ArticlePDF Available

Normal human dermis contains distinct populations of CD11c+BDCA-1+ dendritic cells and CD163+FXIIIA+ macrophages

Authors:

Abstract and Figures

We used a panel of monoclonal antibodies to characterize DCs in the dermis of normal human skin. Staining for the CD11c integrin, which is abundant on many kinds of DCs, revealed cells in the upper dermis. These cells were positive for blood DC antigen-1 (BDCA-1; also known as CD1c), HLA-DR, and CD45, markers that are also expressed by circulating myeloid DCs. A small subset of CD11c+ dermal cells expressed DEC-205/CD205 and DC-lysosomal-associated membrane glycoprotein/CD208 (DC-LAMP/CD208), suggesting some differentiation or maturation. When BDCA-1+ cells were selected from collagenase digests of normal dermis, they proved to be strong stimulators for T cells in a mixed leukocyte reaction. A second major population of cells located throughout the dermis was positive for factor XIIIA (FXIIIA), but lacked CD11c and BDCA-1. They expressed the macrophage scavenger receptor CD163 and stained weakly for HLA-DR and CD45. Isolated CD163+ dermal cells were inactive in stimulating T cell proliferation, but in biopsies of tattoos, these cells were selectively laden with granular pigments. Plasmacytoid DCs were also present in the dermis, marked by CD123 and BDCA-2. In summary, the normal dermis contains typical immunostimulatory myeloid DCs identified by CD11c and BDCA-1, as well as an additional population of poorly stimulatory macrophages marked by CD163 and FXIIIA.
Content may be subject to copyright.
Research article
The Journal of Clinical Investigation http://www.jci.org  Volume 117    Number 9  September 2007  2517
Normal human dermis contains distinct
populations of CD11c
+
BDCA-1
+
dendritic
cells and CD163
+
FXIIIA
+
macrophages
Lisa C. Zaba,
1
Judilyn Fuentes-Duculan,
1
Ralph M. Steinman,
2
James G. Krueger,
1
and Michelle A. Lowes
1
1
Laboratory for Investigative Dermatology and
2
Laboratory of Cellular Physiology and Immunology, The Rockefeller University, New York, New York, USA.
We used a panel of monoclonal antibodies to characterize DCs in the dermis of normal human skin. Staining
for the CD11c integrin, which is abundant on many kinds of DCs, revealed cells in the upper dermis. These
cells were positive for blood DC antigen–1 (BDCA-1; also known as CD1c), HLA-DR, and CD45, markers that
are also expressed by circulating myeloid DCs. A small subset of CD11c
+
dermal cells expressed DEC-205/
CD205 and DC-lysosomal–associated membrane glycoprotein/CD208 (DC-LAMP/CD208), suggesting some
differentiation or maturation. When BDCA-1
+
cells were selected from collagenase digests of normal dermis,
they proved to be strong stimulators for T cells in a mixed leukocyte reaction. A second major population of
cells located throughout the dermis was positive for factor XIIIA (FXIIIA), but lacked CD11c and BDCA-1.
They expressed the macrophage scavenger receptor CD163 and stained weakly for HLA-DR and CD45. Isolat-
ed CD163
+
dermal cells were inactive in stimulating T cell proliferation, but in biopsies of tattoos, these cells
were selectively laden with granular pigments. Plasmacytoid DCs were also present in the dermis, marked
by CD123 and BDCA-2. In summary, the normal dermis contains typical immunostimulatory myeloid DCs
identified by CD11c and BDCA-1, as well as an additional population of poorly stimulatory macrophages
marked by CD163 and FXIIIA.
Introduction
DCs represent a major resident leukocyte population in human 
skin. Two main types of DCs are found in noninflamed skin: epi
-
dermal Langerhans cells (LCs) and dermal DCs (1, 2). LCs express 
Langerin/CD207, an endocytic receptor that localizes to and forms 
Birbeckgranules, as well as the CD1a class I–like molecule that 
presents glycolipids (3). Dermal DCs have long been defined on the 
basis of expression of a clotting factor, the transglutaminase fac
-
tor XIIIA (FXIIIA) (4). Studies that define dermal DCs as FXIIIA
+
have often relied on fluorescence-activated cell sorting(FACS) 
analysis and functional studies using bulk tissue or enzymatically 
manipulated émigrés that “crawl out” of the dermis over a variable 
incubation period, which may increase FXIIIA expression (5, 6).
Currently, so-called myeloid or conventional DCs in many tis
-
suesare oftenidentified on the basis of high expression of HLA-DR 
antigen-presenting molecules and the CD11cintegrin. DCs are 
negative forLin, a cocktail ofantibodies to other cell lineages, 
including T cells (CD3), B cells (CD19 andCD20),monocytes 
(CD14), and granulocytes and NK cells (CD16 and CD56) (7, 8). 
Circulating myeloid DCs can be further classified as 3 mutually 
exclusive subsets — in order of immunostimulatory capacity, blood 
DC antigen–1–positive (BDCA-1
+
), CD16
+
, and BDCA-3
+
(9) — but 
these markers have seen very little use in studies of the skin.
Because a large number of prevalent dermatologic conditions 
fromatopic dermatitistopsoriasis are characterized by extensive 
dermal Tcellinfiltration,and as DCs arepivotal antigen-pre
-
senting cells for T cells, it is important to pursue these distinct 
phenotypicdefinitionsof DCs innormal skinandperipheral 
blood. Here we report that CD11c and FXIIIA marked mutually 
exclusivepopulations, theformer coexpressingBDCA-1(also 
known asCD1c;ref.10),andthelatteruniformlypositively 
for CD163,a scavenger receptor forhemoglobin/haptoglobin 
complexes. CD11c
+
 cells were more typical of DCs, with higher 
HLA-DR expressionandTcell–stimulatingactivity. Surpris
-
ingly, FXIIIA
+
cells behavedmorelikemacrophagessince they 
were weak initiators of T cell responses in the mixed leukocyte 
reaction (MLR) and had numerous phagocytosedpigment–con
-
taining vacuoles in a tattoo.
Results
FXIIIA
+
and CD11c
+
cells are discrete dermal populations. Immuno-
histochemistry of normal human dermis showed distinct 
stainingpatternsforFXIIIA
+
andCD11c
+
cells  (Figure1A). 
Thelarger FXIIIA
+
cellswerelocatedthroughoutthe dermis. 
In contrast, CD11c
+
 myeloid cells were located in the papillary 
andupperreticulardermis. Wecountedtheabsolutenumber 
ofFXIIIA
+
andCD11c
+
cellsusingmatchedtissuesections 
from 15 normalvolunteers (Figure 1B). In the epidermis there 
werenoFXIIIA
+
cellsandameanof4CD11c
+
cellspermm 
(P= 0.04).In thedermis,therewasa mean of83FXIIIA
+
cells 
compared with61CD11c
+
cellspermm(P=0.19).Double-
labeledimmunofluorescenceusingFXIIIAandCD11ccon
-
firmedthatthese2antigenswere notexpressed onthesame 
cell(Figure1C).ApolyclonalFXIIIAantibodyhas  typically 
been used for staining dermal DCs in the past. We used FXIIIA 
affinity-purifiedsheepantibody forallofourdouble-label 
immunofluorescencestudies,aswefoundit toyieldthebest 
Nonstandard abbreviations used: BDCA, blood DC antigen; DC-LAMP, DC-lyso-
somal–associated membrane glycoprotein; DC-SIGN, DC-specific ICAM-3–grabbing 
nonintegrin; FACS, fluorescence-activated cell sorting; FXIIIA, factor XIIIA; LC, 
Langerhans cell; MFI, median fluorescence intensity; MLR, mixed leukocyte reaction; 
MMR, macrophage mannose receptor; PDC, plasmacytoid DC.
Conflict of interest:The authors have declared that no conflict of interest exists.
Citation for this article:
J. Clin. Invest.117:2517–2525 (2007). doi:10.1172/JCI32282.
Related Commentary, page 2382
research article
2518 The Journal of Clinical Investigation http://www.jci.org    Volume 117    Number 9  September 2007
staining. There was almost complete coexpression of the sheep 
FXIIIAantibodyand a commonlyused mousemonoclonal 
(clone AC-1A1),whileothermousemonoclonalantibodiesto 
FXIIIA produced weaker and less consistent staining (data not 
shown). These data indicate that 2 commonlyused markers for 
dermal DCs are actually expressedby comparably abundant but 
different populations.
FXIIIA
+
and CD11c
+
populations are not LCs or plasmacytoid DCs. 
To further characterize these 2 dermal cell populations, we first 
assessed expression of markers used to identify LCs and/or plas
-
macytoid DCs (PDCs), each applied in combination with antibod
-
ies to either FXIIIA or CD11c. Neither FXIIIA nor CD11c colocal
-
ized with the LC markers CD1a (Figure 2, A and B) and Langerin 
(Supplemental Figure 1, A and B; supplemental material available 
online with this article; doi:10.1172/JCI32282DS1), nor the PDC 
markers CD123 (Figure 2, C and D) and BDCA-2 (Supplemental 
Figure 1, C and D). Therefore, FXIIIA
+
and CD11c
+
cells are dis-
tinct from additional LCand PDC populations innormal skin. 
