You are on page 1of 10

Human Chorionic Gonadotropin as a Central

Regulator of Pregnancy Immune Tolerance


Anne Schumacher, Kristina Heinze, Jeanette Witte, Eileen
Poloski, Nadja Linzke, Katja Woidacki and Ana C.
This information is current as Zenclussen
of April 7, 2017.
J Immunol 2013; 190:2650-2658; Prepublished online 8
February 2013;
doi: 10.4049/jimmunol.1202698
http://www.jimmunol.org/content/190/6/2650

Downloaded from http://www.jimmunol.org/ by guest on April 7, 2017


References This article cites 57 articles, 17 of which you can access for free at:
http://www.jimmunol.org/content/190/6/2650.full#ref-list-1
Subscription Information about subscribing to The Journal of Immunology is online at:
http://jimmunol.org/subscription
Permissions Submit copyright permission requests at:
http://www.aai.org/About/Publications/JI/copyright.html
Email Alerts Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright 2013 by The American Association of
Immunologists, Inc. All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
The Journal of Immunology

Human Chorionic Gonadotropin as a Central Regulator of


Pregnancy Immune Tolerance

Anne Schumacher, Kristina Heinze, Jeanette Witte, Eileen Poloski, Nadja Linzke,
Katja Woidacki, and Ana C. Zenclussen
Normal pregnancy is characterized by an early expansion of regulatory T cells (Tregs), which is known to contribute to fetal tol-
erance. However, mechanisms and factors behind Treg expansion are not yet defined. Recently, we proposed that the pregnancy
hormone human chorionic gonadotropin (hCG) efficiently attracts human Tregs to trophoblasts, favoring their accumulation lo-
cally. In this study, we hypothesized that hCG not only acts as a chemoattractant of Tregs but also plays a central role in pregnancy-
induced immune tolerance. Virgin, normal pregnant, and abortion-prone female mice were treated either with 10 IU/ml hCG or
PBS at days 0, 2, 4, and 6 of pregnancy. The hCG effect on Treg frequency and cytokine secretion was determined in Foxp3gfp
females. hCG impact on Treg suppressive capacity was studied in vitro. In vivo, we investigated whether hCG enhances Treg
suppressive capacity indirectly by modulating dendritic cell maturation in an established mouse model of disturbed fetal toler-

Downloaded from http://www.jimmunol.org/ by guest on April 7, 2017


ance. Application of hCG increased Treg frequency in vivo and their suppressive activity in vitro. In females having spontaneous
abortions, hCG provoked not only an augmentation of Treg numbers, but also normalized fetal abortion rates. hCG-generated
Tregs were fully functional and could confer tolerance when adoptively transferred. hCG also retained dendritic cells in a tolero-
genic state that is likely to contribute to both Treg expansion and prevention of abortion. Our results position hCG in a novel, so
far unknown role as modulator of immune tolerance during pregnancy. The Journal of Immunology, 2013, 190: 26502658.

F
etal survival within the maternal uterus is ensured by strong of the fetus (1315). In the 1970s and 1980s, several studies
hormonal changes and the regulation of maternal immune suggested hCG as an important factor modulating T and B cell
responses toward the foreign fetal Ags. The interplay be- responses (16, 17) via the induction of the at that time called
tween hormonal and immunological factors contributes substan- suppressor T cells (18). This concept was no longer followed until
tially to fetal tolerance. Pregnancy-associated hormones like the more recently when Khil and colleagues (19) showed an hCG-
human chorionic gonadotropin (hCG), estrogen, and progesterone mediated inhibitory effect on Th1 responses that was associated
(P) increase during pregnancy and are essential for successful with increased numbers of CD4+CD25+ regulatory T cells (Tregs)
pregnancy outcome (1, 2). hCG is a primate-specific hormone; that in a murine model of autoimmune diabetes. During normal human
is, it is only produced by humans (3) and primates (4), and cannot and murine pregnancy, Tregs expand systemically and locally at
be detected in rodents (5). However, rodents produce the highly the fetalmaternal interface (2022). Disturbed pregnancies are
homologous luteinizing hormone (LH) binding to the same re- associated with a diminished Treg number and activity (2325).
ceptor as hCG (namely, the LH/CG receptor) (6). Because of this Zhou and colleagues (26) recently confirmed the positive associ-
circumstance, hCG effects can be easily studied in the murine ation of Treg expansion with pregnancy establishment as they
system. Immediately after fertilization, hCG is produced by the demonstrated that an increase of Tregs in peripheral blood was
blastocyst and later by the syncytiotrophoblast (7, 8). After a requisite for a successful in vitro fertilization (IVF). We first
reaching its maximum level between the 9th and 12th weeks of introduced Tregs as a useful tool to control disturbed tolerance
pregnancy, hCG concentrations decline until birth (9). In addi- during pregnancy in an abortion-prone (AP) model. We observed
tion to its main function in stimulating P production by the that the transfer of Ag-specific Tregs at early pregnancy stages
corpus luteum, hCG has been described to facilitate trophoblast restored tolerance and thereby prevented fetal rejection (24, 27),
invasion (1012), support angiogenesis, and ensure nourishment which is in agreement with recent findings by Yin and colleagues
(28). Treg transfer induced a tolerant microenvironment directly at
the fetalmaternal interface (29), and Treg protective effects on
Department of Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von- pregnancy were mediated at least in the studied model by IL-10
Guericke University, 39108 Magdeburg, Germany
and programmed death-1, whereas TGF-b and CTLA-4 rather
Received for publication September 26, 2012. Accepted for publication January 11,
2013.
played a secondary role (27, 30). In humans, we found a correla-
tion between hCG and Treg levels, which are both diminished
This work was supported by the Deutsche Forschungsgemeinschaft (Grant ZE526/7-1
to A.C.Z.) and the Medical Faculty of the Otto-von-Guericke University of Magdeburg in patients suffering from spontaneous abortions or extrauterine
(Ph.D. grant to A.S.). pregnancies when compared with normal pregnant (NP) women
Address correspondence and reprint requests to Prof. Ana C. Zenclussen, Experi- (31). We also found hCG to attract Tregs, which express the LH/
mental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University CG receptor, into the fetalmaternal interface (31). Thus, hCG
Magdeburg, Gerhart-Hauptmann-Strasse 35, 39108 Magdeburg, Germany. E-mail
address: ana.zenclussen@med.ovgu.de acts as a chemoattractant to Tregs at early pregnancy stages. This
Abbreviations used in this article: AP, abortion-prone; DC, dendritic cell; hCG, gives rise to a concept in which the very first hormone produced
human chorionic gonadotropin; IVF, in vitro fertilization; LH, luteinizing hormone; by the trophoblast, hCG, contributes to the recruitment of immune
LH/CG, luteinizing hormone/chorionic gonadotropin; NP, normal pregnant; P, pro- cells to the fetalmaternal interface that are pivotal for later tol-
gesterone; Treg, regulatory T cell.
erating the conceptus. In this work, we followed the hypothesis
Copyright 2013 by The American Association of Immunologists, Inc. 0022-1767/13/$16.00 that hCG would not only be a chemoattractant, but also an im-

