ArticlePDF Available

Dendritic cell ontogeny: A human dendritic cell lineage of myeloid origin. Proc Natl Acad Sci USA. 94, 12551-12556

Authors:

Abstract and Figures

Dendritic cells (DC) have been thought to represent a family of closely related cells with similar functions and developmental pathways. The best-characterized precursors are the epidermal Langerhans cells, which migrate to lymphoid organs and become activated DC in response to inflammatory stimuli. Here, we demonstrate that a large subset of DC in the T cell-dependent areas of human lymphoid organs are nonactivated cells and belong to a separate lineage that can be identified by high levels of the interleukin 3 receptor alpha chain (IL-3Ralphahi). The CD34+IL-3Ralphahi DC progenitors are of myeloid origin and are distinct from those that give rise to Langerhans cells in vitro. The IL-3Ralphahi DC furthermore appear to migrate to lymphoid organs independently of inflammatory stimuli or foreign antigens. Thus, DC are heterogeneous with regard to function and ontogeny.
Content may be subject to copyright.
Proc. Natl. Acad. Sci. USA
Vol. 94, pp. 12551–12556, November 1997
Immunology
Dendritic cell ontogeny: A human dendritic cell lineage of
myeloid origin
JOHANNA OLWEUS*,ANDREW BITMANSOUR*, ROGER WARNKE,PETER A. THOMPSON*, JOSE CARBALLIDO§,
LOUIS J. PICKER,AND FRIDTJOF LUND-JOHANSEN§
*Becton Dickinson Immunocytometry Systems, 2350 Qume Drive, San Jose, CA 95131; Department of Pathology, Stanford University, Stanford, CA 94305-5324;
§DNAX Research Institute, 901 California Avenue, Palo Alto, CA 94304; and Southwestern Medical Center, University of Texas, 5323 Harry Hines Boulevard,
Dallas, TX 75235-9072
Edited by Irving L. Weissman, Stanford University School of Medicine, Stanford, CA, and approved September 10, 1997 (received for review April
16, 1997)
ABSTRACT Dendritic cells (DC) have been thought to
represent a family of closely related cells with similar func-
tions and developmental pathways. The best-characterized
precursors are the epidermal Langerhans cells, which migrate
to lymphoid organs and become activated DC in response to
inflammatory stimuli. Here, we demonstrate that a large
subset of DC in the T cell-dependent areas of human lymphoid
organs are nonactivated cells and belong to a separate lineage
that can be identified by high levels of the interleukin 3
receptor
a
chain (IL-3R
a
hi
). The CD34
1
IL-3R
a
hi
DC progen-
itors are of myeloid origin and are distinct from those that give
rise to Langerhans cells in vitro. The IL-3R
a
hi
DC furthermore
appear to migrate to lymphoid organs independently of in-
flammatory stimuli or foreign antigens. Thus, DC are heter-
ogeneous with regard to function and ontogeny.
Dendritic cells (DC) in lymphoid organs are potent antigen-
presenting cells, which play an important role in the initiation
of immune responses (1). Studies showing that epidermal
Langerhans cells are precursors of DC have suggested that the
unique role of DC as ‘‘natures adjuvant’’ is linked to their
developmental pathway. Langerhans cells reside in the epi-
dermis where the cells are capable of antigen uptake but have
low ability for antigen presentation (2). In response to inflam-
matory signals, the cells migrate rapidly to lymphoid tissues
and differentiate into mature, activated DC with potent ability
for stimulation of T cells (3–6). Cells with characteristics of DC
precursors have also been found in other tissues (7), and such
cells also migrate in response to inflammatory mediators (6, 8).
Thus, DC in lymphoid organs have been widely considered to
represent the end stage of a stepwise differentiation and
migration process, which is completed during inflammation
and serves to initiate immune responses (9–12).
Presently, most of the knowledge about the developmental
pathway of DC is based on results obtained by cell culture. Cells
with characteristics of Langerhans cells and DC can be generated
in vitro by culture of CD34
1
cells in the presence of granulocytey
macrophage colony-stimulating factor (GM-CSF) and tumor
necrosis factor
a
(TNF-
a
) (13–19). Results from studies of in vitro
colony formation have further indicated that the GM-CSFyTNF-
a
-responsive progenitors represent a separate DC colony forming
cell (15). These and other observations have supported the view
that DC are a family of closely related cells that constitute a
distinct ‘‘DC lineage.’’ However, progenitors committed to be-
come DC have not yet been identified directly in bone marrow.
The interpretation of results obtained by colony assays and cell
culture is furthermore complicated by the fact that populations of
lymphoid progenitors, granulomonoc ytic progenitors and periph-
eral blood monocytes also assume characteristics of DC in vitro
(14, 18–22).
Primitive hematopoietic progenitors and cells committed to
become lymphocytes, monocytes, granulocytes, and erythroid
cells can be identified as discrete populations of freshly isolated
CD34
1
bone marrow cells using specific cell surface markers
(23–29). Similar characterization of DC progenitors has been
difficult due to the lack of selective markers that identify the cells
at an early stage of differentiation. In the present study, however,
we demonstrate that antibodies to the interleukin 3 receptor
a
chain (IL-3R
a
) selectively react with a large subset of DC in
lymphoid organs and identify their precursors in blood and bone
marrow. The CD34
1
IL-3R
a
hi
progenitors are of myeloid origin
but committed to become DC and distinct from those that give
rise to Langerhans cells. Unlike Langerhans cells, IL-3R
a
hi
DC
home to lymphoid tissue independently of inflammation or
stimulation with foreign antigens.
MATERIALS AND METHODS
Tissue. Tissue from aborted fetuses of gestational age 19–21
weeks was obtained from Advanced Bioscience Resources
(Alameda, CA), a nonprofit organization which provides tissue
in compliance with state and federal laws. Blood donor buffy
coats were obtained from the Stanford Blood Bank (Stanford,
CA). Tonsils and adult lymph nodes were obtained from the
tissue acquisition service and the clinical flow cytometry
laboratory, Department of Pathology, University of Texas
Southwestern Medical Center (Dallas).
Cell Preparation. Mononuclear cell suspensions were ob-
tained by Lymphoprep gradient centrifugation (Nycomed,
Oslo). Antibody-free CD3
1
CD4
1
T cells were isolated to 99%
purity from peripheral blood mononuclear cells (PBMC) using
CD4 Dynabeads in combination with Detachabead reagent
(Dynal, Oslo) after depletion of myeloid cells (CD14
1
and
CD36
1
) by Dynabeads (Dynal). Fetal bone marrow CD34
1
cells were isolated by positive immunomagnetic selection
(Miltenyi Biotech, Auburn, CA), as described (29). Where
noted, subsets of immunostained cells were sorted using a
FACSVantage flow cytometer (Becton Dickinson).
Immunophenotypic Analysis. Multicolor immunofluorescence
staining and analysis was performed by standard methods (see
ref. 29). Primary or secondary antibodies were conjugated to
biotin, fluorescein isothiocyanate (FITC), phycoerythrin (PE),
The publication costs of this article were defrayed in part by page charge
payment. This article must therefore be hereby marked ‘‘advertisement’’ in
accordance with 18 U.S.C. §1734 solely to indicate this fact.
© 1997 by The National Academy of Sciences 0027-8424y97y9412551-6$2.00y0
PNAS is available online at http:yywww.pnas.org.
This paper was submitted directly (Track II) to the Proceedings office.
Abbreviations: DC, dendritic cell; IL-3R
a
, interleukin 3 receptor
a
chain; GM-CSF, granulocyteymacrophage colony-stimulating factor;
G-CSF, granulocyte CSF; M-CSF, macrophage CSF; M-CSFR, M-
CSF receptor; TNF-
a
, tumor necrosis factor
a
; PBMC, peripheral
blood mononuclear cells; FITC, fluorescein isothiocyanate; PE, phy-
coerythrin; FACS, fluorescence-activated cell sorter.
To whom reprint requests should be sent at the present address: J. O.,
Buskerud Central Hospital, Dronninggt. 28, N-3004 Drammen, Nor-
way. e-mail: f-johans@online.no.
12551
peridinin-chlorophyll protein, or allo-phycocyanin. Antibodies
and streptavidin conjugates were from Becton Dickinson, except
CD40, CD86 PE, anti-IL-3R
a
biotin, and anti-IL-3R
a
PE
(PharMingen); CD64 (Meda Rex, West Lebanon, NH); anti-M-
CSFR (Santa Cruz Biotechnology); donkey anti-rat IgG PE,
goat-anti-mouse IgG PE, and goat-anti-human IgM FITC (Jack-
son ImmunoResearch). Cells were analyzed using a FACSCali-
bur flow cytometer (Becton Dickinson).
