Next Article in Journal
The In Vitro Impact of Isoxazole Derivatives on Pathogenic Biofilm and Cytotoxicity of Fibroblast Cell Line
Previous Article in Journal
DNA Methylation as a Diagnostic, Prognostic, and Predictive Biomarker in Head and Neck Cancer
Previous Article in Special Issue
Immune Tolerance of Embryo Implantation and Pregnancy: The Role of Human Decidual Stromal Cell- and Embryonic-Derived Extracellular Vesicles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Endometrial Microbiota and Immune Tolerance in Pregnancy

by
Annalisa Inversetti
1,2,†,
Enrica Zambella
1,†,
Alice Guarano
3,
Marinella Dell’Avanzo
3 and
Nicoletta Di Simone
1,2,*
1
Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
2
IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
3
Humanitas San Pio X, Via Francesco Nava 31, 20159 Milan, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(3), 2995; https://doi.org/10.3390/ijms24032995
Submission received: 30 December 2022 / Revised: 20 January 2023 / Accepted: 1 February 2023 / Published: 3 February 2023
(This article belongs to the Special Issue Endometrium and Pregnancy Immune Tolerance)

Abstract

:
Recent studies have demonstrated that the uterus has its own microbiota. However, there is no consensus on endometrial microbiota composition, thus its role in the healthy uterine environment is still a frontier topic. Endometrial receptivity is key to embryo implantation, and in this specific context immunological tolerance against fetal antigens and the tightly regulated expression of inflammatory mediators are fundamental. According to recent evidence, endometrial microbiota may interact in a very dynamic way with the immune system during the peri-conceptional stage and later during pregnancy. For this reason, a condition of dysbiosis might lead to adverse pregnancy outcomes. The aim of this review is to summarize the evidence on the molecular mechanisms by which the endometrial microbiota may interact with the immune system. For this purpose, the link between dysbiosis and reproductive disorders, such as infertility, recurrent pregnancy loss (RPL), and preterm birth, will be discussed. In conclusion, the most recent findings from molecular analyses will be reported to illustrate and possibly overcome the intrinsic limitations of uterine microbiota detection (low endometrial biomass, high risk of contamination during sampling, and lack of standardization).

1. Introduction

The human microbiome is a collection of different microbes that inhabit the entire human body, and its role in physiology and disease is a topic of increasing interest in the scientific community. The Human Microbiome Project was one of the largest studies to describe all the microorganisms populating different sites (airways, oral cavity, skin, gastrointestinal tract, and urogenital tract) in 300 healthy volunteers from the United States (USA) with the aim of enrolling a sample population that was reasonably heterogenous in terms of ethnicity and other demographic features [1]. Historically, the gastrointestinal microbiota has been more fully investigated compared to the female reproductive tract microbiota, due to the less invasive sampling techniques and the common belief that the upper reproductive tract (URT) is a sterile environment. Nevertheless, the first proof of endometrial colonization in healthy women arrived in 1985, and it has been subsequently demonstrated that the female reproductive tract microbiota represents about 9% of the entire microbiota in women [2,3]. Thanks to the development of gene sequencing technologies, several studies of uterine microecology have been conducted, and the role of microbiota in reproductive health and endometrial disease has been investigated. According to recent evidence, the endometrium enables the growth of symbiotic bacteria which play a dual role:
  • Provide protection from tissue infection by competing with pathogenic bacteria;
  • Ensure immune-cell activation, strengthening the barrier of the endometrium, and promoting endometrial repair and angiogenesis [4,5].
Therefore, the dynamic balance of uterine microecology depends on the relationship between the endometrial microbiota, the immune system, and the endometrium, in both physiological and pathological states [6,7,8,9,10]. Moreover, these three factors all seem to be involved in embryo implantation, which remains one of the most challenging topics. As a matter of fact, the success of this process depends on both embryo quality and endometrial acceptability. Implantation failures occur due to low-quality embryos and poor endometrial receptivity [11,12]. It was demonstrated in many infertile women undergoing treatment with assisted reproductive technology (ART) that pregnancy failure recurs with repeated in vitro fertilization (IVF) cycles, even if the transferred embryos are of high quality [13]. Indeed, several local changes that lead to endometrial decidualization are required to enhance blastocyst implantation [14]. Several components, such as the cellular immune response and microbiome composition, can affect endometrial receptivity [15,16,17,18,19]. The aim of this review is to evaluate the possible role of the endometrial microbiome as an immune modulator in the phase of implantation and during pregnancy. Furthermore, the correlations between endometrial dysbiosis and adverse obstetric outcomes, such as infertility, recurrent pregnancy failure, and preterm labor, are investigated.

2. Endometrial Microbiota and Its Variation during Lifespan

2.1. The Beginning—Initial Studies on the Topic: Emerging Differences Comparing Vaginal and Endometrial Microbiomes

Most of the initial studies detected Lactobacillus (L.) spp., Gardnerella vaginalis, Enterobacter spp., and Mycoplasma hominis through transcervical sampling [20,21]. Later, Mitchell et al. found a great abundance of L. iners, Prevotella spp., and L. crispatus in 95% of vaginal swabs obtained from 58 women undergoing hysterectomy for non-cancer indications [22]. However, less bacterial DNA in upper genital tract (URT; namely, the endocervix and endometrium) samples was obtained [22]. These findings were confirmed by Chen et al., who revealed that the vaginal sites contained about four orders of magnitude more bacteria than the endometrial sites [10]. Moreno and colleagues focused their interest on the similarity—but not perfect correspondence—between the intraindividual vaginal and the endometrial microbiome, highlighting that in approximately 20% of the subjects, the endometrial and vaginal microbiomes differed in terms of either the bacterial taxa identified or the relative abundances in which they were represented in both types of samples [9]. Further studies demonstrated that Lactobacillus was the most represented genus in endometrial samples [23], while other genera commonly detected were Flavobacterium [23], Bifidobacterium, Gardnerella, Prevotella, and Streptococcus [24].

