Next Article in Journal
Y-Box Binding Proteins in mRNP Assembly, Translation, and Stability Control
Next Article in Special Issue
Spermatogonial Stem Cells in Fish: Characterization, Isolation, Enrichment, and Recent Advances of In Vitro Culture Systems
Previous Article in Journal
Effect of Citric Acid on Growth, Ecophysiology, Chloroplast Ultrastructure, and Phytoremediation Potential of Jute (Corchorus capsularis L.) Seedlings Exposed to Copper Stress
Previous Article in Special Issue
Cortisol Directly Stimulates Spermatogonial Differentiation, Meiosis, and Spermiogenesis in Zebrafish (Danio rerio) Testicular Explants
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Opportunities for Endometrial Health by Modifying Uterine Microbial Composition: Present or Future?

by
Nerea M. Molina
1,2,†,
Alberto Sola-Leyva
1,2,†,
Maria Jose Saez-Lara
1,2,3,
Julio Plaza-Diaz
2,3,4,
Aleksandra Tubić-Pavlović
5,
Barbara Romero
2,6,
Ana Clavero
2,6,
Juan Mozas-Moreno
2,6,7,8,
Juan Fontes
2,6 and
Signe Altmäe
1,2,9,*
1
Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, 18071 Granada, Spain
2
Instituto de Investigación Biosanitaria ibs.GRANADA, 18014 Granada, Spain
3
“José Mataix Verdú” Institute of Nutrition and Food Technology (INYTA), Biomedical Research Centre (CIBM), University of Granada, 18100 Granada, Spain
4
Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, 18011 Granada, Spain
5
Clinic of Gynecology and Obstetrics, Clinical Center Niš, 18000 Niš, Serbia
6
Unidad de Reproducción, UGC de Obstetricia y Ginecología, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain
7
Consortium for Biomedical Research in Epidemiology & Public Health (CIBER Epidemiología y Salud Pública-CIBERESP), 28029 Madrid, Spain
8
Departament of Obstetrics and Gynecology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
9
Competence Centre on Health Technologies, 50410 Tartu, Estonia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomolecules 2020, 10(4), 593; https://doi.org/10.3390/biom10040593
Submission received: 26 February 2020 / Revised: 6 April 2020 / Accepted: 9 April 2020 / Published: 11 April 2020
(This article belongs to the Special Issue Relevant Biomolecules for Germ Cells and Fertilization)

Abstract

:
Current knowledge suggests that the uterus harbours its own microbiota, where the microbes could influence the uterine functions in health and disease; however, the core uterine microbial composition and the host-microbial relationships remain to be fully elucidated. Different studies are indicating, based on next-generation sequencing techniques, that microbial dysbiosis could be associated with several gynaecological disorders, such as endometriosis, chronic endometritis, dysfunctional menstrual bleeding, endometrial cancer, and infertility. Treatments using antibiotics and probiotics and/or prebiotics for endometrial microbial dysbiosis are being applied. Nevertheless there is no unified protocol for assessing the endometrial dysbiosis and no optimal treatment protocol for the established dysbiosis. With this review we outline the microbes (mostly bacteria) identified in the endometrial microbiome studies, the current treatments offered for bacterial dysbiosis in the clinical setting, and the future possibilities such as pro- and prebiotics and microbial transplants for modifying uterine microbial composition.

1. Introduction

For a long time it was assumed that the uterus is a sterile organ, with microbial colonisation present only in infection or in a pathological process [1]. The ‘sterile womb’ hypothesis has been challenged by recent studies using next-generation sequencing (NGS) where unique uterine microbial composition was detected. As less than 1% of microbes can grow and form colonies on agar plates [2], analysis of the genomes of the microorganisms, i.e., microbiome, overcomes two common limitations of the traditional culture-based microbe characteristics: nonculturability and genomic diversity [3], opening a new research field in reproductive medicine.
The initial studies of endometrial microbiome suggest its association with reproductive outcomes in assisted reproduction [4,5,6,7,8,9,10,11,12,13] and with different gynaecological pathologies such as chronic endometritis (CE) [14,15,16], endometriosis [13,17,18,19,20,21], dysfunctional endometrial bleeding [22], endometrial polyps [16], and endometrial cancer or hyperplasia [23,24,25]. Nevertheless, causality has been difficult to prove because the reproductive tract represents a polymicrobial niche [26], and it is not clear whether dysbiosis within the uterus is a cause or a consequence of a pathology.
Furthermore, uterine microbial transmission is not clearly established, whereas different routes were proposed, including ascension of bacteria through the cervix, retrograde spread through fallopian tubes, haematogenous spread of oral and/or gut bacteria, through gynaecological procedures (e.g., assisted reproductive technology-related procedures; insertion/removal of the intrauterine devices), sexual habits, and/or with sperm [27,28,29,30,31]. Ascension of radioactively labelled macrospheres from vagina to the uterus by a ‘uterine peristaltic pump’ activity was shown [32,33], highlighting the most probable way for bacterial route into the uterus. Indeed, different studies suggest that microbial composition is highly influenced by the vaginal microbes [8,11,34,35,36].
Evidence for existence of certain microbiota (i.e., microbial community) in the uterus of healthy women is accumulating [14,37,38,39,40,41], but the confirmation of living microbes in the uterus is controversial and, up to now, no consensus on the core uterine microbial composition can be deduced. Uterine microbiome studies are challenged by many factors starting from the hormonal and physiological changes within the menstrual cycle, difficulty in obtaining uterine samples without contaminating the sample with vaginal/cervical bacteria, up to a high contamination risk also during samples processing and sequencing (see Figure 1 for factors). Uterine sample requires invasive sampling methods, and even when avoiding biopsy via the cervix using surgical or explorative procedures, women undergoing these procedures usually present existing medical conditions and often are peri- and postmenopausal, which could contribute to the compositional shifts in the microbiome [42]. Furthermore, the NGS-based studies conducted so far focused on detecting microbial DNA sequences. While this approach provides knowledge of possible taxa present and describes the microbiome, the mere presence of a DNA sequence does not equate with the presence of a live bacteria. Nevertheless, there are studies showing that short bacterial DNA fragments and microbial components can induce a physiological inflammatory response in the host [43,44,45]. Furthermore, other NGS methods are being applied in order to detect active microorganisms, such as metatranscriptomics which analyses microbial RNA transcripts, identifying thereby potential functionally active microorganisms [46,47]. Clearly more knowledge about the microbial composition, dynamics, and function within the uterus in health and disease is warranted in order to better understand the microbial homeostasis and dysbiosis in endometrial functions.
In this review, we summarise the microorganisms identified in microbiome studies in human endometrium, the current treatments offered for bacterial dysbiosis in clinical setting, and the future possibilities for modifying uterine microbial composition.

2. Possible Function of Microbes in the Uterus

It is becoming increasingly evident that microorganisms play an important role in our health and well-being through the production of bioactive molecules shaping a healthy microbiota, which in turn, interacts with our own cells to regulate and influence our metabolism, physiology, and immune functions that ultimately shape our health and resistance to a disease [94,95,96]. The exact role and relationship between microbes and the female reproductive tract remain to be established.
Endometrium seems to be an immunologically suited niche for microbiota with its possible function in modulating inflammatory and immune responses [30,37,97] (Figure 2). The mucosal layers of the female reproductive tract constitute a part of the mucosal immune system, which exhibits a broad repertoire of immune responses [98]. Further support for the possible immune responses in the uterus is provided by the fact that the genital epithelial cells express a wide range of pattern recognition receptors (PRRs) that promote their ability to recognise and differentially respond to various pathogens [98]. The PRRs found in the female reproductive tract include Toll-like receptors (TLRs) and NOD-like receptors, which have important roles in protecting against pathogenic invasion, in tissue adaptation and ultimately in successful reproduction [37,98]. Also pathophysiological effects of uterine microbes on the endometrial epithelium were proposed: (1) the genomic stability of uterine epithelia could be impacted via modulation of transcription factors and other genomic and epigenetic alterations; (2) the integrity of the epithelial barrier could be impaired; and (3) the microbial-secreted metabolites and the inflammation triggered by TLR activation could lead to suppression and/or overgrowth of specific bacteria [30]. The potential molecular functions of the endometrial microbes were linked to cell metabolism, motility, genetic information, immune system, and signalling processes in the NGS studies [5,20]. Regardless of all these hypotheses, future studies are needed to identify the basal uterine microbiota, its diversity and its functions and interactions with the endometrium.

3. Uterine/Endometrial Microbial Composition in Health and Disease

Several studies demonstrated that asymptomatic women harbour commensal microbial communities in their uterus [8,11,16,22,102], and that the uterine microbiome seems to be altered in women who suffer gynaecological pathologies such CE [14,15,16], endometriosis [17,19,20], dysfunctional endometrial bleeding [22], endometrial polyps [16]⁠, endometrial cancer or hyperplasia [16,23,24,25], and infertility [4,8,11,13]⁠ (Table 1). All these studies support the evidence that the uterine microbial composition is clinically relevant and requires further investigation.

3.1. Healthy Women

The uterine microbiome of ‘healthy’ women was mostly investigated as a control group in studies of infertile women or those with a gynaecological pathology [7,8,11,13,14,15,16,17,18,19,22,23,24] (Table 1). Nevertheless, these data suggest that commensal microorganisms can inhabit the upper reproductive tract of healthy women. Regardless of the growing body of literature, the core uterine microbial composition remains an open issue. There are studies concluding that the endometrium has a resident microbiome dominated by Lactobacillus species being similar to that of the vagina [8,11,16], while other studies from surgical procedures (where vaginal bacterial contamination is minimised) demonstrate that Lactobacillus is rare and that Acinetobacter, Pseudomonas and Comamonadaceae dominate [20,25,103]. Also a previous study analysing virgo intacta women identified the obligate anaerobes Jonquetella and Fusobacterium together with Preovotella as the predominant taxa in endometrium [22]. Furthermore, as 40% of the endometrial samples obtained by abdominal hysterectomy did not present any detectable microbes above the negative controls [25], this raises further doubts as to whether there is a unique uterine microbiome in all women, whether these detected bacteria are temporarily or permanently in the uterus, or whether we are dealing with contamination.

3.2. Infertility

Several publications support the theory that alterations in the endometrial microbiome may also impact the reproductive potential of infertile patients and perhaps, correcting microbial dysbiosis would lead to improve success [105]. There are numerous studies trying to associate endometrial microbiome with assisted reproduction outcomes, but just one has found a statistically significant difference in microbiome composition with success in infertility treatment outcomes [11]. In that study, Lactobacillus dominance (>90% of all bacteria) correlated positively with embryo implantation, pregnancy and live birth rates among infertile women undergoing in vitro fertilisation (IVF) [11]. Other studies have not detected any significant associations between endometrial microbiome and the treatment outcomes [4,6,7,8,9,10,12,13].

3.3. Endometriosis

The pathophysiology of endometriosis is still unclear [106]. The ‘Bacterial contamination’ hypothesis in endometriosis was proposed [107], where the inflammatory mediator lipopolysaccharide could be the initial trigger and bacterial contamination its source in the intrauterine environment serving as the primary cause in the growth regulation of endometriosis [108]. Also, considering the altered inflammatory condition in this disease, it is probable that the microbial pathogens activate the immune response by binding to host receptors. Indeed, complex bidirectional interaction between the microbiome and endometriosis is gaining evidence [109]. Nevertheless, studies on endometrial microbiome and endometriosis yielded contradicting results: Streptococcaceae, Moraxellaceae, Staphylococaceae and Enterobacteriaceae families were significantly increased while Lactobacillus species decreased in samples obtained from women with endometriosis [19], and uterine wash samples from women both with and without endometriosis detected Lactobacillus together with Barnesiella, Flavobacterium and Pseudomonas as the most predominant genera [18]. Another study investigating women with endometriosis identified the presence of Pseudomonas, Acinetobacter, Vagococcus and Sphingobium in uterus and revealed that uterine microbiota composition is significantly different in infertile women due to endometriosis [20]. A recently published study analysing endometriotic lesions found that the microbial diversity of lesions was higher compared to eutopic endometrium, where Lactobacillus, Enterococcus, Gardnerella, Pseudomonas, Alishewanella, Ureaplasma and Aerococcus prevailed [17].