Media and Cytokines. Except where specifically noted, cells
were cultured in Yssel’s medium (30) supplemented with
heat-inactivated 10% pooled human AB
1
serum and 10% fetal
FIG. 1. Antibodies to IL-3R
a
selectively stain DC in extrafollicular
regions of human tonsils. (AC) Tonsillar mononuclear cells were
stained with anti-HLA-DR peridinin-chlorophyll protein (PerCP),
CD4 PE, anti-IL-3R
a
biotin 1Streptavidin allo-phycocyanin
(ALPC), and a mixture of FITC-conjugated lineage markers (‘‘lin’’)
for lymphocytes and monocytes (CD3, CD14, CD16, CD19, CD20,
Table 1. Expression of surface molecules on
HLA-DR
1
lin
2
IL-3R
a
hi
DC from mononuclear tonsillar cells,
PBMC, and fetal lymph node (LN) cells, and on CD34
1
IL-3R
a
hi
cells from fetal bone marrow (BM)
Tonsil Fetal LN PBMC Fetal BM
CD1a 22 2 2
CD3 22 2 2
CD4 11y111 11y111 11 11
CD5 22 2 2
CD11b 22 2 2
CD11c 22 2 2
CD13 2y11 2y11
CD14 22 2 2
CD15 2ND 22
CD16 22 2 2
CD19 22 2 2
CD20 22 2 2
CD32 11 1 1
CD33 11(1)1(1)1y11
CD34 2y11 2y1 11y111
CD36 11y111*11y111 11y111 11y1111
CD40 11 1 11 1
CD45RA 11(1)11(1)11(1)11
CD45RO 22 2 2
CD54 11y111 ND 11y111 1y11
CD56 22 2 2
CD58 1ND 1y11 1y11
CD62L 211y11 111
CD64 22 2 2
CD80 22 2 2
CD86 1ND 11(1)
HLA-DR 11 11 11 11
HLA-DQ 2ND 2y12
Cells were stained as described in legends to Figs. 1, 3, and 4. Mean
fluorescence intensity (MFI) levels for the IL-3R
a
hi
populations are
expressed as 2, indicating MFI in the first decade on a four log scale,
which corresponds to isotype control levels. The 1,11, and 111
indicate MFI in the second, third, and fourth decades, respectively.
Aysign means that MFI is on the border between two decades.
Parenthesis means that the MFI is in the upper end of a decade. ND,
not determined. The data are representative of at least three exper-
iments.
*In one out of three experiments tonsillar HLA-DR
1
lin
2
IL-3R
a
hi
DC
were negative for CD36.
CD56, and goat-anti-human IgM). The cells were analyzed by four-
color flow cytometry. Dendritic cells were identified as HLA-
DR
1
CD4
1
lin
2
—i.e., cells that simultaneously satisfy the criteria of
the box regions in Aand B, and are represented by large black dots.
Dashed lines represent isotype control levels. (D) Wright–Giemsa
staining of a cytocentrifuge slide of freshly FACS-sorted tonsillar
HLA-DR
1
lin
2
IL-3R
a
hi
cells. (3600.) (E) FACS-sorted tonsillar
HLA-DR
1
lin
2
IL-3R
a
hi
cells were cultured for 24 h with GM-CSF and
IL-3 and photographed in situ.(3400.) (F) A frozen section of tonsillar
tissue was stained with anti-IL-3R
a
, biotinylated anti-mouse IgG and
streptavidin peroxidase. (3100). Staining was visualized by diamino-
benzidine and hydrogen-peroxide, and the section was counterstained
with methylene blue.
12552 Immunology: Olweus et al. Proc. Natl. Acad. Sci. USA 94 (1997)
bovine serum. Recombinant human cytokines were used as
noted at the following concentrations: GM-CSF (10 ngyml),
IL-3 (10 ngyml), IL-6 (500 unitsyml), IL-7 (50 ngyml) (all from
Collaborative Biomedical Products, Bedford, MA), erythro-
poietin (2.5 unitsyml; CILAG, Schaffhausen, Switzerland),
stem cell factor (40 ngyml; Peprotech, Rocky Hill, NJ), gran-
ulocyte-CSF (G-CSF, 50 ngyml, Amgen Biologicals), and
macrophage-CSF (M-CSF, 10 ngyml;R&DSystems).
Cell Cultures. IL-3R
a
hi
lin
2
HLA-DR
1
cells from tonsil and
PBMC and CD14
hi
monocytes from PBMC were sorted by
fluorescence-activated cell sorter (FACS) and cultured for 36 h
before mixing with T cells to allow maturation of DC precursors.
GM-CSF and IL-3 were added to IL-3R
a
hi
lin
2
HLA-DR
1
cells to
enhance survival. Stimulator cells were washed twice before
coculture with CD4
1
T cells (10
5
per well) in flat-bottomed
96-well plates for 6 days. Bromodeoxyuridine (BrdU; Sigma) (50
m
M) was added 12 h before harvest latex particles (10
5
per well)
were added immediately prior to harvest as a reference for cell
counts (29). The cells were stained with CD3 PE, followed by
fixation, permeabilization, staining with anti-BrdU FITC, and
flow cytometric analysis, as described (29). Cocultures with
stimulator cells derived by culture of fetal bone marrow cells were
pulsed with 1
m
Ci (1 Ci 537 GBq) [
3
H]thymidine for 8 h before
collecting and counting.
RESULTS AND DISCUSSION
IL-3R
a
Is a Selective Marker for a Large Subset of DC in
T Cell-Rich Zones of Human Peripheral Lymphoid Organs.
Human tonsils contain DC that can be identified as cells that
lack lineage markers for monocytes and lymphocytes (lin
2
)
and are positive for HLA-DR and CD4 (HLA-DR
1
CD4
1
lin
2
)
(Fig. 1 Aand B) (31, 32). In an attempt to identify DC-selective
markers, we screened antibodies to leukocyte differentiation
antigens for selective reactivity with this population. Antibod-
ies to IL-3R
a
reacted strongly with more than 85% of HLA-
DR
1
CD4
1
lin
2
cells (0.32–0.37% of tonsillar mononuclear
cells, n53), but weakly with most other cells (n53) (Fig. 1C).
The staining was sufficiently specific to allow a 200-fold
enrichment of HLA-DR
1
CD4
1
lin
2
cells with a single positive
immuno-magnetic selection (n52). HLA-DR
1
lin
2
IL-3R
a
hi
cells were also found in adult cervical, axillar, intramammary,
mesenteric, and femoral lymph nodes. The frequency ranged
from 0.1%–1.8% (average 0.5%, n59). In all cases, cells
staining brightly with the anti-IL-3R
a
were found within the
HLA-DR
1
lin
2
population and constituted the majority of
these cells (on average 60.2 611.2%, n59).
Freshly sorted HLA-DR
1
lin
2
IL-3R
a
hi
cells showed an imma-
ture morphology without cytoplasmic protrusions (Fig. 1D). The
majority of the cells died rapidly in culture, but could be partially
rescued by the addition of cytokines (GM-CSF and IL-3). Under
these conditions, the cells rapidly formed large aggregates of cells
as previously reported for tonsillar DC (32) (Fig. 1E). After 3–5
days the cells were more dispersed and showed multiple long
processes characteristic of DC (data not shown).
IL-3R
a
hi
cells were found almost exclusively in the T
cell-rich extra-follicular regions of the tonsil (Fig. 1F). The
localization as well as the the cytokine requirements and
phenotype of the HLA-DR
1
lin
2
IL-3R
a
hi
cells (Table 1) sug-
gest that they are identical to the ‘‘plasmacytoid T cell’’ DC
that were recently characterized by Grouard et al. (33).
IL-3R
a
hi
DC Are Immature and Appear in Lymphoid Organs
Independently of Stimuli That Cause Up-Regulation of Major
Histocompatibility Complex (MHC) Class II and Costimulatory
Molecules. Originally, it was concluded that DC in lymphoid
tissues such as tonsils are activated, mature antigen-presenting
cells, because isolated cells expressed high levels of MHC class II
and costimulatory molecules (31, 32). However, in those studies
the cells were isolated after 1–2 days of cell culture. As shown in
Fig. 2Aand Table 1, HLA-DR
1
lin
2
IL-3R
a
hi
cells in fresh
preparations of tonsillar mononuclear cells expressed low levels
of CD80 (B7.1), CD86 (B7.2), and HLA-DQ. Overnight culture
of unseparated cells in the absence of cytokines was sufficient to
induce the mature phenotype (Fig. 2A). Sorted HLA-
DR
1
lin
2
IL-3R
a
hi
cells that were allowed to mature in culture for
36 h and kept viable with GM-CSF and IL-3 were potent
stimulators of allogeneic CD4
1
T cells (Fig. 2B). These results are
surprising in view of the hypothesis that DC migrate to lymphoid
organs in response to signals that lead to cell activation. However,
these data are in agreement with those of Grouard et al. (33), who
also studied freshly isolated cells, and with previous reports
FIG. 2. IL-3R
a
hi
DC are immature and appear in lymphoid organs
independently of stimuli that cause up-regulation of major histocompat-
ibility complex class II and costimulatory molecules. (A) Mononuclear
cells from tonsil were stained with anti-HLA-DR, anti-IL-3R
a
and
lineage markers, and either CD86, CD80, HLA-DQ, or isotype control
mAbs before (open bars) and after (filled bars) a 16-h incubation at 37°C
in Yssel’s medium (30). The HLA-DR
1
lin
2
IL-3R
a
hi
population was
analyzed for mean fluorescence intensit y (MFI) staining with the markers
indicated on the figure, and the bars represent MFI after isotype control
levels were subtracted. (B) T cells (10
5
) were cocultured with indicated
numbers of allogeneic IL-3R
a
hi
lin
2
HLA-DR
1
cells from tonsil (stimu-
lator cells). T cell proliferation was measured as the total number of CD3
1
BrdU
1
cells per well at day 6 of coculture. (C) Fetal ly mph node cells were
stained with anti-HLA-DR, anti-IL-3R
a
, and lineage markers (data not
shown) and analyzed by flow cytometry as described in Fig. 1 AC. Data
are representative of three experiments.