2.2. Results from an Infertile Population and Women Undergoing ART Therapy

Sampling of a population undergoing ART therapy gave the unique opportunity to analyze the endometrial microbiome in an interesting temporal window (during embryo transfer) and anatomical site (from the embryo transfer catheter).
Recent studies, reported in detail in Table 1, highlighted a potential association between a state of dysbiosis and the success of the ART procedure. Whether this association is supported by evidence is still a matter of debate, because the role of endometrial microbiome composition in subtypes of infertile patients has not yet been fully elucidated.
Firstly, a certain number of studies on this specific population revealed the possibility of Lactobacillus dominance [25,26,27,28,29].
Subsequent studies [30,31] did not confirm the Lactobacillus dominance that was first hypothesized. Higher diversity seemed to be associated with better outcomes after IVF procedures [31]. In conclusion, these contrasting findings highlighted several aspects: first, the need for a better understanding of endometrial microbiome composition and function in healthy women and its possible role in endometrial functions before studying its impact on the success of ART procedures; second, the relevance of subdividing the study population according to the cause of infertility; third, the need for standardized sampling methods.
Table 1. Endometrial microbiome composition in infertile women, including patients undergoing IVF or with a history of recurrent implantation failure (RIF).
Table 1. Endometrial microbiome composition in infertile women, including patients undergoing IVF or with a history of recurrent implantation failure (RIF).
Author, YearCountryNumber and Type of
Patients
Sample (Method)Findings
Moore, 2000
[32]
USA91 women undergoing IVFET
(Embryo transfer catheter tips)
Live birth rate (LBR):
  • Significantly higher when associated with isolated H2O2-producing Lactobacillus vs. significantly lower with isolated Streptococcus viridans
Moreno, 2016
[24]
Spain35 infertile women undergoing IVF EF
(Aspiration with catheter)
3 poor DNA quality
17 LDM
15 NLDM associated with: significantly lower implantation, pregnancy, ongoing pregnancy and LBR
Tao, 2017
[25]
USA70 women undergoing IVFET
(Embryo transfer catheter tips)
90% Lactobacillus abundance in 33 patients (47.1%)
Kyono, 2018
[26]
Japan102 infertile women:
  • 79 undergoing IVF
  • 23 not undergoing IVF
  • 7 controls
EF
(IUI catheter)
90% Lactobacillus abundance in:
  • 30 IVF patients (38%)
  • 17 non-IVF patients (73.9%)
  • 6 controls (85.7%)
Wee, 2018
[27]
Australia16 infertile women; 15 controlsET
(Biopsy during hysteroscopy)
Lactobacillus more abundant in controls
Kyono, 2019
[30]
Japan92 women undergoing IVFEF
(IUI catheter)
LDM in 47 cases (51.1%); NLDM in 45 cases (48.9%)
Kitaya, 2019
[33]
Japan28 women with history of RIF (RIF group) vs. 18 infertile women undergoing IVF (non-RIF group)EF
(Curette)
Higher α-diversity in endometrium compared to vagina in both groups
LDM:
  • 18/28 (64.3%) RIF group
  • 7/18 (38.9%) infertile group
  • Gardnerella:
  • 11/28 (39.3%) RIF group
  • 5/18 (27.7%) control group
  • Burkholderia:
  • 7/28 (25%) RIF group
  • 0/18 control group
Carosso, 2020
[28]
Italy15 infertile women undergoing IVF pre- and post-COSET
(Embryo transfer catheter tips)
Lactobacillus reduced post-COS
Prevotella significantly increased post-COS
Atopobium significantly
increased post-COS
Kadogami 2020
[34]
Japan392 women with history of RIFEF
(Pipette)
LD:
  • 216/392 (55.1%)
  • NLD:
  • 176/392 (44.9%)
  • with Gardnerella (the most abundant), Atopobium, Streptococcus, and Prevotella
Riganelli, 2020
[35]
Italy 34 infertile women undergoing ARTET
(Pipelle catheter covered by the IUI catheter)
Non-pregnant women (30/34) with increased levels of Lactobacillus species and significantly increased relative abundances of Kocuria dechangensis
Diaz-Martinez, 2021
[36]
Spain48 infertile women undergoing IVFET
(Brush)
Delftia spp., Anaerobacillus spp., and Ralstonia spp. more abundant in endometrium compared to vagina
Lactobacillus spp., Gardnerella spp., Burkholderia spp., and Anaerobacillus spp. more abundant in pregnant women
Streptococcus spp., Ralstonia spp., Prevotella spp., and Delftia spp. more abundant in
non-pregnant women
In RIF patients:
  • Higher abundance of Prevotella, Lactobacillus helveticus, and Sneathia amnii
  • Lower abundance of Ralstonia
Ichiyama, 2021
[37]
Japan 145 women with history of RIF 21 controls ET
(Pipette)
14 genera, including Atopobium, Burkholderia, Delftia, Gardnerella, and Prevotella, in RIF group
vs. Lactobacillus abundances similar in RIF and control groups
Chen 2021,
[38]
China94 infertile women undergoing IVF:
  • 25 with CE
  • 69 without CE
EF
(IUI catheter)
Women affected by chronic endometritis (CE):
  • Lower clinical pregnancy rate compared to non-CE subjects
  • High abundances of
  • Actinobacteria and
  • Fusobacteria
  • Higher relative abundances of Gardnerella in non-pregnant subgroup
Similar relative abundances of Lactobacillus between pregnant and non-pregnant subgroups
Moreno, 2022
[29]
Spain, USA, Turkey, Canada, Japan, Mexico, Malaysia, Argentina342 infertile women undergoing IVFET and EF
(Cannula of Cornier and catheter, respectively)
Lactobacillus strongly correlated with LBR
Gardnerella, Klebsiella, and Streptococcus significantly more abundant in non-pregnant patients
Klebsiella and Staphylococcus correlated with clinical miscarriage
Reschini, 2022
[31]
Italy53 women undergoing IVFEF
(Double-lumen embryo transfer catheter)
Lactobacillus most represented in 16 patients (30%)
90% Lactobacillus abundance in 4 patients (8%)
Higher endometrial biodiversity among pregnant women
Bednarska-Czerwińska, 2022
[39]
Poland142 infertile women undergoing IVFEF
(Endometrial swab)
22 bacterial strains identified:
  • 11 physiological strains (57%), with Lactobacillus most common
  • 8 pathological strains (33%), with Enterobacteriaceae most common
  • 3 pathological and physiological strains
Sezer, 2022
[40]
Turkey26 women with unexplained infertility vs. 26 controls (age-matched) EF
(Endometrial swabs)
QLac/TBM (cutoff of 70.5%) had sensitivity of 84.6% in the infertility diagnosis
Kitaya, 2022
[41]
Japan117 women with history of RIF vs. 55 infertile women (non-RIF group)EF
(Endometrial curette biopsy)
NLDM:
  • 61/117 (52.1%) RIF group
  • 30/55 (54.5%) non-RIF group
  • After lactoferrin supplementation:
  • Higher clinical pregnancy rate and LBR
  • Similar miscarriage rate between the improved * VS/EF microbiota group and the unimproved VS/EF microbiota group
Keburiya, 2022
[42]
Russia130 infertile women undergoing IVF:
  • 39 with the first attempt (group I)
  • 27 with RIF following
  • embryo transfer with ovarian stimulation (group II)
  • 64 with RIF following
frozen-thawed embryo transfer (group III)
ET
(Embryo transfer catheter tips)
Group I:
  • Lactobacilli in moderate or high concentrations and opportunistic microorganisms (G. vaginalis) in moderate or high titers did not significantly affect embryo implantation
Group II:
  • Lower frequency of Lactobacilli, and low and moderate concentrations of opportunistic microorganisms (obligate anaerobes) compared to the first group
Group III:
  • Moderate concentrations of streptococci, enterobacteria, and especially strict anaerobes and Gardnerella
Acronyms: IVF = in vitro fertilization; ET = endometrial tissue; EF = endometrial fluid; LBR = live birth rate; LDM = Lactobacillus-dominated microbiota (>90% Lactobacillus spp); NLDM = non-Lactobacillus-dominated microbiota (<90% Lactobacillus spp. with >10% of other bacteria) [24]; IUI = intrauterine insemination; RIF = repeated implantation failure; COS = controlled ovarian stimulation; LD = Lactobacillus dominant (according to this study [34], >90% of the microbiota represented by Lactobacillus and Bifidobacterium); ART = assisted reproductive technology; CE = chronic endometritis; QLac = quantitative of Lactobacillus; QLac/TBM = ratio between QLac and total bacterial mass; vs. = vaginal secretions. * The increase (10% or more) in Lactobacillus species in microbiota composition [41].

2.3. Microbiome Sampling Direct from the Uterine Cavity during Surgery

The Lactobacillus dominance has also been questioned in different studies in which endometrial sampling was achieved using surgical procedures with direct access to the uterine cavity (open or laparoscopic hysterectomy, hysteroscopy, cesarean section, etc.).
Indeed, Acinetobacter, Pseudomonas, Sphingobium, and Vagococcus were most abundant in 80 endometrial samples from Chinese women undergoing surgery for conditions not related to infection [10]. Another study including 137 Chinese women detected Moraxellaceae, Propionibacteriaceae, Pseudomonadaceae, and Streptococcaceae in the uterine cavity [43]. Later, in 25 Italian females selected for hysterectomy for fibroids, Acinetobacter, Cloacibacterium, Comamonadaceae, and Pseudomonas were dominant in the endometrial microbiota [44]. Regarding sampling during cesarean section, Leoni et al. found a common endometrial microbiota composition among 19 European women [45]. The authors assumed that microbes such as Acinetobacter, Corynebacterium, Cutibacterium, Escherichia, Staphylococcus, and Streptococcus could establish an endometrial microbiome core, while Lactobacillus was found with an abundance rate below 16% [45]. In another study, endometrial microbiome analysis in 10 patients at the time of caesarean delivery showed Escherichia, Acinetobacter, Lactobacillus, and Bacillus as the most frequently detected taxa [46].

2.4. First Meta-Transcriptomic Analysis

More recently, Sola-Leyva et al. [47] analyzed the uterine microbiome in seven healthy women using the meta-transcriptomic technique. The hypothesis was that DNA sequencing could detect live and dead microbes whose genetic material persisted, while RNA detection (transcriptome) could find only microorganisms still alive in the endometrium. They found 85% of bacteria, 10% of fungi, 5% of viruses, and 0.3% of archaea; among the bacteria, they found that, at least at the level of live microorganisms, Lactobacillus were not predominant in the endometrium of healthy young women in the mid-luteal phase, while Clostridium botulinum, Hydrogenophaga sp., Klebsiella pneumoniae, and Pasteurella multocida were the most abundant microorganisms, as shown in Figure 1.

2.5. Age and Hormonal Influence on Endometrial Microbiome Composition

The contrasting findings in terms of endometrial microbiome compositions can be explained not only by the different sites and methods of sampling, but also by the physiological changes in bacterial communities over the female lifespan. Indeed, several factors, such as age, hormonal changes, ethnicity, and intrauterine devices, can modulate the microbiome [48].
Regarding the age factor, Wang et al. studied endometrial and vaginal microbiomes in 145 women and found that in the uterine cavity the “alpha diversity” (e.g., microbial diversity within a single sample) was higher in the youngest women and tended to be slightly lower with advancing age. Conversely, the “beta diversity” (e.g., variability between communities) was lower among patients younger than 20 years old, while the greatest interindividual differences were found in patients aged 41–60 years [49].
Nevertheless, the endometrial microbiome can be influenced by hormonal changes during different menstrual cycle phases. Indeed, according to Pelzer and colleagues, Prevotella spp. could be considered the hallmark of the proliferative phase, while Sneathia spp. could be considered the hallmark of the secretory phase, as shown in Figure 2 [50]. In another study, Lactobacillus seemed to be detected in lower proportions after menstruation and to progressively increase during the follicular phase, with a peak during the luteal phase [34]. In addition, the endometrial microbiome seemed not to be influenced by hormonal changes during the phase of endometrial receptivity, as no differences were found among bacterial communities in endometrial samples collected 2 and 7 days after the luteinizing hormone (LH) peak [24]. Finally, according to the latest publications, the endometrial microbiome is more transcriptionally active in the mid-secretory phase compared to the proliferative one [47].
In addition, endometrial microbial composition can also change during treatment with exogenous hormones. Oral contraceptives and intrauterine devices, such as copper and levonorgestrel-releasing systems, seemed to change vaginal microbiome composition [51,52]. Similarly, it has been demonstrated that exogenous progestin induced decreasing diversity in Lactobacillus spp. phylotypes [50], while controlled ovarian stimulation and exogenous progesterone could modify both the diversity of bacterial taxa and bacterial abundance in the vaginal and endometrial microbiota [28].

3. Endometrial Receptivity and Immune Tolerance in Pregnancy

3.1. Introduction

There are several factors that contribute to a successful pregnancy, and embryo implantation is probably one of the most complicated processes, due to the intricate regulation of combined mediators, such as cytokines, lipids, adhesion molecules, and growth factors. To provide implantation, the endometrial tissue undergoes morphological changes during the mid-secretory phase of the menstrual cycle, also known as the implantation window. Then, the endometrium becomes “receptive” to guarantee the blastocyst’s attachment to the endometrial epithelial cells and to lead the invasion of the endometrial stroma and vasculature [53]. After implantation, because of the bidirectional cellular transport across the maternal–fetal interface, fetal antigens and fetal MHC molecules, which are a combination of both mother (self) and father (foreign), are constantly exposed to maternal circulation. Nevertheless, no maternal immune response against the fetus is triggered.