3.4. Chronic Endometritis

CE is a medical condition implicated in 12–46% cases of infertile patients [110]. Indeed, different studies demonstrated that CE is favourably prevalent in patients affected by infertility, especially in case of repeated IVF failures [111]. In CE, the persistent inflammation of the endometrial mucosa is caused by the presence of bacterial pathogens in the uterine cavity [112]. The most common responsible bacteria for CE are Enterococcus faecalis, Enterobacteriaceae, Streptococcus spp., Staphylococcus spp., Gardnerella vaginalis, Mycoplasma spp. and other pathogens associated with sexually transmitted infections, such as Ureaplasma urealyticum, Chlamydia trachomatis, and Neisseria gonorrhoeae [14].

3.5. Endometrial Polyps

Endometrial polyps, a common gynaecologic disease featured as a localised overgrowth of mucosa, was correlated with CE, and continuous stimulation of biological inflammatory factors are believed to contribute to the disease [113,114]. A previous study found increased Lactobacillus, Bifidobacterium, Gardnerella, Streptococcus, Alteromonas and Euryarchaeota (Archaea) and decreased Pseudomonas and Enterobacteriaceae among endometrial samples from women with endometrial polyps when compared to non-diseased women [16] (Table 1).

3.6. Dysfunctional Menstrual Bleeding

Distinct microbial communities are believed to have a role in gynaecological pathologies such as menorrhagia and dysmenorrhea [22]. The study identified Lactobacillus dominance together with Gardnerella vaginalis, Veillonella spp., Prevotela spp., and Sneathia spp. in the endometrial samples (Table 1), and also detected menstrual cycle dependent changes within the endometrial microbiome during the proliferative and secretory phases of the cycle [22].

3.7. Endometrial Cancer

Prevailing hypotheses for endometrial cancer aetiology are focused on obesity and hormones, and do not consider the potential role of endometrial microbiota [24]. However, studies in other fields, such as cervical cancer, prostate cancer and gastrointestinal tumours, demonstrated that microorganisms play important roles in cancer causation and development [31,115,116,117,118]. In fact, 15% of tumours are estimated to be related to different infectious agents [119]. The first study analysing endometrial microbiome in patients with endometrial cancer revealed a compositional microbiome shift in the cancer cases, distinguishable from the benign cases [23]. The authors suggested that the presence of Atopobium vaginae and Porphyromonas somerae is associated with endometrial cancer [23]. Recently, the same group investigated endometrial cancer risks and they confirmed Porphyromas somerae presence as the most predictive microbial marker of endometrial cancer [24]. Meanwhile, another group studied women undergoing hysterectomy for endometrial tumours and detected Acinetobacter, Pseudomonas, Cloacibacterium, Comamonadaceae, and Escherichia as predominant taxa in the diseased endometrium [25].

4. Current Treatments in Clinical Practice

In the clinical setting, there is a high interest and demand to detect and to improve endometrial dysfunctions in order to treat uterine dysbiosis and to enhance infertility treatment outcomes. However, there is no unified protocol for assessing the endometrial microbial composition, neither for the treatment of uterine dysbiosis.
Pioneering studies based on NGS approaches already developed commercially available tests for assessing endometrial microbiome: the EMMA test by iGenomix® [120] and Endometrial Microbiome Test by Varinos Inc. [121]. The EMMA test is based on the previous study findings where Lactobacillus was dominating in the uterus, and that Lactobacillus dominance correlated with reduced miscarriage and implantation failures and thus improved pregnancy rates in women undergoing IVF [11]. EMMA test classifies endometrial samples into Lactobacillus dominant and non-Lactobacillus dominant profiles. Once the sample is classified as non-Lactobacillus dominant, adequate treatment including antibiotic, probiotics and prebiotics could be applied. In the same line, the Varinos test supports the Lactobacillus dominance in the uterus and categorises endometrial microbiome as Lactobacillus dominant and non-Lactobacillus dominant [9]. Then, choices for intervention are suggested, such as uterine lavage for microbial eradication, eradication treatment with antibiotics, and/or taking probiotics and prebiotics for improving the microbiota. Undoubtedly useful, these tests are based on mostly observational studies and include limited number of patients, resulting in rather limited evidence for testing and subsequent clinical decision-making in a clinical setting [122]. Clearly more research in the field is required starting with establishment of the core uterine microbiome before any treatment recommendations for ‘dysbiosis’ are offered for patients.

4.1. Antibiotics

The use of antibiotics is widely applied in gynaecology and obstetrics [123]. Antibiotics use ranges from diagnostic techniques (e.g., hysterosalpingography, sonography, hysteroscopy, and laparoscopy) to embryo transfer and up to diseases such as endometriosis and endometritis [123]. It is clear that the use of antibiotics produces fluctuation in microorganism communities within the female reproductive tract.
In the infertility clinical setting, a broad-spectrum antibiotic therapy (i.e., amoxicillin or levofloxacin) was recently used to modulate the non-Lactobacillus dominant endometrial environment into Lactobacillus dominant, followed by the combination of antibiotics, prebiotics and/or probiotics, 53% (N = 17) of patients achieved Lactobacillus dominant state in the uterus while showing a higher but not statistically significant pregnancy rates compared to the non-Lactobacillus dominant group [9]. These results are found to be encouraging, and this treatment approach is gaining popularity in the clinics.
CE is the gynaecological condition that is believed to benefit the most from the microbial modulation with antibiotics, as the bacterial infection in this condition is known [124]. Several studies showed that the use of antibiotics in CE has improved the reproductive outcomes. CE patients treated with antibiotics before embryo implantation had significantly better reproductive outcomes compared to those not treated with antibiotics [125], suggesting that the negative impact of CE on reproductive outcomes may be in part attributable to the presence of certain uterine bacteria. In line, McQueen et al. demonstrated that antibiotics treatment among the recurrent pregnancy loss patients with CE improved live birth rate from 7% to 56% after the treatment [126]. In a retrospective study conducted by Cicinell et al. the pregnancy and live birth rates were significantly higher in cured CE women treated with antibiotics when compared with the persistent CE group and even with the non-CE group [111]. Furthermore, Kitaya et al. reported that women with recurrent implantation failure (RIF) treated with antibiotics for CE had higher live birth rates compared with RIF patients with no CE [127]. In addition, the study conducted by Zhang et al. demonstrated that patients diagnosed with RIF and CE when treated with intrauterine antibiotic infusion therapy had significantly higher implantation and pregnancy rates when compared with RIF women without CE and with persistent CE groups [128]. Furthermore, intrauterine infusion of antibiotics after poor oral antibiotic therapy outcomes has also been applied in order to restore the physiological condition of the uterus in CE, and the three patients assessed achieved pregnancy after the treatment [129].
Women with endometriosis is another group of patients that could benefit from the uterine microbial modulation by antibiotics, as the hypothesis of ‘bacterial contamination’ in endometriosis was proposed [107]. Among patients who suffer endometriosis, the prophylactic antibiotic therapy was applied prior to oocyte retrieval with the aim to reduce infection; with the treatment the infection risk reached 0% [130]. In fact, prophylactic administration of antibiotics prior to oocyte retrieval in patients suffering from severe endometriosis is today commonly applied due to the risk of infection [123]. In animal models, mice with endometriosis were treated with broad-spectrum antibiotics and it resulted in a reduction of endometriotic lesions, supporting the possible role of microbes in endometriosis progression [131].
Undoubtedly effective in modulating uterine microbiome, best administration regimens and their effects on endometrial microbiome should be tested further in order to understand the impact on health and to avoid unnecessary antibiotic use.

4.2. Probiotics

Probiotics are defined as ‘live microorganisms that, when administered in adequate amounts, confer a health benefit in the host’, capturing the essence of probiotics, as microbial, viable and beneficial to the health [132]. Probiotic bacteria produce bioactive molecules that could act on the body and promote good health. These formulations might be a safe and effective alternative to antibiotics in restoring the imbalance of the uterine microbiota as seen in female urogenital disorders [133,134]. In the clinical setting, there is a high interest and demand to improve aberrant endometrial microbial environment in order to overcome dysbiosis and to enhance infertility treatment outcomes.
Probiotic interventions were extensively investigated in relation to systemic inflammation [135]. The first probiotics were composed predominantly of members of Lactobacillus and Bifidobacterium, but they showed a lack of precision when targeting a specific biological function [136]. In the male reproductive field, the use of probiotics and synbiotics (formulations including probiotics and prebiotics) was related to improved semen quality [137,138], and in females the protective role of probiotics in relation to vaginal infections was observed [70,139]. A recent study aimed to select potential probiotic strains from vaginal samples to improve the health of the female reproductive tract, and selected the Lactobacillus rhamnosus BPL005 strain for its capacity to reduce pH and produce short chain fatty acids, protective effects against pathogen colonisation and lactate production [140].
Women suffering from endometriosis are shown to benefit from the administration of oral Lactobacillus by reduction in pain [141,142]. In the mouse model, the beneficial effect of Lactobacillus on endometriosis by increasing interleukin-12 levels and the activity of natural killer cells was suggested, and the administration of probiotics resulted in a reduction of endometriotic lesions [143,144,145].
Female genital microbiota modulation could also be used to fight or protect against infection. Recent research has demonstrated how probiotic lactobacilli (Lactobacillus reuteri RC-14 and Lactobacillus rhamnosus GR-1) can improve endometrial epithelial cells barrier function in response to human immunodeficiency virus-1 (HIV-1) [146]. It was also detected that bacterial strains could modulate the immune profile suggesting that the microbiota of the female reproductive tract is an important factor in the acquisition of resistance to the virus [146].
The field of probiotics in modulating uterine microenvironment is very promising, nevertheless we have to bear in mind that probiotics are not medical drugs and they often lack detailed product information, and only a few probiotics were investigated thoroughly in clinical trials [122].

4.3. Prebiotics

Prebiotics target microbes living in/on human with the result of improving health, providing non-viable substrates that serve as nutrients for beneficial microorganisms harboured by the host [147]. In an attempt to modify endometrial microbiome lactoferrin a prebiotic agent with favourable prebiotic activity was administered orally during and after treatment with antibiotics among women undergoing infertility treatment [9]. Among non-Lactobacillus dominant patients treated with lactoferrin for three month after the antibiotics therapy, 67% (6/9) of them reached Lactobacillus dominance in the endometrium [9]. Additionally, lactoferrin administration has demonstrated effective result against bacterial vaginosis, leading to pregnancy and full-term birth in women with a previous medical history of preterm birth [148].
Prebiotics use in the human reproductive field is in its infancy and it is for future studies to unravel its usefulness in modifying microbial niches in female reproductive tract.

4.4. Microbial Transplants

Faecal microbiota transfer (FMT) is increasingly being used for various indications; however, clear evidence for the efficacy of FMT currently exists only for recurrent Clostridioides difficile infection [149]. Over 100 clinical trials using FMT for different conditions are currently ongoing (ClinicalTrials.gov). The problem arises from the fact that FMT is not only being used in clinical trials, but also applied on individual patients with methods that are not publicly documented [149]. In this scenario, the required screening of FMT donors is not always performed in a standardised way, which can cause different side effects and complications among patients. In fact, the Food and Drug Administration (FDA, USA) has communicated a warning regarding the risk of severe bacterial infection after FMT [149]. The hypothesis for the uterine bacterial transmission route originating from the gut exists [30,150], thus FMT was proposed as a promising (future) tool for treatment female reproductive tract diseases [151]. It was shown on broad-spectrum antibiotics-treated endometriosis mice that the FMT from mice with endometriosis resumed the growth of endometriotic lesions suggesting that the gut microbiota could promote endometriosis progression [131]. In humans, differences in gut microbial composition between healthy women and women with endometriosis [152] and polycystic ovary syndrome were reported [153,154,155,156,157,158].
A new area of microbial transplants is arising—vaginal microbiota transplants (VMT), which is opening new frontiers for reproductive health [159]. VMT involves the transfer of cervicovaginal fluid from a ‘healthy’ donor to a patient who aims to restore the most beneficial microenvironment. A pioneering study has tested the use of VMT from healthy donors as therapeutic alternative for patients suffering from symptomatic, intractable and recurrent vaginosis, and reported positive treatment outcomes [160]. Since the uterine colonisation of microorganisms by vaginal-cervical ascension is known [35,36], VMT could open up a future way for managing endometrial dysbiosis. In addition, what about uterine microbiota transfer (UMT)? To conclude, microbial transplants are highly promising ways for modifying uterine microbiota, nevertheless thorough research and testing in randomised, placebo-controlled trials is warranted.