FIG. 3. HLA-DR
1
lin
2
IL-3R
a
hi
DC are present in peripheral blood. (Aand B) PBMC were stained with anti-HLA-DR, anti-IL-3R
a
, and lineage
markers (not shown) and analyzed by flow cytometry as described in Fig. 1 AC. R1 and R2 in Brepresent regions used to sort IL-3R
a
hi
blood cells that
were positive or negative for HLA-DR, respectively. (Cand D) HLA-DR
1
lin
2
IL-3R
a
hi
cells sorted from blood according to R1 in Bwere first cultured
separately with IL-3 and GM-CSF for 36 h and then incubated with allogeneic (C) or autologous (D) CD4
1
T cells. T cell proliferation was measured
as total number of CD3
1
BrdU
1
cells per well at day 6 of coculture with indicated numbers of IL-3R
a
hi
lin
2
HLA-DR
1
cells (
) or CD14
hi
monocytes
(
E
) from the same donor (stimulator cells). Individual displays show data that are representative of three experiments. OLS, ortogonal light scatter.
Immunology: Olweus et al. Proc. Natl. Acad. Sci. USA 94 (1997) 12553
FIG. 4. Proliferating progenitors for IL-3R
a
hi
DC are found as a discrete CD34
1
IL-3R
a
hi
population that is distinct from the cells that give
rise to Langerhans cells in response to GM-CSF and TNF-
a
.(Aand B) Isolated CD34
1
fetal bone marrow cells were stained with CD34 and
anti-IL-3R
a
. The cells were analyzed by flow cytometry as described in Fig. 1 AC. The CD34
1
IL-3R
a
hi
population was defined according to the
region in A(blue dots). CD34
1
IL-3R
a
lo
cells were defined according to the region in B(red dots). OLS, ortogonal light scatter. (C) Freshly sorted
Wright–Giemsa-stained CD34
1
IL-3R
a
hi
cells display mitotic figures. (3600.) (DG) CD34
1
IL-3R
a
hi
cells (blue) and CD34
1
IL-3R
a
lo
cells (red)
were sorted according to the regions in Aand B, respectively, and cultured in the presence of indicated cytokines, stained with CD1a and CD45RA
after 5 days of culture, and analyzed by FACS. L 5B lymphoid cells staining brightly with CD19 (data not shown). None of the cultured
CD34
1
IL-3R
a
hi
cells (Dand E) stained positively with CD19 (data not shown). Dashed lines indicate isotype control levels. (H) Transmission
electron microscograph (35,000) of a CD1a
1
cell sorted from CD34
1
IL-3R
a
lo
cells cultured with GM-CSF and TNF-
a
, as described in G. The
arrows point to areas containing Birbeck granules, (shown in Insets,345,000). (I) CD4
1
T cells (10
5
) were cocultured with indicated numbers of
12554 Immunology: Olweus et al. Proc. Natl. Acad. Sci. USA 94 (1997)
showing that DC in murine spleens were immature immediately
after isolation and differentiated rapidly in vitro (34, 35).
The nonactivated phenotype of DC in tonsils may seem like a
paradox since tonsils are typically removed after repeated in-
flammations. One can further not exclude the possibility that the
isolated cells had migrated to lymphoid tissue in response to
inflammation, but not yet assumed the activated phenotype. To
determine whether the presence of HLA-DR
1
lin
2
IL-3R
a
hi
cells
in lymphoid tissue depends on previous exposure to foreign
antigens or inflammatory stimuli, we examined whether the cells
could be found in fetal lymph nodes. Presumably, these lymph
nodes drain sterile, noninflamed tissues. As shown in Fig. 2C,
HLA-DR
1
lin
2
IL-3R
a
hi
cells were present at high frequencies in
the fetal lymph nodes (2.6 61.1%, n53). These cells were more
frequent than cells expressing high levels of myeloid markers,
such as CD13 and CD33 (1.5 60.2%, n53). The HLA-
DR
1
lin
2
IL-3R
a
hi
cells in fetal lymph nodes expressed the same
combination of markers as those in tonsils (Table 1). This cell type
is therefore most likely capable of migrating to lymphoid tissue
independently of inflammation and foreign antigens and without
initiating immune responses.
IL-3R
a
hi
DC Are Present in Peripheral Blood. HLA-
DR
1
lin
2
IL-3R
a
hi
cells were readily detectable as a population
with low orthogonal light scatter in blood from adult donors, and
constituted 0.47 60.14% (n58) of PBMC (Fig. 3 Aand B, R1).
A second population of IL-3R
a
hi
cells was present among PBMC,
but these cells were HLA-DR
2
(Fig. 3B, R2) and were found to
be basophilic granulocytes (data not shown). The HLA-
DR
1
lin
2
IL-3R
a
hi
blood cells expressed the same combination of
markers and had similar morphology as shown for the tonsillar
counterparts (Table 1 and data not shown). However, an inter-
esting difference was that whereas tonsillar DC did not express
the lymph node homing molecule L-selectin (CD62L), the cells
from PBMC were positive for this marker (Table 1).
Whereas multiple cell types can stimulate allogeneic T cells,
DC are characterized by their higher potency relative to other
antigen-presenting cells (13, 15, 21, 31, 33, 36–39). The cells
also induce proliferation of autologous T cells in vitro (37). The
data in Fig. 3 Cand Ddemonstrate that HLA-DR
1
lin
2
IL-
3R
a
hi
blood cells were up to 100-fold more potent than
monocytes in stimulating both allogeneic and autologous T
cells. Cells with similar characteristics have previously been
identified in blood and were referred to as CD11c
2
DC or
CD33
dim
CD14
2
CD16
2
DC (36, 37).
Precursors of IL-3R
a
hi
DC Are Found Among CD34
1
Bone
Marrow Cells and Are Distinct from the Cells That Give Rise to
Langerhans Cells in Response to GM-CSF and TNF-
a
.IL-3R
a
hi
cells were readily identified as a distinct subset of CD34
1
fetal
bone marrow cells with low orthogonal light scatter (3.1 60.9%,
n55) (Fig. 4 Aand B). The majority of the CD34
1
IL-3R
a
hi
cells
were within the CD34
lo
subset, which contains lineage-committed
progenitors (Fig. 4B) (23). The CD34
1
IL-3R
a
hi
cells had mor-
phology and immunophenotype similar to IL-3R
a
hi
lin
2
HLA-
DR
1
cells in blood and tonsil, but appeared more immature, and
mitotic figures were frequently observed (Fig. 4Cand Table 1).
Previous studies have demonstrated that DC and Langerhans
cells can be derived by culture of CD34
1
cells with GM-CSF and
TNF-
a
(13–19). The subset of CD34
1
cells that contains these
TNF-
a
-dependent progenitors has not yet been identified. We
therefore examined whether they were identical to the CD34
1
IL-
3R
a
hi
cells. The sorted IL-3R
a
hi
population (Fig. 4A) formed
aggregates of cells with DC morphology during culture with
either GM-CSF and IL-3 or GM-CSF and TNF-
a
(data not
shown), and after 5 days the cells were positive for CD1a (Fig. 4
Dand E). However, differentiation into DC occurred indepen-
dently of TNF-
a
, and the CD1a
1
cells co-expressed CD45RA,
which is absent from Langerhans cells (39) (Fig. 4 Dand E).
Tonsillar HLA-DR
1
lin
2
IL-3R
a
hi
cells also obtained a
CD1a
1
CD45RA
1
phenotype when cultured under the same
conditions (data not shown). In contrast, sorted CD34
1
cells with
low levels of the IL-3R
a
(Fig. 4B) gave rise to few CD1a
1
cells
when cultured with GM-CSF and IL-3, but large numbers of
CD45RA
2
CD1a
1
cells in the presence of GM-CSF and TNF-
a
(Fig. 4 Fand G). Consistent with a phenotype of Langerhans
cells, 30–40% of these CD45R A
2
CD1a
1
cells contained Birbeck
granules (Fig. 4H). The frequency of Birbeck granule-positive
cells among CD1a
1
cultured CD34
1
IL-3R
a
hi
cells was less than
4%, and may therefore reflect contamination of sorting gates.
