Anda di halaman 1dari 13

Biomedicine & Pharmacotherapy 88 (2017) 6173

Available online at

ScienceDirect
www.sciencedirect.com

Immune regulatory network in successful pregnancy and reproductive


failures
Mahnaz Ghaebia,b,c , Mohammad Nourid, Aliyeh Ghasemzadehe, Laya Farzadie ,
Farhad Jadidi-Niaragha,b,f , Majid Ahmadia,b , Mehdi Yousea,b,c,*
a
Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
b
Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
c
Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
d
Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
e
Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
f
Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran

A R T I C L E I N F O A B S T R A C T

Article history:
Received 27 November 2016 Maternal immune system must tolerate semiallogenic fetus to establish and maintain a successful
Received in revised form 27 December 2016 pregnancy. Despite the existence of several strategies of trophoblast to avoid recognition by maternal
Accepted 2 January 2017 leukocytes, maternal immune system may react against paternal alloantigenes. Leukocytes are important
components in decidua. Not only T helper (Th)1/Th2 balance, but also regulatory T (Treg) cells play an
Keywords: important role in pregnancy. Although the frequency of Tregs is elevated during normal pregnancies,
Maternal immune system their frequency and function are reduced in reproductive defects such as recurrent miscarriage and
Treg preeclampsia. Tregs are not the sole population of suppressive cells in the decidua. It has recently been
gdT cell shown that regulatory B10 (Breg) cells participate in pregnancy through secretion of IL-10 cytokine.
Breg
Myeloid derived suppressor cells (MDSCs) are immature developing precursors of innate myeloid cells
NKT
MDSCs that are increased in pregnant women, implying their possible function in pregnancy. Natural killer T
Pregnancy (NKT) cells are also detected in mouse and human decidua. They can also affect the fetomaternal
tolerance. In this review, we will discuss on the role of different immune regulatory cells including Treg,
gd T cell, Breg, MDSC, and NKT cells in pregnancy outcome.
2017 Elsevier Masson SAS. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Suppressor and regulatory T cells in pregnancy . . . . . . . . . . . . ... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.1. Treg changes during normal and disturbed pregnancies .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.2. Interaction between Tregs and DCs . . . . . . . . . . . . . . . . ... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.3. Interaction between Treg and NK cells . . . . . . . . . . . . . ... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.4. Interaction between Tregs and TH17 . . . . . . . . . . . . . . . ... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.5. Functional mechanisms of Tregs in normal pregnancy ... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3. Regulatory gdT cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
4. Bregs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
5. NKT cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
6. MDSCs in pregnancy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
7. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8

* Corresponding author at: Drug Applied Research Center, Tabriz University of


Medical Sciences, Tabriz, Iran.
E-mail address: Youseme@tbzmed.ac.ir (M. Youse).

http://dx.doi.org/10.1016/j.biopha.2017.01.016
0753-3322/ 2017 Elsevier Masson SAS. All rights reserved.
62 M. Ghaebi et al. / Biomedicine & Pharmacotherapy 88 (2017) 6173

1. Introduction specic cells show proliferation, IFNg secretion and ability to lyse
target cells following recognition of processed male antigens [6].
Since the fetus is a semiallograft, a successful pregnancy needs Involvement of suppressor cells in the prevention of rejection of
mechanisms to prevent allograft rejection [1]. Polymorphic genes the fetus was initially suggested based on mixed lymphocyte
inherited from the father generate alloantigenes which provoke reactions using splenocytes from either pregnant or virgin females
the maternal immune responses leading to fetal rejection. [25]. The predominance of Th2 type immunity in Th1/Th2 balance
Adaptation of immune system is crucial to prevent rejection of has been proposed as a possible strategy for survival of the fetus in
the semi-allogeneic conceptus in pregnancy [2]. Dysregulation of the uterus [26]. Increased Th2 polarization in maternal peripheral
immune responses may lead to reproductive failures such as blood mononuclear cells (PBMCs) and decreased IFNg secretion
recurrent pregnancy loss (RPL), implantation failure, preterm birth, are associated with the normal pregnancy [27]. Women with RPL
intrauterine fetal growth restriction and preeclampsia [3]. The show increased Th1 to Th2 cell ratio and increased IFNg production
most common pregnancy problems are miscarriage and pre- compared with normal healthy group [28]. In addition, Th17 cells
eclampsia [4,5]. Miscarriage occurs in about 15% of the cases, and are positively associated with idiopathic RPL [29]. The necessity of
recurrent miscarriages in 25% [4]. Although the exact cause of Th2 cytokines in a normal pregnancy is unknown, since studies on
about 50% of recurrent miscarriages remains elusive, however genetically-decient mice which lack the ability to secret Th2
immunological rejection may account for most of these unex- cytokines did not always lead to miscarriage [15,30]. Therefore,
plained cases of pregnancy loss [68]. Several immunological other mechanisms such as the function of regulatory cells regulate
mechanisms have been proposed for tolerance to the semi- alloreactive Th1 cells [15]. Increased number of Treg in normal
allogeneic fetus. It is suggested that miscarriage occurs in mice and pregnancy and impaired function and frequency of these cells
humans when these mechanisms become dysregulated [9]. Since during pregnancy failures demonstrate importance of regulatory
maternal alloreactive lymphocytes are not systemically depleted, cells in pregnancy [15]. Recently the other immune cell popula-
the local mechanisms may act to avoid maternal immune attack tions with regulatory effects have been reported to exist in
[10]. Fetal trophoblast with its special features including low fetomaternal interface and play critical roles in pregnancy
tryptophan levels, the expression of HLA-G/C and so lack of maintenance.
activation of natural killer (NK) cells, absence of classical HLA Class
I and Class II trophoblast expression, high progesterone levels and 2. Suppressor and regulatory T cells in pregnancy
antiidiotype network modulation plays an important role in
immune tolerance to the fetus [1115]. In addition, it has been Based on phenotype, cytokine secretion and tissue origin, three
demonstrated that maternal T cell recognition of fetal antigens subsets of CD4+ regulatory T (Treg) cells with different suppressive
occurs in an indirect manner that means fetal allograft is ignored functions have been distinguished [30]. These are type 1 regulatory
by directly alloreactive T cells that cause acute transplant rejection T cells (Tr1), Th3 cells, and CD4+CD25+ regulatory T-cells (Treg
[16]. Despite the presence of various mechanisms in immune cells) [31]. CD8+/Foxp3+ T cells with regulatory features have also
evasion, maternal adaptive immune system can recognize paternal been described, however their function, ontogeny and regulation
alloantigens [2], as a study published in 2003 has reported that are still unknown [32]. Tr1 cells are CD4+CD25+Foxp3_ T cells
pregnancy induces cytotoxic T cells specic for minor histocom- induced by IL-10 which also exert their suppressive activity by
patibility antigens [17]. Fetal specic adaptive immune responses secretion of IL-10 [33]. They were at rst characterized as
develop as a consequence of normal human pregnancy and unlike suppressors of antigen-specic immune responses in mouse
the murine models, T cells specic for fetal alloantigens are not models of inammatory bowel disease [34]. Th3 cells were
deleted during human pregnancy [6]. So, immunoregulatory initially discovered in mice as inducers of oral tolerance that inhibit
mechanisms are requested to suppress activated fetal-specic T immunity by synthesis of transforming growth factor-b (TGF-b),
lymphocytes. Maternal recognition of embryo-derived paternal IL-10 and IL-4 [35]. Among these three cell subsets,
antigens has been demonstrated to occur in several ways. CD4+CD25+Foxp3+ Treg cells obviously have essential role in a
Trophoblast tissue is the main contact region between mother reproduction. Tregs are described as a distinct cell subset on the
and fetus, but there is a wide micro-chimerism between them basis of surface phenotype and functional properties [36]. They
[18,19]. It has been found that fetal cells are present in mothers constitute 510% of CD4+ T-cells in rodents and 13% of CD4+ T-
blood throughout the pregnancy and for a long time further. cells in humans [37]. Although Tr1 and Th3 cells are present in the
Moreover, maternal cells have been detected in the progeny long uterus [38], the precise role of these cells in pregnancy is unknown
after delivery and remain at adulthood. Trophoblast is clearly as White et al. showed that allogenic pregnancy is not compro-
penetrable, not impenetrable physical barrier through which the mised in IL-10 null mutant mice [39]. On the other hand, Robertson
exchange of cells between mother and fetus takes place [2]. et al. has reported that IL-10 null mutant mice are susceptible to
Interestingly, microvesicles containing fetal antigens are shed from inammation-induced fetal loss [40]. It has been demonstrated
human placenta [20]. It has been demonstrated that leukocytes that HLA-G expressed on EVT cell which modulates alloreactive T
which are important constituents of decidua and myometrium cells induces generation of IL-10-producing Tr1 cells, in an IL10-
affect multiple events during pregnancy, including implantation, dependent pathway [41,42]. Membrane-bound HLA-G and its
maternal tolerance to the semiallogeneic fetus, defense against inhibitory receptor Ig-like transcript (ILT)-4 are upregulated in
infections, and the induction of labor [21]. Leukocytes comprise chronic antigen stimulation in the presence of IL-10, suggesting a
around 3040% of the total endometrial cells in early to positive feedback loop in maintaining maternal tolerance by Tr1
midpregnancy uteri [22]. Leukocytes have been found even in and HLA-G [42]. Recently, DC-10 cells as a subset of human DCs
the proliferative and early secretory phases of menstrual cycle in which produce IL-10 has been characterized in the peripheral
nonpregnant endometria [23]. To elucidate the maternal aware- blood. DC-10 cells express surface HLA-G, ILT2, ILT3, ILT4 and
ness of paternal-derived alloantigens, it has been proposed that secrete IL10 at high levels. In vitro experiments have shown that
antiallogenic T cells are anergized during pregnancy [24]. But the DC-10 cells induce IL10-producing Tr1 cells through IL10-depen-
maternal immune system maintains the ability to respond to dant ILT4/HLA-G pathway [43]. It has been shown that a higher
paternal alloantigens during pregnancy [2]. Fetal-specic cells frequency of DC-10 is present in human decidua than in peripheral
demonstrated an effector memory phenotype and were broadly blood of pregnant women, suggesting the possible role of DC-10 in
functional in a study conducted by Lissauer and coworkers. Fetal conjunction with inducing IL10-producing Tr1 cells in
M. Ghaebi et al. / Biomedicine & Pharmacotherapy 88 (2017) 6173 63

fetomaternal tolerance [44]. The necessity of Th3 cells in been recognized that IDO generates 3-hydroxyanthranillic acid,
pregnancy needs more investigations, because TGFb null mutant which drives the generation of Treg cells [62,69].
mice do not remain alive by the reproductive age and suffer from
non-immune-related reproductive disorders [45]. 2.3. Interaction between Treg and NK cells

2.1. Treg changes during normal and disturbed pregnancies There is increasing evidence that the interaction between NK
and Treg cells could be helpful during pregnancy. Needing to an
It is observed that during pregnancy Treg cells in decidua are immunosuppressive environment for a successful process of
increased [4648], and have a stable suppressive phenotype implantation of the embryo to decidua and tolerance of maternal
[48,49]. Women suffering from repeated miscarriage (RM) were immune system to the fetus may be the reason for this observation
shown to have a reduced frequency [50,51], and function of Treg [70]. Interestingly, 70% of all human decidual lymphocytes are NK
cells [51]. Sasaki et al. [46] observed that the population of cells, termed as uterine or decidual NK (dNK) cells [71]. dNK cells
CD4+CD25bright cells in decidua of normal early pregnancy was are CD56brightCD16CD3 cells, which express killer cell
signicantly higher than peripheral blood and this was not seen in immunoglobulin-like receptors (KIR) and show reduced killing
the patients with spontaneous abortion. Several studies have capacity despite the presence of cytolytic granules [72]. It has been
reported that in the pre-eclamptic patients the frequency of shown that dNK cells have potential to promote Treg cell
CD4+CD25high T cells is signicantly reduced in both the proliferation through interacting with CD14+ cells in decidua.
peripheral blood and decidua compared with normal pregnant The crosstalk between dNK cells and decidual myelomonocytic
women [52,53]. Reduced expression of Foxp3 mRNA in endome- CD14+ cells have been shown in some experiments, which results
trium has also been reported in primary unexplained infertility in the production of IFN-g that induces IDO expression in dCD14+
[54], indicating that defect in T cell differentiation into Treg in cells. The dCD14+ cells could induce Treg cell generation via the
uterus or insufcient migration of Tregs before conception may effects of IDO and TGFb [73]. TGFb is an important cytokine for the
also impair the ability to become pregnant [36]. The massive differentiation of extrathymic Treg cells. Interestingly, dNK cells
increase in circulating Treg cells during early pregnancy, reaching produce TGFb at feto-maternal interface, and this TGFb-producing
highest level in 2nd trimester when trophoblast invasion to cell population decreases in miscarriage. These ndings lead to this
decidua is maximal, suggests that these cells may regulate immune conclusion that dNK cells may do some roles in differentiation of
responses against fetoplacental graft in the uterus [55]. iTregs in decidua [74,75]. Both in vitro and in vivo ndings show
CD4+CD25+ Foxp3+ Treg cells may be benecial as a marker to that cytotoxic activity, cytokine production and proliferation of NK
assess miscarriage risk in pregnant women. Winger and Reed cells can be regulated by Treg cells [76,77]. TGF-b has also been
(2011) identied that Treg values of 1.0% or more seem to dene described as a factor capable of converting NK cells into dNK cells
groups that may require no therapeutic procedure, while values [78] with high proliferation rates in vitro.
below 0.7% may identify patients where therapeutic intervention
may salvage threatened pregnancies [56]. 2.4. Interaction between Tregs and TH17