5. Challenges in Developing Targeted Modulators for Uterine Microbiota

The uterus represents an ideal organ for drug administration, possessing advantages such as the possibility of bypass first-pass metabolism, high permeability for low molecular weight drugs, considerable surface area for absorption, and rich blood supply [161]. However, the effectiveness of the site would depend on intrinsic factors that include pH, temperature, uterine fluid composition, viscosity, enzymatic metabolism, clearance, and others, together with the hormonal fluctuations throughout the menstrual cycle. For instance, uterine pH was shown to change along the menstrual cycle, oscillating between 6.4 and 7 [162] and this could modify the drug-release system since the vast majority of drugs possess an ionisable group (mostly weak bases) [163]. Other biophysical parameters such as oxygen tension (pO2) and temperature are also factors with probable influence on the modulators. pO2 is shown to have cyclical variation and minute-to-minute oscillations within human uterus [164]. Also, the temperature variation is cyclical by day and month, increasing in the luteal phase and is influenced by hormones, density of uterine vascular beds and effectiveness of local heat exchange [164]. It is clear that many factors could influence the drug-release system inside the uterus, and a detailed study of these properties are required in order to develop effective targeted microbiota modulators with the exact dose required, necessary time and right place.
Regardless of the different intrinsic factors, probably one of the most important challenges that arises in modulating the endometrial microbiome is the fact that it is a low microbial biomass niche [26,165]. It is clear that analyses of low-biomass microbial sites are sensitive to contamination (especially from lower genital tract) and data misinterpretation. Thus, researchers face hurdles when describing the baseline microbial communities in endometrium and require well-designed and well-controlled experiments in order to avoid and adjust for the risk of contamination [26,91]. This makes especially necessary to set up standardised detection and description methods for analysing the uterine microbial composition, as well as strategies to re-establish/maintain these microbial populations. The challenge of assessing the true microbial composition in endometrial eu- and dysbiosis, the adequate dosing and evaluating these effects remains for future studies.

6. Conclusions and Future Directions

The conventional approach to target bacterial dysbiosis has been and continues to be the use of antibiotics, which were shown to be both essential and effective for treating infections usually resulting from pathogen proliferation. Nevertheless, the antibiotic drugs lead to unintended off-target effects on microbial community structure which frequently causes adverse effects, making them less appealing as precise therapy to target microbiome [136]. In many cases, its administration aggravates the underlying dysbiosis in long term and may promote resistance [140]. For instance, prescription of antibiotics in a prophylactical way has not resulted in improved pregnancy outcome not even in the cases of high risk of preterm birth on the presence of pathogens [166].
New therapies, such as pro- and prebiotic administration, and microbiota transplants are gaining popularity for improving and maintaining the optimal composition of the microbiota (Figure 3). These approaches attempt to modulate the microbial communities in a way that is beneficial to human health. However, several important questions related to these new clinical strategies remain unresolved, such as indication for prescription, comparative efficacy of monostrain and multistrain probiotics, choice of excipients, and methods of administration and delivery [134]. Furthermore, procedures to develop reliable and reproducible microbiome-based therapeutic approaches represent a challenge [136]. Currently, the pharmacological mechanisms of probiotics and prebiotics’ action are poorly understood, providing not enough evidence to support the use of probiotics for medicinal purposes [134]. Of those commercially available probiotics, it is especially difficult to prove their clinical efficacy as they are based on studies on a small sample size, and the heterogeneity in strains of bacteria used, duration of treatment and the lifestyle of patients, which can also influence the effects of probiotic supplementation [167].
Regarding the human uterus, there are several studies where the effect of probiotic supplementation on the endometrial microbiota was studied, although mainly in combination with antibiotic treatment [9,140,141,142,145]. The use of alternative modulators for uterine microbes is a highly demanded and relevant area of investigation with direct clinical application. Nevertheless, before any treatment strategies could be offered, the core uterine microbial composition needs to be established. In fact, there is an active debate ongoing whether uterus harbours a unique microbiota or not [25]. If the microbiome is certainly present in the uterine environment in the absence of pathologic infection, current data support that it is of low abundance [26,168], and several technical challenges in studies of low-biomass samples exist and make difficult to distinguish microorganisms that are truly present in small quantities from those arisen from contamination [26,91,92]. Thus, the appropriate uterine microenvironment for non-pathological conditions has yet to be established and applying methods for targeted modification of microbial communities is premature.
It is clear that in the case of infection caused by a pathogen, antibiotic treatment is required. However, in the case of prophylactics or suggested dysbiosis based on the molecular detecting methods (endometrial microbiome tests), today is too early to intervene and offer treatment recommendations for patients. In fact, no clinical recommendations are today available for diagnosis of ‘abnormal/unfavourable uterine microbiota’ [122]. To sum up, modulation of uterine microbiota for restoring and maintain microbial composition is a promising field of research and application with high clinical relevance, but we are not there yet and hopefully soon this promising future will be present.

Author Contributions

Conceptualisation, N.M.M., A.S.-L., M.J.S-L., J.P.-D., and S.A.; writing—original draft preparation, N.M.M., A.S.-L. and S.A.; writing—review and editing N.M.M., A.S.-L., M.J.S.-L., J.P.-D., A.T.-P., B.R., A.C., J.M.-M., J.F., and S.A. All authors have read and agreed to the published version of the manuscript.

Funding

This work is supported by the Spanish Ministry of Economy, Industry and Competitiveness (MINECO) and European Regional Development Fund (FEDER): grants RYC-2016-21199 and ENDORE SAF2017-87526-R; Programa Operativo FEDER Andalucía (B-CTS-500-UGR18) and by the University of Granada Plan Propio de Investigación 2016—Excellence actions: Unit of Excellence on Exercise and Health (UCEES)—and Plan Propio de Investigación 2018—Programa Contratos-Puente, and the Junta de Andalucía, Consejería de Conocimiento, Investigación y Universidades, European Regional Development Funds (ref. SOMM17/6107/UGR). A.S.-L. is funded by the Spanish Ministry of Science, Innovation and Universities (PRE2018-0854409).

Acknowledgments

We acknowledge the research support by Copan Italia S.p.A Inc., and Clearblue, SPD Swiss Precision Diagnostics GmbH. This study is part of a PhD Thesis conducted at the Official Doctoral Programme in Biomedicine of the University of Granada, Spain. We are grateful to Ana Yara Postigo-Fuentes for her assistance with English language.

Conflicts of Interest

S.A. has received honoraria for lectures from Merck. The funder had no role in this review. The rest of the authors declare no conflict of interest.

