Next Article in Journal
Efficacy of Green Synthesized Nanoparticles in Photodynamic Therapy: A Therapeutic Approach
Next Article in Special Issue
Potential Significance of Serum Autoantibodies to Endometrial Antigens, α-Enolase and Hormones in Non-Invasive Diagnosis and Pathogenesis of Endometriosis
Previous Article in Journal
Genome-Wide Identification and Expression Analysis of ACTIN Family Genes in the Sweet Potato and Its Two Diploid Relatives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

How Do Microorganisms Influence the Development of Endometriosis? Participation of Genital, Intestinal and Oral Microbiota in Metabolic Regulation and Immunopathogenesis of Endometriosis

Department of Experimental Immunology, Medical University of Lublin, Chodzki Street, 20-093 Lublin, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(13), 10920; https://doi.org/10.3390/ijms241310920
Submission received: 10 May 2023 / Revised: 23 June 2023 / Accepted: 26 June 2023 / Published: 30 June 2023
(This article belongs to the Special Issue The Pathogenesis and Therapy of Endometriosis)

Abstract

:
Microorganisms inhabiting the human body play an extremely key role in its proper functioning, as well as in the development of the immune system, which, by maintaining the immune balance, allows you to enjoy health. Dysbiosis of the intestinal microbiota, or in the oral cavity or reproductive tract, understood as a change in the number and diversity of all microorganisms inhabiting them, may correlate with the development of many diseases, including endometriosis, as researchers have emphasized. Endometriosis is an inflammatory, estrogen-dependent gynecological condition defined by the growth of endometrial cells outside the uterine cavity. Deregulation of immune homeostasis resulting from microbiological disorders may generate chronic inflammation, thus creating an environment conducive to the increased adhesion and angiogenesis involved in the development of endometriosis. In addition, research in recent years has implicated bacterial contamination and immune activation, reduced gastrointestinal function by cytokines, altered estrogen metabolism and signaling, and abnormal progenitor and stem cell homeostasis, in the pathogenesis of endometriosis. The aim of this review was to present the influence of intestinal, oral and genital microbiota dysbiosis in the metabolic regulation and immunopathogenesis of endometriosis.

1. Introduction

Endometriosis is a chronic, inflammatory, estrogen-dependent gynecological condition defined by the growth of endometrial cells outside the uterine cavity [1]. Endometrial cells migrate from their original site, the uterus, to other organs and form endometrial-like tissues in various anatomical locations outside the uterine cavity, especially in the ovaries and peritoneum [1,2]. Although the symptoms of endometriosis are not unique and many of them are similar to the symptoms of other gynecological diseases, it can cause pelvic discomfort and infertility [3]. Literature data estimate that it is diagnosed with a significant delay of 3 to 11 years, which results in disruption of the reproductive cycle in women of reproductive age. The true prevalence of endometriosis cannot be determined due to the lack of effective non-invasive diagnostic techniques; nevertheless, epidemiological data indicate that about 10% of women of childbearing age suffer from endometriosis. Additionally, its incidence increases from 20% to 50% in women experiencing pelvic discomfort or infertility [3,4,5]. Endometriosis is a disease with a complex etiopathogenesis, which involves the participation of genetic and immunological pathways (including inflammation), as well as environmental influences, such as eating habits and nutrients, or antibiotic therapy, which, according to recent research, may be one of the factors modifying the amount of and diversity of microflora. Research indicates that the development of endometriosis involves alterations in multiple biological pathways, mainly the metabolic regulation at the basis of estrogen-dependent disorders [6,7]. The aim of this review was to present the influence of intestinal, oral and genital microbiota dysbiosis in the metabolic regulation and immunopathogenesis of endometriosis.

2. The Importance of Disorders of Immune Homeostasis in the Pathogenesis of Endometriosis

Endometriosis is a chronic inflammatory disease of immune origin characterized by the presence of endometrial-like tissue outside the uterus, commonly found in the pelvic cavity and other abdominal organs. The ectopic tissue exhibits similar characteristics to the endometrium, including the ability to respond to hormonal fluctuations and undergo cyclic shedding and bleeding [8]. Several mechanisms have been proposed for the histologic origins of endometriosis. One theory suggests that fragments of menstrual endometrium pass retrograde through the fallopian tubes and implant on peritoneal surfaces [9]. Another hypothesis is that the peritoneum undergoes metaplasia to form endometriotic lesions within the peritoneal cavity [10]. There are also suggestions that menstrual tissue reaches distant body sites via veins or lymphatic vessels [11], or that circulating blood cells differentiate into endometriotic tissue [12]. However, although various theories have been proposed which posit issues such as genetic predisposition, immune dysfunction and hormonal imbalances as contributing factors, the precise etiology of endometriosis remains unclear. Local inflammation, which plays a major role in endometriosis, causes pain and infertility, as well as affecting the development and progression of the disease [13]. Numerous studies have shown that the peritoneal fluid of patients with this disease contained large amounts of immune cells and macrophages that produce cytokines. What is more, in recent studies, oxidative stress, an imbalance between the production of reactive oxygen species (ROS) and the antioxidant defense system, has emerged as a potential contributor to the pathogenesis of endometriosis. Increased ROS production and reduced antioxidant capacity have been observed in the peritoneal fluid, serum and endometrial tissues of individuals with endometriosis [14,15,16]. Under physiological conditions, cytokines counteract the harmful effects of persistent or excessive inflammatory reactions that can have harmful effects. When the equilibrium between the creation and detoxification of reactive oxidative species (ROS) is interrupted, the relative excess of ROS causes oxidative stress. ROS are reactive oxygen-containing chemical species, such as superoxide (O2), hydroxyl radicals (·OH) and hydrogen peroxide (H2O2). ROS in high concentrations can harm peritoneal mesothelial cells and induce ectopic endometrial implantation [17,18]. It can also alter the shape and function of vascular endothelial cells, such as permeability and adhesion molecule expression, hence increasing the adhesion between inflammatory cells and endothelial cells and leading to prolonged inflammation [19,20]. What is more, ROS can activate NF-κB and protein kinase B (AKT) pathways promoting endometriosis development [20,21]. In pathological conditions, the action of anti-inflammatory mediators may be insufficient or overcompensate for the inflammatory response, leading to inhibition of the immune response, which increases the risk of systemic infection [22]. Elevated concentrations of immune mediators, mainly pro-inflammatory molecules, occur not only locally but also systemically. In damaged endometrial cells, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κβ) is the major transcription factor that is overexpressed. The cytokine-mediated pro-inflammatory environment further enhances this effect. In turn, NF-kβ is involved in the positive regulation of these pro-inflammatory factors [22]. For many years, studies have shown that not only the participation of individual subpopulations of cells of the immune system, but also elevated levels of anti-inflammatory cytokines may be involved in the development of endometriosis.

2.1. The Role of Immune Cells in the Development of Endometriosis

2.1.1. Macrophages

Macrophages can be divided into two groups: M1 macrophages, which have pro-inflammatory effects, and M2 macrophages, which have anti-inflammatory effects and can induce tissue remodeling through profibrotic effects. M2 macrophages also have proangiogenic properties and can induce immunotolerance [23]. An increased number of macrophages was observed in the peritoneal fluid and in the eutopic endometrium of women with endometriosis [24]. However, an increased number of these cells was not observed in peripheral blood in this group of patients [25,26]. Research results suggest that macrophages can directly affect endometrial cells. Co-culture of macrophages with these cells increased cell proliferation, increased invasiveness and led to the activation of the signal transducer and activator of transcription 3 (STAT3) in endometriotic stromal cells [27,28]. The current study demonstrates the ability of macrophages to polarize. Polarity is a spatiotemporally organized field that is time- and tissue-dependent. This phenomenon is associated with both internal, external and environmental tissue signals, including growth factors, cytokines, prostaglandins and pathogen-derived molecules. Polarization is evidenced by changes in phenotype such as its loss, switch or modulation. Depending on the disease state, macrophages show different polarization, which demonstrates their plasticity. In inflammatory diseases, an M1-like polarity is more frequently observed, whereas in chronic bacterial, parasitic or viral diseases, an M2-like polarity predominates [29].

2.1.2. Neutrophils

The number of neutrophils in the peritoneal fluid of endometriosis patients may be increased [30]. When tested in a mouse model, it was observed that reducing the number of neutrophils with the anti-Gr-1 antibody resulted in a reduction in lesion formation. Neutrophils aggravate endometriosis mainly through the expression of pro-inflammatory cytokines such as interleukin 8 (IL-8 or CXCL8), C-X-C motif chemokine ligand 10 (CXCL10) and vascular endothelial growth factor (VEGF), which may promote disease progression [31,32]. One study observed that incubation of neutrophils from disease-free women with plasma or peritoneal fluid from women with endometriosis reduced the rate of apoptosis compared to control women. This study demonstrated the potential presence of apoptotic factors or PF in the plasma of women with endometriosis that are absent in healthy women [33].

2.1.3. Natural Killer Cells

Natural killer (NK) cells are the primary cell population of the immune system and exhibit cytotoxic activity. They are also important for proper tissue homeostasis. NK cell counts appear to be the same in patients with and without endometriosis [24]. The endometrium in healthy women has an immunosuppressive effect on NK cell activity. Most likely, this will facilitate the implantation of the embryo. It has been observed that NK cells show even less cytotoxicity in the peritoneal fluid of endometriosis patients, which may contribute to the transformation of endometrial fragments into lesions [34]. In the natural killer cells in the peritoneal fluid of women with endometriosis, the cytotoxic activating killer-cell immunoglobulin-like receptor (KIR) is inhibited as a result of upregulation of IR2DL1, which may contribute to lower cytotoxicity of these cells [35]. Another mechanism responsible for the reduced cytotoxicity of NK cells may be the expression of TGFB1, which inhibits the expression of NKG2D ligands [36]. The results indicate that IL-6 may be responsible for the immunosuppressive effect against NK cell cytotoxicity to autologous endometrial fragments [37].

2.1.4. T Cells

CD4+ T cells can be subdivided into Th1 cells, which secrete interferon gamma (IFN-γ), and Th2 cells, which secrete IL-4 [38]. In addition, Th17 lymphocytes and regulatory T lymphocytes (Treg) are distinguished [39,40]. Women with endometriosis have been observed to have more Th2 lymphocytes as a result of strong intracellular expression of IL-4 and lack of IL-2 in lymphocytes isolated from ectopic lesions [41]. Th2 cells, through the secretion of IL-4 and IL-13, which mediate the differentiation of resident fibroblasts and the recruitment of fibrocytes to myofibroblasts, may contribute to pelvic fibrosis in women with endometriosis [42]. Studies show that in the case of ectopic endometriosis, a greater number of Th17 cells are present in the peritoneal fluid and peripheral blood compared to women without the disease [43]. Th17 cells express CCR6 (C-C Motif Chemokine Receptor 6), a receptor for the CCL20 (C-C Motif Chemokine Ligand 20) ligand that is produced by endometriosis stromal cells and has a chemotactic effect on Th17 cells [44]. Treg lymphocytes are also more numerous in ectopic lesions. The results of the study showed that in the peritoneal fluid and peripheral blood of patients with endometriosis, their concentrations were higher than in the control group [45,46]. A positive correlation was also found between the number of these cells and disease progression [47].

2.1.5. Tight Junctions

Tight junctions are responsible for regulating and restricting transport between cells, thereby forming a selective barrier between them. They also act as fences by maintaining a tight organization of the plasma membrane of epithelial cells in the apical and basolateral compartments [48]. Tight junctions are mainly composed of claudins, which are found in the apicolateral membranes of epithelial and endothelial cells [49]. Based on function, claudins can be divided into channel-forming (claudin-2, -4, -10, -15, -17 and -21) and barrier-forming (claudin-1, -3, -5, -11, -14 and -18) [50]. Several claudins such as claudin-1, -5, -7 and -11 have been identified in the human endometrium [51,52]. Studies have shown lower expression of claudin-3 and claudin-4 in ectopic endometriosis tissue compared to eutopic endometrium in women with endometriosis at messenger mRNA and protein levels. These changes may play a role in the pathogenesis of this disease [53]. On the other hand, a study by Hoerscher et al. showed that claudin-2 and claudin-3 show similarity in localization in the ectopic compared to the eutopic endometrium [54]. In addition, Gaetje et al. showed that there is reduced expression of claudins in peritoneal endometriosis lesions in comparison with eutopic endometrium. Downregulation of claudin-3 and claudin-7 in endometriotic lesions and reduction of claudin-5 mRNA were observed [51].

2.1.6. Damage-Associated Molecular Patterns (DAMPs) and Pathogen-Associated Molecular Patterns (PAMPs)

Endometriosis is linked to ectopic localized inflammation and an immunocompromised microenvironment. The innate immune system has a variety of pattern recognition receptors (PRRs) that can detect danger-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs) in both intracellular and extracellular environments. Danger-associated molecular patterns reflect host tissue damage. These include heat shock protein 70 (HSP70), adenosine-5′-triphosphate (ATP), high-mobility group box protein 1 (HMGB1) and IL-33. They are released after cell death during inflammation. It has been shown that some DAMPS such as DNA, ATP, HMGB1 and HSP70 are involved in endometriosis [55]. HMGB1 is involved in necrosis and severe cellular stress [56]. It is expressed in epithelial and stroma cells in both endometriosis patients and disease-free patients [57]. Kajihara et al. showed that DAMPs promote the development and progression of endometriosis in association with TLR activation and NF-κB signaling. Their work also suggested that histones may be a new class of DAMP molecules and activate innate immunity during sterile inflammation in endometriosis [55]. The findings suggest that inhibitors of DAMP molecules may represent a potential therapeutic target for preventing or reducing the progression of endometriosis. This is possibly due to the function of these molecules as a link between infection-induced initial cellular damage and activation of innate immune cells as a result of oxidative stress and sterile inflammation [58].
PAMPs have the ability to differentiate between harmful microorganisms and the host’s own cells [59,60,61,62,63,64]. PAMPs are structures found in various microorganisms [65], such as bacterial lipopolysaccharide, lipoprotein, peptidase, flagellin, unmethylated CpG dinucleotide motifs (CpG-DNA), viral double-stranded RNA, fungal cell wall, and others. Upon recognizing PAMPs, PRRs initiate a reaction between the receptor and the ligand, transmitting signals of microbial infection to the cells. This process stimulates the immune response of the host, leading to the elimination of the pathogenic microorganisms [66].

2.1.7. Toll-Like Receptors (TLRs)

Based on the homology of their protein domains, PRRs can be categorized into five groups. These groups include toll-like receptors (TLRs), c-type lectin receptors (CLRs), nod-like receptors (NLRs), retinoic acid-inducible gene I-like receptors (RLRs) and absent in melanoma 2 (AIM2)-like receptors (ALRs). These receptor groups can be further classified into two main classes: membrane-bound receptors and intracellular receptors [67]. The first class primarily consists of TLRs, which are typically found on the cell membrane but may occasionally be located within intracellular compartments (e.g., TLR3). TLR activation triggers a series of events that leads to the transcription of many downstream genes involved in inflammatory response and antimicrobial defense [68]. TLR4, TLR3 and TLR2 are members of the TLR family, mostly found in samples studied for endometriosis. TLR4 is expressed by various immune cells and has been found to be higher in endometrial stromal cells of endometriosis patients. Animal experiments have demonstrated that activation of the LPS/TLR4 pathway, induced by injecting lipopolysaccharide (LPS) into a mouse model of endometriosis, leads to the release of inflammatory factors and promotes the proliferation and invasion of ectopic endometrial stromal cells [69]. TLR3 is involved in innate immunity and its activation induces the production of inflammatory cytokines. TLR3 expression is increased in both ectopic and eutopic endometrium of endometriosis patients, contributing to the inflammatory state and enhanced cell viability [70]. TLR2 plays a role in bacterial infections and the innate immune system. Its expression has been assessed in endometriosis patients, but further research is needed to understand its specific involvement [71]. Overall, more studies are needed to explore the presence and potential use of TLRs as biomarkers for the early detection of endometriosis.