Finally, CD34
1
IL-3R
a
hi
cells cultured with GM-CSF and IL-3
were potent stimulators of allogeneic CD4
1
T cells, and more
potent than CD14
1
macrophages generated by culture of
CD34
1
M-CSFR
1
cells from the same donor in M-CSF (Fig. 4I).
IL-3R
a
hi
DC Are of Myeloid Origin. The distribution of
M-CSF receptor (M-CSFR) and IL-3R
a
among CD34
1
cells
suggested that CD34
1
IL-3R
a
hi
cells may derive from cells in the
M-CSFR
hi
population, which down-regulate the M-CSFR as they
up-regulate the IL-3R
a
(arrow in Fig. 5A). Because M-CSFR
expression on CD34
1
cells is restricted to granulomonocytic
progenitors, this would indicate that IL-3R
a
hi
cells belong to the
granulomonocytic lineage (29). To test this possibility, immature
progenitors (i.e., CD34
hi
, see Fig. 4B) with high levels of M-CSFR
and low levels of IL-3R
a
(region in Fig. 5A) were sorted and
cultured. After 60 h of culture two populations of cells that had
downmodulated the M-CSFR were observed, with high and low
levels of IL-3R
a
, respectively (Fig. 5B). These cells were sorted
and cultured for 5 additional days in medium supplemented with
GM-CSF and IL-3. At this stage, the cultures from the IL-3R
a
lo
population contained CD15
1
granulocytic cells (Fig. 5C) whereas
cultures from the IL-3R
a
hi
population contained CD1a
1
cells
with DC morphology (Fig. 5Dand data not shown). The latter
cells induced strong proliferation of allogeneic T cells compared
with equal numbers of CD14
1
macrophages generated by culture
of CD34
1
M-CSFR
1
cells for 5 days with M-CSF (29) (n52, data
not shown). Addition of TNF-
a
to the medium did not increase
the number of CD34
1
IL-3R
a
hi
cells generated from M-CSFR
hi
myeloid progenitors, further suggesting that the cells are distinct
from the TNF-
a
-dependent Langerhans cell progenitors (data
not shown).
Thymus Contains Small Numbers of IL-3R
a
hi
DC That
Express Low Levels of CD34. Earlier studies have demon-
strated that some DC may share the differentiation pathway of
the T cell lineage and that DC are generated by progenitors in
the thymus (18, 19, 40). We therefore investigated whether
IL-3R
a
hi
DC were present in the thymus. The results showed
that less than 0.1% of the total thymic cell population were
IL-3R
a
hi
DC and that the IL-3R
a
hi
cells constituted only 15 6
2% (n53) of HLA-DR
1
lin
2
subset. This may indicate that
other DC populations than the IL-3R
a
hi
cells are predominant
in thymus. In addition, the IL-3R
a
hi
DC in thymus expressed
low levels of CD34 compared with bone marrow IL-3R
a
hi
cells
from the same donor (data not shown). Similar low levels of
CD34 were found on the cells in fetal lymph nodes (Table 1).
It therefore seems likely that the few IL-3R
a
hi
DC in the
thymus represent cells that have migrated from bone marrow
and are distinct from thymic DC progenitors.
IL-3R
a
hi
DC Constitute a Separate Lineage of DC That Follow
a Differentiation Pathway Distinct from Langerhans Cells. The
data presented here demonstrate that a subset of DC in human
allogeneic DC (
) or macrophages (
E
) from the same donor. The DC were generated by culturing sorted CD34
1
IL-3R
a
hi
cells for 5 days with
GM-CSF and IL-3 (
). The macrophages were generated by culture of CD34
1
M-CSFR
1
cells for 5 days with M-CSF and purified by FACS sorting
of CD14
1
cells. T cell proliferation was measured as incorporation of [
3
H]thymidine at day 6 of coculture. c.p.m; counts per minute. Data are
representative of three experiments.
Immunology: Olweus et al. Proc. Natl. Acad. Sci. USA 94 (1997) 12555
lymphoid organs and blood represent a separate lineage of cells.
The cells can be readily identified and isolated on the basis of their
high levels of the IL-3R
a
. Their precursors in the bone marrow
appear to follow the myeloid differentiation pathway to the
branching point of the granulocytic and monocytic lineages.
Progenitors that have committed to this DC lineage are distinct
from those that give rise to Langerhans cells when cultured in the
presence of GM-CSF and TNF-
a
. Their progeny furthermore
lack several characteristics of Langerhans cells that were previ-
ously considered common to DC. Unlike Langerhans cells, the
IL-3R
a
hi
DC appear to undergo little differentiation during
transit from the bone marrow to lymphoid organs. The IL-3R
a
hi
DC further seem capable of migrating to lymphoid tissue inde-
pendently of inflammatory stimuli. The results concur with
previous reports showing that some DC spend less than 24 h in
transit from the progenitor pool to lymphoid tissue and that DC
turnover occurs constantly during steady state conditions (1,
41–43). Thus, the DC system is constituted by multiple cell types
with distinct developmental pathways and functional properties.
A large proportion of the cells may enter lymphoid tissue without
inducing immune responses.
We thank Drs. Lewis Lan ier, Anne O’Garra, Rene de Waal Malefyt,
and Gosse Adema for critical reading of the manuscript; David Houck
for expert assistance with cell sorting; and Barny Abrams, Dr. Qudrat
Nasraty, and Dr. Kenneth A. Davis for conjugation of antibodies. J.O.
and F.L.-J. were supported by grants from the Norwegian Research
Counsel. DNAX Research Institute is supported by Schering-Plough.
1. Steinman, R. M. (1991) Annu. Rev. Immunol. 9, 271–296.
2. Schuler, G. & Steinman, R. (1985) J. Exp. Med. 161, 526–546.
3. Silberberg-Sinakin, I., Thorbecke, J., Baer, R., Rosenthal, S. &
Berezowsky, V. (1976) Cell. Immunol. 25, 137–151.
4. Macatonia, S., Knight, S., Edwards, A., Griffiths, S. & Fryer, P.
(1987) J. Exp. Med. 166, 1654–1667.
5. Kripke, M., Munn, C., Jeevan, A., Tang, J. & Bucana, C. (1990)
J. Immunol. 145, 2833–2838.
6. Roake, J. A., Rao, A. S., Morris, P. J., Larsen, C. P., Hankins,
D. F. & Austyn, J. M. (1995) J. Exp. Med. 181, 2237–2247.
7. Hart, D. & Fabre, J. (1981) J. Exp. Med. 153, 347–361.
8. MacPherson, G. G., Jenkins, C. D., Stein, M. J. & Edwards, C.
(1995) J. Immunol. 154, 1317–1322.
9. Fearon, D. & Locksley, R. (1996) Science 272, 50–52.
10. Ibrahim, M. A., Chain, B. M. & Katz, D. R. (1995) Immunol.
Today 16, 181–186.
11. Austyn, J. M. (1996) J. Exp. Med. 183, 1287–1292.
12. Lanzavecchia, A. (1996) Curr. Opin. Immunol. 8, 348–354.
13. Caux, C., Dezutter-Dambuyant, D. C., Schmitt, D. & Banche-
reau, J. (1992) Nature (London) 360, 258–261.
14. Reid, C. D., Stackpoole, A., Meager, A. & Tikerpae, J. (1992)
J. Immunol. 149, 2681–2688.
15. Young, J. W., Szabolcs, P. & Moore, M. A. (1995) J. Exp. Med.
182, 1111–1119.
16. Caux, C., Vanbervliet, B., Massacrier, C., Durand, I. & Banche-
reau, J. (1996) Blood 87, 2376–2385.
17. Caux, C., Vanbervliet, B., Massacrier, C., Dezutter-Dambuyant,
D. C., de Saint-Vis, B., Jacquet, C., Yoneda, K., Imamura, S.,
Schmitt, D. & Banchereau, J. (1996) J. Exp. Med. 184, 695–706.
18. Galy, A., Travis, M., Cen, D. & Chen, B. (1995) Immunity 3,
459– 473.
19. Res, P., Martinez, C. E., Jaleco, C. A., Staal, F., Noteboom, E.,
Weijer, K. & Spits, H. (1996) Blood 87, 5196–5206.
20. Inaba, K., Inaba, M., Deguchi, M., Hagi, K., Yasumizu, R.,
Ikehara, S., Muramatsu, S. & Steinman, R. M. (1993) Proc. Natl.
Acad. Sci. USA 90, 3038–3042.
21. Zhou, L. J. & Tedder, T. F. (1996) Proc. Natl. Acad. Sci. USA 93,
2588–2592.
22. Peters, J., Gieseler, R., Thiele, B. & Steinbach, F. (1996) Immu-
nol. Today 17, 273–278.
23. Terstappen, L. W., Huang, S., Safford, M., Lansdorp, P. M. &
Loken, M. R. (1991) Blood 77, 1218–1227.
24. Baum, C. M., Weissman, I. L., Tsukamoto, A. S., Buckle, A. M.
& Peault, B. (1992) Proc. Natl. Acad. Sci. USA 89, 2804–2808.
