Next Article in Journal
FGF2 Dual Warhead Conjugate with Monomethyl Auristatin E and α-Amanitin Displays a Cytotoxic Effect towards Cancer Cells Overproducing FGF Receptor 1
Next Article in Special Issue
Trans-Ethnic Mapping of BANK1 Identifies Two Independent SLE-Risk Linkage Groups Enriched for Co-Transcriptional Splicing Marks
Previous Article in Journal
Comparison of the Effect of Melatonin Treatment before and after Brain Ischemic Injury in the Inflammatory and Apoptotic Response in Aged Rats
Previous Article in Special Issue
Generation of Human Breg-Like Phenotype with Regulatory Function In Vitro with Bacteria-Derived Oligodeoxynucleotides
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

B Regulatory Cells: Players in Pregnancy and Early Life

1
Functional Unit of Clinical Immunology and Primary Immunodeficiencies, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, University of Barcelona, Pediatric Research Institute Sant Joan de Déu, 08950 Barcelona, Spain
2
Functional Unit of Clinical Immunology, Hospital Sant Joan de Déu-Hospital Clínic de Barcelona, Barcelona, Spain
3
Immunology Service, Biomedic Diagnostic Center, Hospital Clínic de Barcelona, Universitat de Barcelona, IDIBAPS, 08036 Barcelona, Spain
4
Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, University of Barcelona, Pediatric Research Institute Sant Joan de Déu, 08950 Barcelona, Spain
*
Authors to whom correspondence should be addressed.
Both authors share co-senior author status.
Int. J. Mol. Sci. 2018, 19(7), 2099; https://doi.org/10.3390/ijms19072099
Submission received: 25 June 2018 / Revised: 13 July 2018 / Accepted: 16 July 2018 / Published: 19 July 2018
(This article belongs to the Special Issue B Cells and Immunological Tolerance)

Abstract

:
Pregnancy and early infancy represent two very particular immunological states. During pregnancy, the haploidentical fetus and the pregnant women develop tolerance mechanisms to avoid rejection; then, just after birth, the neonatal immune system must modulate the transition from the virtually sterile but haploidentical uterus to a world full of antigens and the rapid microbial colonization of the mucosa. B regulatory (Breg) cells are a recently discovered B cell subset thought to play a pivotal role in different conditions such as chronic infections, autoimmunity, cancer, and transplantation among others in addition to pregnancy. This review focuses on the role of Breg cells in pregnancy and early infancy, two special stages of life in which recent studies have positioned Breg cells as important players.

Graphical Abstract

1. The Immune System during Pregnancy and the Neonatal Period

Pregnancy and the neonatal period constitute two special immunological states both for the pregnant women and for the conceptus. On one hand, the mother needs to tolerate the semi-allogenic cells growing in-utero carrying paternal antigens while at the same time needing to be able to deal with infectious microorganisms. As such, it has been proposed that “mammalian pregnancy defies the immune system rules” [1]. Furthermore, the fetus also needs to tolerate the allogenic mother and in particular must transition from this semi-allogenic but virtually sterile maternal uterus to a world full of antigens, including a massive mucosal colonization occurring immediately after birth. Thus, the newborn must develop different strategies to overcome these challenges, including the avoidance of excessive responses, which is critical.

1.1. The Immune System during Pregnancy

Pregnancy is not a state of general immunosuppression but rather of local and systemic immune modulation, during which pregnant women have shown increased susceptibility to influenza, measles, hepatitis E, and Herpes Simplex Virus [2]. Tight immune system regulation and modulation is needed, as both inflammatory and anti-inflammatory signals are needed for successful pregnancies [3,4]. In fact, immune-related causes are thought to explain some of the current ‘idiopathic’ causes of infertility. As a result, women with problems in the immune system have greater probabilities of poor pregnancy outcomes [4]. The diverse combination of hormones throughout pregnancy is crucial, since it modulates the recruitment and limits the presence of different innate and adaptive cell types in the maternal-fetal interphase. Concretely, estradiol, progesterone, and human gonadotropic hormone are the main hormonal regulation players, inhibiting destructive immune responses and inducing tolerance-promoting pathways, such as the human gonadotropic hormone-mediated decrease of the cytotoxic capacity of peripheral natural killer cells in pregnant women [1,5,6].

1.1.1. Natural Killer Cells

Natural killer (NK) cells play a critical role during pregnancy, protecting both the mother and the conceptus [3,4,7,8,9]. NK cells represent a high proportion of the decidual leukocytes, and there is an increase of uterine NK cells, characterized by a CD56brightCD16 phenotype [10]. Cells with this phenotype have cytotoxic capacities in other tissues, but in the decidua they have pregnancy establishment functions [3,7,11,12]. Concretely, NK cells have an increased relative expression of inhibitory receptors and play an important role in the remodeling of decidual blood vessels [4,13,14] and in trophoblastic migration and invasion [3,13,15]. Furthermore, uterine NK cells interact with human leukocyte antigen (HLA)-G and HLA-C, causing their inhibition [3,8,12]. This is of special interest since the poorly polymorphic non-classical HLA-G is expressed in the extra-villous trophoblast [8].

1.1.2. Myeloid-Derived Cells

Macrophages, dendritic cells (DC), and mast cells play a role in implantation and tissue remodeling. Macrophage population is also regulated during the different phases of pregnancy [1,16,17,18]. Concordantly, there is an increased frequency of M2 macrophages, associated with tissue healing and homeostasis in the decidua [7,16,17,18] in relation to tissue remodeling and angiogenesis [5]. Macrophages are also capable of regulating hormonal levels, as they degrade excessive human gonadotropic hormone [1] and are also responsible for pathogen clearance in the endometrium, therefore showing pro- and anti-inflammatory functions [3]. Interestingly, hormonal changes alter the presence and phenotype of circulating monocytes, which have a role in assisting the implantation of the blastocyst and in pregnancy termination [4]. In addition, DC are mainly maintained at a more immature phenotype with regulatory functions [5]. Mast cells are present in the maternal-fetal interface and favor implantation, angiogenesis, and tissue remodeling and are in a quiescent state until term [3,4,5]; besides, they have a different phenotype compared with classical peripheral mast cells [5].

1.1.3. T Cells

T cells are highly regulated during pregnancy. There is an increase in induced T regulatory cells (Treg), regulated by both hormonal changes and alloantigenic exposure [3,7,19,20]. In fact, Treg cell levels are modulated mainly but not exclusively by interleukin (IL)-10 during the menstrual cycle, creating the needed locally tolerant microenvironment [4,8,20]. A decrease in Treg cell markers has been reported in patients with idiopathic infertility [3]; what is more, Treg cells in the endometrium are capable of recognizing HLA-C [8]. Regarding effector T cells, the reduced T helper (Th)1/Th2 ratio is controversial. Knock out mice for Th-2 related cytokines suggest that the reduction or increase of this ratio is a marker of successful or failing pregnancy but not causative of the final outcome [4,20]. However, accumulation of Th1 cells specific for paternal antigens was associated with insufficient generation of Treg cells and, therefore, caused abortion in a murine model [4,8,21]. On the other hand, gamma-delta T cells are increased during pregnancy and are hormone-regulated, presenting with inhibitory functions [4,8]. Besides, memory cluster of differentiation (CD)8+ cells have been found in the fetal-maternal interface with regulatory functions, including CD8+FoxP3+ cells [20]. Other maternal-fetal tolerance mechanisms include increased IL-10 and transforming growth factor (TGF)-β expression [22], an increase of the inhibitory molecule Program Death-Ligand 1 (PD-L1) in the trophoblastic tissue, the lack of class I and II Major Histocompatibility Complex (MHC) expression required for T cell activation by syncytiotrophoblast cells at the major maternal-conceptus interface [8], and, as reported recently, an increase of B regulatory (Breg) cells [23,24].

1.2. The Immune System during the Neonatal Period

Transition from the last stages of pregnancy, through labor, and into the first encounters of the newborn with the real-world requires rapid changes and adaptation of the immune system: from cell autonomous and innate immunity to the adaptive immune system. Although the neonate’s immune system can fight pathogen infections, as a result of the need of tolerance, there is an increased risk for severe infections [25]. During pregnancy, the fetal immune system is active, since the fetus actively generates tolerance to maternal antigens, including specific Treg cells [20]. Immunization through maternal immunoglobulin (Ig)G during late pregnancy is critical, since IgG transfer can confer around three months of broad protection for a variety of infectious diseases including measles, mumps, rubella, and varicella. Altered IgG transfer, observed in very low weight and preterm infants, results in an increased risk of lethal neonatal infection.
The immune system of the newborn has different regulatory mechanisms to promote tolerance. For example, hypoxia during labor can cause tissue damage-enhancing inflammation, making the strong immune bias towards resolution of inflammation and healing very important [26]. Upon Toll-like receptor (TLR)-mediated activation, antigen presenting cells promote Treg cell differentiation; besides, non-inherited maternal antigens challenge also results in CD4+ T cell differentiation into Treg cells. Along with Treg cells, myeloid-derived suppressor cells and erythroid suppressor cells are present in cord blood and regulate CD4+ T cells, CD8+ T cells and NK cell activity. What is more, high adenosine levels in blood after inflammatory events and hypoxic states contribute to the promotion of a tolerogenic state of the immune system through the impairment of MyD88 pathway activation, thus reducing TLR-mediated Th1-polarizing cytokine induction and neutrophil activation [25,27,28,29].
The immune system in the neonatal period shows the following characteristics related to the fact that it is markedly modulated to avoid excessive responses [25,27,28,29,30]: (1) decreased complement system function [31,32]; (2) decreased neutrophil quantity and functions, including respiratory burst [33,34,35,36]; (3) mononuclear presenting cells (monocytes, macrophages, and dendritic cells) have decreased IL-1β, tumor necrosis factor (TNF)-α, and IL-12p70 production, along with normal IL-23 and IL-6 production [37,38,39,40]; (4) decreased Interferon (IFN)-γ production in certain conditions [31,32,41]; (5) interferon responding factor 3 has decreased DNA binding capacity [42]; (6) monocytes produce increased levels of IL-10 and cyclic adenosine monophosphate [37,43]; (7) conventional DC number is decreased and cells are more immature and produce less inflammatory cytokines [44,45], and (8) NK cells have an increased proportion of inhibitory receptors [46,47].
In the neonate, CD4+ T-helper cells are biased towards Th2-like immunity, with a more anti-inflammatory profile. However, in response to some insults such as Bacille-Calmette Guerin (BCG) vaccination, the newborn can develop adult-like Th1 responses, and the Th2 bias can be reverted. Besides, Th17 cell levels are low because of reduced transcription of the RORC transcription factor gene. There is also impaired T cell signaling by the T cell receptor resulting in decreased transcription of CD40L, IL-12, and IFN-γ-related genes. B cells are mostly naïve with a poor repertoire and diminished B cell receptor activity, resulting in decreased antigen response [25,27,28,29]. Accordingly, newborns have an increased risk for severe invasive infections, specifically intracellular pathogen infections requiring Th1 responses, especially Listeria monocytogenes, Salmonella spp., and Mycobacterium infections [25,27,28].

