The biological significance of C5a receptor [(C5aR)2/C5L2], a seven-transmembrane receptor binding C5a and C5adesArg, remains ill-defined. Specific ligation of C5aR2 inhibits C5a-induced ERK1/2 activation, strengthening the view that C5aR2 regulates C5aR1-mediated effector functions. Although C5aR2 and C5aR1 are often coexpressed, a detailed picture of C5aR2 expression in murine cells and tissues is still lacking. To close this gap, we generated a floxed tandem dye (td)Tomato–C5aR2 knock-in mouse that we used to track C5aR2 expression in tissue-residing and circulating immune cells. We found the strongest C5aR2 expression in the brain, bone marrow, and airways. All myeloid-derived cells expressed C5aR2, although with different intensities. C5aR2 expression in blood and tissue neutrophils was strong and homogeneous. Specific ligation of C5aR2 in neutrophils from tdTomato–C5aR2 mice blocked C5a-driven ERK1/2 phosphorylation, demonstrating functionality of C5aR2 in the reporter mice. In contrast to neutrophils, we found tissue-specific differences in C5aR2 expression in eosinophils, macrophages, and dendritic cell subsets. Naive and activated T cells stained negative for C5aR2, whereas B cells from different tissues homogeneously expressed C5aR2. Also, NK cell subsets in blood and spleen strongly expressed C5aR2. Activation of C5aR2 in NK cells suppressed IL-12/IL-18–induced IFN-γ production. Intratracheal IL-33 challenge resulted in decreased C5aR2 expression in pulmonary eosinophils and monocyte-derived dendritic cells. In summary, we provide a detailed map of murine C5aR2 immune cell expression in different tissues under steady-state conditions and upon pulmonary inflammation. The C5aR2 knock-in mouse will help to reliably track and conditionally delete C5aR2 expression in experimental models of inflammation.

Canonical and noncanonical cleavage of C5 results in the generation of C5a, which plays important roles in host defense, immune surveillance, and tissue homeostasis (1, 2). However, C5a also drives detrimental immune responses in autoimmune (3), inflammatory, and neurodegenerative (4) diseases. Strong and sustained production of C5a can lead to downregulation of immune responses in leukocytes, but it may have opposing effects in other cell types (5). These ambivalent biological functions of C5a have been explained by differential expression of C5a receptor (C5aR)1 and C5aR2. The expression and effector functions of C5aR1 are well characterized (68). It belongs to the large family of G protein–coupled receptors (GPCRs). Activation of C5aR1 by its cognate ligand C5a leads to the recruitment of Gαi subunits, resulting in the recruitment of β-arrestin, which is critical for receptor internalization, desensitization, recycling, and, in some cases, G protein–independent signaling (911).

The functional properties and expression patterns of C5aR2 are still enigmatic (5, 12). Like C5aR1, C5aR2 is a seven-transmembrane receptor that belongs to the GPCR family; however, as a result of alterations in the DRY and NPXXY motifs, as well as other properties, it does not couple to G proteins (13) but still binds to β-arrestin. Because C5aR2 lacks coupling to G proteins, it was initially considered a decoy receptor that inhibits the function of C5aR1 by capturing excessive C5a (13). In line with this view, Scola et al. (14) showed that cells that solely express C5aR2 bind C5a independently of C5aR1. This binding leads to ligand degradation, a mechanism by which C5aR2 might block the proinflammatory properties of C5a mediated by activation of C5aR1. Studies by Bamberg et al. (15) supported the view that C5aR2 interacts with β-arrestin, thereby regulating other signaling pathways. Binding of C5a to both receptors triggers their phosphorylation, thereby facilitating their association with β-arrestin. C5aR1/β-arrestin association leads to ERK1/2 phosphorylation, whereas the formation of C5aR2/β-arrestin complexes results in inhibition of ERK1/2 phosphorylation, suggesting that the ratio of C5aR1/C5aR2 expression determines C5a-mediated cellular responses. Also, C5aR2 acts as a negative regulator of C5aR1 in a mouse model of acute lung injury (16). However, the narrowed view of C5aR2 as a sole decoy receptor has been challenged by several findings demonstrating that binding of C5a to C5aR2 induces pro- and anti-inflammatory functions (1719). In neutrophils, C5aR2 deficiency leads to an altered cytokine profile when these cells are stimulated with C5a. In this setting, C5aR2 controlled the C5a-induced upregulation of IL-6, TNF-α, and CR3 (14, 17). Further, C5aR2-deficient mice showed improved survival in the cecal ligation puncture model of sepsis, which is in line with the observation that C5aR2 deficiency results in reduced inflammatory cell infiltration and HMGB-1 production from macrophages and neutrophils (20). Recently, Croker et al. (5) described the specific C5aR2 agonist P32. Stimulation of C5aR2 with this molecule modulated IL-6 release from human macrophages. Further, they observed that stimulation of C5aR2 inhibited the C5a-mediated effector functions of macrophages, as well as neutrophil mobilization without blocking it completely (5).

To better understand the various functions of C5aR2 as a decoy receptor, a regulator of C5aR1 functions, and as a second C5aR that activates cells independently of C5aR1, a detailed understanding of C5aR2 expression and regulation is necessary. In this study, we developed a floxed tandem dye Tomato (tdTomato)-C5aR2 knock-in mouse, which allows reliable tracking of C5aR2 expression in immune and tissue cells. The two loxP sites were introduced to create cell- or tissue-specific C5aR2 knockout mice by mating the floxed tdTomato-C5aR2 knock-in mouse with corresponding Cre deleter strains. Using this reporter mouse, we provide a detailed map of C5aR2 expression in innate and adaptive immune cells residing in the bone marrow (BM), blood, spleen, lung, lamina propria (LP) of the small intestine, visceral adipose tissue (VAT), and brain. Further, we assessed the expression of C5aR2 in the lung and the airways under inflammatory conditions upon IL-33 challenge. Our findings demonstrate differential expression of C5aR2 in myeloid cells and lack of C5aR2 expression in T cells. Surprisingly, we observed C5aR2 expression in B cells and subsets of NK cells. Importantly, C5aR2 activation on NK cells controlled IL-12/IL-18–mediated IFN-γ production. Further, we found IL-33–mediated regulation of C5aR2 expression in pulmonary eosinophils and monocyte-derived dendritic cells (moDCs).

Monoclonal allophycocyanin-Cy7–labeled Ab against CD11b (M1/70), PerCP-Cyanine (Cy)5.5–labeled Ab against CD8a (53-6.7), allophycocyanin or V450-labeled Ab against Ly6G (1A8), V450-labeled Ab against CD45R (RA3-6B2), FITC-labeled Ab against CD90.2 (30-H12), allophycocyanin-H7–labeled Ab against CD19 (1D3), and Brilliant Violet (BV)421-labeled Ab against Siglec-F (E50-2440) and CD43 (1G10) were purchased from BD Biosciences. PerCP-Cy5.5–labeled Ab against Ly6C (RB6-8C5); allophycocyanin-labeled Ab against CD115 (AFS98), CD11c (N418), NK1.1 (PK136), CD4 (GK1.5), and CD11b (M1/70); PE-Cy7–labeled Ab against CD25 (PC61.5) and CD4 (RM4-5); eFluor (eF)450-labeled Ab against CD19 (1D3), CD3e (145-2C11), and CD49b (DX5); Alexa Fluor (AF)488-labeled Ab against CD3 (17A2); allophycocyanin-eF780–labeled Ab against MHC class II (MHCII; M5/144.15.2); BV510-labeled Ab against CD11b (M1/70); AF711-labeled Ab against CD64 (X54-5/7.1); BV421-labeled Ab against F4/80 (BM8); and PerCP-Cy5.5–labeled Ab against CD103 (2E7) were purchased from BioLegend. PerCP-Cy5.5–labeled Ab against CD3 (OKT3), CD5 (53-73), CD27 (LG.7F9), NK1.1 (PK136), TCR-β (H57-597), and CD11b (MI/70); eF780-labeled Ab against CD11c (N418), B220 (RA3-6B2), and CD49b (DX5); eF450-labeled Ab against CD25 (PC61.5) and CD317 (ebio129c); PE-Cy5–labeled Ab against CD127 (A7R34); and PE-Cy7–labeled Ab against IgM (11/41) were purchased from eBioscience (Affymetrix). For surface and intracellular staining, unlabeled rat C5aR2-specific Ab (clone 468705; IgG2B; R&D Systems) was used, the binding of which was detected using a secondary AF647-labeled F(ab)2 anti-rat Ab fragment (Abcam). For intracellular staining of phosphorylated ERK1/2, the AF647-labeled Ab against ERK1/2 (pT202/pY204; BD) was used. Further, we used rabbit unlabeled polyclonal Ab against C5aR2 (catalog number HP8015; Hycult Biotech), the binding of which was detected using a secondary AF647-labeled F(ab)2 anti-rabbit Ab fragment (Abcam).

RBC lysis (RBCL) buffer was prepared using 155 mM NH4Cl, 10 mM KHCO3, and 0.1 mM EDTA (all from Sigma-Aldrich). BSA and a Lamina Propria Dissociation Kit were purchased from Miltenyi Biotec. Recombinant murine GM-CSF and Flt3 were from PeproTech. Liberase TL was from Roche Diagnostics, and DNase I for cell isolation was from Sigma-Aldrich. The KAPA Express Extract Kit was from Peqlab. FBS, RPMI 1640, and HBSS were from PAA. PBS, l-glutamine, penicillin, and streptomycin were from Life Technologies. DNase I for total RNA isolation was from Fermentas. The RevertAid First Strand cDNA Synthesis Kit was from Thermo Scientific. Primers for real-time RT-PCR were from Eurofins Genomics, and all other reagents for RT-PCR were from Bio-Rad. GelRed was purchased from BIOTREND. The Cytofix/Cytoperm kit was purchased from BD Biosciences; C5a was from Hycult Biotech; IL-33 was from eBioscience; DAPI was from Life Technologies; and mounting Medium Fluoromount-G was purchased from SouthernBiotech.

C57BL/6J wild-type (WT) mice were purchased from Janvier Labs. The tdTomato-C5ar2fl/fl and tdTomato-C5ar2fl/+ knock-in mice were generated as described below. All animals were used at 8–12 wk of age and were handled in accordance with the appropriate institutional and national guidelines. Animals were used for organ removal according to protocols approved by local authorities of the Animal Care and Use Committee (Ministerium für Energiewende, Landwirtschaft, Umwelt, Natur, und Digitalisierung, Kiel, Germany). Intratracheal (i.t.) injections of IL-33 were also approved by this committee (V242-81505/2016 [19-2/2017], V242-30278/2016 [50-4/16]). All experiments were performed by certified personnel.

The floxed tdTomato-C5aR2 knock-in mice were generated by gene targeting. A diagram of the targeting strategy is shown in Fig. 1. The targeting vector, the embryonic stem (ES) cells, and the mice were generated by Ozgene (Bentley, Australia). Our strategy was to insert tdTomato immediately following the splice acceptor of exon 2 in-frame with the coding sequence of C5ar2. The self-cleaving peptide derived from porcine teschovirus-1 (P2A) and the cDNA of C5ar2 were also present downstream of tdTomato to permit expression of the endogenous peptide from the knock-in locus. This region was floxed with loxP sites to permit a knockout of the locus by Cre recombinase.