25. Lansdorp, P. M. & Dragowska, W. (1992) J. Exp. Med. 175,
1501–1509.
26. Terstappen, L. W. M. M., Huang, S. & Picker, L. J. (1992) Blood
79, 666– 677.
27. Olweus, J., Lund-Johansen, F. & Terstappen, L. W. M. M. (1994)
Blood 85, 2402–2413.
28. Olweus, J., Terstappen, L., Thompson, P. & Lund-Johansen, F.
(1996) Blood 88, 1594–1607.
29. Olweus, J., Thompson, P. & Lund-Johansen, F. (1996) Blood 88,
3741–3754.
30. Yssel, H., de Vries, J., Koken, M., Van Blitterswijk, W. & Spits,
H. (1984) J. Immunol. Methods 72, 219–224.
31. Hart, D. N. J. & McKenzie, J. L. (1988) J. Exp. Med. 168, 157–170.
32. Cameron, P. U., Lowe, M. G., Sotzik, F., Coughlan, A. F., Crowe,
S. M. & Shortman, K. (1996) J. Exp. Med. 183, 1851–1856.
33. Grouard, G., Rissoan, M., Filgueira, L., Durand, I., Banchereau,
J. & Liu, J. (1997) J. Exp. Med. 185, 1101–1112.
34. Crowley, M., Inaba, K., Witmer-Pack, M., Gezelter, S. & Stein-
man, R. (1990) J. Immunol. Methods 133, 55–66.
35. Inaba, K., Witmer-Pack, M., Inaba, M., Hathcock, K. S., Sakuta,
H., Azuma, M., Yagita, H., Okumura, K., Linsley, P. S., Ikehara,
S., Muramatsu, S., Hodes, R. J. & Steinman, R. M. (1994) J. Exp.
Med. 180, 1849–1860.
36. O’Doherty, U., Peng, M., Gezelter, S., Swiggard, W. J., Betjes, M.,
Bhardwaj, N. & Steinman, R. M. (1994) Immunology 82, 487– 493.
37. Thomas, R. & Lipsky, P. E. (1994) J. Immunol. 153, 4016–4028.
38. Grouard, G., Durand, I., Filgueira, L., Banchereau, J. & Liu, Y.
(1996) Nature (London) 384, 364–367.
39. Pope, M., Betjes, M. G., Hirmand, H., Hoffman, L. & Steinman,
R. M. (1995) J. Invest. Dermatol. 104, 11–17.
40. Ardavin, C., Wu, L., Li, C. L. & Shortman, K. (1993) Nature
(London) 362, 761–763.
41. Pugh, C., MacPherson, G. & Steer, H. (1983) J. Exp. Med. 157,
1758–1779.
42. Fossum, S. (1989) Curr. Top. Pathol. 79, 101–124.
43. Matsuno, K., Ezaki, T., Kudo, S. & Uehara, Y. (1996) J. Exp. Med.
183, 1865–1878.
FIG. 5. IL-3R
a
hi
DC follow the myeloid differentiation pathway to the
branching point of the granulocytic and monocytic lineages. (A) CD34
1
cells were incubated for 12 h in serum-free medium (25) to allow
up-regulation of the M-CSFR, and stained with CD34, anti-M-CSFR, and
anti-IL-3R
a
. The arrow shows the suggested differentiation pathway of
CD34
1
IL-3R
a
hi
cells. The region shows sorting criteria for M-CSFR
hi
y
IL-3R
a
lo
cells. An additional gate was set to include only CD34
hi
cells,
shown in Fig. 4B, to restrict the sort to immature progenitors (29). (B)
After a 60-h culture of CD34
hi
M-CSFR
1
cells in serum-free medium (25)
containing stem cell factor, G-CSF, GM-CSF, IL-3, and IL-6, the cells
were stained with anti-M-CSFR and anti-IL-3R
a
. The regions indicate
criteria for sorting of the two populations that had downmodulated the
M-CSFR during the culture period. (Cand D) After a 5-day secondary
culture of IL-3R
a
lo
M-CSFR
lo
cells (green) and IL-3R
a
hi
M-CSFR
lo
cells
(blue) with IL-3 and GM-CSF, the cells were stained with CD1a and
CD15 and analyzed by FACS. The small subset of CD15
lo
cells in C
represent basophilic granulocytes (27, 29).
12556 Immunology: Olweus et al. Proc. Natl. Acad. Sci. USA 94 (1997)
... T cells are only fully activated via certain signaling interactions with APCs. The ontogeny of DCs has been redefined several times and continues to advance as single cell technology allows for the detection of the dynamic development of cells and detection of transcriptional profiles on minimal subsets, which was reviewed in other places [3,[10][11][12][13]. Initially, it was thought that the precursors of DCs were epidermal Langerhans cells [11]; now, it is known that the origin of DCs begins with the development of hematopoietic stem cells (HSCs) and is derived from the bone marrow [10,11]. ...
... The ontogeny of DCs has been redefined several times and continues to advance as single cell technology allows for the detection of the dynamic development of cells and detection of transcriptional profiles on minimal subsets, which was reviewed in other places [3,[10][11][12][13]. Initially, it was thought that the precursors of DCs were epidermal Langerhans cells [11]; now, it is known that the origin of DCs begins with the development of hematopoietic stem cells (HSCs) and is derived from the bone marrow [10,11]. Granulocytes, monocytes, and DCs share a common progenitor downstream of HSC termed granulocyte-macrophage DC progenitors (GMDPs), from which multiple progenitors of combinations of the three classes further differentiate into, which is summarized in Fig. 1 [14]. ...
... The ontogeny of DCs has been redefined several times and continues to advance as single cell technology allows for the detection of the dynamic development of cells and detection of transcriptional profiles on minimal subsets, which was reviewed in other places [3,[10][11][12][13]. Initially, it was thought that the precursors of DCs were epidermal Langerhans cells [11]; now, it is known that the origin of DCs begins with the development of hematopoietic stem cells (HSCs) and is derived from the bone marrow [10,11]. Granulocytes, monocytes, and DCs share a common progenitor downstream of HSC termed granulocyte-macrophage DC progenitors (GMDPs), from which multiple progenitors of combinations of the three classes further differentiate into, which is summarized in Fig. 1 [14]. ...
Article
Full-text available
Antigen-presenting cells (APCs) are pivotal mediators of immune responses. Their role has increasingly been spotlighted in the realm of cancer immunology, particularly as our understanding of immunotherapy continues to evolve and improve. There is growing evidence that these cells play a non-trivial role in cancer immunity and have roles dependent on surface markers, growth factors, transcription factors, and their surrounding environment. The main dendritic cell (DC) subsets found in cancer are conventional DCs (cDC1 and cDC2), monocyte-derived DCs (moDC), plasmacytoid DCs (pDC), and mature and regulatory DCs (mregDC). The notable subsets of monocytes and macrophages include classical and non-classical monocytes, macrophages, which demonstrate a continuum from a pro-inflammatory (M1) phenotype to an anti-inflammatory (M2) phenotype, and tumor-associated macrophages (TAMs). Despite their classification in the same cell type, each subset may take on an immune-activating or immunosuppressive phenotype, shaped by factors in the tumor microenvironment (TME). In this review, we introduce the role of DCs, monocytes, and macrophages and recent studies investigating them in the cancer immunity context. Additionally, we review how certain characteristics such as abundance, surface markers, and indirect or direct signaling pathways of DCs and macrophages may influence tumor response to immune checkpoint blockade (ICB) therapy. We also highlight existing knowledge gaps regarding the precise contributions of different myeloid cell subsets in influencing the response to ICB therapy. These findings provide a summary of our current understanding of myeloid cells in mediating cancer immunity and ICB and offer insight into alternative or combination therapies that may enhance the success of ICB in cancers.
... Among the several markers that have been immunohistochemically utilized for plasmacytoid dendritic cell identification, blood-derived dendritic cell antigen-2/ CD303 and CD123 represent the most specific and sensitive, and as such are the most commonly used. [5][6][7][11][12][13] Blood-derived dendritic cell antigen-2 is a type II C-type lectin receptor selectively expressed on plasmacytoid dendritic cells. 13 It is involved in antigen capture and in regulation of type I interferon production. ...
... 18 It is highly expressed on the surface of plasmacytoid dendritic cells. However, it should be kept in mind that CD123 can be detected with lower intensity on other cell types such as endothelial cells, 12 other subsets of dendritic cells 10 and activated macrophages, though plasmacytoid dendritic cells can still be differentiated on hematoxylin and eosin staining from the other CD123+ cell types based on their small-to medium-sized plasma cell-like morphology. On the other hand, blood-derived dendritic cell antigen-2 has not been reported to be expressed by other cell types, but it can be partially lost upon the activation of plasmacytoid dendritic cells in inflammatory processes. ...