2. B Cells during Pregnancy and Early Life

The role of B cells during pregnancy and early life has been less studied compared to other subsets of the immune system; however, aberrant B cell numbers and functions have been associated with obstetric complications [48]. B cells have been thought of as mere “antibody-factories” over the years; nowadays, it is known that they have other functions including cytokine production and regulation of T cell responses. B cell development and maturation is a complex and regulated process, initiated at 7- to 8-week gestational age in the fetal liver and continued in the bone marrow after gestational age week 17–18 [49,50,51], leading to different B cell subsets in peripheral blood that include naïve, transitional, marginal zone like B-cells (expressing IgM, IgD, and CD27 in their membrane [49,52]), mature B cells, and plasmablasts [49,50]. During pregnancy, to avoid destructive responses, cellular responses are thought to be diminished and compensated for by increased humoral responses [4,8].

2.1. B Cells during Pregnancy

Maternal antibody production by B cells during pregnancy has been shown to be both protective and harmful. B cells can produce protective antibodies against paternal antigens, such as asymmetric antibodies that bind paternal antigens but do not produce responses against them. These antibodies are increased by progesterone and gonadotropic hormone [5,20,53,54]. In contrast, immunoglobulin production against infectious agents is critical for immune protection of both the mother and the conceptus [48]. However, besides protective antibodies, auto-antibody production can occur after an infection before or during pregnancy, such as anti-phospholipid antibodies; these can be responsible for pregnancy-associated problems. Indeed, pathogenic antibody production and changes in immune parameters are associated with the appearance of pre-eclampsia [54,55]. Pregnancy hormones also regulate B cell population and antibody production during pregnancy [20,48,54]; their response to mitogens and infectious agents is reduced [48]. Fetal trophoblasts positively regulate the generation of IL-10 producing B cells, related to gonadotropic hormone but not to estrogen or progesterone [4,5,8].
Maternal B cells are reduced throughout the course of pregnancy. There is a reduction in maternal pre-pro and immature B cells observed in bone marrow of pregnant mice during gestation while an increase in mature B cells is observed [56,57]. This modification of the B cell compartment is accompanied by an increase in serum IgA, IgM, and IgG3. These observed changes are hormonally driven, but whether by direct effect or by indirect limitation of the availability of IL-7 remains to be deciphered [57]. Related with these observations, alfa fetoprotein at fetal concentrations can induce B cell apoptosis, thus preventing maternal cells from reaching the fetus [53]. In humans, absolute numbers of B cells in peripheral blood are reduced during the third trimester of pregnancy. Of interest, B cells are present in the amniotic fluid in initial phases of pregnancy [58]; additionally, there is an increased frequency of naïve B cells and a reduction in the frequency of transitional and Breg cells. The selective reduction of Breg and transitional B-cell in peripheral blood may be caused by a migration to the uterus, although this has not been confirmed [59].

2.2. B Cells in the Neonatal Period

Neonatal B cells are associated with tolerance and inhibitory mechanisms. It is known that infusion of stem cells from cord blood, rather than adult bone marrow, enables transplantation in patients with increased donor-recipient HLA-mismatch [60], and one of the possible mechanisms explaining this augmented allogenic tolerance is B cell-mediated regulation through Breg cells [61]. Because of maternal antibodies and B cell immaturity, not all vaccines are successful when given at birth, as is the case with oral polio, measles, and rubella vaccination [25,27,28,29]. A few published studies on B cells in the neonate have associated B cells with the Th2 bias: asthmatic mothers of infants with early-allergy had an increase in transitional B cells in the late-pregnancy period, in contrast to non-asthmatic mothers, suggesting that these cells could have a role in the Th1/Th2 bias observed in neonates, which might justify the food allergy [59,62]. B cells [63], and more concretely IL-10 production by B cells [64], have been shown to be important in inflammation in mice. Significantly, murine studies have shown that CD5+ B cells in newborn mice also contribute to a decrease in IL-12p70 production [65]. However, it was not until 2010, thanks to Blair et al., that the first description of the phenotype and function of a human Breg cell subset was published [66].

3. The Breg Subpopulation and Its Role in Health and Disease

Breg cells are a rare B cell subpopulation with regulatory/suppressor functions, and they are one of the peripheral tolerance mechanisms. Breg cells represent less than 10% of total B-cells in circulation and their regulatory activity is mostly but not uniquely performed through IL-10 production. Nevertheless, less than 20% of cells from the different described subsets are IL-10 producers after stimulation [67,68]. There are two theories regarding Breg development: the first states that B cells are a specific lineage with a specific transcription factor that controls the suppressive nature of the cells, while the second suggests that B cells can take on a regulatory phenotype after certain stimuli to suppress inflammation. Inflammation is a potent trigger of Breg cell development and differentiation; Breg cells need a combination of different molecules to become activated, including TLRs, CD40, the B cell receptor, CD80, CD86, and cytokines [67,68].
Currently, there are eight approaches to defining Breg cells in humans [67,68,69,70]; indeed, several markers have been used for the detection and sorting of Breg cells, but as yet, there is no consensus regarding which markers should be used. The different approaches are summarized in Table 1.
The main functions of Breg cells include inhibition of Th1 cells activation, Th17 differentiation and promotion, and maintenance of the Treg cell population [67,68,69]. The major suppressive mechanism for Breg cell function is IL-10 secretion. IL-10 is a suppressor cytokine that can inhibit chemokine and pro-inflammatory cytokine production, thereby inhibiting the effector mechanisms of the immune system. An IL-10 blockade partially inhibits their regulatory function. Although IL-10 is a key player in Breg inhibition of inflammation, other mechanisms have been described. These mechanisms include TGF-β (especially for the differentiation of tolerogenic DCs) and indoleamine 2,3-dioxygenase (IDO) production, cell-to-cell contact by CD80/86 interaction with T cells, PD-L1 inhibition of T follicular helper cells, and CD73-dependent adenosine production [66,68,70,71,75,82,83,84].
The most studied subset of Breg cells is defined by CD24hi and CD38hi expression in B-cells [66,71,85]. Phenotypically, these cells also express IgM, IgD, CD5, CD10, and CD1d [66], resembling transitional B cells [86]. Breg-cells are mainly defined by their regulatory function: Mauri et al. demonstrated that the CD19+CD24hiCD38hi subset is enriched in IL-10 production and can inhibit IFN-γ production [66,87] and block Th1 and Th17 differentiation while maintaining the Treg cell population [71]. Their implication in human immune-related diseases has mostly been studied in autoimmune and allergic diseases [88,89,90,91,92,93,94,95,96,97], persistent infections such as human immunodeficiency virus (HIV) [72], hepatitis B virus (HBV) [98], Mycobacterium tuberculosis [99], cancer [100,101,102,103], transplantation [87,104,105,106,107], and, as demonstrated recently, pregnancy [19,23,24,59,62,88,108].
Viruses, bacteria, helminths [109], and parasites [110] can imbue B cells with regulatory functions. Rapidly after infection, Salmonella and Listeria induce the apparition of IL-10-producing cells in a TLR/MyD88-dependent fashion in mice. Some helminth-derived molecules can directly promote IL-10 stimulation in murine B cells. These B cells can suppress immune responses towards allergens; studies in humans have also shown this bystander regulatory function after helminthic infections. The lack of helminth infections in westernized countries has been proposed as one of the reasons for the increased incidence of allergy and autoimmunity. The role of Breg cells in viral infection has been more studied because of the part they play in HIV infection and in chronic HBV infection [109]. Immature Breg cells inhibit IFN-γ production by CD8+ T cells after HBV virus infection [67,98]. Furthermore, CD24hiCD38hi IL-10-producing cell frequency directly correlates with HIV virus load [111]; furthermore, after in vitro Breg depletion, CD8+ T cell effector function is restored and HIV infected CD4+ cells are cleared in vitro [72].

