visualization and characterization of migratory langerhans cells in murine skin and lymph nodes by...

9
ORIGINAL ARTICLE Visualization and Characterization of Migratory Langerhans Cells in Murine Skin and Lymph Nodes by Antibodies Against Langerin/CD207 Patrizia Stoitzner, Sandra Holzmann, Alexander D. McLellan, n Lennart Ivarsson, Hella St˛ssel, Michaela Kapp, n Ulrike KȄmmerer, n Patrice Douillard,w Eckhart KȄmpgen, n Franz Koch, Sem Saeland,w and Nikolaus Romani Department of Dermatology, University of Innsbruck, Innsbruck, Austria; n Department of Dermatology, University of Wˇrzburg,Wˇrzburg, Germany; wLaboratory for Immunological Research, Schering-Plough, Dardilly, France Dendritic cells are professional antigen-presenting cells that initiate primary immunity. Migration from sites of antigen uptake to lymphoid organs is crucial for the generation of immune responses. We investigated the migratory pathways speci¢cally of epidermal Langer- hans cells by tracing them from the epidermis to the draining lymph nodes. This was possible with a new monoclonal antibody, directed against murine Langer- in/CD207, a type II lectin speci¢c for Langerhans cells. In situ, resident, and activated Langerhans cells express Langerin in the epidermis and on their way through dermal lymphatic vessels. Both emigrated and trypsini- zation-derived Langerhans cells expressed high levels of Langerin intracellularly but reduced it upon prolonged culture periods. Sizeable numbers of Langerin þ cells were found in skin draining lymph nodes but not in mesenteric nodes. Langerin þ cells localized to the T cells areas and rarely to B cell zones. Numbers of Langerin-expressing cells increased after application of a contact sensitizer. In the steady state, Langerhans cells in the skin-draining nodes expressed maturation mar- kers, such as 2A1 and costimulatory molecules CD86 and CD40. These molecules, CD86 and CD40, were further upregulated upon in£ammatory stimuli such as contact sensitization. Thus, the novel anti-Langerin monoclonal antibody permits the unequivocal visuali- zation of migratory Langerhans cells in the lymph nodes for the ¢rst time and thereby allows to dissect the relative immunogenic or tolerogenic contributions of Langerhans cells and other types of dendritic cells. Key words: cell tra⁄cking/dendritic cells/in£ammation/skin/ spleen and lymph nodes. J Invest Dermatol 120:266 ^274, 2003 D endritic cells are the only antigen-presenting cells that are able to initiate primary immune responses (Banchereau and Steinman, 1998). Their functional spectrum comprises antigen uptake and processing capacities (Mellman and Steinman, 2001), which are exerted mostly in peripheral organs and tissues, as well as na- ive and memory T cell sensitizing skills, which are performed in the lymphoid organs.To ful¢ll these tasks they need to be highly motile to bridge the sites of antigen uptake and the sites of T cell stimulation. Emigrating cutaneous dendritic cells carry major histocompatibility complex (MHC)^antigenic peptide complexes on their cell surfaces and present them to T cells in the lymphoid organs (Austyn, 1996; Steinman et al, 1997; Flores-Romo, 2001). Emigration is induced by in£ammatory cytokines, such as tu- mor necrosis factor-a and interleukin-1b (Cumberbatch et al , 1997; Stoitzner et al , 1999) and further modi¢ed by chemotactic agents, such as MIP-3b /CCL19 (Kellermann et al , 1999), SLC/ CCL21 (Saeki et al , 1999), and interleukin-16 (Stoitzner et al , 2001).When infectious agents invade the peripheral tissue in£am- matory mediators are produced by diverse skin cells.They induce the detachment of Langerhans cells from neighboring keratino- cytes, most probably by downregulation of E-cadherin (Schwar- zenberger and Udey, 1996). The released Langerhans cells are able to leave the epidermal compartment and passage the basement membrane possibly by secreting matrix metalloproteinases and digesting the dense collagen meshwork of the epidermal^dermal junction (Kobayashi et al , 1999; Ratzinger et al , 2002). With the help of di¡erent adhesion molecules, e.g., a 6 integrins (Price et al , 1997) and CD44 (Weiss et al , 1997), and enzymes for diges- tion of the extracellular matrix (Ratzinger et al, 2002), dendritic cells migrate further through the dermis until they encounter lymphatic vessels (Larsen et al , 1990; Lukas et al , 1996; Weinlich et al, 1998; Stoitzner et al , 2002). Skin explant culture (Larsen et al , 1990) and contact hypersensitivity models (Enk et al, 1993) have been widely used to investigate morphologic and regulatory aspects of dendritic cell migration.Therefore, we used both mod- els to trace Langerhans cells through the skin into the draining lymph node by means of new monoclonal antibodies (MoAb) that recognize the murine homolog of the human type II lectin Langerin (CD207) (Valladeau et al , 2002). This molecule is ex- pressed on the cell surface and in Birbeck granules, which are the hallmark of Langerhans cells. Langerin appears to be a key structural element in Birbeck granule formation and presumably Reprint requests to: Patrizia Stoitzner, PhD, Department of Dermatol- ogy, University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria. Email: [email protected] Abbreviations: MIP-3b , macrophage in£ammatory protein-3 -beta/ CCL19; SLC, secondary lymphoid tissue chemokine/CCL21; TNCB, 2,4,6-trinitro-1-chlorobenzene. Manuscript received July 26, 2002; revised September 16, 2002; accepted for publication September 28, 2002 0022-202X/03/$15.00 Copyright r 2003 by The Society for Investigative Dermatology, Inc. 266

Upload: nikolaus

Post on 06-Jul-2016

217 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Visualization and Characterization of Migratory Langerhans Cells in Murine Skin and Lymph Nodes by Antibodies Against Langerin/CD207

ORIGINAL ARTICLE

Visualization and Characterization of Migratory LangerhansCells in Murine Skin and Lymph Nodes byAntibodies AgainstLangerin/CD207

Patrizia Stoitzner, Sandra Holzmann, Alexander D. McLellan,n Lennart Ivarsson, Hella St˛ssel, Michaela Kapp,n

Ulrike K'mmerer,n Patrice Douillard,w Eckhart K'mpgen,n Franz Koch, Sem Saeland,w and Nikolaus RomaniDepartment of Dermatology, University of Innsbruck, Innsbruck, Austria; nDepartment of Dermatology, University of Wˇrzburg,Wˇrzburg, Germany;wLaboratory for Immunological Research, Schering-Plough, Dardilly, France

Dendritic cells are professional antigen-presenting cellsthat initiate primary immunity. Migration from sites ofantigen uptake to lymphoid organs is crucial for thegeneration of immune responses. We investigated themigratory pathways speci¢cally of epidermal Langer-hans cells by tracing them from the epidermis to thedraining lymph nodes. This was possible with a newmonoclonal antibody, directed against murine Langer-in/CD207, a type II lectin speci¢c for Langerhans cells.In situ, resident, and activated Langerhans cells expressLangerin in the epidermis and on their way throughdermal lymphatic vessels. Both emigrated and trypsini-zation-derived Langerhans cells expressed high levels ofLangerin intracellularly but reduced it upon prolongedculture periods. Sizeable numbers of Langerinþ cellswere found in skin draining lymph nodes but not inmesenteric nodes. Langerinþ cells localized to the

