ArticlePDF Available

Reciprocal Control of T Helper Cell and Dendritic Cell Differentiation

Authors:

Abstract and Figures

It is not known whether subsets of dendritic cells provide different cytokine microenvironments that determine the differentiation of either type-1 T helper (TH1) or TH2 cells. Human monocyte (pDC1)–derived dendritic cells (DC1) were found to induce TH1 differentiation, whereas dendritic cells (DC2) derived from CD4+CD3–CD11c–plasmacytoid cells (pDC2) induced TH2 differentiation by use of a mechanism unaffected by interleukin-4 (IL-4) or IL-12. The TH2 cytokine IL-4 enhanced DC1 maturation and killed pDC2, an effect potentiated by IL-10 but blocked by CD40 ligand and interferon-γ. Thus, a negative feedback loop from the mature T helper cells may selectively inhibit prolonged TH1 or TH2 responses by regulating survival of the appropriate dendritic cell subset.
Content may be subject to copyright.
oratories (Bar Harbor, ME). All explants were cultured
on 8-mm polycarbonate filters in Transwell plates
(Costar) at the interface of medium (15% fetal bo-
vine serum in Dulbecco’s modified Eagle’s medium,
L-glutamine, penicillin, and streptomycin). For ecto-
derm separation, tissues were incubated with 6%
pancreatin (Gibco-BRL) for 30 min on ice, washed in
10% serum for 20 min, and peeled (13). NIH 3T3 cell
lines were aggregated by culture in hanging drops
(20). Explant cultures shown are representative of at
least four independent experiments. Rat NT-3 and
human g-actin cDNAs were used as probes for the
Northern blot, which was quantified with a multiim-
ager (Fuji).
7. I. Farin˜as, K. R. Jones, C. Backus, X. Y. Wang, L. F.
Reichardt, Nature 369, 658 (1994).
8. A. Patapoutian and L. F. Reichardt, unpublished
observations.
9. D. J. Epstein, M. Vekemans, P. Gros, Cell 67, 767
(1991).
10. T. Franz, Anat. Embryol. 187, 371 (1993).
11. L. C. Schecterson and M. Bothwell, Neuron 9, 449
(1992).
12. G. A. Wilkinson, I. Farin˜as, C. Backus, C. K. Yoshida,
L. F. Reichardt, J. Neurosci. 16, 7661 (1996).
13. A. Neubu¨ser, H. Peters, R. Balling, G. R. Martin, Cell
90, 247 (1997).
14. R. L. Johnson and C. J. Tabin, ibid., p. 979.
15. B. A. Parr, M. J. Shea, G. Vassileva, A. P. McMahon,
Development 119, 247 (1993).
16. A. Kispert, S. Vainio, A. P. McMahon, ibid. 125, 4225
(1998).
17. K. M. Cadigan and R. Nusse, Genes Dev. 11, 3286
(1997).
18. C. van Genderen et al.,ibid. 8, 2691 (1994).
19. I. Farin˜as, G. A. Wilkinson, C. Backus, L. F. Reichardt, A.
Patapoutian. Neuron 21, 325 (1998).
20. T. Serafini et al.,Cell 78, 409 (1994).
21. We thank N. Hong, U. Mu¨ller, A. Neubu¨ser, and
members of Reichardt lab for valuable comments and
A. Saulys for technical help. This work was supported
by research grants from the U.S. Public Health Service
(National Institutes of Health grant MH48200) and
the Howard Hughes Medical Institute. A.P. is a fellow
of the Damon Runyon–Walter Winchell Cancer Re-
search Foundation and L.F.R. is an investigator of the
Howard Hughes Medical Institute.
7 October 1998; accepted 21 January 1999
Reciprocal Control of T Helper
Cell and Dendritic Cell
Differentiation
Marie-Clotilde Rissoan,
1
* Vassili Soumelis,
1
*
Norimitsu Kadowaki,
2
* Geraldine Grouard,
1
Francine Briere,
1
Rene´ de Waal Malefyt,
2
Yong-Jun Liu
1,2
It is not known whether subsets of dendritic cells provide different cytokine
microenvironments that determine the differentiation of either type-1 T helper
(T
H
1) or T
H
2 cells. Human monocyte (pDC1)– derived dendritic cells (DC1) were
found to induce T
H
1 differentiation, whereas dendritic cells (DC2) derived from
CD4
1
CD3
CD11c
plasmacytoid cells (pDC2) induced T
H
2 differentiation by
use of a mechanism unaffected by interleukin-4 (IL-4) or IL-12. The T
H
2 cytokine
IL-4 enhanced DC1 maturation and killed pDC2, an effect potentiated by IL-10
but blocked by CD40 ligand and interferon-g. Thus, a negative feedback loop
from the mature T helper cells may selectively inhibit prolonged T
H
1orT
H
2
responses by regulating survival of the appropriate dendritic cell subset.
The cytokine microenvironment plays a key
role in T helper cell differentiation toward the
T
H
1orT
H
2 cell type during immune respons-
es (16). IL-12 induces T
H
1 differentiation,
whereas IL-4 drives T
H
2 differentiation. Be-
cause T helper cell differentiation requires
the presence of different cytokines at an ini-
tial stage of the T cell– dendritic cell (DC)
interaction (17), we investigated whether
distinct DC lineages or subsets may produce
different cytokines and directly induce T
H
1
or T
H
2 differentiation.