4. Breg Cells in Pregnancy

Breg cells are believed to promote a stable tolerant immune profile in the local microenvironment. Recently, Guzman-Genuino et al. reviewed how the previous knowledge of the role of Breg cells in autoimmunity and transplantation (promoting tolerance) and cancer (promoting tumor growth) could help in the understanding of the role of Breg cells in the establishment and maintenance of pregnancy, where a semi-allogenic mass of cells grows inside a woman [19]. Immunological changes are needed to avoid allogenic reactions that could lead to miscarriage. Body conditions to allow conception and implantation, as well as changes needed to allow embryonic and fetal growth, are regulated by pregnancy hormones. Pregnancy hormones modify immune responses after conception, including Breg cells (Figure 1). Gonadotropic hormone increase, a CD1dhighCD5+ IL-10-producing Breg cell subset, and PD-L1 expression in B cells protect mice from experimental autoimmune encephalomyelitis [19,23,112,113,114,115,116]. Progesterone promotes Th2-like immune responses with a reduction in pro-inflammatory cytokines and increased IL-10 production that is associated with B cell expansion. Human gonadotropic hormone from pregnant women sera increase IL-10 production by B-cells [23,24].
The association of Breg cells with pregnancy success were first observed in mice. In pregnant mice, the increase in CD5+CD1d+ Breg cells is necessary to avoid immunological abortion. In fact, the transfer of Breg cells to abort-prone mice promotes fetal-maternal tolerance by leading to a Treg cell increase and by maintaining DCs in an immature state [108]. In humans, it was observed that women treated during pregnancy with rituximab, a B cell-depleting antibody, presented a higher rate of first-trimester pregnancy loss [117]. A recent review on the effects of rituximab treatment in patients with autoimmune dysregulation within 6 months of conception revealed that patients had a 12% rate of spontaneous abortion, with 40% reported delivery before 37 weeks, and also 39% of newborns had low B cells counts. On the other hand, at 6 months, B cell counts were normalized and infants did not show any clinical adverse effects. These results should be interpreted with caution due to the coexistent use of other immunomodulatory drugs, the effect of maternal condition, and the need for a longer follow-up of the exposed infants [118], but overall, they suggest the important role of B-cells in pregnancy.
CD24hiCD38hi [66] and CD24hiCD27+ [75] have recently been used for the study of Bregs in pregnant women [23,59]. CD24hiCD27hi cells increase in the first trimester of pregnancy but, as observed with murine Breg cells, this increase does not occur in patients who miscarry (Figure 1). In addition, almost 95% of CD24hiCD27hi B cells express the receptor for human gonadotropic hormone [24]. These data highlight the importance of B-cells, specifically Breg, in the mother’s achievement of immune tolerance during the first stages of pregnancy. The importance of this subset of cells in the fetus and the newborn is still to be determined. It has recently been reported that maternal B cells are necessary for the development of perinatal tolerance after mucosal antigen application in a murine model, to protect progeny from experimental allergic airway inflammation. In fact, in the absence of B cells there is an aggravation of the allergic response in the progeny. This tolerance has been related to forkhead box P3 (FoxP3) antigen priming by the IgG contained in the amniotic fluid. However, these observations were made in a non-physiological model; interpretation needs to be made with caution [119]. Moreover, Breg cells (CD24hiCD38hi) have been shown to be reduced during late pregnancy compared with non-pregnant women [59] (Figure 1).

5. Breg Cells in Early Life

CD24hiCD38hi B cells have recently been shown to be at an increased frequency in cord blood of healthy neonates (Figure 2). This B cell subpopulation showed regulatory capacities with a phenotype similar to adult CD24hiCD38hi cells and were preventive of IFN-γ production by T cells when co-cultured [61,120]. Of note, Breg cell frequency has been associated with IFN-γ production after whole blood BCG-stimulation (Esteve-Solé et al., unpublished data). Increased frequency of CD24hiCD38hi B cells in neonates could be ascribed to the immaturity of the system, these cells being ‘only’ transitional B-cells; however, functional studies have confirmed that neonatal CD24hiCD38hi B cells have regulatory functions. Neonatal CD24hiCD38hi B cells produce IL-10 upon stimulation, having a suppressive activity on IFN-γ and IL-4 production by T cells [61,120]. Furthermore, neonatal CD24hiCD38hi B cells have a similar phenotype compared with adult CD24hiCD38hi Breg cells [66,67,71], with increased IgM and decreased CD27 expression. Since CD27 is a memory marker, the decrease can be explained by their very young age [66]. In addition, the presence of elevated levels of IL-10 producing CD24hiCD38hi cells has been associated with good outcomes in neonatal late-onset sepsis [121].
IL-10 blockade experiments performed in neonatal Breg cells revealed that part of their function can be carried out via IL-10-independent mechanisms [61]. Immune regulation by cord blood B-cells seems to be partially mediated by (i) IL-10 production and (ii) cell-to-cell direct contact (mediated by CD80/CD86), but independent of TGF-β, as has also been observed in adults [61,67]. On the other hand, the increased quantity of IgM per cell in neonatal Breg cells compared both to non-Breg (cells with low expression of CD38 and CD24) cells and adult Breg cells [120] could be a new mechanism of action for their inhibitory function (Figure 2). Naturally occurring IgM anti-leukocyte autoantibodies (IgM-ALA) have a suppressor capacity that can inhibit T cell activation and chemotaxis [122,123]. IgM-ALA antibodies are present at birth [123], and they inhibit proinflammatory cells from producing IFN-γ and IL-17 in response to alloantigens in mice [122]. More research should be done to evaluate whether this is a true regulatory mechanism of neonatal Breg function.
One of the studies on neonatal Breg cells revealed an increase in these cells in the neonatal marginal zone-like B cell subset [120]. Circulating marginal zone B cells are representative of splenic marginal zone B cells; these cells are characterized by a pre-diversified Ig repertoire and by initiating T cell-independent responses through TLRs as activation signals. Their responses are mainly directed against encapsulated bacteria, including commensal microbiota [52,124,125], and they play a role in normal pregnancy development [126]. As TLRs activate Bregs [66,75], the greater proportion of marginal zone-like B cells observed may indicate increased regulatory responses after encapsulated bacterial stimulation, thus explaining lower responses to encapsulated bacteria in infants [30]. This could be of importance in the rapid abrogation of unwanted responses to commensal bacteria. It may be hypothesized that during the first contacts with the extra-uterine environment and the adoption of microbiota, the increased proportion of Breg-cells among marginal zone B cells is one of the mechanisms by which the neonatal immune system protects itself from an exacerbated response to the new range of antigens encountered.
Increased Breg levels in umbilical cord blood have been related to lower rates of graft versus host disease (GvHD) in cord-blood transplanted patients [61,127] (Figure 2). In addition, patients developing GvHD present a decreased frequency of Breg cells [61]. However, cord-blood transplantation has also been associated with an increased morbi-mortality due to infections [127]. We hypothesized that, as CD5+ murine B-cells contribute to the reduced production of IL-12 by antigen presenting cells [128], increased neonatal Breg cell subset contributes to the limited Th1 response observed in neonates inhibiting IFN-γ production. Furthermore, the frequency of Breg cells in the neonate can predict the severity of acute bronchiolitis disease after respiratory syncytial virus, thus showing how neonatal Breg cells can modulate microbial pathogenesis [129]. In our group, we recently observed an inverse correlation between the frequency of Breg cells and IFN-γ secretion after whole blood Bacille-Calmette Guerin (BCG) stimulation (unpublished data). It is known that after BCG challenge in neonates, the major providers of IFN-γ are NK cells instead of T cells [130]; therefore, Breg cells could be, at least in part, responsible for this decreased T cell response. This association, together with the fact that high IFN-γ levels are associated with graft versus host disease development [131,132], deserves further investigation and may have implications in clinical practice, especially in stem cell transplantation from cord blood. For example, in IFN-γ receptor 1 deficiency, where baseline levels of IFN-γ in blood are increased [133,134], stem cell transplantation has been difficult to perform with success [135]. We propose that umbilical cord blood transplantation in these patients could help to reduce IFN-γ levels in blood, thus helping the engraftment (Figure 2).

6. Conclusions

B cells in the hallmark of pregnancy and early life have been understudied. However, in recent years there has been growing interest in this lymphocyte population, especially the recently discovered Breg cell subset. Breg cells seem to be an important player permitting pregnancy establishment. The observation of an expanded Breg cell subset in cord blood opens the door for new research and possible treatments, including the study of new mechanisms of action, such as (i) the possible role of the highly increased levels of IgM in neonatal Breg cells in the inhibitory mechanisms, (ii) the definition of the role of Breg in different conditions of health and disease, such as neonatal infection, autoimmunity, and inflammation, as well as the effect of maternal diseases on the Breg cell population in the neonate, and (iii) the evolution of this subset during the first years of life and its relation to childhood immune-mediated diseases. Also, it is advisable to better characterize the role of Breg cells in cord blood transplantation, where they may act as beneficial actors in achieving tolerance or as detrimental actors in susceptibility to infection.
The establishment of pregnancy, its maintenance, and the peripartum period involve complex states, tightly regulated by intricate relationships among the different cell subsets of the immune system. In light of recent discoveries about the Breg cell subset in these situations, there is a need to consider this subset in the overall scheme. However, since few studies have been carried out, more in-depth research must be done to clarify the role of Breg cells and their interaction with their immune system counterparts to reveal the relationships among them and the other generators of tolerance in pregnancy and early life.

Author Contributions

A.E.-S.: conceived and wrote the manuscript and accepted it in its final form. Y.L., A.D.-M., A.M.P.-M., A.V. and J.Y. critically reviewed the manuscript and accepted it in its final form. Specifically: Y.L. in Section 3; A.D.-M. aspects in Section 4; A.M.P.-M. in Section 2; A.V. in Section 5 and J.Y. in Section 1. L.A.: conceived, critically reviewed the manuscript, specifically all the aspects in sections regarding general aspects of neonatal immune system, and accepted it in its final form. M.J.: conceived, critically reviewed the manuscript, specifically all the aspects in sections regarding Breg cells, and accepted it in its final form.

Funding

This study was supported by the projects PI12/01990, PI15/01094, and PFIS0200 (AC16/00025) to LA, integrated in the Plan Nacional de I+D+I and co-financed by the ISCIII—Subdirección General de Evaluación y Formento de la Investigación Sanitaria and the Fondo Europeo de Desarrollo Regional (FEDER), by “Subvencions per a la Intensificació Facultatius Especialistes” del Departament de Salut de la Generalitat de Catalunya. Programa PERIS 2016-2020 (Referencia: SLT006/17/00199)” and by a 2017 Leonardo Grant for Researchers and Cultural Creators, BBVA Foundation to LA.

