B lymphocytes are important players of the adaptive immune system. However, not just activation of B cells but also regulation of B cell signaling is important to prevent hyperactivity and dysregulation of the immune response. Different mechanisms and proteins contribute to this balance. One of these is CD22, a member of the Siglec family. It is an inhibitory coreceptor of the BCR and inhibits B cell activation. Upon BCR stimulation, CD22-dependent inhibition of BCR signaling results in a decreased calcium mobilization. Although some CD22 binding partners have already been identified, the knowledge about the CD22 interactome is still incomplete. In this study, quantitative affinity purification–mass spectrometry enabled the delineation of the CD22 interactome in the B cell line DT40. These data will clarify molecular mechanisms and CD22 signaling events after BCR activation and revealed several new CD22-associated proteins. One new identified interaction partner is the E3 ubiquitin ligase cullin 3, which was revealed to regulate CD22 surface expression and clathrin-dependent CD22 internalization after BCR stimulation. Furthermore cullin 3 was identified to be important for B lymphocytes in general. B cell–specific cullin 3-deficient mice show reduced developing B cells in the bone marrow and a severe pro-B cell proliferation defect. Mature B cells in the periphery are also reduced and characterized by increased CD22 expression and additionally by preactivated and apoptotic phenotypes. The findings reveal novel functions of cullin 3 in B lymphocytes, namely regulating CD22 surface expression and internalization after B cell activation, as well as promoting proliferation of pro-B cells.

B lymphocytes, which are part of the adaptive immune system, develop from pro-B to immature B cells in the bone marrow and leave through the blood stream into the periphery. There, mature B cells are then activated during an immune response. The activation step via the BCR is important to promote B lymphocyte proliferation and further differentiation (1). However, regulation of B cell activation is also crucial for a balanced immune system, as hyperactivation and dysregulation can lead to autoimmunity. To protect B cells from hyperactivity, different regulatory proteins and mechanisms exist. For example, inhibitory membrane receptors, like CD22, are involved in preventing B cell hyperactivation (2, 3). However, their expression has to be controlled and regulated as well to ensure a balance of activation and inhibition in B lymphocytes.

CD22 (Siglec-2) belongs to the sialic acid binding Ig type lectin (Siglec) family, which consists of several transmembrane proteins. These are mostly expressed on immune cells and share some structural features. CD22 is almost exclusively expressed on B lymphocytes and is present on the cell surface from the pre-B cell stage until mature stages (4). CD22 is known to be an important inhibitor on conventional (B2) B cells and negatively regulates BCR signaling (510). Its extracellular domain is responsible for binding the ligand, α2,6-linked sialic acid, via a conserved arginine residue (R130) (11, 12). The cytoplasmic tail of CD22 contains six conserved tyrosines (Y), of which three (Y783, Y843, and Y863) are located in ITIMs (1315). After BCR activation, the inhibitory function of CD22 is mediated via phosphorylation of these ITIMs by the kinase Lyn (14). This in turn leads to SHP-1 binding and activation (16). Thus, SHP-1 inhibits the signaling complex of PLCγ2 (17) and eventually reduces calcium release from the endoplasmic reticulum (5, 7, 8). Additionally, CD22 is involved in activating the plasma membrane Ca2+ ATPase (PMCA) that further removes calcium from the cytosol (18, 19). Besides its inhibiting function, the endocytic capacity of CD22 has often been the focus of studies. On a molecular basis, it has been shown that anti-CD22 directed Ab-binding results in fast CD22 internalization via the clathrin-dependent pathway with rather slow recycling (20, 21). In this scenario AP50, a part of the clathrin-associated AP-2 adaptor complex, binds to the nonphosphorylated tyrosines of YXXØ (Ø is a hydrophobic residue) clathrin-internalization motif within the cytoplasmic tail of CD22 (Y843 and Y863) (21, 22). Moreover, in the steady-state CD22 internalization probably leads to lysosomal degradation (23). This knowledge was then transferred to B cell leukemia treatment by coupling immunotoxins to anti-CD22 Abs (2426) or sialosides (2730). Application of these treatments induces internalization of CD22 together with the bound toxin and results in the killing of leukemic cells. Because understanding the mechanism of CD22 internalization and recycling is important we were interested in finding interactors that might regulate this function. In this study, we searched for new CD22 interacting proteins to further decipher CD22 signaling and the underlying molecular mechanisms. We identified cullin 3 as a CD22-associated protein after anti-IgM treatment that is involved in CD22 internalization.

Cullin 3 belongs to the cullin protein family with seven different family members in mammals that function as E3 ubiquitin ligases (31). Ubiquitination is a posttranslational modification and is defined as the attachment of one ubiquitin molecule or an ubiquitin chain to a protein. This is directed by a guided reaction of ubiquitin-activating enzymes (E1), ubiquitin-conjugating enzymes (E2), and ubiquitin ligases (E3) (32, 33). The attachment of already one ubiquitin moiety (also referred to as monoubiquitination) is sufficient to induce receptor internalization (34). The receptors are then transported to the early endosome (sometimes referred to as sorting endosome) and from there either further to the lysosome for degradation or are recycled back to the cell surface after deubiquitination (3537).

E3 ubiquitin ligases can be subdivided into two classes, the HECT-type E3 and the RING-type E3 ligase complexes (38, 39). Cullin-based complexes belong to the latter one. The covalent attachment of a Nedd8 moiety, an ubiquitin-like protein, activates cullin proteins (40, 41). Cullin 3, like other cullin members, operates as a scaffold protein to form a cullin 3–RING–ubiquitin ligase complex (CRL3). This multisubunit complex consists of a cullin 3, a RING protein, an E2 ubiquitin ligase, and a substrate adaptor that is connected via its BTB/POZ domain to the cullin 3 N terminus (38, 42). The active form of CRL3 is homodimeric, and the dimerization is mediated via the BTB/POZ-substrate adaptor proteins (4345). Protein-interaction domains at the C terminus of the substrate adaptor bind the substrate and thereby recruit it into the CRL3 complex for ubiquitination. Those protein–protein-interaction motifs of BTB/POZ domain proteins are mostly either Kelch, PHR, or zinc finger domains (46). One protein family that often functions as a substrate adaptor in CRL3 is the Kelch-like family (KLHL), with KLHL11 or KLHL6 as examples (45, 47). Ubiquitination of the target protein can have different effects, such as proteasomal or lysosomal degradation or influences on protein localization (48). The function of cullin 3 and its targets were first studied in 1999 and revealed a regulatory role for cullin 3 in Cyclin E expression and therefore controlling S-phase entry in mammalian cells (49). Since then, many studies analyzed the function of cullin 3 in various organisms and cell types. Thus, cullin 3 is important to, for example, degrade MEI-1/Katanin in meiosis-to-mitosis transition in Caenorhabditis elegans or to target Cyclin E to maintain quiescence in murine hepatocytes (50, 51). The important role of cullin 3 is underlined by the fact that a total cullin 3 deletion is embryonic lethal (51), indicating an important developmental role.

During B cell development, cullin 3 is already expressed in the common lymphoid progenitor. The expression level is moderate and constant during B cell development, however the highest expression was observed in germinal center and plasma cells (ImmGen database, http://rstats.immgen.org/Skyline/skyline.html). So far, only one group has studied the function of cullin 3 in B lymphocytes. Using a CUL3flox/flox mouse bred to a CD19Cre mouse revealed hardly any alterations in B lymphocyte development (52). However, the marginal zone B cell population in the spleen and the B1 B cell population in the peritoneal cavity were impaired. They also demonstrated that cullin 3 associates with the BTB-zink finger transcription factor BCL6, which is important to direct germinal center B cell and T follicular helper cell development (52). In this study, we did not only focus on the role of cullin 3 with respect to CD22 but expanded our investigation to analyze general cullin 3 functions in B lymphocytes. Our studies revealed that cullin 3 is not only regulating CD22 expression and internalization after BCR activation but is also important to control pro-B cell proliferation and B cell survival.

Cul3flox/flox mice (51) were purchased from Jackson laboratory (JAX stock no. 028349) and crossed to Flp mice to remove the frt-flanked PGK-neo cassette. Afterwards, Cul3floxΔneo mice were mated with mb1CRE mice (53) to obtain B cell–specific cullin 3 deficient, mice which were further named CUL3fl/fl mb1CRE/+ mice. Control mice (CUL3fl/fl mb1+/+ or CUL3WT/WT mb1CRE/+) were age- and sex-matched littermates with the same genetic background. No differences were observed between the two used controls, CUL3fl/fl mb1+/+ and CUL3WT/WT mb1CRE/+. Mice were originally generated from 129S4/SvJaeSor-derived AK7 embryonic stem cells (51) and were backcrossed to C57BL/6 for seven generations. CD22−/− mice (5) and CD22-Y2,5,6F mice, which have point mutations in the tyrosines of ITIM 2,5,6 (12), were generated as described. Experiments were performed in accordance with the German law for protection of animals, after approval by the animal welfare committee.

Single-cell suspensions of bone marrow, spleen, lymph nodes, or isolated peritoneal cavity were prepared in PBS (Life Technologies) supplemented with 5% FCS (PAN Biotech). Blood was collected in PBS containing 1% heparin (Ratiopharm) from either tail vein or heart. Erythrocytes were depleted using 1× ammonium–chloride–potassium (ACK) lysis buffer, and afterward, cells were washed and stained with Abs conjugated to FITC, AlexaFluor488, PE, PE-CF594, PerCP-Cy5.5, PE-Vio770, allophycocyanin, allophycocyanin-Cy7, BV421, efluor506, or biotin and diluted in PBS containing 0.1% BSA (Carl Roth), 2 mM EDTA (Carl Roth), and 2 mM sodium azide (Sigma-Aldrich). The following Abs were used for cell surface staining: anti-B220 (clone RA3-6B2; eBioscience), anti-CD4 (clone GK1.5; BD Biosciences), anti-CD5 (clone 53-7.3; eBioscience), anti-CD8 (clone 53-6.7; eBioscience), anti-CD19 (clone 6D5; BioLegend), anti-CD21 (clone 7E9; own hybridoma), anti-CD22 (Cy34.1; BD Biosciences), anti-CD23 (clone B3-B4; BD Biosciences), anti-CD24/anti-HSA (clone M1/69; BD Biosciences), anti-CD25 (clone PC61.5; eBioscience), anti-CD43 (clone BV510; BD Biosciences), anti-CD86 (clone GL-1; eBioscience), anti-CD93 (clone AA4.1; ebioscience), anti-CD95/anti–Fas-R (clone Jo2; BD Biosciences), anti-CD117/anti–c-kit (clone 2B8; BioLegend), anti-CD127/anti–IL-7-Rα (clone A7R34; BioLegend), anti-CD268/anti–BAFF-R (clone 7H22-E16; BioLegend), anti-IgD (clone 11-26c; BD Biosciences), anti-IgM (clone II/41; eBioscience), anti-κ–L chain (clone R5240; Pharmingen), anti–MHC class II (MHCII) (clone 25-9-17; BD Biosciences), anti–Siglec-G (clone SH1; BD Biosciences), and α2-6-Sialic acid-PAA [6´SLN(Gc)-PAA, 1MD, kindly provided by N. Bovin]. Fc-block (2.4G2; hybridoma generated in house) was added to prevent unspecific binding via the Fc-part of the Abs to the cell’s Fc receptors. Cells were stained for 30 min at 4°C. To stain dead cells, fixable viability dye (eBioscience) or DAPI (Sigma-Aldrich) was used. Biotinylated Abs were detected by an additional staining of the cells with streptavidin-PE-Vio770 (Miltenyi Biotec). Afterwards, cells were washed. In case of intracellular staining, cells were fixed and permeabilized with Cytofix/Cytoperm kit (BD Biosciences) or by using 4% paraformaldehyde (PFA; Carl Roth) and 70% methanol in PBS (Carl Roth). The following Abs were used for intracellular staining: anti-active caspase3 (clone C92-605; BD Biosciences), anti-pY551/pY511-Btk (clone 24a/BTK; BD Biosciences), and anti-pY759-PLCγ2 (clone K68-689.37; BD Biosciences). In case of cell cycle analyses, DNA staining was performed with 10 μg/ml DAPI (Sigma-Aldrich) in PBS (Life Technologies)/0.1% Triton X-100 (Carl Roth). Afterwards, cells were washed. Data were acquired via FACSCalibur (BD Biosciences) or Cytoflex S (Beckman Coulter) and analyzed using FlowJo Software (Tree Star). Total cell numbers of living cells were determined via trypan blue staining.

Approximately 1–2 × 107 bone marrow cells were resuspended in 0.7 ml RPMI 1640 media (Life Technologies) containing 5% FCS (PAN Biotech) and loaded with 0.7 μM indo-1-AM pluronic acid F-127 (Molecular Probes) by shaking for 25 min at 30°C. Afterwards, 0.7 ml RPMI 1640 media (Life Technologies) containing 10% FCS (PAN Biotech) was added, and cells were incubated by shaking for a further 10 min at 37°C. Cells were washed twice and stained with anti-B220 and anti-HSA as described above. Cells were washed and resuspended in Krebs–Ringer solution, and calcium mobilization was measured at LSR II (BD Biosciences). Basal Ca2+ level was determined for 50 s, then BCR was stimulated using anti-IgM [F(ab)2] (Jackson ImmunoResearch), and Ca2+ mobilization was measured up to 3 min. Indo-1 loading efficiency was determined by stimulation with ionomycin (Sigma-Aldrich). Obtained data were analyzed with FlowJo software (Tree Star). In case of calcium mobilization assays of DT40 cells, 5 × 106 cells were used without additional surface staining. BCR stimulation was performed with anti-chicken IgM (clone M4, kind gift from Prof. J. Wienands, University of Göttingen).

