1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct...

11
A CD1a 1 /CD11c 1 Subset of Human Blood Dendritic Cells Is a Direct Precursor of Langerhans Cells 1 Tomoki Ito,* ² Muneo Inaba, ² Kayo Inaba, Junko Toki, ² Shinji Sogo, § Tomoko Iguchi,* ² Yasushi Adachi, ² Kazuyuki Yamaguchi,* Ryuichi Amakawa,* Jenny Valladeau, Sem Saeland, Shirou Fukuhara,* and Susumu Ikehara 2 * Based on the relative expression of CD11c and CD1a, we have identified three fractions of dendritic cells (DCs) in human peripheral blood, including a direct precursor of Langerhans cells (LCs). The first two fractions were CD11c 1 DCs, comprised of a major CD1a 1 /CD11c 1 population (fraction 1), and a minor CD1a 2 /CD11c 1 component (fraction 2). Both CD11c 1 fractions displayed a monocyte-like morphology, endocytosed FITC-dextran, expressed CD45RO and myeloid markers such as CD13 and CD33, and possessed the receptor for GM-CSF. The third fraction was comprised of CD1a 2 /CD11c 2 DCs (fraction 3) and resembled plasmacytoid T cells. These did not uptake FITC-dextran, were negative for myeloid markers (CD13/CD33), and expressed CD45RA and a high level of IL-3Ra, but not GM-CSF receptors. After culture with IL-3, fraction 3 acquired the characteristics of mature DCs; however, the expression of CD62L (lymph node-homing molecules) remained unchanged, indi- cating that fraction 3 can be a precursor pool for previously described plasmacytoid T cells in lymphoid organs. Strikingly, the CD1a 1 /CD11c 1 DCs (fraction 1) quickly acquired LC characteristics when cultured in the presence of GM-CSF 1 IL-4 1 TGF-b1. Thus, E-cadherin, Langerin, and Lag Ag were expressed within 1 day of culture, and typical Birbeck granules were observed. In contrast, neither CD1a 2 /CD11c 1 (fraction 2) nor CD1a 2 /CD11c 2 (fraction 3) cells had the capacity to differ- entiate into LCs. Furthermore, CD14 1 monocytes only expressed E-cadherin, but lacked the other LC markers after culture in these cytokines. Therefore, CD1a 1 /CD11c 1 DCs are the direct precursors of LCs in peripheral blood. The Journal of Immunology, 1999, 163: 1409 –1419. D endritic cells (DCs) 3 are bone marrow-derived profes- sional APCs that initiate and regulate immune responses (1–3). Recent studies have demonstrated that DCs in situ can exist in different maturational states and pathways of differ- entiation. Subtypes of DCs with some differences in morphology, phenotype, and function include interdigitating cells in lymphoid organs (4), peripheral blood DCs (5, 6), Langerhans cells (LCs) in the epidermis of the skin (7, 8), dermal DCs (9, 10), and thymic DCs (11, 12). In the human tonsil, DCs in the germinal center (13) and plasmacytoid T cells in the T cell zones (14) have recently been identified as new DC subsets. Hemopoietic progenitors that differentiate into various types of DCs are present in the cord blood (15, 16), bone marrow (17), and peripheral blood (18, 19). At least two separate developmental pathways of DCs from CD34 1 progenitors have been shown (16, 19): one pathway involves CD1a 1 /CD14 2 cells with the features of epidermal LCs. The other pathway includes a CD1a 2 /CD14 1 intermediate that can differentiate into monocyte-derived or dermal DCs that lack LC markers. GM-CSF and TNF-a are required in both pathways, but TGF-b1 is found to be important for the for- mation of Birbeck granules and Lag Ag of LCs (20 –22). It has also been found that peripheral blood monocytes cultured with a com- bination of GM-CSF, IL-4, and TNF-a differentiate into mature DCs (23), and that LCs may develop if TGF-b1 is added (24). The identification of DC intermediates in the peripheral blood is, there- fore, important, because it is thought that most blood DCs are either en route from the bone marrow to peripheral tissues, or from nonlymphoid tissues to the regional lymph nodes and spleen. Previously, DCs were purified from the peripheral blood after a 1- to 2-day culture period. These DCs were morphologically, phe- notypically, and functionally different from the DCs present in the circulation (25, 26). Recently, DCs have been isolated from the peripheral blood by immunoselection methods without culture. DCs thus prepared are not the morphologically typical DCs, and, interestingly, there are two subsets with distinct phenotype (27– 29). The nature, origin, and commitment of these DCs require further analysis. In the present study, we have isolated fractions of blood DCs using magnetic beads and a multicolor sorting system. We have distinguished three distinct fractions by extensive surface marker analyses, and have examined their functions and develop- mental capacities. During these investigations, we have identified a distinct subset of DCs that quickly differentiates into LCs. Strunk et al. have reported that, in the peripheral blood, only CD34 1 progenitors expressing the skin-homing receptor differen- tiate into LCs (after 10- to 18-day culture) (19). It has recently been reported that, in the presence of GM-CSF, IL-4, and TGF-b1 for 6 days, human peripheral blood monocytes (CD14 1 cells) can differentiate into LCs characterized by the expression of CD1a, *First Department of Internal Medicine and ² First Department of Pathology, Kansai Medical University, Moriguchi, Osaka, Japan; Department of Zoology, Kyoto Uni- versity, Sakyo, Kyoto, Japan; § Cellular Technology Institute, Otsuka Pharmaceutical Co., Tokushima, Japan; and Schering-Plough, Laboratory for Immunological Re- search, Dardilly, France Received for publication January 11, 1999. Accepted for publication May 20, 1999. 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 a grant from the Ministry of Health and Welfare’s Study Group on Specific Diseases Research and by grants from the Ministry of Ed- ucation, Science, and Culture, and the Japan Private School Promotion Foundation. 2 Address correspondence and reprint requests to Dr. Susumu Ikehara, First Depart- ment of Pathology, Kansai Medical University, 10-15 Fumizono-cho, Moriguchi City, Osaka 570-8506, Japan. E-mail address: [email protected] 3 Abbreviations used in this paper: DC, dendritic cell; EM, electron microscope; LC, Langerhans cell; PerCP, peridini-chlorophyll protein; PI, propidium iodide. Copyright © 1999 by The American Association of Immunologists 0022-1767/99/$02.00

Transcript of 1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct...

Page 1: 1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct ...imed1.kmu-ac.jp/pdf/thesis_dissertation/5.pdf · A CD1a1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct Precursor

A CD1a1/CD11c1 Subset of Human Blood Dendritic Cells Is aDirect Precursor of Langerhans Cells1

Tomoki Ito,* † Muneo Inaba,† Kayo Inaba,‡ Junko Toki,† Shinji Sogo,§ Tomoko Iguchi,*†

Yasushi Adachi,† Kazuyuki Yamaguchi,* Ryuichi Amakawa,* Jenny Valladeau,¶ Sem Saeland,¶

Shirou Fukuhara,* and Susumu Ikehara2*

Based on the relative expression of CD11c and CD1a, we have identified three fractions of dendritic cells (DCs) in humanperipheral blood, including a direct precursor of Langerhans cells (LCs). The first two fractions were CD11c1 DCs, comprised ofa major CD1a1/CD11c1 population (fraction 1), and a minor CD1a2/CD11c1 component (fraction 2). Both CD11c1 fractionsdisplayed a monocyte-like morphology, endocytosed FITC-dextran, expressed CD45RO and myeloid markers such as CD13 andCD33, and possessed the receptor for GM-CSF. The third fraction was comprised of CD1a2/CD11c2 DCs (fraction 3) andresembled plasmacytoid T cells. These did not uptake FITC-dextran, were negative for myeloid markers (CD13/CD33),and expressed CD45RA and a high level of IL-3Ra, but not GM-CSF receptors. After culture with IL-3, fraction 3 acquired thecharacteristics of mature DCs; however, the expression of CD62L (lymph node-homing molecules) remained unchanged, indi-cating that fraction 3 can be a precursor pool for previously described plasmacytoid T cells in lymphoid organs. Strikingly, theCD1a1/CD11c1 DCs (fraction 1) quickly acquired LC characteristics when cultured in the presence of GM-CSF1 IL-4 1

TGF-b1. Thus, E-cadherin, Langerin, and Lag Ag were expressed within 1 day of culture, and typical Birbeck granules wereobserved. In contrast, neither CD1a2/CD11c1 (fraction 2) nor CD1a2/CD11c2 (fraction 3) cells had the capacity to differ-entiate into LCs. Furthermore, CD141 monocytes only expressed E-cadherin, but lacked the other LC markers after culturein these cytokines. Therefore, CD1a1/CD11c1 DCs are the direct precursors of LCs in peripheral blood. The Journal ofImmunology, 1999, 163: 1409 –1419.