The targeting construct was generated by the sequential cloning of four fragments. The final recipient plasmid encoded a neomycin selection cassette. Primers used to amplify the fragments also encoded the restriction enzyme sites required for the assembly of the fragments generated. All fragments were housed in the Surf2 vector backbone (Ozgene). The first fragment was a 4.4-kb fragment encoding the 3′ homology arm. This fragment was amplified from BACsRP23-388G21 and RP23-93F22 using primers 1617_461 (5′-CTAAGGGGTGAATGTTCAGTGAGAAA-3′) and 1617_561 (5′-TAAGCATTGGTAAGACGTCGCAAGAGGGCTTTGGATTTCCTAGGC-3′). The second fragment was a 1.2-kb fragment encoding the P2A and cDNA of C5ar2. This fragment was amplified from BACsRP23-388G21 and RP23-93F22 using primers 1617_44 (5′-TAAGCATTGGTAAGACGTCGACACGTGTCATAACTTCGTATAGCATACATTATACGAAGTTATCTACACCGGCATCTCAGACACCATTTCGTG-3′) and 1617_54 (5′-CTAAACGCGTCTGGAGCTACTAACTTCAGCCTGCTGAAGCAGGCTGGAGACGTGGAGGAGAACCCTGGACCTATGAAATATGAGGGGCTGGGAGACGGGTGTCTGGGGGCCAGAATGATGAACCACACCACCAG-3′); primer 1617_44 encoded a loxP site, and primer 1617_54 encoded the P2A sequence and the coding sequence of exon 1.

The third fragment was a 1.6-kb fragment encoding the coding sequence of tdTomato. This fragment was amplified in one of Ozgene’s proprietary vectors with primers 1617_45 (5′-TAAGCATTGGTAAGGCGCGCCCTTGTACAGCTCGTCCATGCCGTA-3′) and 1617_55 (5′-GCCGCAATGCTGGTACCCTGGAGTGCAGGCACCCTGCGCCACAGTGTTGGTGTCTCTAGCATGGAGCCGCTGGACCCGCCTTGCCCGGGGACAGCAAAGCTGGAGTTTTCCGTTGACTTTCCCTCCTGTCCCCTGCAGGTGTCTGATGGTGAGCAAGGGCGAGGAGGTCATCAAAGA-3′); primer 1617_55 also incorporated the splice acceptor of C5ar2 exon 2. The fourth fragment encoded the 3.6-kb 5′ homology arm. This fragment was amplified from BACsRP23-388G21 and RP23-93F22 using primers 1617_43 (5′-CTAAAGATCTCTGTGCCGGTTATTGCCAACTAAG-3′) and 1617_53 (5′-TAAGCATTGGTAAGGGAGTCAGAATTAGCATTCCAGA-3′).

The four fragments were assembled sequentially as follows. Fragment 1 was excised by digestion with AatII and ligated into fragment 2, which had been digested with the same enzyme. Combined fragments 1 and 2 were excised from the resulting construct by digestion with MluI and ligated into fragment 3, which had been digested with the same enzyme. Combined fragments 1, 2, and 3 were excised from the resulting construct by digestion with NotI and ligated into fragment 4, which had been digested with the same enzyme, resulting in the targeting vector 1617_pTV. This vector was linearized by digestion with SacII prior to electroporation into C57BL/6 Bruce4 ES cells.

For genotyping, we used biopsies from the ear. DNA extraction was performed using the KAPA Express Extract Kit (Peqlab), following the manufacturer’s instructions. To amplify the different DNA fragments, we used following primers: GK 357: 5′-CGTGGAGTTCCTAGAGGCTCA-3′; GK 353: 5′-AATACTCGCTGGTGGTGTGG-3′ (Eurofins Scientific). PCRs were run using the following conditions: 95°C for 3 min, followed by 35 cycles at 95°C for 15 s, 65°C for 15 s, and 72°C for 60 s, followed by 72°C for 120 s. The samples were transferred to a 1.0% sodium borate agarose gel, which was stained for amplification products with GelRed (BIOTREND).

Mice were killed by cervical dislocation under anesthesia, and the heart was perfused with 10 ml of cold PBS before organ removal. For BM cell isolation, femurs and tibias were removed, placed in PBS on ice, and subsequently flushed with PBS. RBCs were removed by incubating the cells in RBCL buffer for 3 min. The reaction was stopped by adding a large volume of PBS. BM-derived DCs (BMDCs) were generated by 9 d-differentiation with GM-CSF (20 ng/ml) as previously described (9, 21). BMDCs were defined as CD11c+CD11b+MCHIIhiCD115 cells. To obtain BM-derived plasmacytoid DCs (BMpDCs), BM cells were differentiated for 9 d in the presence of Flt3 ligand (200 ng/ml). BM-derived macrophages (BMMs) were obtained after 6 d of differentiation in the presence of M-CSF, which is secreted by L929 cells and is used in the form of L929-conditioned medium (22), or as CD11c+CD11b+CD115+MHClo BMMs in a 9 d-differentiation with GM-CSF (20 ng/ml) (21). For pulmonary cell preparation, lungs were harvested and digested using 0.25 mg/ml Liberase TL and 0.5 mg/ml DNase I in pure RPMI medium for 45 min at 37°C, and single-cell suspensions were prepared as described (7). For VAT cell preparations, perigonadal fat tissue was harvested, taking care not to remove the gonads, and minced into small pieces. Tissue was digested using 0.25 mg/ml Liberase TL and 0.5 mg/ml DNase I in pure RPMI 1640 medium for 45 min at 37°C. The cell suspension was filtered through a cell strainer (40 μm Nylon; BD) and washed with 10 ml of RPMI 1640 medium complemented with 10% FCS containing 0.5 mg/ml DNase I. Cells were centrifuged at 350 × g for 10 min at 4°C, resuspended in PBS, transferred into a fresh 15-ml tube, and washed again. Bronchoalveolar lavage fluid (BAL) samples were obtained as described (9). Lavage of the peritoneal cavity was performed with 10 ml of ice-cold PBS. Collected cells were washed once with PBS. RBCs were removed by incubating the cells in RBCL buffer for 3 min and then washed with PBS (7). Blood was collected by cardiac puncture and immediately transferred into tubes containing 10 mM EDTA to prevent coagulation. Diluted blood samples were incubated several times in RBCL buffer to remove RBCs and then washed with PBS (7). Isolation of cells from the spleen and mesenteric lymph nodes (mLNs) was performed by mechanical disruption using a cell strainer (40 μm Nylon; BD) and the plunger of a 5-ml syringe (BD). The cell strainer was flushed three times with 5 ml of PBS. Cells were then incubated in RBCL buffer for 3 min and finally washed with PBS. LP cell suspension was obtained using a Lamina Propria Dissociation Kit (Miltenyi Biotec), following the manufacturer’s recommendations. Briefly, the small intestine was removed from the mice and quickly cleared of feces, residual fat, and Peyer’s patches. Then, they were cut longitudinally and incubated twice in HBSS, once with the addition of EDTA (5 mM) and once without. The tissue was digested using the manufacturer’s enzyme mixture in HBSS with Ca2+ and Mg2+ for 30 min at 37°C and homogenized using a gentleMACS Dissociator. After the last washing step, cell number was determined using a Neubauer chamber, and cells were resuspended in PBS containing 1% BSA. Viability of the collected cells was determined by trypan blue exclusion or a LIVE/DEAD Cell Viability Assay (BD) using flow cytometry (LSR II; BD). Brain tissue was collected, sliced into pieces, and digested with Liberase TL (0.1 mg/ml) and DNase I (0.1 mg/ml) for 30 min at 37°C. Brain homogenate was passed through a cell strainer (40 μm Nylon; BD), and the cell suspension was washed and fractionated on 30–75% discontinuous Percoll gradient (GE Healthcare) for 30 min at 1350 × g without brake. The interphase-containing cells were collected and washed with Dulbecco’s PBS, and the cellular composition was determined by flow cytometry.

Phenotypic characterization of cells was performed using a BD Aria III cell sorter (BD) and a MoFlo Legacy cell sorter (Beckman Coulter). The discriminative markers and gating strategies used to identify different cell types in the different organs and compartments have been described previously (23) and are described briefly in the figure legends and text. Flow cytometric data were analyzed using FlowJo 10 (TreeStar, Ashland, OR).

Cells were collected from the BM, as described above, and stained with DAPI or allophycocyanin-labeled Ab recognizing Ly6G (clone 1A8). The cells were incubated at 4°C for 15 min, transferred to a slide, and mounted with Fluoromount-G. Images were obtained using an Olympus FV 1000 confocal microscope with a 40× oil immersion 0.95 numerical aperture objective. FluoView 2.1c software (Olympus) was used as acquisition software. Image processing was conducted using Imaris software (Bitplane).

BM cells from tdTomato-C5ar2fl/fl reporter and C5ar2−/− mice were left unstimulated or were stimulated for 8 min with C5a (20 nM), with or without treatment with the C5aR2 agonist P32 (100 μM) 30 min prior to C5a addition. The C5aR2 agonist P32 was described recently (5). BM cells were then fixed with 1.8% formaldehyde for 15 min before 500 μl of ice-cold methanol was used to permeabilize the cell membranes. Next, methanol incubation was stopped after 20 min by washing with 500 μl of cold PBS. After resuspending the cell pellet in 100 μl of PBS, cells were stained for 15 min with an AF647-labeled Ab against ERK1/2 (pT202/pY204). Unbound Ab was removed by PBS washing. After pellet resuspension in 300 μl of PBS, samples were analyzed for ERK1/2 phosphorylation by flow cytometry.

The spleen was harvested and dissociated by mechanical disaggregation. After RBC lysis (as described above), cells were counted, and MACS-purified T cells were used for in vitro activation as described (7). To stimulate CD4+ T cells, a 96-well cell culture plate (Greiner Bio-One) was coated with unconjugated anti-CD3 (17A2) and anti-CD28 (37.51; both from eBioscience) in 150 μl of PBS at a final concentration of 2 and 10 μg/ml, respectively. The plate was kept at 37°C, 5% CO2 for ≥2 h before discarding the coating solution and adding the cell suspension. T cells were cultured for 5 d in complete RPMI 1640 culture medium supplemented with 10% FBS, 2 mM l-glutamine, 100 U/ml penicillin, and 100 μg/ml streptomycin. Cells were subsequently analyzed by flow cytometry for tdTomato-C5aR2 expression.

IL-33 (500 ng) was administered i.t. to anesthetized mice on four consecutive days (24). Twenty-four hours after the final IL-33 challenge, BAL and lung samples were collected as described above and used to determine immune cell composition by flow cytometry.

NK cells were purified from the spleen using a cell sorter. NK cells were identified as NKp46+CD3 cells. After sorting, 1 × 105 NK cells were cultured in RPMI 1640 (with 10% FCS, 1% penicillin/streptomycin, and 1% l-glutamine) and stimulated with 10 ng/ml IL-12 and 10 ng/ml IL-18 for 24 h. In some experiments, NK cells were also stimulated with 100 μM of the C5aR2-specific agonist P32. As a readout, IFN-γ production was measured by ELISA.

