Next Article in Journal
The Emerging Role of Virtual Reality as an Adjunct to Procedural Sedation and Anesthesia: A Narrative Review
Next Article in Special Issue
Human Nanoplatelets as Living Vehicles for Tumor-Targeted Endocytosis In Vitro and Imaging In Vivo
Previous Article in Journal
Acute Ischemic Stroke and Transient Ischemic Attack in ST-Segment Elevation Myocardial Infarction Patients Who Underwent Primary Percutaneous Coronary Intervention
Previous Article in Special Issue
Diurnal Oscillations of Fibrinolytic Parameters in Patients with Acute Myocardial Infarction and Their Relation to Platelet Reactivity: Preliminary Insights
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Platelets in the Pathogenesis and Pathophysiology of Adenomyosis

1
Shanghai Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
2
Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai 200011, China
J. Clin. Med. 2023, 12(3), 842; https://doi.org/10.3390/jcm12030842
Submission received: 14 November 2022 / Revised: 4 January 2023 / Accepted: 11 January 2023 / Published: 20 January 2023
(This article belongs to the Special Issue Platelets and the Antiplatelet Drugs: From Bench to Bedside and Back)

Abstract

:
Widely viewed as an enigmatic disease, adenomyosis is a common gynecological disease with bewildering pathogenesis and pathophysiology. One defining hallmark of adenomyotic lesions is cyclic bleeding as in eutopic endometrium, yet bleeding is a quintessential trademark of tissue injury, which is invariably followed by tissue repair. Consequently, adenomyotic lesions resemble wounds. Following each bleeding episode, adenomyotic lesions undergo tissue repair, and, as such, platelets are the first responder that heralds the subsequent tissue repair. This repeated tissue injury and repair (ReTIAR) would elicit several key molecular events crucial for lesional progression, eventually leading to lesional fibrosis. Platelets interact with adenomyotic cells and actively participate in these events, promoting the lesional progression and fibrogenesis. Lesional fibrosis may also be propagated into their neighboring endometrial–myometrial interface and then to eutopic endometrium, impairing endometrial repair and causing heavy menstrual bleeding. Moreover, lesional progression may result in hyperinnervation and an enlarged uterus. In this review, the role of platelets in the pathogenesis, progression, and pathophysiology is reviewed, along with the therapeutic implication. In addition, I shall demonstrate how the notion of ReTIAR provides a much needed framework to tether to and piece together many seemingly unrelated findings and how it helps to make useful predictions.

1. Introduction

Adenomyosis, defined as the presence of endometrial glands and stroma within the myometrium [1], is a uterine disease that affects many women of reproductive age and contributes to dysmenorrhea, pelvic pain, abnormal uterine bleeding (AUB)/heavy menstrual bleeding (HMB), and subfertility [2,3,4,5]. While approximately one third of women with adenomyosis are asymptomatic [6], dysmenorrhea is the most prevalent symptom besides HMB [7]. As a result, adenomyosis negatively impacts the quality of life of the afflicted woman [8]. It is also associated with an increased risk of several adverse pregnancy outcomes [3,9].
Notably, adenomyosis shares many similarities with endometriosis. Both diseases are estrogen-dependent [10], have similar symptomology, and share many molecular aberrations, such as an increased production of proinflammatory cytokines/chemokines [11,12], overexpression of COX-2 [13], increased oxidative stress [14,15,16,17] and angiogenesis [18], epigenetic aberration [19,20], and cancer-driver mutations [21]. Not surprisingly, the management strategies for the two diseases are also similar, and the drugs for treating endometriosis are also used for treating adenomyosis [22]. In fact, few drugs are being developed exclusively for adenomyosis [23].
Yet perhaps the most glaring feature shared by both diseases is cyclic bleeding [24], so much so that one salient commonality conspicuously shared by all current hormonal drugs for treating adenomyosis/endometriosis is that they all arrest cyclic bleeding [24]. However, bleeding is a hallmark of vascular injury and thus tissue injury. In all organisms, following a tissue injury, the evolutionarily conserved tissue repair program will immediately kick in. In other words, adenomyotic lesions thus resemble wounds that undergo repeated tissue injury and repair (ReTIAR) [25]. Consequently, they would experience the well-known four phases in tissue repair: hemostasis, inflammation, proliferation, and remodeling. Among all these phases, platelets are the first to rush to the wounded site, heralding the repair process [26]. Thus, it is quite fitting to review the roles of platelets in adenomyosis, especially in its progression. In addition, since HMB is known to result from impaired endometrial repair [27], it is conceivable that platelets may also have a role in adenomyosis-induced HMB, although research in this area has been surprisingly scanty so far.
Moreover, there are numerous case reports documenting thromboembolism, cerebral infarction, and disseminated intravascular coagulation in women with adenomyosis [28,29,30,31,32,33]. This raises the prospect that women with adenomyosis, especially those with HMB or severe pain, may be hypercoagulable, which, again, hints at the role of platelets.
There have been 3449 papers on adenomyosis as indexed by PubMed (accessed on 14 November 2022), and well over half of them (n = 1941 or 56.3%) were published in the last decade. Many of these publications helped us better understand the pathogenesis and pathophysiology of adenomyosis. We often see very clearly the leaves, twigs, branches, or even a single tree, but unfortunately, we seldom see the forest or the bigger picture. Why is there such an aberration? What is the natural history of the adenomyotic lesion? Is there any theme or framework that can be used to tether to and piece together most, if not all, published findings? Can we make useful predictions to guide our future research?
Without the ReTIAR theme, however, it is difficult to think of any framework that can thether to and piece together many published but seemingly isolated findings. The entire field is like what was depicted in the famous fable on the elephant and the blind men; each of these blind men told a certain truth, but certainly not the whole and the entire truth. However, if we see through the ReTIAR prism, we would realize that the prism is not only useful to assist us in tethering to and piecing together many seemingly unrelated findings, but, more importantly, can also help us to foresee things that we are otherwise completely unaware of.
Within this framework, platelets can be viewed as an important player in adenomyosis. In this review, I shall first provide a brief overview on tissue repair, platelets, and coagulation, and then I will provide a narrative review on the roles of platelets in the pathogenesis and pathophysiology of adenomyosis, piecing together some scattered and seemingly related findings and also attempting to make some useful predictions for future research.

