30
immunology.sciencemag.org/cgi/content/full/5/46/eaax0062/DC1 Supplementary Materials for Gut-resident CX3CR1 hi macrophages induce tertiary lymphoid structures and IgA response in situ Balázs Koscsó, Sravya Kurapati, Richard R. Rodrigues, Jelena Nedjic, Kavitha Gowda, Changsik Shin, Chetna Soni, Azree Zaffran Ashraf, Indira Purushothaman, Maryknoll Palisoc, Sulei Xu, Haoyu Sun, Sathi Babu Chodisetti, Eugene Lin, Matthias Mack, Yuka Imamura Kawasawa, Pingnian He, Ziaur S. M. Rahman, Iannis Aifantis, Natalia Shulzhenko, Andrey Morgun, Milena Bogunovic* *Corresponding author. Email: [email protected] Published 10 April 2020, Sci. Immunol. 5, eaax0062 (2020) DOI: 10.1126/sciimmunol.aax0062 The PDF file includes: Methods Fig. S1. Model of streptomycin-pretreated Salmonella-induced colitis in mice on a Salmonella- resistant background. Fig. S2. Flow cytometry gating strategy to identify MP populations in infected LB. Fig. S3. Response to Salmonella infection in mice lacking Mϕs or cDC2. Fig. S4. Gene expression profiles of inflammatory MP subsets. Fig. S5. Intestinal GC B cells express genes related to IgA class switch. Fig. S6. Response to Salmonella infection in mice after CD4 + T cell depletion. Fig. S7. Development strategy and characterization of Ccr7 fl/fl and Lyz2:Ccr7 –/– mice. Fig. S8. Role of CX3CR1 + Mϕ subsets in the induction of Salmonella-specific T cell responses. Fig. S9. Response to Salmonella infection in Ccr7 –/– mice. References (99104) Other Supplementary Material for this manuscript includes the following: (available at immunology.sciencemag.org/cgi/content/full/5/46/eaax0062/DC1) Table S1. Parametric P values and geometric means of expression levels of cDC and Mϕ genes analyzed in fig. S1B (separate Excel file). Table S2. List of genes and pathways included in pathway analysis (separate Excel file). Table S3. Parametric P values and geometric means of expression levels of each pathway (separate Excel file). Table S4. Parametric P values and geometric means of expression levels of all genes in the analyzed pathways (separate Excel file). Table S5. Raw data file with P values (separate Excel file).

New Supplementary Materials for · 2020. 4. 6. · Sciences, Farmingdale, NY) 6 hours before euthanasia. Colons were removed and washed in HBSS containing 10 μg/ml brefeldin A followed

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

  • immunology.sciencemag.org/cgi/content/full/5/46/eaax0062/DC1

    Supplementary Materials for

    Gut-resident CX3CR1hi macrophages induce tertiary lymphoid structures and IgA

    response in situ

    Balázs Koscsó, Sravya Kurapati, Richard R. Rodrigues, Jelena Nedjic, Kavitha Gowda, Changsik Shin, Chetna Soni, Azree Zaffran Ashraf, Indira Purushothaman, Maryknoll Palisoc, Sulei Xu, Haoyu Sun, Sathi Babu Chodisetti, Eugene Lin,

    Matthias Mack, Yuka Imamura Kawasawa, Pingnian He, Ziaur S. M. Rahman, Iannis Aifantis, Natalia Shulzhenko, Andrey Morgun, Milena Bogunovic*

    *Corresponding author. Email: [email protected]

    Published 10 April 2020, Sci. Immunol. 5, eaax0062 (2020)

    DOI: 10.1126/sciimmunol.aax0062

    The PDF file includes:

    Methods Fig. S1. Model of streptomycin-pretreated Salmonella-induced colitis in mice on a Salmonella-resistant background. Fig. S2. Flow cytometry gating strategy to identify MP populations in infected LB. Fig. S3. Response to Salmonella infection in mice lacking Mϕs or cDC2. Fig. S4. Gene expression profiles of inflammatory MP subsets. Fig. S5. Intestinal GC B cells express genes related to IgA class switch. Fig. S6. Response to Salmonella infection in mice after CD4+ T cell depletion. Fig. S7. Development strategy and characterization of Ccr7fl/fl and Lyz2:Ccr7–/– mice. Fig. S8. Role of CX3CR1+ Mϕ subsets in the induction of Salmonella-specific T cell responses. Fig. S9. Response to Salmonella infection in Ccr7–/– mice. References (99–104)

    Other Supplementary Material for this manuscript includes the following: (available at immunology.sciencemag.org/cgi/content/full/5/46/eaax0062/DC1)

    Table S1. Parametric P values and geometric means of expression levels of cDC and Mϕ genes analyzed in fig. S1B (separate Excel file). Table S2. List of genes and pathways included in pathway analysis (separate Excel file). Table S3. Parametric P values and geometric means of expression levels of each pathway (separate Excel file). Table S4. Parametric P values and geometric means of expression levels of all genes in the analyzed pathways (separate Excel file). Table S5. Raw data file with P values (separate Excel file).

