Skip to main content

Main menu

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons

User menu

  • Subscribe
  • Log in

Search

  • Advanced search
The Journal of Immunology
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons
  • Subscribe
  • Log in
The Journal of Immunology

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Follow The Journal of Immunology on Twitter
  • Follow The Journal of Immunology on RSS

Cutting Edge: Langerin/CD207 Receptor on Dendritic Cells Mediates Efficient Antigen Presentation on MHC I and II Products In Vivo

Juliana Idoyaga, Cheolho Cheong, Koji Suda, Nao Suda, Jae Y. Kim, Haekyung Lee, Chae Gyu Park and Ralph M. Steinman
J Immunol March 15, 2008, 180 (6) 3647-3650; DOI: https://doi.org/10.4049/jimmunol.180.6.3647
Juliana Idoyaga
Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Cheolho Cheong
Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Koji Suda
Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Nao Suda
Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jae Y. Kim
Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Haekyung Lee
Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Chae Gyu Park
Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ralph M. Steinman
Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

The targeted delivery of Ags to dendritic cell (DCs) in vivo greatly improves the efficiency of Ag presentation to T cells and allows an analysis of receptor function. To evaluate the function of Langerin/CD207, a receptor expressed by subsets of DCs that frequently coexpress the DEC205/CD205 receptor, we genetically introduced OVA into the C terminus of anti-receptor Ab H chains. Taking advantage of the new L31 mAb to the extracellular domain of mouse Langerin, we find that the hybrid Ab targets appropriate DC subsets in draining lymph nodes and spleen. OVA is then presented efficiently to CD8+ and CD4+ T cells in vivo, which undergo 4–8 cycles of division in 3 days. Peptide MHC I and II complexes persist for days. Dose response studies indicate only modest differences between Langerin and DEC receptors in these functions. Thus, Langerin effectively mediates Ag presentation.

Dendritic cells (DCs)5 are specialized APCs able to control T cell immunity in vivo (1). Several DC subpopulations are known (2). These typically express distinct receptors for Ag uptake and capacities for Ag processing (3). Many differentially expressed receptors include molecules with C-type lectin domains (4) such as DEC205/CD205 on CD8+ DCs, dermal DCs, and Langerhans cells (LCs) and 33D1/DCIR2 on CD8− cells (2, 3, 5, 6).

Langerin/CD207 is a type II transmembrane protein (5). Originally, Langerin was thought to be LC-specific, but it is expressed on other DCs, especially CD8+ DEC+ DCs in spleen (7) and skin draining lymph nodes (pLN), although at lower levels (6, 8, 9). The expression of Langerin by CD8+DEC+ DCs is primarily seen in BALB/c mice but is much reduced in C57BL/6 mice (8). Langerin recognizes mannose, n-acetylglucosamine, fucose, and sulfated sugars (7, 10, 11). Langerin also binds pathogens such as HIV (12) and Candida albicans (13). Langerin is responsible for Birbeck granule formation within LCs (5). Both Birbeck granules and Langerin are involved in endocytosis (5, 14). In human epidermal LCs, Langerin is predominantly found in an endosomal recycling compartment (14). In murine epidermis, Langerin was found to colocalize with MHC II in situ (15). Abs to Langerin can be internalized (5), but it is not known whether this mediates Ag presentation to T cells.

A new way to assess receptor function, including in vivo, is to deliver Ags within anti-receptor mAbs, which selectively bind to the receptor and initiate Ag uptake, processing, and presentation on MHC products. Depending on the maturation state of the targeted DCs, receptor-mediated targeting enhances or inhibits T cell responses (3, 16, 17, 18). The role of DC subsets expressing select receptors, as well as the role of the receptors themselves in Ag presentation, might therefore be determined by using specific mAbs as Ag carriers.

In this study, we use a newly developed mAb to the extracellular domain of mouse Langerin to begin to study the consequences of Ag uptake via Langerin in vivo (8). We have cloned the H and L chains of this mAb and engineered the H chain to encode OVA. We will show that OVA targeting to Langerin results in efficient presentation to OVA-specific CD4+ and CD8+ transgenic T cells.