References

  1. Evans, J.; Salamonsen, L.A.; Winship, A.; Menkhorst, E.; Nie, G.; Gargett, C.E.; Dimitriadis, E. Fertile ground: Human endometrial programming and lessons in health and disease. Nat. Rev. Endocrinol. 2016, 12, 654–667. [Google Scholar] [CrossRef] [PubMed]
  2. Wade, W. Unculturable bacteria--the uncharacterized organisms that cause oral infections. J. R. Soc. Med. 2002, 95, 81–83. [Google Scholar] [PubMed]
  3. Giudice, L.C. Challenging dogma: The endometrium has a microbiome with functional consequences! Am. J. Obstet. Gynecol. 2016, 215, 682–683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Franasiak, J.M.; Werner, M.D.; Juneau, C.R.; Tao, X.; Landis, J.; Zhan, Y.; Treff, N.R.; Scott, R.T. Endometrial microbiome at the time of embryo transfer: Next-generation sequencing of the 16S ribosomal subunit. J. Assist. Reprod. Genet. 2016, 33, 129–136. [Google Scholar] [CrossRef]
  5. Garcia-Grau, I.; Perez-Villaroya, D.; Bau, D.; Gonzalez-Monfort, M.; Vilella, F.; Moreno, I.; Simon, C. Taxonomical and Functional Assessment of the Endometrial Microbiota in A Context of Recurrent Reproductive Failure: A Case Report. Pathogens 2019, 8, 205. [Google Scholar] [CrossRef] [Green Version]
  6. Hashimoto, T.; Kyono, K. Does dysbiotic endometrium affect blastocyst implantation in IVF patients? J. Assist. Reprod. Genet. 2019, 36, 2471–2479. [Google Scholar] [CrossRef] [Green Version]
  7. Kitaya, K.; Nagai, Y.; Arai, W.; Sakuraba, Y.; Ishikawa, T. Characterization of Microbiota in Endometrial Fluid and Vaginal Secretions in Infertile Women with Repeated Implantation Failure. Mediators Inflamm. 2019, 2019, 4893437. [Google Scholar] [CrossRef] [Green Version]
  8. Kyono, K.; Hashimoto, T.; Nagai, Y.; Sakuraba, Y. Analysis of endometrial microbiota by 16S ribosomal RNA gene sequencing among infertile patients: A single-center pilot study. Reprod. Med. Biol. 2018, 17, 297–306. [Google Scholar] [CrossRef]
  9. 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] [Green Version]
  10. Liu, Y.; Wong, K.K.-W.; Ko, E.Y.-L.; Chen, X.; Huang, J.; Tsui, S.K.-W.; Li, T.C.; Chim, S.S.-C. Systematic Comparison of Bacterial Colonization of Endometrial Tissue and Fluid Samples in Recurrent Miscarriage Patients: Implications for Future Endometrial Microbiome Studies. Clin. Chem. 2018, 64, 1743–1752. [Google Scholar] [CrossRef] [Green Version]
  11. Moreno, I.; Codoñer, F.M.; Vilella, F.; Valbuena, D.; Martinez-Blanch, J.F.; Jimenez-Almazán, J.; Alonso, R.; Alamá, P.; Remohí, J.; Pellicer, A.; et al. Evidence that the endometrial microbiota has an effect on implantation success or failure. Am. J. Obstet. Gynecol. 2016, 215, 684–703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Verstraelen, H.; Vilchez-Vargas, R.; Desimpel, F.; Jauregui, R.; Vankeirsbilck, N.; Weyers, S.; Verhelst, R.; De Sutter, P.; Pieper, D.H.; Van De Wiele, T. Characterisation of the human uterine microbiome in non-pregnant women through deep sequencing of the V1-2 region of the 16S rRNA gene. PeerJ 2016, 4, e1602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Wee, B.A.; Thomas, M.; Sweeney, E.L.; Frentiu, F.D.; Samios, M.; Ravel, J.; Gajer, P.; Myers, G.; Timms, P.; Allan, J.A.; et al. A retrospective pilot study to determine whether the reproductive tract microbiota differs between women with a history of infertility and fertile women. Aust. N. Z. J. Obstet. Gynaecol. 2018, 58, 341–348. [Google Scholar] [CrossRef] [PubMed]
  14. Moreno, I.; Cicinelli, E.; Garcia-Grau, I.; Gonzalez-Monfort, M.; Bau, D.; Vilella, F.; De Ziegler, D.; Resta, L.; Valbuena, D.; Simon, C. The diagnosis of chronic endometritis in infertile asymptomatic women: A comparative study of histology, microbial cultures, hysteroscopy, and molecular microbiology. Am. J. Obstet. Gynecol. 2018, 218, 602.e1–602.e16. [Google Scholar] [CrossRef] [PubMed]
  15. Liu, Y.; Ko, E.Y.-L.; Wong, K.K.-W.; Chen, X.; Cheung, W.-C.; Law, T.S.-M.; Chung, J.P.-W.; Tsui, S.K.-W.; Li, T.-C.; Chim, S.S.-C. Endometrial microbiota in infertile women with and without chronic endometritis as diagnosed using a quantitative and reference range-based method. Fertil. Steril. 2019, 112. [Google Scholar] [CrossRef] [PubMed]
  16. Fang, R.-L.; Chen, L.-X.; Shu, W.-S.; Yao, S.-Z.; Wang, S.-W.; Chen, Y.-Q. Barcoded sequencing reveals diverse intrauterine microbiomes in patients suffering with endometrial polyps. Am. J. Transl. Res. 2016, 8, 1581–1592. [Google Scholar] [PubMed]
  17. Hernandes, C.; Silveira, P.; Rodrigues Sereia, A.F.; Christoff, A.P.; Mendes, H.; Valter de Oliveira, L.F.; Podgaec, S. Microbiome Profile of Deep Endometriosis Patients: Comparison of Vaginal Fluid, Endometrium and Lesion. Diagnostics 2020, 10, 163. [Google Scholar] [CrossRef] [Green Version]
  18. Cregger, M.A.; Lenz, K.; Leary, E.; Leach, R.; Fazleabas, A.; White, B.; Braundmeier, A. Reproductive Microbiomes: Using the Microbiome as a Novel Diagnostic Tool for Endometriosis. Reprod. Immunol. Open Access 2017, 2, 36. [Google Scholar] [CrossRef]
  19. Khan, K.N.; Fujishita, A.; Masumoto, H.; Muto, H.; Kitajima, M.; Masuzaki, H.; Kitawaki, J. Molecular detection of intrauterine microbial colonization in women with endometriosis. Eur. J. Obstet. Gynecol. Reprod. Biol. 2016, 199, 69–75. [Google Scholar] [CrossRef]
  20. Chen, C.; Song, X.; Wei, W.; Zhong, H.; Dai, J.; Lan, Z.; Li, F.; Yu, X.; Feng, Q.; Wang, Z.; et al. The microbiota continuum along the female reproductive tract and its relation to uterine-related diseases. Nat. Commun. 2017, 8, 875. [Google Scholar] [CrossRef] [Green Version]
  21. Li, F.; Chen, C.; Wei, W.; Wang, Z.; Dai, J.; Hao, L.; Song, L.; Zhang, X.; Zeng, L.; Du, H.; et al. The metagenome of the female upper reproductive tract. Gigascience 2018, 7, giy107. [Google Scholar] [CrossRef] [PubMed]
  22. Pelzer, E.S.; Willner, D.; Buttini, M.; Huygens, F. A role for the endometrial microbiome in dysfunctional menstrual bleeding. Antonie van Leeuwenhoek 2018, 111, 933–943. [Google Scholar] [CrossRef] [PubMed]
  23. Walther-António, M.R.S.; Chen, J.; Multinu, F.; Hokenstad, A.; Distad, T.J.; Cheek, E.H.; Keeney, G.L.; Creedon, D.J.; Nelson, H.; Mariani, A.; et al. Potential contribution of the uterine microbiome in the development of endometrial cancer. Genome Med. 2016, 8, 122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Walsh, D.M.; Hokenstad, A.N.; Chen, J.; Sung, J.; Jenkins, G.D.; Chia, N.; Nelson, H.; Mariani, A.; Walther-Antonio, M.R.S. Postmenopause as a key factor in the composition of the Endometrial Cancer Microbiome (ECbiome). Sci. Rep. 2019, 9, 19213. [Google Scholar] [CrossRef] [PubMed]
  25. Winters, A.D.; Romero, R.; Gervasi, M.T.; Gomez-Lopez, N.; Tran, M.R.; Garcia-Flores, V.; Pacora, P.; Jung, E.; Hassan, S.S.; Hsu, C.-D.; et al. Does the endometrial cavity have a molecular microbial signature? Sci. Rep. 2019, 9, 9905. [Google Scholar] [CrossRef]
  26. 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]
  27. Altmäe, S. Uterine Microbiota : A Role Beyond Infection. EMJ Reprod. Heal. 2018, 6, 70–75. [Google Scholar]
  28. Altmäe, S. Commentary: Uterine Microbiota: Residents, Tourists, or Invaders? Front. Immunol. 2018, 9, 1874. [Google Scholar] [CrossRef]
  29. Mändar, R.; Punab, M.; Borovkova, N.; Lapp, E.; Kiiker, R.; Korrovits, P.; Metspalu, A.; Krjutškov, K.; Nõlvak, H.; Preem, J.-K.; et al. Complementary seminovaginal microbiome in couples. Res. Microbiol. 2015, 166, 440–447. [Google Scholar] [CrossRef]
  30. Baker, J.M.; Chase, D.M.; Herbst-Kralovetz, M.M. Uterine Microbiota: Residents, Tourists, or Invaders? Front. Immunol. 2018, 9, 208. [Google Scholar] [CrossRef] [Green Version]
  31. Altmäe, S.; Franasiak, J.M.; Mändar, R. The seminal microbiome in health and disease. Nat. Rev. Urol. 2019, 16, 703–721. [Google Scholar] [CrossRef] [PubMed]
  32. Kunz, G.; Beil, D.; Deiniger, H.; Einspanier, A.; Mall, G.; Leyendecker, G. The uterine peristaltic pump. Normal and impeded sperm transport within the female genital tract. Adv. Exp. Med. Biol. 1997, 424, 267–277. [Google Scholar] [PubMed]
  33. Zervomanolakis, I.; Ott, H.W.; Hadziomerovic, D.; Mattle, V.; Seeber, B.E.; Virgolini, I.; Heute, D.; Kissler, S.; Leyendecker, G.; Wildt, L. Physiology of upward transport in the human female genital tract. Ann. N. Y. Acad. Sci. 2007, 1101, 1–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Mitchell, C.M.; Haick, A.; Nkwopara, E.; Garcia, R.; Rendi, M.; Agnew, K.; Fredricks, D.N.; Eschenbach, D. Colonization of the upper genital tract by vaginal bacterial species in nonpregnant women. Am. J. Obstet. Gynecol. 2015, 212, 611.e1–611.e19. [Google Scholar] [CrossRef] [Green Version]
  35. Romero, R.; Gomez-Lopez, N.; Winters, A.D.; Jung, E.; Shaman, M.; Bieda, J.; Panaitescu, B.; Pacora, P.; Erez, O.; Greenberg, J.M.; et al. Evidence that intra-amniotic infections are often the result of an ascending invasion - a molecular microbiological study. J. Perinat. Med. 2019, 47, 915–931. [Google Scholar] [CrossRef]
  36. Vornhagen, J.; Armistead, B.; Santana-Ufret, V.; Gendrin, C.; Merillat, S.; Coleman, M.; Quach, P.; Boldenow, E.; Alishetti, V.; Leonhard-Melief, C.; et al. Group B streptococcus exploits vaginal epithelial exfoliation for ascending infection. J. Clin. Invest. 2018, 128, 1985–1999. [Google Scholar] [CrossRef]
  37. 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] [Green Version]
  38. Smolnikova, V.; Keburiya, L.; Priputnevich, T.; Muravieva, V.; Kalinina, E.; Sukhikh, G. Influence of Endometrial Microbiota on Reproductive Outcomes in IVF Programs. Am. J. Biomed. Sci. Res. 2019, 4, 197–200. [Google Scholar] [CrossRef]
  39. Egbase, P.E.; Al-Sharhan, M.; Al-Othman, S.; Al-Mutawa, M.; Udo, E.E.; Grudzinskas, J.G. Incidence of microbial growth from the tip of the embryo transfer catheter after embryo transfer in relation to clinical pregnancy rate following in-vitro fertilization and embryo transfer. Hum. Reprod. 1996, 11, 1687–1689. [Google Scholar] [CrossRef] [Green Version]
  40. Moore, D.E.; Soules, M.R.; Klein, N.A.; Fujimoto, V.Y.; Agnew, K.J.; Eschenbach, D.A. Bacteria in the transfer catheter tip influence the live-birth rate after in vitro fertilization. Fertil. Steril. 2000, 74, 1118–1124. [Google Scholar] [CrossRef]
  41. Salim, R.; Ben-Shlomo, I.; Colodner, R.; Keness, Y.; Shalev, E. Bacterial colonization of the uterine cervix and success rate in assisted reproduction: Results of a prospective survey. Hum. Reprod. 2002, 17, 337–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Peric, A.; Weiss, J.; Vulliemoz, N.; Baud, D.; Stojanov, M. Bacterial Colonization of the Female Upper Genital Tract. Int. J. Mol. Sci. 2019, 20, 3405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Schindler, R.; Beck, W.; Deppisch, R.; Aussieker, M.; Wilde, A.; Göhl, H.; Frei, U. Short bacterial DNA fragments: Detection in dialysate and induction of cytokines. J. Am. Soc. Nephrol. 2004, 15, 3207–3214. [Google Scholar] [CrossRef] [PubMed]
  44. Potgieter, M.; Bester, J.; Kell, D.B.; Pretorius, E. The dormant blood microbiome in chronic, inflammatory diseases. FEMS Microbiol. Rev. 2015, 39, 567–591. [Google Scholar] [CrossRef] [Green Version]
  45. Kell, D.B.; Kenny, L.C. A Dormant Microbial Component in the Development of Preeclampsia. Front. Med. 2016, 3, 60. [Google Scholar] [CrossRef] [Green Version]
  46. Knight, R.; Vrbanac, A.; Taylor, B.C.; Aksenov, A.; Callewaert, C.; Debelius, J.; Gonzalez, A.; Kosciolek, T.; McCall, L.-I.; McDonald, D.; et al. Best practices for analysing microbiomes. Nat. Rev. Microbiol. 2018, 16, 410–422. [Google Scholar] [CrossRef] [Green Version]
  47. Bashiardes, S.; Zilberman-Schapira, G.; Elinav, E. Use of Metatranscriptomics in Microbiome Research. Bioinform. Biol. Insights 2016, 10, 19–25. [Google Scholar] [CrossRef] [Green Version]
  48. Takagi, T.; Naito, Y.; Inoue, R.; Kashiwagi, S.; Uchiyama, K.; Mizushima, K.; Tsuchiya, S.; Dohi, O.; Yoshida, N.; Kamada, K.; et al. Differences in gut microbiota associated with age, sex, and stool consistency in healthy Japanese subjects. J. Gastroenterol. 2019, 54, 53–63. [Google Scholar] [CrossRef]
  49. Hopkins, M.J.; Sharp, R.; Macfarlane, G.T. Age and disease related changes in intestinal bacterial populations assessed by cell culture, 16S rRNA abundance, and community cellular fatty acid profiles. Gut 2001, 48, 198–205. [Google Scholar] [CrossRef]
  50. Salazar, N.; Arboleya, S.; Valdés, L.; Stanton, C.; Ross, P.; Ruiz, L.; Gueimonde, M.; de Los Reyes-Gavilán, C.G. The human intestinal microbiome at extreme ages of life. Dietary intervention as a way to counteract alterations. Front. Genet. 2014, 5, 406. [Google Scholar] [CrossRef]
  51. Mändar, R.; Punab, M.; Korrovits, P.; Türk, S.; Ausmees, K.; Lapp, E.; Preem, J.-K.; Oopkaup, K.; Salumets, A.; Truu, J. Seminal microbiome in men with and without prostatitis. Int. J. Urol. 2017, 24, 211–216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Zhou, X.; Brown, C.J.; Abdo, Z.; Davis, C.C.; Hansmann, M.A.; Joyce, P.; Foster, J.A.; Forney, L.J. Differences in the composition of vaginal microbial communities found in healthy Caucasian and black women. ISME J. 2007, 1, 121–133. [Google Scholar] [CrossRef] [PubMed]
  53. Zhou, X.; Hansmann, M.A.; Davis, C.C.; Suzuki, H.; Brown, C.J.; Schütte, U.; Pierson, J.D.; Forney, L.J. The vaginal bacterial communities of Japanese women resemble those of women in other racial groups. FEMS Immunol. Med. Microbiol. 2010, 58, 169–181. [Google Scholar] [CrossRef] [PubMed]