  • Methods

    In vivo cell depletion experiments.

    We previously established that mucosal Ms in the normal intestine express CSF1R, require

    CSF1R signaling for their development, and are selectively depleted by CSF1R blocking

    monoclonal antibody (mAb) with no significant effect on mucosal cDCs, granulocytes or

    mononuclear phagocyte (MP) populations of the mesenteric lymph nodes (MLN) (38). Batf3–/–

    mice were used as an established model lacking cDC1 (40). ItgaxCre

    Notch2fl/fl

    mice were used as

    an established model deficient in cDC2 (36, 37).

    To deplete monocytes mice were injected intraperitoneally with 20 μg of MC-21 CCR2 mAb

    (97) or with control rat IgG on days 1, 2 and 3 of infection with Salmonella. Monocyte depletion

    was confirmed in the blood.

    To deplete CD4+ T cells, mice were injected intraperitoneally with 250 μg of CD4 mAb (clone

    GK1.5, BioXCell, West Lebanon, NH) or with control rat IgG 5 days before infection, and the

    injection was repeated every two days until infection with Salmonella. The injections were

    repeated with the same dose twice every week during the course of the experiment. T cell

    depletion was confirmed in the blood.

    Tamoxifen treatment

    To induce Cre expression, Cx3cr1CreER/WT

    H2-Ab1WT/fl

    or Cx3cr1CreER/WT

    H2-Ab1fl/fl

    mice were

    treated with 0.125 mg/g of body weight of tamoxifen (Sigma) dissolved in corn oil by oral

    gavage 7 days before infection. The treatment was repeated with the same dose on day 3 post-

    infection (Fig. S9J). The model was adapted from previously published studies (66, 98) by

  • titrating down tamoxifen dose and treatment regimen to target only the CX3CR1hi

    M

    population.

    Adoptive OT-II T cell transfer

    For adoptive transfer experiments, OT-II mice were backcrossed to CD45.1+ background to

    create congenic OT-II mice and bred with 129S1 mice. OT-II T cells were purified from spleens

    and lymph nodes of mice from F1 generation of congenic B6 CD45.1+ OT-II mice crossed to

    129S1 mice by negative selection using biotinylated CD11c, F4/80, CD19, NK1.1 and MHCII

    mAbs (Thermo Fisher Scientific) and Streptavidin MicroBeads (Miltenyi Biotec, Auburn, CA).

    Mouse strains backcrossed to Salmonella-resistant background as described above were used as

    recipients. Recipient mice were anesthetized by ketamine/xylazine, and 4-8x106 CFSE-labeled

    (Thermo Fisher Scientific) OT-II T cells were transferred i.v. via retro-orbital injection on the

    day of infection with SL-OVA.

    Tissue preparation

    Single cell suspensions were prepared from MLNs, SB after removing Peyer’s patches and LB

    (cecum and colon) after removing cecal and colonic patches as described previously (72). As cell

    surface CXCR5 expression is undetectable after collagenase treatment we used an alternative

    tissue homogenization approach to detect Tfh cells. Briefly, LB was processed and epithelial

    cells were removed as described (72) followed by cutting the tissue to 2 mm pieces and

    incubation for an additional 45 min in HBSS containing 2% fetal bovine serum (FBS) and EDTA

    in a shaking incubator at 37°C. Tissue pieces were homogenized by pipetting and passed through

    70 μm nylon mesh filters.

  • Immunohistochemistry

    Colons were harvested from mice at days 0, 7, or 10 after infection with SL1344, rinsed in PBS,

    and rolled around a sterile tooth pick before freezing in OCT compound (Sakura Finetek,

    Torrance, CA) on dry ice. 8 μm cross-sections were cut using a cryostat (Leica Biosystems,

    Buffalo Grove, IL). Tissues sections were fixed with 4% PFA or acetone and blocked with goat

    or donkey serum and stained with the following antibodies: anti-mouse CD45R (B220, clone

    RA3-6B2), anti-human/mouse GL7 (clone GL-7), anti-mouse CD4 (clone GK1.5), anti-mouse

    CD45.1 (clone A20), anti-mouse MAdCAM-1 (rabbit polyclonal, R&D Systems, Minneapolis,

    MN) and BacTrace FITC-labeled goat polyclonal anti-Salmonella CSA-1 antibody (Kirkegaard

