Next Article in Journal
Comprehensive Assessment of Medial Knee Joint Instability by Valgus Stress MRI
Previous Article in Journal
Diagnostic Performance of Electromagnetic Navigation Bronchoscopy-Guided Biopsy for Lung Nodules in the Era of Molecular Testing
Previous Article in Special Issue
Transvaginal Strain Elastosonography May Help in the Differential Diagnosis of Endometriosis?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular and Clinical Insights on the Complex Interaction between Oxidative Stress, Apoptosis, and Endobiota in the Pathogenesis of Endometriosis

1
Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no 16, 700115 Iasi, Romania
2
Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street, no 34, 700038 Iasi, Romania
3
Origyn Fertility Center, Palace Street, no 3C, 700032 Iasi, Romania
4
Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, no 20A, 700505 Iasi, Romania
5
Department of Interdisciplinary Research in Science, “Alexandru Ioan Cuza” University, Carol I Avenue, no 11, 700107 Iasi, Romania
*
Authors to whom correspondence should be addressed.
Diagnostics 2021, 11(8), 1434; https://doi.org/10.3390/diagnostics11081434
Submission received: 28 June 2021 / Revised: 5 August 2021 / Accepted: 6 August 2021 / Published: 9 August 2021
(This article belongs to the Collection Diagnosis of Endometriosis: Biomarkers and Clinical Methods)

Abstract

:
Endometriosis (EMS) remains, to date, an intriguing and debilitating gynecological disorder that possesses a multifactorial substrate. Recent studies with the objective of elucidating its etiology highlighted the antagonistic effect of EMS on a multiple of processes involved in homeostasis. Although the current oxidative biomarkers clearly reveal the consequences induced by EMS, its implication in the associated inflammatory reactions could be much more complex. Besides the overproduction of reactive oxygen species (ROS) that leads to an exacerbated oxidative response, it also changes the normal expression of several pro-inflammatory modulators, reflected by the fluctuating activity of several pro- and anti-apoptotic mediators whose expression is impaired. In light of this topic, several studies elucidate the involvement of apoptosis in EMS, being brought controversial findings, even reports with no significant change. Further, some authors reported an abnormal expression of multiple genes that are crucial for the overall functionality of the female reproductive system. Cumulatively, it seems that the subsequent oxidative imbalance and apoptosis process impairment could further disrupt the normal removal of unnecessary biological products. Based on all gathered evidence, we could argue that the related stress state could determine human endobiota impairment, which could further participate in the inflammatory and main antioxidant enzyme changes occurring in EMS. Moreover, a correlation between endobiota integrity, inflammation, and oxidative stress (OS) was suggested in relation to the possible predisposition to pathogen determined infections.

1. Introduction

EMS is a common and enigmatic hormone-dependent gynecological disorder clinically described first in the 1920s. Even though more than one hundred years have passed since it was documented for the first time, its etiology still raises debate and controversy [1]. Clinically, EMS is defined as the presence of vascularized endometrial-like glandular tissue dispersed at ectopic sites and stroma outside the uterus. Despite the occurrence of these lesions within the peritoneal cavity (bladder, ovaries, colon, or peritoneum wall), the latest reports indicate the presence of such structures in the liver, lung, and brain as well [2,3].
Left untreated, EMS causes severe primary dysmenorrhea, chronic pelvic pain, dyspareunia, and infertility [4]. According to the latest issued figures, more than one hundred million women of reproductive age suffer from EMS (between 5–15% females worldwide). Approximately 90% report menstrual reflux, while 5–10% develop EMS [5].
It has been theorized to originate from endometrial fragments shed during menstruation. These lesions reach the peritoneal cavity via the Fallopian tubes, a process that is known as “retrograde menstruation” [6]. However, of the multitude of hypotheses that have been proposed in recent years—immune and stem cell dysfunction, embryonic rest, and coelomic metaplasia [7,8,9]—Sampson’s theory of retrograde menstruation remains the most solid argument of the origin of EMS to date [2,6,10,11].
Halme et al. suggested that most of the women display some degree of reflux of endometrial debris [12]. This argument is supported by the cumulative evidence according to which menstrual effluents could retrogradely scatter into the peritoneal cavity carrying viable endometrial cells [13,14,15,16,17].
Besides the standard clinical panel, EMS-diagnosed women are also at higher risk of developing autoimmune, cardiovascular, or gastrointestinal diseases and cancer [18]. From a mechanistical point of view, EMS could impair the apoptotic process, which could explain the abnormal cytokine storm and abnormal oxidative stress (OS) profile [19].
A disruption of the internal microenvironment could subsequently cause microbiome dysbiosis (e.g., pro-inflammatory cascades). With respect to the plethora of roles of the microbiome in various pathologies [20], endobiota could be the missing link. In this regard, Khan et al. 2018 discussed the new hypothesis known as “bacterial contamination” [21].
Considering the complex mechanisms underlying EMS development and the multiple hypotheses of pathological processes leading to EMS occurrence, we aimed to evaluate the possible relevance of OS, apoptosis, and dysbiosis in EMS. In this way, it is our goal to describe several mechanisms that can elucidate the outcomes and correlation of these particular processes.

2. Materials and Methods

For this work, literature searches were conducted independently using the main scientific databases (Web of Science, Scopus, Google Scholar, Science Direct, Cochrane Database for Systematic Reviews, PubMed/Medline). The initial information acquisition was based on database search using keywords such as “endometriosis”, “oxidative stress”, “apoptosis”, “endobiota”, “vaginal microbiota”, “cervical microbiota”, “gut microbiota”, “animal models”, and “human/patients” in all the relevant combinations. In the second step of selection, all the relevant literature up until June 2021 was categorized based on title, abstract content, and full content, and only the English written articles were further considered. Eight authors (O.-D.I., I.-M.B., R.M., I.S., G.S., D.G. and I.S.) independently inquired the information, with any differences in opinion being solved by common consent with the remaining three authors (B.D., A.C. and C.I.).
Based on all studies selected, in two tables (Table 1, Table 2 and Table 3), we present all main features that define each study.

