ArticlePDF Available

Marginal zone macrophages express a murine homologue of DC-SIGN that captures blood-borne antigens in vivo

Authors:

Abstract and Figures

Antigen-presenting cells are localized in essentially every tissue, where they operate at the interface of innate and acquired immunity by capturing pathogens and presenting pathogen-derived peptides to T cells. C-type lectins are important pathogen recognition receptors and the C-type lectin, dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin (DC-SIGN), is unique in that, in addition to pathogen capture, it regulates adhesion processes such as DC trafficking and T-cell synapse formation. We have isolated a murine homologue of DC-SIGN that is identical to the previously reported murine homologue mSIGNR1. mSIGNR1 is more closely related to the human DC-SIGN homologue L-SIGN than to DC-SIGN itself because mSIGNR1 is specifically expressed by liver sinusoidal endothelial cells, similar to L-SIGN, and not by DCs. Moreover, mSIGNR1 is also expressed by medullary and subcapsular macrophages in lymph nodes and by marginal zone macrophages (MZMs) in the spleen. Strikingly, these MZMs are in direct contact with the bloodstream and efficiently capture specific polysaccharide antigens present on the surface of encapsulated bacteria. We have investigated the in vivo function of mSIGNR1 on MZMs in spleen. We demonstrate here that mSIGNR1 functions in vivo as a pathogen recognition receptor on MZMs that capture blood-borne antigens, which are rapidly internalized and targeted to lysosomes for processing. Moreover, the antigen capture is completely blocked in vivo by the blocking mSIGNR1-specific antibodies. Thus, mSIGNR1, a murine homologue of DC-SIGN, is important in the defense against pathogens and this study will facilitate further investigations into the in vivo function of DC-SIGN and its homologues.
No caption available
… 
No caption available
… 
Content may be subject to copyright.
doi:10.1182/blood-2002-04-1044
Prepublished online June 14, 2002;
van Kooyk
Gangaram-Panday, Gerard C van Duijnhoven, Georg Kraal, Antoon J M van Oosterhout and Yvette
Teunis B H Geijtenbeek, Peter C Groot, Martijn A Nolte, Sandra J van Vliet, Shanti T
that captures blood-born antigens in vivo
Marginal zone macrophages express a murine homologue of DC-SIGN
(973 articles)Phagocytes (5013 articles)Immunobiology Articles on similar topics can be found in the following Blood collections
http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#repub_requests
Information about reproducing this article in parts or in its entirety may be found online at:
http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#reprints
Information about ordering reprints may be found online at:
http://bloodjournal.hematologylibrary.org/site/subscriptions/index.xhtml
Information about subscriptions and ASH membership may be found online at:
articles must include the digital object identifier (DOIs) and date of initial publication.
priority; they are indexed by PubMed from initial publication. Citations to Advance online
prior to final publication). Advance online articles are citable and establish publication
yet appeared in the paper journal (edited, typeset versions may be posted when available
Advance online articles have been peer reviewed and accepted for publication but have not
Copyright 2011 by The American Society of Hematology; all rights reserved.
Washington DC 20036.
by the American Society of Hematology, 2021 L St, NW, Suite 900,
Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
1
Marginal zone macrophages express a murine homologue of DC-
SIGN that captures blood-born antigens in vivo
Running title: A murine homologue of DC-SIGN captures antigens in vivo
Teunis B.H. Geijtenbeek†§, Peter C. Groot¶‡, Martijn A. Nolte †‡, Sandra J. van Vliet, Shanti
T. Gangaram-Panday, Gerard C.F. van Duijnhoven*, Georg Kraal, Antoon J.M. van
Oosterhout and Yvette van Kooyk
Department of Molecular Cell Biology, Vrije Universiteit Medical Center Amsterdam, v.d.
Boechorststraat 7, 1081 BT Amsterdam, The Netherlands.
*Department of Tumor Immunology, University Medical Center St Radboud, Geert
Grooteplein Zuid 30, 6525 GA Nijmegen, The Netherlands.
Department of Pharmacology & Pathophysiology, Faculty of Pharmacy, Utrecht University,
Sorbonnelaan 16, 3584 CA Utrecht, The Netherlands
P.C. Groot and M.A. Nolte contributed equally to this paper.
§To whom correspondence should be addressed:
T.B.H. Geijtenbeek, Department of Molecular Cell Biology, Vrije Universiteit Medical Center
Amsterdam, v.d. Boechorststraat 7, 1081 BT Amsterdam, The Netherlands. Tel: +(31)20-
4448080; Fax: +(31)20-4448081; e-mail: T.Geijtenbeek.Cell@med.vu.nl
Word count. abstract: 244 words, text: 4701 words
Scientific heading: Immunobiology
Copyright 2002 American Society of Hematology
Blood First Edition Paper, prepublished online June 14, 2002; DOI 10.1182/blood-2002-04-1044
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
2
Abstract
Antigen presenting cells are localized in essentially every tissue, where they operate at the
interface of innate and acquired immunity by capturing pathogens and presenting pathogen-
derived peptides to T cells. C-type lectins are important pathogen recognition receptors and
the C-type lectin DC-SIGN is unique in that, in addition to pathogen capture, it regulates
adhesion processes such as DC trafficking and T cell synapse formation. We have isolated a
murine homologue of DC-SIGN that is identical to the previously reported murine homologue
mSIGNR1. mSIGNR1 is more closely related to the human DC-SIGN homologue L-SIGN
than to DC-SIGN self since mSIGNR1 is specifically expressed by liver sinusoidal endothelial
cells, similar as L-SIGN, and not by DC. Moreover, mSIGNR1 is also expressed by medullary
and subcapsular macrophages in lymph nodes, and by marginal zone macrophages (MZM) in
spleen. Strikingly, these MZM are in direct contact with the blood stream and efficiently
capture specific polysaccharide-antigens present on the surface of encapsulated bacteria. We
have investigated the in vivo function of mSIGNR1 on MZM in spleen. We demonstrate here
that mSIGNR1 functions in vivo as a pathogen recognition receptor on MZM that captures
blood-born antigens, which are rapidly internalized and targeted to lysosomes for processing.
Moreover, the antigen-capture is completely blocked in vivo by the blocking mSIGNR1-
specific antibodies. Thus, mSIGNR1, a murine homologue of DC-SIGN, is important in the
defense against pathogens and this study will facilitate further investigations into the in vivo
function of DC-SIGN and its homologues.
e-mail: T.Geijtenbeek.Cell@med.vu.nl
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
3
Introduction
Dendritic cells (DC) are professional antigen presenting cells (APC) that sample foreign
pathogens in the periphery and migrate to the lymphoid tissues where they present processed
antigens to naïve T cells, initiating an immune response1. DC express a repertoire of
specialized receptors to perform their unique functions. We have identified the human DC-
specific adhesion receptor DC-SIGN (CD209), which mediates several key-functions
throughout the lifetime of a DC; DC-SIGN binding to its endothelial ligand ICAM-2 mediates
transendothelial migration of DC2, and DC-SIGN enables primary immune responses by
initiating transient DC-T cell interactions through binding of ICAM-3 on naïve T cells3.
Recently, we demonstrated that DC-SIGN also functions as an antigen receptor by rapidly
internalizing soluble ligands that are targeted to late endosomes/lysosomes; here the
internalized antigen is efficiently processed and subsequently presented to CD4+ T cells4.
Besides playing an important role in the initiation of immune responses, DC-SIGN also
functions as a novel HIV-1 trans-receptor important in the dissemination of HIV-1 from sites
of infection5. Resident mucosal DC capture HIV-1 through the interaction of DC-SIGN with
the HIV-1 envelope glycoprotein gp120, and subsequently migrate to the lymphoid tissues
where DC-SIGN facilitates the transmission of HIV-1 to T cells5. The DC-specific expression
of human DC-SIGN (hDC-SIGN) in situ and in vitro correlates well with both its
immunological functions3 and its role in HIV-1 dissemination5.
The DC-SIGN homologue L-SIGN6, which has also been called DC-SIGN-related (DC-
SIGNR)7, has a similar activity to DC-SIGN, but is not expressed by DC6. Human L-SIGN
(hL-SIGN) is specifically expressed by liver sinusoidal endothelial cells (LSEC), a liver-
resident APC of monocyte origin and in lymph nodes6;8. LSEC interact strongly with
leukocytes and appear to constitute a central mechanism of peripheral immune tolerance in the
liver9. The tissue location and ligand binding properties of hL-SIGN strongly implicate a
physiological role for this receptor in antigen clearance6 as has been reported for other C-type
lectins10;11, as well as in LSEC-leukocyte adhesion.