2.2. Interaction between Tregs and DCs Pro-inammatory IL17-producing Th17 cells are a subset of Th
cells which are implicated in autoimmune as well as inammatory
Activation and expansion of Treg cells need interaction with a diseases. There is an interest on the relationship between Th17 and
specic type of DCs termed tolerogenic DCs [57]. Four subsets of Treg cells [79]. It has been demonstrated that there is some degree
antigen presenting cells have been identied in human decidua of plasticity between Tregs and Th17 cells, meaning that different
[58]. Heikkinen et al. have characterized decidual CD14+ macro- milieus can induce distinct cell subsets [80]. TGFb1 in the absence
phages with features resembling the phenotype of immature DCs of IL-6 simultaneously drives synthesis of both Foxp3 and the Th17
[59]. Mature CD83+ DCs, CD14+ DC-SIGN+ cells and small cell master switch, retinoic acid-related orphan receptor gt
population of immature myeloid DCs are other subtypes present (RORgt) [81]. Foxp3 then directly interacts with RORgt to repress
in early human decidua [60,61]. The IL-10 producing CD14+ Th17 cell differentiation [82]. However, in the presence of IL-6,
decidual cells [59] and immature DCs [60] have ability to induce Foxp3 expression is down-regulated, allowing RORgt to induce IL-
differentiation of Treg cells in the decidua. Tolerogenic DCs (tDC) 17 production and the conversion of naive T cells to Th17 cells [83].
are characterized with their immature or semi-mature phenotype, Treg cells in the absence of adequate TGFb1 can convert
lack of expression of the Th1-inducing cytokine IL-12 and the themselves into Th17 cells [84,85]. The ne balance between Treg
altered expression of co-stimulatory molecules CD80 and CD86 and Th17 cells is associated with maternal tolerance to the fetus
[57]. They also express surface CD8 molecule and indoleamine 2,3- [86,87]. Any disturbances in this balance such as decrease in Treg
dioxygenase (IDO) [62]. Even mature DCs can induce Treg cell and/or increase in Th17 in humans can lead to gestational
proliferation if differentiated appropriately [63]. Plasmacytoid DCs problems, such as preterm birth (PTB) [88], recurrent pregnancy
are able to produce IDO, activate resting Tregs, participate in loss (RPL) [51] or pre-eclampsia (PE) [89]. It is suggested that Treg
converting CD4+CD25 T-cells into CD4+CD25+Foxp3+ Tregs and cells miss their suppressive activity in inammatory conditions
maintaining Treg suppressive activity both in mice [64] and in [90]. Tr1 cells might be crucial in controlling tissue inammation
humans [65]. Expression of IDO may be induced through ligation of through inhibiting Th17 cells in vivo in an IL-10-dependent manner
the costimulatory molecule B7 (CD80/CD86) on DCs by CTLA-4, a [91]. IL-27 has been reported to have immunoregulatory effects by
molecule expressed on Tregs, thereby can also hinder DCs capacity rendering both Foxp3+ and Foxp3 T CD4+ cells to produce IL-10,
to activate effector T cell functions [60,66]. The cytokines in the thus promotes the differentiation of Tr1 cells and prevents Th17
milieu are a pivotal factor in controlling DCs phenotype and then development [92]. Expression of IL-27 has been observed in feto-
signals received by T cells. In the presence of TGFb, IL-10, maternal interface both in humans and mice [93,94].
granulocyte-macrophage colony-stimulating factor (GM-CSF) As mentioned above, TGFb is involved in induction and
and IL-4, differentiated DCs have phenotypic and functional maintenance of Treg in uterus and may be replaced for IL-2 in
features of immature DCs and induce CD4+CD25+ Treg cells this tissue. The other cell sources for TGFb in uterus are epithelial
[67]. Later studies showed direct signaling between Treg cells and cells and maternal lymphomyeloid cells including Th3-type
DCs that gives DCs tolerogenic properties [68]. More recently it has suppressor cells [95].
64 M. Ghaebi et al. / Biomedicine & Pharmacotherapy 88 (2017) 6173

2.5. Functional mechanisms of Tregs in normal pregnancy producing IDO, since CTLA-4Ig was not observed to elevate the IDO
mRNA expression in peripheral blood monocytes whereas
Tr1 and Th3 suppressor T cells exert their suppressive effects via stimulation with IFNg clearly up-regulated the IDO mRNA levels
release of immunosuppressive cytokines, such as IL-10 and TGFb [58]. In addition, placental cells express CTLA-4, conrming
[96]. It seems that Tregs exert their suppressive function mainly in immunoregulatory role for CTLA-4 in uteroplacental interface
a contact dependent manner [97100]. In mice, Tregs seem to [106]. Decidual CD4+CD25high Treg cells express surface CTLA-4
present cytokine-mediated suppression [101]. while CD4+CD25high Tregs from peripheral blood lack the expres-
Immunosuppressive cytokines IL-10 and TGFb have been sion of CTLA-4 in human normal pregnancy [46]. In miscarriage
shown to be released by Tregs thereby suppress the effector cases the expression of CTLA-4 on decidual CD4+CD25high Treg
lymphocytes [102]. It has been emerged that one important cells is downregulated [46]. All these imply the possible role of
mechanism in maternal tolerance to the fetus is tryptophan CTLA-4 molecule on Tregs in maintaining normal pregnancy.
breakdown by the enzyme IDO. DCs catabolize tryptophan, an However on the contrary, blocking CTLA-4 has revealed no
important nutrient for Tregs, by secreting IDO. A drop in contribution on Treg suppressive activity in a pregnancy murine
tryptophan amount leads to suppression of T cell responses. model [107]. Therefore, the precise role of CTLA-4 in protective
Interestingly, CTLA-4 on Treg cells may affect the expression of IDO effects of Tregs in pregnancy remains to be elucidated. Pro-
[66,103105]. In human decidua CD4+CD25+ T cells have been grammed death 1 (PD-1) and its ligand PDL-1 may also play
demonstrated to express high frequency of intracellular CTLA-4 signicant role in tolerance to the fetus [108]. It has been observed
[58]. Thus increase in Treg numbers may stimulate IDO expression that blocking PDL1 in an animal model reduced regulatory function
in APCs then diminish tryptophan concentration. It is possible that and resulted in fetal death [109]. Loss of PDL1 function induces Th1
only a certain DC subtype would be able to respond to CTLA-4 by and Th17 proliferation, and disturbs Th1/Th2/Th17 balance, which

Table 1
The role of immune regulatory cells in normal pregnancy.

Immune Mechanism Role in pregnancy References


cells
Treg Secretion of IL-10 and TGF-b Suppress effector lymphocytes Groux et al. [34]
Weiner [102]
Induction of IDO expression in DC by CTLA-4 Decrease in tryptophan level Munn et al. [104],
Melloret al. [103],
Fingeret al. [105],
Fallarino et al. [66]
PD1PDL1 interaction inhibit Th1 and Th17 proliferation Habicht et al. [109],
Taglauer et al., [108]
DAddio et al. [110]
Up-regulation of galectin-1 Regulate T cell activation and cytokine production Rabinovich et al. [114], Dias-Baruf
Promote apoptosis of activated T cells et al. [115] Matarrese et al.[113]
Inhibit cell growth progression Ilarregui et al. [116]
Inducing tolerogenic DCs
Cell surface TGF-b1 Regulate T cell activation and NK cell function Nakamura et al. [118]
Increasing LIF and HO-1 levels Implantation success Zenclussen et al. [15]
Up-regulate TGB-b, IL-10 and CTLA-4
Breg Production of IL-10 Keep DC in an immature state Fillatreau et al. [156]
Inhibit the production of TNF-a by T cells Schumacher et al. [160]
Rolle et al.[151]
Jensenet al. [146]
NKT Release of IFN-g and IL-4 Regulate the Th1/Th2 balance Tupin et al. [190]
Massive activation of NKT cells induces pregnancy loss Ito et al. [182]
Boyson et al. [199]
MDSC unknown Suppression of the adaptive immunity by limiting the Zhao et al. [211]
proliferation and inltration of DCs and T cells
Secretion of ARG-1, iNOS and IDO and so depletion Suppress T cell activation and function Rodriguez et al. [225]
of arginine, cysteine and tryptophan Condamineet al. [222]
Fletcher et al. [223]
Membrane-bound TGF-b Inhibit T cell responses Li et al. [232]
Up-regulation of Cox2 unknown Zhao et al. [203]
unknown Induce the proliferation and differentiation of Foxp3 + Treg Dilek et al. [233]
cells Schlecker et al. [234]
Expression of PD-L1, CD80/86, and galectin-9 unknown De Wilde et al. [236]
Noman et al.[237]
Down regulating the expression of L-selectin on T Reduce homing to lymph nodes Hanson et al. [235]
cells
unknown Express a predominant but not exclusive Th2-type prole Bartmann, et al. [207]
MCP-1 expression Polarization of T cells into Th2 type Guet al. [239]
inhibit NK cells cytotoxicity Xu et al. [240]
Expression of chemokine TARC Induce Th2 response Tsuda et al. [241]
Engert et al. [242]

Abbreviations: IDO: Indoleamine 2,3-dioxygenase, DC: Dendritic cell, CTLA-4: Cytotoxic T-lymphocyte-associated protein 4, PD-1: Programmed cell death protein 1, PDL-1:
Programmed death-ligand 1, TGFb1: Transforming growth factor beta 1, LIF: Leukemia inhibitory factor, HO-1: Heme oxygenase-1, TNFa: Tumor necrosis factor alpha, IFNg:
Interferon gamma, IL-4: interleukin 4, IL-10: interleukin 10, ARG-1: arginase-1, iNOS: inducible Nitric oxide synthase, COX-2: Cyclooxygenase-2, Foxp3: Forkhead box protein
P3, MCP-1: Monocyte chemoattractant protein-1, TARC: Thymus- and activation-regulated chemokine, MDSC: Myeloid-derived Suppressor Cell, Breg: regulatory B cell, Treg:
regulatory T cell, NKT: Natural killer T, Th1: T helper-1, Th2: T helper-2, Th17: T helper 17, NK: Natural killer.
M. Ghaebi et al. / Biomedicine & Pharmacotherapy 88 (2017) 6173 65

lead to prevention of fetomaternal tolerance [110]. Studies in PD-1 in mice has indicated an importance of LIF in fetomaternal
knockout mice bearing allogeneic pregnancy showed increased tolerance induced by Tregs [120].
abortion rate, but the same rate was not observed in syngeneic HO-1 is important in maternal-fetal tolerance. HO-1 level is low
pregnancies [111] which supports the effects of PD-1 pathway in in placental tissue in abortion-prone mice compared with normal
fetomaternal tolerance. As shown in Table 1, Tregs up-regulate pregnant mice [125]. Foxp3+ Treg cells increase HO-1 level in
immune regulatory molecule galectin 10 [112]. Galectin1 regulates placenta [126] indicating the role of HO-1 in Treg-mediated
T cell activation and cytokine production [113,114], promotes tolerance induction. HO-1 may act in a Treg-dependent manner,
apoptosis of activated T cells and inhibits cell growth [112,113,115]. since HO-1 induction could up-regulate TGB-b, IL-10 and CTLA-4
These ndings suggest galectin1 expressing Treg may play a role in [127] and Treg marker neuropilin-1 [128]. These mechanisms are
the maternal-fetal tolerance. Ilarregui et al. showed that DCs shown in Fig. 1 in brief.
exposed to galectin1 promote IL10-mediated immune tolerance
[116], which is critical in pregnancy. Galectin1-decient mice show 3. Regulatory gdT cells
loss of allogenic pregnancies, supporting a role for galectin1 in
inducing tolerogenic DCs and also generating Tr1 cells in decidua T cells can be divided into two lineages based on the TCR
to maintain allogeneic fetus [117]. Cell surface TGF-b1 is one receptor chains, ab-TCR and gd-TCR expressing cells. The ab-T
contact-dependent mechanism for CD4+CD25+ Treg cells in order cells comprise the bulk of the T cell population in the blood and
to regulate T cell activation and NK cell function [118]. In a study lymphoid tissues while a small portion are TCRgd cells. Unlike
conducted on mice has been shown that the role for surface TGF- ab-T cells, it seems that majority of gdT cells are generated in
b1 may be controversial since TGF-b1 knockout Treg cells could extrathymic sites, only a portion of these cells are originated from
exhibit suppressive function [119]. thymus [129].
Treg transfer studies showed that Tregs create a microenviron- The gdT cells are present in endometrium during pregnancy in
ment rich in TGF-b, leukemia inhibitory factor (LIF) and Heme all mammals [130]. Study on murine system has revealed that the
oxygenase 1 (HO-1) at fetomaternal interface [120]. LIF is essential frequency of uterine gdT cells in allogeneic pregnancy is higher
for implantation success [121,122], however LIF participation in than syngeneic pregnancy and sex hormones have been shown to
elevating maternal tolerance to the embryo by affecting Th1/Th2 affect gdTCR expression in the pregnant uterus [131]. The number
balance is less understood [2]. Although some evidences demon- of gd T cells is increased in peripheral blood and decidua of healthy
strate involvement of LIF in graft acceptance in mouse models pregnant women compared with that of non-pregnant ones [132].
[123,124] suggesting that LIF may have roles in promotion of Regarding the characteristics of decidual gdT cells, it is observed
tolerance. Intriguingly, transfer of Treg to prevent abortion in mice that they are double negative granulated lymphocytes which
is efcient only very early in pregnancy, before implantation occur express neither CD4 nor CD8 [133]. They express CD69 (an
[125]. An augmentation in local levels of LIF following Treg transfer activation marker), CD45RO, CD94 (NK cell receptor) [133], NKG2D
and CTLA-4 and are also CD2-positive. Initial studies have