2.1.8. Lipopolysaccharide

Local and chronic inflammatory reactions in the peritoneal environment play a key role in the pathogenesis of endometriosis. The menstrual blood of women with endometriosis contains a higher concentration of lipopolysaccharide (LPS) compared to women without the disease [72]. Lipopolysaccharide is an inflammatory mediator that is a component of Gram-negative bacterial cells. Under its influence, peritoneal macrophages secrete a variety of compounds such as vascular endothelial cell growth factor (VEGF), interleukin (IL)-6, IL-8, hepatocyte growth factor (HGF) and tumor necrosis factor-alpha (TNF-α) [58,73,74,75]. This LPS-induced secretion effect can be inhibited by the use of neutralizing antibodies for TLR4 and by polymyxin B, which is an LPS antagonist [72]. Khan et al. showed that high LPS content in peritoneal fluid (PF) due to menstrual blood reflux is associated with pelvic inflammatory disease and may contribute to TLR4-mediated progression of endometriosis [69]. Additionally, a study in mice showed that LPS, by generating peritoneal inflammation through activation of the TLR4/NF-κB pathway, exacerbated the development of endometriosis-like lesions [76].

2.2. The Importance of Inflammatory Mediators in Body Fluids (Peripheral Blood, Peritoneal Fluid, Urine) in the Development of Endometriosis

2.2.1. Anti-Inflammatory Cytokines

IL-4 and IL-10 are the main anti-inflammatory cytokines involved in disease progression by stimulating the survival, growth, invasion and immune escape of endometrial lesions [41]. The results of several studies indicate their elevated levels in the lymphocytes and ectopic endometrium of patients with endometriosis [77,78,79]. Particularly high levels of these cytokines were observed in women with advanced endometriosis [80,81,82]. One study showed significantly higher levels of IL-4 in serum and peritoneal fluid in adolescents with endometriosis, which may be used in future work on the development of a biomarker of this disease [80]. A study in the Chinese population showed that the presence of polymorphisms 592A/C and 819T/C of the IL-10 promoter is associated with an increased risk of endometriosis and may be associated with increased disease progression [82,83,84].
Elevated concentrations of these cytokines may result from insufficient control of their pro-inflammatory effects, excessive compensation and inhibition of the immune response. IL-4 is abundant in endometrial tissues and causes activation of p38 mitogen-activated protein kinases, which are a class of mitogen-activated protein kinases (MAPKs), stress-activated protein kinases (SAPK)/Jun amino-terminal kinases (JNK) and p42/44 MAPK signaling, leading to the proliferation of endometriotic stromal cells [85]. The role of IL-10 in endometriosis has been studied in an animal model. Decreasing IL-10 activity in mice with surgically induced endometriosis significantly reduced the size of endometrial lesions. In contrast, IL-10 administration increased endometrial changes in this model [86]. Another anti-inflammatory cytokine is IL-13. Its increased concentration has been demonstrated in the endometrium and in the peritoneal fluid of patients with endometriosis [87]. However, elevated serum levels of it have not been demonstrated. The group of anti-inflammatory cytokines also includes transforming growth factor β1 (TGF-β), which inhibits or reverses the activation of macrophages, thereby reducing the release of pro-inflammatory cytokines and reactive oxygen and nitrogen species [88]. Animal model studies have shown that elevated levels of this cytokine are associated with increased survival, adhesion and proliferation of ectopic endometrial cells during lesion development in endometriosis patients [89,90]. In addition, studies conducted among patients with endometriosis showed an increased level of TGF-β in their peripheral blood [91].

2.2.2. Pro-Inflammatory Cytokines

Studies indicate that elevated levels of pro-inflammatory cytokines can also be observed in the blood serum of women with endometriosis. A study by Borrelli et al. showed elevated levels of IL-8 and monocyte chemoattractant protein-1 (MCP-1/CCL2) in the peripheral blood of patients [92]. A study by Pizzo et al. also showed elevated IL-6 levels in this group of patients [86]. Elevated levels of another pro-inflammatory cytokine, IL-6, in peripheral blood were more common in patients with stage I-II endometriosis [86]. Tumor necrosis factor alpha (TNF-α) also belongs to the group of pro-inflammatory cytokines. Several studies have shown that its serum concentration in patients with endometriosis may be elevated [93]. The development and progression of endometriosis is also influenced by interactions between the two groups of cytokines. The pro-inflammatory cytokine IL-1β increases the secretion of anti-inflammatory IL-4 [94]. The pro-inflammatory cytokine TNF-α and the anti-inflammatory cytokine TGF-β1 significantly increase IL-13 secretion from endometrial epithelial cells (EECs) and endometrial stromal cells (ESCs) in vitro [95]. In contrast, IL-8 and IL-23 produced from ectopic lesions stimulate the differentiation of CD16—NK cells with high levels of cyclooxygenase (COX)-2, which express high levels of anti-inflammatory IL-10 and TGF-β1 [96].

2.2.3. Urinary Markers of Inflammation and Endometriosis

The examination of endometriosis-specific changes in body fluids mainly involves peripheral blood and peritoneal fluid. However, more and more research is focusing on the search for non-invasive, disease-specific markers in the urine. A study by Visnic et al. [97] noticed as many as 17 proteins that were excreted in the urine of endometriosis patients and were involved in the pathogenesis of the disease. Proteins that were elevated in urine included ENG (endoglin), LUM (lumican), TGFB2 (transforming growth factor beta receptor 2), TSPAN1 (tetraspanin-1), CD44, TNC (tenascin), CatG (cathepsin G), DSP (desmoplakin), THBS1 (thrombospondin 1), PCDH1 and PDCH3, (protocadherin-1 and-3), SPARCL1 (SPARC-like protein 1), ZGP1 (zinc-alpha-2-glycoprotein) and ANXA2 (annexin A2) (see Table 1).
These proteins are responsible for cell invasion, proliferation and adhesion, neoangiogenesis, proteolysis and degradation of the extracellular space, processes playing an important role in the pathogenesis of endometriosis [129]. In 2021, a study was conducted evaluating the use of BCL6 (B-cell lymphoma 6) and SIRT11 (NAD-dependent protein deacetylase sirtuin-1) as markers in non-invasive diagnostics of endometriosis, but the results showed that the possibilities of their use are low [130]. Identifying reliable protein markers in urine could potentially provide a non-invasive method for diagnosing and monitoring endometriosis. Several studies have explored the identification of specific protein biomarkers in urine samples from women with endometriosis. These biomarkers are typically proteins that are released or altered in response to the presence of endometriotic lesions. The interaction between protein markers in urine and the microbiota in endometriosis is an area of ongoing research and is not yet fully understood. However, there are a few potential mechanisms by which the microbiota and protein markers in urine may interact in the context of endometriosis: inflammatory response, metabolic alterations and barrier function [131,132]. The presence of endometriotic lesions can trigger an inflammatory response in the reproductive tract, which may lead to changes in the microbiota composition. This altered microbiota, in turn, can contribute to the release of specific proteins into the urine. The inflammatory response and changes in the microbiota may act synergistically, further perpetuating the inflammatory state associated with endometriosis. Dysbiosis or alterations in the microbiota composition observed in endometriosis could influence immune dysregulation, allowing the development and persistence of endometriotic lesions. This immune dysregulation may also affect the expression or release of certain proteins into the urine. Dysbiosis in endometriosis may result in the production of specific metabolites that can impact protein expression or function, leading to their appearance in urine samples. The microbiota plays a role in maintaining the integrity of the epithelial barrier in the reproductive tract. Disruptions in this barrier function could allow the translocation of microbial components or proteins into the urine, potentially leading to the detection of specific protein markers associated with endometriosis [131,132].
A study aimed at examining selected adhesion molecules as urinary biomarkers for diagnosing endometriosis showed no differences in the levels of sVCAM-1, sICAM-1, E-selectin and P-selectin in the urine of patients with endometriosis compared to healthy patients [133]. Studies have also shown that CYFRA 21-1 in urine can be a valuable marker in the diagnosis of endometriosis [133,134]. Chen et al. observed significantly elevated urine histone 4 levels in patients with endometriosis [135]. Elevated levels of soluble fms-like tyrosine kinase (sFlt-1) corrected for creatinine have also been detected in the urine of patients with stage I and II endometriosis [133]. The use of cytokeratin-19 (CK19) as a biomarker was also investigated; however, research results are contradictory. Kuessel et al. showed that there is no significant relationship between urinary CK19 levels and endometriosis [136]. In turn, a study by Tokushige et al. showed that CK19 was expressed only in the urine of women with endometriosis and was absent in the urine samples of healthy patients [137].
Metabolites involved in inflammation and oxidative stress were also found in the urine samples of endometriosis patients. These samples contained higher levels of guanidinosuccinate, creatinine, taurine, valine, 2-hydroxyisovalerate and N-methyl-4-pyridone-5-carboxamide, and decreased lysine levels, compared to disease-free women [138]. An animal-model study showed that the tryptophan metabolites 4,6-dihydroxyquinoline and 5-hydroxy-L-tryptophan were significantly altered in the urine of model endometriosis rats [139]. Regarding estrogen metabolites, a study by Othman et al. found that the urine of women with endometriosis had significantly higher levels of 2-hydroxyestrone than women without the disease, but the ratios of 2-hydroxyestrone/16α-hydroxyestrone were comparable [140].
In 2021, a study was conducted to determine the association of microbial dynamics with estrogens and estrogen metabolites in the urine of endometriosis patients. The results showed that the concentrations of 17β-estradiol and 16-keto-17β-estradiol were higher in these patients. In addition, lower levels of estriol, 2-hydroxyestrone and 2-hydroxyestradiol were observed, mainly in patients using oral contraceptives. Another aim of the study was to analyze the relationship between the species of urinary bacteria and the concentration of estrogens in the urine. In a control group consisting of patients without endometriosis, an association was observed between bacteria in the urine and the concentration of 2-hydroxyestrone in urine samples. In patients with endometriosis, a strong correlation was found between urinary bacteria and the levels of E2, E3, 2-hydroxyestradiol and 2-hydroxyestrone. These results indicate that there is a relationship between abnormal levels of estrogens and their metabolites in the urine and altered composition of the urinary microbiome in patients with endometriosis. These findings suggest that patients with endometriosis may have a unique profile of estrogen metabolites [141].
Metabolic and hormonal disorders can have a significant impact on the microbiota in the context of endometriosis. The microbiota is highly influenced by the host’s metabolism, and changes in nutrient availability can promote the growth of certain bacterial species while suppressing others. This disruption in the metabolic environment can potentially lead to dysbiosis in the reproductive tract microbiota. Hormonal imbalances, particularly concerning estrogen, are not only a hallmark of endometriosis, but can also directly affect the growth of certain bacteria and alter the gene expression of microbes involved in metabolism and virulence. Therefore, endometriosis-related hormonal imbalances have the potential to disrupt the microflora of the reproductive system. Moreover, metabolic and hormonal disturbances in endometriosis may contribute to immune dysregulation that affects the diversity and composition of the microflora and the integrity of the mucosal barrier in the reproductive system. Disruption of barrier function can allow translocation of microbial components and metabolites into surrounding tissues, leading to inflammation and immune activation. This altered barrier function may also affect the composition and diversity of the microflora [131,142,143,144].

3. Influence of Microbiota Dysbiosis of the Reproductive Tract, Intestines and Oral Cavity on the Pathogenesis of Endometriosis

3.1. The Microbiota of the Genital Tract and Endometriosis

Microorganisms and hosts have synergistic links in virtually every niche of the human body, affecting its physiology and physiopathology [145,146,147,148,149]. Similarly, the female genital organs (FGT)—the vagina, cervix, endometrium, fallopian tubes and ovaries—have their own microbiome, accounting for 9% of the total number of bacteria in a woman’s body [150]. Recent molecular studies have shown that the presence of bacteria in the endometrium plays an important role in the proper functioning of the endometrium and the development of pregnancy under normal conditions [151].
The vaginal microbiome tends to play a significant role in the prevention of urogenital diseases such as bacterial vaginosis, inflammatory diseases, sexually transmitted infections and urinary tract infections. This protective role is primarily due to Lactobacillus spp., which is often associated with good gynecological and reproductive health. Lactobacillus spp. produce lactic acid and various bacteriostatic and bactericidal components that contribute to lowering the pH of the vaginal environment to a value less than or equal to 4.5 and promote competitive exclusion [152]. The estrogen-dominant vaginal epithelium and host amylase are essential for the survival and growth of Lactobacillus spp. [153,154,155]. In addition, Lactobacillus spp. help maintain homeostasis by occupying this niche (pathogen exclusion) and producing anti-inflammatory cytokines and antimicrobial peptides from epithelial cells, which strengthens the epithelial cell barrier and gynecological health [156,157,158,159]. The composition and number of microorganisms is extremely diverse in different physiological (menstruation, menopause) (Figure 1) and pathological conditions.
Several studies have proven that Lactobacillus is the dominant genus in the endometrium as well as in the vaginal environment. Mitchell et al. studied the microflora of the vagina and uterus and found that the endometrium is distinguished by the presence of Lactobacillus, the most widespread genus, followed by Gardnerella, Prevotella, Atopobium and Sneathia [163]. However, the composition of the vaginal microflora varies at different stages of menopause (pre-menopausal, perimenopausal and post-menopausal), as well as in pathological conditions such as vaginal atrophy, when the number of Lactobacillus decreases and the number of microorganisms such as Anaerococcus, Peptoniphilus and Prevotella increases [164]. Further research by Verstraelen et al. showed that 90% of the results included in their study were consistent with the presence of a unique microflora dominated by Bacteroides (Bacteroides xylanisolvens, Bacteroides thetaiotaomicron and Bacteroides fragilis) and Pelomonas residing on the endometrium of the human non-pregnant uterus [165].
Research conducted by Chen’s team indicates the presence of many bacterial communities, the most numerous of which are Pseudomonas spp., Acinetobacter spp., Vagococcus spp. and Sphingobium spp. [145]. Similar results were obtained by sequencing endometrial samples from 25 women who underwent total hysterectomy for fibroids or endometrial hyperplasia and it was found that Acinetobacter spp., Pseudomonas spp., Comamonadaceae spp. and Cloacibacterium spp. were the most prevalent taxa in the endometrium [166]. Recent research by the team of Lu et al. in 2021 showed that Rhodococcus spp., Phyllobacterium spp., Sphingomonas spp., Bacteroides spp. and Bifidobacterium spp. were more numerous in the endometrium than Lactobacillus spp. in patients diagnosed with endometrial cancer [167]. Until now, the lack of contamination control, evidence of bacterial viability and transcervical sampling has made it difficult to evaluate the results of these genital tract microbiota studies and their direct impact on the pathogenesis of endometriosis.
In recent years, some scientists have proven the non-sterility of the endometrium. There is evidence that bacteria can enter the uterus as “tourists” or “invaders”, as discussed in a study by Baker et al. (2018). Furthermore, embryos from a woman’s lower genital tract can be transferred in and out of the uterus by the peristaltic pump of the uterus [168], and the peristaltic contractions of the uterine fundus rapidly transport sperm out of the canal cervix to uterus and fallopian tube [169]. The frequency of these contractions changes as the proliferative phase progresses, as was observed using labeled macrospheres over several minutes [169]. In women with infertility and endometriosis, uterine hyperperistalsis has been documented with approximately twice the frequency of contractions [170]. This hyperperistalsis in patients with endometriosis may contribute not only to the transfer of detached endometrial cells to the fallopian tubes and peritoneal cavity, but also to the transfer of microorganisms from the lower genital tract. Mid-cycle uterine dysperistalsis can occur in women, causing undirected and convulsive contractions that can lead to infertility by interfering with sperm transport, but can also result in a high probability of translocation of bacteria into the upper vaginal canal. Several studies have reported some abnormalities of the endometrium in the predominant types of patients with endometriosis. Indeed, Fang et al. compared the bacterial composition of the vagina with the endometrium, as well as the composition of the endometrial microbiome of healthy women, patients with endometrial polyps and patients with chronic endometritis. Proteobacteria spp., Firmicutes spp. and Actinobacteria spp. dominated the intrauterine microbiome in all groups studied. Additionally, although there were significant differences in the vaginal and endometrial microbiome, numbers of Lactobacillus spp., Gardnerella spp., Bifidobacterium spp., Streptococcus spp. and Alteromonas spp. were significantly higher in the healthy group compared to others [171]. Ata et al. conducted a study to compare the composition of the vaginal, cervical and intestinal microflora of women with stage III/IV endometriosis with healthy controls. A difference was identified at the genus level. They found an increased abundance of potentially pathogenic species in the cervical microbiota of women with endometriosis, including Gardnerella spp., Streptococcus spp., Escherichia spp., Shigella spp. and Ureaplasma spp. Surprisingly, they did not find Atopobium spp., a gynecological pathogen, in the vagina and cervix of the endometriosis group [172]. Another study showed a significant incidence of Atopobium vaginae in women. Atopobium may exacerbate intracellular Porphyromonas infection, causing changes in cell regulatory processes and a carcinogenic trigger in women with endometrial cancer. In contrast, they found that A. vaginae was less common in women with mild gynecological disorders, suggesting a possible link at a different stage of action, given that endometriosis is also a benign gynecological disease [173].
Since it is still unknown whether these changes are the cause or effect of the disease, studies in non-human primates (Papio anubis) were conducted to test the hypothesis that the development of endometrial changes causes changes in immune and bacterial dynamics that may promote disease progression. According to the study, the induction of endometriosis decreased peripheral Tregs while increasing the number of Th17 cells in all post-induction harvests, indicating systemic inflammation. After disease induction, the diversity and abundance of the microbiome changed in the vaginal and intestinal microbiota. Consequently, the induction of endometriosis in primates resulted in an immunological shift towards an inflammatory profile, as well as altered mucosal microbial profiles, which may promote inflammation by producing inflammatory mediators [174] (Figure 2).
The vaginal community status type (CST) consists of five CSTs. CST I, II, III and V are associated with a healthy vaginal microbiome and are dominated by Lactobacillus gasseri, Lactobacillus crispatus, Lactobacillus iners and Lactobacillus jensenii. CST IV is associated with vaginal inflammation and dysbiosis. This type consists of more strictly anaerobic bacteria (e.g., Dialister, Gardenerella, Megasphaera, Peptoniphilus, Sneathia, Finegoldia and Prevotella) [160]. The presence of specific species of bacteria in the urogenital (UG) system, such as Lactobacillus spp. or Bifidobacterium infantis, protects mucosal epithelial cells and creates an environment that prevents pathogen survival by producing lactic acid, which lowers vaginal pH [175].
In 2022, Le et al. conducted a study on reproductive olive baboons to assess the impact of endometriosis on mucosal microbial community immunity and dynamics. Comparison of pre-disease samples with samples from baboons with endometriosis showed that disease induction changed the dynamics of the urinary microbiome. As the disease progressed, the levels of a large proportion of species in the urinary tract decreased. The effect of lower diversity of microorganisms due to dysbiosis and inflammation is lower metabolic activity of microorganisms, which can lead to the destabilization of immune homeostasis. This study also analyzed the effect of microbiome dynamics on immune status to determine whether the change in microbiome diversity following endometriosis induction was related to populations of peripheral immune cells. The results showed that urinary dysbiosis in affected animals is accompanied by changes in the levels of peripheral nTregs, iTregs and Th17 cell populations, suggesting that there may be a unique composition of the urinary microbiome as well as a distinct immune profile associated with the induction of endometriosis [173]. Sex hormones contribute to changes in the microbiome of the urinary tract. Decreased estrogen levels during menopause may result in a reduction in the number of commensal species [176]. In patients with endometriosis, an increase in the level of circulating bioactive estrogens may initiate the development and persistence of endometrial changes. Therefore, changes in the composition of the microbiome in these patients may affect estrogen production [177].