Conflicts of Interest

The authors declare no conflict of interest. The founding sponsors had no role in the design of the study, in the collection, analyses, or interpretation of data, in the writing of the manuscript, or in the decision to publish the results.

Abbreviations

BCGBacille-Calmette Guerin
BregB regulatory
CDCluster of diferntiation
DCDendritic cell
FoxP3Forkhead box P3
GvHDGraft versus host disease
GZMBGranzyme B
HBVHepatitis B virus
HIVHuman immunodeficiency virus
HLAHumanl leukocyte antigen
IDOIndoleamine 2,3-dioxygenase
IFNInterferon
IgImmunoglobulin
IgM-ALAIgm anti-leukocyte autoantibodies
ILInterleukin
MHCMajor histocompatibility complex
NKNatural killer
PD-LProgram death-ligand
TGFTransforming growth factor
ThT helper
TIMT cell immunoglobulin and mucin domain
TLRToll-like receptor
TNFTumor necrosis factor
TregT regulatory

References

  1. Schumacher, A.; Costa, S.-D.D.; Zenclussen, A.C. Endocrine factors modulating immune responses in pregnancy. Front. Immunol. 2014, 5, 196. [Google Scholar] [CrossRef] [PubMed]
  2. Sappenfield, E.; Jamieson, D.J.; Kourtis, A.P. Pregnancy and Susceptibility to Infectious Diseases. Infect. Dis. Obstet. Gynecol. 2013, 2013, 1–8. [Google Scholar] [CrossRef] [PubMed]
  3. Krieg, S.; Westphal, L. Immune Function and Recurrent Pregnancy Loss. Semin. Reprod. Med. 2015, 33, 305–311. [Google Scholar] [CrossRef] [PubMed]
  4. Bonney, E.A. Immune Regulation in Pregnancy: A Matter of Perspective? Obstet. Gynecol. Clin. N. Am. 2016, 43, 679–698. [Google Scholar] [CrossRef] [PubMed]
  5. Nair, R.R.; Verma, P.; Singh, K. Immune-endocrine crosstalk during pregnancy. Gen. Comp. Endocrinol. 2017, 242, 18–23. [Google Scholar] [CrossRef] [PubMed]
  6. Schumacher, A. Human Chorionic Gonadotropin as a Pivotal Endocrine Immune Regulator Initiating and Preserving Fetal Tolerance. Int. J. Mol. Sci. 2017, 18, 2166. [Google Scholar] [CrossRef] [PubMed]
  7. Racicot, K.; Kwon, J.Y.; Aldo, P.; Silasi, M.; Mor, G. Understanding the complexity of the immune system during pregnancy. Am. J. Reprod. Immunol. 2014, 72, 107–116. [Google Scholar] [CrossRef] [PubMed]
  8. Adar, T.; Grisaru-Granovsky, S.; Ben Ya’acov, A.; Goldin, E.; Bar-Gil Shitrit, A.; Ben Ya’acov, A.; Goldin, E.; Bar-Gil Shitrit, A. Pregnancy and the Immune System: General Overview and the Gastroenterological Perspective. Dig. Dis. Sci. 2015, 60, 2581–2589. [Google Scholar] [CrossRef] [PubMed]
  9. Zhang, J.; Dunk, C.; Croy, A.B.; Lye, S.J. To serve and to protect: The role of decidual innate immune cells on human pregnancy. Cell Tissue Res. 2016, 363, 249–265. [Google Scholar] [CrossRef] [PubMed]
  10. Pang, G.; Buret, A.; Batey, R.T.; Chen, Q.Y.; Couch, L.; Cripps, A.; Clancy, R. Morphological, phenotypic and functional characteristics of a pure population of CD56+ CD16- CD3- large granular lymphocytes generated from human duodenal mucosa. Immunology 1993, 79, 498–505. [Google Scholar] [PubMed]
  11. González, I.T.; Barrientos, G.; Freitag, N.; Otto, T.; Thijssen, V.L.J.L.; Moschansky, P.; von Kwiatkowski, P.; Klapp, B.F.; Winterhager, E.; Bauersachs, S.; Blois, S.M. Uterine NK Cells Are Critical in Shaping DC Immunogenic Functions Compatible with Pregnancy Progression. PLoS ONE 2012, 7, e46755. [Google Scholar] [CrossRef] [PubMed]
  12. Wang, S.; Li, Y.-P.; Ding, B.; Zhao, Y.-R.; Chen, Z.-J.; Xu, C.-Y.; Fu, Y.-B.; Wang, X.-T. Recurrent miscarriage is associated with a decline of decidual natural killer cells expressing killer cell immunoglobulin-like receptors specific for human leukocyte antigen C. J. Obstet. Gynaecol. Res. 2014, 40, 1288–1295. [Google Scholar] [CrossRef] [PubMed]
  13. Zhang, J.; Dunk, C.E.; Lye, S.J. Sphingosine signalling regulates decidual NK cell angiogenic phenotype and trophoblast migration. Hum. Reprod. 2013, 28, 3026–3037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Hazan, A.D.; Smith, S.D.; Jones, R.L.; Whittle, W.; Lye, S.J.; Dunk, C.E. Vascular-leukocyte interactions: Mechanisms of human decidual spiral artery remodeling in vitro. Am. J. Pathol. 2010, 177, 1017–1030. [Google Scholar] [CrossRef] [PubMed]
  15. Hanna, J.; Goldman-Wohl, D.; Hamani, Y.; Avraham, I.; Greenfield, C.; Natanson-Yaron, S.; Prus, D.; Cohen-Daniel, L.; Arnon, T.I.; Manaster, I.; et al. Decidual NK cells regulate key developmental processes at the human fetal-maternal interface. Nat. Med. 2006, 12, 1065–1074. [Google Scholar] [CrossRef] [PubMed]
  16. Svensson, J.; Jenmalm, M.C.; Matussek, A.; Geffers, R.; Berg, G.; Ernerudh, J. Macrophages at the Fetal-Maternal Interface Express Markers of Alternative Activation and Are Induced by M-CSF and IL-10. J. Immunol. 2011, 187, 3671–3682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Houser, B.L.; Tilburgs, T.; Hill, J.; Nicotra, M.L.; Strominger, J.L. Two Unique Human Decidual Macrophage Populations. J. Immunol. 2011, 186, 2633–2642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Xu, Y.; Romero, R.; Miller, D.; Kadam, L.; Mial, T.N.; Plazyo, O.; Garcia-Flores, V.; Hassan, S.S.; Xu, Z.; Tarca, A.L.; et al. An M1-like Macrophage Polarization in Decidual Tissue during Spontaneous Preterm Labor That Is Attenuated by Rosiglitazone Treatment. J. Immunol. 2016, 196, 2476–2491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Guzman-Genuino, R.M.; Diener, K.R. Regulatory B cells in pregnancy: Lessons from autoimmunity, graft tolerance, and cancer. Front. Immunol. 2017, 8, 1–13. [Google Scholar] [CrossRef] [PubMed]
  20. Zenclussen, A.C. Adaptive Immune Responses During Pregnancy. Am. J. Reprod. Immunol. 2013, 69, 291–303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Rowe, J.H.; Ertelt, J.M.; Xin, L.; Way, S.S. Pregnancy imprints regulatory memory that sustains anergy to fetal antigen. Nature 2012, 490, 102–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Chen, Q.; Zhu, X.; Chen, R.; Liu, J.; Liu, P.; Hu, A.; WU, L.; Hua, H.; Yuan, H. Early Pregnancy Factor Enhances the Generation and Function of CD4+CD25+ Regulatory T Cells. Tohoku J. Exp. Med. 2016, 240, 215–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Muzzio, D.; Zygmunt, M.; Jensen, F. The role of pregnancy-associated hormones in the development and function of regulatory B cells. Front. Endocrinol. 2014, 5, 39. [Google Scholar] [CrossRef] [PubMed]
  24. Rolle, L.; Memarzadeh Tehran, M.; Morell-García, A.; Raeva, Y.; Schumacher, A.; Hartig, R.; Costa, S.-D.; Jensen, F.; Zenclussen, A.C. Cutting Edge: IL-10-Producing Regulatory B Cells in Early Human Pregnancy. Am. J. Reprod. Immunol. 2013, 70, 448–453. [Google Scholar] [CrossRef] [PubMed]
  25. Kollmann, T.R.; Kampmann, B.; Mazmanian, S.K.; Marchant, A.; Levy, O. Protecting the Newborn and Young Infant from Infectious Diseases: Lessons from Immune Ontogeny. Immunity 2017, 46, 350–363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Almanzar, G.; Schönlaub, J.; Hammerer-Lercher, A.; Koppelstaetter, C.; Bernhard, D.; Prelog, M. Influence of the delivery modus on subpopulations and replication of lymphocytes in mothers and newborns. Early Hum. Dev. 2015, 91, 663–670. [Google Scholar] [CrossRef] [PubMed]
  27. Kumar, S.K.M.; Bhat, B.V. Distinct mechanisms of the newborn innate immunity. Immunol. Lett. 2016, 173, 42–54. [Google Scholar] [CrossRef] [PubMed]
  28. Basha, S.; Surendran, N.; Pichichero, M. Immune responses in neonates. Expert Rev. Clin. Immunol. 2014, 10, 1171–1184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Allan Walker, W. Initial intestinal colonization in the human infant and immune homeostasis. Ann. Nutr. Metab. 2013, 63, 8–15. [Google Scholar] [CrossRef] [PubMed]
  30. Levy, O. Innate immunity of the newborn: Basic mechanisms and clinical correlates. Nat. Rev. Immunol. 2007, 7, 379–390. [Google Scholar] [CrossRef] [PubMed]
  31. Maródi, L.; Káposzta, R.; Campbell, D.E.; Polin, R.A.; Csongor, J.; Johnston, R.B. Candidacidal mechanisms in the human neonate. Impaired IFN-gamma activation of macrophages in newborn infants. J. Immunol. 1994, 153, 5643–5649. [Google Scholar] [PubMed]
