Human carotid plaques. Formalin-fixed and paraffin-embedded arterial tissue sections were used after antigen retrieval by heating in microwave oven in buffer pH9. For single labeling, CARD9 rabbit polyclonal antibody (Abcam, Cambridge, UK) was used at 1:500 dilution, incubated for 1 hour and revealed using ABC-peroxidase technique (Vector Laboratories, Burlingame, CA, USA). For double labeling, the sections were first incubated with mouse monoclonal anti-CD68 antibodies diluted at 1:50 (to detect macrophages) or anti-α smooth muscle actin antibodies diluted at 1:100 (to detect smooth muscle cells), both from Dako-Agilent (Trappes, France). Sections were then incubated with species-specific secondary antibodies (1:500 dilution, 45 min at room temperature) conjugated to AlexaFluor 488 or 594 (Fischer Scientific) and mounted on microscope slides using the Prolong Antifade Diamond kit (Thermofisher). Image acquisition was performed on a laser scanning confocal microscope (Leica TCS SP8, Leica Microsystems). Immunostaining studies were performed on normal aorta, atherosclerotic plaques after endarterectomy or on carotid artery obtained post-mortem at autopsy (Ethical Committee CPP Ile de France 2013-13-19).
Animals. Experiments were conducted according to the guidelines formulated by the European Community for experimental animal use (L358-86/609EEC) and were approved by the Ethical Committee of INSERM and the French Ministry of Agriculture (agreement A75-15-32). Card9−/− and Rag2−/−Card9−/− mice (null for the Card9 gene), obtained from Dr. Harry Sokol (Centre de Recherche Saint-Antoine UMR_S 938, Paris, France) have been previously described 11. They were backcrossed for over 10 generations into a C57BL/6J background. Cd36−/− mice (null for the Cd36 gene) C57bl/6 Cd36−/− mice were generated in Dr. Roy Silverstein’s laboratory (Febbraio et al JBC 1999) and were crossed with Card9−/− mice to generate Card9−/−Card36−/− mice. Ten-week old female C57BL/6J Ldlr−/− mice were subjected to medullar aplasia by lethal whole-body irradiation (9.5 grays). The mice were repopulated with an intravenous injection of bone marrow (BM) cells isolated from femurs and tibias of sex-matched C57BL/6J Card9−/− mice or Card9+/+ littermates. After 4 weeks of recovery, mice were fed a pro-atherogenic high-fat diet containing 15% fat, 1.25% cholesterol, and 0% cholate for 8 weeks. During the first 14 days following BM cell transfer, animals were administered an antibiotic in their drinking water (Baytril→, Enrofloxacine).
Pharmacological In Vivo Treatment
Eight-week old male Apoe−/−Card9+/+ and Apoe−/−Card9−/− mice were treated with daily intraperitoneal injections of rapamycin (4 mg/kg body weight) for 6 weeks and were put on a high fat diet. Eight-week old male Apoe−/−Card9+/+ and Apoe−/−Card9−/− mice were treated with metformin (300 mg/kg body weight, drinking water) for 6 weeks and were put on a high fat diet.
Extent and composition of atherosclerotic lesions. Plasma cholesterol was measured using a commercial cholesterol kit (DiaSys® Cholestérol FS*). Quantification of lesion size was performed as described previously 14. Briefly, the basal half of the ventricles and the ascending aorta were perfusion-fixed in situ with 4% paraformaldehyde, then transferred to a PBS-30% sucrose solution, embedded in frozen OCT and stored at − 80°C. Serial 10-µm sections of the aortic sinus with valves (80 per mouse) were cut on a cryostat, as previously described 15. One section out of 5 was used for plaque size quantification after Oil red O staining. In total, 16 sections spanning over 800 µm of the aortic root were used to determine the mean lesion area for each mouse. After PBS flushing, the aorta from the root to the iliac bifurcation was removed and fixed with 10% neutral-buffered formalin. After thorough PBS washing, the adventitial tissue was removed and the aorta was longitudinally opened to expose the luminal surface for en-face visualization of atherosclerotic lesions after Oil Red O staining. Quantification of Oil Red O positive surface area was performed by a blinded operator. Aortic collagen content was detected using Sirius red staining. Necrotic core surface was quantified after Masson’s Trichrome staining. At least 4 sections per mouse were examined for each immunostaining, and appropriate negative controls were used. Morphometric studies were performed using Histolab software (Microvisions)16. For immunostaining on mouse atherosclerotic plaques, we used antibodies raised against Card9 (AA 274–530), MOMA-2 (MAB1852, Merck Milllipore→) and CD3 (A0452, DakoⓇ). The presence of macrophages was determined using specific antibodies, as previously described 16. TUNEL (Terminal dUTP nick end-labeling) staining was performed using In Situ Cell Death Detection Kit (histochemistry staining) and TMR Red kit (Fluorescent staining) (Roche).