Bone marrow cells from CUL3fl/fl mb1CRE/+ and CUL3fl/fl mb1+/+ mice were isolated as described above. Living pro-B cells were sorted (B220pos c-kitpos DAPIneg), and 25,000 pro-B cells per well were plated into a 96-well plate in RPMI 1640 supplemented with 5% FCS, 1,2 mM l-glutamine, 50 μM 2-ME, 100 U/ml penicillin/streptomycin (Pen/Strep), 1 mM sodium pyruvate, and 1× nonessential amino acids (Life Technologies). Cells were either treated with 10 ng/ml recombinant mouse IL-7 (BioLegend) or left untreated and cultivated for 6 d at 37°C and 5% CO2. Three to five technical replicates were analyzed per condition. Each day, living cells were manually counted using trypan blue (Life Technologies) staining and Neubauer chamber. At day 4, an additional flow cytometry staining with anti-B220, anti-IgM, anti-CD25, and anti-CD117/anti-c-kit was performed to determine the percentage of pro-B cells.

To isolate splenic B2 cells, single-cell suspensions were erythrocyte depleted using 1× ACK solution and cells were incubated with supernatants of anti-CD4, anti-CD8, and anti-Thy1 hybridomas (generated in our house) for 30 min at 4°C. Afterwards, T cells were depleted via incubation of cells with baby-rabbit complement (Cedarlane) at 37°C for 45 min.

B cells were isolated from the spleen as described above and resuspended in RPMI 1640 supplemented with 5% FCS, 1,2 mM l-glutamine, 50 μM 2-ME, 100 U/ml Pen/Strep, 1 mM sodium pyruvate, and 1× nonessential amino acids. Cells were split in two cell culture dishes and either treated with 1 μM cullin inhibitor MLN4924 (Enzo) or DMSO as solvent control. MLN4924 is a specific small-molecule inhibitor of NEDD8-activating enzyme E1 and acts by covalent binding to NEDD8. Therefore, the covalent attachment of NEDD8 to members of the Cullin family, which is crucial for their ubiquitin ligase activity, is prevented (54). Further tests showed no difference in the percentage of living cells between MLN4924 treated and solvent control cells (Supplemental Fig. 1A). Cells were incubated (5% CO2, 37°C) for 10 h. Afterwards, 1.4 × 107 cells per condition were washed with PBS and resuspended in 0.35 ml RPMI 1640 containing 10% FCS. Ten micromolar bromoenol lactone (BEL; Cayman Chemical) was added to both conditions, an additional 1 μM MLN4924 was added to the overnight inhibitor-treated sample, and cells were incubated on ice for 10 min. BCR stimulation was introduced by adding 10 μg/ml anti-IgM [F(ab)2] (Jackson ImmunoResearch). Cells were fixed using 2% PFA (Carl Roth) in PBS at different time points and stained with anti-B220, anti-CD22 and anti-κ–L chain as mentioned above. Data were acquired via FACSCalibur (BD Biosciences) and analyzed using FlowJo Software (Treestar).

To generate the CD22-TST-DT40 cell line, chicken DT40 cells (immature B cell line) were transfected by electroporation (260 V) with 10–15 μg pcDNA3.1zeo plasmid containing murine CD22 with a C-terminally fused Twin-Strep-tag (55). Wild-type (WT) DT40 cells, which do not express CD22, were used as control. Cells were cultured at 37°C, 5% CO2 in RPMI 1640 supplemented with 10% FCS (PAN Biotech), 1% chicken serum (Sigma-Aldrich), 2 mM l-glutamine (Life Technologies), 50 μM 2-ME (Life Technologies), 100 U/ml Pen/Strep (Life Technologies), and 0.3 mg/ml Zeocin (only for transfected cells; Thermo Scientific). CD22-TST-WEHI-231 cells were generated by retroviral transduction using Plat-E packaging cell line (56) and pMSCV-puro plasmid containing murine CD22 with a C-terminally fused Twin-Strep-tag (55). Cells were cultured at 37°C, 5% CO2 in RPMI 1640 supplemented with 10% FCS (PAN Biotech), 2 mM l-glutamine (Life Technologies), 50 μM 2-ME (Life Technologies), 100 U/ml Pen/Strep (Life Technologies), and 0.5 μg/ml Puromycin (Thermo Scientific).

Sample preparation.

CD22-TST-DT40 and WT DT40 cells were metabolically labeled using the stable isotope labeling by/with amino acids in cell culture (SILAC) method (57). For this, cells were cultivated for 10 d in SILAC RPMI 1640 (Life Technologies) supplemented with 10% dialyzed FCS (PAN Biotech), 2 mM l-glutamine, 50 μM 2-ME, 100 U/ml Pen/Strep, 200 mg/l l-arginine, and 40 mg/l l-lysine. Arginine and lysine were either supplemented as natural isotopes (light) or heavy amino acid isotopes (13C615N4-arginine, 13C6-lysine; heavy). Light and heavy labeling was switched for CD22-TST-DT40 and WT DT40 cells in the different experiments. A total of 1 × 108 cells of each CD22-TST-DT40 and WT DT40 were starved for 1 h, washed twice with PBS (Life Technologies), and resuspended in PBS. If applicable, BCR was stimulated with 2 μg/ml anti-chicken IgM (clone M4, kind gift from Prof. J. Wienands, University of Göttingen) for 5 min at 37°C. Afterwards, cells were pelleted by centrifugation (1300 rpm, 5 min, 4°C) and lysed in ice-cold lysis buffer (138 mM NaCl, 50 mM Tris [pH 7.8], 10% glycerine, 1 mM sodium-orthovanadate, 0.5 mM EDTA [pH 8], 2% NP40; freshly supplemented with leupeptin, sodium-orthovanadate, PSMF, and aprotinin) for 30 min on ice. CD22-Twin-Strep-tag and bound proteins were purified from cell lysate via Twin-Strep-tag using 180 μl Strep-Tactin Type3 XT bead suspension (IBA Lifesciences). The enrichment was performed according to the manufacturer’s instructions, and proteins were eluted in 12 μl Rotiload (Carl Roth) at 96°C. Eluates of the samples to be compared (e.g., unstimulated CD22-TST-DT40 and unstimulated WT DT40) were combined and separated via SDS-PAGE. Colloidal Coomassie staining of the proteins was performed with PageBlue protein staining solution (Thermo Scientific).

In-gel digestion and peptide extraction.

Each gel lane was excised and cut into 9–10 gel slices. Slices were washed three consecutive times with 10 mM ammonium bicarbonate (ABC) and 5 mM ABC/50% EtOH (v/v). Protein reduction and alkylation was performed with 10 mM DTT at 56°C for 30 min, followed by incubation with 50 mM iodoacetamide at room temperature for 30 min in the dark. Gel slices were then successively washed and dehydrated with 10 mM ACN followed by 100% EtOH three times each. Slices were dried in vacuo for complete dehydration. Tryptic digestion (sequencing grade, 1:50 enzyme to protein ratio; Promega) was performed overnight in 10 mM ABC at 37°C. For peptide extraction, two consecutive times 0.05% (v/v) trifluoroacetic acid in 50% (v/v) acetonitrile were added to each slice and incubated in an ice-cold ultrasonic bath for 10 min. Extracted peptides were dried in vacuo and stored at −80°C until liquid chromatography–mass spectrometry (LC-MS) analysis.

LC-MS analysis.

LC-MS analyses were performed with an Ultimate 3000 RCLCnano HPLC system coupled online to a QExactive Plus mass spectrometer (both Thermo Scientific). The ultrahigh performance liquid chromatography system was equipped with two C18 μ-precolumns (Ø 0.3 × 5 mm; PepMap, Thermo Fisher Scientific) and an Acclaim PepMap analytical column (Ø 75 μm × 500 mm, 2 μm, 100 Å; Dionex LC Packings/Thermo Fisher Scientific). Resuspended peptides (0.1% trifluoroacetic acid, v/v) were loaded and washed for 5 min on a precolumn. Peptide separation was performed at 40°C and a flow rate of 250 nl/min. Peptides were separated using a binary solvent system consisting of solvent A (2% DMSO [v/v] /0.1% formic acid [v/v]) and solvent B (86% acetonitrile [ACN] [v/v], 2% DMSO [v/v], 0.1% formic acid [v/v]). A linear gradient was applied from 3 to 39% solvent B in 30 min. Afterwards, the gradient increased from 39 to 95% solvent B in 5 min and was kept constant at 95% for 5 min. Subsequently, the system was re-equilibrated at 3% solvent B for 14 min.

The QExactive Plus instrument was operated in the positive mode and externally calibrated using standard compounds. Peptides were ionized with a Nanospray Flex ion source (Thermo Scientific) and either stainless steel (Thermo Scientific) or fused silica emitters (New Objective) with a spray voltage of 1.8 and 1.5 kV, respectively. The temperature of the transfer capillary was set to 200°C. Full scans were measured in a scan range between 375 and 1700 m/z and with a resolution of 70,000 (at 200 m/z). The automatic gain control target was set to 3 × 106 ions with a maximum ion collection time of 60 ms. For tandem MS measurements, the top 12 most intense ion signals exceeding the intensity threshold of 5.8 × 103 were selected and subjected to higher-energy collisional dissociation fragmentation with a normalized collision energy of 28. Fragment spectra were measured with a resolution of 35,000 (at 200 m/z), an automatic gain control target of 1 × 105 ions, and a maximum ion collection time of 120 ms. Unassigned and singly charged ions were excluded from fragmentation. The dynamic exclusion time for previously fragmented precursor ions was set to 45 s.

Bioinformatics data analysis.

The Andromeda/MaxQuant software pipeline (version 1.5.3.30) was used for LC-MS raw data analysis (58, 59). The UniPort ProteomeSet of Gallus Gallus including isoforms (17,902 entries, downloaded in February 2015) was extended by the three mouse CD22 isoforms. This database and a set of common contaminations provided by MaxQuant were used to generate peak lists from tandem MS spectra. The MaxQuant database search was performed with the following parameters: sample multiplicity two (Arg10 and Lys8 as heavy labels); tryptic specificity allowing up to three missed cleavages; acetylation of the N termini and oxidation of methionine as variable modifications; carbamidomethylation of cysteine as fixed modification; mass tolerances of 20 ppm for first search and 4.5 ppm for the main search of precursor ions. For identification, a false discovery rate of 1% was applied with a maximal peptide mass of 5600 Da, and a minimum of one unique peptide was required for protein identification. For quantification, the “re-quantify” and “match between runs” options were enabled, and a minimal ratio count of two unique peptides was selected. Perseus (version 1.5.2.6) was used for statistical analysis of MaxQuant data from six biological replicates. Entries identified only by site, reverse, or potential contaminants were removed. Data were further filtered for at least three valid values, normalized SILAC ratios from label-switch experiments were inverted, and log2-transformed mean ratios were calculated. A one sample t test was applied and p values were calculated. Proteins with a p value < 0.05 and a mean log2 ratio greater than 1, 2, or 4 were considered as moderately, significantly, or highly significantly enriched.

First, Twin-Strep-tag affinity purification (see above) or anti-CD22 immunoprecipitation with rabbit anti-CD22 (polyclonal; Pineda) as described before (60) was carried out. Proteins were separated via SDS-PAGE and transferred to nitrocellulose membrane using Trans-Blot Turbo Transfer System (Bio-Rad). In the case of ubiquitin analysis, the nitrocellulose membrane was denatured after protein transfer by incubation for 30 min at 4°C in denaturation buffer (6 M guanidine, 5 mM 2-ME, 1 mM PMSF, 20 mM Tris [pH 7.5]). For analysis, membranes were incubated with one of the following Abs: anti-CD22 (polyclonal; Pineda), anti–cullin 3 (polyclonal C-18 or monoclonal G-8; Santa Cruz Biotechnology), anti-ubiquitin (FK2; Enzo Life Sciences), and anti–Twin-Strep-tag (StrepMAB-Classic, Strep-tagII specific, IBA Lifesciences). If necessary, incubation with HRP-conjugated Abs was performed using anti-rabbit-IgG HRP (Cell Signaling Technology), anti-goat-IgG HRP (Cell Signaling Technology) or anti-mouse IgG HRP (Cell Signaling Technology). Proteins were detected with SuperSignal West Pico PLUS Chemiluminescent Substrate (Thermo Scientific).

CD22-TST-DT40 or CD22-TST-WEHI-231 cells were cultured in DT40 media or WEHI-231 media with 1 μM cullin inhibitor MLN4924 (Enzo Life Sciences) or solvent control for 12–14 h (5% CO2, 37°C). In addition, 10 μM MG-132 and 20 mM NH4Cl were added for 4 h to block proteasome and lysosome activity. A total of 2 × 107 cells were harvested and lysed in ice-cold lysis buffer (138 mM NaCl, 50 mM Tris [pH 7.8], 10% glycerin, 1 mM sodium-orthovanadate, 0.5 mM EDTA pH 8, 1% Triton X-100; freshly supplemented with leupeptin, sodium-orthovanadate, PMSF, aprotinin, iodoacetamide, and N-ethylmaleimide) for 30 min on ice. Twin-Strep-tag purification of CD22 was preformed and samples were analyzed via immunoblotting (both as described above).