  32. Wilson, C.B.; Westall, J.; Johnston, L.; Lewis, D.B.; Dower, S.K.; Alpert, A.R. Decreased production of interferon-gamma by human neonatal cells. Intrinsic and regulatory deficiencies. J. Clin. Investig. 1986, 77, 860–867. [Google Scholar] [CrossRef] [PubMed]
  33. Björkqvist, M.; Jurstrand, M.; Bodin, L.; Fredlund, H.; Schollin, J. Defective Neutrophil Oxidative Burst in Preterm Newborns on Exposure to Coagulase-Negative Staphylococci. Pediatr. Res. 2004, 55, 966–971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Cairo, M.S.; Rucker, R.; Bennetts, G.A.; Hicks, D.; Worcester, C.; Amlie, R.; Johnson, S.; Katz, J.; Thorne, G.M. Improved survival of newborns receiving leukocyte transfusions for sepsis. Pediatrics 1984, 74, 887–892. [Google Scholar] [CrossRef] [PubMed]
  35. Ambruso, D.R.; Bentwood, B.; Henson, P.M.; Johnston, R.B. Oxidative Metabolism of Cord Blood Neutrophils: Relationship to Content and Degranulation of Cytoplasmic Granules. Pediatr. Res. 1984, 18, 1148–1153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Carr, R. Neutrophil Production and Function in Newborn Infants. Br. J. Haematol. 2000, 110, 18–28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Schnurr, M.; Toy, T.; Shin, A.; Wagner, M.; Cebon, J.; Maraskovsky, E. Extracellular nucleotide signaling by P2 receptors inhibits IL-12 and enhances IL-23 expression in human dendritic cells: A novel role for the cAMP pathway. Blood 2005, 105, 1582–1589. [Google Scholar] [CrossRef] [PubMed]
  38. Vanden Eijnden, S.; Goriely, S.; De Wit, D.; Goldman, M.; Willems, F. Preferential production of the IL-12(p40)/IL-23(p19) heterodimer by dendritic cells from human newborns. Eur. J. Immunol. 2006, 36, 21–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Angelone, D.F.; Wessels, M.R.; Coughlin, M.; Suter, E.E.; Valentini, P.; Kalish, L.A.; Levy, O. Innate Immunity of the Human Newborn Is Polarized Toward a High Ratio of IL-6/TNF-α Production In Vitro and In Vivo. Pediatr. Res. 2006, 60, 205–209. [Google Scholar] [CrossRef] [PubMed]
  40. Peters, A.M.; Bertram, P.; Gahr, M.; Speer, C.P. Reduced secretion of interleukin-1 and tumor necrosis factor-alpha by neonatal monocytes. Biol. Neonate 1993, 63, 157–162. [Google Scholar] [CrossRef] [PubMed]
  41. White, G.P.; Watt, P.M.; Holt, B.J.; Holt, P.G. Differential Patterns of Methylation of the IFN- Promoter at CpG and Non-CpG Sites Underlie Differences in IFN- Gene Expression Between Human Neonatal and Adult CD45RO- T Cells. J. Immunol. 2002, 168, 2820–2827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Aksoy, E.; Albarani, V.; Nguyen, M.; Laes, J.-F.; Ruelle, J.-L.; De Wit, D.; Willems, F.; Goldman, M.; Goriely, S. Interferon regulatory factor 3-dependent responses to lipopolysaccharide are selectively blunted in cord blood cells. Blood 2007, 109, 2887–2893. [Google Scholar] [CrossRef] [PubMed]
  43. Levy, O.; Coughlin, M.; Cronstein, B.N.; Roy, R.M.; Desai, A.; Wessels, M.R. The adenosine system selectively inhibits TLR-mediated TNF-alpha production in the human newborn. J. Immunol. 2006, 177, 1956–1966. [Google Scholar] [CrossRef] [PubMed]
  44. De Wit, D.; Tonon, S.; Olislagers, V.; Goriely, S.; Boutriaux, M.; Goldman, M.; Willems, F. Impaired responses to toll-like receptor 4 and toll-like receptor 3 ligands in human cord blood. J. Autoimmun. 2003, 21, 277–281. [Google Scholar] [CrossRef] [PubMed]
  45. Langrish, C.L.; Buddle, J.C.; Thrasher, A.J.; Goldblatt, D. Neonatal dendritic cells are intrinsically biased against Th-1 immune responses. Clin. Exp. Immunol. 2002, 128, 118–123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Wang, Y.; Xu, H.; Zheng, X.; Wei, H.; Sun, R.; Tian, Z. High expression of NKG2A/CD94 and low expression of granzyme B are associated with reduced cord blood NK cell activity. Cell. Mol. Immunol. 2007, 4, 377–382. [Google Scholar] [PubMed]
  47. Dalle, J.-H.; Menezes, J.; Wagner, É.; Blagdon, M.; Champagne, J.; Champagne, M.A.; Duval, M. Characterization of Cord Blood Natural Killer Cells: Implications for Transplantation and Neonatal Infections. Pediatr. Res. 2005, 57, 649–655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Nguyen, T.G.; Ward, C.M.; Morris, J.M. To B or not to B cells-mediate a healthy start to life. Clin. Exp. Immunol. 2013, 171, 124–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Piątosa, B.; Wolska-Kuśnierz, B.; Pac, M.; Siewiera, K.; Gałkowska, E.; Bernatowska, E. B cell subsets in healthy children: Reference values for evaluation of B cell maturation process in peripheral blood. Cytom. Part B Clin. Cytom. 2010, 78B, 372–381. [Google Scholar] [CrossRef] [PubMed]
  50. Leandro, M.J. B-cell subpopulations in humans and their differential susceptibility to depletion with anti-CD20 monoclonal antibodies. Arthritis Res. Ther. 2013, 15 (Suppl. 1), S3. [Google Scholar] [CrossRef] [PubMed]
  51. LeBien, T.W. Fates of human B-cell precursors. Blood 2000, 96, 9–23. [Google Scholar] [PubMed]
  52. Weller, S.; Braun, M.C.; Tan, B.K.; Rosenwald, A.; Cordier, C.; Conley, M.E.; Plebani, A.; Kumararatne, D.S.; Bonnet, D.; Tournilhac, O.; et al. Human blood IgM “memory” B cells are circulating splenic marginal zone B cells harboring a prediversified immunoglobulin repertoire. Blood 2004, 104, 3647–3654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Fettke, F.; Schumacher, A.; Canellada, A.; Toledo, N.; Bekeredjian-Ding, I.; Bondt, A.; Wuhrer, M.; Costa, S.-D.; Zenclussen, A.C. Maternal and Fetal Mechanisms of B Cell Regulation during Pregnancy: Human Chorionic Gonadotropin Stimulates B Cells to Produce IL-10 While Alpha-Fetoprotein Drives Them into Apoptosis. Front. Immunol. 2016, 7. [Google Scholar] [CrossRef] [PubMed]
  54. Fettke, F.; Schumacher, A.; Costa, S.-D.D.; Zenclussen, A.C. B cells: The old new players in reproductive immunology. Front. Immunol. 2014, 5, 285. [Google Scholar] [CrossRef] [PubMed]
  55. Herse, F.; Staff, A.C.; Hering, L.; Müller, D.N.; Luft, F.C.; Dechend, R. AT1-receptor autoantibodies and uteroplacental RAS in pregnancy and pre-eclampsia. J. Mol. Med. 2008, 86, 697–703. [Google Scholar] [CrossRef] [PubMed]
  56. Arck, P.C.; Hecher, K.; Solano, M.E. B Cells in Pregnancy: Functional Promiscuity or Tailored Function? Biol. Reprod. 2015, 92, 12. [Google Scholar] [CrossRef] [PubMed]
  57. Muzzio, D.O.; Soldati, R.; Ehrhardt, J.; Utpatel, K.; Evert, M.; Zenclussen, A.C.; Zygmunt, M.; Jensen, F. B Cell Development Undergoes Profound Modifications and Adaptations During Pregnancy in Mice. Biol. Reprod. 2014, 91, 115. [Google Scholar] [CrossRef] [PubMed]
  58. Gomez-Lopez, N.; Romero, R.; Xu, Y.; Miller, D.; Leng, Y.; Panaitescu, B.; Silva, P.; Faro, J.; Alhousseini, A.; Gill, N.; et al. The immunophenotype of amniotic fluid leukocytes in normal and complicated pregnancies. Am. J. Reprod. Immunol. 2018, 79, e12827. [Google Scholar] [CrossRef] [PubMed]
  59. Lima, J.; Martins, C.; Leandro, M.J.; Nunes, G.; Sousa, M.-J.; Branco, J.C.; Borrego, L.-M. Characterization of B cells in healthy pregnant women from late pregnancy to post-partum: A prospective observational study. BMC Pregnancy Childbirth 2016, 16, 139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Ruggeri, A.; Paviglianiti, A.; Gluckman, E.; Rocha, V. Impact of HLA in cord blood transplantation outcomes. HLA 2016, 87, 413–421. [Google Scholar] [CrossRef] [PubMed]
  61. Sarvaria, A.; Basar, R.; Mehta, R.S.; Shaim, H.; Muftuoglu, M.; Khoder, A.; Sekine, T.; Gokdemir, E.; Kondo, K.; Marin, D.; et al. IL-10+ regulatory B cells are enriched in cord blood and may protect against cGVHD after cord blood transplantation. Blood 2016, 128, 1346–1361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Martins, C.; Lima, J.; Nunes, G.; Borrego, L.M. Pregnancy alters the circulating B cell compartment in atopic asthmatic women, and transitional B cells are positively associated with the development of allergy manifestations in their progeny. Am. J. Reprod. Immunol. 2016, 76, 465–474. [Google Scholar] [CrossRef] [PubMed]