2. A Primer on Tissue Repair, Platelets, and the Coagulation Pathways

Tissue repair, especially dermal tissue repair, has been extensively investigated [34,35]. Following tissue injury, tissue repair or wound healing ensues, which undergoes four distinct but somewhat overlapping phases: hemostasis, inflammation, proliferation, and resolution or remodeling [35]. Upon tissue injury, platelets, as the first-aid cells, are activated and aggregated into the site of the injury, resulting in the formation of a fibrin clot consisting of a network of insoluble fibrin fibers. Besides plugging vessels to stop the hemorrhage, the clot also acts as a provisional matrix to which growth factors bind and through which cells can crawl [36]. The damaged cells also release endogenous molecules, including damage-associated molecular pattern molecules (DAMPs), which may activate pattern recognition receptors (PRRs) and act as activation cues and/or chemotactic factors for other cells in the area [34]. Cells bathed in the serum—the fluid component of clotted blood—which contains many interleukins (ILs), colony-stimulating factors, TNF-α, IFN-γ, and other components, would activate the serum response factor (SRF) that binds and induces transcription of immediate early and other genes, such as c-Fos and early growth response genes [34]. DAMPs, H2O2, Ca2+, chemokines, and other mediators released by injured cells would recruit various inflammatory cells, including neutrophils and macrophages, ushering into the inflammatory phase. Neutrophils also produce neutrophil extracellular traps (NETs) that capture pathogens through a process called NETosis. Early macrophages in the wound, on the other hand, release MCP-1 to recruit more monocytes from the bone marrow and intensify the macrophage response. Macrophages also release pro-inflammatory cytokines, such as IL-6, TNF-α, and IL-1β, to combat infection and carry out phagocytosis of pathogens and tissue debris in the wounding site. At the end of the inflammatory phase, macrophages devour apoptotic neutrophils, ending the inflammatory phase of tissue repair and starting the proliferation phase. They also make transitions into an anti-inflammatory phenotype known as the alternatively activated macrophage or the M2 macrophage [35].
During the proliferation stage of tissue repair, macrophages release growth factors, such as VEGF and PDGF, to induce angiogenesis, which involves the activation of local microvascular endothelial cells. The final stage of tissue repair (i.e., remodeling) consists of the regression of the neovasculature, as well as apoptosis of myofibroblasts. Macrophages also play a phagocytic role where they devour both cell debris and excessive extracellular matrix (ECM), in order to bring the healed tissue to a homeostatic state.
Platelets are anucleated cells originating from cytoplasmic fragmentation of megakaryocytes primarily in the bone marrow. Packaged into several different containers called granules (α-granules, dense granules, and lysosome granules) with a plethora of pre-synthesized bioactive molecules, circulating platelets patrol in the blood of mammals and are critical effectors of hemostasis, blood clotting, and tissue repair [37]. Upon activation following exposure to damaged blood vessels, the surface receptors of platelets undergo a conformational change and bind to exposed sub-endothelial matrix proteins and von Willebrand Factor (vWF), resulting in morphological changes. The conformational change enables ligand binding and subsequent intracellular signaling cascades. The activated platelets create pseudopods, attaching to each other and releasing the contents of granules to initiate the aggregation process. They also bind to fibrinogen, resulting in the platelet plug that can effectively stop the hemorrhage from small injuries. They release ADP from their dense granules and also produce thromboxane A2 (TXA2) from arachidonic acid derived from phospholipids on their membrane. ADP and TXA2 further induce platelet activation [38].
Platelets play a pivotal role in the tissue injury/damage and inflammatory response, destined to initiate the repair of injured tissues. Erratic or uncontrolled platelet activation results in chronic inflammation associated with numeric pathological conditions, including cancer [39], fibrosis [40], and atherothrombosis [41]. Remarkably, platelets have an intricate transcriptome (with an entire repertoire of RNAs produced), activation-dependent post-transcriptional pathways, the capability of influencing extravascular events, and longer life spans than previously appreciated [42].
However, platelets are not merely the cells that mediate hemostasis. Over the years there has been growing evidence that these cells may act as key regulators in immune responses and are involved in the pathogenesis of various immune-mediated diseases, such as irritable bowel disease [43]. They can also function as antigen presenting cells and activate T cells through MHC-I [44]. In addition, platelets from patients with myocardial infarction can activate CD4+/CD69+ T cells and increase the number of regulatory T cells [45].
Upon tissue injury, the initial hemostatic response is triggered by tissue factor (TF) expressed on sub-endothelial pericytes and fibroblasts. Activated Factor VII (fVIIa) then binds to TF to activate fX to fXa, which generates trace amounts of thrombin [46]. There are two major inhibitors that regulate TF-triggered procoagulant responses, thus limiting serine protease actions to the site of vascular injury. TF pathway inhibitor (TFPI) neutralizes fXa when it is in a complex with TF-fVIIa. The other regulator of TF-trigger procoagulant response is antithrombin (AT, also known as AT III), which circulates at a high concentration and neutralizes the initially formed fXa and thrombin. Consequently, the procoagulant triggering reaction only proceeds when TF is exposed at a high enough level to overcome inhibition by TFPI and AT. That is, fVIIa circulates in the blood in search for sites of vascular damage (i.e., where TF is exposed), and trace quantities of fXa and thrombin sound the “alarm” for any potential dangers.
Circulating platelets contribute to localized thrombus formation at the site of vascular injury first by adhering to sub-endothelial collagen-vWF via their glycoprotein (GP) Ib receptors. Thrombin generated by TF-fVIIa/fXa (the “extrinsic pathway”) is capable of activating adherent platelets in its vicinity via protease-activated receptor 1 (PAR1) and PAR4. Thrombin-activated platelets play a pivotal role in subsequent coagulation processes in several ways. First, platelet GPIb receptors bind to fXI, and they also localize fIII to the site of endothelial disruption via its carrier protein vWF. Furthermore, partially activated fV is released from activated platelets. FXI, fVIII, and fV are involved in sustaining procoagulant responses (the “intrinsic pathway”) after thrombin-mediated activation. The serine protease fXIa mediates the activation of fIX to fIXa while fVIIIa serves as a cofactor to fIXa. FVa serves as a cofactor to fXa.
In clinical settings, the activated partial thromboplastin time (aPTT) [16], the prothrombin time (PT), along its international normalized ratio (INR) [17] assays are the three most widely used coagulation tests which correspond, respectively, to the intrinsic (fVIII, fIX, fXII, fXI, fV, fX, prothrombin, and fibrinogen) and extrinsic (prothrombin, fibrinogen, fV, fVII, and fX) pathways of the classical cascade model of hemostasis and measure the levels of individual coagulation factors in these pathways. In contrast, a thrombin time (TT) test measures the rate of the conversion of fibrinogen to fibrin and subsequent clot formation following the addition of thrombin to platelet-poor plasma samples [19]. The TT test may be used to diagnose fibrinogen abnormality when used in conjunction with a prolonged aPTT or PT finding, although it is not recommended for use as a stand-alone diagnostic test [19,20]. While having the advantages of being inexpensive, simple to perform, and easy to automate and validate, it is widely accepted that the aPTT, PT, and TT tests only partially reflect coagulation in a non-physiologic environment and do not capture any temporal or spatial contribution from specific cellular components fundamental to hemostasis [21,22]. Furthermore, the aPTT and PT assays only minimally reflect the levels of natural anticoagulants (e.g., TFPI, AT, and protein C/S) and cannot assess processes such as fibrin polymerization or clot stabilization that occur after the termination stage of these assays [23]. As such, these tests provide information on the initiation of clotting, but not hemostatic capacity in terms of final clot formation and the in vivo hemostatic process.
Apart from PT, aPTT is the most common coagulation test procedure performed in routine laboratories, useful for predicting the propensity for bleeding. The test is traditionally used for identifying quantitative and qualitative abnormalities in the intrinsic (factors VIII, IX, and XI) and common (factors II, V, and X) pathways of coagulation. Short aPTTs used to be dismissed as a laboratory artifact [47], but growing evidence suggests that these are associated with hypercoagulability, manifested as increased thrombin generation [48], and elevated procoagulant factors [49]. Short aPTTs are now considered to represent a procoagulant milieu [50] and have been shown to be associated with venous thromboembolism independent of other risk factors [51]. In patients with chest pain, short aPTTs are associated with increased risk of acute myocardial infarction [52]. They are also associated with impaired fasting glucose level and diabetics [53]. Short aPTTs, along with elevated fibrinogen levels, have also been found to be associated with diabetes [54].