3.2. Immune Cells and Cytokine Involvement According to Reproductive Phase and Gestational Age

The mechanisms of this immune tolerance during pregnancy have been largely investigated. Surprisingly, the immune system is not totally quiescent but in a constant modulation during the entire pregnancy, involving cellular populations that differ according to the different gestational periods, as summarized in Table 2.

3.3. Uterine NK Cells

Other important immune agents include uterine NK (uNK) cells, which account for 60~90% of the decidual immune cells in the early gestational age and then decrease in the middle and late gestation periods [71,72]. The role of peripheral NK cells is the cytolysis of cells lacking self MHC-I, so in the uterus these cells could potentially be responsible for fetal rejection [73]. However, thanks to the different cell-surface receptors expressed by uNKcells and atypical MHC expression by the fetal trophoblast, the cytolysis is inhibited [74,75]. Nevertheless, during implantation, uNK cells are involved in the process of uterine spiral artery remodeling, which is also assisted by endometrial chemokines, such as LIF and IL11, and by cytokines produced by CD8+ T-cells, such as IL-8 and interferon γ (IFNγ) [55,63].
The activity of macrophages and uDCs, as described in Table 2, consists of suppressing local inflammatory responses by CD8+ T-cells against fetal antigens [76], inducing apoptosis of the CD8+ T-cells thanks to Trp depletion and promoting CD4+ T-cell differentiation into Treg cells [77].

3.4. Fetal Escape from Alloantigen Response

There are different mechanisms by which the embryo can escape from the alloantigen immune response. As previously explained, most of the polymorphic MHC class Ia antigens are lacking on the surface of trophoblast cells. Similarly, MHC class II antigens are not expressed. In this context, there is a general down-regulation of most of the MHC class Ia and class II molecules just before implantation occurs [78]. However, the maternal immune tolerance is also maintained when the paternal MHC is artificially re-expressed [79]. This could be explained by the prevalent indirect allogen recognition mediated by maternal antigen-presenting-cells (APCs), bypassing the direct recognition of intact fetal MHC molecules [80,81]. In the uterine environment, thanks to the decidualization process, the maternal APCs, and especially DCs, reduce in number and the remaining ones are less specialized during T-cell activation [60,82,83]. Another factor directly secreted by the embryo is preimplantation factor (PIF); it was demonstrated in several studies that it can modulate immune responses at different levels [84]. It has a major effect on the adaptive immune responses, by binding ligands to CD14 monocytes and neutrophils and to T- and B-cells, enhancing the Th2/Th1 cytokine ratio, and also by reducing IFNγ and stimulating IL-10 secretion. PIF also appears to modulate gene expression, thereby modifying oxidative stress, protein misfolding, and platelet activation [84]. Furthermore, PIF has an effect on the innate immune response, activating mainly the Toll-like receptor (TLR)-4 mediated NALP3 inflammasome complex, reducing IL-1β, IL-18, and IL-33 [85,86] and blocking the release of nitric oxide by macrophages activated by lipopolysaccharides (LPS) [87].

4. How Could Microbiota Modulate Immune Tolerance during Pregnancy?

4.1. Endometrial Microbiome during Embryo Implantation

In recent years, different reviews have summarized the current literature, with the aim of highlighting the endometrial microbial alterations in infertile women [29,48] and in women affected by gynecological disorders, such as endometriosis, endometrial hyperplasia, endometrial cancer, and chronic endometritis [88]. Compared to these previous studies, one of the objectives of our review was to emphasize the role of the endometrial microbiome not only in reproductive and obstetrical disorders, but mainly in physiological processes, such as the fine regulation of the immune balance needed for implantation.
Therefore, thanks to knowledge of the gut and in vitro and in vivo models, a hypothesis could be postulated. First, uterine bacteria or bacterial fragments can influence endometrial receptivity, inducing an inflammatory immune response. Indeed, endometrial epithelial cells represent a physical barrier blocking pathogen invasion, and they can produce antimicrobial peptides (AMPs), which have been linked to key regulatory processes in implantation [89]. In addition, it was demonstrated in a study on human endocervical epithelial cell cultures that commensal bacteria can induce membrane-associated mucin gene expression (MUC1, MUC4, and MUC16) and stabilize the adherent junctions and tight junctions [90].

4.2. Role of Bacteroides Fragilis

Relevant evidence was found regarding the role of Bacteroides (B.) fragilis. B. fragilis is an important Gram-negative anaerobic bacterium commonly found in the lower gastrointestinal tract; however, as stated previously, it belongs to the endometrial microbiome in non-pregnant women [91]. In germ-free mice colonized with B. fragilis strain NCTC9343, this bacterium has been shown to increase the CD4+ population thanks to polysaccharide A (PSA) secretion. Furthermore, PSA activated a signaling link to TLR2 that led to the differentiation of Th1 cells and the establishment of an appropriate Th1/Th2 balance [92]. For this reason, it could be hypothesized that PSA expressed by Bacteroides plays a role in endometrial receptivity due to the immunomodulatory effect. Recently, another study demonstrated in a mouse model that PSA can enhance TLR2 activation at the intestinal mucosal barrier and promote Foxp3+ Treg cells for immunologic tolerance [93].

4.3. Role of Lactobacilli

Another notable component of the endometrial microbiome in healthy women of childbearing age, although not representing more than 90% of the described bacterial taxa, as in the lower genital tract, is Lactobacillus. It is well known that lactic acid can inhibit the production of pro-inflammatory cytokines and chemokines through Toll-like receptor (TLR) activation [94,95] Furthermore, L. delbrueckii and L. rhamnosus seem to decrease the expression of HLA-DR, CD86, CD80, CD83, and IL-12 and to increase the expression of IL-10, IL-2, and IDO in immature DC cultures derived from healthy patients [96]. In conclusion, Lactobacilli could have a tolerogenic effect on the phenotypes of DCs during the maturation process. Similarly, another group demonstrated that L. rhamnosus GR-1 could inhibit pro-inflammatory gene expression in bovine endometrial epithelial cell cultures pre-treated with Escherichia coli, while an insignificant difference was found in the group treated with L. rhamnosus GR-1 alone [97]. Therefore, lactic acid could be an important immune modulator, acting at different levels in the immune response [98]. In particular, several studies on the potential role of Lactobacilli in preventing preterm birth in animal models have been conducted. Yang et al. found that the intraperitoneal administration of the Lactobacillus rhamnosus GR-1 supernatant in pregnant CD1 mice previously treated with an intrauterine injection of LPS via minilaparotomy significantly reduced the rate of preterm-birth (PTB) induced by LPS [99]. These findings were consistent, reporting decreases in anti-inflammatory cytokines generally related to human and murine PTB, in maternal plasma, myometrium, placenta, and amniotic fluid samples [99]. Later, Kim et al. demonstrated that the suppression of proinflammatory cytokines by L. rhamnosus is due to its unique capability to produce heat-resistant but trypsin-sensitive factors in the human myometrial cell line exposed to LPS [100]. Another study evaluated the protective role of Lactobacillus kefiri (Lk48) on PTB. Indeed, the oral prophylactic administration of Lk48 before and during pregnancy effectively reduced LPS-induced PTB and stillbirth in a mouse model. Furthermore, 18 h after the LPS injection, no sign of inflammatory response was found in a histological analysis of endometrial tissues of Lk48-treated mice as compared to controls. Moreover, significantly lower percentages of CD8+ T-cells were found in a flow cytometry analysis of uterus and decidua samples from Lk48-treated mice [101].

5. Endometrial Microbiota Dysbiosis and Adverse Reproductive Outcomes

In recent years, there has been a growing interest in the impact of the endometrial microbiota on reproductive outcomes.

5.1. Recurrent Implantation Failure (RIF)

By analyzing a great number of studies on the topic, which are reported in detail in Table 1, we concluded that a unique consensus on the endometrial microbes related to RIF has not been established yet and that a further evaluation to correlate the inflammatory uterine environment, which is known to be involved in female infertility [102,103] and endometrial dysbiosis, is needed (see Section 2.2).