T cells areas and rarely to B cell zones. Numbers ofLangerin-expressing cells increased after application ofa contact sensitizer. In the steady state, Langerhans cellsin the skin-draining nodes expressed maturation mar-kers, such as 2A1 and costimulatory molecules CD86and CD40. These molecules, CD86 and CD40, werefurther upregulated upon in£ammatory stimuli such ascontact sensitization. Thus, the novel anti-Langerinmonoclonal antibody permits the unequivocal visuali-zation of migratory Langerhans cells in the lymphnodes for the ¢rst time and thereby allows to dissectthe relative immunogenic or tolerogenic contributionsof Langerhans cells and other types of dendritic cells.Key words: cell tra⁄cking/dendritic cells/in£ammation/skin/spleen and lymph nodes. J Invest Dermatol 120:266 ^274,2003

Dendritic cells are the only antigen-presenting cellsthat are able to initiate primary immune responses(Banchereau and Steinman, 1998). Their functionalspectrum comprises antigen uptake and processingcapacities (Mellman and Steinman, 2001), which

are exerted mostly in peripheral organs and tissues, as well as na-ive and memoryT cell sensitizing skills, which are performed inthe lymphoid organs. To ful¢ll these tasks they need to be highlymotile to bridge the sites of antigen uptake and the sites of T cellstimulation. Emigrating cutaneous dendritic cells carry majorhistocompatibility complex (MHC)^antigenic peptide complexeson their cell surfaces and present them toT cells in the lymphoidorgans (Austyn, 1996; Steinman et al, 1997; Flores-Romo, 2001).

Emigration is induced by in£ammatory cytokines, such as tu-mor necrosis factor-a and interleukin-1b (Cumberbatch et al,1997; Stoitzner et al, 1999) and further modi¢ed by chemotacticagents, such as MIP-3b/CCL19 (Kellermann et al, 1999), SLC/

CCL21 (Saeki et al, 1999), and interleukin-16 (Stoitzner et al,2001).When infectious agents invade the peripheral tissue in£am-matory mediators are produced by diverse skin cells. They inducethe detachment of Langerhans cells from neighboring keratino-cytes, most probably by downregulation of E-cadherin (Schwar-zenberger and Udey, 1996). The released Langerhans cells are ableto leave the epidermal compartment and passage the basementmembrane possibly by secreting matrix metalloproteinases anddigesting the dense collagen meshwork of the epidermal^dermaljunction (Kobayashi et al, 1999; Ratzinger et al, 2002). With thehelp of di¡erent adhesion molecules, e.g., a6 integrins (Priceet al, 1997) and CD44 (Weiss et al, 1997), and enzymes for diges-tion of the extracellular matrix (Ratzinger et al, 2002), dendriticcells migrate further through the dermis until they encounterlymphatic vessels (Larsen et al, 1990; Lukas et al, 1996; Weinlichet al, 1998; Stoitzner et al, 2002). Skin explant culture (Larsenet al, 1990) and contact hypersensitivity models (Enk et al, 1993)have been widely used to investigate morphologic and regulatoryaspects of dendritic cell migration.Therefore, we used both mod-els to trace Langerhans cells through the skin into the draininglymph node by means of new monoclonal antibodies (MoAb)that recognize the murine homolog of the human type II lectinLangerin (CD207) (Valladeau et al, 2002). This molecule is ex-pressed on the cell surface and in Birbeck granules, which arethe hallmark of Langerhans cells. Langerin appears to be a keystructural element in Birbeck granule formation and presumably

Reprint requests to: Patrizia Stoitzner, PhD, Department of Dermatol-ogy, University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria.Email: [email protected]

Abbreviations: MIP-3b, macrophage in£ammatory protein-3-beta/CCL19; SLC, secondary lymphoid tissue chemokine/CCL21; TNCB,2,4,6-trinitro-1-chlorobenzene.

Manuscript received July 26, 2002; revised September 16, 2002; acceptedfor publication September 28, 2002

0022-202X/03/$15.00 � Copyright r 2003 by The Society for Investigative Dermatology, Inc.

266

Page 2: Visualization and Characterization of Migratory Langerhans Cells in Murine Skin and Lymph Nodes by Antibodies Against Langerin/CD207

facilitates the uptake of mannans on the cell surface of bacteria(Valladeau et al, 1999, 2000).

Several recent reports attempted to identify migratory Langer-hans cells in the skin draining lymph nodes. Salomon et al (1998)described two CD11cþ MHC class IIþ cell populations in skindraining lymph nodes. One population was comprised of largecells that expressed a mature phenotype and most of the cellswere positive for £uorescein isothiocyanate (FITC) after skinpainting, thus mostly likely representing immigrated Langerhanscells from the periphery. The other dendritic cell population wassmaller in size and segregated into two groups, a CD4þ -mye-loid-derived and a CD8aþ -lymphoid-related one, which tookup intravenously injected FITC and had a more rapid turnover.Ruedl et al (2000) reported a CD11chighCD40high population inthe lymph node that showed some characteristics of Langerhanscells. They were strongly MHC class IIþ , expressed E-cadherin,contained a few Birbeck granules and had a long turnover rate incontrast to the other lymph node populations. Additionally, theytransported £uorescent tracer from the epidermis to the lym-phoid tissue. Henri et al (2001) described a DEC-205/CD205high,CD8a^, MHC class IIhigh population in skin draining lymphnodes, which also expressed high levels of CD40, CD11c, andLangerin, and transported a £uorescent tracer to the lymphoidtissue. We attempted to de¢ne further and characterize theselymph node populations as well as the ¢rst part of the migratorypathway within the skin by means of novel tools, i.e., Langerhanscell-speci¢c anti-mouse Langerin/CD207 antibodies (Valladeauet al, 2002).

MATERIALS AND METHODS

Mice Mice of inbred strains C57BL/6 and BALB/C were purchased fromCharles River Germany (Sulzfeld, Germany) and used at 3^12 mo of age.

Langerin reagents Several di¡erent antibodies against mouse Langerinwere used [HD26, mouse IgG1, and a rabbit immune serum forpreliminary experiments only; HD24, mouse IgG1 (Valladeau et al, 2002)and 929F3, rat IgG1]. The di¡erent anti-Langerin MoAb gave identicalstaining results, both in terms of staining intensity and staining patterns.The anti-Langerin MoAb 929F3 was also available in FITC- and biotin-conjugated forms.

Transfection of COP5 ¢broblasts for control of antibodyspeci¢city Transfections were performed by electroporation of themouse Langerin cDNA (Valladeau et al, 2002) into the murine ¢broblasticCOP5 cell line (Tyndall et al, 1981). Cells transfected with an emptyplasmid served as mock controls.

Skin explant culture Culture medium was RPMI-1640 supplementedwith 10% fetal bovine serum, gentamycin (all from PAA, Linz, Austria),and 2-mercaptoethanol (Sigma, St Louis, MO). Mice were killed and earswere cut o¡ at the base. Ear skin was split in dorsal and ventral halves bymeans of strong forceps and the dorsal halves (i.e., cartilage-free halves)were cultured in 24-well tissue culture plates (one ear per well) asdescribed (Ortner et al, 1996; Romani et al, 1997; Weinlich et al, 1998).Alternatively, epidermis and dermis were separated from each other bymeans of the bacterial enzyme dispase (Boehringer Mannheim,Mannheim, Germany) (Kitano and Okado, 1983), and the epidermalsheets were placed in culture. In most experiments whole skin, epidermis,and dermis were cultured continuously for 48 h. Cells that had emigratedfrom the explants were collected, counted, and further evaluatedphenotypically.