3.2. Gut Microbiota and Endometriosis

The digestive tract is heavily filled with lymphoid structures that contain cells associated with the immune system [178]. It is well known that the intestinal microflora plays an important role in the development of these structures, as well as in the development of immune cell activity. In fact, animal germ-free models have shown that the absence of gut microbiota is correlated with deficiency in secretory IgA and CD8αβ intraepithelial lymphocytes [178]. The gut microbiota also affects the composition of mucosal T lymphocytes (Th1, Th17, Treg, etc.) and dysbiosis can disrupt this delicate balance, causing inflammation and various diseases. These investigations proved that the gut microbiota has a putative role in the function of immune system cells [178]. In addition, commensal microorganisms compete for resources by limiting the colonization of harmful microbes. For example, lactobacilli inhibit the binding of Neisseria gonorrhoeae to the female reproductive system, protecting the host from infection [179]. Commensal bacteria also constantly excite the receptors, resulting in an increase in the number of TLRs and, as a result, enhanced immune surveillance [180]. While certain types of gut bacteria contribute to the development of endometriosis, other microorganisms also help create a healthy barrier in the gut. The impact of gut bacteria on host physiology and immunological processes is mediated through various mechanisms. One such mechanism involves the breakdown of otherwise indigestible nutrients into biologically active metabolites, including short-chain fatty acids (SCFAs) [181,182]. SCFAs such as acetate, propionate, n-butyrate, pentanoic acid (valeric acid) and hexanoic acid (caproic acid) serve as an energy source for enterocytes or are transported into the bloodstream [183]. These SCFAs have been shown to exhibit anti-proliferative effects [184,185,186] and possess anti-inflammatory properties [187] that can extend to distant organs [188]. In lipopolysaccharide-induced macrophages, n-butyrate inhibits the expression of pro-inflammatory cytokines such as tumor necrosis factor α (TNF-α) and IL-6 [189]. SCFAs primarily exert their effects on cells through two main mechanisms. Firstly, they can activate G-protein-coupled receptors, namely, GPR43, GPR41 and GPR109A [190], which are known to downregulate inflammation [191,192]. Secondly, SCFAs can inhibit histone deacetylases [193,194]. Furthermore, it is observed that the fecal matter of mice with endometriosis contained lower levels of short-chain fatty acids and n-butyrate compared to mice without the condition. In a pre-clinical mouse model, treatment with n-butyrate resulted in a decrease in the growth of both mouse and human endometriotic lesions [195].
In addition, microorganisms play an important role in the physiological functions of other mucosal surfaces, such as endometrial remodeling in the uterus and increasing estrogen levels in the blood [180]. Recent studies have shown that the unique EMS microenvironment may favor the propensity of M2 endometrial macrophages to polarize towards M1 polarization and that M2 macrophages paradoxically express pro-inflammatory phenotypes in the early phase, although the pro-inflammatory phenotype of M1 macrophages is promoted when endometrial changes are established [196].
A significant number of Gram-negative bacteria are translocated and infiltrated outside the intestinal cavity as a result of the altered composition of the intestinal microbiota induced by endometriosis, which results in the destruction of intestinal tight junctions and the reduction of tight junction protein 2 (ZO-2) expression [197], resulting in the infiltration of a significant amount of Gram-negative bacteria outside the intestine [198].
According to Harada et al., LPS can activate the macrophage TLR4 in innate immunity, leading to the production of significant levels of TNF-α and IL-8 and the development of an inflammatory environment [199]. TNF-alpha and IL-8 are crucial for endometrial tissue adhesion and angiogenesis induction [200]. The expression of pro-inflammatory phenotypes by M1 macrophages may be facilitated by activation of TLR4 signaling and the inflammatory environment [201]. Firmicutes, Bacteroidetes, Proteobacteria, Actinobacteria, Spirochaetes and Fusobacteria phyla predominate in the population of oral bacteria, and the main properties of these major bacterial taxa have been previously characterized [202,203]. The list of bacterial genera and types in the publicly available Human Oral Microbiota Database (HOMD) is constantly updated. The HOMD study showed that each person has a distinct microflora. The “true” oral microflora mix is difficult to identify because the oral cavity is constantly exposed to the external environment, allowing exogenous microorganisms, air and food to enter [203]. It should be noted that bacterial populations may vary depending on the oral sampling site. Differences in oral microbial diversity have been identified at various sites in the oral cavity, particularly between the mucosa and teeth and between saliva and teeth [204]. A recent study showed that the microbiota of the buccal, gingival and hard palate mucosa were comparable, although saliva, tongue, tonsils and pharynx as well as supragingival and subgingival plaques had altered taxa [205]. So far, saliva is the most perfect oral compartment to study changes in microbial composition in many human diseases, because it contains the most representative oral microflora [206]. The oral microbiome can significantly contribute to the development of various human diseases, most of which are caused by the accumulation of oral bacteria leading to inflammation [207].
Although little is known about the oral microbiome and endometriosis, other inflammatory conditions can affect the oral microbiome. While inflammatory bowel disease (IBD) is a chronic inflammatory condition that affects the digestive tract, patients with IBD can develop oral mucositis [208]. Statistically significant differences in the oral microflora of young IBD patients have been shown to be significantly less than in healthy controls, with a significant reduction in Fusobacteria and Firmicutes phyla [209]. Behçet’s syndrome is a multi-immune systemic disease characterized by an enhanced systemic inflammatory response. Mouth ulceration is a common symptom of this condition. The microbiome of the oral mucosa and saliva of patients with Behçet’s syndrome differed from that of healthy individuals; a significant proportion of Actinobacteria spp. were found in patients with Behçet’s disease and oral ulcers [210]. The extremely inflammatory nature of this condition may affect the composition of the oral microbiome. Dysfunction of the oral microbiome is also associated with other chronic inflammatory diseases such as atherosclerosis and cardiovascular problems. Slocum et al. analyzed the putative pathways through which periodontal infections directly or indirectly stimulate immune dysregulation and, as a result, the progressive inflammation that manifests in cardiovascular disorders [211]. Furthermore, autoimmune diseases have the potential to disrupt the host’s commensal oral microbiota. Rheumatoid arthritis (RA) is an inflammatory disease mainly characterized by inflammation of the joints. The oral and salivary microbiota of RA patients has been shown to be significantly different from that of healthy humans using 16S rRNA sequencing, which closely matches clinical RA measurements such as C-reactive protein levels [212]. The chronic inflammatory nature of RA may play a significant role in the dysbiosis of the oral flora, allowing oral pathogens to colonize the oral region. As a consequence, oral dysbiosis causes changes in the relative abundances of individual components of the bacterial community. Immunological identification of periodontal infection causes an increase in inflammation, which includes the influx of immune cells such as monocytes, B and T lymphocytes, and the synthesis of inflammatory mediators [213,214]. When inflammation becomes persistent, it can contribute to endometriosis. Substantial long-term studies are required to confirm the current evidence implicating oral dysbiosis as an independent risk factor for endometriosis.

4. Metabolic Regulation of Estrogen Metabolism and Microbiota

As mentioned earlier, estrogens are essential for the development and functioning of the female reproductive system and are closely involved in the development of endometriosis [159,215,216,217]. Estrogens affect the microenvironment of the lower female reproductive system by increasing epithelial thickness, glycogen concentration, mucus secretion, promoting lactobacilli abundance and indirectly producing lactic acid [218]. Endometriosis, endometrial cancer and uterine fibroids are proliferative diseases associated with hormonal imbalance [219]. Estrogen promotes ectopic endometrial tissue formation and inflammatory activity in women, and endometriosis has been linked to changes in estrogen signaling [217]. For example, women with endometriosis show an increased pro-inflammatory and anti-apoptotic response to estradiol [220]. This may be due to changes in the expression of the nuclear estrogen receptor. Furthermore, elevated estrogen levels combined with the lack of opposing effects of progesterone create imbalances in progesterone and estrogen production, affecting the epithelium and potentially leading to uncontrolled profiling and chronic inflammation that promotes the development of endometriosis [221,222]. It is well known that the liver binds circulating estrogens to glucuronic acid, which does not bind to estrogen receptors (via glucuronidation). Glucuronic acid-conjugated estrogens are more hydrophilic, allowing them to be excreted in the bile (as bile salts) and then released into the stomach to eliminate conjugated toxins and hormones that are no longer needed [223] (Figure 3).
It has been proven that the intestinal microflora, and especially changes in the intestinal microflora (dysbiosis), are involved in the reactivation of estrogens through the bacterial secretion of the enzymatic β-glucuronidase, detected in some intestinal bacteria, such as Escherichia coli, Bacteroides fragilis and Streptococcus agalactiae [226], which are capable of deconjugating glucuronic acid [227,228]. As a result, active circulating estrogen is reabsorbed, which increases selectivity for beta estrogen receptor (ER) targets [229,230]. Many studies have linked ER variability to estradiol levels, although the mechanism is uncertain [229,231].
β-glucuronidase activity is essential for the formation of potentially harmful and carcinogenic metabolites in the intestines, as well as for the resorption of various chemicals in the circulatory system, such as estrogens [232]. β-glucuronidase promotes estrogen receptor binding, and activation of these receptors promotes proliferation. Changes in gut microbial diversity can disrupt or dysregulate circulating estrogen levels, fueling estrogen-mediated disease, contributing to hyper- or hypoestrogenic states [156,233].
Notably, the microbiota may be responsible for endometriosis by promoting inflammation and hormonal dysregulation (via the estrobolome), affecting cell proliferation/apoptosis, metabolism and oxidative stress, and increasing angiogenesis/vascularity [234,235,236,237,238]. As a result, research on the estrobolome is expected to provide new information that can be used in future interventions and therapies for endometriosis and other estrogen-mediated diseases. The gut microbiota plays a key role in the biotransformation of estrogen, which can affect hormonal balance and estrogen-related conditions such as endometriosis. Although research on the specific interactions between the estrobolome and the microbiome in endometriosis is still limited, there are several potential mechanisms for their interaction. This applies not only to the metabolism of estrogens in the body, but also to the development of inflammation or secondary metabolites. The gut microbiota can influence estrogen metabolism through the production of enzymes that can either increase or decrease estrogen levels. Some bacteria possess the ability to produce β-glucuronidase, an enzyme that can deconjugate estrogen metabolites, allowing them to be reabsorbed into circulation. This process can lead to increased exposure to estrogens and potentially exacerbate estrogen-driven conditions such as endometriosis [227,228]. The gut microbiota can produce metabolites that can influence estrogen metabolism and affect endometriosis. For example, certain bacterial species can produce SCFAs through the fermentation of dietary fibers. SCFAs can influence estrogen metabolism by modulating the expression of estrogen-metabolizing enzymes in the liver and other tissues. These metabolites can potentially impact estrogen levels and the hormonal balance involved in endometriosis [227].

5. Future Directions—The Role of Probiotics and Prebiotics in Endometriosis

The presence of bacteria in both the gut and uterus has been found to be significant in the development and progression of endometriosis. This understanding opens up potential avenues for improving endometriosis care. Modulating the microbiota, either through antibiotics or probiotics, could be a treatment and/or prevention of endometriosis. Probiotics and prebiotics are two components of the emerging field of microbiome-based therapies. While research specifically focused on the role of probiotics and prebiotics in endometriosis is limited, they have been studied in the context of other gynecological conditions and may have potential benefits for endometriosis [239].
Probiotics are live microorganisms that, when administered in adequate amounts, confer health benefits to the host. They are commonly found in certain foods or can be taken as supplements. Probiotics have been investigated for their potential effects on immune regulation, inflammation and gut dysbiosis, all of which are relevant in the context of endometriosis. While the specific strains and mechanisms of action are still being explored, probiotics may offer potential benefits like the modulation of immune response, restoration of gut microbiota and reduction of vaginal dysbiosis [240,241].
Probiotics can help regulate the immune system by promoting anti-inflammatory responses and balancing immune cell activity. This immune modulation could potentially reduce the inflammatory environment associated with endometriosis. Imbalances in the gut microbiota have been associated with various health conditions, including endometriosis. Probiotics may help restore microbial balance in the gut, potentially improving overall gut health and impacting systemic inflammation and immune responses. Some studies have explored the use of probiotics to restore a healthy vaginal microbiota, which could potentially be relevant for endometriosis-related symptoms such as pelvic pain and inflammation [142,240,241,242]. Studies have shown that oral administration of Lactobacillus in women with endometriosis can alleviate endometriosis-associated pain and reduce endometriotic lesions in mice. This effect may be achieved by increasing IL-12 concentration and enhancing NK cell activity, thus reversing immune dysregulation associated with endometriosis. Probiotic treatment with Lactobacillus has also been found to prevent the growth of endometriosis in rat models [243,244,245]. In order to increase the effectiveness of probiotics, it seems important to combine them with prebiotics. Prebiotics are indigestible fibers that serve as food for the beneficial bacteria in your gut. By selectively promoting the growth and activity of beneficial microbes, prebiotics can help improve the composition and function of the gut microbiota. While research focusing specifically on prebiotics in endometriosis is limited, their potential benefits for gut health and immune regulation are promising for future applications. Prebiotics primarily can stimulate the growth of beneficial bacteria, thereby increasing the diversity of microbes in the gut. This increased diversity may contribute to a healthier gut environment and potentially influence systemic inflammation and immune responses. Prebiotics may also support the integrity of the intestinal barrier by reducing the translocation of harmful bacteria and their products into the bloodstream. This can help reduce systemic inflammation and potentially affect symptoms associated with endometriosis [242,246].

