You are on page 1of 9

BIOLOGY OF REPRODUCTION 77, 590–598 (2007)

Published online before print 27 June 2007.


DOI 10.1095/biolreprod.107.060632

Minireview

Dendritic Cells: Key to Fetal Tolerance?1


Sandra M. Blois,2,4,5 Ulrike Kammerer,6 Catalina Alba Soto,7 Mareike C. Tometten,4 Valerie Shaikly,5
Gabriela Barrientos,4 Richard Jurd,5 Daniel Rukavina,8 Angus W. Thomson,9 Burghard F. Klapp,4
Nelson Fernández,3,5 and Petra C. Arck3,4

Downloaded from https://academic.oup.com/biolreprod/article/77/4/590/2629671 by guest on 10 February 2022


University Medicine of Berlin,4 Charité Centrum 12, Internal Medicine and Dermatology, Campus Virchow, 13353
Berlin, Germany
Department of Biological Sciences,5 University of Essex, Wivenhoe Park, Colchester C04 3SQ, United Kingdom
Department of Gynaecology and Obstetrics,6 University of Würzburg, D-97080, Würzburg, Germany
Department of Microbiology,7 Parasitology and Immunology, School of Medicine, University of Buenos Aires,
C1121ABG Buenos Aires, Argentina
Department of Physiology and Immunology,8 Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia
Thomas E. Starzl Transplantation Institute and Department of Surgery,9 University of Pittsburgh Medical Center,
Pittsburgh, Pennsylvania 15213
1
ABSTRACT INTRODUCTION
Pregnancy is a unique event in which a fetus, despite being The question as to why the placenta, despite the expression
genetically and immunologically different from the mother (a of paternally derived fetal alloantigens, is accepted by the
hemi-allograft), develops in the uterus. Successful pregnancy mother remains to be fully explained, although several immune
implies avoidance of rejection by the maternal immune system. tolerance mechanisms have been postulated [1–3].
Fetal and maternal immune cells come into direct contact at the Dendritic cells (DC) represent a population of cells with
decidua, which is a highly specialized mucous membrane that relevance to mucosal sites, such as the skin, airways, gut, and
plays a key role in fetal tolerance. Uterine dendritic cells (DC) decidua. This is because DC have the unique property of being
within the decidua have been implicated in pregnancy mainte-
able to induce both antigen-specific activation and suppression
nance. DC serve as antigen-presenting cells with the unique
ability to induce primary immune responses. Just as lymphocytes
during the immune response. DC may also play a role in
comprise different subsets, DC subsets have been identified that tolerance induction through the generation of T cells with
differentially control lymphocyte function. DC may also act to regulatory properties, which in turn limit the proliferation of
induce immunologic tolerance and regulation of T cell-mediated effector T cells [4, 5] or delete antigen-specific T cells [6, 7].
immunity. Current understanding of DC immunobiology within In the present review, we will discuss the role of DC subsets
the context of mammalian fetal-maternal tolerance is reviewed and their activation signals in the regulation of fetal-maternal
and discussed herein. tolerance. In this context, we will highlight the link between
decidua, dendritic cells, placenta, tolerance
DC and the cellular and molecular network components
involved in the biology of pregnancy, namely T cells,
cytokines, and hormones. These endocrine and immune signals
ultimately determine the maturation, differentiation, and
activation status of DC. In turn, these changes are responsible
for the dual capacity of DC to trigger an immune response that
1
Supported by grants from Charité (to S.M.B. and P.C.A.), DFG (KFO- is associated with fetal rejection and to promote cell tolerance
124/2 to U.K.), and the Croatian Ministry of Science (0376-0377 to at the fetal-maternal interface.
D.R.). S.M.B. is supported by Charité (Habilitations-Stipendium), and
C.A.S. and G.B. are supported by fellowships from CONICET and
DAAD, respectively. S.M.B., P.C.A., and D.R. are part of the EMBIC Immunobiology of Pregnancy
Network of Excellence, which is cofinanced by the European
Commission through the FP6 framework program ‘‘Life Science,
Fetal allograft hypothesis. The fetal-maternal relationship
Genomics and Biotechnology for Health.’’ is a unique immunologic phenomenon. Fetal tissues express
2
Correspondence: Sandra M. Blois, Charité Centrum 12, Internal alloantigens that are encoded by polymorphic MHC genes
Medicine and Dermatology, BMFZ, Raum 2.0547, Augustenburger inherited from the father. However, in a normal pregnancy,
Platz 1, 13353 Berlin, Germany. FAX: 49 30 450 553997; although the mother can raise antibodies against parental gene
e-mail: sandra.blois@charite.de or smblois@essex.ac.uk
3
These authors contributed equally to this work.
products, typically HLA allele-specific antibodies, these do not
elicit fetal rejection.
Initial studies in pregnancy invoking the classical allograft
Received: 5 February 2007. rejection theories implicated CD4 and CD8 T cells. This
First decision: 20 February 2007.
Accepted: 6 June 2007. concept has been expanded by the notion that NK cells are also
Ó 2007 by the Society for the Study of Reproduction, Inc. involved in rejection [8]. In addition, the role of cd T-cell
ISSN: 0006-3363. http://www.biolreprod.org recognition of trophoblasts has been documented in mice [9]
590
DENDRITIC CELLS AND PREGNANCY TOLERANCE 591

and humans [10]. Moreover, NKT cells, most likely with Tolerogenic DC: Different Maturation States or Different
TCRcd, have been implicated in fetal rejection [11], as have Subsets?
NKab T cells [12]. CD4 þ T cells that recognize paternal
antigens but not trophoblasts are presented by antigen- The induction of tolerance by DC appears to contradict the
presenting cells (APC) in decidual tissues [13, 14]. These immunostimulatory function of these cells. However, a role for
observations suggest that the properties of APC in the uterine DC in the induction of peripheral tolerance is supported by
lining are important in modulating local immunity and several studies [23]. Two general hypotheses have been
tolerance. No single mechanism to explain the so-called fetal- proposed to explain how DC might maintain peripheral
maternal immunologic paradox has been identified. As in tolerance. The first hypothesis is based on studies performed
tissue allografts, interactions between the adaptive and innate by Dhodapkar et al. [24] and Steinman et al. [25], which
immune systems, at both the cellular and molecular levels and support the concept that antigen presentation by immature DC
including cytokines, are required to regulate pregnancy to term. induces tolerance, whereas antigen presentation by mature DC
The Th1 to Th2 ratio as a paradigm of fetal-maternal induces immunity. This paradigm has been questioned, since
tolerance. The maternal balance between Th1 and Th2-Th3 mature DC have also been found to induce CD4 T-cell