... [22][23][24][25][26][27][28][29][30][31][32][33][34][35][36] For plasmacytoid dendritic cell content, semi-quantitative scoring systems that evaluate plasmacytoid dendritic cells as a percentage of the total mononuclear infiltrate were more commonly used than absolute plasmacytoid dendritic cell counts in most of the studies. 11,12,[15][16][17][33][34][35][36] Plasmacytoid dendritic cell clusters were defined differently among studies, varying from clusters of at least 5 touching plasmacytoid dendritic cells, 23 ≥10 plasmacytoid dendritic cells, [15][16][17]25,26,30 ≥15 touching CD123 + plasmacytoid dendritic cells 28 to 20 or more cells. 22 Most studies however, defined plasmacytoid dendritic cell clusters as having at least 10 plasmacytoid dendritic cells. ...
Article
Full-text available
Differentiating cutaneous diseases that mimic each other clinically and histopathologically can at times be a challenging task for the dermatopathologist. At the same time, differentiation of entities with overlapping features may be crucial for patient management. Although not seen in normal skin, plasmacytoid dendritic cells usually infiltrate the skin in several infectious, inflammatory/autoimmune and neoplastic entities. Plasmacytoid dendritic cells can be identified in tissue using specific markers such as CD123 and/or blood-derived dendritic cell antigen-2. Plasmacytoid dendritic cells are the most potent producers of type I interferons and their activity may therefore be assessed indirectly in tissue using human myxovirus resistance protein A, a surrogate marker for type I interferon production. In recent years, accumulating evidence has established the utility of evaluating for specific plasmacytoid dendritic cell-related parameters (plasmacytoid dendritic cell content, distribution and clustering and/ or human myxovirus resistance protein A expression) as a diagnostic tool in differentiating cutaneous diseases with overlapping features such as the alopecias, lupus and its mimics, and neoplastic entities. In this review, we provide an update on the current evidence on this topic and on the contexts where this can be a useful adjunct to reach the histopathological diagnosis.
... [4] It is highly expressed on the surface of PDC, which is known to accumulate in the infiltrate of various disorders. [1][2][3] PDC on stimulation secrets large amount of type I interferons (α and β) [5][6][7] LE is an autoimmune disorder with an alteration in the function of Plasmacytoid dendritic cells (PDC). Recentstudies have suggested that CD123 positive PDCs are increased in throughout the spectrum of LE. [1] There are diagnostic difficulties in cases of LE and LPP presenting late in the course of the disease with scarring and loss of follicles and DIF is also often negative. ...
Article
Full-text available
Classification of scarring alopecia poses a major problem, as there is considerable clinicopathologic overlap, particularly between lupus erythematosus (LE) and lichen planopilaris (LPP), especially in later stages. CD123 positive plasmacytoid dendritic cells (PDC) have been shown recently to be present in all forms of LE and are touted to be useful in differentiating LE from other scarring alopecias. Their distribution in non-scarring alopecia is not well documented. This is the first study that examines the PDC in both scarring and non-scarring alopecias. Objective: To study the expression patterns of PDC in cases of both scarring and non-scarring alopecia. Materials and methods: A total of 69 cases of alopecia (48 scarring, 21 non-scarring) were studied for CD123 expression by immunohistochemistry. Results: Among the scarring alopecias, 17/20 LE cases showed PDC in contrast to 1/22 LPP cases. This difference was statistically significant (P = 0.0001). 1/2 cases of folliculitis decalvans showed PDC. None of the cases of unclassified scarring alopecia were positive. In the non-scarring group, 19/20 cases of alopecia areata and a single case of trichotillomania lacked PDC. Conclusion: The finding of CD123 expressing PDC appears to be a promising parameter in distinguishing LE from other forms of alopecia.
... To analyze their potential ability to generate neutrophils, we stained BM-LDCs with an antibody panel including CD34, CD38, CD10, CD123, CD45RA, CD64 and CD115 (refs. 13,14,20,28 ). We identified an SSC lo Lin − CD45 dim CD10 − CD38 + subset with variable expression of CD34 and CD45RA (Fig. 1a), including CD34 + CD45RA − , CD34 + CD45RA + , CD34 dim/− CD45RA + and CD34 dim/− CD45RA − cell subsets. ...
Article
Full-text available
Here we report the identification of human CD66b⁻CD64dimCD115⁻ neutrophil-committed progenitor cells (NCPs) within the SSCloCD45dimCD34⁺ and CD34dim/− subsets in the bone marrow. NCPs were either CD45RA⁺ or CD45RA⁻, and in vitro experiments showed that CD45RA acquisition was not mandatory for their maturation process. NCPs exclusively generated human CD66b⁺ neutrophils in both in vitro differentiation and in vivo adoptive transfer experiments. Single-cell RNA-sequencing analysis indicated NCPs fell into four clusters, characterized by different maturation stages and distributed along two differentiation routes. One of the clusters was characterized by an interferon-stimulated gene signature, consistent with the reported expansion of peripheral mature neutrophil subsets that express interferon-stimulated genes in diseased individuals. Finally, comparison of transcriptomic and phenotypic profiles indicated NCPs represented earlier neutrophil precursors than the previously described early neutrophil progenitors (eNePs), proNeus and COVID-19 proNeus. Altogether, our data shed light on the very early phases of neutrophil ontogeny.
... They can be uniquely identified in blood and affected lymph nodes of NEL by their high levels of interleukin-3 receptor α chain (CD123) combined with other cell markers such as CD45RA, CD68, and HLA-DR. 7,[11][12][13][14][15] PDCs are continuously produced from bone marrow. After leaving the bone marrow, they migrate into T-cell-rich areas of the lymphoid tissues through HEV in lymph nodes. ...
Article
Full-text available
Necrotizing lymphadenitis (NEL) is a self-limited systemic disease exhibiting characteristic clinical features. The pathogenesis of the disease remains unclear, but it may be associated with viral infection. In lymph nodes affected by this disease, innumerable plasmacytoid dendritic cells produce interferon-α when triggered by certain viral stimuli. IFN-α presents antigens causing the transformation of CD8⁺ cells into immunoblasts and apoptosis of CD4⁺ cells. From the perspective of innate immunity, UNC93B1, an endoplasmic reticulum (ER)-resident protein, associates more strongly with TLR9 than TLR7. Homeostasis is maintained under normal conditions. However, in NEL, TLR 7 was observed more than TLR 9, possibly because mutant type UNC93B1 associates more tightly with TLR7. The inhibitory effects against TLR7 by TLR9 were reported to disappear. It is likely that more TLR7 than TLR9 is transported from the ER to endolysosomes. In conclusion, overexpression of TLR7, an innate immune sensor of microbial single-stranded RNA, is inferred. Consequently, NEL may be induced.
... Moreover, although CD34 dim/cells represent neglected, but important, transitional progenitor stages 4 , they have been rarely investigated for a potential ability to generate neutrophils. Therefore, we stained BM-LDCs by a flow-cytometry antibody panel comprising key markers conventionally used to detect either BM or cord blood (CB) CD34 + myeloid and lymphoid progenitors, namely CD34, CD38, CD10, CD123, CD45RA, CD64 and CD115 14,15,21,28 . By doing so, we could identify a lineage negative, SSC low CD45 dim CD10 -CD38 + region displaying variable CD34 and CD45RA levels (panel V of Figure 1A), and including CD34 + CD45RA -, CD34 + CD45RA + , CD34 dim/-CD45RA + and CD34 dim/-CD45RAcell populations. ...
Preprint
We report the identification of human CD66b ⁻ CD64 dim CD115 ⁻ neutrophil-committed progenitors within SSC low CD45 dim CD34 ⁺ and CD34 dim/− bone marrow cells, that we named neutrophil myeloblast (NMs). CD34 ⁺ and CD34 dim/− NMs resulted as either CD45RA ⁺ or CD45RA ⁻ , with CD34 ⁺ CD45RA ⁻ NMs found as selectively expanded in chronic-phase chronic myeloid leukemia patients. By scRNA-seq experiments, CD34 ⁺ and CD34 dim/− NMs were found to consist of combinations of four cell clusters, characterized by different maturation stages and distributed along two differentiation routes. Cell clusters were identified by neutrophil-specific gene profiles, one of them associated to an interferon-stimulated gene (ISG) signature, hence supporting recently identified expansions of mature neutrophil subsets expressing ISGs in blood of diseased individuals. Altogether, our data shed light on the very early phases of neutrophil ontogeny.
Article
Full-text available
Acute myeloid leukemia (AML), the most frequent leukemia in adults, is driven by recurrent somatically acquired genetic lesions in a restricted number of genes. Treatment with tyrosine kinase inhibitors has demonstrated that targeting of prevalent FMS-related receptor tyrosine kinase 3 (FLT3) gain-of-function mutations can provide significant survival benefits for patients, although the efficacy of FLT3 inhibitors in eliminating FLT3-mutated clones is variable. We identified a T cell receptor (TCR) reactive to the recurrent D835Y driver mutation in the FLT3 tyrosine kinase domain (TCRFLT3D/Y). TCRFLT3D/Y-redirected T cells selectively eliminated primary human AML cells harboring the FLT3D835Y mutation in vitro and in vivo. TCRFLT3D/Y cells rejected both CD34⁺ and CD34⁻ AML in mice engrafted with primary leukemia from patients, reaching minimal residual disease-negative levels, and eliminated primary CD34⁺ AML leukemia-propagating cells in vivo. Thus, T cells targeting a single shared mutation can provide efficient immunotherapy toward selective elimination of clonally involved primary AML cells in vivo.
