Skip to content
BY 4.0 license Open Access Published by De Gruyter July 19, 2019

Endometriosis Pathoetiology and Pathophysiology: Roles of Vitamin A, Estrogen, Immunity, Adipocytes, Gut Microbiome and Melatonergic Pathway on Mitochondria Regulation

  • George Anderson EMAIL logo
From the journal Biomolecular Concepts

Abstract

Endometriosis is a common, often painful, condition that has significant implications for a woman’s fertility. Classically, endometriosis has been conceptualized as a local estrogen-mediated uterine condition driven by retrograde menstruation. However, recent work suggests that endometriosis may be a systemic condition modulated, if not driven, by prenatal processes. Although a diverse array of factors have been associated with endometriosis pathophysiology, recent data indicate that the low body mass index and decreased adipogenesis may be indicative of an early developmental etiology with alterations in metabolic function crucial to endometriosis pathoetiology.

The present article reviews the data on the pathoetiology and pathophysiology of endometriosis, suggesting key roles for alterations in mitochondria functioning across a number of cell types and body systems, including the immune system and gut microbiome. These changes are importantly regulated by decreases in vitamin A and its retinoic acid metabolites as well as increases in mitochondria estrogen receptor-beta and the N-acetylserotonin/melatonin ratio across development. This has treatment and future research implications for this still poorly managed condition, as well as for the association of endometriosis with a number of cancers.

Introduction

Endometrial tissue is usually restricted to the uterus. In endometriosis, however, endometrial tissue can grow in other locations, including the ovaries and fallopian tubes, as well as in nearby and more distant tissue. Consequently, the endometrial tissue in these other areas can result in inflammation and scarring [1]. Pelvic pain is usually the main presenting symptom, being chronic in 50% of patients, with 70% experiencing pelvic pain during menstruation. Infertility is common, being evident in approximately 50% of people presenting with endometriosis [2]. Between 5-10% of western women and about 15% of Asian women will experience endometriosis, most typically with a first presentation at 30-40 years, often as a consequence of difficulties in conceiving [1].

Factors linked to endometriosis pathophysiology include: increased oxidative and nitrosative stress (O&NS), chronic immune inflammation, increased immune tolerance, autoimmunity, t helper (Th)17 cells and interleukin (IL)-17, as well as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) associated activation of the aryl hydrocarbon receptor (AhR) [3]. Estrogen at the estrogen receptor-alpha (ERα) can exacerbate symptoms, with estrogen also having regulatory, and symptomatic, effects via the ERβ. As both of these estrogen receptors can be mitochondria-located, there can be significant changes in mitochondria functioning in endometriosis. Raised O&NS in endometrial tissue is overly compensated by an ERβ-mediated increase in mitochondria superoxide dismutase (SOD)2, leading to mitochondria with heightened levels of oxidative phosphorylation and ATP production. Within mitochondria cytochrome P450 (CYP)1B1 is increased [4], as often occurs in many cancers. Increased brain-derived neurotrophic factor (BDNF) [5] and the activation of its receptor, TrkB, are also associated with endometriosis [6]. As such, endometriosis shows alterations in mitochondria functioning, oxidative stress regulation and increased trophic factors, linking endometriosis with tumor-associated pathophysiology.

Endometriosis, like a growing number of other medical conditions [7], shows alterations in the gut microbiome [8]. The raised levels of depression in endometriosis may be linked to this, given the gut microbiome’s role in depression pathoetiology, especially via a decrease in the short-chain fatty acid (SCFA), butyrate [9]. Very high levels of depression are often evident in women with chronic pelvic pain compared to those with endometriosis who are pain free [10]. As such, some of the biological heterogeneity in endometriosis may be mediated by variations in the levels of pain-linked depression- and stress-induced gut dysbiosis and associated increases in gut permeability.

The current article reviews this diverse array of data on the pathoetiology and pathophysiology of endometriosis and proposes an etiological role for prenatal factors, with an increase in the mitochondria N-acetylserotonin (NAS)/melatonin ratio proposed to link the wide array of previously disparate pieces of data pertaining to endometriosis pathophysiology. Decreased adipocyte levels also have a significant role, within the context of a gut-hepatic-adipocyte-hypothalamus axis, with exosomes being important effectors of change by this axis, due primarily to their differential priming of immune cells. Such a frame of reference better integrates wide bodies of data on the pathoetiology and pathophysiology of endometriosis, including its association with heightened cancer risk.

Endometriosis: Pathophysiology

A wide array of pathophysiological processes have been linked to endometriosis, many of which are shared with other conditions, including increased O&NS, and immune-inflammation. This section summarizes the main bodies of data pertaining to endometriosis pathophysiology.

Oxidative and Nitrosative Stress (O&NS)

Increased levels of O&NS are evident in endometriosis, with O&NS facilitating ectopic endometrium implantation [11, 12]. Heightened levels of O&NS positively correlate with symptom severity [13]. Consequently, a wide array of antioxidants and antioxidant inducers, including vitamins C, vitamin E, melatonin, resveratrol, xanthohumol and green tea’s epigallocatechin- 3-gallate can afford some symptomatic relief in endometriosis [11]. Heightened O&NS may be mediated by stress- and pro-inflammatory cytokine-induced gut dysbiosis and increased gut permeability.

Immune Activation

Across different medical conditions, increased O&NS is commonly associated with higher levels of immune-inflammatory activity and pro-inflammatory cytokines, including IL-1β, IL-6, IL-8, IL-17 and IL-18 [14]. This is similar in endometriosis, with heightened IL-1β and IL-18 levels, linked to oxidative stress-induction of the Nod-like receptor family pyrin domain containing (NLRP)3 inflammasome, which correlates with poor survival upon transition to endometriosis-associated ovarian cancer [15]. IL-10 levels are also increased in endometriosis, mediated by increases in regulatory T (Treg) cells, which become increasingly evident over endometriosis progression [16]. These authors also found increased levels of transforming growth factor (TGF-β) in the peritoneal fluid and the serum. An increase in the anti-inflammatory cytokine, IL-37, in the peritoneal fluid has also been shown in endometriosis, with this cytokine thought to contribute to a decrease in inflammatory activity [17]. As such, a mixed immune response is evident in endometriosis, with both pro- and anti-inflammatory components.

A number of immune cell subtypes are altered in endometriosis, including: increased Treg cells in the peritoneal fluid coupled to increased TGF-β and a relatively lower percentage of Th17 cells in both the peritoneal fluid and peripheral blood, with these changes positively correlating with severity [18]. However, other data shows Th17 to be increased in the peritoneal fluid and blood in endometriosis, also in correlation with levels of severity [19]. Such contrasting results are seen to reflect endometriosis heterogeneity, although it may more likely reflect the complex sets of dynamic interactions of immune cell types and factors that act to regulate them, as well as reflecting changes in other systems, such as stress and pro-inflammatory cytokines increasing gut permeability, which can drive wider immune cell changes. Heightened levels of Th17 cell activity also increases the activation of IL-17 producing gammaDelta t cells (γδ-T cells), which are another major contributor to circulating IL-17 levels. This may be particularly relevant in circumstances where gut permeability is increased, given the high levels of γδ-T cells in proximity to the gut [20]. Overall, the heightened levels of pro-and anti-inflammatory cytokines and immune cells may reflect dysregulation in the how inflammatory and anti-inflammatory processes are regulated, as well as alterations in other immune regulatory processes, such the gut microbiome and gut permeability.

Aryl Hydrocarbon Receptor (AhR)

AhR activation is also important to endometriosis pathophysiology, with data suggesting that AhR single nucleotide polymorphisms (SNPs) modulate endometriosis risk [21]. AhR activation can have differential consequences in different cell types, including from different effects arising from exogenous versus endogenous and induced ligands [22]. As well as its regulatory effects, the AhR can significantly interact with, and modulate, many of the pathophysiological factors associated with endometriosis. Polychlorinated biphenyls (PCBs), with a dioxin-like structure that activate the AhR, increase levels of estrogen and 17β-hydroxysteroid dehydrogenase (HSD)-7 activity as well as pro-inflammatory cytokines in a murine model of endometriosis [23]. Antagonism of the AhR prevented such PCBs effects, suggesting a role for environmental toxins, via the AhR, in the pathophysiology of endometriosis. This is supported by similar data in a rodent model of endometriosis [24]. AhR expression on mast cells may also be important, with mast cells in close association with endometrial tissue [25].

Indoleamine 2,3-dioxygenase, Kynurenine and Melatonergic Pathways

Recent data shows endometrial tissue to have increased levels of pro-inflammatory cytokine-induced indoleamine 2,3-dioxygenase (IDO), leading to kynurenine pathway activation. Increased IDO and kynurenine are evident in endometrial tissue, with kynurenine activating the AhR in IL-17 and IL-10 positive mast cells [25]. IDO can also contribute to the metabolism of melatonin [26], contributing to decreased melatonin availability in endometrial tissue. As such, local regulation of AhR ligands by inflammation, as well as environmental factors, may contribute to AhR activity and IDO, with increased IDO expression and activity associated with an increase in the invasiveness of endometrial stromal cells [27]. It should be noted that, as with circulating pro-inflammatory cytokines, environmental toxin effects via the AhR are also likely to increase gut permeability, as well as mediate changes that are common across a number of cancers, including increasing CYP1B1 levels

Notably, pro-inflammatory cytokine-induced IDO drives tryptophan to the production of kynurenine and associated kynurenine pathway products, such as the neuroregulatory kynurenic acid and quinolinic acid, and away from serotonin, NAS and melatonin synthesis. Raised pro-inflammatory cytokines may also inhibit the circadian production of melatonin by the pineal gland [28]. The circadian dysregulation caused by shift work decreases pineal gland melatonin production and increases the risk of endometriosis [29]. Such data indicates a general suppression of melatonergic pathway activity in endometriosis. In a preclinical endometriosis model, the loss of melatonin following pinealectomy exacerbates endometriosis symptomatology [30]. It is this shift away from serotonergic and melatonergic pathways that also forms the biological underpinnings for heightened levels of depression in endometriosis [3, 31].

Gut Microbiome, Gut Permeability and Butyrate

Recent data indicates a role for gut dysbiosis in endometriosis, perhaps in association with alterations in vaginal and cervical microbiota [32], which is supported by preclinical model data [33, 34]. It requires investigation as to whether such gut dysbiosis is associated with an increase in gut permeability. This would seem likely, given that the increase in pro-inflammatory cytokines and pain-associated stress in endometriosis increase gut permeability [35, 36]. Such gut dysregulation is strongly linked to a decrease in the production of the SCFA, Butyrate, which has important roles in gut barrier maintenance, immune dampening, and optimizing mitochondria functioning. Butryate can also induce melatonin and has histone deacetylase (HDAC) inhibitory effects [37, 38]. All of these butyrate effects are likely to modulate endometriosis etiology and course as well as the transition to endometrial and ovarian cancer [39, 40], including inhibiting the self-renewal capacity of endometrial cancer stem cells [41]. This is supported by recent work advocating the utilization of HDAC inhibition in the treatment of endometriosis [42, 43], although there may be some association of HDAC3 inhibition and infertility [44].