Downloaded from https://academic.oup.com/biolreprod/article/77/4/590/2629671 by guest on 10 February 2022


cytokines contributes to the success of pregnancy [15, 16]. It is tolerance [26, 27]. Current data suggest that steady-state DC
now accepted that during pregnancy, local changes in the are sufficiently mature to express intermediate levels of MHC
balance between Th1/Th2-Th3 cytokines occur within the class II and costimulatory molecules but are not yet activated to
uterus and fetal-placental unit. Cytokine shifts influence the extent that they can induce an immune response [27–29].
trophoblast cells and uterine immune cells (macrophages, NK Immunity occurs only in the context of infection-associated
cells, and lymphocytes), which contribute to implantation of signals, which induce full maturation of DC.
the embryo, development of the placenta, and survival of the The second hypothesis suggests that unique DC subsets are
fetus to term. The predominant cytokines at each stage of responsible for the induction of tolerance [30]. For example,
gestation function both to limit maternal immune rejection of murine CD8A þ DC have been shown to act as a specialized
the semi-allogeneic fetus, especially at the fetal-maternal subset of tolerogenic or regulatory DC in experimental systems
interface, and to facilitate the ongoing physiological processes of allograft rejection and in vivo studies [31, 32].
within the maternal reproductive tract. Furthermore, in In support of the subset hypothesis, a DC subset with the
experimental models, cytokine imbalances with inappropriate ability to prime type 1 regulatory T (Treg) cells has recently
activation of macrophages and NK cells have been shown to be been described. This subset produces IL10 upon stimulation,
detrimental to fetal survival [2]. exhibits a stable immature phenotype (CD45RBhigh ITGAXlow
We have previously reported [17] on the decisive roles MHC class IIintermediate CD86low) and plasmacytoid morphol-
played by IL10 and IL12 in driving the acceptance or rejection ogy [33, 34].
of the fetus. A strong, pregnancy-protective role for the Th2 Immature DC express CD200R2, type 2 receptors for
cytokine IL10 is supported by data from mouse models, which CD200, a glycoprotein with pregnancy-protective function,
show that while purified exogenous IL10 administered during which is expressed on trophoblast cells and in certain areas of
pregnancy decreases resorption rates [18], injection of anti- the deciduas on days 8–12 of pregnancy [35]. In DBA/2J-
IL10 antibody enhances these rates. In support of these results, mated CBA/J female mice [18] (a murine model in which
injection of IL12 during early gestation has been found to increased abortion rates can be induced by application of
induce abortion and increase the levels of the proinflammatory lipopolysaccharide, Th1 cytokines or stress exposure), block-
cytokines TNF and IFNG at the fetal-maternal interface [19]. ing CD200 greatly increases fetal rejection and conversely,
administering CD200Fc or CD200 þ splenocytes from BALB/c
Special Role of DC at Mucosal Interfaces mice reduces fetal rejection. Moreover, CD200 triggering of
DC induces a CD4 þIL2RA þ Treg population with suppressive
DC are particularly responsive to signals received from the activity, as demonstrated by increased FOXP3 expression and
tissue microenvironment. In mucosal tissues, DC can acquire inhibition of mixed leukocyte reactions [36]. In line with this
unique features or phenotypes that are necessary for the finding, DBA/2J-mated CBA/J females that are exposed to
regulation of specific local immune functions, which include sound stress show elevated percentages of resident decidual
tolerance to food antigens in the gastrointestinal tract, to DC that express moderate levels of MHC II, CD80/86, and
respiratory antigens in the airways, and protection against ICAM1 on gestation Day 7.5 [17].
pathogenic organisms [20]. The phenotypes of DC are In humans, the number of mature CD83 þ DC increases in
influenced by cytokines and anti-inflammatory mediators the uterine mucosa during the late secretory phase of the
produced by epithelial cells, either under physiological menstrual cycle in comparison to other endometrial phases
conditions or in response to microbial signals [20]. [37]. This is in contrast to first trimester decidua, in which the
Previously, we have proposed [17] that decidual DC are the vast majority of DC express CD209 (previously known as
‘‘gatekeepers’’ of the uterine mucosal immune system, inducing DCSIGN), which is a marker for immature or inactivated DC
tolerance under normal physiological conditions. DC are also [38, 39]. Interestingly, freshly isolated decidual CD209 þ DC
sufficiently responsive to inflammatory stimuli to allow T-cell take up antigen efficiently but are unable to stimulate naı̈ve
priming and protective immunity when necessary. It is well allogeneic T cells. However, when stimulated with an
known that exposure to agents such as prostaglandin E2 (PGE2) inflammatory cytokine cocktail, these cells become mature
[21] polarizes the maturation of myeloid DC into Th2- IL2RA þ/CD83þ DC with potent stimulatory capacity for
promoting DC, while other cytokines, such as transforming allogeneic T cells and are similar to those DC that are sparsely
growth factor b (TGFB), promote tolerogenic DC [22]. Both distributed in early pregnancy decidua [39, 40].
PGE2 and TGFB are present at the fetal-maternal interface in Therefore, it can be hypothesized that the maturation state of
normal pregnancy and may be utilized by DC to regulate DC plays a role in the etiology of spontaneous pregnancy
placental health (Fig. 1). However, the assumption that failure. Indeed, the high abortion rates observed in the CBA/J 3
decidual DC have similar functions to mucosal DC needs to DBA/2J murine model are related to DC that exhibit a more
be further investigated. mature phenotype (higher expression of MHC class II, CD80,
592 BLOIS ET AL.

Downloaded from https://academic.oup.com/biolreprod/article/77/4/590/2629671 by guest on 10 February 2022


FIG. 1. Hypothetical model of how dendritic cells (DC) regulate immune responses directed against the conceptus. During normal pregnancy,
tolerogenic stimuli from trophoblasts (Tro), progesterone (P), prostaglandin E2 (PGE2), vitamin D, and cells in the environment (e.g., NK cells and
macrophages) promote partial activation of the resident DC. This results in the production of anti-inflammatory cytokines (e.g., IL10), which promote the
induction of tolerance at the fetal-maternal interface, thereby activating different mechanisms, such as the production of pregnancy-protective Th2/Th3
cytokines and the generation of Treg, which enhance suppression of the immune system, thereby contributing to fetal tolerance.

and ICAM1), as compared to low-abortion-rate mice [41]. menstrual cycle, and to control trophoblast invasion during
Deciduas from women with recurrent miscarriage at 8 wk of pregnancy [43]. In contrast, during mouse pregnancy, uNK cells
gestation have been shown to contain significantly more CD83 þ accumulate on the mesometrial side of the placenta and play a
DC than gestational age-matched normal controls [42]. The central role in decidualization [45]. Indeed, Il15tm1Imx/tm1Imx
presence of mature DC correlates with higher abortion rates. It mice, which lack uNK cells, present decidual abnormalities,
is conceivable that these activated DC can overcome the which include thickening of the arterial walls with luminal
tolerance to paternal antigens and induce fetal rejection (Fig. 2). narrowing and a hypocellular decidua basalis. However, uNK
cells are not required for successful pregnancy, since IL15/
Dendritic Cells Within the Decidual Cellular Environment mice have normal implantation and nonabortive pregnancies
[46].
Cross-talk between DC and NK cells: two players in Little is known about the potential interactions between DC
innate immunity. Recent data have highlighted the cooperation and NK cells under homeostatic resting conditions. However, it
between DC and NK cells in the control/switch of innate has been shown that interaction between immature DC and
immunity. NCAM1 þþFCGR3 (formerly CD56 þþCD16) activated NK cells results in either DC maturation or cell death
uterine NK (uNK) cells represent the main population of [47]. The mechanisms that determine the death or maturation
lymphoid cells in the uterine mucosa during early pregnancy outcome depend on a dynamic interplay between the ratio of
[43]. However, their number decreases from midgestation DC:NK cells and the DC maturation state [48]. Gerosa et al.
onwards, and they are almost absent at term. Although the [49] have shown that the cross-talk between immature DC and
interaction between trophoblast MHC class I molecules and resting NK cells leads to cell activation only in the presence of
uNK cells is likely to provide the pivotal control for successful microbial stimuli. Furthermore, only under conditions of low
implantation in humans [44], it is unclear how this process is DC:NK ratios do DC-NK cell interactions result in NK
mediated. It is likely that uNK cells play a dual role in activation [50]. The cytokine pattern of DC activation after
reproduction: to monitor mucosal integrity throughout the interaction with uNK cells in response to antigenic stimuli
DENDRITIC CELLS AND PREGNANCY TOLERANCE 593