Materials and Methods

Mice

C57BL/6J and BALB/cJ mice were purchased from Charles River or Jackson Laboratories and used at 6–8 wk of age. OVA-specific, TCR transgenic, OT-I (C57BL/6-Tg(TcraTcrb)1100Mjb/J) and OT-II (C57BL/6-Tg(TcraTcrb)425Cbn/J) mice were used as described (17). All animals were maintained under specific pathogen-free conditions and used under institutional guidelines.

Reagents

Anti-CD16/32, FITC-anti-CD11c, allophycocyanin-anti-CD8, anti-CD4, and PE-Vα2 were from BD Biosciences or eBioscience. Other reagents were RPMI 1640 and DMEM (Invitrogen Life Technologies/GIBCO), ACK (ammonium chloride potassium) lysing buffer (BioSource International), 30% BSA Solution (Sigma-Aldrich), Alexa Fluor 647 labeling kit (Invitrogen Life Technologies/Molecular Probes), and EZ-Link Biotin (Pierce).

Cloning, engineering and production of anti-Langerin Ab

Cloning of V regions from rat mAbs and engineering of hybrid mAbs with mouse IgG1 H and κ light constant regions was described for mAbs III/10 (control Ig), NLDC145 (anti-DEC205) (16), and 33D1 (anti-DCIR2) (3). Similarly, the V regions of anti-mouse Langerin mAb (8) were cloned from RNAs of the L31 hybridoma with the 5′ RACE system (Invitrogen Life Technologies) using rat IgG2a-specific oligonucleotides (5′-CATCCCAGGGTCACCATGGA-3′ and 5′-CTGTTGTTTCAGCTGAGGAGACTG-3′) and rat κ-specific oligonucleotides (5′-CAACCTCACAGGTATAGAG-3′ and 5′-GAAGATGGATACAGTTGGTGCAGCATC-3′) following the manufacturer’s instructions. An OLLAS epitope sequence was inserted as a linker between the H chain and OVA for the detection of hybrid mAbs by an anti-OLLAS tag mAb (19). Hybrid mAbs were produced (3) and receptor binding was verified using Chinese hamster ovary (CHO) transfected cells by FACS.

Adoptive transfer and T cell proliferation responses

OVA-specific transgenic CD8+ or CD4+ T cells were prepared from lymph node and spleen cell suspensions of OT-I or OT-II mice (17). T cell proliferation was evaluated by injecting individual mice with either 2–3 × 106 OT-I or 3–5 × 106 OT-II cells, labeled at 107 cells/ml with 5 μM CFSE (Invitrogen/Molecular Probes) for 10 min at 37°C. Targeting mAbs were injected s.c. 24 h later. CFSE dilution was evaluated 3 days later by FACS after surface staining for CD4 (OT-II) or CD8 (OT-I) and Vα2 (OVA-specific TCR). To address Ag persistence, mAbs were injected 14, 3, or 1 day before T cell transfer.

Evaluation of Ag targeting in vivo

To evaluate mAb targeting to DCs by FACS, animals were injected s.c. with 10 μg of L31-OLLAS-OVA. Spleen and pLN (popliteal, cervical, inguinal, axillary, and brachial) were harvested 24 h later. DCs were enriched by low density on dense BSA (8), and the cells were stained with anti-CD11c-FITC and anti-OLLAS-Alexa Fluor 647. For immunofluorescence, mice were injected s.c. with 25 μg of L31-OLLAS-OVA or control Ig. Twelve hours later, tissue sections were analyzed with a Zeiss LSM 510 system (8).

Statistical analysis

The Student t test was applied to reveal significant differences in fold clonal expansion of transgenic T cells, defined as the ratio of the number of cells with more than one division to the number of transgenic T cells in control mice that had not received Ag. Our results are expressed as the mean ± SE. In the figures, p values of ≤0.05 are labeled with a single asterisk (∗) in contrast to p values of ≤0.01 (∗∗) or ≤0.001 (∗∗∗).