3. Oxidative Stress and EMS

Oxidative Stress and EMS

The implication of OS in EMS was previously described by several studies, which both addressed the screening of OS markers in EMS patients and mechanistical approaches in EMS animal models. In this way, the screening studies showed that several important oxidative markers are modified in EMS patients suggesting that OS could be an important component underlying and/or promoting EMS tissular damage. However, since correlation could not always imply causation, the way in which OS and EMS interact was not yet fully understood.
For instance, several studies showed that the typical OS markers changes suggest that EMS’s antioxidant defense could be impaired. Amreen et al., [22] reported that both superoxide dismutase (SOD) and glutathione peroxidase (GPx) antioxidant activities are significantly decreased in the blood and peritoneal fluid of EMS patients, while Turkyilmaz et al. [23] also found catalase (CAT) activity increased in the serum of 31 EMS-diagnosed women. Moreover, they showed that the pattern in which antioxidant enzymes activity varied is in direct correlation to EMS severity. Furthermore, it was demonstrated that non-enzymatic antioxidants, such as vitamin C, vitamin E, and native and total thiols, are significantly less present in the blood of EMS patients [23,24].
Regarding the effects of ROS production and accumulation in EMS, it was described that the decrease of antioxidant defense could further lead to significant molecular and tissular damage. It was shown that OS could have negative implications on proteins, lipids, and DNA [25]. While Alizadeh et al. [26] found no significant increase in lipid peroxidation (malondialdehyde—MDA) and protein peroxidation markers (carbonyls) in the blood of EMS patients, nor did Elsharkawy et al. [24] in the blood and follicular fluid (FF) of infertile EMS-diagnosed women, other studies bring relevant evidence of molecular damage following oxidative balance impairment. Thus, Nasiri et al. [27] recently evaluated lipid peroxides (LPO) and total antioxidant capacity (TAC) in the serum and FF of women with EMS and reported significantly increased LPO, concomitant with decreased TAC in both fluids, as compared to healthy control women.
However, a correlation between systemic and peritoneal oxidative biomarkers could not be established following Montoya-Estrada et al.’s [28] study in which increased levels of carbonyls and LPO were found in EMS patients peritoneal fluid, while other protein oxidation product (protein dityrosine) levels were increased in the blood, as compared to the control group. Despite the fact that no correlation was found between the hemoglobin content of the peritoneal fluid and carbonyls and LPO and between the OS markers from blood and peritoneal fluid, the study reported a significant increase of MDA levels in the latter, suggesting that, together with the ischemia-modified albumin levels increase, oxidative balance impairment could play a determinant role in EMS-related tissular damage and a possible correlation in the co-occurring pro-inflammatory processes. In this regard, it was shown that several inflammation-related molecules are also regulated dependently or independently of ROS production and accumulation. Thézénas et al. [29] identified amine oxidase 3 and vascular adhesion protein 1 (AOC3/VAP1), in particular alkenal reductase PTGR1 and methionine sulfoxide reductase, as candidates for altered ROS landscape in EMS and possibly influencing the systemic and/or local inflammatory response. Furthermore, they observed important molecular changes in the ectopic lesion (such as iron overload, increased ROS production, and lipid peroxidation) and noted that ROS-derived 4-hydroxy-2-nonenal (4-HNE) over-production led to monocytic interleukin IL-8 release.
The implication of iron in the OS modulation occurring in EMS was also previously suggested on several occasions. In this way, Alizadeh et al. [26] showed that serum iron levels in patients with EMS were significantly increased and suggested that this metal ions could be a potent promoter of OS in EMS. The accumulation of iron ions was also reported by Polak et al. [30] while studying the overall oxidative status of peritoneal fluid of EMS patients suggesting that iron metabolism could be disrupted in this disease. Furthermore, since iron could catalyze many ROS species production and was also implicated in cellular dysfunction, necrosis, and apoptosis, Chen, Hayashi, and co-authors [31,32] investigated the mitochondrial structure and function of endometrial stromal cells (ESCs) in ovarian endometriosis (OE) in relation to mitochondrial and iron metabolism. They found that in the ESCs, mitochondria generated more ROS, while SOD2 was highly expressed in the endometrium, suggesting an abnormal energetic metabolism of ectopic ESCs. SOD2 promoted two distinct processes: Cell proliferation and migration in OE. Furthermore, in an OE murine model, concomitant with the iron accumulation and proteins and DNA alterations due to OS, as evaluated through the levels of 4-NHE and 8-hydroxy-2′-deoxyguanosine (8-OHdG), specific lesions were observed in the intestine, pancreas, and peritoneal wall, and time-dependent ovarian fibrosis development. Further evaluation of fibrotic tissue development in EMS in correlation with OS occurrence was conducted by González-Foruria et al. [33] who described the possible mechanism in which OS may also be implicated in ADAM17/Notch signaling pathway modulation culminating with fibrosis development. They showed that in deep infiltrating endometriosis (DIE), a direct correlation was observed between ADAM17 protein levels and increased advanced oxidation protein products (AOPPs) in the ectopic lesions, as well as the fact that the expression of Notch was abnormal for type-I collagen, which suggested fibrosis development.
Table 1. Summary content regarding the outcomes in which we investigated the oxidative and potential biomarkers.
Table 1. Summary content regarding the outcomes in which we investigated the oxidative and potential biomarkers.
SubjectNumberType of
Sample
Biomarker of
Interest
Reference
Female18 EMS patientsEctopic and
eutopic
samples
Novel OS biomarker-AOC3/VAP1,
alkenal reductase PTGR1 and methionine sulfoxidereductase
[29]
Female133 women; 40
ectopic and 73 eutopic
Ectopic ESCsEctopic ESC ROS ↑
SOD2 ↑
[31]
Female55 EMS patients out of 64Blood and
peritoneal fluid
SOD ↓
GPx ↓
[22]
Female28 EMS patients and 23 controlsPlasma and
peritoneal fluid
Carbonyl ↑
LPO ↑
[28]
Female121 EMS patients and 81 controlsPeritoneal fluidAOPP ↑
ADAM17 ↑
Notch ↑
[33]
Female110 EMS patients and 119 with benign ovarian cystsPeritoneal fluidHaemoglobin ↑
Iron ↑
TOS ↑
TAS ↓
[30]
Female30 EMS patients and 30 controlsFollicular fluid and serumVitamin C ↓
Vitamin E ↓
SOD ↑
MDA ≠
[24]
Female63 EMS patientsFollicular fluid and serumLPO ↑TAC ↑[27]
Female31 EMS patients and 27 controlsSerumSerum native thiols ↓
Total native thiols ↓
CAT ↑
[23]
Female30 EMS patients and 40 controlsSerumIron ↑
MDA ≠
Carbonyl ≠
[26]
Murine modelIron ↑
FSHR ↓
[32]
↑ increased. ↓ decreased. ≠ no significant change.

