Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3147 2023-11-24 02:34:06 |
2 Format correct Meta information modification 3147 2023-11-24 05:15:25 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Begum, M.I.A.; Chuan, L.; Hong, S.; Chae, H. The Pathogenic Mechanisms of Endometriosis. Encyclopedia. Available online: https://encyclopedia.pub/entry/52010 (accessed on 18 May 2024).
Begum MIA, Chuan L, Hong S, Chae H. The Pathogenic Mechanisms of Endometriosis. Encyclopedia. Available at: https://encyclopedia.pub/entry/52010. Accessed May 18, 2024.
Begum, Mst Ismat Ara, Lin Chuan, Seong-Tshool Hong, Hee-Suk Chae. "The Pathogenic Mechanisms of Endometriosis" Encyclopedia, https://encyclopedia.pub/entry/52010 (accessed May 18, 2024).
Begum, M.I.A., Chuan, L., Hong, S., & Chae, H. (2023, November 24). The Pathogenic Mechanisms of Endometriosis. In Encyclopedia. https://encyclopedia.pub/entry/52010
Begum, Mst Ismat Ara, et al. "The Pathogenic Mechanisms of Endometriosis." Encyclopedia. Web. 24 November, 2023.
The Pathogenic Mechanisms of Endometriosis
Edit

Endometriosis (EMS), characterized by the presence of endometrial stromal and glandular tissue outside the uterine cavity, is a relatively common gynecologic disease affecting about 10% of women of reproductive age. While the symptoms of EMS can vary, clinical manifestations such as pelvic pain, heavy or irregular menstrual bleeding, and gastrointestinal symptoms are frequently observed. Apart from the physical discomfort, EMS has been well noticed in infertility.

miRNA endometriosis pathogenic mechanisms peritoneal fluid follicular fluid implantation

1. Introduction

EMS is a common gynecological disorder characterized by pelvic pain, heavy menstruation, and infertility. Roughly half of women affected by EMS encounter difficulties with infertility [1][2]. Multiple studies indicate that women in the early stages of EMS face reduced prospects of natural conception, and there has been a growing prevalence of EMS among women experiencing infertility [3][4]. Among patients with this milder form of the disease, potential mechanisms leading to infertility include persistent inflammation, disrupted ovulation, impaired embryo implantation, and suboptimal receptivity of the endometrial lining [5]. Nonetheless, the precise underlying mechanism of infertility linked to EMS remains elusive.
Although the etiopathogenic mechanisms of EMS have not yet been fully elucidated, the current research indicates that miRNAs are involved in the disease. The differential expression of miRNAs has been repeatedly observed in endometrial tissues and in the extracellular body fluids of women with EMS in contrast to those without the condition [6][7]. This variance in miRNA expression signifies disrupted gene regulation. Profiling miRNAs in individuals with EMS holds the potential to provide valuable insights into understanding the mechanisms driving the development of the disease.
While EMS is often linked to infertility [8], the precise mechanisms driving this association remain incompletely understood. Numerous investigations have been undertaken to shed light on this issue, with various authors proposing diverse mechanisms that could potentially contribute to infertility [9][10][11]. These mechanisms encompass anatomical and microenvironmental factors that may have detrimental effects on oocyte maturation, fertilization, zygote transportation within the fallopian tube, embryo implantation, and even sperms function.

2. Ovary Dysfunction

Patients with EMS exhibited the abnormal secretion of Luteinizing Hormone (LH), potentially leading to ovarian dysfunction [12]. Alterations in folliculogenesis among individuals with EMS may contribute to issues such as ovulatory dysfunction, diminished oocyte quality, reduced fertilization rates, the production of lower-grade embryos, and decreased implantation success [13][14]. In an observational in vitro fertilization (IVF) study involving natural cycles, it was observed that patients with minimal-to-mild EMS had significantly longer follicular phases and lower fertilization rates when compared to women with tubal factor infertility and unexplained infertility [12]. Women with EMS were also found to exhibit a slower rate of follicular growth [15] and smaller dominant follicles compared to women with unexplained infertility [16]. Additionally, EMS patients often experience abnormal follicle development, ovulation, and luteal function [17].

3. Dysregulated Immune Function, Hormonal Imbalance, and Oxygen Species in Follicular Fluid

EMS is linked to inflammatory changes in the follicular fluid (FF) environment. A case-controlled study comparing patients with EMS to those with other causes of infertility has noted an increased percentage of B lymphocytes, Natural Killer (NK) cells, and monocyte–macrophages in the FF. This suggests potential alterations in the immunological function within the FF of individuals with EMS [18]. Elevated concentrations of interleukins IL-6, IL-1b, IL-10, and Tumor Necrosis Factor-alpha (TNF-alpha), along with decreased Vascular Endothelial Growth Factor (VEGF), have been documented in the FF of EMS patients [19][20][21]. Immunological changes in the follicular fluid (FF) and serum of women with EMS may contribute, at least in part, to the pathological changes associated with infertility in these patients [22]. For instance, VEGF, known for enhancing follicular health and vascularization, is found in reduced levels in women with EMS [20], potentially associated with diminished embryo quality and implantation rates [13].
Notably, significantly elevated concentrations of TNF-alpha in granulosa cell cultures from women with EMS have been reported [23], which may also be linked to infertility [24]. A study demonstrated that the use of etanercept, an immunoglobulin fusion protein that hinders TNF-alpha activity, reduced the severity of EMS and the size of endometriotic foci in an animal study [25]. Unlike the effect on EMS, the inhibition of TNF-α did not appear to affect infertility [26]. However, the use of etanercept before in vitro fertilization (IVF) enhanced the success rate in cases of advanced EMS [26]. Cytokine changes may influence alterations in the granulosa cell cycle, as previously observed in EMS patients [27]. For example, elevated levels of IL-10 were shown to prevent the natural downregulation of p27, leading to a G0 phase arrest [28]. Several other cytokines, elevated in the FF of EMS patients, such as IL-6, IL-1b, IL-8, or IL-1a, likely contribute to various cell cycle abnormalities, further contributing to subfertility in these patients [27].
Additionally, increased levels of IL-6 in the preovulatory follicles of EMS patients have been found to result in decreased aromatase activity via the MAPK signal pathway. This reduced aromatase activity leads to the decreased intrafollicular conversion of androstenedione to estrone and the subsequently diminished conversion of androstenedione to testosterone, which is aromatized to estradiol (E2) [29][30]. This decrease in follicular E2 levels may result in fertility issues, including reduced fertilizing capacity [29]. Changes in progesterone levels have also been observed in the FF of EMS patients, suggesting that altered steroidogenesis likely plays a significant role in EMS-associated infertility. However, a direct relationship between infertility and modified progesterone levels has yet to be firmly established [13]. Some researchers have proposed impaired LH production as the primary pathophysiology underlying impaired ovulation [12]. It has been suggested that Gonadotropin-Surge Attenuating Factor (GnSAF), a small polypeptide primarily produced by small follicles in the FF, contributes to decreased LH levels in EMS patients. GnSAF decreases the ability of E2 to sensitize the pituitary to the gonadotropin-releasing hormone, thereby reducing the pituitary’s potential to produce LH. Since estrogen levels are lower in the FF of EMS patients, the antagonistic actions of GnSAF against LH production are likely to result in suboptimal LH levels and impaired ovulation [31].
Various studies have explored changes in the composition of follicular fluid (FF) in women with EMS, including cytokines [32][33], oxidative stress markers [34][35][36][37][38][39], metals [40], growth factors, prostaglandins [41], and macrophage activation patterns [42]. The evidence of oxidative stress (OS) in the follicular microenvironment of these women [34][35][36][37][38][39] suggests that both their peritoneal fluid (PF) and FF may contain substances detrimental to oocyte competence acquisition. In this regard, studies evaluating the effect of FF from infertile women with EMS on the in vitro maturation of bovine oocytes have shown spindle and chromosomal damage [43], which could be prevented by adding antioxidants to the maturation medium, implying a pro-oxidant microenvironment in the ovarian follicles of these women [44]. These alterations may result from OS-induced damage to oocyte cell structures. Evidence also indicates higher levels of 8-Hydroxy-2-Deoxy-Guanosine (8OHdG) in the FF of infertile women with EMS, suggesting oxidative DNA damage in cumulus-oocyte complexes and a possible mechanism contributing to impaired oocyte quality in these patients [35].