Vitamin A and Retinoic Acids

Decreased vitamin A, and its retinoic acid metabolites, may also be relevant to endometriosis pathoetiology and pathophysiology [45]. All-trans retinoic acid (ATRA) dramatically prevents the proliferation of endometrial tissue cysts, coupled to a decrease in local estradiol production [45]. Over the course of the menstrual cycle, changing patterns of steroid exposure modulate expression of retinoid receptors and ATRA synthesis. Local ATRA acts to correctively modulate the endometrial synthesis of many of the factors altered in endometriosis, including cytokines, differentiation, matrix metalloproteinase, secretion, connexin-43, and integrins [46]. ATRA biosynthesis seems impaired in endometriosis lesions, linked to a decrease in cellular retinol-binding protein type 1 (RBP)1 [46]. The regulation of vitamin A metabolites, including ATRA, is driven by endogenous enzymes induced by HDAC inhibitors, thereby suggesting modulation by gut-derived butyrate and other endogenous HDAC inhibitors [47]. As vitamin A increases microbiome-derived butyrate, vitamin A and other positive regulators of butyrate, will increase the protection afforded by ATRA. Vitamin A can increase the melatonergic pathways [48] as well as optimize mitochondria functioning [49]. ATRA prevents TGF-β + IL-6 from inducing Th17 cells [50], favouring increased Treg cells, thereby highlighting a role for vitamin A and its retinoic acid metabolites in the pro-/ anti-inflammatory cytokine balance. Clearly, decreased vitamin A and its metabolites may be an important factor in the pathophysiology of endometriosis, given its regulatory interactions with other altered factors.

Lower vitamin A metabolite levels may arise from increased CYP26. Raised TGF-β levels in the serum and peritoneal fluid in endometriosis [16] would be expected to decrease CYP26, and thereby increase vitamin A metabolite availability [51]. This indicates that increased Treg cells would be co-ordinated with higher vitamin A metabolite availability, which is a link that seems to be broken in endometriosis, possibly from concurrent increases in pro-inflammatory activity and epigenetic HDAC activity [51]. Decreased vitamin A prenatally can significantly alter the development of the post-natal gut [52], and could act as a prenatal pathoetiological prime, in part via the impact of lower vitamin A on mitochondria and cellular regulatory functions.

Uterine Microbiome

There is a growing interest in the composition and role of the uterine microbiome, with likely relevance to endometriosis and its transition to cancer [53]. Cervical lesions seem associated with alterations in the uterine microbiome composition [54], which may vary in endometriosis and over the menstrual cycle [55]. Within individuals, there does seem some continuity within the vagino-uterine microbiome, perhaps arising from cyclical hormonal changes that act to modulate the pH, and therefore the conformation of cervical mucins [56]. A number of lifestyle and dietary factors correlate with the composition of the vagino-uterine microbiome, including herbal medication impacts [55]. The significance of this has still to be determined, including as to any impact on the wider processes and factors associated with endometriosis, such as ATRA regulation, mitochondria functioning and the melatonergic pathways or indeed as to whether there is a gut-uterus axis, maybe linked by dietary impacts or perhaps via exosomes or the micro-metastasis of endometrial cells, which have been found in the lung, brain and other organs in preclinical models [57]. There is some evidence to suggest variations in the ratios of aromatic amino acids and fatty acids synthesized in the vagino-uterine microbiome may have consequences for the functioning or releases of endometrial cells or local immune cells.

14-3-3 and microRNAs

A number of microRNAs show alterations in endometriosis [58], including miR-7, miR-375 and miR-451, all of which regulate the melatonergic pathways and mitochondria functioning. Decreased miR-375 is evident in endometriosis ectopic stromal cells [59]. Decreased miR-375 is linked to raised 14-3-3ζ levels [60] and therefore to 14-3-3ζ-mediated aralkylamine N-acetyltransferase (AANAT) stabilization and NAS synthesis at the start of the melatonergic pathway. Likewise, miR-451 and miR-7 also negatively regulate 14-3-3ζ, with a downregulation of these miRNAs evident in endometriosis and endometriosis-associated ovarian cancers [61, 62]. Such alterations in miRNA-driven changes in 14-3-3ζ-mediated melatonergic pathway initiation may also link to the data showing ectopic endometrial stromal invasion in endometriosis is 14-3-3ζ dependent [63]. This suggests that these miRNAs have a role in co-ordinating the initiation of the melatonergic pathways, and therefore NAS synthesis and associated TrkB activation, with the proliferation and invasion in endometriosis, possibly priming the transition to ovarian cancers.

Low Body Mass Index and Adipocytes

Women with endometriosis have a lower body mass index (BMI) and body fat. A preclinical model suggests that this may be partly mediated by miR-let 7b and miR-342-3p, which alter fat metabolism and drive down adipocyte stem cell levels, thereby readily overlapping with wider endometriosis symptomatology [64]. Transfected primary adipocytes from women with endometriosis, vs healthy controls, led to significant metabolic changes in this murine model, including in mRNA levels of peroxisome proliferator-activated receptor (PPAR)-γ, leptin, adiponectin, IL-6, and hormone-sensitive lipase (HSL), as well as decreased adipocyte stem cell levels [64]. Such data suggests a close association of low BMI and endometriosis pathophysiology, with a mediating role for miR-let 7b and miR-342-3p [64].

In association with this decreased BMI and lower levels of adipocytes, a decrease in serum and peritoneal fluid adiponectin is evident in endometriosis [65], with adiponectin, via its receptors, decreasing the proliferation of endometrial cells [66]. The decrease in adiponectin mRNA, like IL-6 and HSL mRNA, can be mediated by miR-let 7b, with miR-342-3p increasing leptin. Leptin is generally increased in endometriosis, both in the serum and endometrium, with the down-regulation of leptin associated with improved symptomatology [64, 66]. Adipocyte changes may be driven by circulating miRNAs, especially exosomal miR-let 7b and miR-342-3p [64]. Such wider metabolic changes in endometriosis have led to its classification as a systemic disorder [67], although if driven by endometrial tissue-derived exosomal miRNAs, wider alterations in other tissues and organs would be expected.

Estrogen and Estrogen Receptors (ERα, ERβ)

Endometriosis is usually defined as an estrogen-dependent condition, with treatment aimed at lowering estrogen effects, often at the further expense of decreases in fertility. However, SNPs in ERα are not risk factors for endometriosis, although ERα SNPs do modulate stage transition in endometriosis [68], suggesting a role for ERα in the pathophysiology, but not the pathogenesis of endometriosis. Notably, melatonin inhibits the ERα [69], suggesting that melatonin inhibition in endometriosis may potentiate ERα-driven pathophysiology. ERβ is also increased in endometriosis, where it translocates to mitochondria and drives increases in SOD2 and bcl-2, thereby affording protection against apoptosis [70] as well as heightening mitochondria functioning [71]. Endometriosis risk also correlates with CYP1B1 SNPs [4, 72], with CYP1B1 SNPs correlating with ERα and ERβ levels in regard to endometrial cancer risk [73]. CYP1B1 increases the backward conversion of melatonin to NAS [74], suggesting that the NAS/melatonin ratio may be co-ordinated with CYP1B1, ERα and ERβ, all within mitochondria. This indicates that decreased melatonin by AhR-induced CYP1B1 is co-ordinated with estrogen pathophysiology.

Notably, the relatively heightened levels of estrogen in endometriosis, via ERα, may also significantly inhibit the production of adipocytes from bone marrow [75], as well as triglyceride accumulation in adipocytes [76], either directly or via the regulation of endometrial exosomes. It awaits investigation as to whether mitochondria ERβ levels are increased in endometriosis adipocytes, as occurs in endometrial tissue. Heightened ERβ can significantly upregulate mitochondria functioning, including ATP production [77], as well as decrease levels of ERα and estrogen related receptors (ERR)α and ERRβ, which are also significant mitochondria regulators [78]. This may be of some relevance to the alterations in vitamin A and the decrease in vitamin A metabolites and retinol-binding protein (RBP) in endometriosis, given that ERα, but not ERβ, increases RBP-4 [79]. It requires investigation as to whether the increase in mitochondria ERβ occurs in adipocytes in endometriosis and whether this has any implications for the regulation of the retinoic acids, such as via CYP26. The pathophysiological changes occurring in endometriosis are summarized in Figure 1.

Figure 1 The melatonergic pathways integrate endometriosis pathophysiology.
Figure 1

The melatonergic pathways integrate endometriosis pathophysiology.

It still awaits clarification as to the mechanisms underlying adipocyte changes in endometriosis. A number of mechanisms are possible, including: exosome release of miRNAs that drive alterations in in adipocytes [64]; heightened levels of estrogen effects at the ERα initially, before a rise in mitochondria ERβ, as in endometrial tissue [80]; micro-metastasis of endometrial cells [57]; early developmental and ongoing alterations in a gut-liver-hypothalamus-adipocyte axis [81]. Clearly alterations in mitochondria metabolic regulation are of some importance.

The importance of prenatal factors are indicated at the top left-hand side of the figure. Maternal factors, as well as decreased vitamin A and melatonin contribute to suboptimal placenta functioning and foetal growth. This leads to alterations in the gut-liver-hypothalamus-adipocyte axis, leading to decreased adipogenesis and low BMI, with resultant alterations in exosomal content coupled to decreased adiponectin, with miR-342-3p likely mediating effects partly via increased leptin. Such a collection of changes have direct effects on endometrial cell functioning and the interactions of these cells with immune cells. A putative decrease in butyrate and ongoing decreases in vitamin A and its retinoic acid metabolites, contribute to endometriosis, in part via decreased melatonin and suboptimal mitochondria functioning. Increased estrogen activation of microchondria ERβ, possibly via increased ATP production, increases the backward conversion of melatonin to NAS, leading to increased BDNF and TrkB activation, contributing to endometriosis. Similar effects arise from exogenous and induced AhR ligands via their upregulation of CYP1B1. Alterations in miRNAs and 14-3-3 proteins in endometriosis will have significant impacts on the melatonergic pathways. The raised CYP26 metabolism of ATRA may be mediated by altered immune-inflammatory activity and lower butyrate. Decreased ATRA has effects at a number of sites, including AhR activity and miRNA regulation. Endometrial lesion cells may also release exosomes that modulate other cells, including adipocytes, but are not shown for clarity. Abbreviations are shown at the end of the article.

Integrating Pathophysiology within Mitochondria

As the data above indicate, there are a plethora of pathophysiological changes in endometriosis. All of these factors may be integrated (see Figure 1).

Although the feminizing and some tumor effects of estrogen are mediated via ERα, the relevant effects of estrogen in endometriosis may be mostly driven by mitochondria-located ERβ [77, 80]. In other cells ERβ forms a complex with glucose regulated protein (Grp)75, which is a mitochondria matrix chaperone. Once within the mitochondria matrix the ERβ-Grp75 complex is stable and drives a significant increase in mitochondria ATP production and alters mitochondria DNA gene expression [77]. There is a dramatic increase in mitochondria-located ERβ in ectopic endometriotic tissue as well as in estrodiol-primed endometriotic cells [77, 80], which show heightened mitochondria bioenergetics coupled to lower ROS, at least partly mediated by raised SOD2 levels [70, 80]. Heightened mitochondria ERβ levels increases migratory activity, which is attenuated by mitochondria respiration inhibition. Such data highlights the importance of mitochondria in the biological underpinnings of endometriosis, including in the changes linked to increased peritoneal estrogen levels.