Humans have two distinct types of DC pre-
cursors. Peripheral blood monocytes (designat-
ed pDC1) give rise to immature myeloid DCs
after culturing with granulocyte-macrophage
colony-stimulating factor (GM-CSF) and IL-4
(810) or after transmigration through endothe-
lial cells and phagocytosis (11). These imma-
ture cells become mature myeloid DCs (desig-
nated DC1) after stimulation with CD40 ligand
(CD40L) or endotoxin (12, 13). The
CD4
1
CD3
CD11c
plasmacytoid cells (desig-
nated pDC2) from blood or tonsils give rise to
a distinct type of immature DC after culture
with IL-3 (1416 ). These cells differentiate into
mature DCs (designated DC2) after CD40L
stimulation (17). However, unlike pDC1 and
DC1, pDC2 and DC2 display features of the
lymphoid lineage: (i) pDC2 and DC2 express
few myeloid antigens, such as CD11b, CD11c,
CD13, and CD33 (14); (ii) pDC2 cells do not
differentiate into macrophages, following cul-
ture with GM-CSF and M-CSF; (iii) pDC2 and
DC2 have little capacity to phagocytose or
macropinocytose antigens at all stages of their
maturation (14) (iv) like the putative mouse
lymphoid DCs (18), pDC2 cells depend on
IL-3, but not GM-CSF for their survival and
maturation (14) [this can be explained by high
GM-CSF receptor and low IL-3 receptor ex-
pression in pDC1 cells and low GM-CSF re-
ceptor and high IL-3 receptor expression in
pDC2 cells (Fig. 1)]; and (v) pDC2 cells have
high levels of pre–T cell receptor a-chain ex-
pression (19).
Both myeloid DC1 and “lymphoid” DC2
induce strong proliferation of allogeneic naı¨ve
CD4
1
T cells (14). First, we examined the
profile of cytokine production from DC1 and
DC2 after CD40L activation. DC1 produced
large amounts of IL-12 within 24 hours after
CD40L activation (Fig. 2) as reported (12, 13),
whereas DC2 did not (Fig. 2). In addition,
unlike CD40L-activated DC1, CD40L-activat-
ed DC2 produced little IL-1a, IL-1b, IL-6, and
IL-10, but produced comparable amounts of
chemokine IL-8 (Fig. 2) (20). Neither CD40L-
activated DC1 nor DC2 produced detectable
amounts of IL-4 and IL-13. Quantitative poly-
merase chain reaction (PCR) analyses showed
that CD40L activation up-regulated the expres-
sion of mRNA for IL-12p40, IL-1a, and IL-1b
in DC1, but not in DC2 (Table 1) (21). Neither
DC1 nor DC2 transcribed detectable amounts
of IL-4 mRNA, either before or after CD40L
activation (Table 1).
We next examined the nature of primary
allogeneic T cell responses induced by DC1 or
DC2. Naı¨ve CD4
1
CD45RA
1
T cells isolated
from human peripheral blood or umbilical cord
blood were cocultured for 7 days with CD40L-
activated DC1, CD40L-activated DC2, or anti-
bodies to CD3 and CD28 (22). The cultured
cells were counted and restimulated with anti-
CD3 and anti-CD28 for either 4 hours for sin-
gle-cell cytokine analyses by flow cytometry
(Fig. 3B) or 24 hours for cytokine secretion
analyses by enzyme-linked immunosorbent as-
say (ELISA) (Fig. 3A). T cells originally cul-
1
Schering-Plough, Laboratory for Immunological Re-
search, 27 chemin des Peupliers, Boite Postale 11,
69571, Dardilly, France.
2
DNAX Research Institute of
Molecular and Cellular Biology, 901 California Ave-
nue, Palo Alto, CA 94304–1104, USA.
*These authors contributed equally to this work.
To whom correspondence should be addressed. E-
mail: yliu@dnax.org
Fig. 1. Expression of GM-CSF receptor achain
(GM-CSF Ra) and IL-3 receptor achain (IL-3
Ra) on pDC1 and pDC2 (open curve, isotype
control; shaded curve, specific staining).
REPORTS
www.sciencemag.org SCIENCE VOL 283 19 FEBRUARY 1999 1183
tured with DC1 secreted large amounts of in-
terferon-g(IFN-g) (34 to 37 ng/ml, from three
independent experiments), but little IL-4, IL-5,
and IL-10. T cells originally cultured with DC2
secreted little IFN-g(2 to 4 ng/ml), but large
amounts of IL-4 (230 to 1500 pg/ml), IL-5 (300
to 900 pg/ml), and IL-10 (4 to 10 ng/ml). T cells
originally cultured with anti-CD3 and anti-
CD28 secreted mainly IL-2 (Fig. 3A). These
polarized cytokine production profiles were
confirmed by single-cell cytokine analysis us-
ing in situ immunocytology (23) and by immu-
nofluorescence flow cytometry (Fig. 3B) (24).
Thus, myeloid DC1 and “lymphoid” DC2, re-
spectively, induce T
H
1 versus T
H
2 differentia-
tion in vitro.
Because DC2 cells do not produce detect-
able amounts of IL-4, as determined by both
mRNA (to a sensitivity of 10
–12
gram) and
protein analysis, it suggests that the DC2-in-
duced T
H
2 differentiation is IL-4 –independent.
T
H
2 development was not blocked by adding
either polyclonal antibody to IL-4 BDA11 (15
mg/ml) or monoclonal antibody (mAb) to IL-4
MP4-25D2 (5 mg/ml) at the beginning of DC–T
cell coculture (22). These two antibodies to
IL-4 completely inhibited the IL-4 dependent
proliferation of phytohemagglutinin-stimulated
human T cells or CD40- and IL-4 dependent
human B cell proliferation and immunoglobulin
E synthesis. Although addition of antibody to
IL-4 increased the number of IFN-g–producing
cells, it did not block the generation of IL-4
producing cells (Fig. 3B). The DC2-induced
T
H
2 differentiation was not a default mecha-
nism due to an inability of DC2s to produce
IL-12, because polyclonal activation with anti-
bodies to CD3 and CD28 in the absence of
Fig. 2. Cytokine and chemokine secretion by DC 1 after 24 hours of activation with CD40L (open
circles) and DC2 after CD40L activation for 24 hours (solid squares) or 6 days (solid circles). Each
symbol represents an independent experiment.
Fig. 3. DC1 and DC2 in-
duce T
H
1 versus T
H
2 cyto-
kine production, respec-
tively. (A) Quantitation of
T
H
1 and T
H
2 cytokines
by ELISA. Human CD4
1
CD45RO
2
naı¨ve T cells
were cultured for 6 days
with allogeneic CD40L-
activated DC1 or DC2,
or anti-CD3 plus anti-
CD28aCD3/aCD28). Cells
were counted and restim-
ulated with anti-CD3 and
anti-CD28 for 24 hours.