3. Role of Platelets in the Pathogenesis of Adenomyosis

As of now, there are two prevailing theories on the pathogenesis of adenomyosis: metaplasia and invagination [55,56,57]. The former theory posits that the endometrial cells in the muscular layer originate from the metaplasia of Müllerian remnants or stem cells [58,59,60]. In contrast, the latter theory postulates that the direct invasion of endometrium to the muscle layer results from what is called tissue injury and repair that leads to the establishment of lesions [61,62,63]. One important foundation of the invagination theory is the tissue injury and repair (TIAR) hypothesis, proposed by Leyendecker and his associates [61,62], which postulates that injury induced by uterine hyperperistalsis/dysperistalsis causes adenomyosis. The TIAR hypothesis has recently been expanded to lump endometriosis and adenomyosis together as one disease, called archimetriosis [64].
Both theories make a lot of sense. However, so far, there has been no experimental data to support or refute either of the theories or the TIAR hypothesis [65,66]. Neither theory is seemingly backed by any epidemiological data. For the invagination theory in particular, it is unclear as to why and how the hyperperistalsis or injury—arguably the primum movens—occurs. Indeed, uterine peristalsis occurs in all women of reproductive age, but why is there only a fraction of them who develop adenomyosis? What are the risk factors, if any, for this hyperperistalsis/dysperistalsis? Is it possible to intervene or to forestall 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.
In view of extensive epidemiological reports that iatrogenic uterine procedures, such as dilatation and curettage and induced abortion, increase the risk of adenomyosis [67,68,69,70,71], a new hypothesis, termed endometrial-myometrial interface (EMI) disruption (EMID), has been proposed recently [65]. Subsequent animal experiments demonstrate that both mechanically and thermally induced EMID can and does cause adenomyosis in mice [72]. More remarkably, the EMID hypothesis successfully predicted that the risk of developing adenomyosis depends on the mode and severity of EMID and that the risk can be mitigated by perioperative intervention [73]. It also gives a nice explanation as to why iatrogenic uterine procedures are a risk factor for adenomyosis. Of note, the EMID hypothesis also has been validated independently by another mouse model [74].
Tissue injury unavoidably causes disruption of local vasculature as well as the extravasation of blood, leading to platelet aggregation and the formation of clots. Vascular damage results in the loss of perfusion and consequent hypoxia. The tissue hypoxia can be further exacerbated by an influx of inflammatory and stromal cells—all with high metabolic demands for oxygen, which is essential for all aerobic organisms to produce energy via mitochondrial oxidative respiration and to perform other vital biological functions [75]. Once hypoxic, hypoxia-inducible factors (HIFs) are activated [76].
Conceivably, platelets play an important role in the EMID-induced adenomyosis. This is because EMID, induced either mechanically or thermally, would cause tissue and vasculature injury and, as such, platelet aggregation. Yet activated platelets alone can activate HIF-1α—the master regulator of hypoxia, effectively inducing a hypoxic sate in both endometriotic and endometrial stromal cells [77]. In addition, platelets increase the estrogen production in endometriotic stromal cells through upregulation of StAR, HSD3B2, aromatase, and HSD17B1 through the activation of NF-κB and/or TGF-β1 [78]. The increased local estrogen production may facilitate epithelial-mesenchymal transition (EMT), resulting in the invasion of endometrial epithelial cells into the myometrium through disrupted and thus compromised EMI [79].
Interestingly, estrogen has been well documented to be actively involved in tissue repair [80,81]. Numerous studies have shown that estrogen deficiency delays or impairs tissue repair [82,83,84,85,86]. In fact, estrogen is found to be involved in all phases of wound healing [80]. Hence the increased local production of estrogens in adenomyotic lesions [10] may merely reflect the fact that the lesions are indeed wounds.
Consequent to vascular damage, activated platelets can also release an array of cellular growth and angiogenic factors such as PDGF and VEGF, as well as inflammatory mediators, such as IL-8 and IL-1β [87,88,89]. Yet IL-1β released by activated platelets may help to induce de-differentiation of Schwann cells in the EMI region [90]. De-differentiated Schwann cells have recently been implicated in the genesis of adenomyosis induced by EMID [91].

4. Adenomyotic Lesions as Wounds

Like endometriosis, adenomyosis is viewed as an estrogen-dependent disease, characterized by the increased local production of estrogens due to molecular aberrations in steroidogenesis and estrogen-dependent growth of adenomyotic lesions [10]. It also has been recognized as a pelvic inflammatory condition, as manifested by elevated IL-1β, CRH and UCN in adenomyotic nodules [92], NF-κB activation [93,94,95], and the infiltration of macrophages and lymphocytes [96]. In fact, inflammation and coagulation are intricately coupled. Inflammation can activate the coagulation cascade, while coagulation modulates and sustains the inflammatory activity [97,98], establishing a mutually promotional loop. As enormously abundant hematopoietic cells that outnumber leukocytes in the peripheral [99], platelets are now viewed as inflammatory effector cells involved in the activities across the spectrum from acute inflammation to adaptive immunity [100,101]. Consequently, activated platelets are found to play a critical role in initiating inflammation [102]. In some diseases, such as rheumatoid arthritis, platelets can amplify inflammation through collagen-dependent production and release of microparticles [103].
Since ectopic endometrium experiences cyclic bleeding and thus ReTIAR [25], it resembles uncannily to wound healing. This is especially true for endometriotic lesions since scanty research has been done in this regard for adenomyosis. Similar to the inflammation phase in tissue repair, one study published in 2006 reported the rise of neutrophils, and then macrophages in surgically induced endometriosis [104]. In addition, as in tissue repair some immediate early and other genes such as c-Fos are activated in response to SRF [34], lesional expression of c-Fos also has been reported to be elevated at three months after induction of endometriosis in baboons but gradually tapered down later [105].
NETosis is known to play a role in wound healing [106], and NETs have been shown to promote fibroblast-to-myofibroblast transdifferentiation (FMT) and fibrogenesis [107]. This seems to account for elevated plasma NETs in women with endometriosis, especially with deep endometriosis [108]. While elevated NETs have not been reported in adenomyosis, they are very likely to be so. In addition, many DAMP molecules, which are released upon tissue injury or cellular stress and are regarded as endogenous danger signals, such as fibrinogen and two members of the alarmin family, galectins and annexins [109,110], which have also been found to be elevated in adenomyosis [111,112,113,114,115].
In both endometriosis and adenomyosis, EMT has been well recognized to play an important role [79,116,117]. However, EMT is well known to be actively involved in tissue repair as well as fibrosis [118]. Similarly, it has been reported in wound healing that the elevated expression of the CD47—as a “don’t eat me” signal [119]—on murine fibroblasts protect them from being phagocytized and eliminated by macrophages [120], leading to excessive matrix deposition. CD47 interacts with signal regulatory protein α (SIRPα) and regulates the disposal of ineffective normal cells [121]. Thrombospondin-1 (TSP-1) is released by activated platelets and is a ligand of CD47 [122]. TSP-1 also has been shown to participate in fibrogenesis [123].
In endometriosis, the lesional staining of CD47 is indeed elevated and reduces the phagocytosis efficiency of macrophages on endometriotic stromal cells [124]. In addition, TSP-1/CD47/SIRPα collectively enhances cellular viability, reduces apoptosis, and facilitates fibrosis [124]. On the other hand, blocking CD47 ameliorates endometriosis [125]. These findings are consistent with the known role of platelets in tissue repair and are likely to also hold true for adenomyosis.
Yet elevated CD47 expression, along with the activation of Akt, in fibroblasts is a common feature of many fibrotic conditions, induced by the activation of AP1 transcription factor c-Jun in the pathologic fibroblasts [120]. Uncannily similar, both c-Jun and Akt activation have been reported in endometriosis [126,127,128,129]. While c-Jun activation has not been reported in adenomyosis as of now, the activation of Akt has [130]. Consistently, the immunostaining of PP2A, the negative regulator of Akt and of PTEN, and the negative regulator of the PI3K/Akt signaling pathway also have been reported to be reduced in adenomyosis [131].
The α-granules of platelets contain high mobility group box 1 (HMGB1), which is released when platelets are activated [36]. As one of the alarmin/DAMP molecules, elevated plasma HMGB1 levels and lesional expression have been reported in endometriosis [132,133]. It is very likely to be elevated in adenomyotic lesions as well.
In view of the above, it can be seen that adenomyotic lesions are indeed similar to wounds, just as endometriotic lesions. In fact, the role of platelets in the development of adenomyosis has been gradually revealed and recognized in the last few years [134,135], starting with the realization that adenomyotic lesions, just like their endometriotic counterpart [136,137], are practically and fundamentally wounds undergoing ReTIAR [25].
In tissue repair, it is well documented that estrogen is actively involved [80,81]. In fact, estrogen has been shown to be vital to tissue repair, and its deficiency delays or impairs repair [82,83,84,85,86]. Estrogen has been reported to be involved in all phases of tissue repair [80]. Remarkably, endometriotic stromal cells co-cultured with activated platelets display upregulated ERβ [138]. Consistent with the notion that ERβ is shown to play a critical role in tissue repair [139,140], ERβ is overexpressed in adenomyotic lesions [141], as in endometriotic lesions [142,143].
Platelet α-granules contain loads of TSP-1 [144], which are released upon platelet activation. Therefore, the elevated CD47 expression in ectopic endometrium is very likely due, at least in part, to platelet-derived TSP-1. While CD47 overexpression has not been reported in adenomyosis, it is highly likely to be so. Platelets may also be responsible for an increased local production of estrogen and elevated ERβ expression in adenomyotic lesions.
While the role of EMT in facilitating invasion in adenomyosis has been well-documented, few, if any, studies have raised the question of why EMT is involved in the first place. Yet from the ReTIAR perspective, this is actually something to be expected since EMT is known to be vital in wound healing as it is rapidly activated and results in wound closure through re-epithelialization [145], aside from its involvement in development and cancer metastasis. Similarly, myofibroblast activation or FMT is also expected since it leads to tissue contraction, restoring tissue integrity and reducing the wound size [146]. When the repair process goes awry, as in the case of chronic inflammation or recurrent/repeated tissue injury as in adenomyotic lesions, fibrogenesis ensues through prolonged inflammation, EMT, and FMT [147]. Figure 1 presents a schematic diagram that depicts the roles of platelets in adenomyosis [10,35,58,77,78,80,81,134,135,136,137,138,140,141,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,164,165,166,167,168,169,170,171,172,173,174,175].