D endritic cells (DCs)3 are bone marrow-derived profes-sional APCs that initiate and regulate immune responses(1–3). Recent studies have demonstrated that DCs in situ

can exist in different maturational states and pathways of differ-entiation. Subtypes of DCs with some differences in morphology,phenotype, and function include interdigitating cells in lymphoidorgans (4), peripheral blood DCs (5, 6), Langerhans cells (LCs) inthe epidermis of the skin (7, 8), dermal DCs (9, 10), and thymicDCs (11, 12). In the human tonsil, DCs in the germinal center (13)and plasmacytoid T cells in the T cell zones (14) have recentlybeen identified as new DC subsets.

Hemopoietic progenitors that differentiate into various types ofDCs are present in the cord blood (15, 16), bone marrow (17), andperipheral blood (18, 19). At least two separate developmentalpathways of DCs from CD341 progenitors have been shown (16,19): one pathway involves CD1a1/CD142 cells with the features

of epidermal LCs. The other pathway includes a CD1a2/CD141

intermediate that can differentiate into monocyte-derived or dermalDCs that lack LC markers. GM-CSF and TNF-a are required inboth pathways, but TGF-b1 is found to be important for the for-mation of Birbeck granules and Lag Ag of LCs (20–22). It has alsobeen found that peripheral blood monocytes cultured with a com-bination of GM-CSF, IL-4, and TNF-a differentiate into matureDCs (23), and that LCs may develop if TGF-b1 is added (24). Theidentification of DC intermediates in the peripheral blood is, there-fore, important, because it is thought that most blood DCs areeither en route from the bone marrow to peripheral tissues, or fromnonlymphoid tissues to the regional lymph nodes and spleen.

Previously, DCs were purified from the peripheral blood after a1- to 2-day culture period. These DCs were morphologically, phe-notypically, and functionally different from the DCs present in thecirculation (25, 26). Recently, DCs have been isolated from theperipheral blood by immunoselection methods without culture.DCs thus prepared are not the morphologically typical DCs, and,interestingly, there are two subsets with distinct phenotype (27–29). The nature, origin, and commitment of these DCs requirefurther analysis. In the present study, we have isolated fractions ofblood DCs using magnetic beads and a multicolor sorting system.We have distinguished three distinct fractions by extensive surfacemarker analyses, and have examined their functions and develop-mental capacities. During these investigations, we have identifieda distinct subset of DCs that quickly differentiates into LCs.

Strunk et al. have reported that, in the peripheral blood, onlyCD341 progenitors expressing the skin-homing receptor differen-tiate into LCs (after 10- to 18-day culture) (19). It has recentlybeen reported that, in the presence of GM-CSF, IL-4, and TGF-b1for 6 days, human peripheral blood monocytes (CD141 cells) candifferentiate into LCs characterized by the expression of CD1a,

*First Department of Internal Medicine and†First Department of Pathology, KansaiMedical University, Moriguchi, Osaka, Japan;‡Department of Zoology, Kyoto Uni-versity, Sakyo, Kyoto, Japan;§Cellular Technology Institute, Otsuka PharmaceuticalCo., Tokushima, Japan; and¶Schering-Plough, Laboratory for Immunological Re-search, Dardilly, France

Received for publication January 11, 1999. Accepted for publication May 20, 1999.

The costs of publication of this article were defrayed in part by the payment of pagecharges. This article must therefore be hereby markedadvertisementin accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.1 This work was supported by a grant from the Ministry of Health and Welfare’sStudy Group on Specific Diseases Research and by grants from the Ministry of Ed-ucation, Science, and Culture, and the Japan Private School Promotion Foundation.2 Address correspondence and reprint requests to Dr. Susumu Ikehara, First Depart-ment of Pathology, Kansai Medical University, 10-15 Fumizono-cho, Moriguchi City,Osaka 570-8506, Japan. E-mail address: [email protected] Abbreviations used in this paper: DC, dendritic cell; EM, electron microscope; LC,Langerhans cell; PerCP, peridini-chlorophyll protein; PI, propidium iodide.

Copyright © 1999 by The American Association of Immunologists 0022-1767/99/$02.00

Page 2: 1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct ...imed1.kmu-ac.jp/pdf/thesis_dissertation/5.pdf · A CD1a1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct Precursor

E-cadherin, Lag Ag, and Birbeck granules (24). We show in thisstudy, however, that the most direct precursors of LCs in humanblood are a distinctive subset of CD1a1/CD11c1/CD142 DCs.

Materials and MethodsMedia and reagents

The following culture medium was used throughout experiments: RPMI1640 supplemented with 2 mML-glutamine, 100 U/ml penicillin, 100mg/ml streptomycin, and heat-inactivated 10% human AB serum (ICN Bio-medicals, Aurora, OH). Monocyte-conditioned medium was prepared ac-

cording to Reddy et al. (30). Briefly, blood mononuclear cells were layeredonto humang-globulin-coated plates for 1 h, and nonadherent cells werewashed away. The dish-adherent cells were incubated in fresh medium for24 h. The supernatant was harvested and frozen at220°C until use. Thefollowing recombinant human cytokines were purchased from BoehringerMannheim (Indianapolis, IN): GM-CSF (used at a concentration of 100ng/ml), TNF-a (2.5 ng/ml), TGF-b1 (1 ng/ml), IL-3 (10 ng/ml), and IL-4(50 ng/ml). The sources of mAb used in our studies are listed in Table I.FITC-, PE-, PerCP-, PE Cy5-, or biotin-labeled isotype-matched controlswere purchased from Becton Dickinson (Sunnyvale, CA) or PharMingen(San Diego, CA). When unlabeled mAb or biotinylated mAbs were used asthe primary reagents, PE-labeled goat anti-mouse IgG F(ab9)2 (Jackson

Table I. Abs used for phenotypical analysisa

Clone Isotype Source Clone Isotype Source

CD1a BB-5 IgG1 TAG CD42b SZ2 IgG1 IMCD1a BL-6 IgG1 IM CD44 F10-44-2 IgG2a SerotecCD1c M241 IgG1 ANC CD45 HI30 IgG1 PMCD2 T11 IgG1 C CD45RA 2H4 IgG1 CCD3 Leu-4 IgG1 BD CD45RO UCHL1 IgG2a IMCD3 M2AB IgG1 EXA CD49d HP2/1 IgG1 IMCD3 UCHT1 IgG1 PM CD49e SAM1 IgG2b IMCD4 Leu-3a IgG1 BD CD50 HP2/19 IgG2a IMCD4 7E14 IgG1 EXA CD54 Leu-54 IgG2b BDCD5 T1 IgG2a C CD54 D3.6 IgG2b EXACD5 M28623 IgG2a EXA CD56 Leu-19 IgG1 BDCD7 3A1 IgG2b C CD61 SZ21 IgG1 IMCD7 G34 IgG2a EXA CD62L TQ-1 IgG1 CCD8 T8 IgG1 C CD62P CLBThromb/6 IgG1 IMCD9 C3-3A2 IgG1 ANC CD64 10.1 IgG1 PMCD10 J5 IgG2a C CD68 KP-1 IgG1 DakoCD11a HI111 IgG1 PM CD70 BU69 IgG1 ANCCD11b ICRF(44) IgG1 PM CD71 LO1.1 IgG2a BDCD11b Mo1 IgM C CD72 J4-117 IgG2a PMCD11c Leu-M5 IgG2b BD CD80 BB-1 IgM ANCCD11c 3.9 IgG1 Serotec CD83 HB15a IgG2b IMCD13 WM15 IgG1 PM CD86 IT2.2 IgG2b PMCD14 Leu-M3 IgG2b BD CD90 5E10 IgG1 PMCD14 FWKW-1 IgG2b EXA CD95(FAS) UB2 IgG1 MBLCD14 UCHM1 IgG2a ANC CD103 2G5 IgG2a IMCD15 80H5 IgM IM CD114(G-CSFR) LMM741 IgG1 PMCD15s 2H5 IgM PM CD116(GM-CSFR) M5D12 IgM PMCD16 Leu-11a IgG1 BD CD117(C-KIT) 95C3 IgG1 IMCD16 J5511 IgG1 EXA CD121a(IL-1R 1) 6B5 IgG2a PMCD16 3G8 IgG1 PM CD122(IL-2Rb) TIC-1 IgG1 ENDCD18 L130 IgG1 BD CD123(IL-3Ra) 9F5 IgG1 PMCD19 B4 IgG1 C CD130 AM64 IgG1 PMCD19 1G9 IgG1 EXA CD135(Flt3) SF1.340 IgG1 IMCD19 HIB19 IgG1 PM CD152(CTLA-4) BNI3 IgG2a IMCD20 B1 IgG2a C CD154(CD40L) 24-31 IgG1 ANCCD21 BL13 IgG1 IM E-cadherin HECD-1 IgG1 TakaraCD23 9P25 IgG1 IM CLA HECA-452 RatIgM PMCD25(IL-2Ra) 2A3 IgG1 BD Lag IgG1 Dr. ImamuraCD27 M-T271 IgG1 ANC HLA-DR L243 IgG2a BDCD28 KOLT-2 IgG1 Nichirei HLA-DR Tu36 IgG2b PMCD29 4B4 IgG1 C HLA-DQ TU169 IgG2a PMCD30 BerH8 IgG1 PM HLA-DQ 1a3 IgG2a LETCD32 AT10 IgG1 Serotec Glycophorin A KC16 IgG1 IMCD33 My9 IgG2b C TCR-ab WT31 IgG1 BDCD33 Leu-M9 IgG1 BD TCR-gd 11F2 IgG1 BDCD34 HPCA-2 IgG1 BDCD34 QBEnd10 IgG1 IMCD35 AR2 IgG1 EXACD36 FA6.152 IgG1 IMCD38 Leu-17 IgG1 BDCD38 T16 IgG1 IMCD40 5C3 IgG1 PMCD41 SZ22 IgG1 IM