4. Apoptosis and EMS

Apoptosis is a distinctive form of programmed cell death that provides efficient elimination of senescent cells and tissue homeostasis during the menstrual cycle without eliciting inflammatory reactions. While EMS’s severity proved to be directly correlated with a fulminant production of ROS, this pathology also exerts antagonistic effects towards the functionality of apoptosis. Under normal circumstances, apoptosis usually prevents the migration and accumulation of ectopic and eutopic endometrial cells by destroying them before they have the opportunity to form necrotic tissue [34,35].
Several recent studies thoroughly described this correlation and explained the mechanisms in which endometrial cells apoptosis is modulated. On the one hand, it was shown that one of the most important apoptotic pathways in EMS endometrium is the Bcl-2 family-mediated mechanism. Moreover, in an EMS animal model study, Mulyantoro et al. [36] reported the effects of Bcl-2/Bax modulation on the abdominal and pelvic peritoneal tissues EMS implants concluding that the fate of the endometrial implants in the peritoneal tissues could be directly correlated to the Bcl-2/Bax expression. Moreover, Delbandi et al. [37] showed that there could be several differences in this mechanism’s modulation considering the origin of the EMS cells (ectopic versus eutopic). In this way, it seems that the anti-apoptotic signaling molecules (Bcl-2 and Bcl-xL) are expressed with a higher frequency in the ectopic EMS tissues suggesting that the apoptotic signals modulation could be implicated in the fate of the EMS cells. Furthermore, the study also reported the implication of lower caspase-3 pro-apoptotic activity (as an important component of Fas apoptotic pathway) in the eutopic EMS tissues implying that EMS development could be a matter of extrinsic and intrinsic apoptosis modulation. However, the authors suggested that the Fas apoptotic pathway could be implicated in the EMS preventive mechanism induced by immune activation, thus showing an important correlation between apoptosis modulation and inflammatory response.
In this context, Li et al. [38] showed that the upregulation of caspase-3 expression is concomitant with and correlated to the expression of the signal transducer and activator of transcription 1 (STAT1), which is also an important signal molecule in inflammatory response modulation. In addition, in a study of the potential beneficial effect of Kuntai Capsule (KTC) on apoptosis, Zhong et al. [39] showed that the modulation of caspase-8, caspase-9, caspase-3, and cytochrome could promote apoptosis in the EMS tissues. However, the authors pointed that the positive effect of Kuntai Capsule formulation could be effective only on the ectopic EMS tissues, clearly suggesting the differences between the mechanisms underlying ectopic and eutopic apoptosis. Moreover, a similar study pointed out that OS could also be implicated in this equation, since a correlation (yet not causation) could be established between the increased level of serum ROS and the decreased antioxidant enzymes activity (namely, SOD and CAT) and apoptosis rate, Bax, Bcl-2, and caspase-3 expression. Furthermore, the authors described the mechanism in which ROS could be implicated in the mitochondrial apoptotic JNK signaling pathway. We previously discussed the implication of this type of apoptosis in accordance with iron-mediated mitochondrial OS. Thus, it seems that there could be a close interaction between the OS mechanisms occurring as important effects of EMS tissues stimulation and development and the cell proliferation/apoptosis signaling. Caspase-3 was also previously described to be a key modulation point in the EMS apoptotic pathways [40].
On the other hand, Zhong et al. [39] also mentions the importance of traditional Chinese medicine known as the Kuntai Capsule on EMS pathogenesis by normalizing the expression of several pro-inflammatory and apoptotic mediators after three weeks of administration. Meldrum et al. [41] demonstrated that the growth of ectopic endometrium could be sensitive to hormone stimulation. It was previously shown that the impairments of estrogen-mediated cellular signaling could lead to important changes in TNF-α and IL-1β activities [42], thus suggesting that hormonal and inflammatory modulation could be correlated in EMS through the estrogen receptor (ER)β implication. Additionally, the escape of immune surveillance caused by ERβ could further interact with the apoptotic machinery and cytoplasmic inflammasome in the cytoplasm. Further studies showed that estrogen could promote Bcl-2 regulation and thymic stromal lymphopoietin (TSLP) secretion [43] and influence the steroid receptor coactivator (SRC-1) [44] (progesterone pathway). The way in which the latter is implicated in EMS was also correlated to apoptosis mechanisms (TNFα-mediated apoptosis) and inflammation (inflammatory signaling in endometriotic lesions), according to Han et al., 2012 [45] and Han et al., 2015 [42]. In this context, it was shown that p53 levels in ovarian endometriotic tissues are also correlated to the hormonal modulation (secretory phase versus proliferative phase) [46] and that depo-medroxyprogesterone acetate (DMPA), a progesterone derivate, could modulate cell proliferation and apoptosis in EMS women, but only in eutopic lesions cases [47].
However, Song et al. [48] recently showed that Bcl-2 modulation could also be implicated in cell proliferation and apoptosis in both eutopic and ectopic EMS tissues. In this way, the study that evaluated the EMS cell survival reported that the knockdown of YAP in ectopic endometrial stromal cells diminished cell proliferation but increased cell apoptosis concomitant with the decrease in expression of the transcriptional enhancer factor TEF-1 (TEAD1), connective tissue growth factor (CTGF), and Bcl-2. The overexpression of YAP also promoted an exaggerated proliferation and weakened apoptosis of normal endometrial stromal cells suggesting that the Hippo/Yes-associated protein (YAP) pathway is also an important component of apoptosis regulation in both ectopic and eutopic EMS lesion [48].
Other candidates for the potent apoptotic modulators in EMS were described by Bilgic et al. [49]. They showed that downregulation and/or alteration of endocannabinoid receptors could mediate the underlying mechanism of EMS and adenomyosis. Both CB1 and CB2 and also several key enzymes on the endocannabinoid pathway, such as fatty acid amide hydrolase (FAAH), N-acyl phosphatidylethanolamine phospholipase D (NAPE-PLD), diacylglycerol lipase (DAGL), and monoacylglycerol lipase (MAGL), were significantly impaired in endometriotic and adenomyotic tissues leading to a valid model of apoptotic signaling in EMS.
The much more complex TNF-α-mediated apoptosis in EMS still remains to be fully elucidated, since Tian et al. and Long et al. [50,51] described the mechanism through which the activation of this process could also be mediated by death-associated protein kinase 1 (DAPK1), c-Jun, and miRNAs. Another yet fully undiscovered mechanism is the implication of Ca+ and Cav1.3 channels in cell proliferation, aggregation, and apoptosis [52]. However, Yang et al. [52] already described the implication of the Cav1.3 channel, which is expressed in EMS tissue and primary ESCs cells and is highly sensitive to prostaglandin 2 and could modulate caspase 3-mediated apoptosis. The mitogen-activated protein kinase/ERK kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling pathway has also been proposed to fulfil the important role(s) in cell proliferation, apoptosis, and migration. This is the reason why Chen et al. [53] studied these pathways in the EMS context and found that the modulation of these processes could lead to the proliferation, migration, and apoptosis control of the endometrial stromal cells in EMS.
Table 2. Summary content regarding the outcomes in which we investigated the apoptotic and potential markers.
Table 2. Summary content regarding the outcomes in which we investigated the apoptotic and potential markers.
SubjectNumberRegulatory PathwayReference
Experimental Models
BALB/c micen = 33 miceBcl-2 ↓
Bax ↓
[36]
BALB/c nuden = 5 per groupBcl-2 ↑↓[48]
C57BL/6J micen = 3 per groupERβ ↑[44]
Normal (C57BL/6J), ERβ−/− (B6;129P2-Esr2tm1Unc/J), NALP3−/− (B6.129S6-Nlrp3tm1Bhk/J), and SCID (NOD.CB17-Prkdcscid/J). ERBAI and ROSALSL:ERβ, ROSALSL:ERβ:PRCre/+being generated by crossing ROSALSL:ERβ with PRCre+n = 4ERβ ↑↓[42]
Normal C57BL/6J, Tnf−/− (B6;129S-Tnftm1Gkl/J), Mmp9−/− (FVB.CgMmp9tm1Tvu/J), GFP-expressing (C57BL/6-Tg(CAG-GFP)10sb/J) and SCID (NOD.Cg-PrkfcscidB2mtm1UncIl2rgtm1Wjl/Szj), SRC-1 null–was generated by crossing SRC-1-null with GFP-expressingn = 6MMP9 ↑[45]
Sprague- Dawley ratsn = 74caspase-3 ↑
caspase-8 ↑
caspase-9 ↑
cytochrome ↑
[39]
Culture(s)-In Vivo
Human endometrial stromal cellsn = 32; n = 16 peritoneal or OE or both and n = 16 controls; n = 12 dermoid cysts and n = 3 serous cystadenoma and n = 1 simple ovarian cystcaspase-3 ↑
miR-21–5p ↑
[40]
Human endometrial stromal cellsn = 25 OE and n = 5 pelvic EMSBcl-2 ↑↓[43]
Human ectopic and normal endometrial cellsSTAT1 ↑
caspase-3 ↑
[38]
Human eutopic and ectopic ESCs controlsn = 17, n = 11, and n = 15Bcl-2 ↑
Bcl-xL ↑
caspase-3 ↓
VEGF-A ↑
HGF ↑
[37]
Cell line(s)-In Vitro
Female/CRL-7566 cell linen = 20 EMS patients, n = 17 adenomyosis and n = 19 controls; n = 12 proliferative, n = 7 secretory; Endometriosis tissues were divided as: n = 9 cystic and n = 11 non-cysticCB1 ↓
CB2 ↓
[49]
Female/HEK293T, CRL7566, CRL-11731 cell linesn = 10 OE, n = 10 ovarian cancer, and n = 10 controlsmiR-191 ↑↓[50]
Female/CRL-7566 cell linen = 20 eutopic and ectopic endometriummiR-29c ↑↓
c-Jun ↑↓
[51]
Female/hEM15A cell linen = 30 OE and n = 30 controlsCav1.3 ↑↓[52]
Human patients
Femalen = 30 OE and n = 29
controls
p53 ↑
p16 ↑
MDM2 ↓
[46]
Femalen = 28 EMS patients, n = 14 laparoscopy after DMPA injection and n = 14
laparoscopy without DMPA injection
PCNA ≠[47]
Femalen = 30 EMS patients and n = 15 controlsRaf-1 ↑[53]
↑ increased. ↓ decreased. ↑↓ regulatory. ≠ no significant change.