5. Platelets Promote Progression of Adenomyotic Lesions

With the understanding of the natural history of endometriotic lesions in the last few years [25], we now have a better understanding of the natural history of adenomyotic lesions, which are nearly identical to that of endometriosis. Since adenomyotic lesions undergo cyclic bleeding just as eutopic endometrium, they are fundamentally wounds undergoing ReTIAR just like endometriotic lesions [25]. As such, they undergo EMT, FMT, and smooth muscle metaplasia (SMM) and progress ultimately to fibrosis [134,135,136]. Due to FMT and SMM, stromal cells within lesions eventually are differentiated into smooth muscle cells (SMCs) [134,135,136], which are presumably fused into existing myometrium, causing enlarged uterus in women with adenomyosis. Myometrial SMCs from women with adenomyosis may also experience cellular hypertrophy [176], likely due to the activation of the MAPK/ERK and the PI3K/mTOR/Akt pathways [177] and the upregulation of cannabinoid receptor CB1 [178]. Regardless of the source, the resultant enlarged uterus would exhibit increased magnitude and/or frequency of uterine contraction, particularly when oxytocin receptor (OTR) is overexpressed [179]. Additionally, due to the fusion—by no means seamlessly—of newly turned SMCs into the myometrium, the uterine contraction is likely to be out of synchronization [180], resulting possibly in spasm-like contractions.
One strong indication for the involvement of platelets in adenomyosis is the increased immunostaining of TF in both eutopic and ectopic endometrium from women with adenomyosis [181]. As a cellular receptor, TF binds fVII/fVIIa to initiate the coagulation cascade, even though its role in adenomyosis-induced HMB was thought to be of angiogenic in nature [181]. In particular, higher TF staining in eutopic endometrium is found to be associated with the HMB [181]. TF levels in endometrial bleeding sites have been reported to be higher than that in the non-bleeding ones [182], accompanied by abnormally enlarged and distended blood vessels in the bleeding sites suggestive of an angiogenic role of TF [183]. Moreover, lesional staining of plasminogen activator (PA) inhibitor 1 (PAI-1), which inhibits PA, is reported to be elevated in adenomyosis [184]. This may suggest an impaired fibrinolytic system in adenomyosis.
Thrombin can activate proteinase-activated receptors (PARs), which are members of the G protein-coupled receptor (GPCR) family. The PAR family has four members: PAR-1, PAR-2, PAR-3, and PAR-4 [185]. PAR activation induces the activation of mitogen-activated protein kinases (MAPKs), including extracellular signal-regulated kinases (ERK1/2), c-Jun N-terminal kinases (JNKs), and p38 MAPK. PAR-1 and PAR-2 are expressed in eutopic and ectopic endometrium from women with endometriosis [185].
Similar to adenomyosis [181], TF is upregulated in endometriosis as well [186]. Both PAR-1 and PAR-2 have been reported to be expressed in endometriotic stromal cells [186,187,188]. Thrombin stimulation increases the gene expression and secretion of IL-8 and MCP-1 from endometriotic stromal cells, and PAR-1 activation also induces TF expression [187]. In addition, IL-8 also upregulates TF in endometriotic stromal cells [187]. Thus, it seems that a feed-forward loop exists, which promotes inflammation and coagulation back and forth in endometriosis and likely also in adenomyosis.
PAR-2 in endometriotic stromal cells can be activated by the TF-fVIIa complex and the TF-fVIIa-fXa complex, but it also can be activated by mast cell-derived tryptase, which appears to be aplenty as the number of mast cells is increased in endometriosis [189,190]. In addition, it can be induced by TGF-β1 in endometriosis [191]. PAR-2 activation in endometriotic stromal cells induces increased secretion of IL-6 and IL-8 and stimulates cellular proliferation [188]. While neither PAR-1 nor PAR-2 has been reported in adenomyosis so far, given the commonality that both adenomyotic and endometriotic lesions share, it is highly likely that they also should play a role in adenomyosis.
Since activated platelets release copious amount of TGF-β1 [192], which is a prototypical factor in EMT [193], FMT [194,195], and subsequent fibrogenesis [196], platelets can and have been shown to induce EMT, FMT, and SMM in adenomyosis, promoting fibrogenesis [134,135,136]. Aside from adenomyotic cells, many other cells within lesions and the uterus may also participate in the progression of adenomyosis. Stimulated by activated platelets, vascular endothelial cells can be differentiated into myofibroblasts within lesions through endothelial-mesenchymal transition (EndoMT) [166]. Activated platelets can also turn peritoneal mesothelial cells into myofibroblasts through mesothelial-mesenchymal transition (MMT) [197], causing adhesion and the immobility of the uterus in women with adenomyosis. Moreover, macrophages within adenomyotic lesions would interact with adenomyotic cells and become polarized into alternatively activated macrophages (M2 macrophages), and, along with regulatory T cells (Tregs) within lesions, promote lesional fibrogenesis through type II immunity [168,169].
Platelets may also induce the lesional expression of thymic stromal lymphopoietin (TSLP) through platelet-derived IL-1β [198,199] and drive type 2 immunity that facilitates lesional fibrogenesis [169,198], similar to how platelets induces endothelial TSLP expression to promote fibrogenesis in human systemic sclerosis [199]. Consistent with platelet-induced CD69+ T cell activation [45], there is an increase in CD69+ T cells in the peritoneal fluid from women with endometriosis [200,201] and possibly in adenomyosis as well.

5.1. Platelet-Derived Growth Factors

Platelet-derived PDGF, which has been shown to be elevated in endometrial tissues and in the peritoneal fluid of women with endometriosis [202,203,204] and possibly with adenomyosis, are reported to stimulate endometrial cell proliferation, invasiveness, and migration [202,205,206,207]. However, PDGF has been well-documented to be a pro-fibrotic factor [208,209] and has also been implicated in fibrogenesis of endometriosis [169] and likely of adenomyosis as well.

5.2. Serotonin

Activated platelets also release serotonin (5-hydroxytryptamine, or 5-HT), a molecule with pleiotropic functions and also a powerful vasoconstrictor. While its role in adenomyosis or endometriosis has not been fully understood, its pro-fibrotic role suggests that it may play a role in adenomyosis. One notable example for the link between 5-HT and fibrotic diseases is the condition called carcinoid syndrome, which is characterized by tissue fibrosis in various organs and caused by neuroendocrine carcinoid tumors that secrete vast quantities of 5-HT [210]. It also has been reported that 5-HT facilitated collagen production in fibroblasts through 5-HT2B serotonin receptor in a TGF-β-dependent manner [211]. In addition, 5-HT receptor inhibitors stalled fibrogenesis in mouse models of fibrosis, and platelet inhibition led to decreased 5-HT content in the fibrotic skin and decreased dermal thickening in mice [211]. Serotonin also has been shown to aggravate bleomycin-induced pulmonary fibrosis in mice through promoting inflammation, exudation of proteins and cells, oxidative stress, and upregulation of fibrosis-associated genes in the lung tissues [212]. Antagonism of serotonin receptors 5-HT2 and 5-HT2B is shown to attenuate pro-fibrotic phenotype in human adult dermal fibroblasts by blocking TGF-β1 induced non-canonical signaling pathways including STAT3 and ERK1/2 [213]. Antagonism of 5-HT2B also has been shown to hinder TGF-β1-induced valvular myofibroblast differentiation through inhibition of p38 MAPK phosphorylation [214] and also to attenuate myofibroblast differentiation and subsequent fibrotic responses in vitro and in vivo [215]. Whether platelet-derived 5-HT has any pro-fibrogenic role in adenomyosis thus warrants further investigation.