6. Conclusions

Recently, the microbiological aspects of endometriosis have been investigated, but the nature of these interactions is still unclear. Our study gathers the latest knowledge about the involvement of microflora in endometriosis, mainly research on intestinal microflora with clinical results. According to this research, the development and progression of endometriosis may be closely related to the microbiome of the female reproductive system and intestines. This shows that monitoring microbiome diversity can be an effective marker in this clinically difficult disease. More research is needed to investigate the application of this fresh perspective on endometriosis, which may have a potential role in finding preventive, diagnostic and therapeutic options, and which will involve clinical trials.
The results of the laboratory and clinical studies presented in this literature review confirm that the composition of the microbiome of patients with endometriosis differs from the composition of the microbiome of patients without this disease. The main problem with endometriosis is delayed diagnosis. Usually, the average time from onset of symptoms to diagnosis is about 8–10 years. The gold standard for diagnosis is laparoscopy, which has the disadvantage of being invasive. Identifying the composition of the disease-specific microbiome would enable the development of non-invasive diagnostics that could help reduce the incidence of delayed diagnoses, when the disease is usually at a more advanced stage. Endometriosis is a disease that greatly affects the quality of life of patients, so it is important to find new and effective forms of treatment for this disease. Understanding the functional role of microorganisms in endometriosis would also open the door to the use of antibiotics, prebiotics, probiotics and microbial transplants to alter the disease. However, to make this possible, more research is needed on the role of the microbiome in endometriosis.

Author Contributions

Conceptualization, A.S., A.C. and P.M.; methodology, A.S., A.C. and P.M.; validation, P.M., E.G. and A.S.; formal analysis, P.M.; investigation, A.S., A.C., P.M. and E.G.; resources, A.C.; data curation, P.M.; writing—original draft preparation, A.S., A.C. and P.M.; writing—review and editing, P.M. and E.G.; visualization, P.M.; supervision, E.G.; funding acquisition, A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by research grant number PBsd168 of the Medical University of Lublin.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