Amounts of IFN-g, IL-4,
IL-5, IL-10, and IL-2 within
culture supernatants were
collected after 24 hours
and measured by ELISA.
Results represent one of the three independent
experiments. (B) Two-color analysis of IL-4 and
IFN-gexpression by flow cytometry. (Upper pan-
els) DC1–T cell cocultures with control goat im-
munoglobin G antibodies and goat antibody to
IL-12. (Middle panels) DC2–T cell coculture with
control antibody and goat antibody to IL-4. (Low-
er panels) DC2–T cell coculture with anti–IL-12
and IL-12. Some 10
4
cells were analyzed, and the
percentages of each T cell population are indicat-
ed in the plots. Figure 4 represents the results
from one of the four independent experiments
performed.
REPORTS
19 FEBRUARY 1999 VOL 283 SCIENCE www.sciencemag.org1184
IL-12 did not induce T
H
2 differentiation (Fig.
3A). To support this conclusion, we performed
two experiments. First, neutralizing antibody to
IL-12 (AB-219-NA, 25 mg/ml) was added at
the beginning of the DC–T cell cocultures to
see whether anti–IL-12 could induce IL-4 –pro-
ducing T cells in DC1–T cell culture or increase
the number of IL-4 –producing cells in the
DC2–T cell cocultures. Although addition of
antibody to IL-12 decreased the percentage of
IFN-g–producing cells in both DC1–T cell and
DC2–T cell cultures (Fig. 3B), it did not induce
IL-4 –producing cells nor did it significantly
increase IL-4 –producing cell number in
DC1–T cell or DC2–T cell cultures (Fig. 3B).
Second, IL-12 (5 ng/ml) was added at the be-
ginning of DC2–T cell cocultures, to see if it
could block the generation of IL-4 –producing
T cells. Although the addition of IL-12 in-
creased the percentages of IFN-g–producing T
cells, it did not inhibit the number of IL-4
producing cells. However, IL-12 induced the
IL-4 –producing cells to produce IFN-g(Fig.
3B). Thus, DC2 may produce one or more
positive T
H
2 differentiation factors distinct
from IL-4, and its activity can neither be
blocked by IL-12 nor enhanced by anti–IL-12.
A common feature of T cell cytokine-medi-
ated T helper cell differentiation is the positive
autocrine effect. IL-2 promotes the IL-2–pro-
ducing T
H
0 cells, IL-4 promotes the IL-4
producing T
H
2 cells, IFN-gpromotes the IFN-
g–producing T
H
1 cells, IL-10 promotes the IL-
10 –producing regulatory CD4
1
T cells (25),
and TGF-bpromotes the TGF-b–producing
T
H
3 cells (26 ). Because negative feedback reg-
ulation represents a general mechanism used by
living organisms to maintain homeostasis of
physiological processes, the immune system
may need a negative feedback mechanism to
control the balance between T
H
1 and T
H
2 re-
sponses in order to prevent T
H
-mediated auto-
immune inflammatory responses or T
H
2-medi-
ated allergic responses. The studies on the reg-
ulation of DC1 and DC2 maturation allowed us
to identify a potential negative feedback loop in
which IL-4 and IFN-gmay negatively regulate
T
H
1 and T
H
2 development, respectively, by
interfering with the survival and maturation of
pDC1 and pDC2.
We observed that IL-4, IL-10, and CD40L
inhibited the IL-3– dependent proliferation of
pDC2 (Fig. 4A, a) (27). In contrast to CD40L,
which enhanced the survival and maturation of
pDC2, IL-4 and IL-10 decreased pDC2 num-
bers during a 6-day culture period in the pres-
ence IL-3 in a concentration-dependent fashion
(Fig. 4A, b and c) (27). IL-4 and IL-10 have
an additive effect in killing pDC2 (Fig. 4B).
The ability of IL-4 and IL-10 to kill pDC2 by
apoptosis was confirmed by direct culture
morphology, Giemsa staining of cytospin
preparations, and double-staining with an-
nexin–fluorescein isothiocyanate (FITC) and
propidiumiodide.
Because IL-4 and IL-10 are T
H
2 cyto-
kines, we investigated whether CD40L and
IFN-gcould block the negative effect of IL-4
and IL-10 on the survival of pDC2 main-
tained by IL-3. CD40L blocked the killing
effects of IL-4 or IL-10 on pDC2 during a
6-day culture period with IL-3 (Fig. 5A).
CD40L partially rescued pDC2 in the pres-
ence of both IL-4 and IL-10. IFN-galso
blocked the negative effect of IL-4 or IL-10
Fig. 4. (A) IL-4 and IL-10 inhibit the
IL-3–dependent proliferation and sur-
vival of pDC2 in a dose-dependent
fashion. (a) The IL-3–dependent
3
H-
thymidine incorporation by pDC2 at
day 3 of culture is suppressed by IL-4,
IL-10, and CD40L. In contrast to
CD40L, which enhances the survival of
pDC2, IL-4 and IL-10 (b and c) de-
crease in a dose-dependent fashion
the numbers of viable cells after 6
days of culture. Cell viability was de-
termined by trypan blue exclusion.
Results are expressed as means 6SD
of culture triplicates. One representa-
tive of eight independent experiments
is shown. (B) IL-4 and IL-10 have ad-
ditive effects in killing pDC2 over a 6-day time course. Kinetics of cell survival at days 1, 3, and 6 of
culture with IL-3 alone, IL-3 1IL-4, IL-3 1IL-10, and IL-3 1IL-4 1IL-10. The initial input of cells was
45,000 or 25,000 per well. Each symbol represents one independent experiment.
Fig. 5. CD40L and
IFN-grescue pDC2
from cell death in-
duced by IL-4 and IL-
10. (A) CD40L rescues
a large proportion of
DC2 precursors after 6
days of culture with
combinations of IL-3,
IL-4, and IL-10. (B)
IFN-grescues a large
fraction of DC2 pre-
cursors after 6 days of
culture with IL-3 1
IL-4 and IL-3 1IL-10.
It did not rescue DC2
precursors when cultured with IL-3 1IL-4 1IL-10. Results are representative of three independent
experiments.