Splenocyte culture. Splenocytes were cultured in RPMI 1640 supplemented with Glutamax, 10% fetal calf serum (FCS), 0.02 mM β-mercaptoethanol and antibiotics. For cytokine measurements, splenocytes were stimulated with LPS (1 µg/ml) and IFN-γ (100 UI/ml) for 24 hours. IL-1β, IL-10 and TNF-α production in the supernatants were measured using specific ELISA immunoassay kits (BD Biosciences).
Spleen cell recovery and purification. Spleen cells were purified according to standard protocols as follows. CD4+ T cells were negatively selected using a cocktail of anti-CD8a, anti-CD11b, anti-CD45R, anti-DX5, anti-ter 119 antibody-coated magnetic beads, yielding CD4+ cells with > 95% purity (Miltenyi Biotech). CD11c+ cells were positively selected with biotin-conjugated anti-CD11c mAb (7D4, PharMingen) and captured with streptavidin microbeads (Miltenyi Biotec) followed by 2 consecutive magnetic cell separations using LS columns (Miltenyi Biotec), yielding CD11c+ cells with > 80% purity.
CD4 + T cell culture and cytokine assays. Cells were cultured in RPMI 1640 supplemented with Glutamax, 10% FCS, 0.02 mM β-mercaptoethanol and antibiotics. For cytokine measurements, CD4+ T cells were cultured at 1x105 cells/well for 48 hours in anti-CD3-coated microplates (5 µg/ml). In some experiments, CD4+ T cells were stimulated with purified soluble CD3-specific antibody (1 µg/ml, Pharmingen) in the presence of antigen-presenting cells that were purified on CD11c-coated magnetic beads (Miltenyi Biotech). Secretion of IL-17A, IL-22, IL-10 and IFN-γ secretion in the supernatants was measured using analyte-specific ELISAs (BD Biosciences and R&D Systems). T cell proliferation was measured using the Quick Cell proliferation Assay Kit II (Abcam).
Macrophage Experiments
Primary macrophages were derived from mouse BM cells (BMDMs). Tibias and femurs of C57Bl6/J male mice were dissected and their marrow flushed out. Cells were grown in RPMI 1640 medium, 10% FCS, and 15% Macrophage–Colony-Stimulating Factor (M-CSF)-rich L929-conditioned medium for 7 days at 37°C. To analyze oxidized LDL uptake, BMDMs were exposed to human oxidized LDL (25µg/ml) for 24 hours (see oxidation method below). Cells were then washed, fixed and stained using Bodipy (493/503, Thermofischer Scientific D3922). Foam cells were quantified blindly on 6–8 fields and the mean was recorded. To analyze apoptosis susceptibility, macrophages were incubated with OxLDL (200 µg/ml) for 24 h or in a FCS-poor medium (1% FCS, starvation) or with TNF-α (10 ng/ml) and cycloheximide (10 µmol/l) for 6 h or with etoposide (50 µmol/l). Apoptosis was determined by independent experiments using Annexin V- (FITC) apoptosis detection kit with 7-AAD (PerCP) (BD Biosciences) according to the manufacturer's instructions. Intracellular cholesterol (total and ester) quantification was done using Amplex® Red Cholesterol Assay Kit (Invitrogen A12216).
