Next Article in Journal
Molecular Insights into the Specific Targeting of c-MYC G-Quadruplex by Thiazole Peptides
Previous Article in Journal
Transcriptomic–Proteomic Analysis Revealed the Regulatory Mechanism of Peanut in Response to Fusarium oxysporum
Previous Article in Special Issue
Metabolomics Integration in Assisted Reproductive Technologies for Enhanced Embryo Selection beyond Morphokinetic Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Clinical Relevance of Vaginal and Endometrial Microbiome Investigation in Women with Repeated Implantation Failure and Recurrent Pregnancy Loss

by
Xushan Gao
1,*,
Yvonne V. Louwers
1,
Joop S. E. Laven
1 and
Sam Schoenmakers
2
1
Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
2
Division of Obstetrics and Fetal Medicine, Department of Obstetrics and Gynecology, Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(1), 622; https://doi.org/10.3390/ijms25010622
Submission received: 3 November 2023 / Revised: 22 December 2023 / Accepted: 25 December 2023 / Published: 3 January 2024
(This article belongs to the Special Issue Molecular Research in Embryo Implantation)

Abstract

:
Recent studies have investigated if and how the vaginal and endometrial microbiome might affect endometrial receptivity and reproductive health. Although there is no consensus on the existence of a core uterine microbiome yet, evidence shows that the dominance of Lactobacillus spp. in the female reproductive tract is generally associated with eubiosis and improved chances of successful implantation and an ongoing pregnancy. Conversely, vaginal and endometrial dysbiosis can cause local inflammation and an increase of pro-inflammatory cytokines, compromising the integrity and receptivity of the endometrial mucosa and potentially hampering successful embryonic implantation. This review provides a critical appraisal of the influence of the vaginal and endometrial microbiome as parts of the female reproductive tract on fertility outcomes, focusing on repeated implantation failure (RIF) and recurrent pregnancy loss (RPL). It seems that RIF as well as RPL are both associated with an increase in microbiome diversity and a loss of Lactobacillus dominance in the lower female reproductive system.

1. Introduction

Despite the fact that reproductive outcomes in subfertile couples undergoing assisted reproductive treatment (ART) have improved over the last decades [1], the underlying causes of adverse outcomes in early pregnancy are still mostly unknown. Current techniques cannot investigate or visualize essential developmental stages around and after human embryonic implantation. While the intra-uterine environment is safeguarding and allows early embryonic and placental development, it is inaccessible without jeopardizing the early pregnancy itself.
Chronologically, the essential steps to achieve a healthy pregnancy include ovulation, viable sperm cells, fertilization, blastocyst hatching, implantation, placental and embryonic development, and overall adaptation of the maternal immune system to support the early pregnancy. The emergence of ART provided insights into in-vitro fertilization, monitoring of the first embryonic developmental stages, and the possibility of embryo selection for embryo transfer. However, after embryo transfer, a successful pregnancy depends on the crucial embryo-maternal interaction during the period referred to as the black box of pregnancy [2].
The uterine microenvironment and its endometrial lining are crucial during the final pre-implantation stages for embryo apposition, followed by blastocyst hatching, embryo invasion, and endometrial receptivity, all necessary for the successful implantation of the embryo. Synchronization of the transformation into a receptive endometrial layer with the arrival and development of a competent blastocyst plays an important role in this early interaction and is essential for successful implantation. Importantly, the uterine microenvironment is thought to include microbiota and influence a local immune system, which in turn facilitates the embryonic-endometrium crosstalk and therefore acts as a co-determinant for implantation.
Over the last decades, the role of the microbiome in human health and disease, including pregnancy, has gained increased attention.
From the moment of birth, the developing neonatal immune system is initially exposed to microbes through vertical transmission, followed by continuous horizontal transmission from parents, peers, and the environment [3]. During vaginal delivery, the neonatal gut is colonized by the maternal vaginal and gut microbiomes. Offspring delivered via cesarean section are mainly exposed to the maternal skin microbiome, exhibiting a noteworthy reduction in microbiota diversity, structure, and composition compared to those born vaginally [4].
It is suggested that the developing neonatal and infant microbiome is needed for shaping both innate and adaptive immunity. In turn, the developing immune system also influences the shaping of the microbiome itself [5,6]. This interplay has extensively been studied in the context of gut microbiota [7].
The immune system consistently regulates microbes but reacts much stronger when they become invasive or exhibit other pathogen-related behaviors. The gut microbiota interacts with the host immune response and can induce an imbalance in cytokine levels, which could impact placentation and embryonic development, resulting in unexplained pregnancy loss [8,9]. Currently, research is also focusing on the possible role of a leaky gut in the pathophysiology of RPL [10]. Imbalances in the gut microbiome can lead to the production of endotoxins, for example, lipopolysaccharide (LPS). LPS is the main component of the outer membrane of gram-negative bacteria and serves as a marker of increased bacterial translocation from the gut lumen to the circulation via barrier dysfunction. However, in general, LPS is not regarded as inherently harmful. Instead, its endotoxic effects are mainly influenced by the timing of infection and the concentration of LPS, which are mediated through the activation of the innate immune system [11]. Tersigni et al. propose that, as a result of a leaky gut, LPS infiltrates the maternal circulation and activates the endometrial immune system, causing inflammation and leading to RPL [10]. We hypothesize a similar mechanism for the reproductive tract microbiota.
Although more research is needed to determine whether microbial dysbiosis is one of the explanatory variables of early pregnancy complications, current scientific evidence indicates that the association between a perturbed microbiome of the reproductive tract and implantation failure deserves more clinical consideration [12]. The vaginal microbiome is known to play a crucial role in vaginal health. Most research into reproductive outcome has shown that the dominant presence of Lactobacillus species within the female reproductive tract is associated with reproductive success, while adverse reproductive outcome seems to be associated with a non-dominant Lactobacillus environment and a generalized higher microbial diversity and richness [13,14,15,16]. Moreover, depletion of vaginal Lactobacillus spp. is associated with the presence of pro-inflammatory cytokines, which seem to be involved in reproductive failure [17,18]. The microbiota of the female reproductive tract is suggested to be a key player in the local uterine immune pathways operating during pregnancy [19]. This could imply that the consequences and effects of a dysbiotic microbiota on the reproductive outcome, resulting from bacterial invasion from the vagina into the uterine cavity, might be influenced by the local immune system’s response. If the immune system is excessively active during the bacterial invasion, this results in a pro-inflammatory state and potentially leads to either implantation failure or pregnancy loss.
Studies focusing on the influence of the microbiota of the female reproductive tract on endometrial receptivity and how this may impact pregnancy outcomes are increasing. However, many questions remain to be answered. In the current review, we aim to summarize current insights and research on the role of the vaginal and endometrial microbiome and the local immune responses in early pregnancy, focusing on RIF and RPL. We hypothesize an association between the success of early pregnancy, specifically implantation, and the composition of the female genital tract microbiome, immunological pathways, and local inflammation.

2. Role of Microbiota in Modulating Immune Tolerance during Early Pregnancy

2.1. Embryo Implantation

Embryo implantation is a highly organized process that involves a receptive endometrium and a competent blastocyst [20]. The endometrial lining needs to undergo functional remodeling to promote and allow invasion of the blastocyst. In the proliferative phase, the endometrial layer thickens due to the rising levels of estrogens, and after ovulation, the secretory phase starts when endometrial proliferation halts and more endometrial glands filled with glycogen develop under the influence of progesterone in preparation for implantation [21].
After blastocyst hatching, embryo implantation consists of three phases: (1) apposition, during which the blastocyst contacts the implantation site of the endometrium; (2) adhesion, in which trophectoderm cells adhere to the extruding pinopods of the epithelial layer of the endometrium; and (3) invasion, when extra-embryonic trophoblast cells penetrate the endometrial stroma by crossing the basement membrane and invading the endometrial epithelium [22].
To facilitate the endometrial-embryo cross talk, implantation is orchestrated by a synchronized molecular dialogue, directed from the embryo towards the maternal site and vice versa, mediated by cytokines, various growth factors, prostaglandins, matrix-degrading enzymes, and their inhibitors, and adhesion molecules [23]. Asynchrony in the cross talk between a functional blastocyst and a receptive endometrium, either by disruption of the dialogue or timing outside the window of implantation, can interfere with implantation. The pathophysiology of RIF can be attributed to either embryonic factors, such as genetic or chromosomal abnormalities, maternal factors, or a disturbed endometrial-embryo cross talk [24], whereas suboptimal implantation can result in an increased risk of miscarriage or RPL.
According to previous studies, endometrial impairment in selecting good-quality embryos and not rejecting incompetent embryos is suggested to play a role in women with RIF and RPL [25]. Current insights suggest that the endometrium generally accepts all embryos, followed by rejection of those of bad quality while embracing those of good quality [26,27]. According to a recent comprehensive analysis by Brosens et al., the endometrium includes specific checkpoints that must be cleared to reduce maternal investment in a failing pregnancy [26]. For instance, ‘implantation checkpoint’ refers to the implantation window where the human endometrium is prepared to activate both the mechanisms leading to (a) menstrual breakdown and (b) the formation of the decidua of pregnancy [26]. The endometrium of women with RPL is associated with an environment that lacks anti-inflammatory decidual cells and is highly conducive to embryo implantation but is prone to breakdown in pregnancy and is hostile to placental formation. This condition is also named ‘the implantation checkpoint failure’, wherein the endometrium fails to facilitate the development of a normal embryo and does not eliminate the abnormal embryos at implantation [26].

2.2. Immunology of Early Pregnancy

During pregnancy, the maternal immune system facilitates an immunosuppressive environment to prevent the rejection of the developing semi-allogenic fetus and placenta [28], which involves extensive modifications of both the adaptive and innate immune systems. These modifications provide a favorable microenvironment that promotes implantation and supports healthy placentation [23]. Uterine NK cells downregulate their cytotoxic activity to support this immunosuppressive environment for the fetus and placenta [29].
Around the time of blastocyst implantation, mild inflammation is therefore needed to accomplish endometrial receptivity, including the attraction of the developing blastocyst to the epithelium [23]. For this, the T-helper 1/T-helper 2 ratio changes, in which the pro-inflammatory Th1 cells become dominant, secreting cytokines like interleukin-2 (IL-2), IFN-γ, TNF-α, and TNF-β, which promote implantation of the developing embryo [30]. After implantation, the ratio of Th1/Th2 shifts back towards a Th2-dominated anti-inflammatory immune response, leading to the secretion of anti-inflammatory cytokines such as IL-4, IL-6, and IL-10, protecting the fetus and contributing to placental development [31].

2.3. The Role of the Reproductive Tract Microbiota in Early Pregnancy

The composition of the microbiota of the female reproductive tract serves as a proxy for implantation success, as studies show that the general presence of a Lactobacillus dominance microbiome is strongly associated with reproductive success. This might be explained by the fact that Lactobacilli thrive on a certain substrate in the vaginal and endometrial anaerobic environment found in epithelial secretions, which might be similarly crucial for the embryo’s early survival following transfer into the uterine cavity and during the pre-implantation period [32]. Both developing embryos as well as Lactobacilli need glycogen to survive, suggesting that a Lactobacillus-dominant micro-environment in the endometrium, if it exists at all, indicates an optimal environment for the embryo to implant [33].
Lactobacilli protect the local environment against pathogens by promoting tight junction protein expression [34] and the production of lactic acid, as well as some key bacterial metabolites like glycerophospholipids and benzopyran necessary for embryonic development and implantation [35]. Lactic acid lowers the local pH, which provides protection against pathogens and has stimulatory effects on the innate immune system when it is exposed to the gram-negative bacterial lipopolysaccharide [36]. The presence of lactic acid also allows for the breakdown of the endometrial layer, while glycerophospholipids are responsible for the synthesis of prostaglandins and play crucial roles in embryo implantation. Prostaglandins are involved in stimulating angiogenesis during early pregnancy and influence blastocyst transportation by regulating myometrium relaxation and contraction [37,38], and as such, a lack of prostaglandins is associated with lower pregnancy chances [35].
In summary, successful implantation and pregnancy are associated with a Lactobacillus-dominant (LD) microbiome in the female reproductive tract. In contrast, a non-Lactobacillus dominant (NLD) microbiome may contribute to an inflammatory response affecting embryo implantation [39]. Moreover, a lack of glycogen in both the endometrium and the vagina might reduce the number of Lactobacillus species and thereby prevent proper implantation.