Downloaded from https://academic.oup.com/biolreprod/article/77/4/590/2629671 by guest on 10 February 2022


FIG. 2. During failure of gestation, massive infiltration of NK cells to the fetal-maternal interface and the production of IFNG by NK cells, together with
the presence of TNF-producing macrophages stimulate the full activation of DC and the production of inflammatory cytokines, particularly IL12. Antigen
presentation by mature DC induces Th1 cytokine production by lymphocytes, which results in the apoptosis of trophoblast cells and the Th1-type immune
bias of abortion.

could also influence the polarization of T-cell responses. There 55]. These findings indicate that murine DC subsets that differ
may be physiopathologic conditions (i.e., spontaneous abor- in CD8A expression can prime naı̈ve Th cells to produce either
tion) under which disequilibrium of the ratio between uNK Th1 or Th2 cytokines, respectively [52, 56]. The mechanism by
cells and DC results in aberrant cell activation and perturbation which CD8A DC induce Th2 cytokines has not been
of the course of pregnancy (Fig. 2). In particular, it has been established, although IL13 [57] and IL10 [56] are suitable
reported that the vast majority of decidual immature DC are in candidates.
close contact with uNK cells, whereas the small number of The activities of cytokines, such as IL10 and TGFB, as T-
mature decidual DC are clustered with CD3 þ T cells, but not cell polarizing factors are well-documented [58]. In accor-
with uNK cells. Moreover, in vitro studies have shown that dance, we have noted that the intracellular level of IL10 in
decidual CD83 þ DC cocultured with NCAM1 þ autologous uterine DC is significantly higher than the level of IL12 during
uNK cells stimulate the proliferation of NCAM1 þ cells in a most of the gestation period [17]. However, there is a transient
DC-dependent manner, increase KLRC1 inhibitory receptor, increase in the incidence of uterine CD8A þ DC in early
and decrease KLRC2- and KLRK1-activating receptor expres- gestation, namely on Day 5.5, concomitant with a decrease in
sion [51]. the relative number of IL10-producing DC. Our current
paradigm is that DC have remarkable plasticity in their abilities
DC-derived Cytokines and Pregnancy to induce Th1 or Th2 cytokines and to exhibit temporal
Various types of DC-derived molecules with the ability to regulation and subtle discrimination of antigen stimuli [59].
polarize Th-cell responses have been identified. DC from IL12- This prompted us to suggest that in early gestation, fetal
deficient mice fail to induce Th1 responses, which suggests a antigens may be stimulating CD8A þ uterine DC to produce
critical role for IL12 family members (e.g., IL12, IL23, and Th1 cytokines, thus contributing to the inflammatory milieu
IL27) in DC-induced Th1 responses [52, 53]. Consistent with characteristic of the implantation period. At later stages, a
this notion, murine CD8A þ DC, but not CD8A DC, can be regulatory IL10 cytokine pattern predominates, which mediates
induced to produce large amounts of IL12 and IFNG [52, 54, the communication between embryonic cells and maternal
594 BLOIS ET AL.

uterine cells and the maintenance of pregnancy [60–62]. present on immature DC, leads to the generation of
Moreover, recent experiments have shown that uterine DC CD4 þIL2RA þFOXP3þ Treg that have the ability to suppress
from mice with high abortion rates display increased mixed lymphocyte reactions and allograft rejection.
expression of CD8A þ and increased IL12:IL10 ratios com- Other regulatory T cells. In addition to CD4 þ Treg, CD8 þ
pared to mice with a low incidence of fetal rejection [41]. T cells may play a role in oral tolerance [73]. Furthermore,
Compared with mouse DC subtypes, human DC exhibit CD8 þCD28FOXP3þ T-suppressor cells have recently been
considerable plasticity in response to cytokines and pathogens. reported to induce tolerogenic endothelial cells through the
This feature makes it difficult to assign a function to each DC induction of inhibitory receptors and the down-regulation of
lineage. In spite of this, recent findings indicate that myeloid costimulatory and adhesion molecules [74]. In murine
DC produce high levels of IL12 when stimulated with TNF or pregnancy, a CD8A þ cell subpopulation that prevents abortion
CD40 ligand (CD40LG) and stimulate a Th1-skewed cytokine is present between gestation Days 4.5 and 8.5 [2, 75]. These
profile in T cells, whereas fully mature plasmacytoid DC prime CD8A þ cells, which may coexpress the TCRcd, appear to play
T cells towards a Th2 cytokine profile [63]. Even though a crucial role in the regulation of the Th1/Th2 balance [2, 76].
protection from fetal rejection has been traditionally linked to a Our studies suggest that during stress-challenged pregnancy,

Downloaded from https://academic.oup.com/biolreprod/article/77/4/590/2629671 by guest on 10 February 2022