5. Endobiota and EMS

Regarding the microbiota interaction with the reproductive system functions, it was previously shown that the vaginal, cervical, and gut microbiota could be implicated in the pathogenesis of EMS. In this way, Ata et al. [54], in a study on the endobiota status in EMS-diagnosed women, showed the total absence of Atopobium in both vaginal and cervical microbiota of EMS group, and also the presence of Gardnerella, Streptococcus, Escherichia, Shigella, and Ureaplasma in the cervical samples. Moreover, Cregger et al. [55] suggested that the richness and phylogenetic diversity were increased in stage III EMS, as compared to healthy individuals, and that the surgical intervention for EMS tissues neutralization reversed the endobiota characteristics for a limited period of time. In this context, while the bacterial contamination EMS development hypothesis suggests that EMS pathogenesis and inflammation are correlated by the microbial pathogens reaching the endometrial space [56], it was shown that in an EMS mice model, the reduction of the ectopic lesions and the magnitude of the inflammatory response could both be modulated at the same time through antibiotic administration [57]. Furthermore, it is important to mention that both OS-related apoptotic signals and the inflammatory response are modulated differently, considering the origin of the elicitors. In this way, Chen et al. [58] suggested that, during vaginal inflammation due to pathogens invasion, the immune response could also be triggered by ROS overproduction, eventually leading to an apoptotic signal via neutrophilic activation (caspase-3 mediated). A similar mechanism was previously described for apoptotic modulation in the ectopic endothelial stromal cells underlying EMS tumors.
However, despite that the most prevalent microbiota component was the Lactobacillus genus members, Wei et al. [59] showed that as advancing up the reproductive tract, the microflora diversity increases, as they observed by sampling the vagina, posterior vaginal fornix, cervical mucus, endometrium, and peritoneal fluid. Moreover, Riganelli et al. [60] suggested that reproductive system-colonizing Lactobacillus species could play an important role in more than mucosa maintenance, as they observed that the differences between in vitro fertilized pregnant and non-pregnant women mainly reside in the vaginal and endometrial microbiota diversity. Moreover, it is known that lactobacilli are a continuous source of H2O2, which could contribute to intrinsic protective mechanisms, thus the possible implication of this microbiota component in OS and apoptosis modulation [59].
Furthermore, it could be suggested that there could be an interaction between the hormonal fluctuations during the menstrual cycle and reproductive system microbiota. However, disregarding that microbial taxa and potential functions proved to be positively correlated with the menstrual cycle or are over-represented in patients with adenomyosis and/or infertility caused by EMS [61], Akiyama et al. [62] showed that the distribution of microbiota in the cervical mucus of women with and without EMS is very similar and that regardless of the menstrual cycle phase, Lactobacillus spp. were dominant, concomitantly with an increased ratio of Corynebacterium, Enterobacteriaceae, Flavobacterium, Pseudomonas, and Streptococcus in the EMS group. Enterobacteriaceae and Streptococcus were considered to be significant candidates in the EMS group compared to in controls. Furthermore, considering the previously mentioned implications of estrogen in apoptosis, Leonardi et al. [56] thoroughly discussed the role of microbiota in estrogen metabolism in EMS with regards to the changes occurring in the gut microbiota. In this way, it was suggested that EMS development and progression could be very much influenced by the prevalence of the bacteria that produce enzymes for estrogen deconjugation in the gut.
Considering these aspects, further investigations were carried out on animal models in order to elucidate the way in which gut, vaginal, and endometrial microbiota are implicated in the EMS occurrence and development. In this way, using transgenic green fluorescent protein (GFP)+ donor mice and the standard antitumor and immunologic studies mice strain C57BL/6, Hantschel et al. [63] demonstrated that the uterine tissues fragment transplantation receiver mice exhibited significant differences in terms of gut microbiota, alongside the typical tissue morphology of the lesions and ease of differentiating GFP-host tissues. In a similar study, Yuan et al. [64] also described the gut microbiota alterations during EMS development via intraperitoneal injection of endometrial tissues in C57BL/6 mice, as both EMS and mock groups shared an identical microenvironment and richness until day 42. There was a significantly different ratio among Firmicutes/Bacteroidetes in the EMS group, and Bifidobacterium was also increased in this group, suggesting that lesion development and dysbacteriosis could be correlated. Considering this, Hu et al. [65] further analyzed the existence of an inflammatory response of the uterus in FMT-mice and observed that EMS induced by pathogenic bacteria inoculation could be more severe, but reversible. Moreover, considering the molecular changes that can be observed in this context, the authors noted a significant decrease in the short-chain fatty acids (SCFAs) in the feces of mice with dysbiosis and the administered treatment with sodium butyrate/propionate increased the overall concentration in both circulation and uterine tissues. Furthermore, the study reported the increase in pathogen load in the uteri of dysbiosis-associated mice, which could explain the restricted phagocytic ability and responsiveness of neutrophils.
A crucial yet underestimated microbial composition modulator could also be the pathogens encountered in the reproductive system. In this way, several studies reported high human papillomavirus prevalence [66,67,68] in association with EMS. In all these cases, the main aim was to investigate whether sexually transmitted viruses (STDs) or prokaryotes such as human HPVs, herpes viruses, and Chlamydia trachomatis in the lower and upper female genital tract are correlated with EMS injuries. Considering that herpes viruses and Chlamydia trachomatis were not detected in endometriosis lesions, HPV was prevalent in all samples analyzed in both categories: With and without EMS. The risk is higher, even double, in patients with EMS, with all positive patients having been previously detected with an HPV infection. As expected, the risk towards medium (md) and high (hr) HPV was in females with EMS in both lower and upper genital tract. However, Vestergaard et al. [69] contradicts these findings and sustain that no significant differences were observed in terms of virus prevalence in endometriotic lesions.
Table 3. Summary content regarding the outcomes in which we investigated the microbial taxa in EMS.
Table 3. Summary content regarding the outcomes in which we investigated the microbial taxa in EMS.
SubjectNumberRegion AnalyzedMicrobial Taxa DifferencesReference
Female14 stage 3/4 ¾ EMS women and 14
controls
V3-V4 region 16S rRNAAtopobium absence in the vaginal and cervical microbiota
Gardnerella
Streptococcus
Escherichia
Shigella
Ureoplasma
Prevalence of Shigella and Escherichia in the stool
[54]
Female19 women; n = 10 distinct EMS stages and n = 9 controlsV3-V5 region 16S rRNASignificant alterations among bacterial communities depending on the site from which the sample was collected and diagnosis[55]
Female36 EMS women and 14 controlsV4-V5 region 16S rRNAPrevalence of Lactobacillus in the lower third of the vagina and posterior vaginal fornix.
Differences among communities were visible starting from the cervical mucus of EMS patients, gradually increasing towards the reproductive system.
↑ Operational Taxonomic Units in the cervical mucus, endometrium, and peritoneal fluid.
[59]
Female34 womenV4-V4 region 16S rRNALactobacilli-dominant microenvironment in contrast to non-pregnant women, whereas the endometrial bacterial colonization was marker exclusively by a polymicrobial habitat in which lactobacilli were predominant[60]
Female39 EMS women and 30 controlsV5-V6 region 16S rRNASimilar microbial taxa between women with and without EMS.
↑ Lactobacilli spp
↑ Corynebacterium
↑ Enterobacteriaceae in EMS group than in control (p < 0.05)
↑ Flavobacterium
↑ Pseudomonas
↑ Streptococcus in EMS group than in Control (p < 0.05)
[62]
MiceC57BL/6 wild-type mice and transgenic C57BL/6-TgN(ACTB-EGFP)1Osb/J donor mice; 4 per cage-2 EMS and 2 sham-transplanted controlsV4-V5 region16S rRNANo significant difference between groups, being speculated that EMS does not induce a dysbiosis during the acute phase of lesion formation.[63]
MiceC57BL6; 22 from which in 4 distinct intervals (day 7, 14, and 28) fecal sample was collected and the mice were sacrificed for model confirmation; 8 out of 10 in day 42.V4 region 16S rRNASimilar diversity and richness between EMS and mock mice.
↑ Firmicutes/Bacteroidetes ratio in EMS after 42 days
↑ Bifidobacterium in EMS group
[64]
MiceBALB/c miceV4 region 16S rRNA↑ pathogen load in the uteri of gut microbiota-dysbiosis mice[65]
↑ increased.