    & Perry Laboratories, now in Seracare) followed by the secondary reagents. For CXCL13

    staining infected Cx3cr1GFP/WT

    mice were injected i.p. with 0.25 mg brefeldin A (Enzo Life

    Sciences, Farmingdale, NY) 6 hours before euthanasia. Colons were removed and washed in

    HBSS containing 10 μg/ml brefeldin A followed by fixation in 4% PFA and 2% sucrose. Fixed

    colons were rolled around a sterile toothpick before freezing in OCT compound. 8 μm cross-

    sections were cut using a cryostat blocked with donkey serum and stained using anti-mouse

    B220 (clone RA3-6B2) and goat polyclonal anti-mouse CXCL13 Ab (R&D Systems) (64).

    Tissue sections were counterstained with DAPI. Slides were mounted on Prolong Diamond

    (Thermo Fisher Scientific) and imaged using Leica DM4000 (Leica Microsystems, Buffalo

    Grove, IL), Zeiss Axio Observer.Z1 fluorescent microscopes (Carl Zeiss Microscopy, White

    Plains, NY) or Nikon Eclipse Ti confocal microscope. Images were analyzed using Las-X

    (Leica), Zeiss ZEN 2.3 pro or Volocity (PerkinElmer, Waltham, MA) software packages. For

    quantification, tertiary lymphoid structures (TLS) were identified as diffuse non-encapsulated

    aggregates of B220+ cells (follicles) with an area greater than 10

    -3 mm

    2. Follicular area was

  • determined using the tools in the image analyzer software packages listed above. Follicle

    numbers for each colon were determined by calculating the average of follicle numbers counted

    on at least four sections cut 100 μm apart from each other.

    Flow cytometry

    The following primary antibodies were used for flow cytometry: anti-mouse CD45 (clone 30-

    F11), anti-mouse CD11c (clone N418), anti-mouse CD11b (clone M1/70), anti-mouse CD103

    (clone 2E7), anti-mouse MHCII (I-A/I-E) (clone MS/114.15.2), anti-mouse CD115

    (cloneAFS98), anti-mouse F4/80 (clone BM8), anti-mouse Ly6C (clone HK1.4), anti-mouse

    Ly6G (clone 1A8-Ly6g), anti-mouse CD45R (B220) (clone RA3-6B2), anti-mouse CD19 (clone

    eBio1D3), anti-human/mouse GL7 (clone GL-7), anti-mouse CD4 (clone GK1.5), anti-mouse

    CD4 (clone RM4-5), anti-mouse TCRβ (clone H57-597), anti-mouse CD45.1 (clone A20), anti-

    mouse CD25 (clone PC61.5), anti-mouse CD199 (CCR9) (clone eBioCW-1.2), anti-mouse V

    alpha 2 TCR (clone B20.1), anti-mouse CD101 (clone Moushi101) from Thermo Fisher

    Scientific; anti-mouse CD95 (clone Jo2), anti-mouse CXCR5 (clone 2G8) from BD Pharmingen

    (San Diego, CA); anti-mouse CD279 (PD-1) (clone 29F.1A12), anti-mouse CD16/32 (clone 93),

    anti-mouse CD64 (clone X54-5/7.1), anti-mouse XCR1 (clone ZET) from Biolegend (San Diego,

    CA). Samples were acquired on LSRII flow cytometers (BD). For cell sorting a FACSAria III

    instruments were used (101). The purity of sorted populations was confirmed by running the

    collected cells on LSRII flow cytometer (Fig. S2C). Data analysis was performed using FlowJo

    software (FlowJo LLC, Ashland, OR). Gating strategies to identified MP populations, GC B

    cells and Tfh cells are shown on Fig. S2A-B, Fig. S5A and Fig. S6D, respectively. Total cell

    numbers in LB were determined as described previously (72).

  • To compare MP populations in steady state and inflammation with unbiased analysis, control

    (day 0) and infected (day 3) LB samples were analyzed by flow cytometry and 30000 MP events

    from each (see MP subset gate on Fig. S2A) were concatenated. Unsupervised t-SNE analysis

    was performed with FlowJo using the t-SNE plugin set to 10 phenotypic markers (MHCII,

    CD11b, CD16/32, CD64, CX3CR1-GFP, CD11c, CD103, CD101, XCR1, Ly6G), 2500

    iterations, perplexity of 100, and Eta at 200.

    RNA isolation and RT-PCR

    Total RNA was isolated from sorted cells using Trizol as described previously (72). RNA was

    reverse transcribed using RNA to cDNA Ecodry premix (Takara Bio, Kusatsu, Japan). Gene

    expression was assessed using Power SYBR Green PCR Master mix (Life Technologies) on a

    StepOnePlus qPCR instrument. Il10, Tgfb1, Aicda, Ccr7, Cxcl13, Tnf, Tnfsf13, Tnfsf13b and

    Actb PrimeTime qPCR primers were purchased from Integrated DNA Technologies (Skokie, IL).