4. Dysregulated Immune Function, Hormonal Imbalance, and Oxygen Species in Peritoneal Fluid

Altered conditions within the peritoneal fluid (PF) environment of individuals with EMS typically lead to the growth, proliferation, and inflammation of ectopic endometrial tissue [45]. Changes in both humoral and cell-mediated immunity have been observed in the peritoneal environment of EMS patients [46].
A pronounced inflammatory response, accompanied by heightened reactive species and cytokines, can create an unfavorable pelvic environment, which is reflected in the composition of PF in these women [47][48][49][50]. Studies have indicated that changes in the PF composition of women with EMS, encompassing alterations in cellular and humoral mediators [51][52][53][54], including pro-inflammatory cytokines such as TNF-alpha, interleukin (IL)-1β, IL-6, IL-8, IL-10, IL-13, IL-17, IL-33, Monocyte Chemoattractant Protein (MCP)-1, Macrophage Migration Inhibitory Factor (MIF), and Regulated on Activation, Normal T Cell Expressed and Secreted (RANTES) [55][56][57][58][59][60][61], chemokines [62], angiogenic factors [60][63][64], and increased activated macrophages, T-lymphocytes, and NK cells [45], may lead to chronic inflammation, lesion proliferation, and local hormonal imbalances, which, in turn, can result in poor oocyte quality, impaired sperm motility, embryo toxicity, and reduced endometrial receptivity [65].
Increased levels of E2 in the PF of EMS patients have been shown to stimulate the cyclo-oxygenase-2 (COX-2) enzyme, which subsequently upregulates Prostaglandin E2 (PGE2) production. Prostaglandin E2 is the most potent stimulator of aromatase expression in endometriotic tissue [28], leading to elevated E2 production, further promoting its own proliferation and growth [27]. E2 and PGE2 then upregulate COX-2, initiating a positive feedback loop, resulting in a sustained endometriotic state [66]. Pregnancy-associated plasma protein (PAPP-A), produced by the endometrium, ovary, and placenta, exhibits protease activity towards insulin-like growth-factor-binding protein-4 (IGFBP-4), which normally suppresses follicular E2 production. The protease activity of PAPP-A reduces Insulin-like Growth-Factor-Binding Protein-4 (IGFBP-4) levels, leading to increased levels of free Insulin-like Growth Factors (IGF), which synergize with Follicle-Stimulating Hormone (FSH). IGF, in conjunction with LH, increases androstenedione and testosterone production, which are then aromatized under the influence of FSH into Estrone (E1) and E2, respectively [30][67]. Typically, this conversion would occur in the follicular aromatase. However, due to decreased aromatase activity in the follicles of EMS patients, the conversion takes place in the endometriotic tissue where there is an abnormally increased expression of aromatase, further accentuating the endometriotic state. This is supported by a study reporting increased PAPP-A levels in the peritoneal microenvironment of EMS patients, with the degree of elevation correlating with disease severity [68].
The production of abundant amounts of Reactive Oxygen Species (ROS) by elevated numbers of macrophages and polymorphonuclear leukocytes in the PF of EMS patients has been documented [69]. Two studies have also found increased ROS in EMS PF, although the levels were not significantly different from those in disease-free controls [70][71]. Based on these studies, it is believed that the development of oxidative stress (OS) in the local peritoneal environment may be one of the contributing factors in the chain of events leading to EMS-associated infertility [72]. It has also been proposed that redox levels may modulate the severity, dynamics, and progression of the disease. The increase in both the number and activity of macrophages in EMS is accompanied by the release of additional cytokines and other immune mediators, such as nitric oxide (NO) [73]. Nitric oxide is a free radical and a regulator of apoptosis [74]. Low levels of NO play an essential role in ovarian function and implantation. However, higher levels of NO and nitric oxide synthase (NOS) are observed in the endometrium of women with EMS [75].
In the PF of women with EMS-associated infertility, the total antioxidant capacity is reduced, and individual antioxidant enzymes, such as superoxide dismutase, are significantly lower [76]. Moreover, lipid peroxide levels are highest among patients with EMS, suggesting a potential role for ROS in the development of EMS [47]. An imbalance between antioxidants and oxidants has been reported in many studies investigating PF from women with EMS. It has also been theorized that ROS may contribute to the formation of adhesions associated with EMS. Although adhesions due to EMS are known to reduce fertility, the precise mechanism remains incompletely understood [77]. Additionally, alterations in folliculogenesis, likely caused by OS, may impair oocyte quality and have been proposed as a cause of subfertility associated with EMS. Levels of the OS marker, 8-hydroxy 1-deoxyguanosine, were higher in patients with EMS compared to those with tubal, male-factor, or idiopathic infertility [78].
OS has also been shown to induce genomic and mitochondrial DNA damage [44], directly resulting in reduced fertility [79]. It was observed that spermatozoa exhibited heightened DNA fragmentation when exposed to peritoneal fluid (PF) from patients with EMS. Moreover, the degree of fragmentation increased in correlation with the stage of EMS and the duration of infertility [80]. Similarly, oocytes displayed increased DNA damage when subjected to PF from EMS patients, with the extent of damage being influenced by the duration of exposure to the PF [81]. As anticipated, embryos cultivated in the PF of EMS patients also exhibited DNA fragmentation [82]. This elevated DNA damage in sperm, oocytes, and resulting embryos is believed to contribute to the higher rates of miscarriages, as well as fertilization and implantation failures, observed in individuals with EMS [80].