5.3. Conflicting Findings

A recent study found no evidence of platelet aggregation in adenomyotic lesions [216], which is directly at odds with those reported in [134,135]. Yet a close look at the study reveals that the choice of both cases and controls may be the source of the discrepancy. First, the study recruited 17 patients with exclusively severe diffuse adenomyosis who underwent hysterectomy. Apparently, the disease condition was severe enough for these patients to undergo hysterectomy. In other words, the adenomyotic lesions in these patients were well established and, as such, highly fibrotic. As shown previously, ectopic endometrium with higher fibrotic content has significantly less platelet aggregation than that of lower fibrotic content, due possibly to reduced vascularity [217]. Hence adenomyotic lesions in these patients may have had less platelet aggregation than those of early-stage lesions.
For controls, the study used endometrial tissue samples “from 23 patients undergoing hysterectomy (17%), hysteroscopic biopsy (35%) or curettage (48%). Hysterectomy was carried out due to intramural myoma and none of the patients was diagnosed with adenomyosis or endometriosis. The purpose of intervention by biopsy or curettage was diagnostic in the context of infertility (50%) or heavy menstrual bleeding (50%)” [216]. In contrast, the control sample from [134] consisted of endometrial tissue samples through curettage from 20 women with teratoma (n = 1, 5%), cervical intraepithelial neoplasia (CIN)-III (n = 14, 70%), stage Ia1 cervical cancer (n = 3, 15%), and cervical carcinoma in situ (n = 2, 10%), but without any clinical indication or history of adenomyosis or endometriosis. The choice of controls was not perfect, but effectively minimized the risk of having a coagulant endometrium.
While it is understandable that endometrial samples were collected from those who sought medical attention, it is also notable that the majority of the controls (n = 19, or 83%) had either uterine fibroids or problems in fertility or HMB. It is known from the PALM-COIEN classification that coagulopathy is one possible cause for HMB (AUB-C). As such, it is possible that the control samples could also have the tendency of having increased platelet aggregation. Consequently, the tendency of less, but still existent, platelet aggregation in cases coupled with possible involvement in coagulation in controls (fibroids and coagulopathy) may have significantly reduced the signal-to-noise ratio, obscuring the real difference.
The confidence on the role of platelets in adenomyosis can be further bolstered by numerous case reports documenting thromboembolism in women with adenomyosis [28], along with the overexpression of TF in adenomyosis [181]. Additionally, a recent study reported that, compared with women without adenomyosis, women with adenomyosis had higher platelet count and shorter TT and aPTT [218], and another study reported shorter PT and the negative correlation between uterine size and aPTT/TT [111], suggesting that adenomyosis is associated with hypercoagulability. Our recent data also demonstrates that indeed women with adenomyosis who experienced HMB are also in a hypercoagulable state as in endometriosis (Liu et al., submitted for publication).

5.4. Platelet Activation by Thrombin/Thromboxane

While cyclic bleeding [24] inevitably leads to platelet aggregation, there is also evidence for extravasated platelets in endometriosis [151] and possibly in adenomyosis as well. This seems to suggest that the relationship between adenomyotic lesions and platelets are not entirely uni-directional, but, rather, bi-directional. In fact, it has been reported that increased production of thromboxane B2 (TXB2), a metabolite of TXA2, by endometriotic stromal cells stimulated with IL-1β, increases TF expression as well as thrombin concentration in peritoneal fluids from women with endometriosis [151], suggesting that endometriotic and perhaps adenomyotic lesion as well and its microenvironment are conducive to platelet activation and aggregation.
Indeed, endometriotic stromal cells secrete thrombin and TXA2 and induce platelet activation in a density-dependent fashion [219]. Specifically, co-culture of platelets with endometriotic stromal cells results in increased concentration of TXB2, thrombin, and TGF-β1 in a density-dependent manner [219]. Treatment of endometriotic stromal cells with hirudin (a specific thrombin inhibitor) and Ozagrel (a TXA2 synthetase inhibitor), but not apyrase (an adenosine diphosphate (ATP) pathway inhibitor), resulted in significant and substantial suppression of platelet aggregation [219]. Since adenomyotic stromal cells are very similar to their endometriotic counterpart and share many molecular aberrations [93], there is reason to believe that adenomyotic stromal cells would behave similarly and secrete platelet-activating molecules, such as thrombin and TXA2. Thus, platelets and adenomyotic lesions are likely to engage crosstalk, collectively facilitating the progression of adenomyotic lesions and induce a hypercoagulable state in patients with adenomyosis.

5.5. Platelets-Mediated Suppression of Cytotoxicity in NK Cells in Ectopic Endometrium

As a key component of the innate immune system, natural killer (NK) cells are a subset of lymphocytes that provide the first-line defense against pathogens or transformed cells by exerting cytotoxicity and the regulation of cytokine producing effector functions [220,221]. The function of NK cells is tightly regulated by an array of functionally opposing surface receptors, inhibitory receptors that bind major histocompatibility complex (MHC) class I molecules and protect “self”, and activating receptors that bind ligands on virus-infected or tumor cells [222]. Activating and inhibitory receptors can transduce, respectively, positive or negative signals to regulate NK cell cytotoxicity and cytokine release [223]. NK cells may play an important role in peritoneal immune surveillance, possibly eliminating ectopic endometrial cells, with low or absent expression of MHC class I and stress-induced expression for activating NK receptors in women without adenomyosis. However, the role of NK cells in adenomyosis has so far been inconclusive [224].
However, activated platelets within adenomyotic lesions could provide a physical shield to adenomyotic cells, protecting them from cytotoxicity rendered by NK cells as in endometriosis [225]. In fact, platelet coating, as could happen following cyclic bleeding, provides ectopic endometrial cells a physical cloak against NK cells as well as increased MHC-I expression, effectively providing a cloak of “pseudo-self” to coated cells to protect against NK cell lysis [225]. Co-incubation of target cells with platelets reduces the expression of NKG2D ligands MICA and MICB and reduces the NK cell cytotoxicity. In addition, co-incubation of NK cells with platelets also impairs the NK cell cytotoxicity, and this impaired NK cell cytotoxicity is not due to the increased NK cell apoptosis, but, rather, through reduced NK cell degranulation and IFN-γ production, the reduced expression of activating receptors NKG2D and NKp46, and the increased expression of inhibitory receptor KIR2DL1 in NK cells [225]. On the other hand, TGF-β1 neutralization abolishes the aberrant expression of NKG2D, NKp46, and KIR2DL1 and partially restores the impaired NK cell cytotoxicity induced by activated platelets and their releasate [225]. Taken together, these data provide a strong piece of evidence that activated platelets, which are aggregated in ectopic endometrium following cyclic bleeding or simply due to the release of platelet-activating molecules by endometriotic stromal cells [219], impair NK cell cytotoxicity in endometriosis through multiple mechanisms and both soluble and membrane-bound factors are required for NK cell evasion of endometriotic cells. Platelet-derived TGF-β1 may reduce the expression of the activating receptor NKG2D as well as cytotoxicity of NK cells in women with adenomyosis, as in endometriosis [226].