Abbreviations: TAG, BioSource International, Tago Products (Flynn Road, CA); IM, Immunotech (Marseille, France); ANC, Ancell (Bayport, MN); C, Coulter Immunology(Hialeah, FL); BD, Becton Dickinson; EXA, Exalpha (Boston, MA); PM, PharMingen; Serotec, Serotec (Kindlington, U.K.); Nichirei (Tokyo, Japan); Dako (Glostrup, Denmark);MBL, Medical & Biological Laboratories (Nagoya, Aichi, Japan); END, Endogen (Woburn, MA); Takara, Takara Biomedicals (Kusatsu, Shiga, Japan); LET, Leinco Tech-nologies (Manchester, MO).

1410 LC PRECURSORS IN HUMAN PERIPHERAL BLOOD

Page 3: 1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct ...imed1.kmu-ac.jp/pdf/thesis_dissertation/5.pdf · A CD1a1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct Precursor

ImmunoResearch, West Grove, PA), FITC-labeled goat anti-mouse IgGF(ab9)2 (Becton Dickinson), FITC-labeled polyclonal anti-rat Ig (Phar-Mingen), or tri-color-labeled streptavidin (streptavidin-RED670; LifeTechnologies, Glasgow, U.K.) were employed as secondary reagents. ThemAbs against Langerin (DCGM4) (31) and Lag-1 (32) were used to de-termine the differentiation into LCs.

A cell permeabilization kit, FIX & PERM (Caltag, Burlingame, CA),was used for intracellular staining.

Enrichment and analyses of peripheral blood DCs

PBMC were isolated by Lymphoprep (Nycomed Pharma, Oslo, Norway)gradient centrifugation of heparinized blood (50–60 ml) obtained fromeach healthy volunteer. PBMC were incubated with anti-CD3 (HIT3a) andanti-CD14 (M5E2) mAbs (both from PharMingen) for 30 min on ice, andcells binding these mAb were removed using sheep anti-mouse Ig-coatedmagnetic beads (M-450; Dynal, Oslo, Norway). CD32/CD142 cells were

FIGURE 1. Detection and purification ofDC fractions in the peripheral blood. DCfractions identified by four-color EPICSELITE cell sorter (A andB). A DC-enrichedpopulationwasstainedwithFITC-labeledanti-CD1a mAb, PE-labeled anti-CD11c mAb,PerCP-labeled anti-HLA-DR mAb, and bio-tinylated mAbs against lineage markers(CD3, CD7, CD14, CD16, CD19), followedby streptavidin-Red 613. Three fractions ofDC (B), detected as CD1a1/CD11c1 (frac-tion 1), CD1a2/CD11c1 (fraction 2), andCD1a2/CD11c2 (fraction 3), are present inthe lin2/HLA-DR1 fraction (designated as Rin A). These three fractions of DCs were alsoobtained by a two-color FACStar cell sorter(C andD) (details described inMaterials andMethods). Fraction 1 was sorted as lin2/CD1a1 cells after staining with PE (orFITC)-labeled mAbs against lineage markersplus FITC (or PE)-labeled anti-CD1a mAb(R1 in C). Fractions 2 and 3 were purified aslin2/CD1a2/CD11c1 cells (R2 in D) andlin2/CD1a2/CD11c2 cells (R3 in D), re-spectively, after being stained with FITC-la-beled mAbs against lineage markers/CD1aplus PE-labeled anti-CD11c mAb. Figuresrepresentative of more than 10 experimentsare shown.

FIGURE 2. Morphology of DC fractions.Freshly prepared or cultured peripheral bloodDC fractions were histologically examined bylight or electron microscopy. Monocyte-con-ditioned medium was used for the cultures offractions 1 and 2. IL-3-supplemented mediumwas used for fraction 3.A–C, Fraction 1;D–F,fraction 2; G–I, fraction 3. A, D, and G,Freshly prepared DCs stained by May-Giemsasolution (3250).B, E, andH, Freshly preparedDCs examined by EM (33000).C, F, and I,DCs, cultured for 3 days, stained by May-Gi-emsa solution (3250). No Birbeck granuleswere observed in any fractions under theseconditions. Results are representative of eightexperiments.

1411The Journal of Immunology

Page 4: 1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct ...imed1.kmu-ac.jp/pdf/thesis_dissertation/5.pdf · A CD1a1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct Precursor

1412 LC PRECURSORS IN HUMAN PERIPHERAL BLOOD

Page 5: 1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct ...imed1.kmu-ac.jp/pdf/thesis_dissertation/5.pdf · A CD1a1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct Precursor

further incubated with CD4-conjugated microbeads (Miltenyi Biotec., Ber-gisch Gladbach, Germany), and the CD41 cells were then enriched throughMini MACS magnetic separation column (Miltenyi Biotec.). Using thisprotocol, the percentage of DCs (,1% of total PBMC) increased to 20–50%, depending on the individuals (n . 30). The resultant DC-enrichedpopulation (CD41/CD32/CD142 cells) was stained with PE-labeled anti-CD11c (LeuM5), FITC-labeled anti-CD1a (BB-5), PerCP-labeledHLA-DR (L243), and a mixture of biotinylated mAbs against lineagemarkers (CD3; M2AB, binding to a different determinant from that recog-nized by the previous anti-CD3 mAb, CD7; G34, CD14; UCHM1, bindingto a different determinant from that recognized by the previous anti-CD14mAb, CD16; 3G8 and CD19; HIB19), followed by RED613-streptavidin(Life Technologies). The stained cells were analyzed (or sorted forcytologic assay, T cell proliferation assay, and analyses of endocytosis) byan EPICS ELITE flow cytometer (Coulter, Hialeah, FL). Consequently,three phenotypically distinct fractions of DCs were found: CD1a1/CD11c1/lin2/DR1 (fraction 1), CD1a2/CD11c1/lin2/DR1 (fraction 2),and CD1a2/CD11c2/lin2/DR1 cells (fraction 3).