ELISA was used to determine total titer of different Ig isotypes in sera of naive mice. Polysorb plates (Nunc) were coated with 10 μg/ml isotypic-specific unlabeled Abs (anti-IgM, anti-IgG1, anti-IgG2b, anti-IgG2c, anti-IgG3, and anti-IgA; Southern Biotech) diluted in PBS and incubated overnight at 4°C. The next day, plates were blocked with 1% BSA in PBS for 2 h at 37°C. Afterwards, plates were washed three times with PBS. Starting dilution of 1:100–1:200 were chosen for serum samples, followed by a serial dilution of 1:3 in PBS containing 0.1% BSA. mIg Abs of a known concentration (Southern Biotech) served as standards. Serum samples were incubated overnight at 4°C on Polysorb plates. The next day, plates were washed three times with PBS and incubated with isotype specific alkaline phosohatase (AP) conjugated detection Abs (Southern Biotech) for 2 h at 37°C. By addition of the substrate for AP (4-nitrophenyl phosphate disodium salt hexahydrate tablet [Sigma-Aldrich]) dissolved in 20 ml 9.7% solution of 2,2’-iminodiethanol in H2O and 0.5 mM MgCl2 (pH 9.8), binding of AP-conjugated Abs was detected. The OD was measured by ELISA reader (Tecan) at 405 nm and data were analyzed using Soft Max Pro (Molecular Devices).

Murine splenic B cells were isolated via MACS using CD19 microbeads (Miltenyi Biotec) and incubated with anti-CD22-PE (Cy34.1; BD Biosciences) for 1 h at 4°C. After washing, endocytosis was induced by incubating the cells at 37°C for different periods of time. Samples were then washed with 0.2 M glycine/HCl (pH 2.5) to remove extracellular bound Abs and DAPI for dead cell exclusion. Cells were fixed using 2% PFA (Carl Roth) in PBS and analyzed on an LSR II (BD Biosciences). Only intracellular anti-CD22 Ab could be detected. At time point 0 min, an additional sample was prepared without washing off extracellular Abs and was then used as extracellular staining control. The mean fluorescence intensity (MFI) of this sample was set as 100%.

Statistical analyses were performed using GraphPad Prism software. Unpaired t test or Mann–Whitney U test was used to evaluate significance. Statistical data are presented as mean ± SD.

Mass spectrometric data deposition.

All raw data and original MaxQuant result files have been deposited to the ProteomeXchange Consortium (http://proteomecentral.proteomexchange.org) via the Proteomics Identification Database Partner Repository (61) with the dataset identifier PXD013801.

The immature chicken B cell line DT40 (6264) was used to study the CD22 interactome. These cells were chosen because they do not constitutively express any endogenous Siglec isoforms and can be easily transfected using electroporation. Therefore, DT40 cells were transfected with murine CD22 that is C-terminally fused to a Twin-Strep-tag. Afterwards, the correct functioning of murine CD22 in the chicken B cell line was confirmed (Supplemental Fig. 1B–D). We could show that CD22 is able to inhibit BCR-induced Ca2+ signaling (Supplemental Fig. 1B), can bind to α2,6-linked sialic acids on the cell surface (Supplemental Fig. 1C), and is expressed at levels comparable to murine splenic B cells (Supplemental Fig. 1D). In our experimental setup, SILAC DT40 cells were used in their WT form (control) or as stably expressing CD22-Twin-Strep-tag cells. To induce BCR signaling, cells were stimulated with anti-chicken-IgM. Subsequently, the CD22 interactome was analyzed from unstimulated and anti-IgM–stimulated SILAC DT40 cells using quantitative affinity purification–mass spectrometry.

Based on the acquired data, mean log2 SILAC ratios of proteins were calculated and plotted against −log10p values. Proteins exhibiting a mean log2 ratio ≥1 and a p value ˂ 0.05 (n = 6) were deemed significantly enriched with CD22. For unstimulated cells, we found 36 proteins to be enriched with CD22, whereas 64 proteins were enriched with CD22 upon BCR stimulation (Fig. 1A, Supplemental Table I). We were able to confirm already known CD22 interacting proteins, which associate with CD22 upon BCR stimulation. Thus SHP-1, parts of the BCR (including Ig-β, Ig–μ-chain, and Ig–λ-chain), and signaling proteins such as Syk, Grb2, and CD45 were highly enriched following anti-IgM stimulation. Two of the BCR stimulation–induced CD22 interaction partners (i.e., SHP-1 and Ig-β) were also identified in the resting state, suggesting already a weak interaction of CD22 with Ig-β and SHP-1 under unstimulated conditions (mean log2 ratios: Ig-β = 1.69 and SHP-1 = 2.03). However, in BCR-stimulated cells, the association of Ig-β and SHP-1 with CD22 was considerably higher, as shown by the increase of their enrichment factors (mean log2 ratios: Ig-β = 4.66 and SHP-1 = 3.53). Thus, our data confirm previous findings that CD22 is recruited to the BCR signaling complex after anti-IgM stimulation but is only weak associated to the BCR in the resting state (12, 6567). In addition to known CD22-interacting proteins, various new potential CD22 interaction partners including cullin 3, clathrin, BCAP, SERCA2, and Dok3 were identified in this analysis (Fig. 1A). Here, Dok3 and SERCA2 seem to interact with CD22 independent of the cell’s activation status, as they were associated with CD22 regardless of the stimulation condition. However, the majority of so far unreported proteins associated with CD22 were only found after anti-IgM treatment. To further investigate CD22 signaling-dependent mechanisms in B lymphocytes, we decided to examine cullin 3 as novel potential CD22 interaction partner after BCR stimulation.

FIGURE 1.

Cullin 3 is a CD22-interacting protein after BCR stimulation and is important for CD22 ubiquitination. (A) SILAC-based quantitative interactome analysis identifies cullin 3 as a CD22-interacting protein in CD22-TST-DT40 cells upon BCR stimulation. A total of 1 × 108 SILAC CD22-TST-DT40 and WT DT40 cells were analyzed either unstimulated or treated for 5 min with anti-IgM. Twin-Strep-tag affinity purification of CD22 was performed and eluates were analyzed by LC-MS. Volcano plots show the data of six biological replicates per condition. Mean log2 ratios of proteins were plotted against -log10p values. Proteins with a mean log2 ratio of 1 and a p value < 0.05 were deemed CD22 interacting proteins (gray area). Proteins are highlighted according to their mean log2 ratios used as a measure for their level of enrichment with CD22: orange, 2–4-fold; red, 4–16-fold; dark red, 16–64-fold. Blue, known CD22 interaction partners; black, new potential interaction partners of CD22. (B) CD22-TST-DT40 and WT DT40 cells were stimulated with anti-IgM for the indicated time points. Twin-Strep-tag affinity purification of CD22 was performed followed by anti-CD22 and anti-cullin 3 Western blot with specific Abs. TST, Twin-Strep-tag. (C) Splenic B cells from CUL3fl/fl mb1+/+ (control) and CUL3fl/fl mb1CRE/+ mice were isolated via CD19pos MACS isolation and lysed using 2% NP40 lysis buffer. Anti–cullin 3 and anti-actin Western blots with specific Abs were performed. (D) Splenic B cells from WT and CD22−/− mice were stimulated with anti-IgM [F(ab′)2] for the indicated time points. Anti- CD22 immunoprecipitation was performed followed by anti-CD22 and anti–cullin 3 Western blot with specific Abs. (E) Ubiquitination of CD22 is reduced after cullin inhibition. CD22-TST-DT40 cells were treated with 1 μM cullin inhibitor MLN4924 or DMSO for 10 and 4 h with MG-132 and NH4Cl to block proteasomal and lysosomal degradation. Then cells were stimulated with anti-IgM for the indicated time points. Twin-Strep-tag affinity purification was performed followed by anti-CD22 and anti-ubiquitin Western blot with specific Abs. (F) Ubiquitination of CD22 is reduced after cullin inhibition. CD22-TST-WEHI-231 cells were treated with 1 μM cullin inhibitor MLN4924 or DMSO for 10 and 4 h with MG-132 and NH4Cl to block proteasomal and lysosomal degradation. Then cells were stimulated with anti-IgM for the indicated time points. Anti- CD22 immunoprecipitation was performed followed by anti-CD22 and anti-ubiquitin Western blot with specific Abs.

FIGURE 1.

Cullin 3 is a CD22-interacting protein after BCR stimulation and is important for CD22 ubiquitination. (A) SILAC-based quantitative interactome analysis identifies cullin 3 as a CD22-interacting protein in CD22-TST-DT40 cells upon BCR stimulation. A total of 1 × 108 SILAC CD22-TST-DT40 and WT DT40 cells were analyzed either unstimulated or treated for 5 min with anti-IgM. Twin-Strep-tag affinity purification of CD22 was performed and eluates were analyzed by LC-MS. Volcano plots show the data of six biological replicates per condition. Mean log2 ratios of proteins were plotted against -log10p values. Proteins with a mean log2 ratio of 1 and a p value < 0.05 were deemed CD22 interacting proteins (gray area). Proteins are highlighted according to their mean log2 ratios used as a measure for their level of enrichment with CD22: orange, 2–4-fold; red, 4–16-fold; dark red, 16–64-fold. Blue, known CD22 interaction partners; black, new potential interaction partners of CD22. (B) CD22-TST-DT40 and WT DT40 cells were stimulated with anti-IgM for the indicated time points. Twin-Strep-tag affinity purification of CD22 was performed followed by anti-CD22 and anti-cullin 3 Western blot with specific Abs. TST, Twin-Strep-tag. (C) Splenic B cells from CUL3fl/fl mb1+/+ (control) and CUL3fl/fl mb1CRE/+ mice were isolated via CD19pos MACS isolation and lysed using 2% NP40 lysis buffer. Anti–cullin 3 and anti-actin Western blots with specific Abs were performed. (D) Splenic B cells from WT and CD22−/− mice were stimulated with anti-IgM [F(ab′)2] for the indicated time points. Anti- CD22 immunoprecipitation was performed followed by anti-CD22 and anti–cullin 3 Western blot with specific Abs. (E) Ubiquitination of CD22 is reduced after cullin inhibition. CD22-TST-DT40 cells were treated with 1 μM cullin inhibitor MLN4924 or DMSO for 10 and 4 h with MG-132 and NH4Cl to block proteasomal and lysosomal degradation. Then cells were stimulated with anti-IgM for the indicated time points. Twin-Strep-tag affinity purification was performed followed by anti-CD22 and anti-ubiquitin Western blot with specific Abs. (F) Ubiquitination of CD22 is reduced after cullin inhibition. CD22-TST-WEHI-231 cells were treated with 1 μM cullin inhibitor MLN4924 or DMSO for 10 and 4 h with MG-132 and NH4Cl to block proteasomal and lysosomal degradation. Then cells were stimulated with anti-IgM for the indicated time points. Anti- CD22 immunoprecipitation was performed followed by anti-CD22 and anti-ubiquitin Western blot with specific Abs.

Close modal

Cullin 3, an E3 ubiquitin ligase, was found to be associated with CD22 exclusively after anti-IgM stimulation (Fig. 1A). To further validate this interaction in CD22-TST-DT40 cells, CD22-Twin-Strep-tag affinity purifications were performed followed by anti–Twin-Strep-tag and anti–cullin 3 immunoblotting (Fig. 1B). We included a total cell lysate from WT DT40 cells as a positive control for cullin 3 detection. The specificity of the cullin 3 Ab was furthermore proven by analyzing B cell lysates from control mice (CUL3fl/fl mb1+/+) compared with lysates from CUL3fl/fl mb1CRE/+ mice, which have a B cell–specific cullin 3 deletion (Fig. 1C). Following BCR stimulation of CD22-TST-DT40 cells, Western blot analyses support the results from the quantitative affinity purification–mass spectrometry experiments. The results show two characteristic cullin 3 isoform bands at ∼80 and 89 kDa corresponding to nonneddylated and neddylated cullin 3 (68). In unstimulated cells, mainly the lower band (80 kDa) of cullin 3 was detected. This band represents the nonneddylated and therefore inactive form of cullin 3 (68). Upon BCR, the neddylated cullin 3 form (89 kDa) was additionally precipitated with CD22. This observation was further confirmed by coimmunoprecipitation experiments from primary splenic WT B cells. Here, we were able to detect both cullin 3 isoforms (nonneddylated and neddylated) associated with CD22 only after BCR stimulation (Fig. 1D).