Fig. 1. Mechanisms of Treg in induction of fetomaternal tolerance. Tregs secrete IL-10 and TGFb and suppress effector lymphocytes. The interaction between CTLA-4 on Treg
and B7-ligand on DCs, makes DC to produce IDO which deprives T cells from Tryptophan. The crosstalk between dNK and CD14+ myelomonocytic cells results in IFNg
production that induces IDO expression in CD14+ cells. dNK cells release TGFb in fetomaternal interface. DCs, dNK and CD14+ cells through the secretion of TGF-b and IDO can
induce Treg differentiation. Tregs up-regulate immune regulatory molecule Galectin-1. Galectin-1 promotes apoptosis of activated T cells. DCs exposed with galectin-1
show IL-10 mediated immune tolerance. PD-1 is another regulatory molecule on Tregs. Loss of PDL1 function induces Th1 and Th17 proliferation. Tregs create a
microenvironment rich in LIF and HO-1. LIF is essential for implantation success. HO-1 is also important in fetomaternal tolerance.
Abbreviations: TGFB: Transforming growth factor beta, IL-10: Interleukin-10, PDL: Programmed death-ligand, PD-1: Programmed cell death protein 1, LIF: Leukemia
inhibitory factor, HO-1: Heme oxygenase-1, IFNg: Interferon gamma, IDO: Indoleamine 2,3-dioxygenase, CTLA-4: Cytotoxic T-lymphocyte-associated protein 4, Treg:
regulatory T cell, Beff: effector B, Th1: T helper1, Th17: T helper17, dNK: decidual Natural killer, dCD14+ cell: decidual CD14-positive cell, DC: Dendritic cell, Foxp3: Forkhead
box protein P3.
66 M. Ghaebi et al. / Biomedicine & Pharmacotherapy 88 (2017) 6173

suggested that gd T cells existing in uterus produce TGF-b and observed in AP animals. They observed that in addition to
suppress fetal rejection by maternal immune responses [134]. augmentation in frequency of regulatory B10 cells in normal
Further studies showed that decidual gd T cells produce IL-10 and pregnancies, their ability to produce IL-10 mRNA is also increased.
TGF-b at high levels, TNF-a and IFN-g at low levels and little or They transferred regulatory B10 cells to AP female mice and
none of IL-2 and IL-4 [132]. Lots of data from human and murine observed that adoptive transfer of regulatory B10 cells impedes
system demonstrated that decidual gd T cells appear to contribute immunological abortions [146]. Thus, the transfer of regulatory
to Th2 bias in maternal-fetal interface in favor of fetal tolerance B10 cells into AP animals normalizes the abortion rate, which is
[132,135,136]. A population of gdT cells producing IL-10 and TGF-b related to Treg expansion and keeping DCs in an immature state
cytokines has been isolated from tumor-inltrating lymphocytes. [146,160]. Jensen et al. demonstrated that IL-10 produced by
These gdT cells may be involved in inhibition of immune responses regulatory B10 cells is fundamental for keeping DCs in an
to tumor [137]. Findings on IL-10/TGF-b dominant expression in immature state during normal pregnancy. IL-10 prevents mono-
decidual gd T cells have led us to consider them as Treg cells. cytic DCs to develop into IL-12 producing mature DCs, thereby
Decidual gd T cells appeared to induce proliferation and invasion of inhibits them to activate T cells and to induce Th1 differentiation
human rst trimester trophoblast via IL-10 secretion in Fans and [161]. Rolle et al. observed signicantly augmented frequencies of
collegoues work [132], indicating that decidual gd T cells may play CD19+CD24hiCD27+ Breg in normal pregnant compared to non-
an important role in placenta development and function in human. pregnant women. Women suffering from miscarriages showed
It is currently unknown that by which mechanisms IL-10 can signicantly lower percentages of CD19+CD24hiCD27+ Breg than
promote trophoblast invasion and proliferation. women with normal pregnancies at the rst trimester. In addition,
There are two populations of gdT cells, resident and circulating they found that almost all CD19+CD24hiCD27+ cells (95%)
gdT cells. The main difference between the resident and circulating expressed the receptor for hCG. Interestingly, isolated CD19+ B
gd T cells is that they use different Vd chains. The resident gdT cells cells were able to produce IL-10 in response to human recombinant
are Vd1+ but the circulating part is Vd2+ [138]. Vd1+ gdT cells hCG, in vitro. Thus hCG induces IL-10 secretion by B cells and in this
reside in the epithelial surfaces of the intestine, tongue, lung, way inuences the tolerance exerted by Bregs [151]. A clinical
uterus, vagina, and murine skin [138]. In healthy pregnant women, observation in regard to the association between reproductive
there is a dominance of Vd1+ circulating cells, while women with hormones and regulatory B cells is the remission experienced by
recurrent abortions have an accumulation of Vd2+ circulating cells. women suffering MS during pregnancy [162]. Subramanian et al.
Activated gdT cells are much more in women with normal showed that regulatory B cells are involved in the E2-mediated
pregnancies than in recurrent aborters [139]. protection of EAE in mice lacking Tregs [163]. This study supports
the notion that sex hormones have a positive effect on the activity
4. Bregs of regulatory B cells leading to protection against disease. These
results are consistent with former reports demonstrating the role
Pregnancy is a unique condition in which the immune system of of regulatory B cells in protection against EAE through secretion of
mother meets a double challenge, to defend against microbial IL-10 [156,164,165]. B lymphocytes express ER-a receptor for
agents and at the same time must tolerate the semiallogenic fetus Eastrogen- signaling by this receptor is involved in the develop-
[140]. Cytokines produced by immune and nonimmune cells are ment, survival, expansion, and maturation of these cells [166168].
important players in controlling immune response [141]. IL-10 is a Progesterone (P4) has also been reported to affect immunoregula-
suppressive anti-inammatory cytokine that correlates with tion by B cells [169,170]. It is reported that administration of P4
pregnancy outcome and could be used as a therapeutic way in ameliorates the severity of EAE through elevating the numbers of
order to prevent naturally occurring pregnancy loss in a mouse CD19+ cells and level of IL-10 [171].
model [142]. However, IL-10 gene deletion could not induce Recurrent spontaneous abortion is associated with elevated
abortion [143145]. So, necessity of IL-10 in pregnancy success is level of pro-inammatory cytokine TNFa and blocking of TNFa
not clear [146]. It is accepted that trophoblasts or T cells are cellular was suggested as a treatment for recurrent spontaneous abortion
source of IL-10. Recently a new cell source of IL-10 has been [172]. Regulatory B10 cells inhibit secretion of TNFa by activated T
recognized which is known as regulatory B or B10 cells [147149]. cells in several pathological situations [173]. Isolated CD19+ B cells
Two main populations of B cells have been distinguished based on from pregnant women stimulated with CD40L/CpG and secreting
cellular markers, function, and localization [150]. In the past, the IL-10 were reported to inhibit TNFa production by activated CD4+
role of B cells in pregnancy was just attributed to antibody T cells [151]. Interestingly, B cells isolated from pregnant women
production [151]. During normal pregnancies protective antibodies without activation with CD40L/CpG exerted the same effect. In
are increased compared with non-pregnant females [152,153]. In contrast, CD40L/CpG activated and even non-activated B cells were
addition to antibody production, B cells have emerged as not able to inhibit TNFa secretion by T cells [151], suggesting these
regulators of immune responses [154]. B10 cells in particular are cells in pregnant women are activated naturally to produce IL-10,
potent immunoregulatory cell subtypes with ability to produce IL- perhaps with a soluble factor, to apply immunomodulation effects.
10 [147,149,155]. Indeed most of suppressive functions of
regulatory B cells are directly attributed to their capacity to 5. NKT cells
produce IL-10 [147,155157]. It has been suggested that IL-10-
producing B10 cells act to prevent autoimmunity [158] and are Natural killer T (NKT) cells constitute a distinct subgroup of
important mediators of chronic infections [159]. lymphocytes expressing both T-cell and NK-cell markers. These
Jensen et al.showed that the frequency of regulatory B10 cells cells are subdivided into two subgroups including: type I or
was signicantly higher than that observed in nonpregnant CBA/J invariant NKT (iNKT) cells with highly restricted TCR (V a24-J a18,
female mice, but AP (Abnormal Pregnant) mice did not show high V b11) and type II that is a population of CD1d-restricted NKT cells
levels of CD19+CD5+CD1d+ regulatory B10 cells, in fact percentage that express diverse TCRs, referred to as non-invariant NKT (non-
of B10 cells in these animals was comparable with nonpregnant iNKT) cells [174]. In addition, a third group named NKT-like cells
females. Then, they examined IL-10 levels in the spleen of that resembles NKT cells has been recently described. In contrast to
nonpregnant, NP (Normal Pregnant) and AP female mice. IL-10 traditional NKT cells, NKT-like cells (CD3+CD56+, CD3+CD16+)
mRNA level in the spleen of normal pregnant mice was up- are restricted to MHC but not CD1d [175]. NKT-like cells regulate
regulated compared with nonpregnant mice, but that was not both innate and adaptive immune systems [176,177] and serve as a
M. Ghaebi et al. / Biomedicine & Pharmacotherapy 88 (2017) 6173 67

bridge between the two systems [178]. They can modulate the Th1 cell activation in inammation induced preterm birth in a mouse
and Th2 balance [179,180]. NKT cells have been found in both model [200,201].
mouse and human decidua [181183] and affect maternal-fetal
tolerance and successful implantation [184186]. Ito et al. reported 6. MDSCs in pregnancy
that NKT cells were present in decidual tissues of pregnant mice
and induced abortion when stimulated by a specic ligand for MDSCs are a heterogeneous cell population consisting of
Va14, aGalCer [182]. Tsuda et al. reported more NKT cells immature granulocytes, macrophages, and DCs, which are the
accumulated in the human decidua compared with murine intermediates of the normal myeloid development in bone marrow
decidua [183]. NKT cells are not present in the endometrium [202]. They are characterized with myeloid origin, immature state,
during the nonpregnant period [182], but the number of NKT cells T cell suppression [203] and lacking markers of mature myeloid
in the decidua is apparently increased during early pregnancy and lymphoid cells [204]. In the noemal condition, immature
[181,183] and then declines during the late trimester of pregnancy myeloid cells differentiate into macrophages, DCs, and granulo-
[187]. cytes and are scarcely seen in peripheral blood. But differentiation
NKT-like cells were also found in the decidua, and were in myeloid lineage is altered in some pathologies, in particular
substantially increased in the URSA (unexplained RSA) group than malignancies and chronic infection and increased numbers of
in the early pregnancy and nonpregnant groups in peripheral blood immature myeloid cells are detected in circulation [205].
(PB) and decidua [188]. Activation of NKT cells has been observed In mice, these myeloid cells are distinguished by co-expression
in pregnancy loss. Heuvel et al. reported that high frequency of NKT of two surface markers CD11b and Gr-1 (Ly6G/Ly6C) and are
cells in PB correlates with recurrent pregnancy loss or implanta- subdivided into two groups including granulocytic MDSCs (G-
tion failure [189]. MDSCs) dened as CD11b+LY6G+LY6Clow cells and monocytic (M-
NKT cells modulate adaptive immune system by affecting the MDSCs) with CD11b+LY6GLY6Chigh phenotype [206]. In humans,
Th1/Th2 balance, in vivo [190]. NKT cells rapidly secrete IFN-g and very heterogeneous phenotypes of MDSC are described [207].
IL-4 after activation, thereby regulate Th1/Th2 polarity [191]. IFN-g Recently, two human MDSCs have been reported including
released by NKT cells can cause activation of other cell types such CD14CD15+CD66b+ (granulocyte, G-MDSCs) and
as NK cells in decidua, causing NK cell cytotoxicity and cytokine CD14CD15low/ (monocyte, M-MDSCs) [208,209]. MDSCs have
production [192]. The iNKT cells can activate other immune cells, been detected in uteri and placentas of mice [210,211]. More
such as DCs, macrophages, neutrophils, B cells and T cells by recently, MDSCs have been identied in human gestation. In
releasing cytokines [193]. Contact dependent mechanisms be- peripheral blood of pregnant woman, MDSCs appear to play an
tween NKT cells and trophoblasts are also implicated in inducing important role [212,213]. Studies that show progesterone increases
abortion [182]. The change in NKT cells may disturb Th1/Th2 MDSC also conrmed the importance of MDSC during pregnancy in
balance and leads to abortion. In supporting the role of NKT-like mice [214]. Other observations indicate that the presence of
cells during implantation, Miko et al.demonstrated that changes of maternal MDSC is related to the more metastatic activity and
NK and NKT-like cells phenotype during the implantation and early greater aggression of some tumors such as breast cancer, cervical
pregnancy in the same individual may also be important for cancer, lymphomas, malignant melanoma, and leukemias during
successful embryo implantation [184]. Non-iNKT cells have been pregnancy [211].
described in the bone marrow and hepatitis C virus-infected liver An experiment to investigate the role of MDSCs in immune
in humans [194,195]. Organ specic context might dictate ontogeny revealed that granulocytic MDSCs are present in the large
functional capabilities of resident non-iNKT cells, since non-iNKT numbers in pregnant women and in cord blood and drop rapidly
cells residing the bone marrow are Th2 like whereas those present during infancy [205]. Kostlin et al. demonstrated that G-MDSCs,
in infected liver are Th1-biased. Although the iNKT cells present in but not M-MDSCs, are increased in the peripheral blood of
the decidua affect the maintenance of pregnancy in both mice and pregnant women. They observed that percentages of G-MDSCs
humans [181,196], the role of non-iNKT cells in pregnancy remains were elevated in the 1st, 2nd, and 3rd trimester compared with
unknown. Uemura et al. observed that one subclass of CD1d nonpregnant controls and there was no signicant differences
restricted NKT cells that is distinct from the Va24 iNKT cells resides between these three trimesters [212]. On the other hand, Nair et al.
in the decidual tissue and is capable to retain a Th2 environment observed that percentage of CD33+ CD11b+ cells were at highest
during pregnancy. According to their reports, non-iNKT cells level during the 1st trimester of pregnancy then decreased
accumulated in the decidua may interact with the trophoblasts signicantly in 3rd trimester [213]. After delivery, the number of
that express CD1d molecules, having important functions in G-MDSCs decreased within a few days to levels of nonpregnant
pregnancy [185]. Although the decidual non-iNKT cells express controls [212]. Moreover, percentage of M-MDSCs did not change
diverse TCRs, they do not recognize fetal alloantigens. This signicantly during pregnancy [212], while Pan and colleagues
suggests that their recruitment to the decidua might be through observed that the population of M-MDSCs was elevated signi-
interaction with CD1d on trophoblasts rather than recognition of cantly in pregnant women compared with healthy donors and
fetal alloantigens. Pregnancy-associated hormones such as estro- postpartum women. Interestingly, the proportion of M-MDSCs
gen and progesterone may have various effects on these cells [197]. were raised throughout the rst to the second trimesters and
Decidual and peripheral iNKT cells differ in IFN-g and IL-4 declined from the third trimester, indicating dynamics of MDSCs
production. The iNKT clones in decidua showed a bias toward IFN- during gestation. The differences observed from these two studies
g secretion, while peripheral blood CD4+ iNKT population favors to may be due to the genetic background of subjects or cytokine
release IL-4. These ndings are consistent with the previous study milieus produced in the individuals or for other unknown reasons
by Takahashi et al. [198]. RT-PCR results from mouse decidual iNKT [215].
cells indicated the same results [182], proposing Th1-biased iNKT Nair et al. found that functionally suppressive MDSCs decrease
cells participate in both species. Boyson and colleagues demon- in blood and endometrium of miscarriage subjects compared with
strated that activation of iNKT cells leads to both early pregnancy control women undergoing successful pregnancies, especially in
loss and preterm birth, so mechanisms operating in early gestation the rst trimester [213].
are different from late stages of pregnancy [199]. Recently, Li et al. MDSCs do not need to be present in the large numbers to exert
identied the molecular mechanisms concerning the role of iNKT their suppressive functions as it has been previously shown that
68 M. Ghaebi et al. / Biomedicine & Pharmacotherapy 88 (2017) 6173