Table 1 summarizes the expression of various leukocyte markers 
on the FXIIIA
+
 and CD11c
+
 cells.
CD11c
+
cells coexpress the blood DC marker BDCA-1, and a small
fraction also express DC-lysosomal–associated membrane glycoprotein/
CD208 and DEC-205/CD205.BDCA-1also marks apopulation 
of CD11c
+
 myeloid DCs in blood, and we noted coexpression of 
these 2markersinthe dermis as well (Figure 3, Aand B).Two 
othermarkersthatareexpressed bytissueDCsarelysosomal 
marker DC-lysosomalassociated membrane glycoprotein/
CD208 (DC-LAMP/CD208)and endocyticreceptorDEC-205/
CD205.A smallfractionof theCD11c
+
cells expressedthese 2 
markers (Figure 3, C–F). The fractionof CD11c
+
DC-LAMP
+
 cells 
was qualitatively smaller than the fraction of CD11c
+
DEC-205
+
cells.DC-LAMP/CD208is expressedduringDCmaturation, 
andwenoted thatcellspositiveforthismarkerwereoftenin 
dermalaggregateswithotherCD11c
+
DC-LAMP
cells.These 
results indicatethatCD11c
+
cells in thedermis sharefeatures 
with the myeloid DCs in blood, but some are in a more mature 
state of differentiation (11).
FXIIIA
+
cells express the macrophage marker and scavenger receptor
CD163. CD163, a hemoglobin/haptoglobin complex scavenger 
receptor,identifies tissueresident  macrophages, anditwas 
the only marker we studied that was uniformly coexpressed by 
FXIIIA
+
cells andnotby CD11c
+
cells (Figure4, Aand B).Sev-
eral other markers were expressed on a fraction of FXIIIA
+
 and 
CD11c
+
 cells. These included the uptake receptors macrophage 
mannosereceptor/CD206(MMR/CD206; Figure4,CandD), 
DC-specific ICAM-3–grabbing nonintegrin/CD209 (DC-SIGN/
CD209; Figure 4,E and F), CD45, andHLA-DR (Figure 5). In nor
-
mal skin, both MMR/CD206 and DC-SIGN/CD209 were more 
abundantonmacrophages(FXIIIA
+
cells) than DCs(CD11c
+
cells), which is consistent with recent studiesin inflamed skin 
andlymphnodes(11,12). Reciprocally,  CD45andHLA-DR 
were more abundant on CD11c
+
 than on FXIIIA
+
 cells. All these 
observations are consistentwith the interpretation that CD11c
+
cells in the dermis arepart of the DC pathway of differentiation, 
while FXIIIA
+
 cells are more macrophage-like.
Other markers of potential interest. Other markers used for char-
acterization of FXIIIA
+
and CD11c
+
cells wereCD14 (identifies 
monocytes); CD68, CD11b,and RFD7 (commonly used macro
-
phage markers); and CD63 (present onmacrophages and DCs). 
These markers were not helpful in discriminating these 2 popula
-
tions innormal skin (Supplemental Figure 2). CD14,a compo
-
nent of the LPS receptor on monocytes (13), was present on a few 
FXIIIA
+
cells and CD11c
+
cells. We have previously shownthat 
CD68 identifies a subset of CD11c
+
cells that produce IL-20 in 
psoriasis (14), so this is not an exclusive macrophage marker. In 
keeping with this, CD68 also stained both CD11c
+
andFXIIIA
+
Figure 1
FXIIIA
+
and CD11c
+
cells are unique dermal populations. (A) Immunohistochemistry on normal human skin using FXIIIA (left panel) and CD11c
(right panel) antibodies (n = 15). FXIIIA
+
cells were spread throughout the dermis, while CD11c
+
cells were mainly localized to the superficial
dermis. (B) There were similar numbers of CD11c
+
and FXIIIA
+
cells per mm in normal dermis. Error bars indicate SEM. (C) FXIIIA and CD11c
identified 2 discrete populations. White lines denote dermo-epidermal junction. Scale bars: 100 μm.
research article
The Journal of Clinical Investigation http://www.jci.org  Volume 117    Number 9  September 2007  2519
cells. CD11b and RFD7, considered to be common macrophage 
markers (13, 15), were present on both CD11c
+
 and FXIIIA
+
 cells. 
CD63, an MHC class II internalization antigen, marked occasional 
CD11c
+
 and FXIIIA
+
 cells in normal skin.
Isolated CD11c
+
dermal cells contain nonoverlapping major and
minor populations of BDCA-1
+
and BDCA-3
+
cells. To confirm our in 
situdataandassess whether the 3 myeloidDC subsets found 
in blood were also foundin tissue, weperformed 6-color FACS 
on single-cellsuspensions of normal human skin compared with 
peripheral blood, using a custom-madegroup ofmonoclonal 
antibodies to lineage (Lin) that did not containmonoclonalanti
-
bodies to CD14orCD16.PeripheralbloodFACS dot plots are 
shown in Figure 6, Aand B, and the parallelanalysisfrom dermal 
cells is showninFigure6,C and D. Therewere3 discrete Lin
CD11c
+
HLA-DR
+
DC populations in peripheralblood, BDCA-1
+
, 
CD16
hi
, and BDCA-3
+
(Figure 6A), but only 2 clear populations in 
skin, BDCA-1
+
CD16
lo
and BDCA-3
+
CD16
lo
(Figure 6C). In blood, 
2discrete CD11c
+
DC  populations,  BDCA-1
+
and BDCA-3
+
, 
werebothHLA-DR
+
CD14
(Figure6B).Incontrast, BDCA-1
+
cells fromskin increased HLA-DRexpression, and BDCA-3
+
cells 
acquiredlow-levelCD14 expression (Figure 6D). Wealsocon
-
firmed that BDCA-1 and BDCA-3 identified discrete populations 
in normal skin in situ (data not shown).
BDCA-1 and CD163 are superior markers to CD11c and FXIIIA,
respectively, for FACS of isolated dermal cells.To begin totest the 
functional properties of cell populations isolated from dermis, 
werequiredmarkersthatwouldbeoptimalfor FACS.While 
culture conditions may alter cellular surface phenotype, it was 
important to compare DC populations using the new BDCA-1 
andCD163markers with previousstudiesondermalDCs.It 
was  necessarytobalance obtainingsufficientcellstostudy 
with as little manipulation as possible. An overnight culture in 
dispase/collagenase media (16) yielded insufficient cells to per
-
form our MLR experiments. Enzymic alteration of surface epi
-
topes is also possible, although experimentson PBMCs did not 
show loss of FACS staining after culture in dispase collagenase 
Figure 2
FXIIIA
+
and CD11c
+
populations are not LCs or plasmacytoid
DCs. Neither FXIIIA nor CD11c showed coexpression with
Langerhans antigen CD1a (A and B) or plasmacytoid antigen
CD123 (C and D). Scale bar: 100 mm.
Table 1
Characterization of CD11c
+
and FXIIIA
+
cells in normal human
dermis
Antigen Main cell location FXIIIA CD11c
CD1a LCs, IDECs
Langerin/CD207 LCs
CD123 PDCs
BDCA-2 PDCs
BDCA-1 Myeloid DCs +++
DC-LAMP/CD208 Mature DCs +
DEC-205/CD205 Mature DCs +
CD163 Macrophages +++
MMR/CD206 Macrophages, DCs +++ ++
DC-SIGN/CD209 Macrophages, immature DCs +++ ++
CD45 Bone marrow–derived cells + +++
HLA-DR Antigen-presenting cells + ++
CD14 Monocyte-derived cells + +
CD68 Macrophages, DCs + +
CD11b Myeloid cells + ++
RFD7 Macrophages, DCs ++ +
CD63 Macrophages, DCs + +
Qualitative coexpression of FXIIIA and CD11c with additional antibody
is denoted as follows: –, no coexpression; +, some cells show coexpres-
sion; ++, a moderate number of cells show coexpression; +++, most
cells show coexpression. IDEC, inflammatory dendritic epidermal cell.
research article
2520 The Journal of Clinical Investigation http://www.jci.org    Volume 117    Number 9  September 2007
mix (data not shown). This method was modified to allow 1–2 
days ofculture so any lost surface markers could be reexpressed 
and to allow DCsand macrophagesto move out of the scored 
upper and lower dermal surfaces. Comparison ofBDCA-1 ver
-
sus CD163 on thefirst dayofculture(after overnight culture 
in dispase/collagenase) andafter24and 48 hours of culture 
showedasimilarclear distinctionofthe 2 populations(data 
notshown).Cultureofbulk  dermalcellsfromnormalskin 
using this method with and without DC maturation cytokines 
(IL-1β,IL-6,TNF,andPGE
2
)showed thattherewassurpris-
ingly little maturation of myeloid DCs (BDCA-1
+
) as measured 
by HLA-DR and CD83 (data not shown).