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
4
Functional studies into the in vivo function of DC-SIGN have been hampered by the lack of
murine homologues. Therefore we set out to identify and isolate the murine homologue of
DC-SIGN. We have isolated a cDNA clone OtB7 which is homologous to hDC-SIGN and is
identical to murine SIGNR1 (mSIGNR1), a recently reported murine homologue of DC-
SIGN12. Here we describe the cell-specific expression and in vivo function of this murine
homologue of DC-SIGN. We generated antibodies against this homologue and demonstrated
that its cell-specific expression is similar to that of hL-SIGN. mSIGNR1 has a similar binding
activity in vitro to both hDC-SIGN and hL-SIGN. Its high affinity for murine ICAM-2, and its
localization at sites that are in close contact with blood, suggest a role for mSIGNR1 in
lymphocyte migration from the blood into tissues. Moreover, we demonstrate that marginal
zone macrophages (MZM) in spleen efficiently capture blood-born antigens in vivo through
mSIGNR1. mSIGNR1 rapidly internalizes blood-born antigens which are targeted to
lysosomes. These results demonstrate that a murine homologue of DC-SIGN is expressed by
highly phagocytic macrophages in spleen, and functions in vivo as an pathogen recognition
receptor.
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
5
Materials and Methods
Antibodies. The following antibodies were used: N148 (CD11c), anti-ICAM-2 (BD
Pharmingen, NJ), NLDC-40 (anti-DEC-205), M1/70 (CD11b), MECA-32, MOMA-1, ER-
TR913. Polyclonal antisera against mDC-SIGN were raised in rabbits against two mSIGNR1-
specific peptides, KTPNTERQKEQEKILQ and SRFQKYWNRGEPNNI (Eurogentec,
Seraing, Belgium).
Mice. BALB/c mice were bred and maintained in the laboratory animal facilities at the Faculty
of Medicine of the Vrije Universiteit Medical Center Amsterdam under conventional
conditions. Mice were used at ages between 8-15 weeks.
Characterization of the cDNA encoding the murine homologues of hDC-SIGN. The cDNA
encoding mSIGNR1 was amplified by RT-PCR on total RNA derived from lung. The PCR
primers used for isolation of mSIGNR1 were as follows: forward primer,
ATG.AGT.GAC.TCC.ACA.GAA.GCC.AAG.ATG.CAG; reverse primer , AAG.AAG.AAT.
CCC.AGA.GCC.TTT.TTCACG.ATC.C. The PCR product was sequenced and subsequently
cloned into the eukaryotic expression vector pRc/CMV. The homologue was expressed either
by transient transfections of 293T cells, or by stable transfections in the erythroleukemic cell
line K562. We have submitted the full mSIGNR1 cDNA sequence to GenBank under
accession number AF422108.
Generation of murine ICAM-2 and mSIGNR1 fusion proteins. The extracellular domain of
mICAM-2 was isolated by RT-PCR on total liver RNA using primers based on the murine
ICAM-2 cDNA sequence14. The primers were as follows: forward primer
CTA.AGC.TTC.CCC.ACC.TGA.GAT.GTC.TTC.TTT.TGC; reverse primer
CGG.ATC.CGC.CAC.GGG.ACG.TGC.CCG.AAA.G. The identity of the cDNA was
confirmed by sequencing, and the cDNA encoding mICAM-2 was cloned into the Sig-pIgG1-
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
6
Fc vector15. The resulting ICAM-2 Fc chimera contains the extracellular domain of ICAM-2
fused to the IgG1 Fc domain. ICAM-2-Fc was produced in COS cells by co-transfection of
ICAM-2-Sig-pIgG1-Fc and the pEE14 vector. ICAM-2-Fc concentrations in the supernatant
were determined by an anti-IgG1 ELISA.
mSIGNR1 Fc consists of the extracellular portion of mSIGNR1 (amino acid residues 75-225)
fused at the C-terminus to a human IgG1-Fc fragment into the Sig-pIgG1-Fc vector15.
mSIGNR1 Fc was produced in COS cells similarly as described for mICAM-2 Fc.
mSIGNR1-specific ELISA. mSIGNR1-specific antibody ER-TR9 (5 µg/ml) was coated in
ELISA plates for 18 hours at 4°C, followed by blocking with 1% BSA for 30 min. at 4°C.
Soluble mSIGNR1 Fc (1 µg) was added and incubated for 2 hours at 4°C. Unbound protein
was washed away and mSIGNR1 was detected by the mSIGNR1-specific antiserum 698
(1:200), 1 hour at 4°C, followed by peroxidase-conjugated goat-anti- rabbit IgG.
Fluorescent bead adhesion assay. Carboxylate-modified TransFluorSpheres (488/645 nm, 1.0
µm; Molecular Probes, Eugene, OR) were coated with ICAM-2-Fc, ICAM-3-Fc and HIV-1
gp120MN as described previously3;5. The fluorescent bead adhesion assay was performed as
described3. Briefly, 5x106 cells in adhesion buffer (20 mM Tris-HCl pH 8.0, 150 mM NaCl,
1mM CaCl2, 2 mM MgCl2, 0.5% BSA) were pre-incubated with 20µg/ml mAb for 10 minutes
at room temperature. Ligand-coated fluorescent beads (20 beads/cell) were added and the
suspension was incubated for 30 minutes at 37°C. Adhesion was determined using flow
cytometry (FACScan, Becton Dickinson, Oxnard, CA), by measuring the percentage of cells,
which had bound fluorescent beads.
Bone marrow-derived DC. DC were generated as described previously16, with modifications.
Briefly, femurs were dissected, placed in 70% alcohol for 1 min, and washed with PBS.
Marrow was flushed and passed through nylon mesh to remove debris. After washing,
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
7
lymphocytes, granulocytes, and I-A-positive cells were removed by immunomagnetic
depletion against the CD45R, CD4, CD8, and I-A-antigens. Remaining cells were cultured
overnight, and the non-adherent cells were seeded at 2 x 105 cells/ml and 4 ml/well in the
presence of 20 ng/ml rmGM-CSF and 20 ng/ml rmIL-4 in 6-well plates (Costar,
Badhoevedorp, the Netherlands). On day 4, the cultures were refreshed by adding 1 ml of
culture medium supplemented with GM-CSF and IL-4 (both at 10 ng/ml). At day 7, non-
adherent and loosely adherent proliferating DC aggregates were collected and replated in fresh
medium with cytokines(1 x 106 cells/ml).
CD11c+ DC isolation. Spleens were digested with collagenase IV for 40 minutes at 37°C,
followed by erythrocyte lysis. Splenic CD11c+ DC were isolated using MACS CD11c
microbeads (Miltenyi Biotec, Bergisch Gladbach, Germany) according to manufacturer’s
protocol.
Macrophage depletion in vivo. Systemic depletion of macrophages was performed by
treatment with clodronate-filled liposomes, as described previously17. Mice received a single
intravenous injection of 0.2 ml multilamellar liposomes in PBS, containing 2 mg liposome-
encapsulated clodronate (dichloromethylene biphosphonate, a kind gift from Roche
Diagnostics GmbH, Mannheim, Germany). Depletion of macrophages from the spleen was
assessed two days after liposome injection by immunofluorescence analysis.
Antigen capture. For studying the interaction of antigens with MZM, 200 µl FITC-dextran (1
mg/ml, 500 kDa dextran; Molecular Probes, Eugene, OR) was injected intravenously into the
tail veins of naïve mice. Where appropriate mice were treated 30 minutes before dextran
administration with either 200 µl mannan (1 mg/ml) or 200 µl ER-TR9 by intravenous
injection into the tail vein. Mice were sacrificed 40 minutes after dextran injection by
immediate CO2 asphyxiation. The spleen was rapidly removed and directly frozen in Tissue-
Tek (Sakura Finetek, Torrance, CA).
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
8
Immuno-fluorescence analysis. Tissues were collected and frozen in liquid nitrogen.
Cryosections (7 µm) of the tissues were fixed in 100% acetone (10 minutes), washed with
PBS and incubated with the first antibody (10 µg/ml) for 60 minutes at 37°C. The final
staining was performed with secondary antibodies labelled with either FITC or Texas Red for
30 minutes at room temperature.
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
9
Results
Isolation of a murine homologue of DC-SIGN
A 415 nucleotide long cDNA fragment, called OtB7, was isolated by differential gene
expression from a mouse model of allergic asthma, using lung-draining lymph node mRNA
obtained from control and ovalbumin-challenged mice, which display phenotypic
characteristics resembling human asthma18. The cDNA fragment was homologous to hDC-
SIGN, and to two mouse genomic sequences present in the high-throughput division of
GenBank (accession numbers AC073804.1 and AC073706.2). Using a combination of
BLAST searches and multiple alignment analyses we were able to construct a contiguous
19619 bp long genomic sequence composed of overlapping sequences present in both
Genbank entries (Figure 1a). A gene with homology to hDC-SIGN was predicted, and the
1101 bp cDNA encoding the predicted mDC-SIGN homologue (Genbank entry AF422108)
was isolated by RT-PCR from lymph node mRNA. The OtB7 cDNA encodes a type II
transmembrane C-type lectin consisting of 325 amino acid residues. The C-type lectin domain
of the murine homologue has a 74% similarity with that of hDC-SIGN, and the amino acid
residues essential for ligand- as well as Ca2+-binding are identical to those of hDC-SIGN19
(Figure 1b), suggesting a similar ligand specificity. The predicted transmembrane-spanning
region is 85% identical to that of both hDC-SIGN and hL-SIGN. Comparison of the
cytoplasmic region of OtB7 with that of the human homologues demonstrates that OtB7 lacks
the LL sequence motif which is important for internalization of hDC-SIGN4;20. The C-type
lectin domain of hDC-SIGN is separated from the cell-membrane by a stalk of 8 repeats which
form an -helix21, whereas several splice variants of hL-SIGN exist, differing in the number
of repeats from 3 to 96. The stalk of the murine DC-SIGN homologue OtB7 is considerably
shorter and lacks these repeats, however the region contains regularly-spaced hydrophobic
sequences that have been observed in other -helices. The predicted amino acid sequence of
OtB7 is as homologous to hL-SIGN as it is to hDC-SIGN.