5. Sperms Dysfunction

The presence of activated macrophages, cytokines, elevated TNF-alpha levels, heightened growth factors, and oxidative stress (OS) within the peritoneal fluid (PF) of infertile women affected by EMS can potentially exert toxic effects on sperm function [82][83][84][85]. These altered factors may lead to reduced sperm motility [84][86], induce sperm DNA fragmentation [85], promote abnormal sperm acrosome reactions [87], disrupt sperm membrane permeability or integrity [88], impair the interaction between sperm and the uterine tube epithelium [89], and hinder sperm–oocyte fusion [83]. These mechanisms represent additional potential contributors to infertility in individuals with EMS.

6. Implantation Failure and Impaired Endometrial Receptivity

Uterine receptivity refers to the capacity of the endometrium to facilitate the normal implantation of an embryo [90]. When uterine receptivity is compromised, it can lead to various reproductive issues, ranging from the inability of embryos to implant (resulting in infertility) to incomplete or insufficient implantation (which may result in miscarriages). Histologically, uterine receptivity is established during the mid-luteal phase, commonly referred to as the “window of implantation” (WOI). This critical period is characterized by the occurrence of decidualization, a process that takes place in each menstrual cycle independently of the presence of an embryo [91][92]. However, conditions like EMS can disrupt this delicate process. EMS can interfere with uterine receptivity through various mechanisms, including the dysregulation of important signaling pathways and molecules in endometrial stromal cells, alterations in the expression of genes within the endometrium, changes in cell physiology, and the development of vascular abnormalities, among other factors [91].
In a well-functioning endometrium, the signaling of progesterone (P4) and estrogen is intricately synchronized, following a pattern dictated by the phase of the menstrual cycle. This coordination plays a vital role to maintain a regular menstrual cycle, facilitating successful embryo implantation, and fostering the progression of pregnancy [93]. During the proliferative phase, estrogen stimulates the proliferation of epithelial cells, whereas P4 counters the effects of estrogen, marking the onset of the secretory phase. This phase change prompts stromal cells to undergo decidualization. The disruption in the balance of these hormones—characterized by P4 resistance and an overwhelming influence of estrogen dominance—leads to EMS-associated infertility [93][94].
P4 serves as a critical regulatory hormone, preparing the uterus for implantation and sustaining pregnancy. Its effects hinge on the progesterone receptor (PR), with two subtypes (PR-A and PR-B) acting as ligand-activated transcription factors. Their absence, as seen in PR-specific knockout, leads to pregnancy failure due to unchecked estrogen-driven epithelial cell growth and compromised stromal decidualization [95]. Female mice lacking both PRA and PRB experience reproductive issues, underscoring the importance of epithelial PR expression for successful embryo implantation, the containment of estrogen-driven epithelial proliferation, and stromal decidualization. When the endometrium responds inadequately to P4, it is termed P4 resistance, notably seen in EMS as an inability to activate PR [96]. This receptor expression links to various transcriptional pathways. Chromatin-remodeling protein ARID1A, integral to steroid hormone signaling, potentially regulates PR as a target gene. Reduced ARID1A levels are associated with infertile EMS patients’ endometrium [97].
The P4-triggered Ihh-COUP-TFII signaling axis orchestrates essential epithelial–stromal interactions for embryo implantation and stromal decidualization. Indian Hedgehog (Ihh) ligands engage patched receptors (Ptch1 and Ptch2), heightening COUP-TFII expression in stromal cells. COUP-TFII, an orphan nuclear receptor, influences proper embryo implantation and decidualization. Ihh’s orchestration of uterine receptivity and decidualization, via pathways like Ihh-Ptch1-COUP-TFII-Hand2-Bmp2 and Wnt4 signaling, is hindered in EMS [98]. EMS-associated molecular signaling disruptions, such as KRAS activation and elevated SIRT1/BCL6, may impede Ihh signaling in the endometrium. This signaling axis significantly mediates PR, thus impaired Ihh signaling contributes to EMS-associated progesterone resistance [99].
Wnt signaling, particularly the Wnt/β–catenin pathway, significantly contributes to blastocyst activation, uterine development, and decidualization. During mouse uterine decidualization, Wnt signaling dynamically expresses diverse Wnt ligands, Frizzled Receptors (Fzd), inhibitors (Dkk1, Sfrps), and transcriptional activators (β-catenin, etc.). Bone morphogenetic protein 2 (Bmp2), the downstream of PR, acts as a key paracrine factor, transmitting embryonic adhesion signals from the epithelium to stroma, initiating decidualization. Bmp2’s role in decidualization through FK506-binding proteins (Fkbps) and Wnt ligands is evident in conditional knockout mouse models. Uterine β-catenin knockout impairs endometrial decidualization [100]. PR interacts with nuclear transcription factors to regulate decidual processes. FOXO1 collaborates with PR to promote the expression of decidua-related genes (IGFBP1, PRL). Reduced FOXO1 expression in EMS patients’ secretory endometrium is attributed to hyperactive PI3K/AKT pathways [101][102]. Mutations in PR lead to sterility in mice, characterized by diminished or absent ovulation, uterine hyperplasia, the absence of endometrial decidualization, and the restricted development of mammary glands [93].
Estrogen plays a pivotal role in regulating the transition of the endometrium into a receptive state. In a mice model of delayed implantation treated with P4, researchers observed that the receptive window’s duration was extended with lower estrogen levels, but rapidly shortened with higher concentrations [103]. Thus, the estrogen levels dictate the receptive window’s duration in the uterus. EMS is an estrogen-dependent gynecological disorder characterized by excessive estrogen signaling, leading to an elevated production of 17-β estradiol (E2). This affects both the eutopic endometrium and ectopic sites. Dysregulated 17β-HSD1 expression, rather than aromatase anomalies, has been linked to hyperestrogenism [104].
Maintaining an optimal balance between pro- and anti-inflammatory factors at the maternal–fetal interface is crucial for a successful pregnancy. Excessive inflammation is unfavorable for embryo implantation. Both peripheral blood (PB) and peritoneal fluid (PF) in EMS patients contain an array of substances secreted by endometriotic implants and immune cells, such as growth factors, steroid hormones, and inflammatory agents, creating an inflammatory microenvironment [105][106]. The profile of pro- and anti-inflammatory cytokines evolves dynamically in EMS patients. In EMS, the cytokine profile in PF shifts from unfavorable (e.g., IL-1β, IFN-γ, and TNF-alpha) to favorable (e.g., IL-4, IL-10, and TGFβ) for pregnancy as the condition progresses [107][108]. Elevated levels of TNF-alpha, IL-1, IL-6, and IL-17 in PF are generally observed in EMS patients [59]. Research by Lessey et al. revealed that PF negatively affects the endometrium (specifically LIF and integrin αvβ3) in EMS-afflicted women [109], suggesting that inflammatory cytokines in PF directly impact the endometrium. The study also found higher levels of pro-inflammatory cytokines (IL-1α, IL-1β, and IL-6) in the endometrial fluid of EMS patients [110].
The immune cells implicated in the initiation and progression of endometrial lesions encompass macrophages, neutrophils, NK cells, dendritic cells, and regulatory T cells (Treg). These immune cells release chemokines and cytokines, crucial for communication that influences endometrial receptivity for embryo implantation. Emerging evidence suggests the altered immune status in both endometrium and peritoneal environment in EMS patients, contributing to infertility and pregnancy failure [111][112]. Uterine NK (uNK) cells are crucial for successful embryo implantation and pregnancy. They contribute in embryo bio-sensing and impact endometrial fate decisions during implantation [113]. In cases of EMS, elevated CD16+ uNK cells might lead to an increased infertility risk due to an adverse inflammatory environment affecting implantation and decidualization [114].
Methylation changes in the Human Homeobox A10 (HOXA10) genes are significant as their dysregulation can contribute to EMS. HOXA10 expression is regulated by estrogen and progesterone [107]. These genes are pivotal for normal endometrial changes during the menstrual cycle, governing growth, differentiation, and embryo implantation [114][115]. In EMS patients, reduced HOXA10 expression during the secretory phase leads to decreased uterine receptivity and related infertility [113][115].