Table 1. Quantitation of cytokine mRNA by PCR
(expressed as femtograms mRNA per 50 ng cDNA)
(21). Results before (–) and after (1) CD40L acti-
vation are shown after experiment number.
Expt.
number
(CD40L)
IL-12p40 IL-1aIL-1bIL-4
DC1
1 (–) 38 34 41 0
1(1) 12,835 573 2,175 0
2 (–) 7 45 60 2
2(1) 12,460 346 4,165 0
3 (–) 5 44 45 0
3(1) 40,194 441 763 0
DC2
1 (–) 0 12 20 0
1(1) 605 9 16 0
2 (–) 0 37 102 0
2(1) 714 21 50 0
3 (–) 0 7 14 0
3(1) 0 12 45 0
REPORTS
www.sciencemag.org SCIENCE VOL 283 19 FEBRUARY 1999 1185
on pDC2 (Fig. 5B). However, IFN-gdid not
rescue the cells when IL-4 and IL-10 were
both added to the culture. Cells rescued by
either CD40L or IFN-gexpressed high levels
of major histocompatibility complex (MHC
class II DR) and costimulatory molecules
(B7.1/CD80 and B7.2/CD86), and stimulated
the proliferation of allogeneic CD4
1
T cells.
Our study suggests that a negative feed-
back loop may exist in regulating the balance
between T
H
1 versus T
H
2 responses. IL-4, a
key T
H
2 cytokine, kills the pDC2, a profes-
sional antigen-presenting cell subset that in-
duces T
H
2 differentiation. The ability of
CD40L (a potent DC maturation factor) to
prevent IL-4 –induced killing suggests that
IL-4 cannot kill mature DC2 during their
cognate interaction with T cells in established
responses. This may allow the rapid and ef-
ficient development of T
H
2 responses needed
for the host defense. However, overproduc-
tion of IL-4 may inhibit the development of
pDC2. By contrast, IL-4 promotes DC1 mat-
uration together with GM-CSF (12, 13).
These opposing effects of IL-4 on DC1 ver-
sus DC2 may enhance T
H
1 development, but
inhibit T
H
2 development at a late stage of
immune response. The ability of IFN-gto
protect pDC2 from IL-4 –and IL-10 –induced
apoptosis and promote DC2 differentiation
may represent an indirect mechanism to in-
hibit T
H
1 development at later stages of T
H
1
responses. This represents another example
of antagonism between IL-4 and IFN-g(28).
During the last two decades, studies on the
relationship between lineage and function have
been a main focus of B and T lymphocyte
immunology. Mouse DC cells may also have
different lineages with distinct functions (29
35). Whereas the CD8a
CD11c
1
CD11b
1
my-
eloid DC cells are immunogenic for T cells, the
CD8a
1
CD11c
1
CD11b
lymphoid DC cells
may be tolerogenic (31). This concept is sup-
ported by two findings: (i) the lymphoid DC
subset that appears to be localized within the T
cell areas of mouse spleen highly express MHC
class II–self peptide complexes (32); and (ii)
the myeloid subset that appears to be localized
around the marginal zone– bridging channels
migrates into the T cell areas and produces
IL-12 after endotoxin stimulation (33, 34). Our
results here extend the concept regarding the
functional heterogeneity of DC subsets and
suggest two additional mechanisms for T
H
1 and
T
H
2 regulation. DC1 and DC2 stimulate naı¨ve
T helper cells and directly induce their differ-
entiation toward T
H
1orT
H
2. pDC1 and pDC2
provide the potential targets for negative feed-
back regulation by IL-4 and IFN-g. Two im-
portant relationships still need to be established:
(i) the relationship between the mechanisms
regulating immunity/tolerance versus T
H
1/T
H
2
and (ii) the correlation between mouse and
human DC subsets. These studies may ulti-
mately lead to the understanding of the molec-
ular mechanism underlying DC2-induced IL-
4 –independent T
H
2 differentiation and the dis-
tinct functions of DC subsets in normal and
disease states.
References and Notes
1. T. R. Mosmann and R. L. Coffman, Annu. Rev. Immu-
nol. 7, 145 (1989).
2. W. E. Paul and R. A. Seder, Cell 76, 241 (1994).
3. S. Romagnani, Annu. Rev. Immunol. 12, 227 (1994).
4. A. K. Abbas, K. M. Murphy, A. Sher, Nature 383, 787
(1996).
5. S. L. Constant and K. Bottomly, Annu. Rev. Immunol.
15, 297 (1997).
6. A. O’Garra, Immunity 8, 1 (1998).
7. J. Banchereau and R. Steinmann, Nature 392, 245
(1998).
8. F. Sallusto and A. Lanzavecchia, J. Exp. Med. 179,
1109 (1994).
9. N. S. Romani et al.,ibid. 180, 83 (1994).
10. Generation of DC1 from monocytes in vitro. Blood
mononuclear cells were isolated as described (8, 9).
To generate DC1, monocytes were cultured with
GM-CSF (200 ng/ml) and IL-4 (5 ng/ml; Schering-
Plough, Kenilworth, NJ) for 5 days in RPMI-1640
medium supplemented with 10% fetal bovine serum
(FBS), 2 mML-glutamine, and 0.08 mg/ml gentamy-
cin. The resulting immature DC1 were cultured with
CD40L-transfected L cells (one L cell per four DC
cells) for 24 hours.
11. G. J. Randolph, S. Beaulieu, S. Lebecque, R. M. Stein-
man, W. A. Muller, Science 282, 480 (1998).
12. M. D. Cella et al.,J. Exp. Med. 184, 747 (1996).
13. F. Koch et al.,ibid., p. 741.
14. G. Grouard et al.,ibid. 185, 110 (1997).
15. U. O’Doherty et al.,J. Immunol. 82, 487 (1994).
16. J. Olweus et al.,Proc. Natl. Acad. Sci. U.S.A. 94,
12551 (1997).
17. Generation of DC2 from the CD4
1
CD11c
lineage
pDC2 precursors. CD4
1
CD11c
lineage
plasmacytoid
cells were isolated from human tonsils or peripheral
blood as detailed (13). In two experiments, pDC1 and
pDC2 were isolated from the same blood donor.
