23
1 Distribution and Function of Macrophage Galactose-type C-type Lectin 2 (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai 1 , Satoshi Aida 1 , Kengo Saba 1 , Kiwamu Suzuki 1 , Saya Moriyama 1 , Sarawut Oo-puthinan 1 , Makoto Tsuiji 2 , Akiko Morikawa 1 , Yosuke Kumamoto 1 , Daisuke Sugiura 1 , Akihiko Kudo 3 , Yoshihiro Akimoto 3 , Hayato Kawakami 3 , Nicolai V. Bovin 4 , and Tatsuro Irimura 1 Laboratory of Cancer Biology and Molecular Immunology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan 1 ; Department of Microbiology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo 142-8501, Japan 2 ; Division of Microscopic Anatomy, Department of Anatomy, Kyorin University School of Medicine 3 ; Shemyakin & Ovchinnikov Institute of Bio-organic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya 16/10, 117997 Moscow, Russia 4 Running title: Uptake and presentation of glycosylated antigens by MGL2 Address correspondence to: Tatsuro Irimura, Laboratory of Cancer Biology and Molecular Immunology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-0033, Japan; Tel: +81-3-5841-4870; Fax: +81-3-5841-4879; E-Mail: [email protected] Dendritic cells (DCs) express cell surface lectins that are potentially involved in the recognition, uptake, and presentation of glycosylated foreign substances. A unique calcium-type (C-type) lectin, the macrophage galactose (Gal)-type C-type lectin (MGL/CD301) expressed on DCs, is thought to participate in the recognition of molecules from both altered self and pathogens due to its monosaccharide specificity for Gal and N-acetylagalactosamine (GalNAc). Though mice have two MGL genes Mgl1 and Mgl2, their distinct roles have not been previously explored. The present report characterizes the properties of MGL2 by examining its distribution and its role in antigen presentation by DCs. We generated an MGL2-specific monoclonal antibody and examined MGL2 expression in tissues by immunohistochemistry and in isolated cells by flow cytometry. The cells reactive with this antibody were shown to be a portion of MGL1-expressing cells, mostly conventional DCs. Internalization of soluble polyacrylamide with α -GalNAc residues (GalNAc-PAA) by bone marrow-derived DCs (BM-DCs) was mediated by MGL2, as revealed by a comparison of Mgl1 -/- and Mgl2 -/- BM-DCs with wild-type BM-DCs. Biotinylated GalNAc-PAA conjugated to streptavidin (SAv) was more efficiently presented to SAv-primed T cells by BM-DCs than β -N-acetylglucosamine-PAA conjugated to SAv or SAv alone, as shown by thymidine uptake and cytokine production. This is the first report that demonstrates the involvement of GalNAc residues in antigen uptake and presentation by DCs that lead to CD4 + T cell activation. It is estimated that more than half of all proteins produced by eukaryotes are glycosylated (1). Most, though not all, cell surface and secreted proteins are thought to be glycosylated. Therefore, the immune responses to self and foreign proteins are likely to be modulated through the functions of carbohydrate-recognition molecules (i.e., lectins) expressed on antigen presenting cells. Dendritic cells (DCs) are antigen presenting cells that are capable of activating or tolerizing antigen-specific naive T cells (2). Surface lectins are potentially involved in this process as endocytic receptors, signaling receptors, or regulators of cellular localization and trafficking. C-type lectins expressed on DCs and Langerhans cells include type I multi-carbohydrate recognition domain (CRD) lectins, such as the mannose receptor (MR) and DEC-205, and type II single-CRD lectins, such as macrophage galactose (Gal)-type C-type lectin (MGL/CD301), DCIR, DC-SIGN, dectin-1, dectin-2, BDCA-2, and Langerin (3-5). These C-type lectins have previously been shown to be involved with the internalization of antigens and their subsequent presentation to antigen-specific http://www.jbc.org/cgi/doi/10.1074/jbc.M110.113613 The latest version is at JBC Papers in Press. Published on March 19, 2010 as Manuscript M110.113613 Copyright 2010 by The American Society for Biochemistry and Molecular Biology, Inc. by guest on August 11, 2019 http://www.jbc.org/ Downloaded from

Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

  • Upload
    ngoque

  • View
    212

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

1

Distribution and Function of Macrophage Galactose-type C-type Lectin 2 (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic

Cells Kaori Denda-Nagai1, Satoshi Aida1, Kengo Saba1, Kiwamu Suzuki1, Saya Moriyama1, Sarawut

Oo-puthinan1, Makoto Tsuiji2, Akiko Morikawa1, Yosuke Kumamoto1, Daisuke Sugiura1, Akihiko Kudo3, Yoshihiro Akimoto3, Hayato Kawakami3, Nicolai V. Bovin4, and Tatsuro Irimura1

Laboratory of Cancer Biology and Molecular Immunology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan1; Department of Microbiology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo 142-8501, Japan2; Division of Microscopic Anatomy,

Department of Anatomy, Kyorin University School of Medicine3; Shemyakin & Ovchinnikov Institute of Bio-organic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya 16/10, 117997 Moscow,

Russia4 Running title: Uptake and presentation of glycosylated antigens by MGL2

Address correspondence to: Tatsuro Irimura, Laboratory of Cancer Biology and Molecular Immunology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo

Bunkyo-ku, Tokyo 113-0033, Japan; Tel: +81-3-5841-4870; Fax: +81-3-5841-4879; E-Mail: [email protected]

Dendritic cells (DCs) express cell surface

lectins that are potentially involved in the recognition, uptake, and presentation of glycosylated foreign substances. A unique calcium-type (C-type) lectin, the macrophage galactose (Gal)-type C-type lectin (MGL/CD301) expressed on DCs, is thought to participate in the recognition of molecules from both altered self and pathogens due to its monosaccharide specificity for Gal and N-acetylagalactosamine (GalNAc). Though mice have two MGL genes Mgl1 and Mgl2, their distinct roles have not been previously explored. The present report characterizes the properties of MGL2 by examining its distribution and its role in antigen presentation by DCs. We generated an MGL2-specific monoclonal antibody and examined MGL2 expression in tissues by immunohistochemistry and in isolated cells by flow cytometry. The cells reactive with this antibody were shown to be a portion of MGL1-expressing cells, mostly conventional DCs. Internalization of soluble polyacrylamide with α-GalNAc residues (GalNAc-PAA) by bone marrow-derived DCs (BM-DCs) was mediated by MGL2, as revealed by a comparison of Mgl1-/- and Mgl2-/- BM-DCs with wild-type BM-DCs. Biotinylated GalNAc-PAA conjugated to streptavidin (SAv) was more efficiently presented to SAv-primed T cells by BM-DCs than β-N-acetylglucosamine-PAA conjugated to SAv

or SAv alone, as shown by thymidine uptake and cytokine production. This is the first report that demonstrates the involvement of GalNAc residues in antigen uptake and presentation by DCs that lead to CD4+ T cell activation.

It is estimated that more than half of all proteins produced by eukaryotes are glycosylated (1). Most, though not all, cell surface and secreted proteins are thought to be glycosylated. Therefore, the immune responses to self and foreign proteins are likely to be modulated through the functions of carbohydrate-recognition molecules (i.e., lectins) expressed on antigen presenting cells. Dendritic cells (DCs) are antigen presenting cells that are capable of activating or tolerizing antigen-specific naive T cells (2). Surface lectins are potentially involved in this process as endocytic receptors, signaling receptors, or regulators of cellular localization and trafficking. C-type lectins expressed on DCs and Langerhans cells include type I multi-carbohydrate recognition domain (CRD) lectins, such as the mannose receptor (MR) and DEC-205, and type II single-CRD lectins, such as macrophage galactose (Gal)-type C-type lectin (MGL/CD301), DCIR, DC-SIGN, dectin-1, dectin-2, BDCA-2, and Langerin (3-5). These C-type lectins have previously been shown to be involved with the internalization of antigens and their subsequent presentation to antigen-specific

http://www.jbc.org/cgi/doi/10.1074/jbc.M110.113613The latest version is at JBC Papers in Press. Published on March 19, 2010 as Manuscript M110.113613

Copyright 2010 by The American Society for Biochemistry and Molecular Biology, Inc.

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 2: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

2

T cells (4,6-9). However, the involvement of glycoforms in antigen uptake for presentation is not well understood because these previous conclusions have been obtained with anti-lectin antibodies. Among the lectins studied, monosaccharide specificity of MR, DC-SIGN, Langerin, and Dectin-2 is mannose, and dectin-1 is specific for β-glucan (5). The roles of C-type lectins on antigen presenting cells with monosaccharide specificity for Gal or N-acetylgalactosamine (GalNAc) are not thoroughly investigated even though these residues are thought to serve as terminals of glycans involved in tumor immunity, infection, and recognition of altered self.

Examples of Gal and GalNAc epitopes important in tumor immunity are the Tn (GalNAcα-Ser/Thr) and Thomsen-Friedenreich (TF : Galβ1-3GalNAcα-Ser/Thr) antigens in carcinoma-associated mucins (10). Cancer cells express MUC1 which has shorter carbohydrate chains that have been proposed to be recognized by the immune system (11), and MUC1-specific Cytotoxic T Lymphocytes and anti-MUC1 humoral reactions have been detected in breast, pancreatic, and ovarian carcinoma patients. Furthermore, early breast cancer patients with naturally occurring MUC1-specific antibodies have a better prognosis (12). The MUC1-specific immune response might be modulated by its TF/Tn carbohydrate chains, as suggested by a previous report (13). Whether such recognition leads to effective tumor immunity or tumor-induced immune suppression is currently unknown.