5.2. Recurrent Pregnancy Loss (RPL)

Recurrent pregnancy loss (RPL), defined as the loss of two or more pregnancies [104], is a pregnancy complication with an incidence range of 2–5% in couples trying to conceive [105].
Although several risk factors have been associated with RPL, in more than 50% of women RPL is idiopathic [106].
Peuranpää et al. studied endometrial and vaginal microbiota in 47 women with a history of two or more consecutive miscarriages, of whom 36 had experienced unexplained RPL. They found that in RPL patients Lactobacillus crispatus was significantly less abundant and that L. jensenii and G. vaginalis were more abundant compared with the control group, while no significant difference was found for Escherichia coli, Blautia spp., or Faecalibacterium spp. [107]. Furthermore, Lactobacillus iners represented the dominant endometrial bacterium in RPL patients, and, from the fungal analysis, Candida parapsilosis was detected only in endometrial samples of RPL patients, with an average relative abundance rate of 18.2% [107]. Then, another study evaluated 25 RPL patients and found significant differences in the beta diversity levels of microbiota from uterine lavage fluid samples between cases and controls [108]. In addition, the microbiota of the endometrial tissue of the RPL group showed a lower abundance of Lactobacillus spp., and, among the Proteobacteria, Acinetobacter spp. were predominant [108]. Interestingly, when analyzing human Th1/Th2/Th17 cytokines in uterine lavage fluid, the expression levels of IFN-γ and IL-6 were significantly inferior in the RPL group and statistically significant correlations between Aliihoeflea and IL-17A, Acinetobacter and IFN-γ, Serratia and TNF, and Staphylococcus and Serratia and IL-6 were found [108]. Lastly, in a study by Masucci et al., endometrial and vaginal microbiota were analyzed in 40 RPL patients, of whom 15 were HLA-DQ2/DQ8-positive and 25 were HLA DQ2/DQ8-negative, to investigate the pathogenic effect of celiac disease on microbiota [109]. A higher endometrial microbial diversity was found among control groups compared to RPL, with or without genetic predisposition. Furthermore, the Lactobacillus abundance levels were higher in RPL patients (66.2% in HLA-DQ2/DQ8-negative and 72.62% in HLA-DQ2/DQ8-positive patients) compared to controls (48.4%), and L. iners was much higher in both subgroups of RPL women compared to the control group, while L. acidophilus was almost absent [109]. Finally, Gardnerella was more represented in RPL HLA-DQ2/DQ8-positive patients, and Atopobium spp. was detected only in RPL patients, with or without genetic celiac predisposition [109].

5.3. Preterm Birth (PTB)

Analyzing the most recent literature on the correlation between microbiome composition and PTB, great attention has been paid to vaginal microbiome alterations rather than to the endometrial environment. According to a recent overview, conflicting results have been reported due to methodological heterogeneity, the ethnic backgrounds of the included patients, and the associated risk of PTB [110]. To our knowledge, there are no available studies that have directly related endometrial microbiome alterations to PTB. Indeed, due to the cervical shortening in preterm labor or the preterm premature rupture of membranes, the endometrial and vaginal environments have often been considered comparable.

6. Methodological and Sampling Considerations

6.1. Introduction

The last aim of this review is to provide some practical guidelines for physicians working on endometrial microbial analyses. Indeed, the limitations of the previously mentioned studies have already been noted in a recent review [88]. The most ambitious—but fundamental—objective is to standardize the methods used to compare the results of future studies. There are two major challenges in obtaining such results: first, the low microbial biomass of the uterus (especially when compared to the cervix and vagina); second, the high risk of contamination during sampling. To overcome these intrinsic limitations of the uterine microbiome, choosing the correct sampling method is key to reducing this risk. As an alternative to the conventional Pipelle, the preferred instruments used to minimize contamination are the following:
-
A double-sheathed embryo transfer catheter;
-
An intrauterine insemination catheter;
-
A transcervical sheathed brush device.
Lastly, to reduce contamination, the use of appropriate controls is fundamental [111,112]. As has been well explained by Eisenhofer et al. [112], three types of negative controls are recommended to adequately detect contaminants:
(1)
Sampling blank controls, to allow the detection of contaminant DNA introduced during the sampling procedure (e.g., swabs and plastic bags) and any tools used to store or to transport the samples from the sampling site to the laboratory;
(2)
DNA-extraction blank controls, to monitor contaminant DNA contents in the process from extraction to sequencing;
(3)
No-template amplification controls, to monitor contaminant DNA in the process of library preparation and sequencing.
On the other hand, two types of positive controls are recommended:
(1)
DNA-extraction positive controls, to monitor DNA-extraction efficiency: the advice is to use a positive control of known concentration that is relevant to your study and experimental questions;
(2)
Positive amplification controls, using a titration of DNA from a known organism type to be processed during the library preparation step.

6.2. Fluid or Tissue?

In 2018, Liu et al. [113] raised doubts regarding whether the endometrial fluid microbiome and endometrial tissue microbiota fully correspond. Specifically, they identified fewer taxa per 1000 sequencing reads from fluid compared to tissue samples. Further, they reported lower diversity in fluid compared to tissue samples. In conclusion, the endometrial fluid only partially reflects the endometrial microbiome, and a possible explanation for this finding is that the bacteria on the superficial layer of the endometrium are different from those deeper in the endometrial tissue, including the stroma. Thus, it is suggested to include both when performing a study on the endometrial microbiome.

6.3. Storing Methods

Snap-freezing and direct freezing at −80 °C are the gold standards [111]. As an alternative, it is suggested to use storage media that can stabilize nucleic acids at higher temperatures. It is suggested to avoid repeated sequences of “freezing–thawing”.

6.4. Study Planning

Moving from sampling methods to study planning, Molina et al. [111], in 2021, described a list of considerations to follow before starting a study on endometrial microbiota.
Regarding the study design, these are four strong suggestions:
  • To take samples at identical time points;
  • To take samples longitudinally from the same subject (especially if in pregnancy);
  • To use PERMANOVA for the statistical analysis;
  • To make publicly available the following details: the characteristics of the study population, the sample type, the collection method, and the data processing and analysis workflow.
Regarding the study population, especially in those studies involving reproductive outcomes, it seems particularly important to distinguish the phase of the menstrual cycle, since the uterine microbiota undergoes deep changes due to hormonal input [47]. In this context, it seems particularly relevant to take samples at identical time points, as specified above. Further, it also seems relevant to specify parity, since it has been proved that nulliparas and multiparas present different uterine microbiota compositions [48].

6.5. Ethical Considerations

Pain and discomfort frequently accompany the insertion of a catheter in the uterine cavity, both during and after the procedure. Taking nonsteroidal anti-inflammatory drugs, such as ibuprofen, before the procedure can significantly reduce the discomfort. Endometrial sampling should not be performed in case of pregnancy. All patients with the potential for pregnancy should be tested prior to proceeding with endometrial sampling.

7. Conclusions

In the last two decades, an increasing amount of evidence regarding uterine microbiota composition has become available, but a consensus on the endometrial core composition and its relation to the physiological mechanisms that lead to a successful pregnancy has still to be reached. Furthermore, more studies with the aim of unraveling the link between the uterine microbiota, maternal immune response, and clinical reproductive and obstetrical outcomes are needed. The future challenge will be to analyze microbiota in larger patient cohorts and according to standardized methods for sampling, comparative analysis, and interpretation. The aim is to reduce the large amount of variability in results and to understand the microbial roles in various clinical disorders.

Author Contributions

Writing—original draft preparation, A.I. and E.Z.; writing—review and editing, A.I., E.Z., A.G. and M.D.; supervision, N.D.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created.