Purification and characterization of DC fractions

The DC fractions were obtained also by a two-color FACStar cell sorter(Becton Dickinson), as follows. 1) Purification of CD1a1/CD11c1 DCs(fraction 1): after staining the DC-enriched population with PE (or FITC)-labeled mAbs against lineage markers plus FITC (or PE)-labeled anti-CD1a mAb, CD1a1/CD11c1 DCs were sorted as CD1a1/lin2 cells (notethat all CD1a1/lin2 DCs are quite comparable with CD1a1/CD11c1/lin2

DCs when analyzed after staining with anti-CD11c mAb). 2) Purificationof CD1a2/CD11c1 (fraction 2) and CD1a2/CD11c2 (fraction 3) DCs: theDC-enriched population was stained with FITC-labeled mAbs against lin-eage markers and CD1a plus PE-labeled anti-CD11c (LeuM5), and thelin2/CD1a2/CD11c1 and lin2/CD1a2/CD11c2 fractions were then col-lected using a FACStar as fractions 2 and 3, respectively. These fractionscollected using the FACStar were phenotyped. Purity of the sorted cellswas always greater than 96% by reanalysis using a FACScan (BectonDickinson). The sorted fractions, thus prepared, were then stained withPerCP-labeled HLA-DR. The cells bearing HLA-DR (DR1) were gated asDCs, and further analysis was conducted using a FACScan after stainingwith a panel of mAbs (listed in Table I) conjugated with FITC or PE.Before staining, the cells were incubated with an excess amount of unla-beled polyclonal human or mouse Ig to block nonspecific binding oflabeled mAbs.

Culture of DCs

The sorted DCs were cultured in 96-well flat-bottom tissue culture plates at1 3 105 cells/well in medium supplemented with monocyte-conditionedmedium (final concentration 50% v/v) for 1–6 days. In the culture ofCD1a2/CD11c2 DCs (fraction 3), medium supplemented with IL-3 wasused. In some experiments, a mixture of cytokines such as GM-CSF1TGF-b1, GM-CSF1 IL-4, GM-CSF 1 TNF-a, GM-CSF 1 TNF-a 1TGF-b1, or GM-CSF1 IL-4 1 TGF-b1 was used. Half of the mediumwas removed and replenished every 2 days.

Analysis of endocytosis

To assess endocytosis of DC fractions, FITC-dextran (Polysciences, War-rington, PA) was used according to the method described previously (33).Briefly, the cells were incubated with 0.1 mg/ml FITC-dextran at 37°C for15 or 45 min. The results were displayed as mean fluorescence intensityafter subtracting the background in which cells were incubated with FITC-dextran at 4°C.

Cytologic assays

Cells were cytocentrifuged onto a slide and stained with May-Giemsa so-lution, or by mAb to CD68 or Lag, then visualized using the ABC methodusing DAKO LSAB (labeled streptavidin biotin) kit plus hematoxylin anddiaminobenzidine.

For electron-transmission microscopy, cells were fixed with 2.5% glu-taraldehyde in 0.1 M cacodylate buffer (pH 7.4) and postfixed with 1%OsO4. After dehydration with graded ethanol, they were embedded in

Epon. Ultrathin sections were stained with lead citrate and uranyl acetate,and studied using a Hitachi H-600a electron microscope (Hitachi, Ibaragi,Japan).

Mixed leukocyte reaction

Freshly prepared blood DCs, cultured DCs (in the presence of GM-CSFand IL-3), or blood monocytes (purified by magnetic beads as CD141

cells) wereg irradiated at 15 Gy, and graded doses were then added to 23105 allogeneic T cells (collected by magnetic beads as CD31 cells) in96-well flat-bottom culture plates for 6 days. The cells were pulsed with 1mCi of [3H]TdR during the last 16 h of the culture period.

Cell cycle and viability assays

For cell cycle analyses, 105 cells were suspended in 70% ethanol at 4°C for1 h, followed by resuspension in 500ml PBS, 250ml RNase (1 mg/ml;Sigma, St. Louis, MO), and 250ml propidium iodide (PI) (100mg/ml;Molecular Probes, Eugene, OR). Cell viability was determined using An-nexin V-FITC Apoptosis Detection Kit (Genzyme, Cambridge, MA). Thestained cells were analyzed with a FACScan.

ResultsIdentification of three fractions of DCs in peripheral blood

DC-enriched populations prepared from the peripheral blood(CD41/CD32/CD142 cells) were four-color analyzed after stain-ing with PE-labeled anti-CD11c, FITC-labeled anti-CD1a, PerCP-labeled HLA-DR, and a mixture of biotinylated mAbs against lin-eage markers, followed by RED613-streptavidin. lin2/HLA-DR1

cells (DCs) (Fig. 1A) were of medium cell size (between lympho-cytes and monocytes) by light scatter profiles. Two distinct pop-ulations of lin2/DR1 were observed with respect to the expressionof CD11c (Fig. 1B). Furthermore, CD1a (detected by mAb cloneBB-5)-positive cells were newly observed as a blood DC fraction(Fig. 1B). Therefore, three fractions of DCs were identified withthe phenotypes of CD1a1/CD11c1/lin2/DR1 (fraction 1),CD1a2/CD11c1/lin2/DR1 (fraction 2), and CD1a2/CD11c2/lin2/DR1 cells (fraction 3). CD1a has been detected only on epi-dermal LCs (8, 34) and dermal DCs (10) in vivo (or on DCs de-rived from CD341 progenitors (15, 16) or monocyte-derived DCs(23) in the presence of cytokines in vitro), but not on freshly iso-lated blood DCs (27, 28). CD1a1 DCs were also observed when adifferent anti-CD1a mAb (BL-6) was used (data not shown). Theoverall intensity of CD1a expression, however, was lower than thaton CD341 progenitor-derived DCs (data not shown) when deter-mined by either BB-5 or BL-6 Ab.

The percentage of CD1a1/CD11c1 DCs was similar to that ofCD1a2/CD11c2 DCs, and that of CD1a2/CD11c1 DCs was 1/3 to1/10 that of CD1a1/CD11c1 DCs. The frequency of DC fractions,CD1a1/CD11c1 DCs, CD1a2/CD11c1 DCs, and CD1a2/CD11c2 DCs, ranged from 0.56 to 0.18%, 0.14 to 0.03%, and 0.50to 0.13% of PBMCs, respectively, when more than 30 healthyvolunteers were examined. These three fractions of blood DCswere usually purified by a FACStar, as described inMaterials andMethods. Region 1 (R1) in Fig. 1C shows the sorting gate of frac-tion 1 as lin2/CD1a1 cells (note that all CD1a1 cells are CD11c1,as shown in Fig. 1B), and R2 and R3 in Fig. 1D represent thesorting gates of lin2/CD1a2/CD11c1 cells (fraction 2) and lin2/CD1a2/CD11c2 cells (fraction 3), respectively.

FIGURE 3. Expression of surface molecules on freshly isolated fractions of blood DCs. Freshly prepared blood DC fractions (purified by a FACStar)were stained with anti-HLA-DR mAb and the panel of mAbs. The DR1 cells were gated and analyzed by a FACScan. Expression of surface moleculesis shown as shaded histograms. Open histograms in the figures represent negative controls stained with isotype-matched control Abs. Results shown arerepresentative of at least three experiments.

1413The Journal of Immunology

Page 6: 1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct ...imed1.kmu-ac.jp/pdf/thesis_dissertation/5.pdf · A CD1a1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct Precursor

Morphology of peripheral blood DC fractions

Freshly prepared or cultured (for 3 days in monocyte-conditionedmedium or IL-3-supplemented medium) DC fractions were exam-ined by light and electron microscopes (EM). Freshly sortedCD1a1/CD11c1 DCs (fraction 1) (Fig. 2,A andB) and CD1a2/CD11c1 DCs (fraction 2) (Fig. 2,D and E) were found to bemonocyte like, characterized by short cell processes, indented nu-cleus, high nuclear-cytoplasmic ratio, and few cytoplasmic dense

bodies and vesicles, in contrast to typical DCs. After 3-day culture,both fractions showed typical DC features such as long cell pro-cesses, large cell size (Fig. 2,C andF), and a prominent tubular-vesicular system (data not shown).

In contrast to fractions 1 and 2, freshly isolated CD1a2/CD11c2

DCs (fraction 3) displayed lymphoplasmacytoid morphology;that is, a medium size with round nucleus, cytoplasm-contain-ing juxtanuclear Golgi apparatus, and parallelarrays of rough

FIGURE 4. Expression of surface molecules on cultured blood DC fractions. Cultured blood DC fractions were stained with anti-HLA-DR mAb anda panel of mAbs. The DR1 cells were analyzed by a FACScan. The three fractions of DCs were cultured for 3 days with monocyte-conditioned mediumfor fractions 1 and 2, and IL-3-supplemented medium for fraction 3. Results shown are representative of at least three experiments.