References

  1. Giudice, L.C. Clinical practice. Endometriosis. N. Engl. J. Med. 2010, 362, 2389–2398.
  2. Cousins, F.L.; McKinnon, B.D.; Mortlock, S.; Fitzgerald, H.C.; Zhang, C.; Montgomery, G.W.; Gargett, C.E. New concepts on the etiology of endometriosis. J. Obstet. Gynaecol. Res. 2023, 49, 1090–1105.
  3. Akande, V.A.; Hunt, L.P.; Cahill, D.J.; Jenkins, J.M. Differences in time to natural conception between women with unexplained infertility and infertile women with minor endometriosis. Hum. Reprod. 2004, 19, 96–103.
  4. Prescott, J.; Farland, L.V.; Tobias, D.K.; Gaskins, A.J.; Spiegelman, D.; Chavarro, J.E.; Rich-Edwards, J.W.; Barbieri, R.L.; Missmer, S.A. A prospective cohort study of endometriosis and subsequent risk of infertility. Hum. Reprod. 2016, 31, 1475–1482.
  5. Haydardedeoglu, B.; Zeyneloglu, H.B. The impact of endometriosis on fertility. Women’s Health 2015, 11, 619–623.
  6. Bjorkman, S.; Taylor, H.S. MicroRNAs in endometriosis: Biological function and emerging biomarker candidates. Biol. Reprod. 2019, 100, 1135–1146.
  7. Zhang, Y.; Zhang, H.; Yan, L.; Liang, G.; Zhu, C.; Wang, Y.; Ji, S.; He, C.; Sun, J.; Zhang, J. Exosomal microRNAs in tubal fluid may be involved in damage to tubal reproductive function associated with tubal endometriosis. Reprod. Biomed. Online 2023, 47, 103249.
  8. ASRM—Practice Committee of the American Society for Reproductive Medicine. Endometriosis and infertility: A committee opinion. Fertil. Steril. 2012, 98, 591–598.
  9. Rosales, A.A.L.; Rodríguez, J.A.L.; Mendoza, A.A.; Medina, A.L.; González, E.M.; Rodríguez, R.A. Factors Causing Infertility in Women Diagnosed with Endometriosis. Int. J. Med. Sci. Clin. Res. 2022, 2, 1099–1103.
  10. Gupta, S.; Goldberg, J.M.; Aziz, N.; Goldberg, E.; Krajcir, N.; Agarwal, A. Pathogenic mechanisms in endometriosis-associated Infertility. Fertil. Steril. 2008, 90, 247–257.
  11. Gruber, T.M.; Mechsner, S. Pathogenesis of Endometriosis: The Origin of Pain and Subfertility. Cells 2021, 10, 1381.
  12. Cahill, D.J.; Wardle, P.G.; Maile, L.A.; Harlow, C.R.; Hull, M.G. Pituitary–ovarian dysfunction as a cause for endometriosis-associated and unexplained infertility. Hum. Reprod. 1995, 10, 3142–3146.
  13. Pellicer, A.; Albert, C.; Garrido, N.; Navarro, J.; Remohí, J.; Simόn, C. The pathophysiology of endometriosis-associated infertility: Follicular environment and embryo quality. J. Reprod. Fertil. Suppl. 2000, 55, 109–119.
  14. Garrido, N.; Pellicer, A.; Remohi, J.; Simon, C. Uterine and ovarian function in endometriosis. Semin. Reprod. Med. 2003, 21, 183–192.
  15. Doody, M.C.; Gibbons, W.E.; Buttram, V.C., Jr. Linear regression analysis of ultrasound follicular growth series: Evidence for an abnormality of follicular growth in endometriosis patients. Fertil. Steril. 1988, 49, 47–51.
  16. Tummon, I.S.; Maclin, V.M.; Radwanska, E.; Binor, Z.; Dmowski, W.P. Occult ovulatory dysfunction in women with minimal endometriosis or unexplained infertility. Fertil. Steril. 1988, 50, 716–720.
  17. Trinder, J.; Cahill, D.J. Endometriosis and infertility: The debate continues. Hum. Fertil. 2002, 5, S21–S27.
  18. Lachapelle, M.H.; Hemmings, R.; Roy, D.C.; Falcone, T.; Miron, P. Flow cytometric evaluation of leukocyte subpopulations in the follicular fluids of infertile patients. Fertil. Steril. 1996, 65, 1135–1140.
  19. Pellicer, A.; Albert, C.; Mercader, A.; Bonilla-Musoles, F.; Remohi, J.; Simon, C. The follicular and endocrine environment in women with endometriosis: Local and systemic cytokine production. Fertil. Steril. 1998, 70, 425–431.
  20. Garrido, N.; Navarro, J.; Remohi, J.; Simon, C.; Pellicer, A. Follicular hormonal environment and embryo quality in women with endometriosis. Hum. Reprod. Update 2000, 6, 67–74.
  21. Wunder, D.M.; Mueller, M.D.; Birkhauser, M.H.; Bersinger, N.A. Increased ENA-78 in the follicular fluid of patients with endometriosis. Acta Obstet. Gynecol. Scand. 2006, 85, 336–342.
  22. Lucena, E.; Cubillos, J. Immune abnormalities in endometriosis compromising fertility in IVF-ET patients. J. Reprod. Med. 1999, 44, 458–464.
  23. Carlberg, M.; Nejaty, J.; Froysa, B.; Guan, Y.; Soder, O.; Bergqvist, A. Elevated expression of tumour necrosis factor alpha in cultured granulosa cells from women with endometriosis. Hum. Reprod. 2000, 15, 1250–1255.
  24. Naz, R.K.; Butlera, A.; Witta, B.R.; Barad, D.; Menge, A.C. Levels of interferon-gamma and tumor necrosis factor-alpha in sera and cervical mucus of fertile and infertile women: Implication in infertility. J. Reprod. Immunol. 1995, 29, 105–117.
  25. Barrier, B.F.; Bates, G.W.; Leland, M.M.; Leach, D.A.; Robinson, R.D.; Propst, A.M. Efficacy of anti-tumor necrosis factor therapy in the treatment of spontaneous endometriosis in baboons. Fertil. Steril. 2004, 81, 775–779.
  26. Shakiba, K.; Falcone, T. Tumour necrosis factor-a blockers: Potential limitations in the management of advanced endometriosis? A Case Report. Hum. Reprod. 2006, 21, 2417–2420.