only 3% of MDSCs are required to completely block T cell 7. Conclusion


proliferation, in vitro [216,217].
Pan et al. also showed further immunosuppressive effects of Pregnancy is an active immunological process and leukocytes
oestrogen. They reported oestrogen facilitates the expansion and play an important role in gestation. Regulatory cells are present in
activation of M-MDSCs in human pregnancy [215]. fetomaternal interface and they have demonstrated to be potently
All MDSCs are characteristically immunosuppressive and immunosuppressive during pregnancy and in some cases of
function via suppression of T cells and/or NK cells [218,219]. reproductive disturbances their function and/or number are
MDSCs isolated from early septic mice produce proinammatory altered. These ndings suggest that they may be benecial in
cytokines TNF-a, IL-6, and IL-12 while MDSCs derived from late therapeutic approaches to pregnancy associated disorders. How-
septic mice produce TGF-b and IL-10 and were immunosuppres- ever many experiments have been conducted to investigate and
sive [220,221]. The context-specic functions of these MDSCs led clarify the role of different immune cell types in successful and
to speculation that in the feto-maternal interface, the phenotype disturbed pregnancies, the exact mechanisms of their function and
and activity of MDSCs depend on gestation time and the maternal precise role remain to be elucidated.
environment [203]. In the study conducted by Zhao et al. pregnant
mice were recently treated with Gr-1 antibody to deplete MDSCs or References
a low dose of LPS to transform MDSCs into activated neutrophils,
signicant increases in uterine DCs and T cells were observed, [1] S. Saito, A. Shiozaki, Y. Sasaki, A. Nakashima, T. Shima, M. Ito, Regulatory T
cells and regulatory natural killer (NK) cells play important roles in feto-
indicating that MDSCs may mediate the suppression of the maternal tolerance, Seminars in Immunopathology, Springer, 2007, pp. 115
adaptive immunity in the fetoplacental environment by inhibiting 122.
proliferation and inltration of DCs and T cells, in vivo [211]. MDSCs [2] J. Trowsdale, A.G. Betz, Mother's little helpers: mechanisms of maternal-fetal
tolerance, Nat. Immunol. 7 (3) (2006) 241246.
suppress T cell activation and function through secretion of [3] W.A. Hogge, A.L. Byrnes, M.C. Lanasa, U. Surti, The clinical use of karyotyping
arginase-1 (ARG-1), nitric oxide synthase (iNOS) and indoleamine spontaneous abortions, Am. J. Obstet. Gynecol. 189 (2) (2003) 397400.
2,3dioxygenase (IDO) in order to deplete essential nutrients for T [4] D.W. Branch, M. Gibson, R.M. Silver, Recurrent miscarriage, New Engl. J. Med.
363 (18) (2010) 17401747.
cell proliferation, l-arginine (l-Arg), l-cysteine and tryptophan [5] R. Rai, L. Regan, Recurrent miscarriage, Lancet 368 (9535) (2006) 601611.
[222224]. T cells deprivation from l-Arg hinders proliferation by [6] D. Lissauer, K. Piper, O. Goodyear, M.D. Kilby, P.A. Moss, Fetal-specic CD8+
reducing CD3z-chain expression, preventing the expression of cell- cytotoxic T cell responses develop during normal human pregnancy and
exhibit broad functional capacity, J. Immunol. 189 (2) (2012) 10721080.
cycle regulators cyclinD3 and cdk4 and impairing the production of
[7] M.T. Norton, K.A. Fortner, K.H. Oppenheimer, E.A. Bonney, Evidence that CD8
cytokines such as IFN-g [225]. Production of peroxynitrite (PNT) to T-cell homeostasis and function remain intact during murine pregnancy,
nitrosylation of T cell receptors via reactive oxygen species (ROS) Immunology 131 (3) (2010) 426437.
and iNOS is another mechanism of MDSC [222]. PNT could cause [8] I. Sargent, T. Wilkins, C. Redman, Maternal immune responses to the fetus in
early pregnancy and recurrent miscarriage, Lancet 332 (8620) (1988) 1099
nitration of the T cell receptor-CD8 complex that hinders binding to 1104.
MHC class I and makes T cells unresponsive to stimulation by [9] O.B. Christiansen, H.S. Nielsen, A. Kolte, A.T. Pedersen, Research methodology
antigen [203]. NO seems to be essential for successful implantation and epidemiology of relevance in recurrent pregnancy loss, Seminars in
Reproductive Medicine, Copyright 2006, Thieme Medical Publishers, Inc.,
[226], and a link between early pregnancy loss and production of 333 Seventh Avenue, New York, NY 10001, USA, 2006, pp. 005016.
NO by decidual macrophages was assumed [227]. Bartmann et al. [10] N. Rouas-Freiss, R.M.-B. Gonalves, C. Menier, J. Dausset, E.D. Carosella, Direct
results suggest decidua iNOS expression in CD33+ cells is of evidence to support the role of HLA-G in protecting the fetus from maternal
uterine natural killer cytolysis, Proc. Natl. Acad. Sci. 94 (21) (1997) 11520
importance in early pregnancy, while in later pregnancy, CD33+ 11525.
cells likely lose the expression of this enzyme [207]. The iNOS [11] J. Alijotas-Reig, Immunological puzzle related to recurrent miscarriage:
secreted by MDSCs causes conversion of l-arginine to citrulline and overview, Curr. Immunol. Rev. 5 (3) (2009) 175186.
[12] N. Lila, N. Rouas-Freiss, J. Dausset, A. Carpentier, E.D. Carosella, Soluble HLA-G
NO, which represses T cell action through inhibition of Jak/STAT protein secreted by allo-specic CD4+ T cells suppresses the allo-proliferative
signaling, reducing MHC class II expression and inducing T cell response: a CD4+ T cell regulatory mechanism, Proc. Natl. Acad. Sci. 98 (21)
apoptosis [228231]. MDSCs can also inhibit T cell responses (2001) 1215012155.
[13] F.d.S. Nardi, R. Slowik, P.F. Wowk, J.S. da Silva, G.F. Gelmini, T.F. Michelon, J.
through membrane-bound TGF-b [232]. Up-regulation of cyclo-
Neumann, M.d.G. Bicalho, Analysis of HLA-G polymorphisms in couples with
oxygenase 2 (cox2) is another mechanism of MDSC in fetomaternal implantation failure, Am. J. Reprod. Immunol. 68 (6) (2012) 507514.
immune crosstalk [203]. MDSCs are involved in inducing [14] D.A. Somerset, Y. Zheng, M.D. Kilby, D.M. Sansom, M.T. Drayson, Normal
proliferation and differentiation of Foxp3+ Treg cells [233,234]. human pregnancy is associated with an elevation in the immune suppressive
CD25+ CD4+ regulatory T-cell subset, Immunology 112 (1) (2004) 3843.
In addition, MDSCs downregulate the expression of L-selectin on T [15] A.C. Zenclussen, Regulatory T cells in pregnancy, Springer Seminars in
cells, which results in a reduced homing to lymph nodes and Immunopathology, Springer, 2006, pp. 3139.
tumors [235]. Additional mechanisms include expression of [16] A. Erlebacher, D. Vencato, K.A. Price, D. Zhang, L.H. Glimcher, Constraints in
antigen presentation severely restrict T cell recognition of the allogeneic
program death ligand 1(PD-L1), CD80/86, and galectin-9 fetus, J. Clin. Invest. 117 (5) (2007) 13991411.
[236,237]. In Bartmann et al. study, decidual CD33+ cells did [17] R.M. Verdijk, A. Kloosterman, J. Pool, M. van de Keur, A.M. Naipal, A.G. van
express a predominant but not exclusive Th2-type prole. Halteren, A. Brand, T. Mutis, E. Goulmy, Pregnancy induces minor
histocompatibility antigenspecic cytotoxic T cells: implications for stem
Interestingly, MCP-1 was expressed by all CD33+ cells investigated cell transplantation and immunotherapy, Blood 103 (5) (2004) 19611964.
herein [207]. Beside its chemoattractant inuence on monocytes, [18] X.W. Tan, H. Liao, L. Sun, M. Okabe, Z.C. Xiao, G.S. Dawe, Fetal microchimerism
macrophages and lymphocytes [238], MCP-1 is linked with Th2 in the maternal mouse brain: a novel population of fetal progenitor or stem
cells able to cross the bloodbrain barrier? Stem Cells 23 (10) (2005) 1443
response in T-cell immunity [239]. Moreover, MCP-1 inhibits
1452.
perforin production in uterine NK cells [240]. Therefore, decidual [19] Z. Yan, N.C. Lambert, K.A. Guthrie, A.J. Porter, L.S. Loubiere, M.M. Madeleine, A.
MDSCs not only suppress T cell proliferation but also contribute to M. Stevens, H.M. Hermes, J.L. Nelson, Male microchimerism in women
without sons: quantitative assessment and correlation with pregnancy
polarization of T cells into Th2 type immunity and inhibit NK cells
history, Am. J. Med. 118 (8) (2005) 899906.
cytotoxicity. These functions all together promote a successful [20] O.J. Holland, C. Linscheid, H.C. Hodes, T.L. Nauser, M. Gilliam, P. Stone, L.W.
pregnancy. Another chemokine, TARC, which operates in favor of Chamley, M.G. Petroff, Minor histocompatibility antigens are expressed in
Th2 cell responses in the decidua [241] is expressed by the CD33+ syncytiotrophoblast and trophoblast debris: implications for maternal
alloreactivity to the fetus, Am. J. Pathol. 180 (1) (2012) 256266.
cells and not by the CD14+ cells [242] and conrms the role of the [21] P.C. Arck, K. Hecher, Fetomaternal immune cross-talk and its consequences
CD33+ decidual MDSCs in assisting maternal immune environ- for maternal and offspring's health, Nat. Med. 99 (5) (2013) 548556.
ment to maintain pregnancy. [22] A. Munoz-Suano, A.B. Hamilton, A.G. Betz, Gimme shelter: the immune
system during pregnancy, Immunol. Rev. 241 (1) (2011) 2038.
M. Ghaebi et al. / Biomedicine & Pharmacotherapy 88 (2017) 6173 69