6. Conclusions

Based on all aspects discussed throughout this manuscript, we could suggest that endometriosis exerts a pronounced detrimental effect on an organism’s integrity. Overall, clear changes of oxidative biomarkers levels could be observed, the associated repercussions being branched and reflected by an abnormal expression of apoptosis-related mediators that ultimately lead to a dysbiosis. The current knowledge in endometriosis pathophysiology offers some explanation for the crucial roles that oxidative stress, apoptosis, and endobiota possess in various gynecological disorders, further outlining the multifactorial character of endometriosis.

Author Contributions

O.-D.I.; I.-M.B.; R.M.; I.S. (Ioana Scripcariu); G.S.; D.G.; I.S. (Irina Stoian) (conceptualization, data curation, investigation, formal analysis, methodology, writing—original draft); B.D.; A.C.; C.I. (conceptualization, methodology, writing—review and editing); B.D.; A.C.; C.I. (supervision, validation, project administration). All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

The datasets used and analyzed during the current study are available from the corresponding author on reasonable request.

Acknowledgments

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bazot, M.; Bharwani, N.; Huchon, C.; Kinkel, K.; Cunha, T.M.; Guerra, A.; Manganaro, L.; Buñesch, L.; Kido, A.; Togashi, K.; et al. European Society of Urogenital Radiology (ESUR) Guidelines: MR Imaging of Pelvic Endometriosis. Eur. Radiol. 2017, 27, 2765–2775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Vercellini, P.; Viganò, P.; Somigliana, E.; Fedele, L. Endometriosis: Pathogenesis and Treatment. Nat. Rev. Endocrinol. 2014, 10, 261–275. [Google Scholar] [CrossRef]
  3. Zondervan, K.T.; Becker, C.M.; Koga, K.; Missmer, S.A.; Taylor, R.N.; Viganò, P. Endometriosis. Nat. Rev. Dis. Prim. 2018, 4, 9. [Google Scholar] [CrossRef]
  4. Johnson, N.P.; Hummelshoj, L.; Adamson, G.D.; Keckstein, J.; Taylor, H.S.; Abrao, M.S.; Bush, D.; Kiesel, L.; Tamimi, R.; Sharpe-Timms, K.L.; et al. World Endometriosis Society Consensus on the Classification of Endometriosis. Hum. Reprod. 2017, 32, 315–324. [Google Scholar] [CrossRef]
  5. Czyzyk, A.; Podfigurna, A.; Szeliga, A.; Meczekalski, B. Update on Endometriosis Pathogenesis. Minerva Ginecol. 2017, 69, 447–461. [Google Scholar]
  6. 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]
  7. Halme, J.; Hammond, M.G.; Hulka, J.F.; Raj, S.G.; Talbert, L.M. Retrograde Menstruation in Healthy Women and in Patients with Endometriosis. Obstet. Gynecol. 1984, 64, 151–154. [Google Scholar] [PubMed]
  8. Liu, D.T.; Hitchcock, A. Endometriosis: Its Association with Retrograde Menstruation, Dysmenorrhoea and Tubal Pathology. Br. J. Obstet. Gynaecol. 1986, 93, 859–862. [Google Scholar] [CrossRef]
  9. Sourial, S.; Tempest, N.; Hapangama, D.K. Theories on the Pathogenesis of Endometriosis. Int. J. Reprod. Med. 2014, 2014, 179515. [Google Scholar] [CrossRef] [Green Version]
  10. Ahn, S.H.; Monsanto, S.P.; Miller, C.; Singh, S.S.; Thomas, R.; Tayade, C. Pathophysiology and Immune Dysfunction in Endometriosis. Biomed. Res. Int. 2015, 2015, 795976. [Google Scholar] [CrossRef] [Green Version]
  11. Gordts, S.; Koninckx, P.; Brosens, I. Pathogenesis of Deep Endometriosis. Fertil. Steril. 2017, 108, 872–885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Halme, J.; Becker, S.; Hammond, M.G.; Raj, M.H.G.; Raj, S. Increased Activation of Pelvic Macrophages in Infertile Women with Mild Endometriosis. Am. J. Obstet. Gynecol. 1983, 145, 333–337. [Google Scholar] [CrossRef]
  13. Keettel, W.C.; Stein, R.J. The Viability of the Cast-Off Menstrual Endometrium. Am. J. Obstet. Gynecol. 1951, 61, 440–442. [Google Scholar] [CrossRef]
  14. Nisolle, M.; Paindaveine, B.; Bourdon, A.; Berlière, M.; Casanas-Roux, F.; Donnez, J. Histologic Study of Peritoneal Endometriosis in Infertile Women. Fertil. Steril. 1990, 53, 984–988. [Google Scholar] [CrossRef]
  15. Kruitwagen, R.F.P.M.; Poels, L.G.; Willemsen, W.N.P.; de Ronde, I.J.Y.; Jap, P.H.K.; Rolland, R. Endometrial Epithelial Cells in Peritoneal Fluid during the Early Follicular Phase. Fertil. Steril. 1991, 55, 297–303. [Google Scholar] [CrossRef]
  16. Arumugam, K.; Lim, J.M. Menstrual Characteristics Associated with Endometriosis. Br. J. Obstet. Gynaecol. 1997, 104, 948–950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Vercellini, P.; De Giorgi, O.; Aimi, G.; Panazza, S.; Uglietti, A.; Crosignani, P.G. Menstrual Characteristics in Women with and Without Endometriosis. Obstet. Gynecol. 1997, 90, 264–268. [Google Scholar] [CrossRef]