3. Repeated Implantation Failure and Recurrent Pregnancy Loss

3.1. Definition, Prevalence, and Prognosis of Repeated Implantation Failure

Several definitions of RIF have been used over the years in the literature. RIF has been described as an iatrogenic condition resulting from three unsuccessful fresh in vitro fertilization (IVF) cycles [40], failing to achieve a pregnancy after three completed fresh IVF-cycles with their resulting embryo transfers [41], or failure after the transfer of at least four good-quality embryos within a minimum of three fresh or frozen cycles [42]. The prevalence varies depending on the definition of the RIF used. A recent evaluation showed that up to 15% of the 1221 women undergoing IVF treatment suffer from RIF, which is defined as the failure to obtain a clinical pregnancy after at least three embryo transfer attempts [43]. Based on the definition of three or more consecutive failed frozen cycles [42], another group of researchers found that the prevalence of RIF was 5% among 4.429 women (mean age 35.4 years) with up to three successive frozen euploid single blastocyst transfers [44].
The chance of an ongoing pregnancy resulting in a live birth after a diagnosis of RIF remains unclear. A recent study could not identify RIF as a clinical predictive parameter associated with a live birth [45]. However, the cumulative incidence of a subsequent live birth after RIF during a follow-up period of 5.5 years was 49% (95% CI 39–59%) among women under 39 years, with around 70% of these pregnancies resulting from fresh or frozen embryo transfers after IVF or IVF/ICSI [45]. The risk factors for RIF include maternal age, BMI, uterine abnormalities, chromosomal abnormalities, and lifestyle factors such as smoking and excessive alcohol consumption [46,47,48,49].

3.2. Definition, Prevalence, and Prognosis of Recurrent Pregnancy Loss

RPL includes primary and secondary RPL. Primary RPL characterizes women who never had an ongoing pregnancy resulting in a live birth. Secondary RPL refers to women who have had at least one live birth before the recurrent miscarriages. RPL is defined as two or more pregnancy losses before 24 weeks gestation, verified by ultrasound or the presence of chorionic villi on an endometrial biopsy. This includes pregnancies following spontaneous conception as well as after ART. The prevalence of RPL is 1–2% when defined as three consecutive pregnancy losses prior to 20 weeks of gestation [50]. The risk of subsequent pregnancy loss is associated with the number of pregnancy losses; the higher the number of pregnancy losses, the higher the risk of subsequent pregnancy loss. Egerup et al. showed that the number of consecutive early pregnancy losses after the last birth has a significant negative prognostic influence on women with secondary RPL [51]. The authors suggest that an ongoing pregnancy resulting in a live birth in women with secondary RPL ‘neutralizes’ the negative prognostic effect of previous pregnancy losses. In other words, only consecutive pregnancy losses should be counted in the definition of RPL [51]. Recently, Krog et al. analyzed 106 vaginal microbiota samples in women with primary or secondary RPL using the shot-gun sequencing technique [52]. They found a dominance of Lactobacillus cripatus in women with primary RPL and Lactobacillus iners with secondary RPL. Indeed, this implies that a previous successful pregnancy seems to influence the composition of the vaginal microbiota.
Risk factors for RPL include advancing maternal age, obesity, antiphospholipid syndrome, parental chromosomal abnormalities, uterine anomalies, smoking, diabetes mellitus, thyroid disease, and thrombophilia [44,53,54,55]. However, more than 50% of the RPL lacks a clear etiology.

3.3. The Role of Reproductive Tract Microbiota in RIF and RPL

Although the exact mechanism by which pathogenic bacteria contribute to RIF or RPL remains unclear, it is believed that pro-inflammatory immune responses of the host due to either a lack of Lactobacilli, the presence of pathogens in the uterine cavity, or a combination of both factors play a significant role. The interplay between the microbiota and the immune system in the reproductive tract is considered a key factor in understanding the underlying causes of RIF and RPL [56,57].
In addition, several mechanisms for the interaction between the microbiota and the endometrium have been suggested [56,57,58]. During the adhesion process of the embryo to the endometrial lining, inflammatory mediators are carefully regulated [57]. Benner et al. suggested that, during implantation, the immune system is activated via pattern recognition receptors present on the endometrial epithelial cells. Usually, the presence of commensal bacteria can induce similar interaction between the host and microbes, fostering the tolerance of commensals. On the other hand, the presence of pathogenic bacteria and the depletion of commensal bacteria, along with their beneficial molecules, could potentially compromise the integrity of the endometrial mucosal barrier. This interference may alter the mucosal T-cell balance, leading to dysregulation of cytokine levels in immune cell activation and negatively affecting the local immune environment [58]. As a consequence, this cascade of events might hinder the proper invasion of the embryo [56].
In summary, subsequent activation of systemic and local tissue immune responses during pregnancy can lead to inflammation and potential damage to placental tissue and compromise placental function. Vaginal microbiota in bacterial vaginosis are known to interact with and activate vaginal dendritic cells, potentially causing overactivation of the immune system, which has been associated with adverse reproductive outcomes [59].

3.4. Repeated Implantation Failure

3.4.1. Vaginal Microbiome

Various factors can lead to the dysbiotic state of the vaginal microbiome, such as infections, usage of vaginal douches [60], certain diets [61], as well as prolonged use of antibiotics or other medications that eliminate bacteria [62]. The vaginal microbiome in women with RIF showed significantly higher microbial diversity compared to control groups [35]. Several independent studies show high abundances of genera associated with bacterial vaginosis, such as Gardnerella, Prevotella, Atopobium, Megasphaera, Burkholderia, and Sneathia, and low abundances of Lactobacilli in the RIF population compared to controls [13,35,63,64,65,66]. At the species level, women with RIF have a higher relative abundance of L. helveticus, while L. iners, Lactobacillus jensenii, Lactobacillus gasseri, and Lactobacillus agalactiae were more prevalent in women without RIF [63]. However, the presence of L. iners in women with RIF and unexplained infertility suggests an unfavorable role of this species in fertility [67]. Future studies should include a species-level analysis to identify the specific roles of the Lactobacillus species in reproductive outcomes.

3.4.2. Uterine Microbiome

In contrast to the vaginal microenvironment, if any at all, the uterine cavity maintains an extremely low microbial population and harbors 10,000 times fewer bacteria [68,69]. Due to this low biomass, no consensus exists on the composition of healthy endometrial microbiota or even the existence of a core microbiome due to the inaccessibility of the uterine cavity without the risk of contamination [69,70]. A recent study showed that in 141 women with RIF, 121 endometrial samples were non-Lactobacillus dominant with an increased presence of Streptococcus, Staphylococcus, Neisseria, and Klebsiella [71]. These pathogens are known to compromise the integrity of the endometrial epithelial, which can lead to failed implantation [72]. Also, the prevalence of Bacteroides species in the endometrium of non-pregnant women has been linked with RIF [73] A retrospective study has shown a prevalence of up to 66% of chronic endometritis in women with RIF diagnosed by hysteroscopy and biopsy [72]. Another study reported that Lactobacillus was less abundant in chronic endometritis, whereas Dialister, Bifidobacterium, Prevotella, Gardnerella, and Anaerococcus were more abundant in the endometrial microbiota of women with chronic endometritis [74]. However, microbial imbalances in chronic endometritis might also induce an altered immune response, which can cause a decrease in the endometrium’s receptivity, leading to RIF [75]. A dysbiotic vaginal microbiome and its associated pro-inflammatory responses may damage the cervical epithelial barrier, allowing bacterial translocation to the endometrium, which can lead to a local infectious state interfering with early pregnancy [76]. See Table 1 for a summary of the vaginal and endometrial microbiome in women with RIF.
Although an overactive maternal immune system is deemed to be disruptive for implantation and derangements within the endometrial immune environment are suggested to be involved in women with RPL, a comprehensive assessment of the biological basis of the immunology of RIF and RPL is beyond the scope of this review. We aim to discuss the most investigated immunological factors associated with RIF and RPL.

3.4.3. Molecular Immunological Characteristics of RIF

Natural Killer Cells

Natural killer (NK) cells, a subtype of cytotoxic lymphocytes, are key players in innate immunity and play an important role at the fetomaternal interface, making up around 70% of all immune cells [79]. Uterine NK (uNK) cells present in the uterus are involved in the process of successful placentation via angiogenesis and remodeling of the uteroplacental spiral arteries [80,81]. Additionally, NK cells contribute significantly to the establishment and maintenance of pregnancy through interactions with trophoblast cells [82], assisted by the production of cytokines, growth factors, and various chemokines. These factors all have an immunoinhibitory effect and are believed to promote a tolerogenic environment for the embryo [80,81]. Implantation failures have been associated with elevated cytotoxic NK cells in the decidua, a decrease in regulatory NK cells, and dysregulated cytokine production [83,84,85]. Dysregulation of NK cell cytokine production, particularly pro-inflammatory cytokines IFN-γ, and TNF-α, seems to be strongly associated with the immunopathology of RIF and RPL [84]. Activation of IFN-γ and TNF-α not only has inhibitory effects on blastocyst development but may also damage the tight junctions of the endocervical epithelial barrier, leading to a higher susceptibility to infection and inflammation [84].
Furthermore, with a dysregulated immune system, the interaction between uNK cells and the human leukocyte antigens (HLAs) expressed by trophoblasts, which is crucial in preventing trophoblast rejection by the maternal endometrium, becomes ineffective [86].

T Lymphocytes

Balances in diverse T-helper lymphocytes, i.e., Th1, Th2, Th17, and regulatory T cells (Treg), are important to mediate maternal tolerance to the semi-allogenic fetus [87]. The roles of Th1 and Th2 have been discussed before. After implantation, Treg cells are important in the maternal immune tolerance towards the fetus in the first and second trimesters through the production of the immunosuppressive cytokines IL-10 and TGF- β [88]. Th17 cells produce the regulatory cytokine IL-17, which is believed to be essential for the maintenance of a successful pregnancy [89]. An imbalance in the immunological Th17/Treg ratio is thought to be one of the contributing factors to the occurrence of RIF [90].

3.4.4. Reproductive Microbiome and Immunology in RIF

A recent study focused on the immunological markers of endometrial samples in relation to embryo implantation [78]. Endometrial biopsy samples were obtained from 26 women with RIF undergoing IVF treatment and were divided into a non-Lactobacillus dominant (NLD) and Lactobacillus-dominated (LD) microbiota (<90% and ≥90% Lactobacillus, respectively) [78]. The authors observed a significantly positive correlation between the Lactobacillus-dominant group (eubiosis) and the increased levels of cytokines that promote anti-inflammatory characteristics in tissues such as IL-10 and IGF-1 [78]. Moreover, a significantly negative correlation was found between Lactobacillus abundance and the pro-inflammatory cytokines IL-1β and IL-6 [78]. The inflammatory state could be explained by the depletion of Lactobacillus, the presence of pathogens, or a combination of both. In the samples from the group where Lactobacillus was not dominant, a higher level of IL-6 was associated with the presence of anaerobe microbiota such as Gardnerella and Streptococcus [78]. This is in line with another study focusing on uterine immunological changes, which found higher expression levels of IL-6 in women with RIF [91]. In contrast, a study in women with RIF reported lower levels of IL-6 in the endometrial [92] stroma. Based on the discrepancies observed, no definite conclusion can be drawn about the role of the ILs and endometrial microbiota in RIF pathophysiology.
Recent research has shown that in women with RIF, a dysbiotic vaginal microbiome (<90% Lactobacillus) was associated with increased serum levels of pro-inflammatory cytokines, specifically TNF-α and IFN-γ [93]. The presence of Burkholderia, Chryseobacterium, Enterococcus, Escherichia, Gemella, Herbaspirillum, Negativicoccus, and Staphylococcus was linked to an elevated TNF-α/IL-6 ratio. While the abundance of Aerococcus, Burkholderia, Escherichia, Herbaspirillum, Megasphera, Pseudomonas, Ralstonia, and Staphylococcus was associated with an increased IFN-γ/IL-10 ratio [93]. These findings further suggest a correlation between dysbiotic microbiota and an immune system in favor of inflammation.