We proceeded to characterize the relationship between cullin 3 and CD22 to understand the molecular connection between these two proteins. As cullin 3 is an E3 ubiquitin ligase and a scaffold protein for CRL3, ubiquitination analyses of CD22 were conducted. Therefore we performed CD22 immunoprecipitation from BCR-stimulated CD22-TST-DT40 cells, followed by anti-ubiquitin Western blot. The experiments showed that cells that have been treated with a cullin inhibitor (MLN4924) hardly revealed a signal of ubiquitinated CD22 after BCR stimulation (Fig. 1E). In contrast, ubiquitin-specific bands with the molecular mass of CD22 (140 kDa), as well as a weak higher molecular size band, were observed in CD22-TST-DT40 solvent control cells after 5 min of anti-IgM stimulation (Fig. 1E). This signal seems to be reduced after 30 min of stimulation. A similar experimental approach was conducted to investigate CD22 ubiquitination after BCR activation in the murine B cell line WEHI-231. The results of CD22-Twin-Strep-tag–transfected WEHI-231 cells show a similar pattern (Fig. 1F). Cells treated with 1 μM MLN4924 did not reveal a band for ubiquitinated CD22, whereas DMSO control cells showed a clear signal up to 120 min after BCR stimulation.

Ubiquitination of surface receptors leads to internalization (34) and CD22 is known to internalize and recycle after anti-CD22 Ab ligation (20, 21, 69). Thus, it was first analyzed if CD22 internalization and recycling takes also place after indirect CD22 activation by BCR stimulation. Therefore the CD22 internalization and recycling capacity was analyzed using flow cytometry. Isolated splenic B cells from WT mice were treated with BEL to inhibit early endosomes and prevent internalized surface receptors from recycling back to the surface. BEL antagonizes Ca2+ independent PLA2 activity, which is important for endosomal tubule formation and thereby inhibits trafficking from sorting to recycling endosomes (70). As shown in Supplemental Fig. 1E, addition of BEL before anti-IgM stimulation results in a reduced CD22 surface expression with time (starting at 60 min after stimulation), demonstrating CD22 internalization. In comparison, solvent control WT cells did not reveal a decrease of CD22 surface expression after BCR stimulation, indicating that CD22 is slowly internalized after 60 min of IgM stimulation and recycled back to the surface afterward. This experiment was further expanded to study CD22 internalization after BCR stimulation with respect to cullin activity. Hence, splenic WT B cells were treated with BEL and additionally either cullin inhibitor MLN4924 or with a solvent control. The unchanged CD22 surface expression revealed that cullin inhibition abrogated CD22 internalization after BCR stimulation, (Fig. 2A, 2B), whereas control cells showed a significant reduction of CD22 MFI starting 60 min after anti-IgM treatment. CD22 internalization was further investigated in splenic B cells from CD22-Y2,5,6F mice (12). These cells harbor point mutations within the three cytosolic ITIMs and therefore also in the clathrin-dependent internalization motifs (21). Our observations show that these B lymphocytes behaved like cullin inhibitor–treated cells showing unchanged CD22 surface MFI (Fig. 2B). The internalization of the BCR itself, however, was not affected by cullin inhibition and showed a very fast internalization kinetic, as detected by reduced levels of the Ig κ MFI after stimulation (Fig. 2C), in comparison with CD22 levels. In addition, B220 MFI was analyzed and no significance difference between MLN4924 and DMSO control cells was observed (Fig. 2D). Moreover, B220 MFI stayed constant after BCR stimulation. As mentioned before, fast CD22 endocytosis can be achieved by ligating CD22 with an anti-CD22 Ab. This more direct way of activating CD22 endocytosis can be measured by incubating splenic B cells with a fluorochrome-conjugated anti-CD22 Ab. At different time points, nonendocytosed Ab is washed off, and the fluorescent signal from internalized Ab was measured via flow cytometry. As depicted in Supplemental Fig. 1F, inhibition of cullin did not modify CD22 endocytosis rate in this direct activation model.

FIGURE 2.

Cullin inhibition reduces CD22, but not BCR internalization after anti-IgM stimulation. (A) Gating strategy to analyze CD22 internalization of isolated splenic B cells via flow cytometry. B220pos lymphocytes were analyzed concerning CD22 MFI. (BD) Splenic WT or CD22-Y2,5,6F B cells were treated with 1 μM MLN492 or DMSO for 10 h and stimulated with anti-IgM [F(ab′)2] for the indicated time points. CD22 MFI (B), BCR-Ig κ MFI (C), or B220 MFI (D) was analyzed. Mean ± SD of 5–7 (B), 4–7 (C), or 7 (D) biological replicates is shown. *p < 0.05, **p < 0.01 (Mann–Whitney U test).

FIGURE 2.

Cullin inhibition reduces CD22, but not BCR internalization after anti-IgM stimulation. (A) Gating strategy to analyze CD22 internalization of isolated splenic B cells via flow cytometry. B220pos lymphocytes were analyzed concerning CD22 MFI. (BD) Splenic WT or CD22-Y2,5,6F B cells were treated with 1 μM MLN492 or DMSO for 10 h and stimulated with anti-IgM [F(ab′)2] for the indicated time points. CD22 MFI (B), BCR-Ig κ MFI (C), or B220 MFI (D) was analyzed. Mean ± SD of 5–7 (B), 4–7 (C), or 7 (D) biological replicates is shown. *p < 0.05, **p < 0.01 (Mann–Whitney U test).

Close modal

To further examine the role of cullin 3 in B lymphocytes, cullin 3flox/flox mice (51) were obtained and mated to mb1-cre mice to generate a B cell–specific knockout of cullin 3. First, B cell development in the bone marrow (Fig. 3A) and B cell subsets in spleen (Fig. 3B), lymph node, blood, and peritoneal cavity (Supplemental Fig. 2A) were analyzed via flow cytometry in these mice (summarized in Table I). In the bone marrow, we observed a severe reduction of the total B cell count in CUL3fl/fl mb1CRE/+ mice, starting from the pre-B cell stage (Fig. 3A). Peripheral B cell numbers in different lymphatic organs were lowered, too. This affected all subpopulations of B cells, conventional B2 cells, marginal zone B cells, and B1 cells (Fig. 3B). The remaining B cells in bone marrow and spleen were further analyzed regarding their CD22 expression levels. We were able to show that CUL3fl/fl mb1CRE/+ B cells express significantly higher amounts of CD22 on the cell surface during developmental stages in the bone marrow and spleen but also on mature splenic B cells (Fig. 3C). Furthermore, we found generally decreased Ig serum level in naive young CUL3fl/fl mb1CRE/+ mice (Fig. 3D).

FIGURE 3.

B cell–specific cullin 3 deficiency results in reduced B cell numbers, CD22 overexpression, and reduced CD22 internalization after anti-IgM stimulation. (A) Single-cell suspensions from bone marrow were stained with indicated Abs and analyzed by flow cytometry. Dot plots show gating strategy and bar chart shows total cell count of indicated cell population (12–14 mice per genotype). (B) Single-cell suspensions from spleen were stained with indicated Abs and analyzed by flow cytometry. Dot plots show gating strategy and bar chart shows total cell count of indicated cell population (11–14 mice per genotype). (C) CD22 expression was analyzed on indicated B cell populations from bone marrow and spleen single-cell solution by flow cytometry. Bar charts summarize CD22 MFI (11–14 mice per genotype). (D) Ig level in sera of naive mice (age 8–12 wk) were determined via ELISA. Lines display mean, each dot represents one mouse. (E) CD22 internalization after anti-IgM stimulation for indicated time points was analyzed by flow cytometry. Bar chart shows CD22 MFI of seven to nine biological replicates from at least five independent experiments. Data information: in (A)–(C) and (E), data are presented as mean ± SD, or in (D), as mean. **p < 0.01, ***p < 0.001 (Mann–Whitney U test). B1a, B1a cells; B2, conventional (B2) B cells; FO, follicular B cells; Imm., immature B cells; Mat. Rec., mature recirculating B cells; Mat., mature B cells; MZ, marginal zone B cells; Pre, Pre-B cells; Pro, Pro-B cells; T1, transitional B cells T1; T2, transitional B cells T2.

FIGURE 3.

B cell–specific cullin 3 deficiency results in reduced B cell numbers, CD22 overexpression, and reduced CD22 internalization after anti-IgM stimulation. (A) Single-cell suspensions from bone marrow were stained with indicated Abs and analyzed by flow cytometry. Dot plots show gating strategy and bar chart shows total cell count of indicated cell population (12–14 mice per genotype). (B) Single-cell suspensions from spleen were stained with indicated Abs and analyzed by flow cytometry. Dot plots show gating strategy and bar chart shows total cell count of indicated cell population (11–14 mice per genotype). (C) CD22 expression was analyzed on indicated B cell populations from bone marrow and spleen single-cell solution by flow cytometry. Bar charts summarize CD22 MFI (11–14 mice per genotype). (D) Ig level in sera of naive mice (age 8–12 wk) were determined via ELISA. Lines display mean, each dot represents one mouse. (E) CD22 internalization after anti-IgM stimulation for indicated time points was analyzed by flow cytometry. Bar chart shows CD22 MFI of seven to nine biological replicates from at least five independent experiments. Data information: in (A)–(C) and (E), data are presented as mean ± SD, or in (D), as mean. **p < 0.01, ***p < 0.001 (Mann–Whitney U test). B1a, B1a cells; B2, conventional (B2) B cells; FO, follicular B cells; Imm., immature B cells; Mat. Rec., mature recirculating B cells; Mat., mature B cells; MZ, marginal zone B cells; Pre, Pre-B cells; Pro, Pro-B cells; T1, transitional B cells T1; T2, transitional B cells T2.

Close modal
Table I.
Total cell count of lymphocyte populations in lymphatic organs of CUL3fl/fl mb1+/+ and CUL3fl/fl mb1CRE/+ mice
Cullin3fl/fl × mb1+/+Cullin3fl/fl × mb1CRE/+
Bone marrow (×105  
 Pro-B cells (B220pos, IgMneg, c –kitpos CD25neg1.54 ± 0.58 1.29 ± 0.45 
 Pre-B cells (B220pos, IgMneg, c –kitneg, CD25pos13.16 ± 3.48 2.85 ± 1.02*** 
 Immature B cells (B220pos, IgMpos5.78 ± 1.72 1.50 ± 0.61*** 
 Mature, recirculating B cells (B220pos, IgDhigh6.65 ± 3.03 0.62 ± 0.87*** 
Spleen (×105  
 Transitional B cells T1 (B220pos, CD21lo, IgMhi2.80 ± 1.54 0.08 ± 0.05*** 
 Transitional B cells T2 (B220pos, IgDhi, IgMhi1.59 ± 0.83 0.47 ± 0.21*** 
 Follicular, mature B cells (B220pos, CD23med, CD21lo24.38 ± 13.88 2.00 ± 1.01*** 
 Marginal zone B cells (B220pos, CD23lo, CD21hi1.87 ± 1.13 0.04 ± 0.07*** 
 B1a cells (B220lo, CD5lo0.38 ± 0.29 0.06 ± 0.04*** 
 T cells (B220neg, CD5hi27.88 ± 9.48 16.57 ± 4.84* 
 Peritoneal cavity (×105  
 B1a cells (B220lo, CD5lo2.05 ± 1.74 0.32 ± 0.54*** 
 B1b/B2 cells (B220hi, CD5neg3.00 ± 2.69 0.19 ± 0.17** 
 T cells (B220neg, CD5hi4.48 ± 3.67 4.88 ± 4.74 
Lymph node (×104  
 Transitional B cells T1 (B220pos, CD21lo, IgMhi0.64 ± 0.53 0.02 ± 0.01*** 
 Transitional B cells T2 (B220pos, IgDhi, IgMhi1.32 ± 1.07 0.19 ± 0.10*** 
 Mature B cells (B220pos, IgDhi, IgMmed10.34 ± 10.60 0.27 ± 0.17*** 
 B1 cells (B220lo, CD5lo1.35 ± 3.60 0.34 ± 1.00* 
 T cells (B220neg, CD5hi90.10 ± 58.49 94.12 ± 47.03 
Blood (×103  
 B1b/B2 cells (B220hi, CD5neg1.39 ± 1.13 0.04 ± 0.03*** 
 B1a cells (B220lo, CD5lo0.02 ± 0.03 0.00 ± 0.00*** 
Cullin3fl/fl × mb1+/+Cullin3fl/fl × mb1CRE/+
Bone marrow (×105  
 Pro-B cells (B220pos, IgMneg, c –kitpos CD25neg1.54 ± 0.58 1.29 ± 0.45 
 Pre-B cells (B220pos, IgMneg, c –kitneg, CD25pos13.16 ± 3.48 2.85 ± 1.02*** 
 Immature B cells (B220pos, IgMpos5.78 ± 1.72 1.50 ± 0.61*** 
 Mature, recirculating B cells (B220pos, IgDhigh6.65 ± 3.03 0.62 ± 0.87*** 
Spleen (×105  
 Transitional B cells T1 (B220pos, CD21lo, IgMhi2.80 ± 1.54 0.08 ± 0.05*** 
 Transitional B cells T2 (B220pos, IgDhi, IgMhi1.59 ± 0.83 0.47 ± 0.21*** 
 Follicular, mature B cells (B220pos, CD23med, CD21lo24.38 ± 13.88 2.00 ± 1.01*** 
 Marginal zone B cells (B220pos, CD23lo, CD21hi1.87 ± 1.13 0.04 ± 0.07*** 
 B1a cells (B220lo, CD5lo0.38 ± 0.29 0.06 ± 0.04*** 
 T cells (B220neg, CD5hi27.88 ± 9.48 16.57 ± 4.84* 
 Peritoneal cavity (×105  
 B1a cells (B220lo, CD5lo2.05 ± 1.74 0.32 ± 0.54*** 
 B1b/B2 cells (B220hi, CD5neg3.00 ± 2.69 0.19 ± 0.17** 
 T cells (B220neg, CD5hi4.48 ± 3.67 4.88 ± 4.74 
Lymph node (×104  
 Transitional B cells T1 (B220pos, CD21lo, IgMhi0.64 ± 0.53 0.02 ± 0.01*** 
 Transitional B cells T2 (B220pos, IgDhi, IgMhi1.32 ± 1.07 0.19 ± 0.10*** 
 Mature B cells (B220pos, IgDhi, IgMmed10.34 ± 10.60 0.27 ± 0.17*** 
 B1 cells (B220lo, CD5lo1.35 ± 3.60 0.34 ± 1.00* 
 T cells (B220neg, CD5hi90.10 ± 58.49 94.12 ± 47.03 
Blood (×103  
 B1b/B2 cells (B220hi, CD5neg1.39 ± 1.13 0.04 ± 0.03*** 
 B1a cells (B220lo, CD5lo0.02 ± 0.03 0.00 ± 0.00*** 

Total cell counts of B and T cell populations of young, naive mice were determined using indicated cell surface markers and correspond to one femur, whole spleen, peritoneal cavity, one lymph node, or 1 μl blood per analyzed animal. Presented data show mean ± SD of 9–14 mice per genotype. Significant differences are highlighted with bold letters. *p < 0.05, **p < 0.01, ***p < 0.001 (Mann–Whitney U test).

trans, transitional.