Th2-type immune response, myeloid DC predominate in there is a decrease in uterine CD8A þ cells. Since we observed a
human and mouse early pregnancy decidua [17, 38, 40]. low fetal rejection index when CD8A þ cells were restored by
Although these findings apparently contradict the Th1/Th2 dydrogesterone (a progesterone derivate product), it appears
paradigm, they are in line with the new importance given to the that these cells are highly regulated by the adequate levels of
state of activation over the lineage of resident DC. To date, progesterone at the fetal-maternal interface [77]. Moreover,
only one study has examined the expression of IL12 by depletion of CD8A þ cells abrogated the protective effect of
decidual DC, albeit in relation to peripheral blood DC in early dydrogesterone in mice undergoing stress-challenged pregnan-
human pregnancy. The results show that IL12-producing cy. Although they have not been classified as ‘‘professional’’
myeloid DC (ITGAX þ) or lymphoid DC (IL3RAþ) numbers regulatory cells, these uterine CD8A þ cells may be involved in
are significantly reduced in the decidua compared to the the suppression of immune responses.
peripheral blood. In addition, the level of IL12 secreted by Tryptophan 2,3-dioxygenase (TDO2) and indoleamine
decidual myeloid cells stimulated with lipopolysaccharide 2,3-dioxygenase (INDO). Tryptophan catabolism is one of
(LPS) or CD40LG was found to be significantly lower than the many mechanisms involved in tolerance [78]. This essential
that secreted by peripheral blood counterparts [40]. Thus, amino acid is used by vertebrates in protein synthesis and is a
decidual DC can regulate the balance between Th1 and Th2 precursor to serotonin, melatonin, kynurenine, and quinolinic
cytokines, resulting to the maintenance of pregnancy. acid. Moreover, some tryptophan catabolites induce apoptosis
of T cells [79]. Expression of INDO is induced by IFNG in
Consequences of Cross-Talk many tissues and in certain types of activated macrophages and
DC, suggesting a role for INDO in the regulation of immune
Regulatory T cells. Treg block the functions of effector responses [80].
CD4 þ and CD8 þ T cells. As a result, tolerance is achieved Tatsumi et al. [81] have demonstrated that TDO2 is induced
[64]. Naturally occurring (i.e., thymically generated) CD4 þ in endometrial stromal cells concomitant with decidualization,
IL2RA þ Treg appear as mediators to prevent autoimmune which suggests its involvement in the implantation process by
responses to self antigens, whereas Treg induced by antigenic regulating the tryptophan level at the implantation site. On the
stimulation in the periphery (also referred to as ‘Tr1’ cells or other hand, INDO is expressed in the syncytial trophoblast and
‘adaptive regulatory cells’) appear to be involved in the defends the conceptus against rejection by reducing the
maintenance of peripheral tolerance at mucosal surfaces [65, tryptophan level and suppressing T-cell activity. Several
66]. Mucosal tissues are faced with the constant challenge of animal studies that have investigated the role of INDO in
maintaining nonresponsiveness to nonself peptides (antigens), murine pregnancy have given contradictory results. Pharma-
which means that the immune system must employ potent cologic inhibition of INDO activity results in rejection of
regulatory mechanisms. Several studies have demonstrated that allogeneic fetuses by the maternal immune system [13].
Treg can prevent graft rejection [67–69]. A similar situation However, Indotm1Alm/tm1Alm mice do not exhibit fetal rejection
seems to occur during pregnancy, whereby the maternal in allogeneic matings, which suggests that INDO is not an
immune system tolerates paternal antigens expressed by the essential mechanism for tolerance in the context of pregnancy
fetus. Recently, it has been demonstrated that normal human [82]. Moreover, Mackler et al. [83] have shown that INDO is
pregnancy is associated with an increase in the blood Treg cell not expressed in murine trophoblasts but it is expressed in
population. Analysis by stage of pregnancy has shown a stromal cells of the decidua basalis and metrial gland. In
significant increase from the nonpregnant controls to the first contrast, Baban et al. [82] have shown that INDO expression is
trimester and from the first trimester to the second trimester, restricted to the perinuclear regions of primary trophoblast
with no significant change from the second to third trimester. giant cells at the middle stage of mouse gestation. Further
The proportion of CD4 þ IL2RA þ T cells was significantly investigations are needed to clarify the role of INDO in
lower 6–8 wk following delivery than during the third trimester pregnancy maintenance.
but remained higher than in nonpregnant controls [70]. In
addition, decidual CD4 þ IL2RA þ T cells represented 14% of Influence of the Endocrine System During Pregnancy
all CD4 þ T cells in the third trimester [71]. DC production of
IL10 may be critical for the generation of Treg [26, 33]. The Dendritic cells and steroid hormones. An excellent body
presence of decidua-resident, semimature DC subsets with of literature exists regarding the influence of steroid hormone
intermediate expression of MHC and secretion of IL10 [17, 38, pathways on immune cell populations [84, 85]. Progesterone
40] raises the question as to whether the tolerogenic function of probably contributes to the natural suppression of cell-mediated
DC is achieved through the priming of Treg in the decidua. immunity that accompanies pregnancy and allows the fetus to
Moreover, Gorczynski et al. [72] have shown that triggering be tolerated during gestation. Progesterone acts through two
CD200R2, which is a pregnancy protective receptor that is isoforms of the progesterone receptor (PGR) [86]. Until
DENDRITIC CELLS AND PREGNANCY TOLERANCE 595

recently, the consensus has been that PGR is induced by Subsequently, Larsen et al. [99] were the first to show that DC
estrogen, and that many of the effects of progesterone can be trafficked from heart allografts to recipient spleens, which in
attributed to the combined effects of estrogen and progesterone. nonimmunosuppressed hosts is associated with rejection,
Interestingly, it has been demonstrated that progesterone is concomitant with the disappearance of the donor DC. Studies
essential for the induction of uterine decidualization, since this conducted by Lu et al. [100] have revealed that donor-derived
process fails to occur in mice that lack the Pgr gene [87, 88]. myeloid DC persist in and can be propagated from the BM of
Indeed, Liu et al. [89] have reported that estrogen decreases the mice that accepted fully mismatched (liver) allografts without
production of proinflammatory cytokines TNF, IFNG, and immunosuppressive therapy. This cannot be achieved with
IL12 in mature DC. High estrogen levels at the fetal-maternal animals that acutely reject heart grafts from the same donor
interface could influence uterine DC to reduce inflammatory strain [100], unless both donor BM cells and immunosuppres-
cytokine production. sive therapy are administered [101]. Further studies have
Recent studies have implicated progesterone in the shown that immature donor myeloid DC, which are deficient in
modulation of DC differentiation, maturation, and function. surface costimulatory molecules, can induce alloantigen-
Liang et al. [90] have shown that progesterone inhibits immune specific T-cell hyporesponsiveness in vitro [102]. More

Downloaded from https://academic.oup.com/biolreprod/article/77/4/590/2629671 by guest on 10 February 2022


responses by producing more immature bone marrow (BM)- significantly, several groups have shown that when adminis-
derived DC in mice. The addition of progesterone to BM- tered before, during or even after transplantation, immature
derived DC cultures results in significantly increased IL10 donor DC prolong MHC-mismatched allograft (including skin
production and decreased TNF production. In addition, Butts et graft) survival. In some instances, indefinite donor-specific
al. [91] have found that progesterone at concentrations similar graft survival is achieved [103–110]. Recently, it has been
to those occurring during the menstrual cycle or pregnancy has shown that TGFB-treated (‘alternatively-activated’) IL10 þ DC
a crucial effect on rat BM-derived mature DC by modulating resist phenotypic and functional maturation in response to
the ability of these cells to function as stimulators of TLR4 ligation and induce long-term heart allograft survival
proinflammatory responses. In vitro studies have shown that when coadministered with a single dose of CTLA4-Ig. Graft
DC derived from the decidua of pregnant women increase their survival is associated with Treg infiltration and the absence of
CD83 and HLADR expression levels when treated with vasculopathy [111]. There is also evidence that persistence of
progesterone at levels similar to those present during early donor-derived DC may be important for the long-term
pregnancy [92]. maintenance of transplantation tolerance, as well as for the
Vitamin D and PGE2: novel modulators? Experimental prevention of chronic rejection of heart allografts [112].
evidence suggests that vitamin D may play a role in maternal- Collectively, these findings suggest that donor-derived DC,
conceptus cross-talk. Besides its classical activity, the active most likely but not exclusively at an immature/progenitor
form of vitamin D, 1,25-dihydroxyvitamin D3 or 1,25(OH)2D3 stage, can subvert recipient T-cell responses to alloantigens,
has well-known immunomodulatory and antiproliferative through as yet ill-defined mechanisms. Although comparative-
properties on human uNK cells [93]. Female fertility seems ly little research has been performed using syngeneic/
to be markedly reduced in vitamin D-deficient murine models. autologous DC to subvert the indirect pathway, recent animal
Both decidua and trophoblast cells express vitamin D receptor studies support the feasibility of this approach. Thus, Lechler’s
(VDR), as well as CYP27B1 (previously known as 25(OH)D group [109] has shown that indefinite organ graft survival can
1a-hydroxylase), which suggests an autocrine or paracrine role be achieved in the absence of chronic rejection in rats that are
for 1,25(OH)2D3 within these tissues [94]. In particular, receiving pharmacologically modified myeloid DC that express
Zehnder et al. [95] have postulated that local activation of donor and recipient MHC antigens and only a short course of
vitamin D influences implantation, either through the estab- peri-operative cyclosporine (to control the direct pathway).
lished immunomodulatory effects of 1,25(OH)2D3 or via the Significantly, this effect is associated with the induction of
regulation of specific target genes associated with implantation. indirect-pathway Treg. It appears that antigen-specific Treg can
Indeed, 1,25(OH)2D3 regulates developmental target genes, be induced under systemic tolerogenic regimens by plasmacy-
such as the homeobox (Hox) genes. In particular, the Hoxa10 toid DC that acquire alloantigen in the graft and traffic to
gene has been shown to be intimately associated with secondary lymphoid tissues [113].
implantation [96].
Another immunomodulatory molecule that is present at the CONCLUSION
fetal-maternal interface is PGE2. PGE2 suppresses a number of
T-cell reactions (e.g., cellular cytotoxicity and T-cell and killer- Understanding the immunological paradox of maternal
cell activity) and promotes the development of DC with low tolerance towards the fetal allograft is of great importance.
capacities to produce IL12, which supports the generation of The complexity of the uterine network clearly indicates that
Th2 cells. Harizi et al. [97] have demonstrated that exoge- successful pregnancy does not rely entirely on immunological
nously added or endogenously released PGE2 acts on DC by factors. DC appear to be key regulators of pregnancy,
inducing endogenous IL10, which in turn suppresses IL12 involving both innate and adaptive mechanisms. Decidual
production and alters antigen presentation through the DC likely function to co-ordinate the spatial and temporal
inhibition of MHC class II protein expression. As vitamin D immunological shifts necessary for implantation and the
and PGE2 are both present in decidua, these molecules may progression of pregnancy.
inhibit IL12 production by the uterine DC, thereby shifting the
cytokine balance to a tolerogenic state at the fetal-maternal REFERENCES
interface.
1. Medawar PB. Some immunological and endocrine problems raised by
evolution of viviparity in vertebrates. Symp Soc Exp Biol 1953; 7:320–
DC and Promotion of Transplant Tolerance 337.
2. Clark DA, Arck PC, Chaouat G. Why did your mother reject you?
Evidence implicating DC as the main immunological Immunogenetic determinants of the response to environmental selective
stimulus for graft rejection has been provided by the renal pressure expressed at the uterine level. Am J Reprod Immunol 1999; 41:
allograft ‘‘parking’’ experiments of Lechler and Batchelor [98]. 5–22.
596 BLOIS ET AL.