RNA was isolated using TRIzol Reagent, according to the manufacturer’s instructions. After DNase I treatment of the RNA, a reverse-transcription reaction was performed using a RevertAid First Strand cDNA Synthesis Kit. Real-time PCR was done using iQ SYBR Green Supermix on a CFX96 Real-Time System (Bio-Rad) with the following primers: β-actin 5′-GCACCACACCTTCTACAATGAG-3′ and 5′-AAATAGCACAGCCTGGATAGCAAC-3′; C5ar2 5′-CTGGGCCTCTTGCTGACTGTGC-3′ and 5′-GCCCCAGGAAGCCAAAGAGGA-3′. The temperature profile of the RT-PCR was 95°C for 3 min, followed by 40 cycles at 95°C for 5 s, 58°C (β-actin) or 54°C (C5ar2) for 5 s, and 72°C for 30 s, followed by 30 cycles for 5 s with a 1°C temperature increase starting at 65°C to confirm the expected PCR products by melting curve analysis. Real-time RT-PCR data were analyzed using CFX Manager Software 3.1 (Bio-Rad). Amplification products were also separated using a 2% sodium borate agarose gel and were detected by GelRed staining (BIOTREND).

Statistical analysis was performed using GraphPad Prism version 6 (GraphPad). Normal distribution of data was tested using the Kolmogorov–Smirnov and D’Agostino–Pearson tests. Statistical differences between groups were assessed using the nonpaired Student t test (Figs. 7D, 7E, 8) or the paired Student test (Fig. 7F). A p value <0.05 was considered significant (*p < 0.05, **p < 0.01, ***p < 0.001).

We generated a floxed tdTomato-C5aR2 knock-in mouse that allows cell- and tissue-specific tracking and deletion of C5aR2. For this purpose, we cloned a tdTomato cassette directly upstream of the 5′ end of exon 2 of the C5ar2 gene. To allow conditional deletion of the C5ar2 gene, we placed two loxP sites upstream of the tdTomato coding sequence and downstream of exon 2 of the C5ar2 gene (Fig. 1A). We obtained several ES cells that incorporated tdTomato and the two loxP sites into the C5ar2 gene locus. One of such cells was successfully used to produce heterozygous (tdTomato-C5ar2fl/+) and homozygous (tdTomato-C5ar2fl/fl) floxed tdTomato-C5aR2 knock-in mice (Fig. 1B).

FIGURE 1.

Generation and initial characterization of the floxed tdTomato-C5aR2 knock-in mouse. (A) Schematic representation of the gene-targeting strategy. The WT exon 2 of C5ar2 was replaced by a cassette encoding a fusion protein composed of tdTomato-C5aR2 flanked with loxP sites by homologous recombination. The self-cleaving peptide P2A was inserted downstream of tdTomato. (B) PCR-based phenotyping of WT, heterozygous tdTomato-C5ar2fl/+, and homozygous tdTomato-C5ar2fl/fl mice. The primer combination GK357:GK353 amplifies a 225-bp DNA fragment in C57BL/6 WT mice and an 1858-bp DNA fragment in tdTomato-C5ar2fl/fl mice. Both fragments are amplified in tdTomato-C5ar2fl/+ mice. The arrows point to the amplified PCR fragments. (C) Assessment of the tdTomato-C5aR2 signal by flow cytometry using total cell suspensions from blood, spleen, brain, VAT, lung, LP of the small intestine, BAL, and BM from WT and tdTomato-C5ar2fl/fl mice.

FIGURE 1.

Generation and initial characterization of the floxed tdTomato-C5aR2 knock-in mouse. (A) Schematic representation of the gene-targeting strategy. The WT exon 2 of C5ar2 was replaced by a cassette encoding a fusion protein composed of tdTomato-C5aR2 flanked with loxP sites by homologous recombination. The self-cleaving peptide P2A was inserted downstream of tdTomato. (B) PCR-based phenotyping of WT, heterozygous tdTomato-C5ar2fl/+, and homozygous tdTomato-C5ar2fl/fl mice. The primer combination GK357:GK353 amplifies a 225-bp DNA fragment in C57BL/6 WT mice and an 1858-bp DNA fragment in tdTomato-C5ar2fl/fl mice. Both fragments are amplified in tdTomato-C5ar2fl/+ mice. The arrows point to the amplified PCR fragments. (C) Assessment of the tdTomato-C5aR2 signal by flow cytometry using total cell suspensions from blood, spleen, brain, VAT, lung, LP of the small intestine, BAL, and BM from WT and tdTomato-C5ar2fl/fl mice.

Close modal

The initial analysis of the tdTomato signal in different organs using flow cytometry showed that tdTomato+ cells were present in all organs analyzed, with the highest frequency in brain, BM, and BAL. The frequency of tdTomato+ cells was lower in spleen, lung, blood, VAT, and the LP of the small intestine (Fig. 1C). Of note, in lung and the LP of the small intestine, the tdTomato signal was not associated with epithelial cells (data not shown).

In their first description of C5aR2, Ohno et al. (25) stated that C5ar2 mRNA is strongly expressed in granulocytes under steady-state conditions. Since then, several groups determined the potential functions of C5aR2 as a regulator of C5aR1-mediated effects (15) or independent of C5aR1 functions (19, 20). Thus, we determined tdTomato expression in neutrophils from BM, blood, and the peritoneal cavity. In line with previous reports, we found strong expression of tdTomato-C5aR2 in all tested neutrophil populations. When we compared tdTomato-C5aR2 expression in neutrophils from tdTomato-C5ar2fl/+ and tdTomato-C5ar2fl/fl mice, we found that the mean fluorescence intensity (MFI) was higher in tdTomato-C5ar2fl/fl mice (Fig. 2A). Next, we tested whether the tdTomato signal matches C5ar2 mRNA expression. For this purpose, we analyzed sorted neutrophils from blood and BM from WT mice and assessed C5ar2 expression by RT-PCR. In both neutrophil subsets, we found a strong signal for C5ar2 mRNA (Fig. 2B). Further, we assessed C5aR2 protein expression using Ab-based surface staining. Using two commercially available Abs (clone 468705 [R&D Systems] and a polyclonal Ab [Hycult Biotech]), we found a strong and homogeneous signal in BM neutrophils from WT (data not shown) and tdTomato-C5ar2fl/fl mice (Fig. 2C), suggesting C5aR2 expression in such cells. Surprisingly, we also observed a strong signal when we stained BM neutrophils from C5ar2−/− mice, demonstrating that these Abs recognize structures independently of C5aR2 (Fig. 2C). Finally, we stained BM neutrophils from WT and tdTomato-C5ar2fl/fl mice with Ly6G and DAPI and transferred them onto glass slides to analyze the tdTomato signal by fluorescence microscopy. We observed a strong tdTomato signal in Ly6G+ neutrophils from tdTomato-C5ar2fl/fl mice but not from WT mice, and the signal was primarily located in the cytoplasm of these cells (Fig. 2D).

FIGURE 2.

Expression of tdTomato-C5aR2 in naive Ly6G+ neutrophils from BM, blood, and the peritoneal cavity and assessment of C5aR2 function in neutrophils from tdTomato-C5ar2fl/fl mice. Flow cytometric assessment of tdTomato-C5aR2 expression in different neutrophil populations. Data are representative of five independent experiments. (A) tdTomato-C5aR2 expression in neutrophils from BM (top panels), blood (middle panels), and peritoneal lavage fluid (bottom panels), which were identified as Ly6G+ cells. Gray histograms show the WT signal; the black lines represent the signal obtained in tdTomato-C5ar2fl/fl mice (middle panels) and tdTomato-C5ar2fl/+ mice (right panels). (B) C5ar2 mRNA expression in Ly6G+ sorted neutrophils from blood and BM. Data are representative of three independent experiments. (C) C5aR2 surface staining in BM neutrophils using two C5aR2-specific Abs: polyclonal Ab (Hycult Biotech; left panel) and rat mAb (clone 468705; R&D Systems; right panel). The gray histogram shows the signal of the isotype-control Ab, the black line corresponds to the signal obtained with neutrophils from tdTomato-C5ar2fl/fl mice, and the dashed line corresponds to the signal obtained with neutrophils from C5ar2−/− mice. (D) tdTomato-C5aR2 expression in sorted Ly6G+ BM neutrophils from tdTomato-C5ar2fl/fl and WT mice by confocal microcopy (original magnification ×400). Cells were stained for Ly6G (turquoise) and DAPI (blue). The tdTomato signal is shown in red. The data are representative of three independent experiments. (E) Phosphorylation of ERK1/2 in BM neutrophils from tdTomato-C5ar2fl/fl mice (left panel) or C5ar2−/− mice (right panel) in the absence or presence of C5a, with or without C5aR2-specific ligand P32. Data are representative of three independent experiments.

FIGURE 2.

Expression of tdTomato-C5aR2 in naive Ly6G+ neutrophils from BM, blood, and the peritoneal cavity and assessment of C5aR2 function in neutrophils from tdTomato-C5ar2fl/fl mice. Flow cytometric assessment of tdTomato-C5aR2 expression in different neutrophil populations. Data are representative of five independent experiments. (A) tdTomato-C5aR2 expression in neutrophils from BM (top panels), blood (middle panels), and peritoneal lavage fluid (bottom panels), which were identified as Ly6G+ cells. Gray histograms show the WT signal; the black lines represent the signal obtained in tdTomato-C5ar2fl/fl mice (middle panels) and tdTomato-C5ar2fl/+ mice (right panels). (B) C5ar2 mRNA expression in Ly6G+ sorted neutrophils from blood and BM. Data are representative of three independent experiments. (C) C5aR2 surface staining in BM neutrophils using two C5aR2-specific Abs: polyclonal Ab (Hycult Biotech; left panel) and rat mAb (clone 468705; R&D Systems; right panel). The gray histogram shows the signal of the isotype-control Ab, the black line corresponds to the signal obtained with neutrophils from tdTomato-C5ar2fl/fl mice, and the dashed line corresponds to the signal obtained with neutrophils from C5ar2−/− mice. (D) tdTomato-C5aR2 expression in sorted Ly6G+ BM neutrophils from tdTomato-C5ar2fl/fl and WT mice by confocal microcopy (original magnification ×400). Cells were stained for Ly6G (turquoise) and DAPI (blue). The tdTomato signal is shown in red. The data are representative of three independent experiments. (E) Phosphorylation of ERK1/2 in BM neutrophils from tdTomato-C5ar2fl/fl mice (left panel) or C5ar2−/− mice (right panel) in the absence or presence of C5a, with or without C5aR2-specific ligand P32. Data are representative of three independent experiments.

Close modal

To delineate whether the tdTomato construct has any impact on the function of C5aR2, we determined C5a-induced ERK1/2 phosphorylation in BM neutrophils in the presence of the C5aR2-specific agonist P32. Bamberg et al. (15) showed that C5aR2 acts as a negative regulator of ERK1/2 signal transduction. These findings could be corroborated using the C5aR2-selective ligand P32 (5). Stimulation of C5aR2 with P32 in tdTomato-C5ar2fl/fl mice abrogated ERK1/2 phosphorylation after C5a stimulation. This effect was C5aR2 specific, because C5a-mediated ERK1/2 phosphorylation in C5aR2-deficient neutrophils was not reduced by the presence of the C5aR2 agonist (Fig. 2E). These findings demonstrate that C5aR2 in tdTomato-C5ar2fl/fl mice is fully functional.