We foundthatBDCA-1,which colabeled morethan90% of 
CD11c
+
 cells by in situ immunofluorescence (Figure 3B), had a 
muchhighermedian fluorescence intensity(MFI) than did 
CD11c(data notshown). Likewise,CD163, whichshowed 
100% coexpression with FXIIIA in situ (Figure 4A), proved to 
be a much better FACS marker. FXIIIA antibodies bound non
-
specifically to dermal cells in suspension, perhaps as a result 
ofadherence ofplateletfragmentsrichin FXIIIA,whichis 
upregulated during cell culture (5, 6).
Double-label immunofluorescence of BDCA-1 and CD163 
confirmed  that they  were  distinct  populations in  situ 
(Figure 7A), which supports their use as alternative markers 
forCD11c andFXIIIA, respectively,inFACSstudies.Bulk 
dermal single-cell suspensions showed distinctBDCA-1
+
 and 
CD163
hi
populationsbyFACS(Figure 7B).BDCA-1
+
gated 
cells had a higher MFIfor the CD11c than the CD163
hi
 popu-
lation (Figure 7C), most likely because of increased sensitiv
-
ity of FACS compared withimmunohistochemistry.CD163
hi
cells had ahigherMFIforFXIIIAthandidBDCA-1
+
cells 
(Figure7C).BDCA-1
+
gatedcells also hada higherMFI for 
HLA-DR and CD45 thandid CD163
hi
 gated cells (Figure 7C). 
A subsetof BDCA-1
+
cells waspositivefor DCmaturation 
markersCD86, CD83,and DC-LAMP/CD208(Figure7D). 
Because B cells may also express BDCA-1, we confirmed that 
noneoftheBDCA-1
+
cellswere CD19
+
byFACS (datanot 
shown).Thusthedermal cells have a pattern of expression 
similar to in situ characterization.
BDCA-1
+
cells are a major immunostimulatory population from
normal human skin. To test for the immunostimulatory prop-
erties of dermalleukocytes, we focused on FACS-sorted popu
-
lations of BDCA-1
+
 and CD163
hi
 cells released from the der-
mis with collagenase. Data are summarized in Supplemental 
Table3, andFigure8shows representativeFACSplotsof 
the MLR. The sorted cells were 99% pure compared with iso
-
type (Figure 8A). In the MLR induced by in vitro monocyte-
derived mature DCs, 63% of the surviving T cells had under
-
gone extensive proliferation at a stimulator/responder ratio 
of 1:100on day 8 after sorting (Figure 8B). In parallel cultures 
stimulated by BDCA-1
+
 cells, 9.1% of the T cells had prolifer-
ated (1:10 ratio), comparedwith2.1%of CD163
+
cellsand 
1.0%background T cellproliferation (Figure 8C). When these 
sorted BDCA-1
+
 and CD163
+
 populationswere cultured for 2 
days in a DC-maturing cytokine cocktail before setting up the 
MLR, the immunostimulatory capability of BDCA-1
+
cellswas 
increased to 25.2% (1:100 ratio), but the capacity of CD163
+
cellswas unchanged (2.2%; 1:250ratio,lowbecauseof low 
cell survival during the culture period; Figure 8D). BDCA-1
+
sortedcells were cultured for 2days without cytokines, and 
the supernatantfrom thesecellsafter culture also increased T 
cell proliferation (data not shown).
CD163
+
cells phagocytose large particles in a tattoo and have the struc-
tural features of macrophages. Ultrathin sections of a green dye tat-
too werecutfrom tattoo-bearingnormal skin. These sections 
confirmed that the dye was intracytoplasmic and located mostly 
in  cells  clusteredaroundbloodvessels(Figure9A).Electron 
microscopy of the tattoo revealed membrane-bound (Figure 9B, 
blue arrow) tattoo dye particles (red arrow) and microvillous pro
-
jections(greenarrow), confirming the identity of these cellsas 
macrophages. Immunohistochemistryusing BDCA-1 (Figure 9C) 
and CD163 (Figure 9D) showed that dye-laden cells were CD163
+
and notBDCA-1
+
, confirming that typical macrophages rather 
than DCs had ingested the pigment.
Figure 3
CD11c
+
cells are defined by BDCA-1, DC-LAMP/CD208, and DEC-205/
CD205. FXIIIA did not overlap with BDCA-1 (A), DC-LAMP/CD208 (C),
or DEC-205/CD205 (E). Most CD11c
+
cells coexpressed BDCA-1 (B).
Small subsets of CD11c
+
cells coexpressed DC-LAMP/CD208 (D) and
DEC-205/CD205 (F). Scale bar: 100 μm.
research article
The Journal of Clinical Investigation http://www.jci.org  Volume 117    Number 9  September 2007  2521
Discussion
DCs areimportantsentinelsof the cutaneous immune system, 
performing central rolesinboth theinnateandthe acquired 
immune systems.Previous  characterization ofDC  subsetsin 
human  dermis  has been  influenced  by  results with a rabbit 
polyclonal antibody to FXIIIA, which identifies dermal cells with 
a dendritic morphology (4, 17–20).Here wehavecharacterized 
populationsof cells within thenormaldermisand foundthat, 
surprisingly, cells expressing the CD11c integrin and the BDCA-1 
antigen-presenting molecule represent a distinct population func
-
tionally differentiatedalongthe DC pathway, whereas FXIIIA
+
cells are differentiated along the macrophage pathway. Whereas 
mouse skin contains approximately 1 CD11c
+
DC for every 5 mac-
rophages (21), the concentration of CD11c
+
DCs in human skin is 
much higher, closer to a 1:1 ratio.
CD11ccolocalized with several well-recognizedDC markers: 
BDCA-1, DC-LAMP/CD208, and DEC-205/CD205. BDCA-1, 
also known as CD1c(10),is an invariant MHCclassI–like 
antigen receptor molecule that recognizes lipids in mycobac
-
terial cell walls. BDCA-1 is found on immature and mature 
DCs, and also on a subset of B cells. BDCA-1, in the absence of 
the B cell markers CD19 and CD20, would therefore seem to 
be a valuable marker to compare dermal DCs in both normal 
skin (22)and inflammatory skindiseases(23).DC-LAMP/
CD208 is a lysosomal protein that specifically marks mature 
DCs (24). DEC-205/CD205 is a surface receptor that partici
-
pates in DC antigen endocytosis; its expression increases dur
-
ing maturation, and it has been previously demonstrated in 
normal human skin (25). The low frequency of CD11c
+
 DCs 
expressing these 2 antigens observed in normal skin is con
-
sistent with their expected immature DC status. CD11c
+
 cells 
also stained brightly withHLA-DR and CD45, confirming 
their antigen-presenting potential and bone marrow origin, 
respectively. The phenotype of BDCA-1
+
 cells from skin indi-
cates that they are all CD45
hi
HLA-DR
hi
 and that a subset of 
these cells is CD86
+
 and CD83
+
.
Our studiesshow that FXIIIA identifiesa populationof 
tissue-resident macrophages in normal skin. The only anti
-
body that overlapped completely with FXIIIA in situ wasthe 
scavenger receptor CD163, which is selectively expressed on 
monocytes and macrophages (reviewed in ref. 26). The best-
characterized functionof CD163istobindhemoglobin/
haptoglobin complexes, which maybe important in homeo
-
stasis. Therewas low-levelCD45and HLA-DR expression, 
consistent with other tissue macrophages, and limited anti
-
gen-presenting capacity (27). MMR/CD206 and DC-SIGN/
CD209 areC-type lectin receptorsthatare foundonboth 
macrophages and DCs (11, 28), so it was not surprising that 
there was expression of both lectin receptors on both FXIIIA
+
and CD11c
+
 cells in normal human skin.
To evaluate the function of these 2 dominant dermal popu
-
lations, weused 2 approaches. By comparingBDCA-1and 
CD163 cells selected from collagenase digests of normal skin 
asinducersof an allogeneic MLR,we found thatBDCA-1
+
cells were the main immunostimulatory population. These 
BDCA-1
+
cells inducedincreased Tcell proliferation when 
cultured before setting up the MLR, but were not as immuno
-
stimulatory as in vitro–matured DCs, suggesting that there 
werefewmature DCs innormal skincomparedwith skin 
under inflammatory conditions such as psoriasis. This result 
reflects studies by Nestle et al. showing that bulk tissue émigrés 
from normal skin are not as stimulatory as those from patients 
with psoriasis (20). In comparison, CD163
hi
 cells were not immu-
nostimulatory in an allo-MLR, nor were they induced to be stimu
-
latory, although they may possess some antigen-presenting capac
-
ity with upregulation of MHC class II molecules (29).
Skin tattoos provided a second functional study: the phagocytic 
activityof tissue macrophages.Pigment granules were found in 
lysosomal-like cellular structures, and cells containing pigment 
stained uniformly with CD163. The fate of tattoo pigment inject
-
ed into dermal tissues has been studied in the past, and fibroblasts 
were considered to be the primary long-term reservoir of the pig
-
ment granules, with pigment in occasional macrophages (30, 31). 