  18. Kvaskoff, M.; Mu, F.; Terry, K.L.; Harris, H.R.; Poole, E.M.; Farland, L.; Missmer, S.A. Endometriosis: A High-Risk Population for Major Chronic Diseases? Hum. Reprod. Update 2015, 21, 500–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Bulun, S.E.; Yilmaz, B.D.; Sison, C.; Miyazaki, K.; Bernardi, L.; Liu, S.; Kohlmeier, A.; Yin, P.; Milad, M.; Wei, J. Endometriosis. Endocr. Rev. 2019, 40, 1048–1079. [Google Scholar] [CrossRef]
  20. Young, V.B. The Role of the Microbiome in Human Health and Disease: An Introduction for Clinicians. BMJ 2017, 356. [Google Scholar] [CrossRef]
  21. 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]
  22. Amreen, S.; Kumar, P.; Gupta, P.; Rao, P. Evaluation of Oxidative Stress and Severity of Endometriosis. J. Hum. Reprod. Sci. 2019, 12, 40–46. [Google Scholar]
  23. Turkyilmaz, E.; Yildirim, M.; Cendek, B.D.; Baran, P.; Alisik, M.; Dalgaci, F.; Yavuz, A.F. Evaluation of Oxidative Stress Markers and Intra-Extracellular Antioxidant Activities in Patients with Endometriosis. Eur. J. Obstet. Gynecol. Reprod. Biol. 2016, 199, 164–168. [Google Scholar] [CrossRef]
  24. Elsharkawy, S.S.; Abdrabo, S.M.; Khalil, H.M.; Mannaa, F.H. Evaluation of Oxidative Stress Markers in Serum and Follicular Fluid of Women with Infertility Related to Endometriosis. J. Clin. Obstet. Gynecol. Infertility 2020, 4, 1045. [Google Scholar]
  25. Juan, C.A.; Pérez de la Lastra, J.M.; Plou, F.J.; Pérez-Lebeña, E. The Chemistry of Reactive Oxygen Species (ROS) Revisited: Outlining Their Role in Biological Macromolecules (DNA, Lipids and Proteins) and Induced Pathologies. Int. J. Mol. Sci. 2021, 22, 4642. [Google Scholar] [CrossRef] [PubMed]
  26. Alizadeh, M.; Mahjoub, S.; Esmaelzadeh, S.; Hajian, K.; Basirat, Z.; Ghasemi, M. Evaluation of Oxidative Stress in Endometriosis: A Case-Control Study. Casp. J. Intern. Med. 2015, 6, 25–29. [Google Scholar]
  27. Nasiri, N.; Moini, A.; Eftekhari-Yazdi, P.; Karimian, L.; Salman Yazdi, R.; Arabipoor, A. Oxidative Stress Statues in Serum and Follicular Fluid of Women with Endometriosis. Cell J. 2017, 18, 582–587. [Google Scholar] [PubMed]
  28. Montoya-Estrada, A.; Coria-García, C.F.; Cruz-Orozco, O.P.; Aguayo-González, P.; Torres-Ramos, Y.D.; Flores-Herrera, H.; Hicks, J.J.; Medina-Navarro, R.; Guzmán-Grenfell, A.M. Increased Systemic and Peritoneal Oxidative Stress Biomarkers in Endometriosis Are Not Related to Retrograde Menstruation. Redox Rep. 2019, 24, 51–55. [Google Scholar] [CrossRef]
  29. Thézénas, M.-L.; De Leo, B.; Laux-Biehlmann, A.; Bafligil, C.; Elger, B.; Tapmeier, T.; Morten, K.; Rahmioglu, N.; Dakin, S.G.; Charles, P.; et al. Amine Oxidase 3 Is a Novel Pro-Inflammatory Marker of Oxidative Stress in Peritoneal Endometriosis Lesions. Sci. Rep. 2020, 10, 1495. [Google Scholar] [CrossRef] [Green Version]
  30. Polak, G.; Barczyński, B.; Wertel, I.; Kwaśniewski, W.; Bednarek, W.; Derewianka-Polak, M.; Frąszczak, K.; Olajossy, M.; Kotarski, J. Disrupted Iron Metabolism in Peritoneal Fluid May Induce Oxidative Stress in the Peritoneal Cavity of Women with Endometriosis. Ann. Agric. Environ. Med. 2018, 25, 587–592. [Google Scholar] [CrossRef]
  31. Chen, C.; Zhou, Y.; Hu, C.; Wang, Y.; Yan, Z.; Li, Z.; Wu, R. Mitochondria and Oxidative Stress in Ovarian Endometriosis. Free Radic. Biol. Med. 2019, 136, 22–34. [Google Scholar] [CrossRef]
  32. Hayashi, S.; Nakamura, T.; Motooka, Y.; Ito, F.; Jiang, L.; Akatsuka, S.; Iwase, A.; Kajiyama, H.; Kikkawa, F.; Toyokuni, S. Novel Ovarian Endometriosis Model Causes Infertility via Iron-Mediated Oxidative Stress in Mice. Redox Biol. 2020, 37, 101726. [Google Scholar] [CrossRef]
  33. González-Foruria, I.; Santulli, P.; Chouzenoux, S.; Carmona, F.; Chapron, C.; Batteux, F. Dysregulation of the ADAM17/Notch Signalling Pathways in Endometriosis: From Oxidative Stress to Fibrosis. Mol. Hum. Reprod. 2017, 23, 488–499. [Google Scholar] [CrossRef]
  34. Agic, A.; Djalali, S.; Diedrich, K.; Hornung, D. Apoptosis in Endometriosis. Gynecol. Obstet. Investig. 2009, 68, 217–223. [Google Scholar] [CrossRef]
  35. Taniguchi, F.; Kaponis, A.; Izawa, M.; Kiyama, T.; Deura, I.; Ito, M.; Iwabe, T.; Adonakis, G.; Terakawa, N.; Harada, T. Apoptosis and Endometriosis. Front. Biosci. (Elite Ed.) 2011, 3, 648–662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Mulyantoro, I.; Indrapraja, O.; Widjiati, W.; Noerpramana, N.P. Effect of Metformin on Bcl-2/Bax Expression Ratio and Endometrial Implants: A Mouse Model in Endometriosis Study. J. Biomed. Transl. Res. 2020, 6, 53–58. [Google Scholar] [CrossRef]
  37. Delbandi, A.-A.; Mahmoudi, M.; Shervin, A.; Heidari, S.; Kolahdouz-Mohammadi, R.; Zarnani, A.-H. Evaluation of Apoptosis and Angiogenesis in Ectopic and Eutopic Stromal Cells of Patients With Endometriosis Compared to Non-Endometriotic Controls. BMC Womens. Health 2020, 20, 3. [Google Scholar] [CrossRef] [PubMed]