Induction of contact sensitization Murine ear skin was painted with15 ml 1% 2,4,6-trinitro-1-chlorobenzene (TNCB), picryl chloride (KodakEastman, Rochester, NY) on the dorsal and ventral sides. After 24 and 48h epidermal and dermal sheets were prepared. For this purpose, the skinwas £oated dermal side down on 0.5 M ammonium thiocyanate for 20min at 371C. The epidermis was peeled o¡ the dermis and both partswere cut into 5�5 mm pieces and ¢xed in acetone for 20 min.

Immunohistochemistry of epidermal and dermal sheets Epidermaland dermal sheets were incubated overnight at 41C with anti-LangerinMoAb (HD24 and 929F3). Antibodies were visualized by usingbiotinylated anti-mouse and anti-rat immunoglobulin, respectively, and

streptavidin-FITC or streptavidin-Texas Red (all from Amersham LifeScience, Amersham, U.K.; 90 min at 371C). Dendritic cells werecounterstained with anti-I-A/Ediverse (clone 2G9, £uoresceinated, ratIgG2a; BD-Pharmingen, San Diego, CA). Specimens were viewed on aconventional Olympus epi£uorescence microscope.

Isolation of Langerhans cells and dermal dendritic cells frommurine skin Epidermal cells were isolated by standard trypsinization(JHR-trypsin from Sera-lab, Crawley Down, West Sussex, U.K.) (Kochet al, 2001) and dermal cells after removal of the epidermis and thoroughrinsing by digestion in 0.5 mg per ml collagenase P (Roche AppliedScience, Hamburg, Germany) for 30^60 min at 371C. Both cellpopulations were either analyzed by £uorescence-activated cell sorter(FACS) immediately or cultured for 3^4 d in Iscove’s medium (Pan,Hamburg, Germany) supplemented with 10% fetal bovine serum and 200U per ml murine granulocyte-macrophage colony-stimulating factor usingsupernatants of a transfected cell line as the source of cytokine (kindlyprovided by A. Lanzavecchia, Bellinzona, Switzerland).

Preparation of lymph node cell suspensions Cell suspensions fromlymph nodes were obtained by digestion with 0.5 mg per ml collagenaseP (Roche) for 30 min at 371C and subsequent pressing the tissue throughsteel sieves, essentially as described recently for spleen cell suspensions(McLellan et al, 2002). For some experiments, dendritic cells wereenriched by Nycodenz gradient centrifugation (Sigma-Aldrich, Vienna,Austria) as described (McLellan et al, 1995).

Phenotype of Langerhans cells and dermal dendritic cells isolatedfrom skin or lymph nodes Cells were ¢xed and permeabilized with akit (Fix & PermTM) from BD-Pharmingen and stained in sequence withthe rat IgG anti-Langerin 929F3 MoAb or an irrelevant rat IgG as control(Jackson Immuno Research Laboratory, Avondale, PA), anti-ratimmunoglobulin-phycoerythrin (BD-Pharmingen), an excess of rat IgG(100 mg per ml) for blocking residual free antibody binding sites, andcounterstained either with anti-MHC class II (clone 2G9), anti-CD86(clone GL1), or anti-CD40 (clone 3/23) (all FITC conjugated and fromBD-Pharmingen). Nycodenz-enriched cell populations were used foranalyses of lymph node cells.

Detection of emigrated Langerhans cells in skin draining lymphnodes after contact allergen sensitization Mice were sensitized with200 ml (abdomen) or 15 ml (ears) 1% TNCB on both sides. After 24 h,single cell suspensions were prepared from skin draining lymph nodes(auricular and inguinal). Cytocentrifuge smears were prepared from thesecell suspensions, as mentioned above and counterstained with 406-diamidino-2-phenyindole (DAPI) for visualization of nuclei. Langerinþ

cells were counted under the microscope using � 40 objective lenses anda calibrated grid (at least 20 ¢elds). Alternatively, cells from skin draininglymph nodes (auricular and inguinal) were analyzed by FACS.

Immunohistochemistry of lymph node sections

Single enzyme labeling Skin draining (inguinal or auricular) lymph nodesof untreated mice were frozen and cut into 10 mm sections on a cryostat(Frigocut, Leitz,Vienna, Austria).The sections were brie£y ¢xed in acetoneand stained with anti-Langerin (929F3), anti-CD8 (clone TIB105, ratIgG2b), anti-CD45R (B220, TIB146, rat IgM), and irrelevant isotype-matched control rat immunoglobulin. Antibody binding was detected byalkaline phosphatase-conjugated anti-rat immunoglobulin F(ab)2 frag-ments (Jackson). CD3 (clone 2C11, hamster immunoglobulin) was detectedby a biotinylated anti-Syrian and Armenian hamster immunoglobulin(BD-Pharmingen) followed by alkaline phosphatase-conjugated streptavi-din (Jackson).The enzyme was visualized withVector-Blue and nuclei werecounterstained with Nuclear-Fast-Red (both from Vector Laboratories,Burlingame, CA).

Double enzyme labeling Serial frozen 5 mm sections were placed on to3-amino-propyltriethoxy-silane (Sigma) coated slides, air-dried overnight,¢xed in acetone for 10 min, rehydrated in Tris-bu¡ered saline and blockedin 5% goat and 5% rabbit serum inTris-bu¡ered saline for 5 min. Sectionswere incubated in sequence either with rat anti-CD19 and horseradish per-oxidase-rabbit anti-rat immunoglobulin (DAKOCytometrics, Glostrup,Denmark) or with hamster anti-CD3, biotinylated-goat anti-hamster im-munoglobulin (Jackson), and horseradish peroxidase-labeled streptavidin(Sigma). A brown color was revealed by application of the substrate diami-nobenzidine.The second MoAb, rat anti-Langerin (929F3) was followed byhorseradish peroxidase-rabbit anti-rat immunoglobulin (DAKO). The sec-ond enzyme reaction was developed using Histogreen, giving a blue^greencolor. Sections were counterstained with hematoxylin and embedded inorganic mounting media (Vitro-Clud, Langenbrink, Germany).

TRACING MIGRATING LANGERHANS CELLS 267VOL. 120, NO. 2 FEBRUARY 2003

Page 3: Visualization and Characterization of Migratory Langerhans Cells in Murine Skin and Lymph Nodes by Antibodies Against Langerin/CD207

Double immuno£uorescence labeling In some experiments sections werestained with the maturation marker 2A1 [clone 2A1, rat IgG2a, R.M.Steinman, NY (Inaba et al, 1992; Steinman et al, 1997)] followed by FITC-conjugated anti-rat immunoglobulin (BD-Pharmingen). After blockingresidual free antibody binding sites with an excess of rat immunoglobulin(100 mg per ml) counterstaining was done with a biotinylated rat anti-mouse Langerin MoAb (929F3) and streptavidin-Texas Red.

Statistics Appropriate statistic tests (t-test, one-way ANOVA) were chosenand used to analyze data and to determine whether the di¡erences betweenthe results were signi¢cant.

RESULTS

Speci¢city of novel anti-Langerin/CD207 antibodiesSpeci¢city of MoAb HD24 against murine Langerin was shownon mouse Langerin-transfected COP ¢broblasts (Valladeau et al,2002). Other anti-Langerin antibodies were generated byimmunization of rats and the speci¢city of the particular ratMoAb used in this study (929F3) is shown in Fig 1. Langerin-transfected ¢broblasts stained clearly with the 929F3 MoAb incontrast to mock-transfected cells as analyzed by FACS (Fig 1).