  54. Fettweis, J.M.; Brooks, J.P.; Serrano, M.G.; Sheth, N.U.; Girerd, P.H.; Edwards, D.J.; Strauss, J.F.; The Vaginal Microbiome Consortium; Jefferson, K.K.; Buck, G.A. Differences in vaginal microbiome in African American women versus women of European ancestry. Microbiology 2014, 160, 2272–2282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Dunlop, A.L.; Knight, A.K.; Satten, G.A.; Cutler, A.J.; Wright, M.L.; Mitchell, R.M.; Read, T.D.; Mulle, J.; Hertzberg, V.S.; Hill, C.C.; et al. Stability of the vaginal, oral, and gut microbiota across pregnancy among African American women: The effect of socioeconomic status and antibiotic exposure. PeerJ 2019, 7, e8004. [Google Scholar] [CrossRef] [Green Version]
  56. Genc, M.R.; Onderdonk, A. Endogenous bacterial flora in pregnant women and the influence of maternal genetic variation. BJOG 2011, 118, 154–163. [Google Scholar] [CrossRef]
  57. Garcia-Velasco, J.A.; Menabrito, M.; Bruna Catalán, I. What fertility specialists should know about the vaginal microbiome: A review. Reprod. Biomed. Online 2017, 35, 103–112. [Google Scholar] [CrossRef] [Green Version]
  58. Rayanakorn, A.; Katip, W.; Lee, L.H.; Oberdorfer, P. Endophthalmitis with bilateral deafness from disseminated Streptococcus suis infection. BMJ Case Rep. 2019, 12, e228501. [Google Scholar] [CrossRef]
  59. Bracewell-Milnes, T.; Saso, S.; Nikolaou, D.; Norman-Taylor, J.; Johnson, M.; Thum, M.-Y. Investigating the effect of an abnormal cervico-vaginal and endometrial microbiome on assisted reproductive technologies: A systematic review. Am. J. Reprod. Immunol. 2018, 80, e13037. [Google Scholar] [CrossRef]
  60. Wilson, J.D.; Lee, R.A.; Balen, A.H.; Rutherford, A.J. Bacterial vaginal flora in relation to changing oestrogen levels. Int. J. STD AIDS 2007, 18, 308–311. [Google Scholar] [CrossRef]
  61. Dominianni, C.; Sinha, R.; Goedert, J.J.; Pei, Z.; Yang, L.; Hayes, R.B.; Ahn, J. Sex, body mass index, and dietary fiber intake influence the human gut microbiome. PLoS ONE 2015, 10, e0124599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. 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] [PubMed] [Green Version]
  63. Claesson, M.J.; Jeffery, I.B.; Conde, S.; Power, S.E.; O’Connor, E.M.; Cusack, S.; Harris, H.M.B.; Coakley, M.; Lakshminarayanan, B.; O’Sullivan, O.; et al. Gut microbiota composition correlates with diet and health in the elderly. Nature 2012, 488, 178–184. [Google Scholar] [CrossRef] [PubMed]
  64. Javurek, A.B.; Spollen, W.G.; Johnson, S.A.; Bivens, N.J.; Bromert, K.H.; Givan, S.A.; Rosenfeld, C.S. Consumption of a high-fat diet alters the seminal fluid and gut microbiomes in male mice. Reprod. Fertil. Dev. 2017, 29, 1602–1612. [Google Scholar] [CrossRef]
  65. Lynch, S.V.; Pedersen, O. The Human Intestinal Microbiome in Health and Disease. N. Engl. J. Med. 2016, 375, 2369–2379. [Google Scholar] [CrossRef] [Green Version]
  66. Deschasaux, M.; Bouter, K.E.; Prodan, A.; Levin, E.; Groen, A.K.; Herrema, H.; Tremaroli, V.; Bakker, G.J.; Attaye, I.; Pinto-Sietsma, S.-J.; et al. Depicting the composition of gut microbiota in a population with varied ethnic origins but shared geography. Nat. Med. 2018, 24, 1526–1531. [Google Scholar] [CrossRef]
  67. Huang, B.; Fettweis, J.M.; Brooks, J.P.; Jefferson, K.K.; Buck, G.A. The changing landscape of the vaginal microbiome. Clin. Lab. Med. 2014, 34, 747–761. [Google Scholar] [CrossRef] [Green Version]
  68. Fox, C.; Eichelberger, K. Maternal microbiome and pregnancy outcomes. Fertil. Steril. 2015, 104, 1358–1363. [Google Scholar] [CrossRef] [Green Version]
  69. DiGiulio, D.B.; Callahan, B.J.; McMurdie, P.J.; Costello, E.K.; Lyell, D.J.; Robaczewska, A.; Sun, C.L.; Goltsman, D.S.A.; Wong, R.J.; Shaw, G.; et al. Temporal and spatial variation of the human microbiota during pregnancy. Proc. Natl. Acad. Sci. USA 2015, 112, 11060–11065. [Google Scholar] [CrossRef] [Green Version]
  70. Selma-Royo, M.; Tarrazó, M.; García-Mantrana, I.; Gómez-Gallego, C.; Salminen, S.; Collado, M.C. Shaping Microbiota During the First 1000 Days of Life. Adv. Exp. Med. Biol. 2019, 1125, 3–24. [Google Scholar]
  71. McGroarty, J.A.; Tomeczek, L.; Pond, D.G.; Reid, G.; Bruce, A.W. Hydrogen peroxide production by Lactobacillus species: Correlation with susceptibility to the spermicidal compound nonoxynol-9. J. Infect. Dis. 1992, 165, 1142–1144. [Google Scholar] [CrossRef] [PubMed]
  72. Mändar, R.; Türk, S.; Korrovits, P.; Ausmees, K.; Punab, M. Impact of sexual debut on culturable human seminal microbiota. Andrology 2018, 6, 510–512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Vodstrcil, L.A.; Twin, J.; Garland, S.M.; Fairley, C.K.; Hocking, J.S.; Law, M.G.; Plummer, E.L.; Fethers, K.A.; Chow, E.P.F.; Tabrizi, S.N.; et al. The influence of sexual activity on the vaginal microbiota and Gardnerella vaginalis clade diversity in young women. PLoS One 2017, 12, e0171856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Lim, M.Y.; Song, E.-J.; Kim, S.H.; Lee, J.; Nam, Y.-D. Comparison of DNA extraction methods for human gut microbial community profiling. Syst. Appl. Microbiol. 2018, 41, 151–157. [Google Scholar] [CrossRef]
  75. Hallmaier-Wacker, L.K.; Lueert, S.; Roos, C.; Knauf, S. The impact of storage buffer, DNA extraction method, and polymerase on microbial analysis. Sci. Rep. 2018, 8, 6292. [Google Scholar] [CrossRef]
  76. Macklaim, J.M.; Gloor, G.B. From RNA-seq to Biological Inference: Using Compositional Data Analysis in Meta-Transcriptomics. Methods Mol. Biol. 2018, 1849, 193–213. [Google Scholar]
  77. Gill, C.; Van De Wijgert, J.H.H.M.; Blow, F.; Darby, A.C. Evaluation of lysis methods for the extraction of bacterial DNA for analysis of the vaginal microbiota. PLoS ONE 2016, 11, e0163148. [Google Scholar] [CrossRef] [Green Version]
  78. Bjerre, R.D.; Hugerth, L.W.; Boulund, F.; Seifert, M.; Johansen, J.D.; Engstrand, L. Effects of sampling strategy and DNA extraction on human skin microbiome investigations. Sci. Rep. 2019, 9, 17287. [Google Scholar] [CrossRef] [Green Version]
  79. Fricker, A.M.; Podlesny, D.; Fricke, W.F. What is new and relevant for sequencing-based microbiome research? A mini-review. J. Adv. Res. 2019, 19, 105–112. [Google Scholar] [CrossRef]
  80. Greathouse, K.L.; Sinha, R.; Vogtmann, E. DNA extraction for human microbiome studies: The issue of standardization. Genome Biol. 2019, 20, 212. [Google Scholar] [CrossRef] [Green Version]
  81. Salter, S.J.; Cox, M.J.; Turek, E.M.; Calus, S.T.; Cookson, W.O.; Moffatt, M.F.; Turner, P.; Parkhill, J.; Loman, N.J.; Walker, A.W. Reagent and laboratory contamination can critically impact sequence-based microbiome analyses. BMC Biol. 2014, 12, 87. [Google Scholar] [CrossRef] [Green Version]
  82. Glassing, A.; Dowd, S.E.; Galandiuk, S.; Davis, B.; Chiodini, R.J. Inherent bacterial DNA contamination of extraction and sequencing reagents may affect interpretation of microbiota in low bacterial biomass samples. Gut Pathog. 2016, 8, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Kim, D.; Hofstaedter, C.E.; Zhao, C.; Mattei, L.; Tanes, C.; Clarke, E.; Lauder, A.; Sherrill-Mix, S.; Chehoud, C.; Kelsen, J.; et al. Optimizing methods and dodging pitfalls in microbiome research. Microbiome 2017, 5, 52. [Google Scholar] [CrossRef] [PubMed]
  84. Goodrich, J.K.; Di Rienzi, S.C.; Poole, A.C.; Koren, O.; Walters, W.A.; Caporaso, J.G.; Knight, R.; Ley, R.E. Conducting a Microbiome Study. Cell 2014, 158, 250–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Van Der Horst, J.; Buijs, M.J.; Laine, M.L.; Wismeijer, D.; Loos, B.G.; Crielaard, W.; Zaura, E. Sterile paper points as a bacterial DNA-contamination source in microbiome profiles of clinical samples. J. Dent. 2013, 41, 1297–1301. [Google Scholar] [CrossRef] [PubMed]
  86. Laurence, M.; Hatzis, C.; Brash, D.E. Common contaminants in next-generation sequencing that hinder discovery of low-abundance microbes. PLoS One 2014, 9, e97876. [Google Scholar] [CrossRef] [PubMed]
  87. Williams, A.R.W.; Brechin, S.; Porter, A.J.L.; Warner, P.; Critchley, H.O.D. Factors affecting adequacy of Pipelle and Tao Brush endometrial sampling. BJOG An Int. J. Obstet. Gynaecol. 2008, 115, 1028–1036. [Google Scholar] [CrossRef]
  88. Du, J.; Li, Y.; Lv, S.; Wang, Q.; Sun, C.; Dong, X.; He, M.; Ulain, Q.; Yuan, Y.; Tuo, X.; et al. Endometrial sampling devices for early diagnosis of endometrial lesions. J. Cancer Res. Clin. Oncol. 2016, 142, 2515–2522. [Google Scholar] [CrossRef] [Green Version]
  89. Aron-Wisnewsky, J.; Clément, K. The gut microbiome, diet, and links to cardiometabolic and chronic disorders. Nat. Rev. Nephrol. 2016, 12, 169–181. [Google Scholar] [CrossRef]
  90. Clooney, A.G.; Fouhy, F.; Sleator, R.D.; O’ Driscoll, A.; Stanton, C.; Cotter, P.D.; Claesson, M.J. Comparing Apples and Oranges?: Next Generation Sequencing and Its Impact on Microbiome Analysis. PLoS ONE 2016, 11, e0148028. [Google Scholar] [CrossRef]
  91. Weyrich, L.S.; Farrer, A.G.; Eisenhofer, R.; Arriola, L.A.; Young, J.; Selway, C.A.; Handsley-Davis, M.; Adler, C.J.; Breen, J.; Cooper, A. Laboratory contamination over time during low-biomass sample analysis. Mol. Ecol. Resour. 2019, 19, 982–996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Eisenhofer, R.; Minich, J.J.; Marotz, C.; Cooper, A.; Knight, R.; Weyrich, L.S. Contamination in Low Microbial Biomass Microbiome Studies: Issues and Recommendations. Trends Microbiol. 2019, 27, 105–117. [Google Scholar] [CrossRef] [PubMed]
  93. D’Amore, R.; Ijaz, U.Z.; Schirmer, M.; Kenny, J.G.; Gregory, R.; Darby, A.C.; Shakya, M.; Podar, M.; Quince, C.; Hall, N. A comprehensive benchmarking study of protocols and sequencing platforms for 16S rRNA community profiling. BMC Genomics 2016, 17, 55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Marques, F.Z.; Mackay, C.R.; Kaye, D.M. Beyond gut feelings: How the gut microbiota regulates blood pressure. Nat. Rev. Cardiol. 2018, 15, 20–32. [Google Scholar] [CrossRef]
  95. Cerdó, T.; García-Valdés, L.; Altmäe, S.; Ruíz, A.; Suárez, A.; Campoy, C. Role of microbiota function during early life on child’s neurodevelopment. Trends Food Sci. Technol. 2016, 57, 273–288. [Google Scholar] [CrossRef]
  96. Power, M.L.; Quaglieri, C.; Schulkin, J. Reproductive Microbiomes: A New Thread in the Microbial Network. Reprod. Sci. 2017, 24, 1482–1492. [Google Scholar] [CrossRef]
  97. Agostinis, C.; Mangogna, A.; Bossi, F.; Ricci, G.; Kishore, U.; Bulla, R. Uterine Immunity and Microbiota: A Shifting Paradigm. Front. Immunol. 2019, 10, 2387. [Google Scholar] [CrossRef] [Green Version]
  98. Nguyen, P.V.; Kafka, J.K.; Ferreira, V.H.; Roth, K.; Kaushic, C. Innate and adaptive immune responses in male and female reproductive tracts in homeostasis and following HIV infection. Cell. Mol. Immunol. 2014, 11, 410–427. [Google Scholar] [CrossRef] [Green Version]
  99. Wira, C.R.; Fahey, J.V.; Rodriguez-Garcia, M.; Shen, Z.; Patel, M. V Regulation of mucosal immunity in the female reproductive tract: The role of sex hormones in immune protection against sexually transmitted pathogens. Am. J. Reprod. Immunol. 2014, 72, 236–258. [Google Scholar] [CrossRef] [Green Version]
  100. Vallvé-Juanico, J.; Houshdaran, S.; Giudice, L.C. The endometrial immune environment of women with endometriosis. Hum. Reprod. Update 2019, 25, 564–591. [Google Scholar] [CrossRef]
  101. Givan, A.L.; White, H.D.; Stern, J.E.; Colby, E.; Gosselin, E.J.; Guyre, P.M.; Wira, C.R. Flow cytometric analysis of leukocytes in the human female reproductive tract: Comparison of fallopian tube, uterus, cervix, and vagina. Am. J. Reprod. Immunol. 1997, 38, 350–359. [Google Scholar] [CrossRef] [PubMed]
  102. Leoni, C.; Ceci, O.; Manzari, C.; Fosso, B.; Volpicella, M.; Ferrari, A.; Fiorella, P.; Pesole, G.; Cicinelli, E.; Ceci, L.R. Human Endometrial Microbiota at Term of Normal Pregnancies. Genes 2019, 10, 971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Younge, N.; McCann, J.R.; Ballard, J.; Plunkett, C.; Akhtar, S.; Araújo-Pérez, F.; Murtha, A.; Brandon, D.; Seed, P.C. Fetal exposure to the maternal microbiota in humans and mice. JCI insight 2019, 4, e127806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Tao, X.; Franasiak, J.M.; Zhan, Y.; Scott, R.T.; Rajchel, J.; Bedard, J.; Newby, R.; Scott, R.T.; Treff, N.R.; Chu, T. Characterizing the endometrial microbiome by analyzing the ultra-low bacteria from embryo transfer catheter tips in IVF cycles: Next generation sequencing (NGS) analysis of the 16S ribosomal gene. Hum. Microbiome J. 2017, 3, 15–21. [Google Scholar] [CrossRef]
  105. Franasiak, J.M.; Scott, R.T. Introduction: Microbiome in human reproduction. Fertil. Steril. 2015, 104, 1341–1343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Vargas, E.; Aghajanova, L.; Gemzell-Danielsson, K.; Altmäe, S.; Esteban, F.J. Cross-disorder analysis of endometriosis and its comorbid diseases reveals shared genes and molecular pathways and proposes putative biomarkers of endometriosis. Reprod. Biomed. Online 2020, 40, 305–318. [Google Scholar] [CrossRef] [PubMed]
  107. Khan, K.N.; Kitajima, M.; Hiraki, K.; Yamaguchi, N.; Katamine, S.; Matsuyama, T.; Nakashima, M.; Fujishita, A.; Ishimaru, T.; Masuzaki, H. Escherichia coli contamination of menstrual blood and effect of bacterial endotoxin on endometriosis. Fertil. Steril. 2010, 94, 2860–2863.e1-3. [Google Scholar] [CrossRef] [Green Version]
  108. Khan, K.N.; Fujishita, A.; Hiraki, K.; Kitajima, M.; Nakashima, M.; Fushiki, S.; Kitawaki, J. Bacterial contamination hypothesis: A new concept in endometriosis. Reprod. Med. Biol. 2018, 17, 125–133. [Google Scholar] [CrossRef] [Green Version]
  109. Leonardi, M.; Hicks, C.; El-Assaad, F.; El-Omar, E.; Condous, G. Endometriosis and the microbiome: A systematic review. BJOG 2020, 127, 239–249. [Google Scholar] [CrossRef]
  110. Takebayashi, A.; Kimura, F.; Kishi, Y.; Ishida, M.; Takahashi, A.; Yamanaka, A.; Takahashi, K.; Suginami, H.; Murakami, T. The association between endometriosis and chronic endometritis. PLoS ONE 2014, 9, e88354. [Google Scholar] [CrossRef] [Green Version]