MGL is known as a type II transmembrane glycoprotein that contains a single CRD specific for monosaccharides Gal/GalNAc. A single gene encodes human MGL/CD301 (14), whereas mice have two genes MGL1/CD301a (15) and MGL2/CD301b (16). MGL was originally detected on tumoricidal macrophages (15) and was found to be expressed on histiocytic macrophages, but not on Langerhans cells (17,18). Recently, we identified MGL expression on immature DCs in humans and mice (19,20). However, the precise cellular distribution of MGL1 and MGL2 has not been investigated because the MGL-specific monoclonal antibodies (mAbs) used previously (mAb LOM-14 and mAb

ER-MP23) recognize a common epitope between MGL1 and MGL2, although mAb LOM-8.7, specific for MGL1, was also used in some studies (16,21). In the present report, the protein expression of MGL1 and MGL2 is individually shown from investigation with the combined use of mAb LOM-8.7 and novel MGL2-specific mAb URA-1. We describe in detail for the first time the unique characteristics of mAb URA-1. Furthermore, generation of Mgl2-knockout (Mgl2-/-) mice allows for investigating the immunological functions of MGL2 separately from that of MGL1 at the cellular level; once the backcross has been completed, further studies beyond the cellular level may proceed.

Based on available knowledge, MGL is the sole Gal-type lectin among the C-type lectins expressed on DCs. Furthermore, the MGL expressed on immature DCs has been shown to mediate the uptake of antigens containing GalNAc residues (19,20). Mouse and human MGLs have a high affinity for clusters of O-linked GalNAc residues (16,22,23). MGL on immature human monocyte-derived DCs have been shown to recognize and internalize the three tandem repeat peptides of MUC1 carrying Tn (Tn-MUC1) (13). An important question still remaining to be answered is whether human or mouse DCs, after internalizing antigens that have GalNAc residues, are able to elicit antigen-specific T cell responses. Thus, the final part of the report was designed to approach this question on the regulation of immune response in mice. The results indicate that polypeptides associated with mucins and mucin-like molecules with GalNAc residues are taken up by BM-DCs through MGL2 and presented to specific CD4+ T cells. This is the first report demonstrating that GalNAc residues on a complex antigen influence both its uptake by DCs and the subsequent antigen-specific immune response.

EXPERIMENTAL PROCEDURES

Animals–Specific pathogen-free (SPF) female C57BL/6 mice were obtained from CLEA Japan, Inc. (Tokyo, Japan). We used Mgl1-/- mice backcrossed 8 times to C57BL/6 background (24). Mgl2-/- mice were generated as described in the supplemental experimental procedures. Mgl1 or Mgl2 heterozygotes were bred for homozygotes

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 3: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

3

of the deficient Mgl1 or Mgl2 allele and their wild-type (WT) littermates. SPF F344/Du rats were obtained from Charles River Japan, Inc. (Yokohama, Japan). Animals were housed under SPF conditions. All experiments were approved by the Bioscience Committee of the Graduate School of Pharmaceutical Sciences of the University of Tokyo and performed according to the guidelines of the Bioscience Committee of the University of Tokyo.

Preparation of MGL2-specific mAbs–Recombinant proteins corresponding to the extracellular domain (ECD) of MGL1 and MGL2 were prepared as described previously (16). An F344/Du rat was subcutaneously immunized with 100 µg of MGL2-ECD in complete Freund’s adjuvant (BD, Franklin Lakes, NJ). One month later, a second immunization with 100 µg of MGL2-ECD in incomplete Freund’s adjuvant (BD) was given to the same rat, followed by an intraperitoneal booster injection of 100 µg of MGL2-ECD 4 days before fusion. Hybridoma cells were prepared as described previously (25). Cells in the wells that produced antibodies with binding capacity for MGL2-ECD, but not for MGL1-ECD, were subjected to limiting dilution for cloning. The subclass of mAbs was determined using a Rat MonoAB ID/SP Kit (Zymed, San Francisco, CA) according to the manufacturer’s instructions.

Enzyme-linked Immunosorbent Assay (ELISA)–Binding assays for antibodies were carried out as previously described (16).

Immunohistochemical Staining–The binding sites of mAb LOM-8.7 or mAb URA-1 were immunohistochemically detected as previously described with slight modifications (18). Avidin-binding sites were blocked using an Avidin/Biotin Blocking Kit (Vector, Burlingame, CA).

Preparation of Thioglycollate-induced Peritoneal Macrophages and In Vitro Cytokine Stimulation–C57BL/6 mice were injected peritoneally with 4.05% thioglycollate broth (BD). Three days later, peritoneal exudate cells (PEC) were harvested by a lavage with cold, serum-free RPMI 1640 media. Cells were stained with fluorescein isothiocyanate (FITC)-conjugated anti-CD11b (eBioscience, San Diego, CA) and phycoerythrin (PE)-conjugated F4/80

(eBioscience), and the double-positive cells were purified as peritoneal macrophages (PEC-macrophages) using a FACSAria cell sorter (BD) (>95% purity). The isolated cells were cultured in the presence of 10 ng/ml of mouse recombinant IL-4 (Peprotech, Rocky Hill, NJ) or recombinant IFN-γ (Peprotech) in a Petri dish (AGC Techno Glass Co., LTD, Chiba, Japan) overnight. Cells were collected by pipetting with cold Dulbecco’s modified phosphate buffered saline without calcium and magnesium (DPBS-).

Cell Preparations from Bone Marrow (BM), Spleens, and Lungs–BM cells were isolated by flushing femurs and tibias with DPBS-. Spleens were digested for 20 min at 37°C with 1 mg/ml collagenase from Clostridium histolyticum (Sigma, St. Louis, MO) in calcium and magnesium-free Hanks’ balanced salt solution. The digested tissue was resuspended by pipetting. Erythrocytes were lysed by ammonium chloride treatment. Splenocytes were treated with 10 mM EDTA to disrupt T cell-DC complexes.

Lung parenchymal cells were prepared by a modification of the method reported by Corti et al (26). Lung tissue digested with dispase (Invitrogen) was gently teased from the bronchi and transferred to the tube and incubated with gentle stirring with 200 U/ml collagenase (Wako, Osaka, Japan) for 10 min at 37°C. The cell suspension was filtered, centrifuged, and resuspended in DPBS-. Cells were layered onto a 20%/70% Percoll gradient (GE Healthcare) and centrifuged at 1800 rpm for 20 min at room temperature. Cells between the 20% and 70% layers were collected and washed with DPBS-.

Preparation of BM-macrophages–BM cells (2-4 x 106 cells/ml) were cultured in a 6 cm dish (BD) in 5 ml of cRPMI (RPMI 1640 medium supplemented with 10% FCS, 10 mM HEPES, 50 mM 2-ME, 100 U/ml penicillin, and 100 mg/ml streptomycin) and 20% conditioned media of L929 cells (Cell Resource Center for Biomedical Research, Tohoku University) as a source of macrophage colony-stimulating factor. After culturing overnight, non-adherent cells were transferred to a new 10 cm dish (BD) with the same media and 5 ml fresh media and then cultured for 5 days. Adherent cells were collected after treatment with 2.5 mM EDTA on ice for 10 min. Induction of macrophages was verified by flow cytometric analysis of CD11b

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 4: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

4

and F4/80 expression. Preparation and Purification of

BM-DCs–BM-DCs were generated using murine recombinant granulocyte-macrophage colony-stimulating factor (GM-CSF) (provided from Kirin Brewery Co.) as previously described (19). After 5 days culture, non-adherent cells were collected and used as immature BM-DCs. In some experiments, immature BM-DCs were incubated with anti-mouse CD11c microbeads (Miltenyi Biotec, Bergisch Glabach, Germany), and CD11c+ DCs were positively separated by LS columns or AutoMACS (Miltenyi Biotec) according to the manufacturer’s instructions. More than 95% purity was confirmed by flow cytometric analysis using FITC-conjugated anti-CD11c.

Flow Cytometric Analysis–CHO-K1 cells stably transfected with pCMV-Tag4-Mgl1, pCMV-Tag4-Mgl2, or pCMV-Tag4 were incubated with rat IgG, mAb LOM-14 (rat IgG2b), mAb LOM-8.7, or mAb URA-1 (10 µg/ml), followed by FITC-conjugated goat anti-Rat IgG (1/200 dilution, Invitrogen).