A consequence of an ERβ-driven rise in mitochondria ATP levels [77] is an ATP-driven backward conversion of melatonin to NAS [82], as can occur with raised levels of mitochondria CYP1B1 [74]. Although NAS and melatonin have many similar effects, NAS is a BDNF mimic via its activation of TrkB, as well as inducing BDNF. As an increase in both TrkB and BDNF are intimate aspects of endometriosis pathophysiology, such increased synthesis and efflux of NAS, at the expense of melatonin, will drive the proliferation evident in endometriosis. This would suggest that an increase in the NAS/melatonin ratio is a significant mediator of the effects of increased estrogen and the dramatic increase in mitochondria ERβ. Endometriosis is associated with an increased risk of migraines [83], which may also be linked to a raised NAS/ melatonin ratio [3].

Although vitamin A and retinoic acid metabolites can modulate mitochondria functioning per se, ATRA can not only decrease the raised levels of peritoneal estrogen evident in endometriosis, but can also decrease levels of mitochondria ERβ in endometriotic tissue [45]. Dysregulation of the retinoic acids, including from increased CYP26 [84], may therefore determine the mitochondria and estrogen changes occurring in endometriosis. Such changes are likely to correlate with gut dysbiosis and decreased levels of butyrate production (either from decreased vitamin A, or high circulating pro-inflammatory cytokines, or from pain-associated stress), with correlated consequences arising from a decrease butyrate’s immune, mitochondria and melatonin regulatory effects. The loss of butyrate’s HDAC inhibition will lower ATRA levels, thereby further attenuating ATRA’s inhibition of estrogen and mitochondria ERβ.

Heightened pro- and anti-inflammatory cytokine levels in endometriosis will differentially modulate the levels and activity of different immune cells, including mast cells and macrophages as well as t-cell patterning. Raised peritoneal macrophage levels are evident in endometriosis, with ERβ activation in endometrial stromal cells, leading to M2-like macrophage recruitment [85]. These macrophages are proposed to interact with endometrial nerves in the pathogenesis of an endometriosis preclinical model [86]. As such, increased estrogen and mitochondria ERβ, via M2-like macrophage recruitment, may be seen as an attempt to dampen inflammation, complicated by the changes in endometrial nerves. Autocrine and paracrine effects of melatonin are crucial in shifting macrophages from a M1- to M2-like phenotype [87], suggesting that changes in the melatonergic pathways, including within the mitochondria of macrophages and neighbouring cells, may be important to the nature of the immune responses in endometriosis. This requires investigation. Clearly, alterations in mitochondria functioning are relevant across the host of different cells associated with endometriosis, suggesting dynamic intercellular alterations driven by alterations in mitochondria metabolic regulation.

Heightened pro-inflammatory cytokine levels increase IDO, with oxidative stress and stress-induced corticotropin releasing hormone (CRH) increasing TDO, leading to tryptophan metabolism down the kynurenine pathways and away from serotonin, NAS and melatonin synthesis. Consequently, kynurenine levels are increased, leading to AhR activation. The regulatory effects AhR in many cell types can be lost following its activation by exogenous or induced ligands, such as PCBs and kynurenine. PCBs activation of the AhR increases estrogen and 17β-HSD-7 levels as well as pro-inflammatory cytokines, in a murine model of endometriosis [23]. ATRA can have inhibitory effects on AhR-inductions [88]. As such, exogenous and induced ligands of the AhR are likely to contribute to heightened estrogen and cytokine levels and effects, at least transiently, in endometriosis, which the diminished levels of ATRA fail to inhibit. Estrogen can also potentiate the AhR-induction of CYP1B1, thereby decreasing melatonin synthesis, with increasing CYP1B1 levels being evident over endometriosis stages and in the transition to ovarian tumors [74]. Consequently, the attenuation of ATRA’s inhibition of the AhR-induced CYP1B1 contributes to a rise in the NAS/melatonin ratio, and thereby lower melatonin’s optimization of mitochondria functioning. Overall, decreased ATRA and an increased NAS/melatonin ratio driven by exogenous and induced AhR ligands in interaction with estrogen contribute to mitochondria dysregulation in endometriosis.

Altered expression levels of a plethora of miRNA can occur in endometriosis, with their patterning changing over the course of lesion progression as part of co-ordinated cellular plasticity responses [58]. The lower levels of miR-7, miR-375 and miR-451 increase 14-3-3ζ [60], thereby stabilizing AANAT for NAS synthesis, leading to TrkB activation and BDNF induction, especially when coupled to increased CYP1B1’s backward conversion of melatonin to NAS. Decreases in the retinoic acids may also be relevant to these miRNA effects, given that retinoic acids can dramatically increase miR-375 levels, as shown in adipocytes [89]. As such, decreased retinoic acids will also contribute to lower melatonin levels via altered miR-375.

The raised stress and pain levels commonly experienced in endometriosis, coupled to increases in pro-inflammatory cytokines, are highly likely to increase gut dysbiosis and permeability. It will be important to determine when this occurs, including any priming role for pre- and post-natal processes and/or whether it is secondary to symptomatic stress and pro-inflammatory cytokine induction. Lower butyrate levels, and its associated HDAC inhibition, drive many of these gut-associated changes, including a host of processes relevant in endometriosis pathophysiology, including mitochondria, immune, melatonin and ATRA regulation [47]. Any role for the uterine microbiome has still to be investigated, including any possibility of a gut-uterine axis that could link alterations in butyrate to cellular processes acting to regulate the uterine microbiome.

Adipocyte regulation in endometriosis will be important to clarify, including roles for endometrial released exosomes [64], decreased ATRA effects, micro-metastasis and wider gut-liver-hypothalamus-adipocyte changes. Notably, melatonin decreases adipocyte stem cell PPAR-γ and adipogenesis via effects in bone marrow-derived mesenchymal stem cells (90-2), suggesting that an increase in melatonin, or NAS [93], will contribute to the decrease in adipocyte levels and adipocyte stem cells in endometriosis. Melatonin also decreases levels of adipocyte-associated fluxes, including leptin and IL-6 [92], suggesting that alterations in both adipogenesis and adipocyte fluxes may be associated with melatonergic pathway modulation that does not necessarily parallel the melatonergic pathway alterations in endometriotic tissue. This will be important to determine, especially regarding etiology and the conceptualization of endometriosis as a systemic metabolic disorder.

Endometrial lesion tissue processes have overlaps with processes important to adipocyte regulation. TrkB is expressed in murine adipocytes, where it can significantly modulate metabolism and weight [94]. As to whether adipocyte TrkB levels and activity are altered in women with endometriosis requires investigation. Also, 14-3-3ζ is crucial to adipogenesis, as a consequence of large interactome with the wide array of factors required in adipocyte differentiation and functioning, as well as its role in the activation of the melatonergic pathways [95]. CYP1B1 inhibition can also decrease adipogenesis in pluripotent stem cells [96], suggesting a role for the NAS/ melatonin ratio, possibly around bone marrow cells. Decreased adipogenesis can also be driven by alterations in retinoic acids [97] or decreased gut microbiome-derived butyrate [98], again linking to these interconnected processes, as shown in Figure 1. It is of note that it is variations in mitochondria functioning that determine as to whether bone marrow-derived mesenchymal stem cells differential into bone or adipocytes [99], highlighting the role of alterations in mitochondria functioning at another key site in endometriosis pathophysiology.

Overall, it is clear that much of the pathophysiology of endometriosis is intimately associated with the regulation of mitochondria functioning across a host of cell types and systems. Many of the classical changes in endometriosis are intimately linked to alterations in mitochondria functioning, including changes in estrogen ERβ, adiponectin, melatonin and vitamin A. Such processes are also relevant to the pathoetiology of endometriosis.

Integrating Pathoetiology

Sampson’s theory of retrograde menstruation is still widely accepted to reflect core changes driving endometriosis pathoetiology [100]. However, the incidence of retrograde menstruation is far higher than that of endometriosis [101], leading to a revision of Sampson’s theory whereby women with endometriosis may show alterations in their immune system, with perhaps a defect in the recognition of endometrial fragments, akin to autoimmune disorders [102].

More recent work has led to the proposal of a set of genetic and epigenetic changes, perhaps linked to the early pre- and post-natal period [103]. Notably, some endometriosis risk factors are strongly associated with early developmental processes, including SNPs in the AhR and CYP1B1. Likewise factors associated with endometriosis pathophysiology, such as miR-451, vitamin A and the melatonergic pathways have crucial roles in the regulation of placenta and foetal development [52]. It is also widely accepted that alterations in prenatal processes can modulate many postnatal events via changes in the gut and gut microbiome development. As such, there may be scope for perinatal modelling of the etiology of endometriosis, linking to a host of genetic and epigenetic processes [103]. For example, alterations in the gut-liver-hypothalamus-adipocyte may modulate the levels of melatonin and retinoic acids that then act to decrease the differentiation of bone marrow-derived mesenchymal stem cells into adipocytes. Presumably, the low BMI evident in endometriosis can predate endometrial symptomatology, suggesting that early developmental processes underpinning changes in fat regulation may be intimately linked to endometriosis pathoetiology and pathophysiology. Decreased adipogenesis may act to prime endometrial cells, via a decrease in adiponectin, including in regulation of the NAS/melatonin ratio. Further research across these disparate arrays of research may indicate how relevant such early developmentally shaped processes are to the pathoetiology of endometriosis. This is a redefinition of endometriosis from a gynaecological condition to a developmental systemic disorder.

An early developmentally-driven alteration in adipocyte regulation indicates a role for altered adipocyte fluxes and exosomes in endometriosis, which then act to regulate the maturing uterus, either directly or via alterations in uterine-interacting immune cells. Or this could suggest prenatal epiphenomenal alterations in fat and uterine tissue development. A broad body of data shows adipocyte exosomes to modulate a plethora or other tissues, including in the etiology of breast cancer [104]. The alterations in the adipocytes of women with endometriosis would suggest that adipocyte-derived exosomes in endometriosis are distinct and may have differential effects in different tissues, either directly and/ or via immune cells [105].

Different exosomal miRNAs can regulate the macrophage phenotype, e.g. miR-let 7 increases an M2-like phenotype (106-7). This would suggest that the proposed role of M2-like macrophages that infiltrate the uterus in high numbers, may be influenced by their interactions with adipocyte-derived exosomes, as well as fluxes and exosomes derived from endometrial cells [64]. Under inflammatory conditions, macrophages and other immune cells would be expected to show an M1-like inflammatory response to eliminate pathogens and inappropriate cells, which then shifts to a debris-clearing M2-like phenotype associated with homeostasis resolution. The attraction and maintained presence of an M2-like macrophage phenotype that fails to eliminate sources of inflammation can contribute to an immune tolerance, as in endometriosis-associated ovarian cancer [108]. The developmental etiology of adipocyte alterations in endometriosis will be important to determine.

An early developmental etiology to endometriosis is supported by longitudinal data from Sweden, showing that low birth weight for gestational age associates with a subsequent endometriosis diagnosis, which the authors interpret as showing a role for prenatal growth restriction in endometriosis pathoetiology [109]. This may be complicated by the genetic links to endometriosis, as women with endometriosis are more likely to have small for gestational age offspring, as well as increased levels of preeclampsia during pregnancy [110]. However, when maternal endometriosis and other prenatal factors were partialed out, the association of low-birth weight and later endometriosis was still significant, indicating a prenatal etiology to endometriosis. The increased endometriosis risk from AhR SNPs [21] may be mediated by changes occurring prenatally, including within the placenta, where the AhR has a regulatory role that can be dramatically altered by exogenous and induced AhR ligands. Given the lower melatonin levels in pregnancies with endometriosis associated conditions, such as preeclampsia, decreased placental melatonin may contribute to the prenatal etiology of endometriosis. As such, key changes in endometriosis pathophysiology may also be important to any prenatal etiology.