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1202698
The Journal of Immunology 2651

portant modulator of Treg-mediated fetal tolerance by influencing conjugated anti-mouse CD11c (clone HL3), FITC-labeled anti-mouse
their numbers and/or suppressive capacity. To study whether hCG- CD80 (clone 16-10A1), and PE-conjugated anti-mouse I-A/I-E (clone
M5/114.15.2). Foxp3 Ab was purchased from eBioscience (Kranenburg,
mediated effects are of direct or indirect nature, we also concen- Germany). All other Abs were purchased from Becton Dickinson.
trated on the effects of hCG on the maturity state of dendritic cells
(DCs) that act as APCs at the very first step of an adaptive immune FACS and cell culture
response. For in vitro studies, Foxp3+ cells were sorted from spleen or draining
lymph nodes from BALB/c-mated Foxp3gfp transgenic females (treated
Materials and Methods with either PBS or hCG), washed in ice-cold PBS, and kept in RPMI 1640
medium supplemented with 10% FBS (Biochrom, Berlin, Germany) and
Animals 100 ng/ml penicillin/streptomycin (Invitrogen, Karlsruhe, Germany) at 4
Wild type CBA/J female mice and BALB/c and DBA/2J male mice were C. Mononuclear cells were isolated by disaggregating the tissue, filtering it
purchased from Charles River (Germany), maintained in our animal facility, through a 100-mm cell strainer (Becton Dickinson), and lysing the eryth-
and treated according to the institutional guidelines with ministerial ap- rocytes with an NHCl4/NaCl solution. After lysis, cells were stained with
proval (Landesverwaltungsamt Sachsen-Anhalt AZ2/868 and AZ42502-2- PerCp-Cy5.5conjugated anti-mouse CD4 (clone RM4-5) from Becton
1125UniMD). The experiments were conducted by authorized persons Dickinson. CD4 +GFP + (Foxp3 + ) Tregs were sorted by FACS using a
according to the Guide for Care and Use of Animals in Agriculture FACSDiva Flow Cytometer and Cell Sorter from Becton Dickinson.
Research and Teaching. DBA/2J-mated CBA/J females (AP combina- Sorted Tregs were cultured for 24 h in RPMI medium supplemented with
tion) are known to spontaneously develop high abortion rates (24), whereas FBS and penicillin/streptomycin. To induce cytokine secretion, we added
BALB/c-mated CBA/J females (NP combination) represented the control 1 mg/ml ionomycin (Sigma-Aldrich, Steinheim, Germany) and 50 ng/ml
animals. Foxp3gfp transgenic females on a C57BL/6 background, kindly PMA (Sigma-Aldrich) to the culture. After 24 h, supernatants were taken
provided by Dr. Rudensky, were allogeneically mated to BALB/c males. for analysis of IL-10 and TGF-b by ELISA.
In these animals, the complete coding sequence of the GFP (gfp) is
inserted in the first coding exon of the Foxp3 gene, which results in ELISA

Downloaded from http://www.jimmunol.org/ by guest on April 7, 2017


a chimeric GFP-Foxp3 fusion protein (32) that enables the easy identifi-
The amounts of IL-10 and TGF-b were determined in the supernatants of
cation of Foxp3+ Tregs. Animals were paired and checked twice a day for
isolated Tregs from hCG- or PBS-treated pregnant Foxp3gfp females on day
vaginal plug, whose appearance indicated day 0 of pregnancy. Virgin and
10 of pregnancy by using the BD OptEIA ELISA Set for IL-10 provided
pregnant animals were injected i.p. with 10 IU/ml hCG (Sigma Aldrich,
by Becton Dickinson and the Ready-Set-Go Kit for TGF-b provided by
Steinheim, Germany) or PBS (PAA Laboratories, Colbe, Germany). hCG
eBioscience (Frankfurt, Germany). All steps were performed according to
and PBS injections were performed at days 0, 2, 4, and 6 of gestation or
the instructions of the manufacturer.
experiment, and females were sacrificed on day 10 of pregnancy or 4 d
after the last injection in nonpregnant animals.
Magnetic cell isolation and adoptive transfer of Tregs
Sample collection and flow cytometry To investigate whether hCG induces the generation of pregnancy-protective
Blood was obtained by retroorbital puncture under anesthesia. Females Tregs in vivo, we isolated CD4+CD25+ cells by magnetic beads (MACS;
were sacrificed by cervical dislocation; spleen, thymus, para-aortic lymph Miltenyi Biotec, Bergisch Gladbach, Germany) from a mixture of spleen
nodes, and uterine tissue were removed, washed in ice-cold PBS, and kept in and thymus from AP animals (day 12 of pregnancy) previously treated
RPMI 1640 medium at 4C. Pregnant uteri were opened longitudinally, and with either 10 IU/ml hCG or PBS. The purity of the isolated Tregs was
the number of implantations and the abortion rate were determined. 95%. A total of 2 3 105 Tregs, diluted in 200 ml PBS, were adoptively
Fetoplacental units were separated from their implantation sites; deciduae transferred into AP animals by i.v. injection on days 02 of pregnancy. The
were cut in small pieces and collected in HBSS without Ca2+ and Mg2+ used number of 2 3 105 Tregs, adoptively transferred into AP animals, was
(Sigma, Taufkirchen, Germany). Mononuclear cells from blood, spleen, proved to be the minimum cell number essential to confer fetal protection
thymus, para-aortic lymph nodes, uterus, and decidua samples were iso- in this model (24).
lated using our established protocol (24). Thereafter, cells were stained for Suppression assay and conversion assay
extracellular and intracellular markers as described elsewhere (24) and
read on a FACSCalibur (Becton Dickinson, Heidelberg, Germany). The To study the influence of hCG on Treg function, we isolated CD4+CD252
following Abs were used: FITC-conjugated rat anti-mouse CD4 (clone responder T cells and CD4+CD25+ Tregs by magnetic beads from a mix-
RM4-4), PE-labeled anti-mouse Foxp3 (clone FJK-16s), allophycocyanin- ture of spleen and draining lymph nodes (para-aortic plus inguinal lymph

FIGURE 1. hCG increased Treg number and their


capability to secrete suppressive cytokines. Application
of 10 IU/ml hCG in BALB/c-mated Foxp3gfp females
significantly increased the numbers of Foxp3-GFP+
cells (Tregs) in blood [(A); n = 7/group] and decidua
[(B); n = 79/group] when compared with PBS treat-
ment. Moreover, to assess cytokine secretion by Tregs
obtained from either hCG- or PBS-treated pregnant
Foxp3gfp females, we cultured Tregs for 24 h in the
presence of ionomycin and PMA. Tregs obtained from
hCG-treated females secreted increased amounts of
IL-10 [(C); n = 6 per group] and TGF-b [(D); n = 3 per
group] when compared with Tregs obtained from PBS-
treated females. (A and B) Each square represents one
single animal. Data are either presented as medians (A,
B) or means + SD (C, D), and statistical analysis was
carried out by the MannWhitney U test or Student
t test, respectively. *p , 0.05, **p , 0.01.
2652 hCG STIMULATES Treg AND SUPPORTS PREGNANCY

1 mg/ml collagenase type IV (Invitrogen, Karlsruhe, Germany), 0.2 mg/ml


DNase I (Roche Diagnostics, Mannheim, Germany), and 1 mg/ml BSA
(Sigma-Aldrich). Afterward, cell suspensions were filtered through a 100-
mm cell strainer (decidua) or 70- and 40-mm cell strainers (spleen).
Mononuclear cells were obtained by density gradient centrifugation. Iso-
lation of CD11c+ DCs was performed by MACS isolation. Then CD11c+
DCs were cultured for 24 and 48 h with CD4+ T cells from draining lymph
nodes (MACS isolation), and expression of Foxp3 was analyzed by flow
cytometry.
LH/CG receptor detection on Tregs and DCs by flow cytometry
To confirm the presence of the LH/CG receptor on both immune cell
populations, CD4+CD25+ Tregs were isolated from a mixture of spleen and
thymus, whereas CD11c+ DCs were isolated from spleen of virgin CBA/J
females by magnetic beads. Isolated Tregs and DCs were stained using the
polyclonal rabbit anti-mouse LH/CG receptor Ab (Acris, Hiddenhausen,
Germany) for 1 h at room temperature. Then cells were stained using an
FITC-conjugated goat anti-rabbit IgG Ab for 30 min at 4C. Afterward,
cells were analyzed by flow cytometry, and data are presented as histo-
grams using FlowJo 7.6.5 software (Tree Star).