Sequence alignment demonstrates that the putative protein encoded by clone OtB7 is identical
to murine SIGNR1 (Genbank AF373409; results not shown), one of five recently identified
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
10
murine homologues of hDC-SIGN12. We will hereafter refer to OtB7 as murine SIGNR1
(mSIGNR1).
mSIGNR1 binds both murine ICAM-2 and HIV-1 gp120
We raised polyclonal antibodies against two peptides present in the C-terminal region of
mSIGNR1 to investigate the expression of this putative DC-SIGN homologue in different
tissues. The human kidney 293T cell-line was transfected with mSIGNR1 cDNA and
mSIGNR1 was detected on the cell-surface by the mSIGNR1-specific polyclonal antibody 698
(Figure 2a). Thus, as predicted, mSIGNR1 is a type II transmembrane protein since the
antibodies, raised against the C-terminal region, stain intact 293T cells transfected with
mSIGNR1 (Figure 2a). The antibodies stained neither mock- nor hDC-SIGN-transfected cells
(results not shown). The monoclonal antibodies against hDC-SIGN (AZN-D1, -D2 and -D3)
and hL-SIGN (AZN-D2 and -D3) did not cross-react with the murine homologue mSIGNR1
(results not shown).
The C-type lectin domain of mSIGNR1 contains the amino acid residues which have been
shown to be essential in both ligand- and Ca2+-binding by hDC-SIGN19. Therefore we
investigated whether mSIGNR1 was functionally similar to both hDC-SIGN and hL-SIGN.
293T cells transfected with mSIGNR1 cDNA interacted with human ICAM-2, ICAM-3 and
HIV-1 gp120, but not with human ICAM-1, as is also observed for both hDC-SIGN- and hL-
SIGN-expressing cells (Figure 2b). Thus, as predicted by the high homology between C-type
lectin domains, mSIGNR1 has a similar ligand binding activity to both of these human
homologues.
Next we investigated whether mSIGNR1 is able to interact with murine ICAM-2, since mice
do not express a homologue of human ICAM-3. Indeed, mSIGNR1-expressing cells interacted
strongly with murine ICAM-2, in contrast to mock transfected cells (Figure 2c). The adhesion
was similar to that observed for both hDC-SIGN- and hL-SIGN-transfected cells (Figure 2c).
Moreover, the adhesion was inhibited by both the Ca2+-chelator EGTA and the
polycarbohydrate mannan, demonstrating that mSIGNR1 functions as a mannose-binding C-
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
11
type lectin (Figure 2c). The binding site on ICAM-2 for DC-SIGN and its homologues is
conserved between species since both hL-SIGN and hDC-SIGN interact with murine ICAM-2
(Figure 2c), and mSIGNR1 binds to human ICAM-2 (Figure 2b). These data demonstrate that
mSIGNR1 functions as an adhesion molecule with similar specificity to hDC-SIGN and hL-
SIGN.
The expression pattern of mSIGNR1 is similar to that of hL-SIGN
Next, we used our polyclonal antibodies to investigate whether DC express mSIGNR1, since
mRNA encoding mSIGNR1 has been detected by RT-PCR from bone marrow-derived DC22
and splenic CD11c+ DC12. Strikingly, no mSIGNR1 was detected on the cell-surface of either
bone marrow-derived DC or splenic CD11c+ DC (Figure 3a), demonstrating that mSIGNR1,
in contrast to hDC-SIGN, is not expressed by either in vitro- or in vivo-derived murine DC.
Immuno-fluorescence analysis demonstrated that mSIGNR1 is expressed by cells that line the
sinusoids in the liver (Figure 3b).
The mSIGNR1 staining was similar to that observed for hL-SIGN6 and demonstrates that
mSIGRN1 is expressed by LSEC. This is further confirmed by the absence of mSIGNR1
expression from Kupffer cells, which were stained with the marker F4/80 (Figure 3c).
Both hDC-SIGN and hL-SIGN are expressed in lymph nodes, and staining of murine lymph
node tissue sections demonstrated that mSIGNR1 is also highly expressed in lymph nodes
(Figure 3d-g), but not by lymph node-resident DC since neither CD11c nor DEC205 co-
localize with mSIGNR1 expression (Figure 3d and e). The mSIGNR1+ cells co-express the
antigens for SER-4 (Figure 3f) and MOMA-1 (Figure 3g), markers for both medullary and
subcapsular macrophages in lymph nodes23. Thus, mSIGNR1 is specifically expressed by
medullary and subcapsular macrophages in lymph nodes. These data demonstrate that the cell-
specific expression of mSIGNR1 is similar to that of hL-SIGN and this indicates mSIGNR1 is
more closely related to hL-SIGN than to hDC-SIGN.
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
12
mSIGNR1 is specifically expressed by marginal zone macrophages, and is recognized by the
MZM-specific antibody ER-TR9
We next investigated the expression of mSIGNR1 in the spleen, since mSIGNR1 mRNA has
been detected in spleen (results not shown and 12). We performed in situ immuno-fluorescence
analysis using the polyclonal antibodies 698 to identify those spleen cells expressing
mSIGNR1, since no mSIGNR1 was expressed on the cell-surface of splenic CD11c+ DC
(Figure 3a). mSIGNR1 is highly expressed by cells present in the marginal zone bordering the
white pulp (Figure 4a-d). These mSIGNR1+ cells are not DC, since they do not express the
DC markers CD11c and DEC-205 (Figure 4a and b). However, the mSIGNR1+ cells are
specifically stained with the marginal zone macrophage (MZM)-specific antibody ER-TR9
(Figure 4c). Thus, mSIGNR1 is specifically expressed by MZM in the spleen and these cells
are present in the marginal zone bordering the white pulp (Figure 4d). MZM are highly
phagocytotic cells whose dendrites that are in direct contact with the arterial blood stream
(reviewed in 24), and they can be effectively killed by a single intravenous injection of
liposomes containing dichloromethylene diphosphonate25. Indeed, all MZM were depleted
from the spleen after injection of the liposomes, since no staining of the MZM-marker ER-
TR9 is observed (Figure 4e). Moreover, no staining was observed with the polyclonal
antibodies against mSIGNR1, following MZM-depletion from the spleen, confirming that
mSIGNR1 is MZM-specific (Figure 4f).
The antigen of the MZM-specific antibody ER-TR9 has not been identified to date. The
similar expression patterns of the polyclonal antibodies 698 against mSIGNR1, and the MZM-
specific antibody ER-TR9 in spleen (Figure 4), liver and lymph node (results not shown), led
us to investigate whether mSIGNR1 is the antigen for this antibody. Indeed, 293T cells
transfected with mSIGNR1 were stained with the monoclonal antibody ER-TR9, whereas
mock transfected 293T cells are not (Figure 5a). Thus, the receptor recognized by ER-TR9 on
MZM is mSIGNR1. Moreover, this was further confirmed by the specific recognition of the
recombinant mSIGNR1 fusion protein by ER-TR9, in a mSIGNR1-specific sandwich ELISA
(Figure 5b). Comparison of the ER-TR9-specific expression pattern in spleen, lymph node and
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
13
liver with that of the polyclonal antibodies 689 against mSIGNR1 demonstrates that ER-TR9
specifically recognizes mSIGNR1 but not the other murine homologues of DC-SIGN (results
not shown and Figure 4).
mSIGNR1 captures blood-born antigens in vivo
The spleen plays an important role in the defense against pathogens and MZM are important
in this defense (reviewed in 24). MZM are in direct contact with the blood stream and are
highly phagocytotic cells. These cells have been demonstrated to bind specific polysaccharide
antigens present at the surface of encapsulated bacteria26;27. We examined the molecular basis
for this specificity by determining the mechanisms by which these antigens preferentially
targeted to MZM. Polysaccharide antigens rapidly localize to the MZM as demonstrated by
intravenous injection of naïve wild-type mice with the model polysaccharide antigen
fluorescein isothiocyanate (FITC)-dextran (Figure 5c and d). The FITC-dextran staining is
localized in the marginal zone bordering the white pulp (Figure 5c) and co-localizes with the
anti- mSIGNR1 polyclonal antibodies 698 (Figure 5d). Because mSIGNR1 contains a C-type
lectin domain similar to hDC-SIGN which has been shown to bind polycarbohydrates3;28, we
next determined whether mSIGNR1 was integral to this dextran capture by MZM. Naïve mice
were injected with purified ER-TR9 and after 40 minutes FITC-dextran was injected
intravenously. In these mice, the FITC-dextran was not retained in the spleen by MZM (Figure
5e) since no FITC-dextran was detected in the marginal zone. This demonstrates that
mSIGNR1 is responsible for the efficient capture of dextran by MZM. These results were
further confirmed by injecting naïve mice with the yeast-derived polysaccharide mannan prior
to FITC-dextran injection. Strikingly, MZM were then no longer able to capture FITC-dextran
(Figure 5f) because mannan is efficiently bound by mSIGNR1 and blocks its function (Figure
2b and c). Thus, the well-known function of MZM to filter blood-born antigens from blood is
mediated by mSIGNR1, and this mSIGNR1 function is inhibited by ER-TR9, and other
polycarbohydrates such as mannan.