1414 LC PRECURSORS IN HUMAN PERIPHERAL BLOOD

Page 7: 1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct ...imed1.kmu-ac.jp/pdf/thesis_dissertation/5.pdf · A CD1a1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct Precursor

endoplasmic reticulum (Fig. 2,G andH). These cells correspondto plasmacytoid T cells within the T cell zone of lymphoid organs,as recently characterized by Grouard et al. (14). After culture withIL-3, they showed a typical DC morphology (Fig. 2I) with markeddecreases in rough endoplasmic reticulum (data not shown), andthis process was dependent on the presence of IL-3. No Birbeckgranules were found in either of the three presently identified frac-tions of DCs before or after culture with monocyte-conditionedmedium or IL-3 (data not shown).

Phenotype of peripheral blood DC fractions

The fluorometrical characterization of the three fractions of DCswas next conducted. The surface phenotype of the three DC frac-tions before and after culture is highlighted in Figs. 3 and 4. Thecommon features of these three fractions are as follows: none ofthe DC fractions expressed lineage markers for T cells (CD8, TCR,Thy-1/CD90), B cells (CD10, CD20), NK cells (CD56), mono-cytes (CD14), granulocytes (CD15, CD35), or erythrocytes (gly-cophorin A) before or after culture. In addition, they possessedneither CD27 and CD30 (TNF receptor families) nor CD70 (ligandfor CD27). IL-1R type I (CD121a), G-CSFR (CD114), IL-2Rb-chain (CD122), and IL-6Rb (CD130) were negative on all freshlyprepared DC fractions (data not shown). All fractions clearly ex-pressed MHC class II, CD4, CD9, CD36, CD38, CD45, and ad-hesion molecules (CD11a, CD15s, CD18, CD29, CD44, CD49d,CD50, and CD54). Costimulatory molecules (CD80, CD86, andCD40) were dimly expressed on all fractions (fraction 3 little ex-pressed CD40), and were up-regulated after culture. Activationmolecules CD25 and the DC-associated marker CD83 were alsodetected on all fractions after overnight culture, but not on freshlyisolated cells. In addition, CLA (a skin-homing molecule (35),which is constitutively expressed on LCs (36)), and L-selectin(CD62L, known to be a lymph node-homing molecule (37)) werepresent on all fresh DC fractions. The characteristic features ofeach fraction are as follows.

Fraction 1. CD1a1/CD11c1 DCs expressed CD33 and CD13(myeloid-associated markers), CD2high, CD1c, CD49e, CD95(Fas), CD45RO, CD32/CD64 (Fcg receptors), and cytokine recep-tors (GM-CSFR (CD116) and IL-3Ra (moderately positive)). Fur-thermore, they dimly expressed CD5 and CD11b. After culture,CD11b was up-regulated, and CD32/64 was down-regulated.CD62L became undetectable during culture in contrast to freshlyprepared cells. On the basis of the expressions of CD2, CD9,CD11b, CD11c, CD13, CD32, CD33, CD64, and GM-CSFR, frac-tion 1 is thought to be closely associated with the monocyte lin-eage. Actually, the phenotype of fraction 1 was quite similar to thatof blood monocyte-derived DCs and that of CD341 progenitor-derived DCs (but not LC type) (16).Fraction 3. In contrast to fraction 1, CD1a2/CD11c2 DCs ex-pressed dimly or little CD33, CD13, CD1c, CD2, CD49e,CD45RO, CD32, CD64, but were significantly positive forCD45RA. In comparison with the other two fractions, they werehighly positive for CD4, but moderately positive for HLA-DR/DQ.They also possessed IL-3Ra (brightly positive), but not GM-CSFR. Fraction 3 brightly expressed intracytoplasmic CD68, butthe other two fractions were only weakly positive (data notshown). Therefore, fraction 3 is closely related to previously re-ported plasmacytoid T cells, not only morphologically but alsoaccording to phenotypical features. Furthermore, freshly preparedfraction 3 had no CD95. However, CD95 was induced during3-day culture with IL-3, whereas the expression of CD62L re-mained unchanged. When compared with culture in medium alone,this fraction exhibited increased levels of CD83 and HLA-DR/DQin the presence of IL-3 (data not shown).Fraction 2. CD1a2/CD11c1 DCs were a minor population, anduniformly expressed CD11c and CD33, like fraction 1. However,they were found to be heterogeneous in the expression of somesurface molecules such as CD2, CD13, CD45RA, CD45RO, IL-3Ra, and GM-CSFR, although they were morphologically homo-geneous. Fraction 2, in contrast to fraction 1, had no CD1a, CD1c,CD11b, or CD64. However, they had CD11c, CD33, CD13, andGM-CSFR, indicating that they were monocyte-lineage cells.Based on the expression of the CD2 molecule, fraction 2 could be

FIGURE 5. Stimulatory activity of blood DC fractions. Graded doses ofstimulator cells (freshly sorted DC fractions or cultured DC fractions in thepresence of GM-CSF and IL-3 for 3 days) were irradiated and coculturedwith 2 3 105 allogeneic CD31 T cells for 6 days. Cells were then pulsedwith 1 mCi of [3H]TdR during the last 16 h of culture. CD141 monocytes(purified using magnetic beads) were also prepared and used as stimulators.Dashed lines represent freshly prepared DCs and straight lines show cul-tured DCs. Fraction 1 (F), fraction 2 (f), fraction 3 (E), and monocytes(Œ). Vertical bars represent SDs of triplicate cultures. Representative dataof three experiments are shown.

FIGURE 6. Analysis of endocytosis. Fractions of DCs were incubatedwith 0.1 mg/ml FITC-dextran at 37°C for 15 or 45 min. After washingthree times, uptake of FITC-dextran was analyzed by a FACScan. Theresults are shown as mean fluorescence intensity, from which the back-ground fluorescence (incubated at 4°C) is subtracted. CD341 progenitor-derived DCs were obtained by the culture of CD341 progenitor cells col-lected from the human umbilical cord blood samples (according toinstitutional guidelines) for 12 days in the presence of GM-CSF and TNF-a(16, 17). Fraction 1 (F), fraction 2 (f), fraction 3 (E), monocytes (Œ), andCD341 progenitor-derived DCs (M). Results shown are representative ofthree experiments.

1415The Journal of Immunology

Page 8: 1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct ...imed1.kmu-ac.jp/pdf/thesis_dissertation/5.pdf · A CD1a1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct Precursor

divided into two minor subfractions: CD2high and CD2low DCs.After purifying these two populations and staining them with apanel of mAbs, we found that the former were CD13low/CD45RA1/CD45RO2/GM-CSFR1 (bright)/IL-3Ra1 (bright). Onthe other hand, CD2low DCs were CD13high/CD45RA1 (dim)/CD45RO1/GM-CSFR1 (dim)/IL-3Ra1 (dim). Therefore, bothsubpopulations in fraction 2 seem to be related (but not completelymatched) to fractions 1 and 3 on the basis of the expression ofsurface molecules. The physiological features of the heterogeneousfraction are unknown at present.

Finally, CD1a molecules were not induced on the CD1a2 cellseven when both fraction 2 and fraction 3 were cultured with mono-cyte-conditioned medium or IL-3-supplemented medium, respec-tively (Fig. 4), or with any combinations of cytokines examined(GM-CSF, IL-3, TNF-a, and TGF-b1) (data not shown).

Functional characterization of peripheral blood DC fractions

The stimulatory activity for allogeneic T cells was comparedamong the three DC fractions, as shown in Fig. 5. The order in thestimulatory activity of freshly prepared fractions was as follows:fraction 15 fraction 2. fraction 3, being dependent on the orderof the expression of MHC class II molecules. The stimulatory ac-tivities became stronger after culture for 3 days with GM-CSF plusIL-3 (note that fractions 1 and 2 are dependent on GM-CSF, andfraction 3 is dependent on IL-3), indicating that they becomemature DCs.