Prenatal retinoic acid signalling in crucial to fertility [111]. During the first trimester fetal growth is totally dependent upon endometrial secretions mediated by endometrial stromal cells, including a critical role for glycogen, with infertile women generally having very low glycogen as well as retinoic acid levels. Adipocyte-secreted glycogen controls this glycogen metabolism and secretion in endometrial stromal cells, as well as its foetal uptake [112]. This indicates that maternal endometriosis, in association with low adiponectin, retinoic acid signalling and altered exosome content, will not only modulate fertility, but also foetal development, including in the development of the foetal uterus. As to whether such early, primarily first trimester processes, prime adipocyte and uterine changes relevant to low BMI and endometriosis in the offspring requires investigation.

Other perinatal factors are also associated with an increased risk of endometriosis, including cesarean section, premature birth and formula feeding (vs breastfeeding) [113]. Notably, breastfeeding benefits may be mediated via the regulation of the infant’s gut microbiome and the melatonergic pathways [114]. Cesarean section leads to a perturbation of the infant gut microbiota that is partially restored by breastfeeding [115]. As such, perinatal, and perhaps prenatal, endometriosis risk factors may be ameliorated by the regulation of the gut microbiome, including by breastfeeding, highlighting the gut as an important hub upon which risk factors may be interacting in the early developmental pathoetiology of endometriosis.

The induction of endometriosis symptoms in mice leads to alterations in hepatic metabolism [116]. These authors found that inducing endometriosis led to significant alterations in 26 hepatic genes, 6 of which are involved in metabolic regulation, with changes linked to increased metabolism and decreased weight gain. Such data suggests that endometriosis symptomatology can also drive the low BMI and associated adipocyte changes, at least in part via alterations in hepatic metabolic regulation. Such data indicates that local endometrial tissue changes may drive a systemic condition. Notably, hepatic exosomes can significantly modulate adipocyte function [117], whilst the levels of adiponectin release by adipocytes influences the levels and contents of exosomes [118], suggesting that the decreased adipogenesis and adiponectin levels in endometriosis will be associated with an array of alterations in the levels and contents of exosomes, which may interact with adipocyte regulating factors in hepatic exosomes.

As melatonin has inhibitory effects on estrogen ERα signalling, a feedback inhibition of the melatonergic pathways by estrogen may be a core aspect of the local pathoetiological underpinnings of endometriosis. This may be achieved by a number of means, including the potentiation of the AhR’s induction of mitochondria CYP1B1, as is evident in cancer cells, and/or an upregulation of ATP production via mitochondria ERβ [82]. As such, dysregulation in the mutual antagonistic interactions of estrogen and local melatonin production may be an occluded aspect of pathoetiology and pathophysiology.

Overall, the pathoetiology of endometriosis has still to reach an accepted consensus. A growing literature suggests that the prenatal environment is important. As to whether low BMI is upstream or downstream of endometrial changes or concurrently regulated by systemic processes is the subject of current research. A growing body of data suggests that prenatal factors are important mediators of endometriosis, with significant impacts on the melatonergic and vitamin A/retinoic acid pathways, with consequences for the association of endometriosis with a number of cancers.

Treatment implications

The conceptualization of endometriosis as an estrogen-dependent disease has led to treatments that have primarily focussed on the inhibition of estrogen and/or an increase in progesterone. Consequently, combination hormonal contraceptives and progestins are the first-line treatments for endometriosis. These usually have some efficacy in pain management and are reasonably well tolerated. Outwith the long-term side-effects of such treatments, many women experience no, or only partial, improvement in pain. Discontinuation usually leads to symptom recurrence. A number of newer pharmaceuticals, again centred on estrogen-dependent aspect of endometriosis, have shown promise, including GnRH antagonists and CYP19A1/aromatase inhibitors, reviewed in [119]. It should be noted that GnRH is also a significant regulator of the gut, enteric nervous system, IgM responses and gastrointestinal motility, which may all be affected by GnRH antagonism [120].

As indicated by the complexity of factors relevant to the pathoetiology and pathophysiology of endometriosis, a number of novel treatments emerge.

Melatonin 10 mg/day, versus placebo, has shown efficacy in a phase-II clinical trial [121]. Melatonin lowered daily pain and dysmenorrhea by 40%, whilst also significantly reducing BDNF levels (independent of pain regulation). Melatonin also improved sleep quality and reduced analgesic use by 80% (S121), as well as having anti-estrogenic effects. As melatonin lowers the inflammatory content of adipocyte-derived exosomes, there are less detrimental effects of these exosomes in other organs, including the liver [122]. This suggests that melatonin will alleviate any symptomatology associated with adipocyte (and possibly endometrial) exosomes in other organs and immune cells. Given its high safety profile, the use of melatonin, alone or adjunctive, is an immediately practicable treatment that may be intimately linked to endometriosis pathophysiology, as indicated above.

For women with endometriosis, there is a heightened risk of poor pregnancy outcomes, including an increased risk of preeclampsia, neonatal death, still-birth and small for gestational age [110]. Melatonin treatment during pregnancy is likely to lower the likelihood of such poor pregnancy outcomes [123].

Vitamin A and its retinoic acid metabolites, especially ATRA, significantly modulate endometriosis pathophysiology. Diet and supplements to increase vitamin A are likely to afford some benefits and may also improve any concurrent gut dysbiosis and gut permeability, as well as increase the availability of local melatonin [48].

Butyrate supplementation, as with sodium butyrate, is also likely to improve any gut dysbiosis and gut permeability. Butyrate, as a HDAC inhibitor, would be expected to increase the availability of ATRA [47], and therefore increase the benefits of ATRA on mitochondria functioning, possibly via CYP26 inhibition [49,] as well as inducing the melatonergic pathways [48]. Butyrate, like melatonin, will also dampen any heightened immune-inflammatory activity, including that induced by stress-induced gut dysbiosis.

Given the proposed role of ERβ as an important driver of mitochondria alterations in endometriosis [77, 80], the modulation of estrogen effects at the ERβ is a significant treatment target. As the increase in mitochondria ERβ is associated with higher levels of estrogen availability, increasing ATRA, including via butyrate’s HDAC inhibition, will not only decrease peritoneal estrogen levels, but the raised mitochondria ERβ levels in endometriotic tissue [45].

Future Research

Treatment Orientated

What are the processes that are altered in the benefits of melatonin treatment of endometriosis? Are there benefits mediated via changes in oxidative stress, immune-inflammatory activity, gut permeability, exosomes and/ or more directly on endometrial tissue? Research similarly aimed at exploring these processes will also be relevant in regard to butyrate and vitamin A.

Is there an increase in the NAS/melatonin ratio within mitochondria in endometrial tissue? If so, do increases in mitochondria CYP1B1 or ATP production or some other means mediate such ratio changes. Such research would provide a significant focal treatment target.

Are there changes in gut SCFAs in endometriosis, especially in butyrate level? If so, is this due to a decrease in its levels of production or its uptake over intestinal epithelial cells? Do stress and increased pro-inflammatory cytokines drive changes in gut dysbiosis secondary to endometriosis or is gut dysbiosis and a decrease in circulating butyrate causal to some of the mitochondria changes occurring in endometrial tissue and immune responses? Is gut dysbiosis and decreased butyrate production secondary to lower levels of vitamin A.

Pathophysiology Orientated

It will be important to determine as to whether ATRA modulates AhR-mediated increase in mitochondria CYP1B1 in different endometriosis stages and in the tumor transition, whilst exploring any impact of this on the NAS/ melatonin ratio. Similarly, as to the impact of the dramatic increase in mitochondria ERβ on the NAS/melatonin ratio.

It requires determination as to whether the ATRA catabolizing enzymes, CYP26A1 and CYP26A2, are increased in endometriotic tissue.

It requires investigation as to whether the increase in mitochondria ERβ evident in endometrial tissue, also occurs in adipocytes and whether the mitochondria ERβ increase has any implications for the regulation of the retinoic acids, such as via CYP26 regulation.

As well as miR-7, miR-375 and miR-451 regulation of 14-3-3ζ and the melatonergic pathways, it is highly likely that the wide array of dynamic interactions highlighted above are intimately associated with wider alterations in co-ordinated miRNA expression. These will be important to determine, including as to the role of core mitochondria processes in co-ordinating such changes.

Within a conceptualization of endometriosis having a prenatal origin, it will be important to determine alterations in maternal, placenta and foetal fluids as to changes in the many factors highlighted throughout this article, including ATRA, NAS/melatonin, and miRNAs, as well as maternal butyrate circulatory levels and placental 11β-HSD2. Given the higher levels of stress that can be experienced by women with endometriosis, heightened levels of stress and pro-inflammatory cytokines in pregnancy occur, with many of these effects mediated via placental 11β-HSD2 regulation. HDAC inhibition, including from maternal microbiome-derived butyrate, may prevent the effects of cortisol or IL-1β, which decrease placental 11β-HSD2, and therefore the effects of stress in the placenta and foetus [124]. Such prenatal processes will be important to determine.

As AhR SNPs are a susceptibility factor for endometriosis [21], it requires investigation as whether this increased risk is mediated via prenatal factors, including maternal cigarette smoking, which is also an endometriosis risk factor [125], as well as other exogenous and induced AhR ligands, such as factors increasing kynurenine and associated decreases in the melatonergic pathways. A prenatal etiology means that many of the genetic endometriosis risk factors are not necessarily mediating their increased risk effects in endometrial tissue.

Foetal uterus development is highly variable, as measured by growth parameters [126]. It requires investigation as to whether maternal endometriosis-associated factors, such as decreased vitamin A and the retinoic acids or adiponectin [112], have any priming impact on the development of the foetal uterus. This could link foetal uterus development with retinoic acids’ modulation of fertility.

The prenatal and early developmental factors acting on adipogenesis will be important to determine. Such data should give a clearer role as to the influence of low BMI in the pathoetiology of endometriosis.

Conclusions

The pathoetiology and pathophysiology of endometriosis is clearly complex and involves a number of factors and processes over the course of development. Clearly most of the factors and processes associated with endometriosis have an impact on mitochondria functioning in a number of different cells, with this underpinning the changes in miRNAs expressed as well as their presence in exosomes. The processes underpinning low BMI and its association with changes in endometrial cells seems crucial to the conceptualization and treatment of endometriosis. There is a growing consensus that maternal and prenatal factors are important drivers of endometriosis, with significant changes in mitochondria functioning mediated by alterations in melatonergic and vitamin A/retinoic acid pathways regulation, both prenatally and subsequently. Changes in such processes clearly have implications for understanding, and preventing, the association of endometriosis with a number of cancers.