Data analysis and statistics


Data obtained for in vitro assays and cytokine determinations by ELISA are

Downloaded from http://www.jimmunol.org/ by guest on April 7, 2017


presented as means + SD. Statistical analysis was realized by performing the
Student t test. All other data are presented as medians in graphs showing
individual values for each animal. Analysis of statistical differences among
all groups was performed using the nonparametric KruskalWallis test. For
analyzing differences between two particular groups, the MannWhitney
U test was applied. In all cases, p , 0.5 was considered to be statistically
significant.

Results
Application of hCG provoked an increase in Treg number and
their capability to secrete suppressive cytokines
In this study, we investigated whether hCG influences Treg expan-
sion and suppressive function in murine pregnancy. For this, we used
FIGURE 2. hCG application normalized Treg frequencies in AP transgenic Foxp3gfp female mice allogeneically mated to BALB/c
females. Application of 10 IU/ml hCG in AP females (n = 69/organ) males. Foxp3gfp females were treated with hCG or PBS at days 0, 2,
significantly increased the number of CD4+Foxp3+ cells (Tregs) within the 4, and 6 of gestation, and the frequency of Tregs was determined by
CD4+ cell population in thymus (A), para-aortic lymph nodes (B), blood flow cytometry in several organs. We observed a significant ex-
(C), and decidua (D) when compared with PBS-treated AP females (n = 6
pansion of Tregs in blood and decidua of hCG-treated females when
7/organ). No significant differences in the Treg number could be detected
between hCG- (n = 56/organ) and PBS-treated (n = 56/organ) NP
compared with PBS-treated controls (Fig. 1A, 1B). In lymph nodes,
females. Each square represents one single animal, and the lines show the spleen, and thymus, no significant differences among the groups
medians. Statistical analysis was carried out by KruskalWallis test fol- were found (data not shown). To further evaluate whether hCG
lowed by MannWhitney U test. *p , 0.05, **p , 0.01. additionally influences the cytokine secretion pattern of Tregs, we
examined the amount of IL-10 and TGF-b, both known to mediate
nodes) from AP females previously treated with hCG or PBS. Responder Treg suppressive capacity (33, 34). hCG treatment increased, al-
T cells were stained with CFSE (Vybrant CFDA-SE Cell Tracer Kit; though not significantly, the capability of Tregs to secrete IL-10
Molecular Probes, Leiden, The Netherlands) to determine proliferation and and TGF-b (Fig. 1C, 1D) when compared with PBS. Thus, hCG
cultured for 24 and 48 h with or without Tregs in a ratio of 1:1. This ratio provokes an expansion of Tregs and fosters their IL-10 and TGF-b
was determined in previous coculture experiments where we titrated the
number of Tregs to responder T cells and found best results in the ratio of
secretion.
1:1 (24). Proliferation of responder T cells was stimulated by the presence hCG application restored tolerance and diminished fetal
of mitomycin C (Sigma-Aldrich)inactivated splenocytes from DBA/2J
males. Proliferation rate was assessed by flow cytometry. Moreover, rejection by normalizing Treg frequencies
CD11c+ DCs were isolated from spleen and decidua from hormone or We hypothesized that hCG enables fetal survival by promoting
control-treated AP females to study the effect of hCG on DC capability
to promote Treg induction. For this, spleen and decidua tissue were cut Treg expansion. To prove this, we determined the systemic and
into small pieces and enzymatically digested for 20 min at 37C in local number of Tregs in hCG- or PBS-treated NP or AP females.
HBSS medium supplemented with 200 U/ml hyaluronidase (Sigma-Aldrich), hCG application at days 0, 2, 4, and 6 resulted in significantly in-

Table I. hCG increased the number of local Tregs in virgin females

Group n Tregs in Spleen (%) Tregs in Blood (%) Tregs in Uterus (%)

CBA/J + PBS 4 4.08 1.63 12.30


CBA/J + hCG 4 4.52 1.06 17.08
Application of 10 IU/ml hCG in virgin CBA/J females increased the number of Tregs in uterus as compared with PBS
treatment but did not change the levels of Treg in spleen and blood. Data are presented as medians. Statistical analysis was
carried out by the MannWhitney U test. No statistical differences could be detected between hCG- and PBS-treated females.
The Journal of Immunology 2653

creased Treg numbers in thymus, para-aortic lymph nodes, blood, 2J-mated AP CBA/J females and adoptively transferred them into
and decidua (Fig. 2). The levels of Tregs in control pregnant mice AP animals at days 02 of pregnancy. As expected, Tregs obtained
were only slightly augmented (Fig. 2). In virgin CBA/J females, from PBS-treated, DBA/2J-mated CBA/J females were not able to
hCG application increased the number of Tregs in uterus as confer fetal protection to AP females because of their impaired
compared with PBS but did not change the levels of Tregs in spleen functionality (Fig. 4A). By contrast, Tregs obtained from hCG-
and blood (Table I). Thus, hCG seems to expand Tregs in the uterus treated, DBA/2J-mated CBA/J females completely rescued fetuses
regardless of an ongoing pregnancy, whereas systemic expansion from immune rejection after adoptive transfer in AP females (Fig.
of Tregs has been observed only in pregnant female mice. 4A). This was not dependent on the number of implantations
To study the effect of hCG-mediated Treg augmentation on because they were all comparable among the groups (Fig. 4B).
pregnancy, we analyzed the abortion and implantation rates after The adoptive transfer of Tregs from hCG-treated females resulted
hCG or PBS treatment. Negative side effects of hCG on pregnancy in a significant increase in systemic and local Treg levels when
were excluded after observing no differences in the pregnancy compared with the transfer of Tregs from PBS-treated females
outcome when compared with PBS-treated NP females and no (Fig. 4CF).
anomalies in any organ (Fig. 3A). hCG application could impres-
sively prevent fetal rejection in AP females when compared with Tregs generated in vivo after hCG application retained their Ag
AP females treated with PBS (Fig. 3A), and this was not dependent specificity toward paternal Ags
on the number of total implantations (Fig. 3B). As shown in Fig. We next wondered whether Tregs isolated from hCG- or PBS-
3C, AP females displayed fetal resorptions together with healthy treated AP females would suppress proliferation of autologous
embryos, whereas hCG-treated AP females had no resorptions. responder T cells in an MLR in vitro and whether this effect is Ag
These results clearly confirm a positive influence of hCG on preg- specific. As expected, Tregs from both hCG- and PBS-treated