However, our data suggest that macrophages are indeed a signif
-
icant storeof the dermalpigment.Thecells with pigment were 
Figure 4
The macrophage marker CD163 defines FXIIIA
+
cells. FXIIIA
+
cells
expressed macrophage marker CD163 (A), and a subset overlapped with
MMR/CD206 (C) and DC-SIGN/CD209 (E). CD11c did not overlap with
CD163 (B), but a subset overlapped with MMR/CD206 (D) and DC-SIGN/
CD209 (F). Scale bar: 100 μm.
research article
2522 The Journal of Clinical Investigation http://www.jci.org    Volume 117    Number 9  September 2007
CD163
+
and BDCA-1
, wereround, had numerous microvillous 
projections, andthepigment was containedwithin membrane-
bound structures (lysosomes). These characteristics are consistent 
with macrophages (31, 32).
Insummary, we have identified CD11c
+
cellsas immature myeloid 
DCs in normalhuman skinand FXIIIA
+
cells astissue-resident mac-
rophages, not DCs, asthey were previously classified. In future stud
-
ies of cutaneous DCs, we recommend considering the use of 
CD11c/BDCA-1 and CD163 as alternative markers to identify 
dermal DCs and macrophages, respectively, because they are 
morespecific and more useful in flow cytometry applications.
Methods
Skin samples. Skin punch biopsies (6 mm diameter) were obtained 
from 15 normal volunteers under a protocol approved by the Rock-
efeller University Institutional Review Board.Informedconsent 
was obtained. Biopsies were frozen in OTC (Sakura) and stored at 
–80°C for immunohistochemistry and immunofluorescence. Nor-
mal samples from abdominoplasty were processed within 4 hours 
after surgery. These abdominoplasty samples were also the source 
of tattoo material (n = 2).
Immunohistochemistry. Normal skin sections (n = 15) were stained 
with purified mouse  anti-human CD11c  (diluted 1:100)  and 
sheep affinity-purified anti-human FXIIIA (diluted 1:100; Enzyme 
Research Laboratories).Biotin-labeled horse anti-mouse and biotin-
labeled rabbit anti-sheep antibodies (VectorLaboratories)were used 
to detect CD11c and FXIIIA antibodies, respectively. The staining 
signal was amplified withavidin-biotin complex(Vector Labora-
tories) and developed using chromogen 3-amino-9-ethylcarbazole 
(Sigma-Aldrich). The number of positive cells per mm was counted 
manually using computer-assisted image analysis (NIH Image 6.1; 
http://rsb.info.nih.gov/nih-image). For FXIIIA immunostaining, 
several additional antibodies were screened, including AC-1A1 (Lab-
vision), E5014 (Spring Bioscience), and E980.1 (Biogeniex). We chose 
the sheep affinity-purified antibody becauseit gavethe most specific 
cellular staining with the least background epidermal staining.
Antibodies. All antibodies used for immunofluorescence and FACS 
of normal tissue are listed in Supplemental Tables 1 and 2.
Immunofluorescence. Normal skin sections (n = 6) were fixed with 
acetone and blocked in 10% normal goat serum (Vector Laborato-
ries) for 30 minutes.Primary antibodies CD11c and FXIIIA were incu-
batedovernight at 4°C andamplifiedwith theappropriate secondary 
antibody: goatanti-mouse IgG1 conjugatedwith Alexa Fluor 488 or 568, 
or donkey anti-sheepconjugated with AlexaFluor 488 or 568, respective-
ly. For colocalization with CD11c or FXIIIA, sections were then costained 
overnight with a secondantibody, as listed in Supplemental Table 1, and 
amplified with theappropriategoat-antimouse secondaryantibody 
Figure 5
HLA-DR and CD45 mark both CD11c
+
and FXIIIA
+
cells. FXIIIA
+
cells had a
lower expression level of CD45 (A) than did CD11c
+
cells (B). HLA-DR was
also expressed to a lower extent on FXIIIA
+
cells (C) than on CD11c
+
cells
(D). Scale bar: 100 μm.
Figure 6
Cutaneous DCs compared with blood
DCs. (A) There were 3 nonoverlapping
DC populations in peripheral blood,
gating on Lin
CD11c
+
HLA-DR
+
cells:
BDCA-1
+
, CD16
hi
, and BDCA-3
+
. (B)
Blood BDCA-1
+
(left) and BDCA-3
+
(right) cells were both HLA-DR
+
CD14
.
(C) In dermal single-cell suspensions,
BDCA-1
+
cells acquired CD16. (D)
BDCA-1
+
(left) dermal cells increased
HLA-DR expression, and BDCA-3
+
(right) dermal cells acquired low-level
CD14 expression.
research article
The Journal of Clinical Investigation http://www.jci.org  Volume 117    Number 9  September 2007  2523
(Supplemental Table 1). Images were acquired using appropriate filters 
of a Zeiss Axioplan2Imicroscope with Plan Apochromat 20 ×0.7 numer-
ical aperture lens and a Hagamatsu orca ER-cooled charge-coupled device 
camera, controlled by METAVUE software (Universal Imaging). Dermal 
collagen fibers gave green autofluorescence. Antibodies conjugated with 
a fluorochrome often gave background epidermal fluorescence. Images 
ineachfigure are presented assingle-color stainsofgreen andred to 
make apparent the localization of 2 markers on similar or different cells. 
Merged images are shown below the single-color stains. Cells that coex-
press the 2 markers in a similar location are often yellow in color.
Figure 7
BDCA-1 and CD163 are alternative markers for
CD11c and FXIIIA, respectively. (A) BDCA-1 and
CD163 identified discrete populations of dermal
cells. (B) BDCA-1
+
cells (red circle) and CD163
+
cells
(blue circle) were also discrete populations in dermal
single-cell suspensions. (C) FACS histograms gated
on BDCA-1
+
cells (red line), CD163
+
cells (blue line),
or isotype (green line). BDCA-1
+
cells were CD11c
hi
,
FXIIIA
lo
, HLA-DR
hi
, and CD45
hi
. CD163
+
cells were
CD11c
mid
, FXIIIA
hi
, HLA-DR
mid
, and CD45
lo
. (D) A
subset of BDCA-1
+
cells was CD86
hi
, CD83
+
, and
DC-LAMP
hi
. Representative graphs from 3 experi-
ments. Scale bar: 100 μm.
Figure 8
BDCA-1
+
cells are more immunostimulatory. (A) Post-
sort dot plot of dermal cells from normal skin into BDCA-1
+
and CD163
+
populations (red, left and right panels,
respectively) compared with isotype (blue). (B) Posi-
tive control (monocyte-derived mature DC) for MLR on
day 8 after sorting at a 1:100 stimulator/responder ratio.
Gate contains CD3
+
proliferating T cells with left-shifted
CFSE. (C) Using BDCA-1
+
sorted cells as stimula-
tors (1:10 ratio), 9.1% of the T cells proliferated; using
CD163
+
cells (1:10 ratio), 2.1% of live T cells prolifer-
ated. Background proliferation of T cells alone without
stimulation was 1.0%. (D) After cells had been sorted
and cultured for 2 days with cytokines to induce matura-
tion, there was a marked increase in the T cell stimula-
tory capacity of BDCA-1
+
cells (25.2%, 1:100 ratio) ver-
sus CD163 (2.2%, 1:250 ratio). Representative graphs
from 3 experiments.
research article
2524 The Journal of Clinical Investigation http://www.jci.org    Volume 117    Number 9  September 2007
Electron microscopy. Skin was fixed in 2.5% glutaraldehyde and processed 
by routine transmission electron microscopy procedure. Semithin plastic 
sections were stained with toluidine blue for light microscopic evaluation. 
Ultrathin (65-nm) sections were cut with a diamond knife, collected on 
copper grids, and stained with both uranylacetate and lead citrate before 
viewing in a Tecnaispirit electron microscope (FEI Company) equipped 
with an Ultrascan 895 charge-coupled device camera (Gatan).
Skin sample processing. Dermal single-cell suspensionsfrom normal skin 
were obtained using a modified collagenase/dispasemethod (16). Subcu-
taneous fat was excised, and remaining tissue was washed with PBS. The 
dermal layer was heavily scored with a scalpel and incubated in 1 mg/ml 
type1 collagenase (Invitrogen), 1 mg/mldispase (Invitrogen),and 1% 
penicillin-streptomycin solution(Sigma-Aldrich)overnightat 3C. 
The epidermis was peeled off and discarded, and the dermis was trans-
ferred to fresh RPMI1640 supplemented with 10% pooledhumanserum 
(Mediatech Inc.), 0.1% gentamicin reagent solution (Invitrogen), and 1% 
1 M HEPESbuffer(Sigma-Aldrich).The dermis wasincubated 24–48 
hours at 37°C,and the supernatant wascollected and filtered with 40-μm 
cell strainers (BD Biosciences). Cells were then either used immediately 
(for MLR;n= 3) or frozen in RPMI1640 (Invitrogen)and10% DMSO 
(ATCC) for FACS (n = 3).
FACS. Cells were stained with the antibodies listedinSupplemental 
Table2.Briefly, cellswere stained for20 minutesatC, washed with 
FACSwash (PBS 0.1% sodium azide and 2% FBS), and resuspended in 1.3% 
formaldehyde (Fisher Scientific) in FACSwash. To detect DC-LAMP, cells 
were first stained for surface markers, then permeabilized (FacsPerm; BD) 
before intracellular staining. Samples were acquired using FACSCanto or 
LSR-II (both from BD Biosciences) and analyzed with FlowJo (Treestar). 