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
14
MZM rapidly internalize antigens upon binding and we investigated whether mSIGNR1 also
mediates the internalization of ligands. K562 transfectants expressing mSIGNR1 have a high
affinity for FITC-dextran, whereas mock transfected cells do not (Figure 6a), confirming the
in vivo function of mSIGNR1. The dextran interaction with mSIGNR1 in vitro was
specifically blocked by the ER-TR9 antibody (Figure 6a). The fate of the bound dextran was
followed by immunofluorecence analysis. FITC-dextran was rapidly internalized and targeted
to the lysosomes (Figure 6b), demonstrating that mSIGNR1 is responsible for both capture
and internalization of dextran by MZM, and suggesting that mSIGNR1 captures antigens from
blood for degradation and antigen processing.
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
15
Discussion
APC are present in essentially every tissue where they operate at the interface of innate and
acquired immunity by recognizing pathogens and presenting pathogen-derived peptides to T
cells. C-type lectins play an important role as pathogen recognition receptors that capture and
destroy pathogens for antigen processing to permit MHC class II-restricted presentation. It is
becoming clear that not all C-type lectins serve as antigen receptors recognizing pathogens
through carbohydrate structures. The DC-specific C-type lectin hDC-SIGN is unique in that it
regulates adhesion processes, such as DC trafficking through ICAM-2, and T cell synapse
formation through ICAM-3, as well as antigen capture3;4. Moreover, even though several C-
type lectins have been shown to bind HIV-1, hDC-SIGN not only captures HIV-1, but also
protects it in early endosomes allowing HIV-1 transport by peripheral DC to the lymphoid
tissues, where it enhances trans-infection of T cells5.
However, functional studies into the in vivo function of DC-SIGN have been hampered by the
lack of murine homologues. Here we report the identification and isolation of a cDNA called
OtB7 which encodes a murine protein that is homologous to both hDC-SIGN and hL-SIGN
(Figure 1b). During the preparation of this manuscript, five murine DC-SIGN homologues
were identified by RT-PCR from CD11c+ DC although only one, called murine DC-SIGN
(mDC- SIGN) was expressed at high mRNA levels in CD11c+ DC12. In contrast the other
cDNAs, designated mSIGNR1-4, were hardly detectable in DC, but were detected at various
mRNA levels in B and T cells by RT-PCR12. The OtB7 clone that we have isolated from
lymph node cDNA is identical to the DC-SIGN homologue mSIGNR1. We generated
polyclonal antibodies against mSIGNR1 to investigate the cell-specific expression of this
hDC-SIGN homologue, because of the complex mRNA expression patterns of the reported
DC-SIGN homologues. mSIGNR1 is expressed neither by in vitro-generated DC nor by in situ
CD11c+ DC in spleen and lymph nodes (Figure 3 and 4). However, mSIGNR1 is expressed by
LSEC in liver and by medullary and subcapsular macrophages in the lymph nodes (Figure 3).
This specific expression is very similar to that of hL-SIGN, which is expressed by LSEC6;8
and by a specific subpopulation of non-endothelial macrophage-like cells in lymph nodes (A.
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
16
Engering, personal communication), indicating that, based on expression, mSIGNR1 is more
closely related to hL-SIGN than to hDC-SIGN.
Intermediate mRNA levels of mSIGNR1 were detected in murine spleen by RNA
hybridization (data not shown and 12). Here, we demonstrated using mSIGNR1-specific
antibodies that mSIGNR1 is specifically expressed by ER-TR9+ MZM in the spleen (Figure
4). These data were further confirmed by the intravenous injection of liposomes containing
dichloromethylene diphosphonate, which systemically depletes macrophages from the red
pulp and marginal zone25. Injection of the liposomes depleted the spleen of MZM as shown by
the absence of ER-TR9 staining (Figure 4e). No mSIGNR1 was present in MZM-depleted
spleens confirming the specific expression of mSIGNR1 by MZM (Figure 4f). Interestingly,
hDC-SIGN is expressed in spleen in contrast to hL-SIGN (T.G., unpublished results). Thus,
the expression pattern of mSIGNR1 is similar but not identical to that of hL-SIGN, and
mSIGNR1 has attributes of both hDC-SIGN and hL-SIGN suggesting that the distinction
between hL-SIGN and hDC-SIGN may be different and for less defined in the murine system.
The C-type lectin domain of mSIGNR1 has a 74% similarity with that of hDC-SIGN and the
amino acid residues important for both Ca2+- and ligand-binding19 are conserved in the murine
homologue. Indeed, mSIGNR1 has a similar binding activity for hICAM-2, hICAM-3 and
HIV-1 gp120 to both hDC-SIGN and hL-SIGN (Figure 2). Thus, mSIGNR1 can function as
an adhesion receptor. However, mice do not express an ICAM-3 homologue and therefore
binding of mSIGNR1 to ICAM-3 is not physiological. mSIGNR1 also interacts with murine
ICAM-2, which is widely expressed on murine lymphocytes29, and could therefore function as
the leukocyte ligand for mSIGNR1 mediating contact between mSIGNR1-postive cells and
leukocytes in mice. Strikingly, mSIGNR1 is abundantly expressed in liver and spleen by
LSEC and MZM respectively. Both cell-types are in direct contact with the blood and are
important in lymphocyte interactions. LSEC-leukocyte interactions appear to constitute a
central mechanism of peripheral immune surveillance in the liver30. The expression and
ligand-specificity of mSIGNR1 suggests a physiological role for this receptor in LSEC-
leukocyte adhesion through binding of ICAM-2 on lymphocytes, similar to hL-SIGN6. The
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
17
strategic localization of MZM in the spleen resembles that of LSEC in the liver, and the
splenic marginal zone is important as a gateway for migrating lymphocytes (reviewed in 24).
MZM interact with incoming lymphocytes and antigens in the marginal sinus since part of the
arterial blood supply opens into the wider space of the marginal sinus resulting in a blood flow
with lower resistance. The only entry into the white pulp is via the marginal zone, and
lymphocytes entering the marginal zone are either directed into the white pulp or will leave
the marginal zone through the red pulp side. Little is known about the cellular interactions
important for this lymphocyte migration but carbohydrate interactions are known to be
important in this migration to the white pulp since polysaccharides such as mannan inhibit this
process26;31;32. Strikingly, here we demonstrate that the mSIGNR1-dependent binding of
ICAM-2 is inhibited by mannan suggesting that mSIGNR1 provides the interactions that
enable lymphocyte migration into the white pulp. This hypothesis is further supported by the
clustering of MZM with lymphocytes26 and the high expression of ICAM-2 on murine
lymphocytes29.
Besides being a gateway for migrating lymphocytes, the splenic marginal zone is also
important as a defense against pathogens. Because of their position adjacent to the marginal
sinuses, MZM are amongst the first cells that can interact with blood-born antigens and are
presumed to have a critical role in host defense against bacterial pathogen (reviewed in 24).
MZM are specifically stained by the monoclonal antibody ER-TR913 and although its antigen
had not been identified, results suggested that the ER-TR9-antigen is involved in antigen
capture. Strikingly, mSIGNR1 is specifically expressed by MZM and we demonstrate here
that mSIGNR1 is the antigen of ER-TR9 (Figure 5a and b). Thus, the C-type lectin mSIGNR1
may have a function as a pathogen recognition receptor. C-type lectins play an important role
as pathogen recognition receptors that capture and destroy pathogens for antigen processing to
permit MHC class II-restricted presentation. Recently, we have demonstrated that hDC-SIGN
functions as a pathogen recognition receptor in vitro that rapidly internalizes captured antigen
into lysosomal compartments for processing and antigen presentation4. Therefore, we
investigated whether mSIGNR1 is involved in pathogen recognition and capture in vivo.
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
18
MZM are highly phagocytotic cells as demonstrated by their depletion after injection of
liposomes containing dichloromethylene diphosphonate (Figure 4) and their capacity to filter
pathogens from blood27. Indeed, bacterial antigens rapidly localize to the MZM as
demonstrated by intravenous injection of naïve wild-type mice with the model antigen, FITC-
dextran (Figure 5c). The FITC-dextran is specifically captured by the MZM in the marginal
zone and the FITC-staining co-localizes with antibodies against mSIGNR1 (Figure 5d).