These cells were cultured with IL-3 in the presence or
absence of CD40L-transfected L cells for 6 days or
with IL-3 alone for 5 days, followed by culture with
CD40L-transfected L cells for 24 hours.
18. D. Saunderset al.,J. Exp. Med. 184, 2185 (1996).
19. L. Bruno, P. Res, M. Dessing, M. Cella, H. Spits, ibid.
185, 875 (1997).
20. Quantitation of cytokine secretion by ELISA. The
presence of cytokines in culture supernatants of DC1
and DC2 after 24 hours of CD40L activation or in T
cell cultures for 24 hours with anti-CD3 and anti-
CD28 was determined by ELISA. ELISA kits for IL-1a,
IL-1b, IL-5, IL-6, IL-10, IL-12, IL-13, and IFN-gwere
obtained from R&D Systems (Minneapolis, MN); for
IL-2 and IL-4 from Cayman Chemical (Ann Arbor, MI).
21. Quantitation of mRNA expression. RNA isolation was
according to P. Chomcznski and N. Sacchi [Anal. Bio-
chem. 162, 156 (1987)]. The reverse transcription was
performed with SuperScriptII (Gibco-BRL, Rockville,
MD). We analyzed 50 ng of cDNA for the expression of
cytokine genes by the Fluorogenic 59-nuclease PCR
assay [P. M. Holland et al.,Proc. Natl. Acad. Sci. U.S.A.
88, 7276 (1991)], using a Perkin-Elmer ABI Prism 7700
Sequence Detection System (SDS; ABI–Perkin-Elmer,
Foster City, CA). Reactions were incubated for 2 min at
50°C, denatured for 10 min at 95°C and subjected to 40
two-step amplification cycles with annealing/extension
at 60°C for 1 min followed by denaturation at 95°C for
15 s. The following amplicons were used and analyzed
with 6-carboxy-fluorescein–labeled predeveloped Tag-
man assay reagents (Perkin-Elmer, Foster City, CA):
IL-1a, IL-1b, IL-4, and IL-12p40. Cytokine amplicons
spanned at least one intron/exon boundary. An 18S
ribosomal RNA amplicon was analyzed with a labeled
probe (Perkin-Elmer, Foster City, CA) and used as an
internal control for quantitation of the total amount of
cDNA in a multiplex reaction. Seven 10-fold dilutions of
plasmids (10 ng/ml) containing cytokine cDNAs were
used to create a standard curve for quantitation of
cytokine cDNA using the SDS software, then these
values were adjusted for the amount of total cDNA.
Values are expressed as femtogram of cDNA per 50 ng
input total RNA.
22. Purification of naı¨ve T cells and DC–T cell cocultures.
CD4
1
CD45RA T cells were incubated with a cocktail
of mAbs, including IOM2 (CD14); ION16 (CD16);
ION2 (HLA-DR) (Immunotech); NKH1 (CD56); OKT8
(CD8) (Ortho); 4G7 (CD19); UCHL1 (CD45RO); and
mAb 89 (CD40). This was followed by incubation
with anti-mouse immunoglobin-coated magnetic
beads and magnetic depletion. This was repeated two
times to create .96% pure CD4
1
naı¨ve T cells. T
cells were cocultured with allogeneic DC1 or DC2 at
a 2:1, 4 :1, and 8 :1 ratios in Yssel’s medium (Irvine
Scientific, Santa Ana, CA) containing 10% FBS in
24-well culture plates for 6 days with or without: (i)
polyclonal antibody to IL-4 (BDA11, 15 mg/ml; R&D
Systems); (ii) mAb to IL-4 (MP4-25D2; Pharmingen);
(iii) antibody to IL-12 (AB-219-NA, 25 mg/ml; goat
polyclonal immunoglobin G, R&D Systems); and (iv)
IL-12 (5 ng/ml; R&D Systems). T cells were also
cultured with anti-CD3 (5 mg/ml) and anti-CD28 (1
mg/ml; Pharmingen) coated on culture plates for 6
days. After 6 days of priming, T cells were restimu-
lated with anti-CD3 and anti-CD28 for either 4 or 24
hours.
23. Immunostaining of cytokines on cytospins. Cytospin
slides were fixed in cold paraformaldehyde for 5 min
and washed in Hank’s balanced salt solution. For
IL-12p40 staining, the slides were incubated with
mAb 609 (5 mg/ml; R&D Systems) or an isotype
control mAb for 45 min. The binding of antibody was
revealed by a Vectastain ABC kit ( Vector Laboratory,
Burlingame, CA). DAB chromogen was used to reveal
the peroxidase activity. For IFN-gstaining, slides
were incubated with mAb B27 (J. Abrams, DNAX
Research Institute of Molecular and Cellular Biology)
followed by staining with the Vectastain ABC kit.
24. Flow cytometry analysis of intracellular cytokines. After
6 days of DC–T cell coculture, T cells were reactivated
with anti-CD3 and anti-CD28 for 5 hours. Brefeldin A (1
mg/ml; Sigma) was added into the cultures for 2 hours
before the staining to prevent cytokine secretion. Cells
were washed and fixed with paraformaldehyde and
incubated with the following antibodies (Pharmingen):
anti-IL-4 –phycoerythrin (PE) plus anti-IFN-g–FITC; anti-
IL-10–PE plus anti-IFN-g–FITC; and anti-IL-4–FITC plus
anti-IL-10–PE. Samples were analyzed on a FACScan
(Becton-Dickinson).
25. H. Groux et al.,Nature 389, 737 (1997).
26. R. A. Seder et al.,J. Immunol. 160, 5719 (1998).
27. pDC2 were cultured in complete medium alone (RPMI-
1640 medium supplemented with 10% FBS, 2 mM
L-glutamine, and 40 mg/ml gentamycin) or with differ-
ent cytokines. Cytokines used were IL-3 (10 ng/ml), IL-4
(50 U/ml), IL-10 (100 ng/ml), and IL-13 (50 ng/ml) from
Schering-Plough Research Institute, Kenilworth, NJ, and
IFN-g(25 ng/ml) and IL-2 (20 U/ml) from Genzyme,
Boston, MA. Irradiated CD40L-transfected L cells were
used at a concentration of one L cell per four DC2 cells.