The capacity of the DC fractions for endocytosis was examinedby uptake of FITC-dextran. As shown in Fig. 6, freshly isolatedfractions 1 and 2 had a potent ability to endocytose FITC-dextranduring a 45-min incubation compared with CD341 progenitor-derived mature DCs, although this capacity was less than

FIGURE 7. Differentiation of fraction 1 DCs to LCs in the presence of GM-CSF, IL-4, and TGF-b1. A, Induction of LC-specific molecules on fraction1 DCs. DCs (fraction 1) and CD141 monocytes were cultured in the presence of GM-CSF, IL-4, and TGF-b1 for 6 days, and cells harvested daily andstained with anti-E-cadherin, anti-Langerin (DCGM-4), or anti-CD1a (BB-5) and anti-HLA-DR mAbs. The DR1 cells were gated and analyzed by aFACScan. The expression of E-cadherin, Langerin, or CD1a is shown as shaded histograms. Note that E-cadherin and Langerin were induced on fraction1 DCs already after 1 day of culture. Open histograms represent negative controls stained with isotype-matched control Abs.B, Histocytochemicalexamination of Lag in fraction 1 DCs cultured in GM-CSF, IL-4, and TGF-b1. Cultures were cytocentrifuged and stained with mAb against Lag. Originalmagnification:3250. Results shown are representative of three experiments.

1416 LC PRECURSORS IN HUMAN PERIPHERAL BLOOD

Page 9: 1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct ...imed1.kmu-ac.jp/pdf/thesis_dissertation/5.pdf · A CD1a1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct Precursor

monocytes. The endocytic activity of fractions 1 and 2 decreasedafter culture for 3 days with monocyte-conditioned medium (datanot shown). Fraction 3, which had been freshly isolated or cultured(with IL-3), showed little FITC-dextran uptake.

CD1a1/CD11c1 peripheral blood DCs rapidly differentiateinto LCs

It should be noted that no Birbeck granules were found in CD1a1/CD11c1 DCs (fraction 1) before or after culture with monocyte-conditioned medium. To further characterize fraction 1, we com-pared the effects of combinations of cytokines (GM-CSF1 TGF-b1, GM-CSF1 IL-4, GM-CSF 1 TNF-a, GM-CSF 1 IL-4 1TGF-b1, and GM-CSF1 TNF-a 1 TGF-b1) on their maturationand differentiation. The expression of E-cadherin, the LC-re-stricted Langerin (detected by mAb DCGM-4), and Lag Ag waskinetically examined. In the presence of GM-CSF1 IL-4 1 TGF-b1, E-cadherin and Langerin were detected (Fig. 7A) already after1 day of culture, and intracellular Lag was also slightly positiveboth in FACS (data not shown) and cytochemical (Fig. 7B) exam-inations. Expression of the LC-specific molecules increased at 3days, as shown in Fig. 7. Finally, fraction 1 showed DC morphol-ogy, and typical Birbeck granules were observed 6 days after cul-ture (Fig. 8). In addition, the intensity of CD1a expression in-creased concomitantly (Fig. 7A).

It should be noted that, regardless of the time point examined,differentiation to LCs from fraction 1 was only observed with thecombination of GM-CSF1 IL-4 1 TGF-b1, but not with anycytokine alone nor any combinations tested. Of importance, nei-ther fraction 2 nor 3 had the capacity to differentiate into LCs when

cultured with GM-CSF1 IL-4 1 TGF-b1 or other cytokinestested (GM-CSF, IL-3, TNF-a, and TGF-b1) (data not shown).Furthermore, induction of LC-specific molecules was not observedin CD141 monocytes during culture with the same cytokine com-bination (GM-CSF1 IL-4 1 TGF-b1). On day 6, E-cadherin andCD1a became positive, as has been reported by Geissmann et al.(24); however, the more sensitive Lag and Langerin markers werenot detected in our culture condition (Fig. 7). Furthermore, whenwe examined the frequency of dividing cells (evaluated by PIstaining) and viable cells (evaluated by PI and annexin V staining)before and after the culture, the percentages of dividing cells wereless than 1% (before and after 1-, 3-, and 6-day culture), and thepercentages of viable cells were more than 90% throughout theculture period (data not shown). In addition, 15 Gy irradiation didnot alter LC differentiation from fraction 1 (data not shown). Thesefindings clearly demonstrate that 1) fraction 1 has the capacity todifferentiate into LCs without cell proliferation, and 2) fraction 1 isthe direct/immediate precursor of LCs, compared with monocytes.

As shown in Fig. 9, 3 days after culture in the presence ofGM-CSF, IL-4, and TGF-b1, fraction 1 DCs expressed higher in-tensities of cutaneous lymphocyte-associated Ag and CD1a, withlower HLA-DR, CD54, CD40/CD80/CD86, and CD83 (mostlynegative, a few positive) than those cultured with monocyte-con-ditioned medium. Activation-associated markers, CD25 andCD71, were not induced, and CD2 was down-regulated in contrastto the cells before culture. Furthermore, CD23 (FceRII) was in-duced on fraction 1 under this condition (but not with monocyte-conditioned medium), as reported by Schmitt et al. (38) on LCs.These findings suggest that the combination of IL-4, GM-CSF, and

FIGURE 8. Morphology of fraction 1 DCs culturedfor 6 days in the presence of GM-CSF, IL-4, and TGF-b1. Typical Birbeck granules (arrow andinset) are ob-served (;20–25% of cultured DCs) by EM examina-tions. Original magnification:left, 36,000; right,312,000;inset, 320,000.

FIGURE 9. Phenotype of fraction 1DCs after culture with GM-CSF, IL-4,and TGF-b1. Fraction 1 DCs were cul-tured with GM-CSF, IL-4, and TGF-b1for 3 days, and the cells were harvestedand stained with the mAbs listed in thefigure. The DR1 cells were gated and an-alyzed by a FACScan. Shaded histogramsrepresent expression of the surface mol-ecules, and open histograms representisotype controls. Representative results ofthree experiments are shown.

1417The Journal of Immunology

Page 10: 1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct ...imed1.kmu-ac.jp/pdf/thesis_dissertation/5.pdf · A CD1a1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct Precursor

TGF-b1 is essential for differentiation of fraction 1 into LCs ratherthan terminal maturation and/or activation.

Our results represent the first observation showing that LCs canoriginate from DCs circulating in peripheral blood, as their directprecursors.

DiscussionWe have identified three fractions of DCs in the peripheral bloodusing a panel of mAb. Two of these fractions, fraction 1 (CD1a1/CD11c1 DCs) and fraction 2 (CD1a2/CD11c1 DCs), showedmore potent APC activity and monocyte-like morphology. Frac-tion 3 (CD1a2/CD11c2 DCs) displayed poor endocytic activityand a lymphoplasmacytoid morphology. All fractions were con-firmed to exhibit dendritic morphology and acquired CD83 duringculture.

As previously reported, less mature APCs in the blood, withlittle expression of CD11c and CD33, have been speculated to beon the way from the bone marrow to nonlymphoid tissues such asthe skin, and more mature potent APCs bearing CD11c and CD33are thought to be en route to the lymph nodes from the skin (27).When freshly isolated, these DCs display immature dendritic mor-phology and express CD4 (5, 27), but not CD83 (26, 39), in agree-ment with our results (Figs. 2 and 3). Notably, DCs in the bloodhave been found within a CD1a-negative fraction (26–28, 39).However, we have identified one fraction of DCs as CD1a low-positive cells (fraction 1). The difference in detection of CD1abetween our present results and previous studies could be due tothe following: 1) in previous studies, T cells were depleted bySRBC-rosette formation, and this caused the depletion of someDCs bearing CD1a because this DC population coexpresses CD2(binding site of SRBC) (5, 26, 27); 2) part of the CD1a1 DCfraction coexpressing CD11b might be depleted along with my-elomonocyte-lineage cells when anti-CD11b mAb was used in im-munoselection (29, 39, 40); 3) the level of CD1a expression on thisfraction is much lower than that on the CD341 progenitor-derivedDCs or monocyte-derived DCs; therefore, CD1alow1 DCs wereclassified as CD1a-negative cells; in this context, Brown et al.reported that CD1a was weakly expressed on a small amount ofblood DCs (41); and 4) different anti-CD1a mAbs employed indetection, this being considered to be a main reason. In our study,a difference in the intensity of CD1a was actually observed whentwo anti-CD1a mAbs (clones BB-5 and BL-6) were compared. TheCD11c1/CD331 DC subset in the blood that O’Doherty et al. (27)and Thomas et al. (28) previously identified might contain fraction2 (CD1a2/CD11c1 DCs) and a part of fraction 1 (CD1a1/CD11c1

DCs). In their studies, CD1alow1 DCs might be classified as CD1a-negative cells.