  27. Saito, H.; Seino, T.; Kaneko, T.; Nakahara, K.; Toya, M.; Kurachi, H. Endometriosis and oocyte quality. Gynecol. Obstet. Invest. 2002, 53 (Suppl. S1), 46–51.
  28. Perrin, G.Q.; Johnson, H.M.; Subramaniam, P.S. Mechanism of interleukin-10 inhibition of T-helper cell activation by superantigen at the level of the cell cycle. Blood 1999, 93, 208–216.
  29. Yoshida, S.; Harada, T.; Iwabe, T.; Taniguchi, F.; Mitsunari, M.; Yamauchi, N.; Deura, I.; Horie, S.; Terakawa, N. A combination of interleukin-6 and its soluble receptor impairs sperm motility: Implications in infertility associated with endometriosis. Hum. Reprod. 2004, 19, 1821–1825.
  30. Ulukus, M.; Cakmak, H.; Arici, A. The role of endometrium in endometriosis. J. Soc. Gynecol. Investig. 2006, 13, 467–476.
  31. Dafopoulos, K.; Kotsovassilis, C.G.; Milingos, S.; Kallitsaris, A.; Galazios, G.; Zintzaras, E.; Sotiros, P.; Messinis, I.E. Changes in pituitary sensitivity to GnRH in estrogentreated post-menopausal women: Evidence that gonadotrophin surge attenuating factor plays a physiological role. Hum. Reprod. 2004, 19, 1985–1992.
  32. Singh, A.K.; Dutta, M.; Chattopadhyay, R.; Chakravarty, B.; Chaudhury, K. Intrafollicular interleukin-8, interleukin-12, and adrenomedullin are the promising prognostic markers of oocyte and embryo quality in women with endometriosis. J. Assist. Reprod. Genet. 2016, 33, 1363–1372.
  33. Wu, G.; Bersinger, N.A.; Mueller, M.D.; von Wolff, M. Intrafollicular inflammatory cytokines but not steroid hormone concentrations are increased in naturally matured follicles of women with proven endometriosis. J. Assist. Reprod. Genet. 2017, 34, 357–364.
  34. Choi, Y.S.; Cho, S.; Seo, S.K.; Park, J.H.; Kim, S.H.; Lee, B.S. Alteration in the intrafollicular thiol-redox system in infertile women with endometriosis. Reproduction 2015, 149, 155–162.
  35. Da Broi, M.G.; de Albuquerque, F.O.; de Andrade, A.Z.; Cardoso, R.L.; Jordão Junior, A.A.; Navarro, P.A. Increased concentration of 8-hydroxy-2′-deoxyguanosine in follicular fluid of infertile women with endometriosis. Cell Tissue. Res. 2016, 366, 231–242.
  36. Huang, B.; Li, Z.; Ai, J.; Zhu, L.; Li, Y.; Jin, L.; Zhang, H. Antioxidant capacity of follicular fluid from patients undergoing in vitro fertilization. Int. J. Clin. Exp. Pathol. 2014, 7, 2273–2282.
  37. Liu, F.; He, L.; Liu, Y.; Shi, Y.; Du, H. The expression and role of oxidative stress markers in the serum and follicular fluid of patients with endometriosis. Clin. Exp. Obstet. Gynecol. 2013, 40, 372–376.
  38. Singh, A.K.; Chattopadhyay, R.; Chakravarty, B.; Chaudhury, K. Markers of oxidative stress in follicular fluid of women with endometriosis and tubal infertility undergoing IVF. Reprod. Toxicol. 2013, 42, 116–124.
  39. 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.
  40. Liu, M.; Wu, K.; Wu, Y. The emerging role of ferroptosis in female reproductive disorders. Biomed. Pharmacother. 2023, 166, 115415.
  41. Minami, T.; Tsuzuki, Y.; Tanaka, Y.; Kitawaki, J.; Mori, T. The Tpl2-MEK pathway plays a critical role in spheroid-cultured endometriotic stromal cells. Am. J. Reprod. Immunol. 2023, 89, e13689.
  42. Lamaita, R.M.; Pontes, A.; Belo, A.V.; Caetano, J.P.; Andrade, S.P.; Cândido, E.B.; Carneiro, M.M.; Silva-Filho, A.L. Evaluation of N-acetilglucosaminidase and myeloperoxidase activity in patients with endometriosis-related infertility undergoing intracytoplasmic sperm injection. J. Obstet. Gynaecol. Res. 2012, 38, 810–816.
  43. Da Broi, M.G.; Malvezzi, H.; Paz, C.C.; Ferriani, R.A.; Navarro, P.A. Follicular fluid from infertile women with mild endometriosis may compromise the meiotic spindles of bovine metaphase II oocytes. Hum. Reprod. 2014, 29, 315–323.
  44. Giorgi, V.S.; Da Broi, M.G.; Paz, C.C.; Ferriani, R.A.; Navarro, P.A. N-Acetyl-Cysteine and l-Carnitine Prevent Meiotic Oocyte Damage Induced by Follicular Fluid from Infertile Women with Mild Endometriosis. Reprod. Sci. 2016, 23, 342–351.
  45. Lebovic, D.I.; Mueller, M.D.; Taylor, R.N. Immunobiology of endometriosis. Fertil. Steril. 2001, 75, 1–10.
  46. Oosterlynck, D.J.; Meuleman, C.; Waer, M.; Koninckx, P.R.; Vandeputte, M. Immunosuppressive activity of peritoneal fluid in women with endometriosis. Obstet. Gynecol. 1993, 82, 206–212.
  47. Szczepańska, M.; Koźlik, J.; Skrzypczak, J.; Mikołajczyk, M. Oxidative stress may be a piece in the endometriosis puzzle. Fertil. Steril. 2003, 79, 1288–1293.
  48. Didzoiokaite, G.; Bitiute, G.; Gudaite, J.; Kvedariene, V. Oxidative Stress as a Potential Underlying Cause of Minimal and Mild Endometriosis-Related Infertility. Int. J. Mol. Sci. 2023, 24, 3809.
  49. Gupta, S.; Agarwal, A.; Krajcir, N.; Alvarez, J.G. Role of oxidative stress in endometriosis. Reprod. Biomed. Online 2006, 13, 126–134.