Acknowledgments

Figures were created with Biorender.com.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. NIH HMP Working Group; Peterson, J.; Garges, S.; Giovanni, M.; McInnes, P.; Wang, L.; Schloss, J.; Bonazzi, V.; McEwen, J.; Wetterstrand, K.; et al. The NIH Human Microbiome Project. Genome Res. 2009, 19, 2317–2323. [Google Scholar] [CrossRef] [PubMed]
  2. Heinonen, P.K.; Teisala, K.; Punnonen, R.; Miettinen, A.; Lehtinen, M.; Paavonen, J. Anatomic sites of upper genital tract infection. Obstet. Gynecol. 1985, 66, 384–390. Available online: https://pubmed.ncbi.nlm.nih.gov/3160987/ (accessed on 24 November 2022). [PubMed]
  3. Sirota, I.; Zarek, S.M.; Segars, J.H. Potential Influence of the Microbiome on Infertility and Assisted Reproductive Technology. Semin. Reprod. Med. 2014, 32, 35. [Google Scholar] [CrossRef] [PubMed]
  4. Macpherson, A.J.; de Agüero, M.G.; Ganal-Vonarburg, S.C. How nutrition and the maternal microbiota shape the neonatal immune system. Nat. Rev. Immunol 2017, 17, 508–517. [Google Scholar] [CrossRef]
  5. Hooper, L.; Littman, D.R.; Macpherson, A.J. Interactions between the microbiota and the immune system. Science 2012, 336, 1268–1273. [Google Scholar] [CrossRef]
  6. Agostinis, C.; Mangogna, A.; Bossi, F.; Ricci, G.; Kishore, U.; Bulla, R. Uterine Immunity and Microbiota: A Shifting Paradigm. Front. Immunol. 2019, 10, 2387. [Google Scholar] [CrossRef]
  7. Critchley, H.O.D.; Babayev, E.; Bulun, S.E.; Clark, S.; Garcia-Grau, I.; Gregersen, P.K.; Kilcoyne, A.; Kim, J.-Y.J.; Lavender, M.; Marsh, E.E.; et al. Menstruation: Science and society. Am. J. Obstet. Gynecol. 2020, 223, 624–664. [Google Scholar] [CrossRef]
  8. Ilicic, M.; Zakar, T.; Paul, J.W. The Regulation of Uterine Function during Parturition: An Update and Recent Advances. Reprod. Sci. 2020, 27, 3–28. [Google Scholar] [CrossRef]
  9. Moreno, I.; Simon, C. Relevance of assessing the uterine microbiota in infertility. Fertil. Steril. 2018, 110, 337–343. [Google Scholar] [CrossRef]
  10. Chen, C.; Song, X.; Wei, W.; Zhong, H.; Dai, J.; Lan, Z.; Li, F.; Yu, X.; Feng, Q.; Wang, Z.; et al. The microbiota continuum along the female reproductive tract and its relation to uterine-related diseases. Nat. Commun. 2017, 8, 875. [Google Scholar] [CrossRef] [Green Version]
  11. Idelevich, A.; Vilella, F. Mother and Embryo Cross-Communication. Genes 2020, 11, 376. [Google Scholar] [CrossRef] [PubMed]
  12. Ticconi, C.; di Simone, N.; Campagnolo, L.; Fazleabas, A. Clinical consequences of defective decidualization. Tissue Cell 2021, 72, 101586. [Google Scholar] [CrossRef] [PubMed]
  13. Benner, M.; Ferwerda, G.; Joosten, I.; van der Molen, R.G. How uterine microbiota might be responsible for a receptive, fertile endometrium. Hum. Reprod. Update 2018, 24, 393–415. [Google Scholar] [CrossRef]
  14. Rashid, N.A.; Lalitkumar, S.; Lalitkumar, P.G.; Gemzell-Danielsson, K. Endometrial receptivity and human embryo implantation. Am. J. Reprod. Immunol. 2011, 66 (Suppl. 1), 23–30. [Google Scholar] [CrossRef] [PubMed]
  15. Hashimoto, T.; Kyono, K. Does dysbiotic endometrium affect blastocyst implantation in IVF patients? J. Assist. Reprod. Genet. 2019, 36, 2471–2479. [Google Scholar] [CrossRef] [PubMed]
  16. Kayama, H.; Okumura, R.; Takeda, K. Interaction Between the Microbiota, Epithelia, and Immune Cells in the Intestine. Annu. Rev. Immunol. 2020, 38, 23–48. [Google Scholar] [CrossRef]
  17. Fu, M.; Zhang, X.; Liang, Y.; Lin, S.; Qian, W.; Fan, S. Alterations in Vaginal Microbiota and Associated Metabolome in Women with Recurrent Implantation Failure. mBio 2020, 11, e03242-19. [Google Scholar] [CrossRef]
  18. Laniewski, P.; Gomez, A.; Hire, G.; So, M.; Herbst-Kralovetz, M.M. Human Three-Dimensional Endometrial Epithelial Cell Model to Study Host Interactions with Vaginal Bacteria and Neisseria gonorrhoeae. Infect. Immun. 2017, 85, e01049-16. [Google Scholar] [CrossRef]
  19. Pekmezovic, M.; Mogavero, S.; Naglik, J.R.; Hube, B. Host-Pathogen Interactions during Female Genital Tract Infections. Trends Microbiol. 2019, 27, 982–996. [Google Scholar] [CrossRef]
  20. Eschenbach, D.A.; Rosene, K.; Tompkins, L.S.; Watkins, H.; Gravett, M.G. Endometrial cultures obtained by a triple-lumen method from afebrile and febrile postpartum women. J. Infect. Dis. 1986, 153, 1038–1045. [Google Scholar] [CrossRef]
  21. Hemsell, D.L.; Obregon, V.L.; Heard, M.C. Endometrial bacteria in asymptomatic, nonpregnant women. J. Reprod. Med. 1989, 34, 872–874. Available online: https://europepmc.org/article/med/2585386 (accessed on 30 November 2022).
  22. Mitchell, C.M.; Haick, A.; Nkwopara, E.; Garcia, R.; Rendi, M.; Agnew, K.; Fredricks, D.N.; Eschenbach, D. Colonization of the upper genital tract by vaginal bacterial species in nonpregnant women. Am. J. Obstet. Gynecol. 2015, 212, 611.e1–611.e9. [Google Scholar] [CrossRef] [PubMed]
  23. Franasiak, M.F.; Werne, M.D.; Juneau, C.R.; Tao, X.; Landis, J.; Zhan, Y.; Treff, N.R.; Scott, R.T. Endometrial microbiome at the time of embryo transfer: Next-generation sequencing of the 16S ribosomal subunit. J. Assist. Reprod. Genet. 2016, 33, 129–136. [Google Scholar] [CrossRef] [PubMed]
  24. Moreno, I.; Codoñer, F.; Vilella, F.; Valbuena, D.; Martinez-Blanch, J.; Jimenez-Almazán, J.; Alonso, R.; Alamá, P.; Remohí, J.; Pellicer, A.; et al. Evidence that the endometrial microbiota has an effect on implantation success or failure. Am. J. Obstet. Gynecol. 2016, 215, 684–703. [Google Scholar] [CrossRef]
  25. Tao, X.; Franasiak, J.M.; Zhan, Y.; Scott, R.T., III; Rajchel, J.; Bedard, J.; Newby, R., Jr.; Scott, R.T.; Treff, N.R.; Chu, T. Characterizing the endometrial microbiome by analyzing the ultra-low bacteria from embryo transfer catheter tips in IVF cycles: Next generation sequencing (NGS) analysis of the 16S ribosomal gene. Hum. Microb. J. 2017, 3, 15–21. [Google Scholar] [CrossRef]
  26. Kyono, K.; Hashimoto, T.; Nagai, Y.; Sakuraba, Y. Analysis of endometrial microbiota by 16S ribosomal RNA gene sequencing among infertile patients: A single-center pilot study. Reprod. Med. Biol. 2018, 17, 297–306. [Google Scholar] [CrossRef]
  27. Wee, B.A.; Thomas, M.; Sweeney, E.L.; Frentiu, F.D.; Samios, M.; Ravel, J.; Gajer, P.; Myers, G.; Timms, P.; Allan, J.; et al. A retrospective pilot study to determine whether the reproductive tract microbiota differs between women with a history of infertility and fertile women. Aust. N. Z. J. Obstet. Gynaecol. 2018, 58, 341–348. [Google Scholar] [CrossRef]
  28. Carosso, A.; Revelli, A.; Gennarelli, G.; Canosa, S.; Cosma, S.; Borella, F.; Tancredi, A.; Paschero, C.; Boatti, L.; Zanotto, E.; et al. Controlled ovarian stimulation and progesterone supplementation affect vaginal and endometrial microbiota in IVF cycles: A pilot study. J. Assist. Reprod. Genet. 2020, 37, 2315–2326. [Google Scholar] [CrossRef]
  29. Moreno, I.; Garcia-Grau, I.; Perez-Villaroya, D.; Gonzalez-Monfort, M.; Bahçeci, M.; Barrionuevo, M.J.; Taguchi, S.; Puente, E.; Dimattina, M.; Lim, M.W.; et al. Endometrial microbiota composition is associated with reproductive outcome in infertile patients. Microbiome 2022, 10, 1. [Google Scholar] [CrossRef]
  30. Kyono, K.; Hashimoto, T.; Kikuchi, S.; Nagai, Y.; Sakuraba, Y. A pilot study and case reports on endometrial microbiota and pregnancy outcome: An analysis using 16S rRNA gene sequencing among IVF patients, and trial therapeutic intervention for dysbiotic endometrium. Reprod. Med. Biol. 2019, 18, 72–82. [Google Scholar] [CrossRef]
  31. Reschini, M.; Benaglia, L.; Ceriotti, F.; Borroni, R.; Ferrari, S.; Castiglioni, M.; Guarneri, D.; Porcaro, L.; Vigano’, P.; Somigliana, E.; et al. Endometrial microbiome: Sampling, assessment, and possible impact on embryo implantation. Sci. Rep. 2022, 12, 8467. [Google Scholar] [CrossRef] [PubMed]
  32. Moore, D.E.; Soules, M.R.; Klein, N.A.; Fujimoto, V.Y.; Agnew, K.J.; Eschenbach, D.A. Bacteria in the transfer catheter tip influence the live-birth rate after in vitro fertilization. Fertil. Steril. 2000, 74, 1118–1124. [Google Scholar] [CrossRef] [PubMed]