Results and Discussion

Preparation of anti-Langerin-OLLAS-OVA hybrid mAb

To examine the capacity of the Langerin receptor to process and present proteins through the MHC I and II pathways, we genetically engineered mAbs against Langerin (8), DEC (16), DCIR2 (3), and a nonreactive control Ig (ISO) to express OVA. To track Ab expression and binding, we added a linker tag sequence between the C terminus of the Ab H chain and OVA named OLLAS, recognized by an anti-OLLAS mAb (19) (Fig. 1⇓A). In each case, the fusion H chain had a molecular mass of ∼110 kDa (Fig. 1⇓B) and reacted with anti-mouse Ig, anti-OVA, and anti-OLLAS mAbs by Western blotting (Fig. 1⇓C). By FACS, each fusion mAb bound appropriately to CHO transfectants expressing receptor (Fig. 1⇓D, gray histograms), but not to untransfected CHO (CHO/Neo; Fig. 1⇓D, black histograms), while the control Ig-OVA did not bind to Langerin, DEC, or DCIR2 (Fig. 1⇓D). Thus, each mAb to three endocytic receptors can be engineered to express both OVA as an Ag and OLLAS as a sensitive tag.

FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

Genetic engineering of OVA into the anti-Langerin mAb H chain. A, Diagram of engineered mAbs fused to OVA, with OLLAS tag between H chain and OVA. mCκ, mouse Cκ; mIgG1, mouse IgG1. B, Coomassie-stained 10% SDS-PAGE reducing gel comparing chimeric anti-Langerin (Lang), anti-DEC, anti-DCIR2, and control Ig -OLLAS-OVA, with molecular mass in kDa. C, Western blots of hybrid mAbs using HRP-conjugated anti-mouse IgG1 (left), anti-OVA (middle), or anti-OLLAS biotin followed by HRP-conjugated streptavidin (right). D, Binding of mAbs to CHO/mouse Langerin (mLang), CHO/mouse DEC (mDEC), CHO/mouse DCIR2 (mDCIR2) (gray), and control CHO/Neo cells (black), using graded doses (0.02–2 μg) of recombinant mAbs and anti-OLLAS-Alexa Fluor 647 (αOLLAS). ISO stands for isotype control.

Anti-Langerin targets selectively and systemically to DCs in vivo

To assess targeting of anti-Langerin to DCs in vivo, we injected BALB/c mice s.c. in the footpads with anti-Langerin-OLLAS-OVA or control mAb and detected OLLAS by FACS analysis and immunofluorescence. The isotype control mAb (ISO-OLLAS-OVA, where ISO is isotype control) did not bind to DCs (Fig. 2⇓A, top), whereas anti-Langerin-OLLAS-OVA targeted all subsets of Langerin+ cells, e.g., both CD11chigh (∼25 ± 5% of Langerin+ cells) and CD11clow (∼70 ± 5% of Langerin+ cells) DCs in spleen and in pLN (Fig. 2⇓A). By FACS, the DC labeling by anti-OLLAS tag mAb was selective for those CD11c+ cells that also were labeled with anti-Langerin (data not shown). By immunofluorescence of tissue sections, anti-OLLAS also selectively stained cells reactive with anti-Langerin in the T cell area of pLN (Fig. 2⇓B). These results indicate that s.c. injection of hybrid anti-Langerin Abs targets Langerin+ DCs selectively and systemically (spleen) in vivo.

FIGURE 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2.

Detection of recombinant mAbs after targeting Langerin in vivo. A, BALB/c mice were injected s.c. with 10 μg of anti-Langerin or control Ig -OLLAS-OVA. Twenty-four hours later, DCs were enriched and analyzed by FACS using anti-OLLAS-Alexa Fluor 647 mAb. The selectively targeted CD11chigh and CD11clow DCs are indicated by an arrowhead and arrow, respectively, where the latter are the tissue-derived Langerin+ cells. B, Twelve hours postinoculation with 25 μg of anti-Langerin-OLLAS-OVA to BALB/c mice, lymph node sections were stained with anti-OLLAS biotin and streptavidin-Alexa Fluor 555 (red), anti-Langerin Alexa Fluor 647 (green), and anti-B220-FITC followed by anti-FITC Alexa Fluor 488 (blue). ISO stands for isotype control.