Previous studies reported important functional roles for C5aR2 in macrophages. For example, C5a-induced G-CSF production is markedly reduced in LPS-activated macrophages from C5ar2−/− mice compared with WT mice (26). Also, C5a-induced HMGB-1 production in macrophages depends on the presence of C5aR2 (20). We determined the expression of C5aR2 in macrophages from M-CSF– and GM-CSF–stimulated BM cell cultures, peritoneal cavity, LP of the small intestine, airways, lung tissue, VAT, and brain. All tested macrophage populations expressed tdTomato-C5aR2, although the expression levels were different (Fig. 3A, 3B, 3D–H). Peritoneal macrophages strongly and homogeneously expressed tdTomato-C5aR2 (Fig. 3D). In contrast, tdTomato-C5aR2 expression was much lower in M-CSF– or GM-CSF–derived BMMs: ∼40% of M-CSF BMMs (Fig. 3A) and 20% of GM-CSF BMMs (Fig. 3B) stained positive for tdTomato-C5aR2. Macrophages from the LP of the small intestine (Fig. 3E), lung and airways (Fig. 3F), and VAT (Fig. 3G), as well as macrophages or microglia cells from the brain (Fig. 3H), showed a heterogeneous expression pattern of tdTomato-C5aR2, with a tdTomato+ and a tdTomato population. Around 80–85% of LP (Fig. 3E) and airway (Fig. 3F) macrophages, as well as microglia cells (Fig. 3H), were tdTomato-C5aR2+. Approximately 60–65% of the brain macrophages (Fig. 3H) and 55–60% of pulmonary macrophages (Fig. 3F) were tdTomato-C5aR2+, whereas only 20–25% of the VAT macrophages stained positive for tdTomato-C5aR2 (Fig. 3G). These findings demonstrate tissue-specific differences in macrophage tdTomato-C5aR2 expression.

FIGURE 3.

Differential expression of tdTomato-C5aR2 in tissue macrophages. Flow cytometric assessment of tdTomato-C5aR2 expression in different macrophage populations. Gray histograms show the WT signal; black lines correspond to the signals in homozygous tdTomato-C5ar2fl/fl mice. (A) M-CSF–derived BM macrophages (M-CSF BMMs) were identified as F4/80+CD11b+ cells. (B) The GM-CSF–derived BM monocyte/macrophage population (GM-CSF BMMs) was identified among the CD11c+CD11b+ cells as the CD115+MHCIIlo population. (C) C5aR2 surface staining (left panel) and intracellular staining (right panel) in GM-CSF BMMs using a polyclonal C5aR2-specific Ab (Hycult Biotech). The gray histogram shows the signal of the isotype-control Ab, the black line corresponds to the signal obtained with cells from tdTomato-C5ar2fl/fl mice, and the dashed line shows the signal obtained with cells from C5ar2−/− mice. (D) In peritoneal lavage fluid, macrophages were characterized as F4/80+CD11b+ cells. C5aR2 surface staining in peritoneal macrophages (right panel) using polyclonal C5aR2-specific Ab (Hycult Biotech). The gray histogram shows the signal of the isotype-control Ab, the black line corresponds to the signal obtained with cells from tdTomato-C5ar2fl/fl mice, and the dashed line shows the signal obtained with cells from C5ar2−/−mice. (E) Macrophages from the LP of the small intestine were identified as lineageCD11c+CD11b+MHCII+F4/80+ cells. (F) Pulmonary macrophages were identified as SiglecF+CD11c+ cells associated with lung tissue (lungs; left panels) or from the airways (BAL; right panels). (G) In VAT, macrophages were identified as Siglec-FF4/80+CD11b+ cells. (H) In brain, microglial cells were identified as CD45Rlo cells, and macrophages were identified as CD45hiF4/80+CD11b+ cells. Data are representative of five independent experiments.

FIGURE 3.

Differential expression of tdTomato-C5aR2 in tissue macrophages. Flow cytometric assessment of tdTomato-C5aR2 expression in different macrophage populations. Gray histograms show the WT signal; black lines correspond to the signals in homozygous tdTomato-C5ar2fl/fl mice. (A) M-CSF–derived BM macrophages (M-CSF BMMs) were identified as F4/80+CD11b+ cells. (B) The GM-CSF–derived BM monocyte/macrophage population (GM-CSF BMMs) was identified among the CD11c+CD11b+ cells as the CD115+MHCIIlo population. (C) C5aR2 surface staining (left panel) and intracellular staining (right panel) in GM-CSF BMMs using a polyclonal C5aR2-specific Ab (Hycult Biotech). The gray histogram shows the signal of the isotype-control Ab, the black line corresponds to the signal obtained with cells from tdTomato-C5ar2fl/fl mice, and the dashed line shows the signal obtained with cells from C5ar2−/− mice. (D) In peritoneal lavage fluid, macrophages were characterized as F4/80+CD11b+ cells. C5aR2 surface staining in peritoneal macrophages (right panel) using polyclonal C5aR2-specific Ab (Hycult Biotech). The gray histogram shows the signal of the isotype-control Ab, the black line corresponds to the signal obtained with cells from tdTomato-C5ar2fl/fl mice, and the dashed line shows the signal obtained with cells from C5ar2−/−mice. (E) Macrophages from the LP of the small intestine were identified as lineageCD11c+CD11b+MHCII+F4/80+ cells. (F) Pulmonary macrophages were identified as SiglecF+CD11c+ cells associated with lung tissue (lungs; left panels) or from the airways (BAL; right panels). (G) In VAT, macrophages were identified as Siglec-FF4/80+CD11b+ cells. (H) In brain, microglial cells were identified as CD45Rlo cells, and macrophages were identified as CD45hiF4/80+CD11b+ cells. Data are representative of five independent experiments.

Close modal

We also determined surface and intracellular C5aR2 expression in GM-CSF BMMs and surface expression in peritoneal macrophages using the two anti-C5aR2 Abs (as described above). In GM-CSF BMMs from tdTomato-C5ar2fl/fl mice, we found strong C5aR2 surface and intracellular staining, which was like the signal obtained using cells from C5ar2−/− mice (Fig. 3C, data not shown). In line with this observation, peritoneal macrophages from tdTomato-C5ar2fl/fl and C5ar2−/− mice stained positive using the Abs against C5aR2. These data confirm our findings with neutrophils (Fig. 2C) and provide additional evidence that the two commercially available Abs used in this study recognize C5aR2, as well as other structures on neutrophils and macrophages and, thus, are not specific for C5aR2.

Little is known about the biological role or function of C5aR2 in eosinophils, which play important roles in allergic diseases such as allergic asthma. Previously, we found that C5aR2 drives eosinophilic airway inflammation in different models of experimental allergic asthma (27). In this study, we assessed the expression of tdTomato-C5aR2 in eosinophils from blood, lung, LP of the small intestine, BM, and VAT. BM and VAT are considered reservoirs of eosinophils (28, 29). In all tested eosinophil populations, we found a clear tdTomato signal (Fig. 4). In eosinophils from BM (Fig. 4A), blood (Fig. 4B), lung (Fig. 4C), and LP of the small intestine (Fig. 4D), we observed two distinct populations. In BM, lung, and in the LP of the small intestine (65–80%) as well as in the blood (90–95%), most eosinophils expressed tdTomato-C5aR2; however, a fraction of the eosinophils were clearly tdTomato-C5aR2 (Fig. 4A–D). In VAT (Fig. 4D), dtTomato-C5aR2 expression was homogeneous but weaker than in all other compartments.

FIGURE 4.

tdTomato-C5aR2 is differentially expressed in eosinophils from different organs. Flow cytometric assessment of tdTomato-C5aR2 expression in different eosinophil populations. Gray histograms show the WT signal; black lines correspond to the signal in homozygous tdTomato-C5ar2fl/fl mice. Eosinophils were identified as Siglec-F+ cells in BM (A) and blood (B) and as Siglec-F+CD11c cells in the lung (C), SiglecF+F4/80lo cells in the LP of the small intestine (D), and Siglec-F+F4/80int cells in VAT (E). Data are representative of five independent experiments.

FIGURE 4.

tdTomato-C5aR2 is differentially expressed in eosinophils from different organs. Flow cytometric assessment of tdTomato-C5aR2 expression in different eosinophil populations. Gray histograms show the WT signal; black lines correspond to the signal in homozygous tdTomato-C5ar2fl/fl mice. Eosinophils were identified as Siglec-F+ cells in BM (A) and blood (B) and as Siglec-F+CD11c cells in the lung (C), SiglecF+F4/80lo cells in the LP of the small intestine (D), and Siglec-F+F4/80int cells in VAT (E). Data are representative of five independent experiments.

Close modal

Several DC subsets in lymphoid and nonlymphoid compartments can be defined based on genetic and phenotypic markers (30). These include conventional DCs (cDCs), inflammatory moDCs, and pDCs. In mice, cDCs can be further classified into CD11b+-like and CD8+-like subsets. Following this differentiation scheme, CD11b+ cDCs are composed of splenic CD11b+CD8 DCs and pulmonary CD11b+ cDCs. The CD8-like cDC subset comprises splenic CD8+CD11b and pulmonary CD103+CD11b cDCs (31). At steady-state, only a very small fraction of CD11b+ DCs in the lung are moDCs, which increase under inflammatory conditions (32). Similarly, CD11b+ and CD103+ cDC subpopulations have been described in the LP of the small intestine (33). In contrast, DCs in the VAT are poorly defined. We identified a F4/80Siglec-FMHCII+CD11c+CD11b+ population in the VAT that we consider CD11b+ cDCs. However, we did not find any CD8-like cDC equivalent (data not shown). Finally, we observed a mixed BMDC/BMM population, as recently described (21), comprising 20–30% CD11c+CD11b+CD115MHCII+ BMDCs and 70–80% CD11c+CD11b+CD115+ BMMs when BM cells were differentiated in GM-CSF–supplemented medium.

The expression of tdTomato-C5aR2 within the DC compartment was heterogeneous. In the lung, a minor subpopulation of CD11b+ cDCs (15–20%) and a large subset of moDCs (60–65%) stained positive for tdTomato-C5aR2, whereas CD103+ cDCs did not express tdTomato-C5aR2 (Fig. 5A). Similarly, at best, a very minor fraction of GM-CSF–derived BMDCs expressed tdTomato-C5aR2 (Fig. 5B). Also, both splenic DC subsets stained negative for the tdTomato signal (Fig. 5C). In the LP of the small intestine, CD11b+ and CD103+ cDCs expressed tdTomato-C5aR2, although the expression levels were low (Fig. 5D). The VAT DC subset did not express tdTomato (Fig. 5E). Similarly, pDCs from spleen, the LP of the small intestine, lung, and VAT, as well as BMpDCs, stained negative for tdTomato (Fig. 5F).

FIGURE 5.

Subsets of mucosal surface–associated DCs express tdTomato-C5aR2. Flow cytometric assessment of tdTomato-C5aR2 expression in different DC populations. Gray histograms show the WT signal; black lines correspond to the signal in homozygous tdTomato-C5ar2fl/fl mice. (A) After excluding Siglec-F+ cells (eosinophils and alveolar macrophages) and lineage+ cells (CD19, CD49b, CD3e, Ly6G; data not shown), DCs were identified as CD11c+MHCII+ cells. In the DC population, CD103+ and CD11b+ cDCs could be separated. The latter population was further subdivided into CD11b+CD64 cDCs and CD64+ moDCs. (B) tdTomato-C5aR2 expression in GM-CSF–derived BMDCs (GM-CSF BMDCs), which were identified among the CD11c+CD11b+ DCs (data not shown) as CD115MHCII+ inflammatory DCs. (C) tdTomato-C5aR2 expression in CD11b+CD8 and CD11bCD8+ DCs from spleen. (D) LP DCs were identified as CD11c+CD11bMHCII+CD103+ cDCs and CD11c+CD11b+CD103loF4/80 DCs. (E) VAT DCs were identified as F4/80Siglec-FCD11c+MHCII+ cells. (F) tdTomato-C5aR2 expression in different pDC populations identified as CD11bCD11c+mPDCA+ cells in the spleen. In LP, lung, and VAT, the cells were further identified by positive staining for B220 and Siglec-H. In BM-derived cultures in the presence of Flt3 ligand, BMpDCs were identified as CD11bB220+mPDCA+ cells. Data are representative of five independent experiments

FIGURE 5.