Downloaded from http://www.jimmunol.org/ by guest on April 7, 2017


nancy outcome in this model by restoring the levels of Tregs to the animals had a strong suppressive impact on the proliferation rate
values observed in the normal pregnancy combination. of responder T cells when compared with proliferation of responder
hCG application generated fully functional Tregs that can T cells without Tregs (Fig. 5A). Interestingly, we found that Tregs
confer tolerance when adoptively transferred from hCG-treated female mice had a significantly increased sup-
pressive capacity compared with Tregs obtained from PBS-treated
To know whether Tregs generated after hCG injection are fully females (Fig. 5A), which confirms that hCG contributes to Treg
functional, we next isolated Tregs from hCG- or PBS-treated, DBA/ suppressive capacity. To further examine whether the effect of
hCG on Treg function is specific for paternal Ags, we determined
the proliferation rate of responder T cells obtained from a third-
party combination (CBA/J 3 C57BL/6) in the presence of Tregs
from hCG- or PBS-treated AP females. Tregs from both sources
had no effect on the proliferation rate of responder T cells when
compared with their proliferation without Tregs (Fig. 5B). These
results reveal the important influence of hCG on Treg function-
ality: hCG applied to pregnant animals fosters the expansion of
Tregs that are specific against paternal Ags and play, therefore, an
important role in tolerating these Ags.

Tregs and DCs expressed the LH/CG receptor


After we proved an influence of hCG on Treg expansion and
functionality, we wondered whether this effect is mediated di-
rectly by hCG binding to the LH/CG receptor on Treg cell surface
or indirectly by modulation of APCs, that is, DCs, which then
may induce Treg generation. To test both possibilities, we
checked for the presence of the LH/CG receptor on both murine
Tregs and DCs. Interestingly, 97% of Tregs and 69.4% of DCs
(Fig. 6) isolated from virgin CBA/J females expressed the LH/
CG receptor on their surface, making them susceptible to hCG
signaling.
hCG retained DCs in an immature state, and in vitro these cells
favor Treg generation/expansion
FIGURE 3. hCG prevented fetal rejection in AP females. (A) PBS- To evaluate the possibility that the influence of hCG on Treg
treated AP females (n = 10) showed an increased abortion rate when number and function is mediated indirectly via altering the phe-
compared with PBS-treated NP females (n = 6). Fetal rejection in AP notype and activity of other immune cells, especially cells of the
females could be completely prevented by application of 10 IU/ml hCG innate immune system presenting Ags, we investigated the influ-
(n = 12). No differences have been observed between hCG- (n = 6) and ence of hCG on the phenotype of DCs. According to their mat-
PBS-treated NP females. (B) The number of implantations was comparable uration state, DCs support activation of the immune system or
among all groups. (C) Representative pictures of whole implantation sites
suppress immune responses. Mature DCs have been shown to
revealed that PBS-treated AP females showed fetal resorptions among
healthy fetuses, whereas hCG-treated AP females, as well as hCG- and activate T cell responses (35, 36) that might result in fetal rejection
PBS-treated NP females, displayed only healthy implantations. Each whereas immature tolerogenic DCs induce the generation of Tregs
square represents one single animal, and the lines show the medians. favoring fetal protection (37). DCs from hCG-treated AP females
Statistical analysis was carried out by KruskalWallis test followed by were characterized for their maturation state and compared with
MannWhitney U test. ***p , 0.001. DCs obtained from PBS-treated control females. We observed that
2654 hCG STIMULATES Treg AND SUPPORTS PREGNANCY

FIGURE 4. hCG induced fully functional Tregs


that conferred tolerance and further Treg augmen-
tation after their adoptive transfer. (A) Adoptive
transfer of Tregs isolated from hCG-treated AP
females (n = 12) in AP females (n = 11) rescued
females from abortion. By contrast, adoptively
transferred Tregs from PBS-treated AP females (n =
10) in AP females (n = 7) had no effect on the
abortion rate. (B) The number of implantations was
comparable among all groups. AP females (n = 68/
organ) adoptively transferred with Tregs from hCG-
treated AP females (n = 69/organ) showed in-
creased numbers of CD4+Foxp3+ cells (Tregs)
within the CD4+ cell population in thymus (C),
para-aortic lymph nodes (D), blood (E), and decidua
(F) when compared with AP females (n = 7/organ)

Downloaded from http://www.jimmunol.org/ by guest on April 7, 2017


adoptively transferred with Tregs from PBS-treated
AP females (n = 67/organ). Each square represents
one single animal, and the lines show the medians.
Statistical analysis was carried out by Kruskal
Wallis test followed by MannWhitney U test. *p ,
0.05, **p , 0.01, ***p , 0.001.

hCG significantly decreased the total number of CD11c+ cells at context, it was already demonstrated that stimulation of mono-
the fetalmaternal interface but not in the spleen (Fig. 7A, 7B). cytes with hCG augmented the production of IL-8 (38) known to
Moreover, the number of mature DCs determined by the expres- attract leukocytes (39). Moreover, hCG is a potent attractor of
sion of CD80 and MHC class II was significantly reduced in de- neutrophils, monocytes, and lymphocytes at very low doses (40).
cidua but not spleen after hCG treatment as compared with PBS In this work, we hypothesized that hCG may also be of central
treatment (Fig. 7CF). In virgin CBA/J females, hCG slightly importance in promoting tolerance by directly influencing Treg
decreased the number of total CD11c+ DCs and mature CD11c+ number and functionality. Because this may also occur indirectly
MHCII+ DCs in spleen and uterus when compared with PBS, but by interaction with, for example, DCs, we extended our study to
did not alter the levels of CD11c+CD80+ DCs in both organs these cells as well.
(Tables II, III). We tested our hypothesis in the murine system knowing that hCG
To test whether uterine DCs from hCG-treated animals favor the is commonly used in superovulation protocols for genetic engi-
generation or expansion of Tregs, we cocultured DCs obtained neering of mouse strains and efficiently binds to the endogenous
from spleen or decidua of either hCG- or PBS-treated AP females murine LH/CG receptor (41). We first took advantage of the
with CD4+ T cells and determined the number of CD4+ cells Foxp3gfp transgenic animals that allowed us to easily isolate and
expressing Foxp3. Indeed, we observed an upregulation of Foxp3 characterize Foxp3+ Tregs after hormonal treatment in an allo-
expression in CD4+ T cells when decidual DCs from hCG-treated geneic pregnancy setting. We observed that hCG significantly
AP females were present in the culture (Fig. 8A). This effect was increased the number of Tregs in the periphery, as well as locally,
not observed when DCs were of splenic origin (Fig. 8B). Alto- suggesting an accumulation of Tregs at the fetalmaternal inter-
gether, our results confirm an effect of hCG on DC phenotype face according to our observations in human pregnancy. Further-
and at least in vitro, these cells contribute to augmented Treg more, hCG application contributed to Treg suppressive capacity
frequency. by elevating the secretion of IL-10 and TGF-b. Having these
results, we wondered whether the positive effect of hCG on Treg
Discussion number and function may favor fetal survival. For this, we next
Although it is well-known that both the endocrine and the immune used a model of disturbed tolerance, in which we previously
systems substantially contribute to successful pregnancy outcome, demonstrated diminished Treg number and activity, and proved
the interplay between both systems and the major players at that Treg reconstitution can diminish the abortion rates to normal
molecular and cellular levels are still under investigation. We had levels (24, 27). hCG not only restored the Treg levels to normality,
already reported that hCG produced mainly by the trophoblast is but also completely prevented abortion. In particular, we proved
one of the factors attracting human Tregs efficiently to the fetal a significant increase of thymic Tregs after hCG application,
maternal interface contributing to their local accumulation. This suggesting that this hormone supports the expansion of naturally
local accumulation of Tregs might also be favored by other im- occurring Tregs produced in the thymus. Because more Tregs
mune cells secreting hCG at the fetalmaternal interface. In this were also found in para-aortic lymph nodes, blood, and decidua,
The Journal of Immunology 2655