  38. Li, J.; Abudula, M.; Fan, X.; Wang, F.; Chen, Y.; Liu, L. Sunitinib Induces Primary Ectopic Endometrial Cell Apoptosis Through Up-Regulation of STAT1 In Vitro. J. Clin. Lab. Anal. 2020, 34, e23482. [Google Scholar] [CrossRef] [PubMed]
  39. Zhong, R.; Ma, A.; Zhu, J.; Li, G.; Xie, S.; Li, Z.; Gui, Y.; Zhu, Y. Kuntai Capsule Inhibited Endometriosis via Inducing Apoptosis in a Rat Model. Evid. Based Complement. Altern. Med. 2016, 2016, 5649169. [Google Scholar] [CrossRef] [PubMed]
  40. Park, J.; Lee, S.; Kim, M.-K.; Lee, J.; Yun, B.; Park, J.; Seo, S.K.; Cho, S.; Choi, Y. Saponin Extracts Induced Apoptosis of Endometrial Cells from Women with Endometriosis Through Modulation of miR-21-5p. Reprod. Sci. 2017, 25, 193371911771126. [Google Scholar] [CrossRef]
  41. Meldrum, D.R.; Chang, R.J.; Lu, J.; Vale, W.; Rivier, J.; Judd, H.L. “Medical Oophorectomy” Using a Long-Acting gGnRHrh Agonist-A Possible New Approach to the Treatment of Endometriosis. J. Clin. Endocrinol. Metab. 1982, 54, 1081–1083. [Google Scholar] [CrossRef] [PubMed]
  42. Han, S.J.; Jung, S.Y.; Wu, S.-P.; Hawkins, S.M.; Park, M.J.; Kyo, S.; Qin, J.; Lydon, J.P.; Tsai, S.Y.; Tsai, M.-J.; et al. Estrogen Receptor β Modulates Apoptosis Complexes and the Inflammasome to Drive the Pathogenesis of Endometriosis. Cell 2015, 163, 960–974. [Google Scholar] [CrossRef] [Green Version]
  43. Yang, H.; Kai, C.; Mei, J.; Zhou, W.-J.; Liu, L.; Yao, L.; Meng, Y.; Wang, M.; Ha, S.; Lai, Z.; et al. Estrogen Restricts the Apoptosis of Endometrial Stromal Cells by Promoting TSLP Secretion. Mol. Med. Rep. 2018, 18, 4410–4416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Cho, Y.J.; Lee, J.E.; Park, M.J.; O’Malley, B.W.; Han, S.J. Bufalin Suppresses Endometriosis Progression by Inducing Pyroptosis and Apoptosis. J. Endocrinol. 2018, 237, 255–269. [Google Scholar] [CrossRef] [Green Version]
  45. Han, S.J.; Hawkins, S.M.; Begum, K.; Jung, S.Y.; Kovanci, E.; Qin, J.; Lydon, J.P.; DeMayo, F.J.; O’Malley, B.W. A New Isoform of Steroid Receptor Coactivator-1 Is Crucial for Pathogenic Progression of Endometriosis. Nat. Med. 2012, 18, 1102–1111. [Google Scholar] [CrossRef] [Green Version]
  46. Sang, L.; Fang, Q.-J.; Zhao, X.-B. A Research on the Protein Expression of p53, p16, and MDM2 in Endometriosis. Medicine 2019, 98, e14776. [Google Scholar] [CrossRef] [PubMed]
  47. Tingthanatikul, Y.; Tawarasumida, Y.; Lertvikool, S.; Wongkularb, A.; Sophonsritsuk, A.; Weerakiet, S.; Sroyraya, M.; Chansela, P.; Changklungmoa, N.; Songkoomkrong, S.; et al. DMPA Suppresses Cell Proliferation and Enhances Cell Apoptosis of Eutopic Endometrium in Women with Endometriosis: A Randomized Controlled Study. J. Med. Assoc. Thai. 2016, 99, 751–756. [Google Scholar]
  48. Song, Y.; Fu, J.; Zhou, M.; Xiao, L.; Feng, X.; Chen, H.; Huang, W. Activated Hippo/Yes-Associated Protein Pathway Promotes Cell Proliferation and Anti-Apoptosis in Endometrial Stromal Cells of Endometriosis. J. Clin. Endocrinol. Metab. 2016, 101, 1552–1561. [Google Scholar] [CrossRef] [PubMed]
  49. Bilgic, E.; Meydanli, E.; Köse, S.; Aydin, M.; Karaismailoğlu, E.; Akar, I.; Usubutun, A.; Korkusuz, P. Endocannabinoids Modulate Apoptosis in Endometriosis and Adenomyosis. Acta Histochem. 2017, 119, 523–532. [Google Scholar] [CrossRef] [PubMed]
  50. Tian, X.; Xu, L.; Wang, P. MiR-191 Inhibits TNF-α Induced Apoptosis of Ovarian Endometriosis and Endometrioid Carcinoma Cells by Targeting DAPK1. Int. J. Clin. Exp. Pathol. 2015, 8, 4933–4942. [Google Scholar]
  51. Long, M.; Wan, X.; La, X.; Gong, X.; Cai, X. MiR-29c Is Downregulated in the Ectopic Endometrium and Exerts Its Effects on Endometrial Cell Proliferation, Apoptosis and Invasion by Targeting C-Jun. Int. J. Mol. Med. 2015, 35, 1119–1125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Yang, Y.; Yuan, Y.; Ma, X.; Xing, F. Ca2+ Channel Subunit a 1D Inhibits Endometriosis Cell Apoptosis and Mediated by Prostaglandin E2. Ginekol. Pol. 2019, 90, 669–674. [Google Scholar] [CrossRef] [Green Version]
  53. Chen, Q.; Hang, Y.; Zhang, T.; Tan, L.; Li, S.; Jin, Y. USP10 Promotes Proliferation and Migration and Inhibits Apoptosis of Endometrial Stromal Cells in Endometriosis through AC-Tivating the Raf-1/MEK/ERK Pathway. Am. J. Physiol. Physiol. 2018, 315, C863–C872. [Google Scholar] [CrossRef] [PubMed]
  54. 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]
  55. Cregger, M.; 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. [Google Scholar] [CrossRef]
  56. Leonardi, M.; Hicks, C.; El-Assaad, F.; El-Omar, E.; Condous, G. Endometriosis and the Microbiome: A Systematic Review. BJOG An. Int. J. Obstet. Gynaecol. 2020, 127, 239–249. [Google Scholar] [CrossRef]