Subsets of mucosal surface–associated DCs express tdTomato-C5aR2. Flow cytometric assessment of tdTomato-C5aR2 expression in different DC populations. Gray histograms show the WT signal; black lines correspond to the signal in homozygous tdTomato-C5ar2fl/fl mice. (A) After excluding Siglec-F+ cells (eosinophils and alveolar macrophages) and lineage+ cells (CD19, CD49b, CD3e, Ly6G; data not shown), DCs were identified as CD11c+MHCII+ cells. In the DC population, CD103+ and CD11b+ cDCs could be separated. The latter population was further subdivided into CD11b+CD64 cDCs and CD64+ moDCs. (B) tdTomato-C5aR2 expression in GM-CSF–derived BMDCs (GM-CSF BMDCs), which were identified among the CD11c+CD11b+ DCs (data not shown) as CD115MHCII+ inflammatory DCs. (C) tdTomato-C5aR2 expression in CD11b+CD8 and CD11bCD8+ DCs from spleen. (D) LP DCs were identified as CD11c+CD11bMHCII+CD103+ cDCs and CD11c+CD11b+CD103loF4/80 DCs. (E) VAT DCs were identified as F4/80Siglec-FCD11c+MHCII+ cells. (F) tdTomato-C5aR2 expression in different pDC populations identified as CD11bCD11c+mPDCA+ cells in the spleen. In LP, lung, and VAT, the cells were further identified by positive staining for B220 and Siglec-H. In BM-derived cultures in the presence of Flt3 ligand, BMpDCs were identified as CD11bB220+mPDCA+ cells. Data are representative of five independent experiments

Close modal

Currently available data suggest that C5aR2 is expressed in human and murine T lymphocytes. C5aR2 expression was shown in human naive and central memory T cells (34). In mice, it was recently shown that C5aR2 might be involved in T cell functions (14, 34); however, only one study showed C5ar2 expression in murine T cells, at least at the mRNA level (6). To gain insight into the expression of C5aR2 in CD4+ and CD8+ T cells, we determined tdTomato-C5aR2 expression in naive CD4+ and CD8+ T cells from spleen, blood, and mLNs, as well as CD3+ T cells from the lung. Further, we assessed tdTomato-C5aR2 expression in splenic CD4+ T cells in response to stimulation with CD3/CD28. We found no tdTomato-C5aR2 expression in naive CD4+ or CD8+ T cells from different tissues (Fig. 6A–D). Also, CD3/CD28 stimulation did not result in the induction of tdTomato-C5aR2 (Fig. 6E). This observation was confirmed at the mRNA level using naive CD4+ T cells purified from spleen, in which we found no amplification of C5ar2 transcripts (Fig. 6F).

FIGURE 6.

B lymphocytes, but not T lymphocytes, express tdTomato-C5aR2. Flow cytometric assessment of tdTomato-C5aR2 expression in splenic (A) and blood-derived (B) CD19+ B cells (upper panels) and CD4+ or CD8+ T lymphocytes (lower panels). (C) tdTomato-C5aR2 expression in CD19+ B cells (upper panels) and CD4+ or CD8+ T lymphocytes (lower panels) from mLNs. (D) tdTomato-C5aR2 expression in pulmonary CD3+ T lymphocytes (top panels), CD3CD49b+ NK cells (middle panel), and CD19+ B cells (bottom panels). (E) tdTomato-C5aR2 expression in purified naive splenic CD4+ T lymphocytes stimulated in vitro for 5 d in the presence of anti-CD3– and anti-CD28–specific Abs. In (A)–(E), gray histograms show the WT signal; black lines correspond to the signal in tdTomato-C5ar2fl/fl mice. (F) C5ar2 mRNA expression in sorted CD3+ T cells and CD19+ B cells from spleen. Data are representative of five independent experiments.

FIGURE 6.

B lymphocytes, but not T lymphocytes, express tdTomato-C5aR2. Flow cytometric assessment of tdTomato-C5aR2 expression in splenic (A) and blood-derived (B) CD19+ B cells (upper panels) and CD4+ or CD8+ T lymphocytes (lower panels). (C) tdTomato-C5aR2 expression in CD19+ B cells (upper panels) and CD4+ or CD8+ T lymphocytes (lower panels) from mLNs. (D) tdTomato-C5aR2 expression in pulmonary CD3+ T lymphocytes (top panels), CD3CD49b+ NK cells (middle panel), and CD19+ B cells (bottom panels). (E) tdTomato-C5aR2 expression in purified naive splenic CD4+ T lymphocytes stimulated in vitro for 5 d in the presence of anti-CD3– and anti-CD28–specific Abs. In (A)–(E), gray histograms show the WT signal; black lines correspond to the signal in tdTomato-C5ar2fl/fl mice. (F) C5ar2 mRNA expression in sorted CD3+ T cells and CD19+ B cells from spleen. Data are representative of five independent experiments.

Close modal

For human B cells, C5ar2 mRNA expression has been shown on plasma cells and memory B cells (15). Detailed information about C5aR2 expression in mouse B cells is lacking. When we assessed tdTomato-C5aR2 expression in CD19+ B cells from spleen, blood, mLN, and lung, we found a clear and homogeneous tdTomato signal in B cells from all tissues (Fig. 6A–C) with the exception of the lung, in which the signal was weak (Fig. 6D). C5ar2 expression was confirmed at the mRNA level in splenic B cells, in which we consistently amplified a faint band (Fig. 6F).

Previously, human NK cells were found to express C5ar2 mRNA (15) and C5aR2 protein intracellularly (15, 35). Furthermore, mouse NK cells have been reported to express low levels of C5ar2 mRNA, which was upregulated upon Escherichia coli infection (36). In this study, we found a clear tdTomato-C5aR2 signal in NK cells under steady-state conditions in spleen and blood (Fig. 7A, 7B), whereas NK cells from the spleen stained tdTomato-C5aR2 (Fig. 6D). Interestingly, only 20–25% of splenic NK cells and 30–35% of blood NK cells stained positive for tdTomato-C5aR2. C5ar2 expression was confirmed at the mRNA level using purified splenic CD49+CD3 NK cells (Fig. 7C). To assess the functional role of C5aR2 expression in NK cells, we first determined the maturation profile of NK cells from BM, spleen, and lung. As shown in Fig. 7D, we found the expected low frequency of mature CD11b+CD27 NK cells in the BM and an increased frequency of such cells in spleen and lung. C5aR2 deficiency had no impact on the distribution of mature NK cells (Fig. 7D). We also analyzed other maturation stages of NK cells (CD11bCD27, CD11bCD27+, CD11b+CD27+) and found no differences between WT and C5ar2−/− mice (data not shown). One important function of NK cells is to produce IFN-γ in response to IL-12 and IL-18 stimulation (37, 38). When we treated purified NK cells from WT and C5ar2−/− mice with IL-12 and IL-18, we found a significantly enhanced production of IFN-γ in C5aR2-deficient NK cells (Fig. 7E), suggesting that C5aR2-activation suppresses IL-12/IL-18–mediated IFN-γ production. Thus, we stimulated purified splenic NK cells from WT mice with IL-12/IL-18 in the presence or absence of the specific C5aR2 agonist P32. Indeed, such treatment significantly suppressed IL-12/IL-18–driven production of IFN-γ (Fig. 7F).

FIGURE 7.

C5aR2 expression in NK cells and its role in NK cell activation. Flow cytometric assessment of tdTomato-C5aR2 expression in splenic (A) and blood-derived (B) CD3CD49b+ NK cells. Gray histograms show the WT signal; black lines correspond to the signal in tdTomato-C5ar2fl/fl mice. (C) C5ar2 mRNA expression in sorted CD3CD49+ NK cells from spleen. (D) Frequency of CD11b+CD27 NK cells in the BM (left panel), spleen (middle panel), and lung (right panel) from WT and C5ar2−/− mice. (E) INF-γ production from sorted splenic NK cells of WT and C5ar2−/− mice 24 h after stimulation with IL-12/IL-18. (F) INF-γ production from sorted WT splenic NK cells 24 h after stimulation with IL-12/IL-18, with or without the C5aR2 agonist P32. Shown is the relative decrease in IL-12/IL-18–mediated IFN-γ production in response to C5aR2 stimulation by the C5aR2 agonist P32. Data in (A)–(D) are representative of five independent experiments. *p < 0.05, ***p < 0.001, unpaired Student t test (D and E), paired Student t test (F).

FIGURE 7.

C5aR2 expression in NK cells and its role in NK cell activation. Flow cytometric assessment of tdTomato-C5aR2 expression in splenic (A) and blood-derived (B) CD3CD49b+ NK cells. Gray histograms show the WT signal; black lines correspond to the signal in tdTomato-C5ar2fl/fl mice. (C) C5ar2 mRNA expression in sorted CD3CD49+ NK cells from spleen. (D) Frequency of CD11b+CD27 NK cells in the BM (left panel), spleen (middle panel), and lung (right panel) from WT and C5ar2−/− mice. (E) INF-γ production from sorted splenic NK cells of WT and C5ar2−/− mice 24 h after stimulation with IL-12/IL-18. (F) INF-γ production from sorted WT splenic NK cells 24 h after stimulation with IL-12/IL-18, with or without the C5aR2 agonist P32. Shown is the relative decrease in IL-12/IL-18–mediated IFN-γ production in response to C5aR2 stimulation by the C5aR2 agonist P32. Data in (A)–(D) are representative of five independent experiments. *p < 0.05, ***p < 0.001, unpaired Student t test (D and E), paired Student t test (F).

Close modal

IL-33 is abundantly expressed in lung epithelial cells and regulates the development of Th2 immunity in experimental allergic asthma (39). In this study, we administered IL-33 i.t. and determined the impact of this treatment on the frequency of tdTomato-C5aR2+ immune cells in the lung and airways. Further, we assessed the change in tdTomato-C5aR2 expression in such cells. Administration of IL-33 into WT and tdTomato-C5ar2fl/fl mice resulted in similar accumulation of inflammatory cells in the airways and the lung tissue (data not shown). However, IL-33 treatment reduced the frequency and the expression levels of tdTomato-C5aR2 in lung eosinophils and moDCs but not in macrophages or CD11b+ cDCs (Fig. 8A, 8C, 8E). Further, the frequency of tdTomato-C5aR2+ airway macrophages or their tdTomato-C5aR2 expression was not altered (Fig. 8B, 8D, 8F).

FIGURE 8.

Impact of i.t. IL-33 stimulation on C5aR2 expression in pulmonary and airway immune cells. Flow cytometric assessment of tdTomato-C5aR2 expression in different airway and pulmonary immune cells. The gray histograms show the WT signal; the black lines show the signal obtained with heterozygous tdTomato-C5ar2fl/+ mice upon IL-33 treatment, and the dashed lines correspond to the signal obtained upon PBS control treatment. Flow cytometric analysis of the tdTomato-C5aR2 signal in pulmonary eosinophils, macrophages, CD11b+ cDCs, and moDCs (A) and in airway macrophages (B). Quantitative evaluation of tdTomato-C5aR2 expression in pulmonary eosinophils, macrophages, CD11+ cDCs, and moDCs (C) and in airway macrophages (D). Shown is the MFI of the tdTomato signal normalized to WT control fluorescence (ΔMFI) in cells from mice treated with PBS or IL-33. Data are mean ± SEM. Quantitative evaluation of tdTomato-C5aR2+ pulmonary cell populations (E) and airway macrophages (F). Shown is the frequency of tdTomato-C5aR2+ cells in response to PBS or IL-33 treatment. Data are mean ± SEM. *p < 0.05, *p < 0.05, ***p < 0.001, Student t test.