Cells isolated from mouse tissues and primary cultured cells were incubated with anti-mouse CD16/CD32 (1/100 dilution of ammonium sulfate precipitated hybridoma culture supernatant; the 2.4G2 hybridoma was purchased from ATCC) and 5% normal mouse serum to reduce nonspecific binding 5 min before the addition of the first antibodies. Cells were then incubated with biotin-, FITC-, PE-, PE-Cy5-, and/or allophycocyanin (APC)-conjugated antibodies for 30 min. Biotinylated antibodies were visualized with APC- or PE-Cy7- labeled streptavidin (SAv) (BioLegend, San Diego, CA). To exclude dead cells, all cells were resuspended in DPBS- containing 0.1% BSA and 0.1% sodium azide (FCM buffer) containing 7-amino-actinomycin D (eBioscience). A biotin-conjugated anti-MGL1 mAb LOM-8.7, a biotin-conjugated anti-MGL2 mAb URA-1, and an APC-conjugated anti-MGL2 mAb URA-1 were prepared using purified antibodies from the hybridoma culture supernatant; the following additional antibodies were used: FITC-conjugated anti-CD11c (BioLegend); PE-conjugated anti-CD4, CD40, Gr-1 (BioLegend), CD8 (Immunotech), CD11b, B220, CD86, major histocompatibility complex (MHC) class II,

F4/80 (eBioscience), mPDCA-1, and DEC-205 (Miltenyi Biotec); PE-Cy5-conjugated anti-CD3 and CD19 (eBioscience); isotype controls bio-rat IgG2a, PE-rat IgG2a, rat IgG2b, APC-rat IgG2a (eBioscience), and FITC hamster IgG (Santa Cruz Biotechnology, Santa Cruz, CA ).

All procedures were performed on ice. Antibodies and reagents were diluted in FCM buffer. The cells were rinsed twice with FCM buffer at the end of each incubation period. Samples were analyzed on an EPICS XL flow cytometer (Beckman Coulter, Fullerton, CA) or a FACSAria cell sorter. Data were analyzed using FlowJo software (Tree Star, Ashland, OR).

Carbohydrate Binding and Internalization Assessed by Flow Cytometric Analysis–The binding and uptake assays were performed as previously described (19).

Antigen Presentation Assays–Mice were immunized subcutaneously in the hind limb footpad and the tail base with 10-50 µg SAv (Sigma) emulsified in complete Freund’s adjuvant. WT or Mgl1 or Mgl2 heterozygotes were used for the immunization when Mgl1-/- or Mgl2-/- BM-DCs were used as stimulators in the antigen presentation assays. Seven days after the immunization, inguinal, popliteal, and iliac lymph nodes (LNs) were removed and single cell suspensions were prepared. T cells were purified with nylon wool columns (Wako). In some experiments, CD4+ T cells or CD8+ T cells were negatively selected by MACS. Briefly, nylon wool-purified T cells or LN cells were incubated with bio-anti-B220, CD11b, and CD4 or CD8 antibodies, followed by SAv-microbeads (Miltenyi Biotec). Then, cells were applied to LS columns or AutoMACS. Over 95% purity was confirmed using flow cytometry.

SAv of various concentrations indicated in figures or figure legends was pre-incubated with 1 µg/ml biotinylated (bio)-α-GalNAc-soluble polyacrylamide polymers (PAA) (27), bio-β-N-acetylglucosamine (GlcNAc)-PAA, bio-60 mer MUC1 tandem repeat synthetic peptide (non-glycosylated: AHGVTSAPDTRPAPGSTAPP x 3), or enzymatically glycosylated bio-MUC1 60 mer (9Tn, AHGVT*SAPDTRPAPGS*T*APP x 3, in which * indicates a GalNAc residue, or 14Tn, AHGVT*S*(except for the amino terminus) APDT*RPAPGS*T*APP x 3), kindly provided

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 5: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

5

by Dr. H. Clausen (University of Copenhagen, Denmark) (28). Immature BM-DCs or CD11c+ BM-DCs were cultured overnight with SAv complexes as antigens in cDMEM supplemented with GM-CSF. Antigen-pulsed BM-DCs were treated with 50 µg/ml mitomycin C (Kyowa Hakko Kirin Co., Tokyo, Japan). SAv-primed T cells (3 x 105 cells) were then co-cultured with antigen-pulsed BM-DCs in a 96-well U-bottom plate for 3 days. 3H-Thymidine (GE Healthcare) was added during the last 16 h. Data are shown as the mean cpm of triplicate cultures. The assays were judged to have failed due to a mismatch of MHC between T cells and DCs when the levels of 3H-thymidine uptake by T cells co-cultured with unpulsed BM-DCs of Mgl2+/+ or Mgl2-/- mice were similar to that of T cells stimulated with Concanavalin A.

To determine cytokine production levels, culture supernatants were collected 3 days after co-culturing of SAv-primed T cells with Ag-pulsed BM-DCs. IFN-γ (eBioscience), IL-4, IL-10, and IL-17A (Biolegend) were measured using cytokine ELISA kits according to the manufacturer’s instructions.

Statistics–Results of Carbohydrate binding and internalization assays and antigen presentation assays were statistically evaluated using the student’s t-test. P values less than 0.05 were considered significant. RESULTS

Preparation and Characterization of MGL2-specific mAb URA-1–Hybridomas secreting mAb specific for MGL2-ECD, but not for MGL1-ECD, were prepared, and a specific mAb URA-1(IgG2a) was chosen. The reactivity of this mAb with MGL1-ECD and MGL2-ECD was compared with that of mAb LOM-8.7 (MGL1-specific) and mAb LOM-14 (cross-reactive between MGL1 and MGL2) (Fig. 1A). The mAb URA-1 was shown to be specific for MGL2. The specificity of mAb URA-1 was also confirmed by flow cytometric analysis using CHO cells stably transfected with cDNAs of full length Mgl1 or Mgl2 (Fig. 1B). Because the stem domains of MGL1 and MGL2 have highly homologous amino acid sequences, the sequence within the CRD was thought to be responsible for the specificity of mAb URA-1. This mAb bound

to MGL2-CRD as well as MGL2-ECD (Supplementary Fig. 1A). The 61st amino acid within the CRD of MGL2 was previously shown to be important for the carbohydrate recognition of MGL2 (29). When this Leu was mutated to Val, which is the amino acid in MGL1 at the same position, the reactivity of mAb URA-1 to MGL2 diminished (Supplementary Fig. 1A). A similarly drastic effect was observed when the 110th Phe was substituted with Ile and when the 113th Asp was substituted with Gly (Supplementary Fig. 1A). To compare its blocking activity with that of mAb LOM-8.7 (25), we investigated the inhibition of ligand binding by mAb URA-1 using bio-Lewis X (LeX)-PAA (for MGL1) and bio-β−GalNAc-PAA (for MGL2). As shown in Supplementary Fig. 1B, mAb URA-1 inhibited the binding of β-GalNAc-PAA to MGL2-ECD, but not LeX-PAA to MGL1-ECD.

Immunohistochemical Distribution of MGL1 and MGL2 in Normal Mouse Tissues–The binding sites for mAb LOM-8.7 and mAb URA-1 were compared by immunohistochemistry. Both mAbs bound to cells in the connective tissues of almost all organs except for the brain; this profile was similar to the one previously shown using cross-reactive mAb LOM-14 (18). The number of cells that reacted with mAb URA-1 was similar to the number that reacted with mAb LOM-8.7 in most organs, including the thymus, large intestine, esophagus, stomach, heart, trachea, kidney, and skin (Fig. 2A and data not shown). The mAb binding profiles to sections of the thymus, a representative organ, are shown in Fig. 2A. The binding sites of these mAbs were present along the capsule and septa and scattered in the cortex and medulla. In spleens, MGL1+

cells (i.e. cells reactive with mAb LOM-8.7) had a more widespread distribution than MGL2+ cells (i.e. cells reactive with mAb URA-1) (Fig. 2B). The variations and distributions of the mAb-reactive cells in peripheral LNs have been published separately (30). In lung alveolar spaces, the number of MGL2+ cells appeared to be slightly greater than the number of MGL1+ cells (Fig. 2C).

Identification of DCs Expressing MGL1 and/or MGL2 in BM, Spleens, and Lungs–The surface expression of MGL1 and MGL2 on cells from the BM, spleens, and lungs were analyzed by flow cytometry using mAb LOM-8.7 and mAb

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 6: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

6

URA-1 after excluding dead cells and CD3+ or CD19+ cells. The mAbs were found to bind subsets of CD11c+ cells (Fig. 3A and 3B). BM and spleen were populated with more MGL1+

cells than MGL2+ cells. All MGL2+ cells appeared to co-express MGL1 in the BM, spleens, and lungs, whereas a significant portion of MGL1 single-positive cells were observed in the BM and spleens (Fig. 3C). In the BM, MGL1 and MGL2 double-positive cells represented a homogeneous population that was CD11c+CD4-CD8-CD11b+B220-MHCII+F4/80low, whereas MGL1 single-positive cells contained at least 2 populations, which were CD11c+B220+ cells and CD11c+B220- cells (Fig. 3D). In spleens, MGL1 and MGL2 double-positive cells were CD11c+CD4±CD8-CD11b+MHCII+F4/80low, and MGL1 single-positive cells contained at least 2 populations of CD11clowCD4+CD8±CD11b- B220+MHCIIlow and CD11c+CD4±CD8-CD11b+B220-MHCII+F4/80low cells (Fig. 3E). We also confirmed that CD11clow MGL1 single-positive cells in the spleen expressed mPDCA-1 (data not shown). These cells were also positive for B220 and should be classified as plasmacytoid DCs (pDCs) (31-33). In the lung, alveolar macrophages with high levels of autofluorescence did not express MGL1 or MGL2, as previously reported (17). MGL1 and MGL2 double-positive cells in the lung were CD11c+CD4-CD8-CD11b+MHCII+F4/80low (Fig. 3F). Although the number of cells in this organ with immunohistochemical reactivity with mAb URA-1 appeared to be slightly greater than the number of cells stained with mAb LOM-8.7, flow cytometry revealed a cell population that was reactive to both mAbs. These results collectively indicate that cells reactive to both mAb LOM-8.7 and mAb URA-1 correspond to CD8- conventional DCs (cDCs), and MGL1 single-positive cells are pDCs and cDCs.