  1. Conflict of interest: Author states no conflict of interest

List of abbreviations

AANAT

aralkylamine N-acetyltransferase

AhR

aryl hydrocarbon receptor

ATRA

all-trans retinoic acid

BDNF

brain-derived neurotrophic factor

BMI

body mass index

CRH

corticotropin releasing hormone

CYP

cytochrome P 450

ER

estrogen receptor

GnRH

gonadotropin-releasing hormone

GRP

glucose regulated protein

HDAC

histone deacetylase

HSD

hydroxysteroid dehydrogenase

HSL

hormone-sensitive lipase

IDO

indoleamine 2,3-dioxygenase

Ig

immunoglobulin

IL

interleukin

miR

microRNA

NAS

N-acetylserotonin

NLRP3

Nod-like receptor family pyrin domain containing inflammasome

O&NS

oxidative and nitrosative stress

PCBs

polychlorinated biphenyls

PPAR

peroxisome proliferator-activated receptor

RBP

retinol-binding protein

SCFA

short-chain fatty acids

SNP

single nucleotide polymorphism

SOD

superoxide dismutase

TCDD

2,3,7,8-tetrachlorodibenzo-p-dioxin

TDO

tryptophan 2,3-dioxygenase

TGF

transforming growth factor

Th

T helperTreg regulatory t cell

TrkB

tyrosine kinase receptor-B

References

1 Bulun SE. Endometriosis. N Engl J Med. 2009;360:268-279. doi.org/10.1056/NEJMra080469010.1056/NEJMra0804690Search in Google Scholar PubMed

2 Bulletti C, Coccia ME, Battistoni S, Borini A. Endometriosis and infertility. J Assist Reprod Genet. 2010;27(8):441-7. doi. org/10.1007/s10815-010-9436-110.1007/s10815-010-9436-1Search in Google Scholar PubMed PubMed Central

3 Anderson G, Maes M. Melatonin: A Natural Homeostatic Regulator – Interactions with Immune Inflammation and Trytophan Catabolite Pathways in the Modulation of Migraine and Endometriosis. J Nat Prod Res Updates. 2015;1:7-17.Search in Google Scholar

4 Piccinato CA, Neme RM, Torres N, Sanches LR, Cruz Derogis PB, Brudniewski HF, E Silva JC, et al. Increased expression of CYP1A1 and CYP1B1 in ovarian/peritoneal endometriotic lesions. Reproduction. 2016;151(6):683-92. doi: 10.1530/REP-15-0581.10.1530/REP-15-0581Search in Google Scholar PubMed

5 Wessels JM, Kay VR, Leyland NA, Agarwal SK, Foster WG. Assessing brain-derived neurotrophic factor as a novel clinical marker of endometriosis. Fertil Steril. 2016;105(1):119-28.e1-5. doi: 10.1016/j.fertnstert.2015.09.003.10.1016/j.fertnstert.2015.09.003Search in Google Scholar PubMed

6 Dewanto A, Dudas J, Glueckert R, Mechsner S, Schrott-Fischer A, Wildt L, et al. Localization of TrkB and p75 receptors in peritoneal and deep infiltrating endometriosis: an immunohistochemical study. Reprod Biol Endocrinol. 2016;14(1):43. doi: 10.1186/s12958-016-0178-5.10.1186/s12958-016-0178-5Search in Google Scholar PubMed PubMed Central

7 Anderson G, Maes M. How Immune-inflammatory Processes Link CNS and Psychiatric Disorders: Classification and Treatment Implications. CNS Neurol Disord Drug Targets. 2017;16(3):266-278. doi: 10.2174/187152731566616112214465 9.10.2174/1871527315666161122144659Search in Google Scholar PubMed

8 Laschke MW, Menger MD. The gut microbiota: a puppet master in the pathogenesis of endometriosis? Am J Obstet Gynecol. 2016;215(1):68.e1-4. doi: 10.1016/j.ajog.2016.02.036.10.1016/j.ajog.2016.02.036Search in Google Scholar PubMed

9 Zalar B, Haslberger A, Peterlin B. The Role of Microbiota in Depression - a brief review. Psychiatr Danub. 20180;30(2):136141. doi: 10.24869/psyd.2018.10.24869/psyd.2018Search in Google Scholar

10 Lorençatto C, Petta CA, Navarro MJ, Bahamondes L, Matos A. Depression in women with endometriosis with and without chronic pelvic pain. Acta Obstet Gynecol Scand. 2006;85(1):8892. doi.org/10.1080/0001634050045611810.1080/00016340500456118Search in Google Scholar PubMed

11 Harlev A, Gupta S, Agarwal A. Targeting oxidative stress to treat endometriosis. Expert Opin Ther Targets 2015: 1-18.10.1517/14728222.2015.1077226Search in Google Scholar PubMed

12 Yeo SG, Won YS, Lee HY, Kim YI, Lee JW, Park DC. Increased expression of pattern recognition receptors and nitric oxide synthase in patients with endometriosis. Int J Med Sci. 2013;10(9):1199-208. doi.org/10.7150/ijms.516910.7150/ijms.5169Search in Google Scholar PubMed PubMed Central

13 Amreen S, Kumar P, Gupta P, Rao P. Evaluation of Oxidative Stress and Severity of Endometriosis. J Hum Reprod Sci. 2019;12(1):40-46. doi: 10.4103/jhrs.JHRS_27_17.10.4103/jhrs.JHRS_27_17Search in Google Scholar PubMed PubMed Central

14 de Melo LGP, Nunes SOV, Anderson G, Vargas HO, Barbosa DS, Galecki P, et al. Shared metabolic and immune-inflammatory, oxidative and nitrosative stress pathways in the metabolic syndrome and mood disorders. Prog Neuropsychopharmacol Biol Psychiatry. 2017;78:34-50. doi: 10.1016/j. pnpbp.2017.04.027.10.1016/j.pnpbp.2017.04.027Search in Google Scholar PubMed

15 Chang CM, Wang ML, Lu KH, Yang YP, Juang CM, Wang PH, et al. Integrating the dysregulated inflammasome-based molecular functionome in the malignant transformation of endometriosis-associated ovarian carcinoma. Oncotarget. 2017;9(3):3704-3726. doi: 10.18632/oncotarget.23364.10.18632/oncotarget.23364Search in Google Scholar PubMed PubMed Central

16 Sikora J, Smycz-Kubańska M, Mielczarek-Palacz A, Bednarek I, Kondera-Anasz Z. The involvement of multifunctional TGF-β and related cytokines in pathogenesis of endometriosis. Immunol Lett. 2018;201:31-37. doi: 10.1016/j.imlet.2018.10.011.10.1016/j.imlet.2018.10.011Search in Google Scholar PubMed

17 Kaabachi W, Kacem O, Belhaj R, Hamzaoui A, Hamzaoui K. Interleukin-37 in endometriosis. Immunol Lett. 2017;185:52-55. doi: 10.1016/j.imlet.2017.03.012.10.1016/j.imlet.2017.03.012Search in Google Scholar PubMed

18 Khan KN, Yamamoto K, Fujishita A, Muto H, Koshiba A, Kuroboshi H, et al. Differential levels of regulatory T-cells and T-helper-17 cells in women with early and advanced endometriosis. J Clin Endocrinol Metab. 2019;1:jc.2019-00350. doi: 10.1210/jc.2019-00350.10.1210/jc.2019-00350Search in Google Scholar PubMed

19 Gogacz M, Winkler I, Bojarska-Junak A, Tabarkiewicz J, Semczuk A, Rechberger T, et al. Increased percentage of Th17 cells in peritoneal fluid is associated with severity of endometriosis. J Reprod Immunol. 2016;117:39-44. doi: 10.1016/j. jri.2016.04.289.10.1016/j.jri.2016.04.289Search in Google Scholar PubMed

20 Akitsu A, Iwakura Y. Interleukin-17-producing γδ T (γδ17) cells in inflammatory diseases. Immunology. 2018;155(4):418-426. doi: 10.1111/imm.12993.10.1111/imm.12993Search in Google Scholar PubMed PubMed Central

21 Zheng NN, Bi YP, Zheng Y, Zheng RH. Meta-analysis of the association of AhR Arg554Lys, AhRR Pro185Ala, and ARNT Val189Val polymorphisms and endometriosis risk in Asians. J Assist Reprod Genet. 2015;32(7):1135-44. doi: 10.1007/s10815-015-0505-3.10.1007/s10815-015-0505-3Search in Google Scholar PubMed PubMed Central

22 Beischlag TV, Anderson G, Mazzoccoli G. Glioma: Tryptophan Catabolite and Melatoninergic Pathways Link microRNA, 14-33, Chromosome 4q35, Epigenetic Processes and other Glioma Biochemical Changes. Curr Pharm Des. 2016;22(8):1033-48.10.2174/1381612822666151214104941Search in Google Scholar PubMed

23 Huang Q, Chen Y, Chen Q, Zhang H, Lin Y, Zhu M, et al. Dioxin-like rather than non-dioxin-like PCBs promote the development of endometriosis through stimulation of endocrine-inflammation interactions. Arch Toxicol. 2017;91(4):1915-1924. doi: 10.1007/s00204-016-1854-0.10.1007/s00204-016-1854-0Search in Google Scholar PubMed

24 Chiappini F, Sánchez M, Miret N, Cocca C, Zotta E, Ceballos L, et al. Exposure to environmental concentrations of hexachlorobenzene induces alterations associated with endometriosis progression in a rat model. Food Chem Toxicol. 2019;123:151-161. doi: 10.1016/j.fct.2018.10.056.10.1016/j.fct.2018.10.056Search in Google Scholar PubMed

25 Mariuzzi L, Domenis R, Orsaria M, Marzinotto S, Londero AP, Bulfoni M, et al. Functional expression of aryl hydrocarbon receptor on mast cells populating human endometriotic tissues. Lab Invest. 2016;96(9):959-971. doi: 10.1038/ labinvest.2016.74.10.1038/labinvest.2016.74Search in Google Scholar PubMed PubMed Central

26 Ferry G, Ubeaud C, Lambert PH, Bertin S, Cogé F, Chomarat P, et al. Molecular evidence that melatonin is enzymatically oxidized in a different manner than tryptophan: investigations with both indoleamine 2,3-dioxygenase and myeloperoxidase. Biochem J. 2005;388(1):205-15. doi:10.1042/BJ2004207510.1042/BJ20042075Search in Google Scholar PubMed PubMed Central

27 Mei J, Li MQ, Ding D, Li DJ, Jin LP, Hu WG, et al. Indoleamine 2,3-dioxygenase-1 (IDO1) enhances survival and invasiveness of endometrial stromal cells via the activation of JNK signaling pathway. Int J Clin Exp Pathol. 2013;6(3):431-44.Search in Google Scholar PubMed

28 Pontes GN, Cardoso EC, Carneiro-Sampaio MM, Markus RP. Pineal melatonin and the innate immune response: the TNF-alpha increase after cesarean section suppresses nocturnal melatonin production. J Pineal Res. 2007;43(4):365-71.10.1111/j.1600-079X.2007.00487.xSearch in Google Scholar PubMed

29 Marino JL, Holt VL, Chen C, Davis S. Shift work, hCLOCK T3111C polymorphism, and endometriosis risk. Epidemiology. 2008;19(3):477-84. doi: 10.1097/EDE.0b013e31816b7378.10.1097/EDE.0b013e31816b7378Search in Google Scholar PubMed PubMed Central

30 Koc O, Gunduz B, Topcuoglu A, Bugdayci G, Yilmaz F, Duran B. Effects of pinealectomy and melatonin supplementation on endometrial explants in a rat model. Eur J Obstet Gynecol Reprod Biol. 2010 Nov;153(1):72-6. doi: 10.1016/j. ejogrb.2010.06.012.10.1016/j.ejogrb.2010.06.012Search in Google Scholar PubMed

31 Anderson G, Maes M. Oxidative/nitrosative stress and immuno-inflammatory pathways in depression: treatment implications. Curr Pharm Des. 2014;20(23):3812-47.10.2174/13816128113196660738Search in Google Scholar PubMed

32 Ata B, Yildiz S, Turkgeldi E, Brocal VP, Dinleyici EC, Moya A, et al. The Endobiota Study: Comparison of Vaginal, Cervical and Gut Microbiota Between Women with Stage 3/4 Endometriosis and Healthy Controls. Sci Rep. 2019;9(1):2204. doi: 10.1038/ s41598-019-39700-6.10.1038/s41598-019-39700-6Search in Google Scholar PubMed PubMed Central