Targeting proteins to DCs and LCs in vivo via anti-Langerin

C57BL/6 mice were injected with graded doses of each OLLAS-OVA fusion mAb 1 day after inoculating CFSE-labeled OT-I or OT-II T cells, the latter to monitor Ag presentation in vivo. After 3 days, pLN and spleen were evaluated for T cell proliferation by CFSE dilution and the total number of expanded CFSE-low cells. Anti-DEC, anti-Langerin, and anti-DCIR2- OLLAS-OVA induced responses by MHC I-restricted CD8+ OT-I cells, as well as MHC II-restricted OT-II cells, in both spleen and pLN. Virtually all T cells entered cell cycle and underwent 4–8 cell divisions after a single dose of 3 μg of anti-Langerin-OLLAS-OVA (Fig. 3⇓A). Similar results were obtained when we inoculated fusion mAbs lacking the OLLAS tag, indicating that the OLLAS sequence did not affect Ag presentation in vivo (data not shown). We also reproduced prior work (3) showing that 33D1 anti-DCIR2, which selectively targets CD8− DCs, was more efficient in inducing CD4+ T cell expansion while anti-DEC was more efficient for CD8+ T cells (Fig. 3⇓B). Anti-Langerin-OVA was intermediate in its behavior, slightly less effective than anti-DEC for triggering OT-I expansion but more effective for OT-II T cells (Fig. 3⇓B). The observations with OT-II cells in C57BL/6 mice were confirmed on DO11.10 T cells in BALB/c mice (data not shown). Taken together, Langerin delivers OVA Ag for efficient presentation on MHC I and II products.

FIGURE 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3.

Anti-Langerin-OVA induces strong in vivo proliferation of CD8+ and CD4+ transgenic T cells. A, C57BL/6 mice were injected i.v. with 2–5 × 106 CFSE-labeled OT-I or OT-II T cells and, 24 h later, with graded doses of the indicated OLLAS-OVA fusion mAb s.c. Three days after the hybrid mAb injection, pLN and spleen were harvested and expansion of CD8+ or CD4+Vα2 cells was evaluated by FACS for CFSE dilution. Panels are typical of 2–4 experiments. B, Bar graphs to quantify T cells undergoing more than one division (fold clonal expansion; see Materials and Methods) on day 3 after the inoculation of 3 μg of anti-Langerin, anti-DEC, or anti-DCIR2 -OLLAS-OVA. Means and SE values are shown for six experiments. The Greek letter alpha (α) stands for “anti,” and ISO stands for isotype control.

Long-lasting presentation of OVA delivered via anti-Langerin

To evaluate the persistence of presented Ag, we injected the different OVA fusion mAbs and 1, 3, or 14 days later we injected CFSE-labeled, OVA-specific OT-I or OT-II cells. Presentation to CD8+ T cells after one dose of anti-Langerin-OLLAS-OVA was long-lasting (Fig. 4⇓), being vigorous in pLN and spleen 14 days after mAb injection. In contrast, as shown previously (3) presentation by CD8− DCs via anti-DCIR2 persisted for 3 days (Fig. 4⇓). Functional peptide MHC complexes persisted to a slightly greater extent at day 14 when OVA was targeted via Langerin rather than DEC (Fig. 4⇓).

FIGURE 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 4.

Prolonged MHC I and II presentation after anti-Langerin targeting. C57BL/6 mice were inoculated with the indicated OLLAS-OVA fusion mAb. One, 3, or 14 days later 2–5 × 106 CFSE-labeled OT-I or OT-II cells were transferred. Proliferation in pLN and spleen was followed by FACS 3 days later. One of two similar experiments is shown. The Greek letter alpha (α) stands for “anti,” and ISO stands for isotype control.