  63. Katz, S.I.; Parker, D.; Turk, J.L. B-cell suppression of delayed hypersensitivity reactions. Nature 1974, 251, 550–551. [Google Scholar] [CrossRef] [PubMed]
  64. Fillatreau, S.; Sweenie, C.H.; McGeachy, M.J.; Gray, D.; Anderton, S.M. B cells regulate autoimmunity by provision of IL-10. Nat. Immunol. 2002, 3, 944–950. [Google Scholar] [CrossRef] [PubMed]
  65. Holt, P.G.; Jones, C.A. The development of the immune system during pregnancy and early life. Allergy 2000, 55, 688–697. [Google Scholar] [CrossRef] [PubMed]
  66. Blair, P.A.; Noreña, L.Y.; Flores-Borja, F.; Rawlings, D.J.; Isenberg, D.A.; Ehrenstein, M.R.; Mauri, C. CD19(+)CD24(hi)CD38(hi) B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic Lupus Erythematosus patients. Immunity 2010, 32, 129–140. [Google Scholar] [CrossRef] [PubMed]
  67. Mauri, C.; Menon, M. Human regulatory B cells in health and disease: Therapeutic potential. J. Clin. Investig. 2017, 127, 772–779. [Google Scholar] [CrossRef] [PubMed]
  68. Rosser, E.C.; Mauri, C. Regulatory B cells: Origin, phenotype, and function. Immunity 2015, 42, 607–612. [Google Scholar] [CrossRef] [PubMed]
  69. Mauri, C.; Menon, M. The expanding family of regulatory B cells. Int. Immunol. 2015, 27, 479–486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Floudas, A.; Amu, S.; Fallon, P.G. New Insights into IL-10 Dependent and IL-10 Independent Mechanisms of Regulatory B Cell Immune Suppression. J. Clin. Immunol. 2016, 36, 25–33. [Google Scholar] [CrossRef] [PubMed]
  71. Flores-Borja, F.; Bosma, A.; Ng, D.; Reddy, V.; Ehrenstein, M.R.; Isenberg, D.A.; Mauri, C. CD19+CD24hiCD38hi B Cells Maintain Regulatory T Cells While Limiting TH1 and TH17 Differentiation. Sci. Transl. Med. 2013, 5, 173ra23. [Google Scholar] [CrossRef] [PubMed]
  72. Siewe, B.; Stapleton, J.T.; Martinson, J.; Keshavarzian, A.; Kazmi, N.; Demarais, P.M.; French, A.L.; Landay, A. Regulatory B cell frequency correlates with markers of HIV disease progression and attenuates anti-HIV CD8+ T cell function in vitro. J. Leukoc. Biol. 2013, 93, 811–818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Menon, M.; Blair, P.A.; Isenberg, D.A.; Mauri, C. A Regulatory Feedback between Plasmacytoid Dendritic Cells and Regulatory B Cells Is Aberrant in Systemic Lupus Erythematosus. Immunity 2016, 44, 683–697. [Google Scholar] [CrossRef] [PubMed]
  74. Bosma, A.; Abdel-Gadir, A.; Isenberg, D.A.; Jury, E.C.; Mauri, C. Lipid-Antigen Presentation by CD1d + B Cells Is Essential for the Maintenance of Invariant Natural Killer T Cells. Immunity 2012, 36, 477–490. [Google Scholar] [CrossRef] [PubMed]
  75. Iwata, Y.; Matsushita, T.; Horikawa, M.; Dilillo, D.J.; Yanaba, K.; Venturi, G.M.; Szabolcs, P.M.; Bernstein, S.H.; Magro, C.M.; Williams, A.D.; et al. Characterization of a rare IL-10-competent B-cell subset in humans that parallels mouse regulatory B10 cells. Blood 2011, 117, 530–541. [Google Scholar] [CrossRef] [PubMed]
  76. Lindner, S.; Dahlke, K.; Sontheimer, K.; Hagn, M.; Kaltenmeier, C.; Barth, T.F.E.; Beyer, T.; Reister, F.; Fabricius, D.; Lotfi, R.; et al. Interleukin 21-induced granzyme b-expressing b cells infiltrate tumors and regulate t cells. Cancer Res. 2013, 73, 2468–2479. [Google Scholar] [CrossRef] [PubMed]
  77. Van de Veen, W.; Stanic, B.; Yaman, G.; Wawrzyniak, M.; Söllner, S.; Akdis, D.G.; Rückert, B.; Akdis, C.A.; Akdis, M. IgG4 production is confined to human IL-10-producing regulatory B cells that suppress antigen-specific immune responses. J. Allergy Clin. Immunol. 2013, 131, 1204–1212. [Google Scholar] [CrossRef] [PubMed]
  78. Matsumoto, M.; Baba, A.; Yokota, T.; Nishikawa, H.; Ohkawa, Y.; Kayama, H.; Kallies, A.; Nutt, S.L.; Sakaguchi, S.; Takeda, K.; et al. Interleukin-10-producing plasmablasts exert regulatory function in autoimmune inflammation. Immunity 2014, 41, 1040–1051. [Google Scholar] [CrossRef] [PubMed]
  79. Saze, Z.; Schuler, P.J.; Hong, C.-S.; Cheng, D.; Jackson, E.K.; Whiteside, T.L. Adenosine production by human B cells and B cell-mediated suppression of activated T cells. Blood 2013, 122, 9–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Nouël, A.; Pochard, P.; Simon, Q.; Ségalen, I.; Le Meur, Y.; Pers, J.O.; Hillion, S. B-Cells induce regulatory T cells through TGF-β/IDO production in A CTLA-4 dependent manner. J. Autoimmun. 2015, 59, 53–60. [Google Scholar] [CrossRef] [PubMed]
  81. Liu, J.; Zhan, W.; Kim, C.J.; Clayton, K.; Zhao, H.; Lee, E.; Cao, J.C.; Ziegler, B.; Gregor, A.; Yue, F.Y.; et al. IL-10-producing B cells are induced early in HIV-1 infection and suppress HIV-1-specific T cell responses. PLoS ONE 2014, 9, e89236. [Google Scholar] [CrossRef] [PubMed]
  82. Goode, I.; Xu, H.; Ildstad, S.T. Regulatory B Cells: The New “It” Cell. Transplant. Proc. 2014, 46, 3–8. [Google Scholar] [CrossRef] [PubMed]
  83. Kaku, H.; Cheng, K.F.; Al-Abed, Y.; Rothstein, T.L. A novel mechanism of B cell-mediated immune suppression through CD73 expression and adenosine production. J. Immunol. 2014, 193, 5904–5913. [Google Scholar] [CrossRef] [PubMed]
  84. Ray, A.; Wang, L.; Dittel, B.N. IL-10-independent regulatory B-cell subsets and mechanisms of action. Int. Immunol. 2015, 27, 531–536. [Google Scholar] [CrossRef] [PubMed]
  85. Mauri, C.; Ehrenstein, M.R. The “short” history of regulatory B cells. Trends Immunol. 2008, 29, 34–40. [Google Scholar] [CrossRef] [PubMed]
  86. Sims, G.P.; Ettinger, R.; Shirota, Y.; Yarboro, C.H.; Illei, G.G.; Lipsky, P.E. Identification and characterization of circulating human transitional B cells. Blood 2016, 105, 4390–4399. [Google Scholar] [CrossRef] [PubMed]
  87. Khoder, A.; Sarvaria, A.; Alsuliman, A.; Chew, C.; Sekine, T.; Cooper, N.; Mielke, S.; de Lavallade, H.; Muftuoglu, M.; Fernandez Curbelo, I.; et al. Regulatory B cells are enriched within the IgM memory and transitional subsets in healthy donors but are deficient in chronic GVHD. Blood 2014, 124, 2034–2045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Martins, C.; Lima, J.; Nunes, G.; Borrego, L.M. Regulatory T and B cells in asthmatic women: Variations from pregnancy to postpartum. J. Investig. Allergol. Clin. Immunol. 2017, 27, 46–57. [Google Scholar] [CrossRef] [PubMed]
  89. Noh, J.; Noh, G.; Lee, S.J.; Lee, J.H.; Kim, A.; Kim, H.S.; Choi, W.S. Tolerogenic effects of interferon-gamma with induction of allergen-specific interleukin-10-producing regulatory B cell (Br1) changes in non-IgE-mediated food allergy. Cell. Immunol. 2012, 273, 140–149. [Google Scholar] [CrossRef] [PubMed]
  90. Amu, S.; Saunders, S.P.; Kronenberg, M.; Mangan, N.E.; Atzberger, A.; Fallon, P.G. Regulatory B cells prevent and reverse allergic airway inflammation via FoxP3-positive T regulatory cells in a murine model. J. Allergy Clin. Immunol. 2010, 125, 1114.e8–1124.e8. [Google Scholar] [CrossRef] [PubMed]
  91. Matsushita, T.; Horikawa, M.; Iwata, Y.; Tedder, T.F. Regulatory B Cells (B10 Cells) and Regulatory T Cells Have Independent Roles in Controlling Experimental Autoimmune Encephalomyelitis Initiation and Late-Phase Immunopathogenesis. J. Immunol. 2010, 185, 2240–2252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Duddy, M.; Niino, M.; Adatia, F.; Hebert, S.; Freedman, M.; Atkins, H.; Kim, H.J.; Bar-Or, A. Distinct effector cytokine profiles of memory and naive human B cell subsets and implication in multiple sclerosis. J. Immunol. 2007, 178, 6092–6099. [Google Scholar] [CrossRef] [PubMed]
  93. Knippenberg, S.; Smolders, J.; Thewissen, M.; Peelen, E.; Tervaert, J.W.C.; Hupperts, R.; Damoiseaux, J. Effect of vitamin D 3 supplementation on peripheral B cell differentiation and isotype switching in patients with multiple sclerosis. Mult. Scler. J. 2011, 17, 1418–1423. [Google Scholar] [CrossRef] [PubMed]