5.6. Platelets and Coagulation in Adenomyosis-Induced HMB

It has been well-documented that HMB results from impaired endometrial repair [27,227]. As such, platelets and/or coagulation must be involved in endometrial repair and thus HMB. In fact, coagulopathy is one known cause for AUB and has been designated in the FIGO PALM-COIEN classification [228].
Coagulation pathways should be and are involved in adenomyosis in general and adenomyosis-induced HMB (ADM-HMB) in particular for the following reasons. First, coagulation is set off by tissue injury, or more precisely, vascular injury. Platelets are known to play an important role in the development of adenomyosis [134,135]. In fact, anti-platelet therapy has been shown to have therapeutic effect in mouse models of adenomyosis [229], similar to that in endometriosis [230,231]. Thus, the hypercoagulation in adenomyosis agrees with the aggravation and activation of platelets within adenomyotic lesions, which, in turn, induce PAI-1 expression in ectopic endometrium [136], as shown in adenomyosis [184].
Second, extrinsic compression of blood vessels by an enlarged uterus may cause venous stenosis and thus stasis, as well as hypercoagulability, as shown in this study. Adenomyotic stromal cells may also secrete thrombin and TXA2, similar to endometriotic stromal cells [219]. Thus, a positive feed-forward loop could be established, contributing to the hypercoagulability in women with adenomyosis [218], especially in those who complained of HMB. In addition, patients with adenomyosis often have elevated CA125 levels, especially those with a large uterus [232]. High CA125 levels may be associated with cerebral infarction [233], and they correlate with D-dimer levels under a high tumor burden [234]. Furthermore, red blood cells may also be caught in the process of thrombus formation and contribute to the mass of the thrombus; as such, severe anemia might impair normal hemostasis. This would suggest that patients who take iron supplements may have shorter aPTT and higher plasma FDP levels than those without.
Third, irrespective of the cause, HMB ultimately results from impaired endometrial repair [235], which is intricately linked with coagulation as well as fibrinolysis. It is not surprising that coagulopathy is one of the causes for AUB (AUB-C). In fact, the stoppage of menstrual bleed loss (MBL) can be attained through endometrial hemostasis by platelet aggregation, fibrin deposition, and thrombus formation, which are regulated at the molecular level by coordinated and intricate interactions of hormonal, immunological, and hemostatic factors.
Patients with adenomyosis who were experiencing HMB had a significantly higher platelet count and significantly higher plasma fibrinogen and D-dimer levels, but they had shorter PT and aPTT as compared with women without adenomyosis (Liu et al., submitted for publication). Within the ADM-HMB patients, patients who complained of MBL of 100 mL or higher had significantly higher plasma D-dimer and fibrin-degradation products (FDP) levels but shorter aPTT and lower plasma fibrinogen levels than those who had MBL of less than 100 mL (Liu et al., submitted for publication). These data strongly suggest that women with adenomyosis who complained of HMB are in a hypercoagulable state as compared with women without adenomyosis, more so in those who complained of excessive MBL.
APTT and PT are often used to evaluate the intrinsic and extrinsic pathways of coagulation, respectively, both in conjunction with the common pathway [236]. Hence, these finding suggests that, compared with women without adenomyosis, the enhanced hypercoagulation in adenomyosis [218] and women with adenomyosis who complained of HMB may be attributable to the activation of both intrinsic and extrinsic coagulation pathways. Fibrinogen is a known coagulation factor associated with hypercoagulation [237] and the release of TXA2. Once thrombin is generated, fibrinogen activates platelets to produce TXA2, resulting in more platelet activation, which, in turn, induces PAI-1 expression in ectopic endometrium [136], as demonstrated recently in adenomyosis [184]. This may establish a vicious cycle in maintaining platelet activation, the activation of the coagulation cascade, and then higher plasma fibrinogen levels and shorter aPTTs in adenomyosis.
The elevated fibrinogen levels in ADM-HMB patients as compared with women without adenomyosis (Liu et al., submitted for publication) is suggestive of a heightened coagulable state in the former group. In contrast, within ADM-HMB patients who experienced excessive HMB were found to have decreased fibrinogen levels as compared with those of moderate-heavy MBL. Since thrombin converts soluble plasma fibrinogen into fibrin, the reduced fibrinogen levels may suggest a more efficient conversion of fibrinogen to fibrin, thus increased fibrinolytic activity probably due to the increased thrombin production, since thrombin acts as a serine protease that converts soluble fibrinogen into insoluble strands of fibrin, as well as catalyzing many other coagulation-related reactions [238].
These notions are consistent with the report that both eutopic and ectopic endometrium in women with adenomyosis exhibit elevated expression of TF and that the TF immunoreactivity in eutopic endometrium correlated positively with the amount of MBL [181]. Adenomyotic lesions may also release TF-positive macrovesicles, which may lead to the direct initiation of the coagulation cascade since TF activates the extrinsic pathway of the coagulation cascade, resulting in thrombin generation, platelet aggregation, and clot formation. These findings also are consistent with numerous case reports documenting increased thrombotic propensity in women with adenomyosis [28,29,30,31,32].
TF activation increases the production of thrombin, which is known to induce VEGF expression in several cell types [239,240] and to induce VEGF secretion from human endometrial stromal cells undergoing decidualization [241]. Since VEGF overexpression also results in vascular permeability, persistent thrombin generation could lead to enlarged, fragile, and leaky vessels, overwhelming TF-thrombin mediated hemostasis and, as such, causing HMB. This is especially true since thrombin and a PAR-1 agonist increase the production of MMP-1 and active MMP-2 in endometrial stromal cells [242], raising the possibility that thrombin may facilitate tissue degradation or impair endometrial integrity via PAR-1, increasing the amount of MBL.
There are several implications. First, the realization of the hypercoagulability in patients with adenomyosis who complained of HMB should help healthcare providers to take precautionary or even prophylactic measures when seeing such patients due to increased risk of thrombotic events [28,29,30,31,32]. In addition, the conceivable increase in thrombin levels in adenomyosis patients who complained of excessive HMB may also signal possible impaired decidualization since thrombin can reduce the secretion of prolactin, a key marker of decidualization, alter the morphological transformation of decidualizing endometrial stromal cells, and activate genes involved in matrix degradation and proinflammatory chemokines [243]. Moreover, the hypercoagulability in these patients, the heightened coagulation and fibrinolytic activity in adenomyosis who complained of excessive HMB s, along with the anticipated alleviation of hypercoagulation after removal or perhaps containment of adenomyotic lesions as in endometriosis [244], suggest that coagulation parameters could be used for monitoring the treatment response/efficacy.

6. Platelets and Coagulation in Adenomyosis-Induced Dysmenorrhea

As of now, the molecular mechanisms underlying adenomyosis-induced dysmenorrhea are not fully elucidated, but they are thought to be attributable to increased uterine contractility, hyperinnervation, increased lesion-derived pain mediators, and central sensitization. Through platelet-derived TGF-β1, activated platelets can promote the progression of adenomyosis through EMT, FMT, SMM and fibrogenesis [134,135] as well as EndoMT and MMT [166,197]. The increasing lesional fibrosis is correlated with the severity of dysmenorrhea [245,246]. Platelet activation and the resultant induction of TF in adenomyotic lesions [93,181] would result in the release of thrombin in the lesion and then to its neighboring myometrium, which may facilitate, through PAR-1 [247,248], uterine contractions [249,250], contributing to dysmenorrhea.
In addition, histamine and serotonin released by platelet dense granules when activated have been shown to increase uterine contractility [251,252,253,254]. As adenomyotic lesions are located within the myometrium and platelet-derived histamine and serotonin are likely to permeate into neighboring myometrium, eliciting increased uterine contractility and, as such, contributing to dysmenorrhea.
Activated platelets also release platelet-activating factor (PAF) [144], which may be involved in dysmenorrhea and pelvic pain [255]. Indeed, in women with endometriosis, a condition associated with NSAID-resistant dysmenorrhea, peritoneal PAF synthesis is enhanced [256]. In contrast, PAF acetylhydrolase, which hydrolyzes PAF and related oxidized phospholipids, is reported to be reduced in the peritoneal fluid [257]. Intraperitoneal injections of a PAF receptor agonist CPAF and PGF evoked visceral pain and pelvic hyperalgesia through reducing uterine perfusion and increased uterine contractility [163,164,258,259,260,261,262,263,264,265,266,267,268].