3.5. Recurrent Pregnancy Loss

3.5.1. Vaginal Microbiome

A dominance of Lactobacillus spp. is found to play a protective role in the vaginal microbiota in early pregnancy, while vaginal dysbiosis is associated with RPL in ART patients [30,94,95,96]. The vaginal microbiota from women with depleted levels of Lactobacillus spp. displayed increased diversity of potential pathogens such as Streptococcus, Prevotella, Ureaplasma, Peptoniphilus, Dialaster, Megasphaera, Sneathia sanguinegen, Gardnerella, and Atopobium [17,30,75,94,95,96,97,98,99,100,101]. Several of these microbes are classified as part of the community state type (CST) IV, which is associated with a dysbiotic vaginal microbiota [30].
A recent study did not find a difference in vaginal microbiota, including L. iners, G. vaginalis, Atopobium vaginae, and Bifidobacterium breve, between women with RPL (n = 88) and healthy controls (n = 17) in a Japanese population [57], although this study is limited by its small sample size. Another recent study, comparing the vaginal microbiota of women with a normal pregnancy that ended through an induced abortion and women with unexplained RPL, showed that the latter group had an increased incidence of Pseudomonas, Roseburia, Collinsella aerofaciens, and Arthrobacter [94]. Therefore, analyses of the vaginal microbiome could act as a starting point for diagnostic investigations in RPL, especially in women undergoing assisted reproductive technology (ART) treatment.
Several studies have presented associations between the specific vaginal presence of Atopobium and RPL. One study found a high abundance of Atopobium in the vaginal microbiome samples of women with RPL as compared to controls [97], and another study reports that a relative abundance of >0.01% Atopobium could be used as a potential microbial biomarker to predict spontaneous miscarriages in the first trimester [102]. The potential immune-invasive role of Atopobium could be involved in disrupting the physicochemical barrier of the vaginal mucosa, thereby causing the release of pro-inflammatory cytokines and invasion of other anaerobes [102]. However, more research is needed on how the bacterial load of Atopobium affects pregnancy loss before it can be implemented as a diagnostic test.

3.5.2. Uterine Microbiome

Analyses in the microbiota of endometrial tissue and uterine lavage fluid showed significantly different microbes as compared with the lower genital tract, with relatively higher abundances of Acinetobacter, Anaerobacillus, Erysipelothrix, Bacillus, and Hydrogenophilus spp. in the RPL group compared to controls [103]. Another study collected endometrial samples by endometrial curette, which showed the dominance of L. iners instead of L. crispatus in the RPL group compared with controls [99]. L. iners has been reported to be associated with endometrial dysbiosis and adverse reproductive outcomes, including subfertility [104] and a history of pregnancy loss [105]. Recently, it was shown that 22 different significant taxa exist in endometrial fluid versus endometrial tissue, indicating that uterine lavage may not entirely reflect the microbial composition within endometrial tissue [106]. Endometrial samples from women with RPL were found to have a higher bacterial diversity and richness with species such as Aliihoeflea, Acinetobacter, Staphylococcus, and Serratia [106]. Another prospective study showed a relative dominance of Ureaplasma species in the endometrial microbiome tissue as an independent risk factor for a subsequent miscarriage after an embryo transfer with an euploid karyotype [107]. Furthermore, the composition of the endometrial microbiota during a menstrual cycle appears significantly different between women with RPL and controls [77]. Women with recurrent pregnancy loss (RPL) have a distinct endometrial microbiome characterized by maintained diversity and richness throughout the menstrual cycle, while in the control group, the microbiota decreases in diversity near ovulation and remains stable during the luteal phase [77]. This observation serves as a distinguishing factor between women who experience RPL and those who do not miscarry.
To summarize, the decreased abundance of Lactobacillus spp. and increased richness and diversity of the endometrial microbiome in women with RPL appear to be associated with miscarriages.
See Table 2 for a summary of the vaginal and endometrial microbiome in women with RPL.

3.5.3. Molecular Immunological Characteristics of RPL

NK Cells

Peripheral NK (pNK) cells before pregnancy seem significantly increased in women with RPL compared with controls, and this also differs between women with primary and secondary RPL [108,109,110]. Women experiencing primary RPL demonstrate significantly higher levels of pNK cells when compared to women who have had a previous live birth and nulliparous control women [111]. However, there is no consensus on whether the increased level of NK cells represents cytotoxicity in women with RPL before pregnancy [112,113]. Therefore, testing preconceptional pNK cell activity to predict subsequent miscarriage in women with RPL is not recommended [55].

Cytokines

In addition, increased levels of NK cells, increased plasma levels of tumor necrosis factor-α (TNF-α), interferon-gamma (IFN-γ), and IL-6 seem to be associated with RPL [109,114]. TNF-α promotes trophoblast invasion and induces vascular endothelial growth factor (VEGF) important for placental development [115]. TNF-α and IFN-γ are Th1 cytokines commonly present in acute and chronic inflammatory conditions. Their presence poses a potential risk, as they may cause direct damage to the placenta and fetus or indirectly activate cytotoxic cells, including NK or T cells [116]. However, one study did not find differences in the levels of IFN-γ and TNF-α between women with RPL and a successfully completed pregnancy compared to those who experienced another miscarriage [117].

T lymphocytes

An imbalance between Th17 cells and Treg cells also plays a role in RPL [87,118]. In the event of a bacterial invasion, Th17 is activated and contributes to inflammation by secreting cytokines such as IL-17A, IL-22, and IL-26 [118]. A higher Th17/Treg cell ratio was found in studies with unexplained RPL compared to fertile controls [119,120]. These findings suggest that an imbalance in Th1/Th2 and Th1/Treg cell ratios might have a negative impact on implantation and the maintenance of pregnancy.

3.5.4. Reproductive Tract Microbiome and Immunology in RPL

Uterine and peripheral NK cells are considered members of the innate immune system and play an active role in pregnancy support. Chemokines, cytokines, growth factors, and angiogenic factors regulate trophoblast migration and promote placental growth. NK cells also play an important immunoregulatory role in the defense against infections [55]. In women with RPL, high levels of peripheral NK cells and an increased abundance of G. vaginalis and gram-negative anaerobes are observed in the vaginal microbiota, suggesting a potential link between the microbial composition, local inflammation, alterations in immune parameters, and the occurrence of miscarriage [95].
The pro-inflammatory cytokine IL-2, primarily produced by Th1 cells, and the anti-inflammatory IL-10, mainly secreted by Th2 cells, both play important roles in promoting successful implantation [87,121]. Studies focusing on the vaginal compartment found greater levels of IL-2 and lower levels of IL-10 in women with unexplained RPL compared to those with viable pregnancies [122]. Earlier research has discovered a connection between RPL and the down-regulation of IL-10 [123]. However, the mechanisms responsible for the reduced production of IL-10 at the maternal-fetal interface remain unclear. Pro-inflammatory cytokines such as IL-6, IL-1β, and TNF-α seem to be associated with a decrease in Lactobacillus and an increase in Prevotella and Streptococcus in women with euploid miscarriages [17].
Endometrial samples from women with RPL were found to have a higher bacterial diversity and richness and were associated with decreased IFN-γ and IL-6 pro-inflammatory cytokine concentrations [103]. Also, statistically significant correlations between Aliihoeflea and IL-17A, Acinetobacter and IFN-γ, Serratia and TNF-α, and Staphylococcus and Serratia and IL-6 were found [103].

4. Summary

This review provides an overview of the interaction of the reproductive tract microbiota, focusing on the vagina and uterine cavity, and the local immune responses in women experiencing RIF and RPL. Importantly, there seems to be a detectable difference in vaginal microbiome composition between healthy controls and women with RIF or RPL before conception. Women with RIF or RPL fail to attenuate their endometrial microbiome diversity and richness throughout the menstrual cycle [77].
A Lactobacillus-dominant vaginal microbiota seems to be beneficial for embryo implantation, whereas a non-Lactobacillus-dominant microbiota seems to impair implantation success. The pathogens identified in the dysbiotic vaginal microbiota of women with RIF were also found to be present in the dysbiotic vaginal microbiota of women with RPL. Among these pathogens, Gardnerella, Prevotella, Atopobium, Megasphaera, and Sneathia were observed to have high abundances. In contrast, the increased abundance of genera found in the endometrial microbiota is different among women with RIF and RPL.
Studies have discovered positive correlations between a higher abundance of Lactobacillus and anti-inflammatory cytokines, while a negative association has been observed between Lactobacillus and pro-inflammatory cytokines in the endometrial microbiota of women with RIF [78,91]. Additionally, pro-inflammatory cytokines have been positively linked to Gardnerella and Streptococcus [78].
Moreover, the innate and adaptive immune systems seem to be dysregulated in women with RPL. Predominant Th1 and Th17 immunity and decreased Th2 and Treg cells are associated with the RPL of fetuses with a normal karyotype [124]. Failures to maintain adequate Th1/Th2 and Th17/Treg cell balance have been associated with RIF and RPL [119,120].
It seems that, compared to eubiosis (Figure 1), vaginal and/or endometrial dysbiosis is associated with RIF (Figure 2) and RPL (Figure 3), and that in both, similar mechanisms are involved. Moreover, inflammation and dysregulated activation of the immune system seem to affect the integrity of the endometrial mucosa, leading to RIF, and subsequently interfere with the receptivity of the endometrium and the processes of implantation and placentation, leading to RPL.
The reproductive microbiome does not necessarily have to be limited to the reproductive tract, since recent evidence suggests that an imbalance of gut microbes, or gut microbial dysbiosis, can also lead to adverse pregnancy outcomes [8,125]. A study found a correlation between decreased gut microbiome diversity and an increased ratio of Firmicutes to Bacteriodetes in women with first-trimester miscarriages, indicating that the pro-inflammatory effect of the gut microbiome is probably caused by holistic dysbiosis rather than the dominance of a particular pathogen [125].
The gut microbiome is a crucial factor for regulating and shaping the immune response, and gut microbial dysbiosis can trigger inflammation [9]. Also, the Th1/Th17-mediated pro-inflammatory state in unexplained RPL is associated with gut dysbiosis [125]. The extent of the impact of the gut microbiota on reproductive organs is being increasingly recognized, revealing a greater influence than was previously understood. A pathological state of the intestinal system, the so-called leaky gut, is one in which imbalances in the gut microbiota can lead to the production of toxins that infiltrate the maternal circulation and activate the endometrial immune system, leading to inflammation and RPL [10]. The gut microbiome also affects hormonal homeostasis and the coagulation system, which are involved in implantation [67,126]. In the presence of increased abundances of Gram-negative bacteria in the gut microbiota, causing inflammation and the systematic release of pro-inflammatory cytokines, the secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) could be impaired, resulting in subfertility [127]. Future research will help to clarify the relationship between gut microbiota and reproductive outcomes.