For more information, please contact the first author of this publication.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Maybin, J.A.; Critchley, H.O.D. Menstrual Physiology: Implications for Endometrial Pathology and Beyond. Hum. Reprod. Update 2015, 21, 748–761. [Google Scholar] [CrossRef] [Green Version]
  2. Karamian, A.; Paktinat, S.; Esfandyari, S.; Nazarian, H.; Ziai, S.A.; Zarnani, A.-H.; Salehpour, S.; Hosseinirad, H.; Karamian, A.; Novin, M.G. Pyrvinium Pamoate Induces In-Vitro Suppression of IL-6 and IL-8 Produced by Human Endometriotic Stromal Cells. Hum. Exp. Toxicol. 2021, 40, 649–660. [Google Scholar] [CrossRef] [PubMed]
  3. Nnoaham, K.E.; Hummelshoj, L.; Webster, P.; d’Hooghe, T.; de Cicco Nardone, F.; de Cicco Nardone, C.; Jenkinson, C.; Kennedy, S.H.; Zondervan, K.T. World Endometriosis Research Foundation Global Study of Women’s Health consortium Impact of Endometriosis on Quality of Life and Work Productivity: A Multicenter Study across Ten Countries. Fertil. Steril. 2011, 96, 366–373.e8. [Google Scholar] [CrossRef] [Green Version]
  4. Rocha, A.L.L.; Reis, F.M.; Taylor, R.N. Angiogenesis and Endometriosis. Obstet. Gynecol. Int. 2013, 2013, e859619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Husby, G.K.; Haugen, R.S.; Moen, M.H. Diagnostic Delay in Women with Pain and Endometriosis. Acta Obstet. Et Gynecol. Scand. 2003, 82, 649–653. [Google Scholar] [CrossRef] [PubMed]
  6. Sourial, S.; Tempest, N.; Hapangama, D.K. Theories on the Pathogenesis of Endometriosis. Int. J. Reprod. Med. 2014, 2014, e179515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Youseflu, S.; Jahanian Sadatmahalleh, S.; Mottaghi, A.; Kazemnejad, A. The Association of Food Consumption and Nutrient Intake with Endometriosis Risk in Iranian Women: A Case-Control Study. Int. J. Reprod. BioMed. 2019, 17, 661–670. [Google Scholar] [CrossRef]
  8. Christodoulakos, G.; Augoulea, A.; Lambrinoudaki, I.; Sioulas, V.; Creatsas, G. Pathogenesis of Endometriosis: The Role of Defective ‘Immunosurveillance’. Eur. J. Contracept. Reprod. Health Care 2007, 12, 194–202. [Google Scholar] [CrossRef]
  9. Sampson, J.A. Peritoneal endometriosis due to the menstrual dissemination of endometrial tissue into the peritoneal cavity. Am. J. Obstet. Gynecol. 1927, 14, 422–469. [Google Scholar] [CrossRef]
  10. Ferguson, B.R.; Bennington, J.L.; Haber, S.L. Histochemistry of mucosubstances and histology of mixed Müllerian pelvic lymph node glandular inclusions. Evidence for histogenesis by Müllerian metaplasia of coelomic epithelium. Obstet. Gynecol. 1969, 33, 617–625. [Google Scholar]
  11. Sampson, J.A. Metastatic or Embolic Endometriosis, due to the Menstrual Dissemination of Endometrial Tissue into the Venous Circulation. Am. J. Pathol. 1927, 3, 93–110. [Google Scholar] [PubMed]
  12. Sasson, I.E.; Taylor, H.S. Stem cells and the pathogenesis of endometriosis. Ann. N. Y. Acad. Sci. 2008, 1127, 106–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Lousse, J.-C.; Langendonckt, A.V.; Defrere, S.; Ramos, R.G.; Colette, S.; Donnez, J. Peritoneal Endometriosis Is an Inflammatory Disease. FBE 2012, 4, 23–40. [Google Scholar] [CrossRef]
  14. Gazvani, R.; Templeton, A. Peritoneal Environment, Cytokines and Angiogenesis in the Pathophysiology of Endometriosis. Reproduction 2002, 123, 217–226. [Google Scholar] [CrossRef] [PubMed]
  15. Van Langendonckt, A.; Casanas-Roux, F.; Donnez, J. Oxidative Stress and Peritoneal Endometriosis. Fertil. Steril. 2002, 77, 861–870. [Google Scholar] [CrossRef]
  16. Sikora, J.; Mielczarek-Palacz, A.; Kondera-Anasz, Z. Role of oxidative stress and mitochondrial abnormalities in endometriosis development. Front. Biosci. 2018, 23, 1010–1024. [Google Scholar]
  17. Silva, E.J.R.; do Amara, V.F.; Mendonça, J.M.; Nakao, L.S.; Neto, O.P.; Ferriani, R.A. Serum markers of oxidative stress and endometriosis. Clin. Exp. Obstet. Gynecol. 2014, 41, 371–374. [Google Scholar] [CrossRef]
  18. Harlev, A.; Gupta, S.; Agarwal, A. Targeting oxidative stress to treat endometriosis. Expert Opin. Ther. Targets 2015, 19, 1447–1464. [Google Scholar] [CrossRef]
  19. Andrisani, A.; Donà, G.; Brunati, A.M.; Clari, G.; Armanini, D.; Ragazzi, E.; Ambrosini, G.; Bordin, L. Increased oxidation-related glutathionylation and carbonic anhydrase activity in endometriosis. Reprod. Biomed. Online 2014, 28, 773–779. [Google Scholar] [CrossRef] [Green Version]
  20. Nagata, M. Inflammatory cells and oxygen radicals. Curr. Drug Targets Inflamm. Allergy 2005, 4, 503–504. [Google Scholar] [CrossRef] [PubMed]
  21. Donnez, J.; Binda, M.M.; Donnez, O.; Dolmans, M.-M. Oxidative stress in the pelvic cavity and its role in the pathogenesis of endometriosis. Fertil. Steril. 2016, 106, 1011–1017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Lin, Y.-H.; Chen, Y.-H.; Chang, H.-Y.; Au, H.-K.; Tzeng, C.-R.; Huang, Y.-H. Chronic Niche Inflammation in Endometriosis-Associated Infertility: Current Understanding and Future Therapeutic Strategies. Int. J. Mol. Sci. 2018, 19, 2385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ning, F.; Liu, H.; Lash, G.E. The Role of Decidual Macrophages During Normal and Pathological Pregnancy. Am. J. Reprod. Immunol. 2016, 75, 298–309. [Google Scholar] [CrossRef] [PubMed]
  24. Zhong, Q.; Yang, F.; Chen, X.; Li, J.; Zhong, C.; Chen, S. Patterns of Immune Infiltration in Endometriosis and Their Relationship to R-AFS Stages. Front. Genet. 2021, 12, 631715. [Google Scholar] [CrossRef]
  25. May, K.E.; Villar, J.; Kirtley, S.; Kennedy, S.H.; Becker, C.M. Endometrial Alterations in Endometriosis: A Systematic Review of Putative Biomarkers. Hum. Reprod. Update 2011, 17, 637–653. [Google Scholar] [CrossRef] [Green Version]
  26. Yang, H.; Zhu, L.; Wang, S.; Lang, J.; Xu, T. Noninvasive Diagnosis of Moderate to Severe Endometriosis: The Platelet-Lymphocyte Ratio Cannot Be a Neoadjuvant Biomarker for Serum Cancer Antigen 125. J. Minim. Invasive Gynecol. 2015, 22, 373–377. [Google Scholar] [CrossRef]
  27. Itoh, F.; Komohara, Y.; Takaishi, K.; Honda, R.; Tashiro, H.; Kyo, S.; Katabuchi, H.; Takeya, M. Possible Involvement of Signal Transducer and Activator of Transcription-3 in Cell-Cell Interactions of Peritoneal Macrophages and Endometrial Stromal Cells in Human Endometriosis. Fertil. Steril. 2013, 99, 1705–1713. [Google Scholar] [CrossRef]
  28. Shao, J.; Zhang, B.; Yu, J.-J.; Wei, C.-Y.; Zhou, W.-J.; Chang, K.-K.; Yang, H.-L.; Jin, L.-P.; Zhu, X.-Y.; Li, M.-Q. Macrophages Promote the Growth and Invasion of Endometrial Stromal Cells by Downregulating IL-24 in Endometriosis. Reproduction 2016, 152, 673–682. [Google Scholar] [CrossRef] [Green Version]
  29. Atri, C.; Guerfali, F.Z.; Laouini, D. Role of Human Macrophage Polarization in Inflammation during Infec-tious Diseases. Int. J. Mol. Sci. 2018, 19, 1801. [Google Scholar] [CrossRef] [Green Version]
  30. Milewski, Ł.; Dziunycz, P.; Barcz, E.; Radomski, D.; Roszkowski, P.I.; Korczak-Kowalska, G.; Kamiński, P.; Malejczyk, J. Increased Levels of Human Neutrophil Peptides 1, 2, and 3 in Peritoneal Fluid of Patients with Endometriosis: Association with Neutrophils, T Cells and IL-8. J. Reprod. Immunol. 2011, 91, 64–70. [Google Scholar] [CrossRef]
  31. Takamura, M.; Koga, K.; Izumi, G.; Urata, Y.; Nagai, M.; Hasegawa, A.; Harada, M.; Hirata, T.; Hirota, Y.; Wada-Hiraike, O.; et al. Neutrophil Depletion Reduces Endometriotic Lesion Formation in Mice. Am. J. Reprod. Immunol. 2016, 76, 193–198. [Google Scholar] [CrossRef] [PubMed]
  32. Kim, J.-Y.; Lee, D.-H.; Joo, J.-K.; Jin, J.-O.; Wang, J.-W.; Hong, Y.-S.; Kwak, J.-Y.; Lee, K.-S. ORIGINAL ARTICLE: Effects of Peritoneal Fluid from Endometriosis Patients on Interferon-γ-Induced Protein-10 (CXCL10) and Interleukin-8 (CXCL8) Released by Neutrophils and CD4+ T Cells. Am. J. Reprod. Immunol. 2009, 62, 128–138. [Google Scholar] [CrossRef] [PubMed]
  33. Kwak, J.-Y.; Park, S.-W.; Kim, K.-H.; Na, Y.-J.; Lee, K.-S. Modulation of Neutrophil Apoptosis by Plasma and Peritoneal Fluid from Patients with Advanced Endometriosis. Hum. Reprod. 2002, 17, 595–600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Somigliana, E.; Viganò, P.; Gaffuri, B.; Candiani, M.; Busacca, M.; Blasio, A.M.D.; Vignali, M. Modulation of NK Cell Lytic Function by Endometrial Secretory Factors: Potential Role in Endometriosis. Am. J. Reprod. Immunol. 1996, 36, 295–300. [Google Scholar] [CrossRef]
  35. Matsuoka, S.; Maeda, N.; Izumiya, C.; Yamashita, C.; Nishimori, Y.; Fukaya, T. Expression of Inhibitory-Motif Killer Immunoglobulin-Like Receptor, KIR2DL1, Is Increased in Natural Killer Cells from Women with Pelvic Endometriosis. Am. J. Reprod. Immunol. 2005, 53, 249–254. [Google Scholar] [CrossRef]
  36. Du, Y.; Liu, X.; Guo, S.-W. Platelets Impair Natural Killer Cell Reactivity and Function in Endometriosis through Multiple Mechanisms. Hum. Reprod. 2017, 32, 794–810. [Google Scholar] [CrossRef] [Green Version]
  37. Kang, Y.-J.; Jeung, I.C.; Park, A.; Park, Y.-J.; Jung, H.; Kim, T.-D.; Lee, H.G.; Choi, I.; Yoon, S.R. An Increased Level of IL-6 Suppresses NK Cell Activity in Peritoneal Fluid of Patients with Endometriosis via Regulation of SHP-2 Expression. Hum. Reprod. 2014, 29, 2176–2189. [Google Scholar] [CrossRef] [Green Version]
  38. Mosmann, T.R.; Coffman, R.L. TH1 and TH2 Cells: Different Patterns of Lymphokine Secretion Lead to Different Functional Properties. Annu. Rev. Immunol. 1989, 7, 145–173. [Google Scholar] [CrossRef]
  39. Park, H.; Li, Z.; Yang, X.O.; Chang, S.H.; Nurieva, R.; Wang, Y.-H.; Wang, Y.; Hood, L.; Zhu, Z.; Tian, Q.; et al. A Distinct Lineage of CD4 T Cells Regulates Tissue Inflammation by Producing Interleukin 17. Nat. Immunol. 2005, 6, 1133–1141. [Google Scholar] [CrossRef] [Green Version]
  40. Harrington, L.E.; Hatton, R.D.; Mangan, P.R.; Turner, H.; Murphy, T.L.; Murphy, K.M.; Weaver, C.T. Interleukin 17-Producing CD4+ Effector T Cells Develop via a Lineage Distinct from the T Helper Type 1 and 2 Lineages. Nat. Immunol. 2005, 6, 1123–1132. [Google Scholar] [CrossRef]
  41. Antsiferova, Y.S.; Sotnikova, N.Y.; Posiseeva, L.V.; Shor, A.L. Changes in the T-Helper Cytokine Profile and in Lymphocyte Activation at the Systemic and Local Levels in Women with Endometriosis. Fertil. Steril. 2005, 84, 1705–1711. [Google Scholar] [CrossRef] [PubMed]
  42. Borthwick, L.A.; Wynn, T.A.; Fisher, A.J. Cytokine Mediated Tissue Fibrosis. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2013, 1832, 1049–1060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Gogacz, M.; Winkler, I.; Bojarska-Junak, A.; Tabarkiewicz, J.; Semczuk, A.; Rechberger, T.; Adamiak, A. Increased Percentage of Th17 Cells in Peritoneal Fluid Is Associated with Severity of Endometriosis. J. Reprod. Immunol. 2016, 117, 39–44. [Google Scholar] [CrossRef] [PubMed]
  44. Hirata, T.; Osuga, Y.; Takamura, M.; Kodama, A.; Hirota, Y.; Koga, K.; Yoshino, O.; Harada, M.; Takemura, Y.; Yano, T.; et al. Recruitment of CCR6-Expressing Th17 Cells by CCL 20 Secreted from IL-1β-, TNF-α-, and IL-17A-Stimulated Endometriotic Stromal Cells. Endocrinology 2010, 151, 5468–5476. [Google Scholar] [CrossRef]
  45. Chen, P.; Zhang, Z.; Chen, Q.; Ren, F.; Li, T.; Zhang, C.; Wang, D. Expression of Th1 and Th2 Cytokine-Associated Transcription Factors, T-Bet and GATA-3, in the Eutopic Endometrium of Women with Endometriosis. Acta Histochem. 2012, 114, 779–784. [Google Scholar] [CrossRef]
  46. Olkowska-Truchanowicz, J.; Bocian, K.; Maksym, R.B.; Białoszewska, A.; Włodarczyk, D.; Baranowski, W.; Ząbek, J.; Korczak-Kowalska, G.; Malejczyk, J. CD4+ CD25+ FOXP3+ Regulatory T Cells in Peripheral Blood and Peritoneal Fluid of Patients with Endometriosis. Hum. Reprod. 2013, 28, 119–124. [Google Scholar] [CrossRef] [Green Version]
  47. Li, M.-Q.; Wang, Y.; Chang, K.-K.; Meng, Y.-H.; Liu, L.-B.; Mei, J.; Wang, Y.; Wang, X.-Q.; Jin, L.-P.; Li, D.-J. CD4+Foxp3+ Regulatory T Cell Differentiation Mediated by Endometrial Stromal Cell-Derived TECK Promotes the Growth and Invasion of Endometriotic Lesions. Cell Death Dis. 2014, 5, e1436. [Google Scholar] [CrossRef] [Green Version]
  48. Zihni, C.; Mills, C.; Matter, K.; Balda, M.S. Tight junctions: From simple barriers to multifunctional molecular gates. Nat. Rev. Mol. Cell Biol. 2016, 17, 564–580. [Google Scholar] [CrossRef]
  49. Günzel, D.; Yu, A.S.L. Claudins and the modulation of tight junction permeability. Physiol. Rev. 2013, 93, 525–569. [Google Scholar] [CrossRef] [Green Version]
  50. Loeffelmann, A.C.; Hoerscher, A.; Riaz, M.A.; Zeppernick, F.; Meinhold-Heerlein, I.; Konrad, L. Claudin-10 Expression Is Increased in Endometriosis and Adenomyosis and Mislocalized in Ectopic Endometriosis. Diagnostics 2022, 12, 2848. [Google Scholar] [CrossRef]
  51. Gaetje, R.; Holtrich, U.; Engels, K.; Kissler, S.; Rody, A.; Karn, T.; Kaufmann, M. Differential expression of claudins in human endometrium and endometriosis. Gynecol. Endocrinol. 2008, 24, 442–449. [Google Scholar] [CrossRef] [PubMed]
  52. Horné, F.; Dietze, R.; Berkes, E.; Oehmke, F.; Tinneberg, H.-R.; Meinhold-Heerlein, I.; Konrad, L. Impaired Localization of Claudin-11 in Endometriotic Epithelial Cells Compared to Endometrial Cells. Reprod. Sci. 2019, 26, 1181–1192. [Google Scholar] [CrossRef] [PubMed]
  53. Pan, X.-Y.; Li, X.; Weng, Z.-P.; Wang, B. Altered expression of claudin-3 and claudin-4 in ectopic endometrium of women with endometriosis. Fertil. Steril. 2009, 91, 1692–1699. [Google Scholar] [CrossRef] [PubMed]
  54. Hoerscher, A.; Horné, F.; Dietze, R.; Berkes, E.; Oehmke, F.; Tinneberg, H.-R.; Meinhold-Heerlein, I.; Konrad, L. Localization of claudin-2 and claudin-3 in eutopic and ectopic endometrium is highly similar. Arch. Gynecol. Obstet. 2020, 301, 1003–1011. [Google Scholar] [CrossRef] [PubMed]
  55. Kajihara, H.; Yamada, Y.; Kanayama, S.; Furukawa, N.; Noguchi, T.; Haruta, S.; Yoshida, S.; Sado, T.; Oi, H.; Kobayashi, H. New insights into the pathophysiology of endometriosis: From chronic inflammation to danger signal. Gynecol. Endocrinol. 2011, 27, 73–79. [Google Scholar] [CrossRef]
  56. Yu, Y.; Tang, D.; Kang, R. Oxidative stress-mediated HMGB1 biology. Front. Physiol. 2015, 6, 93. [Google Scholar] [CrossRef] [Green Version]
  57. Yun, B.H.; Chon, S.J.; Choi, Y.S.; Cho, S.; Lee, B.S.; Seo, S.K. Pathophysiology of Endometriosis: Role of High Mobility Group Box-1 and Toll-Like Receptor 4 Developing Inflammation in Endometrium. PLoS ONE 2016, 11, e0148165. [Google Scholar] [CrossRef] [Green Version]
  58. Khan, K.N.; Kitajima, M.; Imamura, T.; Hiraki, K.; Fujishita, A.; Sekine, I.; Ishimaru, T.; Masuzaki, H. Toll-like receptor 4-mediated growth of endometriosis by human heat-shock protein 70. Hum. Reprod. 2008, 23, 2210–2219. [Google Scholar] [CrossRef] [Green Version]
  59. Brubaker, S.W.; Bonham, K.S.; Zanoni, I.; Kagan, J.C. Innate Immune Pattern Recognition: A Cell Biological Perspective. Annu. Rev. Immunol. 2015, 33, 257–290. [Google Scholar] [CrossRef] [Green Version]
  60. Kawai, T.; Akira, S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 2011, 34, 637–650. [Google Scholar] [CrossRef] [Green Version]
  61. Mnich, M.E.; van Dalen, R.; Van Sorge, N.M. C-Type Lectin Receptors in Host Defense Against Bacterial Pathogens. Front. Cell. Infect. Microbiol. 2020, 10, 309. [Google Scholar] [CrossRef] [PubMed]
  62. Kim, Y.K.; Shin, J.-S.; Nahm, M.H. NOD-Like Receptors in Infection, Immunity, and Diseases. Yonsei Med. J. 2016, 57, 5–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Iurescia, S.; Fioretti, D.; Rinaldi, M. Targeting cytosolic nucleic acid-sensing pathways for cancer immunotherapies. Front. Immunol. 2018, 9, 711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Caneparo, V.; Landolfo, S.; Gariglio, M.; De Andrea, M. The Absent in Melanoma 2-Like Receptor IFN-Inducible Protein 16 as an Inflammasome Regulator in Systemic Lupus Erythematosus: The Dark Side of Sensing Microbes. Front. Immunol. 2018, 9, 1180. [Google Scholar] [CrossRef]