Langerhans cells express Langerin in the steady state in theskin MoAb against mouse Langerin speci¢cally stainLangerhans cells. This is best visualized in epidermal sheets as¢rst shown by Valladeau et al (2002). Here, we con¢rm thatvirtually all Langerhans cells, as identi¢ed by their MHC class IIexpression, display strong Langerin reactivity in intracellulargranular structures. Inversely, all Langerinþ cells expressedMHC II (Fig 2A). In native dermis we observed only some rareLangerinþ cells, possibly Langerhans cells in transit (data notshown).

Migratory Langerhans cells express Langerin in situ Murineskin explants were cultured for 48 h and epidermal and dermalsheets were prepared for immunohistochemistry at the end ofthe culture period. Inspection of epidermal sheets con¢rmed the

previously described phenomenon (Larsen et al, 1990; Lukas et al,1996; Weinlich et al, 1998): the density of Langerhans cellsremaining in the epidermis was greatly reduced. The fewremaining Langerhans cells appeared activated by their stronglyincreased MHC class II expression. They uniformly expressedLangerin at a staining intensity indistinguishable to that ofLangerhans cells in untreated epidermis (Fig 2B). Thecorresponding dermal sheets showed migratory dendritic cells in‘‘cords’’ (Larsen et al, 1990), i.e., string-like accumulations ofstrongly MHC class IIþ cells. Approximately 60% of the MHCclass IIþ cells in the ‘‘cords’’ expressed Langerin (Fig 2C),indicating that they were Langerhans cells from the epidermis.Langerin staining was predominantly intracellular. Thispercentage varied to a large extent from ‘‘cord’’ to ‘‘cord’’. The vastmajority of migrating dendritic cells were mature as determinedby two criteria. First, even conventional £uorescence microscopyrevealed the prominent cell surface localization and the absenceof intracellular MHC II molecules, which may be regarded as asign of maturation (Pierre et al, 1997). Secondly, and morespeci¢cally, most emigrating Langerinþ Langerhans cells in the‘‘cords’’coexpressed the maturation marker 2A1 (Fig 2E,F).

Migratory Langerhans cells express Langerin in vivo Anin£ammatory situation in the skin in vivo was induced by theepicutaneous application of the contact allergen TNCB. After 24h only few lymph vessels ¢lled with emigrating cutaneousdendritic cells were visible. ‘‘Cords’’ were more pronounced after48 h, albeit to a lesser degree as compared with the skin explantculture model. Again, a large part of the migratory cells wereLangerhans cells as de¢ned by their marked expression ofLangerin (Fig 2D).

Origin and phenotype of dendritic cells that emigrate out ofskin explants As opposed to the human system (Lenz et al,1993; Larregina et al, 2001) it has not been possible so far to drawan unequivocal distinction between Langerhans cells and dermaldendritic cells that migrate from murine whole skin explants.Wetherefore used Langerin antibodies to analyze dendritic cells thathad ‘‘crawled out’’ from murine whole skin explants over a periodof 48 h. Double-labeling of cytospins revealed that virtually allLangerin-expressing cells were also strongly MHC class IIþ andexpressed the maturation marker 2A1 (Inaba et al, 1992) (data notshown). Conversely, however, not all MHC class IIþ cells wereanti-Langerin reactive. Control specimens of emigrant dendriticcells from epidermal sheets that had been obtained by dispasetreatment before the onset of culture, were virtually allLangerinþ (Fig 3A). Dendritic cells from the correspondingdermal sheets were largely Langerin^ (Fig 3A), yet, a substantialsubpopulation of Langerinþ dendritic cells was consistentlyfound. As expected, dendritic cells from whole skin explantsrepresented a mixture of Langerinþ (i.e., Langerhans cells) andLangerin^ (i.e., dermal dendritic cells) MHC class IIþ cells(Fig 3A). Both populations occurred in approximately equalproportions, Langerhans cells being slightly more frequent. Amean of 62% of all MHC class IIþ cells coexpressed Langerinon cytospins. By FACS, similar percentages of intracellularlyLangerinþ cells were determined for populations of dendriticcells (i.e., MHC class IIþ cells) emigrated from dispase-procuredepidermal sheets, corresponding dermal sheets and from wholeskin explants (Fig 3B). In preparations of dermal cells isolatedby collagenase digestion of native dermis we found that anaverage of 10% of the MHC class II-expressing cells werepositive for Langerin (Fig 3C). Interestingly, after a cultureperiod of 2 d the proportion of Langerin-expressing cells in theMHC class IIþ fraction rose signi¢cantly over 50% anddecreased afterwards.

Retention of Langerin in Langerhans cells duringmaturation Based on its speci¢city it is tempting to useLangerin as a tracer for Langerhans cells from the epidermis. Yet,Langerin is endocytosed quickly after antigen uptake (Valladeau

Figure1. The new rat MoAb (929F3) is speci¢c for Langerin. Fibro-blasts were transfected either with a mock gene or the mouse Langeringene. In FACS analyses the new MoAb 929F3 stained speci¢cally the Lan-gerin-transfected cells (right panel) and not the control cells (left panel).Thinline, isotype control; bold line, Langerin staining.

268 STOITZNER ETAL THE JOURNAL OF INVESTIGATIVE DERMATOLOGY

Page 4: Visualization and Characterization of Migratory Langerhans Cells in Murine Skin and Lymph Nodes by Antibodies Against Langerin/CD207

et al, 1999) and is downregulated upon Langerhans cell culture(Valladeau et al, 2002). Birbeck granules disappear at theultrastructural level (Schuler and Steinman, 1985). We thereforewondered for how long Langerhans cells, that were generatedby di¡erent isolation procedures, would express Langerin.Emigrated Langerhans cells obtained by epidermal explantculture and Langerhans cells isolated by trypsinization of nativeepidermis and cultured together with keratinocytes were stainedside by side for Langerin and MHC II and analyzed on cytospinsor by FACS. On cytospins, migratory Langerhans cells identi¢edby their MHC II expression, retained Langerin expression during4 d of skin explant culture and only few Langerhans cellscompletely lost their Langerin expression (data not shown).Cytospins of day 4 Langerhans cells obtained by trypsinization

of native epidermis showed a more pronounced decrease inLangerin expression with more cells actually losing anti-Langerin reactivity, as described by Valladeau et al (2002). Time-course experiments were evaluated by more sensitive FACSanalyses. We observed a decrease in intracellular Langerinstaining intensity in the Langerhans cells generated both ways(Fig 4) with a more pronounced reduction in the Langerhanscells obtained by trypsinization.

Langerhans cells can be detected in the T cell area of skindraining lymph nodes in the steady state As intracellularLangerin expression is retained in migratory Langerhans forsome time, we used it as a Langerhans cell tracer molecule andsearched for Langerinþ cells in the lymph nodes. Langerin-

Figure 2. Langerin is expressed in resident and migrating Langerhans cells. In epidermal (rows A, B) and dermal sheets (rows C, D, E + F), Langer-hans cells were stained with anti-Langerin HD24 MoAb (middle column) and with anti-MHC class II (left column) or the maturation marker 2A1 as indicated.Double exposures of both colors are depicted in the right column. Langerin is expressed in all MHC class II+ cells, i.e., Langerhans cells, in both native (A) andcultured (B) epidermis. About 60% of migrating MHC class II+ cells in the lymphatic vessels in the dermis express Langerin (C). In an in vivo in£ammatorysituation after application of contact allergen lymphatic vessels also contained Langerin (929F3 MoAb)/MHC class II-double positive cells (D). Almost allLangerin+ cells (green £uorescence in E, F; using a FITC-conjugated 929F3 anti-Langerin) in the lymphatic vessels expressed the maturation marker 2A1(red £uorescence in E, F). Mature, Langerin^ cells, i.e., migrating dermal dendritic cells are marked with arrowheads. Isotype controls were always clearlynegative. Scale bar ¼ 20 mm for all pictures.