Fraction 1 is morphologically similar to monocytes and, afterculture, the cells resemble monocyte-derived DCs. Furthermore,fraction 1 has myelomonocyte-associated Ags, and has the capac-ity to uptake FITC-dextran. These findings indicate that fraction 1is a homogeneous subset, and most likely a CD14-negative mono-cyte-associated or monocyte-derived DC. Strikingly, cells in frac-tion 1, but not fraction 2 or 3, rapidly became cells with typical LCcharacteristics in the presence of GM-CSF, IL-4, and TGF-b1.E-cadherin (Fig. 7A) and Lag (Fig. 7B) were detected after short-term culture, and the newly described LC-specific Ag, Langerin(31), was concomitantly induced (Fig. 7A). Furthermore, the dif-ferentiation of LCs from fraction 1 was confirmed by the appear-ance of Birbeck granules (Fig. 8). DCs bearing CD1a are inducedfrom CD141 blood monocytes after culture with GM-CSF andIL-4 (23, 33). Geissmann et al. (24) have recently reported thatblood monocytes differentiate into LCs after a 6-day culture in the

additional presence of TGF-b1. We did not observe acquisition ofa complete LC phenotype from CD141 monocytes in our culturecondition (Fig. 7). Furthermore, the differentiation of LCs fromfraction 1 occurred during a very short-term culture. In addition,the possibility that a dividing or a distinct subpopulation in fraction1 selectively differentiates into LCs was ruled out by the following:there was,1% of dividing cells, there was a low cell mortalityrate, and a low rate of apoptosis throughout the short-term culture.Therefore, fraction 1 is a distinct direct/immediate precursor ofLCs in the peripheral blood, and might be an intermediate stagebetween monocytes and LCs.

LCs are myeloid-lineage DCs that originate from the bone mar-row. However, little is known about their migration/differentiationpathway. Our results might help elucidate this question, as fraction1 is the direct precursor of LCs (in response to GM-CSF1 IL-4 1TGF-b1) and a bipotent cell that can be induced to differentiateinto non-LCs such as dermal DCs (in response to monocyte-con-ditioned medium). This indicates that common precursors for theLC lineage and the monocyte-associated DC lineage are present inthe peripheral blood. It can be speculated that part of this fractionmigrates to the epidermis to differentiate into LCs, and that theother migrates to nonlymphoid tissues such as the dermis and dif-ferentiate into dermal DCs. This process might be dependent uponcytokines produced by regional microenvironments.

Fraction 3 (CD1a2/CD11c2 DCs) is another homogeneous sub-set in the peripheral blood, and distinct from fraction 1, based onits morphology, the expression of lineage-associated Ags, lack ofability to uptake FITC-dextran, and dependency on IL-3. From ourdetailed studies on blood DCs, the features of fraction 3 corre-spond completely to those of plasmacytoid T cells in the T zone ofthe lymphoid organ (found to be close to or within the high en-dothelial venule) that could be derived from CD41/CD11c2 bloodDCs (14). Fraction 3 possesses high levels of the lymph node-homing molecule; CD62L, and its expression, remains unchangedduring culture, although it is down-regulated on fraction 1 (Figs. 3and 4). These findings support the possibility that fraction 3 in theblood may be a precursor pool for plasmacytoid T cells that di-rectly migrate to the T zone in the lymphoid tissues via the highendothelial venule. Furthermore, fraction 3 requires IL-3, but notGM-CSF, for cell maturation and development, whereas typicalmyeloid-lineage DCs are known to be dependent on GM-CSF,indicating that fraction 3 is not in the monocyte-associated DClineage. Grouard et al. have pointed out that plasmacytoid T cellsin the lymphoid organs are lymphoid-derived DCs for this reason(14). However, fraction 3 also expresses several myelo-monocyticmarkers (CD9; Fig. 3, CD36 and CD68; data not shown); there-fore, it is not evident at the moment whether they indeed are lym-phoid-derived DCs. This issue will require further studies on thesecells. Our results clearly indicate that fraction 3 is a different lin-eage from that previously examined in in vitro differentiation stud-ies of CD341 progenitors to LCs or dermal DCs.

Although examination of the minor fraction, fraction 2 (CD1a2/CD11c1 DCs), has been hampered by the limitation of cells col-lected from the peripheral blood, this fraction has similar featuresto fraction 1 in their morphology, allostimulatory activity, and theexpression of several myelo-monocytic Ags, suggesting that cellsin this fraction are monocyte-associated or monocyte-derived DCs,or that they are in an intermediate stage preceding fraction 1. Fur-thermore, fraction 2 is phenotypically similar to recently reportedDCs located in the germinal center (GCDC) (13); therefore, thepossibility exists that fraction 2 becomes GCDCs, which have beenpresumed to be derived from blood CD41/CD11c1 DCs.

In summary, we have characterized three fractions of DCs in thehuman peripheral blood using a panel of mAbs. It is noted that

1418 LC PRECURSORS IN HUMAN PERIPHERAL BLOOD

Page 11: 1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct ...imed1.kmu-ac.jp/pdf/thesis_dissertation/5.pdf · A CD1a1/CD11c1 Subset of Human Blood Dendritic Cells Is a Direct Precursor

fraction 1 has the capacity to become LCs. These results suggestthat blood DC fractions (subsets) are different not only in theirmaturational stages, but also in their lineage or differentiation path-ways. Our studies of the DC fractions were conducted usinghealthy volunteers. Therefore, the findings in this study also offerbasic data for the diagnosis of some immune-related diseases. Weare now proceeding with experiments related to these points.

AcknowledgmentsWe thank Y. Tokuyama and M. Shinkawa for their skillful technical as-sistance, F. Ishida (Central Research Center, Kansai Medical University)for sorting cells on a FACStar, and K. Ando for manuscript preparation.We thank Dr. S. Imamura and Dr. K. Yoneda (Faculty of Medicine, KyotoUniversity) for anti-Lag 1 Ab.

References1. Steinman, R. M. 1991. The dendritic cell system and its role in immunogenicity.

Annu. Rev. Immunol. 9:271.2. Banchereau, J., and R. M. Steinman. 1998. Dendritic cells and the control of

immunity. Nature 392:245.3. Inaba, K., J. P. Metlay, M. T. Crowley, and R. M. Steinman. 1990. Dendritic cells

pulsed with protein antigen in vitro can prime antigen-specific, MHC-restricted Tcells in situ.J. Exp. Med. 172:631.

4. Bjorck, P., L. F.-Romo, and Y.-J. Liu. 1997. Human interdigitating dendritic cellsdirectly stimulate CD40-activated naive B cells.Eur. J. Immunol. 27:1266.

5. O’Doherty, U., R. M. Steinman, M. Peng, P. U. Cameron, S. Gezelter,I. Kopeloff, W. J. Swiggard, M. Pope, and N. Bhardwaj. 1993. Dendritic cellsfreshly isolated from human blood express CD4 and mature into typical immu-nostimulatory dendritic cells after culture in monocyte-conditioned medium.J. Exp. Med. 178:1067.

6. Thomas, R., L. S. Davis, and P. E. Lipsky. 1993. Isolation and characterizationof human peripheral blood dendritic cells.J. Immunol. 150:821.

7. Rowden, G., M. G. Lewis, and A. K. Sullivan. 1977. Ia antigen expression onhuman epidermal Langerhans cells.Nature 268:247.

8. Romani, N., A. Lenz, H. Stossel, U. Stanzl, O. Majdic, P. Fritsch, and G. Schuler.1989. Cultured human Langerhans cells resemble lymphoid dendritic cells inphenotype and function.J. Invest. Dermatol. 93:600.

9. Cerio, R., C. E. M. Griffiths, K. D. Cooper, B. J. Nickoloff, and J. T. Headington.1989. Characterization of factor XIIIa positive dermal dendritic cells in normaland inflamed skin.Br. J. Dermatol. 121:421.

10. Nestle, F. O., X.-G. Zheng, C. B. Thompson, L. A. Turka, and B. J. Nickoloff.1993. Characterization of dermal dendritic cells obtained from normal humanskin reveals phenotypic and functionally distinctive subsets.J. Immunol. 151:6535.

11. Ardavin, C., L. Wu, C.-L. Li, and K. Shortman. 1993. Thymic dendritic cells andT cells develop simultaneously in the thymus from a common precursor popu-lation. Nature 362:761.