As KLHL6 was shown to be a substrate adaptor for cullin 3 (47, 71) and was identified in the quantitative CD22 interactome study upon BCR stimulation (Fig. 1A), B cell populations of KLHL6−/− mice were investigated regarding their CD22 expression level. These, however, did not show CD22 overexpression (Supplemental Fig. 2B), as observed in B cell–specific cullin 3 deficient mice (Fig. 3C). Furthermore, the overexpression phenotype of CD22 in CUL3fl/fl mb1CRE/+ mice is not observed for other B cell surface receptors, like CD19, Siglec-G, IgM, or IgD (or IgM/IgD ratio), as their expression pattern is mostly unaltered on bone marrow and splenic B cells of CUL3fl/fl mb1CRE/+ mice (Supplemental Fig. 3A, 3B). However, some receptors seem to be influenced by a cullin 3 deficiency. It turned out that besides CD22, CD21 and CD23 expression levels are also enhanced on follicular B cells in the spleen (Supplemental Fig. 3B).

As inhibition of cullin leads to altered CD22 internalization after BCR stimulation in murine splenic B cells from WT mice (Fig. 2B), this experiment was also applied to the CUL3fl/fl mb1CRE/+ strain. Because of the low numbers of remaining splenic B cells available, only two time points, unstimulated and 120 min anti-IgM stimulation, were analyzed (Fig. 3E). The results of CUL3fl/fl mb1CRE/+ B cells resemble MLN4924-treated WT B cells, as no significant CD22 MFI reduction was observed. In contrast, control B cells (CUL3WT/WT mb1CRE/+) showed a moderate, but highly significant, decrease of CD22 on the cell surface (Fig. 3E).

Subsequent experiments were conducted to analyze the impaired B cell development in CUL3fl/fl mb1CRE/+ mice. First, flow-sorted pro-B cells were cultured for 6 d with or without IL-7 treatment (Fig. 4A left). The addition of 10 ng/ml IL-7 resulted in proliferation of CUL3fl/fl mb1+/+ pro-B cells. Signaling via IL-7– IL-7-Rα is important for early B cell development in the bone marrow to ensure correct Ig gene rearrangement and differentiation from pro- to pre-B cells (72, 73). In contrast to WT controls, CUL3fl/fl mb1CRE/+ pro-B cells did not show this expansion in cell numbers, instead cell numbers decreased with time. Overall, we detected a significant defect in the proliferation capacity of CUL3fl/fl mb1CRE/+ pro-B cells. The kinetics of these cells resemble more the untreated samples. The percentages of pro-B cells at day 4 within the cultures show no major difference between CUL3fl/fl mb1CRE/+ and CUL3fl/fl mb1+/+, indicating no escape from proliferation by further differentiation (Fig. 4A right). Furthermore, the expression level of IL-7-Rα, which is the receptor to IL-7, was examined (Fig. 4B). However, no differences were detected in the IL-7-Rα expression on bone marrow B cells between CUL3fl/fl mb1CRE/+ and CUL3fl/fl mb1+/+ mice. Others were able to show a regulatory role of cullin 3 in mitotic cell cycle entry and delayed mitotic entry in absence of cullin 3 (74). Such a possible defect in the pro-B cell subset could contribute to the proliferation defect, that we observed. Therefore we were interested in cell cycle phase distribution of pro-B cells from CUL3fl/fl mb1CRE/+ mice. Our results demonstrate significant enrichment of CUL3fl/fl mb1CRE/+ pro-B cells in the G2/M cell cycle phase and a tendency of reduced percentage in G1 phase (Fig. 4D), indicating a dysregulated cell cycle in pro-B cells.

FIGURE 4.

Cullin 3–deficient B lineage cells show an early proliferation defect, an apoptotic phenotype, and altered expression levels of Fas and BAFF-R. (A) Proliferation of sorted pro-B cells (B220pos c-kitpos DAPIneg) was analyzed after IL-7 treatment (left). Twenty five thousand cells per well were either treated with 10 ng/ml mouse rIL-7 or left untreated and cultivated for 6 d. Each day, living cells were counted. The data show mean value of three to five technical replicates per condition. The experiment is representative for one out of three independent experiments. The bar chart (right) shows results from flow cytometry analysis at day 4 to determine the percentage of living pro-B cells (B220pos IgMneg c-kitpos CD25neg) in the culture (right). The data display mean values of three independent experiments with technical replicates each. (B) Single-cell suspensions from bone marrow were analyzed to determine the IL-7-Rα expression on pro (B220pos IgMneg c-kitpos CD25neg), pre (B220pos IgMneg CD25pos c-kitneg), immature (B220pos IgMpos), and mature recirculating (B220pos IgDpos) B cells by flow cytometry. Bar chart shows IL-7-Rα MFI (9–11 mice per genotype). (C) Single-cell suspensions from bone marrow were analyzed to determine the percentage of activeCaspase3pos pro and pre (B220pos IgMneg), immature (B220pos IgMpos) and mature recirculating (B220pos IgDpos) B cells by flow cytometry. Each dot represents one mouse. Data shows the result of four independent experiments. (D) Single-cell suspensions from bone marrow were analyzed by flow cytometry to determine the percentages of G1-, S-, and G2/M-phase pro-B cells (B220pos IgMneg c-kitpos CD25neg). The histogram (left) represents the gating strategy to set the gates for the different cell cycle stage according to the DNA content. Bar chart (right) shows percentage of G1-, S-, and G2/M-phase pro-B cells (B220pos IgMneg c-kitpos CD25neg). Each dot represents one mouse. Data shows the result of six independent experiments. (E) Single-cell suspensions from bone marrow were analyzed to determine the Fas receptor expression on bone marrow (left) and splenic (right) B cell populations by flow cytometry. Pro (B220pos IgMneg c-kitpos CD25neg), pre (B220pos IgMneg CD25pos c-kitneg), immature (B220pos IgMpos), and mature recirculating (B220pos IgDpos) B cells in the bone marrow as well as T1 (B220pos CD21lo IgMhi), T2 (B220pos IgDhi IgMhi), and mature (B220pos IgDpos IgMlo-med) B cells in the spleen were analyzed. Bar charts show Fas receptor MFI (9–11 and 5–6 mice per genotype for bone marrow and spleen). (F) Single-cell suspensions from bone marrow were analyzed to determine the BAFF-R expression on bone marrow (left) and splenic (right) B cell populations by flow cytometry. Pro (B220pos IgMneg c-kitpos CD25neg), pre (B220pos IgMneg CD25pos c-kitneg), immature (B220pos IgMpos), and mature recirculating (B220pos IgDpos) B cells in the bone marrow as well as T1 (B220pos CD21lo IgMhi), T2 (B220pos IgDhi IgMhi), and mature (B220pos IgDpos IgMlo-med) B cells in the spleen were analyzed. Bar charts show BAFF-R MFI (9–11 and 5–6 mice per genotype for bone marrow and spleen). In (A)–(F), data are presented as mean ± SD. *p < 0.05, **p < 0.01, ***p < 0.001 (Mann–Whitney U test).

FIGURE 4.

Cullin 3–deficient B lineage cells show an early proliferation defect, an apoptotic phenotype, and altered expression levels of Fas and BAFF-R. (A) Proliferation of sorted pro-B cells (B220pos c-kitpos DAPIneg) was analyzed after IL-7 treatment (left). Twenty five thousand cells per well were either treated with 10 ng/ml mouse rIL-7 or left untreated and cultivated for 6 d. Each day, living cells were counted. The data show mean value of three to five technical replicates per condition. The experiment is representative for one out of three independent experiments. The bar chart (right) shows results from flow cytometry analysis at day 4 to determine the percentage of living pro-B cells (B220pos IgMneg c-kitpos CD25neg) in the culture (right). The data display mean values of three independent experiments with technical replicates each. (B) Single-cell suspensions from bone marrow were analyzed to determine the IL-7-Rα expression on pro (B220pos IgMneg c-kitpos CD25neg), pre (B220pos IgMneg CD25pos c-kitneg), immature (B220pos IgMpos), and mature recirculating (B220pos IgDpos) B cells by flow cytometry. Bar chart shows IL-7-Rα MFI (9–11 mice per genotype). (C) Single-cell suspensions from bone marrow were analyzed to determine the percentage of activeCaspase3pos pro and pre (B220pos IgMneg), immature (B220pos IgMpos) and mature recirculating (B220pos IgDpos) B cells by flow cytometry. Each dot represents one mouse. Data shows the result of four independent experiments. (D) Single-cell suspensions from bone marrow were analyzed by flow cytometry to determine the percentages of G1-, S-, and G2/M-phase pro-B cells (B220pos IgMneg c-kitpos CD25neg). The histogram (left) represents the gating strategy to set the gates for the different cell cycle stage according to the DNA content. Bar chart (right) shows percentage of G1-, S-, and G2/M-phase pro-B cells (B220pos IgMneg c-kitpos CD25neg). Each dot represents one mouse. Data shows the result of six independent experiments. (E) Single-cell suspensions from bone marrow were analyzed to determine the Fas receptor expression on bone marrow (left) and splenic (right) B cell populations by flow cytometry. Pro (B220pos IgMneg c-kitpos CD25neg), pre (B220pos IgMneg CD25pos c-kitneg), immature (B220pos IgMpos), and mature recirculating (B220pos IgDpos) B cells in the bone marrow as well as T1 (B220pos CD21lo IgMhi), T2 (B220pos IgDhi IgMhi), and mature (B220pos IgDpos IgMlo-med) B cells in the spleen were analyzed. Bar charts show Fas receptor MFI (9–11 and 5–6 mice per genotype for bone marrow and spleen). (F) Single-cell suspensions from bone marrow were analyzed to determine the BAFF-R expression on bone marrow (left) and splenic (right) B cell populations by flow cytometry. Pro (B220pos IgMneg c-kitpos CD25neg), pre (B220pos IgMneg CD25pos c-kitneg), immature (B220pos IgMpos), and mature recirculating (B220pos IgDpos) B cells in the bone marrow as well as T1 (B220pos CD21lo IgMhi), T2 (B220pos IgDhi IgMhi), and mature (B220pos IgDpos IgMlo-med) B cells in the spleen were analyzed. Bar charts show BAFF-R MFI (9–11 and 5–6 mice per genotype for bone marrow and spleen). In (A)–(F), data are presented as mean ± SD. *p < 0.05, **p < 0.01, ***p < 0.001 (Mann–Whitney U test).

Close modal

Additional analyses focused on investigating indications for ongoing apoptosis in cullin 3-deficient B cells, which would also explain the observed phenotype. Therefore, intracellular staining of active caspase3, which is an indicator of ongoing apoptosis, was performed in bone marrow B cells and analyzed via flow cytometry (Fig. 4C). Early developmental B cell stages showed a clear tendency of more active caspase3 and mature recirculating CUL3fl/fl mb1CRE/+ B cells exhibited a significantly stronger caspase3 activation compared with control cells. Further experiments, regarding the expression level of BAFF-R and Fas-R, additional receptors critical for survival and apoptosis regulation, were performed to further analyze the mechanism of the developmental defect of B cells in CUL3fl/fl mb1CRE/+ mice. Thus, flow cytometry analysis of bone marrow and splenic B cell populations were conducted. Developing B cells in the bone marrow did not reveal a higher Fas-R expression; this only applied to the mature recirculating B cell pool (Fig. 4E, Supplemental Fig. 3C). However, splenic subpopulations, beginning at transitional T2 stage, had an enhanced Fas-R surface expression (Fig. 4E, Supplemental Fig. 3C). Furthermore, immature CUL3fl/fl mb1CRE/+ B cells in the bone marrow showed a reduced BAFF-R expression (Fig. 4F, Supplemental Fig. 3C). This observation was also confirmed in the splenic B cell populations (Fig. 4F, Supplemental Fig. 3C). In summary, pro-B cells display an proliferation defect with a tendency to more apoptosis induction and accumulation in the G2/M cell cycle phase cells. Higher Fas-R and reduced BAFF-R expression, however, seemed to mainly affect the more mature stages of B cell development in bone marrow and all B cell populations in the spleen, which could have direct consequences for B cell survival.