5. Challenges and Future Perspectives

Microbiome studies are complicated by the diversity in the execution of analytical steps, which can cause heterogeneity due to differences in local protocols, sampling methods, primers used, sequencing techniques, and bioinformatics pipelines, making comparisons between studies difficult, especially across different ethnicities. To obtain comprehensive insights into the role of Lactobacillus spp. in reproductive health, studies should incorporate a species-level analysis of the vaginal and uterine microbiota, as microbiota dominated by Lactobacillus may not always indicate the presence of the beneficial L. crispatus, but rather an intermediate state towards a dysbiotic condition, often associated with the presence of L. iners or L. gasseri [128].
Furthermore, conducting species-level analysis allows for better comparisons between the studies conducted in different ethnicities. Lactobacilli species and their proportions are different among different ethnicities. Black African and African-American women carry more often L. iners compared to Caucasian women, who carry more likely L. crispatus [129].
Also, the obstetric history is of importance since the dominance of L. crispatus in the vaginal microbiota could change to L. iners in Caucasian women who gave birth [52]. Indeed, one study found that the abundance of L. crispatus is negatively correlated with the number of previous deliveries. In other words, a nullipara has a two-times higher mean relative abundance of L. crispatus than women with one prior delivery (58% vs. 26%) [130]. Future studies should take into account the obstetric history as well as the number of previous pregnancies at the time of sample collection.
Finally, during natural conception, the microbial content present in sperm may enter the uterine cavity and potentially interact with the local microbiota of the endometrial environment [19]. The role of the seminal microbiome in sperm health lacks consensus. Some studies indicate that Lactobacillus, commonly abundant in semen, is often found to be less abundant in men with lower seminal quality [131,132]. Others found conflicting results and showed an increase in Lactobacillus in patients with azoospermia [133]. Prevotella, on the other hand, consistently correlates with reduced sperm quality, primarily affecting sperm motility [131,132]. Although more research is needed to investigate whether the dysbiotic vaginal microbiota in women, which is linked to unfavorable fertility outcomes, could be reflected in the seminal microbiome of their partners,
Last but not least, to understand the molecular mechanisms simultaneously interacting around the timing of implantation, the use of different ex vivo-omics could offer more insights. Combining analyses and integrating techniques including metabolomics, transcriptomics, proteomics, and metagenomics for a single sample or condition provides a more comprehensive understanding of the taxonomic composition, local gene expression, and local molecular mechanisms involved in RIF or RPL.
A metatranscriptomics approach in microbiome research could be used to uncover specific information regarding transcriptionally active bacteria. Metatranscriptomics of chorionic villi tissue and decidua of women with RPL have shown upregulation of pro-inflammatory genes and dysregulation of genes involved in angiogenesis. These results indicate a link between inflammation and abnormal placental development in women with RPL [134,135]. Another study reported dysregulation of genes involved in the extracellular matrix and its remodeling, crucial for proper implantation and placentation during trophoblast invasion [136]. Combining these omics data can lead to a better understanding of the relationship between taxonomic composition and local gene expression patterns.
In the decidual tissue of women with RPL, proteomics showed overexpression of proteins related to the inhibition of decidual cell growth and oxidative stress pathways. Oxidative stress, caused by chronic inflammation, can interfere with mitochondrial function and cause DNA damage, thereby affecting embryonic development [137].
Future research should also focus on the use of blastoids, endometrial, and placental organoid models to mimic the physiological microbial-immunological and endometrial-embryo cross talk which provides the opportunity to study the local interaction with bacterial products, gene expression, and immunological pathways and non-invasively offer new insights into the pathophysiology of RIF and RPL.

6. Conclusions

In conclusion, the current review provides an updated overview of the vaginal and endometrial bacterial communities and their interaction with the local immune system in RIF and RPL. A state of microbiota dysbiosis with an overabundance of pathogenic species or the absence of Lactobacillus-dominated vaginal and endometrial microbiomes might trigger inflammation, hinder the process of embryonic implantation, and interfere with early pregnancy. To attain a better understanding of the role of Lactobacillus spp. and other microbes within the reproductive tract microbiota in reproductive health and disease, it is crucial to include species-level analyses of the vaginal and uterine microbiota in studies. This approach not only provides a more detailed examination but also facilitates more accurate comparisons among different studies conducted in diverse ethnic and obstetric populations. Future research should also be conducted in larger sample sizes and incorporate omics technologies and organoids to further enhance our understanding of reproductive health and disease.

Author Contributions

X.G. and S.S. wrote this article. Y.V.L. and J.S.E.L. reviewed and edited this article. 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

Not applicable.

Acknowledgments

The authors wish to thank M.F.M. Engel from the Erasmus MC Medical Library for developing and updating the search strategies.

Conflicts of Interest

X.G., Y.V.L. and S.S. have nothing to declare. J.S.E.L. reports unrestricted research grants from Ansh Labs (Webster, TX, USA), from Ferring (Hoofddorp, The Netherlands), from Dutch Heart Association (Utrecht, The Netherlands), from Zon MW (Amsterdam, The Netherlands), from Roche Diagnostics (Rothkreuz, Switzerland), and consultancy fees from Ferring (Hoofddorp, The Netherlands), from Titus Healthcare (Hoofddorp, The Netherlands), from Gedeon Richter (Groot-Bijgaarden, Belgium), and is an unpaid board member and president of the AE-PCOS Society, outside the submitted work.