  111. Cicinelli, E.; Matteo, M.; Trojano, G.; Mitola, P.C.; Tinelli, R.; Vitagliano, A.; Crupano, F.M.; Lepera, A.; Miragliotta, G.; Resta, L. Chronic endometritis in patients with unexplained infertility: Prevalence and effects of antibiotic treatment on spontaneous conception. Am. J. Reprod. Immunol. 2018, 79. [Google Scholar] [CrossRef] [PubMed]
  112. Cicinelli, E.; De Ziegler, D.; Nicoletti, R.; Colafiglio, G.; Saliani, N.; Resta, L.; Rizzi, D.; De Vito, D. Chronic endometritis: Correlation among hysteroscopic, histologic, and bacteriologic findings in a prospective trial with 2190 consecutive office hysteroscopies. Fertil. Steril. 2008, 89, 677–684. [Google Scholar] [CrossRef] [PubMed]
  113. Al-Jefout, M.; Black, K.; Schulke, L.; Berbic, M.; Luscombe, G.; Tokushige, N.; Manconi, F.; Markham, R.; Fraser, I.S. Novel finding of high density of activated mast cells in endometrial polyps. Fertil. Steril. 2009, 92, 1104–1106. [Google Scholar] [CrossRef] [PubMed]
  114. El-Hamarneh, T.; Hey-Cunningham, A.J.; Berbic, M.; Al-Jefout, M.; Fraser, I.S.; Black, K. Cellular immune environment in endometrial polyps. Fertil. Steril. 2013, 100, 1364–1372. [Google Scholar] [CrossRef]
  115. Mayrand, M.-H.; Duarte-Franco, E.; Rodrigues, I.; Walter, S.D.; Hanley, J.; Ferenczy, A.; Ratnam, S.; Coutlée, F.; Franco, E.L.; Canadian Cervical Cancer Screening Trial Study Group. Human papillomavirus DNA versus Papanicolaou screening tests for cervical cancer. N. Engl. J. Med. 2007, 357, 1579–1588. [Google Scholar] [CrossRef] [Green Version]
  116. Mira-Pascual, L.; Cabrera-Rubio, R.; Ocon, S.; Costales, P.; Parra, A.; Suarez, A.; Moris, F.; Rodrigo, L.; Mira, A.; Collado, M.C. Microbial mucosal colonic shifts associated with the development of colorectal cancer reveal the presence of different bacterial and archaeal biomarkers. J. Gastroenterol. 2015, 50, 167–179. [Google Scholar] [CrossRef]
  117. Audirac-Chalifour, A.; Torres-Poveda, K.; Bahena-Román, M.; Téllez-Sosa, J.; Martínez-Barnetche, J.; Cortina-Ceballos, B.; López-Estrada, G.; Delgado-Romero, K.; Burguete-García, A.I.; Cantú, D.; et al. Cervical Microbiome and Cytokine Profile at Various Stages of Cervical Cancer: A Pilot Study. PLoS ONE 2016, 11, e0153274. [Google Scholar] [CrossRef]
  118. Wroblewski, L.E.; Peek, R.M.; Wilson, K.T. Helicobacter pylori and gastric cancer: Factors that modulate disease risk. Clin. Microbiol. Rev. 2010, 23, 713–739. [Google Scholar] [CrossRef] [Green Version]
  119. Shahanavaj, K.; Gil-Bazo, I.; Castiglia, M.; Bronte, G.; Passiglia, F.; Carreca, A.P.; del Pozo, J.L.; Russo, A.; Peeters, M.; Rolfo, C. Cancer and the microbiome: Potential applications as new tumor biomarker. Expert Rev. Anticancer Ther. 2015, 15, 317–330. [Google Scholar] [CrossRef] [Green Version]
  120. Igenomix Foundation. EMMA Endometrial Microbiome Metagenomic Analysis: A screening test to evaluate the endometrium at the microbiological level. Available online: https://www.igenomix.com/genetic-solutions/emma-clinics/ (accessed on 23 February 2020).
  121. Varinos Inc. Innovate Reproductive Health by Genomic Testing. Available online: https://www.varinos.com/english (accessed on 23 February 2020).
  122. Haahr, T.; Jensen, J.S.; Humaidan, P. Research and business – the yin and yang in modern medicine. Reprod. Biomed. Online 2020. [Google Scholar] [CrossRef]
  123. Pereira, N.; Hutchinson, A.P.; Lekovich, J.P.; Hobeika, E.; Elias, R.T. Antibiotic Prophylaxis for Gynecologic Procedures prior to and during the Utilization of Assisted Reproductive Technologies: A Systematic Review. J. Pathog. 2016, 2016, 4698314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Kitaya, K.; Takeuchi, T.; Mizuta, S.; Matsubayashi, H.; Ishikawa, T. Endometritis: New time, new concepts. Fertil. Steril. 2018, 110, 344–350. [Google Scholar] [CrossRef] [PubMed]
  125. Cicinelli, E.; Matteo, M.; Tinelli, R.; Pinto, V.; Marinaccio, M.; Indraccolo, U.; De Ziegler, D.; Resta, L. Chronic endometritis due to common bacteria is prevalent in women with recurrent miscarriage as confirmed by improved pregnancy outcome after antibiotic treatment. Reprod. Sci. 2014, 21, 640–647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. McQueen, D.B.; Bernardi, L.A.; Stephenson, M.D. Chronic endometritis in women with recurrent early pregnancy loss and/or fetal demise. Fertil. Steril. 2014, 101, 1026–1030. [Google Scholar] [CrossRef] [PubMed]
  127. Kitaya, K.; Matsubayashi, H.; Takaya, Y.; Nishiyama, R.; Yamaguchi, K.; Takeuchi, T.; Ishikawa, T. Live birth rate following oral antibiotic treatment for chronic endometritis in infertile women with repeated implantation failure. Am. J. Reprod. Immunol. 2017, 78, e12719. [Google Scholar] [CrossRef] [PubMed]
  128. Zhang, Y.; Xu, H.; Liu, Y.; Zheng, S.; Zhao, W.; Wu, D.; Lei, L.; Chen, G. Confirmation of chronic endometritis in repeated implantation failure and success outcome in IVF-ET after intrauterine delivery of the combined administration of antibiotic and dexamethasone. Am. J. Reprod. Immunol. 2019, 82, e13177. [Google Scholar] [CrossRef]
  129. Sfakianoudis, K.; Simopoulou, M.; Nikas, Y.; Rapani, A.; Nitsos, N.; Pierouli, K.; Pappas, A.; Pantou, A.; Markomichali, C.; Koutsilieris, M.; et al. Efficient treatment of chronic endometritis through a novel approach of intrauterine antibiotic infusion: A case series. BMC Womens. Health 2018, 18, 197. [Google Scholar] [CrossRef] [Green Version]
  130. Weinreb, E.B.; Cholst, I.N.; Ledger, W.J.; Danis, R.B.; Rosenwaks, Z. Should all oocyte donors receive prophylactic antibiotics for retrieval? Fertil. Steril. 2010, 94, 2935–2937. [Google Scholar] [CrossRef]
  131. Chadchan, S.B.; Cheng, M.; Parnell, L.A.; Yin, Y.; Schriefer, A.; Mysorekar, I.U.; Kommagani, R. Antibiotic therapy with metronidazole reduces endometriosis disease progression in mice: A potential role for gut microbiota. Hum. Reprod. 2019, 34, 1106–1116. [Google Scholar] [CrossRef] [Green Version]
  132. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [Green Version]
  133. Khalesi, S.; Bellissimo, N.; Vandelanotte, C.; Williams, S.; Stanley, D.; Irwin, C. A review of probiotic supplementation in healthy adults: Helpful or hype? Eur. J. Clin. Nutr. 2019, 73, 24–37. [Google Scholar] [CrossRef] [PubMed]
  134. Trush, E.A.; Poluektova, E.A.; Beniashvilli, A.G.; Shifrin, O.S.; Poluektov, Y.M.; Ivashkin, V.T. The Evolution of Human Probiotics: Challenges and Prospects. Probiotics Antimicrob. Proteins 2020. [Google Scholar] [CrossRef] [PubMed]
  135. Pelzer, E.; Gomez-Arango, L.F.; Barrett, H.L.; Nitert, M.D. Review: Maternal health and the placental microbiome. Placenta 2017, 54, 30–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Kashyap, P.C.; Chia, N.; Nelson, H.; Segal, E.; Elinav, E. Microbiome at the Frontier of Personalized Medicine. Mayo Clin. Proc. 2017, 92, 1855–1864. [Google Scholar] [CrossRef] [Green Version]
  137. Maretti, C.; Cavallini, G. The association of a probiotic with a prebiotic (Flortec, Bracco) to improve the quality/quantity of spermatozoa in infertile patients with idiopathic oligoasthenoteratospermia: A pilot study. Andrology 2017, 5, 439–444. [Google Scholar] [CrossRef] [Green Version]
  138. Valcarce, D.G.; Genovés, S.; Riesco, M.F.; Martorell, P.; Herráez, M.P.; Ramón, D.; Robles, V. Probiotic administration improves sperm quality in asthenozoospermic human donors. Benef. Microbes 2017, 8, 193–206. [Google Scholar] [CrossRef]
  139. López-Moreno, A.; Aguilera, M. Probiotics Dietary Supplementation for Modulating Endocrine and Fertility Microbiota Dysbiosis. Nutrients 2020, 12, 757. [Google Scholar] [CrossRef] [Green Version]
  140. Chenoll, E.; Moreno, I.; Sánchez, M.; Garcia-Grau, I.; Silva, Á.; González-Monfort, M.; Genovés, S.; Vilella, F.; Seco-Durban, C.; Simón, C.; et al. Selection of New Probiotics for Endometrial Health. Front. Cell. Infect. Microbiol. 2019, 9, 114. [Google Scholar] [CrossRef]
  141. Khodaverdi, S.; Mohammadbeigi, R.; Khaledi, M.; Mesdaghinia, L.; Sharifzadeh, F.; Nasiripour, S.; Gorginzadeh, M. Beneficial effects of oral lactobacillus on pain severity in women suffering from endometriosis: A pilot placebo-controlled randomized clinical trial. Int. J. Fertil. Steril. 2019, 13, 178–183. [Google Scholar]
  142. Itoh, H.; Uchida, M.; Sashihara, T.; Ji, Z.-S.; Li, J.; Tang, Q.; Ni, S.; Song, L.; Kaminogawa, S. Lactobacillus gasseri OLL2809 is effective especially on the menstrual pain and dysmenorrhea in endometriosis patients: Randomized, double-blind, placebo-controlled study. Cytotechnology 2011, 63, 153–161. [Google Scholar] [CrossRef] [Green Version]
  143. Uchida, M.; Kobayashi, O. Effects of Lactobacillus gasseri OLL2809 on the induced endometriosis in rats. Biosci. Biotechnol. Biochem. 2013, 77, 1879–1881. [Google Scholar] [CrossRef] [PubMed]
  144. Somigliana, E.; Viganò, P.; Rossi, G.; Carinelli, S.; Vignali, M.; Panina-Bordignon, P. Endometrial ability to implant in ectopic sites can be prevented by interleukin-12 in a murine model of endometriosis. Hum. Reprod. 1999, 14, 2944–2950. [Google Scholar] [CrossRef] [PubMed]
  145. Itoh, H.; Sashihara, T.; Hosono, A.; Kaminogawa, S.; Uchida, M. Lactobacillus gasseri OLL2809 inhibits development of ectopic endometrial cell in peritoneal cavity via activation of NK cells in a murine endometriosis model. Cytotechnology 2011, 63, 205–210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Dizzell, S.; Nazli, A.; Reid, G.; Kaushic, C. Protective Effect of Probiotic Bacteria and Estrogen in Preventing HIV-1-Mediated Impairment of Epithelial Barrier Integrity in Female Genital Tract. Cells 2019, 8, 1120. [Google Scholar] [CrossRef] [Green Version]
  147. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [Green Version]
  148. Otsuki, K.; Imai, N. Effects of lactoferrin in 6 patients with refractory bacterial vaginosis. Biochem. Cell Biol. 2017, 95, 31–33. [Google Scholar] [CrossRef]
  149. Stallmach, A.; Steube, A.; Grunert, P.; Hartmann, M.; Biehl, L.M.; Vehreschild, M.J.G.T. Fecal Microbiota Transfer. Dtsch. Arztebl. Int. 2020, 117, 31–38. [Google Scholar] [CrossRef] [Green Version]
  150. Donnet-Hughes, A.; Perez, P.F.; Doré, J.; Leclerc, M.; Levenez, F.; Benyacoub, J.; Serrant, P.; Segura-Roggero, I.; Schiffrin, E.J. Potential role of the intestinal microbiota of the mother in neonatal immune education. Proc. Nutr. Soc. 2010, 69, 407–415. [Google Scholar] [CrossRef] [Green Version]
  151. Quaranta, G.; Sanguinetti, M.; Masucci, L. Fecal Microbiota Transplantation: A Potential Tool for Treatment of Human Female Reproductive Tract Diseases. Front. Immunol. 2019, 10, 2653. [Google Scholar] [CrossRef]
  152. Ata, B.; Yildiz, S.; Turkgeldi, E.; Brocal, V.P.; Dinleyici, E.C.; Moya, A.; Urman, B. The Endobiota Study: Comparison of Vaginal, Cervical and Gut Microbiota Between Women with Stage 3/4 Endometriosis and Healthy Controls. Sci. Rep. 2019, 9, 2204. [Google Scholar] [CrossRef] [Green Version]
  153. Lindheim, L.; Bashir, M.; Münzker, J.; Trummer, C.; Zachhuber, V.; Leber, B.; Horvath, A.; Pieber, T.R.; Gorkiewicz, G.; Stadlbauer, V.; et al. Alterations in Gut Microbiome Composition and Barrier Function Are Associated with Reproductive and Metabolic Defects in Women with Polycystic Ovary Syndrome (PCOS): A Pilot Study. PLoS ONE 2017, 12, e0168390. [Google Scholar] [CrossRef]
  154. Liu, R.; Zhang, C.; Shi, Y.; Zhang, F.; Li, L.; Wang, X.; Ling, Y.; Fu, H.; Dong, W.; Shen, J.; et al. Dysbiosis of Gut Microbiota Associated with Clinical Parameters in Polycystic Ovary Syndrome. Front. Microbiol. 2017, 8, 324. [Google Scholar] [CrossRef]
  155. Insenser, M.; Murri, M.; Del Campo, R.; Martínez-García, M.Á.; Fernández-Durán, E.; Escobar-Morreale, H.F. Gut Microbiota and the Polycystic Ovary Syndrome: Influence of Sex, Sex Hormones, and Obesity. J. Clin. Endocrinol. Metab. 2018, 103, 2552–2562. [Google Scholar] [CrossRef]
  156. Torres, P.J.; Siakowska, M.; Banaszewska, B.; Pawelczyk, L.; Duleba, A.J.; Kelley, S.T.; Thackray, V.G. Gut Microbial Diversity in Women With Polycystic Ovary Syndrome Correlates With Hyperandrogenism. J. Clin. Endocrinol. Metab. 2018, 103, 1502–1511. [Google Scholar] [CrossRef]
  157. Zhou, L.; Ni, Z.; Cheng, W.; Yu, J.; Sun, S.; Zhai, D.; Yu, C.; Cai, Z. Characteristic gut microbiota and predicted metabolic functions in women with PCOS. Endocr. Connect. 2020, 9, 63–73. [Google Scholar] [CrossRef]
  158. Jobira, B.; Frank, D.N.; Pyle, L.; Silveira, L.J.; Kelsey, M.M.; Garcia-Reyes, Y.; Robertson, C.E.; Ir, D.; Nadeau, K.J.; Cree-Green, M. Obese adolescents with PCOS have altered biodiversity and relative abundance in gastrointestinal microbiota. J. Clin. Endocrinol. Metab. 2020. [Google Scholar] [CrossRef]
  159. DeLong, K.; Zulfiqar, F.; Hoffmann, D.E.; Tarzian, A.J.; Ensign, L.M. Vaginal Microbiota Transplantation: The Next Frontier. J. Law. Med. Ethics 2019, 47, 555–567. [Google Scholar] [CrossRef]
  160. Lev-Sagie, A.; Goldman-Wohl, D.; Cohen, Y.; Dori-Bachash, M.; Leshem, A.; Mor, U.; Strahilevitz, J.; Moses, A.E.; Shapiro, H.; Yagel, S.; et al. Vaginal microbiome transplantation in women with intractable bacterial vaginosis. Nat. Med. 2019, 25, 1500–1504. [Google Scholar] [CrossRef]
  161. Leyva-Gómez, G.; Prado-Audelo, D.; Ortega-Peña, S.; Mendoza-Muñoz, N.; Urbán-Morlán, Z.; González-Torres, M.; Carmen, G.D.; Figueroa-González, G.; Reyes-Hernández, O.D.; Cortés, H.; et al. Modifications in Vaginal Microbiota and Their Influence on Drug Release: Challenges and Opportunities. Pharmaceutics 2019, 11, 217. [Google Scholar]
  162. Feo, L.G. The pH of the human uterine cavity in situ. Am. J. Obstet. Gynecol. 1955, 70, 60–64. [Google Scholar] [CrossRef]