Cell surface expression of MGL1 and MGL2 on induced macrophages and BM-DCs–The reactivity of these mAbs to BM-macrophages and PEC-macrophages was examined using flow cytometry. The mAb LOM-8.7 bound to BM-macrophages (Fig. 4A) and PEC-macrophages (Fig. 4B) at significant levels. In contrast, the levels of mAb URA-1 binding to BM-macrophages (Fig. 4A) and PEC-macrophages (Fig. 4B) were very low. As

Raes et al. reported, the expression of Mgl1 and Mgl2 mRNA may be induced by alternative activation of macrophages (34), PEC-macrophages were cultured overnight in the presence of IL-4 or IFN-γ, and mAb binding was examined. Classical activation of macrophages by IFN-γ was confirmed by the observed increase of MHC class II surface expression using flow cytometry (data not shown), and alternative activation of macrophages by IL-4 was confirmed by the induction of mRNA expression of Mgl1, Mgl2, Arginase-1, and MR (Supplementary Fig. 2). Surface MGL1 expression was slightly elevated after an overnight culture of PEC-macrophages with IL-4, but mAb URA-1 binding slightly decreased compared with cells cultured without cytokines (Fig. 4C). When intracellular bindings of these antibodies were examined by flow cytometry or cytostaining following permeabilization, the induction of intracellular proteins corresponding to these lectins was not observed (data not shown).

Both mAb LOM-8.7 and mAb URA-1 bound at significant levels to the surface of immature BM-DCs expressing low to moderate levels of MHC class II, whereas their bindings to mature BM-DCs expressing high levels of MHC class II were low (Fig. 5A and C). The mRNA level of Mgl1 examined by RT-PCR was previously reported to correlate with the surface binding of mAb LOM-14 to BM-DCs; the bindings were high at immature stages and low at mature stages (19). A similar mRNA expression pattern was observed for Mgl2 (data not shown).

MGL2 is required for the binding and uptake of GalNAc-PAA by BM-DCs–We previously reported that GalNAc-PAA was efficiently bound and internalized by immature BM-DCs (19). To understand whether MGL1 or MGL2 alone was sufficient for the recognition of GalNAc-PAA, the binding and uptake of GalNAc-PAA was examined using CHO cells transfected with Mgl1 or Mgl2 cDNA. FITC-GalNAc-PAA, but not FITC-GlcNAc-PAA, was bound and internalized by both cell types (Fig. 6A). These results demonstrated that MGL1 or MGL2 alone on CHO cells was sufficient to bind and internalize GalNAc-PAA.

To determine whether the binding and uptake of FITC-GalNAc-PAA was mediated by MGL1 or MGL2 expressed on DCs, BM-DCs

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 7: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

7

were prepared from the BM cells of Mgl1-/-, Mgl2-/-, or WT littermate mice. Mgl2-/- mice were generated as described in Supplementary Fig. 3. The mice bred normally and were healthy under SPF conditions at least up to 6 months. There was no remarkable phenotype in the development of immune cells according to the examination of peripheral blood smears and flow cytometric analysis of thymocytes (data not shown). The absence of MGL1 or MGL2 was confirmed on BM-DCs of Mgl1-/- or Mgl2-/- mice, respectively (Fig. 5B, D). Unexpectedly, MGL2 expression on Mgl1-/- BM-DCs (geometric mean fluorescence intensity (MFI) = 1679) was approximately half of WT littermate (Mgl1+/+) BM-DCs (MFI = 2690) (Fig. 5A, B, E), and MGL1 expression on Mgl2-/- BM-DCs (MFI = 399) was significantly lower than that of BM-DCs from WT littermate (Mgl2+/+) mice (geometric MFI = 1717) (Fig. 5C, D, F). The relative mRNA expression levels of Mgl1 in Mgl2-/- mice or Mgl2 in Mgl1-/- mice normalized to β-actin were lower than that of WT littermates, as measured by real-time RT-PCR (data not shown). However, the mechanism of reduced mRNA expression levels of MGL1 or MGL2 on Mgl2-/- or Mgl1-/- cells, respectively, is currently unknown.

BM-DCs prepared from Mgl1+/+ (Fig. 6B) or Mgl2+/+ mice (Fig. 6C) bound and internalized FITC-GalNAc-PAA, but not FITC-GlcNAc-PAA. The level of binding and the rate of uptake of FITC-GalNAc-PAA by Mgl1-/- BM-DCs were compared with those of Mgl1+/+ BM-DCs (Fig. 6B). FITC-GalNAc-PAA binding was slightly lower than Mgl1+/+ BM-DCs, and FITC-GalNAc-PAA uptake was approximately half that of Mgl1+/+ BM-DCs at each time point. These results appear to correlate with the cell surface levels of MGL2 on Mgl1-/- BM-DCs. In contrast to the MFI of FITC-GalNAc-PAA binding and uptake by Mgl1-/- BM-DCs, that of Mgl2-/- BM-DCs was reduced to almost the same levels of the controls (Fig. 6C). These results strongly suggest that cell surface expression of MGL2 on BM-DCs is required for the binding and internalization of FITC-GalNAc-PAA.

Presentation of antigens linked to GalNAc residues internalized through MGL2 on BM-DCs–To investigate whether antigens containing terminal GalNAc residues were

presented by MHC molecules to T cells after internalization, we developed an antigen presentation assay using bio-GalNAc-PAA conjugated with SAv as a model antigen. Mice were immunized with SAv, and SAv-primed T cells were harvested. T cell proliferation was efficiently induced in a dose-dependent manner when the cells were co-cultured with BM-DCs pre-incubated with bio-GalNAc-PAA conjugated with SAv (Fig. 7A). BM-DCs pre-incubated with SAv alone or bio-GlcNAc-PAA conjugated with SAv did not induce efficient proliferation of SAv-primed T cells. Because BM-DCs pre-incubated with bio-GalNAc-PAA alone did not induce efficient proliferation of SAv-primed T cells (Supplementary Fig. 4A) and because BM-DCs pre-incubated with GalNAc-SAv did not induce efficient proliferation of ovalbumin (OVA)-primed T cells (Supplementary Fig. 4B), we concluded that the efficient proliferation of SAv-primed T cells after co-culturing with GalNAc-SAv-pulsed BM-DCs was antigen specific.

SAv-primed T cells were shown to produce IFN-γ and IL-17A when co-cultured with BM-DCs pre-incubated with GalNAc-SAv (Fig. 7B). IL-4 and IL-10 were below the detectable levels (data not shown).

To test whether glycoproteins containing terminal GalNAc residues were able to induce efficient antigen-specific T cell proliferation, bio-MUC1 peptides with enzymatically-attached GalNAc residues were used. Flow cytometry confirmed that immature BM-DCs bound and internalized complexes of bio-9Tn-MUC1 peptides and FITC-SAv (data not shown). SAv conjugated with MUC1 peptides containing 9 or 14 GalNAc residues, but not with non-glycosylated MUC1 peptides, induced efficient proliferation of SAv-primed T cells in a dose dependent manner (Fig. 7C).

When SAv-primed T cells were separated as CD4+ cells and CD8+ cells and used for the antigen presentation assays, only SAv-primed CD4+ T cells were able to proliferate and secrete IL-17A (Fig. 7D and Supplementary Fig. 5). This CD4+-specific antigen presentation was not likely due to the immunization method; BM-DCs were able to cross-present SAv to CD8+ T cells when treated with CpG-oligodeoxynucleotide (ODN) (35) (Supplementary Fig. 5B).

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 8: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

8

The involvement of MGL1 or MGL2 in antigen presentation was directly examined using BM-DCs prepared from Mgl1-/-, Mgl2-/-, or WT littermate mice. Mgl1-/- BM-DCs pulsed with bio-GalNAc-PAA conjugated with SAv induced efficient T cell proliferation compared to SAv alone or bio-GlcNAc-PAA conjugated with SAv (Fig. 7E). In contrast, Mgl2-/- BM-DCs pulsed with bio-GalNAc-PAA conjugated with SAv did not induce T cell proliferation (Fig. 7F). As indicated by the surface expression levels of CD40 and CD86 analyzed using flow cytometry, the lack of MGL1 or MGL2 did not influence the maturation of BM-DCs after antigen uptake (data not shown); the increased antigen uptake and presentation due to GalNAc residues having high affinity to MGL2 is likely the basis for the efficient CD4+ T cell response.

DISCUSSION

The organ distribution of cells expressing mouse MGL2, a novel isolectin of MGL, was examined using a specific mAb URA-1, and the role of this lectin in the uptake and presentation of glycosylated antigens was assessed using DCs from Mgl2-/- mice. Site-directed mutagenesis of the MGL2-CRD clearly demonstrated that the Leu-61, Phe-110, and Asp-113 amino acid residues were required for mAb URA-1 binding (Supplementary Fig. 1A). Also, the results indicated that this antibody recognized a conformational epitope. As a consequence, denaturation of MGL2 by SDS treatment during Western blotting analysis abolished the mAb reactivity (data not shown). Leu-61, one of the amino acid residues required for antibody binding, was previously shown to be involved in ligand binding (29), supporting our finding that mAb URA-1 was capable of blocking the binding of ligands to MGL2 (Supplementary Fig. 1B).