Prior observations had shown that Langerin acts as an endocytic receptor in vitro with cultured human LCs. In this study we show that Langerin mediates Ag presentation in vivo by targeting the receptor selectively within the new L31 mAb to the Langerin extracellular domain (8). Langerin delivers Ag to both CD4+ and CD8+ T cells, and the presentation is prolonged for several days. When anti-Langerin-OVA is compared with anti-DEC-OVA where DEC and Langerin expression is to a large extent confined to the same subsets of DCs, e.g., CD8+ DCs and LCs, anti-Langerin is slightly less efficient than anti-DEC for presentation on MHC I and more efficient for MHC II, and Langerin-based presentation also seems to persist slightly longer. In contrast, when anti-Langerin-OVA is compared with anti-DCIR2-OVA, presentation of anti-Langerin is less efficient for MHC II but more efficient to MHC I. Although the OT-I and OT-II transgenic T cells have been valuable in this study for carrying out quantitative comparisons of the efficacy of different endocytic receptors, the functional consequences of Ag presentation are best conducted with other more clinically relevant Ags and the infrequent T cells in the polyclonal repertoire.

Acknowledgments

We thank Judy Adams for preparing figures, Lucio Verani for help with references, Alison North and the Bio-Imaging Resource Center of Rockefeller University (New York, NY) for help with confocal microscopy, and the Northeast Biodefense Center Protein Expression Core (Albany, NY) for expression and purification of mAb L31.

Disclosures

The authors have no financial conflict of interest.

Footnotes

  • The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

  • ↵1 This work was supported by the Center for AIDS Vaccine Discovery, National Institutes of Health Grants AI 13013, AI 40874, and AI 057158 (to R.M.S.) and AI 057158 (to C.G.P.), and the Canadian Histiocytosis Association.

  • ↵2 J.I. and C.C. contributed equally to this work.

  • ↵3 C.G.P. and R.M.S. are equally contributing senior authors.

  • ↵4 Address correspondence and reprint requests to Dr. Chae Gyu Park or Dr. Ralph M. Steinman, Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065. E-mail addresses: steinma{at}mail.rockefeller.edu and parkc{at}mail.rockefeller.edu

  • ↵5 Abbreviations used in this paper: DC, dendritic cell; CHO, Chinese hamster ovary; LC, Langerhans cell; pLN, skin draining lymph node.

  • Received November 29, 2007.
  • Accepted January 25, 2008.
  • Copyright © 2008 by The American Association of Immunologists