3. Chaouat G, Menu E, Kinsky R, Brezin C. Immunologically mediated producing IL-10 mediate tolerance induced by respiratory exposure to
abortions: one or several pathways? Res Immunol 1990; 141:188–195. antigen. Nat Immunol 2001; 2:725–731.
4. Jonuleit H, Schmitt E, Schuler G, Knop J, Enk AH. Induction of 27. Menges M, Rossner S, Voigtlander C, Schindler H, Kukutsch NA,
interleukin 10-producing, nonproliferating CD4( þ) T cells with regula- Bogdan C, Erb K, Schuler G, Lutz MB. Repetitive injections of dendritic
tory properties by repetitive stimulation with allogeneic immature human cells matured with tumor necrosis factor alpha induce antigen-specific
dendritic cells. J Exp Med 2000; 192:1213–1222. protection of mice from autoimmunity. J Exp Med 2002; 195:15–21.
5. Mahnke K, Knop J, Enk AH. Induction of tolerogenic DCs: ‘you are 28. Steinman RM. Some interfaces of dendritic cell biology. APMIS 2003;
what you eat’. Trends Immunol 2003; 24:646–651. 111:675–697.
6. Bonifaz L, Bonnyay D, Mahnke K, Rivera M, Nussenzweig MC, 29. Lutz MB, Schuler G. Immature, semi-mature and fully mature dendritic
Steinman RM. Efficient targeting of protein antigen to the dendritic cell cells: which signals induce tolerance or immunity? Trends Immunol
receptor DEC-205 in the steady state leads to antigen presentation on 2002; 23:445–449.
major histocompatibility complex class I products and peripheral CD8 þ 30. Reid SD, Penna G, Adorini L. The control of T cell responses by
T cell tolerance. J Exp Med 2002; 196:1627–1638. dendritic cell subsets. Curr Opin Immunol 2000; 12:114–121.
7. Hawiger D, Inaba K, Dorsett Y, Guo M, Mahnke K, Rivera M, Ravetch 31. Fazekas de St Groth B. The evolution of self-tolerance: a new cell arises
JV, Steinman RM, Nussenzweig MC. Dendritic cells induce peripheral T to meet the challenge of self-reactivity. Immunol Today 1998; 19:448–
cell unresponsiveness under steady state conditions in vivo. J Exp Med 454.
2001; 194:769–779. 32. Grohmann U, Fallarino F, Bianchi R, Belladonna ML, Vacca C, Orabona

Downloaded from https://academic.oup.com/biolreprod/article/77/4/590/2629671 by guest on 10 February 2022