The combined use of MGL2-specific mAb URA-1 and MGL1-specific mAb LOM-8.7 enabled us to examine the protein expression patterns of MGL1 and MGL2, respectively, for the first time. Both MGL1 and MGL2 were detected in the connective tissues of almost all organs; previous studies using cross-reactive mAb LOM-14 reported similar observations (18). However, cells expressing MGL2 were limited to a portion of MGL1-expressing cells, mostly cDCs.

In contrast, MGL1 was expressed on macrophages, cDCs, and pDCs. The fact that the distribution of MGL2 is more limited to cDCs than MGL1 strongly implies that the function of this molecule is closely linked to the role of cDCs. MGL2-positive cells were rare in BMs and spleens but were present in peripheral organs, including skin, LNs (30), large intestine (36), and lungs. It is known that splenic cDCs are mainly derived from precursor cells residing in the spleen and are formed independently of BM-derived or blood precursor cells under steady-state conditions (37,38). Therefore, the expression of MGL2 was likely to be induced after precursor cells migrated into peripheral tissues. Auffray and co-workers reported that when Gr-1-CX3CR1+ resting monocytes extravasated and invaded surrounding tissues in response to tissue damage, Mgl2 mRNA expression was upregulated (39). The expression of MGL1, but not MGL2, was observed on pDCs, which is consistent with the report by Cisse and co-workers showing that Mgl1 is predominantly expressed on pDCs compared to other cells in the spleen (40). Human MGL was reported to be expressed on CD11c+DR+Lin- DCs from fresh blood, but not on CD11c-DR+Lin- pDCs (41). Considering that the primary sequence and the carbohydrate specificity of mouse MGL2 are similar to those of human MGL, mouse MGL2 should represent the counterpart to human MGL.

It was previously reported that Mgl1 and Mgl2 gene expression was induced upon alternative activation of macrophages by Th2-associated cytokines such as IL-4 and/or IL-13 (34). Based on these observations, MGL1 and MGL2 were expected to serve as surface markers identifying alternatively activated macrophages. However, we clearly demonstrated in the present paper that MGL1 and MGL2 protein expression was not significantly upregulated on thioglycollate-induced PEC-macrophages after stimulation with IL-4 in vitro, even though the mRNA levels were elevated. It is possible that MGL1 and MGL2 protein expression may be upregulated under certain situations in vivo, including allergy and helminth infection, but this possibility remains to be further elucidated.

In the present study, we demonstrated that complexes of protein antigens having GalNAc

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 9: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

9

residues were efficiently internalized through MGL2 on immature BM-DCs and presented to antigen-specific CD4+ T cells. Such uptake and presentation were not likely to be mediated by MGL1, as shown by the comparison between BM-DCs from Mgl1-/- and Mgl2-/- mice. Recombinant MGL1 was previously shown to have affinity for GalNAc residues linked to a peptide (23). Furthermore, MGL1 or MGL2 alone on CHO cells was sufficient to bind and internalize GalNAc-PAA to a similar extent (Fig. 6A). However, when BM-DCs from Mgl1-/- and Mgl2-/- mice were compared for their binding and uptake of GalNAc-PAA, the decrease in GalNAc-PAA binding and uptake (Fig. 6B and 6C) corresponded to the surface levels of MGL2, but not those of MGL1. It appeared that cell surface expression of MGL2 on BM-DCs was required for the internalization of GalNAc-PAA. Because LeX-PAA, which is expected to mimic the ligand for MGL1, showed very low binding to MGL1 expressed on cell surfaces (data not shown), the involvement of MGL1 expressed on BM-macrophages or BM-DCs with the binding and uptake of carbohydrate ligands remains unclear.

It is widely accepted that DCs are capable of presenting antigens to both CD4+ and CD8+ T cells. However, BM-DCs pulsed with GalNAc-SAv stimulated SAv-primed CD4+ but not CD8+ T cells (Fig. 7D and Supplementary Fig. 5). Thus, we attempted to confirm that such cross-presentation did occur in the experimental system we employed. CpG-ODN treatment before antigen uptake was previously shown to induce cross-presentation by BM-DCs (35). Based on this report, BM-DCs were treated overnight with CpG-ODN and then incubated with SAv at a relatively high concentration (100 µg/ml) for 2 hr. The CpG-treated and SAv-pulsed BM-DCs induced IL-17A production from both CD4+ and CD8+ SAv-primed T cells (Supplementary Fig. 5B). The antigen presentation by CpG-treated BM-DCs pulsed with GalNAc-SAv was not examined because CpG-treatment induced maturation of BM-DCs and decreased the expression levels of MGL2. However, these results strongly suggest that antigens internalized through MGL2 are efficiently presented on MHC class II, but not on MHC class I.

MGL2 binds ligands at neutral pH, and the affinity was significantly lowered under acidic conditions (a half-maximal ligand binding pH 5.7 for MGL2) (16). Because the GalNAc-PAA internalized by immature BM-DCs has previously been shown to co-localize with LAMP-1 and MHC class II (19), ligands internalized through MGL2 should be transported and released in the late endosomes/lysosomes. These processes might be mediated by the tyrosine-based uptake motif (YENF) in the cytoplasmic tail. The role of the cytoplasmic motif in uptake and intracellular trafficking of MGL2 remains to be elucidated. OVA conjugated to anti-DEC-205 mAb was targeted to DCs in vivo and presented on MHC class I as well as MHC class II (42,43). On the other hand, antigens targeted to DEC-205 on BM-DCs were not presented on MHC class I (42). The possibility of antigen cross-presentation as a result of uptake through MGL2 should further be examined in vivo or by using cells isolated from mouse tissue, although MGL2 was not expressed on CD8+ cDCs (Fig. 3), which is thought to be an important subset for cross-presentation.

In vivo targeting is an important for achieving therapeutic immunization. To explore this possibility, we injected GalNAc-SAv subcutaneously and attempted to examine antigen-specific T cell proliferation in vitro. However, SAv itself inhibited T cell proliferation in vitro by unknown reasons, and this system was found unsuited to study the enhancement of immune responses (Denda-Nagai unpublished data). Novel strategies without SAv should be developed to investigate the use of glycoforms in immunotherapy.

To date, only a limited number of reports have demonstrated that glycosylated antigens are efficiently taken up for presentation by DCs through C-type lectins. Mannosylated-BSA or mannosylated peptides were presented more efficiently than non-mannosylated forms by monocyte-derived DCs (44). Such efficient uptake of mannosylated antigens was likely to be mediated by MR and/or DC-SIGN. Soluble OVA, known to induce a T cell response, was reported to be efficiently internalized by MR for cross-presentation (45,46). In the present study, we demonstrated that tumor antigen MUC1 with GalNAc residues was recognized and internalized by immature BM-DCs, and SAv conjugated to

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 10: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

10

MUC1 with GalNAc was presented to T cells. Furthermore, human MGL on monocyte-derived DCs is involved in the binding and uptake of Tn-MUC1 (13). Altered O-glycans, such as the exposed GalNAc residues on this mucin, are known to be associated with tumor cells. MUC1 that has a sialylated TF or sialylated Tn, both of which are also expressed by carcinoma cells, is known to impair maturation of DCs and induce IL-10highIL-12low regulatory DCs; however, the receptor required for the recognition of sialylated MUC1 is unknown (47,48). On the other hand, antigens having GalNAc residues did not impair the maturation of BM-DCs (data not shown) and did not seem to induce regulatory DCs; we did not observe IL-10 production after co-culturing T cells and GalNAc-SAv-pulsed BM-DCs in the present study. Thus, the question of whether MGL might be involved in tumor surveillance or induction of self-tolerance in the absence/presence of adequate stimuli in vivo should be answered through further investigation.

In conclusion, we demonstrated that MGL2 was preferentially expressed on cDCs and involved in the efficient uptake and presentation of antigens with GalNAc residues. Targeting of MGL by the addition of GalNAc residues to antigens should contribute to novel vaccine development.

Acknowledgements

We thank Ms. Kyoko Sakai and Ms. Miki

Noji for their assistance in the preparation of this manuscript and Ms. Miho Mori and Ms. Satomi Yoshinaga for animal care. We thank Dr. Kayo Inaba for her advice in generating BM-DCs and antigen presentation assays, Dr. Henrik Clausen for providing biotinylated MUC1 peptides, Dr. Stephan Hedrick for providing Mgl1-/- mice, and Kirin Brewery Co., Ltd. for supplying GM-CSF. We also thank Dr. Gen Yamada, Center for Animal Resources and Development, Kumamoto University, for his kind help in generating Mgl2-/- mice.

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 11: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

11

REFERENCES

1. Apweiler, R., Hermjakob, H., and Sharon, N. (1999) Biochim Biophys Acta 1473, 4-8 2. Banchereau, J., and Steinman, R. M. (1998) Nature 392, 245-252 3. Cambi, A., and Figdor, C. G. (2003) Curr Opin Cell Biol 15, 539-546 4. Figdor, C. G., van Kooyk, Y., and Adema, G. J. (2002) Nat Rev Immunol 2, 77-84 5. Robinson, M. J., Sancho, D., Slack, E. C., LeibundGut-Landmann, S., and Reis e Sousa, C.