Appropriate isotypes were used.
FACS and MLR. Dermal cells from single-cell suspensions of normal skin 
were stained withBDCA-1, CD19, andCD163 antibodyandsortedon 
a FACSAria (BD Biosciences) using a low-pressure setting. Two popula-
tions were obtained: BDCA-1
+
CD19
 and CD163
+
. A post-sort collection 
was performed to confirm the purity of each stimulating population. For 
some experiments, sorted cells were cultured for 2 days with and without 
cytokines for maturing DCs ex-vivo (IL-1β, IL-6, TNF-α, and PGE
2
), then 
washed and prepared for the MLR.
Responding T cells were obtained from a normal volunteer by density 
centrifugation over Ficoll-Paque Plus (Amersham Biosciences), followed 
by T cell purification using a T cell–negative selection kit (Dynal). T cells 
were labeled with 10μm CFSE and cocultured with either BDCA-1
+
or 
CD163
+
 sorted cells at ratios of 1:10, 1:50, and 1:100 (depending on cell 
yield). Tcells withouta stimulator populationwere used as a negative 
control, andmonocyte-derivedmature DCswereadded  to  Tcellsas 
a positivecontrol. The process for making mature DCs was previously 
described (33). T cell proliferation was analyzed on day 8 after sorting. The 
cultures were harvested, stained with 250 ng/ml propidium iodide (PI) to 
label dead cells, and CD3-APC (BD) for 15 minutes at room temperature. 
PI-negative cells were gated and then plotted as CFSE versus CD3
+
 cells, 
where proliferating cells diluted their content of CFSE and moved to the 
left of the nonproliferating cells. The CFSE-low cells were quantified as a 
percentage of live cells in the culture (34).
Statistics. A 2-tailed paired Student’s ttestwas usedto compare CD11c and 
FXIIIA
+
 cells in normal skin sections. Results are shown as mean ± SEM. 
A Pvalue less than 0.05 was considered significant.
Acknowledgments
This research was supported byNIH grants R01AI-49572, AI-49832, 
UL1 RR024143, and AI40045 (R.M. Steinman). M.A. Lowes is sup
-
ported by NIH grant 1 K23 AR052404-01A1, and L. Zaba is sup
-
ported by NIH MSTP grant GM07739. We thank plastic surgeons 
A.N.LaBruna and D.M.Senderoff for their generous donation 
ofabdominoplastysurgical waste,H.Shioand A.Khatcherian 
for their technical  assistance  on  electron  microscopy  and 
immunohistochemistry of the tattoo, respectively, and A. Piperno 
for the kind gift of monoclonal antibody DEC-205/CD205.We 
also appreciate the assistance and advice of the Flow Cytometry 
CoreFacility (S. Mazel) andBio-imaging Resource Center (A. 
North) at Rockefeller University. We thank Patricia Gilleaudea and 
Mary Whalen-Sullivan for excellent care of our patients.
Received for publication March 7, 2007, and accepted in revised 
form June 6, 2007.
Address correspondenceto: Michelle A. Lowes,Laboratoryfor 
Investigative Dermatology, The Rockefeller University, 1230 York 
Avenue, New York, New York 10021, USA. Phone: (212) 327-7576; 
Fax: (212) 327-8353; E-mail: lowesm@rockefeller.edu.
Figure 9
CD163
+
cells phagocytose large particles and have the structural fea-
tures of macrophages. (A) Tattoo skin section (0.5 μm) stained with
toludine blue. Cells containing green tattoo dye in their cytoplasm
(black arrow) surrounded a blood vessel. (B) Electron microscopy of
a tattoo showed that dye particles (red arrow) were membrane bound
(blue arrow) within the cytoplasm of a cell with multiple microvillus pro-
trusions (green arrow). (C and D) Cells containing green tattoo dye
particles stained for CD163 (D) but not BDCA-1 (C). Scale bar: 10 μm
(A, C, and D); 200 nm (B).
research article
The Journal of Clinical Investigation http://www.jci.org  Volume 117    Number 9  September 2007  2525
1. Larregina, A.T., and Falo, L.D. 2005. Changing par-
adigms in cutaneous immunology: adapting with 
dendritic cells. 
J. Invest. Dermatol.124:1–12.
2. Valladeau, J., and Saeland, S. 2005. Cutaneous den-
dritic cells. Semin. Immunol.17:273–283.
3. Hunger, R.E., et al. 2004. Langerhans cells utilize 
CD1a and langerin to efficiently present nonpep-
tide antigens to T cells. 
J. Clin. Invest.113:701–708. 
doi:10.1172/JCI200419655.
4. Cerio, R., Griffiths, C.E., Cooper, K.D., Nickoloff, 
B.J., and Headington, J.T. 1989. Characterization of 
factor XIIIa positive dermal dendritic cells innormal 
and inflamed skin. 
Br. J. Dermatol.121:421–431.
5. Henriksson, P., Becker, S., Lynch, G., and 
McDonagh, J. 1985. Identification of intracellular 
factor XIII in human monocytes and macrophages. 
J. Clin. Invest.76:528–534.
6. Torocsik, D., Bardos, H., Nagy, L., andAdany, R. 
2005. Identification of factor XIII-A as a marker of 
alternative macrophage activation. Cell. Mol. Life Sci.
62:2132–2139.
7. Freudenthal, P.S., and Steinman, R.M. 1990. The 
distinct surface of human blood dendritic cells, as 
observed after an improved isolation method. Proc.
Natl. Acad. Sci. U. S. A.87:7698–7702.
8. Summers, K.L.,Hock,  B.D.,McKenzie,J.L., and 
Hart, D.N. 2001.Phenotypic characterizationof 
five dendritic cell subsets in human tonsils. Am. J.
Pathol.159:285–295.
9. MacDonald, K.P.,etal.  2002.Characterization 
of human blood  dendritic cell subsets. 
Blood.
100:4512–4520.
10. Brigl, M.,and Brenner, M.B. 2004. CD1:antigen pre-
sentation and T cell function. Annu. Rev. Immunol.
22:817–890.
11. Granelli-Piperno, A., et al. 2005. Dendritic cell-spe
-
cific intercellularadhesion molecule3-grabbing 
nonintegrin/CD209 is abundant on macrophages 
in the normal  human  lymph  node and is  not 
required for dendritic cell stimulation of the mixed 
leukocyte reaction. 
J. Immunol.175:4265–4273.
12. Krutzik, S.R., et al. 2005. TLR activation triggers 
the rapid differentiation of monocytes into macro-
phages and dendritic cells. Nat. Med.11:653–660.
13. Gordon, S., and Taylor, P.R. 2005. Monocyte and 
macrophage heterogeneity.
Nat. Rev. Immunol.
5:953–964.
14. Wang, F.,et al. 2006. Prominent production of IL-20 
by CD68+/CD11c+ myeloid-derivedcells inpso
-
riasis: gene regulation and cellular effects. J. Invest.
Dermatol.126:1590–1599.
15. Tormey, V.J., et al. 1997. T-cell cytokines may con
-
trol the balance of functionally distinctmacro
-
phage populations. 
Immunology.90:463–469.
16. Angel, C.E., et al. 2006. Cutting edge: CD1a+ anti-
gen-presenting cells in human dermis respond rap-
idly to CCR7 ligands. J. Immunol.176:5730–5734.
17. Nestle, F.O., Zheng, X.G., Thompson, C.B., Turka, 
L.A., andNickoloff, B.J. 1993.Characterization 
ofdermal dendritic cellsobtained fromnormal 
human skin reveals phenotypic andfunctionally 
distinctive subsets. 
J. Immunol.151:6535–6545.
18. Meunier,  L.,Gonzalez-Ramos,  A., and Cooper, 
K.D.  1993. Heterogeneous populations ofclass 
II MHC+ cells in human dermal cell suspensions. 
Identificationof  asmall  subset  responsible  for 
potent dermal antigen-presenting cell activity with 
features analogous to Langerhans cells. 
J. Immunol.
151:4067–4080.
19. Deguchi, M., Aiba, S., Ohtani, H., Nagura, H., and 
Tagami, H. 2002. Comparison of the distribution 
andnumbers of antigen-presentingcells among 
T-lymphocyte-mediated dermatoses: CD1a+, factor 
XIIIa+, and CD68+ cells in eczematous dermatitis, 
psoriasis, lichen planusand graft-versus-host disease. 
Arch. Dermatol. Res.294:297–302.
20. Nestle, F.O., Turka, L.A., and Nickoloff, B.J. 1994. 
Characterization of dermal dendritic cells in pso
-
riasis. Autostimulation  of  T  lymphocytes and 
induction  of  Th1typecytokines.  J. Clin. Invest.
94:202–209.
21. Dupasquier, M., Stoitzner, P., van Oudenaren, A., 
Romani, N., and Leenen, P.J. 2004. Macrophages 
and dendritic cells constitute a major subpopula
-
tion of cells in the mouse dermis. J. Invest. Dermatol.
123:876–879.
22. Narbutt, J., et al. 2006. The number and distribu-
tion of blood dendritic cells in the epidermis and 
dermis of healthy human subjects. Folia Histochem.