33 Yuan M, Li D, Zhang Z, Sun H, An M, Wang G. Endometriosis induces gut microbiota alterations in mice. Hum Reprod. 2018;33(4):607-616. doi: 10.1093/humrep/dex372.10.1093/humrep/dex372Search in Google Scholar PubMed

34 Chadchan SB, Cheng M, Parnell LA, Yin Y, Schriefer A, Mysorekar IU, et al. Antibiotic therapy with metronidazole reduces endometriosis disease progression in mice: a potential role for gut microbiota. Hum Reprod. 2019;30pii:dez041. doi: 10.1093/humrep/dez041.10.1093/humrep/dez041Search in Google Scholar PubMed PubMed Central

35 Anderson G, Seo M, Berk M, Carvalho AF, Maes M. Gut Permeability and Microbiota in Parkinson’s Disease: Role of Depression, Tryptophan Catabolites, Oxidative and Nitrosative Stress and Melatonergic Pathways. Curr Pharm Des. 2016;22(40):6142-6151.10.2174/1381612822666160906161513Search in Google Scholar PubMed

36 Torres-Reverón A, Rivera-Lopez LL, Flores I, Appleyard CB. Antagonizing the corticotropin releasing hormone receptor 1 with antalarmin reduces the progression of endometriosis. PLoS One. 2018;13(11):e0197698. doi: 10.1371/journal. pone.0197698.10.1371/journal.pone.0197698Search in Google Scholar PubMed PubMed Central

37 Parada Venegas D, De la Fuente MK, Landskron G, González MJ, Quera R, Dijkstra G, et al. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front Immunol. 2019;10:277. doi: 10.3389/fimmu.2019.00277.10.3389/fimmu.2019.00277Search in Google Scholar PubMed PubMed Central

38 Jin CJ, Engstler AJ, Sellmann C, Ziegenhardt D, Landmann M, Kanuri G, et al. Sodium butyrate protects mice from the development of the early signs of non-alcoholic fatty liver disease: role of melatonin and lipid peroxidation. Br J Nutr. 2016:1-12.10.1017/S0007114516004025Search in Google Scholar PubMed

39 Takai N, Desmond JC, Kumagai T, Gui D, Said JW, Whittaker S, et al. Histone deacetylase inhibitors have a profound antigrowth activity in endometrial cancer cells. Clin Cancer Res. 2004;10(3):1141-9.10.1158/1078-0432.CCR-03-0100Search in Google Scholar PubMed

40 Yabushita H, Noguchi M, Nakanishi M. Emergence of an irreversible differentiated subclone from the poorly differentiated ovarian adenocarcinoma cell line AMOC-2 treated with sodium butyrate. J Obstet Gynaecol Res. 1997;23(6):493-502.10.1111/j.1447-0756.1997.tb00878.xSearch in Google Scholar PubMed

41 Kato K, Kuhara A, Yoneda T, Inoue T, Takao T, Ohgami T, et al. Sodium butyrate inhibits the self-renewal capacity of endometrial tumor side-population cells by inducing a DNA damage response. Mol Cancer Ther. 2011;10(8):1430-9. doi: 10.1158/1535-7163.MCT-10-1062.10.1158/1535-7163.MCT-10-1062Search in Google Scholar PubMed

42 Kawano Y, Nasu K, Hijiya N, Tsukamoto Y, Amada K, Abe W, et al. CCAAT/enhancer-binding protein α is epigenetically silenced by histone deacetylation in endometriosis and promotes the pathogenesis of endometriosis. J Clin Endocrinol Metab. 2013;98(9):E1474-82. doi: 10.1210/jc.2013-1608.10.1210/jc.2013-1608Search in Google Scholar PubMed

43 Grimstad FW, Decherney A. A Review of the Epigenetic Contributions to Endometriosis. Clin Obstet Gynecol. 2017;60(3):467-476. doi: 10.1097/GRF.0000000000000298.10.1097/GRF.0000000000000298Search in Google Scholar PubMed

44 Kim TH, Yoo JY, Choi KC, Shin JH, Leach RE, Fazleabas AT, et al. Loss of HDAC3 results in nonreceptive endometrium and female infertility. Sci Transl Med. 2019;11(474):pii: eaaf7533. doi: 10.1126/scitranslmed.aaf7533.10.1126/scitranslmed.aaf7533Search in Google Scholar PubMed PubMed Central

45 Yamagata Y, Takaki E, Shinagawa M, Okada M, Jozaki K, Lee L, et al. Retinoic acid has the potential to suppress endometriosis development. J Ovarian Res. 2015;8:49. doi: 10.1186/s13048-015-0179-6.10.1186/s13048-015-0179-6Search in Google Scholar PubMed PubMed Central

46 Pierzchalski K, Taylor RN, Nezhat C, Jones JW, Napoli JL, Yang G, et al. Retinoic acid biosynthesis is impaired in human and murine endometriosis. Biol Reprod. 2014;91(4):84. doi: 10.1095/biolreprod.114.119677.10.1095/biolreprod.114.119677Search in Google Scholar PubMed PubMed Central

47 Li Y, Wang L, Ai W, He N, Zhang L, Du J, et al. Regulation of retinoic acid synthetic enzymes by WT1 and HDAC inhibitors in 293 cells. Int J Mol Med. 2017;40(3):661-672. doi: 10.3892/ ijmm.2017.3051.10.3892/ijmm.2017.3051Search in Google Scholar PubMed PubMed Central

48 Fu Z, Kato H, Kotera N, Sugahara K, Kubo T. Regulation of the expression of serotonin N-acetyltransferase gene in Japanese quail (Coturnix japonica): II. Effect of vitamin A deficiency. J Pineal Res. 1999;27(1):34-41.10.1111/j.1600-079X.1999.tb00594.xSearch in Google Scholar PubMed

49 Mu Q, Yu W, Zheng S, Shi H, Li M, Sun J, et al. RIP140/ PGC-1α axis involved in vitamin A-induced neural differentiation by increasing mitochondrial function. Artif Cells Nanomed Biotechnol. 2018;46(sup1):806-816. doi: 10.1080/21691401.2018.1436552.10.1080/21691401.2018.1436552Search in Google Scholar PubMed

50 Mucida D, Park Y, Kim G, Turovskaya O, Scott I, Kronenberg M, et al. Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science. 2007;317(5835):256-6010.1126/science.1145697Search in Google Scholar PubMed

51 Takeuchi H, Yokota A, Ohoka Y, Iwata M. Cyp26b1 regulates retinoic acid-dependent signals in T cells and its expression is inhibited by transforming growth factor-β. PLoS One. 2011 Jan 7;6(1):e16089. doi: 10.1371/journal.pone.0016089.10.1371/journal.pone.0016089Search in Google Scholar PubMed PubMed Central

52 Anderson G, Seo S. Integrating Autism Spectrum Disorder Pathophysiology: Mitochondria, Vitamin A, CD38, Oxytocin, Serotonin and Melatonergic Alterations in the Placenta and Gut. Curr Pharm Des. In press.Search in Google Scholar

53 Garcia-Grau I, Simon C, Moreno I. Uterine microbiome-low biomass and high expectations. Biol Reprod. 2018. doi: 10.1093/biolre/ioy257.10.1093/biolre/ioy257Search in Google Scholar PubMed

54 Kwon M, Seo SS, Kim MK, Lee DO, Lim MC. Compositional and Functional Differences between Microbiota and Cervical Carcinogenesis as Identified by Shotgun Metagenomic Sequencing. Cancers (Basel). 2019;11(3). pii: E309. doi: 10.3390/cancers11030309.10.3390/cancers11030309Search in Google Scholar PubMed PubMed Central

55 Chen C, Song X, Wei W, Zhong H, Dai J, Lan Z, et al. The microbiota continuum along the female reproductive tract and its relation to uterine-related diseases. Nat Commun. 2017;8(1):875. doi: 10.1038/s41467-017-00901-0.10.1038/s41467-017-00901-0Search in Google Scholar PubMed PubMed Central

56 Brunelli R, Papi M, Arcovito G, Bompiani A, Castagnola M, Parasassi T, et al. Globular structure of human ovulatory cervical mucus. FASEB J. 2007;21(14):3872-6.10.1096/fj.07-8189comSearch in Google Scholar PubMed

57 Samani EN, Mamillapalli R, Li F, Mutlu L, Hufnagel D, Krikun G, et al. Micrometastasis of endometriosis to distant organs in a murine model. Oncotarget. 2017;10(23):2282-2291. doi: 10.18632/oncotarget.16889.10.18632/oncotarget.16889Search in Google Scholar PubMed PubMed Central

58 Bjorkman S, Taylor HS. MicroRNAs in endometriosis: biological function and emerging biomarker candidates.Biol Reprod. 2019;100(5):1135-1146. doi: 10.1093/biolre/ioz014.10.1093/biolre/ioz014Search in Google Scholar PubMed PubMed Central

59 Rekker K, Tasa T, Saare M, Samuel K, Kadastik Ü, Karro H, et al. Differentially-Expressed miRNAs in Ectopic Stromal Cells Contribute to Endometriosis Development: The Plausible Role of miR-139-5p and miR-375. Int J Mol Sci. 2018;19(12):pii: E3789. doi: 10.3390/ijms19123789.10.3390/ijms19123789Search in Google Scholar PubMed PubMed Central

60 Tsukamoto Y, Nakada C, Noguchi T, Tanigawa M, Nguyen LT, Uchida T, et al. MicroRNA-375 is downregulated in gastric carcinomas and regulates cell survival by targeting PDK1 and 14-3-3zeta. Cancer Res. 2010;70(6):2339-49. doi: 10.1158/0008-5472.CAN-09-2777.10.1158/0008-5472.CAN-09-2777Search in Google Scholar PubMed

61 Wu RL, Ali S, Bandyopadhyay S, Alosh B, Hayek K, Daaboul MF, et al. Comparative Analysis of Differentially Expressed miRNAs and their Downstream mRNAs in Ovarian Cancer and its Associated Endometriosis. J Cancer Sci Ther. 2015;7(7):258-265.Search in Google Scholar PubMed

62 Zhou X, Hu Y, Dai L, Wang Y, Zhou J, Wang W, et al. MicroRNA-7 inhibits tumor metastasis and reverses epithelial-mesenchymal transition through AKT/ERK1/2 inactivation by targeting EGFR in epithelial ovarian cancer. PLoS One. 2014;9(5):e96718. doi: 10.1371/journal.pone.0096718.10.1371/journal.pone.0096718Search in Google Scholar PubMed PubMed Central

63 Wan L, Zou Y, Wan LH, Wang LQ, Huang MZ, Wu J, et al. Tanshinone IIA inhibits the proliferation, migration and invasion of ectopic endometrial stromal cells of adenomyosis via 14-3-3ζ downregulation. Arch Gynecol Obstet. 2015;292(6):1301-9. doi: 10.1007/s00404-015-3766-2.10.1007/s00404-015-3766-2Search in Google Scholar PubMed

64 Zolbin MM, Mamillapalli R, Nematian SE, Goetz L, Taylor HS. Adipocyte alterations in endometriosis: reduced numbers of stem cells and microRNA induced alterations in adipocyte metabolic gene expression. Reprod Biol Endocrinol. 2019;17(1):36. doi: 10.1186/s12958-019-0480-0.10.1186/s12958-019-0480-0Search in Google Scholar PubMed PubMed Central