  65. Zindel, J.; Kubes, P. DAMPs, PAMPs, and LAMPs in Immunity and Sterile Inflammation. Annu. Rev. Pathol. Mech. Dis. 2020, 15, 493–518. [Google Scholar] [CrossRef] [Green Version]
  66. Saijo, Y.; EPi, L.; Yasuda, S. Pattern recognition receptors and signaling in plant–microbe interactions. Plant J. 2018, 93, 592–613. [Google Scholar] [CrossRef]
  67. He, S.; He, J.; Chen, J.; Chen, F.; Ou, S. Research advancement of innate immunity and pattern recognition receptors. Chin. J. Anim. Nutr. 2017, 29, 3844–3851. [Google Scholar]
  68. Mortaz, E.; Adcock, I.M.; Tabarsi, P.; Darazam, I.A.; Movassaghi, M.; Garssen, J.; Jamaati, H.; Velayati, A. Pattern recognitions receptors in immunodeficiency disorders. Eur. J. Pharmacol. 2017, 808, 49–56. [Google Scholar] [CrossRef]
  69. Azuma, Y.; Taniguchi, F.; Nakamura, K.; Nagira, K.; Khine, Y.M.; Kiyama, T.; Uegaki, T.; Izawa, M.; Harada, T. Lipopolysaccharide promotes the development of murine endometriosis-like lesions via the nuclear factor-kappa B pathway. Am. J. Reprod. Immunol. 2017, 77, e12631. [Google Scholar] [CrossRef] [Green Version]
  70. Herington, J.L.; Bruner-Tran, K.L.; A Lucas, J.; Osteen, K.G. Immune interactions in endometriosis. Expert Rev. Clin. Immunol. 2011, 7, 611–626. [Google Scholar] [CrossRef] [Green Version]
  71. Sobstyl, M.; Niedźwiedzka-Rystwej, P.; Grywalska, E.; Korona-Głowniak, I.; Sobstyl, A.; Bednarek, W.; Roliński, J. Toll-Like Receptor 2 Expression as a New Hallmark of Advanced Endometriosis. Cells 2020, 9, 1813. [Google Scholar] [CrossRef] [PubMed]
  72. 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.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Khan, K.N.; Masuzaki, H.; Fujishita, A.; Kitajima, M.; Kohno, T.; Sekine, I.; Matsuyama, T.; Ishimaru, T. Regulation of hepatocyte growth factor by basal and stimulated macrophages in women with endometriosis. Hum. Reprod. 2005, 20, 49–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Iba, Y.; Harada, T.; Horie, S.; Deura, I.; Iwabe, T.; Terakawa, N. Lipopolysaccharide-promoted proliferation of endometriotic stromal cells via induction of tumor necrosis factor α and interleukin-8 expression. Fertil. Steril. 2004, 82 (Suppl. S3), 1036–1042. [Google Scholar] [CrossRef] [Green Version]
  75. 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]
  76. García-Gómez, E.; Vázquez-Martínez, E.R.; Reyes-Mayoral, C.; Cruz-Orozco, O.P.; Camacho-Arroyo, I.; Cerbón, M. Regulation of Inflammation Pathways and Inflammasome by Sex Steroid Hormones in Endometriosis. Front. Endocrinol. 2020, 10, 935. [Google Scholar] [CrossRef] [Green Version]
  77. Suen, J.-L.; Chang, Y.; Chiu, P.-R.; Hsieh, T.-H.; Hsi, E.; Chen, Y.-C.; Chen, Y.-F.; Tsai, E.-M. Serum Level of IL-10 Is Increased in Patients with Endometriosis, and IL-10 Promotes the Growth of Lesions in a Murine Model. Am. J. Pathol. 2014, 184, 464–471. [Google Scholar] [CrossRef]
  78. Fan, Y.-Y.; Chen, H.-Y.; Chen, W.; Liu, Y.-N.; Fu, Y.; Wang, L.-N. Expression of Inflammatory Cytokines in Serum and Peritoneal Fluid from Patients with Different Stages of Endometriosis. Gynecol. Endocrinol. 2018, 34, 507–512. [Google Scholar] [CrossRef]
  79. Drosdzol-Cop, A.; Skrzypulec-Plinta, V.; Stojko, R. Serum and Peritoneal Fluid Immunological Markers in Adolescent Girls with Chronic Pelvic Pain. Obstet. Gynecol. Surv. 2012, 67, 374. [Google Scholar] [CrossRef]
  80. Chang, K.-K.; Liu, L.-B.; Jin, L.-P.; Zhang, B.; Mei, J.; Li, H.; Wei, C.-Y.; Zhou, W.-J.; Zhu, X.-Y.; Shao, J.; et al. IL-27 Triggers IL-10 Production in Th17 Cells via a c-Maf/RORγt/Blimp-1 Signal to Promote the Progression of Endometriosis. Cell Death Dis. 2017, 8, e2666. [Google Scholar] [CrossRef] [Green Version]
  81. Zhang, X.; Hei, P.; Deng, L.; Lin, J. Interleukin-10 Gene Promoter Polymorphisms and Their Protein Production in Peritoneal Fluid in Patients with Endometriosis. Mol. Hum. Reprod. 2007, 13, 135–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Malutan, A.M.; Drugan, C.; Walch, K.; Drugan, T.; Ciortea, R.; Mihu, D. The Association between Interleukin-10 (IL-10) −592C/A, −819T/C, −1082G/A Promoter Polymorphisms and Endometriosis. Arch. Gynecol. Obstet. 2017, 295, 503–510. [Google Scholar] [CrossRef] [PubMed]
  83. Xie, J.; Wang, S.; He, B.; Pan, Y.; Li, Y.; Zeng, Q.; Jiang, H.; Chen, J. Association of Estrogen Receptor Alpha and Interleukin-10 Gene Polymorphisms with Endometriosis in a Chinese Population. Fertil. Steril. 2009, 92, 54–60. [Google Scholar] [CrossRef]
  84. Kaabachi, W.; Kacem, O.; Belhaj, R.; Hamzaoui, A.; Hamzaoui, K. Interleukin-37 in Endometriosis. Immunol. Lett. 2017, 185, 52–55. [Google Scholar] [CrossRef] [PubMed]
  85. Pizzo, A.; Salmeri, F.M.; Ardita, F.V.; Sofo, V.; Tripepi, M.; Marsico, S. Behaviour of Cytokine Levels in Serum and Peritoneal Fluid of Women with Endometriosis. Gynecol. Obstet. Investig. 2003, 54, 82–87. [Google Scholar] [CrossRef] [PubMed]
  86. Chegini, N.; Roberts, M.; Ripps, B. Differential Expression of Interleukins (IL)-13 and IL-15 in Ectopic and Eutopic Endometrium of Women with Endometriosis and Normal Fertile Women. Am. J. Reprod. Immunol. 2003, 49, 75–83. [Google Scholar] [CrossRef]
  87. Li, M.O.; Wan, Y.Y.; Sanjabi, S.; Robertson, A.-K.L.; Flavell, R.A. Transforming Growth Factor-Β Regulation of Immune Responses. Annu. Rev. Immunol. 2006, 24, 99–146. [Google Scholar] [CrossRef]
  88. Young, V.J.; Ahmad, S.F.; Duncan, W.C.; Horne, A.W. The Role of TGF-β in the Pathophysiology of Peritoneal Endometriosis. Hum. Reprod. Update 2017, 23, 548–559. [Google Scholar] [CrossRef] [Green Version]
  89. Chegini, N. TGF-β System: The Principal Profibrotic Mediator of Peritoneal Adhesion Formation. Semin. Reprod. Med. 2008, 26, 298–312. [Google Scholar] [CrossRef]
  90. Sikora, J.; Smycz-Kubańska, M.; Mielczarek-Palacz, A.; Bednarek, I.; Kondera-Anasz, Z. The Involvement of Multifunctional TGF-β and Related Cytokines in Pathogenesis of Endometriosis. Immunol. Lett. 2018, 201, 31–37. [Google Scholar] [CrossRef]
  91. Borrelli, G.M.; Abrão, M.S.; Mechsner, S. Can Chemokines Be Used as Biomarkers for Endometriosis? A Systematic Review. Hum. Reprod. 2014, 29, 253–266. [Google Scholar] [CrossRef] [PubMed]
  92. Xavier, P.; Belo, L.; Beires, J.; Rebelo, I.; Martinez-de-Oliveira, J.; Lunet, N.; Barros, H. Serum Levels of VEGF and TNF-α and Their Association with C-Reactive Protein in Patients with Endometriosis. Arch. Gynecol. Obstet. 2006, 273, 227–231. [Google Scholar] [CrossRef] [PubMed]
  93. Wu, R.-F.; Yang, H.-M.; Zhou, W.-D.; Zhang, L.-R.; Bai, J.-B.; Lin, D.-C.; Ng, T.-W.; Dai, S.-J.; Chen, Q.-H.; Chen, Q.-X. Effect of Interleukin-1β and Lipoxin A4 in Human Endometriotic Stromal Cells: Proteomic Analysis. J. Obstet. Gynaecol. Res. 2017, 43, 308–319. [Google Scholar] [CrossRef]
  94. Mei, J.; Xie, X.-X.; Li, M.-Q.; Wei, C.-Y.; Jin, L.-P.; Li, D.-J.; Zhu, X.-Y. Indoleamine 2,3-Dioxygenase-1 (IDO1) in Human Endometrial Stromal Cells Induces Macrophage Tolerance through Interleukin-33 in the Progression of Endometriosis. Int. J. Clin. Exp. Pathol. 2014, 7, 2743–2757. [Google Scholar]
  95. Mei, J.; Zhou, W.-J.; Zhu, X.-Y.; Lu, H.; Wu, K.; Yang, H.-L.; Fu, Q.; Wei, C.-Y.; Chang, K.-K.; Jin, L.-P.; et al. Suppression of Autophagy and HCK Signaling Promotes PTGS2high FCGR3− NK Cell Differentiation Triggered by Ectopic Endometrial Stromal Cells. Autophagy 2018, 14, 1376–1397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Višnić, A.; Jurešić, G.; Domitrović, R.; Klarić, M.; Šepić, T.S.; Barišić, D. Proteins in urine—Possible biomarkers of endometriosis. J. Reprod. Immunol. 2023, 157, 103941. [Google Scholar] [CrossRef]
  97. ENG—Endoglin—Homo Sapiens (Human)|UniProtKB|UniProt. Available online: https://www.uniprot.org/uniprotkb/P17813/entry (accessed on 4 May 2023).
  98. Nolan-Stevaux, O.; Zhong, W.; Culp, S.; Shaffer, K.; Hoover, J.; Wickramasinghe, D.; Ruefli-Brasse, A. Endoglin Requirement for BMP9 Signaling in Endothelial Cells Reveals New Mechanism of Action for Selective Anti-Endoglin Antibodies. PLoS ONE 2012, 7, e50920. [Google Scholar] [CrossRef] [Green Version]
  99. Castonguay, R.; Werner, E.D.; Matthews, R.G.; Presman, E.; Mulivor, A.W.; Solban, N.; Sako, D.; Pearsall, R.S.; Underwood, K.W.; Seehra, J.; et al. Soluble Endoglin Specifically Binds Bone Morphogenetic Proteins 9 and 10 via Its Orphan Domain, Inhibits Blood Vessel Formation, and Suppresses Tumor Growth. J. Biol. Chem. 2011, 286, 30034–30046. [Google Scholar] [CrossRef] [Green Version]
  100. Chen, X.; Wang, J.; Tu, F.; Yang, Q.; Wang, D.; Zhu, Q. Endoglin Promotes Cell Migration and Invasion in Endometriosis by Regulating EMT. Ginekol. Pol. 2021, 1–16. [Google Scholar] [CrossRef]
  101. LUM—Lumican—Homo Sapiens (Human)|UniProtKB|UniProt. Available online: https://www.uniprot.org/uniprotkb/P51884/entry (accessed on 4 May 2023).
  102. Sahar, T.; Nigam, A.; Anjum, S.; Waziri, F.; Jain, S.K.; Wajid, S. Differential Expression of Lumican, Mimecan, Annexin A5 and Serotransferrin in Ectopic and Matched Eutopic Endometrium in Ovarian Endometriosis: A Case-Control Study. Gynecol. Endocrinol. 2021, 37, 56–60. [Google Scholar] [CrossRef]
  103. TGFBR2—TGF-Beta Receptor Type-2—Homo Sapiens (Human)|UniProtKB|UniProt. Available online: https://www.uniprot.org/uniprotkb/P37173/entry (accessed on 4 May 2023).
  104. TSPAN1—Tetraspanin-1—Homo Sapiens (Human)|UniProtKB|UniProt. Available online: https://www.uniprot.org/uniprotkb/O60635/entry (accessed on 4 May 2023).
  105. Shin, H.; Yang, W.; Chay, D.B.; Lee, E.; Chung, J.; Kim, H.; Kim, J. Tetraspanin 1 Promotes Endometriosis Leading to Ovarian Clear Cell Carcinoma. Mol. Oncol. 2021, 15, 987–1004. [Google Scholar] [CrossRef] [PubMed]
  106. CD44—CD44 Antigen—Homo Sapiens (Human)|UniProtKB|UniProt. Available online: https://www.uniprot.org/uniprotkb/P16070/entry (accessed on 4 May 2023).
  107. Yoshida, T.; Matsuda, Y.; Naito, Z.; Ishiwata, T. CD44 in Human Glioma Correlates with Histopathological Grade and Cell Migration. Pathol. Int. 2012, 62, 463–470. [Google Scholar] [CrossRef] [PubMed]
  108. Pazhohan, A.; Amidi, F.; Akbari-Asbagh, F.; Seyedrezazadeh, E.; Aftabi, Y.; Abdolalizadeh, J.; Khodarahmian, M.; Khanlarkhani, N.; Sobhani, A. Expression and Shedding of CD44 in the Endometrium of Women with Endometriosis and Modulating Effects of Vitamin D: A Randomized Exploratory Trial. J. Steroid Biochem. Mol. Biol. 2018, 178, 150–158. [Google Scholar] [CrossRef] [PubMed]
  109. TNC—Tenascin—Homo Sapiens (Human)|UniProtKB|UniProt. Available online: https://www.uniprot.org/uniprotkb/P24821/entry (accessed on 4 May 2023).
  110. Tan, O.; Ornek, T.; Seval, Y.; Sati, L.; Arici, A. Tenascin Is Highly Expressed in Endometriosis and Its Expression Is Upregulated by Estrogen. Fertil. Steril. 2008, 89, 1082–1089. [Google Scholar] [CrossRef]
  111. CTSG—Cathepsin G—Homo Sapiens (Human)|UniProtKB|UniProt. Available online: https://www.uniprot.org/uniprotkb/P08311/entry (accessed on 4 May 2023).
  112. Thorpe, M.; Fu, Z.; Chahal, G.; Akula, S.; Kervinen, J.; de Garavilla, L.; Hellman, L. Extended Cleavage Specificity of Human Neutrophil Cathepsin G: A Low Activity Protease with Dual Chymase and Tryptase-Type Specificities. PLoS ONE 2018, 13, e0195077. [Google Scholar] [CrossRef] [Green Version]
  113. Grzywa, R.; Gorodkiewicz, E.; Burchacka, E.; Lesner, A.; Laudański, P.; Łukaszewski, Z.; Sieńczyk, M. Determination of Cathepsin G in Endometrial Tissue Using a Surface Plasmon Resonance Imaging Biosensor with Tailored Phosphonic Inhibitor. Eur. J. Obstet. Gynecol. Reprod. Biol. 2014, 182, 38–42. [Google Scholar] [CrossRef]
  114. DSP—Desmoplakin—Homo Sapiens (Human)|UniProtKB|UniProt. Available online: https://www.uniprot.org/uniprotkb/P15924/entry (accessed on 4 May 2023).
  115. Baranov, V.; Malysheva, O.; Yarmolinskaya, M. Pathogenomics of Endometriosis Development. Int. J. Mol. Sci. 2018, 19, 1852. [Google Scholar] [CrossRef] [Green Version]
  116. THBS1—Thrombospondin 1—Homo Sapiens (Human)|UniProtKB|UniProt. Available online: https://www.uniprot.org/uniprotkb/A8MZG1/entry (accessed on 4 May 2023).
  117. Manero, M.G.; Olartecoechea, B.; Royo, P.; Alcázar, J.L. Thrombospondin-1 Serum Levels Do Not Correlate with Pelvic Pain in Patients with Ovarian Endometriosis. J. Ovarian Res. 2009, 2, 18. [Google Scholar] [CrossRef] [Green Version]
  118. Tan, X.-J.; Lang, J.-H.; Zheng, W.-M.; Leng, J.-H.; Zhu, L. Ovarian Steroid Hormones Differentially Regulate Thrombospondin-1 Expression in Cultured Endometrial Stromal Cells: Implications for Endometriosis. Fertil. Steril. 2010, 93, 328–331. [Google Scholar] [CrossRef]
  119. PCDH1—Protocadherin-1—Homo Sapiens (Human)|UniProtKB|UniProt. Available online: https://www.uniprot.org/uniprotkb/Q08174/entry (accessed on 4 May 2023).
  120. Li, M.; Xin, X.-Y.; Jin, Z.-S.; Hua, T.; Wang, H.-B.; Wang, H.-B. Transcriptomic Analysis of Stromal Cells from Patients with Endometrial Carcinoma. Int. J. Clin. Exp. Pathol. 2017, 10, 9853–9857. [Google Scholar]
  121. SPARCL1—SPARC-like Protein 1—Homo Sapiens (Human)|UniProtKB|UniProt. Available online: https://www.uniprot.org/uniprotkb/Q14515/entry (accessed on 4 May 2023).
  122. Yusuf, N.; Inagaki, T.; Kusunoki, S.; Okabe, H.; Yamada, I.; Matsumoto, A.; Terao, Y.; Takeda, S.; Kato, K. SPARC Was Overexpressed in Human Endometrial Cancer Stem-like Cells and Promoted Migration Activity. Gynecol. Oncol. 2014, 134, 356–363. [Google Scholar] [CrossRef] [PubMed]
  123. AZGP1—Zinc-Alpha-2-Glycoprotein—Homo Sapiens (Human)|UniProtKB|UniProt. Available online: https://www.uniprot.org/uniprotkb/P25311/entry#function (accessed on 4 May 2023).
  124. Woźny, Ł.A.; Morawiecka-Pietrzak, M.; Jaszczura, M.; Ziora, K.; Grzeszczak, W. The new adipokine zinc-α2-glycoprotein (ZAG) as a link between adipose tissue and kidney? [Czy nowa adipocytokina cynkowa α2-glikoproteina (ZAG) stanowi ogniwo między tkanką tłuszczową a nerkami?]. Endokrynol. Pol. 2019, 70, 171–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Anastasiu, C.V.; Moga, M.A.; Elena Neculau, A.; Bălan, A.; Scârneciu, I.; Dragomir, R.M.; Dull, A.-M.; Chicea, L.-M. Biomarkers for the Noninvasive Diagnosis of Endometriosis: State of the Art and Future Perspectives. Int. J. Mol. Sci. 2020, 21, 1750. [Google Scholar] [CrossRef] [Green Version]
  126. Kovács, Z.; Glover, L.; Reidy, F.; MacSharry, J.; Saldova, R. Novel Diagnostic Options for Endometriosis—Based on the Glycome and Microbiome. J. Adv. Res. 2021, 33, 167–181. [Google Scholar] [CrossRef]
  127. ANXA2—Annexin A2—Homo Sapiens (Human)|UniProtKB|UniProt. Available online: https://www.uniprot.org/uniprotkb/P07355/entry (accessed on 4 May 2023).
  128. Deng, L.; Gao, Y.; Li, X.; Cai, M.; Wang, H.; Zhuang, H.; Tan, M.; Liu, S.; Hao, Y.; Lin, B. Expression and Clinical Significance of Annexin A2 and Human Epididymis Protein 4 in Endometrial Carcinoma. J. Exp. Clin. Cancer Res. 2015, 34, 96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Sansone, A.M.; Hisrich, B.V.; Young, R.B.; Abel, W.F.; Bowens, Z.; Blair, B.B.; Funkhouser, A.T.; Schammel, D.P.; Green, L.J.; Lessey, B.A.; et al. Evaluation of BCL6 and SIRT1 as Non-Invasive Diagnostic Markers of Endometriosis. Curr. Issues Mol. Biol. 2021, 43, 1350–1360. [Google Scholar] [CrossRef]
  130. Jiang, I.; Yong, P.J.; Allaire, C.; Bedaiwy, M.A. Intricate Connections between the Microbiota and Endometriosis. Int. J. Mol. Sci. 2021, 22, 5644. [Google Scholar] [CrossRef]
  131. Qin, R.; Tian, G.; Liu, J.; Cao, L. The Gut Microbiota and Endometriosis: From Pathogenesis to Diagnosis and Treatment. Front. Cell. Infect. Microbiol. 2022, 12, 1069557. [Google Scholar] [CrossRef]