    Additional primers used: IaF (5’-CCAGGCATGGTTGAGATAGAGATAG-3’), CaR (5’-

    GAGCTGGTGGGAGTGTCAGTG-3’), ImF (5’-CTCTGGCCCTGCTTATTGTTG-3’) (49).

    The primers IaF and CaR were used to detect IgA germline transcript (aGT), and ImF and CaR

    were used to detect rearranged, post switch IgA transcript (PST). Gene expression levels were

    calculated by normalization to Actb, and shown as fold change over the population described in

    figure legends.

    RNA sequencing

    Total RNA was isolated from cell populations sorted from LB of control mice and mice infected

    with SL1344 (day 5). Samples for each population were prepared by pooling cells sorted from 5

    mice. In total, 5 samples were prepared, representing 5 independent cell sorting experiment. The

  • cDNA libraries were prepared using the QuantSeq 3’mRNA-Seq Library Prep Kit FWD for

    Illumina (Lexogen, Vienna, Austria) as per the manufacturer’s instructions. Briefly, total RNA

    was reverse transcribed using oligo (dT) primers. The second cDNA strand was synthesized by

    random priming in a manner that DNA polymerase is efficiently stopped when reaching the next

    hybridized random primer, and only the fragment close to the 3’ end gets captured for later

    indexed adapter ligation and PCR amplification. The processed libraries were assessed for size

    distribution and concentration using BioAnalyzer High Sensitivity DNA Kit (Agilent

    Technologies, Santa Clara, CA). Pooled libraries were diluted to 2 nM in EB buffer (Qiagen,

    Germantown, MD) and then denatured using the Illumina protocol. The denatured libraries were

    diluted to 10 pM in pre-chilled hybridization buffer, loaded onto a TruSeq v2 Rapid flow cell on

    an Illumina HiSeq 2500 and run for 50 cycles using a single-read recipe according to the

    manufacturer's instructions. De-multiplexed sequencing reads were generated using Illumina

    bcl2fastq (released version 2.18.0.12) allowing no mismatches in the index read.

    Sequences were processed to remove adapter, poly A and low-quality bases by BBTools

    (https://jgi.doe.gov/data-and-tools/bbtools/) using bbduk parameters of k=13, ktrim=r,

    forcetrimleft=12, useshortkmers=t, mink=5, qtrim=r, trimq=15, minlength=20. Reads were

    aligned to mouse genome and transcriptome (ENSEMBL NCBIM37) using Tophat (v2.1.1)

    (102) with default parameters. Number of reads per million for mouse genes were counted using

    HTSeq (v 0.6.0) (103) and quantile normalized.

    In order to better understand differences between M populations and cDCs we focused our

    analyses on specific pathways known to be associated with MP. Genes and pathways for analysis

    were selected based on their described roles in physiological processes linked to MP and enteric

    https://jgi.doe.gov/data-and-tools/bbtools/

  • infections using NCBI BioSystems and Kyoto Encyclopedia of Genes and Genomes (KEGG)

    databases. The list of genes and pathways is shown in Table S2.

    The expression of pathways was compared between cell populations using a previously

    described approach with minor modifications (104). Specifically, the gene expression per sample

    was normalized by the gene’s median expression across all samples. Then for each sample, the

    geometric mean of all genes in a pathway was calculated to represent the expression of the

    pathway in the sample (Table S3, S4). These values (per pathway per sample) were used for

    class comparison analysis in BRBArrayTools (https://brb.nci.nih.gov/BRB-ArrayTools/) to

    identify differentially expressed pathways between cell populations. The values of geometric

    means of the pathway per cell populations were used to generate heat maps in R using heatmap.2

    (gplots), row scaling, and the pathways were manually organized.

    Individual gene expression was compared between cell populations using BRBArrayTools

    software. Adjustment for multiple hypotheses was performed by calculating False Discovery

    Rate using Benjamini-Hochberg Procedure. The expression levels of differentially expressed

    genes (FDR < 1%) obtained by comparing (day 0) SB cDC2 vs Per. MFs was used for

    hierarchical clustering of samples in BRB with centered correlation and average linkage.

    Individual heat maps per pathway illustrate the geometric mean of gene expression per cell

    population. The heat maps were generated in R using heatmap.2 (gplots), row scaling, and the

    genes were organized as per decreasing (CX3CR1hi

    D.5 SL / CX3CR1lo

    D.5 SL) fold change.