References

  1. Andersen, A.N.; Goossens, V.; Bhattacharya, S.; Ferraretti, A.P.; Kupka, M.S.; De Mouzon, J.; Nygren, K.G.; European Society of Human Reproduction and Embryology (ESHRE). Assisted reproductive technology and intrauterine inseminations in Europe, 2005: Results generated from European registers by ESHRE: ESHRE. The European IVF Monitoring Programme (EIM), for the European Society of Human Reproduction and Embryology (ESHRE). Hum. Reprod. 2009, 24, 1267–1287. [Google Scholar] [CrossRef]
  2. Macklon, N.S.; Geraedts, J.P.; Fauser, B.C. Conception to ongoing pregnancy: The ‘black box’ of early pregnancy loss. Hum. Reprod. Update 2002, 8, 333–343. [Google Scholar] [CrossRef] [PubMed]
  3. Gensollen, T.; Iyer, S.S.; Kasper, D.L.; Blumberg, R.S. How colonization by microbiota in early life shapes the immune system. Science 2016, 352, 539–544. [Google Scholar] [CrossRef] [PubMed]
  4. Senn, V.; Bassler, D.; Choudhury, R.; Scholkmann, F.; Righini-Grunder, F.; Vuille-Dit-Bile, R.N.; Restin, T. Microbial Colonization From the Fetus to Early Childhood-A Comprehensive Review. Front. Cell. Infect. Microbiol. 2020, 10, 573735. [Google Scholar] [CrossRef] [PubMed]
  5. Alexander, K.L.; Targan, S.R.; Elson, C.O., III. Microbiota activation and regulation of innate and adaptive immunity. Immunol. Rev. 2014, 260, 206–220. [Google Scholar] [CrossRef] [PubMed]
  6. Postler, T.S.; Ghosh, S. Understanding the Holobiont: How Microbial Metabolites Affect Human Health and Shape the Immune System. Cell Metab. 2017, 26, 110–130. [Google Scholar] [CrossRef]
  7. Yao, Y.; Cai, X.; Ye, Y.; Wang, F.; Chen, F.; Zheng, C. The Role of Microbiota in Infant Health: From Early Life to Adulthood. Front. Immunol. 2021, 12, 708472. [Google Scholar] [CrossRef]
  8. Qi, X.; Yun, C.; Pang, Y.; Qiao, J. The impact of the gut microbiota on the reproductive and metabolic endocrine system. Gut Microbes 2021, 13, 1894070. [Google Scholar] [CrossRef]
  9. Schirmer, M.; Smeekens, S.P.; Vlamakis, H.; Jaeger, M.; Oosting, M.; Franzosa, E.A.; Ter Horst, R.; Jansen, T.; Jacobs, L.; Bonder, M.J.; et al. Linking the Human Gut Microbiome to Inflammatory Cytokine Production Capacity. Cell 2016, 167, 1125–1136.e8. [Google Scholar] [CrossRef]
  10. Tersigni, C.; D’Ippolito, S.; Di Nicuolo, F.; Marana, R.; Valenza, V.; Masciullo, V.; Scaldaferri, F.; Malatacca, F.; de Waure, C.; Gasbarrini, A.; et al. Recurrent pregnancy loss is associated to leaky gut: A novel pathogenic model of endometrium inflammation? J. Transl. Med. 2018, 16, 102. [Google Scholar] [CrossRef]
  11. Freudenberg, M.A.; Kalis, C.; Chvatchko, Y.; Merlin, T.; Gumenscheimer, M.; Galanos, C. Role of interferons in LPS hypersensitivity. J. Endotoxin Res. 2003, 9, 308–312. [Google Scholar] [CrossRef] [PubMed]
  12. Cimadomo, D.; Craciunas, L.; Vermeulen, N.; Vomstein, K.; Toth, B. Definition, diagnostic and therapeutic options in recurrent implantation failure: An international survey of clinicians and embryologists. Hum. Reprod. 2021, 36, 305–317. [Google Scholar] [CrossRef] [PubMed]
  13. Bernabeu, A.; Lledo, B.; Diaz, M.C.; Lozano, F.M.; Ruiz, V.; Fuentes, A.; Lopez-Pineda, A.; Moliner, B.; Castillo, J.C.; Ortiz, J.A.; et al. Effect of the vaginal microbiome on the pregnancy rate in women receiving assisted reproductive treatment. J. Assist. Reprod. Genet. 2019, 36, 2111–2119. [Google Scholar] [CrossRef] [PubMed]
  14. 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] [PubMed]
  15. Miyagi, M.; Mekaru, K.; Tanaka, S.E.; Arai, W.; Ashikawa, K.; Sakuraba, Y.; Nakamura, R.; Oishi, S.; Akamine, K.; Aoki, Y. Endometrial and vaginal microbiomes influence assisted reproductive technology outcomes. JBRA Assist. Reprod. 2022, 27, 267. [Google Scholar] [CrossRef] [PubMed]
  16. Moreno, I.; Codoner, F.M.; Vilella, F.; Valbuena, D.; Martinez-Blanch, J.F.; Jimenez-Almazan, J.; Alonso, R.; Alama, P.; Remohi, 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]
  17. Grewal, K.; Lee, Y.S.; Smith, A.; Brosens, J.J.; Bourne, T.; Al-Memar, M.; Kundu, S.; MacIntyre, D.A.; Bennett, P.R. Chromosomally normal miscarriage is associated with vaginal dysbiosis and local inflammation. BMC Med. 2022, 20, 38. [Google Scholar] [CrossRef]
  18. Manhanzva, M.T.; Abrahams, A.G.; Gamieldien, H.; Froissart, R.; Jaspan, H.; Jaumdally, S.Z.; Barnabas, S.L.; Dabee, S.; Bekker, L.G.; Gray, G.; et al. Inflammatory and antimicrobial properties differ between vaginal Lactobacillus isolates from South African women with non-optimal versus optimal microbiota. Sci. Rep. 2020, 10, 6196. [Google Scholar] [CrossRef]
  19. Koedooder, R.; Mackens, S.; Budding, A.; Fares, D.; Blockeel, C.; Laven, J.; Schoenmakers, S. Identification and evaluation of the microbiome in the female and male reproductive tracts. Hum. Reprod. Update 2019, 25, 298–325. [Google Scholar] [CrossRef]
  20. Bischof, P.; Campana, A. Trophoblast differentiation and invasion: Its significance for human embryo implantation. Early Pregnancy 1997, 3, 81–95. [Google Scholar]
  21. Norwitz, E.R.; Schust, D.J.; Fisher, S.J. Implantation and the survival of early pregnancy. N. Engl. J. Med. 2001, 345, 1400–1408. [Google Scholar] [CrossRef] [PubMed]
  22. Carson, D.D.; Bagchi, I.; Dey, S.K.; Enders, A.C.; Fazleabas, A.T.; Lessey, B.A.; Yoshinaga, K. Embryo implantation. Dev. Biol. 2000, 223, 217–237. [Google Scholar] [CrossRef] [PubMed]
  23. Dekel, N.; Gnainsky, Y.; Granot, I.; Racicot, K.; Mor, G. The role of inflammation for a successful implantation. Am. J. Reprod. Immunol. 2014, 72, 141–147. [Google Scholar] [CrossRef] [PubMed]
  24. Franasiak, J.M.; Alecsandru, D.; Forman, E.J.; Gemmell, L.C.; Goldberg, J.M.; Llarena, N.; Margolis, C.; Laven, J.; Schoenmakers, S.; Seli, E. A review of the pathophysiology of recurrent implantation failure. Fertil. Steril. 2021, 116, 1436–1448. [Google Scholar] [CrossRef] [PubMed]
  25. Teklenburg, G.; Salker, M.; Heijnen, C.; Macklon, N.S.; Brosens, J.J. The molecular basis of recurrent pregnancy loss: Impaired natural embryo selection. Mol. Hum. Reprod. 2010, 16, 886–895. [Google Scholar] [CrossRef]
  26. Brosens, J.J.; Bennett, P.R.; Abrahams, V.M.; Ramhorst, R.; Coomarasamy, A.; Quenby, S.; Lucas, E.S.; McCoy, R.C. Maternal selection of human embryos in early gestation: Insights from recurrent miscarriage. Semin. Cell Dev. Biol. 2022, 131, 14–24. [Google Scholar] [CrossRef]
  27. Teklenburg, G.; Salker, M.; Molokhia, M.; Lavery, S.; Trew, G.; Aojanepong, T.; Mardon, H.J.; Lokugamage, A.U.; Rai, R.; Landles, C.; et al. Natural selection of human embryos: Decidualizing endometrial stromal cells serve as sensors of embryo quality upon implantation. PLoS ONE 2010, 5, e10258. [Google Scholar] [CrossRef]
  28. Mor, G.; Cardenas, I. The immune system in pregnancy: A unique complexity. Am. J. Reprod. Immunol. 2010, 63, 425–433. [Google Scholar] [CrossRef]
  29. Gaynor, L.M.; Colucci, F. Uterine Natural Killer Cells: Functional Distinctions and Influence on Pregnancy in Humans and Mice. Front. Immunol. 2017, 8, 467. [Google Scholar] [CrossRef]
  30. Al-Memar, M.; Bobdiwala, S.; Fourie, H.; Mannino, R.; Lee, Y.S.; Smith, A.; Marchesi, J.R.; Timmerman, D.; Bourne, T.; Bennett, P.R.; et al. The association between vaginal bacterial composition and miscarriage: A nested case-control study. BJOG 2020, 127, 264–274. [Google Scholar] [CrossRef]
  31. Mor, G.; Aldo, P.; Alvero, A.B. The unique immunological and microbial aspects of pregnancy. Nat. Rev. Immunol. 2017, 17, 469–482. [Google Scholar] [CrossRef] [PubMed]
  32. Koedooder, R.; Singer, M.; Schoenmakers, S.; Savelkoul, P.H.M.; Morre, S.A.; de Jonge, J.D.; Poort, L.; Cuypers, W.S.S.; Budding, A.E.; Laven, J.S.E.; et al. The ReceptIVFity cohort study protocol to validate the urogenital microbiome as predictor for IVF or IVF/ICSI outcome. Reprod. Health 2018, 15, 202. [Google Scholar] [CrossRef] [PubMed]
  33. 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] [PubMed]
  34. Delgado-Diaz, D.J.; Jesaveluk, B.; Hayward, J.A.; Tyssen, D.; Alisoltani, A.; Potgieter, M.; Bell, L.; Ross, E.; Iranzadeh, A.; Allali, I.; et al. Lactic acid from vaginal microbiota enhances cervicovaginal epithelial barrier integrity by promoting tight junction protein expression. Microbiome 2022, 10, 141. [Google Scholar] [CrossRef] [PubMed]
  35. 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. [Google Scholar] [CrossRef] [PubMed]
  36. Witkin, S.S.; Alvi, S.; Bongiovanni, A.M.; Linhares, I.M.; Ledger, W.J. Lactic acid stimulates interleukin-23 production by peripheral blood mononuclear cells exposed to bacterial lipopolysaccharide. FEMS Immunol. Med. Microbiol. 2011, 61, 153–158. [Google Scholar] [CrossRef] [PubMed]
  37. Baryla, M.; Kaczynski, P.; Goryszewska, E.; Riley, S.C.; Waclawik, A. Prostaglandin F(2alpha) stimulates adhesion, migration, invasion and proliferation of the human trophoblast cell line HTR-8/SVneo. Placenta 2019, 77, 19–29. [Google Scholar] [CrossRef]
  38. Kaczynski, P.; Goryszewska, E.; Baryla, M.; Waclawik, A. Prostaglandin F2alpha stimulates angiogenesis at the embryo-maternal interface during early pregnancy in the pig. Theriogenology 2020, 142, 169–176. [Google Scholar] [CrossRef]
  39. Lebedeva, O.P.; Popov, V.N.; Syromyatnikov, M.Y.; Starkova, N.N.; Maslov, A.Y.; Kozarenko, O.N.; Gryaznova, M.V. Female reproductive tract microbiome and early miscarriages. Apmis 2023, 131, 61–76. [Google Scholar] [CrossRef]
  40. Polanski, L.T.; Baumgarten, M.N.; Quenby, S.; Brosens, J.; Campbell, B.K.; Raine-Fenning, N.J. What exactly do we mean by ‘recurrent implantation failure’? A systematic review and opinion. Reprod. Biomed. Online 2014, 28, 409–423. [Google Scholar] [CrossRef]
  41. Tan, B.K.; Vandekerckhove, P.; Kennedy, R.; Keay, S.D. Investigation and current management of recurrent IVF treatment failure in the UK. BJOG 2005, 112, 773–780. [Google Scholar] [CrossRef] [PubMed]
  42. Coughlan, C.; Ledger, W.; Wang, Q.; Liu, F.; Demirol, A.; Gurgan, T.; Cutting, R.; Ong, K.; Sallam, H.; Li, T.C. Recurrent implantation failure: Definition and management. Reprod. Biomed. Online 2014, 28, 14–38. [Google Scholar] [CrossRef] [PubMed]
  43. Busnelli, A.; Reschini, M.; Cardellicchio, L.; Vegetti, W.; Somigliana, E.; Vercellini, P. How common is real repeated implantation failure? An indirect estimate of the prevalence. Reprod. Biomed. Online 2020, 40, 91–97. [Google Scholar] [CrossRef] [PubMed]
  44. Pirtea, P.; De Ziegler, D.; Tao, X.; Sun, L.; Zhan, Y.; Ayoubi, J.M.; Seli, E.; Franasiak, J.M.; Scott, R.T., Jr. Rate of true recurrent implantation failure is low: Results of three successive frozen euploid single embryo transfers. Fertil. Steril. 2021, 115, 45–53. [Google Scholar] [CrossRef] [PubMed]
  45. Koot, Y.E.M.; Hviid Saxtorph, M.; Goddijn, M.; de Bever, S.; Eijkemans, M.J.C.; Wely, M.V.; van der Veen, F.; Fauser, B.; Macklon, N.S. What is the prognosis for a live birth after unexplained recurrent implantation failure following IVF/ICSI? Hum. Reprod. 2019, 34, 2044–2052. [Google Scholar] [CrossRef] [PubMed]
  46. Vomstein, K.; Voss, P.; Molnar, K.; Ainsworth, A.; Daniel, V.; Strowitzki, T.; Toth, B.; Kuon, R.J. Two of a kind? Immunological and clinical risk factors differ between recurrent implantation failure and recurrent miscarriage. J. Reprod. Immunol. 2020, 141, 103166. [Google Scholar] [CrossRef] [PubMed]
  47. Orvieto, R.; Meltcer, S.; Nahum, R.; Rabinson, J.; Anteby, E.Y.; Ashkenazi, J. The influence of body mass index on in vitro fertilization outcome. Int. J. Gynaecol. Obstet. 2009, 104, 53–55. [Google Scholar] [CrossRef] [PubMed]
  48. Waylen, A.L.; Metwally, M.; Jones, G.L.; Wilkinson, A.J.; Ledger, W.L. Effects of cigarette smoking upon clinical outcomes of assisted reproduction: A meta-analysis. Hum. Reprod Update 2009, 15, 31–44. [Google Scholar] [CrossRef]