8. Aoki K, Kajiura S, Matsumoto Y, Ogasawara M, Okada S, Yagami Y, C, Uyttenhove C, Fioretti MC, Puccetti P. IL-6 inhibits the tolerogenic
Gleicher N. Preconceptional natural-killer-cell activity as a predictor of function of CD8 alpha þ dendritic cells expressing indoleamine 2,3-
miscarriage. Lancet 1995; 345:1340–1342. dioxygenase. J Immunol 2001; 167:708–714.
9. Arck PC, Ferrick DA, Steele-Norwood D, Croitoru K, Clark DA. 33. Groux H, Fournier N, Cottrez F. Role of dendritic cells in the generation
Regulation of abortion by gamma delta T cells. Am J Reprod Immunol of regulatory T cells. Semin Immunol 2004; 2:99–106.
1997; 37:87–93. 34. Wakkach A, Fournier N, Brun V, Breittmayer JP, Cottrez F, Groux H.
10. Nagaeva O, Jonsson L, Mincheva-Nilsson L. Dominant IL-10 and TGF- Characterization of dendritic cells that induce tolerance and T regulatory
beta mRNA expression in gamma delta T cells of human early pregnancy 1 cell differentiation in vivo. Immunity 2003; 18:605–617.
decidua suggests immunoregulatory potential. Am J Reprod Immunol 35. Clark DA, Keil A, Chen Z, Markert U, Manuel J, Gorczynski RM.
2002; 48:9–17. Placental trophoblast from successful human pregnancies expresses the
11. Clark DA, Croitoru K. TH1/TH2,3 imbalance due to cytokine-producing tolerance signaling molecule, CD200 (OX-2). Am J Reprod Immunol
NK, gamma delta T and NK-gamma delta T cells in murine pregnancy 2003; 50:187–195.
decidua in success or failure of pregnancy. Am J Reprod Immunol 2001; 36. Gorczynski RM, Lee L, Boudakov I. Augmented Induction of
45:257–265. CD4þCD25þ Treg using monoclonal antibodies to CD200R. Transplan-
12. Ito K, Karasawa M, Kawano T, Akasaka T, Koseki H, Akutsu Y, Kondo tation 2005; 79:1180–1183.
E, Sekiya S, Sekihawa K, Harada M, Yamashita M, Nakayama T, et al. 37. Rieger L, Honig A, Sutterlin M, Kapp M, Dietl J, Ruck P, Kammerer U.
Involvement of decidual Valpha14 NKT cells in abortion. Proc Natl Antigen-presenting cells in human endometrium during the menstrual
Acad Sci U S A 2000; 97:740–744. cycle compared to early pregnancy. J Soc Gynecol Investig 2004; 11:
13. Munn DH, Zhou M, Attwood JT, Bondarev I, Conway SJ, Marshall B, 488–493.
Brown C, Mellor AL. Prevention of allogeneic fetal rejection by 38. Gardner L, Moffett A. Dendritic cells in the human decidua. Biol Reprod
tryptophan catabolism. Science 1998; 281:1191–1193. 2003; 69:1438–1446.
14. Aluvihare VR, Kallikourdis M, Betz AG. Regulatory T cells mediate 39. Kammerer U, Eggert AO, Kapp M, McLellan AD, Geijtenbeek TB, Dietl
maternal tolerance to the fetus. Nat Immunol 2004; 3:266–271. J, van Kooyk Y, Kampgen E. Unique appearance of proliferating
15. Dealtry GB, O’Farrell MK, Fernandez N. The Th2 cytokine environment antigen-presenting cells expressing DC-SIGN (CD209) in the decidua of
of the placenta. Int Arch Allergy Immunol 2000; 123:107–119. early human pregnancy. Am J Pathol 2003; 162:887–896.
16. Chaouat G, Ledee-Bataille N, Dubanchet S, Zourbas S, Sandra O, Martal 40. Miyazaki S, Tsuda H, Sakai M, Hori S, Sasaki Y, Futatani T, Miyawaki
J. TH1/TH2 paradigm in pregnancy: paradigm lost? Cytokines in T, Saito S. Predominance of Th2-promoting dendritic cells in early
pregnancy/early abortion: reexamining the TH1/TH2 paradigm. Int Arch human pregnancy decidua. J Leukoc Biol 2003; 74:514–522.
Allergy Immunol 2004; 134:93–119. 41. Blois S, Tometten M, Kandil J, Hagen E, Klapp BF, Margni RA, Arck
17. Blois SM, Alba Soto CD, Tometten M, Klapp BF, Margni RA, Arck PC. PC. Intercellular adhesion molecule-1/LFA-1 cross talk is a proximate
Lineage, maturity, and phenotype of uterine murine dendritic cells mediator capable of disrupting immune integration and tolerance
throughout gestation indicate a protective role in maintaining pregnancy. mechanism at the feto-maternal interface in murine pregnancies. J
Biol Reprod 2004; 70:1018–1023. Immunol 2005; 174:1820–1829.
18. Chaouat G, Assal Meliani A, Martal J, Raghupathy R, Elliott JF, 42. Askelund K, Liddell HS, Zanderigo AM, Fernando NS, Khong TY,
Mosmann T, Wegmann TG. IL-10 prevents naturally occurring fetal loss Stone PR, Chamley LW. CD83(þ)dendritic cells in the decidua of
in the CBA 3 DBA/2 mating combination, and local defect in IL-10 women with recurrent miscarriage and normal pregnancy. Placenta 2004;
production in this abortion-prone combination is corrected by in vivo 25:140–145.
injection of IFN-tau. J Immunol 1995; 154:4261–4268. 43. Moffett-King A. Natural killer cells and pregnancy. Nat Rev Immunol
19. Knackstedt MK, Zenclussen AC, Hertwig K, Hagen E, Dudenhausen 2002; 2:656–663.
JW, Clark DA, Arck PC. Th1 cytokines and the prothrombinase fgl2 in 44. Bulmer JN, Johnson PM. Immunohistological characterization of the
stress-triggered and inflammatory abortion. Am J Reprod Immunol 2003; decidual leucocytic infiltrate related to endometrial gland epithelium in
49:210–220. early human pregnancy. Immunology 1985; 55:35–44.
20. Mowat AM. Anatomical basis of tolerance and immunity to intestinal 45. Croy BA. Granulated metrial gland cells—interesting cells found in the
antigens. Nat Rev Immunol 2003; 3:331–341. pregnant uterus. Am J Reprod Immunol 1990; 1:19–21.
21. Kalinski P, Hilkens CM, Snijders A, Snijdewint FG, Kapsenberg ML. 46. Barber EM, Pollard JW. The uterine NK cell population requires IL-15
Dendritic cells, obtained from peripheral blood precursors in the presence but these cells are not required for pregnancy nor the resolution of a
of PGE2, promote Th2 responses. Adv Exp Med Biol 1997; 417:363– Listeria monocytogenes infection. J Immunol 2003; 171:37–46.
367. 47. Moretta L. Lymphocyte effector mechanisms in innate and adaptive
22. Rutella S, Danese S, Leone G. Tolerogenic dendritic cells: Cytokine immunity. Curr Opin Immunol 2005; 3:303–305.
modulation comes of age. Blood 2006; 108:1435–1440. 48. Zitvogel L. Dendritic and natural killer cells cooperate in the control/
23. Hackstein H, Morelli AE, Thomson AW. Designer dendritic cells for switch of innate immunity. J Exp Med 2002; 195:9–14.
tolerance induction: guided not misguided missiles. Trends Immunol 49. Gerosa F, Baldani-Guerra B, Nisii C, Marchesini V, Carra G, Trinchieri
2001; 8:437–442. G. Reciprocal activating interaction between natural killer cells and
24. Dhodapkar MV, Young JW, Chapman PB, Cox WI, Fonteneau JF, dendritic cells. J Exp Med 2002; 195:327–333.
Amigorena S, Houghton AN, Steinman RM, Bhardwaj N. Paucity of 50. Piccioli D, Sbrana S, Melandri E, Valiante NM. Contact-dependent
functional T-cell memory to melanoma antigens in healthy donors and stimulation and inhibition of dendritic cells by natural killer cells. J Exp
melanoma patients. Clin Cancer Res 2000; 6:4831–4838. Med 2002; 195:335–341.
25. Steinman RM. Dendritic cells and the control of immunity: enhancing the 51. Juretic K, Strbo N, Crncic TB, Laskarin G, Rukavina D. An insight into
efficiency of antigen presentation. Mt Sinai J Med 2001; 68:160–166. the dendritic cells at the maternal-fetal interface. Am J Reprod Immunol
26. Akbari O, DeKruyff RH, Umetsu DT. Pulmonary dendritic cells 2004; 52:350–355.
DENDRITIC CELLS AND PREGNANCY TOLERANCE 597

52. Maldonado-Lopez R, De Smedt T, Michel P, Godfroid J, Pajak B, 76. Szekeres-Bartho J, Kinsky R, Kapovic M, Chaouat G. Complete Freund
Heirman C, Thielemans K, Leo O, Urbain J, Moser M. CD8alpha þ and adjuvant treatment of pregnant females influences resorption rates in
CD8alpha- subclasses of dendritic cells direct the development of distinct CBA/J 3 DBA/2 matings via progesterone-mediated immunomodulation.
T helper cells in vivo. J Exp Med 1999; 189:587–592. Am J Reprod Immunol 1991; 26:82–83.
53. Trinchieri G. Interleukin-12 and the regulation of innate resistance and 77. Blois SM, Joachim R, Kandil J, Margni R, Tometten M, Klapp BF, Arck
adaptive immunity. Nat Rev Immunol 2003; 3:133–146. PC. Depletion of CD8 þ cells abolishes the pregnancy protective effect of
54. Pulendran B, Lingappa J, Kennedy MK, Smith J, Teepe M, Rudensky A, progesterone substitution with dydrogesterone in mice by altering the
Maliszewski CR, Maraskovsky E. Developmental pathways of dendritic Th1/Th2 cytokine profile. J Immunol 2004; 172:5893–5899.
cells in vivo: distinct function, phenotype, and localization of dendritic 78. Mellor AL, Chandler P, Lee GK, Johnson T, Keskin DB, Lee J, Munn
cell subsets in FLT3 ligand-treated mice. J Immunol 1997; 159:2222– DH. Indoleamine 2,3-dioxygenase, immunosuppression and pregnancy. J
2231. Reprod Immunol 2002; 57:143–150.
55. Reis e Sousa C, Hieny S, Scharton-Kersten T, Jankovic D, Charest H, 79. Fallarino F, Grohmann U, Vacca C, Orabona C, Spreca A, Fioretti MC,
Germain RN, Sher A. In vivo microbial stimulation induces rapid CD40 Puccetti P. T cell apoptosis by kynurenines. Adv Exp Med Biol 2003;
ligand-independent production of interleukin 12 by dendritic cells and 527:183–190.
their redistribution to T cell areas. J Exp Med 1997; 186:1819–1829. 80. Moffett JR, Namboodiri MA. Tryptophan and the immune response.
56. Pulendran B, Smith JL, Caspary G, Brasel K, Pettit D, Maraskovsky E, Immunol Cell Biol 2003; 81:247–265.
Maliszewski CR. Distinct dendritic cell subsets differentially regulate the 81. Tatsumi K, Higuchi T, Fujiwara H, Nakayama T, Egawa H, Itoh K, Fujii