TRACING MIGRATING LANGERHANS CELLS 269VOL. 120, NO. 2 FEBRUARY 2003

Page 5: Visualization and Characterization of Migratory Langerhans Cells in Murine Skin and Lymph Nodes by Antibodies Against Langerin/CD207

expressing cells can be found in skin draining lymph nodes fromuntreated mice as recently shown by Henri et al (2001). Therefore,we ¢rst compared Langerin expression in skin-draining vsmesenteric lymph nodes of untreated mice. Lymph node cellswere prepared, enriched for dendritic cells by a Nycodenzgradient, stained for Langerin and analyzed by FACS. We founda signi¢cant population of Langerinþ cells in skin draininglymph nodes (Fig 5A). By contrast, in mesenteric lymph nodesonly very few Langerinþ cells occurred (Fig 5A). Byimmunohistochemistry of lymph node sections this di¡erencewas con¢rmed in that much higher numbers of Langerin-expressing cells were visible in the skin-draining nodes than inthe mesenteric lymph nodes (Fig 5B).

To locate more precisely these migratory skin dendritic cells insitu, we stained sections of skin draining lymph nodes ofuntreated mice with anti-Langerin and markers that de¢ne the Tand B cell areas (CD3 for T cells and CD45R/B220 for B cells).Langerinþ cells were found in the Tcell areas. By contrast, B cellfollicles did not harbor Langerin-reactive cells in general butrarely a few cells could be observed (Fig 6a^c). On consecutivesections CD11c and CD205/DEC-205 localized to the same areas(data not shown). To underscore these ¢ndings we conducteddouble-labeling of lymph node sections. Langerin-expressing

cells colocalized with CD3þ T cells (Fig 6d) but were excludedfrom B cell areas (de¢ned by anti-CD19 labeling) (Fig 6e).

Increased numbers of Langerhans cells can be detectedin the skin draining lymph nodes after contactsensitization Next, we investigated Langerin expression inskin draining lymph nodes 24 h after application of a contactsensitizer (1% TNCB) to abdominal and ear skin. Cytospins ofunenriched suspensions of lymph node cells were prepared andstained for Langerin. In lymph nodes from control mice thathad been treated with the vehicle only (acetone/olive oil) fewLangerinþ cells were detected by immuno£uorescencemicroscopy. The numbers of Langerinþ cells per inguinallymph node in vehicle-treated mice was on average 486072700.In inguinal lymph nodes from sensitized animals the number

Figure 3. Origin of migratory skin dendritic cells. From skin explantcultures of epidermis, dermis, and whole skin, cutaneous dendritic cellsemigrated into the medium during a 48 h incubation. (A) Emigrated cellswere double labeled for Langerin and MHC class II on cytospins and dou-ble-positive cells were scored (mean 7 SD, n ¼ 4, npo0.01, nnpo0.001).(B) By FACS, these migratory cells were double labeled for Langerin andMHC class II and analyzed (one representative experiment of at least three).Nearly all epidermal cells were double positive for both markers (left panel)in contrast to the dermal cells (middle panel). Mixed populations of Langer-hans cells and dermal dendritic cells emigrated from complete skin (rightpanel). (C) Dermal cells were prepared by collagenase digestion and cul-tured up to 3 d stained with anti-Langerin and MHC class II MoAb. ByFACS Langerin+ cells of all MHC class II expressing cells were determinedand are presented in percent (mean7SD, n¼ 5, npo0.001). An increase inLangerin+ cells in the dendritic cell population in the dermis could be bestobserved after 2 d of culture. For all experiments we used 929F3 MoAb.

Figure 4. Loss in intensity of Langerin expression in Langerhanscells during prolonged culture periods. Langerhans cells were gener-ated either by trypsinization of the epidermis and subsequent culture for4 d (upper panels) or by migration out of epidermal sheets during 4 d (lowerpanels). Cells were double labeled for Langerin (929F3 MoAb) and MHCclass II and analyzed by FACS. This experiment is representative of fourindependent experiments.

270 STOITZNER ETAL THE JOURNAL OF INVESTIGATIVE DERMATOLOGY

Page 6: Visualization and Characterization of Migratory Langerhans Cells in Murine Skin and Lymph Nodes by Antibodies Against Langerin/CD207

of Langerin-expressing cells increased up to an average54350722960 (Fig 7A) thus inducing an 11-fold increase inimmigrated Langerhans cells. In FACS analyses of skin draininglymph node cells (auricular and inguinal) from vehicle- orTNCB-treated mice we observed a pronounced increase in thepercentages of Langerinþ cells (Fig 7B). When thesepercentages were multiplied with the total cell numbersobtained in the hemocytometer we observed again a rise inabsolute Langerhans cell numbers in skin draining lymph nodesafter TNCB application (Fig 7C); however, this was not as

pronounced, which may be explained by the di¡erentexperimental settings.

Phenotype of Langerinþ cells in skin draining lymphnodes Langerinþ cells in the nodes of untreated mice werefurther analyzed, particularly with regard to their state ofmaturation. Cells were isolated from skin draining lymph nodes,enriched for dendritic cells with a Nycodenz-gradient. CD11cþ

and CD11c^ cell populations were analyzed for Langerin andMHC class II expression by FACS. No Langerin staining wasfound in the CD11c^ fraction emphasizing the speci¢city ofLangerin for dendritic cells. All Langerin-expressing cells weredetectable in the CD11cþ fraction representing lymph nodedendritic cells. Furthermore, Langerinþ cells expressed highlevels of MHC class II, which has been described to be a markerfor migratory Langerhans cells in skin draining lymph nodes(Ruedl et al, 2000; Henri et al, 2001) (Fig 8a). Almost allmigrating Langerinþ cells in the lymphatic vessels of the skinexpressed the maturation marker 2A1 (Fig 2E,F). We weretherefore interested whether the Langerinþ cells that arrive inthe lymph node in the ‘‘steady state’’ were already mature.Indeed, by immuno£uorescence on lymph node sections wenoted that most, but not all, Langerinþ cells coexpressedthe maturation marker 2A1� (Fig 8b). Thus, a distinct minor

Figure 5. Langerin+ cells are found in skin draining lymph nodes.(A) Cells from skin-draining and mesenteric lymph nodes were isolated,enriched on a Nycodenz-gradient and stained for Langerin. In skin drain-ing lymph nodes (left panel) we found substantial numbers of Langerin+

cells in contrast to mesenteric lymph nodes (right panel). Langerin stainingbold line, isotype staining, dashed line. One representative experiment ofthree is shown. (B) By immunohistochemistry high numbers of Langerin+

cells (blue color) can be appreciated in an auricular node (left panel) but onlyfew scattered Langerin+ cells in a mesenteric node (right panel). One repre-sentative experiment of three is shown. Panel B, original magni¢cation �60. For all experiments we used 929F3 MoAb.

Figure 6. Langerin+ cells localize to theTcell areas of skin draininglymph nodes. Lymph node sections were immunolabeled to determinethe localization of Langerin+ cells (929F3 MoAb). On consecutive sections(top row) it becomes apparent that Langerin+ cells (b) are in theTcell area asde¢ned by anti-CD3 reactivity (c) but not in the B cell follicles as de¢nedby B220 labeling (a). Double labeling of Langerin (blue cells) and CD3 (d)and CD19 (e) (brown cells) con¢rms the mutual exclusion of B cells andLangerin-reactive cells (e). Original magni¢cation: (a^c) � 40; (d,e) �240.