12. Wu, L., L. Chung-Leung, and K. Shortman. 1996. Thymic dendritic cell precur-sors: relationship to the T lymphocyte lineage and phenotype of the dendritic cellprogeny.J. Exp. Med. 184:903.

13. Grouard, G., I. Durand, L. Filgueira, J. Banchereau, and Y.-J. Liu. 1996. Den-dritic cells capable of stimulating T cells in germinal centres.Nature 384:364.

14. Grouard, G., M.-C. Rissoan, L. Filgueira, I. Durand, J. Banchereau, and Y.-J. Liu.1997. The enigmatic plasmacytoid T cells develop into dendritic cells with in-terleukin (IL)-3 and CD40-ligand.J. Exp. Med. 185:1101.

15. Caux, C., C. Dezutter-Dambuyant, D. Schmitt, and J. Banchereau. 1992. GM-CSF and TNF-a cooperate in the generation of dendritic Langerhans cells.Nature360:258.

16. Caux, C., B. Vanbervliet, C. Massacrier, C. Dezutter-Dambuyant,B. de Saint-Vis, C. Jacquet, K. Yoneda, S. Imamura, D. Schmitt, andJ. Banchereau. 1996. CD341 hematopoietic progenitors from human cord blooddifferentiate along two independent dendritic cell pathways in response to GM-CSF1 TNFa. J. Exp. Med. 184:695.

17. Reid, C. D. L., A. Stackpoole, A. Meager, and J. Tikerpae. 1992. Interactions oftumor necrosis factor with granulocyte-macrophage colony-stimulating factorand other cytokines in the regulation of dendritic cell growth in vitro from earlybipotent CD341 progenitors in human bone marrow.J. Immunol. 149:2681.

18. Romani, N., S. Gruner, D. Brang, E. Kampgen, A. Lenz, B. Trockenbachter,G. Konwalinka, P. O. Fritsch, R. M. Steinman, and G. Schuler. 1994. Prolifer-ating dendritic cell progenitors in human blood.J. Exp. Med. 180:83.

19. Strunk, D., C. Egger, G. Leitner, D. Hanau, and G. Stingl. 1997. A skin homingmolecule defines the Langerhans cell progenitor in human peripheral blood.J. Exp. Med. 185:1131.

20. Strobl, H., E. Riedl, C. Scheinecker, C. Bello-Fernandez, W. F. Pickl,K. Rappersberger, O. Majdic, and W. Knapp. 1996. TGF-b1 promotes in vitrodevelopment of dendritic cells from CD341 hematopoietic progenitors.J. Im-munol. 157:1499.

21. Borkowski, T. A., J. J. Letterio, A. G. Farr, and M.C. Udey. 1996. A role forendogenous transforming growth factorb1 in Langerhans cell biology: the skinof transforming growth factorb1 null mice is devoid of epidermal Langerhanscells.J. Exp. Med. 184:2417.

22. Strobl, H., C. Bello-Fernandez, E. Riedl, W. F. Pickl, O. Majdic, S. D. Lyman,and W. Knapp. 1997. Flt3 ligand in cooperation with TGF-b1 potentiates in vitrodevelopment of Langerhans type dendritic cells in serum-free medium.Blood90:1425.

23. Zhou, L.-J., and T. F. Tedder. 1996. CD141 blood monocytes can differentiateinto functionally mature CD831 dendritic cells.Proc. Natl. Acad. Sci. USA 93:2588.

24. Geissmann, F., C. Prost, J.-P. Monnet, M. Dy, N. Brousse, and O. Hermine. 1998.Transforming growth factorb1, in the presence of granulocyte/macrophage col-ony-stimulating factor and interleukin 4, induces differentiation of human periph-eral blood monocytes into dendritic Langerhans cells.J. Exp. Med. 187:961.

25. Freudenthal, P. S., and R. M. Steinman. 1990. The distinct surface of humanblood dendritic cells, as observed after an improved isolation method.Proc. Natl.Acad. Sci. USA 87:7698.

26. Zhou, L.-J., and T. F. Tedder. 1995. Human blood dendritic cells selectivelyexpress CD83, a member of the immunoglobulin superfamily.J. Immunol. 154:3821.

27. O’Doherty, U., M. Peng, S. Gezelter, W. J. Swiggard, M. Betjes, N. Bhardwaj,and R. M. Steinman. 1994. Human blood contains two subsets of dendritic cells,one immunologically mature and the other immature.Immunology 82:487.

28. Thomas, R., and P. E. Lipsky. 1994. Human peripheral blood dendritic cell sub-sets: isolation and characterization of precursor and mature antigen-presentingcells.J. Immunol. 153:4016.

29. Fanger, N. A., D. Voigtlaender, C, Liu, S. Swink, K. Wardwell, J. Fisher,R. F. Graziano, L. C. Pfefferkorn, and P. M. Guyre. 1997. Characterization ofexpression, cytokine regulation, and effector function of the high affinity IgGreceptor FcgRI(CD64) expressed on human blood dendritic cells.J. Immunol.158:3090.

30. Reddy, A., M. Sapp, M. Feldman, M. Subklewe, and N. Bhardwaj. 1997. Amonocyte conditioned medium is more effective than defined cytokines in me-diating the terminal maturation of human dendritic cells.Blood 90:3640.

31. Valladeau, J., V. Duvert-Frances, C. Dezutter-Dambuyant, C. Vincent, J.-J. Pin,C. Massacrier, J. Vincent J. Davoust, and S. Saeland. 1998. A monoclonal anti-body against Langerin, a protein specific of Langerhans cells, is internalized incoated pits and Birbeck granules.J. Leukocyte Biol. Suppl. 2:25.

32. Kashihara, M., M. Ueda, Y. Horiguchi, F. Furukawa, M. Hanaoka, andS. Imamura. 1986. A monoclonal antibody specifically reactive to human Lang-erhans cells.J. Invest. Dermatol. 87:602.

33. Sallusto, F., and A. Lanzavecchia. 1994. Efficient presentation of soluble antigenby cultured human dendritic cells is maintained by granulocyte/macrophage col-ony-stimulating factor plus interleukin 4 and down-regulated by tumor necrosisfactor-a. J. Exp. Med. 179:1109.

34. Shibaki, A., L. Meunier, C. Ra, S. Shimada, A. Ohkawara, and K. D. Cooper.1995. Differential responsiveness of Langerhans cell subsets of varying pheno-typic states in normal human epidermis.J. Invest. Dermatol. 104:42.

35. Berg, E. L., T. Yoshino, L. S. Rott, M. K. Robinson, R. A. Warnock,T. K. Kishimoto, L. J. Picker, and E. C. Butcher. 1991. The cutaneous lympho-cyte antigen is skin lymphocyte homing receptor for the vascular lectin endothe-lial cell-leukocyte adhesion molecule 1.J. Exp. Med. 174:1461.

36. Koszik, F., D. Strunk, I. Simonitsch, L. J. Picker, G. Stingl, and E. Payer. 1994.Expression of monoclonal antibody HECA-452-defined E-selectin ligands onLangerhans cells in normal and diseased skin.J. Invest. Dermatol. 102:773.

37. Nakache, M., E. L. Berg, P. R. Streeter, and E. C. Butcher. 1989. The mucosalvascular addressin is a tissue-specific endothelial cell adhesion molecule for cir-culating lymphocytes.Nature 337:179.

38. Schmitt, D. A., T. Bieber, J.-P. Cazenave, and D. Hanau. 1990. Fc receptors ofhuman Langerhans cells.J. Invest. Dermatol. 94:15S.

39. Fearnley, D. B., A. D. McLellan, S. I. Mannering, B.D. Hock, and D. N. J. Hart.1997. Isolation of human blood dendritic cells using the CMRF-44 monoclonalantibody: implications for studies on antigen-presenting cell function and immu-notherapy.Blood 89:3708.

40. McLellan. A. D., R. V. Sorg, L. A. Williams, and D. N. J. Hart. 1996. Humandendritic cells activate T lymphocytes via a CD40:CD40 ligand-dependent path-way. Eur. J. Immunol. 26:1204.

41. Brown, K. A., P. Bedford, M. Macey, D. A. McCarthy, F. Leroy, A. J. Vora,A. J. Stagg, D. C. Dumonde, and S. C. Knight. 1997. Human blood dendriticcells: binding to vascular endothelium and expression of adhesion molecules.Clin. Exp. Immunol. 107:601.

1419The Journal of Immunology