As CD22 is regulating B cell activation (59), it was important to study B cell activation status in cullin 3–deficient B lymphocytes, which among other phenotypes show an enhanced CD22 expression. To study basal and BCR-induced activation level, calcium mobilization assays of pre-B/immature B cells from the bone marrow were performed. After determining basal cellular calcium concentration for 50 s, anti-IgM stimulation was applied, and the changes in calcium concentration were measured for additional 150 s. The result showed reproducible higher basal calcium level (measured as indo-1 ratio of Ca2+ bound to Ca2+ unbound dye) before BCR stimulation in CUL3fl/fl mb1CRE/+ mice (Fig. 5A). Additionally it was observed that upon anti-IgM treatment in different concentrations, the CUL3fl/fl mb1CRE/+ B cells were not able to reach the cytosolic calcium level of control cells. This preactivation was then further analyzed on splenic B cells by staining of typical activation markers. The experiments revealed a higher expression level of the activation marker MHCII and CD86 (Fig. 5B, 5C). Additional experiments showed that proteins of the BCR signaling cascade, such as PLCγ2 and BTK, also have a higher basal activation, which increased even further after BCR stimulation (Fig. 5D, 5E).

FIGURE 5.

Cullin 3–deficient B cells show a preactivated phenotype and impaired Ca2+ signaling. (A) Immature and pre-B cells (B220pos HSApos) from bone marrow from indicated genotype were loaded with Indo-1. Ca2+ baseline was measured under unstimulated conditions for 50 s, afterward BCR was stimulated with anti-IgM [F(ab′)2] in indicated concentrations. Ca2+ mobilization was continued to 3 min. Data shown are representative of five individual experiments with five mice per genotype. (B) Splenic B220pos cells of indicated genotype were analyzed regarding to activation marker expression. Histograms show CD86 (left) and MHCII (right) expression from one representative experiment. (C) Bar charts summarize CD86 MFI (left) and MHCII MFI (right) of splenic B220pos cells of 9–11 mice per genotype. (D) B220pos cells from spleen were analyzed with phosphospecific Abs for BTK and PLCγ to determine the phosphorylation level of the indicated protein with and without BCR stimulation. Histograms show p-BTK (upper) and p-PLCγ (lower) of the indicated genotypes from one representative experiment. (E) Bar charts summarize p-BTK MFI (upper) and p-PLCγ MFI (lower) of the indicated genotypes with and without BCR stimulation. Data are generated from five mice per genotype. In (C) and (E), data are presented as mean ± SD. *p < 0.05, **p < 0.01, ***p < 0.001 (Mann–Whitney U test).

FIGURE 5.

Cullin 3–deficient B cells show a preactivated phenotype and impaired Ca2+ signaling. (A) Immature and pre-B cells (B220pos HSApos) from bone marrow from indicated genotype were loaded with Indo-1. Ca2+ baseline was measured under unstimulated conditions for 50 s, afterward BCR was stimulated with anti-IgM [F(ab′)2] in indicated concentrations. Ca2+ mobilization was continued to 3 min. Data shown are representative of five individual experiments with five mice per genotype. (B) Splenic B220pos cells of indicated genotype were analyzed regarding to activation marker expression. Histograms show CD86 (left) and MHCII (right) expression from one representative experiment. (C) Bar charts summarize CD86 MFI (left) and MHCII MFI (right) of splenic B220pos cells of 9–11 mice per genotype. (D) B220pos cells from spleen were analyzed with phosphospecific Abs for BTK and PLCγ to determine the phosphorylation level of the indicated protein with and without BCR stimulation. Histograms show p-BTK (upper) and p-PLCγ (lower) of the indicated genotypes from one representative experiment. (E) Bar charts summarize p-BTK MFI (upper) and p-PLCγ MFI (lower) of the indicated genotypes with and without BCR stimulation. Data are generated from five mice per genotype. In (C) and (E), data are presented as mean ± SD. *p < 0.05, **p < 0.01, ***p < 0.001 (Mann–Whitney U test).

Close modal
FIGURE 6.

Model of cullin 3–dependent CD22 internalization after BCR stimulation. One to thirty minutes after BCR activation, CD22 is quickly phosphorylated. Within the first minutes, CRL3 ubiquitinates CD22, which is a sign for receptor internalization. Y843 and Y863 (part of the clathrin-dependent internalization motif) are phosphorylated and prevent AP50 binding. From 30 min after BCR stimulation, CD22 phosphorylation declines. Clathrin-associated adaptor complex binds via AP50 to Y843 or Y863 and CD22 is internalized. Receptor recycling is facilitated by deubiquitination in the early endosome.

FIGURE 6.

Model of cullin 3–dependent CD22 internalization after BCR stimulation. One to thirty minutes after BCR activation, CD22 is quickly phosphorylated. Within the first minutes, CRL3 ubiquitinates CD22, which is a sign for receptor internalization. Y843 and Y863 (part of the clathrin-dependent internalization motif) are phosphorylated and prevent AP50 binding. From 30 min after BCR stimulation, CD22 phosphorylation declines. Clathrin-associated adaptor complex binds via AP50 to Y843 or Y863 and CD22 is internalized. Receptor recycling is facilitated by deubiquitination in the early endosome.

Close modal

In this study, we performed a global CD22 interactome analysis to identify CD22 interacting proteins in BCR unstimulated and stimulated cells. We identified various new CD22 interacting proteins that have not been described in the context of CD22 so far. Among those, we focused on investigating the connection between CD22 and cullin 3, an E3 ubiquitin ligase. E3 ubiquitin ligases are involved in regulating membrane receptor expression via ubiquitination followed by internalization (34, 48, 75). This process is mediated in either a clathrin-dependent or a clathrin-independent way (7679). Our study revealed that after anti-IgM stimulation, CD22 is ubiquitinated in a cullin-dependent manner and later on internalized and recycled to the surface. We think that cullin 3, which was identified as CD22 interaction partner in our screening, is the cullin protein promoting CD22 ubiquitination. However, as we used a global cullin inhibitor for our functional ubiquitination assays, we cannot exclude the involvement of other cullin family members. In our analysis, we also observed some differences in CD22 ubiquitination kinetics between CD22-Twin-Strep-tag–transfected cell lines from chicken and mouse origin. Both systems, however, show a cullin-dependent CD22 ubiquitination in response to BCR activation, which is a new important finding in the field of Siglec biology.

Subsequent analyses focused on revealing the downstream effects of CD22 ubiquitination and led to our model (Fig. 6). We propose that upon BCR stimulation, CD22 is activated within the first minutes of B cell activation through tyrosine phosphorylation by kinases. Thereby a CD22 mediated signaling cascade is initiated. CD22 is then ubiquitinated presumably by CRL3, peaking at 120 min in the murine system (earlier in DT40 cells). However, CD22 stays on the surface for some time and is not endocytosed right away. This correlates with our observation of a delay in clathrin-dependent internalization of CD22. This process is initiated by binding of AP50 to CD22. AP50 is part of the clathrin-associated AP-2 adaptor complex and binds to CD22 via Y843 and Y863 (part of the internalization motifs YXXØ), which are located in the cytoplasmic tail of CD22. However, the tyrosines of CD22 become phosphorylated shortly after BCR activation, which blocks AP50 binding to CD22 (21, 80). Only after ∼30 min, when this phosphorylation diminishes, clathrin-dependent internalization is initiated (21, 80). Then CD22 molecules return to the cell surface by a mechanism called receptor recycling rather than being degraded in the lysosome (Fig. 6). Our experiments could show for the first time, to our knowledge, that CD22 internalization upon BCR stimulation is dependent on the ubiquitin machinery. The molecular reason of this rather slow internalization process can be explained with the need for the inhibitory signal of CD22 after BCR stimulation. By sustaining CD22 localization to the cell surface after B cell activation, instead of fast CD22 internalization, the mechanism ensures adequate regulation of BCR signaling. However, this inhibitory signal needs to be turned off eventually, and this newly determined CRL3-dependent internalization of CD22 might be the molecular way to guarantee this.

This newly identified role of cullin 3 in activated B cells raised the question about its function in steady-state B lymphocytes. To address this, we crossed CUL3flox/flox mice to the B cell–specific cre-deleter strain mb1cre/+ to obtain highly efficient B cell–specific cullin 3 deletion already in early developmental B cell stages (53). Our analyses demonstrate that a cullin 3 deficiency not only causes defects in CD22 internalization upon BCR stimulation but also leads to constitutive higher expression levels of CD22 in unstimulated B cells. This again points to an important regulatory role of cullin 3 with respect to CD22. However, we do not conclude that this additional regulatory mechanism of cullin 3 in the steady-state is necessarily mediated by the same cellular mechanism as the internalization of CD22 in activated B cells. Although cullin 3 is clearly involved in controlling CD22 surface expression in unstimulated and stimulated B cells, the mechanisms might be different. Besides CD22, CD21 and CD23 surface receptor levels also seem to be elevated, indicating an additional role for cullin 3 in CD21 and CD23 expression.

Cullin 3 is known to be already expressed during B cell development in the bone marrow. Hence, we were also interested in other B cell related functions of cullin 3. Despite previously published results that showed hardly any developmental defects in CUL3fl/fl CD19CRE/+ B cells (52), we observed a much stronger opposing phenotype. We would like to highlight that instead of using CUL3fl/fl CD19CRE/+ mice, which were used in the other study, we chose to mate CUL3fl/fl with the mb1CRE/+ deleter strain. A gene deletion under the mb1 promotor was shown to be more efficient, compared with the CD19cre/+, and is therefore better suited to study B cell related functions (53). We chose the mb1cre/+ system to guarantee an earlier and more efficient deletion of the loxP flanked DNA segment of cullin 3 and therefore avoid cells that escape this excision process (53). This more stringent approach revealed an important function of cullin 3 during B cell development, as B cell numbers were heavily impaired in CUL3fl/fl mb1CRE/+ mice. This phenotype was not observed by Mathew et al. (52), probably because of inefficient and incomplete deletion of CUL3 using the CD19cre/+ system. So-called “deletion escapers” could fill up the B cell niche, as they might have a survival advantage compared with cullin 3–deficient cells. This idea is supported by our observations, as we only detected few B cells in the periphery of CUL3fl/fl mb1CRE/+ mice.

The surviving B cells were characterized by an apoptotic and at the same time preactivated phenotype. However, we do not think that cullin 3–deficient B cells are functionally unresponsive, as IgM and IgD levels were comparable to WT cells, and anergic B cells would be characterized by an IgM reduction and higher IgD expression (8185). Furthermore, we do not conclude that CD22 overexpression causes the preactivated and apoptotic phenotype, because CD22-deficient B cells also show signs of preactivation and higher turnover (5). Therefore, this might rather be a direct or indirect effect of the cullin 3 deletion. However, whether the augmented CD22 expression is relevant for any of the B cell phenotypes in CUL3fl/fl mb1CRE/+ mice will be further analyzed using CD22−/− × CUL3fl/fl mb1CRE/+ in the future. The crossing of CD22−/− and CUL3fl/fl mb1CRE/+ is still ongoing, as CD22 and mb1 genes are both located on chromosome 7 with a distance of only 5.77 cM.

Developing B cells in the bone marrow showed a drastic reduction in the CUL3fl/fl mb1CRE/+ mice. This phenotype was observed starting at the pre-B cell stage. Hence, we addressed the question if the reduction of pre-B cells is caused by a proliferative or differentiational defect. Here we focused on IL-7 signaling, which is important for B cell development from pro- to pre-B cells in the bone marrow (72, 73). In fact, our data clearly demonstrate that CUL3fl/fl mb1CRE/+ pro-B cells fail to proliferate in presence of IL-7 but die instead. As IL-7R expression was unaltered, we suggest that a cell intrinsic problem causes this defect and propose that cullin 3 is important for pro-B cell proliferation. Moreover, cullin 3 was published to be involved in cell cycle control of hepatocytes (51), which might be one of its molecular function in B cells, too. Others have shown a regulatory role for cullin 3 in mitotic cell cycle entry and that cullin 3 knockdown cells undergo a delayed mitotic entry (74). In these experiments Cul3 small interfering RNA transfected HeLa cells showed an accumulation in G2/M phase (35.9% compared with 21.5% in control cells) (74). We observed this to a similar extent in CUL3fl/fl mb1CRE/+ pro-B cells. This accumulation of G2/M cell cycle phase might contribute to less pro-B cell proliferation.

Altogether, we showed that cullin 3 is a novel CD22-interacting protein that controls not only CD22 surface expression under steady-state conditions but also induces CD22 internalization by ubiquitination after BCR stimulation. Moreover, characterizing B cell–specific cullin 3 deficient mice (CUL3fl/fl mb1CRE/+), we revealed the importance of cullin 3 for normal B cell development. To our knowledge, this study provides a first insight into the role of cullin 3 on B cell development and additionally shows its importance for CD22 surface expression and receptor internalization after B cell activation.