    In vitro B cell cultures

  • MP populations were sorted from the LB of Cx3cr1GFP/WT

    mice harvested at day 6 of infection. B

    cells were purified from spleens of naïve CD45.1+ congenic mice using EasySep mouse B cell

    isolation kit (STEMCELL Technologies, Vancouver, Canada). B cells were cultured with MP

    subsets in RPMI-1640 medium supplemented with 10% FBS, 5.5x10-5

    M 2-ME, 10 mM HEPES,

    1 mM sodium pyruvate (RPMI), 100 units/ml penicillin, 100 μg/ml streptomycin, 0.25 μg/ml

    amphotericin B and 100 μg/ml gentamicin in presence or absence of SL1344. At day 4 of

    culture, supernatants were analyzed to assess IgG and IgA levels with ELISA and cells were

    collected and analyzed by flow cytometry to detect GC B cells.

    In vitro antigen presentation assays

    MP subsets were sorted from the LB of Cx3cr1GFP/WT

    mice harvested at day 6 after infection. T

    cells were purified from MLNs and spleens of congenic OT-II mice using EasySep mouse CD4+

    T cell isolation kit (STEMCELL). MP subsets were incubated with HK SL-OVA in RPMI in

    presence of rGM-CSF/rM-CSF (Ms) or rFlt3L (DCs) for 2 hours. Isolated OT-II T cells labeled

    with CFSE were added to the wells at an APC:T cell ratio of 1:1 in RPMI with 100 units/ml

    penicillin, 100 μg/ml streptomycin, 0.25 μg/ml amphotericin B and 100 μg/ml gentamicin. T cell

    activation and proliferation were assessed by flow cytometry after culturing for 4 days at 37°C

    and 5% CO2. Bone marrow derived (BM) DCs and Ms were prepared as described previously

    (105). BM Ms or BM DCs cells were cultured in RPMI with HK SL-OVA for 18 h at 37C and

    5% CO2. Thereafter, OT-II T cells labeled with CFSE were added to SL-OVA loaded Ms or

    DCs at a 1:1 ratio. After 72 hours of co-culture, OT-II T cell activation and proliferation was

    assessed by flow cytometry.

    Ex vivo Transwell assay

  • For B cell migration assays 96-well Transwell support system was used (Corning, NY, USA)

    with 5 um pore size. CX3CR1hi

    Ms were sorted from the LB of Cx3cr1GFP/WT

    mice harvested at

    day 12 of infection cultured for 3 hours in the lower chamber of Transwell plate in presence or

    absence of CXCL13 Ab (50 g/ml) (64). Spleens were collected from naïve CD45.1+ congenic

    mice and after homogenization splenocytes were re-suspended in RPMI-1640 containing 10 mM

    HEPES, 0.5% fatty acid-free BSA (Sigma), 100 units/ml penicillin, 100 μg/ml streptomycin,

    0.25 μg/ml amphotericin B and 100 μg/ml gentamicin and incubated in 37C water bath for 30

    min. Splenocytes were added to the upper chamber of the Transwell plate at 1:1 ratio and

    incubated for 3 hours at 37C and 5% CO2. For input wells, splenocytes were added to the lower

    well without CX3CR1hi

    Ms. After the incubation, cells from the lower well were collected and

    the numbers of migrated B cells were analyzed using flow cytometry.

  • Fig. S1. Model of streptomycin-pretreated Salmonella-induced colitis in mice on a

    Salmonella-resistant background. WT mice were infected with SL1344 as described in

    Methods.

    (A) Salmonella CFU in the lumen, intestinal mucosa and isolated organized gut associated

    lymphoid tissue (GALT) from various intestinal regions (D.1 p.i.) (n=5).

    (B) Body weight loss during Salmonella-induced colitis (n=8).

    (C) Lipocalin-2 concentrations in fecal pellets of WT mice with Salmonella-induced colitis

    (n=4).

    (D) Neutrophil counts in various intestinal regions shown as % of total viable cells (D.14 p.i.)

    (n=3-10).

    (E) Counts of myeloid cell subsets in the small bowel (SB) and large bowel (LB) (n=5-8).

  • Graphs show mean±SEM, data is representative or combination of 2 independent experiments.

    Statistical analysis: two-way ANOVA. *p

  • Fig. S2. Flow cytometry gating strategy to identify MP populations in infected LB.

    Cx3cr1GFP/WT

    , Notch2fl/fl

    (Cont.), ItgaxCre

    Notch2fl/fl

    (Itgax:Notch2–/–

    ) and

    ItgaxCre

    Notch2fl/fl

    Cx3cr1GFP/WT

    mice were infected with SL1344.