65 Takemura Y, Osuga Y, Harada M, Hirata T, Koga K, Yoshino O, et al. Concentration of adiponectin in peritoneal fluid is decreased in women with endometriosis. Am J Reprod Immunol. 2005;54(4):217-21.10.1111/j.1600-0897.2005.00300.xSearch in Google Scholar PubMed

66 Bohlouli S, Rabzia A, Sadeghi E, Chobsaz F, Khazaei M. in vitro Anti-Proliferative Effect of Adiponectin on Human Endometriotic Stromal Cells through AdipoR1 and AdipoR2 Gene Receptor Expression. Iran Biomed J. 2016;20(1):12-7.Search in Google Scholar PubMed

67 Alderman MH III, Yoder N, Taylor HS. The systemic effects of endometriosis.In: Seminars in reproductive medicine: Thieme Medical Publishers; 2017. p.263–70.10.1055/s-0037-1603582Search in Google Scholar PubMed

68 Zhao L, Gu C, Huang K, Fan W, Li L, Ye M, et al. Association between oestrogen receptor alpha (ESR1) gene polymorphisms and endometriosis: a meta-analysis of 24 case-control studies. Reprod Biomed Online. 2016;33(3):335-49. doi: 10.1016/j. rbmo.2016.06.003.10.1016/j.rbmo.2016.06.003Search in Google Scholar PubMed

69 Chuffa LG, Seiva FR, Fávaro WJ, Teixeira GR, Amorim JP, Mendes LO, et al. Melatonin reduces LH, 17 beta-estradiol and induces differential regulation of sex steroid receptors in reproductive tissues during rat ovulation. Reprod Biol Endocrinol. 2011;9:108. http://dx.doi.org/10.1186/1477-7827-9-10810.1186/1477-7827-9-108Search in Google Scholar PubMed PubMed Central

70 Liao TL, Lee YC, Tzeng CR, Wang YP, Chang HY, Lin YF, et al. Mitochondrial translocation of estrogen receptor β affords resistance to oxidative insult-induced apoptosis and contributes to the pathogenesis of endometriosis. Free Radic Biol Med. 2019 Jan 24;134:359-373. doi: 10.1016/j. freeradbiomed.2019.01.022.10.1016/j.freeradbiomed.2019.01.022Search in Google Scholar PubMed

71 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 May 20;136:22-34. doi: 10.1016/j. freeradbiomed.2019.03.027.10.1016/j.freeradbiomed.2019.03.027Search in Google Scholar PubMed

72 Tong X, Li Z, Wu Y, Fu X, Zhang Y, Fan H. COMT 158G/A and CYP1B1 432C/G polymorphisms increase the risk of endometriosis and adenomyosis: a meta-analysis. Eur J Obstet Gynecol Reprod Biol. 2014;179:17-21. doi: 10.1016/j. ejogrb.2014.04.039.10.1016/j.ejogrb.2014.04.039Search in Google Scholar PubMed

73 Zhu ZY, Mu YQ, Fu XM, Li SM, Zhao FX. Association of CYP1B1 gene polymorphisms and the positive expression of estrogen alpha and estrogen beta with endometrial cancer risk. Eur J Gynaecol Oncol. 2011;32(2):188-91.Search in Google Scholar PubMed

74 Yu Z, Tian X, Peng Y, Sun Z, Wang C, Tang N, et al. Mitochondrial cytochrome P450 (CYP) 1B1 is responsible for melatonin-induced apoptosis in neural cancer cells. J Pineal Res. 2018;65(1):e12478. doi: 10.1111/jpi.12478.10.1111/jpi.12478Search in Google Scholar PubMed

75 Gavin KM, Sullivan TM, Kohrt WM, Majka SM, Klemm DJ. Ovarian Hormones Regulate the Production of Adipocytes From Bone Marrow-Derived Cells. Front Endocrinol (Lausanne). 2018;9:276. doi: 10.3389/fendo.2018.0027610.3389/fendo.2018.00276Search in Google Scholar PubMed PubMed Central

76 Luo F, Huang WY, Guo Y, Ruan GY, Peng R, Li XP. 17β-estradiol lowers triglycerides in adipocytes via estrogen receptor α and it may be attenuated by inflammation. Lipids Health Dis. 2017;16(1):182. doi: 10.1186/s12944-017-0575-6.10.1186/s12944-017-0575-6Search in Google Scholar PubMed PubMed Central

77 Song IS, Jeong YJ, Jeong SH, Kim JE, Han J, Kim TH, et al. Modulation of Mitochondrial ERβ Expression Inhibits Triple-Negative Breast Cancer Tumor Progression by Activating Mitochondrial Function. Cell Physiol Biochem. 2019;52(3):468-485. doi: 10.33594/000000034.10.33594/000000034Search in Google Scholar PubMed

78 Cavallini A, Resta L, Caringella AM, Dinaro E, Lippolis C, Loverro G. Involvement of estrogen receptor-related receptors in human ovarian endometriosis. Fertil Steril. 2011;96(1):102-6. doi: 10.1016/j.fertnstert.2011.04.032.10.1016/j.fertnstert.2011.04.032Search in Google Scholar PubMed

79 Jung US, Jeong KJ, Kang JK, Yi K, Shin JH, Seo HS, et al. Effects of estrogen receptor α and β on the expression of visfatin and retinol-binding protein 4 in 3T3-L1 adipocytes. Int J Mol Med. 2013;32(3):723-8. doi: 10.3892/ijmm.2013.1440.10.3892/ijmm.2013.1440Search in Google Scholar PubMed

80 Chen C, Zhou Y, Hu C, Wang Y, Yan Z, Li Z, et al. Mitochondria and oxidative stress in ovarian endometriosis. Free Radic Biol Med. 2019;136:22-34. doi: 10.1016/j. freeradbiomed.2019.03.027.10.1016/j.freeradbiomed.2019.03.027Search in Google Scholar PubMed

81 Zhuang P, Shou Q, Lu Y, Wang G, Qiu J, Wang J, et al. Arachidonic acid sex-dependently affects obesity through linking gut microbiota-driven inflammation to hypothalamus-adipose-liver axis. Biochim Biophys Acta Mol Basis Dis. 2017;1863(11):2715-2726. doi: 10.1016/j.bbadis.2017.07.003.10.1016/j.bbadis.2017.07.003Search in Google Scholar PubMed

82 Souza-Teodoro LH, Dargenio-Garcia L, Petrilli-Lapa CL, Souza Eda S, Fernandes PA, Markus RP, et al. Adenosine triphosphate inhibits melatonin synthesis in the rat pineal gland. J Pineal Res. 2016;60(2):242-9. doi: 10.1111/jpi.12309.10.1111/jpi.12309Search in Google Scholar PubMed

83 Yang MH, Wang PH, Wang SJ, Sun WZ, Oyang YJ, Fuh JL. Women with endometriosis are more likely to suffer from migraines: a population-based study. PLoS One. 2012;7:e33941.10.1371/journal.pone.0033941Search in Google Scholar PubMed PubMed Central

84 Stevison F, Jing J, Tripathy S, Isoherranen N. Role of Retinoic Acid-Metabolizing Cytochrome P450s, CYP26, in Inflammation and Cancer. Adv Pharmacol. 2015;74:373-412. doi: 10.1016/bs.apha.2015.04.006.10.1016/bs.apha.2015.04.006Search in Google Scholar PubMed PubMed Central

85 Gou Y, Li X, Li P, Zhang H, Xu T, Wang H, et al. Estrogen receptor β upregulates CCL2 via NF-κB signaling in endometriotic stromal cells and recruits macrophages to promote the pathogenesis of endometriosis. Hum Reprod. 2019;34(4):646-658. doi: 10.1093/humrep/dez019.10.1093/humrep/dez019Search in Google Scholar PubMed

86 Liang Y, Xie H, Wu J, Liu D, Yao S. Villainous role of estrogen in macrophage-nerve interaction in endometriosis. Reprod Biol Endocrinol. 2018;16(1):122. doi: 10.1186/s12958-018-0441-z.10.1186/s12958-018-0441-zSearch in Google Scholar PubMed PubMed Central

87 Muxel SM, Pires-Lapa MA, Monteiro AW, Cecon E, Tamura EK, Floeter-Winter LM, et al. NF-κB drives the synthesis of melatonin in RAW 264.7 macrophages by inducing the transcription of the arylalkylamine-N-acetyltransferase (AA-NAT) gene. PLoS One. 2012;7(12):e52010. doi: 10.1371/journal. pone.0052010.10.1371/journal.pone.0052010Search in Google Scholar PubMed PubMed Central

88 Ohno M, Ikenaka Y, Ishizuka M. All-trans retinoic acid inhibits the recruitment of ARNT to DNA, resulting in the decrease of CYP1A1 mRNA expression in HepG2 cells. Biochem Biophys Res Commun. 2012;417(1):484-9. doi: 10.1016/j.bbrc.2011.11.146.10.1016/j.bbrc.2011.11.146Search in Google Scholar PubMed

89 Perri M, Caroleo MC, Liu N, Gallelli L, De Sarro G, Kagechika H, et al. 9-cis Retinoic acid modulates myotrophin expression and its miR in physiological and pathophysiological cell models. Exp Cell Res. 2017;354(1):25-30. doi: 10.1016/j. yexcr.2017.03.022.10.1016/j.yexcr.2017.03.022Search in Google Scholar PubMed

90 Basoli V, Santaniello S, Cruciani S, Ginesu GC, Cossu ML, Delitala AP, et al. Melatonin and Vitamin D Interfere with the Adipogenic Fate of Adipose-Derived Stem Cells. Int J Mol Sci. 2017;18(5). pii: E981. doi: 10.3390/ijms18050981.10.3390/ijms18050981Search in Google Scholar PubMed PubMed Central

91 Rhee YH, Ahn JC. Melatonin attenuated adipogenesis through reduction of the CCAAT/enhancer binding protein beta by regulating the glycogen synthase 3 beta in human mesenchymal stem cells. J Physiol Biochem. 2016;72(2):145-55. doi: 10.1007/s13105-015-0463-3.10.1007/s13105-015-0463-3Search in Google Scholar PubMed

92 Zhang L, Su P, Xu C, Chen C, Liang A, Du K, et al. Melatonin inhibits adipogenesis and enhances osteogenesis of human mesenchymal stem cells by suppressing PPARγ expression and enhancing Runx2 expression. J Pineal Res. 2010;49(4):364-72. doi: 10.1111/j.1600-079X.2010.00803.x.10.1111/j.1600-079X.2010.00803.xSearch in Google Scholar PubMed

93 Ng TB, Wong CM. Effects of pineal indoles and arginine vasotocin on lipolysis and lipogenesis in isolated adipocytes. J Pineal Res. 1986;3(1):55-66.10.1111/j.1600-079X.1986.tb00726.xSearch in Google Scholar PubMed

94 Nakagomi A, Okada S, Yokoyama M, Yoshida Y, Shimizu I, Miki T, et al. Role of the central nervous system and adipose tissue BDNF/TrkB axes in metabolic regulation. NPJ Aging Mech Dis. 2015;1:15009. doi: 10.1038/npjamd.2015.9.10.1038/npjamd.2015.9Search in Google Scholar PubMed PubMed Central