  50. Ruder, E.H.; Hartman, T.J.; Blumberg, J.; Goldman, M.B. Oxidative stress and antioxidants: Exposure and impact on female fertility. Hum. Reprod. Update 2008, 14, 345–357.
  51. Eisermann, J.; Gast, M.J.; Pineda, J.; Odem, R.R.; Collins, J.L. Tumor necrosis factor in peritoneal fluid of women undergoing laparoscopic surgery. Fertil. Steril. 1988, 50, 573–579.
  52. Chen, S.; Liu, Y.; Zhong, Z.; Wei, C.; Liu, Y.; Zhu, X. Peritoneal immune microenvironment of endometriosis: Role and therapeutic perspectives. Front. Immunol. 2023, 14, 1134663.
  53. Cheong, Y.C.; Shelton, J.B.; Laird, S.M.; Richmond, M.; Kudesia, G.; Li, T.C.; Ledger, W.L. IL-1, IL-6 and TNF-alpha concentrations in the peritoneal fluid of women with pelvic adhesions. Hum. Reprod. 2002, 17, 69–75.
  54. Jørgensen, H.; Hill, A.S.; Beste, M.T.; Kumar, M.P.; Chiswick, E.; Fedorcsak, P.; Isaacson, K.B.; Lauffenburger, D.A.; Griffith, L.G.; Qvigstad, E. Peritoneal fluid cytokines related to endometriosis in patients evaluated for infertility. Fertil. Steril. 2017, 107, 1191–1199.
  55. Ahn, S.H.; Edwards, A.K.; Singh, S.S.; Young, S.L.; Lessey, B.A.; Tayade, C. IL-17A Contributes to the Pathogenesis of Endometriosis by Triggering Proinflammatory Cytokines and Angiogenic Growth Factors. J. Immunol. 2015, 195, 2591–2600.
  56. Bersinger, N.A.; von Roten, S.; Wunder, D.M.; Raio, L.; Dreher, E.; Mueller, M.D. PAPP-A and osteoprotegerin, together with interleukin-8 and RANTES, are elevated in the peritoneal fluid of women with endometriosis. Am. J. Obstet. Gynecol. 2006, 195, 103–108.
  57. Harada, T.; Yoshioka, H.; Yoshida, S.; Iwabe, T.; Onohara, Y.; Tanikawa, M.; Terakawa, N. Increased interleukin-6 levels in peritoneal fluid of infertile patients with active endometriosis. Am. J. Obstet. Gynecol. 1997, 176, 593–597.
  58. Sikora, J.; Mielczarek-Palacz, A.; Kondera-Anasz, Z. Imbalance in cytokines from interleukin-1 family–role in pathogenesis of endometriosis. Am. J. Reprod. Immunol. 2012, 68, 138–145.
  59. Wang, X.M.; Ma, Z.Y.; Song, N. Inflammatory cytokines IL-6, IL- 10, IL-13, TNF-α and peritoneal fluid flora were associated with infertility in patients with endometriosis. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 2513–2518.
  60. Yoshino, O.; Osuga, Y.; Koga, K.; Hirota, Y.; Tsutsumi, O.; Yano, T.; Morita, Y.; Momoeda, M.; Fujiwara, T.; Kugu, K.; et al. Concentrations of interferon-gamma-induced protein-10 (IP-10), an antiangiogenic substance, are decreased in peritoneal fluid of women with advanced endometriosis. Am. J. Reprod. Immunol. 2003, 50, 60–65.
  61. Huang, X.; Wu, L.; Pei, T.; Liu, D.; Liu, C.; Luo, B.; Xiao, L.; Li, Y.; Wang, R.; Ouyang, Y.; et al. Single-cell transcriptome analysis reveals endometrial immune microenvironment in minimal/mild endometriosis. Clin. Exp. Immunol. 2023, 212, 285–295.
  62. Margari, K.M.; Zafiropoulos, A.; Hatzidaki, E.; Giannakopoulou, C.; Arici, A.; Matalliotakis, I. Peritoneal fluid concentrations of β-chemokines in endometriosis. Eur. J. Obstet. Gynecol. Reprod. Biol. 2013, 169, 103–107.
  63. Cosín, R.; Gilabert-Estellés, J.; Ramón, L.A.; Gómez-Lechón, M.J.; Gilabert, J.; Chirivella, M.; Braza-Boïls, A.; España, F.; Estellés, A. Influence of peritoneal fluid on the expression of angiogenic and proteolytic factors in cultures of endometrial cells from women with endometriosis. Hum Reprod. 2010, 25, 398–405.
  64. Kianpour, M.; Nematbakhsh, M.; Ahmadi, S.M.; Jafarzadeh, M.; Hajjarian, M.; Pezeshki, Z.; Safari, T.; Eshraghi-Jazi, F. Serum and peritoneal fluid levels of vascular endothelial growth factor in women with endometriosis. Int. J. Fertil. Steril. 2013, 7, 96–99.
  65. Miller, J.E.; Ahn, S.H.; Monsanto, S.P.; Khalaj, K.; Koti, M.; Tayade, C. Implications of immune dysfunction on endometriosis associated infertility. Oncotarget 2017, 8, 7138–7147.
  66. Attar, E.; Bulun, S.E. Aromatase and other steroidogenic genes in endometriosis: Translational aspects. Hum. Reprod. Update 2006, 12, 49–56.
  67. Fortune, J.E.; Rivera, G.M.; Yang, M.Y. Follicular development: The role of the follicular microenvironment in selection of the dominant follicle. Anim. Reprod. Sci. 2004, 82–83, 109–126.
  68. Arici, A.; Matalliotakis, I.; Goumenou, A.; Koumantakis, G.; Fragouli, Y.; Mahutte, N.G. Increased pregnancy-associated plasma protein-A (PAPP-A) concentrations in peritoneal fluid of women with endometriosis. Am. J. Reprod. Immunol. 2003, 49, 70–74.
  69. Zeller, J.M.; Henig, I.; Radwanska, E.; Dmowski, W.P. Enhancement of human monocyte and peritoneal macrophage chemiluminescence activities in women with endometriosis. Am. J. Reprod. Immunol. Microbiol. 1987, 13, 78–82.
  70. Bedaiwy, M.A.; Falcone, T.; Sharma, R.K.; Goldberg, J.M.; Attaran, M.; Nelson, D.R.; Agarwal, A. Prediction of endometriosis with serum and peritoneal fluidmarkers: A prospective controlled trial. Hum. Reprod. 2002, 17, 426–431.