FIGURE 8.

Impact of i.t. IL-33 stimulation on C5aR2 expression in pulmonary and airway immune cells. Flow cytometric assessment of tdTomato-C5aR2 expression in different airway and pulmonary immune cells. The gray histograms show the WT signal; the black lines show the signal obtained with heterozygous tdTomato-C5ar2fl/+ mice upon IL-33 treatment, and the dashed lines correspond to the signal obtained upon PBS control treatment. Flow cytometric analysis of the tdTomato-C5aR2 signal in pulmonary eosinophils, macrophages, CD11b+ cDCs, and moDCs (A) and in airway macrophages (B). Quantitative evaluation of tdTomato-C5aR2 expression in pulmonary eosinophils, macrophages, CD11+ cDCs, and moDCs (C) and in airway macrophages (D). Shown is the MFI of the tdTomato signal normalized to WT control fluorescence (ΔMFI) in cells from mice treated with PBS or IL-33. Data are mean ± SEM. Quantitative evaluation of tdTomato-C5aR2+ pulmonary cell populations (E) and airway macrophages (F). Shown is the frequency of tdTomato-C5aR2+ cells in response to PBS or IL-33 treatment. Data are mean ± SEM. *p < 0.05, *p < 0.05, ***p < 0.001, Student t test.

Close modal

C5a exerts critical roles in the regulation of innate and adaptive immune responses through activation of its cognate receptors C5aR1 and C5aR2. C5aR1 signaling pathways intersect with TLR (40, 41) and FcγR signaling (3). Also, they modulate and control T cell proliferation and differentiation (34, 42). The role of C5aR2 in C5a-driven effector and regulatory functions is less well understood (12, 43). Initially, C5aR2 was considered solely as a decoy receptor counterbalancing the proinflammatory effector functions mediated by C5aR1 (13); however, more recent data show that C5aR2 exerts pro- and anti-inflammatory properties independently of C5aR1 (1719). For example, C5aR2 selectively modulates LPS-driven IL-6 production from human macrophages (5). Previous studies suggested that C5aR2 is broadly expressed on several myeloid and lymphoid cells together with C5aR1 (43); however, C5aR2 expression levels are lower than those of C5aR1 (15, 19, 25). Until now, a detailed picture of C5aR2 expression in murine cells and tissues was lacking. Taking advantage of the floxed tdTomato-C5aR2 reporter knock-in mouse, we provide a detailed map of C5aR2-expressing immune cells in different tissues under steady-state conditions and upon pulmonary inflammation. We also evaluated tdTomato-C5aR2 expression in lung and intestinal epithelial cells. In contrast to C5aR1 and C5aR2 expression in human intestinal epithelial cells and a colonic cell line (44), we found no evidence for tdTomato-C5aR2 in intestinal or pulmonary epithelial cells.

First, we assessed the distribution of C5aR2 expression in neutrophils from different organs. In a previous article, neutrophils were identified as C5aR2+ cells; however, the same article stated that cells from the BM were negative for C5aR2 (25). This view was challenged by later studies showing C5ar2 mRNA expression, as well as protein expression, in BM neutrophils (19). In line with these studies, we found that neutrophils from BM, blood, and the peritoneal cavity were strongly positive for tdTomato-C5aR2. tdTomato expression mirrored C5ar2 mRNA expression, demonstrating that the reporter mouse strain is a reliable tool to determine C5aR2 expression. Moreover, C5aR2 expressed by neutrophils in tdTomato-C5ar2fl/fl mice is fully functional, as evidenced by the fact that the recently described C5aR2 agonist P32 (5) suppressed C5a-mediated ERK1/2 phosphorylation downstream of C5aR1 signaling in neutrophils from the reporter mice. Also, we demonstrate that the C5aR2 reporter mouse allows tracking of C5aR2 expression by immunofluorescence microscopy. Importantly, we tested two commercially available Abs that are reported to specifically recognize C5aR2. To our surprise, these Abs showed strong signals with neutrophils or macrophages from WT mice, as well as from C5aR2-deficient mice, clearly demonstrating that they recognize structures other than C5aR2. This finding further supports the view that tracking of C5aR2 expression independently of Ab staining is required to provide a reliable picture about C5aR2 expression in tissues.

In the next step, we assessed the distribution of tdTomato-C5aR2+ cells in blood, spleen, brain, VAT, lung, LP of the small intestine, BAL, and BM. We found tdTomato-C5aR2+ cells in all tested organs, with the highest abundance in brain and BM.

Previous studies found important roles for anaphylatoxin receptors in the development of neurodegenerative diseases in mice and humans (10, 45). In line with these observations, we already observed strong expression of tdTomato-C5aR2 in naive mice. In a recent study, Fonseca et al. (46) elegantly showed the upregulation of C5aR1 and C5aR2 in human Alzheimer’s disease patients using anaphylatoxin receptor Abs. To ensure the specificity of the staining and identify C5aR1- and C5aR2-expressing cells in that study, the investigators combined several Ab clones, confirming the difficulties that we observed using C5aR2 Abs in mice. This finding is in line with a recent study in which the investigators encountered problems with the reliable use of C5aR2 Abs in the spinal cord, because all Abs showed staining in C5ar2−/− mice (47). Using the tdTomato-C5aR2 reporter mouse, we observed C5aR2 receptor expression in resident microglia cells and macrophages. The strong tdTomato signal that we observed in both populations is in line with studies indicating that C5aR2 is expressed in microglia cells (45, 48, 49). Further, another study showed that astrocytes and neurons also express C5aR2 (46). A detailed and comprehensive picture of C5aR2 expression in the multiple cell types in the brain and neuronal tissue is beyond the scope of the current study and will be addressed in a future analysis.

In allergic asthma models, it was shown that the C5aR2 is required for full manifestation of the allergic phenotype, including the development of airway hyperresponsiveness and airway eosinophilia (27). In this article, we show that tdTomato-C5aR2 is strongly expressed in eosinophils from the BM, blood, lung, and LP of the small intestine and, to a lesser extent, the VAT. Interestingly, our findings suggest that the expression levels of C5aR2 in eosinophils are somewhat lower than in neutrophils, particularly in BM. In line with our observations in mice, human eosinophils were reported to express C5aR2, at least at the mRNA level (15). Pulmonary challenge with IL-33, a strong proinflammatory cytokine involved in the early response of the lung mucosal surface to airway insults had a strong impact on C5aR2 expression in eosinophils and moDCs. In fact, C5aR2 expression was decreased, as well as the frequency of C5aR2+ eosinophils and moDCs. We have previously shown that pulmonary eosinophils and moDCs also express C5aR1 (7) and that eosinophils upregulate C5aR1 during allergic asthma (50). Given that C5aR2 can control and counteract C5aR1-driven cell activation (18), these findings suggest that eosinophils and moDCs may become more sensitive to C5a-mediated cell activation in a pulmonary environment with high IL-33 levels.

It was shown that C5aR2 expression in human monocytes was upregulated in patients with Mycobacterium tuberculosis infection (51), suggesting a role for this receptor in monocyte/macrophage-mediated control of this pathogen. In line with an important role for C5aR2 in macrophage function, it was shown that atherosclerosis and vascular remodeling is attenuated in C5aR2-deficient mice. In vitro assays further revealed that proinflammatory and proatherosclerotic mediators from macrophages were reduced significantly when C5aR2 was depleted (52). Further, C5aR2 activation in macrophages has been suggested to regulate the development of obesity, because C5aR2 deficiency increased macrophage numbers in white adipose tissues, thereby contributing to obesity-associated pathologies (53). We found tdTomato-C5aR2 signals in all macrophage populations under steady-state conditions, including those from VAT, although the expression was lower compared with that of macrophages from other compartments. In future work, tdTomato-C5aR2 reporter mice might prove useful to track C5aR2 expression in VAT macrophages under obese conditions.

Investigating macrophages from BM cell culture, we found that only one third of these cells expressed tdTomato-C5aR2 with a relatively low fluorescence intensity. Recently, it was hypothesized that, in this setting, C5aR2 might play a more important role than C5aR1 in regulating IL-1β responses (54). Clearly, specific deletion of C5aR2 using the floxed tdTomato-C5aR2 reporter mouse will help to clarify the exact role of C5aR2 in NLRP3 inflammasome activation in macrophages.

In the initial article describing C5aR2, Ohno et al. (25) found that C5aR2 is expressed in immature, but not in mature, DCs. This view was challenged by Gerard and colleagues (15), who showed that C5aR2 expression is absent in immature DCs but was upregulated upon TLR4 or TLR9 stimulation in these cells. Using the tdTomato-C5aR2 reporter mouse, we demonstrate that the C5aR2 expression pattern strongly depends on the DC subset investigated. Although moDCs and CD11b+CD103 DCs from lung express low levels of tdTomato-C5aR2 under steady-state conditions, the receptor is barely detectable in other DC subpopulations. Also, GM-CSF–differentiated DCs from BM were largely negative for C5aR2, which is in line with the observations by Ohno et al. (25). This weak expression of C5aR2 in DCs might explain the minor influence of C5aR2 activation on the process of splenic DC–driven naive T cell differentiation into Th1 or Th17 effector or regulatory T cells (55).

Taking into account the recently discovered important role for C5aR2 in complement-driven inflammasome activation of human CD4+ T cells (34) and considering the ongoing controversy regarding the expression of C5aR1 in murine T cells (68, 42), we tested T cell subsets from different sources for tdTomato-C5aR2 expression. Several articles have highlighted the role of C5aR1 for the regulation of T cell survival, proliferation, and differentiation into Th1 (40), Th2 (56), or Th17 (55, 57, 58) effector cells. Importantly, C5aR2 was also recently shown to regulate Th1 lineage commitment through its impact on NLRP3 activation in human CD4+ T cells (34). Similar to what we had previously observed for C5aR1 using GFP-C5aR1 reporter mice (7), we found that T cells completely lacked tdTomato-C5aR2 and C5ar2 mRNA expression, even after CD3/CD28 stimulation. Our findings suggest that the differences that we observed previously for C5aR1 expression between human and mouse T cells also apply to the expression pattern of C5aR2.

With regard to C5aR2 expression in murine B cells, very limited information is available. In the human system, C5aR2 expression was shown in plasma and memory B cells (15). In this article, we demonstrate that naive mouse B cells from spleen, blood, and mLNs homogeneously express C5aR2. These data were corroborated by C5ar2 mRNA expression in splenic B cells. These results are in sharp contrast to our findings in GFP-C5aR1 reporter mice (7); B cells from different lymphoid organs did not express C5aR1. The expression of C5aR2 in murine B cells independently from C5aR1 suggests an exclusive functional role for C5a on mouse B cells through C5aR2.