Figure 7. Increased numbers of Langerin+ cells in skin draininglymph nodes after contact sensitization with TNCB. Mice were trea-ted with the contact sensitizer TNCB or vehicle (acetone/olive oil) on theshaved abdominal or ear skin. After 24 h skin draining lymph node cellswere prepared and stained for Langerin (929F3 MoAb). (A) On cytospins,nuclei were counterstained with DAPI and the Langerin+ cells werecounted (mean7SD, n¼ 5, npo0.001). (B) By FACS, the percentage ofLangerin+ cells in total lymph node suspensions was determined. One re-presentative experiment of 11. (C) Absolute numbers of Langerhans cellsdetermined by relating FACS-derived percentages with hemocytometercounts (mean7SD, n¼11, npo0.0001).

TRACING MIGRATING LANGERHANS CELLS 271VOL. 120, NO. 2 FEBRUARY 2003

Page 7: Visualization and Characterization of Migratory Langerhans Cells in Murine Skin and Lymph Nodes by Antibodies Against Langerin/CD207

(o 3% of Langerinþ cells) subset of immature (i.e., 2A1)Langerhans cells was consistently detected in the lymph nodes.FACS analyses con¢rmed that almost all Langerinþ cells in theskin draining lymph node coexpressed the costimulatorymolecules CD86 and to a lesser extent CD40 (Fig 8c). Forcomparison, we induced a strong in£ammation in the skin byepicutaneous application of the contact allergen TNCB andexamined the maturation state of immigrated Langerhans cellsin the skin draining lymph nodes 24 and 48 h thereafter. Weobserved a slight but consistent increase in cells coexpressingLangerin and CD86 or Langerin and CD40; however, this wasnot statistically signi¢cant (Fig 8c). Additionally, the intensity of

these two costimulatory molecules on Langerinþ cells in thelymph nodes was consistently enhanced after application ofcontact allergen (Fig 8d).

DISCUSSION

Until now, the hallmark for murine Langerhans cells was the ten-nis-racket-shaped Birbeck granule that could only be visualizedby transmission electron microscopy (Birbeck et al, 1961; Wol¡,1967; Hashimoto, 1971). Langerin/CD207, a type II transmem-brane protein belonging to the family of Ca2þ -dependent(C-type) lectin receptors, was identi¢ed as a main molecularcomponent of Birbeck granules, ¢rst in humans (Valladeau et al,1999, 2000) and then in the mouse (Valladeau et al, 2002). Thedevelopment of monoclonal anti-mouse Langerin antibodies(Valladeau et al, 2002) made it possible for the ¢rst time to tracemurine Langerhans cells along their migration pathway.We showhere that Langerhans cells express Langerin during the entire mi-gration process from the epidermis through lymphatic vesselsinto the lymph nodes.

Origin of dendritic cells emigrating out of whole skinexplants Novel anti-mouse Langerin MoAb (Valladeau et al,2002) make it possible for the ¢rst time to determine the relativeproportions of Langerhans cells and dermal dendritic cells inmurine whole skin explant cultures. As expected, dendritic cells‘‘crawling out’’ from epidermal sheets are virtually all Langerinþ ,and dendritic cells originating from dermal sheets are largelyLangerin^. Still, surprisingly many Langerinþ cells occurred inthe dermal emigrant populations as compared with humandermal populations where this percentage rarely exceeded 5% ofall dendritic cells (Lenz et al, 1993). This is in contrast to the¢nding that immuno£uorescence of native dermis revealed onlyfew scattered Langerinþ cells. One explanation for thisdiscrepancy might be the minute pieces of epidermis thatinevitably adhere to the dermal sheet after dispase separation orhair follicles as observed by histologic hematoxylin and eosinstaining. Another source for Langerinþ Langerhans cells in thedermis could be emigrating Langerhans cells already on theirway out of the skin in the steady state, but one might notexpect such high numbers. A further possibility is thatLangerinþ cells in the dermal preparations are immigratingLangerhans cell precursors, expressing already MHC class II butnot yet Langerin. They would develop into Langerin-expressingcells during the emigration out of the dermal explants. Thispossibility is underscored by our observation that dermal cellsuspensions isolated by enzymatic digestion and cultured for 2 dcontained over ¢ve times more Langerinþ cells as compared

Figure 8. Phenotype of Langerin+ cells in skin draining lymphnodes. (a) Skin draining lymph node cells were isolated, enriched with aNycodenz-gradient, and analyzed by FACS for MHC class II and Langerinexpression. Langerin+ cells were only found in the CD11c+, i.e., dendriticcell fraction and expressed high levels of MHC class II. (b) Double labelingof skin draining lymph node sections with the maturation marker 2A1(green £uorescence) and Langerin (red £uorescence) shows that nearly allLangerin+ cells are mature (yellow £uorescence in A). (A) Overview,double exposure. Some mature non-Langerhans cells are marked witharrowheads, (B,C) Same ¢eld, single labeling. Higher magni¢cation of2A1 (E), Langerin (F), and overlap (D). Scale bars: (A) 50 mm; (B,C) 100mm; (D^F) 20 mm. (c) Skin draining lymph node cells were enriched witha Nycodenz-gradient and examined for expression of costimulatory mole-cules CD86 and CD40. A high percentage of Langerin+ cells expressedthese markers and numbers increased after induction of a contact hypersen-sitivity reaction (mean7SD, n¼ 5). (d) By FACS analysis an increase inintensity of CD86 (left panel) and CD40 (right panel) expression was ob-served in TNCB-treated mice (bold line) in contrast to untreated mice (thinline). Dotted lines are isotype controls. One representative experiment of¢ve is shown. For all experiments we used 929F3 MoAb.

272 STOITZNER ETAL THE JOURNAL OF INVESTIGATIVE DERMATOLOGY

Page 8: Visualization and Characterization of Migratory Langerhans Cells in Murine Skin and Lymph Nodes by Antibodies Against Langerin/CD207

with the onset of culture. Such a precursor population migratingout of human skin explants expressing CD14þ and Langerin hasbeen described by Larregina et al (2001); however, preliminaryexperiments did not reveal the presence of CD14þ cells infreshly isolated dermal cell suspensions.

Langerhans cells can be traced to the draining lymph nodesby their Langerin expression As Langerin is expressedintracellularly in Langerhans cells for quite a long time, we wereable to track migrating Langerhans cells to their destination inthe lymphoid organs. Henri et al (2001) observed that migratoryskin dendritic cells still express Langerin in the skin draininglymph nodes. Here we extend these initial ¢ndings in thatLangerinþ cells were found in the T cell areas and were almostalways absent from B cell follicles. No Langerin expressioncould be detected in the CD11c^ cells in the lymph nodesunderlining that Langerin is an exclusive marker for dendriticcells. Further characterization of Langerin-expressing cells in thelymph nodes revealed that these cells express high levels of MHCclass II as has been described for immigrated Langerhans cells inthe lymph nodes (Ruedl et al, 2000; Henri et al, 2001).Furthermore, we demonstrate that substantial numbers ofLangerinþ cells were only found in skin draining lymph nodes,whereas just few scattered cells were visible in the mesentericlymph nodes. This emphasizes the validity of Langerin as amarker for epidermis-derived Langerhans cells. Weak Langerinstaining in the mesenteric lymph nodes was shown by Valladeauet al (2002). They also described Langerinþ cells in epithelia ofthe lung and the tonsils. Similar Langerin-expressing cells in theepithelium of the gut (A. Kaser, personal communication) wouldbe a possible source for the Langerinþ cells in the mesentericlymphoid tissue.