  49. Zeadna, A.; Son, W.Y.; Moon, J.H.; Dahan, M.H. A comparison of biochemical pregnancy rates between women who underwent IVF and fertile controls who conceived spontaneouslydagger. Hum. Reprod. 2015, 30, 783–788. [Google Scholar] [CrossRef]
  50. Ford, H.B.; Schust, D.J. Recurrent pregnancy loss: Etiology, diagnosis, and therapy. Rev. Obstet. Gynecol. 2009, 2, 76–83. [Google Scholar]
  51. Egerup, P.; Kolte, A.M.; Larsen, E.C.; Krog, M.; Nielsen, H.S.; Christiansen, O.B. Recurrent pregnancy loss: What is the impact of consecutive versus non-consecutive losses? Hum. Reprod 2016, 31, 2428–2434. [Google Scholar] [CrossRef] [PubMed]
  52. Krog, M.C.; Peker, N.; Hugerth, L.W.; Vomstein, K.; Bliddal, S.; Reinhardt Nielsen, J.; Slot, A.; Vauvert Hviid, K.; Engstrand, L.; Schuppe-Koistinen, I. O-262 The vaginal and faecal microbiome in women with recurrent pregnancy loss (RPL) before pregnancy according to the reproductive outcome after referral. Hum. Reprod. 2023, 38, dead093-316. [Google Scholar] [CrossRef]
  53. Kaiser, J.; Branch, D.W. Recurrent Pregnancy Loss: Generally Accepted Causes and Their Management. Clin. Obstet. Gynecol. 2016, 59, 464–473. [Google Scholar] [CrossRef] [PubMed]
  54. RPL, E.G.G.o.; 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, 1–12. [Google Scholar] [CrossRef]
  55. RPL, E.G.G.o.; Bender Atik, R.; Christiansen, O.B.; Elson, J.; Kolte, A.M.; Lewis, S.; Middeldorp, S.; McHeik, S.; Peramo, B.; Quenby, S.; et al. ESHRE guideline: Recurrent pregnancy loss: An update in 2022. Hum. Reprod. Open 2023, 2023, hoad002. [Google Scholar] [CrossRef]
  56. Al-Nasiry, S.; Ambrosino, E.; Schlaepfer, M.; Morre, S.A.; Wieten, L.; Voncken, J.W.; Spinelli, M.; Mueller, M.; Kramer, B.W. The Interplay Between Reproductive Tract Microbiota and Immunological System in Human Reproduction. Front. Immunol. 2020, 11, 378. [Google Scholar] [CrossRef]
  57. Bardos, J.; Fiorentino, D.; Longman, R.E.; Paidas, M. Immunological Role of the Maternal Uterine Microbiome in Pregnancy: Pregnancies Pathologies and Alterated Microbiota. Front. Immunol. 2019, 10, 2823. [Google Scholar] [CrossRef]
  58. 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]
  59. Ravel, J.; Moreno, I.; Simón, C. Bacterial vaginosis and its association with infertility, endometritis, and pelvic inflammatory disease. Am. J. Obstet. Gynecol. 2021, 224, 251–257. [Google Scholar] [CrossRef]
  60. Schwebke, J.R.; Richey, C.M.; Weiss, H.L. Correlation of behaviors with microbiological changes in vaginal flora. J. Infect. Dis. 1999, 180, 1632–1636. [Google Scholar] [CrossRef]
  61. Neggers, Y.H.; Nansel, T.R.; Andrews, W.W.; Schwebke, J.R.; Yu, K.F.; Goldenberg, R.L.; Klebanoff, M.A. Dietary intake of selected nutrients affects bacterial vaginosis in women. J. Nutr. 2007, 137, 2128–2133. [Google Scholar] [CrossRef] [PubMed]
  62. Ahrens, P.; Andersen, L.O.; Lilje, B.; Johannesen, T.B.; Dahl, E.G.; Baig, S.; Jensen, J.S.; Falk, L. Changes in the vaginal microbiota following antibiotic treatment for Mycoplasma genitalium, Chlamydia trachomatis and bacterial vaginosis. PLoS ONE 2020, 15, e0236036. [Google Scholar] [CrossRef] [PubMed]
  63. Diaz-Martinez, M.D.C.; Bernabeu, A.; Lledo, B.; Carratala-Munuera, C.; Quesada, J.A.; Lozano, F.M.; Ruiz, V.; Morales, R.; Llacer, 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] [PubMed]
  64. Haahr, T.; Jensen, J.S.; Thomsen, L.; Duus, L.; Rygaard, K.; Humaidan, P. Abnormal vaginal microbiota may be associated with poor reproductive outcomes: A prospective study in IVF patients. Hum. Reprod. 2016, 31, 795–803. [Google Scholar] [CrossRef] [PubMed]
  65. 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]
  66. Kong, Y.; Liu, Z.; Shang, Q.; Gao, Y.; Li, X.; Zheng, C.; Deng, X.; Chen, T. The Disordered Vaginal Microbiota Is a Potential Indicator for a Higher Failure of in vitro Fertilization. Front. Med. 2020, 7, 217. [Google Scholar] [CrossRef]
  67. Patel, N.; Patel, N.; Pal, S.; Nathani, N.; Pandit, R.; Patel, M.; Patel, N.; Joshi, C.; Parekh, B. Distinct gut and vaginal microbiota profile in women with recurrent implantation failure and unexplained infertility. BMC Women’s Health 2022, 22, 113. [Google Scholar] [CrossRef]
  68. 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]
  69. 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, e611–e619. [Google Scholar] [CrossRef]
  70. O’Callaghan, J.L.; Turner, R.; Dekker Nitert, M.; Barrett, H.L.; Clifton, V.; Pelzer, E.S. Re-assessing microbiomes in the low-biomass reproductive niche. BJOG 2020, 127, 147–158. [Google Scholar] [CrossRef]
  71. Zou, Y.; Liu, X.; Chen, P.; Wang, Y.; Li, W.; Huang, R. The endometrial microbiota profile influenced pregnancy outcomes in patients with repeated implantation failure: A retrospective study. J. Reprod. Immunol. 2023, 155, 103782. [Google Scholar] [CrossRef] [PubMed]
  72. Cicinelli, E.; Matteo, M.; Tinelli, R.; Lepera, A.; Alfonso, R.; Indraccolo, U.; Marrocchella, S.; Greco, P.; Resta, L. Prevalence of chronic endometritis in repeated unexplained implantation failure and the IVF success rate after antibiotic therapy. Hum. Reprod. 2015, 30, 323–330. [Google Scholar] [CrossRef] [PubMed]
  73. Vitale, S.G.; Ferrari, F.; Ciebiera, M.; Zgliczyńska, M.; Rapisarda, A.M.C.; Vecchio, G.M.; Pino, A.; Angelico, G.; Knafel, A.; Riemma, G.; et al. The Role of Genital Tract Microbiome in Fertility: A Systematic Review. Int. J. Mol. Sci. 2021, 23, 180. [Google Scholar] [CrossRef] [PubMed]
  74. Liu, Y.; Ko, E.Y.; Wong, K.K.; Chen, X.; Cheung, W.C.; Law, T.S.; Chung, J.P.; Tsui, S.K.; Li, T.C.; Chim, S.S. Endometrial microbiota in infertile women with and without chronic endometritis as diagnosed using a quantitative and reference range-based method. Fertil. Steril. 2019, 112, 707–717.e1. [Google Scholar] [CrossRef]
  75. Chen, P.; Chen, P.; Guo, Y.; Fang, C.; Li, T. Interaction Between Chronic Endometritis Caused Endometrial Microbiota Disorder and Endometrial Immune Environment Change in Recurrent Implantation Failure. Front. Immunol. 2021, 12, 748447. [Google Scholar] [CrossRef] [PubMed]
  76. Borgdorff, H.; Gautam, R.; Armstrong, S.D.; Xia, D.; Ndayisaba, G.F.; van Teijlingen, N.H.; Geijtenbeek, T.B.; Wastling, J.M.; van de Wijgert, J.H. Cervicovaginal microbiome dysbiosis is associated with proteome changes related to alterations of the cervicovaginal mucosal barrier. Mucosal. Immunol. 2016, 9, 621–633. [Google Scholar] [CrossRef]
  77. Vomstein, K.; Reider, S.; Bottcher, B.; Watschinger, C.; Kyvelidou, C.; Tilg, H.; Moschen, A.R.; Toth, B. Uterine microbiota plasticity during the menstrual cycle: Differences between healthy controls and patients with recurrent miscarriage or implantation failure. J. Reprod. Immunol. 2022, 151, 103634. [Google Scholar] [CrossRef]
  78. Cela, V.; Daniele, S.; Obino, M.E.R.; Ruggiero, M.; Zappelli, E.; Ceccarelli, L.; Papini, F.; Marzi, I.; Scarfo, G.; Tosi, F.; et al. Endometrial Dysbiosis Is Related to Inflammatory Factors in Women with Repeated Implantation Failure: A Pilot Study. J. Clin. Med. 2022, 11, 2481. [Google Scholar] [CrossRef]
  79. Tuckerman, E.; Mariee, N.; Prakash, A.; Li, T.C.; Laird, S. Uterine natural killer cells in peri-implantation endometrium from women with repeated implantation failure after IVF. J. Reprod. Immunol. 2010, 87, 60–66. [Google Scholar] [CrossRef]
  80. 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]
  81. Vacca, P.; Mingari, M.C.; Moretta, L. Natural killer cells in human pregnancy. J. Reprod. Immunol. 2013, 97, 14–19. [Google Scholar] [CrossRef]
  82. Moffett, A.; Colucci, F. Uterine NK cells: Active regulators at the maternal-fetal interface. J. Clin. Investig. 2014, 124, 1872–1879. [Google Scholar] [CrossRef]
  83. Chen, X.; Mariee, N.; Jiang, L.; Liu, Y.; Wang, C.C.; Li, T.C.; Laird, S. Measurement of uterine natural killer cell percentage in the periimplantation endometrium from fertile women and women with recurrent reproductive failure: Establishment of a reference range. Am. J. Obstet. Gynecol. 2017, 217, 680.e1–680.e6. [Google Scholar] [CrossRef] [PubMed]
  84. Fukui, A.; Kwak-Kim, J.; Ntrivalas, E.; Gilman-Sachs, A.; Lee, S.K.; Beaman, K. Intracellular cytokine expression of peripheral blood natural killer cell subsets in women with recurrent spontaneous abortions and implantation failures. Fertil. Steril. 2008, 89, 157–165. [Google Scholar] [CrossRef] [PubMed]
  85. Saito, S.; Nakashima, A.; Myojo-Higuma, S.; Shiozaki, A. The balance between cytotoxic NK cells and regulatory NK cells in human pregnancy. J. Reprod. Immunol. 2008, 77, 14–22. [Google Scholar] [CrossRef] [PubMed]
  86. King, A. Uterine leukocytes and decidualization. Hum. Reprod. Update 2000, 6, 28–36. [Google Scholar] [CrossRef] [PubMed]
  87. Wang, W.; Sung, N.; Gilman-Sachs, A.; Kwak-Kim, J. T Helper (Th) Cell Profiles in Pregnancy and Recurrent Pregnancy Losses: Th1/Th2/Th9/Th17/Th22/Tfh Cells. Front. Immunol. 2020, 11, 2025. [Google Scholar] [CrossRef]
  88. Sasaki, Y.; Sakai, M.; Miyazaki, S.; Higuma, S.; Shiozaki, A.; Saito, S. Decidual and peripheral blood CD4+CD25+ regulatory T cells in early pregnancy subjects and spontaneous abortion cases. Mol. Hum. Reprod. 2004, 10, 347–353. [Google Scholar] [CrossRef]
  89. Kaminski, V.L.; Ellwanger, J.H.; Matte, M.C.C.; Savaris, R.F.; Vianna, P.; Chies, J.A.B. IL-17 blood levels increase in healthy pregnancy but not in spontaneous abortion. Mol. Biol. Rep. 2018, 45, 1565–1568. [Google Scholar] [CrossRef]
  90. Liang, P.Y.; Diao, L.H.; Huang, C.Y.; Lian, R.C.; Chen, X.; Li, G.G.; Zhao, J.; Li, Y.Y.; He, X.B.; Zeng, Y. The pro-inflammatory and anti-inflammatory cytokine profile in peripheral blood of women with recurrent implantation failure. Reprod. Biomed. Online 2015, 31, 823–826. [Google Scholar] [CrossRef]
  91. Amjadi, F.; Zandieh, Z.; Mehdizadeh, M.; Aghajanpour, S.; Raoufi, E.; Aghamajidi, A.; Aflatoonian, R. The uterine immunological changes may be responsible for repeated implantation failure. J. Reprod. Immunol. 2020, 138, 103080. [Google Scholar] [CrossRef]
  92. Rajaei, S.; Zarnani, A.H.; Jeddi-Tehrani, M.; Tavakoli, M.; Mohammadzadeh, A.; Dabbagh, A.; Mirahmadian, M. Cytokine profile in the endometrium of normal fertile and women with repeated implantation failure. Iran. J. Immunol. 2011, 8, 201–208. [Google Scholar] [PubMed]
  93. Hortal, M.; Lledo, B.; Ortiz, J.A.; Fabregat, A.; Moliner, B.; Bernabeu, A.; Bernabeu, R. O-261 The relationship of abnormal vaginal microbiome (dysbiosis of vaginal microbiome) and proinflammatory cytokines in recurrent implantation failure patients. Hum. Reprod. 2023, 38, dead093.315. [Google Scholar] [CrossRef]
  94. Fan, T.; Zhong, X.M.; Wei, X.C.; Miao, Z.L.; Luo, S.Y.; Cheng, H.; Xiao, Q. The alteration and potential relationship of vaginal microbiota and chemokines for unexplained recurrent spontaneous abortion. Medicine 2020, 99, e23558. [Google Scholar] [CrossRef] [PubMed]
  95. Kuon, R.J.; Togawa, R.; Vomstein, K.; Weber, M.; Goeggl, T.; Strowitzki, T.; Markert, U.R.; Zimmermann, S.; Daniel, V.; Dalpke, A.H.; et al. Higher prevalence of colonization with Gardnerella vaginalis and gram-negative anaerobes in patients with recurrent miscarriage and elevated peripheral natural killer cells. J. Reprod. Immunol. 2017, 120, 15–19. [Google Scholar] [CrossRef]
  96. Zhang, F.; Zhang, T.; Ma, Y.; Huang, Z.; He, Y.; Pan, H.; Fang, M.; Ding, H. Alteration of vaginal microbiota in patients with unexplained recurrent miscarriage. Exp. Ther. Med. 2019, 17, 3307–3316. [Google Scholar] [CrossRef] [PubMed]