(2006) Nat Immunol 7, 1258-1265 6. Carter, R. W., Thompson, C., Reid, D. M., Wong, S. Y., and Tough, D. F. (2006) Cell Immunol

239, 87-91 7. Idoyaga, J., Cheong, C., Suda, K., Suda, N., Kim, J. Y., Lee, H., Park, C. G., and Steinman, R. M.

(2008) J Immunol 180, 3647-3650 8. Tacken, P. J., de Vries, I. J., Torensma, R., and Figdor, C. G. (2007) Nat Rev Immunol 7, 790-802 9. van Vliet, S. J., Aarnoudse, C. A., Broks-van den Berg, V. C., Boks, M., Geijtenbeek, T. B., and

van Kooyk, Y. (2007) Eur J Immunol 37, 2075-2081 10. Springer, G. F. (1997) J Mol Med 75, 594-602 11. Denda-Nagai, K., and Irimura, T. (2000) Glycoconj J 17, 649-658 12. von Mensdorff-Pouilly, S., Verstraeten, A. A., Kenemans, P., Snijdewint, F. G., Kok, A., Van

Kamp, G. J., Paul, M. A., Van Diest, P. J., Meijer, S., and Hilgers, J. (2000) J Clin Oncol 18, 574-583

13. Napoletano, C., Rughetti, A., Agervig Tarp, M. P., Coleman, J., Bennett, E. P., Picco, G., Sale, P., Denda-Nagai, K., Irimura, T., Mandel, U., Clausen, H., Frati, L., Taylor-Papadimitriou, J., Burchell, J., and Nuti, M. (2007) Cancer Res 67, 8358-8367

14. Suzuki, N., Yamamoto, K., Toyoshima, S., Osawa, T., and Irimura, T. (1996) J Immunol 156, 128-135

15. Sato, M., Kawakami, K., Osawa, T., and Toyoshima, S. (1992) J Biochem 111, 331-336 16. Tsuiji, M., Fujimori, M., Ohashi, Y., Higashi, N., Onami, T. M., Hedrick, S. M., and Irimura, T.

(2002) J Biol Chem 277, 28892-28901 17. Imai, Y., Akimoto, Y., Mizuochi, S., Kimura, T., Hirano, H., and Irimura, T. (1995) Immunology

86, 591-598 18. Mizuochi, S., Akimoto, Y., Imai, Y., Hirano, H., and Irimura, T. (1997) Glycobiology 7, 137-146 19. Denda-Nagai, K., Kubota, N., Tsuiji, M., Kamata, M., and Irimura, T. (2002) Glycobiology 12,

443-450 20. Higashi, N., Fujioka, K., Denda-Nagai, K., Hashimoto, S., Nagai, S., Sato, T., Fujita, Y.,

Morikawa, A., Tsuiji, M., Miyata-Takeuchi, M., Sano, Y., Suzuki, N., Yamamoto, K., Matsushima, K., and Irimura, T. (2002) J Biol Chem 277, 20686-20693

21. Dupasquier, M., Stoitzner, P., Wan, H., Cerqueira, D., van Oudenaren, A., Voerman, J. S., Denda-Nagai, K., Irimura, T., Raes, G., Romani, N., and Leenen, P. J. (2006) J Leukoc Biol 80, 838-849

22. Iida, S., Yamamoto, K., and Irimura, T. (1999) J Biol Chem 274, 10697-10705 23. Yamamoto, K., Ishida, C., Shinohara, Y., Hasegawa, Y., Konami, Y., Osawa, T., and Irimura, T.

(1994) Biochemistry 33, 8159-8166 24. Onami, T. M., Lin, M. Y., Page, D. M., Reynolds, S. A., Katayama, C. D., Marth, J. D., Irimura,

T., Varki, A., Varki, N., and Hedrick, S. M. (2002) Mol Cell Biol 22, 5173-5181 25. Kimura, T., Imai, Y., and Irimura, T. (1995) J Biol Chem 270, 16056-16062 26. Corti, M., Brody, A. R., and Harrison, J. H. (1996) Am J Respir Cell Mol Biol 14, 309-315 27. Bovin, N. V. (1998) Glycoconj J 15, 431-446 28. Wandall, H. H., Hassan, H., Mirgorodskaya, E., Kristensen, A. K., Roepstorff, P., Bennett, E. P.,

Nielsen, P. A., Hollingsworth, M. A., Burchell, J., Taylor-Papadimitriou, J., and Clausen, H. (1997) J Biol Chem 272, 23503-23514

29. Oo-Puthinan, S., Maenuma, K., Sakakura, M., Denda-Nagai, K., Tsuiji, M., Shimada, I.,

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 12: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

12

Nakamura-Tsuruta, S., Hirabayashi, J., Bovin, N. V., and Irimura, T. (2008) Biochim Biophys Acta 1780, 89-100

30. Kumamoto, Y., Denda-Nagai, K., Aida, S., Higashi, N., and Irimura, T. (2009) PLoS One 4, e5619 31. Asselin-Paturel, C., Boonstra, A., Dalod, M., Durand, I., Yessaad, N., Dezutter-Dambuyant, C.,

Vicari, A., O'Garra, A., Biron, C., Briére, F., and Trinchieri, G. (2001) Nat Immunol 2, 1144-1150 32. Krug, A., French, A. R., Barchet, W., Fischer, J. A., Dzionek, A., Pingel, J. T., Orihuela, M. M.,

Akira, S., Yokoyama, W. M., and Colonna, M. (2004) Immunity 21, 107-119 33. Nakano, H., Yanagita, M., and Gunn, M. D. (2001) J Exp Med 194, 1171-1178 34. Raes, G., Brys, L., Dahal, B. K., Brandt, J., Grooten, J., Brombacher, F., Vanham, G., Noel, W.,

Bogaert, P., Boonefaes, T., Kindt, A., Van den Bergh, R., Leenen, P. J., De Baetselier, P., and Ghassabeh, G. H. (2005) J Leukoc Biol 77, 321-327

35. Datta, S. K., Redecke, V., Prilliman, K. R., Takabayashi, K., Corr, M., Tallant, T., DiDonato, J., Dziarski, R., Akira, S., Schoenberger, S. P., and Raz, E. (2003) J Immunol 170, 4102-4110

36. Saba, K., Denda-Nagai, K., and Irimura, T. (2009) Am J Pathol 174, 144-152 37. Naik, S. H., Metcalf, D., van Nieuwenhuijze, A., Wicks, I., Wu, L., O'Keeffe, M., and Shortman,

K. (2006) Nat Immunol 7, 663-671 38. Varol, C., Landsman, L., Fogg, D. K., Greenshtein, L., Gildor, B., Margalit, R., Kalchenko, V.,

Geissmann, F., and Jung, S. (2007) J Exp Med 204, 171-180 39. Auffray, C., Fogg, D., Garfa, M., Elain, G., Join-Lambert, O., Kayal, S., Sarnacki, S., Cumano, A.,

Lauvau, G., and Geissmann, F. (2007) Science 317, 666-670 40. Cisse, B., Caton, M. L., Lehner, M., Maeda, T., Scheu, S., Locksley, R., Holmberg, D., Zweier, C.,

den Hollander, N. S., Kant, S. G., Holter, W., Rauch, A., Zhuang, Y., and Reizis, B. (2008) Cell 135, 37-48

41. Clark, G., Munster, D., Yusuf, S., and Hart, D. N. (2005) Cell Immunol 236, 21-28 42. Bonifaz, L., Bonnyay, D., Mahnke, K., Rivera, M., Nussenzweig, M. C., and Steinman, R. M.

(2002) J Exp Med 196, 1627-1638 43. Bonifaz, L. C., Bonnyay, D. P., Charalambous, A., Darguste, D. I., Fujii, S., Soares, H., Brimnes,

M. K., Moltedo, B., Moran, T. M., and Steinman, R. M. (2004) J Exp Med 199, 815-824 44. Tan, M. C., Mommaas, A. M., Drijfhout, J. W., Jordens, R., Onderwater, J. J., Verwoerd, D.,

Mulder, A. A., van der Heiden, A. N., Scheidegger, D., Oomen, L. C., Ottenhoff, T. H., Tulp, A., Neefjes, J. J., and Koning, F. (1997) Eur J Immunol 27, 2426-2435

45. Burgdorf, S., Kautz, A., Bohnert, V., Knolle, P. A., and Kurts, C. (2007) Science 316, 612-616 46. Burgdorf, S., Lukacs-Kornek, V., and Kurts, C. (2006) J Immunol 176, 6770-6776 47. Monti, P., Leone, B. E., Zerbi, A., Balzano, G., Cainarca, S., Sordi, V., Pontillo, M., Mercalli, A.,

Di Carlo, V., Allavena, P., and Piemonti, L. (2004) J Immunol 172, 7341-7349 48. Rughetti, A., Pellicciotta, I., Biffoni, M., Backstrom, M., Link, T., Bennet, E. P., Clausen, H., Noll,

T., Hansson, G. C., Burchell, J. M., Frati, L., Taylor-Papadimitriou, J., and Nuti, M. (2005) J Immunol 174, 7764-7772

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 13: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

13

Footnotes

This work was supported in part by grants-in-aid from the Ministry of Education, Science, Sports and Culture of Japan, the Research Association for Biotechnology, and the Program for Promotion of Fundamental Studies in Health Sciences of the Pharmaceutical and Medical Device Agency. Daisuke Sugiura is a recipient of the JSPS Research Fellowship for Young Scientists.