References

  1. ↵
    Trombetta, E. S., I. Mellman. 2005. Cell biology of antigen processing in vitro and in vivo. Annu. Rev. Immunol. 23: 975-1028.
    OpenUrlCrossRefPubMed
  2. ↵
    Shortman, K., Y. J. Liu. 2002. Mouse and human dendritic cell subtypes. Nat. Rev. Immunol. 2: 151-161.
    OpenUrlCrossRefPubMed
  3. ↵
    Dudziak, D., A. O. Kamphorst, G. F. Heidkamp, V. Buchholz, C. Trumpfheller, S. Yamazaki, C. Cheong, K. Liu, H. W. Lee, C. G. Park, et al 2007. Differential antigen processing by dendritic cell subsets in vivo. Science 315: 107-111.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    Figdor, C. G., Y. van Kooyk, G. J. Adema. 2002. C-type lectin receptors on dendritic cells and Langerhans cells. Nat. Rev. Immunol. 2: 77-84.
    OpenUrlCrossRefPubMed
  5. ↵
    Valladeau, J., O. Ravel, C. Dezutter-Dambuyant, K. Moore, M. Kleijmeer, Y. Liu, V. Duvert-Frances, C. Vincent, D. Schmitt, J. Davoust, et al 2000. Langerin, a novel C-type lectin specific to Langerhans cells, is an endocytic receptor that induces the formation of Birbeck granules. Immunity 12: 71-81.
    OpenUrlCrossRefPubMed
  6. ↵
    Kissenpfennig, A., S. Henri, B. Dubois, C. Laplace-Builhe, P. Perrin, N. Romani, C. H. Tripp, P. Douillard, L. Leserman, D. Kaiserlian, et al 2005. Dynamics and function of Langerhans cells in vivo: dermal dendritic cells colonize lymph node areas distinct from slower migrating Langerhans cells. Immunity 22: 643-654.
    OpenUrlCrossRefPubMed
  7. ↵
    Takahara, K., Y. Omatsu, Y. Yashima, Y. Maeda, S. Tanaka, T. Iyoda, B. Clausen, K. Matsubara, J. Letterio, R. M. Steinman, et al 2002. Identification and expression of mouse Langerin (CD207) in dendritic cells. Int. Immunol. 14: 433-444.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    Cheong, C., J. Idoyaga, Y. Do, M. Pack, S. H. Park, H. Lee, Y. S. Kang, J. H. Choi, J. Y. Kim, A. Bonito, et al 2007. Production of monoclonal antibodies that recognize the extracellular domain of mouse Langerin/CD207. J. Immunol. Methods 324: 48-62.
    OpenUrlCrossRefPubMed
  9. ↵
    Douillard, P., P. Stoitzner, C. H. Tripp, V. Clair-Moninot, S. Ait-Yahia, A. D. McLellan, A. Eggert, N. Romani, S. Saeland. 2005. Mouse lymphoid tissue contains distinct subsets of langerin/CD207 dendritic cells, only one of which represents epidermal-derived Langerhans cells. J. Invest. Dermatol. 125: 983-994.
    OpenUrlCrossRefPubMed
  10. ↵
    Galustian, C., C. G. Park, W. Chai, M. Kiso, S. A. Bruening, Y. S. Kang, R. M. Steinman, T. Feizi. 2004. High and low affinity carbohydrate ligands revealed for murine SIGN-R1 by carbohydrate array and cell binding approaches, and differing specificities for SIGN-R3 and langerin. Int. Immunol. 16: 853-866.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    Stambach, N. S., M. E. Taylor. 2003. Characterization of carbohydrate recognition by langerin, a C-type lectin of Langerhans cells. Glycobiology 13: 401-410.
    OpenUrlAbstract/FREE Full Text
  12. ↵
    de Witte, L., A. Nabatov, M. Pion, D. Fluitsma, M. A. de Jong, T. de Gruijl, V. Piguet, Y. van Kooyk, T. B. Geijtenbeek. 2007. Langerin is a natural barrier to HIV-1 transmission by Langerhans cells. Nat. Med. 13: 367-371.
    OpenUrlCrossRefPubMed
  13. ↵
    Takahara, K., Y. Yashima, Y. Omatsu, H. Yoshida, Y. Kimura, Y. S. Kang, R. M. Steinman, C. G. Park, K. Inaba. 2004. Functional comparison of the mouse DC-SIGN, SIGNR1, SIGNR3 and Langerin, C-type lectins. Int. Immunol. 16: 819-829.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Mc Dermott, R., U. Ziylan, D. Spehner, H. Bausinger, D. Lipsker, M. Mommaas, J. P. Cazenave, G. Raposo, B. Goud, H. de la Salle, et al 2002. Birbeck granules are subdomains of endosomal recycling compartment in human epidermal Langerhans cells, which form where Langerin accumulates. Mol. Biol. Cell 13: 317-335.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Tirlapur, U. K., W. J. Mulholland, B. J. Bellhouse, M. Kendall, J. F. Cornhill, Z. Cui. 2006. Femtosecond two-photon high-resolution 3D imaging, spatial-volume rendering and microspectral characterization of immunolocalized MHC-II and mLangerin/CD207 antigens in the mouse epidermis. Microsc. Res. Tech. 69: 767-775.
    OpenUrlCrossRefPubMed
  16. ↵
    Hawiger, D., K. Inaba, Y. Dorsett, M. Guo, K. Mahnke, M. Rivera, J. V. Ravetch, R. M. Steinman, M. C. Nussenzweig. 2001. Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivo. J. Exp. Med. 194: 769-780.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    Bonifaz, L. C., D. P. Bonnyay, A. Charalambous, D. I. Darguste, S. Fujii, H. Soares, M. K. Brimnes, B. Moltedo, T. M. Moran, R. M. Steinman. 2004. In vivo targeting of antigens to maturing dendritic cells via the DEC-205 receptor improves T cell vaccination. J. Exp. Med. 199: 815-824.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Ramakrishna, V., J. F. Treml, L. Vitale, J. E. Connolly, T. O’Neill, P. A. Smith, C. L. Jones, L. Z. He, J. Goldstein, P. K. Wallace, et al 2004. Mannose receptor targeting of tumor antigen pmel17 to human dendritic cells directs anti-melanoma T cell responses via multiple HLA molecules. J. Immunol. 172: 2845-2852.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Park, S. H., C. Cheong, J. Idoyaga, J. Y. Kim, J. H. Choi, Y. Do, H. Lee, J. H. Jo, Y. S. Oh, W. Im, et al. Generation and application of new rat monoclonal antibodies against synthetic FLAG and OLLAS tags for improved immunodetection. J. Immunol. Methods. In Press.
PreviousNext
Back to top