  • (A) Flow cytometry dot plots show gating strategy to identify myeloid cells in the infected (D.3

    p.i.) large bowel (LB) of Cx3cr1GFP/WT

    mice.

    (B) Flow cytometry dot plots show gating strategy to identify mononuclear phagocyte (MP)

    populations in normal (D.0) and infected (D.10 p.i.) large bowel (LB) of Cx3cr1GFP/WT

    mice.

    (C) Representative flow cytometry dot plots show the purity of MP populations sorted from

    normal (D.0) LB, SB and peritoneal lavage and from infected LB (D.5 p.i.).

    (D) Flow cytometry dot plots show percentage (%) of CCR2+ cells (identified as RFP

    + cells)

    among blood monocyte and LB MP populations in Ccr2RFP/WT

    Cx3cr1GFP/WT

    mice infected with

    Salmonella (D.8 p.i.).

    (E) Graphs show myeloid cell counts in the LB before and after infection with SL1344 in

    Notch2fl/fl

    (Cont.) and ItgaxCre

    Notch2fl/fl

    (Itgax:Notch2–/–

    ) mice (n=2-4).

    (F) Flow cytometry dot plots show MP populations in the LB and SB of Notch2fl/fl

    Cx3cr1GFP/WT

    (Cont.) and ItgaxCre

    Notch2fl/fl

    Cx3cr1GFP/WT

    (Itgax:Notch2–/–

    ) mice infected with Salmonella (D.3

    p.i.).

    Graphs show mean±SEM, data is representative of 2 independent experiments. Statistical

    analysis: two-way ANOVA. *p

  • Fig. S3. Response to Salmonella infection in mice lacking Mϕs or cDC2. WT or Cx3cr1GFP/WT

    mice were injected with CSF1R mAb to deplete Ms (CSF1R Ab) or control IgG before

    infection with SL1344 (A). ItgaxCre

    Notch2fl/fl

    (Itgax:Notch2–/–

    ) mice were used as a cDC2-

    deficient model, their Notch2fl/fl

    (Cont.) littermates were used as control (D-J). Mice were

    rescued with ampicillin (G-H).

    (A) Graphs show myeloid cell counts in the LB before and after infection with SL1344 in

    Cx3cr1GFP/WT

    mice injected with CSF1 mAb to deplete Ms (∆M) or control IgG (Cont.) (n=5-

    8).

    (B) Salmonella CFU in the mesenteric lymph nodes (MLNs) and spleen of WT mice at indicated

    time points p.i (n=7-14).

    (C) Numbers of Salmonella (SL)-specific IgA-producing cells in the LB and MLNs of WT mice

    before and at indicated time points after infection as measured by ELISpot (n=2-8).

    (D) Survival curves of control and Itgax:Notch2–/–

    mice (n=8-9).

    (E) Weight loss as percent of body weight at D.0 by control and Itgax:Notch2–/–

    mice (n=8-9).

    (F) Salmonella CFU in fecal pellets in control and Itgax:Notch2–/–

    mice (D.1 p.i.) (n=6-7).

    (G) Lipocalin-2 concentrations in fecal pellets of infected control and Itgax:Notch2–/–

    mice (n=8-

    9).

    (H) Salmonella (SL)-specific antibody titers in fecal pellets (mucosal IgA) and blood (serum IgA

    and IgG) of control and Itgax:Notch2–/–

    mice (n=8-9).

    (I) Numbers of TLS follicles (TLF) per colonic section in control and Itgax:Notch2–/–

    mice (D.10

    p.i.) (n=3).

    (J) Area of follicles in colonic sections of control and Itgax:Notch2–/–

    mice (D.10 p.i.) (n=3).

  • Graphs show mean±SEM and are representative or combination of at least 2 independent

    experiments. Statistical analysis: Student’s t-test or two-way ANOVA. *p

  • Fig. S4. Gene expression profiles of inflammatory MP subsets. Cx3cr1GFP/WT

    mice were

    infected with SL1344.

    (A) RNAseq heat maps show expression levels of genes related to the indicated pathways in MP

    subsets isolated by FACS from Salmonella-infected LB (D.5 p.i.). Because intestinal MP subsets

    were sorted based on their expression of the antigen-presenting molecule MHCII, to highlight the

  • differences in the expression of genes from the Antigen presentation/MHCI and Antigen

    presentation/MHCII pathways among LB MP subsets, they are compared to MHCII– peritoneal

    Ms isolated from normal mice on the same genetic background. Each sample represents the

    geometric mean of 5 independent samples.

  • Fig. S5. Intestinal GC B cells express genes related to IgA class switch. Mice were infected

    with WT Salmonella SL1344.