  71. Wang, Y.; Sharma, R.K.; Falcone, T.; Goldberg, J.; Agarwal, A. Importance of reactive oxygen species in the peritoneal fluid of women with endometriosis or idiopathic infertility. Fertil. Steril. 1997, 68, 826–830.
  72. Dai, Y.; Lin, X.; Liu, N.; Shi, L.; Zhuo, F.; Huang, Q.; Gu, W.; Zhao, F.; Zhang, Y.; Zhang, Y.; et al. Integrative analysis of transcriptomic and metabolomic profiles reveals abnormal phosphatidylinositol metabolism in follicles from endometriosis-associated infertility patients. J. Pathol. 2023, 260, 248–260.
  73. Dong, M.; Shi, Y.; Cheng, Q.; Hao, M. Increased nitric oxide in peritoneal fluid from women with idiopathic infertility and endometriosis. J. Reprod. Med. 2001, 46, 887–891.
  74. Chung, H.T.; Pae, H.O.; Choi, B.M.; Billiar, T.R.; Kim, Y.M. Nitric oxide as a bioregulator of apoptosis. Biochem. Biophys. Res. Commun. 2001, 282, 1075–1079.
  75. Khorram, O.; Lessey, B.A. Alterations in expression of endometrial endothelial nitric oxide synthase and alpha(v)beta (3) integrin in women with endometriosis. Fertil. Steril. 2002, 78, 860–864.
  76. Osman, H.G.; El-Refaey, A.A.A.; Aziz, A.A.F.A.; El-Sokkaryl, A.M.A.; El-Saeed, R.A. Leptin and antioxidant profile in infertile women with endometriosis. J. Endometr. 2010, 2, 135–143.
  77. Alpay, Z.; Saed, G.M. Diamond MP. Female infertility and free radicals: Potential role in adhesions and endometriosis. J. Soc. Gynecol. Investig. 2006, 13, 390–398.
  78. Aitken, R.J.; Krausz, C. Oxidative stress, DNA damage and the Y chromosome. Reproduction 2001, 122, 497–506.
  79. Guerin, P.; El Mouatassim, S.; Menezo, Y. Oxidative stress and protection against reactive oxygen species in the pre-implantation embryo and its surroundings. Hum. Reprod. Update 2001, 7, 175–189.
  80. Mansour, G.; Goldberg, J.; Agarwal, A.; Sharma, R.; Mahfouz, R.; Falcone, T. Correlation between sperm DNA damage, stage of endometriosis and the duration of infertility. In Proceedings of the American Society of Reproductive Medicine 63rd Annual Meeting, Washington, DC, USA, 13–17 October 2007.
  81. Mansour, G.; Agarwal, A.; Radwan, E.; Sharma, R.; Goldberg, J.; Falcone, T. DNA damage in metaphase II oocytes is induced by peritoneal fluid from endometriosis patients. In Proceedings of the American Society of Reproductive Medicine 63rd Annual Meeting, Washington, DC, USA, 13–17 October 2007.
  82. Mansour, G.; Radwan, E.; Sharma, R.; Agarwal, A.; Falcone, T.; Goldberg, J. DNA damage to embryos incubated in the peritoneal fluid of patients with endometriosis: Role in infertility. In Proceedings of the American Society of Reproductive Medicine 63rd Annual Meeting, Washington, DC, USA, 13–17 October 2007.
  83. Aeby, T.C.; Huang, T.; Nakayama, R.T. The effect of peritoneal fluid from patients with endometriosis on human sperm function in vitro. Am. J. Obstet. Gynecol. 1996, 174, 1779–1783.
  84. Liu, Y.; Luo, L.; Zhao, H. Changes of cytokines levels in peritoneal fluids of patients with endometriosis and its effect on reproductive activity. J. Tongji. Med. Univ. 2000, 20, 163–165.
  85. Mansour, G.; Aziz, N.; Sharma, R.; Falcone, T.; Goldberg, J.; Agarwal, A. The impact of peritoneal fluid from healthy women and from women with endometriosis on sperm DNA and its relationship to the sperm deformity index. Fertil. Steril. 2009, 92, 61–67.
  86. Oral, E.; Arici, A.; Olive, D.L.; Huszar, G. Peritoneal fluid from women with moderate or severe endometriosis inhibits sperm motility: The role of seminal fluid components. Fertil. Steril. 1996, 66, 787–792.
  87. Arumugam, K. Endometriosis and infertility: Raised iron concentration in the peritoneal fluid and its effect on the acrosome reaction. Hum. Reprod. 1994, 9, 1153–1157.
  88. Said, T.M.; Agarwal, A.; Falcone, T.; Sharma, R.K.; Bedaiwy, M.A.; Li, L. Infliximab may reverse the toxic effects induced by tumor necrosis factor alpha in human spermatozoa: An in vitro model. Fertil. Steril. 2005, 83, 1665–1673.
  89. Reeve, L.; Lashen, H.; Pacey, A.A. Endometriosis affects sperm-endosalpingeal interactions. Hum. Reprod. 2005, 20, 448–451.
  90. Lessey, B.A.; Young, S.L. What exactly is endometrial receptivity? Fertil. Steril. 2019, 11, 611–617.
  91. Ticconi, C.; Di Simone, N.; Campagnolo, L.; Fazleabas, A. Clinical consequences of defective decidualization. Tissue Cell 2021, 72, 101586.
  92. Lédée, N.; Petitbarat, M.; Prat-Ellenberg, L.; Dray, G.; Cassuto, G.-N.; Chevrier, L.; Kazhalawi, A.; Vezmar, K.; Chaouat, G. The uterine immune profile: A method for individualizing the management of women who have failed to implant an embryo after IVF/ICSI. J. Reprod. Immunol. 2020, 142, 103207.
  93. Marquardt, R.M.; Kim, T.H.; Shin, J.H.; Jeong, J.W. Progesterone and estrogen signaling in the endometrium: What goes wrong in endometriosis? Int. J. Mol. Sci. 2019, 20, 3822.
  94. Kapoor, R.; Stratopoulou, C.A.; Dolmans, M.M. Pathogenesis of endometriosis: New insights into prospective therapies. Int. J. Mol. Sci. 2021, 22, 11700.
