to 1/12800 in DB. The mouse anti-ovalbumin IgG1 monoclonal antibody (Clone L71, catalogue no. 7093, Ams Biotechnology, Abingdon, UK) was used as serially diluted standard (range 0–1000 pg/ml) in DB. After a washing step performed as described above, 20 ml of diluted standards and samples were added to corresponding wells. The plate was then incubated for 90 min at room temperature. The washing step was repeated and 20 μL of detection antibody, either the phosphatase alkaline-conjugated goat anti-mouse IgE (SouthernBiotech, catalogue no. 1110-04, ImTec Diagnostics, Antwerp, Belgium), either the phosphatase alkaline-conjugated goat anti-mouse IgG1 (SouthernBiotech, catalogue no. 1071-04, ImTec Diagnostics, Antwerp, Belgium) diluted 1/500 in DB, were added to each corresponding well. The plate were then incubated for 90 min at room temperature. Following a last wash, 40 μl of substrate (1 x phosphate tablet (Catalogue no. S0942, Merck, Overijse, Belgium) dissolved in 10 mL of substrate buffer (1 mM 2-Amino-2-methyle-1-propanole, 0,1 mM MgCl2×6H2O) were added to each well. After a last incubation step for 60 min at 37°C, the absorbance was measured at 405 nm using an ELISA plate reader (SpectraMax Plus 384 Microplate Reader and SoftMax Pro 7 Software, Molecular Devices, Berks, UK). The OVA-specific IgE or IgG1 antibodies concentrations were determined for each sample using the corresponding formulated standard curve.
For PN specific assays, same protocol was used with adapted antigen. Peanut protein, extracted from defatted peanut flour10 (Bell Plantation, Tifton, GA, USA), diluted at 12.5 ng/ml in carbonate-bicarbonate buffer (Catalogue no. C3041, Merck, Overijse, Belgium) was used for the coating overnight. Mouse sera were diluted 1/10 in DB to determine PN-specific IgE antibody levels, and 1/100 to determine PN-specific IgG1 antibody levels. Both detection antibodies (goat anti-mouse IgE-phosphatase alkaline conjugated and goat anti-mouse IgG1-phosphatase alkaline conjugated (SouthernBiotech, ImTec Diagnostics, Antwerp, Belgium) were diluted 1/1000 in DB. The absorbance was measured at 405 nm using an ELISA plate reader (SpectraMax Plus 384 Microplate Reader and SoftMax Pro 7 Software, Molecular Devices, Berks, UK).
Total IgE and IgG1 ELISA
Total IgE and IgG1 were quantified from serum samples by sandwich ELISA. The same protocol as for allergen ELISA was used, with adapted reagents. Rat anti-mouse IgE at 60 ng/well or rat anti-mouse IgG1 at 20 ng/well (SouthernBiotech, catalogue no. 1130-01 and 1144-01 respectively, ImTec Diagnostics, Antwerp, Belgium) was diluted in carbonate buffer and used as capture antibody. Mouse sera were serially diluted from 1/10 to 1/320 to determine the total IgE antibodies concentration, and the mouse IgE isotype control (SouthernBiotech, catalogue no. 0114-01, ImTec Diagnostics, Antwerp, Belgium) was used as the serially diluted standard in DB (range 0–250 pg/ml). To determine the total IgG1 antibody concentrations, mouse sera were serially diluted from 1/500 to 1/16000 in DB, and the Mouse IgG1 isotype control (SouthernBiotech, catalogue no. 0102-01, ImTec Diagnostics, Antwerp, Belgium) was used as serially diluted standard in DB (range 0–2000 pg/ml). Each detection antibody, either the phosphatase alkaline-conjugated goat anti-mouse IgE (SouthernBiotech, catalogue no. 1110-04, ImTec Diagnostics, Antwerp, Belgium) or the phosphatase alkaline-conjugated goat anti-mouse IgG1 (SoutherBiotech, catalogue no. 1071-04, ImTec Diagnostics, Antwerp, Belgium), was diluted 1/500 in DB for the assays. The total IgE or IgG1 concentration was calculated the same way as for the OVA-specific IgE/IgG1 concentration.
Flow cytometry for Ig-coated bacteria
Quantification of Ig-coated bacteria was determined from frozen fecal samples or tissue contents by flow cytometry. Samples were homogenized in 500 µl ice-cold PBS by mixing at max speed on a ThermoMixer for 20 min at 4°C. Samples were topped up with 500 µl PBS followed by centrifugation at 100 × g for 5 min at 4°C. The supernatant was passed through a 70 µm strainer (Pluriselect, catalogue no. 43-10070-70, ImTec diagnostics, Antwerp, Belgium) followed by centrifugation at 10000 × g for 5 min at 4°C. The pellet was resuspended with 1 mL PBS for measurement of the optical density at 600 nm and quantification of the bacteria. Approximately 109 bacteria were used per staining. Samples were incubated with 500 µl PBS with 5% goat serum (Gibco, catalogue no. 11540526, Fisher Scientific, Erembodegem-Aalst, Belgium) at 4°C for 20 min. After a centrifugation at 10000 × g for 5 min at 4°C, pellets were resuspended in PBS with 5% goat serum and the appropriate antibody: FITC-conjugated anti-mouse IgA (Clone mA-6E1, eBioscience, catalogue no. 11-4204-83, Life Technologies, Merelbeke, Belgium), PE-conjugated anti-mouse IgE (Clone RME-1, catalogue no. 406908, Biolegend Europe BV, Amsterdam, Netherlands), or PE-conjugated IgG1 isotype (Clone RTK2071, catalogue no. 400408, Biolegend Europe BV, Amsterdam, Netherlands). After incubating for 30 min at 4°C, samples were washed in 1 mL PBS. Bacterial pellets were resuspended in 200 µl DNA staining solution (0.9 % NaCl in 0.1 M HEPES, pH 7.2) with 1:4000 SYTO™ 60 Red Fluorescent Nucleic Acid Stain (5 mM Solution in DMSO, cat no. S11342, Invitrogen) and incubated for 20 min at 4°C. Samples were washed in 1 mL PBS and resuspended in 200 µl PBS for aquisition on a NovoCyte Quanteon Flow Cytometer (ACEA Biosciences). FCS files were imported into FlowJo (BD Biosciences) for analysis. IgE-coated bacteria were quantified using a negative gate constructed with the corresponding isotype control sample (Extended Data Fig. 2d).
p-nitrophenyl-based enzyme assays
The enzyme assays were performed as per the previously described protocol58.
Statistical analysis
Unless otherwise specifed, all statistical analyses, were performed in GraphPad Prism version 8.0 using either an unpaired t-test, a Mann–Whitney test, or a two-way ANOVA, with P values adjusted using the Benjamini-Hochberg method. For each experiment, n indicates the number of mice per group.
Data availability
The raw fastq files from 16S rRNA gene sequencing have been deposited in the European Nucleotide Archive (ENA) at EMBL-EBI under accession numbers PRJEB53451 (https://www.ebi.ac.uk/ena/browser/view/PRJEB53451) and PRJEB51707 (https://www.ebi.ac.uk/ena/browser/view/PRJEB51707). The mass cytometry datasets for colonic lamina propria have been uploaded to the FlowRepository database under accession number FR-FCM-Z5G2 (https://flowrepository.org/).
Code availability
The analysis pipelines used in this study can be found at https://github.com/DII-LIH-Luxembourg.