We thank the Proteomics Identification Database team for data deposition to the ProteomeXchange Consortium.

This work was supported by the German Research Foundation (DFG) (TRR 130-P04 and TRR 130-C02 to L.N. and B.W.). Research in the Warscheid group was supported by the DFG under Germany’s Excellence Strategy (Centre for Integrative Biological Signalling Studies Grant EXC2189, Project Identifier 390939984).

The raw data and original MaxQuant results presented in this article have been submitted to the ProteomeXchange Consortium under identifier PXD013801.

The online version of this article contains supplemental material.

Abbreviations used in this article:

ABC

ammonium bicarbonate

AP

alkaline phosohatase

BEL

bromoenol lactone

CRL3

cullin 3–RING–ubiquitin ligase complex

LC-MS

liquid chromatography–mass spectrometry

MFI

mean fluorescence intensity

MHCII

MHC class II

Pen/Strep

penicillin/streptomycin

SILAC

stable isotope labeling by/with amino acids in cell culture

WT

wild-type.

1
Nutt
,
S. L.
,
P. D.
Hodgkin
,
D. M.
Tarlinton
,
L. M.
Corcoran
.
2015
.
The generation of antibody-secreting plasma cells.
Nat. Rev. Immunol.
15
:
160
171
.
2
Crocker
,
P. R.
,
P.
Redelinghuys
.
2008
.
Siglecs as positive and negative regulators of the immune system.
Biochem. Soc. Trans.
36
:
1467
1471
.
3
Crocker
,
P. R.
,
A.
Varki
.
2001
.
Siglecs in the immune system.
Immunology
103
:
137
145
.
4
Torres
,
R. M.
,
C. L.
Law
,
L.
Santos-Argumedo
,
P. A.
Kirkham
,
K.
Grabstein
,
R. M.
Parkhouse
,
E. A.
Clark
.
1992
.
Identification and characterization of the murine homologue of CD22, a B lymphocyte-restricted adhesion molecule.
J. Immunol.
149
:
2641
2649
.
5
Nitschke
,
L.
,
R.
Carsetti
,
B.
Ocker
,
G.
Köhler
,
M. C.
Lamers
.
1997
.
CD22 is a negative regulator of B-cell receptor signalling.
Curr. Biol.
7
:
133
143
.
6
Sato
,
S.
,
P. J.
Jansen
,
T. F.
Tedder
.
1997
.
CD19 and CD22 expression reciprocally regulates tyrosine phosphorylation of Vav protein during B lymphocyte signaling.
Proc. Natl. Acad. Sci. USA
94
:
13158
13162
.
7
Sato
,
S.
,
A. S.
Miller
,
M.
Inaoki
,
C. B.
Bock
,
P. J.
Jansen
,
M. L. K.
Tang
,
T. F.
Tedder
.
1996
.
CD22 is both a positive and negative regulator of B lymphocyte antigen receptor signal transduction: altered signaling in CD22-deficient mice.
Immunity
5
:
551
562
.
8
O’Keefe
,
T. L.
,
G. T.
Williams
,
S. L.
Davies
,
M. S.
Neuberger
.
1996
.
Hyperresponsive B cells in CD22-deficient mice.
Science
274
:
798
801
.
9
Otipoby
,
K. L.
,
K. B.
Andersson
,
K. E.
Draves
,
S. J.
Klaus
,
A. G.
Farr
,
J. D.
Kerner
,
R. M.
Perlmutter
,
C. L.
Law
,
E. A.
Clark
.
1996
.
CD22 regulates thymus-independent responses and the lifespan of B cells.
Nature
384
:
634
637
.
10
Meyer
,
S. J.
,
A. T.
Linder
,
C.
Brandl
,
L.
Nitschke
.
2018
.
B cell Siglecs-news on signaling and its interplay with ligand binding.
Front. Immunol.
9
:
2820
.
11
May
,
A. P.
,
R. C.
Robinson
,
M.
Vinson
,
P. R.
Crocker
,
E. Y.
Jones
.
1998
.
Crystal structure of the N-terminal domain of sialoadhesin in complex with 3′ sialyllactose at 1.85 A resolution.
Mol. Cell
1
:
719
728
.
12
Müller
,
J.
,
I.
Obermeier
,
M.
Wöhner
,
C.
Brandl
,
S.
Mrotzek
,
S.
Angermüller
,
P. C.
Maity
,
M.
Reth
,
L.
Nitschke
.
2013
.
CD22 ligand-binding and signaling domains reciprocally regulate B-cell Ca2+ signaling.
Proc. Natl. Acad. Sci. USA
110
:
12402
12407
.
13
Otipoby
,
K. L.
,
K. E.
Draves
,
E. A.
Clark
.
2001
.
CD22 regulates B cell receptor-mediated signals via two domains that independently recruit Grb2 and SHP-1.
J. Biol. Chem.
276
:
44315
44322
.
14
Smith
,
K. G. C.
,
D. M.
Tarlinton
,
G. M.
Doody
,
M. L.
Hibbs
,
D. T.
Fearon
.
1998
.
Inhibition of the B cell by CD22: a requirement for Lyn.
J. Exp. Med.
187
:
807
811
.
15
Blasioli
,
J.
,
S.
Paust
,
M. L.
Thomas
.
1999
.
Definition of the sites of interaction between the protein tyrosine phosphatase SHP-1 and CD22.
J. Biol. Chem.
274
:
2303
2307
.
16
Poe
,
J. C.
,
M.
Fujimoto
,
P. J.
Jansen
,
A. S.
Miller
,
T. F.
Tedder
.
2000
.
CD22 forms a quaternary complex with SHIP, Grb2, and Shc. A pathway for regulation of B lymphocyte antigen receptor-induced calcium flux.
J. Biol. Chem.
275
:
17420
17427
.
17
Ono
,
M.
,
H.
Okada
,
S.
Bolland
,
S.
Yanagi
,
T.
Kurosaki
,
J. V.
Ravetch
.
1997
.
Deletion of SHIP or SHP-1 reveals two distinct pathways for inhibitory signaling.
Cell
90
:
293
301
.
18
Chen
,
J.
,
P. A.
McLean
,
B. G.
Neel
,
G.
Okunade
,
G. E.
Shull
,
H. H.
Wortis
.
2004
.
CD22 attenuates calcium signaling by potentiating plasma membrane calcium-ATPase activity.
Nat. Immunol.
5
:
651
657
.
19
Chen
,
J.
,
H.
Wang
,
W.-P.
Xu
,
S.-S.
Wei
,
H. J.
Li
,
Y.-Q.
Mei
,
Y.-G.
Li
,
Y.-P.
Wang
.
2016
.
Besides an ITIM/SHP-1-dependent pathway, CD22 collaborates with Grb2 and plasma membrane calcium-ATPase in an ITIM/SHP-1-independent pathway of attenuation of Ca2+i signal in B cells.
Oncotarget
7
:
56129
56146
.
20
Tateno
,
H.
,
H.
Li
,
M. J.
Schur
,
N.
Bovin
,
P. R.
Crocker
,
W. W.
Wakarchuk
,
J. C.
Paulson
.
2007
.
Distinct endocytic mechanisms of CD22 (Siglec-2) and Siglec-F reflect roles in cell signaling and innate immunity.
Mol. Cell. Biol.
27
:
5699
5710
.
21
John
,
B.
,
B. R.
Herrin
,
C.
Raman
,
Y. N.
Wang
,
K. R.
Bobbitt
,
B. A.
Brody
,
L. B.
Justement
.
2003
.
The B cell coreceptor CD22 associates with AP50, a clathrin-coated pit adapter protein, via tyrosine-dependent interaction.
J. Immunol.
170
:
3534
3543
.
22
Boll
,
W.
,
H.
Ohno
,
Z.
Songyang
,
I.
Rapoport
,
L. C.
Cantley
,
J. S.
Bonifacino
,
T.
Kirchhausen
.
1996
.
Sequence requirements for the recognition of tyrosine-based endocytic signals by clathrin AP-2 complexes.
EMBO J.
15
:
5789
5795
.
23
Shan
,
D.
,
O. W.
Press
.
1995
.
Constitutive endocytosis and degradation of CD22 by human B cells.
J. Immunol.
154
:
4466
4475
.
24
Wayne
,
A. S.
,
N. N.
Shah
,
D.
Bhojwani
,
L. B.
Silverman
,
J. A.
Whitlock
,
M.
Stetler-Stevenson
,
W.
Sun
,
M.
Liang
,
J.
Yang
,
R. J.
Kreitman
, et al
.
2017
.
Phase 1 study of the anti-CD22 immunotoxin moxetumomab pasudotox for childhood acute lymphoblastic leukemia.
Blood
130
:
1620
1627
.
25
Kreitman
,
R. J.
,
C.
Dearden
,
P. L.
Zinzani
,
J.
Delgado
,
L.
Karlin
,
T.
Robak
,
D. E.
Gladstone
,
P.
le Coutre
,
S.
Dietrich
,
M.
Gotic
, et al
.
2018
.
Moxetumomab pasudotox in relapsed/refractory hairy cell leukemia.
Leukemia
32
:
1768
1777
.
26
Kantarjian
,
H. M.
,
Y.
Su
,
E. J.
Jabbour
,
H.
Bhattacharyya
,
E.
Yan
,
J. C.
Cappelleri
,
D. I.
Marks
.
2018
.
Patient-reported outcomes from a phase 3 randomized controlled trial of inotuzumab ozogamicin versus standard therapy for relapsed/refractory acute lymphoblastic leukemia.
Cancer
124
:
2151
2160
.
27
Prescher
,
H.
,
A.
Schweizer
,
E.
Kuhfeldt
,
L.
Nitschke
,
R.
Brossmer
.
2017
.
New human CD22/Siglec-2 ligands with a triazole glycoside.
ChemBioChem
18
:
1216
1225
.
28
Schweizer
,
A.
,
M.
Wöhner
,
H.
Prescher
,
R.
Brossmer
,
L.
Nitschke
.
2012
.
Targeting of CD22-positive B-cell lymphoma cells by synthetic divalent sialic acid analogues.
Eur. J. Immunol.
42
:
2792
2802
.
29
Chen
,
W. C.
,
G. C.
Completo
,
D. S.
Sigal
,
P. R.
Crocker
,
A.
Saven
,
J. C.
Paulson
.
2010
.
In vivo targeting of B-cell lymphoma with glycan ligands of CD22.
Blood
115
:
4778
4786
.
30
Peng
,
W.
,
J. C.
Paulson
.
2017
.
CD22 ligands on a natural N-glycan scaffold efficiently deliver toxins to B-lymphoma cells.
J. Am. Chem. Soc.
139
:
12450
12458
.
31
Sarikas
,
A.
,
T.
Hartmann
,
Z.-Q.
Pan
.
2011
.
The cullin protein family.
Genome Biol.
12
:
220
.
32
Varshavsky
,
A.
1997
.
The ubiquitin system.
Trends Biochem. Sci.
22
:
383
387
.
33
Hershko
,
A.
,
A.
Ciechanover
.
1992
.
The ubiquitin system for protein degradation.
Annu. Rev. Biochem.
61
:
761
807
.
34
Terrell
,
J.
,
S.
Shih
,
R.
Dunn
,
L.
Hicke
.
1998
.
A function for monoubiquitination in the internalization of a G protein-coupled receptor.
Mol. Cell
1
:
193
202
.
35
Sorkin
,
A.
,
M.
von Zastrow
.
2009
.
Endocytosis and signalling: intertwining molecular networks.
Nat. Rev. Mol. Cell Biol.
10
:
609
622
.
36
Katzmann
,
D. J.
,
C. J.
Stefan
,
M.
Babst
,
S. D.
Emr
.
2003
.
Vps27 recruits ESCRT machinery to endosomes during MVB sorting.
J. Cell Biol.
162
:
413
423
.
37
Raiborg
,
C.
,
H.
Stenmark
.
2009
.
The ESCRT machinery in endosomal sorting of ubiquitylated membrane proteins.
Nature
458
:
445
452
.
38
Petroski
,
M. D.
,
R. J.
Deshaies
.
2005
.
Function and regulation of cullin-RING ubiquitin ligases.
Nat. Rev. Mol. Cell Biol.
6
:
9
20
.
39
Glickman
,
M. H.
,
A.
Ciechanover
.
2002
.
The ubiquitin-proteasome proteolytic pathway: destruction for the sake of construction.
Physiol. Rev.
82
:
373
428
.
40
Lammer
,
D.
,
N.
Mathias
,
J. M.
Laplaza
,
W.
Jiang
,
Y.
Liu
,
J.
Callis
,
M.
Goebl
,
M.
Estelle
.
1998
.
Modification of yeast Cdc53p by the ubiquitin-related protein rub1p affects function of the SCFCdc4 complex.
Genes Dev.
12
:
914
926
.
41
Liakopoulos
,
D.
,
G.
Doenges
,
K.
Matuschewski
,
S.
Jentsch
.
1998
.
A novel protein modification pathway related to the ubiquitin system.
EMBO J.