  97. Jiao, X.; Zhang, L.; Du, D.; Wang, L.; Song, Q.; Liu, S. Alteration of vaginal microbiota in patients with recurrent miscarriage. J. Obstet. Gynaecol. 2022, 42, 248–255. [Google Scholar] [CrossRef]
  98. Zhao, F.; Chen, Y.; Gao, J.; Wu, M.; Li, C.; Wang, Z.; Huang, N.; Cui, L.; Du, M.; Ying, C. Characterization of Vaginal Microbiota in Women With Recurrent Spontaneous Abortion That Can Be Modified by Drug Treatment. Front. Cell. Infect. Microbiol. 2021, 11, 680643. [Google Scholar] [CrossRef]
  99. Peuranpaa, P.; Holster, T.; Saqib, S.; Kalliala, I.; Tiitinen, A.; Salonen, A.; Hautamaki, H. Female reproductive tract microbiota and recurrent pregnancy loss: A nested case-control study. Reprod. Biomed. Online 2022, 45, 1021–1031. [Google Scholar] [CrossRef]
  100. Skafte-Holm, A.; Humaidan, P.; Bernabeu, A.; Lledo, B.; Jensen, J.S.; Haahr, T. The Association between Vaginal Dysbiosis and Reproductive Outcomes in Sub-Fertile Women Undergoing IVF-Treatment: A Systematic PRISMA Review and Meta-Analysis. Pathogens 2021, 10, 295. [Google Scholar] [CrossRef]
  101. Mori, R.; Hayakawa, T.; Hirayama, M.; Ozawa, F.; Yoshihara, H.; Goto, S.; Kitaori, T.; Ozaki, Y.; Sugiura-Ogasawara, M. Cervicovaginal microbiome in patients with recurrent pregnancy loss. J. Reprod. Immunol. 2023, 157, 103944. [Google Scholar] [CrossRef]
  102. Chen, S.; Xue, X.; Zhang, Y.; Zhang, H.; Huang, X.; Chen, X.; Deng, G.; Luo, S.; Gao, J. Vaginal Atopobium is Associated with Spontaneous Abortion in the First Trimester: A Prospective Cohort Study in China. Microbiol. Spectr. 2022, 10, e0203921. [Google Scholar] [CrossRef] [PubMed]
  103. 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]
  104. Campisciano, G.; Iebba, V.; Zito, G.; Luppi, S.; Martinelli, M.; Fischer, L.; De Seta, F.; Basile, G.; Ricci, G.; Comar, M. Lactobacillus iners and gasseri, Prevotella bivia and HPV Belong to the Microbiological Signature Negatively Affecting Human Reproduction. Microorganisms 2020, 9, 39. [Google Scholar] [CrossRef]
  105. Nasioudis, D.; Forney, L.J.; Schneider, G.M.; Gliniewicz, K.; France, M.; Boester, A.; Sawai, M.; Scholl, J.; Witkin, S.S. Influence of Pregnancy History on the Vaginal Microbiome of Pregnant Women in their First Trimester. Sci. Rep. 2017, 7, 10201. [Google Scholar] [CrossRef] [PubMed]
  106. Liu, Y.; Wong, K.K.; Ko, E.Y.; Chen, X.; Huang, J.; Tsui, S.K.; Li, T.C.; Chim, S.S. 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]
  107. Shi, Y.; Yamada, H.; Sasagawa, Y.; Tanimura, K.; Deguchi, M. Uterine endometrium microbiota and pregnancy outcome in women with recurrent pregnancy loss. J. Reprod. Immunol. 2022, 152, 103653. [Google Scholar] [CrossRef]
  108. Karami, N.; Boroujerdnia, M.G.; Nikbakht, R.; Khodadadi, A. Enhancement of peripheral blood CD56(dim) cell and NK cell cytotoxicity in women with recurrent spontaneous abortion or in vitro fertilization failure. J. Reprod. Immunol. 2012, 95, 87–92. [Google Scholar] [CrossRef]
  109. Lee, S.K.; Na, B.J.; Kim, J.Y.; Hur, S.E.; Lee, M.; Gilman-Sachs, A.; Kwak-Kim, J. Determination of clinical cellular immune markers in women with recurrent pregnancy loss. Am. J. Reprod. Immunol. 2013, 70, 398–411. [Google Scholar] [CrossRef] [PubMed]
  110. Seshadri, S.; Sunkara, S.K. Natural killer cells in female infertility and recurrent miscarriage: A systematic review and meta-analysis. Hum Reprod Update 2014, 20, 429–438. [Google Scholar] [CrossRef] [PubMed]
  111. Shakhar, K.; Ben-Eliyahu, S.; Loewenthal, R.; Rosenne, E.; Carp, H. Differences in number and activity of peripheral natural killer cells in primary versus secondary recurrent miscarriage. Fertil. Steril. 2003, 80, 368–375. [Google Scholar] [CrossRef] [PubMed]
  112. Katano, K.; Suzuki, S.; Ozaki, Y.; Suzumori, N.; Kitaori, T.; Sugiura-Ogasawara, M. Peripheral natural killer cell activity as a predictor of recurrent pregnancy loss: A large cohort study. Fertil. Steril. 2013, 100, 1629–1634. [Google Scholar] [CrossRef] [PubMed]
  113. Matsubayashi, H.; Shida, M.; Kondo, A.; Suzuki, T.; Sugi, T.; Izumi, S.; Hosaka, T.; Makino, T. Preconception peripheral natural killer cell activity as a predictor of pregnancy outcome in patients with unexplained infertility. Am. J. Reprod. Immunol. 2005, 53, 126–131. [Google Scholar] [CrossRef] [PubMed]
  114. Calleja-Agius, J.; Jauniaux, E.; Pizzey, A.R.; Muttukrishna, S. Investigation of systemic inflammatory response in first trimester pregnancy failure. Hum. Reprod. 2012, 27, 349–357. [Google Scholar] [CrossRef] [PubMed]
  115. Yang, X.; Tian, Y.; Zheng, L.; Luu, T.; Kwak-Kim, J. The Update Immune-Regulatory Role of Pro- and Anti-Inflammatory Cytokines in Recurrent Pregnancy Losses. Int. J. Mol. Sci. 2022, 24, 132. [Google Scholar] [CrossRef] [PubMed]
  116. Robinson, D.P.; Klein, S.L. Pregnancy and pregnancy-associated hormones alter immune responses and disease pathogenesis. Horm. Behav. 2012, 62, 263–271. [Google Scholar] [CrossRef] [PubMed]
  117. Bates, M.D.; Quenby, S.; Takakuwa, K.; Johnson, P.M.; Vince, G.S. Aberrant cytokine production by peripheral blood mononuclear cells in recurrent pregnancy loss? Hum. Reprod. 2002, 17, 2439–2444. [Google Scholar] [CrossRef]
  118. Lee, S.K.; Kim, J.Y.; Lee, M.; Gilman-Sachs, A.; Kwak-Kim, J. Th17 and regulatory T cells in women with recurrent pregnancy loss. Am. J. Reprod. Immunol. 2012, 67, 311–318. [Google Scholar] [CrossRef]
  119. Lee, S.K.; Kim, J.Y.; Hur, S.E.; Kim, C.J.; Na, B.J.; Lee, M.; Gilman-Sachs, A.; Kwak-Kim, J. An imbalance in interleukin-17-producing T and Foxp3(+) regulatory T cells in women with idiopathic recurrent pregnancy loss. Hum. Reprod. 2011, 26, 2964–2971. [Google Scholar] [CrossRef]
  120. Sereshki, N.; Gharagozloo, M.; Ostadi, V.; Ghahiri, A.; Roghaei, M.A.; Mehrabian, F.; Andalib, A.A.; Hassanzadeh, A.; Hosseini, H.; Rezaei, A. Variations in T-helper 17 and Regulatory T Cells during The Menstrual Cycle in Peripheral Blood of Women with Recurrent Spontaneous Abortion. Int. J. Fertil. Steril. 2014, 8, 59–66. [Google Scholar]
  121. Daher, S.; de Arruda Geraldes Denardi, K.; Blotta, M.H.; Mamoni, R.L.; Reck, A.P.; Camano, L.; Mattar, R. Cytokines in recurrent pregnancy loss. J. Reprod. Immunol. 2004, 62, 151–157. [Google Scholar] [CrossRef] [PubMed]
  122. Xu, L.; Huang, L.; Lian, C.; Xue, H.; Lu, Y.; Chen, X.; Xia, Y. Vaginal Microbiota Diversity of Patients with Embryonic Miscarriage by Using 16S rDNA High-Throughput Sequencing. Int. J. Genom. 2020, 2020, 1764959. [Google Scholar] [CrossRef] [PubMed]
  123. Plevyak, M.; Hanna, N.; Mayer, S.; Murphy, S.; Pinar, H.; Fast, L.; Ekerfelt, C.; Ernerudh, J.; Berg, G.; Matthiesen, L.; et al. Deficiency of decidual IL-10 in first trimester missed abortion: A lack of correlation with the decidual immune cell profile. Am. J. Reprod. Immunol. 2002, 47, 242–250. [Google Scholar] [CrossRef] [PubMed]
  124. Piccinni, M.P.; Robertson, S.A.; Saito, S. Editorial: Adaptive Immunity in Pregnancy. Front. Immunol. 2021, 12, 770242. [Google Scholar] [CrossRef] [PubMed]
  125. Liu, Y.; Chen, H.; Feng, L.; Zhang, J. Interactions between gut microbiota and metabolites modulate cytokine network imbalances in women with unexplained miscarriage. NPJ Biofilms Microbiomes 2021, 7, 24. [Google Scholar] [CrossRef] [PubMed]
  126. Baker, J.M.; Al-Nakkash, L.; Herbst-Kralovetz, M.M. Estrogen-gut microbiome axis: Physiological and clinical implications. Maturitas 2017, 103, 45–53. [Google Scholar] [CrossRef] [PubMed]
  127. Demir, B.; Guven, S.; Guven, E.S.; Atamer, Y.; Gul, T. Serum IL-6 level may have role in the pathophysiology of unexplained infertility. Am. J. Reprod. Immunol. 2009, 62, 261–267. [Google Scholar] [CrossRef]
  128. Verstraelen, H.; Verhelst, R.; Claeys, G.; De Backer, E.; Temmerman, M.; Vaneechoutte, M. Longitudinal analysis of the vaginal microflora in pregnancy suggests that L. crispatus promotes the stability of the normal vaginal microflora and that L. gasseri and/or L. iners are more conducive to the occurrence of abnormal vaginal microflora. BMC Microbiol. 2009, 9, 116. [Google Scholar] [CrossRef]
  129. France, M.T.; Ma, B.; Gajer, P.; Brown, S.; Humphrys, M.S.; Holm, J.B.; Waetjen, L.E.; Brotman, R.M.; Ravel, J. VALENCIA: A nearest centroid classification method for vaginal microbial communities based on composition. Microbiome 2020, 8, 166. [Google Scholar] [CrossRef]
  130. Kervinen, K.; Holster, T.; Saqib, S.; Virtanen, S.; Stefanovic, V.; Rahkonen, L.; Nieminen, P.; Salonen, A.; Kalliala, I. Parity and gestational age are associated with vaginal microbiota composition in term and late term pregnancies. EBioMedicine 2022, 81, 104107. [Google Scholar] [CrossRef]
  131. Baud, D.; Pattaroni, C.; Vulliemoz, N.; Castella, V.; Marsland, B.J.; Stojanov, M. Sperm Microbiota and Its Impact on Semen Parameters. Front. Microbiol. 2019, 10, 234. [Google Scholar] [CrossRef] [PubMed]
  132. Weng, S.L.; Chiu, C.M.; Lin, F.M.; Huang, W.C.; Liang, C.; Yang, T.; Yang, T.L.; Liu, C.Y.; Wu, W.Y.; Chang, Y.A.; et al. Bacterial communities in semen from men of infertile couples: Metagenomic sequencing reveals relationships of seminal microbiota to semen quality. PLoS ONE 2014, 9, e110152. [Google Scholar] [CrossRef] [PubMed]
  133. Chen, H.; Luo, T.; Chen, T.; Wang, G. Seminal bacterial composition in patients with obstructive and non-obstructive azoospermia. Exp. Ther. Med. 2018, 15, 2884–2890. [Google Scholar] [CrossRef] [PubMed]
  134. Li, J.; Wang, L.; Ding, J.; Cheng, Y.; Diao, L.; Li, L.; Zhang, Y.; Yin, T. Multiomics Studies Investigating Recurrent Pregnancy Loss: An Effective Tool for Mechanism Exploration. Front. Immunol. 2022, 13, 826198. [Google Scholar] [CrossRef]
  135. Banerjee, P.; Jana, S.K.; Pasricha, P.; Ghosh, S.; Chakravarty, B.; Chaudhury, K. Proinflammatory cytokines induced altered expression of cyclooxygenase-2 gene results in unreceptive endometrium in women with idiopathic recurrent spontaneous miscarriage. Fertil. Steril. 2013, 99, 179–187.e2. [Google Scholar] [CrossRef]
  136. Pan, H.T.; Ding, H.G.; Fang, M.; Yu, B.; Cheng, Y.; Tan, Y.J.; Fu, Q.Q.; Lu, B.; Cai, H.G.; Jin, X.; et al. Proteomics and bioinformatics analysis of altered protein expression in the placental villous tissue from early recurrent miscarriage patients. Placenta 2018, 61, 1–10. [Google Scholar] [CrossRef]
  137. Yin, X.J.; Hong, W.; Tian, F.J.; Li, X.C. Proteomic analysis of decidua in patients with recurrent pregnancy loss (RPL) reveals mitochondrial oxidative stress dysfunction. Clin. Proteomics 2021, 18, 9. [Google Scholar] [CrossRef]
Figure 1. Eubiotic vaginal and endrometrial microbiota.
Figure 1. Eubiotic vaginal and endrometrial microbiota.
Ijms 25 00622 g001
Figure 2. Alterations of the vaginal and endometrial microbiota in women with RIF.
Figure 2. Alterations of the vaginal and endometrial microbiota in women with RIF.
Ijms 25 00622 g002
Figure 3. Alterations of the vaginal and endometrial microbiota in women with RPL.
Figure 3. Alterations of the vaginal and endometrial microbiota in women with RPL.
Ijms 25 00622 g003
Table 1. Summary of the vaginal and endometrial microbiome in women with RIF.
Table 1. Summary of the vaginal and endometrial microbiome in women with RIF.
First Author (Year)Study Group CountrySpecimen TypeComposition: Phylum/Genus/SpeciesConclusion
VAGINA
Kitaya et al. (2019)RIF patients (n = 28)JapanVaginal swabLactobacillus (>90%) dominance in the RIF group. There were no significant differences in the community composition of the vaginal microbiome between the RIF group and the control group.
Control n = 18 (undergoing first IVF attempt)
Ichiyama et al. (2021) [65]Women with RIF (n = 145)JapanVaginal swabsAtopobium, Megasphaera, Gardneralla, Prevotella ↑*The vaginal microbiota of the RIF group had lower levels of Lactobacillus than in the control group.