7. Putting Pieces Together

The adoption of the view that adenomyotic lesions are wounds undergoing ReTIAR has important implications for both research and clinical management of adenomyosis. It immediately provides a broader perspective on the big picture of the natural history of adenomyotic lesions. Several key molecular processes, such as EMT, FMT, SMM, and fibrogenesis, are now in plain sight, and with these we can see not just leaves, twigs, branches, and trees, but also the entire forest—in broad strokes, at the very least.
The ReTIAR framework also provides a dynamic view of the lesional progression and underscores the importance of lesional microenvironment (Figure 1). Indeed, many cells in adenomyotic lesions may change their identity—epithelial cells become mesenchymal cells and thus stromal/fibroblasts, and fibroblasts can be turned into myofibroblasts. Many cells, including platelets, within the lesional microenvironment are not merely innocent bystanders but are actually active aiders and abettors that co-conspire with adenomyotic cells to facilitate lesional progression.
The ReTIAR prism not only provides a much needed backbone with which we can tether to and piece together many seemingly unrelated findings, but also help to guide us to make useful predictions. For example, given the role of CD47 in tissue repair and in endometriosis, and particularly given that platelets, when activated, can release TSP-1 [144], we can deduce that platelet-derived TSP-1 can upregulate CD47 expression in adenomyotic lesions and reduce the phagocytosis efficiency of macrophages on adenomyotic stromal cells, as in endometriosis [124]. In addition, TSP-1/CD47/SIRPα collectively enhance cellular viability, reduce apoptosis, and facilitate fibrosis, as in endometriosis [124].
Platelet granules contain a variety of important adhesive, inflammatory, angiogenic, and pro-thrombogenic molecules [269], and their roles in the development of adenomyosis have not been investigated so far. Many other scattered and seemingly unrelated findings can actually be pieced together. For example, plasma levels of lysophosphatidic acid (LPA) have been reported to be elevated in women with adenomyosis, along with some of its receptors LPARs [270]. In endometriosis, it has been reported that LPA facilitates the invasiveness of endometriotic epithelial cells through LPAR1 and LPAR3 [271]. In adenomyosis, it has been reported recently that estrogen-increased SGK1 promotes endometrial stromal cell invasion by regulating LPAR2 [272], which also is shown to play a role in ovarian endometrioma [273]. However, activated platelets release LPA [144]. In addition, SGK1, which is a target of TGF-β1 [274], has been shown to be a powerful regulator of platelet dense granule biogenesis, platelet secretion, and thrombus formation [275]. In fact, platelet-derived LPA promotes proliferation in tumor cells [276].
Similarly, the sphingosine 1 phosphate (S1P) signaling pathway has been reported to be dysregulated in adenomyosis, manifesting as elevated S1P receptor S1P3 but not S1P2 expression [277]. In endometriosis, S1P also increased IL-6 expression and cellular proliferation [278] and induced alternatively activated macrophages [279]. In particular, several S1Ps are overexpressed in both ovarian endometrioma and deep endometriotic lesions and appear to be involved in TGF-β1-induced fibrogenesis [280]. Again, activated platelets release S1P [144], which can also induce TF expression in endothelial cells [281].
The dynamic progression of adenomyotic lesions would help us in the clinical management of adenomyosis. For example, with the understanding that the PGE2 signaling is important for early lesions but is detrimental to older lesions [282,283], one should refrain from the use of COX-2 inhibitors for treatment purposes for patients with well-established or advanced adenomyosis. In addition, one may capitalize on the knowledge of progressive fibrogenesis of adenomyosis and employ elastography to better diagnose adenomyosis [245].

8. Therapeutic Implications

In view of the facilitating role of platelets in adenomyosis progression, naturally, one may think that anti-platelet therapy can have therapeutic potentials. Remarkably, in traditional Chinese medicine (TCM), adenomyosis- or endometriosis-related symptoms such as dysmenorrhea, pain, and infertility have always been recognized as being caused by “blood stasis” or, in modern medical parlance, aberrant coagulation, and the treatment has been invariably the use of herbs, in various concoctions, that are now known to be anti-platelet or anti-thrombotic. Indeed, it has been reported that platelet depletion resulted in a significantly reduced lesion size and improved hyperalgesia in mice with induced endometriosis [151]. The treatment with a recombinant P-selectin in a mouse with induced endometriosis resulted in soluble P-selectin treatment markedly reduced the lesion size in the mouse through decreased platelet aggregation and angiogenesis, improved general hyperalgesia, and the reduced extent of macrophages infiltration, resulting in reduced fibrotic tissue content [230]. In addition, treatment with Ozagrel, a TXA2 synthase inhibitor, yields significant reduction in lesion growth along with improved hyperalgesia in mice with induced endometriosis [231]. Other anti-platelet compounds, such as scutellarin [284], andrographolide [285], and sodium tanshinone IIA [286], also show therapeutic potentials in preclinical studies [287,288]. Similar to endometriosis, the treatment with the anti-platelet drug Ozagrel, as well as platelet depletion, dose-dependently reduced platelet aggregation and the number of macrophages; suppressed myometrial infiltration; improved generalized hyperalgesia; reduced uterine contractility; lowered plasma corticosterone levels; and reduced the lesional staining of COX-2, phosphorylated NF-κB p65 subunit, oxytocin receptor, and TRPV1, but it elevated the lesional staining of PR-B and collagens and slowed down the process of fibrogenesis of mouse with induced adenomyosis [229]. Similarly, andrographolide [180], epigallocatechin-3-gallate [289], resveratrol [290], quercetin [291], leonurine [292], berberine [293], and valproic acid [180,294] all have been demonstrated to be therapeutic potentials in mice with induced adenomyosis. In addition, andrographolide [93], tanshinone IIA [295], and valproic acid [296] have demonstrated their therapeutic potential through in vitro studies.
Andrographolide [297], berberine [298], resveratrol [299], quercetin [300], leonurine [301], tanshinone IIA [302,303], valproic acid [304], and even danazol [305] are known to be anti-platelet. Consistent with the preclinical studies, valproic acid, andrographolide, and danazol ring have shown clinically to be promising in treating adenomyosis [306,307,308,309]. Other compounds have been tested in preclinical studies, but not in humans yet.

9. Summary and Perspective

Growing evidence indicates that, in a nutshell, adenomyotic lesions are fundamentally wounds undergoing ReTIAR due to cyclic bleeding. Platelets are rapidly deployed to and aggregate at the wounded site to initiate hemostasis, inaugurating the tissue repair process of inflammation, proliferation, and tissue remodeling [36]. Activated platelets secret a plethora of bioactive molecules, including various cytokines/chemokines and growth factors [192]. As such, the involvement of platelets in adenomyosis appears to be, in retrospect, rather obvious. Yet platelets do not just passively impact adenomyotic lesions. Adenomyotic stromal cells may also produce potent platelet-activating molecules, such as thrombin and TXA2 [219], and collagens [136], which, coupled with increased angiogenesis and thus vascular permeability, may further lead to platelet aggregation. Consequently, adenomyotic lesions and platelets engage active cross-talks to maintain lesion growth and facilitate lesional progression and fibrogenesis [136,137,151].
Because of the involvement of platelets and TF activation in adenomyosis, women with adenomyosis are in a hypercoagulable state [218], and this may account for the increased risk of thrombosis as reported in the literature [28]. This seems to suggest that adenomyotic lesions and platelet activation are mutually causative, or at least they are intimately entwined.
From the perspective ReTIAR, the natural history of adenomyotic lesions can be easily grasped, although a lot of details are still in need of ironing out. With this prism, we can see the important role of platelets and other immune cells in the development of adenomyosis, as well as adenomyosis-induced HMB and dysmenorrhea. Importantly, the notion of ReTIAR provides a much needed framework so that we can tether to and piece together many seemingly unrelated findings. Importantly, the ReTIAR also helps us to make useful predictions, for example, the role of platelet-derived TSP-1, PAF, HMGB1, and S1P, which otherwise seem to be utterly unrelated. More importantly, this ReTIAR notion also gives us clues to look for and round up other culprits that caused adenomyosis, which are otherwise unsuspected or overlooked, and, as such, at large. These would include, but not limited to, histamine, serotonin, and thrombin, which are very likely to be involved in the progression of adenomyosis and adenomyosis-induced HMB and/or dysmenorrhea. Of course, future studies are warranted to interrogate these suspects.
While the involvement of platelets in the progression of adenomyosis is gaining support, their roles in interacting with other immune cells in the context of lesional progression are still poorly understood. For example, platelets seem to work with Treg cells to form a type 2 immunity in lesional microenvironment that is conducive to lesional progression and fibrogenesis [169]. In other words, we have just scratched the surface. How platelets work with other immune cells, what their underlying molecular mechanisms are, and how to devise novel therapeutics to treat adenomyosis more effectively are unresolved questions that warrant future research.
Regardless, the ReTIAR notion and the roles of platelets in adenomyosis, as elaborated above, not only help us to piece together many seemingly unrelated findings, but also help us to see things that we were not aware of before. As demonstrated in several studies, anti-platelet therapeutics appears to be promising in treating adenomyosis, although their real efficacy would await much research. Still, given the spectacular failure of clinical trials of non-hormonal drugs in treating endometriosis/adenomyosis [23,310], it may be time to re-appraise the strategy, For example, anti-angiogenesis and anti-inflammation are often advocated, but the normal physiology of endometrium also requires angiogenesis and inflammation. Hence, a direct assault on angiogenesis and inflammation is likely to disrupt normal angiogenesis and inflammation needed for endometrial repair and, as such, cause collateral damage. On the other hand, as platelets are situated at the crossroads of proinflammatory and the resolution pathways during inflammation [311], the anti-platelet or anti-coagulation approach appears to more rational and beneficial.