  94. Saussine, A.; Tazi, A.; Feuillet, S.; Rybojad, M.; Juillard, C.; Bergeron, A.; Dessirier, V.; Bouhidel, F.; Janin, A.; Bensussan, A.; et al. Active chronic sarcoidosis is characterized by increased transitional blood B cells, increased IL-10-producing regulatory B cells and high BAFF levels. PLoS ONE 2012, 7, e43588. [Google Scholar] [CrossRef] [PubMed]
  95. Watanabe, R.; Ishiura, N.; Nakashima, H.; Kuwano, Y.; Okochi, H.; Tamaki, K.; Sato, S.; Tedder, T.F.; Fujimoto, M. Regulatory B Cells (B10 Cells) Have a Suppressive Role in Murine Lupus: CD19 and B10 Cell Deficiency Exacerbates Systemic Autoimmunity. J. Immunol. 2010, 184, 4801–4809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Carter, N.A.; Rosser, E.C.; Mauri, C. Interleukin-10 produced by B cells is crucial for the suppression of Th17/Th1 responses, induction of T regulatory type 1 cells and reduction of collagen-induced arthritis. Arthritis Res. Ther. 2012, 14, R32. [Google Scholar] [CrossRef] [PubMed]
  97. Noh, G.; Lee, J.H. Regulatory B cells and allergic diseases. Allergy Asthma Immunol. Res. 2011, 3, 168–177. [Google Scholar] [CrossRef] [PubMed]
  98. Das, A.; Ellis, G.; Pallant, C.; Lopes, A.R.; Khanna, P.; Peppa, D.; Chen, A.; Blair, P.; Dusheiko, G.; Gill, U.; et al. IL-10-producing regulatory B cells in the pathogenesis of chronic hepatitis B virus infection. J. Immunol. 2012, 189, 3925–3935. [Google Scholar] [CrossRef] [PubMed]
  99. Zhang, M.; Zheng, X.; Zhang, J.; Zhu, Y.; Zhu, X.; Liu, H.; Zeng, M.; Graner, M.W.; Zhou, B.; Chen, X. CD19(+)CD1d(+)CD5(+) B cell frequencies are increased in patients with tuberculosis and suppress Th17 responses. Cell. Immunol. 2012, 274, 89–97. [Google Scholar] [CrossRef] [PubMed]
  100. Han, S.; Feng, S.; Ren, M.; Ma, E.; Wang, X.; Xu, L.; Xu, M. Glioma cell-derived placental growth factor induces regulatory B cells. Int. J. Biochem. Cell Biol. 2014, 57, 63–68. [Google Scholar] [CrossRef] [PubMed]
  101. Schioppa, T.; Moore, R.; Thompson, R.G.; Rosser, E.C.; Kulbe, H.; Nedospasov, S.; Mauri, C.; Coussens, L.M.; Balkwill, F.R. B regulatory cells and the tumor-promoting actions of TNF- during squamous carcinogenesis. Proc. Natl. Acad. Sci. USA 2011, 108, 10662–10667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Biragyn, A.; Lee-Chang, C. A new paradigm for an old story: The role of regulatory B cells in cancer. Front. Immunol. 2012, 3, 206. [Google Scholar] [CrossRef] [PubMed]
  103. Horikawa, M.; Minard-Colin, V.; Matsushita, T.; Tedder, T.F. Regulatory B cell production of IL-10 inhibits lymphoma depletion during CD20 immunotherapy in mice. J. Clin. Investig. 2011, 121, 4268–4280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Zhao, G.; Moore, D.J.; Lee, K.M.; Kim, J.I.; Duff, P.E.; O’Connor, M.R.; Hirohashi, T.; Lei, J.; Yang, M.; Markmann, J.F.; et al. An unexpected counter-regulatory role of IL-10 in B-lymphocyte-mediated transplantation tolerance. Am. J. Transplant. 2010, 10, 796–801. [Google Scholar] [CrossRef] [PubMed]
  105. Rowe, V.; Banovic, T.; MacDonald, K.P.; Kuns, R.; Don, A.L.; Morris, E.S.; Burman, A.C.; Bofinger, H.M.; Clouston, A.D.; Hill, G.R. Host B cells produce IL-10 following TBI and attenuate acute GVHD after allogeneic bone marrow transplantation. Blood 2006, 108, 2485–2492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Newell, K.A.; Asare, A.; Kirk, A.D.; Gisler, T.D.; Bourcier, K.; Suthanthiran, M.; Burlingham, W.J.; Marks, W.H.; Sanz, I.; Lechler, R.I.; et al. Identification of a B cell signature associated with renal transplant tolerance in humans. J. Clin. Investig. 2010, 120, 1836–1847. [Google Scholar] [CrossRef] [PubMed]
  107. Shabir, S.; Girdlestone, J.; Briggs, D.; Kaul, B.; Smith, H.; Daga, S.; Chand, S.; Jham, S.; Navarrete, C.; Harper, L.; et al. Transitional B Lymphocytes Are Associated With Protection From Kidney Allograft Rejection: A Prospective Study. Am. J. Transplant. 2015, 15, 1384–1391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Jensen, F.; Muzzio, D.; Soldati, R.; Fest, S.; Zenclussen, A.C. Regulatory B10 cells restore pregnancy tolerance in a mouse model. Biol. Reprod. 2013, 89, 90. [Google Scholar] [CrossRef] [PubMed]
  109. Shen, P.; Fillatreau, S. Suppressive functions of B cells in infectious diseases. Int. Immunol. 2015, 27, 513–519. [Google Scholar] [CrossRef] [PubMed]
  110. Soares, R.R.; Antinarelli, L.M.R.; Abramo, C.; Macedo, G.C.; Coimbra, E.S.; Scopel, K.K.G. What do we know about the role of regulatory B cells (Breg) during the course of infection of two major parasitic diseases, malaria and leishmaniasis? Pathog. Glob. Health 2017, 111, 107–115. [Google Scholar] [CrossRef] [PubMed]
  111. Jiao, Y.; Wang, X.; Zhang, T.; Lijun, S.; Wang, R.; Li, W.; Ji, Y.; Wu, H.; Liu, C. Regulatory B cells correlate with HIV disease progression. Microbiol. Immunol. 2014, 58, 449–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Bodhankar, S.; Wang, C.; Vandenbark, A.A.; Offner, H. Estrogen-induced protection against experimental autoimmune encephalomyelitis is abrogated in the absence of B cells. Eur. J. Immunol. 2011, 41, 1165–1175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Zhang, J.; Benedek, G.; Bodhankar, S.; Lapato, A.; Vandenbark, A.A.; Offner, H. IL-10 producing B cells partially restore E2-mediated protection against EAE in PD-L1 deficient mice. J. Neuroimmunol. 2015, 285, 129–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Zhang, J.; Lapato, A.; Bodhankar, S.; Vandenbark, A.A.; Offner, H. Treatment with IL-10 producing B cells in combination with E2 ameliorates EAE severity and decreases CNS inflammation in B cell-deficient mice. Metab. Brain Dis. 2015, 30, 1117–1127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Benedek, G.; Zhang, J.; Bodhankar, S.; Nguyen, H.; Kent, G.; Jordan, K.; Manning, D.; Vandenbark, A.A.; Offner, H. Estrogen induces multiple regulatory B cell subtypes and promotes M2 microglia and neuroprotection during experimental autoimmune encephalomyelitis. J. Neuroimmunol. 2016, 293, 45–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Benedek, G.; Zhang, J.; Nguyen, H.; Kent, G.; Seifert, H.; Vandenbark, A.A.; Offner, H. Novel feedback loop between M2 macrophages/microglia and regulatory B cells in estrogen-protected EAE mice. J. Neuroimmunol. 2017, 305, 59–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Chakravarty, E.F.; Murray, E.R.; Kelman, A.; Farmer, P.; Saji, F.; Samejima, Y.; Kamiura, S.; Koyoma, M.; Boothby, L.; Doering, P.; et al. Pregnancy outcomes after maternal exposure to rituximab. Blood 2011, 117, 1499–1506. [Google Scholar] [CrossRef] [PubMed]
  118. Das, G.; Damotte, V.; Gelfand, J.M.; Bevan, C.; Cree, B.A.C.; Do, L.; Green, A.J.; Hauser, S.L.; Bove, R. Rituximab before and during pregnancy: A systematic review, and a case series in MS and NMOSD. Neurol. Neuroimmunol. Neuroinflamm. 2018, 5, e453. [Google Scholar] [CrossRef] [PubMed]
  119. Happle, C.; Jirmo, A.C.; Meyer-Bahlburg, A.; Habener, A.; Hoymann, H.G.; Hennig, C.; Skuljec, J.; Hansen, G. B cells control maternofetal priming of allergy and tolerance in a murine model of allergic airway inflammation. J. Allergy Clin. Immunol. 2018, 141, 685.e6–696.e6. [Google Scholar] [CrossRef] [PubMed]
  120. Esteve-Solé, A.; Teixidó, I.; Deyà-Martínez, A.; Yagüe, J.; Plaza-Martín, A.M.; Juan, M.; Alsina, L. Characterization of the Highly Prevalent Regulatory CD24hiCD38hi B-Cell Population in Human Cord Blood. Front. Immunol. 2017, 8, 201. [Google Scholar] [CrossRef] [PubMed]
  121. Li, S.; Ma, F.; Hao, H.; Wang, D.; Gao, Y.; Zhou, J.; Li, F.; Lin, H.C.; Xiao, X.; Zeng, Q. Marked elevation of circulating CD19+CD38hiCD24hitransitional B cells give protection against neonatal sepsis. Pediatr. Neonatol. 2018, 59, 286–304. [Google Scholar] [CrossRef] [PubMed]