Cholesterol Efflux Assays
BMDMs were obtained by differentiation of BM cells in Dulbecco’s modified Eagle medium (DMEM) supplemented with 10% fetal bovine serum, 2 mM glutamine, 20% L929 cell-conditioned media (as a source of M-CSF), and penicillin-streptomycin for 5 days. BMDMs were loaded with 50 µg/ml [3H]cholesterol-labeled acetylated LDL (acLDL, 1 µCi/mL) for 48 hours in serum-free DMEM supplemented with 50 mM glucose, 2 mM glutamine, 0.2% BSA (RGGB) and 100 µg/ml penicillin / streptomycin. The labeling medium was then removed and cells were washed twice in PBS and then equilibrated in RGGB for additional 16–24 hours. To measure cholesterol efflux, cells were incubated 4 hours at 37°C in the presence of 60µg/ml lipid-free apoAI (Sigma), or 30 µg/ml HDL-PL (density = 1.063–1.21 g/ml), isolated from normolipidemic plasma by preparative ultracentrifugation, as cellular cholesterol acceptor. Finally, culture media was harvested and cleared of cellular debris by brief centrifugation. Fractional cholesterol efflux (expressed as a percentage) was calculated as the amount of radio-label detected in the supernatants divided by total radio-label in each well (radioactivity in the supernatant plus radioactivity in the cells) obtained after lipid extraction from cells in a mixture of 3:2 hexane:isopropanol (3:2 vol/vol). The background cholesterol efflux obtained in the absence of any acceptor was subtracted from the efflux obtained with samples.
Flow Cytometry
Blood and spleen samples were collected at sacrifice for analysis of leukocyte subsets. Myeloid cells were identified as CD45 + CD11b+. Monocytes were identified as CD11b + CD115+. Among them, classical monocytes were Gr1high (or Ly6Chigh) and non-classical monocytes were Gr1low (or Ly6Clow). Neutrophils were identified as CD11b + CD115-Gr1+ (or CD11b + CD115-Ly6G+). B220 + IgM + B lymphocytes, CD4 + and CD8 + T lymphocyte subsets were also analyzed. Antibodies raised against CD11b, CD115, Gr1 (Ly6C and G), B220, CD4, CD8a, NK1.1, CD45, F4/80, CD3ε, MHC II, IgM, CD11c and CD36 were used for immunostaining and are listed in supplementary table 1.
Forward scatter (FSC) and side scatter (SSC) were used to gate live cells excluding red blood cells, debris, and cell aggregates in total blood cells and splenocytes preparations. Cells were acquired using a BD LSRII Fortessa flow cytometer (BD Biosciences) and analyzed with FlowJo™ (TreeStar, Inc.).
Quantitative real-time PCR.
Quantitative real-time PCR was performed, after Trizol RNA extraction and RT-PCR, on an ABI Prism 7700 (Applied Biosystems™) in triplicate for each sample. Expression of all genes has been normalised to the expression level of Gapdh. Relative expression was calculated using the 2-delta-delta CT method followed by geometric average, as recommended 17 ,18. The following primer sequences were used: Card9 (F: 5’- GAC CCT CTT AGT CCC AAT CTG − 3’; R: 5’- CTC GTC GTC ATT CTC ATA GTC TG -3’), Mrs1 (F: 5’- CCG TGA ATC TAC AGC AAA GCA − 3’; R: 5’- CCC AGT CCT TCA GTC TGA GG -3’), Scarb1 (F: 5’- CCT CCT GTT GCT GGT GCC CAT CAT − 3’; R: 5’- GCA CTG GTG GGC TGT CCG CTG AGA − 3’), Cd36 (F: 5’- TGG CCA AGC TAT TGC GAC ATG ATT A -3’; R: 5’- CGG GGA TTC CTT TAA GGT CGA TTT C -3’), Abcg1 (F: 5’- GAC AGC CAT CCC CGT CCT GCT CTT − 3’; R: 5’- CTC CCG CAG GAT GGC CTC TGA CTT − 3’), and Abca1 (F: 5’- GGC GGA CCT CCT GTG GTG TTT − 3’; R: 5’- GAA TCT CCG GGC TTT AGG GTC CAT − 3’).