pregnancy is further underlined by a study of Mansour and col-


leagues (42), who lately showed that intrauterine injection of
hCG before embryo transfer in patients undergoing IVF/
intracytoplasmic sperm injection significantly improved implan-
tation and pregnancy rates. Unfortunately, in that study, the
authors did not analyze the number and activity of Tregs (42).
However, increased pregnancy rates after IVF have been shown to
be associated with increased Treg numbers in peripheral blood
(26). Moreover, based on our human data showing hCG as a po-
tent chemoattractor for Tregs (31), the Egyptian IVF-ET Center
conducted a clinical trial investigating the effect of uterine in-
jection of hCG on endometrial Tregs. Upcoming results will prove
whether the intrauterine application of hCG around implantation
time may increase endogenous endometrial Treg levels, and
thereby favor the implantation process. This would support our
assumption that normal progressing pregnancies with normal hCG
levels ensure the generation and/or expansion of cells that are
fundamental for pregnancy maintenance (43).
To prove the generation of functionally active, pregnancy-
protective Tregs after hCG application, we adoptively trans-

Downloaded from http://www.jimmunol.org/ by guest on April 7, 2017


ferred Tregs isolated from hCG-treated AP females (having oth-
erwise dysfunctional Tregs) into AP mice. These cells had the
capacity to completely protect from fetal rejection, whereas Tregs
isolated from PBS-treated AP females did not. Our data raise the
possibility that transferred Tregs induce the generation of new
Tregs by infectious tolerance or by conversion from naive T cells,
as it has been described by others (4446). In vitro, Tregs from
hCG-treated females had a significantly increased capacity to in-
hibit responder T cell proliferation when compared with Tregs
from PBS-treated females. This confirms our in vivo data on hCG-
mediated generation of pregnancy-protective Tregs. In agreement
FIGURE 5. hCG promoted the in vivo generation of Treg that retained with this, Khil and colleagues (19) showed that application of
their Ag-specificity in vitro. (A) Both, Treg isolated from hCG- and PBS- hCG in a murine model for autoimmune diabetes prevented the
treated AP females significantly suppressed proliferation of autologous onset of the disease. In this model, hCG application resulted in an
responder T cells (Tresp). Interestingly, Treg from hCG-treated females increase of Tregs in spleen and pancreatic lymph nodes, and re-
showed a significant increased suppressive capacity when compared with duced the number of effector T cells. Moreover, hCG elevated IL-
Treg from PBS-treated females. (B) Neither Tregs from hCG- nor PBS-
10 and TGF-b expression in splenic cells, which indicates an
treated AP females were able to suppress the proliferation of allologous
responder T cells from a third-party combination (Tthird party). In addition,
augmented suppressive Treg function (19). In our model, hCG
no differences could be observed between the suppressive capacities from also provoked Tregs to secrete more IL-10 and TGF-b. Fuchs and
hCG- or PBS-treated females. Suppression assays were performed in colleagues (18, 47) demonstrated an hCG-dependent generation of
duplicates and repeated at least three times to ensure reproducibility. Data suppressor T cells, leading to inhibited mitogen-induced activation
are presented as means plus SD. Statistical analysis between two different of B cells. Previous data from our group confirmed an Ag-specific
groups was performed using the Student t test. *p , 0.05, **p , 0.01, Treg function in the AP model (27). Thus, we wondered whether
***p . 0.001, ****p . 0.0001. activation of Tregs can be mediated by hCG alone or is also de-
pendent on the presence of specific paternal Ags. We could clearly
and knowing that hCG also fosters their migration (37), we pro- show that neither Tregs obtained from PBS- nor from hCG-treated
pose that Tregs previously activated by specific paternal Ags in AP females significantly suppressed proliferation of responder
draining lymph nodes actively migrate to the fetalmaternal in- T cells from a third-party combination, confirming that there is
terface to support fetal survival. The positive effect of hCG on a need for specific Ag stimulation of Tregs in addition to their

FIGURE 6. Tregs and DCs expressed the LH/CG


receptor. Ninety-seven percent of CD4+CD25+ Tregs
and 69.4% of CD11c+ DCs isolated from virgin CBA/J
females expressed the LH/CG receptor on their cell
surface. Negative controls are included to exclude un-
specific binding of the second Ab. Data are presented as
histograms using FlowJo software.
2656 hCG STIMULATES Treg AND SUPPORTS PREGNANCY

FIGURE 7. hCG reduced the number of DCs and


retained them in an immature state. Application of
10 IU/ml hCG in AP females (n = 57/organ)
resulted in a significant reduction of total CD11c+
DCs in decidua (B), but had no effect on the number
of splenic CD11c+ DCs (A) when compared with
PBS-treated females (n = 57/organ). The number
of mature CD11c+CD80+ and CD11c+MHCII+ DCs
in decidua was significantly decreased after hCG
treatment (n = 78) as compared with PBS treat-
ment (n = 7) (D, F). By contrast, hCG had no impact
on the number of mature DCs in spleen (n = 45/

Downloaded from http://www.jimmunol.org/ by guest on April 7, 2017


treatment) (C, E). Each square represents one single
animal, and the lines show the medians. Statistical
analysis was carried out by the MannWhitney U
test. *p , 0.05, **p , 0.01.

activation by hCG. According to these data, we did not observe pregnancy hormones may also influence DC phenotype and ac-
a significant increase in the Treg number in uterine tissue of virgin tivity. In terms of an hCG-mediated effect on DCs, in vitro data
CBA/J females injected with hCG when compared with CBA/J are quite controversial (48, 53, 54) and in vivo data are not
females treated with PBS. Therefore, we assume that although available yet.
hCG has a strong impact on Treg expansion and functionality, the Because the maturation state of DCs is important for their ca-
presence of specific paternal Ags is essential to ensure complete pability to induce immunity or tolerance (35), we were interested
Treg induction during pregnancy. to study the influence of hCG on DC phenotype and function.
Next, we focused on learning whether hCG mediates Treg in- Blois and colleagues (55) showed that during pregnancy, the
duction directly or indirectly via DCs. Because both Tregs and DCs number of CD11c+ DCs in decidua strongly increases when
expressed the LH/CG receptor, both ways are possible. According compared with the peripheral blood. The majority of murine DCs
to our murine data, the presence of the LH/CG receptor has been at the fetalmaternal interface are of myeloid origin, having an
proved on human DCs (48) and human Tregs (31). Several studies immature tolerogenic phenotype, and produce high amounts of IL-
suggest an impact of tolerogenic DCs on Treg generation (49, 50). 10 (56). In this regard, the secretion of IL-10 seems to be an es-
We recently demonstrated an involvement of the pregnancy- sential mechanism for DC function at the fetalmaternal interface
protective enzyme heme oxygenase-1 for DC-mediated Treg in- as IL-10 directly inhibits effector T cells (57) and promotes Treg
duction and function during murine pregnancy (37). In addition, generation (36). Segerer and colleagues (54), as well as Wan and
it has been shown that P induces an immature phenotype in bone colleagues (53), nicely showed in vitro that hCG inhibits upreg-
marrowderived DCs (51) and estrogen reduces the production ulation of MHC class II molecules on DCs, reduces their T cell
of inflammatory cytokines by mature DCs (52), suggesting that stimulatory capacity, and induces IL-10 production. By contrast,