Downloaded from https://academic.oup.com/biolreprod/article/77/4/590/2629671 by guest on 10 February 2022


class of immune response in vivo. Proc Natl Acad Sci U S A 1999; 96: S, Fujita J. Induction of tryptophan 2,3-dioxygenase in the mouse
1036–1041. endometrium during implantation. Biochem Biophys Res Commun 2000;
57. McKenzie GJ, Emson CL, Bell SE, Anderson S, Fallon P, Zurawski G, 274:166–170.
Murray R, Grencis R, McKenzie AN. Impaired development of Th2 cells 82. Baban B, Chandler P, McCool D, Marshall B, Munn DH, Mellor AL.
in IL-13-deficient mice. Immunity 1998; 9:423–432. Indoleamine 2,3-dioxygenase expression is restricted to fetal trophoblast
58. Rincon M, Flavell RA. T-cell subsets: transcriptional control in the Th1/ giant cells during murine gestation and is maternal genome specific. J
Th2 decision. Curr Biol 1997; 7:R729–732. Reprod Immunol 2004; 61:67–77.
59. Groux H, Bigler M, de Vries JE, Roncarolo MG. Interleukin-10 induces a 83. Mackler AM, Barber EM, Takikawa O, Pollard JW. Indoleamine 2,3-
long-term antigen-specific anergic state in human CD4 þ T cells. J Exp dioxygenase is regulated by IFN-gamma in the mouse placenta during
Med 1996; 184:19–29. Listeria monocytogenes infection. J Immunol 2003; 170:823–830.
60. d’Ostiani CF, Del Sero G, Bacci A, Montagnoli C, Spreca A, Mencacci 84. Moser M, De Smedt T, Sornasse T, Tielemans F, Chentoufi AA, Muraille
A, Ricciardi-Castagnoli P, Romani L. Dendritic cells discriminate E, Van Mechelen M, Urbain J, Leo O. Glucocorticoids down-regulate
between yeasts and hyphae of the fungus Candida albicans. Implications dendritic cell function in vitro and in vivo. Eur J Immunol 1995; 25:
for initiation of T helper cell immunity in vitro and in vivo. J Exp Med 2818–2824.
2000; 191:1661–1674. 85. Xing N, Maldonado ML, Bachman LA, McKean DJ, Kumar R, Griffin
61. Lin H, Mosmann TR, Guilbert L, Tuntipopipat S, Wegmann TG. MD. Distinctive dendritic cell modulation by vitamin D(3) and
Synthesis of T helper 2-type cytokines at the maternal-fetal interface. J glucocorticoid pathways. Biochem Biophys Res Commun 2002; 297:
Immunol 1993; 151:4562–4573. 645–652.
62. Piccinni MP, Beloni L, Livi C, Maggi E, Scarselli G, Romagnani S. 86. O’Malley BW, Tsai MJ. Molecular pathways of steroid receptor action.
Defective production of both leukemia inhibitory factor and type 2 T- Biol Reprod 1992; 46:163–167.
helper cytokines by decidual T cells in unexplained recurrent abortions. 87. Lydon JP, DeMayo FJ, Funk CR, Mani SK, Hughes AR, Montgomery
Nat Med 1998; 4:1020–1024. CA Jr, Shyamala G, Conneely OM, O’Malley BW. Mice lacking
63. Arck PC, Troutt AB, Clark DA. Soluble receptors neutralizing TNF- progesterone receptor exhibit pleiotropic reproductive abnormalities.
alpha and IL-1 block stress-triggered murine abortion. Am J Reprod Genes Dev 1995; 9:2266–2278.
Immunol 1997; 37:262–266. 88. Paria BC, Tan J, Lubahn DB, Dey SK, Das SK. Uterine decidual
64. Rissoan MC, Soumelis V, Kadowaki N, Grouard G, Briere F, de Waal response occurs in estrogen receptor-alpha-deficient mice. Endocrinology
Malefyt R, Liu YJ. Reciprocal control of T helper cell and dendritic cell 1999; 140:2704–2710.
differentiation. Science 1999; 283:1183–1186. 89. Liu HY, Buenafe AC, Matejuk A, Ito A, Zamora A, Dwyer J,
65. Sakaguchi S. Naturally arising CD4 þ regulatory t cells for immunologic Vandenbark AA, Offner H. Estrogen inhibition of EAE involves effects
self-tolerance and negative control of immune responses. Annu Rev on dendritic cell function. J Neurosci Res 2002; 70:238–248.
Immunol 2004; 22:531–562. 90. Liang J, Sun L, Wang Q, Hou Y. Progesterone regulates mouse dendritic
66. Shevach EM. CD4þ CD25þ suppressor T cells: more questions than cells differentiation and maturation. Int Immunopharmacol 2006; 5:830–
answers. Nat Rev Immunol 2002; 2:389–400. 838.
67. Read S, Powrie F. CD4(þ) regulatory T cells. Curr Opin Immunol 2001; 91. Butts CL, Shukair SA, Duncan KM, Bowers E, Horn C, Belyavskaya E,
13:644–649. Tonelli L, Sternberg EM. Progesterone inhibits mature rat dendritic cells
68. Graca L, Cobbold SP, Waldmann H. Identification of regulatory T cells in a receptor-mediated fashion. Int Immunol 2007; 3:287–296.
in tolerated allografts. J Exp Med 2002; 195:1641–1646. 92. Ivanova E, Kyurkchiev D, Altankova I, Dimitrov J, Binakova E,
69. Wood KJ, Sakaguchi S. Regulatory T cells in transplantation tolerance. Kyurkchiev S. CD83 monocyte-derived dendritic cells are present in
Nat Rev Immunol 2003; 3:199–210. human decidua and progesterone induces their differentiation in vitro.
70. Somerset DA, Zheng Y, Kilby MD, Sansom DM, Drayson MT. Normal Am J Reprod Immunol 2005; 4:199–205.
human pregnancy is associated with an elevation in the immune 93. Evans KN, Nguyen L, Chan J, Innes BA, Bulmer JN, Kilby MD,
suppressive CD25 þ CD4 þ regulatory T-cell subset. Immunology Hewison M. Effects of 25-hydroxyvitamin D3 and 1,25-dihydroxyvita-
2004; 112:38–43. min D3 on cytokine production by human decidual cells. Biol Reprod
71. Heikkinen J, Mottonen M, Alanen A, Lassila O. Phenotypic character- 2006; 75:816–822.
ization of regulatory T cells in the human decidua. Clin Exp Immunol 94. Diaz L, Sanchez I, Avila E, Halhali A, Vilchis F, Larrea F. Identification
2004; 136:373–378. of a 25-hydroxyvitamin D3 1alpha-hydroxylase gene transcription
72. Gorczynski RM. Thymocyte/splenocyte-derived CD4 þCD25 þTreg product in cultures of human syncytiotrophoblast cells. J Clin Endocrinol
stimulated by anti-CD200R2 derived dendritic cells suppress mixed Metab 2000; 85:2543–2549.
leukocyte cultures and skin graft rejection. Transplantation 2006; 81: 95. Zehnder D, Evans KN, Kilby MD, Bulmer JN, Innes BA, Stewart PM,
1027–1034. Hewison M. The ontogeny of 25-hydroxyvitamin D(3) 1alpha-hydrox-
73. Weiner HL. Oral tolerance for the treatment of autoimmune diseases. ylase expression in human placenta and decidua. Am J Pathol 2002; 161:
Annu Rev Med 1997; 48:341–351. 105–114.
74. Manavalan JS, Kim-Schulze S, Scotto L, Naiyer AJ, Vlad G, Colombo 96. Taylor HS, Vanden Heuvel GB, Igarashi P. A conserved Hox axis in the
PC, Marboe C, Mancini D, Cortesini R, Suciu-Foca N. Alloantigen mouse and human female reproductive system: late establishment and
specific CD8 þCD28 FOXP3 þ T suppressor cells induce ILT3 þ persistent adult expression of the Hoxa cluster genes. Biol Reprod 1997;
ILT4 þ tolerogenic endothelial cells, inhibiting alloreactivity. Int 57:1338–1345.
Immunol 2004; 16:1055–1068. 97. Harizi H, Juzan M, Pitard V, Moreau JF, Gualde N. Cyclooxygenase-2-
75. Arck PC, Merali F, Chaouat G, Clark DA. Inhibition of immunopro- issued prostaglandin e(2) enhances the production of endogenous IL-10,
tective CD8 þ T cells as a basis for stress-triggered substance P-mediated which down-regulates dendritic cell functions. J Immunol 2002; 168:
abortion in mice. Cell Immunol 1996; 171:226–230. 2255–2263.
598 BLOIS ET AL.