Cytobiol.44:61–63.
23. Fivenson, D.P., and Nickoloff, B.J. 1995. Distinc
-
tive dendritic cell subsets expressing factor XIIIa, 
CD1a, CD1b and CD1c in mycosis fungoides and 
psoriasis. 
J. Cutan. Pathol.22:223–228.
24. de Saint-Vis, B., et al. 1998. A novel lysosome-asso
-
ciatedmembrane glycoprotein, DC-LAMP, induced 
upon DC maturation, is transiently expressed in 
MHC class II compartment. 
Immunity.9:325–336.
25. Ebner, S., et al. 2004. Expression of C-type lectin 
receptorsby subsets of dendritic cellsin humanskin. 
Int. Immunol.16:877–887.
26. Fabriek, B.O., Dijkstra, C.D., and van den Berg, T.K. 
2005. The macrophage scavenger receptor CD163. 
Immunobiology.210:153–160.
27. Janeway, C., Travers, P., Walport, M., and Shlom-
chik, M. 2004. Immunobiology: the immune system in
health and disease.6th edition. Garland Science. New 
York, New York, USA. 800 pp.
28. McGreal, E.P., Miller, J.L., andGordon, S. 2005. 
Ligand recognition by antigen-presenting cell C-type 
lectin receptors.Curr. Opin. Immunol.17:18–24.
29. Steinman, R.M., and Hemmi, H. 2006. Dendritic 
cells:translating innateto adaptiveimmunity. 
Curr. Top. Microbiol. Immunol.311:17–58.
30. Lea, P.J., and Pawlowski, A. 1987. Human tattoo. 
Electron microscopic assessment of epidermis,epi-
dermal-dermal junction, and dermis. 
Int. J. Dermatol.
26:453–458.
31. Fujita, H., Nishii, Y., Yamashita, K., Kawamata, S., 
and Yoshikawa, K. 1988. The uptake and long-term 
storage of Indiaink particles and latex beads by 
fibroblasts in the dermis and subcutis of mice, with 
specialregard to the non-inflammatory defense reac
-
tion by fibroblasts. Arch. Histol. Cytol.51:285–294.
32. Taylor,C.R., Anderson, R.R., Gange, R.W., Michaud, 
N.A., and Flotte, T.J. 1991.Light and electron micro-
scopicanalysis of tattoostreated by Q-switched 
ruby laser. 
J. Invest. Dermatol.97:131–136.
33. Lee, A.W., et al. 2002. A clinical grade cocktail of 
cytokines  and PGE2  results in  uniform  matu-
ration  of human monocyte-derived dendritic 
cells:implicationsforimmunotherapy.
Vaccine.
20(Suppl. 4):A8–A22.
34. Parish,  C.R., andWarren,H.S.2001. Use of  the 
intracellular  fluorescent  dye  CFSE  to  monitor 
lymphocyte migration andproliferation. In Cur-
rent protocols in immunology. John Wiley & Sons Inc. 
Hoboken, New Jersey, USA. Unit 4.9.1.
... While Langerhans cells are characterized by the expression of Langerin (3), to date, no specific marker exclusively expressed on all dermal dendritic subsets has been reported. However, dermal dendritic cells can be identified and distinguished from dermal monocytes and macrophages by a low CD14 expression and a high CD11c expression (4,5). Yet, the primary and common function of all cutaneous dendritic cell subsets includes endocytosis/phagocytosis, processing and presenting antigens to naïve T cells (6). ...
... The aim of this study was to explore and validate immature dendritic cells (iDCs) derived from the monocytic cell line THP-1 as suitable surrogates for dermal dendritic cells upon integration into a human full-thickness skin model. The ability of THP-1-derived iDCs to identify sensitizers such as NiSO 4 and DNCB and to upregulate the DC activation markers CD54 and CD86 has been recently shown (41). Furthermore, the capability to phagocytose pathogen-derived membrane components and to potentially induce T cell activation via upregulation of IL-12p40 upon sensitizer treatment has been proven (41). ...
... Subsequently, the next logically step was to prove whether our iDCs might be suitable surrogates for dermal dendritic cells. Dermal dendritic cells can be identified and distinguished from dermal monocytes and macrophages by a low CD14 expression and a high CD11c expression (4,5). However, in contrast to Langerhans cells, no exclusive cell-specific marker expressed on all dermal dendritic subsets has been reported so far. ...
Article
Full-text available
We have integrated dermal dendritic cell surrogates originally generated from the cell line THP-1 as central mediators of the immune reaction in a human full-thickness skin model. Accordingly, sensitizer treatment of THP-1-derived CD14⁻, CD11c⁺ immature dendritic cells (iDCs) resulted in the phosphorylation of p38 MAPK in the presence of 1-chloro-2,4-dinitrobenzene (DNCB) (2.6-fold) as well as in degradation of the inhibitor protein kappa B alpha (IκBα) upon incubation with NiSO4 (1.6-fold). Furthermore, NiSO4 led to an increase in mRNA levels of IL-6 (2.4-fold), TNF-α (2-fold) and of IL-8 (15-fold). These results were confirmed on the protein level, with even stronger effects on cytokine release in the presence of NiSO4: Cytokine secretion was significantly increased for IL-8 (147-fold), IL-6 (11.8-fold) and IL-1β (28.8-fold). Notably, DNCB treatment revealed an increase for IL-8 (28.6-fold) and IL-1β (5.6-fold). Importantly, NiSO4 treatment of isolated iDCs as well as of iDCs integrated as dermal dendritic cell surrogates into our full-thickness skin model (SM) induced the upregulation of the adhesion molecule clusters of differentiation (CD)54 (iDCs: 1.2-fold; SM: 1.3-fold) and the co-stimulatory molecule and DC maturation marker CD86 (iDCs ~1.4-fold; SM:~1.5-fold) surface marker expression. Noteworthy, the expression of CD54 and CD86 could be suppressed by dexamethasone treatment on isolated iDCs (CD54: 1.3-fold; CD86: 2.1-fold) as well as on the tissue-integrated iDCs (CD54: 1.4-fold; CD86: 1.6-fold). In conclusion, we were able to integrate THP-1-derived iDCs as functional dermal dendritic cell surrogates allowing the qualitative identification of potential sensitizers on the one hand, and drug candidates that potentially suppress sensitization on the other hand in a 3D human skin model corresponding to the 3R principles (“replace”, “reduce” and “refine”).
... Consistent with prior observations in human skin (84), SSc saSE tissues contain greater numbers of CD163 + and CD206 + macrophages than HC tissues (8), which is also reflected in increased CD163 and CD206 surface marker levels in SSc tissues (Fig. S15). Flow cytometry analyses also confirmed that macrophages comprised ~5-10% of cells in the saSE tissue, which is consistent with reported cell proportions in human skin (6,85). ...
Preprint
Systemic sclerosis (SSc) is an autoimmune disease characterized by skin fibrosis, internal organ involvement and vascular dropout. We previously developed and phenotypically characterized an in vitro 3D skin-like tissue model of SSc, and now analyze the transcriptomic (scRNA-seq) and epigenetic (scATAC-seq) characteristics of this model at single-cell resolution. SSc 3D skin-like tissues were fabricated using autologous fibroblasts, macrophages, and plasma from SSc patients or healthy control (HC) donors. SSc tissues displayed increased dermal thickness and contractility, as well as increased alpha-SMA staining. Single-cell transcriptomic and epigenomic analyses identified keratinocytes, macrophages, and five populations of fibroblasts (labeled FB1 - 5). Notably, FB1 APOE-expressing fibroblasts were 12-fold enriched in SSc tissues and were characterized by high EGR1 motif accessibility. Pseudotime analysis suggests that FB1 fibroblasts differentiate from a TGF-beta-responsive fibroblast population and ligand-receptor analysis indicates that the FB1 fibroblasts are active in macrophage crosstalk via soluble ligands including FGF2 and APP. These findings provide characterization of the 3D skin-like model at single cell resolution and establish that it recapitulates subsets of fibroblasts and macrophage phenotypes observed in skin biopsies.
... Aberrant polarization of CD4 + T helper lymphocytes induce secretion of TNF-α, IFN-γ, IL-17, and IL-22, activating keratinocytes, thus amplifying inflammation in psoriatic plaques [23]. Cutaneous DCs and keratinocytes are able to perceive tissue damage derived from different sources, through receptors recognizing molecular patterns deriving from pathogens or host-derived molecules exposed following tissue damage [24][25][26][27]. Skin DCs consist of various populations located in the epidermis or dermis and respond to antigens and allergens with the production of chemokines, inflammatory cytokines, and biocidal molecules such as nitric oxide (NO). ...
Article
Full-text available
Human skin has long been known as a protective organ, acting as a mechanical barrier towards the external environment. More recent is the acquisition that in addition to this fundamental role, the complex architecture of the skin hosts a variety of immune and non-immune cells playing preeminent roles in immunological processes aimed at blocking infections, tumor progression and migration, and elimination of xenobiotics. On the other hand, dysregulated or excessive immunological response into the skin leads to autoimmune reactions culminating in a variety of skin pathological manifestations. Among them is psoriasis, a multifactorial, immune-mediated disease with a strong genetic basis. Psoriasis affects 2–3% of the population; it is associated with cardiovascular comorbidities, and in up to 30% of the cases, with psoriatic arthritis. The pathogenesis of psoriasis is due to the complex interplay between the genetic background of the patient, environmental factors, and both innate and adaptive responses. Moreover, an autoimmune component and the comprehension of the mechanisms linking chronic skin inflammation with systemic and joint manifestations in psoriatic patients is still a major challenge. The understanding of these mechanisms may offer a valuable chance to find targetable molecules to treat the disease and prevent its progression to severe systemic conditions.