Table II. hCG decreases the number of total and CD11c+MHCII+ DCs in spleen

CD11c+ in Spleen CD11c+CD80+ in Spleen CD11c+MHCII+ in


Group n (%) (%) Spleen (%)

CBA/J + PBS 4 3.38 0.97 1.69


CBA/J + hCG 4 2.81 1.13 1.18
Application of 10 IU/ml hCG in virgin CBA/J females reduces the number of total CD11c+ and mature CD11c+MHCII+ DCs
in spleen as compared with PBS but did not change the levels of CD11c+CD80+ DCs. Data are presented as medians. Statistical
analysis was carried out by the MannWhitney U test. No statistical differences could be detected between hCG- and PBS-
treated females.
The Journal of Immunology 2657

Table III. hCG decreases the number of total and CD11c1 MHCII1 DCs in uterus

CD11c1 CD11c1CD801 in CD11c1MHCII1


Group n in Uterus (%) Uterus (%) in Uterus (%)

CBA/J 1 PBS 4 1.07 0.09 0.62


CBA/J 1 hCG 4 0.89 0.09 0.38
Application of 10 IU/ml hCG in virgin CBA/J females reduces the number of total CD11c1 and mature CD11c1MHCII1
DCs in uterus as compared to PBS but did not change the levels of CD11c1CD801 DCs. Data are presented as medians.
Statistical analysis was carried out by the MannWhitney U test. No statistical differences could be detected between hCG- and
PBS-treated females.

Yoshimura and colleagues (48) revealed that hCG induces the maternal T cell responses toward the fetus and supporting gen-
maturation of myeloid and lymphoid DCs in conjunction with eration of Tregs. Coculture of decidual CD11c+ cells and CD4+
increases in the expression of adhesion/costimulatory molecules, T cells further reveal that DCs from hCG-treated AP females in-
their stimulatory activities in MLR, and cytokine secretion. In this deed provoked the generation or expansion of Tregs by upregu-
article, we provide in vivo evidence for a strong impact of hCG lating Foxp3 expression in CD4+ T cells. According to our
that reduces the number of total and mature DCs directly at the observation that hCG seems to have no influence on the phenotype
fetalmaternal interface, but not in the periphery. Regarding the of splenic DCs, we could not detect Foxp3 upregulation in CD4+
impact of a reduction of total DCs for pregnancy outcome, Plaks T cells in the presence of splenic DCs from hCG-treated AP
and colleagues (58) nicely showed that a depletion of uterine DCs females. Thus, we suppose that hCG directly modulates DCs at the

Downloaded from http://www.jimmunol.org/ by guest on April 7, 2017


by applying diphtheria toxin to CD11cDTR mice resulted in an fetalmaternal interface, and this favors a local Treg expansion.
impaired decidualization and implantation of embryos. However, Our work provides strong evidence that hCG, the most important
DC depletion in this model took place either before pregnancy or pregnancy hormone, secreted by the trophoblast, contributes to
at very early gestation days. In this study, we can only make fetal tolerance not only by attracting Tregs to the fetalmaternal
conclusions about the number of total CD11c+ DCs on day 10 of interface, but also by augmenting the number and suppressive
pregnancy after hCG or PBS treatment. Moreover, although hCG capacity of Tregs. hCG has a direct effect on Tregs but also acts
reduced the number of total CD11c+ DCs, there are still plenty of indirectly by maintaining DCs in an immature, tolerogenic state.
DCs present at the fetalmaternal interface that can do their job. Our observations offer a possible explanation for improved
Furthermore, we could prove that hCG especially reduced the pregnancy outcomes of IVF patients treated with hCG after em-
number of mature CD11c+ DCs within the whole CD11c+ DC bryo transfer. This contributes to the basic knowledge of Treg
population. Thus, we would assume that hCG favors the presence generation and functionality during pregnancy and may help to
of a population consisting of immature tolerogenic DCs that have establish therapies to prevent pregnancy failures.
been shown to promote fetal survival by reducing alloreactive
Disclosures
The authors have no financial conflicts of interest.

References
1. Chambers, S. P., and A. G. Clarke. 1979. Measurement of thymus weight,
lumbar node weight and progesterone levels in syngeneically pregnant, alloge-
neically pregnant, and pseudopregnant mice. J. Reprod. Fertil. 55: 309315.
2. Stewart, D. R., J. W. Overstreet, S. T. Nakajima, and B. L. Lasley. 1993. En-
hanced ovarian steroid secretion before implantation in early human pregnancy.
J. Clin. Endocrinol. Metab. 76: 14701476.
3. Policastro, P., C. E. Ovitt, M. Hoshina, H. Fukuoka, M. R. Boothby, and
I. Boime. 1983. The beta subunit of human chorionic gonadotropin is encoded by
multiple genes. J. Biol. Chem. 258: 1149211499.
4. Crawford, R. J., G. W. Tregear, and H. D. Niall. 1986. The nucleotide sequences
of baboon chorionic gonadotropin beta-subunit genes have diverged from the
human. Gene 46: 161169.
5. Tepper, M. A., and J. L. Roberts. 1984. Evidence for only one beta-luteinizing
hormone and no beta-chorionic gonadotropin gene in the rat. Endocrinology 115:
385391.
6. Salesse, R., N. Genty, and J. Garnier. 1983. Lutropin is processed much more
rapidly than human choriogonadotropin by porcine Leydig cells in primary
culture. Biol. Cell 49: 187190.
7. Lopata, A., and D. L. Hay. 1989. The potential of early human embryos to form
blastocysts, hatch from their zona and secrete HCG in culture. Hum. Reprod. 4
(8Suppl.): 8794.
8. Hoshina, M., M. Boothby, R. Hussa, R. Pattillo, H. M. Camel, and I. Boime.
FIGURE 8. hCG provoked the in vivo generation of tolerogenic DCs 1985. Linkage of human chorionic gonadotrophin and placental lactogen bio-
that induced Tregs in vitro. CD11c+ DCs isolated from the decidua of synthesis to trophoblast differentiation and tumorigenesis. Placenta 6: 163172.
hCG-treated AP females significantly induced the generation/expansion of 9. Braunstein, G. D., J. Rasor, H. Danzer, D. Adler, and M. E. Wade. 1976. Serum
CD4+Foxp3+ cells within the CD4+ population when compared with human chorionic gonadotropin levels throughout normal pregnancy. Am. J.
Obstet. Gynecol. 126: 678681.
CD11c+ DCs isolated from PBS-treated females (B). By contrast, CD11c+ 10. Fluhr, H., D. Bischof-Islami, S. Krenzer, P. Licht, P. Bischof, and M. Zygmunt.
DCs isolated from the spleen of hCG-treated females had no influence on 2008. Human chorionic gonadotropin stimulates matrix metalloproteinases-2
Treg generation/expansion when compared with CD11c+ DCs isolated and -9 in cytotrophoblastic cells and decreases tissue inhibitor of
from PBS-treated AP females (A). Conversion assays were performed in metalloproteinases-1, -2, and -3 in decidualized endometrial stromal cells. Fertil.
Steril. 90(4Suppl.): 13901395.
duplicates and repeated at least three times to ensure reproducibility. Data
11. Kornyei, J. L., Z. M. Lei, and C. V. Rao. 1993. Human myometrial smooth
are presented as means + SD. Statistical analysis between two different muscle cells are novel targets of direct regulation by human chorionic gonado-
groups was performed using the Student t test. *p , 0.05. tropin. Biol. Reprod. 49: 11491157.
2658 hCG STIMULATES Treg AND SUPPORTS PREGNANCY