[23] U. Thiruchelvam, I. Dranseld, P.T. Saunders, H.O. Critchley, The importance [49] J. Mjsberg, J. Svensson, E. Johansson, L. Hellstrm, R. Casas, M.C. Jenmalm, R.
of the macrophage within the human endometrium, J. Leukoc. Biol. 93 (2) Boij, L. Matthiesen, J.-I. Jnsson, G. Berg, Systemic reduction of functionally
(2013) 217225. suppressive CD4dimCD25highFoxp3+ Tregs in human second trimester
[24] A. Tafuri, J. Alferink, P. Moller, G.J. Hammerling, B. Arnold, T cell awareness of pregnancy is induced by progesterone and 17b-estradiol, J. Immunol. 183 (1)
paternal alloantigens during pregnancy, Science 270 (5236) (1995) 630. (2009) 759769.
[25] G. Chaouat, G.A. Voisin, Regulatory T cell subpopulations in pregnancy I. [50] L. Arruvito, M. Sanz, A.H. Banham, L. Fainboim, Expansion of CD4+ CD25+ and
Evidence for suppressive activity of the early phase of MLR, J. Immunol. 122 FOXP3+ regulatory T cells during the follicular phase of the menstrual cycle:
(4) (1979) 13831388. implications for human reproduction, J. Immunol. 178 (4) (2007) 25722578.
[26] R. Raghupathy, Th 1-type immunity is incompatible with successful [51] W.-J. Wang, C.-F. Hao, G.-J. Yin, S.-H. Bao, L.-H. Qiu, Q.-D. Lin, Increased
pregnancy, Immunol. Today 18 (10) (1997) 478482. prevalence of T helper 17 (Th17) cells in peripheral blood and decidua in
[27] S. Saito, A. Nakashima, T. Shima, M. Ito, Review article: th1/Th2/Th17 and unexplained recurrent spontaneous abortion patients, J. Reprod. Immunol.
regulatory T-Cell paradigm in pregnancy, Am. J. Reprod. Immunol. 63 (6) 84 (2) (2010) 164170.
(2010) 601610. [52] D. Darmochwal-Kolarz, S. Saito, J. Rolinski, J. Tabarkiewicz, B. Kolarz, B.
[28] J. Kwak-Kim, H. Chung-Bang, S. Ng, E. Ntrivalas, C. Mangubat, K. Beaman, A. Leszczynska-Gorzelak, J. Oleszczuk, Activated t lymphocytes in pre-
Beer, A. Gilman-Sachs, Increased T helper 1 cytokine responses by circulating Eclampsia, Am. J. Reprod. Immunol. 58 (1) (2007) 3945.
T cells are present in women with recurrent pregnancy losses and in infertile [53] Y. Sasaki, D. Darmochwal-Kolarz, D. Suzuki, M. Sakai, M. Ito, T. Shima, A.
women with multiple implantation failures after IVF, Hum. Reprod. 18 (4) Shiozaki, J. Rolinski, S. Saito, Proportion of peripheral blood and decidual CD4
(2003) 767773. + CD25bright regulatory T cells in pre-eclampsia, Clin. Exp. Immunol. 149 (1)
[29] S. Lee, J. Kim, S. Hur, C. Kim, B. Na, M. Lee, A. Gilman-Sachs, J. Kwak-Kim, An (2007) 139145.
imbalance in interleukin-17-producing T and Foxp3+ regulatory T cells in [54] M.J. Jasper, K.P. Tremellen, S.A. Robertson, Primary unexplained infertility is
women with idiopathic recurrent pregnancy loss, Hum. Reprod. 26 (11) associated with reduced expression of the T-regulatory cell transcription
(2011) 29642971. factor Foxp3 in endometrial tissue, Mol. Hum. Reprod. 12 (5) (2006) 301308.
[30] G. Chaouat, N. Lde-Bataille, S. Zourbas, S. Ostojic, S. Dubanchet, J. Martal, R. [55] R. Pijnenborg, J. Bland, W. Robertson, I. Brosens, Uteroplacental arterial
Frydman, Cytokines, implantation and early abortion: re-examining the Th1/ changes related to interstitial trophoblast migration in early human
Th2 paradigm leads to question the single pathway, single therapy concept, pregnancy, Placenta 4 (4) (1983) 397413.
Am. J. Reprod. Immunol. 50 (3) (2003) 177186. [56] E.E. Winger, J.L. Reed, Low circulating CD4+ CD25+ Foxp3+ T regulatory cell
[31] F. Jadidi-Niaragh, M. Youse, A. Memarian, M. Hojjat-Farsangi, J. Khoshnoodi, levels predict miscarriage risk in newly pregnant women with a history of
S.M. Razavi, M. Jeddi-Tehrani, F. Shokri, Increased frequency of CD8+ and CD4 failure, Am. J. Reprod. Immunol. 66 (4) (2011) 320328.
+ regulatory T cells in chronic lymphocytic leukemia: association with [57] R.M. Steinman, D. Hawiger, M.C. Nussenzweig, Tolerogenic dendritic cells*,
disease progression, Cancer Invest. 31 (2) (2013) 121131. Annu. Rev. Immunol. 21 (1) (2003) 685711.
[32] M. Gol-Ara, F. Jadidi-Niaragh, R. Sadria, G. Azizi, A. Mirshaey, The role of [58] J. Heikkinen, M. Mttnen, A. Alanen, O. Lassila, Phenotypic characterization
different subsets of regulatory T cells in immunopathogenesis of rheumatoid of regulatory T cells in the human decidua, Clin. Exp. Immunol. 136 (2) (2004)
arthritis, Arthritis 2012 (2012) 116. 373378.
[33] J. Alijotas-Reig, E. Llurba, J.M. Gris, Potentiating maternal immune tolerance [59] J. Heikkinen, M. Mttnen, J. Komi, A. Alanen, O. Lassila, Phenotypic
in pregnancy: a new challenging role for regulatory T cells, Placenta 35 (4) characterization of human decidual macrophages, Clin. Exp. Immunol. 131
(2014) 241248. (3) (2003) 498505.
[34] H. Groux, A. O'Garra, M. Bigler, M. Rouleau, S. Antonenko, J.E. de Vries, M.G. [60] L. Gardner, A. Moffett, Dendritic cells in the human decidua, Biol. Reprod. 69
Roncarolo, A CD4+; T-cell subset inhibits antigen-specic T-cell responses (4) (2003) 14381446.
and prevents colitis, Nature 389 (6652) (1997) 737742. [61] U. Kmmerer, A.O. Eggert, M. Kapp, A.D. McLellan, T.B. Geijtenbeek, J. Dietl, Y.
[35] Y.Y. Wan, R.A. Flavell, YinYangfunctions of transforming growth factor-b van Kooyk, E. Kmpgen, Unique appearance of proliferating antigen-
and T regulatory cells in immune regulation, Immunol. Rev. 220 (1) (2007) presenting cells expressing DC-SIGN (CD209) in the decidua of early human
199213. pregnancy, Am. J. Pathol. 162 (3) (2003) 887896.
[36] L.R. Guerin, J.R. Prins, S.A. Robertson, Regulatory T-cells and immune [62] A.E. Morelli, A.W. Thomson, Tolerogenic dendritic cells and the quest for
tolerance in pregnancy: a new target for infertility treatment? Hum. Reprod. transplant tolerance, Nat. Rev. Immunol. 7 (8) (2007) 610621.
Update 15 (5) (2009) 517535. [63] S. Yamazaki, T. Iyoda, K. Tarbell, K. Olson, K. Velinzon, K. Inaba, R.M. Steinman,
[37] E.M. Shevach, CD4+ CD25+ suppressor T cells: more questions than answers, Direct expansion of functional CD25+ CD4+ regulatory T cells by antigen-
Nat. Rev. Immunol. 2 (6) (2002) 389400. processing dendritic cells, J. Exp. Med. 198 (2) (2003) 235247.
[38] S.A. Robertson, Control of the immunological environment of the uterus, Rev. [64] M.D. Sharma, B. Baban, P. Chandler, D.-Y. Hou, N. Singh, H. Yagita, M. Azuma, B.
Reprod. 5 (3) (2000) 164174. R. Blazar, A.L. Mellor, D.H. Munn, Plasmacytoid dendritic cells from mouse
[39] C.A. White, M. Johansson, C.T. Roberts, A.J. Ramsay, S.A. Robertson, Effect of tumor-draining lymph nodes directly activate mature Tregs via indoleamine
interleukin-10 null mutation on maternal immune response and 2, 3-dioxygenase, J. Clin. Invest. 117 (9) (2007) 25702582.
reproductive outcome in mice, Biol. Reprod. 70 (1) (2004) 123131. [65] W. Chen, X. Liang, A.J. Peterson, D.H. Munn, B.R. Blazar, The indoleamine 2, 3-
[40] S.A. Robertson, A.S. Care, R.J. Skinner, Interleukin 10 regulates inammatory dioxygenase pathway is essential for human plasmacytoid dendritic cell-
cytokine synthesis to protect against lipopolysaccharide-induced abortion induced adaptive T regulatory cell generation, J. Immunol. 181 (8) (2008)
and fetal growth restriction in mice, Biol. Reprod. 76 (5) (2007) 738748. 53965404.
[41] S. Gregori, C.F. Magnani, M.-G. Roncarolo, Role of human leukocyte antigen-G [66] F. Fallarino, U. Grohmann, K.W. Hwang, C. Orabona, C. Vacca, R. Bianchi, M.L.
in the induction of adaptive type 1 regulatory T cells, Hum. Immunol. 70 (12) Belladonna, M.C. Fioretti, M.-L. Alegre, P. Puccetti, Modulation of tryptophan
(2009) 966969. catabolism by regulatory T cells, Nat. Immunol. 4 (12) (2003) 12061212.
[42] A. Naji, S. Le Rond, A. Durrbach, I. Krawice-Radanne, C. Creput, M. Daouya, J. [67] K. Sato, N. Yamashita, M. Baba, T. Matsuyama, Modied myeloid dendritic
Caumartin, J. LeMaoult, E.D. Carosella, N. Rouas-Freiss, CD3+ CD4low and CD3 cells act as regulatory dendritic cells to induce anergic and regulatory T cells,
+ CD8low are induced by HLA-G: novel human peripheral blood suppressor Blood 101 (9) (2003) 35813589.
T-cell subsets involved in transplant acceptance, Blood 110 (12) (2007) 3936 [68] C. Oderup, L. Cederbom, A. Makowska, C.M. Cilio, F. Ivars, Cytotoxic T
3948. lymphocyte antigen-4-dependent down-modulation of costimulatory
[43] S. Gregori, D. Tomasoni, V. Pacciani, M. Scirpoli, M. Battaglia, C.F. Magnani, E. molecules on dendritic cells in CD4+ CD25+ regulatory T-cell-mediated
Hauben, M.-G. Roncarolo, Differentiation of type 1 T regulatory cells (Tr1) by suppression, Immunology 118 (2) (2006) 240249.
tolerogenic DC-10 requires the IL-10dependent ILT4/HLA-G pathway, Blood [69] Y. Belkaid, G. Oldenhove, Tuning microenvironments: induction of regulatory
116 (6) (2010) 935944. t cells by dendritic cells, Immunity 29 (3) (2008) 362371.
[44] G. Amodio, A. Mugione, A.M. Sanchez, P. Vigan, M. Candiani, E. Somigliana, [70] I. Pedroza-Pacheco, A. Madrigal, A. Saudemont, Interaction between natural
M.G. Roncarolo, P. Panina-Bordignon, S. Gregori, HLA-G expressing DC-10 and killer cells and regulatory T cells: perspectives for immunotherapy, Cell. Mol.
CD4+ T cells accumulate in human decidua during pregnancy, Hum. Immunol. 10 (3) (2013) 222229.
Immunol. 74 (4) (2013) 406411. [71] A. King, T. Burrows, S. Verma, S. Hiby, Y. Loke, Human uterine lymphocytes,
[45] W.V. Ingman, R.L. Robker, K. Woittiez, S.A. Robertson, Null mutation in Hum. Reprod. Update 4 (5) (1998) 480485.
transforming growth factor b1 disrupts ovarian function and causes oocyte [72] A. Moffett-King, Natural killer cells and pregnancy, Nat. Rev. Immunol. 2 (9)
incompetence and early embryo arrest, Endocrinology 147 (2) (2006) 835 (2002) 656663.
845. [73] P. Vacca, C. Cantoni, M. Vitale, C. Prato, F. Canegallo, D. Fenoglio, N. Ragni, L.
[46] Y. Sasaki, M. Sakai, S. Miyazaki, S. Higuma, A. Shiozaki, S. Saito, Decidual and Moretta, M.C. Mingari, Crosstalk between decidual NK and CD14+
peripheral blood CD4+ CD25+ regulatory T cells in early pregnancy subjects myelomonocytic cells results in induction of Tregs and immunosuppression,
and spontaneous abortion cases, Mol. Hum. Reprod. 10 (5) (2004) 347353. Proc. Natl. Acad. Sci. 107 (26) (2010) 1191811923.
[47] T. Tilburgs, D. Roelen, B. Van Der Mast, J. Van Schip, C. Kleijburg, G. de Groot- [74] D.A. Clark, G. Vince, K.C. Flanders, H. Hirte, P. Starkey, Immunology: CD56+
Swings, H. Kanhai, F. Claas, S. Scherjon, Differential distribution of CD4+ CD25 lymphoid cells in human rst trimester pregnancy decidua as a source of
bright and CD8+ CD28- T-cells in decidua and maternal blood during human novel transforming growth factor-b2-related immunosuppressive factors,
pregnancy, Placenta 27 (2006) 4753. Hum. Reprod. 9 (12) (1994) 22702277.
[48] T. Tilburgs, D.L. Roelen, B.J. van der Mast, G.M. de Groot-Swings, C. Kleijburg, [75] S. Higuma-Myojo, Y. Sasaki, S. Miyazaki, M. Sakai, A. Siozaki, N. Miwa, S. Saito,
S.A. Scherjon, F.H. Claas, Evidence for a selective migration of fetus-specic Cytokine prole of natural killer cells in early human pregnancy, Am. J.
CD4+ CD25bright regulatory T cells from the peripheral blood to the decidua Reprod. Immunol. 54 (1) (2005) 2129.
in human pregnancy, J. Immunol. 180 (8) (2008) 57375745.
70 M. Ghaebi et al. / Biomedicine & Pharmacotherapy 88 (2017) 6173