17
:
2208
2214
.
42
Pintard
,
L.
,
A.
Willems
,
M.
Peter
.
2004
.
Cullin-based ubiquitin ligases: Cul3-BTB complexes join the family.
EMBO J.
23
:
1681
1687
.
43
Choo
,
Y. Y.
,
T.
Hagen
.
2012
.
Mechanism of cullin3 E3 ubiquitin ligase dimerization.
PLoS One
7
: e41350.
44
Wimuttisuk
,
W.
,
J. D.
Singer
.
2007
.
The Cullin3 ubiquitin ligase functions as a Nedd8-bound heterodimer.
Mol. Biol. Cell
18
:
899
909
.
45
Canning
,
P.
,
C. D. O.
Cooper
,
T.
Krojer
,
J. W.
Murray
,
A. C. W.
Pike
,
A.
Chaikuad
,
T.
Keates
,
C.
Thangaratnarajah
,
V.
Hojzan
,
V.
Ayinampudi
, et al
.
2013
.
Structural basis for Cul3 protein assembly with the BTB-Kelch family of E3 ubiquitin ligases. [Published erratum appears in 2013 J. Biol. Chem. 388: 28304.]
J. Biol. Chem.
288
:
7803
7814
.
46
Stogios
,
P. J.
,
G. S.
Downs
,
J. J. S.
Jauhal
,
S. K.
Nandra
,
G. G.
Privé
.
2005
.
Sequence and structural analysis of BTB domain proteins.
Genome Biol.
6
:
R82
.
47
Bertocci
,
B.
,
D.
Lecoeuche
,
D.
Sterlin
,
J.
Kühn
,
B.
Gaillard
,
A.
De Smet
,
F.
Lembo
,
C.
Bole-Feysot
,
N.
Cagnard
,
T.
Fadeev
, et al
.
2017
.
Klhl6 deficiency impairs transitional B cell survival and differentiation.
J. Immunol.
199
:
2408
2420
.
48
Komander
,
D.
,
M.
Rape
.
2012
.
The ubiquitin code.
Annu. Rev. Biochem.
81
:
203
229
.
49
Singer
,
J. D.
,
M.
Gurian-West
,
B.
Clurman
,
J. M.
Roberts
.
1999
.
Cullin-3 targets cyclin E for ubiquitination and controls S phase in mammalian cells.
Genes Dev.
13
:
2375
2387
.
50
Pintard
,
L.
,
T.
Kurz
,
S.
Glaser
,
J. H.
Willis
,
M.
Peter
,
B.
Bowerman
.
2003
.
Neddylation and deneddylation of CUL-3 is required to target MEI-1/Katanin for degradation at the meiosis-to-mitosis transition in C. elegans.
Curr. Biol.
13
:
911
921
.
51
McEvoy
,
J. D.
,
U.
Kossatz
,
N.
Malek
,
J. D.
Singer
.
2007
.
Constitutive turnover of cyclin E by Cul3 maintains quiescence.
Mol. Cell. Biol.
27
:
3651
3666
.
52
Mathew
,
R.
,
M. P.
Seiler
,
S. T.
Scanlon
,
A. P.
Mao
,
M. G.
Constantinides
,
C.
Bertozzi-Villa
,
J. D.
Singer
,
A.
Bendelac
.
2012
.
BTB-ZF factors recruit the E3 ligase cullin 3 to regulate lymphoid effector programs.
Nature
491
:
618
621
.
53
Hobeika
,
E.
,
S.
Thiemann
,
B.
Storch
,
H.
Jumaa
,
P. J.
Nielsen
,
R.
Pelanda
,
M.
Reth
.
2006
.
Testing gene function early in the B cell lineage in mb1-cre mice.
Proc. Natl. Acad. Sci. USA
103
:
13789
13794
.
54
Soucy
,
T. A.
,
P. G.
Smith
,
M. A.
Milhollen
,
A. J.
Berger
,
J. M.
Gavin
,
S.
Adhikari
,
J. E.
Brownell
,
K. E.
Burke
,
D. P.
Cardin
,
S.
Critchley
, et al
.
2009
.
An inhibitor of NEDD8-activating enzyme as a new approach to treat cancer.
Nature
458
:
732
736
.
55
Schmidt
,
T. G. M.
,
L.
Batz
,
L.
Bonet
,
U.
Carl
,
G.
Holzapfel
,
K.
Kiem
,
K.
Matulewicz
,
D.
Niermeier
,
I.
Schuchardt
,
K.
Stanar
.
2013
.
Development of the Twin-Strep-tag® and its application for purification of recombinant proteins from cell culture supernatants.
Protein Expr. Purif.
92
:
54
61
.
56
Morita
,
S.
,
T.
Kojima
,
T.
Kitamura
.
2000
.
Plat-E: an efficient and stable system for transient packaging of retroviruses.
Gene Ther.
7
:
1063
1066
.
57
Ong
,
S.-E.
,
B.
Blagoev
,
I.
Kratchmarova
,
D. B.
Kristensen
,
H.
Steen
,
A.
Pandey
,
M.
Mann
.
2002
.
Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics.
Mol. Cell. Proteomics
1
:
376
386
.
58
Cox
,
J.
,
M.
Mann
.
2008
.
MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification.
Nat. Biotechnol.
26
:
1367
1372
.
59
Cox
,
J.
,
N.
Neuhauser
,
A.
Michalski
,
R. A.
Scheltema
,
J. V.
Olsen
,
M.
Mann
.
2011
.
Andromeda: a peptide search engine integrated into the MaxQuant environment.
J. Proteome Res.
10
:
1794
1805
.
60
Waisman
,
A.
,
M.
Kraus
,
J.
Seagal
,
S.
Ghosh
,
D.
Melamed
,
J.
Song
,
Y.
Sasaki
,
S.
Classen
,
C.
Lutz
,
F.
Brombacher
, et al
.
2007
.
IgG1 B cell receptor signaling is inhibited by CD22 and promotes the development of B cells whose survival is less dependent on Ig alpha/beta.
J. Exp. Med.
204
:
747
758
.
61
Perez-Riverol
,
Y.
,
A.
Csordas
,
J.
Bai
,
M.
Bernal-Llinares
,
S.
Hewapathirana
,
D. J.
Kundu
,
A.
Inuganti
,
J.
Griss
,
G.
Mayer
,
M.
Eisenacher
, et al
.
2019
.
The PRIDE database and related tools and resources in 2019: improving support for quantification data.
Nucleic Acids Res.
47
:
D442
D450
.
62
Buerstedde
,
J.-M.
,
S.
Takeda
.
1991
.
Increased ratio of targeted to random integration after transfection of chicken B cell lines.
Cell
67
:
179
188
.
63
Kubista
,
H.
,
T.
Hawkins
,
S. E.
Moss
.
1998
.
Characterisation of calcium signalling in DT40 chicken B-cells.
Biochim. Biophys. Acta
1448
:
299
310
.
64
Winding
,
P.
,
M. W.
Berchtold
.
2001
.
The chicken B cell line DT40: a novel tool for gene disruption experiments.
J. Immunol. Methods
249
:
1
16
.
65
Gasparrini
,
F.
,
C.
Feest
,
A.
Bruckbauer
,
P. K.
Mattila
,
J.
Müller
,
L.
Nitschke
,
D.
Bray
,
F. D.
Batista
.
2016
.
Nanoscale organization and dynamics of the siglec CD22 cooperate with the cytoskeleton in restraining BCR signalling.
EMBO J.
35
:
258
280
.
66
Collins
,
B. E.
,
O.
Blixt
,
N. V.
Bovin
,
C. P.
Danzer
,
D.
Chui
,
J. D.
Marth
,
L.
Nitschke
,
J. C.
Paulson
.
2002
.
Constitutively unmasked CD22 on B cells of ST6Gal I knockout mice: novel sialoside probe for murine CD22.
Glycobiology
12
:
563
571
.
67
Han
,
S.
,
B. E.
Collins
,
P.
Bengtson
,
J. C.
Paulson
.
2005
.
Homomultimeric complexes of CD22 in B cells revealed by protein-glycan cross-linking.
Nat. Chem. Biol.
1
:
93
97
.
68
Wu
,
J.-T.
,
H.-C.
Lin
,
Y.-C.
Hu
,
C.-T.
Chien
.
2005
.
Neddylation and deneddylation regulate Cul1 and Cul3 protein accumulation.
Nat. Cell Biol.
7
:
1014
1020
.
69
O’Reilly
,
M. K.
,
H.
Tian
,
J. C.
Paulson
.
2011
.
CD22 is a recycling receptor that can shuttle cargo between the cell surface and endosomal compartments of B cells.
J. Immunol.
186
:
1554
1563
.
70
de Figueiredo
,
P.
,
A.
Doody
,
R. S.
Polizotto
,
D.
Drecktrah
,
S.
Wood
,
M.
Banta
,
M. S.
Strang
,
W. J.
Brown
.
2001
.
Inhibition of transferrin recycling and endosome tubulation by phospholipase A2 antagonists.
J. Biol. Chem.
276
:
47361
47370
.
71
Choi
,
J.
,
K.
Lee
,
K.
Ingvarsdottir
,
R.
Bonasio
,
A.
Saraf
,
L.
Florens
,
M. P.
Washburn
,
S.
Tadros
,
M. R.
Green
,
L.
Busino
.
2018
.
Loss of KLHL6 promotes diffuse large B-cell lymphoma growth and survival by stabilizing the mRNA decay factor roquin2.
Nat. Cell Biol.
20
:
586
596
.
72
Corcoran
,
A. E.
,
A.
Riddell
,
D.
Krooshoop
,
A. R.
Venkitaraman
.
1998
.
Impaired immunoglobulin gene rearrangement in mice lacking the IL-7 receptor.
Nature
391
:
904
907
.
73
von Freeden-Jeffry
,
U.
,
P.
Vieira
,
L. A.
Lucian
,
T.
McNeil
,
S. E.
Burdach
,
R.
Murray
.
1995
.
Lymphopenia in interleukin (IL)-7 gene-deleted mice identifies IL-7 as a nonredundant cytokine.
J. Exp. Med.
181
:
1519
1526
.
74
Moghe
,
S.
,
F.
Jiang
,
Y.
Miura
,
R. L.
Cerny
,
M.-Y.
Tsai
,
M.
Furukawa
.
2012
.
The CUL3-KLHL18 ligase regulates mitotic entry and ubiquitylates Aurora-A.
Biol. Open
1
:
82
91
.
75
Piper
,
R. C.
,
I.
Dikic
,
G. L.
Lukacs
.
2014
.
Ubiquitin-dependent sorting in endocytosis.
Cold Spring Harb. Perspect. Biol.
6
:
a016808
a016808
.
76
Mayor
,
S.
,
R. E.
Pagano
.
2007
.
Pathways of clathrin-independent endocytosis.
Nat. Rev. Mol. Cell Biol.
8
:
603
612
.
77
Mayor
,
S.
,
R. G.
Parton
,
J. G.
Donaldson
.
2014
.
Clathrin-independent pathways of endocytosis.
Cold Spring Harb. Perspect. Biol.
6
: a016758.
78
Maxfield
,
F. R.
,
T. E.
McGraw
.
2004
.
Endocytic recycling.
Nat. Rev. Mol. Cell Biol.
5
:
121
132
.
79
Doherty
,
G. J.
,
H. T.
McMahon
.
2009
.
Mechanisms of endocytosis.
Annu. Rev. Biochem.
78
:
857
902
.
80
Fujimoto
,
M.
,
Y.
Kuwano
,
R.
Watanabe
,
N.
Asashima
,
H.
Nakashima
,
S.
Yoshitake
,
H.
Okochi
,
K.
Tamaki
,
J. C.
Poe
,
T. F.
Tedder
,
S.
Sato
.
2006
.
B cell antigen receptor and CD40 differentially regulate CD22 tyrosine phosphorylation.
J. Immunol.
176
:
873
879
.
81
Duty
,
J. A.
,
P.
Szodoray
,
N.-Y.
Zheng
,
K. A.
Koelsch
,
Q.
Zhang
,
M.
Swiatkowski
,
M.
Mathias
,
L.
Garman
,
C.
Helms
,
B.
Nakken
, et al
.
2009
.
Functional anergy in a subpopulation of naive B cells from healthy humans that express autoreactive immunoglobulin receptors.
J. Exp. Med.
206
:
139
151
.
82
Merrell
,
K. T.
,
R. J.
Benschop
,
S. B.
Gauld
,
K.
Aviszus
,
D.
Decote-Ricardo
,
L. J.
Wysocki
,
J. C.
Cambier
.
2006
.
Identification of anergic B cells within a wild-type repertoire.
Immunity
25
:
953
962
.
83
Quách
,
T. D.
,
N.
Manjarrez-Orduño
,
D. G.
Adlowitz
,
L.
Silver
,
H.
Yang
,
C.
Wei
,
E. C.
Milner
,
I.
Sanz
.
2011
.
Anergic responses characterize a large fraction of human autoreactive naive B cells expressing low levels of surface IgM.
J. Immunol.
186
:
4640
4648
.
84
Zikherman
,
J.
,
R.
Parameswaran
,
A.
Weiss
.
2012
.
Endogenous antigen tunes the responsiveness of naive B cells but not T cells.
Nature
489
:
160
164
.
85
Goodnow
,
C. C.
,
J.
Crosbie
,
H.
Jorgensen
,
R. A.
Brink
,
A.
Basten
.
1989
.
Induction of self-tolerance in mature peripheral B lymphocytes.
Nature
342
:
385
391
.

The authors have no financial conflicts of interest.

Supplementary data