  33. Kitaya, K.; Nagai, Y.; Arai, W.; Sakuraba, Y.; Ishikawa, T. Characterization of Microbiota in Endometrial Fluid and Vaginal Secretions in Infertile Women with Repeated Implantation Failure. Mediat. Inflamm. 2019, 2019, 4893437. [Google Scholar] [CrossRef] [PubMed]
  34. Kadogami, D.; Nakaoka, Y.; Morimoto, Y. Use of a vaginal probiotic suppository and antibiotics to influence the composition of the endometrial microbiota. Reprod. Biol. 2020, 20, 307–314. [Google Scholar] [CrossRef] [PubMed]
  35. Riganelli, L.; Iebba, V.; Piccioni, M.; Illuminati, I.; Bonfiglio, G.; Neroni, B.; Calvo, L.; Gagliardi, A.; Levrero, M.; Merlino, L.; et al. Structural Variations of Vaginal and Endometrial Microbiota: Hints on Female Infertility. Front. Cell. Infect. Microbiol. 2020, 10, 350. [Google Scholar] [CrossRef]
  36. Diaz-martínez, M.D.C.; Bernabeu, A.; Lledó, B.; Carratalá-Munuera, C.; Quesada, J.; Lozano, F.; Ruiz, V.; Morales, R.; Llácer, J.; Ten, J.; et al. Impact of the Vaginal and Endometrial Microbiome Pattern on Assisted Reproduction Outcomes. J. Clin. Med. 2021, 10, 4063. [Google Scholar] [CrossRef]
  37. Ichiyama, T.; Kuroda, K.; Nagai, Y.; Urushiyama, D.; Ohno, M.; Yamaguchi, T.; Nagayoshi, M.; Sakuraba, Y.; Yamasaki, F.; Hata, K.; et al. Analysis of vaginal and endometrial microbiota communities in infertile women with a history of repeated implantation failure. Reprod. Med. Biol. 2021, 20, 334–344. [Google Scholar] [CrossRef]
  38. Chen, W.; Wei, K.; He, X.; Wei, J.; Yang, L.; Li, L.; Chen, T.; Tan, B. Identification of Uterine Microbiota in Infertile Women Receiving in vitro Fertilization with and without Chronic Endometritis. Front. Cell. Dev. Biol. 2021, 9, 693267. [Google Scholar] [CrossRef]
  39. ABednarska-Czerwińska; Czerwiński, M.; Morawiec, E.; Łach, A.; Ziaja, A.; Kusaj, A.; Strączyńska, P.; Sagan, D.; Boroń, D.; Grabarek, B.O. Marking the Profile of the Microflora of the Endometrium and Uterine Cervix in Women as a Potential Factor Determining the Effectiveness of In Vitro Fertilization. J. Clin. Med. 2022, 11, 3348. [Google Scholar] [CrossRef]
  40. Sezer, O.; Çalışkan, C.S.; Celik, S.; Kilic, S.S.; Kuruoglu, T.; Ustun, G.U.; Yurtcu, N. Assessment of vaginal and endometrial microbiota by real-time PCR in women with unexplained infertility. J. Obstet. Gynaecol. Res. 2022, 48, 129–139. [Google Scholar] [CrossRef]
  41. Kitaya, K.; Ishikawa, T. Genital tract dysbiosis in infertile women with a history of repeated implantation failure and pilot study for reproductive outcomes following oral enteric coating lactoferrin supplementation. Arch. Gynecol. Obstet. 2022, 306, 1761–1769. [Google Scholar] [CrossRef] [PubMed]
  42. Keburiya, L.K.; Smolnikova, V.Y.; Priputnevich, T.V.; Muravieva, V.V.; Gordeev, A.B.; Trofimov, D.Y.; Shubina, E.S.; Kochetkova, T.O.; Rogacheva, M.S.; Kalinina, E.A.; et al. Does the uterine microbiota affect the reproductive outcomes in women with recurrent implantation failures? BMC Womens Health 2022, 22, 168. [Google Scholar] [CrossRef] [PubMed]
  43. Li, F.; Chen, C.; Wei, W.; Wang, Z.; Dai, J.; Hao, L.; Song, L.; Zhang, X.; Zeng, L.; Du, H.; et al. The metagenome of the female upper reproductive tract. Gigascience 2018, 7, giy107. [Google Scholar] [CrossRef] [PubMed]
  44. Winters, A.D.; Romero, R.; Gervasi, M.T.; Gomez-Lopez, N.; Tran, M.R.; Garcia-Flores, V.; Pacora, P.; Jung, E.; Hassan, S.S.; Hsu, C.-D.; et al. Does the endometrial cavity have a molecular microbial signature? Sci. Rep. 2019, 9, 9905. [Google Scholar] [CrossRef] [PubMed]
  45. Leoni, C.; Ceci, O.; Manzari, C.; Fosso, B.; Volpicella, M.; Ferrari, A.; Fiorella, P.; Pesole, G.; Cicinell, E.; Ceci, L.R. Human Endometrial Microbiota at Term of Normal Pregnancies. Genes 2019, 10, 971. [Google Scholar] [CrossRef]
  46. Younge, N.; McCann, J.R.; Ballard, J.; Plunkett, C.; Akhtar, S.; Araújo-Pérez, F.; Murtha, A.; Brandon, D.; Seed, P.C. Fetal exposure to the maternal microbiota in humans and mice. JCI Insight 2019, 4, e127806. [Google Scholar] [CrossRef]
  47. Sola-Leyva, A.; Andrés-León, E.; Molina, N.; Terron-Camero, L.C.; Plaza-Díaz, J.; Sáez-Lara, M.J.; Gonzalvo, M.C.; Sánchez, R.; Ruíz, S.; Martínez, L.; et al. Mapping the entire functionally active endometrial microbiota. Hum. Reprod. 2021, 36, 1021–1031. [Google Scholar] [CrossRef]
  48. Toson, B.; Simon, C.; Moreno, I. The Endometrial Microbiome and Its Impact on Human Conception. Int. J. Mol. Sci. 2022, 23, 485. [Google Scholar] [CrossRef]
  49. Wang, J.; Li, Z.; Ma, X.; Du, L.; Jia, Z.; Cui, X.; Yu, L.; Yang, J.; Xiao, L.; Zhang, B.; et al. Translocation of vaginal microbiota is involved in impairment and protection of uterine health. Nat. Commun. 2021, 12, 4191. [Google Scholar] [CrossRef]
  50. Pelzer, E.S.; Willner, D.; Buttini, M.; Huygens, F. A role for the endometrial microbiome in dysfunctional menstrual bleeding. Antonie Van Leeuwenhoek 2018, 111, 933–943. [Google Scholar] [CrossRef]
  51. Brooks, J.P.; Edwards, D.J.; Blithe, D.L.; Fettweis, J.M.; Serrano, M.G.; Sheth, N.U.; Strauss, J.F.; Buck, G.A.; Jefferson, K.K. Effects of combined oral contraceptives, depot medroxyprogesterone acetate and the levonorgestrel-releasing intrauterine system on the vaginal microbiome. Contraception 2017, 95, 405–413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Achilles, S.L.; Austin, M.N.; Meyn, L.A.; Mhlanga, F.; Chirenje, Z.M.; Hillier, S.L. Impact of contraceptive initiation on vaginal microbiota. Am. J. Obstet. Gynecol. 2018, 218, 622.e1–622.e10. [Google Scholar] [CrossRef] [PubMed]
  53. Governini, L.; Luongo, F.P.; Haxhiu, A.; Piomboni, P.; Luddi, A. Main actors behind the endometrial receptivity and successful implantation. Tissue Cell 2021, 73, 101656. [Google Scholar] [CrossRef] [PubMed]
  54. Gnainsky, Y.; Granot, I.; Aldo, P.; Barash, A.; Or, Y.; Mor, G.; Dekel, N. Biopsy-induced inflammatory conditions improve endometrial receptivity: The mechanism of action. Reproduction 2015, 149, 75–85. [Google Scholar] [CrossRef]
  55. Soares, M.J.; Chakraborty, D.; Kubota, K.; Renaud, S.J.; Rumi, M.A.K. Adaptive mechanisms controlling uterine spiral artery remodeling during the establishment of pregnancy. Int. J. Dev. Biol. 2014, 58, 247–259. [Google Scholar] [CrossRef]
  56. Tilburgs, T.; Roelen, D.L.; van der Mast, B.J.; de Groot-Swings, G.M.; Kleijburg, C.; Scherjon, S.A.; Claas, F. Evidence for a selective migration of fetus-specific CD4+CD25bright regulatory T cells from the peripheral blood to the decidua in human pregnancy. J. Immunol. 2008, 180, 5737–5745. [Google Scholar] [CrossRef]
  57. Saito, S.; Nakashima, A.; Shima, T.; Ito, M. Th1/Th2/Th17 and regulatory T-cell paradigm in pregnancy. Am. J. Reprod. Immunol. 2010, 63, 601–610. [Google Scholar] [CrossRef]
  58. Svensson-Arvelund, J.; Ernerudh, J. The Role of Macrophages in Promoting and Maintaining Homeostasis at the Fetal-Maternal Interface. Am. J. Reprod. Immunol. 2015, 74, 100–109. [Google Scholar] [CrossRef]
  59. Gori, S.; Soczewski, E.; Fernández, L.; Grasso, E.; Gallino, L.; Merech, F.; Colado, A.; Borge, M.; Leirós, C.P.; Salamone, G.; et al. Decidualization Process Induces Maternal Monocytes to Tolerogenic IL-10-Producing Dendritic Cells (DC-10). Front. Immunol. 2020, 11, 1571. [Google Scholar] [CrossRef]
  60. Robertson, S.A.; Moldenhauer, L.M. Immunological determinants of implantation success. Int. J. Dev. Biol. 2014, 58, 205–217. [Google Scholar] [CrossRef]
  61. van der Zwan, A.; Bi, K.; Norwitz, E.R.; Crespo, Â.C.; Claas, F.H.J.; Strominger, J.L.; Tilburgs, T. Mixed signature of activation and dysfunction allows human decidual CD8+ T cells to provide both tolerance and immunity. Proc. Natl. Acad. Sci. USA 2018, 115, 385–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. McIntire, R.H.; Hunt, J.S. Antigen presenting cells and HLA-G—A review. Placenta 2005, 26 (Suppl. A), S104–S109. [Google Scholar] [CrossRef] [PubMed]
  63. Scaife, P.J.; Bulmer, J.N.; Robson, S.C.; Innes, B.A.; Searle, R.F. Effector activity of decidual CD8+ T lymphocytes in early human pregnancy. Biol. Reprod. 2006, 75, 562–567. [Google Scholar] [CrossRef] [PubMed]