    (A) Gating strategy to identify GC B cells in normal and infected (D.10 p.i.) LB of WT mice as

    CD19+B220

    +GL7

    +CD95

    + cells. Gated on viable DAPI

    –CD45

    + singlets.

    (B) Expression of Aicda, IgA germline transcript (aGT) and mature rearranged IgA (Iµ-Cα)

    transcript (PST) by GL7+ and GL7

    – CD19

    +B220

    + B cells (Fig. S5A) purified by FACS from

    infected LB (D.10 p.i.) as measured by qPCR (n=3-4).

    (C) Flow cytometry dot plots show proportions of B cells (thick gates) and CD4+ T cells among

    total lymphocytes in tertiary lymphoid structures (follicles) (TLFs), mucosa, and colonic and

    cecal patches of infected LB isolated from a WT mouse (D.14 p.i.). Gated on CD45+CD11b

    CD11c– cells.

    Graphs show mean±SEM and are representative of at least 2 independent experiments. Statistical

    analysis: Student’s t test or two-way ANOVA. *p

  • Fig. S6. Response to Salmonella infection in mice after CD4+ T cell depletion. H2-Ab1

    fl/fl

    (Cont.), Lyz2Cre/WT

    H2-Ab1fl/fl

    (Lyz2:MHCII–/–

    ), WT mice treated with control IgG (Cont.) or CD4

    Ab to deplete CD4+ T cells were infected with SL1344.

  • (A) Statistical summary of flow cytometry data show proportions of MHCII+ cells among MP

    populations and B220+ B cells in the LB and MLNs (for B cells only) isolated from infected

    Cont. and Lyz2:MHCII

    –/– mice (D.7 p.i.) (n=5).

    (B) Bone marrow derived (BM) Ms and DCs from Cont. and Lyz2:MHCII–/–

    mice were co-

    cultured with OT-II cells in the presence or absence of 2x106 CFU of heat-killed (HK) SL1344-

    OVA (SL-OVA). Flow cytometry dot plots show percentages of proliferating CFSE low cells

    among OT-II Vα2+CD4

    + T cells on D.4 of co-culture.

    (C) Summary of B. Data is representative of 2 independent experiments (n=2).

    (D) Gating strategy to identify Tfh cells as TCRβ+CD4

    +CXCR5

    + PD1

    + cells in the LB from

    SL1344-infected WT mice. Gated on viable DAPI–CD45

    + singlets.

    (E) Survival curves of mice treated with IgG control or CD4 Ab (n=8).

    (F) Weight loss as percent of D.0 in mice treated with IgG control or CD4 Ab (n=8).

    (G) Lipocalin-2 concentrations in fecal pellets collected from mice treated with IgG control or

    CD4 Ab (n=8).

    (H) Salmonella-specific antibody titers in fecal pellets (mucosal IgA) and blood (serum IgA and

    IgG) of mice treated with IgG control or CD4 Ab (n=8).

    (I) Numbers of Salmonella-specific IgA-producing cells in the LB of mice treated with IgG

    control or CD4 Ab (D.7 p.i.) as measured by ELISpot (n=5-6).

    (J) Number of follicles per colonic section in mice treated with IgG control or CD4 Ab (D.10

    p.i.) (n=8-9).

    (K) Area of follicles in colonic sections of mice treated with IgG control or CD4 Ab (D.10 p.i.)

    (n=8-9).

  • Graphs show mean±SEM and are representative of at least 2 independent experiments. Statistical

    analysis: Student’s t-test or two-way ANOVA. *p

  • Fig. S7. Development strategy and characterization of Ccr7fl/fl

    and Lyz2:Ccr7–/–

    mice.

    (A, B) Gene targeting strategy for generation of Ccr7fl/fl

    animals. A Pgk-NEO cassette flanked by

    Frt sites was inserted in the intronic region between exon II and exon III. LoxP sites are flanking

    exon III, which allows for Cre recombinase mediated deletion of the targeted locus. Primer sets

  • P1/P2 and the outlined probe were used for screening of ES colonies by PCR and Southern Blot.

    Grey bars depict the end of 5’ and 3’ homology arms.

    (C) PCR screen with primer combination P1/P2. Recombination frequency is 3.5% (12/346

    screened clones).

    (D) Ccr7 expression by cDC1 and Ms isolated by FACS from LB of SL1344-infected Ccr7fl/fl

    (Cont.) and Lyz2Cre

    Ccr7fl/fl

    (Lyz2:Ccr7–/–

    ) mice (D.5 p.i.) as measured by qPCR. Data shows fold

    change over control (n=4-8).

    (E) Absolute numbers of MP subsets in LB of infected Ccr7fl/fl

    Cx3cr1GFP/WT

    (Cont.) and

    Lyz2Cre

    Ccr7fl/fl

    Cx3cr1GFP/WT

    (Lyz2:Ccr7–/–

    ) mice (D.5 p.i.) (n=4-7).