[76] W.H. Fridman, F. Pags, C. Sauts-Fridman, J. Galon, The immune contexture [101] A. Dario, L.W. Vignali, J. Creg, How regulatory T cells work, Nat. Rev. Immunol.
in human tumours: impact on clinical outcome, Nat. Rev. Cancer 12 (4) (2012) 8 (7) (2008) 523532.
298306. [102] H.L. Weiner, Induction and mechanism of action of transforming growth
[77] N. Ralainirina, A. Poli, T. Michel, L. Poos, E. Andrs, F. Hentges, J. Zimmer, factor-b-secreting Th3 regulatory cells, Immunol. Rev. 182 (1) (2001) 207
Control of NK cell functions by CD4+ CD25+ regulatory T cells, J. Leukoc. Biol. 214.
81 (1) (2007) 144153. [103] J. Mellor, P. Chandler, K. Smith, H. Molina, D. Mao, D.H. Munn, Prevention of
[78] D.B. Keskin, D.S. Allan, B. Rybalov, M.M. Andzelm, J.N. Stern, H.D. Kopcow, L.A. Tcelldriven complement activation and inammation by tryptophan
Koopman, J.L. Strominger, TGFb promotes conversion of CD16+ peripheral catabolism during pregnancy, Nat. Immunol. 2 (1) (2001) 6468.
blood NK cells into CD16- NK cells with similarities to decidual NK cells, Proc. [104] D.H. Munn, M. Zhou, J.T. Attwood, I. Bondarev, S.J. Conway, B. Marshall, C.
Natl. Acad. Sci. 104 (9) (2007) 33783383. Brown, A.L. Mellor, Prevention of allogeneic fetal rejection by tryptophan
[79] M. Youse, A.A. Movassaghpour, K. Shamsasenjan, G. Ghalamfarsa, S. catabolism, Science 281 (5380) (1998) 11911193.
Sadreddini, F. Jadidi-Niaragh, M. Hojjat-Farsangi, The skewed balance [105] E.B. Finger, J.A. Bluestone, When ligand becomes receptortolerance via B7
between Tregs and Th17 in chronic lymphocytic leukemia, Future Oncol. 11 signaling on DCs, Nat. Immunol. 3 (11) (2002) 10561057.
(10) (2015) 15671582. [106] K. Kaufman, J. Bowen, A. Tsai, J. Bluestone, J. Hunt, C. Ober, The CTLA-4 gene is
[80] M. Kleinewietfeld, D.A. Haer, The plasticity of human Treg and Th17 cells expressed in placental broblasts, Mol. Hum. Reprod. 5 (1) (1999) 8487.
and its role in autoimmunity, Seminars in Immunology, Elsevier, 2013, pp. [107] P.O. Wafula, A. Teles, A. Schumacher, K. Pohl, H. Yagita, H.D. Volk, A.C.
305312. Zenclussen, PD-1 but not CTLA-4 blockage abrogates the protective effect of
[81] F. Jadidi-Niaragh, A. Mirshaey, The deviated balance between regulatory T regulatory t cells in a pregnancy murine model, Am. J. Reprod. Immunol. 62
cell and Th17 in autoimmunity, Immunopharmacol. Immunotoxicol. 34 (5) (5) (2009) 283292.
(2012) 727739. [108] E.S. Taglauer, T.M. Yankee, M.G. Petroff, Maternal PD-1 regulates
[82] G. Azizi, F. Jadidi-Niaragh, A. Mirshaey, Th17Cells in Immunopathogenesis accumulation of fetal antigen-specic CD8+ T cells in pregnancy, J. Reprod.
and treatment of rheumatoid arthritis, Int. J. Rheum. Dis. 16 (3) (2013) 243 Immunol. 80 (1) (2009) 1221.
253. [109] A. Habicht, S. Dada, M. Jurewicz, B.T. Fife, H. Yagita, M. Azuma, M.H. Sayegh, I.
[83] F. Jadidi-Niaragh, G. Ghalamfarsa, A. Memarian, H. Asgarian-Omran, S.M. Guleria, A link between PDL1 and T regulatory cells in fetomaternal tolerance,
Razavi, A. Sarrafnejad, F. Shokri, Downregulation of IL-17-producing T cells is J. Immunol. 179 (8) (2007) 52115219.
associated with regulatory T cell expansion and disease progression in [110] F. DAddio, L.V. Riella, B.G. Mfarrej, L. Chabtini, L.T. Adams, M. Yeung, H. Yagita,
chronic lymphocytic leukemia, Tumor Biol. 34 (2) (2013) 929940. M. Azuma, M.H. Sayegh, I. Guleria, The link between the PDL1 costimulatory
[84] L. Xu, A. Kitani, I. Fuss, W. Strober, Cutting edge: regulatory T cells induce CD4 pathway and Th17 in fetomaternal tolerance, J. Immunol. 187 (9) (2011)
+ CD25- Foxp3- T cells or are self-induced to become Th17 cells in the absence 45304541.
of exogenous TGF-b, J. Immunol. 178 (11) (2007) 67256729. [111] I. Guleria, A. Khosroshahi, M.J. Ansari, A. Habicht, M. Azuma, H. Yagita, R.J.
[85] F. Jadidi-Niaragh, A. Mirshaey, Th17 cell, the new player of Noelle, A. Coyle, A.L. Mellor, S.J. Khoury, A critical role for the programmed
neuroinammatory process in multiple sclerosis, Scand. J. Immunol. 74 (1) death ligand 1 in fetomaternal tolerance, J. Exp. Med. 202 (2) (2005) 231237.
(2011) 113. [112] M.I. Garn, C.-C. Chu, D. Golshayan, E. Cernuda-Morolln, R. Wait, R.I. Lechler,
[86] V.R. Aluvihare, M. Kallikourdis, A.G. Betz, Regulatory T cells mediate maternal Galectin-1: a key effector of regulation mediated by CD4+ CD25+ T cells,
tolerance to the fetus, Nat. Immunol. 5 (3) (2004) 266271. Blood 109 (5) (2007) 20582065.
[87] J.H. Lee, B. Ulrich, J. Cho, J. Park, C.H. Kim, Progesterone promotes [113] P. Matarrese, A. Tinari, E. Mormone, G.A. Bianco, M.A. Toscano, B. Ascione, G.A.
differentiation of human cord blood fetal T cells into T regulatory cells but Rabinovich, W. Malorni, Galectin-1 sensitizes resting human T lymphocytes
suppresses their differentiation into Th17 cells, J. Immunol. 187 (4) (2011) to Fas (CD95)-mediated cell death via mitochondrial hyperpolarization,
17781787. budding, and ssion, J. Biol. Chem. 280 (8) (2005) 69696985.
[88] M. Kouck, K. Mal9 ckov, T. Cindrov-Davies, A. Germanov, A. Parzek, M. [114] G. Rabinovich, A. Ariel, R. Hershkoviz, J. Hirabayashi, K. Kasai, O. Lider, Specic
Kalousov, Z. Hjek, T. Zima, Low levels of circulating T-regulatory inhibition of T-cell adhesion to extracellular matrix and proinammatory
lymphocytes and short cervical length are associated with preterm labor, J. cytokine secretion by human recombinant galectin-1, Immunology 97 (1)
Reprod. Immunol. 106 (2014) 110117. (1999) 100106.
[89] E. Soto-Vega, H. Solleiro-Villavicencio, M. Vargas-Rojas, Th1, Th2, Th17 and [115] M. Dias-Baruf, H. Zhu, M. Cho, S. Karmakar, R.P. McEver, R.D. Cummings,
Treg levels in umbilical cord blood of preeclamptic women.(HUM1P. 300), J. Dimeric galectin-1 induces surface exposure of phosphatidylserine and
Immunol. 194 (1 Suppl) (2015) 52 (2552.25). phagocytic recognition of leukocytes without inducing apoptosis, J. Biol.
[90] C. Pasare, R. Medzhitov, Toll pathway-dependent blockade of CD4+ CD25+ T Chem. 278 (42) (2003) 4128241293.
cell-mediated suppression by dendritic cells, Science 299 (5609) (2003) [116] J.M. Ilarregui, D.O. Croci, G.A. Bianco, M.A. Toscano, M. Salatino, M.E.
10331036. Vermeulen, J.R. Geffner, G.A. Rabinovich, Tolerogenic signals delivered by
[91] S. Huber, N. Gagliani, E. Esplugues, W. O'Connor, F.J. Huber, A. Chaudhry, M. dendritic cells to T cells through a galectin-1-driven immunoregulatory
Kamanaka, Y. Kobayashi, C.J. Booth, A.Y. Rudensky, Th17 cells express circuit involving interleukin 27 and interleukin 10, Nat. Immunol. 10 (9)
interleukin-10 receptor and are controlled by Foxp3- and Foxp3+ regulatory (2009) 981991.
CD4+ T cells in an interleukin-10-dependent manner, Immunity 34 (4) (2011) [117] S.M. Blois, J.M. Ilarregui, M. Tometten, M. Garcia, A.S. Orsal, R. Cordo-Russo,
554565. M.A. Toscano, G.A. Bianco, P. Kobelt, B. Handjiski, A pivotal role for galectin-1
[92] G. Murugaiyan, A. Mittal, R. Lopez-Diego, L.M. Maier, D.E. Anderson, H.L. in fetomaternal tolerance, Nat. Med. 13 (12) (2007) 14501457.
Weiner, IL-27 is a key regulator of IL-10 and IL-17 production by human CD4+ [118] K. Nakamura, A. Kitani, W. Strober, Cell contact?dependent
T cells, J. Immunol. 183 (4) (2009) 24352443. immunosuppression by CD4+ CD25+ regulatory T cells is mediated by cell
[93] G. Chaouat, M. Petitbarat, R. Bulla, S. Dubanchet, K. Valdivia, N. Lede, T. surfacebound transforming growth factor b, J. Exp. Med. 194 (5) (2001)
Steffen, J.C. Jensenius, F. Tedesco, Early regulators in abortion and 629644.
implications for a preeclampsia model, J. Reprod. Immunol. 82 (2) (2009) [119] C.A. Piccirillo, J.J. Letterio, A.M. Thornton, R.S. McHugh, M. Mamura, H.
132141. Mizuhara, E.M. Shevach, CD4+ CD25+ regulatory T cells can mediate
[94] A.E. Mas, M. Petitbarat, S. Dubanchet, S. Fay, N. Lede, G. Chaouat, Immune suppressor function in the absence of transforming growth factor b1
regulation at the interface during early steps of murine implantation: production and responsiveness, J. Exp. Med. 196 (2) (2002) 237246.
involvement of two new cytokines of the IL-12 family (IL-23 and IL-27) and of [120] A.C. Zenclussen, K. Gerlof, M.L. Zenclussen, S. Ritschel, A. Zambon Bertoja, S.
TWEAK, Am. J. Reprod. Immunol. 59 (4) (2008) 323338. Fest, S. Hontsu, S. Ueha, K. Matsushima, J. Leber, Regulatory T cells induce a
[95] L. Gabryov, J.R. Christensen, X. Wu, A. Kissenpfennig, B. Malissen, A. privileged tolerant microenvironment at the fetal-maternal interface, Eur. J.
O'Garra, Integrated T-cell receptor and costimulatory signals determine TGF- Immunol. 36 (1) (2006) 8294.
b-dependent differentiation and maintenance of Foxp3+ regulatory T cells, [121] J. Sherwin, T. Freeman, R. Stephens, S. Kimber, A. Smith, I. Chambers, S. Smith,
Eur. J. Immunol. 41 (5) (2011) 12421248. A. Sharkey, Identication of genes regulated by leukemia-inhibitory factor in
[96] D.A. Clark, G. Chaouat, Regulatory T cells and reproduction: how do they do the mouse uterus at the time of implantation, Mol. Endocrinol. 18 (9) (2004)
it? J. Reprod. Immunol. 96 (1) (2012) 17. 21852195.
[97] B. Birebent, R. Lorho, H. Lechartier, S. de Guibert, M. Alizadeh, N. Vu, A. [122] C.L. Stewart, P. Kaspar, L.J. Brunet, H. Bhatt, I. Gadi, F. Kntgen, S.J.
Beauplet, N. Robillard, G. Semana, uppressive properties of human CD4+ Abbondanzo, Blastocyst implantation depends on maternal expression of
CD25+ regulatory T cells are dependent on CTLA-4 expression, Eur. J. leukaemia inhibitory factor, Nature 359 (6390) (1992) 7679.
Immunol. 34 (12) (2004) 34853496. [123] S. Akita, H. Ishihara, R.M. Abdur, T. Fujii, Leukemia inhibitory factor gene
[98] D. Dieckmann, H. Plottner, S. Berchtold, T. Berger, G. Schuler, Ex vivo isolation improves skin allograft survival in the mouse model1, Transplantation 70 (7)
and characterization of CD4+ CD25+ T cells with regulatory properties from (2000) 10261031.
human blood, J. Exp. Med. 193 (11) (2001) 13031310. [124] S.M. Metcalfe, A.D.S. Poorni, Transplantation tolerance: gene expression
[99] H. Jonuleit, E. Schmitt, M. Stassen, A. Tuettenberg, J. Knop, A.H. Enk, proles comparing allotolerance vs. allorejection, Int. Immunopharmacol. 5
Identication and functional characterization of human CD4+ CD25+ T cells (1) (2005) 3339.
with regulatory properties isolated from peripheral blood, J. Exp. Med. 193 [125] A.C. Zenclussen, K. Gerlof, M.L. Zenclussen, A. Sollwedel, A.Z. Bertoja, T. Ritter,
(11) (2001) 12851294. K. Kotsch, J. Leber, H.-D. Volk, Abnormal T-cell reactivity against paternal
[100] M.K. Levings, R. Sangregorio, M.-G. Roncarolo, Human CD25+ CD4+ T antigens in spontaneous abortion: adoptive transfer of pregnancy-induced
regulatory cells suppress naive and memory T cell proliferation and can be CD4+ CD25+ T regulatory cells prevents fetal rejection in a murine abortion
expanded in vitro without loss of function, J. Exp. Med. 193 (11) (2001) 1295 model, Am. J. Pathol. 166 (3) (2005) 811822.
1302.
M. Ghaebi et al. / Biomedicine & Pharmacotherapy 88 (2017) 6173 71