  163. Manallack, D.T. The pK(a) Distribution of Drugs: Application to Drug Discovery. Perspect. Medicin. Chem. 2007, 1, 25–38. [Google Scholar] [CrossRef]
  164. Ng, K.Y.B.; Mingels, R.; Morgan, H.; Macklon, N.; Cheong, Y. In vivo oxygen, temperature and pH dynamics in the female reproductive tract and their importance in human conception: A systematic review. Hum. Reprod. Update 2018, 24, 15–34. [Google Scholar] [CrossRef]
  165. Selway, C.A.; Eisenhofer, R.; Weyrich, L.S. Microbiome applications for pathology: Challenges of low microbial biomass samples during diagnostic testing. J. Pathol. Clin. Res. 2020. [Google Scholar] [CrossRef]
  166. Tita, A.T.N.; Cliver, S.P.; Goepfert, A.R.; Conner, M.; Goldenberg, R.L.; Hauth, J.C.; Andrews, W.W. Impact of interconception antibiotics on the endometrial microbial flora. Am. J. Obstet. Gynecol. 2007, 196, 226.e1–226.e16. [Google Scholar] [CrossRef] [Green Version]
  167. Kristensen, N.B.; Bryrup, T.; Allin, K.H.; Nielsen, T.; Hansen, T.H.; Pedersen, O. Alterations in fecal microbiota composition by probiotic supplementation in healthy adults: A systematic review of randomized controlled trials. Genome Med. 2016, 8, 52. [Google Scholar] [CrossRef] [Green Version]
  168. Łaniewski, P.; Ilhan, Z.E.; Herbst-Kralovetz, M.M. The microbiome and gynaecological cancer development, prevention and therapy. Nat. Rev. Urol. 2020, 17, 232–250. [Google Scholar] [CrossRef]
Figure 1. Dynamics and factors that could influence the endometrial microbiome and its analysis, including technical factors in the experimental design and individual/participant/group factors that could fluctuate the microbial composition in the uterus. Age: microbiome changes along lifespan [48], and the diversity and composition decline in the elderly [49,50]. Medical history: genital and extragenital diseases [19,22,23,51], including history of sexually transmitted infections can influence microbiome analysis. Ethnicity and different geographic regions: the ethnic origin of individuals seems to be an important factor to consider in microbiome research [52,53,54,55], as African American and Hispanic women show a trend to be more colonised with a non-Lactobacillus species in the upper genital tract than Caucasian women [34]. Genetic variation: host genetics can affect microbiome composition and maintenance [56,57]. Cultural habits: these habits showed leading to microbial variations [58]. Menstrual cycle and hormones: fluctuations in circulating oestrogen and progesterone levels might influence endometrial microbiome [19,57,59,60]. BMI: body mass index, especially obesity has been associated with altered intestinal microbiome composition [61,62]. Diet: it was shown that the gut microbiome not just metabolises ingested food but is itself shaped by the mode and type of food consumption [63,64]. Physical activity: exercising has shown to impact microbial composition [65,66]. Childbirth and pregnancy [67,68,69,70]. Sexual habits: the concept of seminovaginal microbiome, i.e., the partners share their microbial communities, has gained support [29]. Spermicidal agents could disrupt endometrial microbiota [71]; also sexual debut and activity can influence genital microbiome [72,73]. Study protocol: different protocols (collection technique, sample storage, extraction method, choice of primers) can lead to different results [74,75,76,77,78,79,80]. High risk of contamination: laboratory and reagent contamination can critically impact sequence-based microbiome analyses [81,82,83,84]; negative controls consisting of blanks are recommended [81,82,85,86]. Sampling method: an important point to consider consists in avoiding the surface of the vagina and the walls of cervical canal when taking the endometrial sample [87,88]. Sequencing: different sequencing platforms and sequencing filtering and processing can result in different outcomes [46,89,90]. Sample processing: amplicon sequencing has shown to cause great variance [75,90]. Low-biomass: high risk of misinterpretation of the results [26,91,92]. Analyses and interpretation: statistical analyses and interpretation can induce bias [84,90,93].
Figure 1. Dynamics and factors that could influence the endometrial microbiome and its analysis, including technical factors in the experimental design and individual/participant/group factors that could fluctuate the microbial composition in the uterus. Age: microbiome changes along lifespan [48], and the diversity and composition decline in the elderly [49,50]. Medical history: genital and extragenital diseases [19,22,23,51], including history of sexually transmitted infections can influence microbiome analysis. Ethnicity and different geographic regions: the ethnic origin of individuals seems to be an important factor to consider in microbiome research [52,53,54,55], as African American and Hispanic women show a trend to be more colonised with a non-Lactobacillus species in the upper genital tract than Caucasian women [34]. Genetic variation: host genetics can affect microbiome composition and maintenance [56,57]. Cultural habits: these habits showed leading to microbial variations [58]. Menstrual cycle and hormones: fluctuations in circulating oestrogen and progesterone levels might influence endometrial microbiome [19,57,59,60]. BMI: body mass index, especially obesity has been associated with altered intestinal microbiome composition [61,62]. Diet: it was shown that the gut microbiome not just metabolises ingested food but is itself shaped by the mode and type of food consumption [63,64]. Physical activity: exercising has shown to impact microbial composition [65,66]. Childbirth and pregnancy [67,68,69,70]. Sexual habits: the concept of seminovaginal microbiome, i.e., the partners share their microbial communities, has gained support [29]. Spermicidal agents could disrupt endometrial microbiota [71]; also sexual debut and activity can influence genital microbiome [72,73]. Study protocol: different protocols (collection technique, sample storage, extraction method, choice of primers) can lead to different results [74,75,76,77,78,79,80]. High risk of contamination: laboratory and reagent contamination can critically impact sequence-based microbiome analyses [81,82,83,84]; negative controls consisting of blanks are recommended [81,82,85,86]. Sampling method: an important point to consider consists in avoiding the surface of the vagina and the walls of cervical canal when taking the endometrial sample [87,88]. Sequencing: different sequencing platforms and sequencing filtering and processing can result in different outcomes [46,89,90]. Sample processing: amplicon sequencing has shown to cause great variance [75,90]. Low-biomass: high risk of misinterpretation of the results [26,91,92]. Analyses and interpretation: statistical analyses and interpretation can induce bias [84,90,93].
Biomolecules 10 00593 g001
Figure 2. Endometrial microbiota interplay in the uterus. Uterine microbes could impact the genomic stability of uterine epithelia through modulation of transcription factors and other genomic and epigenetic alterations; also, microbial-secreted metabolites may support the growth of specific species and suppress the growth of other bacteria; and the consumption of a limited resource can starve the pathogenic invaders. In short, the endometrium is an immunologically suited niche for microbes: the endometrial immune system needs to be well adapted to withstand the continuous threat caused by microbial colonisation of the large endometrial mucosal surface, separated from host tissue by only a single layer of epithelial cells. Thus, tissue invasion of microbes must be limited in order to prevent potentially harmful inflammation or imbalance in the symbiotic relationship. Endometrial microbial homeostasis is probably regulated in three different ways: (1) a single layer of columnar epithelial cells forming a strong barrier through tight junctions anatomically limiting exposure of resident bacteria to the systemic immune system; (2) immune mediators such as infection-controlling molecules (antimicrobial peptides, AMPs) that are present in the endometrial mucosal surface and the endometrial fluid [99] and could restrict direct contact between epithelia and microbes; and (3) a rapid detection (epithelial cells express pattern recognition receptors (PRRs) that recognise and act to pathogens) and killing of bacteria upon a barrier breach by the endometrial lymphocytes that are present throughout all stages of the menstrual cycle [100,101].
Figure 2. Endometrial microbiota interplay in the uterus. Uterine microbes could impact the genomic stability of uterine epithelia through modulation of transcription factors and other genomic and epigenetic alterations; also, microbial-secreted metabolites may support the growth of specific species and suppress the growth of other bacteria; and the consumption of a limited resource can starve the pathogenic invaders. In short, the endometrium is an immunologically suited niche for microbes: the endometrial immune system needs to be well adapted to withstand the continuous threat caused by microbial colonisation of the large endometrial mucosal surface, separated from host tissue by only a single layer of epithelial cells. Thus, tissue invasion of microbes must be limited in order to prevent potentially harmful inflammation or imbalance in the symbiotic relationship. Endometrial microbial homeostasis is probably regulated in three different ways: (1) a single layer of columnar epithelial cells forming a strong barrier through tight junctions anatomically limiting exposure of resident bacteria to the systemic immune system; (2) immune mediators such as infection-controlling molecules (antimicrobial peptides, AMPs) that are present in the endometrial mucosal surface and the endometrial fluid [99] and could restrict direct contact between epithelia and microbes; and (3) a rapid detection (epithelial cells express pattern recognition receptors (PRRs) that recognise and act to pathogens) and killing of bacteria upon a barrier breach by the endometrial lymphocytes that are present throughout all stages of the menstrual cycle [100,101].
Biomolecules 10 00593 g002
Figure 3. Current and future strategies for modifying uterine microbial composition. Intrauterine drug delivery represents an attractive alternative to achieve local and systemic effects due to the high contact surface exposed, the mucoadhesion of the epithelium, and the high absorption of drugs into the bloodstream. Several strategies for modifying endometrial microbial composition are being applied, nevertheless the core microbial composition is not established. The standard protocols for detecting uterine microbes and treatment protocols of dysbiosis are yet to be established.
Figure 3. Current and future strategies for modifying uterine microbial composition. Intrauterine drug delivery represents an attractive alternative to achieve local and systemic effects due to the high contact surface exposed, the mucoadhesion of the epithelium, and the high absorption of drugs into the bloodstream. Several strategies for modifying endometrial microbial composition are being applied, nevertheless the core microbial composition is not established. The standard protocols for detecting uterine microbes and treatment protocols of dysbiosis are yet to be established.
Biomolecules 10 00593 g003
Table 1. Predominant taxa in endometrial microbiome for different gynaecological disorders revealed by next-generation sequencing studies.
Table 1. Predominant taxa in endometrial microbiome for different gynaecological disorders revealed by next-generation sequencing studies.
Gynaecological ConditionPredominant Taxa
HealthyAcinetobacter [102,103], Bacillus [103], Barnesiella [23], Bifidobacterium [11], Blautia [23], Corynebacterium [102], Desulfosporosinus [16], Enterobacter [16], Escherichia [102,103], Fusobacterium [22], Gardnerella [11], Jonquetella [22], Lactobacillus [8,11,16,19], Parabacteroides [23], Prevotella [11,22], Propionibacterium [102], Pseudomonas [16], Ralstonia [16], Shigella [23], Staphylococcus [23,102], Streptococcus [11,102]
InfertilityAtopobium [6,8,9,10], Bacteroides [12], Betaproteobacteria [12], Bifidobacterium [5,8,9,10,11,13,104], Burkholderia [7], Chitinophagaceae [12], Corynebacterium [104], Escherichia/Shigella [10,12], Flavobacterium [4], Gardnerella [5,6,7,8,9,10,11,13], Lactobacillus [4,5,6,7,8,9,10,11,13,104], Megasphaera [9,10], Pelomonas [12], Prevotella [8,9,10,11,13], Pseudoalteromonas [5], Rhodanobacter [5], Sneathia [8,9], Staphylococcus [8,9,10,104], Streptococcus [6,7,8,9,11,104]
EndometriosisAcinetobacter [20], Barnesiella [18], Comamonadaceae [20], Enterobacteriaceae [19], Flavobacterium [18], Gardnerella [17], Lactobacillus [17,18,20], Moraxellaceae [19], Prevotella [17], Pseudomonas [18,20], Sphingobium [20], Staphylococaceae [19], Streptococcaceae [17,19], Vagococcus [20]
Chronic endometritisAlteromonas [16], Anaerococcus [15], Atopobium [15], Bifidobacterium [14,15,16], Dialister [15], Gardnerella [14,15,16], Lactobacillus [14,16], Magasphaera [14], Parvimonas [14], Prevotella [14,15], Propionibacterium [14], Streptococcus [14,16], Veillonella [14]
Endometrial polypsAlteromonas [16], Bifidobacterium [16], Euryarchaeota (Archaea) [16], Gardnerella [16], Lactobacillus [16], Streptococcus [16]
Dysfunctional menstrual bleedingGardnerella [22], Lactobacillus [22], Prevotella [22], Sneathia [22], Veilonella [22]
Endometrial cancerAcinetobacter [25], Anaerostipes [23], Anaerotruncus [23], Arthrospira [23], Atopobium [23], Bacteroides [23], Cloacibacterium [25], Comamonadaceae [25], Dialister [23], Escherichia [25], Peptoniphilus [23], Porphyromonas [23,24], Pseudomonas [25], Ruminococcus [23], Treponema [23]