Abbreviations used are: APC, allophycocyanin; BM, bone marrow; cDC, conventional DC; CRD, carbohydrate recognition domain; DC, dendritic cell; DPBS, Dulbecco’s modified phosphate buffered saline; ECD, extracellular domain; ELISA, enzyme-linked immunosorbent assay; FITC, fluorescein isothiocyanate; Gal, galactose; GalNAc, N-acetylgalactosamine; GlcNAc, N-acetylglucosamin; GM-CSF, granulocyte-macrophage colony-stimulating factor; LeX, Lewis X; LN, lymph node; mAb, monoclonal antibody; MFI, mean fluorescence intensity; MGL, macrophage galactose-type calcium-type lectin; MHC, major histocompatibility complex; MR, mannose receptor; ODN, oligodeoxynucleotide; OVA, ovalbumin; PAA, polyacrylamide polymer; pDC, plasmacytoid DC; PE, phycoerythrin; PEC, peritoneal exudate cells; TF, Thomsen-Friedenreich; SAv, streptavidin; SPF, specific pathogen free; and WT, wild-type.

Figure Legends

Figure 1: Specificity of mAb URA-1 for MGL2 compared with mAb LOM-14 and mAb LOM-8.7. (A) Binding of mAb LOM-14 (anti-MGL1/MGL2: open triangle), mAb LOM-8.7 (anti-MGL1: open circle), and mAb URA-1 (anti-MGL2: open square) to MGL1-ECD or MGL2-ECD in the ELISA are shown. Hybridoma culture supernatants were serially diluted as indicated. Data are shown as the mean ± SD of triplicates. (B) Binding profiles of the same mAbs to CHO cells transfected with Mgl1 or Mgl2 cDNA using flow cytometric analysis are shown. The binding profiles of control rat IgG are shown by gray-filled lines. The binding profiles of these three mAbs indicated above are shown by black lines. Figure 2: Immunohistochemical localizations of binding sites for mAb LOM-8.7 and mAb URA-1 in various tissues. (A) Thymus. (B) Spleen. (C) Lung. The distribution of cells stained with these antibodies indicated by arrowheads was limited within connective tissues in these organs. The apparent relative incidence of mAb LOM-8.7- and mAb URA-1-positive cells was similar in thymi. The apparent relative incidence was greater with mAb LOM-8.7 than with mAb URA-1 in spleens, whereas it was slightly greater with mAb URA-1 than with mAb LOM-8.7 in lungs. Scale bars represent 50 µm for (A) and (C) and 100 µm for (B). Figure 3: Expression of MGL1 and MGL2 on cells from BM, spleens, and lungs analyzed by flow cytometry. (A) Staining profiles of anti-CD11c and bio-LOM-8.7 followed by PE-Cy7-SAv are shown. (B) Staining profiles of anti-CD11c and APC-URA-1 are shown. (C) Staining profiles of bio-LOM-8.7 followed by PE-Cy7-SAv and APC-URA-1 are shown. In panels (D)–(F), MGL1 and MGL2 double-positive cells and MGL1 single-positive cells gated in (C) were further analyzed for the expression of CD11c and other surface markers: (D) cells from BM, (E) cells from spleens, and (F) cells from lungs. In all panels, cells were analyzed after excluding dead cells and CD3+ or CD19+ cells. The data are representative of three or more independent experiments. Figure 4: Expression of MGL1 and MGL2 on surface of macrophages analyzed by flow cytometry. (A) BM-macrophages. (B) PEC-macrophages. (C) PEC-macrophages cultured overnight in the presence or absence of indicated cytokines. In all panels, the profiles obtained with isotype controls are shown as gray-filled lines, and the profiles with mAbs LOM-8.7 and URA-1 are shown as black lines. Geometric MFI for the mAb binding and isotype control are shown in each panel and at the right of each panel, respectively.

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 14: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

14

Figure 5: Expression of MGL1 and MGL2 on surface of CD11c+ BM-DCs. (A) BM-DCs were prepared from BM cells of WT littermate of Mgl1-/- mice (Mgl1+/+). (B) BM-DCs were prepared from BM cells of Mgl1-/- mice. (C) BM-DCs were prepared from BM cells of WT littermate of Mgl2-/- mice (Mgl2+/+). (D) BM-DCs were prepared from BM cells of Mgl2 -/- mice. In all panels, CD11c+ cells were obtained by MACS, and bindings of mAbs LOM-8.7 and URA-1 were determined by flow cytometry. The data are representative of three or more independent experiments. (E) MGL2 expression profiles of Mgl1+/+

BM-DCs (black line derived from the right panel in A) and Mgl1-/- BM-DCs (gray-filled line derived from the right panel in B) are shown as overlaid histograms. (F) MGL1 expression profiles of Mgl2+/+

BM-DCs (black line derived from the middle panel in C) and Mgl2-/- BM-DCs (gray-filled line derived from the middle panel in D) are shown as overlaid histograms. In panels (E) and (F), MFI for the mAb binding are shown under each panel. Figure 6: Binding and uptake of GalNAc-PAA by CHO cells transfected with Mgl1 or Mgl2 cDNA and by CD11c+ BM-DCs. (A) Flow cytometric profiles of CHO transfectants with FITC-PAA are shown. The cells were incubated with FITC-GlcNAc-PAA (dotted lines) or FITC-GalNAc-PAA (thin lines) on ice for 30 min to estimate the amount bound to cell surfaces. The relative amounts of FITC-GalNAc-PAA internalized by the cells were estimated as follows. The cells were incubated with FITC-GalNAc-PAA at 37°C for 30 min, treated with EDTA to remove cell surface-associated FITC-GalNAc-PAA, and then analyzed (bold lines). The cells incubated with FITC-GalNAc-PAA on ice for 30 min and then treated with EDTA were also analyzed as a control (gray-filled). (B) BM-DCs were prepared from BM cells of Mgl1-/- or WT littermate (Mgl1+/+) mice. (C) BM-DCs were prepared from BM cells of Mgl2-/- or WT littermate (Mgl2+/+) mice. In panels (B) and (C), CD11c+ cells were obtained by MACS. Binding and uptake of FITC-PAA were examined using flow cytometry by the same methods as in (A). Differences in the geometric MFI of FITC after uptake at 37°C and binding on ice are shown. In all panels the data are representative of three independent experiments. Figure 7: T cell activation through antigen presentation by BM-DCs pulsed with bio-GalNAc-PAA conjugated to SAv. (A) SAv-primed T cells were co-cultured with immature BM-DCs pulsed with bio-GalNAc-PAA conjugated to SAv (GalNAc-SAv: closed circle), bio-GlcNAc-PAA conjugated to SAv (GlcNAc-SAv: closed triangle), SAv (open square), or no antigen (open circle). The DC to T cell ratio was 4:1. T cell proliferation was measured by [3H]-thymidine uptake. Data are shown as the mean cpm ± SD of triplicate cultures. **p < 0.01 and ***p < 0.001 for GalNAc-SAv compared to GlcNAc-SAv at different SAv concentrations. (B) Production of cytokines was measured with culture supernatants of SAv-primed T cells co-cultured with CD11c+ BM-DCs. IFN-γ and IL-17A were detected and quantified. BM-DCs were pulsed with GalNAc-SAv (black bar), GlcNAc-SAv (dark gray bar), or no antigen (open bar). The DC to T cell ratio was 10:1. Data are shown as the mean concentration (pg/ml) ± SD of triplicate cultures. *p < 0.05, and ***p < 0.001 for GalNAc-SAv compared to GlcNAc-SAv. (C) SAv-primed T cells were co-cultured with immature BM-DCs pulsed with bio-14Tn-MUC1 and SAv (14Tn-MUC1-SAv: closed diamond), bio-9Tn-MUC1 and SAv (9Tn-MUC1-SAv: closed circle), bio-MUC1 and SAv (MUC1-SAv: closed triangle), SAv (open square), or no antigen (open circle). The DC to T cell ratio was 4:1. T cell proliferation was measured by [3H]-thymidine uptake, and the data are shown as the mean cpm ± SD of triplicate cultures. **p < 0.01 and ***p < 0.001 for 14Tn-MUC1-SAv or 9Tn-MUC1-SAv compared to MUC1-SAv, respectively. (D) Nylon wool (NW)-purified, CD4+, and CD8+ T cells obtained from SAv-immunized mice were compared for their proliferation response to GalNAc-PAA-mediated antigen presentation by co-culturing them with CD11C+ BM-DCs pulsed with GalNAc-SAv (black bar), GlcNAc-SAv (dark gray bar), SAv (light gray bar), or no antigen (open bar). The DC to T cell ratio was 4:1. T cell proliferation was measured by [3H]-thymidine uptake, and the data are shown as the mean cpm ± SD of triplicate cultures. **p < 0.01, ***p < 0.001, and n.s. (not significant) for GalNAc-SAv compared to no antigen, GlcNAc-SAv, or SAv. (E) SAv-primed T cells were co-cultured with CD11c+ BM-DCs from Mgl1-/- or WT littermate (Mgl1+/+) mice. (F) SAv-primed T cells were co-cultured with CD11c+ BM-DCs from Mgl2-/- or WT littermate (Mgl2+/+) mice. In panels (E) and

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 15: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

15

(F), CD11c+ BM-DCs were pulsed with GalNAc-SAv (black bar), GlcNAc-SAv (dark gray bar), SAv (light gray bar), or no antigen (open bar). The DC to T cell ratio was 75:1. T cell proliferation was measured by [3H]-thymidine uptake, and the data are shown as the mean cpm ± SD of triplicate cultures. *p < 0.05, **p < 0.01, ***p < 0.001, and n.s. (not significant) for GalNAc-SAv compared to no antigen, GlcNAc-SAv, or SAv.