... These data are supported by a study in humans showing that CD14+ cells were a transient population of monocyte-derived macrophages [50]. CD163 has been proposed to be a good marker for dermal macrophages, as it specifically identifies skin-specific macrophages that are not recently migrated monocytes [51]. Analysis of the location of these different mononuclear phagocyte populations in the dermis have shown that DCs can be found closer to the epidermis (around 0-20 μm beneath the dermo-epidermal junction) and macrophages are located deeper in the skin (around 40-60 μm beneath the dermo-epidermal junction) [52]. ...
Article
Full-text available
Skin is an organ with a dynamic ecosystem that harbors pathogenic and commensal microbes, which constantly communicate amongst each other and with the host immune system. The skin acts as a physical barrier to prevent the invasion of foreign pathogens while providing a home to the commensal microbiota. The harsh physical landscape of skin, particularly the desiccated, nutrient-poor, acidic environment, also contributes to the adversity that pathogens face when colonizing human skin. The paper review how skin contributes to barrier immunity. The review also discusses specialized immune cells that are resident in steady-state skin including mononuclear phagocytes, such as Langerhans cells, dermal macrophages and dermal dendritic cells in addition to the resident memory T cells.
... Pig skin contains a population of dermal macrophages with a phenotype CD172a + CD163 + SLA-DR −/lo , CD14 + , CD16 + , DC-SIGN/CD209 lo , MR/CD206 lo , CD1 −/lo [25,169]. A phenotypically similar population, MHC-II lo CD163 hi , has been described in human dermis [278]. CD163 is also expressed on two populations of dermal dendritic cells (DDCs): one CD163 hi , with CD163 levels similar to those of macrophages, and other CD163 lo . ...
Article
Full-text available
Besides its importance as a livestock species, pig is increasingly being used as an animal model for biomedical research. Macrophages play critical roles in immunity to pathogens, tissue development, homeostasis and tissue repair. These cells are also primary targets for replication of viruses such as African swine fever virus, classical swine fever virus, and porcine respiratory and reproductive syndrome virus, which can cause huge economic losses to the pig industry. In this article, we review the current status of knowledge on porcine macrophages, starting by reviewing the markers available for their phenotypical characterization and following with the characteristics of the main macrophage populations described in different organs, as well as the effect of polarization conditions on their phenotype and function. We will also review available cell lines suitable for studies on the biology of porcine macrophages and their interaction with pathogens.
... Moreover, the development and manufacturing of needle technologies have grown substantially, and understanding of the role of skin APCs has helped in the selection of devices for efficient immunisation. Explanations for the improvement of immune induction by ID vaccination include the targeting of specific APCs in the skin: Langerhans cells (LCs; CD1a + and CD207 + ) [19,20] in the epidermal layer and macrophage cells (CD163 + ), and dermal dendritic cells (DDCs; CD1c + ) in the dermis [21]. It has been shown that in vitro targeting of different APCs elicits multiple immune responses: CD4 T cells, CD8 T cells, and humoral responses [22,23]. ...
Article
Full-text available
For intradermal (ID) immunisation, novel needle-based delivery systems have been proposed as a better alternative to the Mantoux method. However, the penetration depth of needles in the human skin and its effect on immune cells residing in the different layers of the skin has not been analyzed. A novel and user-friendly silicon microinjection needle (Bella-muTM) has been developed, which allows for a perpendicular injection due to its short needle length (1.4-1.8 mm) and ultrashort bevel. We aimed to characterize the performance of this microinjection needle in the context of the delivery of a particle-based outer membrane vesicle (OMV) vaccine using an ex vivo human skin explant model. We compared the needles of 1.4 and 1.8 mm with the conventional Mantoux method to investigate the depth of vaccine injection and the capacity of the skin antigen-presenting cell (APC) to phagocytose the OMVs. The 1.4 mm needle deposited the antigen closer to the epidermis than the 1.8 mm needle or the Mantoux method. Consequently, activation of epidermal Langerhans cells was significantly higher as determined by dendrite shortening. We found that five different subsets of dermal APCs are able to phagocytose the OMV vaccine, irrespective of the device or injection method. ID delivery using the 1.4 mm needle of a OMV-based vaccine allowed epidermal and dermal APC targeting, with superior activation of Langerhans cells. This study indicates that the use of a microinjection needle improves the delivery of vaccines in the human skin.
... Moreover, development and manufacturing of microneedle technologies have grown substantially, an understanding of the role of skin APCs has helped in the selection of devices for efficient immunization. Benefits have been demonstrated by targeting specific APCs in the skin: Langerhans cells (LCs; CD1a + and CD207 + ) [15,16] in the epidermal layer and macrophage cells (CD163 + ), and dermal dendritic cells (DDCs; CD1c + ) in the dermis [17]. It has been shown that in vitro targeting of different APCs elicits multiple immune responses: ...
Thesis
Les stratégies vaccinales sont au cœur du développement d’un vaccin. Elles s’orientent autour de l’administration et de la formulation d’un antigène (Ag). Par exemple, l’environnement immunitaire de la peau est très riche en cellules présentatrices d’Ag (CPA) contrairement au muscle, ce qui la rend très prometteuse pour l’injection d’un vaccin. Cependant, la méthode conventionnelle Mantoux est difficile à utiliser, elle demande une injection très minutieuse, parallèle à l’épiderme. Les premiers travaux de cette thèse consistent à mettre en avant l’utilisation de nouvelles microaiguilles Bella-muTM, qui permet une application simple de la voie ID. Elle induit un ciblage des CPA de la peau avec l’activation des cellules de Langerhans et la prise en charge de l’Ag par les CPA du derme. Par ailleurs, la vaccination VIH est très complexe à cause de son extrême variabilité, ce qui bloque le développement d’un vaccin capable d’induire une réponse protectrice. Une formulation à base d’un peptide très conservé entre les souches formulées avec du Squalène montre une bonne qualité de réponse immunitaire adaptative avec la production d’anticorps neutralisant à large spectre contrairement à l’aluminium. Le deuxième projet de la thèse utilise la microscopie 3D pour étudier les différents éléments de la réponse immunitaire innée induits par ces deux adjuvants qui impactent la réponse humorale dans le ganglion. En conclusion, le choix des stratégies vaccinales tel que la voie d’administration et la formulation qui va déterminer les évènements de la réponse innée mise en place après l’injection qui impact la qualité de la réponse adaptative et donc l’efficacité du vaccin.
Article
Full-text available
Acne vulgaris, one of the most common skin diseases, is a chronic cutaneous inflammation of the upper pilosebaceous unit (PSU) with complex pathogenesis. Inflammation plays a central role in the pathogenesis of acne vulgaris. During the inflammatory process, the innate and adaptive immune systems are coordinately activated to induce immune responses. Understanding the infiltration and cytokine secretion of differential cells in acne lesions, especially in the early stages of inflammation, will provide an insight into the pathogenesis of acne. The purpose of this review is to synthesize the association of different cell types with inflammation in early acne vulgaris and provide a comprehensive understanding of skin inflammation and immune responses.
Chapter
The presentation of hematolymphoid and histiocytic diseases can vary widely between children and adults as can age-related differential diagnoses and the consequences of treatment. Providing up to date information and classification for diseases across the hematopoietic, lymphoid and histiocytic systems, the book also covers morphologic, immunophenotypic, cytogenetic, and molecular genetic features of routine and diagnostically challenging disorders in pediatric patients. Over 200 high-quality colour illustrations aid accurate diagnosis and correct interpretations when evaluating peripheral blood, bone marrow, lymph nodes and other tissues specimens. This comprehensive yet practical guide also discusses useful ancillary tests and introduces newly recognised entities identified in the current literature, such as GATA-2 haploinsufficiency, B-ALL with DUX4-IGH and ERG deletion, and ALK-positive histiocytosis. An online version of the book with expandable figures can be accessed on Cambridge Core, via the code printed on the inside of the cover.