Share and Cite

MDPI and ACS Style

Molina, N.M.; Sola-Leyva, A.; Saez-Lara, M.J.; Plaza-Diaz, J.; Tubić-Pavlović, A.; Romero, B.; Clavero, A.; Mozas-Moreno, J.; Fontes, J.; Altmäe, S. New Opportunities for Endometrial Health by Modifying Uterine Microbial Composition: Present or Future? Biomolecules 2020, 10, 593. https://doi.org/10.3390/biom10040593

AMA Style

Molina NM, Sola-Leyva A, Saez-Lara MJ, Plaza-Diaz J, Tubić-Pavlović A, Romero B, Clavero A, Mozas-Moreno J, Fontes J, Altmäe S. New Opportunities for Endometrial Health by Modifying Uterine Microbial Composition: Present or Future? Biomolecules. 2020; 10(4):593. https://doi.org/10.3390/biom10040593

Chicago/Turabian Style

Molina, Nerea M., Alberto Sola-Leyva, Maria Jose Saez-Lara, Julio Plaza-Diaz, Aleksandra Tubić-Pavlović, Barbara Romero, Ana Clavero, Juan Mozas-Moreno, Juan Fontes, and Signe Altmäe. 2020. "New Opportunities for Endometrial Health by Modifying Uterine Microbial Composition: Present or Future?" Biomolecules 10, no. 4: 593. https://doi.org/10.3390/biom10040593

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