Finally, we determined tdTomato-C5aR2 expression in NK cells from different tissues. Interestingly, we found that only subpopulations of NK cells expressed the receptor in blood and spleen. In a previous report, low levels of C5ar2 mRNA expression were reported for NK cells from spleen, which was upregulated upon exposure to E. coli (15, 35, 36). Also, C5aR2 mRNA expression has been shown in human NK cells (15). More recently, this finding has been confirmed by another group, who also demonstrated intracellular expression of C5aR2 (35). The expression of C5aR2 in NK cells contrasts with previous findings obtained with GFP-C5aR1 reporter mice (7). In that study, we found no C5aR1 expression in NK cells from different tissues, suggesting that, similar to what we have observed in B cells, C5aR2 is the C5aR receptor that is exclusively expressed in mouse NK cells. Assessing the functional consequence of the expression of C5aR2, we found that NK cells, which lack C5aR2, are much more potent IFN-γ producers than their WT counterparts in response to IL-12/IL-18 stimulation. Further, specific ligation of C5aR2 with the C5aR2 agonist P32 suppressed IFN-γ production in WT NK cells. These findings demonstrate that C5aR2 activation controls IL-12/IL-18–induced IFN-γ production from NK cells, which plays an important role during early infection with intracellular pathogens; these data suggest that the complement system regulates early immune responses during such infections through NK cell control.

In summary, we provide a detailed map of C5aR2 expression in different cells from the innate and adaptive immune system. Our data clearly show C5aR2 expression in neutrophils, eosinophils, macrophages, moDCs, CD11b+CD103 cDCs, B cells, and NK cells from different tissues. In contrast, we demonstrate that most of the DC subsets, as well as naive and activated T cells, do not express C5aR2. Also, we show a novel function for C5aR2 in NK cells and a novel role for IL-33 as regulator of C5aR2 expression in pulmonary eosinophils and moDCs. The floxed tdTomato-C5aR2 reporter mouse will prove useful to track C5aR2 expression and to conditionally delete this receptor in experimental disease settings to further delineate the enigmatic role of this anaphylatoxin receptor in health and disease.

This work was supported by German Research Foundation International Research Training Group 1911 Projects A1, A7/B1, and B2 (to Y.L., J.K., and C.M.K., respectively) and the National Council for Scientific and Technological Development-Brazil (to L.N.A.).

Abbreviations used in this article:

AF

Alexa Fluor

BAL

bronchoalveolar lavage fluid

BM

bone marrow

BMDC

BM-derived DC

BMM

BM-derived macrophage

BMpDC

BM-derived plasmacytoid DC

BV

Brilliant Violet

C5aR

C5a receptor

cDC

conventional DC

DC

dendritic cell

eF

eFluor

ES

embryonic stem

GPCR

G protein–coupled receptor

i.t.

intratracheal(ly)

LP

lamina propria

MFI

mean fluorescence intensity

MHCII

MHC class II

mLN

mesenteric lymph node

moDC

monocyte-derived DC

pDC

plasmacytoid DC

RBCL

RBC lysis

tdTomato

tandem dye Tomato

VAT

visceral adipose tissue

WT

wild-type.