  64. Zenclussen, A.C.; Gerlof, K.; Zenclussen, L.; Ritschel, S.; Bertoja, A.Z.; Fest, S.; Hontsu, S.; Ueha, S.; Matsushima, K.; Leber, J.; et al. Regulatory T cells induce a privileged tolerant microenvironment at the fetal-maternal interface. Eur. J. Immunol. 2006, 36, 82–94. [Google Scholar] [CrossRef] [PubMed]
  65. Rudensky, A.Y. Regulatory T cells and Foxp3. Immunol. Rev. 2011, 241, 260–268. [Google Scholar] [CrossRef] [PubMed]
  66. Shevach, E.M. CD4+ CD25+ suppressor T cells: More questions than answers. Nat. Rev. Immunol. 2002, 2, 389–400. [Google Scholar] [CrossRef] [PubMed]
  67. Ghiringhelli, F.; Ménard, C.; Terme, M.; Flament, C.; Taieb, J.; Chaput, N.; Puig, P.E.; Novault, S.; Escudier, B.; Vivier, E.; et al. CD4+ CD25+ regulatory T cells inhibit natural killer cell functions in a transforming growth factor-beta-dependent manner. J. Exp. Med. 2005, 202, 1075–1085. [Google Scholar] [CrossRef] [PubMed]
  68. Taams, L.S.; van Amelsfort, J.M.R.; Tiemessen, M.M.; Jacobs, K.M.G.; Jong, E.C.; Akbar, A.N.; Bijlsma, J.W.J.; Lafeber, F.P.J.G. Modulation of monocyte/macrophage function by human CD4+CD25+ regulatory T cells. Hum. Immunol. 2005, 66, 222–230. [Google Scholar] [CrossRef]
  69. Lim, H.W.; Hillsamer, P.; Banham, A.H.; Kim, C.H. Cutting edge: Direct suppression of B cells by CD4+ CD25+ regulatory T cells. J. Immunol. 2005, 175, 4180–4183. [Google Scholar] [CrossRef]
  70. Zhang, Y.; Wang, Y.; Li, M.Q.; Duan, J.; Fan, D.X.; Jin, L.P. IL-25 promotes Th2 bias by upregulating IL-4 and IL-10 expression of decidual γδT cells in early pregnancy. Exp. Ther. Med. 2018, 15, 1855–1862. [Google Scholar] [CrossRef]
  71. RSearle, F.; Jones, R.K.; Bulmer, J.N. Phenotypic analysis and proliferative responses of human endometrial granulated lymphocytes during the menstrual cycle. Biol. Reprod. 1999, 60, 871–878. [Google Scholar] [CrossRef] [PubMed]
  72. Vacca, P.; Moretta, L.; Moretta, A.; Mingari, M.C. Origin, phenotype and function of human natural killer cells in pregnancy. Trends Immunol. 2011, 32, 517–523. [Google Scholar] [CrossRef]
  73. Benichou, G.; Yamada, Y.; Aoyama, A.; Madsen, J.C. Natural killer cells in rejection and tolerance of solid organ allografts. Curr. Opin. Organ. Transplant. 2011, 16, 47–53. [Google Scholar] [CrossRef]
  74. Faas, M.M.; de Vos, P. Uterine NK cells and macrophages in pregnancy. Placenta 2017, 56, 44–52. [Google Scholar] [CrossRef]
  75. Ishitani, A.; Sageshima, N.; Lee, N.; Dorofeeva, N.; Hatake, K.; Marquardt, H.; Geraghty, D.E. Protein expression and peptide binding suggest unique and interacting functional roles for HLA-E, F, and G in maternal-placental immune recognition. J. Immunol. 2003, 171, 1376–1384. [Google Scholar] [CrossRef] [PubMed]
  76. Munn, D.H.; Mellor, A.L. Indoleamine 2,3 dioxygenase and metabolic control of immune responses. Trends Immunol. 2013, 34, 137–143. [Google Scholar] [CrossRef] [PubMed]
  77. Mondanelli, G.; Bianchi, R.; Pallotta, M.T.; Orabona, C.; Albini, E.; Iacono, A.; Belladonna, M.L.; Vacca, C.; Fallarino, F.; Macchiarulo, A.; et al. A Relay Pathway between Arginine and Tryptophan Metabolism Confers Immunosuppressive Properties on Dendritic Cells. Immunity 2017, 46, 233–244. [Google Scholar] [CrossRef] [PubMed]
  78. Guleria, I.; Sayegh, M.H. Maternal acceptance of the fetus: True human tolerance. J. Immunol. 2007, 178, 3345–3351. [Google Scholar] [CrossRef]
  79. Rogers, A.M.; Boime, I.; Connolly, J.; Cook, J.; Russell, J. Maternal—Fetal tolerance is maintained despite transgene—Driven trophoblast expression of MHC class I, and defects in Fas and its ligand. Eur. J. Immunol. 1998, 28, 3479–3487. [Google Scholar] [CrossRef]
  80. Erlebacher, A.; Vencato, D.; Price, K.A.; Zhang, D.; Glimcher, L.H. Constraints in antigen presentation severely restrict T cell recognition of the allogeneic fetus. J. Clin. Investig. 2007, 117, 1399–1411. [Google Scholar] [CrossRef]
  81. Erlebacher, A. Immune surveillance of the maternal/fetal interface: Controversies and implications. Trends Endocrinol. Metab. 2010, 21, 428–434. [Google Scholar] [CrossRef] [Green Version]
  82. Sakaguchi, S. Regulatory T cells: Key controllers of immunologic self-tolerance. Cell 2000, 101, 455–458. [Google Scholar] [CrossRef]
  83. Collins, M.K.; Tay, C.S.; Erlebacher, A. Dendritic cell entrapment within the pregnant uterus inhibits immune surveillance of the maternal/fetal interface in mice. J. Clin. Investig. 2009, 119, 2062–2073. [Google Scholar] [CrossRef]
  84. Barnea, E.R.; Hayrabedyan, S.; Todorova, K.; Almogi-Hazan, O.; Or, R.; Guingab, J.; McElhinney, J.; Fernandez, N.; Barder, T. PreImplantation factor (PIF*) regulates systemic immunity and targets protective regulatory and cytoskeleton proteins. Immunobiology 2016, 221, 778–793. [Google Scholar] [CrossRef]
  85. Di Simone, N.; Di Nicuolo, F.; Marana, R.; Castellani, R.; Ria, F.; Veglia, M.; Scambia, G.; Surbek, D.; Barnea, E.; Mueller, M. Synthetic PreImplantation Factor (PIF) prevents fetal loss by modulating LPS induced inflammatory response. PLoS ONE 2017, 12, e0180642. [Google Scholar] [CrossRef]
  86. Mueller, M.; Zhou, J.; Yang, L.; Gao, Y.; Wu, F.; Schoeberlein, A.; Surbek, D.; Barnea, E.R.; Paidas, M.; Huang, Y. PreImplantation factor promotes neuroprotection by targeting microRNA let-7. Proc. Natl. Acad. Sci. USA 2014, 111, 13882–13887. [Google Scholar] [CrossRef]
  87. Barnea, E.R.; Kirk, D.; Todorova, K.; McElhinney, J.; Hayrabedyan, S.; Fernández, N. PIF direct immune regulation: Blocks mitogen-activated PBMCs proliferation, promotes TH2/TH1 bias, independent of Ca(2+). Immunobiology 2015, 220, 865–875. [Google Scholar] [CrossRef]
  88. Medina-Bastidas, D.; Camacho-Arroyo, I.; García-Gómez, E. Current findings in endometrial microbiome: Impact on uterine diseases. Reproduction 2022, 163, R81–R96. [Google Scholar] [CrossRef]
  89. Frew, L.; Stock, S.J. Antimicrobial peptides and pregnancy. Reproduction 2011, 141, 725–735. [Google Scholar] [CrossRef]
  90. Radtke, A.L.; Quayle, A.J.; Herbst-Kralovetz, M.M. Microbial products alter the expression of membrane-associated mucin and antimicrobial peptides in a three-dimensional human endocervical epithelial cell model. Biol. Reprod. 2012, 87, 1–10. [Google Scholar] [CrossRef]
  91. Verstraelen, H.; Vilchez-Vargas, R.; Desimpel, F.; Jauregui, R.; Vankeirsbilck, N.; Weyers, S.; Verhelst, R.; De Sutter, P.; Pieper, D.H.; Van De Wiele, T. Characterisation of the human uterine microbiome in non-pregnant women through deep sequencing of the V1-2 region of the 16S rRNA gene. PeerJ 2016, 4, e1602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Mazmanian, S.K.; Cui, H.L.; Tzianabos, A.O.; Kasper, D.L. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 2005, 122, 107–118. [Google Scholar] [CrossRef] [PubMed]
  93. Round, J.L.; Lee, S.M.; Li, J.; Tran, G.; Jabri, B.; Chatila, T.A.; Mazmanian, S.K. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science 2011, 332, 974–977. [Google Scholar] [CrossRef] [PubMed]
  94. do Carmo, M.S.; Noronha, F.M.F.; Arruda, M.O.; Costa, P.D.S.; Bomfim, M.R.Q.; Monteiro, A.S.; Ferro, T.A.F.; Fernandes, E.S.; Girón, J.A.; Monteiro-Neto, V. Lactobacillus fermentum ATCC 23271 Displays In vitro Inhibitory Activities against Candida spp. Front. Microbiol. 2016, 7, 1722. [Google Scholar] [CrossRef] [PubMed]
  95. Rautava, S.; Collado, M.C.; Salminen, S.; Isolauri, E. Probiotics modulate host-microbe interaction in the placenta and fetal gut: A randomized, double-blind, placebo-controlled trial. Neonatology 2012, 102, 178–184. [Google Scholar] [CrossRef]
  96. Esmaeili, S.A.; Mahmoudi, M.; Rezaieyazdi, Z.; Sahebari, M.; Tabasi, N.; Sahebkar, A.; Rastin, M. Generation of tolerogenic dendritic cells using Lactobacillus rhamnosus and Lactobacillus delbrueckii as tolerogenic probiotics. J. Cell Biochem. 2018, 119, 7865–7872. [Google Scholar] [CrossRef]
  97. Liu, J.; Feng, X.; Li, B.; Sun, Y.; Jin, T.; Feng, M.; Ni, Y.; Liu, M. Lactobacillus rhamnosus GR-1 Alleviates Escherichia coli-Induced Inflammation via NF-κB and MAPKs Signaling in Bovine Endometrial Epithelial Cells. Front. Cell Infect. Microbiol. 2022, 12, 809674. [Google Scholar] [CrossRef]