Article
Since their discovery, the identity of plasmacytoid dendritic cells (pDCs) has been at the center of a continuous dispute in the field, and their classification as dendritic cells (DCs) has been recently re-challenged. pDCs are different enough from the rest of the DC family members to be considered a lineage of cells on their own. Unlike the exclusive myeloid ontogeny of cDCs, pDCs may have dual origin developing from myeloid and lymphoid progenitors. Moreover, pDCs have the unique ability to quickly secrete abundant levels of type I interferon (IFN-I) in response to viral infections. In addition, pDCs undergo a differentiation process after pathogen recognition that allows them to activate T cells, a feature that has been shown to be independent of presumed contaminating cells. Here, we aim to provide an overview of the historic and current understanding of pDCs and argue that their classification as either lymphoid or myeloid may be an oversimplification. Instead, we propose that the capacity of pDCs to link the innate and adaptive immune response by directly sensing pathogens and activating adaptive immune responses justify their inclusion within the DC network.
Article
Full-text available
Polymorphonuclear neutrophils are no longer considered as a homogeneous population of terminally differentiated and short‐lived cells that belong to the innate immune system only. In fact, data from the past decades have uncovered that neutrophils exhibit large phenotypic heterogeneity and functional versatility that render them more plastic than previously thought. Hence, their precise role as effector cells in inflammation, in immune response and in other pathophysiological processes, including tumors, needs to be better evaluated. In such a complex scenario, common knowledge of the differentiation of neutrophils in bone marrow refers to lineage precursors, starting from the still poorly defined myeloblasts, and proceeding sequentially to promyelocytes, myelocytes, metamyelocytes, band cells, segmented neutrophils, and mature neutrophils, with each progenitor stage being more mature and better characterized. Thanks to the development and utilization of cutting‐edge technologies, novel information about neutrophil precursors at stages earlier than the promyelocytes, hence closer to the hematopoietic stem cells, is emerging. Accordingly, this review discusses the main findings related to the very early precursors of human neutrophils and provides our perspectives on human neutropoiesis.
Article
Human plasmacytoid dendritic cells (pDCs) are interleukin-3 (IL-3)-dependent cells implicated in autoimmunity, but the role of IL-3 in pDC biology is poorly understood. We found that IL-3-induced Janus kinase 2-dependent expression of SLC7A5 and SLC3A2, which comprise the large neutral amino acid transporter, was required for mammalian target of rapamycin complex 1 (mTORC1) nutrient sensor activation in response to toll-like receptor agonists. mTORC1 facilitated increased anabolic activity resulting in type I interferon, tumor necrosis factor, and chemokine production and the expression of the cystine transporter SLC7A11. Loss of function of these amino acid transporters synergistically blocked cytokine production by pDCs. Comparison of in vitro-activated pDCs with those from lupus nephritis lesions identified not only SLC7A5, SLC3A2, and SLC7A11 but also ectonucleotide pyrophosphatase-phosphodiesterase 2 (ENPP2) as components of a shared transcriptional signature, and ENPP2 inhibition also blocked cytokine production. Our data identify additional therapeutic targets for autoimmune diseases in which pDCs are implicated.
Article
Full-text available
Initiation of an adoptive immune response against pathogenic organisms, such as bacteria and fungi, may involve phagocytic activity of dendritic cells (DC) or their immature precursors as a prelude to antigen processing and presentation. After intravenous injection of rats with particulate matter, particle-laden cells were detected in the peripheral hepatic lymph. Since it has been known there is a constant efflux of DC from nonlymphoid organs into the draining peripheral lymph, we examined whether these particle-laden cells belonged to the DC or macrophage lineage. The majority of particle-laden cells in lymph showed immature monocyte-like cytology, and the amount of ingested particles was small relative to typical macrophages. We identified these particle-laden cells as DC based on a number of established criteria: (a) they had a phenotype characteristic of rat DC, that is, major histocompatibility complex class Ihigh+ and IIhigh+, intercellular adhesion molecule 1+ and 80% positive with the rat DC-specific mAb OX62; (b) they showed strong stimulating capacity in primary allogeneic mixed leukocyte reaction; (c) in vitro, they had little phagocytic activity; and (d) the kinetics of translocation was similar to that of lymph DC in that they migrated to the thymus-dependent area of the regional nodes. Furthermore, bromodeoxyuridine feeding studies revealed that most of the particle-laden DC were recently produced by the terminal division of precursor cells, at least 45% of them being <5.5 d old. The particle-laden DC, defined as OX62+ latex-laden cells, were first found in the sinusoidal area of the liver, in the liver perfusate, and in spleen cell suspensions, suggesting that the site of particle capture was mainly in the blood marginating pool. It is concluded that the particle-laden cells in the hepatic lymph are recently produced immature DC that manifest a temporary phagocytic activity for intravascular particles during or after the terminal division and that the phagocytic activity is downregulated at a migratory stage when they translocate from the sinusoidal area to the hepatic lymph.
Article
Full-text available
A subset of CD4+CD11c−CD3− blood cells was recently shown to develop into dendritic cells when cultured with monocyte conditioned medium. Here, we demonstrate that CD4+ CD11c−CD3− cells, isolated from tonsils, correspond to the so-called plasmacytoid T cells, an obscure cell type that has long been observed by pathologists within secondary lymphoid tissues. They express CD45RA, but not markers specific for known lymphoid- or myeloid-derived cell types. They undergo rapid apoptosis in culture, unless rescued by IL-3. Further addition of CD40-ligand results in their differentiation into dendritic cells that express low levels of myeloid antigens CD13 and CD33.
Article
Full-text available
Human dendritic cells (DC) can now be generated in vitro in large numbers by culturing CD34+ hematopoietic progenitors in presence of GM-CSF+TNF alpha for 12 d. The present study demonstrates that cord blood CD34+ HPC indeed differentiate along two independent DC pathways. At early time points (day 5-7) during the culture, two subsets of DC precursors identified by the exclusive expression of CD1a and CD14 emerge independently. Both precursor subsets mature at day 12-14 into DC with typical morphology and phenotype (CD80, CD83, CD86, CD58, high HLA class II). CD1a+ precursors give rise to cells characterized by the expression of Birbeck granules, the Lag antigen and E-cadherin, three markers specifically expressed on Langerhans cells in the epidermis. In contrast, the CD14+ progenitors mature into CD1a+ DC lacking Birbeck granules, E-cadherin, and Lag antigen but expressing CD2, CD9, CD68, and the coagulation factor XIIIa described in dermal dendritic cells. The two mature DC were equally potent in stimulating allogeneic CD45RA+ naive T cells. Interestingly, the CD14+ precursors, but not the CD1a+ precursors, represent bipotent cells that can be induced to differentiate, in response to M-CSF, into macrophage-like cells, lacking accessory function for T cells. Altogether, these results demonstrate that different pathways of DC development exist: the Langerhans cells and the CD14(+)-derived DC related to dermal DC or circulating blood DC. The physiological relevance of these two pathways of DC development is discussed with regard to their potential in vivo counterparts.
Article
These studies address the hypothesis that Ag-bearing epidermal Langerhans cells migrate to the regional lymph node during contact sensitization and function as APC. Skin from C3H mice was grafted onto BALB/c nude mice, and 7 or 14 days later, the recipients were sensitized with FITC through the grafts. APC from lymph nodes draining the site of sensitization were capable of sensitizing C3H recipients to FITC. Because sensitization is MHC restricted, only cells reaching the lymph node from the grafted skin could have induced contact hypersensitivity in C3H mice. Examination of the FITC+ draining lymph node cells by immunofluorescence and immunoelectron microscopy demonstrated that all were Ia+, most were F4/80+, and some contained Birbeck granules. These studies demonstrate that Ia+, FITC+ cells from the skin, at least some of which are Langerhans cells, leave the skin after epicutaneous sensitization with FITC and participate in the initiation of the contact hypersensitivity response within the regional lymph node.