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 16: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

AMGL1 MGL2 LOM-14

LOM-8.7URA-1

DilutionAbso

rban

ce 4

05 n

m 1.00.80.60.40.20.0

10-1 10-2 10-3 10-4 10-5

Dilution

1.00.80.60.40.20.0

1.2

10-1 10-2 10-3 10-4 10-5

B

Cel

lsCHO-mock

CHO-MGL1

Cel

ls

CHO-MGL2

Cel

ls

LOM-14(MGL1/MGL2)

0

100

200

300

10-1 100 101 102 1030

100

200

300

LOM-8.7 (MGL1)

0

100

200

300

0

100

200

300

10-1 100 101 102 1030

100

200

300

URA-1(MGL2)

0

100

200

300

0

100

200

300

10-1 100 101 102 1030

100

200

3000

100

200

300

Denda-Nagai et al.Figure 1

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 17: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

A

C

B

rat IgG2a LOM-8.7 (MGL1) URA-1 (MGL2)

Denda-Nagai et al.Figure 2

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 18: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

Denda-Nagai et al.Figure 3

Isot

ype

MG

L1BM

Spleen

Lung

A

Isot

ype

MG

L2

MG

L2

Isot

ype

BM

Spleen0.16 43.7

55.50.63

0.44 7.99

90.11.45

1.06 98.3

0.60

0.47 46

52.60.94

0.62 11.4

87.30.62

0.3 37.4

61.40.9

0.95 98.7

0.320

0.3 70.2

29.10.45

MGL1+MGL2+

2.11 32.8

59.45.64

2.96 29.4

62.15.58

0.35 8.92

83.57.27

4.95 61.3

30.33.46

0.41 6.99

85.47.21

4.18 64

28.43.42

1.22 13.4

79.16.33

5.77 63.5

27.43.39

MGL1+MGL2–

Lung0 6.23

91.52.29

2.58 97.4

00

0.18 11.5

86.91.44

0 0.96

96.62.44

0.44 38.5

59.11.95

2.06 97.9

00

0.39 43.6

54.51.54

0 1.55

96.42.06

MGL1+MGL2+

D

E

F

CD4 CD8 CD11b CD40 B220 CD86 MHCII F4/800 0.71

75.823.5

0 3.63

75.620.7

MGL1+MGL2+

0 3.06

73.223.8

0 0.21

77.722.1

23.5 76.3

00.26

0 9.13

6822.8

20.2 77.5

0.581.75

5.73 34.1

44.615.5

MGL1+MGL2–

0.87 57.1

36.45.64

0.27 6.1

86.86.86

3.32 12.7

81.32.68

0.11 1.47

917.47

0.91 78.5

14.65.98

0.13 1.7

90.87.36

3.45 28.4

64.63.5

2.12 4.92

87.25.77

CD11c

Isotype

0.025 0

0.04899.9

0.36 0.034

0.3199.3

MGL1

0.61

14

0.053

1.28

1.81

IsotypeIsotype

7.29e-3 2.11e-4

0.04799.9

0.014 7.92e-4

0.09799.90.18 0.044

0.1299.7CD11c

0.25 1.37

3.3795

1.68 13.7

36.548.11.36 0.66

12.285.7

0.32 0.011

0.2399.4

0.29 0.057

4.6895

0.062 0.64

46.3532.05 1.25

11.385.4CD11c

B C

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 19: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

0102 103 104 1050

100

200

300

0102 103 104 1050

100

200

300

0102 103 104 1050

100

200

300

0102 103 104 1050

100

200

300

0102 103 104 1050

100

200

300

LOM-8.7(MGL1)

URA-1(MGL2)

0102 103 104 1050

100

200

300C

ells

0102 103 104 1050

100

200

300

Cel

lsA

B

C

0102 103 104 1050

100

200

300

Cel

lsnone

0102 103 104 1050

100

200

300

Cel

lsIL-4

0102 103 104 1050

100

200

300

Cel

lsIFN-γ

MFI (Isotype Ctrl)

86.7

81.3

175

174

158

1310

1925

1009

1058

707

115

358

538

440

510

Denda-Nagai et al.Figure 4

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 20: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

0102 103 104 105

0102

103

104

105 63.6 0.48

0.1135.80102 103 104 105

0102

103

104

105 9.29 54.3

22.513.90102 103 104 105

0102

103

104

105 6.85 46.2

32.414.6

AM

HC

II

Mgl1+/+

0102 103 104 105

0102

103

104

105 71.8 0.54

0.1427.50102 103 104 105

0102

103

104

105 66.9 1.01

0.2731.80102 103 104 105

0102

103

104

105 13.4 56.2

17.113.3

MH

CIIB

Mgl1–/–

0102 103 104 105

0102

103

104

105 66.4 0.53

0.3432.70102 103 104 105

0102

103

104

105 11.2 63.7

16.78.280102 10 3 104 105

0102

103

104

105 7.63 66.3

16.59.59

C

MH

CII

Mgl2+/+

rat IgG2a LOM-8.7(MGL1)

URA-1(MGL2)

0102 103 104 105

0102

103

104

105 54.8 0.48

0.4544.20102 103 104 105

0102

103

104

105 29.6 23.5

22.824.0

D

MH

CII

Mgl2–/–

0102 103 104 105

0102

103

104

105 55.7 0.71

0.4743.1

Cel

ls

0102 103 104 1050

20

40

60

80

100

E URA-1(MGL2)

Mgl1+/+: 2690Mgl1–/–: 1679

LOM-8.7(MGL1)

0102 103 104 1050

30

60

90

120

Cel

ls

Mgl2+/+: 1717Mgl2–/–: 399

F

Denda-Nagai et al.Figure 5

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 21: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

AC

ells

MFI

B 3000

2500

2000

1000

0

1500

500

GlcNAc-PAAMgl1+/+

GalNAc-PAAMgl1+/+Mgl1–/– Mgl1–/–

4°C, 30 min37°C, 30 min37°C, 60 min37°C, 120 min

MFI

C 3000

2500

2000

1000

0

1500

500

GlcNAc-PAAMgl2+/+

GalNAc-PAAMgl2+/+Mgl2–/– Mgl2–/–

4°C, 30 min37°C, 30 min37°C, 60 min37°C, 120 min

CHO-mock

10-1 100 101 102 1030

100

200

300

CHO-MGL2

10-1 100 101 102 1030

100

200

300

CHO-MGL1

10-1 100 101 102 1030

100

200

300

Denda-Nagai et al.Figure 6

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 22: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

0

20000

40000

60000

80000

100000

0.4 10 50

cpm

20Streptavidin conc. (µg/ml)

**

*****

GalNAc-SAvGlcNAc-SAvSAvno antigen

A

14Tn-MUC1-SAv

MUC1-SAvSAvno antigen

9Tn-MUC1-SAv

0

10000

20000

30000

40000

50000

60000

70000

0.4 2 10

cpm

0Streptavidin conc. (µg/ml)

***

*** **

***** **

CD

NW CD4 CD80

20000

40000

60000

80000

cpm

**

n.s.

*********** no antigen

SAvGlcNAc-SAvGalNAc-SAv

Fno antigenSAvGlcNAc-SAvGalNAc-SAv

0

20000

40000

60000

Mgl2+/+ Mgl2–/–

cpm

n.s.

*********E

Mgl1+/+ Mgl1–/–

cpm

0

10000

20000

30000

40000

************* no antigen

SAvGlcNAc-SAvGalNAc-SAv

Bno antigenGlcNAc-SAvGalNAc-SAv

400

300

200

100

0

IFN

-γ (p

g/m

l)

* 900

600

IL-1

7A (p

g/m

l)

800700

500400300200100

0

***

Denda-Nagai et al.Figure 7

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 23: Distribution and Function of Macrophage Galactose-type C ... · (MGL2/CD301b): Efficient Uptake and Presentation of Glycosylated Antigens by Dendritic Cells Kaori Denda-Nagai1, Satoshi

IrimuraAkihiko Kudo, Yoshihiro Akimoto, Hayato Kawakami, Nicolai V. Bovin and TatsuroOo-puthinan, Makoto Tsuiji, Akiko Morikawa, Yosuke Kumamoto, Daisuke Sugiura,

Kaori Denda-Nagai, Satoshi Aida, Kengo Saba, Kiwamu Suzuki, Saya Moriyama, Sarawutdendritic cells

(MGL2/CD301b): Efficient uptake and presentation of glycosylated antigens by Distribution and function of macrophage galactose-type C-type lectin 2

published online March 19, 2010J. Biol. Chem. 

  10.1074/jbc.M110.113613Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

Supplemental material:

  http://www.jbc.org/content/suppl/2010/03/19/M110.113613.DC1

by guest on August 11, 2019

http://ww

w.jbc.org/

Dow

nloaded from