Transcriptomic Analysis On Human Monocytes
The study was approved by the Committee for the Protection of Human Subjects in Biomedical Research (C10-14 Prédisposition génétique aux infections fongiques sévères). RNA sequencing libraries were prepared from 100 to 200 ng of total RNA using the Illumina® Stranded Total RNA Prep, Ligation with Ribo-Zero Plus library preparation kit, which allows performing a strand specific sequencing. This protocol includes a first step of enzymatic depletion of abundant transcripts from multiple species (including human cytoplasmic & mitochondria rRNA, mouse rRNA, rat rRNA, bacteria Gram +/- rRNA, human beta globin transcripts) using specific probes. cDNA synthesis was then performed and resulting fragments were used for dA-tailing followed by ligation of RNA Index Anchors. PCR amplification with indexed primers (IDT for Illumina RNA UD Indexes) was finally achieved, with 13 cycles, to generate the final cDNA libraries. Individual library quantification and quality assessment were performed using Qubit fluorometric assay (Invitrogen) with dsDNA HS (High Sensitivity) Assay Kit and LabChip GX Touch using a High Sensitivity DNA chip (Perkin Elmer). Libraries were then equimolarly pooled and quantified by qPCR using the KAPA library quantification kit (Roche). Sequencing was carried out on the NovaSeq 6000 instrument from Illumina using paired-end 2 x 100 bp, to obtain around 100 million clusters (200 million raw paired-end reads) per sample. Raw and normalized counts are provided in Supplementary Table 2.
We performed the gene set enrichment analysis using clusterProfiler v4.0.5 19 with selected pathways from Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases, and Benjamini-Hochberg correction was applied. Used keywords were: “apopto*”, “atheroscleros*”, “NF-kappa B”, “TNF”. All pathways with adjusted p-value below 0.05 were considered as significantly enriched. For each category, median expression of all gene included in core enrichment were calculated for each patient before plotting the heatmap.
Single-cell Analysis Of Card9/card9 Expression Patterns
The murine scRNA-seq dataset was generated by integrating single-cell gene expression data of aortic cells from steady-state or atherosclerotic vessels from several published studies 20–26. Data were integrated using canonical correlation analysis in Seurat v3 27. Human scRNA-seq data were mined from reference 28 and correspond to single-cell analysis of total cells from atherosclerotic coronary arteries dissected from explanted hearts of transplant recipients (n = 4 patients) and analyzed in Seurat v3. Expression of Card9/CARD9 was mapped using the FeaturePlot function in Seurat. For clarity, minimum and maximum gene expression cutoffs were applied, and cells with detectable transcripts were brought to the front of plots using the “order = TRUE” argument within the Seurat v3 FeaturePlot function.
Western-blot
Proteins from BMDMs were extracted by pipetting in ice-cold lysis buffer (NaCl 150 mM, HEPES 20 mM, EDTA 1 mM, EGTA 1 mM, NP-40 0.25% (Vol/Vol)) supplemented with protease (1 tab/10ml, Roche) and phosphatase (Na4VO3 2 mM) inhibitors). Protein extracts were briefly sonicated (5 pulses, amplitude 20, Vibracell 75021) and quantified by BCA (Thermofisher Scientific). Equal protein amounts were separated by SDS-PAGE (10% acrylamide) and transferred onto nitrocellulose membrane (0.2 µm, Biorad). For LC3b detection, SDS-PAGE separated proteins were transferred onto polyvinylidene difluoride membranes.
Membranes were then blocked with Tris Buffered Saline supplemented with 0.1% Tween-20 (TBST) and 2% BSA (2h at room temperature), then incubated with primary antibodies (overnight, 4ºC) diluted following the manufacturer’s recommendations. After three washes in TBST, membranes were incubated with species-specific horseradish peroxidase-conjugated secondary antibodies (1:8,000 dilution, 45 min at room temperature). After three washes in TBST, the peroxidase activity was detected using Clarity Western ECL Substrate (Biorad) using Cytiva’s ImageQuant Fluor 800. The migration position of transferred proteins was compared to the PageRuler Prestain Protein Ladder (10 to 170 kDa, Thermo Fisher Scientific). Densitometric analysis was performed using ImageJ software (NIH). Phosphorylated protein signals were normalized on total protein levels, whereas non-phosphorylated proteins were normalized on β-actin protein levels. Primary and HRP-coupled secondary antibodies used for immunoblotting experiments are listed in supplementary table 1. Uncropped Western blots are available as supplementary material.