12. Kayisli, U. A., B. Selam, O. Guzeloglu-Kayisli, R. Demir, and A. Arici. 2003. 37. Schumacher, A., P. O. Wafula, A. Teles, T. El-Mousleh, N. Linzke,
Human chorionic gonadotropin contributes to maternal immunotolerance and M. L. Zenclussen, S. Langwisch, K. Heinze, I. Wollenberg, P. A. Casalis, et al.
endometrial apoptosis by regulating Fas-Fas ligand system. J. Immunol. 171: 2012. Blockage of heme oxygenase-1 abrogates the protective effect of regula-
23052313. tory T cells on murine pregnancy and promotes the maturation of dendritic cells.
13. Berndt, S., S. Perrier dHauterive, S. Blacher, C. Pequeux, S. Lorquet, C. Munaut, PLoS ONE 7: e42301.
M. Applanat, M. A. Herve, N. Lamande, P. Corvol, et al. 2006. Angiogenic ac- 38. Kosaka, K., H. Fujiwara, K. Tatsumi, S. Yoshioka, Y. Sato, H. Egawa,
tivity of human chorionic gonadotropin through LH receptor activation on en- T. Higuchi, T. Nakayama, M. Ueda, M. Maeda, and S. Fujii. 2002. Human
dothelial and epithelial cells of the endometrium. FASEB J. 20: 26302632. chorionic gonadotropin (HCG) activates monocytes to produce interleukin-8 via
14. Zygmunt, M., F. Herr, S. Keller-Schoenwetter, K. Kunzi-Rapp, K. Munstedt, a different pathway from luteinizing hormone/HCG receptor system. J. Clin.
C. V. Rao, U. Lang, and K. T. Preissner. 2002. Characterization of human Endocrinol. Metab. 87: 51995208.
chorionic gonadotropin as a novel angiogenic factor. J. Clin. Endocrinol. Metab. 39. Ushigoe, K., M. Irahara, M. Fukumochi, M. Kamada, and T. Aono. 2000. Pro-
87: 52905296. duction and regulation of cytokine-induced neutrophil chemoattractant in rat
15. Fujimoto, A., Y. Osuga, T. Fujiwara, T. Yano, O. Tsutsumi, M. Momoeda, ovulation. Biol. Reprod. 63: 121126.
K. Kugu, K. Koga, Y. Morita, O. Wada, and Y. Taketani. 2002. Human chorionic 40. Reinisch, N., B. A. Sitte, C. M. Kahler, and C. J. Wiedermann. 1994. Human
gonadotropin combined with progesterone for luteal support improves pregnancy chorionic gonadotrophin: a chemoattractant for human blood monocytes, neu-
rate in patients with low late-midluteal estradiol levels in IVF cycles. J. Assist. trophils and lymphocytes. J. Endocrinol. 142: 167170.
Reprod. Genet. 19: 550554. 41. Luo, C., J. Zuniga, E. Edison, S. Palla, W. Dong, and J. Parker-Thornburg. 2011.
16. Kaye, M. D., and W. R. Jones. 1971. Effect of human chorionic gonadotropin on
Superovulation strategies for 6 commonly used mouse strains. J. Am. Assoc. Lab.
in vitro lymphocyte transformation. Am. J. Obstet. Gynecol. 109: 10291031.
Anim. Sci. 50: 471478.
17. Hammarstrom, L., T. Fuchs, and C. I. Smith. 1979. The immunodepressive effect
42. Mansour, R., N. Tawab, O. Kamal, Y. El-Faissal, A. Serour, M. Aboulghar, and
of human glucoproteins and their possible role in the nonrejection process during
G. Serour. 2011. Intrauterine injection of human chorionic gonadotropin before
pregnancy. Acta Obstet. Gynecol. Scand. 58: 417422.
embryo transfer significantly improves the implantation and pregnancy rates in
18. Fuchs, T., L. Hammarstrom, C. I. Smith, and J. Brundin. 1980. In vitro induction
of murine suppressor T-cells by human chorionic gonadotropin. Acta Obstet. in vitro fertilization/intracytoplasmic sperm injection: a prospective randomized
Gynecol. Scand. 59: 355359. study. Fertil. Steril. 96: 13701374, e1.
19. Khil, L. Y., H. S. Jun, H. Kwon, J. K. Yoo, S. Kim, A. L. Notkins, and 43. Samstein, R. M., S. Z. Josefowicz, A. Arvey, P. M. Treuting, and A. Y. Rudensky.
J. W. Yoon. 2007. Human chorionic gonadotropin is an immune modulator and 2012. Extrathymic generation of regulatory T cells in placental mammals mit-