95 Mugabo Y, Sadeghi M, Fang NN, Mayor T, Lim GE. Elucidation of the 14-3-3ζ interactome reveals critical roles of RNA-splicing factors during adipogenesis. J Biol Chem. 2018;293(18):6736-6750. doi: 10.1074/jbc.M117.816272.10.1074/jbc.M117.816272Search in Google Scholar PubMed PubMed Central

96 Fan CF, Zhu AN, Huang TT, Li L, Wang SQ. Tetramethoxystilbene, a selective CYP1B1 inhibitor, suppresses adipogenesis of C3H10T1/2 pluripotent stem cells. Nan Fang Yi Ke Da Xue Xue Bao. 2015 Jan;35(1):72-6.Search in Google Scholar PubMed

97 Cruz ACC, Cardozo FTGS, Magini RS, Simões CMO. Retinoic acid increases the effect of bone morphogenetic protein type 2 on osteogenic differentiation of human adipose-derived stem cells. J Appl Oral Sci. 2019;27:e20180317. doi: 10.1590/1678-7757-2018-0317.10.1590/1678-7757-2018-0317Search in Google Scholar PubMed PubMed Central

98 Tugnoli B, Bernardini C, Forni M, Piva A, Stahl CH, Grilli E. Butyric acid induces spontaneous adipocytic differentiation of porcine bone marrow-derived mesenchymal stem cells. In Vitro Cell Dev Biol Anim. 2019;55(1):17-24. doi: 10.1007/s11626-018-0307-x.10.1007/s11626-018-0307-xSearch in Google Scholar PubMed

99 Yang F, Yang L, Li Y, Yan G, Feng C, Liu T, et al. Melatonin protects bone marrow mesenchymal stem cells against iron overload-induced aberrant differentiation and senescence. J Pineal Res. 2017t;63(3). doi: 10.1111/jpi.12422.10.1111/jpi.12422Search in Google Scholar PubMed

100 Sampson JA. Metastatic or Embolic Endometriosis, due to the Menstrual Dissemination of Endometrial Tissue into the Venous Circulation. Am J Pathol. 1927;3:93-110.43.Search in Google Scholar PubMed

101 Herington JL, Bruner-Tran KL, Lucas JA, Osteen KG. Immune interactions in endometriosis. Expert Rev Clin Immunol. 2011;7:611-26.10.1586/eci.11.53Search in Google Scholar PubMed PubMed Central

102 Zhang T, De Carolis C, Man GCW, Wang CC. The link between immunity, autoimmunity and endometriosis: a literature update. Autoimmun Rev. 2018;17(10):945-955. doi: 10.1016/j. autrev.2018.03.017.10.1016/j.autrev.2018.03.017Search in Google Scholar PubMed

103 Koninckx PR, Ussia A, Adamyan L, Wattiez A, Gomel V, Martin DC. Pathogenesis of endometriosis: the genetic/epigenetic theory. Fertil Steril. 2019;111(2):327-340. doi: 10.1016/j. fertnstert.2018.10.013.10.1016/j.fertnstert.2018.10.013Search in Google Scholar PubMed

104 Wang S, Su X, Xu M, Xiao X, Li X, Li H, et al. Exosomes secreted by mesenchymal stromal/stem cell-derived adipocytes promote breast cancer cell growth via activation of Hippo signaling pathway. Stem Cell Res Ther. 2019;10(1):117. doi: 10.1186/s13287-019-1220-2.10.1186/s13287-019-1220-2Search in Google Scholar PubMed PubMed Central

105 Flaherty SE 3rd, Grijalva A, Xu X, Ables E, Nomani A, et al. A lipase-independent pathway of lipid release and immune modulation by adipocytes. Science. 2019;363(6430):989-993. doi: 10.1126/science.aaw2586.10.1126/science.aaw2586Search in Google Scholar PubMed PubMed Central

106 Park JE, Dutta B, Tse SW, Gupta N, Tan CF, Low JK, et al. Hypoxia-induced tumor exosomes promote M2-like macrophage polarization of infiltrating myeloid cells and microRNA-mediated metabolic shift. Oncogene. 2019. doi: 10.1038/ s41388-019-0782-x.10.1038/s41388-019-0782-xSearch in Google Scholar PubMed

107 Li J, Xue H, Li T, Chu X, Xin D, Xiong Y, et al. Exosomes derived from mesenchymal stem cells attenuate the progression of atherosclerosis in ApoE-/- mice via miR-let7 mediated infiltration and polarization of M2 macrophage. Biochem Biophys Res Commun. 2019;510(4):565-572. doi: 10.1016/j. bbrc.2019.02.005.10.1016/j.bbrc.2019.02.005Search in Google Scholar PubMed

108 Kanlikilicer P, Bayraktar R, Denizli M, Rashed MH, Ivan C, Aslan B, et al. Exosomal miRNA confers chemo resistance via targeting Cav1/p-gp/M2-type macrophage axis in ovarian cancer. EBioMedicine. 2018;38:100-112. doi: 10.1016/j. ebiom.2018.11.004.10.1016/j.ebiom.2018.11.004Search in Google Scholar PubMed PubMed Central

109 Gao M, Allebeck P, Mishra GD, Koupil I. Developmental origins of endometriosis: a Swedish cohort study. J Epidemiol Community Health. 2019;73(4):353-359. doi: 10.1136/jech-2018-211811.10.1136/jech-2018-211811Search in Google Scholar PubMed PubMed Central

110 Lalani S, Choudhry AJ, Firth B, Bacal V, Walker M, Wen SW, et al. Endometriosis and adverse maternal, fetal and neonatal outcomes, a systematic review and meta-analysis. Hum Reprod. 2018;33(10):1854-1865. doi: 10.1093/humrep/dey269.10.1093/humrep/dey269Search in Google Scholar PubMed PubMed Central

111 Teletin M, Vernet N, Ghyselinck NB, Mark M. Roles of Retinoic Acid in Germ Cell Differentiation. Curr Top Dev Biol. 2017;125:191-225. doi: 10.1016/bs.ctdb.2016.11.013.10.1016/bs.ctdb.2016.11.013Search in Google Scholar PubMed

112 Duval F, Dos Santos E, Maury B, Serazin V, Fathallah K, Vialard F, et al. Adiponectin regulates glycogen metabolism at the human fetal–maternal interface J Mol Endocrinol. 2018;61(3):139-152. doi: 10.1530/JME-18-0013.10.1530/JME-18-0013Search in Google Scholar PubMed

113 Liu S, Cui H, Zhang Q, Hua K. Influence of early-life factors on the development of endometriosis. Eur J Contracept Reprod Health Care. 2019:1-6. doi: 10.1080/13625187.2019.1602723.10.1080/13625187.2019.1602723Search in Google Scholar PubMed

114 Anderson G, Vaillancourt C, Maes M, Reiter RJ. Breastfeeding and the gut-brain axis: is there a role for melatonin? Biomol Concepts. 2017;8(3-4):185-195. doi: 10.1515/bmc-2017-0009.10.1515/bmc-2017-0009Search in Google Scholar PubMed

115 Liu Y, Qin S, Song Y, Feng Y, Lv N, Xue Y, et al. The Perturbation of Infant Gut Microbiota Caused by Cesarean Delivery Is Partially Restored by Exclusive Breastfeeding. Front Microbiol. 2019;10:598. doi: 10.3389/fmicb.2019.00598.10.3389/fmicb.2019.00598Search in Google Scholar PubMed PubMed Central

116 Goetz LG, Mamillapalli R, Taylor HS. Low Body Mass Index in Endometriosis Is Promoted by Hepatic Metabolic Gene Dysregulation in Mice. Biol Reprod. 2016;95(6):115.10.1095/biolreprod.116.142877Search in Google Scholar PubMed PubMed Central

117 Wang S, Xu M, Li X, Su X, Xiao X, Keating A, et al. Exosomes released by hepatocarcinoma cells endow adipocytes with tumor-promoting properties. J Hematol Oncol. 2018;11(1):82. doi: 10.1186/s13045-018-0625-1.10.1186/s13045-018-0625-1Search in Google Scholar PubMed PubMed Central

118 Obata Y, Kita S, Koyama Y, Fukuda S, Takeda H, Takahashi M, et al. Adiponectin/T-cadherin system enhances exosome biogenesis and decreases cellular ceramides by exosomal release. JCI Insight. 2018;3(8):pii:99680. doi: 10.1172/jci.insight.99680.10.1172/jci.insight.99680Search in Google Scholar PubMed PubMed Central

119 Barra F, Grandi G, Tantari M, Scala C, Facchinetti F, Ferrero S. A comprehensive review of hormonal and biological therapies for endometriosis: latest developments. Expert Opin Biol Ther. 2019;19(4):343-360. doi: 10.1080/14712598.2019.1581761.10.1080/14712598.2019.1581761Search in Google Scholar PubMed

120 Ohlsson B. Gonadotropin-Releasing Hormone and Its Role in the Enteric Nervous System. Front Endocrinol (Lausanne). 2017;8:110. doi: 10.3389/fendo.2017.0011010.3389/fendo.2017.00110Search in Google Scholar PubMed PubMed Central

121 Schwertner A, Conceição Dos Santos CC, Costa GD, Deitos A, de Souza A, et al. Efficacy of melatonin in the treatment of endometriosis: a phase II, randomized, double-blind, placebo-controlled trial. Pain. 2013 Jun;154(6):874-81. doi: 10.1016/j. pain.2013.02.025.10.1016/j.pain.2013.02.025Search in Google Scholar PubMed

122 Rong B, Feng R, Liu C, Wu Q, Sun C. Reduced delivery of epididymal adipocyte-derived exosomal resistin is essential for melatonin ameliorating hepatic steatosis in mice. J Pineal Res. 2019;66(4):e12561. doi: 10.1111/jpi.12561.10.1111/jpi.12561Search in Google Scholar PubMed

123 Marseglia L, D’Angelo G, Manti S, Reiter RJ, Gitto E. Potential Utility of Melatonin in Preeclampsia, Intrauterine Fetal Growth Retardation, and Perinatal Asphyxia. Reprod Sci. 2016;23(8):970-7. Doi: 10.1177/1933719115612132.10.1177/1933719115612132Search in Google Scholar PubMed

124 Togher KL, Kenny LC, O’Keeffe GW. Class-Specific Histone Deacetylase Inhibitors Promote 11-Beta Hydroxysteroid Dehydrogenase Type 2 Expression in JEG-3 Cells. Int J Cell Biol. 2017;2017:6169310. doi: 10.1155/2017/6169310.10.1155/2017/6169310Search in Google Scholar PubMed PubMed Central

125 Wolff EF, Sun L, Hediger ML, Sundaram R, Peterson CM, Chen Z, Buck Louis GM. In utero exposures and endometriosis: the Endometriosis, Natural History, Disease, Outcome (ENDO) Study. Fertil Steril. 2013;99(3):790-5. doi: 10.1016/j. fertnstert.2012.11.013.10.1016/j.fertnstert.2012.11.013Search in Google Scholar PubMed PubMed Central

126 Mrkaić AG, Petrović AS, Nezhat FR, Trandafilović M, Vlajković S, Vasović LP. Some features of the developmental uterus in human fetuses. J Matern Fetal Neonatal Med. 2014;27(15):150712. doi: 10.3109/14767058.2013.860587.10.3109/14767058.2013.860587Search in Google Scholar PubMed

Received: 2019-05-11
Accepted: 2019-07-01
Published Online: 2019-07-19

© 2019 George Anderson, published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 Public License.

Downloaded on 5.6.2023 from https://www.degruyter.com/document/doi/10.1515/bmc-2019-0017/html
Scroll to top button