    Bar graphs show mean±SEM (n=3-7) and are combination of 2 independent experiments.

    Statistical analysis: Student’s t test. *p

  • Fig. S8. Role of CX3CR1+ Mϕ subsets in the induction of Salmonella-specific T cell

    responses. WT, H2-Ab1fl/fl

    (Cont.) and Lyz2:MHCII–/–

    mice were infected with SL1344 or

    SL1344-OVA as indicated.

    (A) Gating strategy to identify adoptively transferred CD45.1+ OT-II cells in LB of WT recipient

    mice infected with either WT SL1344 (SL) or SL1344-OVA (SL-OVA).

    (B) Control and Lyz2:MHCII–/–

    mice were infected with SL1344-OVA one day prior to receiving

    CFSE-labeled congenic CD45.1+ OT-II T cells. Flow cytometry dot plots show percentages of

    proliferating CFSE low cells among total CD45.1+Vα2

    +CD4

    +CCR9

    – (LB) and

    CD45.1+Vα2

    +CD4

    + (MLNs) T cells in the LB and MLNs on D.5 p.i.

    (C) Statistical summary of B.

  • Bar graphs show mean±SEM (n=4-10) and are combination of 2 independent experiments.

    Statistical analysis: Student’s t test or two-way ANOVA. ***p

  • Fig. S9. Response to Salmonella infection in Ccr7–/–

    mice. Cx3cr1GFP/WT

    control (Cont.) and

    Cx3cr1GFP/WT

    Ccr7–/–

    mice, WT (Cont.) and Ccr7–/–

    mice or Cx3cr1CreER/WT

    H2-ab1WT/fl

    and

  • Cx3cr1CreER/WT

    H2-ab1fl/fl

    mice were infected with SL1344. Mice were rescued with ampicillin

    (B, C).

    (A) Absolute numbers of MP subsets in LB of infected Cx3cr1GFP/WT

    control and Cx3cr1GFP/WT

    Ccr7–/–

    mice (D.5 p.i.) (n=4).

    (B) Lipocalin-2 concentrations in fecal pellets of Cont. and Ccr7–/–

    mice (n=5-6).

    (C) Salmonella-specific antibody titers in fecal pellets (mucosal IgA) and blood (serum IgA and

    IgG) of Cont. and Ccr7–/–

    mice (n=7-8).

    (D) Numbers of follicles per colonic section in Cont. and Ccr7–/–

    mice before and after infection

    (n=3-6).

    (E) Area of follicles in colonic sections of control and Ccr7–/–

    mice after infection (D.10 p.i.)

    (n=3-6).

    (F) Kinetics of Tnfsf13a expression by MP subsets FACS-isolated from the normal (D.0) and

    infected LB as measured by qPCR. Data shows fold change over CX3CR1hi

    Ms (D.0) (n=2-3).

    (G) Gating strategy to identify a B220+ subset among CX3CR1

    hi Ms in the LB of SL1344-

    infected Cx3cr1GFP/WT

    mice. Gated on viable CD45+CD11b

    lo/hiCD11c

    lo/hiMHCII

    +CX3CR1

    hi

    population.

    (H) CX3CR1hi

    Ms and cDC1 isolated from LB of infected Cx3cr1GFP/WT

    mice (D.5 p.i.) were co-

    cultured in vitro with CFSE-labeled OT-II CD4+ T cells in the presence of 2x10

    6 CFU of heat-

    killed (HK) SL1344-OVA (SL-OVA) or OVA protein. Flow cytometry dot plots show

    percentages of proliferating CFSE low cells among OT-II Vα2+CD4

    + T cells on day 4 of co-

    culture.

    (I) Summary of H (n=1-3).

  • (J) Graphical summary of the experiment to selectively deplete CX3CR1hi

    Ms in

    Cx3cr1CreER

    H2-ab1fl/fl

    mice. Mice were treated with tamoxifen (Tam) 7 days before and 3 days

    after infection with SL1344.

    (K) Statistical summary of flow cytometry data show proportions of MHCII+ cells among B220

    +

    B cells in the infected LB isolated from tamoxifen-treated Cx3cr1CreER/WT

    H2-ab1WT/fl

    (Cre:WT/fl)

    or Cx3cr1CreER/WT

    H2-ab1fl/fl

    (fl/fl) mice (Cre:fl/fl) (D.7 p.i.) (n=3-6).

    Bar graphs show mean±SEM and are representative or combination of at least 2 independent

    experiments. Statistical analysis: Student’s t-test or two-way ANOVA. *p