Article
The aim of the present study was to determine whether stem cell factor (SCF) and erythropoietin (EPO) act differently on defined subsets of progenitor cells, and if potential differences correlate with the receptor density on each subset. To investigate this possibility directly, we optimized conditions for the identification and purification of homogeneous progenitor cell subpopulations from human bone marrow. Populations containing 40% and 44% colony forming cells (CFCs) with 99% and 95% purity for the granulomonocytic and erythroid lineage, respectively, were sorted on the basis of differential expression of CD34, CD64, and CD71. In addition, a population containing 67% CFCs, of which 29–43% were CFU-MIX, was sorted from CD34hi CD38loCD50+ cells. Purified progenitor cell subsets were compared directly for responsiveness to SCF and EPO using a short-term proliferation assay. Expression of the receptors for SCF and EPO were then examined on each subset using a flow cytometer modified for high- sensitivity fluorescence measurements. The results show that EPO induces extensive proliferation of erythroid progenitor cells, but has no effect on the proliferation or survival of primitive or granulomonocytic progenitors, even when used in combination with other cytokines. The majority of erythroid progenitor cells furthermore stained positively with anti-EPO receptor (EPO-R) monoclonal antibodies, whereas other progenitor cells were negative. SCF alone induced extensive proliferation of erythroid progenitor cells, and had a stronger synergistic effect on primitive than on granulo-monocytic progenitors. In spite of these differences in SCF activity, there were no significant differences in SCF-R expression between the progenitor subsets. These results suggest that the selective action of EPO on erythropoiesis is determined by lineage-restricted receptor expression, whereas there are additional cell-type specific factors that influence progenitor cell responses to SCF.
Article
Multiparameter flow cytometry was applied on normal human bone marrow (BM) cells to study the lineage commitment of progenitor cells ie, CD34+ cells. Lineage commitment of the CD34+ cells into the erythroid lineage was assessed by the coexpression of high levels of the CD71 antigen, the myeloid lineage by coexpression of the CD33 antigen and the B-lymphoid lineage by the CD10 antigen. Three color immunofluorescence experiments showed that all CD34+ BM cells that expressed the CD71, CD33, and CD10 antigens, concurrently stained brightly with anti-CD38 monoclonal antibodies (MoAbs). In addition, the CD38 antigen was brightly expressed on early T lymphocytes in human thymus, characterized by CD34, CD5, and CD7 expression. Only 1% of the CD34+ cells, 0.01% of nucleated cells in normal BM, did not express the CD38 antigen. The CD34+, CD38- cell population lacked differentiation markers and were homogeneous primitive blast cells by morphology. In contrast the CD34+, CD38 bright cell populations were heterogeneous in morphology and contained myeloblasts and erythroblasts, as well as lymphoblasts. These features are in agreement with properties expected from putative pluripotent hematopoietic stem cells; indeed, the CD34 antigen density decreased concurrently with increasing CD38 antigen density suggesting an upregulation of the CD38 antigen on differentiation of the CD34+ cells. Further evidence for a strong enrichment of early hematopoietic precursors in the CD34+, CD38- cell fraction was obtained from culture experiments in which CD34+ cell fractions with increasing density of the CD38 antigen were sorted singularly and assayed for blast colony formation. On day 14 of incubation, interleukin-3 (IL-3), IL-6, and GM-CSF, G-CSF, and erythropoietin (Epo) were added in each well. Twenty-five percent of the single sorted cells that expressed CD34 but lacked CD38 antigen gave rise to primitive colonies 28 to 34 days after cell sorting. The ability to form primitive colonies decreased rapidly with increasing density of the CD38 antigen. During 120 days of culture, up to five sequential generations of colonies were obtained after replating of the first-generation primitive colonies. This study provides direct evidence for the existence of a single class of progenitors with extensive proliferative capacity in human BM and provides an experimental approach for their purification, manipulation, and further characterization.
Article
Using multidimensional flow cytometry we have defined and quantified the human T-cell differentiation pathway, focusing on those events occurring among the most immature thymocytes and putative bone marrow (BM) T-precursors. Early thymocytes were found to express the CD34 antigen and consisted of a mean 1.2% of cells within human pediatric (n = 9) and 2.0% in fetal thymi (n = 4). All CD34+ thymocytes were atypical blast by morphology, expressed intracytoplasmatic, but not cell surface, CD3, and were cell surface CD2+, CD5+, CD7+, CD38+, CD45+, CD45RA+, CD49d+, and LECAM-1(Leu8)high. CD34high thymocytes lacked surface expression of CD4 and CD8, but as CD34 expression diminished there was a coordinate increase in CD4 levels, followed by the appearance of CD8. The expression of CD1 and CD10 also increased concomitant with the loss of CD34, whereas expression of LECAM-1 diminished with CD34 downregulation. The differential expression of these antigens on early thymocytes (as well as the number of thymocytes displaying these patterns) was highly reproducible among the nine pediatric and four fetal specimens examined, suggesting a precise, stereotyped regulation of early differentiation events. Cell populations with antigen expression patterns suggestive of pluripotent stem cell (CD34high, CD38-), or non-T-lineage committed stem cells (CD34+, CD33+ or CD34+, CD19+) were not identified in either fetal or pediatric thymi (sensitivity = 1/10(4)). The presence of cells with the antigenic profile of the earliest CD34+ thymocytes was explored in human BM. Putative BM T-cell precursors with the appropriate phenotype (CD34+, CD7+, CD5+, CD2+, LECAM-1high) were readily identified in fetal specimens (constituting +/- 2% of the CD34+ population), but could not be reliably detected in adults. In contrast with thymi, only 13% of these cells expressed cytoplasmatic CD3, suggesting the presence of the immediate precursor of the putative prothymocyte population. This was further supported by the detection of CD34bright, CD7+, CD2-, CD5-, LECAM-1moderate cells in fetal specimens. Our results document the flow of cell surface differentiation during T-lymphopoiesis and suggest that T-lineage features are first acquired in the BM. The ability to reproducibly identify and isolate T-cell precursor populations of precisely defined maturational stage in marrow and thymus by multiparameter flow cytometry will facilitate characterization of the molecular events controlling T-lineage differentiation.
Article
A major unresolved paradox in immunology remains: how do we avoid harm, despite the abundant opportunities for induction of immune responses against self-proteins? Here, Mohammad Ibrahim, Benjamin Chain and David Katz extend Janeway's proposed explanation, arguing that adaptive immune responses are initiated not only by conserved microbial products, but also by microenvironmental tissue injury. They suggest that the key step is local dendritic cell activation, followed by upregulation of T-cell costimulatory molecules on these cells, and migration, leading to antigen presentation
Article
Dendritic cells isolated from thymus and tonsil were tested for susceptibility to HIV-1 strains that are tropic for macrophages or for T cell lines. DCs were purified by cell sorting and before infection expressed high levels of CD4 and HLA-DR and lacked markers for T, B, NK cells, or macrophages. Viral entry and reverse transcription was found after pulsing with strains of HIV-1 that could infect macrophages. During the first 36 h the PCR signals for gag sequences increased in DCs and macrophages. In contrast little if any viral DNA was found after pulsing macrophages or DCs with HIV-1 that was able to infect T cell lines. DCs pulsed with HIV-1 were able to transmit infection to responding T cells during an allogeneic or superantigen response. Selection for virus able to infect lymphoid DCs and other DCs expressing CD4 and its transfer to T cells during subsequent immune responses may provide a mechanism for the observed predominance of macrophage-tropic HIV-1 after in vivo transmission.
Article
B7-2 is a recently discovered, second ligand for the CTLA-4/CD28, T cell signaling system. Using the GL-1 rat monoclonal antibody (mAb), we monitored expression of B7-2 on mouse leukocytes with an emphasis on dendritic cells. By cytofluorography, little or no B7-2 was detected on most cell types isolated from spleen, thymus, peritoneal cavity, skin, marrow, and blood. However, expression of B7-2 could be upregulated in culture. In the case of epidermal and spleen dendritic cells, which become highly immunostimulatory for T cells during a short period of culture, the upregulation of B7-2 was dramatic and did not require added stimuli. Lipopolysaccharide did not upregulate B7-2 levels on dendritic cells, in contrast to macrophages and B cells. By indirect immunolabeling, the level of staining with GL-1 mAb exceeded that seen with rat mAbs to several other surface molecules including intercellular adhesion molecule 1, B7-1, CD44, and CD45, as well as new hamster mAbs to CD40, CD48, and B7-1/CD80. Of these accessory molecules, B7-2 was a major species that increased in culture, implying a key role for B7-2 in the functional maturation of dendritic cells. B7-2 was the main (> 90%) CTLA-4 ligand on mouse dendritic cells. When we applied GL-1 to tissue sections of a dozen different organs, clear-cut staining with B7-2 antigen was found in many. B7-2 staining was noted on liver Kupffer cells, interstitial cells of heart and lung, and profiles in the submucosa of the esophagus. B7-2 staining was minimal in the kidney and in the nonlymphoid regions of the gut, and was not observed at all in the brain. In the tongue, only rare dendritic cells in the oral epithelium were B7-2+, but reactive cells were scattered about the interstitial spaces of the muscle. In all lymphoid tissues, Gl-1 strongly stained certain distinct regions that are occupied by dendritic cells and by macrophages. For dendritic cells, these include the thymic medulla, splenic periarterial sheaths, and lymph node deep cortex; for macrophages, the B7-2-rich regions included the splenic marginal zone and lymph node subcapsular cortex. Splenic B7-2+ cells were accessible to labeling with GL-1 mAb given intravenously. Dendritic cell stimulation of T cells (DNA synthesis) during the mixed leukocyte reaction was significantly (35-65%) blocked by GL-1.(ABSTRACT TRUNCATED AT 400 WORDS)