1
Guo
,
R. F.
,
P. A.
Ward
.
2005
.
Role of C5a in inflammatory responses.
Annu. Rev. Immunol.
23
:
821
852
.
2
Guo
,
R. F.
,
N. C.
Riedemann
,
P. A.
Ward
.
2004
.
Role of C5a-C5aR interaction in sepsis.
Shock
21
:
1
7
.
3
Karsten
,
C. M.
,
J.
Köhl
.
2012
.
The immunoglobulin, IgG Fc receptor and complement triangle in autoimmune diseases.
Immunobiology
217
:
1067
1079
.
4
Fonseca
,
M. I.
,
R. R.
Ager
,
S. H.
Chu
,
O.
Yazan
,
S. D.
Sanderson
,
F. M.
LaFerla
,
S. M.
Taylor
,
T. M.
Woodruff
,
A. J.
Tenner
.
2009
.
Treatment with a C5aR antagonist decreases pathology and enhances behavioral performance in murine models of Alzheimer’s disease.
J. Immunol.
183
:
1375
1383
.
5
Croker
,
D. E.
,
P. N.
Monk
,
R.
Halai
,
G.
Kaeslin
,
Z.
Schofield
,
M. C.
Wu
,
R. J.
Clark
,
M. A.
Blaskovich
,
D.
Morikis
,
C. A.
Floudas
, et al
.
2016
.
Discovery of functionally selective C5aR2 ligands: novel modulators of C5a signalling.
Immunol. Cell Biol.
94
:
787
795
.
6
Strainic
,
M. G.
,
E. M.
Shevach
,
F.
An
,
F.
Lin
,
M. E.
Medof
.
2013
.
Absence of signaling into CD4+ cells via C3aR and C5aR enables autoinductive TGF-β1 signaling and induction of Foxp3+ regulatory T cells.
Nat. Immunol.
14
:
162
171
.
7
Karsten
,
C. M.
,
Y.
Laumonnier
,
B.
Eurich
,
F.
Ender
,
K.
Bröker
,
S.
Roy
,
A.
Czabanska
,
T.
Vollbrandt
,
J.
Figge
,
J.
Köhl
.
2015
.
Monitoring and cell-specific deletion of C5aR1 using a novel floxed GFP-C5aR1 reporter knock-in mouse.
J. Immunol.
194
:
1841
1855
.
8
Dunkelberger
,
J.
,
L.
Zhou
,
T.
Miwa
,
W. C.
Song
.
2012
.
C5aR expression in a novel GFP reporter gene knockin mouse: implications for the mechanism of action of C5aR signaling in T cell immunity.
J. Immunol.
188
:
4032
4042
.
9
Schmudde
,
I.
,
H. A.
Ströver
,
T.
Vollbrandt
,
P.
König
,
C. M.
Karsten
,
Y.
Laumonnier
,
J.
Köhl
.
2013
.
C5a receptor signalling in dendritic cells controls the development of maladaptive Th2 and Th17 immunity in experimental allergic asthma.
Mucosal Immunol.
6
:
807
825
.
10
Klos
,
A.
,
A. J.
Tenner
,
K. O.
Johswich
,
R. R.
Ager
,
E. S.
Reis
,
J.
Köhl
.
2009
.
The role of the anaphylatoxins in health and disease.
Mol. Immunol.
46
:
2753
2766
.
11
Kemper
,
C.
,
J.
Köhl
.
2013
.
Novel roles for complement receptors in T cell regulation and beyond.
Mol. Immunol.
56
:
181
190
.
12
Kemper
,
C.
2016
.
Targeting the Dark Horse of complement: the first generation of functionally selective C5aR2 ligands.
Immunol. Cell Biol.
94
:
717
718
.
13
Okinaga
,
S.
,
D.
Slattery
,
A.
Humbles
,
Z.
Zsengeller
,
O.
Morteau
,
M. B.
Kinrade
,
R. M.
Brodbeck
,
J. E.
Krause
,
H. R.
Choe
,
N. P.
Gerard
,
C.
Gerard
.
2003
.
C5L2, a nonsignaling C5A binding protein.
Biochemistry
42
:
9406
9415
.
14
Scola
,
A. M.
,
K. O.
Johswich
,
B. P.
Morgan
,
A.
Klos
,
P. N.
Monk
.
2009
.
The human complement fragment receptor, C5L2, is a recycling decoy receptor.
Mol. Immunol.
46
:
1149
1162
.
15
Bamberg
,
C. E.
,
C. R.
Mackay
,
H.
Lee
,
D.
Zahra
,
J.
Jackson
,
Y. S.
Lim
,
P. L.
Whitfeld
,
S.
Craig
,
E.
Corsini
,
B.
Lu
, et al
.
2010
.
The C5a receptor (C5aR) C5L2 is a modulator of C5aR-mediated signal transduction.
J. Biol. Chem.
285
:
7633
7644
.
16
Wang
,
R.
,
B.
Lu
,
C.
Gerard
,
N. P.
Gerard
.
2016
.
C5L2, the second C5a anaphylatoxin receptor, suppresses LPS-induced acute lung injury.
Am. J. Respir. Cell Mol. Biol.
55
:
657
666
.
17
Gao
,
H.
,
T. A.
Neff
,
R. F.
Guo
,
C. L.
Speyer
,
J. V.
Sarma
,
S.
Tomlins
,
Y.
Man
,
N. C.
Riedemann
,
L. M.
Hoesel
,
E.
Younkin
, et al
.
2005
.
Evidence for a functional role of the second C5a receptor C5L2.
FASEB J.
19
:
1003
1005
.
18
Croker
,
D. E.
,
R.
Halai
,
G.
Kaeslin
,
E.
Wende
,
B.
Fehlhaber
,
A.
Klos
,
P. N.
Monk
,
M. A.
Cooper
.
2014
.
C5a2 can modulate ERK1/2 signaling in macrophages via heteromer formation with C5a1 and β-arrestin recruitment.
Immunol. Cell Biol.
92
:
631
639
.
19
Chen
,
N. J.
,
C.
Mirtsos
,
D.
Suh
,
Y. C.
Lu
,
W. J.
Lin
,
C.
McKerlie
,
T.
Lee
,
H.
Baribault
,
H.
Tian
,
W. C.
Yeh
.
2007
.
C5L2 is critical for the biological activities of the anaphylatoxins C5a and C3a.
Nature
446
:
203
207
.
20
Rittirsch
,
D.
,
M. A.
Flierl
,
B. A.
Nadeau
,
D. E.
Day
,
M.
Huber-Lang
,
C. R.
Mackay
,
F. S.
Zetoune
,
N. P.
Gerard
,
K.
Cianflone
,
J.
Köhl
, et al
.
2008
.
Functional roles for C5a receptors in sepsis.
Nat. Med.
14
:
551
557
.
21
Helft
,
J.
,
J.
Böttcher
,
P.
Chakravarty
,
S.
Zelenay
,
J.
Huotari
,
B. U.
Schraml
,
D.
Goubau
,
C.
Reis e Sousa
.
2015
.
GM-CSF mouse bone marrow cultures comprise a heterogeneous population of CD11c(+)MHCII(+) macrophages and dendritic cells.
Immunity
42
:
1197
1211
.
22
Weischenfeldt
,
J.
,
B.
Porse
.
2008
.
Bone marrow-derived macrophages (BMM): isolation and applications.
CSH Protoc.
2008
:
pdb.prot5080
.
23
Quell
,
K. M.
,
C. M.
Karsten
,
A.
Kordowski
,
L. N.
Almeida
,
D.
Briukhovetska
,
A. V.
Wiese
,
J.
Sun
,
F.
Ender
,
K.
Antoniou
,
T.
Schröder
, et al
.
2017
.
Monitoring C3aR expression using a floxed tdTomato-C3aR reporter knock-in mouse.
J. Immunol.
199
:
688
706
.
24
Kondo
,
Y.
,
T.
Yoshimoto
,
K.
Yasuda
,
S.
Futatsugi-Yumikura
,
M.
Morimoto
,
N.
Hayashi
,
T.
Hoshino
,
J.
Fujimoto
,
K.
Nakanishi
.
2008
.
Administration of IL-33 induces airway hyperresponsiveness and goblet cell hyperplasia in the lungs in the absence of adaptive immune system.
Int. Immunol.
20
:
791
800
.
25
Ohno
,
M.
,
T.
Hirata
,
M.
Enomoto
,
T.
Araki
,
H.
Ishimaru
,
T. A.
Takahashi
.
2000
.
A putative chemoattractant receptor, C5L2, is expressed in granulocyte and immature dendritic cells, but not in mature dendritic cells.
Mol. Immunol.
37
:
407
412
.
26
Bosmann
,
M.
,
M. D.
Haggadone
,
F. S.
Zetoune
,
J. V.
Sarma
,
P. A.
Ward
.
2013
.
The interaction between C5a and both C5aR and C5L2 receptors is required for production of G-CSF during acute inflammation.
Eur. J. Immunol.
43
:
1907
1913
.
27
Zhang
,
X.
,
I.
Schmudde
,
Y.
Laumonnier
,
M. K.
Pandey
,
J. R.
Clark
,
P.
König
,
N. P.
Gerard
,
C.
Gerard
,
M.
Wills-Karp
,
J.
Köhl
.
2010
.
A critical role for C5L2 in the pathogenesis of experimental allergic asthma.
J. Immunol.
185
:
6741
6752
.
28
Wu
,
D.
,
A. B.
Molofsky
,
H. E.
Liang
,
R. R.
Ricardo-Gonzalez
,
H. A.
Jouihan
,
J. K.
Bando
,
A.
Chawla
,
R. M.
Locksley
.
2011
.
Eosinophils sustain adipose alternatively activated macrophages associated with glucose homeostasis.
Science
332
:
243
247
.
29
Rothenberg
,
M. E.
,
S. P.
Hogan
.
2006
.
The eosinophil.
Annu. Rev. Immunol.
24
:
147
174
.
30
Guilliams
,
M.
,
F.
Ginhoux
,
C.
Jakubzick
,
S. H.
Naik
,
N.
Onai
,
B. U.
Schraml
,
E.
Segura
,
R.
Tussiwand
,
S.
Yona
.
2014
.
Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny.
Nat. Rev. Immunol.
14
:
571
578
.
31
Mayer
,
C. T.
,
P.
Ghorbani
,
A.
Nandan
,
M.
Dudek
,
C.
Arnold-Schrauf
,
C.
Hesse
,
L.
Berod
,
P.
Stüve
,
F.
Puttur
,
M.
Merad
,
T.
Sparwasser
.
2014
.
Selective and efficient generation of functional Batf3-dependent CD103+ dendritic cells from mouse bone marrow.
Blood
124
:
3081
3091
.
32
Hoffmann
,
F.
,
F.
Ender
,
I.
Schmudde
,
I. P.
Lewkowich
,
J.
Köhl
,
P.
König
,
Y.
Laumonnier
.
2016
.
Origin, localization, and immunoregulatory properties of pulmonary phagocytes in allergic asthma.
Front. Immunol.
7
:
107
.
33
Varol
,
C.
,
A.
Vallon-Eberhard
,
E.
Elinav
,
T.
Aychek
,
Y.
Shapira
,
H.
Luche
,
H. J.
Fehling
,
W. D.
Hardt
,
G.
Shakhar
,
S.
Jung
.
2009
.
Intestinal lamina propria dendritic cell subsets have different origin and functions.
Immunity
31
:
502
512
.
34
Arbore
,
G.
,
E. E.
West
,
R.
Spolski
,
A. A.
Robertson
,
A.
Klos
,
C.
Rheinheimer
,
P.
Dutow
,
T. M.
Woodruff
,
Z. X.
Yu
,
L. A.
O’Neill
, et al
.
2016
.
T helper 1 immunity requires complement-driven NLRP3 inflammasome activity in CD4+ T cells.
Science
352
:
aad1210
.
35
Min
,
X.
,
C.
Liu
,
Y.
Wei
,
N.
Wang
,
G.
Yuan
,
D.
Liu
,
Z.
Li
,
W.
Zhou
,
K.
Li
.
2014
.
Expression and regulation of complement receptors by human natural killer cells.
Immunobiology
219
:
671
679
.
36
Fusakio
,
M. E.
,
J. P.
Mohammed
,
Y.
Laumonnier
,
K.
Hoebe
,
J.
Köhl
,
J.
Mattner
.
2011
.
C5a regulates NKT and NK cell functions in sepsis.
J. Immunol.
187
:
5805
5812
.
37
Haeberlein
,
S.
,
H.
Sebald
,
C.
Bogdan
,
U.
Schleicher
.
2010
.
IL-18, but not IL-15, contributes to the IL-12-dependent induction of NK-cell effector functions by Leishmania infantum in vivo.
Eur. J. Immunol.
40
:
1708
1717
.
38
Okamura
,
H.
,
S.
Kashiwamura
,
H.
Tsutsui
,
T.
Yoshimoto
,
K.
Nakanishi
.
1998
.
Regulation of interferon-gamma production by IL-12 and IL-18.
Curr. Opin. Immunol.
10
:
259
264
.
39
Drake
,
L. Y.
,
H.
Kita
.
2017
.
IL-33: biological properties, functions, and roles in airway disease.
Immunol. Rev.
278
:
173
184
.
40
Hawlisch
,
H.
,
Y.
Belkaid
,
R.
Baelder
,
D.
Hildeman
,
C.
Gerard
,
J.
Köhl
.
2005
.
C5a negatively regulates toll-like receptor 4-induced immune responses.
Immunity
22
:
415
426
.
41
Liang
,
S.
,
J. L.
Krauss
,
H.
Domon
,
M. L.
McIntosh
,
K. B.
Hosur
,
H.
Qu
,
F.
Li
,
A.
Tzekou
,
J. D.
Lambris
,
G.
Hajishengallis
.
2011
.
The C5a receptor impairs IL-12-dependent clearance of Porphyromonas gingivalis and is required for induction of periodontal bone loss.
J. Immunol.
186
:
869
877
.
42
Strainic
,
M. G.
,
J.
Liu
,
D.
Huang
,
F.
An
,
P. N.
Lalli
,
N.
Muqim
,
V. S.
Shapiro
,
G. R.
Dubyak
,
P. S.
Heeger
,
M. E.
Medof
.
2008
.
Locally produced complement fragments C5a and C3a provide both costimulatory and survival signals to naive CD4+ T cells.
Immunity
28
:
425
435
.
43
Li
,
R.
,
L. G.
Coulthard
,
M. C.
Wu
,
S. M.
Taylor
,
T. M.
Woodruff
.
2013
.
C5L2: a controversial receptor of complement anaphylatoxin, C5a.
FASEB J.
27
:
855
864
.
44
Cao
,
Q.
,
S. M.
McIsaac
,
A. W.
Stadnyk
.
2012
.
Human colonic epithelial cells detect and respond to C5a via apically expressed C5aR through the ERK pathway.
Am. J. Physiol. Cell Physiol.
302
:
C1731
C1740
.
45
Woodruff
,
T. M.
,
R. R.
Ager
,
A. J.
Tenner
,
P. G.
Noakes
,
S. M.
Taylor
.
2010
.
The role of the complement system and the activation fragment C5a in the central nervous system.
Neuromolecular Med.
12
:
179
192
.
46
Fonseca
,
M. I.
,
S. O.
McGuire
,
S. E.
Counts
,
A. J.
Tenner
.
2013
.
Complement activation fragment C5a receptors, CD88 and C5L2, are associated with neurofibrillary pathology.
J. Neuroinflammation
10
:
25
.
47
Biggins
,
P. J.
,
F. H.
Brennan
,
S. M.
Taylor
,
T. M.
Woodruff
,
M. J.
Ruitenberg
.
2017
.
The alternative receptor for complement component 5a, C5aR2, conveys neuroprotection in traumatic spinal cord injury.
J. Neurotrauma
34
:
2075
2085
.
48
Gavrilyuk
,
V.
,
S.
Kalinin
,
B. S.
Hilbush
,
A.
Middlecamp
,
S.
McGuire
,
D.
Pelligrino
,
G.
Weinberg
,
D. L.
Feinstein
.
2005
.
Identification of complement 5a-like receptor (C5L2) from astrocytes: characterization of anti-inflammatory properties.
J. Neurochem.
92
:
1140
1149
.
49
Nomaru
,
H.
,
K.
Sakumi
,
A.
Katogi
,
Y. N.
Ohnishi
,
K.
Kajitani
,
D.
Tsuchimoto
,
E. J.
Nestler
,
Y.
Nakabeppu
.
2014
.
Fosb gene products contribute to excitotoxic microglial activation by regulating the expression of complement C5a receptors in microglia.
Glia
62
:
1284
1298
.
50
Ender
,
F.
,
A. V.
Wiese
,
I.
Schmudde
,
J.
Sun
,
T.
Vollbrandt
,
P.
König
,
Y.
Laumonnier
,
J.
Köhl
.
2017
.
Differential regulation of C5a receptor 1 in innate immune cells during the allergic asthma effector phase.
PLoS One
12
:
e0172446
.
51
Sabio y García
,
C. A.
,
N.
Yokobori
,
J. I.
Basile
,
L.
Balboa
,
A.
González
,
B.
López
,
V.
Ritacco
,
S.
Barrera
,
M. D.
Sasiain
.
2017
.
C5aR contributes to the weak Th1 profile induced by an outbreak strain of Mycobacterium tuberculosis.
Tuberculosis (Edinb.)
103
:
16
23
.
52
Selle
,
J.
,
Y.
Asare
,
J.
Köhncke
,
S.
Alampour-Rajabi
,
G.
Shagdarsuren
,
A.
Klos
,
C.
Weber
,
J.
Jankowski
,
E.
Shagdarsuren
.
2015
.
Atheroprotective role of C5ar2 deficiency in apolipoprotein E-deficient mice.
Thromb. Haemost.
114
:
848
858
.
53
Gauvreau
,
D.
,
A.
Gupta
,
A.
Fisette
,
F. Q.
Tom
,
K.
Cianflone
.
2013
.
Deficiency of C5L2 increases macrophage infiltration and alters adipose tissue function in mice.
PLoS One
8
:
e60795
.
54
Haggadone
,
M. D.
,
J. J.
Grailer
,
F.
Fattahi
,
F. S.
Zetoune
,
P. A.
Ward
.
2016
.
Bidirectional crosstalk between C5a receptors and the NLRP3 inflammasome in macrophages and monocytes.
Mediators Inflamm.
2016
:
1340156
.
55
Weaver
,
D. J.
 Jr.
,
E. S.
Reis
,
M. K.
Pandey
,
G.
Köhl
,
N.
Harris
,
C.
Gerard
,
J.
Köhl
.
2010
.
C5a receptor-deficient dendritic cells promote induction of Treg and Th17 cells.
Eur. J. Immunol.
40
:
710
721
.
56
Köhl
,
J.
,
R.
Baelder
,
I. P.
Lewkowich
,
M. K.
Pandey
,
H.
Hawlisch
,
L.
Wang
,
J.
Best
,
N. S.
Herman
,
A. A.
Sproles
,
J.
Zwirner
, et al
.
2006
.
A regulatory role for the C5a anaphylatoxin in type 2 immunity in asthma.
J. Clin. Invest.
116
:
783
796
.
57
Hashimoto
,
M.
,
K.
Hirota
,
H.
Yoshitomi
,
S.
Maeda
,
S.
Teradaira
,
S.
Akizuki
,
P.
Prieto-Martin
,
T.
Nomura
,
N.
Sakaguchi
,
J.
Köhl
, et al
.
2010
.
Complement drives Th17 cell differentiation and triggers autoimmune arthritis.
J. Exp. Med.
207
:
1135
1143
.
58
Lajoie
,
S.
,
I. P.
Lewkowich
,
Y.
Suzuki
,
J. R.
Clark
,
A. A.
Sproles
,
K.
Dienger
,
A. L.
Budelsky
,
M.
Wills-Karp
.
2010
.
Complement-mediated regulation of the IL-17A axis is a central genetic determinant of the severity of experimental allergic asthma.
Nat. Immunol.
11
:
928
935
.

The authors have no financial conflicts of interest.