Treatment of naïve mice with purified anti-SIGNR1 antibody ER-TR9 prior to the FITC-
dextran injection completely abrogates dextran binding by MZM (Figure 5e), demonstrating
that mSIGNR1 mediates the dextran capture by MZM. The anti-SIGNR1 antibody ER-TR9
also inhibits the in vitro binding of mSIGNR1 to dextran-FITC (Figure 6a) confirming that
ER-TR9 indeed inhibits the in vivo observed antigen uptake by mSIGNR1. Injection of the
yeast-derived polysaccharide mannan prior to FITC-dextran injection also prevents dextran-
binding by MZM, demonstrating that mSIGNR1 also binds mannan in vivo (Figure 5f) and
that mannan blocks the in vivo function of mSIGNR1. These data demonstrate that mSIGNR1
is involved in pathogen recognition by MZM in vivo. Moreover, antigens captured by
mSIGNR1 are rapidly internalized and targeted to lysosomes for degradation (Figure 6). Rapid
internalization of mSIGNR1 upon ligand binding (Figure 6b) suggests that ER-TR9 blocks
binding in vivo through downregulation of mSIGNR1 on the surface of MZM. The
cytoplasmic region of mSIGNR1 lacks the di-leucine motif present in hDC-SIGN, which
mediates the internalization of hDC-SIGN. However, the murine receptor contains a tri-acid
cluster (DDD) in its cytoplasmic region that functions as an internalization motif in DEC-205
where it is responsible for targeting to lysosomes for antigen processing33. The observed
lysosomal targeting of internalized ligand-mSIGNR1 complexes is likely be due to this tri-
acidic cluster. Thus, mSIGNR1 is important in the defense against pathogens by capturing
pathogens from the blood, which are then rapidly internalized into lysosomes and processed.
MZM do not express MHC class II molecules and it is thought that pathogen degradation
products are shed from the cell-surface and taken up by marginal zone B cells after
opsonization by complement. The marginal zone B cells migrate into the follicle and present
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
19
the captured antigens to elicit an immune response34. Thus, the function of mSIGNR1 links
innate immunity with adaptive immunity. Furthermore, the affinity of mSIGNR1 for HIV-1
gp120 suggests that mSIGNR1 may be able to interact with both pathogens containing
dextran- and mannan-like structures, and viruses containing viral components with structures
similar to gp120.
In conclusion, we have isolated a murine homologue of hDC-SIGN, mSIGNR1, and
investigated its cell-specific protein expression using antibodies against mSIGNR1. We have
demonstrated that mSIGNR1 functions as a pathogen recognition receptor in vivo and thus is
important in the defense against pathogens. This in vivo model and the identification of
blocking anti-mSIGNR1 antibodies will facilitate studies into the in vivo function of DC-
SIGN.
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
20
Acknowledgements
We are grateful to E. van Kesteren-Hendrix and N. van Rooijen for liposome-depleted tissues.
We thank J. Samsom, W. Jansen and R. Mebius for helpful discussions and providing
antibodies, and L. Colledge for suggestions and editing of the manuscript.
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
21
References.
1. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature
1998;392:245-252.
2. Geijtenbeek TBH, Krooshoop DJEB, Bleijs DA et al. DC-SIGN-ICAM-2 interaction
mediates dendritic cell trafficking. Nature Immunology 2000;1:353-357.
3. Geijtenbeek TBH, Torensma R, van Vliet SJ et al. Identification of DC-SIGN, a novel
dendritic cell-specific ICAM-3 receptor that supports primary immune responses. Cell
2000;100:575-585.
4. Engering A, Geijtenbeek TBH, van Vliet SJ et al. The dendritic cell-specific adhesion
receptor DC-SIGN internalizes antigen for presentation to T cells. J Immunol. 2002;168:2118-
2126.
5. Geijtenbeek TBH, Kwon DS, Torensma R, et al. DC-SIGN, a dendritic cell-specific HIV-1-
binding protein that enhances trans-infection of T cells. Cell 2000;100:587-597.
6. Bashirova AA, Geijtenbeek TBH, van Duijnhoven GCF et al. A dendritic cell-specific
intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN)-related protein is highly
expressed on human liver sinusoidal endothelial cells and promotes hiv-1 infection. J Exp
Med. 2001;193:671-678.
7. Soilleux EJ, Barten R, Trowsdale J. Cutting Edge: DC-SIGN; a Related Gene, DC-SIGNR;
and CD23 Form a Cluster on 19p13. J Immunol. 2000;165:2937-2942.
8. Pohlmann S, Soilleux EJ, Baribaud F et al. DC-SIGNR, a DC-SIGN homologue expressed
in endothelial cells, binds to human and simian immunodeficiency viruses and activates
infection in trans. Proc Natl Acad Sci U S A. 2001;98:2670-2675.
9. Limmer A, Ohl J, Kurts C et al. Efficient presentation of exogenous antigen by liver
endothelial cells to CD8+ T cells results in antigen-specific T-cell tolerance. Nat Med.
2000;6:1348-1354.
10. Mahnke K, Guo M, Lee S et al. The dendritic cell receptor for endocytosis, DEC-205,
can recycle and enhance antigen presentation via major histocompatibility complex class II-
positive lysosomal compartments. J Cell Biol. 2000;151:673-684.
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
22
11. Stahl PD, Ezekowitz RA. The mannose receptor is a pattern recognition receptor
involved in host defense. Curr Opin Immunol. 1998;10:50-55.
12. Park CG, Takahara K, Umemoto E et al. Five mouse homologues of the human
dendritic cell C-type lectin, DC- SIGN. Int Immunol. 2001;13:1283-1290.
13. Dijkstra CD, Van Vliet E, Dopp EA, van der Lelij AA, Kraal G. Marginal zone
macrophages identified by a monoclonal antibody: characterization of immuno- and enzyme-
histochemical properties and functional capacities. Immunology.1985;55:23-30.
14. Xu H, Tong IL, De Fougerolles AR, Springer TA. Isolation, characterization, and
expression of mouse ICAM-2 complementary and genomic DNA. J Immunol. 1992;149:2650-
2656.
15. Fawcett J, Holness CL, Needham LA et al. Molecular cloning of ICAM-3, a third ligand
for LFA-1, constitutively expressed on resting leukocytes. Nature 1992;360:481-484.
16. Inaba K, Inaba M, Romani N et al. Generation of large numbers of dendritic cells from
mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating
factor. J Exp Med.1992;176:1693-1702.
17. Van Rooijen N, Sanders A. Liposome mediated depletion of macrophages: mechanism
of action, preparation of liposomes and applications. J Immunol Methods. 1994;174:83-93.
18. Van Oosterhout AJ, Hofstra CL, Shields R et al. Murine CTLA4-IgG treatment inhibits
airway eosinophilia and hyperresponsiveness and attenuates IgE upregulation in a murine
model of allergic asthma. Am J Respir Cell Mol Biol. 1997;17:386-392.
19. Geijtenbeek TBH, van Duijnhoven GCF, van Vliet SJ et al. Identification of different
binding sites in the dendritic cell- specific receptor DC-SIGN for intercellular adhesion
molecule 3 and HIV- 1. J Biol Chem. 2002;277:11314-11320.
20. Sol-Foulon N, Moris A, Nobile C et al. HIV-1 nef induced ip-regulation of DC-SIGN
in dendritic cells promotes lymphocyte clustering and viral spread. Immunity 2002;16:145-
155.
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
23
21. Curtis BM, Scharnowske S, Watson AJ. Sequence and expression of a membrane-
associated C-type lectin that exhibits CD4-independent binding of human immunodeficiency
virus envelope glycoprotein gp120. Proc Natl Acad Sci U S A. 1992;89:8356-8360.
22. Baribaud F, Pohlmann S, Sparwasser T et al. Functional and antigenic characterization
of human, rhesus macaque, pigtailed macaque, and murine dc-sign. J Virol.2001;75:10281-9.
23. Kraal G, Janse M. Marginal metallophilic cells of the mouse spleen identified by a
monoclonal antibody. Immunology. 1986;58:665-669
24. Kraal G. Cells in the marginal zone of the spleen. Int Rev Cytol.1992;132:31-74.
25. van Rooijen N, van Nieuwmegen R. Elimination of phagocytic cells in the spleen after
intravenous injection of liposome-encapsulated dichloromethylene diphosphonate. An
enzyme-histochemical study. Cell Tissue Res. 1984;238:355-358.
26. Lyons AB, Parish CR. Are murine marginal-zone macrophages the splenic white pulp
analog of high endothelial venules? Eur J Immunol. 1995;25:3165-3172.
27. Humphrey JH, Grennan D. Different macrophage populations distinguished by means of
fluorescent polysaccharides. Recognition and properties of marginal-zone macrophages. Eur J
Immunol. 1981;11:221-228.
28. Feinberg H, Mitchell DA, Drickamer K, Weis WI. Structural Basis for Selective
Recognition of Oligosaccharides by DC- SIGN and DC-SIGNR. Science 2001;294:2163-
2166.
29. Xu H, Bickford JK, Luther E et al. Characterization of murine intercellular adhesion
molecule-2. J Immunol. 1996;156:4909-4914.
30. Knolle PA, Gerken G. Local control of the immune response in the liver. Immunol Rev.
2000;174:21-34.
31. Weston SA, Parish CR. Evidence that mannose recognition by splenic sinusoidal cells
plays a role in the splenic entry of lymphocytes. Eur J Immunol. 1992;22:1975-1981.