  57. Chadchan, S.B.; Cheng, M.; Parnell, L.A.; Yin, Y.; Schriefer, A.; Mysorekar, I.U.; Kommagani, R. Antibiotic Therapy with Metronidazole Reduces Endometriosis Disease Progression in Mice: A Potential Role for Gut Microbiota. Hum. Reprod. 2019, 34, 1106–1116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Chen, Z.; Zhang, Z.; Zhang, H.; Xie, B. Analysis of the Oxidative Stress Status in Nonspecific Vaginitis and Its Role in Vaginal Epithelial Cells Apoptosis. Biomed. Res. Int. 2015, 2015, 795656. [Google Scholar] [CrossRef] [Green Version]
  59. Wei, W.; Zhang, X.; Tang, H.; Zeng, L.; Wu, R. Microbiota Composition and Distribution Along the Female Reproductive Tract of Women with Endometriosis. Ann. Clin. Microbiol. Antimicrob. 2020, 19, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Riganelli, L.; Iebba, V.; Piccioni, M.; Illuminati, I.; Bonfiglio, G.; Neroni, B.; Calvo, L.; Gagliardi, A.; Levrero, M.; Merlino, L.; et al. Structural Variations of Vaginal and Endometrial Microbiota: Hints on Female Infertility. Front. Cell. Infect. Microbiol. 2020, 10, 350. [Google Scholar] [CrossRef] [PubMed]
  61. Agostinis, C.; Mangogna, A.; Bossi, F.; Ricci, G.; Kishore, U.; Bulla, R. Uterine Immunity and Microbiota: A Shifting Paradigm. Front. Immunol. 2019, 10, 2387. [Google Scholar] [CrossRef] [Green Version]
  62. Akiyama, K.; Nishioka, K.; Khan, K.N.; Tanaka, Y.; Mori, T.; Nakaya, T.; Kitawaki, J. Molecular Detection of Microbial Colonization in Cervical Mucus of Women with and without Endometriosis. Am. J. Reprod. Immunol. 2019, 82, e13147. [Google Scholar] [CrossRef]
  63. Hantschel, J.; Weis, S.; Schäfer, K.-H.; Menger, M.D.; Kohl, M.; Egert, M.; Laschke, M.W. Effect of Endometriosis on the Fecal Bacteriota Composition of Mice during the Acute Phase of Lesion Formation. PLoS ONE 2019, 14, e0226835. [Google Scholar] [CrossRef] [Green Version]
  64. Yuan, M.; Li, D.; Zhang, Z.; Sun, H.; An, M.; Wang, G. Endometriosis Induces Gut Microbiota Alterations in Mice. Hum. Reprod. 2018, 33, 607–616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Hu, X.; Mu, R.; Xu, M.; Yuan, X.; Jiang, P.; Guo, J.; Cao, Y.; Zhang, N.; Fu, Y. Gut Microbiota Mediate the Protective Effects on Endometritis Induced by Staphylococcus aureus in Mice. Food Funct. 2020, 11, 3695–3705. [Google Scholar] [CrossRef]
  66. Oppelt, P.; Renner, S.P.; Strick, R.; Valletta, D.; Mehlhorn, G.; Fasching, P.A.; Beckmann, M.W.; Strissel, P.L. Correlation of High-Risk Human Papilloma Viruses but Not of Herpes Viruses or Chlamydia trachomatis with Endometriosis Lesions. Fertil. Steril. 2010, 93, 1778–1786. [Google Scholar] [CrossRef] [PubMed]
  67. Heidarpour, M.; Derakhshan, M.; Derakhshan-Horeh, M.; Kheirollahi, M.; Dashti, S. Prevalence of High-Risk Human Papillomavirus Infection in Women with Ovarian Endometriosis. J. Obstet. Gynaecol. Res. 2017, 43, 135–139. [Google Scholar] [CrossRef] [Green Version]
  68. Rocha, R.M.; Souza, R.P.; Gimenes, F.; Consolaro, M.E.L. The High-Risk Human Papillomavirus Continuum Along the Female Reproductive Tract and Its Relationship to Infertility and Endometriosis. Reprod. Biomed. Online 2019, 38, 926–937. [Google Scholar] [CrossRef]
  69. Vestergaard, A.L.; Knudsen, U.B.; Munk, T.; Rosbach, H.; Bialasiewicz, S.; Sloots, T.P.; Martensen, P.M.; Antonsson, A. Low Prevalence of DNA Viruses in the Human Endometrium and Endometriosis. Arch. Virol. 2010, 155, 695–703. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Doroftei, B.; Ilie, O.-D.; Balmus, I.-M.; Ciobica, A.; Maftei, R.; Scripcariu, I.; Simionescu, G.; Grab, D.; Stoian, I.; Ilea, C. Molecular and Clinical Insights on the Complex Interaction between Oxidative Stress, Apoptosis, and Endobiota in the Pathogenesis of Endometriosis. Diagnostics 2021, 11, 1434. https://doi.org/10.3390/diagnostics11081434

AMA Style

Doroftei B, Ilie O-D, Balmus I-M, Ciobica A, Maftei R, Scripcariu I, Simionescu G, Grab D, Stoian I, Ilea C. Molecular and Clinical Insights on the Complex Interaction between Oxidative Stress, Apoptosis, and Endobiota in the Pathogenesis of Endometriosis. Diagnostics. 2021; 11(8):1434. https://doi.org/10.3390/diagnostics11081434

Chicago/Turabian Style

Doroftei, Bogdan, Ovidiu-Dumitru Ilie, Ioana-Miruna Balmus, Alin Ciobica, Radu Maftei, Ioana Scripcariu, Gabriela Simionescu, Delia Grab, Irina Stoian, and Ciprian Ilea. 2021. "Molecular and Clinical Insights on the Complex Interaction between Oxidative Stress, Apoptosis, and Endobiota in the Pathogenesis of Endometriosis" Diagnostics 11, no. 8: 1434. https://doi.org/10.3390/diagnostics11081434

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