Nontransfected or CD32-transfected L cells were used
as controls. We cultured 3 310
4
cells per well in
triplicate in 96-well plates. After 3 days, cells were
pulsed with 1 mCi of
3
H-thymidine for 12 hours before
harvesting and of counting. Viable and dead cell num-
bers at 1, 3, and 6 days after culture were determined
by trypan blue exclusion assay.
28. U. Boehm, T. Klamp, M. Groot, J. C. Howard, Annu.
Rev. Immunol. 15, 749 (1997).
29. C. Ardavin, L. Wu, C. L. Li, K. Shortman, Nature 362,
761 (1993).
30. L. Wu, C.-H. Li, K. Shortman, J. Exp. Med. 184, 903
(1996).
31. G. Suss and K. Shortman, ibid. 183, 1789 (1996).
32. K. Inaba et al.,ibid. 186, 665 (1997).
33. T. De Smedt et al.,ibid. 184, 1413 (1996).
34. C. R. e Sousa et al.,ibid. 186, 1819 (1997).
35. B. Pulendran et al.,J. Immunol. 159, 2222 (1997).
36. We thank L. Lanier, R. Coffman, A. O’Garra, H. Kanzler,
and K. Moore for critical reading of the manuscript, S.
Ho, P. Larenas, B. Bennett, and I. Durand for technical
help, and J. Katheiser for editorial assistance. DNAX
Research Institute of Molecular and Cellular Biology is
supported by Schering-Plough Corporation.
10 November 1998; accepted 11 January 1999
REPORTS
19 FEBRUARY 1999 VOL 283 SCIENCE www.sciencemag.org1186
... In addition, one must consider whether different DC subsets are endowed with unique capacities to induce Th1, Th2, or Treg responses. In humans, monocyte-derived CD11c + DCs predominantly polarize naive T cells toward a Th1 profile [45], whereas the CD11c -DC subset induces T cells to predominantly produce Th2 cytokines [46]. The extent of T-cell polarization by CD11c -DCs may be related to their differentiation/maturation stages. ...
Article
Full-text available
Dendritic cells (DCs) are a heterogeneous group of antigen-presenting cells crucial for fostering allograft tolerance while simultaneously supporting host defense against infections and cancer. Within the tumor microenvironment, DCs can either mount an immune response against cancer cells or foster immunotolerance, presenting a dual role. In immunocompromised individuals, posttransplant malignancies pose a significant health concern, with DCs serving as vital players in immune responses against cancer cells. Both recipient- and donor-derived DCs play a critical role in the rejection process, infiltrating the transplanted organ and sustaining T-cell responses. The use of immunosuppressive drugs represents the predominant approach to control this immunological barrier in transplanted organs. Evidence has shed light on the immunopharmacology of these drugs and novel strategies for manipulating DCs to promote allograft survival. Therefore, comprehending the mechanisms underlying this intricate microenvironment and the effects of immunosuppressive therapy on DCs is crucial for developing targeted therapies to reduce graft failure rates. This review will delve into the fundamental immunobiology of DCs and provide a detailed exploration of their clinical significance concerning alloimmune responses and posttransplant malignancies.
... Dendritic cells are a key factor in the pathogenesis of SLE with abnormalities in the immune system, among which plasmacytoid dendritic cells (pDCs) are closely related to the progression of SLE [37,38] . After pDCs are activated, Th cells can be made to develop into Th2 cells, and then B cells will be activated to secrete a large number of autoantibodies [39] . ...
Preprint
Full-text available
Background: Systemic lupus erythematosus (SLE) is an autoimmune inflammatory connective tissue disease involving multiple organs. As one of the serious complications of SLE, sepsis (SEP) has a high risk of death. Here, the goal of this study was to identify vulnerable biomarkers that could be used to diagnose SLE and SEP. Methods: We used the Limma R software tool and the Gene Expression Omnibus (GEO) database to find differentially expressed genes (DEGs) in SLE. Additionally, genes associated with oxidative stress and immune system function were chosen from the MSigDB database and the Genecard database, respectively. Weighted gene Coexpression network analysis (WGCNA) was used to identify the important module genes associated with SEP. With the help of WGCNA, machine learning, and logistic regression, immunological and oxidative stress-related hub genes were discovered and validated by an external validation set. The analysis was put to the test using consensus clustering. Immune cell infiltration was investigated in SLE and SEP patients. Results: We obtained 957 genes from the GSE6163 dataset and 2559 genes from the significant module of WGCNA, which yielded 46 genes after taking intersection with immune and oxidative stress-related genes. According to the enrichment analysis's findings, the two diseases share a lot of similar immunological and inflammation-related pathways. Machine learning was utilized to pick 11 hub genes, and ROC was employed to evaluate the diagnostic effectiveness. Furthermore, the expression profiles of the hub genes revealed by logistic regression modeling have a significant diagnostic value. Moreover, consensus clustering revealed a favorable correlation between the severity of immunological and oxidative stress and disease activity in SLE and SEP. Analysis of immune infiltration revealed a more consistent immune cell infiltration behavior between SLE and SEP. Conclusion: In this study, the expression of 11 potential hub genes, including TLR2, IL1RN, IRF9, ISG20, TXK, SH2D1A, IL7R, CD28, ITK, CD3E, and CCR7, were thoroughly analyzed using bioinformatics. An efficient logistic regression model was created, and it was possible to identify a correlation between the progression of SLE and SEP disease and the expression of immunological and oxidative stress by consensus clustering. In addition, there is a similar immunoinvasive behavior between the two diseases. It is helpful to identify the biological markers with potential diagnostic value.
... Even extremely low antigen doses are taken up by DCs and exposed in the lymph nodes after a journey through tissues and lymphatic pathways, which can take up to a few days. Rissoan (1999) have showed that DCs with and without the CD11 marker (CD11 + and CD11 − , respectively) produced IFN-γ and IL-4, respectively; the first class was named DC1, and the second, DC2. The selective production of IFN-γ and IL-4 by DC1 and DC2, respectively, seemed to have improved the understanding of lymphocyte response regulation: DC1 would activate the Th1 circuit by producing IFNγ, whereas DC2 would activate Th2 by producing IL-4. ...