  98. Li, H.; Zang, Y.; Wang, C.; Li, H.; Fan, A.; Han, C.; Xue, F. The Interaction Between Microorganisms, Metabolites, and Immune System in the Female Genital Tract Microenvironment. Front. Cell Infect. Microbiol. 2020, 10, 609488. [Google Scholar] [CrossRef]
  99. Yang, S.; Li, W.; Challis, J.R.G.; Reid, G.; Kim, S.O.; Bocking, A.D. Probiotic Lactobacillus rhamnosus GR-1 supernatant prevents lipopolysaccharide-induced preterm birth and reduces inflammation in pregnant CD-1 mice. Am. J. Obstet. Gynecol. 2014, 211, 44.e1–44.e12. [Google Scholar] [CrossRef]
  100. Kim, B.; Shynlova, O.; Lye, S. Probiotic Lactobacillus rhamnosus GR-1 is a unique prophylactic agent that suppresses infection-induced myometrial cell responses. Sci. Rep. 2019, 9, 4698. [Google Scholar] [CrossRef]
  101. Ventimiglia, M.S.; Valeff, N.J.; Albán, M.P.; Paturlanne, J.M.; Juriol, L.; Quadrana, F.; Cecotti, M.; Malamud, M.; Dibo, M.J.; Serradell, M.D.L.; et al. Probiotic Lactobacillus kefiri prevents endotoxin-induced preterm birth and stillbirth in mice. Reproduction 2021, 161, 657–667. [Google Scholar] [CrossRef] [PubMed]
  102. Azkargorta, M.; Escobes, I.; Iloro, I.; Osinalde, N.; Corral, B.; Ibañez-Perez, J.; Exposito, A.; Prieto, B.; Elortza, F.; Matorras, R. Differential proteomic analysis of endometrial fluid suggests increased inflammation and impaired glucose metabolism in non-implantative IVF cycles and pinpoints PYGB as a putative implantation marker. Hum. Reprod. 2018, 33, 1898–1906. [Google Scholar] [CrossRef]
  103. Kitaya, K.; Tada, Y.; Hayashi, T.; Taguchi, S.; Funabiki, M.; Nakamura, Y. Comprehensive endometrial immunoglobulin subclass analysis in infertile women suffering from repeated implantation failure with or without chronic endometritis. Am. J. Reprod. Immunol. 2014, 72, 386–391. [Google Scholar] [CrossRef] [PubMed]
  104. The ESHRE Guideline Group on RPL; Bender Atik, R.; Christiansen, O.B.; Elson, J.; Kolte, A.M.; Lewis, S.; Middeldorp, S.; Nelen, W.; Peramo, B.; Quenby, S.; et al. ESHRE guideline: Recurrent pregnancy loss. Hum. Reprod. Open. 2018, 2018, hoy004. [Google Scholar] [CrossRef]
  105. Van Dijk, M.M.; Kolte, A.M.; Limpens, J.; Kirk, E.; Quenby, S.; van Wely, M.; Goddijn, M. Recurrent pregnancy loss: Diagnostic workup after two or three pregnancy losses? A systematic review of the literature and meta-analysis. Hum. Reprod. Update 2020, 26, 356–367. [Google Scholar] [CrossRef]
  106. Carp, H.J. Recurrent Pregnancy Loss Causes, Controversies, and Treatment, 3rd ed.; CRC Press: London, UK, 2020; Volume 29, Available online: https://www.routledge.com/Recurrent-Pregnancy-Loss-Causes-Controversies-and-Treatment/Carp/p/book/9781138325654 (accessed on 19 January 2023).
  107. Peuranpää, P.; Holster, T.; Saqib, S.; Kalliala, I.; Tiitinen, A.; Salonen, A.; Hautamäki, H. Female reproductive tract microbiota and recurrent pregnancy loss: A nested case-control study. Reprod. Biomed. Online 2022, 45, 1021–1031. [Google Scholar] [CrossRef]
  108. Liu, F.-T.; Yang, S.; Yang, Z.; Zhou, P.; Peng, T.; Yin, J.; Ye, Z.; Shan, H.; Yu, Y.; Li, R. An Altered Microbiota in the Lower and Upper Female Reproductive Tract of Women with Recurrent Spontaneous Abortion. Microbiol. Spectr. 2022, 10, e0046222. [Google Scholar] [CrossRef] [PubMed]
  109. Masucci, L.; D’Ippolito, S.; De Maio, F.; Quaranta, G.; Mazzarella, R.; Bianco, D.M.; Castellani, R.; Inversetti, A.; Sanguinetti, M.; Gasbarrini, A.; et al. Celiac Disease Predisposition and Genital Tract Microbiota in Women Affected by Recurrent Pregnancy Loss. Nutrients 2023, 15, 221. [Google Scholar] [CrossRef]
  110. Peelen, M.J.; Luef, B.M.; Lamont, R.F.; de Milliano, I.; Jensen, J.S.; Limpens, J.; Hajenius, P.J.; Jørgensen, J.S.; Menon, R. The influence of the vaginal microbiota on preterm birth: A systematic review and recommendations for a minimum dataset for future research. Placenta 2019, 79, 30–39. [Google Scholar] [CrossRef]
  111. Molina, N.M.; Sola-Leyva, A.; Haahr, T.; Aghajanova, L.; Laudanski, P.; Castilla, J.A.; Altmäe, S. Analysing endometrial microbiome: Methodological considerations and recommendations for good practice. Hum. Reprod. 2021, 36, 859–879. [Google Scholar] [CrossRef]
  112. Eisenhofer, R.; Minich, J.J.; Marotz, C.; Cooper, A.; Knight, R.; Weyrich, L.S. Contamination in Low Microbial Biomass Microbiome Studies: Issues and Recommendations. Trends Microbiol. 2019, 27, 105–117. [Google Scholar] [CrossRef] [PubMed]
  113. Liu, Y.; Wong, K.W.K.; Ko, E.Y.-L.; Chen, X.; Huang, J.; Tsui, S.K.-W.; Li, T.C.; Chim, S.S.-C. Systematic Comparison of Bacterial Colonization of Endometrial Tissue and Fluid Samples in Recurrent Miscarriage Patients: Implications for Future Endometrial Microbiome Studies. Clin. Chem. 2018, 64, 1743–1752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Composition of the endometrial microbiome according to a recent meta-transcriptomic study by Sola-Leyva et al. [47] and description of the cervical and vaginal microbiome by Chen et al. [10].
Figure 1. Composition of the endometrial microbiome according to a recent meta-transcriptomic study by Sola-Leyva et al. [47] and description of the cervical and vaginal microbiome by Chen et al. [10].
Ijms 24 02995 g001
Figure 2. Endometrial microbiota changes under hormonal influence in different phases of the menstrual cycle [50].
Figure 2. Endometrial microbiota changes under hormonal influence in different phases of the menstrual cycle [50].
Ijms 24 02995 g002
Table 2. Immune cells and cytokine involvement according to the reproductive phase.
Table 2. Immune cells and cytokine involvement according to the reproductive phase.
TimingImmune Cell AssetsCytokines
Mid-ovulatory Increasing levels of:
  • IL-6
  • IL-8
  • IL-15
  • GM-CSF
  • TNF-α [54]
ImplantationIncreasing numbers of NK cells [55] (progesterone-mediated). Increasing numbers of Treg cells [56,57]. Shift to M2 macrophages [58], induced by growth factors released by the trophoblast uDCs expressing fewer CD83 and CD86 costimulatory molecules [59]Thanks to progesterone, decreasing levels of GM-CSF and IL-1 [60] and increasing levels of IL-8 [60]. Thanks to estrogen, increasing levels of:
  • CCL-3
  • CCL-4
  • CCL-5 [61]
Thanks to M2 macrophages, increase in PGE2 and IL10 [62]. For uterine spiral artery remodeling, increase in:
  • LIF
  • IL11
  • IL-8
  • IFNγ [55,63]
First and second trimesterTreg effects [64,65,66,67,68,69] (peak during second trimester):
  • Decreasing:
  • Th1/Th17 responses
  • B-cell proliferation
  • Antibody production
  • NK cytotoxicity
  • Increasing:
  • Decidual T-cells and
  • Macrophage levels
Treg effects [64,65,66,67,68,69] Increasing levels of:
  • IL-10
  • LIF
  • TGF-β
  • HO-1
  • IL-25 [70]
  • IL-4
  • IL-10
Acronyms: IL = interleukin; GM-CSF = granulocyte–macrophage colony-stimulating factor; TNF = tumor necrosis factor; NK = natural killer; Treg = regulatory T; M2 macrophages = wound-healing phenotype of macrophages; uDCs = uterine dendritic cells; PGE2 = prostaglandin E2; LIF = leukemia inhibitory factor; IFNγ = interferon γ; CCL = chemokine C-C motif ligand; TGF = transforming growth factor; HO = heme oxygenase.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Inversetti, A.; Zambella, E.; Guarano, A.; Dell’Avanzo, M.; Di Simone, N. Endometrial Microbiota and Immune Tolerance in Pregnancy. Int. J. Mol. Sci. 2023, 24, 2995. https://doi.org/10.3390/ijms24032995

AMA Style

Inversetti A, Zambella E, Guarano A, Dell’Avanzo M, Di Simone N. Endometrial Microbiota and Immune Tolerance in Pregnancy. International Journal of Molecular Sciences. 2023; 24(3):2995. https://doi.org/10.3390/ijms24032995

Chicago/Turabian Style

Inversetti, Annalisa, Enrica Zambella, Alice Guarano, Marinella Dell’Avanzo, and Nicoletta Di Simone. 2023. "Endometrial Microbiota and Immune Tolerance in Pregnancy" International Journal of Molecular Sciences 24, no. 3: 2995. https://doi.org/10.3390/ijms24032995

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