Healthy controls (n = 21)Lactobacillus ↓*The dysbiotic microbiota from the RIF group consisted of significantly higher levels of 4 other genera.
Diaz-Martinez et al. (2021) [63]IVF patients with a RIF history (n = 23) SpainVaginal swabsGenusThere was no significant difference in Lactobacillus between the groups with RIF in history and without.
Streptococcus
IVF patients without RIF in history (n = 25)Preovotella spp., Ureaplasma spp. and Dialister spp. ↑
Species
L. helveticus
Patel et al. (2022) [67]Women with RIF (n = 10), unexplained infertility (n = 10) and healthy controls (n = 10)IndiaVaginal swabsThe dominance of Lactobacillaceae in all the groups Lactobacillus iners AB-1 dominance in the unexplained infertility group.There was no difference in lactobacillus between the group with RIF, unexplained infertility, and healthy controls.
Leptotrichia and Sneathia ↑ in control group *
ENDOMETRIUM
Kitaya et al. (2019)RIF patients (n = 28)JapanEndometrial fluidGenusThe endometrial fluid microbiota showed a significant difference in community composition between the RIF group and the control group. Burkholderia was only detected in the RIF group.
Lactobacillus (>90%) dominance Gardnerella
Control (infertile patients undergoing first IVF attempt n = 18) Burkholderia ↑*
Ichiyama et al. (2021) [65]Women with RIF (n = 145)JapanEndometrial tissue with PipetteAtopobium, Megasphaera, Gardnerella, Prevotella, Schlegelella, Delftia, Burkholderia, Sphingobacterium, Dietzia, Enterococcus, Micrococcus, Ralstonia, Leucobacter, and Hydrogenophaga ↑*The dysbiotic microbiota from the RIF group consisted of significantly higher levels of 14 other genera.
Healthy controls (n = 21)
Diaz-Martinez et al. (2021) [63]Patients with a RIF history (n = 23) SpainTranscervical endometrial tissue (Tao Brush IMC Endometrial Sampler)GenusThe RIF group had significantly different compositions of the endometrial fluid microbiota. A higher abundance of Prevotella was detected in RIF patients.
Prevotella and Sneathia amnii ↑*
and without RIF in history (n = 25)Species
L. helveticus ↑*
Keburiya et al. (2022) Women with RIF (n = 91)RussiaEndometrial tissue (Embryo transfer catheter tips) Obligate anaerobes (streptococci, enterobacteria) were found in the RIF group but with low concentrations and were insignificant. G. Vaginalis was significantly higher in the naïve IVF patients but did not have a significant impact on embryo implantation.
There was no significant difference in Lactobacillus between the RIF group and naïve IVF patients.
Women with first IVF attempt (n = 39)
Chen et al. (2022) [75]Women with RIF (n = 75)ChinaTranscervical endometrial tissue PhylumThere was a significant difference in community composition at the genus level of the RIF group compared to the control group. The dominance of Sphingomonas was found in the endometrial microbiota of the RIF group
Dominance of Firmicutes, Proteobacteria, Bacteroidetes, Cyanobacteria, and Actinobacteria
Healthy controls (n = 36) Genus
Sphingomonas, Brevundimonas, DMER64,
Methylobacterium, Rhodoferax, Caulobacter ↑*
Lactobacillus, Pseudomonas, Delftia ↓*
Vomstein et al. (2022) [77]Women with RIF (n = 20)FinlandTranscervical endometrial tissuePhylumWomen with RIF fail to attenuate their endometrial microbiome diversity and richness throughout the menstrual cycle
Firmicutes further decrease after ovulation.
Healthy controls (n = 10)Proteobacteria increase after ovulation.
Bacteroidetes peak in the follicular phase.
Gardnerella and Dialister were increased around ovulation
Cela et al. (2022) [78]Women with RIF (n = 26)ItalyTranscervical endometrial tissueGenusLactobacillus abundance is found to be negatively correlated with anti-inflammatory cytokines and positively correlated with pro-inflammatory cytokines.
With LD (n = 13)Lactobacillus ↓*
And NLDM (n = 13)Gardnerella, Streptococcus and Bifidobacterium
Zou et al. (2023) [71] Women with (n = 141)ChinaTranscervical endometrial tissueGenusDiverse pathogenic bacteria were found in high abundance in the endometrial microbiota of women with RIF.
Streptococcus, Staphylococcus, Neisseria, and Klebsiella
Iwami et al. (2023) RIF patients with endometrial microbiota data (n = 131): n = 30 (23%) with abnormal microbiota. JapanEndometrial tissue with pipelleGenusTreatment with probiotics in women with RIF and abnormal endometrial microbiota might improve IVF outcomes.
Streptococcus, Gardnerella, Atopobium and Bifidobacterium
Lozano et al. (2023)Women with RIF (n = 27) SpainTranscervical endometrial tissue (Tao Brush IUMC Endometrial sampler) GenusLactobacillus is adversely associated with pathogenic bacteria (Prevotella, Dialister, and Streptococcus), and this dysbiosis might be the cause of an increased risk of implantation failure.
Lactobacillus ↑* (control 97.96% vs. RIF 92.27%)
Women without RIF (n = 18)Prevotella ↑* (control 0.00% vs. RIF 2.19%)
Dialister ↑* (control 0.06% vs. RIF 0.15%)
Streptococcus ↑* (control 0.05% vs. RIF 0.18%)
↑ = increased abundance of the bacteria, ↓ = decreased abundance of the bacteria * = significant difference between the study group and the control group. Abbreviations: LD: Lactobacillus-dominant; NLDM: Non- Lactobacillus-dominant microbiome.
Table 2. Summary of the vaginal and endometrial microbiome in women with RPL.
Table 2. Summary of the vaginal and endometrial microbiome in women with RPL.
VAGINA
First Author (Year)Study GroupCountrySpecimen TypeComposition: Phylum/Genus/SpeciesConclusion
Kuon et al. (2017) [95]Women with RPL (n = 243)GermanyVaginal swabs GenusRPL patients with elevated peripheral natural killer cells suffer more often from colonization by G. vaginalis and gram-negative anaerobes.
Group B Streptococcus
Enterobacteriaceae
Species
Gardnerella vaginalis
Zhang et al. (2019) [96]History of recurrent miscarriages (n = 10)ChinaVaginal swabsPhylumThe dysbiotic microbiota from the RPL group consisted of an increased abundance of Atopobium, Prevotella, and Streptococcus and a decreased abundance of Lactobacillus.
Firmicutes ↑*
Actinobacteria ↓*
Bacteroidetes ↓*
Healthy controls (n = 10)Genus
Atopobium, Prevotella and Streptococcus ↑*
Lactobacillus
Al-Memar et al. (2020) [30]Women with first term miscarriages (n = 64)UKVaginal swabs GenusA reduced abundance of Lactobacillus was found in first-trimester miscarriages compared to women with viable pregnancies.
Community State Type IV was more often found in miscarriages.
Streptococcus, Prevotella, Ureaplasma, Peptoniphilus, and Dialaster
Women with full term pregnancies (n = 83) Lactobacillus spp. ↓
Fan et al. (2020) [94]Women with unexplained recurrent spontaneous abortion (n = 31) ChinaVaginal swabs PhylumThe vaginal microbiota of women with unexplained RPL has much higher alpha diversity and a higher abundance of Proteobacteria.
Proteobacteria ↑*
Normal pregnancy induced abortion (n = 27)Genus
Gammaproteobacteria, Proteobacteria, Pseudomonas, Moraxella, Ruminococcus, Collinsella aerofaciens, Alteromonadaceae, Cellvibrio, Arthrobacter, Roseburia, and Micrococcaceae
Peuranpaa et al. (2022) [99]Women with RPL (n = 47)FinlandVaginal swabsSpeciesThe vaginal microbiota of women with RPL has a higher abundance of G. vaginalis compared with healthy controls.
Healthy controls (n = 39)Gardnerella vaginalis
Caliskan et al. (2022)Women with recurrent miscarriages (n = 25) TurkeyVaginal swabsGenusA decrease in Lactobacillus spp. And an increase in anaerobic microorganisms were found in the vaginal microbiota of women with RPL.
Lactobacillus
Healthy controls (n = 25)Enterobacterium spp., Eubacterium spp.
Megasphaera spp. and Sneathia spp. ↑
Jiao et al. (2022) [97]History of recurrent miscarriages (n = 16) ChinaVaginal swabsGenusA decrease in Lactobacillus and an increase in the abundance of Atopobium and Prevotella were found in the vaginal microbiota of women with RPL.
Lactobacillus ↓*
Healthy controls (n = 20)Gardnerella ↓*
Atopobium ↑*
Liu et al. (2022) [103]Women with RPL (n = 25, with 50% chronic endometritis)ChinaVaginal swabsGenusLactobacillus was the dominant genus in the RPL and control groups.
No significant difference was found between the vaginal microbiota of the two groups.
Lactobacillus (>90%)
Bidifidobacterium
Healthy controls (n = 25, with 24% chronic endometritis)Gardnerella spp. ↓
Atopobium
Ncib et al. (2022)Women with unexplained RPL (n = 65)Italia Vaginal swabsGenusThere is a high prevalence of aerobic vaginitis-causing bacteria in women with RPL (65%).
Enterococcus,
Healthy controls with at least two live births and no miscarriages (n = 50)Staphylococcus,
Streptococcus
Mori et al. (2023) [101]Patients with unexplained RPL (n = 88)JapanVaginal swabs GenusThere is no difference in Lactobacillus and bacterial diversity between patients with RPL and live birth.
L. iners dominance of the vaginal microbiota in the Japanese study population.
Healthy controls with no history of miscarriage (n = 17)Dominance of Lactobacillus iners, Gardnerella vaginalis, Atopobium vaginae and Bifidobacterium breve.
ENDOMETRIUM
First Author (Year)Study groupCountrySamplingComposition: Phylum/Genus/SpeciesConclusion
Verstraelen et al. (2016)19 women with RIF (n = 11), RPL (n = 7) or both (n = 1)BelgiumTranscervical endometrial tissue (Tao Brush IUMC Endometrial sampler)Phylum90% of the women with either RPL or RIF had a uterine microbiota with a predominance of the phylum Bacteroidetes.
Bacteroidetes ↑
Proteobacteria ↑
Species
Bacteroides xylanisolvens, Bacteroides thetaiotaomicron, Bacteroides fragilis
Peuranpaa et al. (2022) [99]Women with RPL (n = 47)FinlandTranscervical endometrial tissueSpeciesL. crispatus is less abundant in women with RPL compared to controls (17.2% versus 45.6%).
G. vaginalis is more abundant in the RPL group.
L. crispatus
Healthy controls (n = 39)L. iners dominance
Gardnerella vaginalis
Liu et al. (2022) [103]Women with RPL (n = 25, with 50% chronic endometritis)ChinaTranscervical endometrial tissuePhylumA lower abundance of Lactobacillus spp. was found, and Acinetobacter spp. were predominant.
Healthy controls (n = 25, with 24% chronic endometritis)Proteobacteria ↑
Genus
Lactobacillus (<10%)
Anaerobacillus ↑*
Erysipelothrix ↑*
Hydrogenophilus↑*
Shi et al. (2022) [107]Patients with RPL (n = 63)JapanTranscervical endometrial tissueGenusA higher abundance of Ureaplasma species in the endometrium was associated with an increased risk of miscarriage with an euploid karyotype.
Ureaplasma spp. ↑
↑ = increased abundance of the bacteria, ↓ = decreased abundance of the bacteria, * = significant difference between the study group and the control group. Abbreviations: LD: Lactobacillus-dominant; NLDM: Non- Lactobacillus-dominant microbiome.
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

Gao, X.; Louwers, Y.V.; Laven, J.S.E.; Schoenmakers, S. Clinical Relevance of Vaginal and Endometrial Microbiome Investigation in Women with Repeated Implantation Failure and Recurrent Pregnancy Loss. Int. J. Mol. Sci. 2024, 25, 622. https://doi.org/10.3390/ijms25010622

AMA Style

Gao X, Louwers YV, Laven JSE, Schoenmakers S. Clinical Relevance of Vaginal and Endometrial Microbiome Investigation in Women with Repeated Implantation Failure and Recurrent Pregnancy Loss. International Journal of Molecular Sciences. 2024; 25(1):622. https://doi.org/10.3390/ijms25010622

Chicago/Turabian Style

Gao, Xushan, Yvonne V. Louwers, Joop S. E. Laven, and Sam Schoenmakers. 2024. "Clinical Relevance of Vaginal and Endometrial Microbiome Investigation in Women with Repeated Implantation Failure and Recurrent Pregnancy Loss" International Journal of Molecular Sciences 25, no. 1: 622. https://doi.org/10.3390/ijms25010622

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