  122. Lobo, P.I.; Bajwa, A.; Schlegel, K.H.; Vengal, J.; Lee, S.J.; Huang, L.; Ye, H.; Deshmukh, U.; Wang, T.; Pei, H.; et al. Natural IgM anti-leukocyte autoantibodies attenuate excess inflammation mediated by innate and adaptive immune mechanisms involving Th-17. J Immunol. 2012, 188, 1675–1685. [Google Scholar] [CrossRef] [PubMed]
  123. Lobo, P.I.; Schlegel, K.H.; Spencer, C.E.; Okusa, M.D.; Chisholm, C.; Mchedlishvili, N.; Park, A.; Christ, C.; Burtner, C. Naturally Occurring IgM Anti-Leukocyte Autoantibodies (IgM-ALA) Inhibit T Cell Activation and Chemotaxis. J. Immunol. 2008, 180, 1780–1791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Cerutti, A.; Cols, M.; Puga, I. Marginal zone B cells: Virtues of innate-like antibody-producing lymphocytes. Nat. Rev. Immunol. 2013, 13, 118–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Allman, D.; Pillai, S. Peripheral B cell subsets. Curr. Opin. Immunol. 2008, 20, 149–157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Muzzio, D.O.; Ziegler, K.B.; Ehrhardt, J.; Zygmunt, M.; Jensen, F. Marginal zone B cells emerge as a critical component of pregnancy well-being. Reproduction 2016, 151, 29–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Lin, S.-J.; Yan, D.-C.; Lee, Y.-C.; Hsiao, H.-S.; Lee, P.-T.; Liang, Y.-W.; Kuo, M.-L. Umbilical cord blood immunology: Relevance to stem cell transplantation. Clin. Rev. Allergy Immunol. 2012, 42, 45–57. [Google Scholar] [CrossRef] [PubMed]
  128. Sun, C.M.; Deriaud, E.; Leclerc, C.; Lo-Man, R. Upon TLR9 signaling, CD5+ B cells control the IL-12-dependent Th1-priming capacity of neonatal DCs. Immunity 2005, 22, 467–477. [Google Scholar] [CrossRef] [PubMed]
  129. Zhivaki, D.; Lemoine, S.; Lim, A.; Morva, A.; Vidalain, P.-O.; Schandene, L.; Casartelli, N.; Rameix-Welti, M.-A.; Hervé, P.-L.; Dériaud, E.; et al. Respiratory Syncytial Virus Infects Regulatory B Cells in Human Neonates via Chemokine Receptor CX3CR1 and Promotes Lung Disease Severity. Immunity 2017, 46, 301–314. [Google Scholar] [CrossRef] [PubMed]
  130. Watkins, M.L.V.; Semple, P.L.; Abel, B.; Hanekom, W.A.; Kaplan, G.; Ress, S.R. Exposure of cord blood to Mycobacterium bovis BCG induces an innate response but not a T-cell cytokine response. Clin. Vaccine Immunol. 2008, 15, 1666–1673. [Google Scholar] [CrossRef] [PubMed]
  131. Resende, R.G.; Correia-Silva, J.D.F.; Silva, T.A.; Xavier, S.G.; Bittencourt, H.; Gomez, R.S.; Abreu, M.H.N.G. Saliva and blood interferon gamma levels and IFNG genotypes in acute graft-versus-host disease. Oral Dis. 2012, 18, 816–822. [Google Scholar] [CrossRef] [PubMed]
  132. Rottman, M.; Soudais, C.; Vogt, G.; Renia, L.; Emile, J.-F.; Decaluwe, H.; Gaillard, J.-L.; Casanova, J.-L. IFN-γ Mediates the Rejection of Haematopoietic Stem Cells in IFN-γR1-Deficient Hosts. PLoS Med. 2008, 5, e26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Bustamante, J.; Boisson-Dupuis, S.S.S.; Abel, L.; Casanova, J.-L. Mendelian susceptibility to mycobacterial disease: Genetic, immunological, and clinical features of inborn errors of IFNgamma immunity. Semin. Immunol. 2014, 26, 454–470. [Google Scholar] [CrossRef] [PubMed]
  134. Fieschi, C.; Dupuis, S.; Picard, C.; Smith, C.I.; Holland, S.M.; Casanova, J.L. High levels of interferon gamma in the plasma of children with complete interferon gamma receptor deficiency. Pediatrics 2001, 107, E48. [Google Scholar] [CrossRef] [PubMed]
  135. Olbrich, P.; Martínez-Saavedra, M.T.; Perez-Hurtado, J.M.; Sanchez, C.; Sanchez, B.; Deswarte, C.; Obando, I.; Casanova, J.-L.; Speckmann, C.; Bustamante, J.; et al. Diagnostic and therapeutic challenges in a child with complete Interferon-γ Receptor 1 deficiency. Pediatr. Blood Cancer 2015, 62, 2036–2039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Breg cells in pregnancy. Breg cells are modulated throughout pregnancy, with a major role in pregnancy establishment. Pregnancy-related hormones play a major role in regulating Breg cell frequency and functions. PR: progesterone receptor, ER: estrogen receptor; LH/hCRG: human chorionic gonadotropin receptor.
Figure 1. Breg cells in pregnancy. Breg cells are modulated throughout pregnancy, with a major role in pregnancy establishment. Pregnancy-related hormones play a major role in regulating Breg cell frequency and functions. PR: progesterone receptor, ER: estrogen receptor; LH/hCRG: human chorionic gonadotropin receptor.
Ijms 19 02099 g001
Figure 2. Breg cells in the neonate. Breg cell frequency is increased in neonates, providing improved tolerance to allogenic antigens (i.e., after cord blood stem cell transplantation). Besides, there is a remarkable increase of IgM expression, which may be a new inhibitory mechanism of Bregs in neonates. Finally, Breg cells inhibit Th1 cell responses, which may be related to the reported low interferon (IFN)-γ production after mycobacterial challenge. M. tb: Mycobacterium tuberculosis, Th1: T helper 1.
Figure 2. Breg cells in the neonate. Breg cell frequency is increased in neonates, providing improved tolerance to allogenic antigens (i.e., after cord blood stem cell transplantation). Besides, there is a remarkable increase of IgM expression, which may be a new inhibitory mechanism of Bregs in neonates. Finally, Breg cells inhibit Th1 cell responses, which may be related to the reported low interferon (IFN)-γ production after mycobacterial challenge. M. tb: Mycobacterium tuberculosis, Th1: T helper 1.
Ijms 19 02099 g002
Table 1. Described B regulatory (Breg) cell subsets. DC = dendritic cells; NK = natural killer; IL = interleukin; PD-L = program death-ligand; Ig = immunoglobulin; TIM1 = T cell immunoglobulin and mucin domain; TGF = transforming growth factor.
Table 1. Described B regulatory (Breg) cell subsets. DC = dendritic cells; NK = natural killer; IL = interleukin; PD-L = program death-ligand; Ig = immunoglobulin; TIM1 = T cell immunoglobulin and mucin domain; TGF = transforming growth factor.
NamePhenotypic MarkersFunctionReferences
Immature B-cellsCD24hiCD38hiperform their action on CD4 and CD8 T cells, plasmacitoid DCs, and invariant NK T cells by IL-10 secretion, and PD-L1, CD80, CD86, and CD1d ligation[66,71,72,73,74]
B10 cellsCD24hiCD27hiproduce IL-10 and regulate monocytes and effector CD4 T cells[75]
Granzyme B+ (GZMB) cellsCD38+CD1d+IgM+CD147+regulatory function on CD4 T cells by IL10, indoleamine 2,3-dioxygenase (IDO), and GZMB[76]
Br1CD25hiCD71hiCD73lowproduce IL-10 and allergen-specific IgG4, thus suppressing allergen-specific CD4 cells and maintaining allergen tolerance[77]
PlasmablastsCD27intCD38hiproduce IL-10, but their target cell type is not known yet[78]
-CD39+CD73+regulate CD4 and CD8 T cells by adenosine formation, thus reducing inflammation by adenosine triphosphate[79]
Induced Breg cells-produce TGF-ß and IDO to suppress CD4 T cells. These cells are developed after T cell cytotoxic T-lymphocyte antigen 4 interaction[80]
-Express TIM1inhibit CD8 and CD4 T cells by producing IL-10[81]

Share and Cite

MDPI and ACS Style

Esteve-Solé, A.; Luo, Y.; Vlagea, A.; Deyà-Martínez, Á.; Yagüe, J.; Plaza-Martín, A.M.; Juan, M.; Alsina, L. B Regulatory Cells: Players in Pregnancy and Early Life. Int. J. Mol. Sci. 2018, 19, 2099. https://doi.org/10.3390/ijms19072099

AMA Style

Esteve-Solé A, Luo Y, Vlagea A, Deyà-Martínez Á, Yagüe J, Plaza-Martín AM, Juan M, Alsina L. B Regulatory Cells: Players in Pregnancy and Early Life. International Journal of Molecular Sciences. 2018; 19(7):2099. https://doi.org/10.3390/ijms19072099

Chicago/Turabian Style

Esteve-Solé, Ana, Yiyi Luo, Alexandru Vlagea, Ángela Deyà-Martínez, Jordi Yagüe, Ana María Plaza-Martín, Manel Juan, and Laia Alsina. 2018. "B Regulatory Cells: Players in Pregnancy and Early Life" International Journal of Molecular Sciences 19, no. 7: 2099. https://doi.org/10.3390/ijms19072099

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop