Cracking the enigma of adenomyosis: an update on its pathogenesis and pathophysiology

in Reproduction
Author:
Sun-Wei Guo Shanghai Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China

Search for other papers by Sun-Wei Guo in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0002-8511-7624

Correspondence should be addressed to S-W Guo; Email: hoxa10@outlook.com
Free access

Sign up for journal news

In brief

Traditionally viewed as enigmatic and elusive, adenomyosis is a fairly common gynecological disease but is under-recognized and under-researched. This review summarizes the latest development on the pathogenesis and pathophysiology of adenomyosis, which have important implications for imaging diagnosis of the disease and for the development of non-hormonal therapeutics.

Abstract

Traditionally considered as an enigmatic disease, adenomyosis is a uterine disease that affects many women of reproductive age and is a contributing factor for pelvic pain, heavy menstrual bleeding (HMB), and subfertility. In this review, the new development in the pathogenesis and pathophysiology of adenomyosis has been summarized, along with their clinical implications. After reviewing the progress in our understanding of the pathogenesis and describing the prevailing theories, in conjunction with their deficiencies, a new hypothesis, called endometrial–myometrial interface disruption (EMID), which is backed by extensive epidemiologic data and demonstrated by a mouse model, is reviewed, along with recent data implicating the role of Schwann cells in the EMI area in the genesis of adenomyosis. Additionally, the natural history of adenomyotic lesions is elaborated and underscores that, in essence, adenomyotic lesions are fundamentally wounds undergoing repeated tissue injury and repair (ReTIAR), which progress to fibrosis through epithelial–mesenchymal transition, fibroblast-to-myofibroblast transdifferentiation, and smooth muscle metaplasia. Increasing lesional fibrosis propagates into the neighboring EMI and endometrium. The increased endometrial fibrosis, with ensuing greater tissue stiffness, results in attenuated prostaglandin E2, hypoxia signaling and glycolysis, impairing endometrial repair and causing HMB. Compared with adenomyosis-associated HMB, the mechanisms underlying adenomyosis-associated pain are less understood but presumably involve increased uterine contractility, hyperinnervation, increased lesional production of pain mediators, and central sensitization. Viewed through the prism of ReTIAR, a new imaging technique can be used to diagnose adenomyosis more accurately and informatively and possibly help to choose the best treatment modality.

Abstract

In brief

Traditionally viewed as enigmatic and elusive, adenomyosis is a fairly common gynecological disease but is under-recognized and under-researched. This review summarizes the latest development on the pathogenesis and pathophysiology of adenomyosis, which have important implications for imaging diagnosis of the disease and for the development of non-hormonal therapeutics.

Abstract

Traditionally considered as an enigmatic disease, adenomyosis is a uterine disease that affects many women of reproductive age and is a contributing factor for pelvic pain, heavy menstrual bleeding (HMB), and subfertility. In this review, the new development in the pathogenesis and pathophysiology of adenomyosis has been summarized, along with their clinical implications. After reviewing the progress in our understanding of the pathogenesis and describing the prevailing theories, in conjunction with their deficiencies, a new hypothesis, called endometrial–myometrial interface disruption (EMID), which is backed by extensive epidemiologic data and demonstrated by a mouse model, is reviewed, along with recent data implicating the role of Schwann cells in the EMI area in the genesis of adenomyosis. Additionally, the natural history of adenomyotic lesions is elaborated and underscores that, in essence, adenomyotic lesions are fundamentally wounds undergoing repeated tissue injury and repair (ReTIAR), which progress to fibrosis through epithelial–mesenchymal transition, fibroblast-to-myofibroblast transdifferentiation, and smooth muscle metaplasia. Increasing lesional fibrosis propagates into the neighboring EMI and endometrium. The increased endometrial fibrosis, with ensuing greater tissue stiffness, results in attenuated prostaglandin E2, hypoxia signaling and glycolysis, impairing endometrial repair and causing HMB. Compared with adenomyosis-associated HMB, the mechanisms underlying adenomyosis-associated pain are less understood but presumably involve increased uterine contractility, hyperinnervation, increased lesional production of pain mediators, and central sensitization. Viewed through the prism of ReTIAR, a new imaging technique can be used to diagnose adenomyosis more accurately and informatively and possibly help to choose the best treatment modality.

Introduction

First documented by German pathologist Carl von Rokitansky in 1860 (Benagiano & Brosens 2006), adenomyosis is defined as the presence of endometrial glands and stroma within the myometrium (Bird et al. 1972). It is a uterine disease that affects many women of reproductive age and is a contributing factor for dysmenorrhea, pelvic pain, abnormal uterine bleeding (AUB), and subfertility (Farquhar & Brosens 2006, Vercellini et al. 2014, Harada et al. 2016, Gordts et al. 2018).

In spite of its long history, adenomyosis is a under-researched disease (Wang et al. 2022b), but the seemingly neglect may be changing. In the last 10 years, there has been a growing interest in adenomyosis as seen by a swell of excellent reviews on the pathogenesis and pathophysiology of adenomyosis in recent years (Benagiano et al. 2012, Vannuccini et al. 2017, Garcia-Solares et al. 2018, Vannuccini & Petraglia 2019, Zhai et al. 2020, Bulun et al. 2021, Donnez et al. 2021, Stratopoulou et al. 2021b, Khan et al. 2022).

This manuscript intends to review some recent development in adenomyosis pathogenesis and pathophysiology. Given the vastly diverse areas in adenomyosis research spanning from epidemiology, pathogenesis, and pathophysiology to clinical management, it is perhaps more productive not to rehash what have been reviewed or just compile more published results. Fittingly or not, the state of adenomyosis research is often likened to the fable of ‘the elephant and the blind men’, in that researchers around the world are groping to untie the Gordian knot of adenomyosis pathogenesis/pathophysiology, with each reporting certain important and novel finding of their own. However, it is apparent that the whole and entire truth is still amiss. At the core of this state is the seeming lack of a big picture. For a good review, it is perhaps more profitable to have a clear framework and then piece together most, if not all, published results. Indeed, if the blind men in that fable were told that the creature they were trying to fathom is a mammal, perhaps they could have built on this, pieced together all their findings, and reached a much more revealing conclusion that was closer to the truth.

With this in mind, I shall provide an overview of new developments in the quest for the pathogenesis and pathophysiology of adenomyosis and then use the perspective that we gained from these findings to understand the pathophysiology of adenomyosis-associated heavy menstrual bleeding (HMB) and pain, and then mention, very briefly, how the new understanding of the natural history of adenomyotic lesions can help devise better imaging diagnostic and non-hormonal therapeutics. Due to space limitation, adenomyosis-associated infertility will not be reviewed here but can be found in a recent review (Khan et al. 2022). In particular, I shall capitalize on the intrinsic similarity that both adenomyosis and endometriosis share in common and extrapolate findings in endometriosis to adenomyosis. It is important to note that, because of the emphasis on a major framework, inevitably, this review has, by necessity, to pick and choose those data that are deemed to be relevant and not superfluous. By so doing, however, this may engender the risk of being biased in favor of the data generated from this author’s own lab, but by no means other published data would be slighted entirely. It just means that we have to comb through the vast sea of publications to get a firm grip of the nutshell of the matter.

Etiology and pathogenesis

The prevailing theories

There are currently two prevailing and competing theories: invagination and metaplasia (Garcia-Solares et al. 2018). The invagination theory was built primarily upon the tissue injury and repair (TIAR) hypothesis proposed by Leyendecker and his colleagues (Leyendecker et al. 2009, 2015, Leyendecker & Wildt 2011). The word ‘metaplasia’ was created following anatomical and histological observations of the unexpected appearance of foreign tissues in ectopic sites (Slack 1986) and is now commonly used to refer to the conversion from one type of differentiated cell to another (Slack & Tosh 2001).

In essence, the TIAR theory postulates that repeated and sustained myometrial overstretching due to hyperperistalsis of unknown origin would cause injury to the myocytes and fibroblasts in the junctional zone (JZ), an anatomic region originally defined by the MRI that overlaps with the endometrial–myometrial interface (EMI). This event, termed microtrauma, would focally activate the TIAR system with increased inflammation and local production of estrogens, which, in turn, induce more inflammation and more estrogen production, establishing a feed-forward loop that further induces uterine hyperperistalsis through ERα induction of the oxytocin (OT)/OT receptor (OTR) system (Leyendecker et al. 2009, Leyendecker & Wildt 2011). The chronic hyperperistalsis in the JZ facilitates repetitive autotraumatization, causing the disruption of the muscular fibers in the EMI, eventually leading to the invagination of endometrial basal layer into the myometrium, and thus adenomyosis. In fact, this theory is recently extended to encompass endometriosis as well, which is subsumed, along with adenomyosis, into the term ‘archimetrosis’ to indicate the common origin (Leyendecker et al. 2022).

The metaplasia theory posits that adenomyotic lesions may originate from metaplasia of displaced embryonic pluripotent Müllerian remnants (Ferenczy 1998). The underlying mechanisms are completely unclear. Alternatively, endometrial stem/progenitor cells or their niche cells may behave aberrantly for some unknown reason and their differentiating progeny cells move toward the myometrium, rather than functionalis, resulting in adenomyosis (Gargett 2007).

Despite the appeal, unfortunately, so far there have been no experimental data whatsoever to support or refute either of the theories or the TIAR hypothesis (Wang et al. 2022b). Of particular note is that neither theory seems to be backed by any epidemiological data. For the invagination theory, it is unclear as to why and how this injury – which amounts to primum movens – occurs. Indeed, uterine peristalsis occurs in all women of reproductive age, but why is there only a fraction of them develop adenomyosis? What are the risk factors, if any, for this hyperperistalsis/dysperistalsis? Is it possible to intervene or prevent it? What can be done to mitigate the risk of hyperperistalsis in the first place? It is also unclear why and how the stem cells are recruited and then turned into endometrial epithelial and stromal cells that respond to hormonal fluctuations and become adenomyotic lesions.

A good theory has to satisfy at least three basic requirements: falsifiability, explanatory power, and predictivity. While both theories could explain some phenomena, they have to be falsifiable and have predictive power. Hence, a better alternative would be to dramatically revamp the existing theories and start anew.

An emerging hypothesis

A large body of epidemiological data has shown, quite consistently, that iatrogenic uterine procedures, such as dilatation and curettage and induced abortion, increase the risk of developing adenomyosis later in life (Parazzini et al. 1997, Levgur et al. 2000, Taran et al. 2010). With this in mind, the hypothesis of endometrial–myometrial disruption (EMID) has been proposed (Guo 2020b). This hypothesis predicted that, first, the magnitude of risk of developing adenomyosis resulting from EMID depends on the severity and mode of disruption; and secondly, perioperative intervention may help reduce the risk of adenomyosis (Guo 2020b). This may explain why not all women who underwent iatrogenic uterine procedures develop adenomyosis, simply because different women may simply have experienced different modes or degrees of EMID. If EMID is extensive and severe enough, there should be an elevated risk of developing adenomyosis as opposed to minor or no EMID.

Additionally, the EMID, as tissue trauma, would cause tissue injury, eliciting the release of substance P (Mantyh 1991) and PGE2 (Kirchhoff et al. 2009), and activate the hypothalamic–pituitary–adrenal (HPA)/sympathetic–adrenal–medullary (SAM) axes, resulting in increased release of catecholamines such as adrenaline/noradrenaline, which, in turn, may suppress cell-mediated immunity (Goldfarb et al. 2011), hampering the capability of immune cells to remove endometrial cells that invaded into the myometrium through breached EMI. Indeed, in mice that received the EMID procedure, the systemic levels of substance P, PGE2, adrenaline, and noradrenaline are all elevated as compared with those receiving a sham surgery (Wang et al. unpublished data). It has been reported that both adrenaline and noradrenaline suppress IL-12 production in a dose-dependent fashion and at physiological concentrations while dose-dependently increasing the production of IL-10, potentiating Th2-regulated responses (humoral immunity) (Elenkov et al. 1996). Yet, IL-12 is known to be a key inducer of differentiation of uncommitted T helper (Th) cells toward the Th1 phenotype, which regulates cellular immunity, whereas IL-10 inhibits Th1 functions and potentiates Th2-regulated responses (i.e. humoral immunity). Both catecholamines also reduced NK cell cytotoxicity (Ben-Eliyahu et al. 2000).

In addition, PGE2 has been shown to reduce IL-12 production (Stolina et al. 2000) and CD4+ and CD8+ T cell numbers and CD4+ T cell cytotoxicity (Yang et al. 2003), increase IL-10 levels (Stolina et al. 2000), as well as regulatory T cell inhibitory capacity and numbers (Baratelli et al. 2005, Sharma et al. 2005).

The abundance of adrenergic nerve fibers in the uterus, especially in the EMI region (Alm & Lundberg 1988, Bae et al. 2001), seems to give credence to this view. This would indicate that perioperative intervention to counter either the action of the receptor for substance P, neurokinin receptor 1 (NK1R), or to contain the release of PGE2 by inhibiting COX-2 or the adrenaline receptors would reduce the risk of adenomyosis resulting from EMID (Hao et al. 2020). Indeed, perioperative administration of an NK1R inhibitor or a β-blocker plus a COX-2 inhibitor did reduce the risk of developing adenomyosis in mice (Hao et al. 2020).

Pursuit of this hypothesis, a mouse model mimicking iatrogenic uterine procedures has been established (Hao et al. 2020). Aside from being consistent with epidemiologic data and demonstrating the falsifiability of the EMID hypothesis, this model also demonstrates that indeed the risk of adenomyosis did depend precisely on the severity and mode of EMID, and, more importantly, perioperative intervention can significantly reduce the risk of developing adenomyosis (Hao et al. 2020). Hopefully, the insight gained from understanding of how EMID increases the risk of developing adenomyosis can help us unravel the adenomyosis pathogenesis due to other causes.

It should be noted that the EMID hypothesis has also been validated independently recently by Hiraoka et al. using multiple punctures onto the EMI in mouse (Hiraoka et al. 2022). Thus, as long as the EMID is severe enough, it will cause adenomyosis.

The EMID hypothesis and its experimental evidence provide a good explanation for primum movens. It should be noted that the nascent lesional focus, once formed, could still face possible removal by immune cells, and the successful establishment of an adenomyotic lesion would depend on whether it can resist the removal. However, once the lesion is securely established, and barring any extraneous factors or lifestyle/behavioral changes that either boost the immunity or decelerate the progression (such as caloric restriction or eustress (Yin et al. 2018, 2020)), the established lesion should progress mostly on cyclic bleeding (Guo 2018b). Conceivably, the lesional progression could, in fact, be facilitated by psychogenic stress (Long et al. 2016b, Guo et al. 2017), high-fat diet (Heard et al. 2016), lower dairy consumption (Nodler et al. 2020), surgery (Liu et al. 2016a, Long et al. 2016a), history of adverse early life events (Long et al. 2020), and perhaps other factors yet to be identified.

After an adenomyotic focus is successfully established, it would undergo cyclic bleeding similar to the eutopic endometrium (Brosens 1997) and, as such, effectively become a wound that undergoes repeated tissue injury and repair (ReTIAR) (Guo 2018b), progressing to fibrosis through epithelial–mesenchymal transition (EMT), fibroblast-to-myofibroblast transdifferentiation (FMT), and smooth muscle metaplasia (SMM) (Liu et al. 2016b, Shen et al. 2016). In other words, once the primum movens is effectively applied, everything else will be set in motion, more or less on its own, resulting in adenomyosis as we see it.

What is special about the EMI?

While EMID induction of adenomyosis has been experimentally demonstrated, one lingering issue is why and how. Of course, injury resulting from the EMID would lead to platelet aggregation, inflammation, and hypoxia, resulting in the release of copious inflammatory cytokines such as IL-1β and growth factors such as TGF-β1, as well as increased local estrogen production (Guo 2020b, Qi et al. 2020). The increased estrogen levels may promote EMT and enhance the invasiveness of endometrial epithelial cells, facilitating the invasion of these cells to cross the breached EMI and into the myometrium, establishing the adenomyotic lesion (Chen et al. 2010).

It should be noted that the EMI is more than just a demarcation area that separates endometrium from myometrium or a physical barrier that impedes or block the invasion of endometrial epithelial cells into the myometrium. In fact, it is the region where the endometrial stem cells reside (Cousins et al. 2018, Tempest et al. 2020). It is also the location from which uterine contraction is started (Brosens et al. 2010). More importantly, EMI, unlike endometrium or myometrium, is densely innervated with peripheral nerves (Krantz 1959, Quinn 2007, Barcena de Arellano et al. 2013b, Zhang et al. 2014), maintained by ensheathing Schwann cells (SCs) that are derived from neural crest (Jessen et al. 2015). SCs are the most important type of glial cells in the peripheral nerve system (PNS) and are distributed nearly ubiquitously in the human body (Alvarez-Suarez et al. 2020). Their abundance, coupled with their remarkable plasticity capable of dedifferentiating and re-differentiating following injury to the nerve, endows their important roles in repairing tissues and in promoting cancer progression (Jessen et al. 2015, Carr & Johnston 2017, Stierli et al. 2019, Sun et al. 2022).

Following injury, SCs rapidly dedifferentiate to an unmyelinated progenitor state that promotes axonal regeneration and repair (Carr & Johnston 2017). Of course, there are nuances when it comes to injured SCs (called bridge cells), intact region, and distal region (Clements et al. 2017). Elevated levels of TGF-β, which is aplenty in the wounded tissues due to activated platelets, also stimulate the targeted migration of SCs across the bridge, providing a conducive substrate for successful nerve regeneration and tissue repair (Clements et al. 2017). Dedifferentiated SCs (dSCs) overexpress genes encoding for secreted factors intimately associated with tissue repair, such as growth factors, TGF-β signaling, chemotaxis and inflammation, migration and adhesion, extracellular matrix (ECM) remodeling, and angiogenesis (Parfejevs et al. 2018). In particular, dSCs can release TGF-β3 that facilitates fibroblast proliferation and thus promotes wound healing (Ou et al. 2022). TGF-β signaling reprograms bridge SCs that are dedifferentiated to invasive mesenchymal-like cells (Clements et al. 2017). In the context of EMID, injury-induced dSCs can induce EMT in neighboring endometrial epithelial cells by releasing TGF-β1 (Wang et al. unpublished data).

Since EMID causes not only tissue injury but also nerve injury, naturally SCs should be viewed as a high-value suspect in causing adenomyosis. Indeed, when peripheral nerves are injured, SCs can dedifferentiate into immature SCs and acquire stemness (Lee & Wolfe 2000, Jessen et al. 2015, Jessen & Mirsky 2016). In addition, SCs can also transdifferentiate into endoneurial fibroblasts (Joseph et al. 2004), chromaffin cells (Kastriti et al. 2019), melanocytes (Adameyko et al. 2009), mesenchymal cells (Kaukua et al. 2014), and parasympathetic, sympathetic, enteric, GABAergic, glycinergic, serotoninergic, and cholinergic neurons (Dyachuk et al. 2014, Espinosa-Medina et al. 2014, Su et al. 2014, Wang et al. 2016, Milichko & Dyachuk 2020). In view of their pleiotropic roles after injury, there is reason to believe that EMID may cause injury to peripheral nerves residing in the EMI region, resulting in SC dedifferentiation. Inundated by inflammatory cytokines such as IL-1β (Shaw & Martin 2009), estrogens resulting from a hypoxia microenvironment (Qi et al. 2020), and growth factors such as TGF-β1 released copiously by activated platelets and other immune cells (Guo 2020b), it is conceivable the dedifferentiated SCs might be coaxed and differentiated into endometrial epithelial cells. Furthermore, these SC-turned endometrial epithelial and stromal cells (through EMT) may form the original focus of adenomyotic lesion. That is, SCs residing in the EMI, when injured, would become dedifferentiated and turn into endometrial epithelial and stromal cells, forming an original adenomyotic lesion in disrupted myometrium.

To test the hypothesis that SCs are involved in inducing adenomyosis resulting from EMID, we treated mice perioperatively with either MEK/ERK or JNK inhibitors or vehicle 4 h before and 24, 48, and 72 h after the EMID procedure since Ras/Raf/ERK and c-Jun N-terminal kinase (JNK) are two important signaling pathways involved in the dedifferentiation of SCs (Jessen et al. 2015, Nocera & Jacob 2020) and inhibition of either pathway can prevent SCs from dedifferentiation (Harrisingh et al. 2004, Blom et al. 2014). We found that EMID resulted in progressive SCs dedifferentiation, concomitant with increased abundance of epithelial cells in the myometrium and subsequent EMT (Wang et al. 2022c). This EMID-induced change was abrogated significantly with perioperative administration of JNK or MEK/ERK inhibitors. Consistently, perioperative administration of a JNK or a MEK/ERK inhibitor reduced the incidence by nearly 33.5 and 14.3%, respectively, in conjunction with reduced myometrial infiltration of adenomyosis and alleviation of adenomyosis-associated hyperalgesia (Wang et al. 2022c). Both treatments significantly decelerated adenomyosis establishment and progression of EMT, FMT, and fibrogenesis in adenomyotic lesions. These data provide the first piece of evidence, strongly implicating the involvement of SCs in the pathogenesis of adenomyosis induced by EMID (Wang et al. 2022c).

It is also possible that terminal SCs residing in the EMI are also involved in uterine peristalsis. This is because the terminal SC area is known to play a role in neuromuscular junctions that are involved in muscle contraction (Alvarez-Suarez et al. 2020). In addition, SC-derived extracellular vesicles may also help to form the lesion by recruiting endometrial stem cells (Wang et al. 2022a). However, data on this area are very scanty, and further investigations are warranted.

Figure 1 provides a schematic illustration of possible mechanisms underlying EMID-induced adenomyosis. It should be emphasized that EMID is just one cause of adenomyosis. There are adenomyosis cases that are not caused by EMID.

Figure 1
Figure 1

Schematic illustration of the formation of adenomyotic lesions due to the endometrial–myometrial interface disruption (EMID). Iatrogenic procedures cause disruption at the endometrial–myometrial interface (EMI), resulting in tissue injury, including nerve injury. The tissue injury leads to platelet aggregation, inflammation, and the induction of HIF-1α, effectively causing tissue hypoxia. As a result, genes involved in estrogen production are upregulated, resulting in increased local production of estrogen and subsequent induction of both ERα and ERβ, which, in turn, leading to the induction of the OT/OTR signaling and increased uterine peristalsis. In addition, tissue hypoxia activates TGF-β1, VEGF, COX-2, and SDF-1 signaling pathways, leading to increased angiogenesis, vasculature, and recruitment of BMDSCs to the wounding site. The induction of COX-2 would also increase the production of PGH2 and TXA2, which also enhances uterine peristalsis. Moreover, the TGF-β1 signaling pathway induces EMT, leading to the invasion of endometrial epithelial cells to the EMI and further down to the myometrium. Tissue injury also would activate the HPA/SAM axes, leading to the release of catecholamines and PGE2, which collectively result in impaired cell-mediated immunity and, as such, enhances the survival of displaced and dispersed endometrial cells within the myometrium. Nerve injury results in the release of substance P and the induction of NK1R. It also leads to dedifferentiation of ensheathing Schwann cells, which are turned into endometrial epithelial cells under the influence of IL-1β and growth factors such as TGF-β1. These newly minted endometrial epithelial cells can also undergo EMT and differentiate into endometrial stromal cells. All these events ultimately lead to the formation of adenomyotic lesions in the myometrium. BMDSC, bone marrow-derived stem cells; COX-2, cyclooxygenase-2; E2, 17β-estradiol; EMI, endometrial–myometrial interface; EMT, epithelial–mesenchymal transition; ERα, estrogen receptor α; ERβ, estrogen receptor β; HIF-1α, hypoxia-inducible factor 1α; HPA, hypothalamic–pituitary–adrenal; OT, oxytocin; OTR, oxytocin receptor; P450 aromatase, aromatase; PGE2, prostaglandin E2; SAM, sympathetic–adrenal–medullary; SDF-1, stromal cell-derived factor 1; SF-1, steroidogenic factor-1; TGF-β1, transforming growth factor β1; TXA2, thromboxane A2; VEGF, vascular endothelial growth factor.

Citation: Reproduction 164, 5; 10.1530/REP-22-0224

Heterogeneity in pathogenesis

Growing evidence suggests that different subtypes of adenomyosis appear to have different symptomology and likely pathogenesis. Internal adenomyosis, that is, lesions proximal to the endometrium, has been shown to be more likely to be associated with HMB, while external adenomyosis distal to the endometrium but proximal to the uterine serosa is more closely linked with deep endometriosis (Kishi et al. 2012, Khan et al. 2019, Bourdon et al. 2021). It has been documented recently that there is a close link between external/extrinsic adenomyosis and deep endometriosis (Chapron et al. 2017, Marcellin et al. 2018, 2020, Khan et al. 2019, Bourdon et al. 2021), implying that the two disease entities may have a causal relationship or that the two entities may represent the same disease (Donnez et al. 2019). Equally likely, one disease entity could be simply caused by the other due to sheer physical proximity of each other and the invasiveness of ectopic endometrium (Gaetje et al. 1995). Of particular interest is that intrinsic/internal adenomyosis is found to contain almost tall columnar cell type with abundant stromal cells around glands reminiscent of the endometrial gland cells and stroma supporting their origin from endometrium, yet, in contrast, in extrinsic/external adenomyosis, the glandular epithelial cells are found to be mostly thin with scanty CD10-positive stromal cells that closely similar to their coexisting deep endometriotic lesions (Khan et al. 2019).

So far, preliminary mutation data seem to hint that deep endometriotic lesions adjacent to adenomyosis exhibited a higher KRAS mutation frequency than its neighboring adenomyotic lesions (Anglesio et al. 2017). Assuming a clonal relationship exists, this seems to suggest that external/extrinsic adenomyotic lesions may be colonized by their adjacent deep endometriotic lesions. At this moment, however, these data are too scarce to be conclusive.

The recent discovery of the three-dimensional histoarchitecture of the endometrium (Tempest et al. 2020, Yamaguchi et al. 2021) would lend support to the notion that EMID resulting from iatrogenic uterine procedures would likely cause intrinsic/internal, but less likely extrinsic/external, adenomyosis simply because of physical proximity. This seems to be supported by published data reporting that internal adenomyosis is associated with a history of uterine procedures (Kishi et al. 2012, Kobayashi et al. 2021).

One mouse experiment provides support for this notion. CD-1 mice neonatally fed with tamoxifen can develop adenomyosis (Parrott et al. 2001). As a selective estrogen receptor modulator, the action of tamoxifen is mediated through ERα and ERβ (Jordan 2006), as well as non-genomic mechanisms, such as through the G-protein coupled receptor 30 (GPR30) signaling pathway (Ignatov et al. 2010, Mo et al. 2013, Rouhimoghadam et al. 2018, Liu et al. 2021). In a manner identical to the tamoxifen induction of adenomyosis, mice neonatally fed with tamoxifen and diarylpropionitrile (DPN, an ERβ agonist) yielded adenomyosis in 100 and 50% of ICR mice, respectively (Cao et al. 2022), while those fed with propylpyrazoletriol (PPT, an ERα agonist) or G-1 (a GPR30 agonist) did not develop adenomyosis. Of particular note, adenomyotic lesions in the DPN group were found to be restricted to the serosal layer reminiscent of extrinsic/external adenomyosis in humans (Kishi et al. 2012, Bazot & Darai 2018) and were quite different from those induced by tamoxifen. These findings not only demonstrate that neonatal feeding of tamoxifen does not induce adenomyosis through a single ER but also seem to suggest that the pathogenesis of extrinsic/external adenomyosis may be different from other subtypes of adenomyosis.

Pathophysiology

Adenomyosis can cause HMB, dysmenorrhea and other pelvic pains, and infertility. From the perspective of healthcare providers, it suffices to understand the pathophysiology for the purpose of management. As of now, the pathophysiology underlying adenomyosis-induced symptomology is still poorly understood. To understand pathophysiology, the crux is, first and foremost, to understand the natural history of adenomyotic lesions, at least in broad strokes, since these lesions conceivably serve as the root causes for all the symptoms manifested.

The natural history of adenomyotic lesions

In the last 6–7 years, the natural history of adenomyotic lesions, at least in silhouette form, has emerged. In a nutshell, as adenomyotic lesions experience cyclic bleeding just as eutopic endometrium, they are fundamentally wounds undergoing ReTIAR (Guo 2018b). Similar to endometriotic lesions, they undergo EMT, FMT, and SMM and progress ultimately to fibrosis (Liu et al. 2016b, Shen et al. 2016, Zhang et al. 2016). Due to FMT and SMM, stromal cells within lesions eventually are turned into smooth muscle cells (SMCs) (Liu et al. 2016b, Shen et al. 2016, Zhang et al. 2016), which are fused into existing myometrium, causing enlarged uterus in women with adenomyosis.

Of course, myometrial SMCs from women with adenomyosis also exhibit cellular hypertrophy (Mehasseb et al. 2010), most likely due to the activation of the MAPK/ERK and the PI3K/mTOR/AKT pathways (Streuli et al. 2015) and, curiously, the upregulation of cannabinoid receptor CB1 (Wang et al. 2021a). Irrespective of the sources, the enlarged uterus is likely to result in increased magnitude of uterine contraction, especially when oxytocin receptor (OTR) is overexpressed (Guo et al. 2013). In addition, due to the fusion of newly turned SMCs into the myometrium, the uterine contraction is likely to be out of synchronization, resulting in dysperistalsis (Mao et al. 2011).

Aside from ectopic endometrial cells, many other cells within lesions and the uterus also participate in the progression of adenomyosis. Platelets, for example, can induce EMT, FMT, and SMM in ectopic endometrium, facilitating fibrogenesis (Liu et al. 2016b, Shen et al. 2016, Zhang et al. 2016). Vascular endothelial cells, stimulated by activated platelets, can be turned into myofibroblasts within lesions through endothelial–mesenchymal transition (Yan et al. 2020b). Activated platelets can also promote mesothelial–mesenchymal transition in peritoneal mesothelial cells (Yan et al. 2020a), causing adhesion and immobility of the uterus in women with adenomyosis. In addition, alternatively activated macrophages and regulatory T cells (Tregs) within lesions could promote lesional fibrogenesis through type II immunity (Bacci et al. 2009, Duan et al. 2018, Xiao et al. 2020).

Among all the molecular/cellular events involved in adenomyosis development, EMT appears to be the most documented. Many factors/molecules have been reported to be involved in EMT in the context of adenomyosis: estrogen (Chen et al. 2010), β-catenin (Oh et al. 2013), HGF (Khan et al. 2015), Notch1 (Qi et al. 2015), platelet-derived TGF-β1 (Liu et al. 2016b, Shen et al. 2016), macrophages (An et al. 2017a,b), ILK (Zhou et al. 2018), eIF3e (Cai et al. 2019), FAK (Zheng et al. 2018), and Talin1 (Wang et al. 2021b), among others. However, it is well-known that, aside from its involvement in development and cancer metastasis, EMT is known to be vital in wound healing, where it is rapidly activated and results in wound closure through re-epithelialization (Savagner 2015). Along the same vein, FMT or myofibroblast activation also is critical in tissue repair, as it leads to tissue contraction, restoring tissue integrity and reducing the wound size (Hinz 2016). Thus, from the ReTIAR perspective, the involvement of EMT in adenomyosis should be considred as obligatory.

Ectopic endometrial stromal cells can secrete neurotrophic factors, such as NGF, NT-3, TrkB, TXA2, and GDNF (Barcena de Arellano et al. 2011, 2013a, Greaves et al. 2015, Yan et al. 2017a), and axon guidance molecules such as Semaphorin 3E and SLIT/ROBO (Greaves et al. 2014, Asally et al. 2019), resulting in increased nerve fiber density and hyperinnervation in lesions (Wang et al. 2009a,b, Zhang et al. 2010). Moreover, eutopic endometrium from women with endometriosis, and possibly with adenomyosis as well, can promote neuroangiogenesis through exosome release (Sun et al. 2019).

As a consequence of hyperinnervation in endometrium and myometrium from women with adenomyosis (Zhang et al. 2009, 2010, Lertvikool et al. 2014, Choi et al. 2015), any minute or seemingly innoxious nociceptive stimulation, or elevation in pain mediators, or hyperperistalsis/dysperistalsis would be transduced, through the ascending transmission from primary afferent terminals to dorsal root ganglia and then to the central nervous system (CNS), resulting in the perception of pain. This, coupled with loss of GABAergic inhibition that inhibits the transfer of nociceptive signals from primary afferent fibers to the CNS in adenomyosis (Chen et al. 2014), would exacerbate the pain sensation.

Conversely, with the increase in nerve fibers, especially elevated density of sensory nerve fibers, there would be an increase in the secretion of neuropeptides. Yet, several neuropeptides, such as substance P and calcitonin gene-related peptide (CGRP), can facilitate EMT, FMT, and SMM in ectopic endometrium (Liu et al. 2019, Yan et al. 2019), accelerating lesional fibrosis. In other words, adenomyotic lesions and their surrounding nerves are partners in crime that cause pain in women with adenomyosis (Yan et al. 2017b).

Pain is a potent stressor, and HMB and/or infertility also would cause anxiety, discomfort, depression, and stress of various degrees. Yet, chronic or persistent stress would inevitably activate the HPA/SAM axes, yielding the release of catecholamines. Adrenaline and noradrenaline would activate the adrenergic β2 (ADRB2) and cAMP-responsive element-binding protein (CREB) on ectopic endometrium, accelerating the lesional progression (Long et al. 2016b, Guo et al. 2017). Chronic stress can also suppress dopamine receptor D2 (DRD2) expression in ectopic endometrium (Guo et al. 2017). In contrast, eustress, or the opposite of stress, increases the lesional expression of DRD2 and retards lesional progression (Yin et al. 2020). This seems to echo the reports that dopamine or DRD2 agonists could block stress-facilitated angiogenesis and proliferation in cancers (Moreno-Smith et al. 2011, 2013). Thus, a vicious cycle of adenomyosis–pain–stress–lesional progression–more pain could be established.

Aside from platelets, immune cells, endothelial cells, and sensory nerves, dSCs may also participate in facilitating lesional progression. This is because that SCs can become dedifferentiated not only because of injury but also due to other pathological stimuli, such as hypoxia (Demir et al. 2016) and STAT3 activation (Benito et al. 2017), which have been reported to be involved in adenomyosis (Goteri et al. 2009, Guo et al. 2021, Hiraoka et al. 2022). Indeed, dSCs release many secreted factors that are actively involved in tissue repair and promote myofibroblast differentiation by paracrine modulation of TGF-β signaling (Parfejevs et al. 2018), likely facilitating progression and fibrogenesis adenomyotic lesions. Research in this area has been ongoing, and the precise role of SCs in adenomyosis will be hopefully clarified.

Adenomyosis is widely regarded to exhibit progesterone resistance (Vannuccini et al. 2017), manifesting as progesterone unresponsiveness in ectopic endometrium and possibly in eutopic endometrium as well. Overexpression of ERβ, coupled with reduced expression of ERα, may play a role in decreased progesterone receptor (PR) expression and thus is responsible for progesterone resistance (Bulun et al. 2019). One proximate cause is the promoter hypermethylation of PR-B and thus its silence (Jichan et al. 2010), which might be caused by chronic and prolonged exposure to inflammatory cytokines such as TNF-α (Wu et al. 2008). KRAS mutation, likely a result of lesional progression (Guo 2018a, 2020a), also has been reported to be linked with progesterone resistance in adenomyosis (Inoue et al. 2019).

Aside from these possible causes, progressive fibrogenesis would lead to reduced vascular density and PR expression (Liu et al. 2018b) and, as such, may also contribute to progesterone resistance. In addition, increasing lesional fibrosis may yield epigenetic aberrations (Liu et al. 2018b). These epigenetic aberrations (Liu & Guo 2012, Liu et al. 2012) can be refractory to medical treatment or rectification and reduced lesional vascularity would pose a great challenge to deliver therapeutic agents to the target tissues. These aberrations, in conjunction with KRAS mutation, reduced PR-B expression or even silencing and other epigenetic aberrations would explain why adenomyosis is typically resistant to medical treatment, in general, and to progesterone/progestins, in particular.

Figure 2 is a schematic sketch of known and possible mechanisms underlying the natural history of adenomyosis. Note that the natural history is similar to that of endometriotic lesions but not identical, simply due to the different lesional microenvironments and also to the closer proximity to myometrium (which may be easier to cause dysperistalsis) and to endometrium (which may be more likely to impact on endometrial repair).

Figure 2
Figure 2

Schematic illustration of the natural history of ectopic endometrium. The arrows indicate the crosstalk between adenomyotic lesions and other cells in the lesional microenvironment. Pain, HMB, and/or infertility would constitute psychological stress, activating the HPA/SAM axes, resulting in the release of adrenaline/noradrenaline, which, when reaching adenomyotic lesions, downregulate DRD2 but upregulate ADRB2 in adenomyotic lesions, promoting the progression of adenomyosis. Many cells in the lesional microenvironment interact with adenomyotic lesions, and cytokines/chemokines, growth factors, and neuropeptides released by these cells, such as platelets (anuclear), immune cells (such as macrophages and lymphocytes), sensory nerve fibers, and Schwann cells can facilitate the development of adenomyotic lesions. Type 2 immunity also promotes fibrogenesis of adenomyotic lesions. ADRB2, adrenoceptor β2; DRD2, dopamine receptor D2; EMT, epithelial–mesenchymal transition; EndoMT, endothelial–mesenchymal transition; FMT, fibroblast-to-myofibroblast transdifferentiation; HMB, heavy menstrual bleeding; MMT, mesothelial–mesenchymal transition; PR, progesterone receptor; SMM, smooth muscle metaplasia.

Citation: Reproduction 164, 5; 10.1530/REP-22-0224

Redefine adenomyosis

Despite the first description dating back over 160 years ago, the definition of adenomyosis is still the one made by Bird et al. half a century ago, that is, ‘the benign invasion of endometrium into the myometrium, producing a diffusely enlarged uterus which microscopically exhibits ectopic, nonneoplastic, endometrial glands, and stroma surrounded by hypertrophic-hyperplastic musculature’ (Bird et al. 1972). Obviously, this definition was made before the advent of imaging techniques and was based entirely on post-hysterectomy samples. With the increasing popularity of imaging diagnostics, the increased thickness (>12 mm) of the JZ progressively became pathognomonic of adenomyosis and, for a number of years, represented a sort of a golden standard for non-invasive identification of the disease (see, for example, Novellas et al. 2011).

However, using the JZ thickness in defining adenomyosis can be fraught with many problems. First, the JZ is defined on MRI and is not really an anatomically definable area. Secondly, in about 20% of premenopausal women, the JZ is simply undefinable on MRI (Harada et al. 2016). As such, the importance of JZ thickness has been downplayed and a ‘sole criterion’-based diagnosis has been called into question, with a broader approach being proposed (Bazot & Darai 2018). Finally, how the JZ thickness relates to adenomyosis itself is poorly understood.

Moreover, as the natural history of adenomyotic lesions is unveiled, perhaps we could define adenomyosis as a condition that started with the deposition of endometrial stroma and epithelium within the myometrium, which undergoes cyclic bleeding and thus repeated tissue injury and repair, resulting in gradual and progressive smooth muscle metaplasia and fibrogenesis (Guo 2018b ). One advantage of this definition is that it seems to capture the essence of the natural history of the disease and highlights the dynamic and progressive nature of adenomyotic lesions.

Heavy menstrual bleeding

Per the FIGO AUB classification ‘PALM-COEIN’, adenomyosis is known to be a structural cause of AUB, which includes the symptom of HMB (Munro et al. 2011). HMB is defined as excessive menstrual blood loss (MBL) that disrupts physical, social, emotional, and/or material quality of life ((NICE) 2018). HMB has been objectively defined as MBL greater than 80 mL (Hallberg et al. 1966, Higham & Shaw 1999). Unfortunately, the mechanisms underlying adenomyosis-induced HMB are poorly understood (Critchley et al. 2020).

In the absence of pregnancy, endometrial cyclooxygenase-2 (COX-2) is upregulated following the progesterone withdrawal in the late secretory phase, resulting in subsequent increased levels of prostaglandins (PGs), namely, PGE2 and PGF2 α. This signifies a pivotal event for subsequent menstrual induction (Critchley et al. 1999, Sugino et al. 2004). Increased endometrial PGE2 signaling, coupled with endometrial generation of PGF2 α, a vasoconstrictor, leads to local hypoxia in the upper functional layer of the endometrium and the activation of hypoxia-inducible factor 1α (HIF-1α), the master regulator of cellular hypoxia. This defining event establishes an endometrial microenvironment that is conducive to tissue repair and angiogenesis (Critchley et al. 2006, Maybin et al. 2011, 2018). It is well documented that the disruption of the hypoxia signaling or the PGE2 signaling in endometrium would impair endometrial repair, causing HMB (Maybin et al. 2018, Critchley et al. 2020).

In many fibrotic diseases, PGE2 is known to be anti-fibrotic (Huang et al. 2007, 2009, Penke et al. 2014, Wettlaufer et al. 2016). With the progression of adenomyosis and fibrogenesis (Liu et al. 2016b , Shen et al. 2016, Hao et al. 2020, Yang et al. 2021), the lesional stiffness or rigidity has been observed to increase (Liu et al. 2018a ), which would result in the downregulation of COX-2 expression and consequent decrease in PGE2 production and signaling (Liu et al. 2010a ). Indeed, it has been shown recently that endometriotic stromal cells cultured in stiffer substrates demonstrate the downregulation of COX-2 and E-series receptor type 2 (EP2) and EP4 (Huang et al. 2022b ). Since adenomyotic and endometriotic lesions, as ectopic endometrium, share the same and defining hallmark of cyclic bleeding (Brosens 1997), they also share similarities but are not necessarily identical due to different microenvironments and molecular processes that underpin lesional progression (Guo 2018b ). Consequently, this raises the possibility that, as adenomyotic lesions become progressively fibrotic and thus increasingly stiffened, the behavior of neighboring eutopic endometrial cells may likely change.

Given the natural history of adenomyotic lesions as outlined above and in light of the documented mechanisms underlying HMB as well as the disrupted PGE2 signaling in fibrosis, it is conceivable that women with adenomyosis who complain of HMB may have a greater extent of lesional fibrosis and thus greater stiffness, as well as reduced PGE2 signaling. This can be manifested by reduced immunostaining of HIF-1α, COX-2, EP2, and EP4 in adenomyotic lesions and their neighboring JZ and eutopic endometrium. This may lead to attenuation of local PG signaling with reduced expression of COX-2, EP2, and EP4 and thus reduced PGE2, which, in turn, may result in suppression of HIF-1α and subsequent impaired endometrial repair and, finally, HMB.

Indeed, it has been recently demonstrated that, in women with adenomyosis who complained of excessive MBL, the lesional stiffness, as measured by transvaginal ultrasonic elastography, is significantly higher than in those who complained of moderate–heavy MBL (Huang et al. 2022a). Consistent with the increased lesional stiffness, the extent of lesional fibrosis and the extent of tissue fibrosis in neighboring EMI and eutopic endometrium are also significantly higher. In addition, in adenomyotic lesions and their neighboring EMI and eutopic endometrial tissues, the expression of HIF-1α, COX-2, EP2, and EP4 is significantly reduced, concomitant with increased tissue fibrosis (Huang et al. 2022a ).

These findings strongly indicate reduced hypoxia and reduced PGE2 production and signaling in adenomyosis lesions, as well as in their neighboring EMI and endometrial tissues, as lesions become highly fibrotic. This reduced hypoxia and PGE2 signaling appears to be first initiated presumably in adenomyosis lesions and then propagates to their neighboring EMI and then eutopic endometrium. Given the recent report that adenomyosis lesions are directly connected with their neighboring endometrial glands (Yamaguchi et al. 2021), this propagation could conceivably be conducted through cellular communications in a paracrine or autocrine manner. There is an apparent gradient in the extent of tissue fibrosis, with the adenomyosis lesion having the highest fibrotic content, followed by EMI and then endometrium, suggesting that the progression of adenomyotic lesions impacted the same signaling pathways in both EMI and eutopic endometrium (Huang et al. 2022a). In light of the well-documented role of PGE2 and hypoxia signaling pathways in endometrial repair and HMB (Critchley et al. 2006, Maybin et al. 2011, 2018), these data strongly suggest that the increased lesional fibrosis may contribute to disturbed PGE2 signaling in eutopic endometrium and thus possibly reduced hypoxia necessary for endometrial repair, leading to HMB in women with adenomyosis.

Recent investigations further reveal that glycolysis plays an important role in endometrial repair, and decreased glycolysis at menstruation impairs endometrial repair, causing HMB (Mao et al. 2022). This is not surprising, considering that a complete endometrial repair would require, by necessity, increased (but transitory) inflammation, followed by cellular proliferation and angiogenesis (Critchley et al. 2020) – all of which require obligatorily more nutrients and energy expenditure (Vinaik et al. 2020), with the goal of a full repair to prepare for endometrial differentiation and decidualization that set the stage for possible pregnancy. Somehow, endometrium would modulate their cellular bioenergetics in response to increased demands and shift to glycolysis-based metabolism. Glycolysis has several distinct advantages over more traditional metabolism through oxidative phosphorylation (OXPHOS): first, it produces the energy currency ATP faster in a given time span despite lower efficiency in one cycle, as well as macromolecules necessary for fast dividing cells and, simultaneously, reduced oxidative stress. The anabolic pathways branching out from the glycolytic sequence produce the intermediate products necessary for cellular proliferation, including amino acids and lipid precursors (Levine & Puzio-Kuter 2010, Cairns et al. 2011). Consequently, fast proliferating endometrial cells can increase the availability of macromolecules for growth needs through glycolysis by facilitating glucose uptake and slowing the entry of pyruvate into mitochondria (Vander Heiden et al. 2009). Secondly, glycolysis can offer much needed protection against oxidative stress (Cairns et al. 2011) by reducing reactive oxygen species (ROS) – common byproducts of OXPHOS and through re-channeling glycolytic intermediates into the pentose phosphate pathway (PPP) that generates essential nucleotide precursors and at the same time reduces nicotinamide adenine dinucleotide phosphate hydride that is necessary for the activity of antioxidant enzymes. Excessive ROS and/or impaired ROS detoxification cause oxidative damage and are thus detrimental to cells and thus to tissue repair (Cano Sanchez et al. 2018) and conceivably can compromise or impair normal endometrial repair.

Since HIF-1α can regulate its target genes involved in glycolysis, including genes encoding for key enzymes in the glycolysis such as glucose transporter 1 (GLUT1), pyruvate kinase isoform M2 (PKM2), lactate dehydrogenase A (LDHA), hexokinase 2 (HK2), and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) (Semenza 2013, Xie et al. 2019, Zhang et al. 2019, Wu et al. 2020), the reduced hypoxia signaling in endometrium resulting from fibrotic adenomyotic lesions (Huang et al. 2022a) conceivably can impair glycolysis and, as such, impair endometrial repair and cause adenomyosis-induced HMB (Mao et al. 2022).

The possible mechanisms underlying adenomyosis-associated HMB are depicted in Fig. 3. Note that since tissue factor is overexpressed in adenomyosis (Liu et al. 2011), patients may also exhibit hypercoagulability as in endometriosis (Wu et al. 2015, Ding et al. 2018), causing HMB.

Figure 3
Figure 3

Diagram depicting possible mechanisms of adenomyosis-associated heavy menstrual bleeding (HMB). Increasing lesional fibrosis would propagate to neighboring endometrial–myometrial interface and then eutopic endometrium, attenuating the PGE2 and hypoxia signaling by downregulation of COX-2, EP2, 4, and HIF-1α. As a result, glycolysis, which is vital for normal endometrial repair, is impaired, resulting in reduced supply of energy and macromolecules and reduced inflammation necessary for endometrial repair. Collectively, reduced PGE2 and hypoxia signaling, in conjunction with diminished supply of energy and macromolecules and attenuated inflammation, results in impaired endometrial repair and, as such, HMB. The upward arrows indicate upregulation/overexpression or increase. The directional arrows mean ‘lead to’ or ‘result in’.

Citation: Reproduction 164, 5; 10.1530/REP-22-0224

These findings provide a good explanation as to why different subtypes of adenomyosis have different symptoms. Indeed, one early study reported that shallower penetration of adenomyosis lesions into the myometrium was associated with higher incidence of HMB (Bird et al. 1972). This is because adenomyosis lesions that are closer to the endometrium are more likely than those more distant ones to cause HMB as lesions progress, simply because of the ease of propagation due to physical proximity. They also explain why internal adenomyosis is more likely to be associated with HMB while external adenomyosis is more often associated with deep endometriosis (and thus pain) (Bourdon et al. 2021).

It is also possible that HMB could further exacerbate endometrial repair impairment as bleeding may cause local iron overload, which may attenuate the proliferation of endometrial stromal cells by inducing autophagy (Zhou et al. 2022) and thus exacerbating impaired endometrial repair. Of course, whether this is true would await further investigation.

Pain

Among all complaints from women with adenomyosis, dysmenorrhea and other types of pain top the list of complaints and are the most debilitating that substantially and significantly reduced the quality of life in affected women. According to the recently revised definition by the International Association for the Study of Pain, pain is ‘an unpleasant sensory and emotional experience associated with, or resembling that associated with, actual or potential tissue damage’ (Raja et al. 2020). As such, pain is always a personal experience, influenced, to varying degrees, by not only biological but also a myriad of psychological and social factors (Vader et al. 2021). In addition, pain, which is always subjective, and nociception, which could be quantitated, are different phenomena, and the former cannot be inferred exclusively from activity in sensory neurons (Vader et al. 2021).

With this understanding, it is then easy to understand why there is no direct correlation between the disease stage and severity of pelvic symptoms in women with endometriosis (Vercellini et al. 2007), and likely in women with adenomyosis as well. Indeed, with a kaleidoscopic variation in subtype (internal, external, intramural, or unclassified, or focal vs diffuse), size, depth of infiltration, presence or absence of adhesion, and co-occurrence with or without endometriosis, uterine fibroids or other co-morbidity, on top of age, phase of menstrual cycle, gravidity, parity, hormonal status, and previous history of uterine surgery, along with different developmental trajectories including, but not limited to, in utero and early childhood development that may shape one’s neuronal wiring and pain threshold, variation in symptomology and severity is something to be expected. This is especially true that adenomyosis-associated pain may be accompanied by histological changes in the CNS, as shown in endometriosis (As-Sanie et al. 2012, 2016).

However, since adenomyosis is known to be associated with dysmenorrhea and other types of pain, it must have certain attributes that induce these pain behaviors. In addition, the disease stage or lesion type may not fully capture the essence of an adenomyotic lesion. For example, if we quantitate the extent of lesional fibrosis, then we can find its correlation with the severity of dysmenorrhea (Nie et al. 2022), albeit imperfect correlation.

There are several attributes that are known to contribute to dysmenorrhea and other types of pain in women with adenomyosis. First, the myometrium of women with adenomyosis exhibits elevated expression of OTR, which can result in increased frequency and magnitude of uterine contractions (Nie et al. 2010, Guo et al. 2013). As eluded to above, the newly formed SMCs, differentiated from FMT and SMM, would be fused into existing myometrium, and this is likely to cause irregular contractions (Guo et al. 2013), possibly akin to spams. Of course, adenomyotic lesions may also secrete certain molecules that facilitate the proliferation of myometrial SMCs (Huang et al. 2021a,b, Wang et al. 2021c). Irrespective of sources, there is a net increase in myometrial SMCs, leading to uterine enlargement. This, coupled with hyperinnervation in endometrium and myometrium (Wang et al. 2009a,b, Zhang et al. 2010, Choi et al. 2015) as well as reduced suppression of the transfer of nociceptive signals to the CNS in adenomyosis (Chen et al. 2014), would easily entail patients to perceive dysmenorrhea.

Secondly, adenomyotic lesions may secrete neurotrophic factors that promote hyperinnervation. These factors include neurotrophins such as NGF and its receptor TrkA, brain-derived neurotrophic factor (BDNF) and its receptor TrkB, neurotrophin-3 (NT-3) which binds to TrkC, and neurotrophin-4/5 (NT-4) which also binds to TrkB. All these neurotrophins can bind to p75. These factors support and direct neuritogenesis.

Mouse models of adenomyosis show elevated expression of NGF and its high-affinity receptor p75 and low-affinity receptor TrkA in lesions, eutopic endometrium, and myometrium (Green et al. 2003, Li et al. 2011, Barcena de Arellano et al. 2012, Carrarelli et al. 2017). In particular, adenomyotic stromal cells secrete NGF (Li et al. 2015). TrkA expression is also found to be elevated in dorsal root ganglia from mice with induced adenomyosis (Li et al. 2011). However, there is also a study reporting no difference in the expression of NGF, NT-3, and p75 in eutopic endometrium and myometrium (Barcena de Arellano et al. 2012).

Ectopic endometrial stromal cells are known to secrete thromboxane A2 (TXA2) that induces neurite outgrowth (Yan et al. 2017a). In patients with adenomyosis who were treated with levonorgestrel-releasing intrauterine system (LNG-IUS), the expression of NGF and its cognate receptors p75 and TrkA in endometrium and myometrium was significantly decreased as compared to those of hormonally untreated patients (Choi et al. 2010).

Lastly, it is likely that women with adenomyosis may exhibit central sensitization as in those with endometriosis (He et al. 2010). Thus, once there is central sensitization, any stimulus, be it noxious or innocuous, would easily elicit the perception of pain.

Overall, however, our understanding on the mechanisms underlying adenomyosis-associated pain is still fragmentary. For example, transient receptor potential vanilloid type 1 (TRPV1), a pain signal integrator, is reported to be highly expressed in adenomyotic lesions (Nie et al. 2010). But what kind of role TRPV1 plays in adenomyosis-associated pain is largely unknown since TRPV1 is typically expressed in primary nociceptive nerves such as Aδ- and C-fibers, serving as a sensor of noxious stimuli. In addition, whether nerves and adenomyotic lesions form a partnership in facilitating lesional progression, as in endometriosis (Yan et al. 2017b), has not been fully elucidated. We recently reported that certain neuropeptide receptors, such as NK1R (receptor for substance P), receptor activity modifying protein 1 (RAMP-1) and calcitonin receptor-like receptor (CRLR), the two receptors for CGRP, and neurotransmitter receptor ADRB2 are all elevated in adenomyotic lesions (Xu et al. 2021). In contrast, the expression of α7 nicotinic acetylcholine receptor (α7nAChR) is reduced (Xu et al. 2021). Given the report that women with endometriosis have reduced vagal tone (Hao et al. 2021) and α7nAChR activation appears to be therapeutic (Hao et al. 2022), it seems that a great deal of work is still needed to unravel the mystery of adenomyosis-associated pain.

Clinical significance

Imaging diagnosis

In the last three decades, MRI and ultrasound have gradually become the mainstay for the diagnosis of adenomyosis, completely replacing histological evaluation following hysterectomy as the main diagnostic tool. In particular, overall, transvaginal ultrasound (TVUS) and MRI have similar diagnostic sensitivity and specificity (Chapron et al. 2020), although the latter is less operator-dependent, more subjective, and relies less on the capacity of the observer to diagnose. However, TVUS remains to be the first-line technique in gynecological work-up because it is widely available, easier than MRI to operate but less expensive than MRI, and also allows a dynamic examination to explore organ mobility and site-specific tenderness.

Thus, both TVUS and MRI are excellent imaging modalities for diagnosing adenomyosis. However, there is still ample room for improvement in terms of diagnostic accuracy. In addition, neither of the two imaging modalities produces features that directly correlate with the severity of symptoms and the response to hormonal treatment or prognosis (Chapron et al. 2020, Habiba et al. 2020) and neither of them is helpful in predicting the response to hormonal treatment. Moreover, no universally accepted classification system exists (Gordts et al. 2008, 2018, Habiba et al. 2020).

As alluded to above, fibrosis is one important pathognomonic feature of adenomyosis, as in endometriosis (Guo 2018b, Vigano et al. 2018). Incidentally, uterine fibroids also have excessive ECM deposition (Stewart 2015). The extent of fibrosis in either adenomyosis or fibroids can conceivably determine the lesional stiffness or rigidity, which could be evaluated by palpation through tissue deformation. Hence, lesional stiffness contains information inherently embedded within adenomyotic lesions, revealing just how advanced the lesion is. Unfortunately, neither TVUS nor MRI can directly measure the lesional stiffness.

Elastography is an emergent imaging technology that recently becomes commercially available. It generates images of tissue stiffness, by ultrasound elastography (UE) (Cui et al. 2022) or magnetic resonance elastography (MRE) (Mariappan et al. 2010). Thus, it is akin to the traditional palpation in clinical examination but it is less subjective, requires little experience, and provides better spatial localization information (Shiina et al. 2015). As of now, the application of MRE in gynecology has been scanty, but UE has been gaining tracks in gynecology. Perhaps, the greatest advantage of elastography is that it has a much wider dynamic range than CT, ultrasound, and MRI (Mariappan et al. 2010).

UE has been shown to be used to diagnose adenomyosis (Liu et al. 2018a) and deep endometriosis (Ding et al. 2020). Remarkably, the average lesional stiffness in adenomyosis is significantly higher than that of uterine fibroids, which, in turn, is higher than normal myometrium (Liu et al. 2018a). More importantly, lesional stiffness as measured by UE correlated positively with the extent of lesional fibrosis and negatively with lesional staining levels of PR and vascularity (Liu et al. 2018a, Ding et al. 2020). Thus, once lesional stiffness is quantitated by UE, the possible lesional response to hormonal treatment can be determined, since lower PR expression is associated with poor response to progesterone treatment (Flores et al. 2018). Preliminary data show that women with ‘soft’ lesions are more likely to respond to dienogest treatment (Ding et al. unpublished data). Therefore, UE can not only improve diagnostic accuracy but, more importantly, help physicians to decide on the best treatment modality.

Development of novel therapeutics

While hysterectomy is a definitive treatment for adenomyosis, whether or not to undergo this procedure can be an agonizing and difficult decision to make for women who wish to preserve their fertility or hold the view that uterus is an iconic symbol of womanhood. Total hysterectomy is also reported to increase the risk of cardiovascular disease, depression, and dementia (Madueke-Laveaux et al. 2021). Traditionally, progestins, oral contraceptives, LNG-IUS, and GnRH agonists, along with uterine artery embolization (UAE) and conservative surgery involving endomyometrial ablation, laparoscopic myometrial electrocoagulation or excision were the mainstay therapeutics (Wood 2001). Fortunately, the arsenals for combating adenomyosis have been expanded greatly during the last decade. There are several excellent reviews on this topic already (Vannuccini et al. 2018, Cope et al. 2020, Stratopoulou et al. 2021a).

While hormonal drugs are the mainstay of current therapeutics, a recent survey of 1420 patients with endometriosis in Austria, Germany, and Switzerland found that an overwhelming proportion of them hold a negative attitude toward hormonal drugs, and the majority of them disliked the side effects of these drugs, especially amongst young, city-dwelling, educated women (Burla et al. 2021). Remarkably, 95.3% of the surveyees expressed preference for plant-based products, preferably in oral form (Burla et al. 2021). This demand contrasts sharply with resounding failure in clinical trials on non-hormonal drugs in endometriosis and adenomyosis (Guo & Evers 2013, Guo 2014) and highlights the medical need that has not been fulfilled.

As of writing, 83 clinical trials on adenomyosis have been registered at ClinicalTrials.gov (accessed on June 23, 2022). With only two exceptions (NCT01821001 and NCT03749109), all trial drugs are hormonal. Trial NCT01821001 is on the use of vaginal ring containing bromocriptine, a DRD2 agonist. A pilot study found that this therapy is effective in improving HMB and dysmenorrhea in women with adenomyosis (Andersson et al. 2019). A recent study on 18 patients with diffuse adenomyosis who complained of HMB also reports that treatment with vaginal bromocriptine for 6 months resulted in a significant decrease in JZmax and a reduced number of women with asymmetric myometrial wall thickness as evaluated by ultrasound (Andersson et al. 2020). Another trial (NCT03749109) on the use of vaginal ring containing Quinagolide, also a DRD2 agonist, to treat adenomyosis is recently completed (www.ClinicalTrials.gov, accessed on September 9, 2022) but not yet published as of writing.

While the mechanism of action for bromocriptine has been thought to suppress prolactin (Cope et al. 2020), there could be some other possible mechanisms. Chronic stress can activate the HPA/SAM axes, causing the release of adrenaline and noradrenaline, which, in turn, can activate the expression of ADRB2 and suppress DRD2 expression in endometriosis (Guo et al. 2017), and possibly in adenomyosis as well. Dopamine or DRD2 agonists could block the stress-facilitated angiogenesis and proliferation of adenomyotic cells, as in cancer (Moreno-Smith et al. 2011, 2013). Alternatively, DRD2 agonists may be anti-fibrotic (Jiang et al. 2014, Han et al. 2015) and thus can stall lesional fibrogenesis. Indeed, DRD2 can act as a negative regulator of inflammation (Zhang et al. 2012, Jiang et al. 2014). Incidentally, increased local prolactin production is also seen in uterine fibroids, which may stimulate transdifferentiation of myometrial cells to myofibroblasts, which, in turn, promotes fibrogenesis in fibroids (DiMauro et al. 2022). Taken together, the mechanisms of action for DRD2 agonists seem to be in need of further elucidation.

Given the role of platelets in the development of adenomyosis (Liu et al. 2016b, Shen et al. 2016, Zhang et al. 2016), anti-platelet therapy has been proposed and shown to be promising in mouse with induced adenomyosis (Zhu et al. 2016). In addition, since andrographolide is known to be anti-platelet (Lu et al. 2011), it has been proposed as a potential therapeutics (Li et al. 2013), and its clinical use appears to be very encouraging (Liu et al. 2015). Moreover, many compounds that are extracted from traditional Chinese medicine herbs, such as resveratrol, scutellarin, tanshinone IIA, quercetin, leonurine, and tetramethylpyrazine, all share the commonality of being anti-platelet or anti-thrombotic, and they have demonstrated promising potential in mouse models of endometriosis and adenomyosis (Zhu et al. 2015, Nie & Liu 2017a,b, Nie et al. 2018, Ding et al. 2019, Luo et al. 2020, Huang et al. 2022c).

Similar to endometriosis, adenomyosis has many epigenetic aberrations (Liu & Guo 2012, Liu et al. 2012). Based on the desirable effect of histone deacetylase inhibitors in ectopic endometrium (Wu & Guo 2006, 2007, Wu et al. 2007, Liu & Guo 2011, Mao et al. 2011), a pilot clinical study reported on the use of valproic acid (VPA) to treat adenomyosis (Liu & Guo 2008). An expanded case series also provided encouraging results (Liu et al. 2010b). Unfortunately, since clinical trials typically demand a great deal of resources, further research in this direction has not been continued. Incidentally or not, VPA also possesses anti-platelet and anti-fibrotic capabilities (Larsson et al. 2016, Costalonga et al. 2017).

In endometriosis area, many drug trials fail simply because of safety concerns (e.g. ulipristal and AKR1C3 inhibitor) (Guo 2014, Saunders & Horne 2021). Yet, many anti-platelet compounds extracted from herbs, such as tanshinone IIA, quercetin, and tetramethylpyrazine, are known to have excellent safety profiles. For example, both tanshinone IIA and tetramethylpyrazine are officially listed in the Chinese Pharmacopoeia for the treatment of menstrual disorders and blood circulation diseases and prevention of inflammation (Kuang et al. 2005). It may be desirable to select those compounds that are, first, known to be safe and target known molecular pathways in the development of adenomyosis.

Conclusions

Adenomyosis has been traditionally viewed as an enigmatic disease and, as such, defying effective management. Fortunately, research published in the last decade has helped us to unveil some pathogenesis and pathophysiology of the disease. We now know that EMID can increase the risk of adenomyosis, and Schwann cells within the EMI may play an important role. In addition, the pathogenesis of adenomyosis is surely multifactorial, and EMID is just one such cause.

We also have a fairly good grip on the natural history of adenomyotic lesions by now through the identification of several underlying key molecular processes such as EMT, FMT, SMM, and fibrogenesis. This knowledge also led to our better understanding of adenomyosis-associated HMB. Along the way, we have learned several important lessons from this investigation: (1) lesional progress is a dynamic process; (2) cell identity within lesions is not immutable; and (3) lesional microenvironment is also important for lesional progression – many cells within the microenvironment also participate in lesional progression. Of course, there are still many t’s that need to be crossed and many i’s need to be dotted. Nonetheless, this skeletal knowledge has helped us to piece together many observations that are seemingly unrelated and to capitalize on emerging imaging technology such as elastography for better diagnosis of the disease. The addition of elastographic information on adenomyosis and on co-existent pathology should help to enhance the future classification system for adenomyosis. The development of novel therapeutics would take time, but the grip of the natural history of ectopic endometrium has already helped us understand why clinical trials on endometriosis/adenomyosis fail (Guo & Groothuis 2018).

Of course, there is still a long way to go before the day when a precision medicine approach can effectively alleviate adenomyosis-associated symptoms and when a safe preoperative or perioperative intervention can be instituted to mitigate or even eliminate the risk of adenomyosis. Until then, effects on molecular, genetic/epigenetic, clinical, and epidemiologic investigation on adenomyosis need to be doubled down.

Declaration of interest

S W G provided consultancy advice for MSD R&D, Chugai Pharmaceutical Co., and BioHaven Pharmaceuticals, but these activities had no bearing on this work.

Funding

This study was funded in part by grant 82071623 from the National Natural Science Foundation of China, an Excellence in Centers of Clinical Medicine grant (2017ZZ01016) from the Science and Technology Commission of Shanghai Municipality, and Clinical Research Plan grant SHDC2020CR2062B from Shanghai Shenkang Center for Hospital Development.

References

  • Adameyko I, Lallemend F, Aquino JB, Pereira JA, Topilko P, Muller T, Fritz N, Beljajeva A, Mochii M & Liste I et al.2009 Schwann cell precursors from nerve innervation are a cellular origin of melanocytes in skin. Cell 139 366379. (https://doi.org/10.1016/j.cell.2009.07.049)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Alm P & Lundberg LM 1988 Co-existence and origin of peptidergic and adrenergic nerves in the guinea pig uterus. Retrograde tracing and immunocytochemistry, effects of chemical sympathectomy, capsaicin treatment and pregnancy. Cell and Tissue Research 254 517530. (https://doi.org/10.1007/BF00226501)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Alvarez-Suarez P, Gawor M & Proszynski TJ 2020 Perisynaptic Schwann cells – the multitasking cells at the developing neuromuscular junctions. Seminars in Cell and Developmental Biology 104 3138. (https://doi.org/10.1016/j.semcdb.2020.02.011)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • An M, Li D, Yuan M, Li Q, Zhang L & Wang G 2017a Different macrophages equally induce EMT in endometria of adenomyosis and normal. Reproduction 154 7992. (https://doi.org/10.1530/REP-17-0174)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • An M, Li D, Yuan M, Li Q, Zhang L & Wang G 2017b Interaction of macrophages and endometrial cells induces epithelial-mesenchymal transition-like processes in adenomyosis. Biology of Reproduction 96 4657. (https://doi.org/10.1095/biolreprod.116.144071)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Andersson JK, Khan Z, Weaver AL, Vaughan LE, Gemzell-Danielsson K & Stewart EA 2019 Vaginal bromocriptine improves pain, menstrual bleeding and quality of life in women with adenomyosis: a pilot study. Acta Obstetricia et Gynecologica Scandinavica 98 13411350. (https://doi.org/10.1111/aogs.13632)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Andersson JK, Pozzi Mucelli R, Epstein E, Stewart EA & Gemzell-Danielsson K 2020 Vaginal bromocriptine for treatment of adenomyosis: impact on magnetic resonance imaging and transvaginal ultrasound. European Journal of Obstetrics, Gynecology, and Reproductive Biology 254 3843. (https://doi.org/10.1016/j.ejogrb.2020.08.040)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Anglesio MS, Papadopoulos N, Ayhan A, Nazeran TM, Noe M, Horlings HM, Lum A, Jones S, Senz J & Seckin T et al.2017 Cancer-associated mutations in endometriosis without cancer. New England Journal of Medicine 376 18351848. (https://doi.org/10.1056/NEJMoa1614814)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Asally R, Markham R & Manconi F 2019 The expression and cellular localisation of neurotrophin and neural guidance molecules in peritoneal ectopic lesions. Molecular Neurobiology 56 40134022. (https://doi.org/10.1007/s12035-018-1348-6)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • As-Sanie S, Harris RE, Napadow V, Kim J, Neshewat G, Kairys A, Williams D, Clauw DJ & Schmidt-Wilcke T 2012 Changes in regional gray matter volume in women with chronic pelvic pain: a voxel-based morphometry study. Pain 153 10061014. (https://doi.org/10.1016/j.pain.2012.01.032)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • As-Sanie S, Kim J, Schmidt-Wilcke T, Sundgren PC, Clauw DJ, Napadow V & Harris RE 2016 Functional connectivity is associated with altered brain chemistry in women with endometriosis-associated chronic pelvic pain. Journal of Pain 17 113. (https://doi.org/10.1016/j.jpain.2015.09.008)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bacci M, Capobianco A, Monno A, Cottone L, Di Puppo F, Camisa B, Mariani M, Brignole C, Ponzoni M & Ferrari S et al.2009 Macrophages are alternatively activated in patients with endometriosis and required for growth and vascularization of lesions in a mouse model of disease. American Journal of Pathology 175 547556. (https://doi.org/10.2353/ajpath.2009.081011)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bae SE, Corcoran BM & Watson ED 2001 Immunohistochemical study of the distribution of adrenergic and peptidergic innervation in the equine uterus and the cervix. Reproduction 122 275282. (https://doi.org/10.1530/rep.0.1220275)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Baratelli F, Lin Y, Zhu L, Yang SC, Heuze-Vourc'h N, Zeng G, Reckamp K, Dohadwala M, Sharma S & Dubinett SM 2005 Prostaglandin E2 induces FOXP3 gene expression and T regulatory cell function in human CD4+ T cells. Journal of Immunology 175 14831490. (https://doi.org/10.4049/jimmunol.175.3.1483)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barcena de Arellano ML, Arnold J, Vercellino F, Chiantera V, Schneider A & Mechsner S 2011 Overexpression of nerve growth factor in peritoneal fluid from women with endometriosis may promote neurite outgrowth in endometriotic lesions. Fertility and Sterility 95 11231126. (https://doi.org/10.1016/j.fertnstert.2010.10.023)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barcena de Arellano ML, Wagner MF, Oldeweme J, Arnold J, Ebert A, Schneider A & Mechsner S 2012 Neurotrophin expression is not affected in uteri of women with adenomyosis. Journal of Molecular Neuroscience 47 495504. (https://doi.org/10.1007/s12031-012-9757-x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barcena de Arellano ML, Arnold J, Lang H, Vercellino GF, Chiantera V, Schneider A & Mechsner S 2013a Evidence of neurotrophic events due to peritoneal endometriotic lesions. Cytokine 62 253261. (https://doi.org/10.1016/j.cyto.2013.03.003)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barcena de Arellano ML, Oldeweme J, Arnold J, Schneider A & Mechsner S 2013b Remodeling of estrogen-dependent sympathetic nerve fibers seems to be disturbed in adenomyosis. Fertility and Sterility 100 801809. (https://doi.org/10.1016/j.fertnstert.2013.05.013)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bazot M & Darai E 2018 Role of transvaginal sonography and magnetic resonance imaging in the diagnosis of uterine adenomyosis. Fertility and Sterility 109 389397. (https://doi.org/10.1016/j.fertnstert.2018.01.024)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Benagiano G & Brosens I 2006 History of adenomyosis. Best Practice and Research: Clinical Obstetrics and Gynaecology 20 449463. (https://doi.org/10.1016/j.bpobgyn.2006.01.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Benagiano G, Habiba M & Brosens I 2012 The pathophysiology of uterine adenomyosis: an update. Fertility and Sterility 98 572579. (https://doi.org/10.1016/j.fertnstert.2012.06.044)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ben-Eliyahu S, Shakhar G, Page GG, Stefanski V & Shakhar K 2000 Suppression of NK cell activity and of resistance to metastasis by stress: a role for adrenal catecholamines and beta-adrenoceptors. Neuroimmunomodulation 8 154164. (https://doi.org/10.1159/000054276)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Benito C, Davis CM, Gomez-Sanchez JA, Turmaine M, Meijer D, Poli V, Mirsky R & Jessen KR 2017 STAT3 controls the long-term survival and phenotype of repair Schwann cells during nerve regeneration. Journal of Neuroscience 37 42554269. (https://doi.org/10.1523/JNEUROSCI.3481-16.2017)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bird CC, McElin TW & Manalo-Estrella P 1972 The elusive adenomyosis of the uterus – revisited. American Journal of Obstetrics and Gynecology 112 583593. (https://doi.org/10.1016/0002-9378(7290781-8)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Blom CL, Martensson LB & Dahlin LB 2014 Nerve injury-induced c-Jun activation in Schwann cells is JNK independent. BioMed Research International 2014 392971. (https://doi.org/10.1155/2014/392971)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bourdon M, Oliveira J, Marcellin L, Santulli P, Bordonne C, Maitrot Mantelet L, Millischer AE, Plu Bureau G & Chapron C 2021 Adenomyosis of the inner and outer myometrium are associated with different clinical profiles. Human Reproduction 36 349357. (https://doi.org/10.1093/humrep/deaa307)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brosens IA 1997 Endometriosis – a disease because it is characterized by bleeding. American Journal of Obstetrics and Gynecology 176 263267. (https://doi.org/10.1016/s0002-9378(9770482-4)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brosens I, Derwig I, Brosens J, Fusi L, Benagiano G & Pijnenborg R 2010 The enigmatic uterine junctional zone: the missing link between reproductive disorders and major obstetrical disorders? Human Reproduction 25 569574. (https://doi.org/10.1093/humrep/dep474)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bulun SE, Yilmaz BD, Sison C, Miyazaki K, Bernardi L, Liu S, Kohlmeier A, Yin P, Milad M & Wei J 2019 Endometriosis. Endocrine Reviews 40 10481079. (https://doi.org/10.1210/er.2018-00242)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bulun SE, Yildiz S, Adli M & Wei JJ 2021 Adenomyosis pathogenesis: insights from next-generation sequencing. Human Reproduction Update 27 10861097. (https://doi.org/10.1093/humupd/dmab017)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Burla L, Kalaitzopoulos DR, Metzler JM, Scheiner D & Imesch P 2021 Popularity of endocrine endometriosis drugs and limited alternatives in the present and foreseeable future: a survey among 1420 affected women. European Journal of Obstetrics, Gynecology, and Reproductive Biology 262 232238. (https://doi.org/10.1016/j.ejogrb.2021.05.040)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cai X, Shen M, Liu X & Nie J 2019 The possible role of eukaryotic translation initiation factor 3 subunit e (eIF3e) in the epithelial-mesenchymal transition in adenomyosis. Reproductive Sciences 26 377385. (https://doi.org/10.1177/1933719118773490)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cairns RA, Harris IS & Mak TW 2011 Regulation of cancer cell metabolism. Nature Reviews: Cancer 11 8595. (https://doi.org/10.1038/nrc2981)

  • Cano Sanchez M, Lancel S, Boulanger E & Neviere R 2018 Targeting oxidative stress and mitochondrial dysfunction in the treatment of impaired wound healing: a systematic review. Antioxidants 7 98. (https://doi.org/10.3390/antiox7080098)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cao Y, Wang X, Liu X, Harada T & Guo S-W 2022 Neonatal feeding of an estrogen receptor β (ERβ) agonist induces external adenomyosis-like lesions in ICR mouse. Reproductive and Developmental Medicine In press. (https://doi.org/10.1097/RD9.0000000000000012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Carr MJ & Johnston AP 2017 Schwann cells as drivers of tissue repair and regeneration. Current Opinion in Neurobiology 47 5257. (https://doi.org/10.1016/j.conb.2017.09.003)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Carrarelli P, Yen CF, Funghi L, Arcuri F, Tosti C, Bifulco G, Luddi A, Lee CL & Petraglia F 2017 Expression of inflammatory and neurogenic mediators in adenomyosis. Reproductive Sciences 24 369375. (https://doi.org/10.1177/1933719116657192)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chapron C, Tosti C, Marcellin L, Bourdon M, Lafay-Pillet MC, Millischer AE, Streuli I, Borghese B, Petraglia F & Santulli P 2017 Relationship between the magnetic resonance imaging appearance of adenomyosis and endometriosis phenotypes. Human Reproduction 32 13931401. (https://doi.org/10.1093/humrep/dex088)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chapron C, Vannuccini S, Santulli P, Abrao MS, Carmona F, Fraser IS, Gordts S, Guo SW, Just PA & Noel JC et al.2020 Diagnosing adenomyosis: an integrated clinical and imaging approach. Human Reproduction Update 26 392411. (https://doi.org/10.1093/humupd/dmz049)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chen YJ, Li HY, Huang CH, Twu NF, Yen MS, Wang PH, Chou TY, Liu YN, Chao KC & Yang MH 2010 Oestrogen-induced epithelial-mesenchymal transition of endometrial epithelial cells contributes to the development of adenomyosis. Journal of Pathology 222 261270. (https://doi.org/10.1002/path.2761)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chen Y, Zhu B, Zhang H, Ding D, Liu X & Guo SW 2014 Possible loss of GABAergic inhibition in mice with induced adenomyosis and treatment with epigallocatechin-3-gallate attenuates the loss with improved hyperalgesia. Reproductive Sciences 21 869882. (https://doi.org/10.1177/1933719113518984)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Choi YS, Cho S, Lim KJ, Jeon YE, Yang HI, Lee KE, Heena K, Seo SK, Kim HY & Lee BS 2010 Effects of LNG-IUS on nerve growth factor and its receptors expression in patients with adenomyosis. Growth Factors 28 452460. (https://doi.org/10.3109/08977194.2010.511619)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Choi YJ, Chang JA, Kim YA, Chang SH, Chun KC & Koh JW 2015 Innervation in women with uterine myoma and adenomyosis. Obstetrics and Gynecology Science 58 150156. (https://doi.org/10.5468/ogs.2015.58.2.150)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Clements MP, Byrne E, Camarillo Guerrero LF, Cattin AL, Zakka L, Ashraf A, Burden JJ, Khadayate S, Lloyd AC & Marguerat S et al.2017 The wound microenvironment reprograms Schwann cells to invasive mesenchymal-like cells to drive peripheral nerve regeneration. Neuron 96 98 .e7114.e7. (https://doi.org/10.1016/j.neuron.2017.09.008)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cope AG, Ainsworth AJ & Stewart EA 2020 Current and future medical therapies for adenomyosis. Seminars in Reproductive Medicine 38 151156. (https://doi.org/10.1055/s-0040-1719016)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Costalonga EC, de Freitas LJ, Aragone DDSP, Silva FMO & Noronha IL 2017 Anti-fibrotic effects of valproic acid in experimental peritoneal fibrosis. PLoS ONE 12 e0184302. (https://doi.org/10.1371/journal.pone.0184302)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cousins FL, O DF & Gargett CE 2018 Endometrial stem/progenitor cells and their role in the pathogenesis of endometriosis. Best Practice and Research: Clinical Obstetrics and Gynaecology 50 2738. (https://doi.org/10.1016/j.bpobgyn.2018.01.011)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Critchley HO, Jones RL, Lea RG, Drudy TA, Kelly RW, Williams AR & Baird DT 1999 Role of inflammatory mediators in human endometrium during progesterone withdrawal and early pregnancy. Journal of Clinical Endocrinology and Metabolism 84 240248. (https://doi.org/10.1210/jcem.84.1.5380)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Critchley HO, Osei J, Henderson TA, Boswell L, Sales KJ, Jabbour HN & Hirani N 2006 Hypoxia-inducible factor-1alpha expression in human endometrium and its regulation by prostaglandin E-series prostanoid receptor 2 (EP2). Endocrinology 147 744753. (https://doi.org/10.1210/en.2005-1153)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Critchley HOD, Maybin JA, Armstrong GM & Williams ARW 2020 Physiology of the endometrium and regulation of menstruation. Physiological Reviews 100 11491179. (https://doi.org/10.1152/physrev.00031.2019)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cui XW, Li KN, Yi AJ, Wang B, Wei Q, Wu GG & Dietrich CF 2022 Ultrasound elastography. Endoscopic Ultrasound 1 1 252274. (https://doi.org/10.4103/EUS-D-21-00151)

  • Demir IE, Tieftrunk E, Schorn S, Saricaoglu ÖC, Pfitzinger PL, Teller S, Wang K, Waldbaur C, Kurkowski MU & Wormann SM et al.2016 Activated Schwann cells in pancreatic cancer are linked to analgesia via suppression of spinal astroglia and microglia. Gut 65 10011014. (https://doi.org/10.1136/gutjnl-2015-309784)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • DiMauro A, Seger C, Minor B, Amitrano AM, Okeke I, Taya M, Rackow AR, Kumar D, Kottman RM & Bhagavath B et al.2022 Prolactin is expressed in uterine leiomyomas and promotes signaling and fibrosis in myometrial cells. Reproductive Sciences 29 25252535. (https://doi.org/10.1007/s43032-021-00741-w)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ding D, Liu X & Guo SW 2018 Further evidence for hypercoagulability in women with ovarian endometriomas. Reproductive Sciences 25 15401548. (https://doi.org/10.1177/1933719118799195)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ding D, Cai X, Zheng H, Guo SW & Liu X 2019 Scutellarin suppresses platelet aggregation and stalls lesional progression in mouse with induced endometriosis. Reproductive Sciences 26 14171428. (https://doi.org/10.1177/1933719118817661)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ding D, Chen Y, Liu X, Jiang Z, Cai X & Guo SW 2020 Diagnosing deep endometriosis using transvaginal elastosonography. Reproductive Sciences 27 14111422. (https://doi.org/10.1007/s43032-019-00108-2)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Donnez J, Dolmans MM & Fellah L 2019 What if deep endometriotic nodules and uterine adenomyosis were actually two forms of the same disease? Fertility and Sterility 111 454456. (https://doi.org/10.1016/j.fertnstert.2018.12.018)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Donnez J, Stratopoulou CA & Dolmans MM 2021 Uterine adenomyosis: from disease pathogenesis to a new medical approach using GnRH antagonists. International Journal of Environmental Research and Public Health 18 9941. (https://doi.org/10.3390/ijerph18199941)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Duan J, Liu X, Wang H & Guo SW 2018 The M2a macrophage subset may be critically involved in the fibrogenesis of endometriosis in mice. Reproductive Biomedicine Online 37 254268. (https://doi.org/10.1016/j.rbmo.2018.05.017)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dyachuk V, Furlan A, Shahidi MK, Giovenco M, Kaukua N, Konstantinidou C, Pachnis V, Memic F, Marklund U & Muller T et al.2014 Neurodevelopment. Parasympathetic neurons originate from nerve-associated peripheral glial progenitors. Science 345 8287. (https://doi.org/10.1126/science.1253281)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Elenkov IJ, Papanicolaou DA, Wilder RL & Chrousos GP 1996 Modulatory effects of glucocorticoids and catecholamines on human interleukin-12 and interleukin-10 production: clinical implications. Proceedings of the Association of American Physicians 108 374381.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Espinosa-Medina I, Outin E, Picard CA, Chettouh Z, Dymecki S, Consalez GG, Coppola E & Brunet JF 2014 Neurodevelopment. Parasympathetic ganglia derive from Schwann cell precursors. Science 345 8790. (https://doi.org/10.1126/science.1253286)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Farquhar C & Brosens I 2006 Medical and surgical management of adenomyosis. Best Practice and Research: Clinical Obstetrics and Gynaecology 20 603616. (https://doi.org/10.1016/j.bpobgyn.2006.01.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ferenczy A 1998 Pathophysiology of adenomyosis. Human Reproduction Update 4 312322. (https://doi.org/10.1093/humupd/4.4.312)

  • Flores VA, Vanhie A, Dang T & Taylor HS 2018 Progesterone receptor status predicts response to progestin therapy in endometriosis. Journal of Clinical Endocrinology and Metabolism 103 45614568. (https://doi.org/10.1210/jc.2018-01227)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gaetje R, Kotzian S, Herrmann G, Baumann R & Starzinski-Powitz A 1995 Invasiveness of endometriotic cells in vitro. Lancet 346 14631464. (https://doi.org/10.1016/s0140-6736(9592474-4)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Garcia-Solares J, Donnez J, Donnez O & Dolmans MM 2018 Pathogenesis of uterine adenomyosis: invagination or metaplasia? Fertility and Sterility 109 371379. (https://doi.org/10.1016/j.fertnstert.2017.12.030)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gargett CE 2007 Uterine stem cells: what is the evidence? Human Reproduction Update 13 87101. (https://doi.org/10.1093/humupd/dml045)

  • Goldfarb Y, Sorski L, Benish M, Levi B, Melamed R & Ben-Eliyahu S 2011 Improving postoperative immune status and resistance to cancer metastasis: a combined perioperative approach of immunostimulation and prevention of excessive surgical stress responses. Annals of Surgery 253 798810. (https://doi.org/10.1097/SLA.0b013e318211d7b5)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gordts S, Brosens JJ, Fusi L, Benagiano G & Brosens I 2008 Uterine adenomyosis: a need for uniform terminology and consensus classification. Reproductive Biomedicine Online 17 244248. (https://doi.org/10.1016/s1472-6483(1060201-5)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gordts S, Grimbizis G & Campo R 2018 Symptoms and classification of uterine adenomyosis, including the place of hysteroscopy in diagnosis. Fertility and Sterility 109 380 .e1388.e1. (https://doi.org/10.1016/j.fertnstert.2018.01.006)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Goteri G, Lucarini G, Montik N, Zizzi A, Stramazzotti D, Fabris G, Tranquilli AL & Ciavattini A 2009 Expression of vascular endothelial growth factor (VEGF), hypoxia inducible factor-1alpha (HIF-1alpha), and microvessel density in endometrial tissue in women with adenomyosis. International Journal of Gynecological Pathology 28 157163. (https://doi.org/10.1097/PGP.0b013e318182c2be)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Greaves E, Collins F, Esnal-Zufiaurre A, Giakoumelou S, Horne AW & Saunders PT 2014 Estrogen receptor (ER) agonists differentially regulate neuroangiogenesis in peritoneal endometriosis via the repellent factor SLIT3. Endocrinology 155 40154026. (https://doi.org/10.1210/en.2014-1086)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Greaves E, Temp J, Esnal-Zufiurre A, Mechsner S, Horne AW & Saunders PT 2015 Estradiol is a critical mediator of macrophage-nerve cross talk in peritoneal endometriosis. American Journal of Pathology 185 22862297. (https://doi.org/10.1016/j.ajpath.2015.04.012)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Green AR, Edwards RE, Greaves P & White IN 2003 Comparison of the effect of oestradiol, tamoxifen and raloxifene on nerve growth factor-alpha expression in specific neonatal mouse uterine cell types using laser capture microdissection. Journal of Molecular Endocrinology 30 111. (https://doi.org/10.1677/jme.0.0300001)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guo SW 2014 An overview of the current status of clinical trials on endometriosis: issues and concerns. Fertility and Sterility 101 183 .e4190.e4. (https://doi.org/10.1016/j.fertnstert.2013.08.050)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guo SW 2018a Cancer driver mutations in endometriosis: variations on the major theme of fibrogenesis. Reproductive Medicine and Biology 17 369397. (https://doi.org/10.1002/rmb2.12221)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guo SW 2018b Fibrogenesis resulting from cyclic bleeding: the Holy Grail of the natural history of ectopic endometrium. Human Reproduction 33 353356. (https://doi.org/10.1093/humrep/dey015)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guo SW 2020a Cancer-associated mutations in endometriosis: shedding light on the pathogenesis and pathophysiology. Human Reproduction Update 26 423449. (https://doi.org/10.1093/humupd/dmz047)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guo SW 2020b The pathogenesis of adenomyosis vis-a-vis endometriosis. Journal of Clinical Medicine 9 485. (https://doi.org/10.3390/jcm9020485)

  • Guo SW & Evers JLH 2013 Lack of transparency of clinical trials on endometriosis. Obstetrics and Gynecology 121 12811290. (https://doi.org/10.1097/AOG.0b013e318291f299)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guo SW & Groothuis PG 2018 Is it time for a paradigm shift in drug research and development in endometriosis/adenomyosis? Human Reproduction Update 24 577598. (https://doi.org/10.1093/humupd/dmy020)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guo SW, Mao X, Ma Q & Liu X 2013 Dysmenorrhea and its severity are associated with increased uterine contractility and overexpression of oxytocin receptor (OTR) in women with symptomatic adenomyosis. Fertility and Sterility 99 231240. (https://doi.org/10.1016/j.fertnstert.2012.08.038)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guo SW, Zhang Q & Liu X 2017 Social psychogenic stress promotes the development of endometriosis in mouse. Reproductive Biomedicine Online 34 225239. (https://doi.org/10.1016/j.rbmo.2016.11.012)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guo S, Zhang D, Lu X, Zhang Q, Gu R, Sun B & Sun Y 2021 Hypoxia and its possible relationship with endometrial receptivity in adenomyosis: a preliminary study. Reproductive Biology and Endocrinology 19 7. (https://doi.org/10.1186/s12958-020-00692-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Habiba M, Gordts S, Bazot M, Brosens I & Benagiano G 2020 Exploring the challenges for a new classification of adenomyosis. Reproductive Biomedicine Online 40 569581. (https://doi.org/10.1016/j.rbmo.2020.01.017)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hallberg L, Hogdahl AM, Nilsson L & Rybo G 1966 Menstrual blood loss – a population study. Variation at different ages and attempts to define normality. Acta Obstetricia et Gynecologica Scandinavica 45 320351. (https://doi.org/10.3109/00016346609158455)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Han F, Konkalmatt P, Chen J, Gildea J, Felder RA, Jose PA & Armando I 2015 MiR-217 mediates the protective effects of the dopamine D2 receptor on fibrosis in human renal proximal tubule cells. Hypertension 65 11181125. (https://doi.org/10.1161/HYPERTENSIONAHA.114.05096)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hao M, Liu X & Guo SW 2020 Adenomyosis in mice resulting from mechanically or thermally induced endometrial-myometrial interface disruption and its possible prevention. Reproductive Biomedicine Online 41 925942. (https://doi.org/10.1016/j.rbmo.2020.07.023)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hao M, Liu X, Rong P, Li S & Guo SW 2021 Reduced vagal tone in women with endometriosis and auricular vagus nerve stimulation as a potential therapeutic approach. Scientific Reports 11 1345. (https://doi.org/10.1038/s41598-020-79750-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hao M, Liu X & Guo SW 2022 Activation of alpha7 nicotinic acetylcholine receptor retards the development of endometriosis. Reproductive Biology and Endocrinology 20 85. (https://doi.org/10.1186/s12958-022-00955-w)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Harada T, Khine YM, Kaponis A, Nikellis T, Decavalas G & Taniguchi F 2016 The impact of adenomyosis on women’s fertility. Obstetrical and Gynecological Survey 71 557568. (https://doi.org/10.1097/OGX.0000000000000346)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Harrisingh MC, Perez-Nadales E, Parkinson DB, Malcolm DS, Mudge AW & Lloyd AC 2004 The Ras/Raf/ERK signalling pathway drives Schwann cell dedifferentiation. EMBO Journal 23 30613071. (https://doi.org/10.1038/sj.emboj.7600309)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • He W, Liu X, Zhang Y & Guo SW 2010 Generalized hyperalgesia in women with endometriosis and its resolution following a successful surgery. Reproductive Sciences 17 10991111. (https://doi.org/10.1177/1933719110381927)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Heard ME, Melnyk SB, Simmen FA, Yang Y, Pabona JM & Simmen RC 2016 High-fat diet promotion of endometriosis in an immunocompetent mouse model is associated with altered peripheral and ectopic lesion redox and inflammatory status. Endocrinology 157 28702882. (https://doi.org/10.1210/en.2016-1092)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Higham JM & Shaw RW 1999 Clinical associations with objective menstrual blood volume. European Journal of Obstetrics, Gynecology, and Reproductive Biology 82 7376. (https://doi.org/10.1016/s0301-2115(9800224-3)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hinz B 2016 The role of myofibroblasts in wound healing. Current Research in Translational Medicine 64 171177. (https://doi.org/10.1016/j.retram.2016.09.003)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hiraoka T, Hirota Y, Aikawa S, Iida R, Ishizawa C, Kaku T, Hirata T, Fukui Y, Akaeda S & Matsuo M et al.2022 Constant activation of STAT3 contributes to the development of adenomyosis in females. Endocrinology 163 bqac044. (https://doi.org/10.1210/endocr/bqac044)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang S, Wettlaufer SH, Hogaboam C, Aronoff DM & Peters-Golden M 2007 Prostaglandin E(2) inhibits collagen expression and proliferation in patient-derived normal lung fibroblasts via E prostanoid 2 receptor and cAMP signaling. American Journal of Physiology: Lung Cellular and Molecular Physiology 292 L405L413. (https://doi.org/10.1152/ajplung.00232.2006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang SK, White ES, Wettlaufer SH, Grifka H, Hogaboam CM, Thannickal VJ, Horowitz JC & Peters-Golden M 2009 Prostaglandin E(2) induces fibroblast apoptosis by modulating multiple survival pathways. FASEB Journal 23 43174326. (https://doi.org/10.1096/fj.08-128801)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang JH, Duan H, Wang S & Wang YY 2021a Estrogen 17betaestradiol accelerates the proliferation of uterine junctional zone smooth muscle cells via the let7a/Lin28B axis in adenomyosis. Molecular Medicine Reports 23 337. (https://doi.org/10.3892/mmr.2021.11976)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang JH, Duan H, Wang S, Wang YY & Lv CX 2021b Upregulated microRNA let-7a accelerates apoptosis and inhibits proliferation in uterine junctional zone smooth muscle cells in adenomyosis under conditions of a normal activated hippo-YAP1 axis. Reproductive Biology and Endocrinology 19 81. (https://doi.org/10.1186/s12958-021-00753-w)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang Q, Liu X, Critchley H, Fu Z & Guo SW 2022a How does the extent of fibrosis in adenomyosis lesions contribute to heavy menstrual bleeding? Reproductive Medicine and Biology 21 e12442. (https://doi.org/10.1002/rmb2.12442)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang Q, Liu X & Guo SW 2022b Higher fibrotic content of endometriotic lesions is associated with diminished prostaglandin E2 signaling. Reproductive Medicine and Biology 21 e12423. (https://doi.org/10.1002/rmb2.12423)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang S, Xiao F, Guo SW & Zhang T 2022c Tetramethylpyrazine retards the progression and fibrogenesis of endometriosis. Reproductive Sciences 29 11701187. (https://doi.org/10.1007/s43032-021-00813-x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ignatov A, Ignatov T, Roessner A, Costa SD & Kalinski T 2010 Role of GPR30 in the mechanisms of tamoxifen resistance in breast cancer MCF-7 cells. Breast Cancer Research and Treatment 123 8796. (https://doi.org/10.1007/s10549-009-0624-6)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Inoue S, Hirota Y, Ueno T, Fukui Y, Yoshida E, Hayashi T, Kojima S, Takeyama R, Hashimoto T & Kiyono T et al.2019 Uterine adenomyosis is an oligoclonal disorder associated with KRAS mutations. Nature Communications 10 5785. (https://doi.org/10.1038/s41467-019-13708-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jessen KR & Mirsky R 2016 The repair Schwann cell and its function in regenerating nerves. Journal of Physiology 594 35213531. (https://doi.org/10.1113/JP270874)