... The DCs can communicate with other immune cells in two manners. First, they can communicate through intercellular interactions using specific functional proteins and cell surface markers, such as TLRs [70], lectin-like receptors [71][72][73][74] and, second, they can communicate using soluble factors [70,75,76]. The PAMPs present on the pathogen surface are specific to the PRRs present on the DC surface [77]. ...
Article
Full-text available
In view of the severe downsides of conventional cancer therapies, the quest of developing alternative strategies still remains of critical importance. In this regard, antigen cross-presentation, usually employed by dendritic cells (DCs), has been recognized as a potential solution to overcome the present impasse in anti-cancer therapeutic strategies. It has been established that an elevated cytotoxic T lymphocyte (CTL) response against cancer cells can be achieved by targeting receptors expressed on DCs with specific ligands. Glycans are known to serve as ligands for C-type lectin receptors (CLRs) expressed on DCs, and are also known to act as a tumor-associated antigen (TAA), and, thus, can be harnessed as a potential immunotherapeutic target. In this scenario, integrating the knowledge of cross-presentation and glycan-conjugated nanovaccines can help us to develop so called ‘glyco-nanovaccines’ (GNVs) for targeting DCs. Here, we briefly review and analyze the potential of GNVs as the next-generation anti-tumor immunotherapy. We have compared different antigen-presenting cells (APCs) for their ability to cross-present antigens and described the potential nanocarriers for tumor antigen cross-presentation. Further, we discuss the role of glycans in targeting of DCs, the immune response due to pathogens, and imitative approaches, along with parameters, strategies, and challenges involved in cross-presentation-based GNVs for cancer immunotherapy. It is known that the effectiveness of GNVs in eradicating tumors by inducing strong CTL response in the tumor microenvironment (TME) has been largely hindered by tumor glycosylation and the expression of different lectin receptors (such as galectins) by cancer cells. Tumor glycan signatures can be sensed by a variety of lectins expressed on immune cells and mediate the immune suppression which, in turn, facilitates immune evasion. Therefore, a sound understanding of the glycan language of cancer cells, and glycan–lectin interaction between the cancer cells and immune cells, would help in strategically designing the next-generation GNVs for anti-tumor immunotherapy.
Article
Full-text available
LTX-315 is a synthetic cationic oncolytic peptide with potent anticancer activity but limited toxicity for non-malignant cells. LTX-315 induces both immunogenic tumor cell death and generation of tumor-specific immune responses in multiple experimental tumor models. Given the central role of dendritic cell (DC) maturation in the induction of antigen-specific immunity, we investigated the effect of LTX-315 treatment on the maturation of tumor-infiltrating DCs (TiDCs) and the generation of anti-melanoma immunity. We found that LTX-315 treatment induces the maturation of DCs, both indirectly through the release of cancer cell-derived damage-associated molecular patterns (DAMPs)/alarmins and nucleic acids (DNA and RNA) capable of triggering distinct Toll-like receptor (TLR) signaling, and, directly by activating TLR7. The latter results in the ignition of multiple intracellular signaling pathways that promotes DC maturation, including NF-κB, mitogen activated protein kinases (MAPKs), and inflammasome signaling, as well as increased type 1 interferon production. Critically, the effects of LTX-315 on DCs the consequent promotion of anti-melanoma immunity depend on the cytosolic signal transducer myeloid differentiation response gene 88 (MyD88). These results cast light on the mechanisms by which LTX-315 induces DC maturation and hence elicits anticancer immunity, with important implications for the use of LTX-315 as an anticancer immunotherapeutic.
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.
Chapter
Interactions between the immune and nervous systems are involved in many disease processes. Modulation of inflammation can provide an important opportunity to enhance regeneration within the central nervous system. This authoritative book defines the key cellular players in mounting an inflammatory response and highlights critical factors in the target organ that influence the nature of that response and its capacity either to damage or protect the brain. Several key clinical areas are highlighted – particularly autoimmune diseases of the nervous system including multiple sclerosis, as well as microbiological and traumatic challenges; the book therefore provides both a summary of the basic science background as well as practical, clinical-friendly guidelines to management. The book will be of interest to a wide range of physicians, including neurologists, neurosurgeons, neurorehabilitationists, infectious disease physicians, and clinical neuroscientists, as well as neuroscientists and immunologists.
Article
Full-text available
We investigated the possibility that T helper cells might enhance the stimulatory function of dendritic cells (DCs). We found that ligation of CD40 by CD40L triggers the production of extremely high levels ofbioactive IL-12. Other stimuli such as microbial agents, TNF-cx or LPS are much less effective or not at all. In addition, CD40L is the most potent stimulus in upregu-lating the expression ofICAM-1, CD80, and CD86 molecules on DCs. These effects of CD40 ligation result in an increased capacity of DCs to trigger proliferative responses and IFN-~/pro-duction by T cells. These findings reveal a new role for CD40-CD40L interaction in regulating DC function and are relevant to design therapeutic strategies using cultured DCs.
Article
Full-text available
The 5' -* 3' exonuclease activity of the thermostable enzyme Thermus aquaticus DNA polymerase may be employed in a polymerase chain reaction product detection system to generate a specific detectable signal concomitantly with amplification. An oligonucleotide probe, nonextendable at the 3' end, labeled at the 5' end, and designed to hybridize within the target sequence, is introduced into the polymerase chain reaction assay. Annealing of probe to one of the polymerase chain reaction product strands during the course of amplification generates a substrate suitable for exonuclease activity. During amplification, the 5' -- 3' exonuclease activity of T. aquaticus DNA polymerase degrades the probe into smaller fragments that can be differentiated from undegraded probe. The assay is sensitive and specific and is a significant improvement over more cumbersome detection methods.