32. Weston SA, Parish CR. Modification of lymphocyte migration by mannans and
phosphomannans. Different carbohydrate structures control entry of lymphocytes into spleen
and lymph nodes. J Immunol. 1991;146:4180-4186.
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
24
33. Mahnke K, Guo M, Lee S et al. The dendritic cell receptor for endocytosis, DEC-205,
can recycle and enhance antigen presentation via major histocompatibility complex class II-
positive lysosomal compartments. J Cell Biol. 2000;151:673-684.
34. van Rooijen N. Antigen processing and presentation in vivo: the microenvironment as a
crucial factor. Immunol Today 1990;11:436-439.
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
25
Legends
Figure 1. Clone OtB7 is identical to the murine DC-SIGN homologue SIGNR1
a. Schematic representation of the genomic structure of clone OtB7. Exons are numbered and
depicted as black bars. The genomic sequence was constructed by a combination of BLAST
searches and multiple alignment analyses, and is composed of overlapping sequences present
in the Genbank entries (AC073804.1 and AC073706.2).
b. Clone OtB7 is homologous to both human DC-SIGN and L-SIGN as demonstrated by the
amino acid sequence alignments of clone OtB7 (Genbank AF422108) with human DC-SIGN
(AAK20997) and L-SIGN (AAK20998). Amino acid residues involved in direct ligand
binding, as was shown for DC-SIGN 19, are denoted by filled arrows.
Figure 2. mSIGNR1 binds both mouse ICAM-2 and HIV-1 gp120, similar to both hDC-
SIGN and hL-SIGN.
a. Polyclonal antibodies against mSIGNR1 stain 293T cells transfected with mSIGNR1
cDNA. 293T cells were transfected with different cDNAs and subsequently stained with
antibodies against hDC-SIGN (AZN-D1; filled histogram) and mSIGNR1 (698; filled
histogram). Isotype controls are shown as open histograms. One representative experiment out
of three is shown.
b. mSIGNR1 binds human ICAM-2, ICAM-3 and HIV-1 gp120 but not human ICAM-1,
similar to hDC-SIGN and hL-SIGN. The adhesion of 293T transfected with mSIGNR1 to the
ligands was determined using the fluorescent bead adhesion assay. Specificity was determined
in the presence of blocking antibodies (AZN-D2, 20 µg/ml), mannan (5 µg/ml) or EGTA (5
mM). Cells were also pre-incubated with the polyclonal antibody against mSIGNR1 (1:10).
One representative experiment out of three is shown.
c. mSIGNR1 binds murine ICAM-2 with high affinity, similar to hDC-SIGN and hL-SIGN.
The adhesion assay was performed as described in Figure 2b. One representative experiment
out of three is shown.
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
26
Figure 3. The expression pattern of mSIGNR1 is similar to that of hL-SIGN.
a. mSIGNR1 is not expressed by bone-marrow-derived DC or by splenic CD11c+ DC. Bone-
marrow derived DC and splenic CD11c+ DC were stained with the anti-mSIGNR1 antibody
698. Bone marrow DC were more than 90% pure as determined by CD11c staining (data not
shown). Dotted and open histogram represent isotype and mSIGNR1 staining, respectively.
Splenic DC were double stained with CD11c and the mSIGNR1-specific antibody 698.
b,c. Immuno-fluorescence staining of murine liver tissue sections with the polyclonal antibody
against mSIGNR1 698 (green, b and c) and double staining with F4/80 (red, c) (magnification
200x).
d-g. Immuno-fluorescence double staining of murine lymph node tissue sections with the
polyclonal antibody against mSIGNR1 698 (green) and in red CD11c (d), DEC-205 (e), SER-
4 (f), MOMA-1 (g). (magnification 200x).
Figure 4. mSIGNR1 is specifically expressed by splenic marginal zone macrophages.
a-d. Immuno-fluorescence double staining of murine spleen tissue sections with the polyclonal
antibody against mSIGNR1 698 (green) and in red CD11c (a), DEC-205 (b), ER-TR9 (c),
B220 (d) (magnification 200x).
e, f. Immuno-fluorescence of double staining of murine spleen tissue sections after depletion
with dichloromethylene diphosphonate liposomes. Double staining with (e) CD11c (red) and
ER-TR9 (green); and (f) CD11b (red) and polyclonal mSIGNR1 antibody 698 (green)
(magnification 200x).
Figure 5. mSIGNR1 efficiently captures polysaccharide antigens in vivo.
a. The MZM-specific antibody ER-TR9 stains 293T cells transfected with mSIGNR1 cDNA.
293T cells were transfected with mSIGNR1 cDNA and subsequently stained either with the
mSIGNR1-specific antibodies 698 or ER-TR9 (filled histograms). Isotype controls are
represented by open histograms.
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
27
b. Recombinant mSIGNR1 is specifically detected by ER-TR9 using a sandwich ELISA. ER-
TR9-coated wells are incubated with recombinant mSIGNR1 and binding is detected by the
mSIGNR1-specific antibody 698. Standard deviation < 0.05.
c-d. MZM efficiently capture dextran-FITC in vivo. Naïve mice were injected intravenously
with FITC-dextran. Spleens were isolated and immuno-fluorescence analyses of murine
spleen tissue sections were performed. Double stainings with FITC-dextran (green) and B220
(red, c) or mSIGNR1-specific antibody 698 (red, d) are shown (magnification 200x).
e-f. Dextran capture by MZM in vivo is blocked by the mSIGNR1-specific antibody ER-TR9
and mannan. Naïve mice were treated with either purified ER-TR9 (e) or the yeast-derived
polysaccharide mannan (f) prior to intravenous injection with FITC-dextran. Double stainings
were performed with FITC-dextran (green) and the mSIGNR1-specific antibody 698 (red)
(magnification 200x).
Figure 6. mSIGNR1 mediates rapid internalization of captured dextran.
a. Dextran is efficiently captured by mSIGNR1. K562 cells expressing mSIGNR1 and mock
transfected K562 cells were incubated with FITC-dextran (1 µg/ml) at 37°C and unbound
dextran was washed away after 45 minutes. Specificity was determined in the presence of the
antibody ER-TR9. Binding was analyzed by flow cytometry. Standard deviation < 5%.
b. Dextran is rapidly internalized by SIGNR1. The fate of bound FITC-dextran was followed
by analyzing FITC-staining of K562 cells expressing mSIGNR1 after incubation with FITC-
dextran using immuno-fluorescence microscopy (magnification 200x).
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
28
Figure 1
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
29
Figure 2
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
30
Figure 3
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
31
Figure 4
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
32
Figure 5
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
33
Figure 6.
For personal use only. by guest on May 29, 2013. bloodjournal.hematologylibrary.orgFrom
... DC-SIGN is a receptor expressed by DCs ex vivo and in vivo (33,34). DC-SIGN receptors in mice have eight orthologs, including SIGN-R1 to SIGN-R8 (35), and dextran uptake via the DC-SIGN family receptors SIGN-R1 and SIGN-R3 has been reported in mice (36,37). The DC-SIGN signalosome is a complex composed of the scaffold proteins LSP1, KSR1, and CNK and is required for the constitutive association of DC-SIGN with RAF1 and the association or isolation of the compounds with more proinflammatory effects (16). ...
Article
Full-text available
The binding of tumor necrosis factor-like cytokine 1A (TL1A) to death receptor 3 (DR3) plays an important role in the interaction between dendritic cells (DCs) and T cells and contributes to intestinal inflammation development. However, the mechanism by which DCs expressing TL1A mediate helper T (Th) cell differentiation in the intestinal lamina propria (LP) during the pathogenesis of inflammatory bowel disease remains unclear. In this study, we found that TL1A/DR3 promoted Th1 and Th17 cell differentiation in T-T and DC-T cell interaction-dependent manners. TL1A-deficient CD4+ T cells failed to polarize into Th1/Th17 cells and did not cause colonic inflammation in a T cell transfer colitis model. Notably, TL1A was located in the cytoplasm and nuclei of DCs, positively regulated the DC-specific ICAM-grabbing nonintegrin/RAF1/nuclear factor κB signaling pathway, enhanced the antigen uptake ability of DCs, and promoted TLR4-mediated DC activation, inducing naive CD4+ T cell differentiation into Th1 and Th17 cells. Our work reveals that TL1A plays a regulatory role in inflammatory bowel disease pathogenesis.
... In the spleen, S.Tm is found within tissue resident macrophages: red-pulp macrophages identified as F4/ 80 1 cells and marginal zone macrophages identified as Siglec1 1 cells (58,75). Marginal zone macrophages express SIGNR1, which binds to bacteria and other pathogens (76)(77)(78). Tissue resident red-pulp macrophages express natural resistance-associated macrophage protein 1 (NRAMP1), which is associated with protection against S.Tm (39). Mice lacking functional NRAMP1 (e.g., C57BL6/J) are susceptible to S.Tm infection, while mice with a functional Nramp1 gene (e.g., 129sv mice [79]) can provide a model of chronic infection. ...