[126] B.-M. Choi, H.-O. Pae, Y.-R. Jeong, Y.-M. Kim, H.-T. Chung, Critical role of heme [153] A.C. Zenclussen, T. Gentile, R. Margni, G. Kortebani, A. Mazzolli, Asymmetric
oxygenase-1 in Foxp3-mediated immune suppression, Biochem. Biophys. antibodies and pregnancy, Am. J. Reprod. Immunol. 45 (5) (2001) 289294.
Res. Commun. 327 (4) (2005) 10661071. [154] T.W. LeBien, T.F. Tedder, B lymphocytes: how they develop and function,
[127] A.C. Zenclussen, S. Fest, P. Busse, R. Joachim, B.F. Klapp, P.C. Arck, Questioning Blood 112 (5) (2008) 15701580.
the Th1/Th2 paradigm in reproduction: peripheral levels of IL-12 are down- [155] C. Mauri, A. Bosma, Immune regulatory function of B cells, Annu. Rev.
regulated in miscarriage patients, Am. J. Reprod. Immunol. 48 (4) (2002) Immunol. 30 (2012) 221241.
245251. [156] S. Fillatreau, C.H. Sweenie, M.J. McGeachy, D. Gray, S.M. Anderton, B cells
[128] A. Sollwedel, A.Z. Bertoja, M.L. Zenclussen, K. Gerlof, U. Lisewski, P. Wafula, B. regulate autoimmunity by provision of IL-10, Nat. Immunol. 3 (10) (2002)
Sawitzki, C. Woiciechowsky, H.-D. Volk, A.C. Zenclussen, Protection from 944950.
abortion by heme oxygenase-1 up-regulation is associated with increased [157] M. Horikawa, V. Minard-Colin, T. Matsushita, T.F. Tedder, Regulatory B cell
levels of Bag-1 and neuropilin-1 at the fetal-maternal interface, J. Immunol. production of IL-10 inhibits lymphoma depletion during CD20
175 (8) (2005) 48754885. immunotherapy in mice, J. Clin. Invest. 121 (11) (2011) 42684280.
[129] L. Mincheva-Nilsson, Pregnancy and gamma/delta T cells: taking on the hard [158] C. Mauri, D. Gray, N. Mushtaq, M. Londei, Prevention of arthritis by
questions, Reprod. Biol. Endocrinol. 1 (1) (2003) 1. interleukin 10producing B cells, J. Exp. Med. 197 (4) (2003) 489501.
[130] E.N. Meeusen, R.J. Bischof, C.S. Lee, Comparative T-cell responses during [159] L. Majlessi, R. Lo-Man, C. Leclerc, Regulatory B and T cells in infections,
pregnancy in large animals and humans, Am. J. Reprod. Immunol. 46 (2) Microbes Infect. 10 (9) (2008) 10301035.
(2001) 169179. [160] A. Schumacher, P.O. Wafula, A. Teles, T. El-Mousleh, N. Linzke, M.L.
[131] J. Szekeres-Bartho, A. Barakonyi, B. Polgar, Z. Faust, T. Palkovics, L. Szereday, Zenclussen, S. Langwisch, K. Heinze, I. Wollenberg, P.A. Casalis, H.-D. Volk,
The role of g/d t cells in Progesterone-Mediated immunomodulation during S. Fest, A.C. Zenclussen, Blockage of heme oxygenase-1 abrogates the
pregnancy: a review, Am. J. Reprod. Immunol. 42 (1) (1999) 4448. protective effect of regulatory t cells on murine pregnancy and promotes the
[132] D.-X. Fan, J. Duan, M.-Q. Li, B. Xu, D.-J. Li, L.-P. Jin, The decidual gamma-delta T maturation of dendritic cells, PLoS One 7 (8) (2012) e42301.
cells up-regulate the biological functions of trophoblasts via IL-10 secretion [161] P. Allavena, L. Piemonti, D. Longoni, S. Bernasconi, A. Stoppacciaro, L. Ruco, A.
in early human pregnancy, Clin. Immunol. 141 (3) (2011) 284292. Mantovani, IL-10 prevents the differentiation of monocytes to dendritic cells
[133] M.-N. Lucia, M. Kling, S. Hammarstrm, O. Nagaeva, K.-G. Sundqvist, M.-L. but promotes their maturation to macrophages, Eur. J. Immunol. 28 (1)
Hammarstrm, V. Baranov, gdT cells of human early pregnancy decidua: (1998) 359369.
evidence for local proliferation, phenotypic heterogeneity, and extrathymic [162] K. Patas, J.B. Engler, M.A. Friese, S.M. Gold, Pregnancy and multiple sclerosis:
differentiation pathway, J. Reprod. Immunol. 34 (1) (1997) 52. feto-maternal immune cross talk and its implications for disease activity, J.
[134] T. Suzuki, K. Hiromatsu, Y. Ando, T. Okamoto, Y. Tomoda, Y. Yoshikai, Reprod. Immunol. 97 (1) (2013) 140146.
Regulatory role of gamma delta T cells in uterine intraepithelial lymphocytes [163] S. Subramanian, M. Yates, A.A. Vandenbark, H. Offner, Oestrogen-mediated
in maternal antifetal immune response, J. Immunol. 154 (9) (1995) 4476 protection of experimental autoimmune encephalomyelitis in the absence of
4484. Foxp3+ regulatory T cells implicates compensatory pathways including
[135] P.C. Arck, D.A. Ferrick, D. Steele-Norwood, K. Croitoru, D.A. Clark, Regulation regulatory B cells, Immunology 132 (3) (2011) 340347.
of abortion by glT cells, Am. J. Reprod. Immunol. 37 (1) (1997) 8793. [164] M. Kala, S.N. Rhodes, W.-H. Piao, F.-D. Shi, D.I. Campagnolo, T.L. Vollmer, B
[136] P.C. Arck, D.A. Ferrick, D. Steele-Norwood, K. Croitoru, D.A. Clark, Murine T cell cells from glatiramer acetate-treated mice suppress experimental
determination of pregnancy outcome: i. Effects of strain, ab T cell receptor, autoimmune encephalomyelitis, Exp. Neurol. 221 (1) (2010) 136145.
gd T cell receptor, and gd T cell subsets, Am. J. Reprod. Immunol. 37 (6) (1997) [165] T. Matsushita, K. Yanaba, J.-D. Bouaziz, M. Fujimoto, T.F. Tedder, Regulatory B
492502. cells inhibit EAE initiation in mice while other B cells promote disease
[137] A. Hanninen, P.C. Harrison, gd T cells as mediators of mucosal tolerance: the progression, J. Clin. Invest. 118 (10) (2008) 34203430.
autoimmune diabetes model, Immunol. Rev. 173 (1) (2000) 109119. [166] D.A. Gonzlez, B.B. Daz, M.d.C.R. Prez, A.G. Hernndez, B.N.D. Chico, A.C. de
[138] Z.W. Chen, Comparative biology of gd T cells, Sci. Prog. 85 (4) (2002) 347358. Len, Sex hormones and autoimmunity, Immunol. Lett. 133 (1) (2010) 613.
[139] J. Szekeres-Bartho, A. Barakonyi, E. Miko, B. Polgar, T. Palkovics, The role of [167] C.M. Grimaldi, D.J. Michael, B. Diamond, Cutting edge: expansion and
g/dT cells in the feto-maternal relationship, Seminars in Immunology, activation of a population of autoreactive marginal zone B cells in a model of
Elsevier, 2001, pp. 229233. estrogen-induced lupus, J. Immunol. 167 (4) (2001) 18861890.
[140] A.J. Nahmias, J. Schollin, C. Abramowsky, Evolutionarydevelopmental [168] P.W. Kincade, K.L. Medina, K.J. Payne, M.I.D. Rossi, K.S.R. Tudor, Y. Yamashita,
perspectives on immune system interactions among the pregnant woman, T. Xx, Early B-lymphocyte precursors and their regulation by sex steroids,
placenta, and fetus, and responses to sexually transmitted infectious agents, Immunol. Rev. 175 (1) (2000) 128137.
Ann. N. Y. Acad. Sci. 1230 (1) (2011) 2547. [169] A. Canellada, S. Blois, T. Gentile, R.A. Margni Idehu, In vitro modulation of
[141] J.G. Cannon, Inammatory cytokines in nonpathological states, Physiology 15 protective antibody responses by estrogen, progesterone and interleukin-6,
(6) (2000) 298303. Am. J. Reprod. Immunol. 48 (5) (2002) 334343.
[142] G. Chaouat, A.A. Meliani, J. Martal, R. Raghupathy, J. Elliott, J. Elliot, T. [170] D. Kyurkchiev, E. Ivanova-Todorova, S.D. Kyurkchiev, New target cells of the
Mosmann, T. Wegmann, IL-10 prevents naturally occurring fetal loss in the immunomodulatory effects of progesterone, Reprod. Biomed. Online 21 (3)
CBA x DBA/2 mating combination, and local defect in IL-10 production in this (2010) 304311.
abortion-prone combination is corrected by in vivo injection of IFN-tau, J. [171] M. Yates, Y. Li, P. Chlebeck, T. Proctor, A. Vandenbark, H. Offner, Progesterone
Immunol. 154 (9) (1995) 42614268. treatment reduces disease severity and increases IL-10 in experimental
[143] H. Lin, T.R. Mosmann, L.e.a. Guilbert, S. TuntiPopipat, T.G. Wegmann, autoimmune encephalomyelitis, J. Neuroimmunol. 220 (1) (2010) 136139.
Synthesis of T helper 2-type cytokines at the maternal-fetal interface, J. [172] P.A. Carpentier, A.L. Dingman, T.D. Palmer, Placental TNF-a signaling in
Immunol. 151 (9) (1993) 45624573. illness-induced complications of pregnancy, Am. J. Pathol. 178 (6) (2011)
[144] I. Roth, D.B. Corry, R.M. Locksley, J.S. Abrams, M.J. Litton, S.J. Fisher, Human 28022810.
placental cytotrophoblasts produce the immunosuppressive cytokine [173] P.A. Blair, K.A. Chavez-Rueda, J.G. Evans, M.J. Shlomchik, A. Eddaoudi, D.A.
interleukin 10, J. Exp. Med. 184 (2) (1996) 539548. Isenberg, M.R. Ehrenstein, C. Mauri, Selective targeting of B cells with
[145] L. Svensson, M. Arvola, M.-A. Sllstrm, R. Holmdahl, R. Mattsson, The Th2 agonistic anti-CD40 is an efcacious strategy for the generation of induced
cytokines IL-4 and IL-10 are not crucial for the completion of allogeneic regulatory T2-like B cells and for the suppression of lupus in MRL/lpr mice, J.
pregnancy in mice, J. Reprod. Immunol. 51 (1) (2001) 37. Immunol. 182 (6) (2009) 34923502.
[146] F. Jensen, D. Muzzio, R. Soldati, S. Fest, A.C. Zenclussen, Regulatory B10 cells [174] Y. Yazdani, M. Mohammadnia-Afrouzi, M. Youse, E. Anvari, G. Ghalamfarsa,
restore pregnancy tolerance in a mouse model, Biol. Reprod. 89 (4) (2013) 90. H. Hasannia, S. Sadreddini, F. Jadidi-Niaragh, Myeloid-derived suppressor
[147] D.J. DiLillo, T. Matsushita, T.F. Tedder, B10 cells and regulatory B cells balance cells in B cell malignancies, Tumor Biol. 36 (10) (2015) 73397353.
immune responses during inammation, autoimmunity, and cancer, Ann. N. [175] F. Jadidi-Niaragh, M. Jeddi-Tehrani, B. Ansaripour, S.M. Razavi, R.A. Sharian,
Y. Acad. Sci. 1183 (1) (2010) 3857. F. Shokri, Reduced frequency of NKT-like cells in patients with progressive
[148] K. Yanaba, J.-D. Bouaziz, S. Sato, T.F. Tedder, A regulatory B cell subset with a chronic lymphocytic leukemia, Med. Oncol. 29 (5) (2012) 35613569.
unique CD1dhiCD5+ phenotype controls T cell-dependent inammatory [176] Y. Jiang, X. Cui, C. Cui, J. Zhang, F. Zhou, Z. Zhang, Y. Fu, J. Xu, Z. Chu, J. Liu, The
responses (97.1), J. Immunol. 182 (1 Suppl) (2009) 97 197.1. function of CD3, BioMed Res. Int. 2014 (2014) 19.
[149] A. Yoshizaki, T. Miyagaki, D.J. DiLillo, T. Matsushita, M. Horikawa, E.I. [177] G. Ghalamfarsa, A. Hadinia, M. Youse, F. Jadidi-Niaragh, The role of natural
Kountikov, R. Spolski, J.C. Poe, W.J. Leonard, T.F. Tedder, Regulatory B cells killer T cells in B cell malignancies, Tumor Biol. 34 (3) (2013) 13491360.
control T-cell autoimmunity through IL-21-dependent cognate interactions, [178] L. Van Kaer, V.V. Parekh, L. Wu, Invariant natural killer T cells: bridging innate
Nature 491 (7423) (2012) 264268. and adaptive immunity, Cell Tissue Res. 343 (1) (2011) 4355.
[150] J.W. Tung, M.D. Mrazek, Y. Yang, L.A. Herzenberg, L.A. Herzenberg, [179] D.I. Godfrey, M. Kronenberg, Going both ways: immune regulation via CD1d-
Phenotypically distinct B cell development pathways map to the three B dependent NKT cells, J. Clin. Invest. 114 (10) (2004) 13791388.
cell lineages in the mouse, Proc. Natl. Acad. Sci. 103 (16) (2006) 62936298. [180] W. Sun, P.B. Subrahmanyam, J.E. East, T.J. Webb, Connecting the dots: articial
[151] L. Rolle, M. Memarzadeh Tehran, A. Morell-Garca, Y. Raeva, A. Schumacher, R. antigen presenting cell-mediated modulation of natural killer T cells, J.
Hartig, S.D. Costa, F. Jensen, A.C. Zenclussen, Cutting Edge: IL-10-producing Interferon Cytokine Res. 32 (11) (2012) 505516.
regulatory B cells in early human pregnancy, Am. J. Reprod. Immunol. 70 (6) [181] J.E. Boyson, B. Rybalov, L.A. Koopman, M. Exley, S.P. Balk, F.K. Racke, F. Schatz,
(2013) 448453. R. Masch, S.B. Wilson, J.L. Strominger, CD1d and invariant NKT cells at the
[152] A.C. Eblen, C. Gercel-Taylor, L.B. Shields, J.S. Sanlippo, S.T. Nakajima, D.D. human maternalfetal interface, Proc. Natl. Acad. Sci. 99 (21) (2002) 13741
Taylor, Alterations in humoral immune responses associated with recurrent 13746.
pregnancy loss, Fertil. Steril. 73 (2) (2000) 305313.
72 M. Ghaebi et al. / Biomedicine & Pharmacotherapy 88 (2017) 6173