In this issue

The Journal of Immunology: 180 (6)
The Journal of Immunology
Vol. 180, Issue 6
15 Mar 2008
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Advertising (PDF)
  • Back Matter (PDF)
  • Editorial Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word about The Journal of Immunology.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Cutting Edge: Langerin/CD207 Receptor on Dendritic Cells Mediates Efficient Antigen Presentation on MHC I and II Products In Vivo
(Your Name) has forwarded a page to you from The Journal of Immunology
(Your Name) thought you would like to see this page from the The Journal of Immunology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Cutting Edge: Langerin/CD207 Receptor on Dendritic Cells Mediates Efficient Antigen Presentation on MHC I and II Products In Vivo
Juliana Idoyaga, Cheolho Cheong, Koji Suda, Nao Suda, Jae Y. Kim, Haekyung Lee, Chae Gyu Park, Ralph M. Steinman
The Journal of Immunology March 15, 2008, 180 (6) 3647-3650; DOI: 10.4049/jimmunol.180.6.3647

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Cutting Edge: Langerin/CD207 Receptor on Dendritic Cells Mediates Efficient Antigen Presentation on MHC I and II Products In Vivo
Juliana Idoyaga, Cheolho Cheong, Koji Suda, Nao Suda, Jae Y. Kim, Haekyung Lee, Chae Gyu Park, Ralph M. Steinman
The Journal of Immunology March 15, 2008, 180 (6) 3647-3650; DOI: 10.4049/jimmunol.180.6.3647
del.icio.us logo Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results and Discussion
    • Acknowledgments
    • Disclosures
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Cutting Edge: T Cell Responses to B.1.1.529 (Omicron) SARS-CoV-2 Variant Induced by COVID-19 Infection and/or mRNA Vaccination Are Largely Preserved
  • Cutting Edge: SARS-CoV-2 Infection Induces Robust Germinal Center Activity in the Human Tonsil
  • Cutting Edge: Enhanced Antitumor Immunity in ST8Sia6 Knockout Mice
Show more CUTTING EDGE

Similar Articles

Navigate

  • Home
  • Current Issue
  • Next in The JI
  • Archive
  • Brief Reviews
  • Pillars of Immunology
  • Translating Immunology

For Authors

  • Submit a Manuscript
  • Instructions for Authors
  • About the Journal
  • Journal Policies
  • Editors

General Information

  • Advertisers
  • Subscribers
  • Rights and Permissions
  • Accessibility Statement
  • FAR 889
  • Privacy Policy
  • Disclaimer

Journal Services

  • Email Alerts
  • RSS Feeds
  • ImmunoCasts
  • Twitter

Copyright © 2022 by The American Association of Immunologists, Inc.

Print ISSN 0022-1767        Online ISSN 1550-6606