Article
Full-text available
Bacterial pathogens can invade the tissue and establish a protected intracellular niche at the site of invasion that can spread locally (e.g., microcolonies) or to systemic sites (e.g., granulomas). Invasion of the tissue and establishment of intracellular infection are rare events that are difficult to study in the in vivo setting but have critical clinical consequences, such as long-term carriage, reinfections, and emergence of antibiotic resistance. Here, I discuss Salmonella interactions with its host macrophage during early stages of infection and their critical role in determining infection outcome. The dynamics of host-pathogen interactions entail highly heterogenous host immunity, bacterial virulence, and metabolic cross talk, requiring in vivo analysis at single-cell resolution. I discuss models and single-cell approaches that provide a global understanding of the establishment of a protected intracellular niche within the tissue and the host-pathogen landscape at infection bottlenecks during early stages of infection. Studying cellular host-pathogen interactions in vivo can improve our knowledge of the trajectory of infection between the initial inoculation with a dose of pathogens and the appearance of symptoms of disease.
... Two specific subsets of macrophages are also found here: MZ macrophages (MZMs) and m arginal metallophilic macrophages (MMMs) (76,87). The MZMs line the outer ring of the WP and are characterized by expression of C-type lectin SIGNR1 and type I scavenger receptor MARCO (85,88). The MMMs form the inner ring, located closer to the WP (89,90). ...
Article
Full-text available
Mesenchymal stromal cells (MSCs) have demonstrated therapeutic potential in inflammatory models of human disease. However, clinical translation has fallen short of expectations, with many trials failing to meet primary endpoints. Failure to fully understand their mechanisms of action is a key factor contributing to the lack of successful commercialisation. Indeed, it remains unclear how the long-ranging immunomodulatory effects of MSCs can be attributed to their secretome, when MSCs undergo apoptosis in the lung shortly after intravenous infusion. Their apoptotic fate suggests that efficacy is not based solely on their viable properties, but also on the immune response to dying MSCs. The secondary lymphoid organs (SLOs) orchestrate immune responses and play a key role in immune regulation. In this review, we will discuss how apoptotic cells can modify immune responses and highlight the importance of MSC-immune cell interactions in SLOs for therapeutic outcomes.
Article
Blood‐borne pathogen (BBP) infections can rapidly progress to life‐threatening sepsis and must therefore be promptly eliminated by the host's immune system. Intravascular macrophages of the liver sinusoid, splenic marginal zone and red pulp and perisinusoidal macrophage protrusions in the bone marrow (BM) directly phagocytose BBPs in the blood as an innate immune response. The liver, spleen and BM thereby work together as the blood defence system (BDS) in response to BBPs by exerting their different immunological roles. The liver removes the vast majority of these invading organisms via innate immunity, but their complete elimination is not possible without the actions of antibodies. Splenic marginal zone B cells promptly produce IgM and IgG antibodies against BBPs. The splenic marginal zone transports antigenic information from the innate to the adaptive immune systems. The white pulp of the spleen functions as adaptive immune tissue and produces specific and high‐affinity antibodies with an immune memory against BBPs. The BM works to maintain immune memory by supporting the survival of memory B cells, memory T cells and long‐lived plasma cells (LLPCs), all of which have dedicated niches. Furthermore, BM perisinusoidal naïve follicular B cells promptly produce IgM antibodies against BBPs in the BM sinusoid and the IgG memory B cells residing in the BM rapidly transform to plasma cells which produce high‐affinity IgG antibodies upon reinfection. This review describes the complete immune defence characteristics of the BDS against BBPs through the collaboration of the liver, spleen and BM with combined different immunological roles.
Chapter
Emerging evidence suggests that dendritic cells play a major role in the orchestration of the immune response to bacteria. This volume introduces the reader to the complex world of dendritic cells and describes how the intimate interplay between dendritic cells, bacteria and the environment dictates either the induction of immunity or tolerance to the encountered microorganisms. It discusses how this can allow organisms to tolerate beneficial bacteria and to react against pathogens, as well as the strategies pathogenic bacteria have evolved to escape dendritic cell patrolling. Expert contributors discuss everything from bacterial capture and recognition to their killing, processing and the induction of adaptive immunity. Particular focus is on the tissue context in which bacteria are handled by dendritic cells and on possible defects therein, which may potentially lead to chronic infection or inflammation. Graduate students and researchers will find this an invaluable overview of current dendritic cell biology research.
Chapter
Human macrophages (MΦ) represent the crucial innate elements for the orchestration of adaptive immune responses, since they are present in all tissues of the body and display a large spectrum of phenotypic and functional features. These cells present a high level of plasticity and could be derived from the embryogenic precursors as well as circulatory blood monocytes. A series of different studies have uncovered how tissue-derived signals, soluble factors, and/or antigen stimulation may contribute to epigenetic, transcriptomic, and functional landscape of human MΦ. In this chapter, MΦ differentiation properties will be discussed, mostly focusing on how the new experimental and technological approaches are changing the bimodal human MΦ paradigm towards a much more diverse, tissue-adapted, and specialized panorama.
Chapter
Macrophages populate the kidney in several waves during kidney development, out of which some remain there and, together with dendritic cells, form a peritubular network of resident mononuclear phagocytes. Little information exists on how these cells contribute to homeostasis in a healthy kidney. During the different phases of kidney injury and repair, resident and infiltrating macrophages both amplify changing local microenvironments. In an early injury, they empower host defense to infections or enhance sterile inflammation, while later they enhance the resolution of inflammation and healing responses. In chronic kidney disease, macrophages contribute to continued inflammatory tissue remodeling and tissue fibrosis. The different functional properties of kidney macrophages associate with different gene expression patterns, surface markers, and cytokine secretion profiles, which have led to different phenotype classifications. A universal classification concept that integrates all different views on kidney macrophages still does not exist. However, numerous interventional studies demonstrate that modulating macrophage recruitment and activation patterns can modulate the outcome of kidney disease models. This chapter gives an overview of general concepts and main findings of macrophages in the kidney, a dynamic field with inputs from many different research domains.
Chapter
Osteoclasts are giant multinucleated cells derived from myeloid precursor cells in the bone marrow and have a unique bone-destroying capacity. Osteoclasts are essential for homeostatic bone remodeling in health, and the receptor activator of nuclear factor-kappa B ligand (RANKL) is the critical cytokine that induces osteoclastogenesis. However, several factors can disrupt this essential physiological process, including menopause-associated hormonal changes, age-related factors, trauma, drugs, and infectious diseases, which lead to the development of various bone disorders in both women and men. Here we discuss the historical background and the recent advances in understanding osteoclast differentiation by focusing on the RANKL signaling. Also, we review the major osteolytic diseases emphasizing our current knowledge of the underlying pathophysiological mechanisms.
Chapter
Secondary lymphoid organs (SLOs) are well-organized structures that control immune responses. These tissues allow for the capture, processing, and presentation of antigens, leading to successful elimination of pathogens and induction of adaptive immunity. Among lymphoid organs, the spleen and lymph nodes are strategically located to efficiently trap foreign antigens derived from the bloodstream and peripheral tissues, respectively. SLOs contain many different cells of the immune system, including antigen‐presenting cells and lymphocytes. The cross talk between these cell types is crucial for maintaining local and systemic homeostasis and inducing rapid immune responses that protect against pathogenic infection. Among antigen-presenting cells, several macrophage populations can be found in the different compartments of the spleen and lymph node. These cells are often positioned in strategic locations within lymphoid organs, allowing them to act as sentinels at vulnerable sites. As such, these cells are involved in scavenging blood-borne debris, iron recycling by phagocytosis of red blood cells, as well as playing an important role in host defense by bridging the innate and adaptive arms of the immune system. In this chapter, we will describe the various specialized macrophage populations within the lymph node and spleen microenvironment, as well as review the importance of these cells in shaping the nature and quality of immunity and inflammation.
Article
Full-text available
Contact between dendritic cells (DC) and resting T cells is essential to initiate a primary immune response. Here, we demonstrate that ICAM-3 expressed by resting T cells is important in this first contact with DC. We discovered that instead of the common ICAM-3 receptors LFA-1 and alphaDbeta2, a novel DC-specific C-type lectin, DC-SIGN, binds ICAM-3 with high affinity. DC-SIGN, which is abundantly expressed by DC both in vitro and in vivo, mediates transient adhesion with T cells. Since antibodies against DC-SIGN inhibit DC-induced proliferation of resting T cells, our findings predict that DC-SIGN enables T cell receptor engagement by stabilization of the DC-T cell contact zone.
Article
Full-text available
The discovery of dendritic cell (DC)-specific intercellular adhesion molecule (ICAM)-3–grabbing nonintegrin (DC-SIGN) as a DC-specific ICAM-3 binding receptor that enhances HIV-1 infection of T cells in trans has indicated a potentially important role for adhesion molecules in AIDS pathogenesis. A related molecule called DC-SIGNR exhibits 77% amino acid sequence identity with DC-SIGN. The DC-SIGN and DC-SIGNR genes map within a 30-kb region on chromosome 19p13.2-3. Their strong homology and close physical location indicate a recent duplication of the original gene. Messenger RNA and protein expression patterns demonstrate that the DC-SIGN–related molecule is highly expressed on liver sinusoidal cells and in the lymph node but not on DCs, in contrast to DC-SIGN. Therefore, we suggest that a more appropriate name for the DC-SIGN–related molecule is L-SIGN, liver/lymph node–specific ICAM-3–grabbing nonintegrin. We show that in the liver, L-SIGN is expressed by sinusoidal endothelial cells. Functional studies indicate that L-SIGN behaves similarly to DC-SIGN in that it has a high affinity for ICAM-3, captures HIV-1 through gp120 binding, and enhances HIV-1 infection of T cells in trans. We propose that L-SIGN may play an important role in the interaction between liver sinusoidal endothelium and trafficking lymphocytes, as well as function in the pathogenesis of HIV-1.