Downloaded from http://www.jimmunol.org/ by guest on April 7, 2017


can prevent autoimmune diabetes in NOD mice. Diabetologia 50: 21472155. igates maternal-fetal conflict. Cell 150: 2938.
20. Aluvihare, V. R., M. Kallikourdis, and A. G. Betz. 2004. Regulatory T cells 44. Chen, W., W. Jin, N. Hardegen, K. J. Lei, L. Li, N. Marinos, G. McGrady,
mediate maternal tolerance to the fetus. Nat. Immunol. 5: 266271. and S. M. Wahl. 2003. Conversion of peripheral CD4+CD25- naive T cells to
21. Somerset, D. A., Y. Zheng, M. D. Kilby, D. M. Sansom, and M. T. Drayson. CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor
2004. Normal human pregnancy is associated with an elevation in the immune Foxp3. J. Exp. Med. 198: 18751886.
suppressive CD25+ CD4+ regulatory T-cell subset. Immunology 112: 3843. 45. Grohmann, U., C. Orabona, F. Fallarino, C. Vacca, F. Calcinaro, A. Falorni,
22. Thuere, C., M. L. Zenclussen, A. Schumacher, S. Langwisch, U. Schulte-Wrede, P. Candeloro, M. L. Belladonna, R. Bianchi, M. C. Fioretti, and P. Puccetti. 2002.
A. Teles, S. Paeschke, H. D. Volk, and A. C. Zenclussen. 2007. Kinetics of reg- CTLA-4-Ig regulates tryptophan catabolism in vivo. Nat. Immunol. 3: 1097
ulatory T cells during murine pregnancy. Am. J. Reprod. Immunol. 58: 514523. 1101.
23. Sasaki, Y., M. Sakai, S. Miyazaki, S. Higuma, A. Shiozaki, and S. Saito. 2004. 46. Munn, D. H., M. D. Sharma, J. R. Lee, K. G. Jhaver, T. S. Johnson, D. B. Keskin,
Decidual and peripheral blood CD4+CD25+ regulatory T cells in early preg- B. Marshall, P. Chandler, S. J. Antonia, R. Burgess, et al. 2002. Potential reg-
nancy subjects and spontaneous abortion cases. Mol. Hum. Reprod. 10: 347353. ulatory function of human dendritic cells expressing indoleamine 2,3-dioxy-
24. Zenclussen, A. C., K. Gerlof, M. L. Zenclussen, A. Sollwedel, A. Z. Bertoja, genase. Science 297: 18671870.
T. Ritter, K. Kotsch, J. Leber, and H. D. Volk. 2005. Abnormal T-cell reactivity 47. Fuchs, T., L. Hammarstrom, C. I. Smith, and J. Brundin. 1981. In vitro induction
against paternal antigens in spontaneous abortion: adoptive transfer of of human suppressor T cells by a chorionic gonadotropin preparation. J. Reprod.
pregnancy-induced CD4+CD25+ T regulatory cells prevents fetal rejection in Immunol. 3: 7584.
a murine abortion model. Am. J. Pathol. 166: 811822. 48. Yoshimura, T., M. Inaba, K. Sugiura, T. Nakajima, T. Ito, K. Nakamura,
25. Jasper, M. J., K. P. Tremellen, and S. A. Robertson. 2006. Primary unexplained H. Kanzaki, and S. Ikehara. 2003. Analyses of dendritic cell subsets in preg-
infertility is associated with reduced expression of the T-regulatory cell tran- nancy. Am. J. Reprod. Immunol. 50: 137145.
scription factor Foxp3 in endometrial tissue. Mol. Hum. Reprod. 12: 301308. 49. Chen, W., X. Liang, A. J. Peterson, D. H. Munn, and B. R. Blazar. 2008. The
26. Zhou, J., Z. Wang, X. Zhao, J. Wang, H. Sun, and Y. Hu. 2012. An increase of indoleamine 2,3-dioxygenase pathway is essential for human plasmacytoid
Treg cells in the peripheral blood is associated with a better in vitro fertilization dendritic cell-induced adaptive T regulatory cell generation. J. Immunol. 181:
treatment outcome. Am. J. Reprod. Immunol. 68: 100106. 53965404.
27. Schumacher, A., P. O. Wafula, A. Z. Bertoja, A. Sollwedel, C. Thuere, 50. Sharma, M. D., B. Baban, P. Chandler, D. Y. Hou, N. Singh, H. Yagita,
I. Wollenberg, H. Yagita, H. D. Volk, and A. C. Zenclussen. 2007. Mechanisms M. Azuma, B. R. Blazar, A. L. Mellor, and D. H. Munn. 2007. Plasmacytoid
of action of regulatory T cells specific for paternal antigens during pregnancy. dendritic cells from mouse tumor-draining lymph nodes directly activate mature
Obstet. Gynecol. 110: 11371145. Tregs via indoleamine 2,3-dioxygenase. J. Clin. Invest. 117: 25702582.
28. Yin, Y., X. Han, Q. Shi, Y. Zhao, and Y. He. 2012. Adoptive transfer of CD4 51. Liang, J., L. Sun, Q. Wang, and Y. Hou. 2006. Progesterone regulates mouse
+CD25+ regulatory T cells for prevention and treatment of spontaneous abor- dendritic cells differentiation and maturation. Int. Immunopharmacol. 6: 830
tion. Eur. J. Obstet. Gynecol. Reprod. Biol. 161: 177181. 838.
29. Zenclussen, A. C., K. Gerlof, M. L. Zenclussen, S. Ritschel, A. Zambon Bertoja, 52. Liu, H. Y., A. C. Buenafe, A. Matejuk, A. Ito, A. Zamora, J. Dwyer,
S. Fest, S. Hontsu, S. Ueha, K. Matsushima, J. Leber, and H. D. Volk. 2006.
A. A. Vandenbark, and H. Offner. 2002. Estrogen inhibition of EAE involves
Regulatory T cells induce a privileged tolerant microenvironment at the fetal-
effects on dendritic cell function. J. Neurosci. Res. 70: 238248.
maternal interface. Eur. J. Immunol. 36: 8294.
53. Wan, H., M. A. Versnel, L. M. Leijten, C. G. van Helden-Meeuwsen, D. Fekkes,
30. Wafula, P. O., A. Teles, A. Schumacher, K. Pohl, H. Yagita, H. D. Volk, and
P. J. Leenen, N. A. Khan, R. Benner, and R. C. Kiekens. 2008. Chorionic go-
A. C. Zenclussen. 2009. PD-1 but not CTLA-4 blockage abrogates the protective
nadotropin induces dendritic cells to express a tolerogenic phenotype. J. Leukoc.
effect of regulatory T cells in a pregnancy murine model. Am. J. Reprod.
Immunol. 62: 283292. Biol. 83: 894901.
31. Schumacher, A., N. Brachwitz, S. Sohr, K. Engeland, S. Langwisch, 54. Segerer, S. E., N. Muller, J. van den Brandt, M. Kapp, J. Dietl, H. M. Reichardt,
M. Dolaptchieva, T. Alexander, A. Taran, S. F. Malfertheiner, S. D. Costa, et al. L. Rieger, and U. Kammerer. 2009. Impact of female sex hormones on the
2009. Human chorionic gonadotropin attracts regulatory T cells into the fetal- maturation and function of human dendritic cells. Am. J. Reprod. Immunol. 62:
maternal interface during early human pregnancy. J. Immunol. 182: 54885497. 165173.
32. Fontenot, J. D., J. P. Rasmussen, L. M. Williams, J. L. Dooley, A. G. Farr, and 55. Blois, S. M., C. D. Alba Soto, M. Tometten, B. F. Klapp, R. A. Margni, and
A. Y. Rudensky. 2005. Regulatory T cell lineage specification by the forkhead P. C. Arck. 2004. Lineage, maturity, and phenotype of uterine murine dendritic
transcription factor foxp3. Immunity 22: 329341. cells throughout gestation indicate a protective role in maintaining pregnancy.
33. Groux, H., A. OGarra, M. Bigler, M. Rouleau, S. Antonenko, J. E. de Vries, and Biol. Reprod. 70: 10181023.
M. G. Roncarolo. 1997. A CD4+ T-cell subset inhibits antigen-specific T-cell 56. Shortman, K., and Y. J. Liu. 2002. Mouse and human dendritic cell subtypes.
responses and prevents colitis. Nature 389: 737742. Nat. Rev. Immunol. 2: 151161.
34. Weiner, H. L. 2001. Induction and mechanism of action of transforming growth 57. Groux, H., M. Bigler, J. E. de Vries, and M. G. Roncarolo. 1996. Interleukin-10
factor-beta-secreting Th3 regulatory cells. Immunol. Rev. 182: 207214. induces a long-term antigen-specific anergic state in human CD4+ T cells. J.
35. Lutz, M. B., and G. Schuler. 2002. Immature, semi-mature and fully mature dendritic Exp. Med. 184: 1929.
cells: which signals induce tolerance or immunity? Trends Immunol. 23: 445449. 58. Plaks, V., T. Birnberg, T. Berkutzki, S. Sela, A. BenYashar, V. Kalchenko,
36. Wakkach, A., N. Fournier, V. Brun, J. P. Breittmayer, F. Cottrez, and H. Groux. G. Mor, E. Keshet, N. Dekel, M. Neeman, and S. Jung. 2008. Uterine DCs are
2003. Characterization of dendritic cells that induce tolerance and T regulatory 1 crucial for decidua formation during embryo implantation in mice. J. Clin. In-
cell differentiation in vivo. Immunity 18: 605617. vest. 118: 39543965.

You might also like