  95. Fernandez-Valdivia, R.; Jeong, J.; Mukherjee, A.; Soyal, S.; Li, J.; Ying, Y.; Demayo, F.; Lydon, J. A mouse model to dissect progesterone signaling in the female reproductive tract and mammary gland. Genesis 2010, 48, 106–113.
  96. McKinnon, B.; Mueller, M.; Montgomery, G. Progesterone Resistance in Endometriosis: An Acquired Property? Trends Endocrinol. Metab. 2018, 29, 535–548.
  97. Kim, H.; Kim, T.; Yoo, J.; Young, S.; Lessey, B.; Ku, B.; Jeong, J. ARID1A and PGR proteins interact in the endometrium and reveal a positive correlation in endometriosis. Biochem. Biophys. Res. Commun. 2021, 550, 151–157.
  98. Zhou, Y.; Peng, Y.; Xia, Q.; Yan, D.; Zhang, H.; Zhang, L.; Chen, Y.; Zhao, X.; Li, J. Decreased Indian hedgehog signaling activates autophagy in endometriosis and adenomyosis. Reproduction 2021, 161, 99–109.
  99. Yoo, J.; Kim, T.; Fazleabas, A.; Palomino, W.; Ahn, S.; Tayade, C.; Schammel, D.; Young, S.; Jeong, J.; Lessey, B. KRAS Activation and over-expression of SIRT1/BCL6 Contributes to the Pathogenesis of Endometriosis and Progesterone Resistance. Sci. Rep. 2017, 7, 6765.
  100. Zhang, L.; Patterson, A.; Zhang, L.; Teixeira, J.; Pru, J. Endometrial stromal beta-catenin is required for steroid-dependent mesenchymal-epithelial cross talk and decidualization. Reprod. Biol. Endocrinol. 2012, 10, 75.
  101. Vasquez, Y.; Mazur, E.; Li, X.; Kommagani, R.; Jiang, L.; Chen, R.; Lanz, R.; Kovanci, E.; Gibbons, W.; DeMayo, F. FOXO1 is required for binding of PR on IRF4, novel transcriptional regulator of endometrial stromal decidualization. Mol. Endocrinol. 2015, 29, 421–433.
  102. Yin, X.; Pavone, M.; Lu, Z.; Wei, J.; Kim, J. Increased activation of the PI3K/AKT pathway compromises decidualization of stromal cells from endometriosis. J. Clin. Endocrinol. Metab. 2012, 97, E35–E43.
  103. Ma, W.; Song, H.; Das, S.; Paria, B.; Dey, S. Estrogen is a critical determinant that specifies the duration of the window of uterine receptivity for implantation. Proc. Natl. Acad. Sci. USA 2003, 100, 2963–2968.
  104. Anupa, G.; Sharma, J.; Roy, K.; Sengupta, J.; Ghosh, D. An assessment of the multifactorial profile of steroid-metabolizing enzymes and steroid receptors in the eutopic endometrium during moderate to severe ovarian endometriosis. Reprod. Biol. Endocrinol. 2019, 17, 111.
  105. Kalu, E.; Sumar, N.; Giannopoulos, T.; Patel, P.; Croucher, C.; Sherriff, E.; Bansal, A. Cytokine profiles in serum and peritoneal fluid from infertile women with and without endometriosis. J. Obstet. Gynaecol. Res. 2007, 33, 490–495.
  106. Skrzypczak, J.; Szczepańska, M.; Puk, E.; Kamieniczna, M.; Kurpisz, M. Peritoneal fluid cytokines and sICAM-1 in minimal endometriosis: Search for discriminating factors between infertility and/or endometriosis. Eur. J. Obstet. Gynecol. Reprod. Biol. 2005, 122, 95–103.
  107. Banerjee, P.; Jana, S.; Pasricha, P.; Ghosh, S.; Chakravarty, B.; Chaudhury, K. Proinflammatory cytokines induced altered expression of cyclooxygenase-2 gene results in unreceptive endometrium in women with idiopathic recurrent spontaneous miscarriage. Fertil. Steril. 2013, 99, 179–187.
  108. Raghupathy, R. Th1-type immunity is incompatible with successful pregnancy. Immunol. Today 1997, 18, 478–482.
  109. Illera, M.; Juan, L.; Stewart, C.; Cullinan, E.; Ruman, J.; Lessey, B. Effect of peritoneal fluid from women with endometriosis on implantation in the mouse model. Fertil. Steril. 2000, 74, 41–48.
  110. Llarena, N.; Richards, E.; Priyadarshini, A.; Fletcher, D.; Bonfield, T.; Flyckt, R. Characterizing the endometrial fluid cytokine profile in women with endometriosis. J. Assist. Reprod. Genet. 2020, 37, 2999–3006.
  111. Patel, B.; Lenk, E.; Lebovic, D.; Shu, Y.; Yu, J.; Taylor, R. Pathogenesis of endometriosis: Interaction between Endocrine and inflammatory pathways. Best Pract. Res. Clin. Obstet. Gynaecol. 2018, 50, 50–60.
  112. Lessey, B.; Lebovic, D.; Taylor, R. Eutopic endometrium in women with endometriosis: Ground zero for the study of implantation defects. Semin. Reprod. Med. 2013, 31, 109–124.
  113. Kong, C.; Ordoñez, A.; Turner, S.; Tremaine, T.; Muter, J.; Lucas, E.; Salisbury, E.; Vassena, R.; Tiscornia, G.; Fouladi-Nashta, A.; et al. Embryo biosensing by uterine natural killer cells determines endometrial fate decisions at implantation. FASEB J. 2021, 35, e21336.
  114. Giuliani, E.; Parkin, K.; Lessey, B.; Young, S.; Fazleabas, A. Characterization of uterine NK cells in women with infertility or recurrent pregnancy loss and associated endometriosis. Am. J. Reprod. Immunol. 2014, 72, 262–269.
  115. Koukoura, O.; Sifakis, S.; Spandidos, D.A. DNA methylation in endometriosis (Review). Mol. Med. Rep. 2016, 13, 2939–2948.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 173
Revisions: 2 times (View History)
Update Date: 24 Nov 2023
1000/1000