To enhance the migration of Langerhans cells into the lymphnodes we induced contact hypersensitivity reactions with TNCBand observed an increase in Langerinþ cells in skin draininglymph nodes. Other studies have shown that higher numbers ofdendritic cells can be found in the lymph nodes after applicationof di¡erent contact sensitizers (Knight et al, 1985; Macatonia et al,1986; Botham et al, 1987). Dendritic cells were identi¢ed by theirforward/side scatter pro¢les in the FACS, their expression ofMHC class II and other dendritic cell markers (CD11c), or bythe presence of sensitizing trackers such as FITC. These assaysdid not allow an unequivocal discrimination between migratoryLangerhans cells and migratory dermal dendritic cells and theirrelative contributions to sensitization. Additionally, the assaysmay not always permit a clear-cut distinction betweenmigratory cells or lymph node-resident MHC class IIþ cellsbecause free FITC may di¡use through blood and lymph tospleen and lymph nodes, respectively, where it would stainresident dendritic cells (Pior et al, 1999; and own observations).The MoAb against Langerin used here are tools to visualizeselectively immigrated Langerhans cells in the skin draininglymph nodes and to study the relative contributions ofLangerhans cells to the induction of immune responses.

Immigrated Langerhans cells in the skin draining lymphnodes are largely mature in the steady-state situation Inthe steady-state, immature or partially mature Langerhans cellsare thought to carry self antigens to the lymph nodes therebymaintaining peripheral tolerance (Steinman and Nussenzweig,2002). Because of the lack of a Langerhans cell-speci¢c marker itwas not possible to de¢ne the maturation stage of immigratedLangerhans cells in the steady-state vs the in£ammatorysituation. Here we show for the ¢rst time that Langerhans cellstra⁄cking into the lymph node in the steady state alreadyexpress maturation marker 2A1, costimulatory molecules CD86and CD40, and high levels of MHC class II. This is in somecontrast to recent observations by Geissmann et al (2002) whoreported that Langerinþ cells were largely immature in humanlymph nodes, albeit in a di¡erent situation, i.e., a chronicin£ammation.We observed a small but consistent increase in the

expression of the CD86 and CD40 after application of contactallergen, i.e., in response to in£ammation. In keeping with ourdata, Ruedl et al (2001) showed that CD40þ/CD11cþ sortedlymph node cells, possibly Langerhans cells, expressed CD80and CD86 in the steady state and upregulated them afterapplication of a £uorescent tracer, i.e., in response toin£ammation, too. In addition, the lymph node dendritic cellsstill took up and processed antigens to some degree alsoindicating an incomplete maturation state (Koch et al, 1995;Ruedl et al, 2001).

In a mouse model of tolerance induction by injection of theanti-DEC-205 MoAb coupled to a protein, tolerance was brokenafter simultaneous injection of anti-CD40 MoAb, whichincreased surface expression of CD86 and CD40 on CD11cþ

lymph node dendritic cells (Hawiger et al, 2001). Menges et al(2002) demonstrated that partially mature dendritic cellsexpressed lower levels of CD40 molecules and prevented thedevelopment of experimental autoimmune encephalomyelitis inmice in contrast to fully matured dendritic cells whenadoptively transferred.

Migration is closely linked to the maturation process.Upregulation of CCR7, for instance, is a prerequisite formigration to occur (Caux et al, 2002). Therefore, it is unlikelythat Langerhans cells arriving in the lymph node in the steadystate would still express an immature phenotype, even thoughGeissmann et al (2002) recently suggested a way of uncouplingmigration (CCR7 expression) from maturation (CD83, CD86,and CD208 expression). We observed here that, in response toin£ammation, higher numbers of more mature Langerhans cellsarrive in the lymph node. This would increase the frequency ofencounters with T cells and thus the generation of immunity.Thus, a combination of migration levels and the stage ofdendritic cell maturation might determine the outcome of theresponse in the lymph node. This new antibody will allow togain further insights into these functional states of Langerhanscells.

The continued support of Dr Peter Fritsch, Chairman of the Department of Derma-tology is greatly appreciated.This work was supported by a grant from the AustrianScience Fund to N. Romani (FWF grant no. P-14949-MED).

REFERENCES

Austyn JM: New insights into the mobilization and phagocytic activity of dendriticcells. J Exp Med 183:1287^1292, 1996

Banchereau J, Steinman RM: Dendritic cells and the control of immunity. Nature392:245^252, 1998

Birbeck MS, Breathnach AS, Everall JD: An electron microscopic study of basal mel-anocytes and high level clear cells (Langerhans cell) in vitiligo. J Invest Dermatol37:51^64, 1961

Botham PA, Rattray NJ, Walsh ST, Riley EJ: Control of the immune response tocontact sensitizing chemicals by cutaneous antigen-presenting cells. Br J Derma-tol 117:1^9, 1987

Caux C, Vandervliet B, Massacrier C, et al: Regulation of dendritic cell recruitmentby chemokines.Transplantation 73:S7^S11, 2002

Cumberbatch M, Dearman RJ, Kimber I: Langerhans cells require signals from bothtumour necrosis factor-a and interleukin-1b for migration. Immunology92:388^395, 1997

Enk AH, Angeloni VL, Udey MC, Katz SI: An essential role for Langerhans cell-derived IL-1 b in the initiation of primary immune responses in skin. J Immu-nol 150:3698^3704, 1993

Flores-Romo L: In vivo maturation and migration of dendritic cells. Immunology102:255^262, 2001

Geissmann F, Dieu-Nosjean MC, Dezutter C, et al: Accumulation of immature Lan-gerhans cells in human lymph nodes draining chronically in£amed skin. J ExpMed 196:417^430, 2002

Hashimoto K: Langerhans cell granule. An endocytic organelle. Arch Dermatol104:148^160, 1971

Hawiger D, Inaba K, Dorsett Y, et al: Dendritic cells induce peripheral T cell unre-sponsiveness under steady state conditions in vivo. J Exp Med 194:769^779, 2001

Henri S,Vremec D, Kamath A, et al: The dendritic cell populations of mouse lymphnodes. J Immunol 167:741^748, 2001

TRACING MIGRATING LANGERHANS CELLS 273VOL. 120, NO. 2 FEBRUARY 2003

Page 9: Visualization and Characterization of Migratory Langerhans Cells in Murine Skin and Lymph Nodes by Antibodies Against Langerin/CD207

Inaba K, Inaba M, Romani N, et al: Generation of large numbers of dendritic cellsfrom mouse bone marrow cultures supplemented with granulocyte/macro-phage colony-stimulating factor. J Exp Med 176:1693^1702, 1992

Kellermann SA, Hudak S, Oldham ER, Liu YJ, McEvoy LM: The CC chemokinereceptor-7 ligands 6Ckine and macrophage in£ammatory protein-3b are po-tent chemoattractants for in vitro- and in vivo-derived dendritic cells. J Immu-nol 162:3859^3864, 1999

Kitano Y, Okado N: Separation of the epidermal sheet by dispase. Br J Dermatol108:555^560, 1983

Knight SC, Krejci J, Malkovsky M, Colizzi V, Gautam A, Asherson GL: The role ofdendritic cells in the initiation of immune responses to contact sensitizers. I. Invivo exposure to antigen. Cell Immunol 94:427^434, 1985

Kobayashi Y, Matsumoto M, Kotani M, Makino T: Possible involvement of matrixmetalloproteinase-9 in Langerhans cell migration and maturation. J Immunol163:5989^5993, 1999