  132. Proestling, K.; Wenzl, R.; Yotova, I.; Hauser, C.; Husslein, H.; Kuessel, L. Investigating Selected Adhesion Molecules as Urinary Biomarkers for Diagnosing Endometriosis. Reprod. BioMedicine Online 2020, 40, 555–558. [Google Scholar] [CrossRef]
  133. Kuessel, L.; Jaeger-Lansky, A.; Pateisky, P.; Rossberg, N.; Schulz, A.; Schmitz, A.A.P.; Staudigl, C.; Wenzl, R. Cytokeratin-19 as a Biomarker in Urine and in Serum for the Diagnosis of Endometriosis–a Prospective Study. Gynecol. Endocrinol. 2014, 30, 38–41. [Google Scholar] [CrossRef]
  134. Chen, X.; Liu, H.; Sun, W.; Guo, Z.; Lang, J. Elevated Urine Histone 4 Levels in Women with Ovarian Endometriosis Revealed by Discovery and Parallel Reaction Monitoring Proteomics. J. Proteom. 2019, 204, 103398. [Google Scholar] [CrossRef] [PubMed]
  135. Cho, S.H.; Oh, Y.J.; Nam, A.; Kim, H.Y.; Park, J.H.; Kim, J.H.; Park, K.H.; Cho, D.J.; Lee, B.S. Evaluation of Serum and Urinary Angiogenic Factors in Patients with Endometriosis. Am. J. Reprod. Immunol. 2007, 58, 497–504. [Google Scholar] [CrossRef] [PubMed]
  136. Tokushige, N.; Markham, R.; Crossett, B.; Ahn, S.B.; Nelaturi, V.L.; Khan, A.; Fraser, I.S. Discovery of a Novel Biomarker in the Urine in Women with Endometriosis. Fertil. Steril. 2011, 95, 46–49. [Google Scholar] [CrossRef] [PubMed]
  137. Vicente-Muñoz, S.; Morcillo, I.; Puchades-Carrasco, L.; Payá, V.; Pellicer, A.; Pineda-Lucena, A. Nuclear Magnetic Resonance Metabolomic Profiling of Urine Provides a Noninvasive Alternative to the Identification of Biomarkers Associated with Endometriosis. Fertil. Steril. 2015, 104, 1202–1209. [Google Scholar] [CrossRef] [Green Version]
  138. Wu, X.; Zhao, C.; Zhang, A.; Zhang, J.; Wang, X.; Sun, X.; Sun, Z.; Wang, X. High-Throughput Metabolomics Used to Identify Potential Therapeutic Targets of Guizhi Fuling Wan against Endometriosis of Cold Coagulation and Blood Stasis. RSC Adv. 2018, 8, 19238–19250. [Google Scholar] [CrossRef] [Green Version]
  139. Othman, E.R.; Markeb, A.A.; Khashbah, M.Y.; Abdelaal, I.I.; ElMelegy, T.T.; Fetih, A.N.; Van der Houwen, L.E.; Lambalk, C.B.; Mijatovic, V. Markers of Local and Systemic Estrogen Metabolism in Endometriosis. Reprod. Sci. 2021, 28, 1001–1011. [Google Scholar] [CrossRef]
  140. Le, N.; Cregger, M.; Brown, V.; Mola, J.L.D.; Bremer, P.; Nguyen, L.; Groesch, K.; Wilson, T.; Diaz-Sylvester, P.; Braundmeier-Fleming, A. Association of Microbial Dynamics with Urinary Estrogens and Estrogen Metabolites in Patients with Endometriosis. PLoS ONE 2021, 16, e0261362. [Google Scholar] [CrossRef]
  141. Ser, H.-L.; Au Yong, S.-J.; Shafiee, M.N.; Mokhtar, N.M.; Ali, R.A.R. Current Updates on the Role of Microbiome in Endometriosis: A Narrative Review. Microorganisms 2023, 11, 360. [Google Scholar] [CrossRef]
  142. Chadchan, S.B.; Naik, S.K.; Popli, P.; Talwar, C.; Putluri, S.; Ambati, C.R.; Lint, M.A.; Kau, A.L.; Stallings, C.L.; Kommagani, R. Gut Microbiota and Microbiota-Derived Metabolites Promotes Endometriosis. Cell Death Discov. 2023, 9, 28. [Google Scholar] [CrossRef]
  143. Colonetti, T.; Saggioratto, M.C.; Grande, A.J.; Colonetti, L.; Junior, J.C.D.; Ceretta, L.B.; Roever, L.; Silva, F.R.; da Rosa, M.I. Gut and Vaginal Microbiota in the Endometriosis: Systematic Review and Meta-Analysis. BioMed Res. Int. 2023, 2023, e2675966. [Google Scholar]
  144. 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] [PubMed] [Green Version]
  145. Green, K.A.; Zarek, S.M.; Catherino, W.H. Gynecologic Health and Disease in Relation to the Microbiome of the Female Reproductive Tract. Fertil. Steril. 2015, 104, 1351–1357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Payne, M.S.; Bayatibojakhi, S. Exploring Preterm Birth as a Polymicrobial Disease: An Overview of the Uterine Microbiome. Front. Immunol. 2014, 5, 595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. 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] [PubMed] [Green Version]
  148. Davies, J. In a Map for Human Life, Count the Microbes, Too. Science 2001, 291, 2316. [Google Scholar] [CrossRef] [PubMed]
  149. Moreno, I.; Simon, C. Deciphering the Effect of Reproductive Tract Microbiota on Human Reproduction. Reprod. Med. Biol. 2019, 18, 40–50. [Google Scholar] [CrossRef] [Green Version]
  150. 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] [Green Version]
  151. Ravel, J.; Gajer, P.; Abdo, Z.; Schneider, G.M.; Koenig, S.S.K.; McCulle, S.L.; Karlebach, S.; Gorle, R.; Russell, J.; Tacket, C.O.; et al. Vaginal Microbiome of Reproductive-Age Women. Proc. Natl. Acad. Sci. USA 2011, 108, 4680–4687. [Google Scholar] [CrossRef] [Green Version]
  152. Ma, B.; Forney, L.J.; Ravel, J. Vaginal Microbiome: Rethinking Health and Disease. Annu. Rev. Microbiol. 2012, 66, 371–389. [Google Scholar] [CrossRef] [Green Version]
  153. Muzny, C.A.; Laniewski, P.; Schwebke, J.R.; Herbst-Kralovetz, M.M. Host–Vaginal Microbiota Interactions in the Pathogenesis of Bacterial Vaginosis. Curr. Opin. Infect. Dis. 2020, 33, 59. [Google Scholar] [CrossRef]
  154. Nunn, K.L.; Clair, G.C.; Adkins, J.N.; Engbrecht, K.; Fillmore, T.; Forney, L.J. Amylases in the Human Vagina. mSphere 2020, 5, e00943-20. [Google Scholar] [CrossRef]
  155. Yarbrough, V.L.; Winkle, S.; Herbst-Kralovetz, M.M. Antimicrobial Peptides in the Female Reproductive Tract: A Critical Component of the Mucosal Immune Barrier with Physiological and Clinical Implications. Hum. Reprod. Update 2015, 21, 353–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Belkaid, Y.; Hand, T.W. Role of the Microbiota in Immunity and Inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Shreiner, A.B.; Kao, J.Y.; Young, V.B. The Gut Microbiome in Health and in Disease. Curr. Opin. Gastroenterol. 2015, 31, 69. [Google Scholar] [CrossRef] [PubMed]
  158. Lobionda, S.; Sittipo, P.; Kwon, H.Y.; Lee, Y.K. The Role of Gut Microbiota in Intestinal Inflammation with Respect to Diet and Extrinsic Stressors. Microorganisms 2019, 7, 271. [Google Scholar] [CrossRef] [Green Version]
  159. Muhleisen, A.L.; Herbst-Kralovetz, M.M. Menopause and the Vaginal Microbiome. Maturitas 2016, 91, 42–50. [Google Scholar] [CrossRef]
  160. Smith, S.B.; Ravel, J. The Vaginal Microbiota, Host Defence and Reproductive Physiology. J. Physiol. 2017, 595, 451–463. [Google Scholar] [CrossRef] [Green Version]
  161. Farage, M.; Maibach, H. Lifetime Changes in the Vulva and Vagina. Arch. Gynecol. Obstet. 2006, 273, 195–202. [Google Scholar] [CrossRef]
  162. Mitchell, C.M.; Haick, A.; Nkwopara, E.; Garcia, R.; Rendi, M.; Agnew, K.; Fredricks, D.N.; Eschenbach, D. Colonization of the Upper Genital Tract by Vaginal Bacterial Species in Nonpregnant Women. Am. J. Obstet. Gynecol. 2015, 212, 611.e1–611.e9. [Google Scholar] [CrossRef] [Green Version]
  163. Brotman, R.M.; Shardell, M.D.; Gajer, P.; Fadrosh, D.; Chang, K.; Silver, M.I.; Viscidi, R.P.; Burke, A.E.; Ravel, J.; Gravitt, P.E. Association between the Vaginal Microbiota, Menopause Status, and Signs of Vulvovaginal Atrophy. Menopause 2014, 21, 450. [Google Scholar] [CrossRef] [Green Version]
  164. Verstraelen, H.; Vilchez-Vargas, R.; Desimpel, F.; Jauregui, R.; Vankeirsbilck, N.; Weyers, S.; Verhelst, R.; Sutter, P.D.; Pieper, D.H.; Wiele, T.V.D. 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]
  165. Winters, A.D.; Romero, R.; Gervasi, M.T.; Gomez-Lopez, N.; Tran, M.R.; Garcia-Flores, V.; Pacora, P.; Jung, E.; Hassan, S.S.; Hsu, C.-D.; et al. Does the Endometrial Cavity Have a Molecular Microbial Signature? Sci. Rep. 2019, 9, 9905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Lu, W.; He, F.; Lin, Z.; Liu, S.; Tang, L.; Huang, Y.; Hu, Z. Dysbiosis of the endometrial microbiota and its association with inflammatory cytokines in endometrial cancer. Int. J. Cancer 2021, 148, 1708–1716. [Google Scholar] [CrossRef] [PubMed]
  167. Baker, J.M.; Chase, D.M.; Herbst-Kralovetz, M.M. Uterine Microbiota: Residents, Tourists, or Invaders? Front. Immunol. 2018, 9, 208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Zervomanolakis, I.; Ott, H.; 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]
  169. Kissler, S.; Hamscho, N.; Zangos, S.; Wiegratz, I.; Schlichter, S.; Menzel, C.; Doebert, N.; Gruenwald, F.; Vogl, T.; Gaetje, R.; et al. Uterotubal Transport Disorder in Adenomyosis and Endometriosis—A Cause for Infertility. BJOG Int. J. Obstet. Gynaecol. 2006, 113, 902–908. [Google Scholar] [CrossRef]
  170. 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]
  171. 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]
  172. 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] [Green Version]
  173. Le, N.; Cregger, M.; Fazleabas, A.; Braundmeier-Fleming, A. Effects of Endometriosis on Immunity and Mucosal Microbial Community Dynamics in Female Olive Baboons. Sci. Rep. 2022, 12, 1590. [Google Scholar] [CrossRef]
  174. Gajer, P.; Brotman, R.M.; Bai, G.; Sakamoto, J.; Schütte, U.M.E.; Zhong, X.; Koenig, S.S.K.; Fu, L.; Ma, Z.; Zhou, X.; et al. Temporal Dynamics of the Human Vaginal Microbiota. Sci. Transl. Med. 2012, 4, 132ra52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Amabebe, E.; Anumba, D.O.C. The Vaginal Microenvironment: The Physiologic Role of Lactobacilli. Front. Med. 2018, 5, 181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. 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]
  177. Hooper, L.V.; Littman, D.R.; Macpherson, A.J. Interactions between the Microbiota and the Immune System. Science 2012, 336, 1268–1273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Spurbeck, R.R.; Arvidson, C.G. Inhibition of Neisseria gonorrhoeae Epithelial Cell Interactions by Vaginal Lactobacillus Species. Infect. Immun. 2008, 76, 3124–3130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. 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]
  180. Li, Z.; Quan, G.; Jiang, X.; Yang, Y.; Ding, X.; Zhang, D.; Wang, X.; Hardwidge, P.R.; Ren, W.; Zhu, G. Effects of metabolites derived from gut microbiota and hosts on pathogens. Front. Cell. Infect. Microbiol. 2018, 8, 314. [Google Scholar] [CrossRef] [Green Version]
  181. Kho, Z.Y.; Lal, S.K. The Human Gut Microbiome—A Potential Controller of Wellness and Disease. Front. Microbiol. 2018, 9, 1835. [Google Scholar] [CrossRef] [Green Version]
  182. Den Besten, G.; van Eunen, K.; Groen, A.K.; Venema, K.; Reijngoud, D.-J.; Bakker, B.M. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 2013, 54, 2325–2340. [Google Scholar] [CrossRef] [Green Version]
  183. Fu, H.; Shi, Y.Q.; Mo, S.J. Effect of short-chain fatty acids on the proliferation and differentiation of the human colonic adenocarcinoma cell line Caco-2. Chin. J. Dig. Dis. 2004, 5, 115–117. [Google Scholar] [CrossRef]
  184. Semaan, J.; El-Hakim, S.; Ibrahim, J.-N.; Safi, R.; Elnar, A.A.; El Boustany, C. Comparative effect of sodium butyrate and sodium propionate on proliferation, cell cycle and apoptosis in human breast cancer cells MCF-7. Breast Cancer 2020, 27, 696–705. [Google Scholar] [CrossRef]
  185. Bindels, L.B.; Porporato, P.; Dewulf, E.M.; Verrax, J.; Neyrinck, A.M.; Martin, J.C.; Scott, K.P.; Calderon, P.B.; Feron, O.; Muccioli, G.G.; et al. Gut microbiota-derived propionate reduces cancer cell proliferation in the liver. Br. J. Cancer 2012, 107, 1337–1344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Maslowski, K.M.; Vieira, A.T.; Ng, A.; Kranich, J.; Sierro, F.; Yu, D.; Schilter, H.C.; Rolph, M.S.; Mackay, F.; Artis, D.; et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 2009, 461, 1282–1286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Parada Venegas, D.; De La Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front. Immunol. 2019, 10, 277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Chen, G.; Ran, X.; Li, B.; Li, Y.; He, D.; Huang, B.; Fu, S.; Liu, J.; Wang, W. Sodium Butyrate Inhibits Inflammation and Maintains Epithelium Barrier Integrity in a TNBS-induced Inflammatory Bowel Disease Mice Model. EBioMedicine 2018, 30, 317–325. [Google Scholar] [CrossRef] [Green Version]
  189. Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly-Y, M.; Glickman, J.N.; Garrett, W.S. The Microbial Metabolites, Short-Chain Fatty Acids, Regulate Colonic Treg Cell Homeostasis. Science 2013, 341, 569–573. [Google Scholar] [CrossRef] [Green Version]
  190. Bhatt, B.; Zeng, P.; Zhu, H.; Sivaprakasam, S.; Li, S.; Xiao, H.; Dong, L.; Shiao, P.; Kolhe, R.; Patel, N.; et al. Gpr109a Limits Microbiota-Induced IL-23 Production to Constrain ILC3-Mediated Colonic Inflammation. J. Immunol. 2018, 200, 2905–2914. [Google Scholar] [CrossRef] [Green Version]
  191. Ang, Z.; Ding, J.L. GPR41 and GPR43 in Obesity and Inflammation—Protective or Causative? Front. Immunol. 2016, 7, 28. [Google Scholar] [CrossRef] [Green Version]
  192. Park, J.; Kim, M.; Kang, S.; Jannasch, A.; Cooper, B.; Patterson, J.; Kim, C. Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR–S6K pathway. Mucosal Immunol. 2014, 8, 80–93. [Google Scholar] [CrossRef] [Green Version]
  193. Licciardi, P.V.; Ververis, K.; Karagiannis, T.C. Histone Deacetylase Inhibition and Dietary Short-Chain Fatty Acids. ISRN Allergy 2011, 2011, 869647. [Google Scholar] [CrossRef] [Green Version]
  194. Chadchan, S.B.; Popli, P.; Ambati, C.R.; Tycksen, E.; Han, S.J.; E Bulun, S.; Putluri, N.; Biest, S.W.; Kommagani, R. Gut microbiota–derived short-chain fatty acids protect against the progression of endometriosis. Life Sci. Alliance 2021, 4, e202101224. [Google Scholar] [CrossRef] [PubMed]
  195. Vallvé-Juanico, J.; Santamaria, X.; Vo, K.C.; Houshdaran, S.; Giudice, L.C. Macrophages Display Proinflammatory Phenotypes in the Eutopic Endometrium of Women with Endometriosis with Relevance to an Infectious Etiology of the Disease. Fertil. Steril. 2019, 112, 1118–1128. [Google Scholar] [CrossRef] [PubMed]
  196. Ni, Z.; Sun, S.; Bi, Y.; Ding, J.; Cheng, W.; Yu, J.; Zhou, L.; Li, M.; Yu, C. Correlation of Fecal Metabolomics and Gut Microbiota in Mice with Endometriosis. Am. J. Reprod. Immunol. 2020, 84, e13307. [Google Scholar] [CrossRef] [PubMed]
  197. Meroni, M.; Longo, M.; Dongiovanni, P. Alcohol or Gut Microbiota: Who Is the Guilty? Int. J. Mol. Sci. 2019, 20, 4568. [Google Scholar] [CrossRef] [Green Version]
  198. Harada, T.; Iwabe, T.; Terakawa, N. Role of Cytokines in Endometriosis. Fertil. Steril. 2001, 76, 1–10. [Google Scholar] [CrossRef]
  199. Nothnick, W.B. Treating Endometriosis as an Autoimmune Disease. Fertil. Steril. 2001, 76, 223–231. [Google Scholar] [CrossRef]
  200. Wanderley, C.W.; Colón, D.F.; Luiz, J.P.M.; Oliveira, F.F.; Viacava, P.R.; Leite, C.A.; Pereira, J.A.; Silva, C.M.; Silva, C.R.; Silva, R.L.; et al. Paclitaxel Reduces Tumor Growth by Reprogramming Tumor-Associated Macrophages to an M1 Profile in a TLR4-Dependent Manner. Cancer Res. 2018, 78, 5891–5900. [Google Scholar] [CrossRef] [Green Version]
  201. Dewhirst, F.E.; Chen, T.; Izard, J.; Paster, B.J.; Tanner, A.C.R.; Yu, W.-H.; Lakshmanan, A.; Wade, W.G. The Human Oral Microbiome. J. Bacteriol. 2010, 192, 5002–5017. [Google Scholar] [CrossRef] [Green Version]
  202. Paster, B.J.; Boches, S.K.; Galvin, J.L.; Ericson, R.E.; Lau, C.N.; Levanos, V.A.; Sahasrabudhe, A.; Dewhirst, F.E. Bacterial Diversity in Human Subgingival Plaque. J. Bacteriol. 2001, 183, 3770–3783. [Google Scholar] [CrossRef] [Green Version]
  203. Zaura, E.; Keijser, B.J.; Huse, S.M.; Crielaard, W. Defining the Healthy “Core Microbiome” of Oral Microbial Communities. BMC Microbiol. 2009, 9, 259. [Google Scholar] [CrossRef] [Green Version]