Microbiota Analysis
Stool collection and DNA extraction
Fecal samples were homogenized and 0.2g aliquots were stored at − 80°C for further analysis. DNA was extracted from fecal samples as previously described 29. Briefly, following microbial lysis by mechanical and chemical methods, nucleic acids were precipitated in isopropanol for 10 min at room temperature, incubated for 15 min on ice and centrifuged at 20,000g for 30 min at 4°C. Pellets were resuspended in 450 µl of PBS and 50 µl of potassium acetate. After RNase treatment and DNA precipitation, nucleic acids were recovered via centrifugation at 20,000g for 30 min at 4°C. The pelleted DNA was resuspended in 80 µl of trypsin-EDTA buffer.
Sequencing
Microbiota analysis was performed by amplicon sequencing of the V3-V4 region of the 16S ribosomal RNA gene. This region was amplified using the following primers – 16S sense 5′-TACGGRAGGCAGCAG-3′ and anti-sense 5′-CTACCNGGGTATCTAAT-3′ – according to an optimized and standardized 16S amplicon library preparation protocol (Metabiote, GenoScreen, Lille, France). Briefly, PCR of the 16S DNA was performed with 5ng of genomic DNA according to the manufacturer’s protocol (Metabiote), with bar-coded primers (Metabiote MiSeq Primers) to a final concentration of 0.2 µmol/l, with an annealing temperature of 50°C for 30 cycles. Purification of the PCR products was performed with Agencourt AMPure XP-PCR purification system (Beckman Coulter, Brea, CA, USA), and quantified following the manufacturer’s instructions. The samples were multiplexed at equal concentrations. Sequencing was performed on an Illumina MiSeq platform (Illumina, San Diego, CA, USA) using a 250 bp paired-end sequencing protocol at GenoScreen. Raw paired-end reads were subjected to the following processes: (1) quality filtering using the PRINSEQ-lite PERL script 30, by truncating the bases from the 3′ end, that did not exhibit a quality < 30, based on the Phred algorithm and (2) searching for and removing both forward and reverse primer sequences using CutAdapt, with no mismatches allowed in the primer sequences. Only sequences where perfect matching forward and reverse primers were detected were included.
16S sequence analysis
Sequences were quality filtered using the dada2 software package (version 1.12.1) 31 in the R programming language (R version 3.6.1) to produce amplicon sequence variants (ASVs). Taxonomic classification was performed using the Silva reference database (version 132) 32. Bacterial ASVs that could not be assigned to Phylum-level taxonomy were excluded. Alpha diversity was estimated using the number of observed species and the Shannon diversity index. Raw sequence data are accessible in the Sequence Read Archive (accession number pending). Beta diversity analysis was performed on proportion-normalized data using the Bray-Curtis index. Assessment for significant differences between clusters was performed using PERMANOVA with the adonis function in the vegan package (version 2.5-6) in R with 99999 permutations.
Differential abundance was tested using linear discriminant analysis with effect size (Lefse) using default settings 33.
Ldl Isolation And Oxidation
LDL from normal human pooled sera was prepared by ultracentrifugation and dialyzed against PBS containing 100 µM EDTA. The LDL pool was then diluted to 2 g/l with PBS into a final volume of 3 ml. LDLs were mildly oxidized by UV-C for 2 h in the presence of 5 µM CuSO4 as previously reported 34. Oxidized LDL contained 4.2 to 7.4 nmoles of TBARS (thiobarbituric acid-reactive substances) /µg apoB. Relative electrophoretic mobility (REM) and 2,4,6-trinitrobenzenesulfonic acid (TNBS) reactive amino groups were 1.2–1.3 times and 85–92% of native LDL, respectively.
Statistical analysis. Graphs and statistical analyses were performed using Prism software (Graphpad). Values are expressed as mean ± s.e.m. Differences between values were examined using the non-parametric Mann-Whitney test. When 3 or more experimental groups were compared, Kruskal-Wallis test was used. Statistical significance was reached when P < 0.05.