98. Lechler RI, Batchelor JR. Restoration of immunogenicity to passenger 106. Gao JX, Madrenas J, Zeng W, Cameron M, Zhang Z, Wang JJ, Zhong R.
cell depleted kidney allografts by the addition of donor strain dendritic CD-40 deficient dendritic cells producing interleukin-10, but not
cells. J Exp Med 1982; 155:31–41. interleukin-12, induce T-cell hyporesponsiveness in vitro and prevent
99. Larsen CP, Morris PJ, Austyn JM. Migration of dendritic leukocytes acute allograft rejection. Immunology 1999; 98:159–170.
from cardiac allografts into host spleens. A novel pathway for initiation 107. O’Connell PJ, Li W, Wang Z, Specht SM, Logar AJ, Thomson AW.
of rejection. J Exp Med 1990; 171:307–314. Immature and mature CD8alpha þ dendritic cells prolong the survival of
100. Lu L, Rudert WA, Qian S, McCaslin D, Fu F, Rao AS, Trucco M, Fung vascularized heart allografts. J Immunol 2002; 168:143–154.
JJ, Starzl TE, Thomson AW. Growth of donor-derived dendritic cells 108. Hayamizu K, Huie P, Sibley RK, Strober S. Monocyte-derived dendritic
from the bone marrow of murine liver allgraft recipients in response to cell precursors facilitate tolerance to heart allografts after total lymphoid
granulocyte/macrophage colony stimulating factor. J Exp Med 1995; irradiation. Transplantation 1998; 66:1285–1291.
182:379–387. 109. Mirenda V, Berton I, Read J, Cook T, Smith J, Dorling A, Lechler RI.
101. Khanna A, Steptoe RJ, Antonysamy MA, Li W, Thomson AW. Donor Modified dendritic cells coexpressing self and allogeneic major
bone marrow potentiates the effect of tacrolimus on nonvascularized histocompatability complex molecules: an efficient way to induce
heart allograft survival: association with microchimerism and growth of indirect pathway regulation. J Am Soc Nephrol 2004; 15:987–997.
donor dendritic cell progenitors from recipient bone marrow. Transplan-
110. Lutz MB, Suri RM, Niimi M, Ogilvie AL, Kukutsch NA, Rossner S,
tation 1998; 65:479–485.
Schuler G, Austyn JM. Immature dendritic cells generated with low
102. Lu L, McCaslin D, Starzl TE, Thomson AW. Bone marrow-derived

Downloaded from https://academic.oup.com/biolreprod/article/77/4/590/2629671 by guest on 10 February 2022


doses of GM-CSF in the absence of IL-4 are maturation resistant and
dendritic cell progenitors (NLDC 145þ, MHC class II þ, B7–1dim, B7–2)
prolong allograft survival in vivo. Eur J Immunol 2000; 30:1813–1822.
induce alloantigen-specific hyporesponsiveness in murine T lympho-
cytes. Transplantation 1995; 60:1539–1545. 111. Lan YY, Wang Z, Raimondi G, Wu W, Colvin BL, DeCreus A,
103. Rastellini C, Lu L, Ricordi C, Starzl TE, Rao AS, Thomson AW. Thomson AW. Alternatively-activated’ dendritic cells preferentially
Granulocyte/macrophage colony-stimulating factor-stimulated hepatic secrete IL-10, expand Foxp3þ CD4þ T cells and induce long-term organ
dendritic cell progenitors prolong pancreatic islet allograft survival. allograft survival in combination with CTLA4-Ig. J Immunol 2006; 177:
Transplantation 1995; 60:1366–1370. 5868–5877.
104. Fu F, Li Y, Qian S, Lu L, Chambers F, Starzl TE, Fung JJ, Thomson 112. Demetris A, Murase N, Ye Q, Galvao F, Richert C, Saad R, Pham S,
AW. Costimulatory molecule-deficient dendritic cell progenitors (MHC Duquesnoy R, Zeevi A, Fung J, Starzl T. Analysis of chronic rejection
class II þ, CD80dim, CD86) prolong cardiac allograft survival in and obliterative arteriolopathy. Possible contributions of donor antigen
nonimmunosuppressed recipients. Transplantation 1996; 62:659–665. presenting cells and lymphatic disruption. Am J Path 1997; 150:563–
105. Roelen DL, Schuurhuis DH, van den Boogaardt DE, Koekkoek K, van 578.
Miert PP, van Schip JJ, Laban S, Rea D, Melief CJ, Offringa R, 113. Ochando JC, Homma C, Yang Y, Hidalgo A, Garin A, Tacke F, Angeli
Ossendorp F, Claas FH. Prolongation of skin graft survival by V, Li Y, Boros P, Ding Y, Jessberger R, Trinchieri G, et al. Alloantigen-
modulation of the alloimmune response with alternatively activated presenting plasmacytoid dendritic cells mediate tolerance to vascularized
dendritic cells. Transplantation 2003; 76:1608–1615. grafts. Nat Immunol 2006; 7:652–662.

You might also like