  • Jessen KR, Mirsky R & Lloyd AC 2015 Schwann cells: development and role in nerve repair. Cold Spring Harbor Perspectives in Biology 7 a020487. (https://doi.org/10.1101/cshperspect.a020487)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jiang X, Konkalmatt P, Yang Y, Gildea J, Jones JE, Cuevas S, Felder RA, Jose PA & Armando I 2014 Single-nucleotide polymorphisms of the dopamine D2 receptor increase inflammation and fibrosis in human renal proximal tubule cells. Hypertension 63 e74e80. (https://doi.org/10.1161/HYPERTENSIONAHA.113.02569)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jichan N, Xishi L & Guo SW 2010 Promoter hypermethylation of progesterone receptor isoform B (PR-B) in adenomyosis and its rectification by a histone deacetylase inhibitor and a demethylation agent. Reproductive Sciences 17 9951005. (https://doi.org/10.1177/1933719110377118)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jordan VC 2006 Tamoxifen (ICI46,474) as a targeted therapy to treat and prevent breast cancer. British Journal of Pharmacology 147 (Supplement 1) S269S276. (https://doi.org/10.1038/sj.bjp.0706399)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Joseph NM, Mukouyama YS, Mosher JT, Jaegle M, Crone SA, Dormand EL, Lee KF, Meijer D, Anderson DJ & Morrison SJ 2004 Neural crest stem cells undergo multilineage differentiation in developing peripheral nerves to generate endoneurial fibroblasts in addition to Schwann cells. Development 131 55995612. (https://doi.org/10.1242/dev.01429)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kastriti ME, Kameneva P, Kamenev D, Dyachuk V, Furlan A, Hampl M, Memic F, Marklund U, Lallemend F & Hadjab S et al.2019 Schwann cell precursors generate the majority of chromaffin cells in Zuckerkandl organ and some sympathetic neurons in paraganglia. Frontiers in Molecular Neuroscience 12 6. (https://doi.org/10.3389/fnmol.2019.00006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kaukua N, Shahidi MK, Konstantinidou C, Dyachuk V, Kaucka M, Furlan A, An Z, Wang L, Hultman I & Ahrlund-Richter L et al.2014 Glial origin of mesenchymal stem cells in a tooth model system. Nature 513 551554. (https://doi.org/10.1038/nature13536)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Khan KN, Kitajima M, Hiraki K, Fujishita A, Nakashima M & Masuzaki H 2015 Involvement of hepatocyte growth factor-induced epithelial-mesenchymal transition in human adenomyosis. Biology of Reproduction 92 35. (https://doi.org/10.1095/biolreprod.114.124891)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Khan KN, Fujishita A, Koshiba A, Kuroboshi H, Mori T, Ogi H, Itoh K, Nakashima M & Kitawaki J 2019 Biological differences between intrinsic and extrinsic adenomyosis with coexisting deep infiltrating endometriosis. Reproductive Biomedicine Online 39 343353. (https://doi.org/10.1016/j.rbmo.2019.03.210)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Khan KN, Fujishita A & Mori T 2022 Pathogenesis of human adenomyosis: current understanding and its association with infertility. Journal of Clinical Medicine 11 4057. (https://doi.org/10.3390/jcm11144057)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kirchhoff C, Biberthaler P, Mutschler WE, Faist E, Jochum M & Zedler S 2009 Early down-regulation of the pro-inflammatory potential of monocytes is correlated to organ dysfunction in patients after severe multiple injury: a cohort study. Critical Care 13 R88. (https://doi.org/10.1186/cc7914)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kishi Y, Suginami H, Kuramori R, Yabuta M, Suginami R & Taniguchi F 2012 Four subtypes of adenomyosis assessed by magnetic resonance imaging and their specification. American Journal of Obstetrics and Gynecology 207 114.e1114.e7. (https://doi.org/10.1016/j.ajog.2012.06.027)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kobayashi H, Matsubara S & Imanaka S 2021 Clinicopathological features of different subtypes in adenomyosis: focus on early lesions. PLoS ONE 16 e0254147. (https://doi.org/10.1371/journal.pone.0254147)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Krantz KE 1959 Innervation of the human uterus. Annals of the New York Academy of Sciences 75 770784. (https://doi.org/10.1111/j.1749-6632.1959.tb44589.x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kuang L, Zhang K & aCP Commission 2005 Pharmacopoeia of the People’s Republic of China. Beijing, China: People’ s Medical Publishing House.

  • Larsson P, Alwis I, Niego B, Sashindranath M, Fogelstrand P, Wu MC, Glise L, Magnusson M, Daglas M & Bergh N et al.2016 Valproic acid selectively increases vascular endothelial tissue-type plasminogen activator production and reduces thrombus formation in the mouse. Journal of Thrombosis and Haemostasis 14 24962508. (https://doi.org/10.1111/jth.13527)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lee SK & Wolfe SW 2000 Peripheral nerve injury and repair. Journal of the American Academy of Orthopaedic Surgeons 8 243252. (https://doi.org/10.5435/00124635-200007000-00005)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lertvikool S, Sukprasert M, Pansrikaew P, Rattanasiri S & Weerakiet S 2014 Comparative study of nerve fiber density between adenomyosis patients with moderate to severe pain and mild pain. Journal of the Medical Association of Thailand 97 791797.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Levgur M, Abadi MA & Tucker A 2000 Adenomyosis: symptoms, histology, and pregnancy terminations. Obstetrics and Gynecology 95 688691. (https://doi.org/10.1016/s0029-7844(9900659-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Levine AJ & Puzio-Kuter AM 2010 The control of the metabolic switch in cancers by oncogenes and tumor suppressor genes. Science 330 13401344. (https://doi.org/10.1126/science.1193494)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Leyendecker G & Wildt L 2011 A new concept of endometriosis and adenomyosis: tissue injury and repair (TIAR). Hormone Molecular Biology and Clinical Investigation 5 125142. (https://doi.org/10.1515/HMBCI.2011.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Leyendecker G, Wildt L & Mall G 2009 The pathophysiology of endometriosis and adenomyosis: tissue injury and repair. Archives of Gynecology and Obstetrics 280 529538. (https://doi.org/10.1007/s00404-009-1191-0)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Leyendecker G, Bilgicyildirim A, Inacker M, Stalf T, Huppert P, Mall G, Bottcher B & Wildt L 2015 Adenomyosis and endometriosis. Re-visiting their association and further insights into the mechanisms of auto-traumatisation. An MRI study. Archives of Gynecology and Obstetrics 291 917932. (https://doi.org/10.1007/s00404-014-3437-8)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Leyendecker G, Wildt L, Laschke MW & Mall G 2022 Archimetrosis: the evolution of a disease and its extant presentation: pathogenesis and pathophysiology of archimetrosis (uterine adenomyosis and endometriosis ). Archives of Gynecology and Obstetrics In press.(https://doi.org/10.1007/s00404-022-06597-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li Y, Zhang SF, Zou SE, Xia X & Bao L 2011 Accumulation of nerve growth factor and its receptors in the uterus and dorsal root ganglia in a mouse model of adenomyosis. Reproductive Biology and Endocrinology 9 30. (https://doi.org/10.1186/1477-7827-9-30)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li B, Chen M, Liu X & Guo SW 2013 Constitutive and tumor necrosis factor-alpha-induced activation of nuclear factor-kappaB in adenomyosis and its inhibition by andrographolide. Fertility and Sterility 100 568577. (https://doi.org/10.1016/j.fertnstert.2013.04.028)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li Y, Zou S, Xia X & Zhang S 2015 Human adenomyosis endometrium stromal cells secreting more nerve growth factor: impact and effect. Reproductive Sciences 22 10731082. (https://doi.org/10.1177/1933719114561559)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu X & Guo SW 2008 A pilot study on the off-label use of valproic acid to treat adenomyosis. Fertility and Sterility 89 246250. (https://doi.org/10.1016/j.fertnstert.2006.11.009)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu X & Guo SW 2011 Valproic acid alleviates generalized hyperalgesia in mice with induced adenomyosis. Journal of Obstetrics and Gynaecology Research 37 696708. (https://doi.org/10.1111/j.1447-0756.2011.01655.x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu X & Guo SW 2012 Aberrant immunoreactivity of deoxyribonucleic acid methyltransferases in adenomyosis. Gynecologic and Obstetric Investigation 74 100108. (https://doi.org/10.1159/000337718)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu F, Mih JD, Shea BS, Kho AT, Sharif AS, Tager AM & Tschumperlin DJ 2010a Feedback amplification of fibrosis through matrix stiffening and COX-2 suppression. Journal of Cell Biology 190 693706. (https://doi.org/10.1083/jcb.201004082)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu X, Yuan L & Guo SW 2010b Valproic acid as a therapy for adenomyosis: a comparative case series. Reproductive Sciences 17 904912. (https://doi.org/10.1177/1933719110373807)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu X, Nie J & Guo SW 2011 Elevated immunoreactivity to tissue factor and its association with dysmenorrhea severity and the amount of menses in adenomyosis. Human Reproduction 26 337345. (https://doi.org/10.1093/humrep/deq311)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu X, Nie J & Guo SW 2012 Elevated immunoreactivity against class I histone deacetylases in adenomyosis. Gynecologic and Obstetric Investigation 74 5055. (https://doi.org/10.1159/000336409)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu X, Yu S & Guo SW 2014 A pilot study on the use of andrographolide to treat symptomatic adenomyosis. Gynecology and Minimally Invasive Therapy 3 119126. (https://doi.org/10.1016/j.gmit.2014.12.002)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu X, Long Q & Guo SW 2016a Surgical history and the risk of endometriosis: a hospital-based case-control study. Reproductive Sciences 23 12171224. (https://doi.org/10.1177/1933719116632921)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu X, Shen M, Qi Q, Zhang H & Guo SW 2016b Corroborating evidence for platelet-induced epithelial-mesenchymal transition and fibroblast-to-myofibroblast transdifferentiation in the development of adenomyosis. Human Reproduction 31 734749. (https://doi.org/10.1093/humrep/dew018)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu X, Ding D, Ren Y & Guo SW 2018a Transvaginal elastosonography as an imaging technique for diagnosing adenomyosis. Reproductive Sciences 25 498514. (https://doi.org/10.1177/1933719117750752)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu X, Zhang Q & Guo SW 2018b Histological and immunohistochemical characterization of the similarity and difference between ovarian endometriomas and deep infiltrating endometriosis. Reproductive Sciences 25 329340. (https://doi.org/10.1177/1933719117718275)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu X, Yan D & Guo SW 2019 Sensory nerve-derived neuropeptides accelerate the development and fibrogenesis of endometriosis. Human Reproduction 34 452468. (https://doi.org/10.1093/humrep/dey392)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu L, Liu S, Luo H, Chen C, Zhang X, He L & Tu G 2021 GPR30-mediated HMGB1 upregulation in CAFs induces autophagy and tamoxifen resistance in ERalpha-positive breast cancer cells. Aging 13 1617816197. (https://doi.org/10.18632/aging.203145)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Long Q, Liu X & Guo SW 2016a Surgery accelerates the development of endometriosis in mice. American Journal of Obstetrics and Gynecology 215 320.e1320.e15. (https://doi.org/10.1016/j.ajog.2016.02.055)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Long Q, Liu X, Qi Q & Guo SW 2016b Chronic stress accelerates the development of endometriosis in mouse through adrenergic receptor beta2. Human Reproduction 31 25062519. (https://doi.org/10.1093/humrep/dew237)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Long Q, Liu X & Guo SW 2020 Early maternal separation accelerates the progression of endometriosis in adult mice. Reproductive Biology and Endocrinology 18 63. (https://doi.org/10.1186/s12958-020-00600-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lu WJ, Lee JJ, Chou DS, Jayakumar T, Fong TH, Hsiao G & Sheu JR 2011 A novel role of andrographolide, an NF-kappa B inhibitor, on inhibition of platelet activation: the pivotal mechanisms of endothelial nitric oxide synthase/cyclic GMP. Journal of Molecular Medicine 89 12611273. (https://doi.org/10.1007/s00109-011-0800-0)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Luo M, Cai X, Yan D, Liu X & Guo SW 2020 Sodium tanshinone IIA sulfonate restrains fibrogenesis through induction of senescence in mice with induced deep endometriosis. Reproductive Biomedicine Online 41 373384. (https://doi.org/10.1016/j.rbmo.2020.04.006)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Madueke-Laveaux OS, Elsharoud A & Al-Hendy A 2021 What we know about the long-term risks of hysterectomy for benign indication – a systematic review. Journal of Clinical Medicine 10 5335. (https://doi.org/10.3390/jcm10225335)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mantyh PW 1991 Substance P and the inflammatory and immune response. Annals of the New York Academy of Sciences 632 263271. (https://doi.org/10.1111/j.1749-6632.1991.tb33114.x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mao X, Wang Y, Carter AV, Zhen X & Guo SW 2011 The retardation of myometrial infiltration, reduction of uterine contractility, and alleviation of generalized hyperalgesia in mice with induced adenomyosis by levo-tetrahydropalmatine (l-THP) and andrographolide. Reproductive Sciences 18 10251037. (https://doi.org/10.1177/1933719111404610)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mao C, Liu X & Guo S-W 2022 Decreased glycolysis at menstruation delays endometrial repair and is associated with increased menstrual blood loss. Reproductive Sciences.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Marcellin L, Santulli P, Bortolato S, Morin C, Millischer AE, Borghese B & Chapron C 2018 Anterior focal adenomyosis and bladder deep infiltrating endometriosis: is there a link? Journal of Minimally Invasive Gynecology 25 896901. (https://doi.org/10.1016/j.jmig.2018.02.002)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Marcellin L, Santulli P, Bourdon M, Maignien C, Campin L, Lafay-Pillet MC, Millischer AE, Bordonne C, Borghese B & Dousset B et al.2020 Focal adenomyosis of the outer myometrium and deep infiltrating endometriosis severity. Fertility and Sterility 114 818827. (https://doi.org/10.1016/j.fertnstert.2020.05.003)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mariappan YK, Glaser KJ & Ehman RL 2010 Magnetic resonance elastography: a review. Clinical Anatomy 23 497511. (https://doi.org/10.1002/ca.21006)

  • Maybin JA, Hirani N, Jabbour HN & Critchley HO 2011 Novel roles for hypoxia and prostaglandin E2 in the regulation of IL-8 during endometrial repair. American Journal of Pathology 178 12451256. (https://doi.org/10.1016/j.ajpath.2010.11.070)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Maybin JA, Murray AA, Saunders PTK, Hirani N, Carmeliet P & Critchley HOD 2018 Hypoxia and hypoxia inducible factor-1alpha are required for normal endometrial repair during menstruation. Nature Communications 9 295. (https://doi.org/10.1038/s41467-017-02375-6)

    • PubMed