Article
Full-text available
Dendritic cells (DC) capture microorganisms that enter peripheral mucosal tissues and then migrate to secondary lymphoid organs, where they present these in antigenic form to resting T cells and thus initiate adaptive immune responses. Here, we describe the properties of a DC-specific C-type lectin, DC-SIGN, that is highly expressed on DC present in mucosal tissues and binds to the HIV-1 envelope glycoprotein gp120. DC-SIGN does not function as a receptor for viral entry into DC but instead promotes efficient infection in trans of cells that express CD4 and chemokine receptors. We propose that DC-SIGN efficiently captures HIV-1 in the periphery and facilitates its transport to secondary lymphoid organs rich in T cells, to enhance infection in trans of these target cells.
Article
Antigen processing and presentation in in vitro is an increasingly well understood phenomenon. However, in vivo, a large number of variables conspire to obscure and confuse. In this article, Nico van Rooijen attempts to bring order to events that occur in the spleen after antigenic challenge: starting with the large body of reliable in vitro data he incorporates information on splenic anatomy, cell trafficking and the cellular microenvironment to arrive at a physiological model for antigen handling in vivo.
Article
Contact between dendritic cells (DC) and resting T cells is essential to initiate a primary immune response. Here, we demonstrate that ICAM-3 expressed by resting T cells is important in this first contact with DC. We discovered that instead of the common ICAM-3 receptors LFA-1 and alphaDbeta2, a novel DC-specific C-type lectin, DC-SIGN, binds ICAM-3 with high affinity. DC-SIGN, which is abundantly expressed by DC both in vitro and in vivo, mediates transient adhesion with T cells. Since antibodies against DC-SIGN inhibit DC-induced proliferation of resting T cells, our findings predict that DC-SIGN enables T cell receptor engagement by stabilization of the DC-T cell contact zone.
Article
Antigen-presenting, major histocompatibility complex (MHC) class II-rich dendritic cells are known to arise from bone marrow. However, marrow lacks mature dendritic cells, and substantial numbers of proliferating less-mature cells have yet to be identified. The methodology for inducing dendritic cell growth that was recently described for mouse blood now has been modified to MHC class II-negative precursors in marrow. A key step is to remove the majority of nonadherent, newly formed granulocytes by gentle washes during the first 2-4 d of culture. This leaves behind proliferating clusters that are loosely attached to a more firmly adherent "stroma." At days 4-6 the clusters can be dislodged, isolated by 1-g sedimentation, and upon reculture, large numbers of dendritic cells are released. The latter are readily identified on the basis of their distinct cell shape, ultrastructure, and repertoire of antigens, as detected with a panel of monoclonal antibodies. The dendritic cells express high levels of MHC class II products and act as powerful accessory cells for initiating the mixed leukocyte reaction. Neither the clusters nor mature dendritic cells are generated if macrophage colony-stimulating factor rather than granulocyte/macrophage colony-stimulating factor (GM-CSF) is applied. Therefore, GM-CSF generates all three lineages of myeloid cells (granulocytes, macrophages, and dendritic cells). Since > 5 x 10(6) dendritic cells develop in 1 wk from precursors within the large hind limb bones of a single animal, marrow progenitors can act as a major source of dendritic cells. This feature should prove useful for future molecular and clinical studies of this otherwise trace cell type.
Article
DC-SIGN, a human C-type lectin, is expressed on the surface of dendritic cells (DC), while a closely related human gene, DC-SIGNR or L-SIGN, is found on sinusoidal endothelial cells of liver and lymph node. Both DC-SIGN and DC-SIGNR/L-SIGN can bind ICAM-3 and HIV gp120, and transmit HIV to susceptible cells in trans. Here, we report the cloning of five mouse genes homologous to human DC-SIGN and DC-SIGNR/L-SIGN. Only one gene, named mouse DC-SIGN, is highly expressed in DC, and is not found in a panel of mouse macrophage and lymphocyte cell lines. The other four genes, named mouse SIGNR1 (SIGN-Related gene 1), SIGNR2, SIGNR3 and SIGNR4, are expressed at lower levels in various cells according to RT-PCR and Northern blot analyses on RNA. All the genes of mouse DC-SIGN and SIGNRs map to adjacent regions of chromosome 8 A1.2‐1.3. However, like human DC-SIGN, only the mouse DC-SIGN gene is closely juxtaposed to the CD23 gene, while the other four SIGNR genes are located close to each other in a neighboring region. mRNAs of mouse DC-SIGN and three SIGNR genes encode type II transmembrane proteins (DC-SIGN, 238 amino acids; SIGNR1, 325 amino acids; SIGNR3, 237 amino acids; SIGNR4, 208 amino acids), but the SIGNR2 gene only encodes a carbohydrate recognition domain (CRD) without a cytosolic domain and a transmembrane domain (SIGNR2, 178 amino acids). Amino acid sequence similarities between the CRD of human DC-SIGN and the mouse homologues are 67% for DC-SIGN, 69% for SIGNR1, 65% for SIGNR2, 68% for SIGNR3 and 70% for SIGNR4 respectively. However, the membrane proximal neck domains in the mouse genes are much shorter than their counterparts in human DC-SIGN and DC-SIGNR/L-SIGN. This family of mouse C-type lectins is therefore complex, but only one of the new genes, DC-SIGN, is juxtaposed to CD23 and is expressed at high levels in DC.
Article
The mannose receptor recognizes the patterns of carbohydrates that decorate the surfaces and cell walls of infectious agents. This macrophage and dendritic cell pattern-recognition receptor mediates endocytosis and phagocytosis. The mannose receptor is the prototype of a new family of multilectin receptor proteins (membrane-spanning receptors containing eight-ten lectin-like domains, which appear to play a key role in host defense) and provides a link between innate and adaptive immunity. Recent advances include the identification of three new members of the mannose receptor family, additional work on defining the molecular requirements for sugar binding, a role for the mannose receptor in antigen presentation of lipoglycan antigens and evidence that the mannose receptor is associated with a signal transduction pathway leading to cytokine production.
Article
Intercellular adhesion molecule-2 (ICAM-2), a cell surface glycoprotein, is a second counter-receptor for lymphocyte function-associated Ag-1 (LFA-1). We report here the isolation and characterization of the cDNA and the gene that encode murine ICAM-2 (Accession numbers X65493 and X65490, respectively). The deduced sequence of the cDNA has 60% amino acid identity with its human counterpart and has the same expression pattern in cells and tissues. Furthermore, COS cells transfected with mouse ICAM-2 complementary and genomic DNA bind to purified human LFA-1, demonstrating the conservation of the function of ICAM-2 as a ligand for LFA-1 and conservation across species of sequences that are critical for binding to human LFA-1. COS cells transfected with the ICAM-2 cDNA do not react with mAb PA3, previously suggested to define ICAM-2 in the mouse. The mouse ICAM-2 gene was isolated and its structural organization determined. The gene is present in a single copy in the mouse genome and contains four exons spanning about 5.0 kb of DNA. The exon/intron architecture correlates to the structural domains of the protein and resembles that of other Ig superfamily members. The gene for ICAM-2, which is constitutively expressed in endothelial cells, has several conserved sequence motifs in its promoter region, including a direct repeat, and lacks transcription factor-binding sites present in the ICAM-1 gene, which is inducible in endothelial cells.
Article
The co-ordinated function of effector and accessory cells in the immune system is assisted by adhesion molecules on the cell surface that stabilize interactions between different cell types. Leukocyte function-associated antigen 1 (LFA-1) is expressed on the surface of all white blood cells and is a receptor for intercellular adhesion molecules (ICAM) 1 and 2 (ref. 3) which are members of the immunoglobulin superfamily. The interaction of LFA-1 with ICAMs 1 and 2 provides essential accessory adhesion signals in many immune interactions, including those between T and B lymphocytes and cytotoxic T cells and their targets. In addition, both ICAMs are expressed at low levels on resting vascular endothelium; ICAM-1 is strongly upregulated by cytokine stimulation and plays a key role in the arrest of leukocytes in blood vessels at sites of inflammation and injury. Recent work has indicated that resting leukocytes express a third ligand, ICAM-3, for LFA-1 (refs 11, 12). ICAM-3 is potentially the most important ligand for LFA-1 in the initiation of the immune response because the expression of ICAM-1 on resting leukocytes is low. We report the expression cloning of a complementary DNA, pICAM-3, encoding a protein constitutively expressed on all leukocytes, which binds LFA-1. ICAM-3 is closely related to ICAM-1, consists of five immunoglobulin domains, and binds LFA-1 through its two N-terminal domains.