Article
Full-text available
The fate of India ink particles and polystyrene latex beads injected into the dermis and subcutis of the skin of the auricle and back in mice was observed with the naked eye, light microscopy and electron microscopy. The tattoo patterns made by injected ink particles remained essentially unchanged for life as observed with the naked eye. India ink particles and latex beads were endocytosed by fibroblasts as well as macrophages in the dermis and subcutis. Numerous ink particles or small latex beads (0.22 micron in diameter) were packed into vacuoles 0.1-10.0 micron in diameter which occupied a large volume of the cytoplasm of the cell body and processes of fibroblasts, whereas numerous particles and larger beads (0.22 and 2.0 micron) were taken up into the cell body of macrophages in the vicinity. Most fibroblasts, characterized by long cell processes and well developed rough endoplasmic reticulum, are easily distinguished from macrophages, the latter being round or oval in shape, and having many lysosomes and numerous irregularly shaped microvillous projections. It is believed that fibroblasts taking up and storing the ink particles or latex beads move poorly and are almost fixed in the connective tissue: the tattoos therefore do not change markedly. It is emphasized that the uptake and long-term storage of ink particles and latex beads by the dermal and subcutaneous fibroblasts represent a specific non-inflammatory defense mechanism that protects the living body, without immune reactions, against injuries and invasions by non-toxic foreign agencies. The histiocyte, a term proposed by KIYONO (1914) for a fixed macrophage on the basis of his studies using vital dye staining, is considered to include, in addition to true macrophages, fibroblasts showing endocytotic activities for small foreign bodies such as acid dyes, ink particles, and latex beads.
Article
The relative contribution of dermal-derived immunocompetent cells to the overall immunologic response in skin has been hampered by the lack of appropriate isolation techniques. In this report, we provide a purification schema that reliably yields highly purified populations of dermal dendritic cells (DDC). These cells are motile, express high levels of class II MHC antigens that decorate their cytoplasmic dendritic processes, and lack numerous B cell, T cell, and natural killer cell antigens. Using a broad panel of 45 different antibodies, an extensive phenotypic analysis was completed, revealing distinctive profiles for subsets of DDC. Despite homogeneous light scatter profile and cytologic appearance, three subsets of DDC could be distinguished by phenotypic and functional criteria. All DDC, but not epidermal Langerhans cells, express factor XIIIa. By triple color cell staining the relative distribution of factor XIIIa positive DDC is as follows: subset 1, 65% to 70% of total DDC express neither CD1a nor CD14; subset 2, 15% to 20% of total DDC express CD1a but not CD14; and subset 3, 10% to 15% of total DDC express CD14 but not CD1a. The CD14-negative subset of DDC were shown to be as potent stimulators of allogeneic mixed lymphocyte reactions as Langerhans cells or blood-derived dendritic cells. However, DDC subsets differed in their ability to support autologous T cell proliferation in response to the mitogenic lectin PHA or bacterial-derived superantigen. In these assays, subsets 1 and 2 were significantly more potent as antigen-presenting cells compared with subset 3. Thus, normal skin contains at least three separate populations of DDC, which have distinctive phenotypic markers and immunologic capabilities.
Article
Ultrathin serial sections of human biopsy specimens, taken at 24 hours, I month, and 1, 3, and 40 years post-tattooing were examined under the electron microscope. The ink particles found in cells were measured and compared with control ink particles embedded in agar. Freshly tattooed skin showed an inflammatory reaction followed by ultrastructural necrosis. The time of healing, about I month from introduction of ink to complete healing, hits been divided into three phases: inflammatory reaction and necrosis, format/on of basement membrane, and normal epidermis and dermis. Once the skin showed normal ultrastructure, ink particles were found only in dermal fibroblasts.
Article
The papillary dermis of psoriasis and mycosis fungoides (MF) lesions is characterized by prominent collections of cells with dendritic morphology. Immunophenotypically distinct populations of cutaneous dendritic cells have been identified as CD1a+, FXIIIa-Langerhans cells (LC) and CD1a-, FXIIIa+ dermal dendritic cells (DDC). In this study, antibodies against the human GDI cluster of antigens (i.e. CD1a, CD1b and CD1c) and the DDC) marker (FXIIIa) were used to further characterize the subsets of dendritic cells in normal skin as compared to neonatal foreskin, psoriasis and MF by both immunoperoxidase and double immunofluorescence techniques. Normal skin and foreskin epidermis and dermis contained few CD1b+ or CD1c+ cells along with normal numbers of CD1a+ LC and FXIIIa+ DDC. Both MF and psoriasis were characterized by CD1a+ cells in the epidermis and dermis. FXIIIa+ cells were greatly expanded in the upper dermis of MF lesions and to a lesser degree in psoriasis as has been previously described by our group. MF contained significantly increased epidermal and dermal CD1b+ (15.7/5 high power fields [HPF] and 59.7/5 HPF respectively) and CD1c+ dendritic cells (33.8/5 HPF and 95.9/5 HPF respectively), while in psoriasis these cells were not statistically different from normal skin. Double immunofluorescence studies revealed that some (<25%) FXIIIa+ cells co-expressed CD1b and CD1c in MF>psoriasis> foreskin, while FXIIIa+ DDC never co-expressed CD1a. Thus, in contrast to normal skin in which epidermal or dermal dendritic cells rarely express CD1b and CD1c antigens, these members of the CD1 family are upregulated on both LC and DDC in benign and malignant inflammatory states. Upregulation of CD1b and CD1c on MF epidermal and dermal dendritic cells, as compared to psoriasis, foreskin and normal skin, may be useful in the immunophenotypic recognition of MF, as well as in helping to understand its immunobiology.
Article
Short-pulse laser exposures can be used to alter pigmented structures in tissue by selective photothermolysis. Potential mechanisms of human tattoo pigment lightening with Q-switched ruby laser were explored by light and electron microscopy. Significant variation existed between and within tattoos. Electron microscopy of untreated tattoos revealed membrane-bound pigment granules, predominantly within fibroblasts and macrophages, and occasionally in mast cells. These granules contained pigment particles ranging from 2-in diameter. Immediately after exposure, dose-related injury was observed in cells containing pigment. Some pigment particles were smaller and lamellated. At fluences greater than or equal to 3 J/cm2, dermal vacuoles and homogenization of collagen bundles immediately adjacent to extracellular pigment were occasionally observed. A brisk neutrophilic infiltrate was apparent by 24 h. Eleven days later, the pigment was again intracellular. Half of the biopsies at 150 d revealed a mild persistent lymphocytic infiltrate. There was no fibrosis except for one case of clinical scarring. These findings confirm that short-pulse radiation can be used to selectively disrupt cells containing tattoo pigments. The physial alteration of pigment granules, redistribution, and elimination appear to account for clinical lightening of the tattoos.
Article
Prior studies have identified a subset of dendritic cells in human blood, as well as their stimulatory function for T-cell-mediated immune responses. However research has been limited by difficulties in isolation, since dendritic cells make up only 0.1-1% of blood mononuclear cells. We present a protocol that reliably yields preparations that are greater than 80-90% pure. The method relies on the sequential depletion of the major cell types in blood and simultaneously provides T cells, monocytes, and B plus natural killer cells for comparison with dendritic cells. The last step in the procedure is the removal of residual contaminants on the basis of expression of a CD45R epitope. The enrichment of dendritic cells is evident by three criteria, each of which is related to the surface of these antigen-presenting cells. (i) All dendritic cells are motile, constantly forming large lamellipodia or veils. (ii) When analyzed with a large panel of monoclonal antibodies and the FACS, the cells express high levels of all known polymorphic major histocompatibility complex gene products, as well as a distinct combination of receptors and adhesion molecules. Unlike monocytes, for example, dendritic cells lack Fc receptors and the colony-stimulating factor 1 receptor (c-fms) but express much higher levels of ICAM-1 and LFA-3 adhesins. (iii) In functional assays, dendritic cells are at least 100 times more potent than monocytes or lymphocytes in stimulating the primary mixed leukocyte reaction. These properties help make the trace subset of dendritic cells more amenable to further functional and clinical studies.
Article
The immunocytochemical identification and characterization of indigenous dermal dendritic cells (dermal dendrocytes) using a rabbit polyclonal antibody to clotting enzyme factor XIII subunit A (FXIIIa) was carried out on normal and inflamed human cutaneous tissue. The immunophenotype of FXIIIa positive dendritic cells was analysed with a panel of 18 monoclonal antibodies using immunoperoxidase and double immunofluorescence staining techniques. The antibody against FXIIIa detected highly dendritic dermal cells located particularly in the upper reticular and papillary dermis. Double fluorescence microscopy showed that FXIIIa positive cells were bone marrow derived (HLe-I+) and co-expressed monocyte, macrophage or antigen presenting cell markers (HLA-DR+, LFA-I+, HLA-DQ+, OKM5+, Mo I+, Mono-I+, Leu M3+). No labelling was obtained with cell markers for Langerhans cells (CDI), T lymphocytes (CD2), granulocytes (LeuMI) fibroblasts (Te7), intercellular adhesion molecule-I (ICAM-I) or endothelial cells (Factor VIII related antigen). Gamma interferon induced increased expression of HLA-DR and co-expression of ICAM-I on FXIIIa+ dermal dendritic cells in normal skin in organ culture. Moreover, in benign inflammatory dermatoses such as atopic eczema and psoriasis there was an increased number of FXIIIa+, DR+, ICAM-I+ cells in the upper dermis and foci of FXIIIa+ cells in the epidermis closely associated with lymphocytes. FXIIIa positive cells in human skin represent a specific population of bone-marrow dermal dendritic cells, distinct from Langerhans cells, that share some features common to mononuclear phagocytes (monocyte/macrophages). In addition, the detection of HLA-DQ on 48% of FXIIIa+ cells and the lack of OKMI in combination with high OKM5 expression suggests an antigen-presenting cell phenotype.