Koch F, Trockenbacher B, K'mpgen E, et al: Antigen processing in populations ofmature murine dendritic cells is caused by subsets of incompletely maturedcells. J Immunol 155:93^100, 1995

Koch F, K'mpgen E, Schuler G, Romani N: Isolation, enrichment and culture ofmurine epidermal Langerhans cells. In: Stagg AJ, Robinson S (eds). DendriticCell Protocols. Totowa, NJ: Humana Press, 2001: pp 43^62

Larregina AT, Morelli AE, Spencer LA, Logar AJ, Watkins SC, Thomson AW, FaloLD Jr: Dermal-resident CD14þ cells di¡erentiate into Langerhans cells. NatureImmunol 2:1151^1158, 2001

Larsen CP, Steinman RM, Witmer-Pack M, Hankins DF, Morris PJ, Austyn JM:Migration and maturation of Langerhans cells in skin transplants and explants.J Exp Med 172:1483^1493, 1990

Lenz A, Heine M, Schuler G, Romani N: Human and murine dermis contain den-dritic cells. Isolation by means of a novel method and phenotypical and func-tional characterization. J Clin Invest 92:2587^2596, 1993

Lukas M, St˛ssel H, Hefel L, et al: Human cutaneous dendritic cells migrate throughdermal lymphatic vessels in a skin organ culture model. J Invest Dermatol106:1293^1299, 1996

Macatonia SE, Edwards AJ, Knight SC: Dendritic cells and the initiation of contactsensitivity to £uorescein isothiocyanate. Immunology 59:509^514, 1986

McLellan AD, Starling GC, Hart DNJ: Isolation of human blood dendritic cells bydiscontinuous Nycodenz gradient centrifugation. J Immunol Methods 184:81^89,1995

McLellan AD, Kapp M, Eggert A, et al: Anatomic location and T-cell stimulatoryfunctions of mouse dendritic cell subsets de¢ned by CD4 and CD8 expression.Blood 99:2084^2093, 2002

Mellman I, Steinman RM: Dendritic cells Specialized and regulated antigen proces-sing machines. Cell 106:255^258, 2001

Menges M, R˛ssner S, Voigtl'nder C, et al: Repetitive injections of dendritic cellsmatured with tumor necrosis factor a induce antigen-speci¢c protection ofmice from autoimmunity. J Exp Med 195:15^21, 2002

Ortner U, Inaba K, Koch F, Heine M, Miwa M, Schuler G, Romani N: An im-proved isolation method for murine migratory cutaneous dendritic cells. J Im-munol Methods 193:71^79, 1996

Pierre P,Turley SJ, Gatti E, et al: Developmental regulation of MHC class II transportin mouse dendritic cells. Nature 388:787^792, 1997

Pior J,Vogl T, Sorg C, Macher E: Free hapten molecules are dispersed by way of thebloodstream during contact sensitization to £uorescein isothiocyanate. J InvestDermatol 113:888^893, 1999

Price AA, Cumberbatch M, Kimber I, Ager A: a6 integrins are required forLangerhans cell migration from the epidermis. J Exp Med 186:1725^1735,1997

Ratzinger G, Stoitzner P, Ebner S, et al: Matrix metalloproteinases 9 and 2 are neces-sary for the migration of Langerhans cells and dermal dendritic cells from hu-man and murine skin. J Immunol 168:4361^4371, 2002

Romani N, Bhardwaj N, Pope M, et al: Dendritic cells. In: Herzenberg LA, WeirDM, Herzenberg L, Blackwell C (eds).Weir’s Handbook of Experimental Immunol-ogy. Oxford: Blackwell Science, 1997: pp 1561^1564

Ruedl C, Koebel P, Bachmann M, Hess M, Karjalainen K: Anatomical origin ofdendritic cells determines their life span in peripheral lymph nodes. J Immunol165:4910^4916, 2000

Ruedl C, Koebel P, Karjalainen K: In vivo-matured Langerhans cells continue to takeup and process native proteins unlike in vitro-matured counterparts. J Immunol166:7178^7182, 2001

Saeki H, Moore AM, Brown MJ, Hwang ST: Cutting edge. Secondary lymphoid-tissue chemokine (SLC) and CC chemokine receptor 7 (CCR7) participate inthe emigration pathway of mature dendritic cells from the skin to regionallymph nodes. J Immunol 162:2472^2475, 1999

Salomon B, Cohen JL, Masurier C, Klatzmann D: Three populations of mouselymph node dendritic cells with di¡erent origins and dynamics. J Immunol160:708^717, 1998

Schuler G, Steinman RM: Murine epidermal Langerhans cells mature into potentimmunostimulatory dendritic cells in vitro. J Exp Med 161:526^546, 1985

Schwarzenberger K, Udey MC: Contact allergens and epidermal proin£ammatorycytokines modulate Langerhans cell E-cadherin expression in situ. J Invest Der-matol 106:553^558, 1996

Steinman RM, Nussenzweig MC: Avoiding horror autotoxicus. The importance ofdendritic cells in peripheral T cell tolerance. Proc Natl Acad Sci USA 99:351^358,2002

Steinman RM, Pack M, Inaba K: Dendritic cells in the T-cell areas of lymphoidorgans. Immunol Rev 156:25^37, 1997

Stoitzner P, Zanella M, Ortner U, et al: Migration of Langerhans cells and dermaldendritic cells in skin organ cultures: augmentation by TNF-a and IL-1b.J Leukocyte Biol 66:462^470, 1999

Stoitzner P, Ratzinger G, Koch F, et al: Interleukin-16 supports the migration of Lan-gerhans cells, partly in a CD4-independent way. J Invest Dermatol 116:641^649,2001

Stoitzner P, Pfaller K, St˛ssel H, Romani N: A close-up view of migrating Langer-hans cells in the skin. J Invest Dermatol 118:117^125, 2002

Tyndall C, La Mantia G,Thacker CM, Favaloro J, Kamen R: A region of the poly-oma virus genome between the replication origin and late protein coding se-quences is required in cis for both early gene expression and viral DNAreplication. Nucleic Acids Res 9:6231^6238, 1981

Valladeau J, Duvert-Frances V, Pin JJ, et al: The monoclonal antibody DCGM4 recog-nizes Langerin, a protein speci¢c of Langerhans cells, and is rapidly interna-lized from the cell surface. Eur J Immunol 29:2695^2704, 1999

Valladeau J, Ravel O, Dezutter-Dambuyant C, et al: Langerin, a novel C-type lectinspeci¢c to Langerhans cells, is an endocytic receptor that induces the formationof Birbeck granules. Immunity 12:71^81, 2000

Valladeau J, Clair-Moninot V, Dezutter-Dambuyant C, et al: Identi¢cation of mouseLangerin/CD207 in Langerhans cells and some dendritic cells of lymphoid tis-sues. J Immunol 168:782^792, 2002

Weinlich G, Heine M, St˛ssel H, et al: Entry into a¡erent lymphatics and maturationin situ of migrating cutaneous dendritic cells. J Invest Dermatol 110:441^448,1998

Weiss JM, Sleeman J, Renkl AC, et al: An essential role for CD44 variant isoforms inepidermal Langerhans cell and blood dendritic cell function. J Cell Biol137:1137^1147, 1997

Wol¡ K:The ¢ne structure of the Langerhans cell granule. J Cell Biol 35:466^473, 1967

274 STOITZNER ETAL THE JOURNAL OF INVESTIGATIVE DERMATOLOGY