  204. Segata, N.; Haake, S.K.; Mannon, P.; Lemon, K.P.; Waldron, L.; Gevers, D.; Huttenhower, C.; Izard, J. Composition of the Adult Digestive Tract Bacterial Microbiome Based on Seven Mouth Surfaces, Tonsils, Throat and Stool Samples. Genome Biol. 2012, 13, R42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Yang, F.; Zeng, X.; Ning, K.; Liu, K.-L.; Lo, C.-C.; Wang, W.; Chen, J.; Wang, D.; Huang, R.; Chang, X.; et al. Saliva Microbiomes Distinguish Caries-Active from Healthy Human Populations. ISME J. 2012, 6, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. He, J.; Li, Y.; Cao, Y.; Xue, J.; Zhou, X. The Oral Microbiome Diversity and Its Relation to Human Diseases. Folia Microbiol. 2015, 60, 69–80. [Google Scholar] [CrossRef] [PubMed]
  207. Shanahan, F. The Microbiota in Inflammatory Bowel Disease: Friend, Bystander, and Sometime-Villain. Nutr. Rev. 2012, 70, S31–S37. [Google Scholar] [CrossRef]
  208. Docktor, M.J.; Paster, B.J.; Abramowicz, S.; Ingram, J.; Wang, Y.E.; Correll, M.; Jiang, H.; Cotton, S.L.; Kokaras, A.S.; Bousvaros, A. Alterations in Diversity of the Oral Microbiome in Pediatric Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2012, 18, 935–942. [Google Scholar] [CrossRef]
  209. Seoudi, N.; Bergmeier, L.A.; Drobniewski, F.; Paster, B.; Fortune, F. The Oral Mucosal and Salivary Microbial Community of Behçet’s Syndrome and Recurrent Aphthous Stomatitis. J. Oral Microbiol. 2015, 7, 27150. [Google Scholar] [CrossRef] [Green Version]
  210. Slocum, C.; Kramer, C.; Genco, C.A. Immune Dysregulation Mediated by the Oral Microbiome: Potential Link to Chronic Inflammation and Atherosclerosis. J. Intern. Med. 2016, 280, 114–128. [Google Scholar] [CrossRef]
  211. Zhang, X.; Zhang, D.; Jia, H.; Feng, Q.; Wang, D.; Liang, D.; Wu, X.; Li, J.; Tang, L.; Li, Y.; et al. The Oral and Gut Microbiomes Are Perturbed in Rheumatoid Arthritis and Partly Normalized after Treatment. Nat. Med. 2015, 21, 895–905. [Google Scholar] [CrossRef]
  212. Abusleme, L.; Dupuy, A.K.; Dutzan, N.; Silva, N.; Burleson, J.A.; Strausbaugh, L.D.; Gamonal, J.; Diaz, P.I. The Subgingival Microbiome in Health and Periodontitis and Its Relationship with Community Biomass and Inflammation. ISME J. 2013, 7, 1016–1025. [Google Scholar] [CrossRef] [Green Version]
  213. Shaik-Dasthagirisaheb, Y.B.; Huang, N.; Gibson, F.C. Inflammatory Response to Porphyromonas Gingivalis Partially Requires Interferon Regulatory Factor (IRF) 3. Innate Immun. 2014, 20, 312–319. [Google Scholar] [CrossRef] [Green Version]
  214. Eyster, K.M. The Estrogen Receptors: An Overview from Different Perspectives. Estrogen Recept. Methods Protoc. 2016, 1366, 1–10. [Google Scholar] [CrossRef]
  215. Chantalat, E.; Valera, M.-C.; Vaysse, C.; Noirrit, E.; Rusidze, M.; Weyl, A.; Vergriete, K.; Buscail, E.; Lluel, P.; Fontaine, C.; et al. Estrogen Receptors and Endometriosis. Int. J. Mol. Sci. 2020, 21, 2815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Laux-Biehlmann, A.; d’Hooghe, T.; Zollner, T.M. Menstruation Pulls the Trigger for Inflammation and Pain in Endometriosis. Trends Pharmacol. Sci. 2015, 36, 270–276. [Google Scholar] [CrossRef] [PubMed]
  217. Galvankar, M.; Singh, N.; Modi, D. Estrogen Is Essential but Not Sufficient to Induce Endometriosis. J. Biosci. 2017, 42, 251–263. [Google Scholar] [CrossRef] [PubMed]
  218. Zhang, Q.; Shen, Q.; Celestino, J.; Milam, M.R.; Westin, S.N.; Lacour, R.A.; Meyer, L.A.; Shipley, G.L.; Davies, P.J.A.; Deng, L.; et al. Enhanced Estrogen-Induced Proliferation in Obese Rat Endometrium. Am. J. Obstet. Gynecol. 2009, 200, 186.e1–186.e8. [Google Scholar] [CrossRef] [Green Version]
  219. Reis, F.M.; Petraglia, F.; Taylor, R.N. Endometriosis: Hormone Regulation and Clinical Consequences of Chemotaxis and Apoptosis. Hum. Reprod. Update 2013, 19, 406–418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. van Weelden, W.J.; Massuger, L.F.A.G.; Enitec; Pijnenborg, J.M.A.; Romano, A. Anti-Estrogen Treatment in Endometrial Cancer: A Systematic Review. Front. Oncol. 2019, 9, 359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. Rodriguez, A.C.; Blanchard, Z.; Maurer, K.A.; Gertz, J. Estrogen Signaling in Endometrial Cancer: A Key Oncogenic Pathway with Several Open Questions. Horm. Cancer 2019, 10, 51–63. [Google Scholar] [CrossRef] [Green Version]
  222. Palmisano, B.T.; Zhu, L.; Stafford, J.M. Role of Estrogens in the Regulation of Liver Lipid Metabolism. In Sex and Gender Factors Affecting Metabolic Homeostasis, Diabetes and Obesity; Mauvais-Jarvis, F., Ed.; Advances in Experimental Medicine and Biology; Springer International Publishing: Cham, Switzerland, 2017; pp. 227–256. ISBN 978-3-319-70178-3. [Google Scholar]
  223. Parida, S.; Sharma, D. The Microbiome–Estrogen Connection and Breast Cancer Risk. Cells 2019, 8, 1642. [Google Scholar] [CrossRef] [Green Version]
  224. The Gut Microbiome and the Estrobolome: How Gut Microbes Affect Estrogen Metabolism—Vibrant Wellness. Available online: https://www.vibrant-wellness.com/the-gut-microbiome-and-the-estrobolome-how-gut-microbes-affect-estrogen-metabolism/ (accessed on 4 May 2023).
  225. Sui, Y.; Wu, J.; Chen, J. The Role of Gut Microbial β-Glucuronidase in Estrogen Reactivation and Breast Cancer. Front. Cell Dev. Biol. 2021, 9, 631552. [Google Scholar] [CrossRef]
  226. Baker, J.M.; Al-Nakkash, L.; Herbst-Kralovetz, M.M. Estrogen–Gut Microbiome Axis: Physiological and Clinical Implications. Maturitas 2017, 103, 45–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Ervin, S.M.; Li, H.; Lim, L.; Roberts, L.R.; Liang, X.; Mani, S.; Redinbo, M.R. Gut Microbial β-Glucuronidases Reactivate Estrogens as Components of the Estrobolome That Reactivate Estrogens. J. Biol. Chem. 2019, 294, 18586–18599. [Google Scholar] [CrossRef] [PubMed]
  228. Flores, R.; Shi, J.; Fuhrman, B.; Xu, X.; Veenstra, T.D.; Gail, M.H.; Gajer, P.; Ravel, J.; Goedert, J.J. Fecal Microbial Determinants of Fecal and Systemic Estrogens and Estrogen Metabolites: A Cross-Sectional Study. J. Transl. Med. 2012, 10, 253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  229. Looijer-van Langen, M.; Hotte, N.; Dieleman, L.A.; Albert, E.; Mulder, C.; Madsen, K.L. Estrogen Receptor-β Signaling Modulates Epithelial Barrier Function. Am. J. Physiol. Gastrointest. Liver Physiol. 2011, 300, G621–G626. [Google Scholar] [CrossRef]
  230. 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]
  231. Beaud, D.; Tailliez, P.; Anba-Mondoloni, J. Genetic Characterization of the β-Glucuronidase Enzyme from a Human Intestinal Bacterium, Ruminococcus Gnavus. Microbiology 2005, 151, 2323–2330. [Google Scholar] [CrossRef]
  232. Kitawaki, J.; Kado, N.; Ishihara, H.; Koshiba, H.; Kitaoka, Y.; Honjo, H. Endometriosis: The Pathophysiology as an Estrogen-Dependent Disease. J. Steroid Biochem. Mol. Biol. 2002, 83, 149–155. [Google Scholar] [CrossRef]
  233. Laschke, M.W.; Menger, M.D. The Gut Microbiota: A Puppet Master in the Pathogenesis of Endometriosis? Am. J. Obstet. Gynecol. 2016, 215, 68.e1–68.e4. [Google Scholar] [CrossRef]
  234. Hakansson, A.; Molin, G. Gut Microbiota and Inflammation. Nutrients 2011, 3, 637–682. [Google Scholar] [CrossRef] [Green Version]
  235. Morotti, M.; Vincent, K.; Becker, C.M. Mechanisms of Pain in Endometriosis. Eur. J. Obstet. Gynecol. Reprod. Biol. 2017, 209, 8–13. [Google Scholar] [CrossRef]
  236. Symons, L.K.; Miller, J.E.; Kay, V.R.; Marks, R.M.; Liblik, K.; Koti, M.; Tayade, C. The Immunopathophysiology of Endometriosis. Trends Mol. Med. 2018, 24, 748–762. [Google Scholar] [CrossRef] [PubMed]
  237. Wang, Y.; Nicholes, K.; Shih, I.-M. The Origin and Pathogenesis of Endometriosis. Annu. Rev. Pathol. Mech. Dis. 2020, 15, 71–95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Bodke, H.; Jogdand, S. Role of Probiotics in Human Health. Cureus 2022, 14, e31313. [Google Scholar] [CrossRef]
  240. George Kerry, R.; Patra, J.K.; Gouda, S.; Park, Y.; Shin, H.-S.; Das, G. Benefaction of Probiotics for Human Health: A Review. J. Food Drug Anal. 2018, 26, 927–939. [Google Scholar] [CrossRef] [Green Version]
  241. Hashem, N.M.; Gonzalez-Bulnes, A. The Use of Probiotics for Management and Improvement of Reproductive Eubiosis and Function. Nutrients 2022, 14, 902. [Google Scholar] [CrossRef]
  242. 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]
  243. 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] [Green Version]
  244. 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]
  245. Davani-Davari, D.; Negahdaripour, M.; Karimzadeh, I.; Seifan, M.; Mohkam, M.; Masoumi, S.J.; Berenjian, A.; Ghasemi, Y. Prebiotics: Definition, Types, Sources, Mechanisms, and Clinical Applications. Foods 2019, 8, 92. [Google Scholar] [CrossRef] [Green Version]
  246. You, S.; Ma, Y.; Yan, B.; Pei, W.; Wu, Q.; Ding, C.; Huang, C. The Promotion Mechanism of Prebiotics for Probiotics: A Review. Front. Nutr. 2022, 9, 2223. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Microbiological differentiation of female reproductive tracts in various physiological conditions; based on [160,161,162]. The age frames of girls in particular stages of development are an individual feature; however, in order to standardize the changes in the composition and number of microorganisms of the reproductive tract, let us assume that the period of infancy and childhood applies to girls up to the age of 8; early puberty is 8–13 years old; late puberty is 14–17 years old; reproductive age is 18–40 years; and postmenopause covers women over the age of 55. In addition, in the diagram, the microorganisms are listed in descending order of predominance.
Figure 1. Microbiological differentiation of female reproductive tracts in various physiological conditions; based on [160,161,162]. The age frames of girls in particular stages of development are an individual feature; however, in order to standardize the changes in the composition and number of microorganisms of the reproductive tract, let us assume that the period of infancy and childhood applies to girls up to the age of 8; early puberty is 8–13 years old; late puberty is 14–17 years old; reproductive age is 18–40 years; and postmenopause covers women over the age of 55. In addition, in the diagram, the microorganisms are listed in descending order of predominance.
Ijms 24 10920 g001
Figure 2. The interaction of bacteria with the endometrial epithelium. Changes in the vaginal and intestinal diversity of the microbiome resulted in an immunological shift towards an inflammatory profile, which may promote inflammation by producing inflammatory mediators; based on [174].
Figure 2. The interaction of bacteria with the endometrial epithelium. Changes in the vaginal and intestinal diversity of the microbiome resulted in an immunological shift towards an inflammatory profile, which may promote inflammation by producing inflammatory mediators; based on [174].
Ijms 24 10920 g002
Figure 3. Metabolism of estrogen in the human body; based on [224,225].
Figure 3. Metabolism of estrogen in the human body; based on [224,225].
Ijms 24 10920 g003
Table 1. Characteristics of selected proteins with biomarker molecule potential found in the urine of patients with endometriosis.
Table 1. Characteristics of selected proteins with biomarker molecule potential found in the urine of patients with endometriosis.
GeneName of ProteinProtein IDAmino
Acids
Mass
(kDa)
FunctionReferences
ENGEndoglinP1781365870.578
  • Vascular endothelium glycoprotein that plays an important role in the regulation of angiogenesis;
  • Acts as TGF-beta coreceptor and is involved in the TGF-beta/BMP signaling cascade that ultimately leads to the activation of SMAD transcription factors;
  • ENG was significantly higher in the ectopic endometriotic tissues than that in eutopic endometriotic tissue.
[98,99,100,101]
LUMLumicanP5188433838.429
  • Lumican demonstrated higher immunoreactivity and relative expression in the endometrium of women with PCOS compared to that of women with regular menstrual cycles.
[102,103]
TGFB2Transforming Growth Factor Beta Receptor 2P3717356764.568
  • Transmembrane serine/threonine kinase forming with the TGF-beta type I serine/threonine kinase receptor, TGFBR1, the non-promiscuous receptor for the TGF-beta cytokines TGFB1, TGFB2 and TGFB3.
[88,104]
TSPAN1Tetraspanin-1O6063524126.301
  • SPAN1 overexpression enhanced cell growth and invasion of ovarian clear cell carcinoma;
  • TSPAN1 promoted endometriotic cell growth through AMPK activity.
[105,106]
CD44CD44 AntigenP1607074281.538
  • CD44 plays a role in cell–cell interactions, cell adhesion and migration, helping them to sense and respond to changes in the tissue microenvironment;
  • The concentration of soluble CD44 in the serum and endometrial fluid of endometriosis patients was higher than in healthy women.
[107,108,109]
TNCTenascinP248212201240.853
  • Tenascin is a high-molecular-weight ECM protein and plays a critical role in tissue regeneration, hyperplastic and neoplastic processes;
  • The modulation of tenascin as an extracellular matrix protein by E(2) in endometriotic stromal cells may be one of the factors playing a role in the development of endometriosis.
[110,111]
CatGCathepsin GP0831125528.837
  • Serine protease with trypsin- and chymotrypsin-like specificity;
  • The level of cathepsin G ascertained in endometrium tissue samples was over twice as high for the group of patients suffering from endometriosis as compared to the control group.
[112,113,114]
DSPDesmoplakinP159242871331.774
  • Major high-molecular-weight protein of desmosomes;
  • Regulates profibrotic gene expression in cardiomyocytes via activation of the MAPK14/p38 MAPK signaling cascade and increase in TGFB1 protein abundance.
[115,116]
THBS1Thrombospondin 1A8MZG19410.053
  • Thrombospondin-1 serum levels correlate with pelvic pain in patients with ovarian endometriosis.
[117,118,119]
PCDH1Protocadherin-1Q081741060114.743
  • Involved in cell–cell interaction processes and in cell adhesion;
  • PCDHs mediate cell growth, cell cycle arrest, apoptosis and migration of endometrial cancer, underlining their critical roles in endometrial carcinogenesis.
[120,121]
SPARCL1SPARC-like Protein 1Q1451566475.208
  • SPARC enhanced fibronectin expression and promoted migration activity;
  • SPARC was expressed in endometrial cancer tissues.
[122,123]
AZGP1Zinc-alpha-2-glycoproteinP2531129834.259
  • ZAG activity is controlled by hormones, the immune system and, interestingly, polyunsaturated fats and essential fatty acids;
  • Elevated ZAG may not be the most specific biomarker for endometriosis, but it may point to other mechanisms potentially more diagnostic.
[124,125,126,127]
ANXA2Annexin A2P0735533938.604
  • Calcium-regulated membrane-binding protein whose affinity for calcium is greatly enhanced by anionic phospholipids;
  • ANXA2 inhibition abrogated endometrial tissue growth, metastasis and angiogenesis in an adenomyosis nude mice model and significantly alleviated hyperalgesia.
[96,128]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sobstyl, A.; Chałupnik, A.; Mertowska, P.; Grywalska, E. How Do Microorganisms Influence the Development of Endometriosis? Participation of Genital, Intestinal and Oral Microbiota in Metabolic Regulation and Immunopathogenesis of Endometriosis. Int. J. Mol. Sci. 2023, 24, 10920. https://doi.org/10.3390/ijms241310920

AMA Style

Sobstyl A, Chałupnik A, Mertowska P, Grywalska E. How Do Microorganisms Influence the Development of Endometriosis? Participation of Genital, Intestinal and Oral Microbiota in Metabolic Regulation and Immunopathogenesis of Endometriosis. International Journal of Molecular Sciences. 2023; 24(13):10920. https://doi.org/10.3390/ijms241310920

Chicago/Turabian Style

Sobstyl, Anna, Aleksandra Chałupnik, Paulina Mertowska, and Ewelina Grywalska. 2023. "How Do Microorganisms Influence the Development of Endometriosis? Participation of Genital, Intestinal and Oral Microbiota in Metabolic Regulation and Immunopathogenesis of Endometriosis" International Journal of Molecular Sciences 24, no. 13: 10920. https://doi.org/10.3390/ijms241310920

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