[182] K. Ito, M. Karasawa, T. Kawano, T. Akasaka, H. Koseki, Y. Akutsu, E. Kondo, S. [208] T. Condamine, I. Ramachandran, J.-I. Youn, D.I. Gabrilovich, Regulation of
Sekiya, K. Sekikawa, M. Harada, Involvement of decidual V(14 NKT cells in tumor metastasis by myeloid-derived suppressor cells, Annu. Rev. Med. 66
abortion, Proc. Natl. Acad. Sci. 97 (2) (2000) 740744. (2015) 97.
[183] H. Tsuda, M. Sakai, T. Michimata, K. Tanebe, S. Saito, S. Hayakawa, [209] P. Serani, Myeloid derived suppressor cells in physiological and pathological
Characterization of NKT cells in human peripheral blood and decidual conditions: the good, the bad, and the ugly, Immunol. Res. 57 (13) (2013)
lymphocytes, Am. J. Reprod. Immunol. 45 (5) (2001) 295302. 172184.
[184] E. Miko, Z. Manfai, M. Meggyes, A. Barakonyi, F. Wilhelm, A. Varnagy, J. Bodis, [210] O. Fainaru, G. Paz, S. Hantisteanu, M. Hallak, Myeloid cell alterations in the
Z. Illes, J. Szekeres-Bartho, L. Szereday, Possible role of natural killer and mouse placenta precede the onset of labor and delivery, Am. J. Obstet.
natural killer T-like cells in implantation failure after IVF, Reprod. Biomed. Gynecol. 210 (1) (2014) 73 e1-73. e7.
Online 21 (6) (2010) 750756. [211] H. Zhao, F. Kalish, S. Schulz, Y. Yang, R.J. Wong, D.K. Stevenson, Unique roles of
[185] Y. Uemura, M. Suzuki, T.-Y. Liu, Y. Narita, S. Hirata, H. Ohyama, O. Ishihara, S. inltrating myeloid cells in the murine uterus during early to midpregnancy,
Matsushita, Role of human non-invariant NKT lymphocytes in the J. Immunol. 194 (8) (2015) 37133722.
maintenance of type 2 T helper environment during pregnancy, Int. [212] N. Kstlin, H. Kugel, B. Spring, A. Leiber, A. Marm, M. Henes, N. Rieber, D.
Immunol. 20 (3) (2008) 405412. Hartl, C.F. Poets, C. Gille, Granulocytic myeloid derived suppressor cells
[186] M. van den Heuvel, C. Peralta, K. Hatta, V. Han, D. Clark, Decline in number of expand in human pregnancy and modulate T-cell responses, Eur. J. Immunol.
elevated blood CD3 (+) CD56 (+) NKT cells in response to intravenous 44 (9) (2014) 25822591.
immunoglobulin treatment correlates with successful pregnancy (vol 58, pg [213] R.R. Nair, P. Sinha, A. Khanna, K. Singh, Reduced myeloid-derived suppressor
447, 2007), Am. J. Reprod. Immunol. 58 (6) (2007) 547. cells in the blood and endometrium is associated with early miscarriage, Am.
[187] S. Wang, C. Li, H. Kawamura, H. Watanabe, T. Abo, Unique sensitivity to J. Reprod. Immunol. 73 (6) (2015) 479486.
a-galactosylceramide of NKT cells in the uterus, Cell. Immunol. 215 (1) [214] R.G. Spallanzani, T. Dalotto-Moreno, X.L.R. Iraolagoitia, A. Ziblat, C.I. Domaica,
(2002) 98105. D.E. Avila, L.E. Rossi, M.B. Fuertes, M.A. Battistone, G.A. Rabinovich, Expansion
[188] J. Yuan, J. Li, S.-Y. Huang, X. Sun, Characterization of the subsets of human of CD11b+ Ly6G+ Ly6Cint cells driven by medroxyprogesterone acetate in
NKT-like cells and the expression of Th1/Th2 cytokines in patients with mice bearing breast tumors restrains NK cell effector functions, Cancer
unexplained recurrent spontaneous abortion, J. Reprod. Immunol. 110 (2015) Immunol. Immunother. 62 (12) (2013) 17811795.
8188. [215] T. Pan, L. Zhong, S. Wu, Y. Cao, Q. Yang, Z. Cai, X. Cai, W. Zhao, N. Ma, W. Zhang,
[189] M.J. Van Den Heuvel, C.G. Peralta, K. Hatta, V.K. Han, D.A. Clark, Decline in 17b-Oestradiol enhances the expansion and activation of myeloid-derived
number of elevated blood CD3+ CD56+ NKT cells in response to intravenous suppressor cells via signal transducer and activator of transcription (STAT)- 3
immunoglobulin treatment correlates with successful pregnancy, Am. J. signalling in human pregnancy, Clin. Exp. Immunol. 185 (1) (2016) 8697.
Reprod. Immunol. 58 (5) (2007) 447459. [216] L.A. Mauti, M.-A. Le Bitoux, K. Baumer, J.-C. Stehle, D. Golshayan, P. Provero, I.
[190] E. Tupin, Y. Kinjo, M. Kronenberg, The unique role of natural killer T cells in Stamenkovic, Myeloid-derived suppressor cells are implicated in regulating
the response to microorganisms, Nat. Rev. Microbiol. 5 (6) (2007) 405417. permissiveness for tumor metastasis during mouse gestation, J. Clin. Invest.
[191] M. Taniguchi, T. Nakayama, Recognition and Function of Va14 NKT Cells, 121 (7) (2011) 27942807.
Seminars in Immunology, Elsevier, 2000, pp. 543550. [217] A. Mazzoni, V. Bronte, A. Visintin, J.H. Spitzer, E. Apolloni, P. Serani, P.
[192] N.Y. Smyth, D.G. Crowe, K. Pellicci, J.M. Kyparissoudis, K. Kelly, H. Takeda, Zanovello, D.M. Segal, Myeloid suppressor lines inhibit T cell responses by an
Sequential production of interferon-g by NK1. 1+ T cells and natural killer NO-dependent mechanism, J. Immunol. 168 (2) (2002) 689695.
cells is essential for the antimetastatic effect of a-galactosylceramide, Blood [218] P. Filipazzi, V. Huber, L. Rivoltini, Phenotype, function and clinical
99 (4) (2002) 12591266. implications of myeloid-derived suppressor cells in cancer patients,
[193] P.J. Brennan, M. Brigl, M.B. Brenner, Invariant natural killer T cells: an innate Cancer Immunol. Immunother. 61 (2) (2012) 255263.
activation scheme linked to diverse effector functions, Nat. Rev. Immunol. 13 [219] A.J. Montero, C.M. Diaz-Montero, C.E. Kyriakopoulos, V. Bronte, S.
(2) (2013) 101117. Mandruzzato, Myeloid-derived suppressor cells in cancer patients: a
[194] M.A. Exley, Q. He, O. Cheng, R.-J. Wang, C.P. Cheney, S.P. Balk, M.J. Koziel, clinical perspective, J. Immunother. 35 (2) (2012) 107115.
Cutting edge: compartmentalization of Th1-like noninvariant CD1d-reactive [220] A.G. Cuenca, M.J. Delano, K.M. Kelly-Scumpia, C. Moreno, P.O. Scumpia, D.M.
T cells in hepatitis C virus-infected liver, J. Immunol. 168 (4) (2002) 1519 LaFace, P.G. Heyworth, P.A. Efron, L.L. Moldawer, A paradoxical role for
1523. myeloid-derived suppressor cells in sepsis and trauma, Mol. Med. 17 (34)
[195] M.A. Exley, S.M.A. Tahir, O. Cheng, A. Shaulov, R. Joyce, D. Avigan, R. Sackstein, (2011) 281.
S.P. Balk, Cutting edge: a major fraction of human bone marrow lymphocytes [221] J. Dai, M. El Gazzar, G.Y. Li, J.P. Moorman, Z.Q. Yao, Myeloid-derived
are Th2-like CD1d-reactive T cells that can suppress mixed lymphocyte suppressor cells: paradoxical roles in infection and immunity, J. Innate
responses, J. Immunol. 167 (10) (2001) 55315534. Immun. 7 (2) (2014) 116126.
[196] Y. Dang, J. Beckers, C.R. Wang, K. Heyborne, Natural killer 1.1+ ab T cells in the [222] T. Condamine, D.I. Gabrilovich, Molecular mechanisms regulating myeloid-
periimplantation uterus, Immunology 101 (4) (2000) 484491. derived suppressor cell differentiation and function, Trends Immunol. 32 (1)
[197] M.-P. Piccinni, M.-G. Giudizi, R. Biagiotti, L. Beloni, L. Giannarini, S. (2011) 1925.
Sampognaro, P. Parronchi, R. Manetti, F. Annunziato, C. Livi, Progesterone [223] M. Fletcher, M.E. Ramirez, R.A. Sierra, P. Raber, P. Thevenot, A.A. Al-Khami, D.
favors the development of human T helper cells producing Th2-type Sanchez-Pino, C. Hernandez, D.D. Wyczechowska, A.C. Ochoa, l-Arginine
cytokines and promotes both IL-4 production and membrane CD30 depletion blunts antitumor T-cell responses by inducing myeloid-derived
expression in established Th1 cell clones, J. Immunol. 155 (1) (1995) 128133. suppressor cells, Cancer Res. 75 (2) (2015) 275283.
[198] T. Takahashi, M. Nieda, Y. Koezuka, A. Nicol, S.A. Porcelli, Y. Ishikawa, K. [224] P.C. Rodrguez, A.C. Ochoa, Arginine regulation by myeloid derived
Tadokoro, H. Hirai, T. Juji, Analysis of human Va24+ CD4+ NKT cells activated suppressor cells and tolerance in cancer: mechanisms and therapeutic
by a-glycosylceramide-pulsed monocyte-derived dendritic cells, J. Immunol. perspectives, Immunol. Rev. 222 (1) (2008) 180191.
164 (9) (2000) 44584464. [225] P.C. Rodriguez, D.G. Quiceno, A.C. Ochoa, L-arginine availability regulates T-
[199] J.E. Boyson, N. Nagarkatti, L. Nizam, M.A. Exley, J.L. Strominger, Gestation lymphocyte cell-cycle progression, Blood 109 (4) (2007) 15681573.
stage-dependent mechanisms of invariant natural killer T cell-mediated [226] J. Sengupta, L. Dhawan, P. Lalitkumar, D. Ghosh, Nitric oxide in blastocyst
pregnancy loss, Proc. Natl. Acad. Sci. U. S. A. 103 (12) (2006) 45804585. implantation in the rhesus monkey, Reproduction 130 (3) (2005) 321332.
[200] L. Li, J. Yang, Y. Jiang, J. Tu, D.J. Schust, Activation of decidual invariant natural [227] E. Haddad, A. Duclos, M. Baines, Early embryo loss is associated with local
killer T cells promotes lipopolysaccharide-induced preterm birth, Mol. Hum. production of nitric oxide by decidual mononuclear cells, J. Exp. Med. 182 (4)
Reprod. 21 (4) (2015) 369381. (1995) 11431151.
[201] L.-P. Li, Y.-C. Fang, G.-F. Dong, Y. Lin, S. Saito, Depletion of invariant NKT cells [228] R.M. Bingisser, P.A. Tilbrook, P.G. Holt, U.R. Kees, Macrophage-derived nitric
reduces inammation-induced preterm delivery in mice, J. Immunol. 188 (9) oxide regulates T cell activation via reversible disruption of the Jak3/STAT5
(2012) 46814689. signaling pathway, J. Immunol. 160 (12) (1998) 57295734.
[202] M. Motallebnezhad, F. Jadidi-Niaragh, E.S. Qamsari, S. Bagheri, T. Gharibi, M. [229] O. Harari, J.K. Liao, Inhibition of MHC II gene transcription by nitric oxide and
Youse, The immunobiology of myeloid-derived suppressor cells in cancer, antioxidants, Curr. Pharm. Des. 10 (8) (2004) 893898.
Tumor Biol. 37 (2) (2016) 13871406. [230] J.B. Mannick, A. Hausladen, L. Liu, D.T. Hess, M. Zeng, Q.X. Miao, L.S. Kane, A.J.
[203] A.-M. Zhao, H.-J. Xu, X.-M. Kang, L.-M. Lu, New insights into myeloid-derived Gow, J.S. Stamler, Fas-induced caspase denitrosylation, Science 284 (5414)
suppressor cells and their roles in feto-maternal immune cross-talk, J. (1999) 651654.
Reprod. Immunol. 113 (2016) 3541. [231] L. Rivoltini, M. Carrabba, V. Huber, C. Castelli, L. Novellino, P. Dalerba, R.
[204] D.I. Gabrilovich, S. Nagaraj, Myeloid-derived suppressor cells as regulators of Mortarini, G. Arancia, A. Anichini, S. Fais, Immunity to cancer: attack and
the immune system, Nat. Rev. Immunol. 9 (3) (2009) 162174. escape in T lymphocytetumor cell interaction, Immunol. Rev. 188 (1) (2002)
[205] S. Gantt, A. Gervassi, H. Jaspan, H. Horton, The role of myeloid-derived 97113.
suppressor cells in immune ontogeny, Front. Immunol. 5 (2014) 387. [232] H. Li, Y. Han, Q. Guo, M. Zhang, X. Cao, Cancer-expanded myeloid-derived
[206] A.R. Arocena, L.I. Onofrio, A.V. Pellegrini, A.E. Carrera Silva, A., Paroli, R.C., suppressor cells induce anergy of NK cells through membrane-bound TGF-
Cano, M.P., Aoki, S. Gea, Myeloid-derived suppressor cells are key players in b1, J. Immunol. 182 (1) (2009) 240249.
the resolution of inammation during a model of acute infection, Eur. J. [233] N. Dilek, N. Poirier, C. Usal, B. Martinet, G. Blancho, B. Vanhove, Control of
Immunol. 44 (1) (2014) 184194. transplant tolerance and intragraft regulatory T cell localization by myeloid-
[207] C. Bartmann, M. Junker, S.E. Segerer, S.F. Husler, M. Krockenberger, U. derived suppressor cells and CCL5, J. Immunol. 188 (9) (2012) 42094216.
Kmmerer, CD33+/HLA-DRneg and CD33+/HLA-DR+/ cells: rare [234] E. Schlecker, A. Stojanovic, C. Eisen, C. Quack, C.S. Falk, V. Umansky, A.
populations in the human decidua with characteristics of MDSC, Am. J. Cerwenka, Tumor-inltrating monocytic myeloid-derived suppressor cells
Reprod. Immunol. 75 (5) (2016) 539556.
M. Ghaebi et al. / Biomedicine & Pharmacotherapy 88 (2017) 6173 73

mediate CCR5-dependent recruitment of regulatory T cells favoring tumor [239] L. Gu, S. Tseng, R.M. Horner, C. Tam, M. Loda, B.J. Rollins, Control of TH2
growth, J. Immunol. 189 (12) (2012) 56025611. polarization by the chemokine monocyte chemoattractant protein-1, Nature
[235] E.M. Hanson, V.K. Clements, P. Sinha, D. Ilkovitch, S. Ostrand-Rosenberg, 404 (6776) (2000) 407411.
Myeloid-derived suppressor cells down-regulate L-selectin expression on [240] X. Xu, Q. Wang, B. Deng, H. Wang, Z. Dong, X. Qu, B. Kong, Monocyte
CD4+ and CD8+ T cells, J. Immunol. 183 (2) (2009) 937944. chemoattractant protein-1 secreted by decidual stromal cells inhibits NK
[236] V. De Wilde, N. Van Rompaey, M. Hill, J.-F. Lebrun, P. Lemaitre, F. Lhomm, C. cells cytotoxicity by up-regulating expression of SOCS3, PLoS One 7 (7) (2012)
Kubjak, B. Vokaer, G. Oldenhove, L.-M. Charbonnier, Endotoxin-Induced e41869.
myeloid-Derived suppressor cells inhibit alloimmune responses via heme [241] H. Tsuda, T. Michimata, S. Hayakawa, K. Tanebe, M. Sakai, M. Fujimura, K.
oxygenase-1, Am. J. Transplant. 9 (9) (2009) 20342047. Matsushima, S. Saito, A th2 chemokine, TARC, produced by trophoblasts and
[237] M.Z. Noman, G. Desantis, B. Janji, M. Hasmim, S. Karray, P. Dessen, V. Bronte, S. endometrial gland cells, regulates the inltration of CCR4+ t lymphocytes
Chouaib, PD-L1 is a novel direct target of HIF-1a, and its blockade under into human decidua at early pregnancy, Am. J. Reprod. Immunol. 48 (1)
hypoxia enhanced MDSC-mediated T cell activation, J. Exp. Med. 211 (5) (2002) 18.
(2014) 781790. [242] S. Engert, L. Rieger, M. Kapp, J.C. Becker, J. Dietl, U. Kmmerer, Proling
[238] S.L. Deshmane, S. Kremlev, S. Amini, B.E. Sawaya, Monocyte chemoattractant chemokines, cytokines and growth factors in human early pregnancy
protein-1 (MCP-1): an overview, J. Interferon Cytokine Res. 29 (6) (2009) decidua by protein array, Am. J. Reprod. Immunol. 58 (2) (2007) 129137.
313326.

Anda mungkin juga menyukai