Article
Full-text available
Dendritic cells (DC), the most efficient antigen-presenting cells, are well equipped for activation of naive CD4+ T cells by their expression of high levels of major histocompatibility complex and costimulator molecules. We now demonstrate that some DC are equally well equipped for killing these same T cells. Murine splenic DC consist of both conventional CD8alpha- DC and a major population of CD8alpha+ DC. Whereas CD8- DC induce a vigorous proliferative response in CD4 T cells, CD8+ DC induce a lesser response that is associated with marked T cell apoptosis. By using various mixtures of T cells and DC from Fas-mutant lpr/lpr mice and Fas-ligand (FasL) mutant gld/gld mice, we show this death is due to interaction of Fas on activated T cells with FasL on CD8+ DC. Furthermore, we show by direct surface staining that CD8+ DC, but not CD8- DC, express FasL at high levels. These findings indicate that FasL+ CD8+ DC are a specialized subgroup of DC with a role in the regulation of the response of primary peripheral T cells.
Article
Full-text available
Dendritic cells (DC) are described as "nature's adjuvant," since they have the capacity to sensitize T cells in vivo upon first encounter with the antigen. The potent accessory properties of DC appear to develop sequentially. In particular, the ability to process antigens and to sensitize native T cells develops in sequence, a process termed "maturation" that is well described in vitro. Here, we obtain evidence for maturation in vivo in response to the bacterial product lipopolysaccharide (LPS). Before LPS treatment, many DC are found at the margin between the red and white pulp. These cells lack the M342 and DEC-205 markers, but process soluble proteins effectively. 6 h after LPS, DC with the M342 and DEC-205 markers are found in increased numbers in the T cell areas. These cells have a reduced capacity to process proteins, but show increases in the B7 costimulator and T cell stimulatory capacity. 48 h after LPS, the number of DC in the spleen is reduced markedly. We interpret these findings to mean that LPS can cause DC in the marginal zone to mature and to migrate into and then out of the T cell areas.
Article
We have shown previously that dendritic cells (DC) produce IL-12 upon interaction with CD4+ T cells. Here we ask how this IL-12 production is induced and regulated. Quantitative PCR and in situ hybridization for IL-12 p40 and an ELISA specific for the p70 heterodimer were used to determine IL-12 production. We demonstrate that ligation of either CD40 or MHC class II molecules independently trigger IL-12 production in DC, and that IL-12 production is downregulated by IL-4 and IL-10. The levels of bioactive IL-12 that can be released by triggering with an anti-CD40 mAb or with a T cell hybridoma are high (range 260-4700 pg/ml from 1 X 10(6) DC in 72 h). The CD40-mediated pathway indicates that IL-12 production is induced in DC upon interaction with activated, CD40 ligand-expressing helper T cells, even in the absence of cognate antigen recognition. Side-by-side comparison of IL-12 production, and blocking experiments employing an anti-CD40 ligand mAb, suggest that the CD40-mediated pathway is quantitatively more significant than induction via the MHC class II molecule. The importance of the CD40/CD40 ligand interaction for IL-12 induction in DC likely contributes to the recent finding that mice lacking the CD40 ligand are impaired in mounting Th1 type cell-mediated immune responses.
Article
A large body of evidence suggests the existence of polarized human T cell responses, reminiscent of Th1 and Th2 subsets described for mouse T cells. Human Th1-like cells preferentially develop during infections by intracellular bacteria, protozoa, and viruses, whereas Th2-like cells predominate during helminthic infestations and in response to common environmental allergens. The cytokine profile of “natural immunity” evoked by different offending agents in the context of different host genetic backgrounds appears to be a critical factor in determining the phenotype of the subsequent specific response. Strongly polarized human Th1-type and Th2-type responses not only play different roles in protection, they can also promote different immunopathological reactions. Th1-type responses appear to be involved in organ specific autoimmunity, in contact dermatitis, and in some chronic inflammatory disorders of unknown etiology. In contrast, in genetically predisposed hosts, Th2-type responses against common environmental allergens are responsible for triggering of allergic atopic disorders. Altered profiles of lymphokine production may account for immune dysfunctions in some primary or acquired immunodeficiency syndromes. The role of lymphokines produced by T cells in the pathogenesis of systemic autoimmune disorders is less clear. Further work is also required to better clarify the role of T cell-derived lymphokines in protecting against tumors or in favoring their development.
Article
Using granulocyte/macrophage colony-stimulating factor (GM-CSF) and interleukin 4 we have established dendritic cell (DC) lines from blood mononuclear cells that maintain the antigen capturing and processing capacity characteristic of immature dendritic cells in vivo. These cells have typical dendritic morphology, express high levels of major histocompatibility complex (MHC) class I and class II molecules, CD1, Fc gamma RII, CD40, B7, CD44, and ICAM-1, and lack CD14. Cultured DCs are highly stimulatory in mixed leukocyte reaction (MLR) and are also capable of triggering cord blood naive T cells. Most strikingly, these DCs are as efficient as antigen-specific B cells in presenting tetanus toxoid (TT) to specific T cell clones. Their efficiency of antigen presentation can be further enhanced by specific antibodies via FcR-mediated antigen uptake. Incubation of these cultured DCs with tumor necrosis factor alpha (TNF-alpha) or soluble CD40 ligand (CD40L) for 24 h results in an increased surface expression of MHC class I and class II molecules, B7, and ICAM-1 and in the appearance of the CD44 exon 9 splice variant (CD44-v9); by contrast, Fc gamma RII is markedly and sometimes completely downregulated. The functional consequences of the short contact with TNF-alpha are in increased T cell stimulatory capacity in MLR, but a 10-fold decrease in presentation of soluble TT and a 100-fold decrease in presentation of TT-immunoglobulin G complexes.
Article
Dendritic cells, a minor cell population in lymphoid tissues, are specialized for presentation of antigenic peptides to T lymphocytes. Thymic dendritic cells are involved in the deletion of self-reactive T lymphocytes. Although all dendritic cells are ultimately of bone-marrow origin, it has not been clear whether thymic dendritic cells are produced in the adult thymus from a precursor cell or whether they migrate there preformed from the periphery. Recently we isolated from adult mouse thymus a population of early T precursors that could still form B lymphocytes, but not erythroid or myeloid cells, when transferred intravenously. Here we show that these thymic lymphoid precursor cells, as well as bone-marrow haematopoietic stem cells, are able to form both dendritic cells and T-cell progeny when transferred into an irradiated thymus. Such linked development may ensure that developing T cells are negatively selected predominantly by self antigens presented on newly formed thymic dendritic cells.