Funding

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

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

S.W.G. provided consultancy advice for MSD R&D, Chugai Pharmaceutical Co., and BioHaven Pharmaceuticals, and is a member of the Scientific Advisory Board of Heranova BioSciences, but these activities had no bearing on this work.

References

  1. Bird, C.C.; McElin, T.W.; Manalo-Estrella, P. The elusive adenomyosis of the uterus—Revisited. Am. J. Obstet. Gynecol. 1972, 112, 583–593. [Google Scholar] [CrossRef]
  2. Farquhar, C.; Brosens, I. Medical and surgical management of adenomyosis. Best. Pract. Res. Clin. Obstet. Gynaecol. 2006, 20, 603–616. [Google Scholar] [CrossRef] [PubMed]
  3. Harada, T.; Khine, Y.M.; Kaponis, A.; Nikellis, T.; Decavalas, G.; Taniguchi, F. The impact of adenomyosis on women’s fertility. Obstet. Gynecol. Surv. 2016, 71, 557–568. [Google Scholar] [CrossRef]
  4. Vercellini, P.; Consonni, D.; Dridi, D.; Bracco, B.; Frattaruolo, M.P.; Somigliana, E. Uterine adenomyosis and in vitro fertilization outcome: A systematic review and meta-analysis. Hum. Reprod. 2014, 29, 964–977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Gordts, S.; Grimbizis, G.; Campo, R. Symptoms and classification of uterine adenomyosis, including the place of hysteroscopy in diagnosis. Fertil. Steril. 2018, 109, 380–388.e1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Benson, R.C.; Sneeden, V.D. Adenomyosis: A reappraisal of symptomatology. Am. J. Obstet. Gynecol. 1958, 76, 1044–1057. [Google Scholar] [CrossRef]
  7. Li, X.; Liu, X.; Guo, S.W. Clinical profiles of 710 premenopausal women with adenomyosis who underwent hysterectomy. J. Obstet. Gynaecol. Res. 2014, 40, 485–494. [Google Scholar] [CrossRef]
  8. Alcalde, A.M.; Martinez-Zamora, M.A.; Gracia, M.; Ros, C.; Rius, M.; Castelo-Branco, C.; Carmona, F. Assessment of Quality of Life, Sexual Quality of Life, and Pain Symptoms in Deep Infiltrating Endometriosis Patients With or Without Associated Adenomyosis and the Influence of a Flexible Extended Combined Oral Contraceptive Regimen: Results of a Prospective, Observational Study. J. Sex. Med. 2022, 19, 311–318. [Google Scholar] [CrossRef]
  9. Harada, T.; Taniguchi, F.; Amano, H.; Kurozawa, Y.; Ideno, Y.; Hayashi, K.; Harada, T.; Japan, E.; Children’s Study, G. Adverse obstetrical outcomes for women with endometriosis and adenomyosis: A large cohort of the Japan Environment and Children’s Study. PLoS ONE 2019, 14, e0220256. [Google Scholar] [CrossRef] [Green Version]
  10. Kitawaki, J. Adenomyosis: The pathophysiology of an oestrogen-dependent disease. Best. Pract. Res. Clin. Obstet. Gynaecol. 2006, 20, 493–502. [Google Scholar] [CrossRef]
  11. Wu, M.Y.; Ho, H.N. The role of cytokines in endometriosis. Am. J. Reprod. Immunol. 2003, 49, 285–296. [Google Scholar] [CrossRef] [PubMed]
  12. Ulukus, M.; Ulukus, E.C.; Tavmergen Goker, E.N.; Tavmergen, E.; Zheng, W.; Arici, A. Expression of interleukin-8 and monocyte chemotactic protein 1 in women with endometriosis. Fertil. Steril. 2008, 91, 687–693. [Google Scholar] [CrossRef] [PubMed]
  13. Ota, H.; Igarashi, S.; Sasaki, M.; Tanaka, T. Distribution of cyclooxygenase-2 in eutopic and ectopic endometrium in endometriosis and adenomyosis. Hum. Reprod. 2001, 16, 561–566. [Google Scholar] [CrossRef] [Green Version]
  14. Ota, H.; Igarashi, S.; Hatazawa, J.; Tanaka, T. Immunohistochemical assessment of superoxide dismutase expression in the endometrium in endometriosis and adenomyosis. Fertil. Steril. 1999, 72, 129–134. [Google Scholar] [CrossRef] [PubMed]
  15. Ota, H.; Igarashi, S.; Hatazawa, J.; Tanaka, T. Endothelial nitric oxide synthase in the endometrium during the menstrual cycle in patients with endometriosis and adenomyosis. Fertil. Steril. 1998, 69, 303–308. [Google Scholar] [CrossRef]
  16. Ota, H.; Igarashi, S.; Kato, N.; Tanaka, T. Aberrant expression of glutathione peroxidase in eutopic and ectopic endometrium in endometriosis and adenomyosis. Fertil. Steril. 2000, 74, 313–318. [Google Scholar] [CrossRef]
  17. Ota, H.; Igarashi, S.; Tanaka, T. Xanthine oxidase in eutopic and ectopic endometrium in endometriosis and adenomyosis. Fertil. Steril. 2001, 75, 785–790. [Google Scholar] [CrossRef]
  18. Goteri, G.; Lucarini, G.; Montik, N.; Zizzi, A.; Stramazzotti, D.; Fabris, G.; Tranquilli, A.L.; Ciavattini, A. Expression of vascular endothelial growth factor (VEGF), hypoxia inducible factor-1alpha (HIF-1alpha), and microvessel density in endometrial tissue in women with adenomyosis. Int. J. Gynecol. Pathol. Off. J. Int. Soc. Gynecol. Pathol. 2009, 28, 157–163. [Google Scholar] [CrossRef]
  19. Liu, X.; Guo, S.W. Aberrant immunoreactivity of deoxyribonucleic acid methyltransferases in adenomyosis. Gynecol. Obstet. Investig. 2012, 74, 100–108. [Google Scholar] [CrossRef]
  20. Liu, X.; Nie, J.; Guo, S.W. Elevated immunoreactivity against class I histone deacetylases in adenomyosis. Gynecol. Obstet. Investig. 2012, 74, 50–55. [Google Scholar] [CrossRef]
  21. Inoue, S.; Hirota, Y.; Ueno, T.; Fukui, Y.; Yoshida, E.; Hayashi, T.; Kojima, S.; Takeyama, R.; Hashimoto, T.; Kiyono, T.; et al. Uterine adenomyosis is an oligoclonal disorder associated with KRAS mutations. Nat. Commun. 2019, 10, 5785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Vannuccini, S.; Luisi, S.; Tosti, C.; Sorbi, F.; Petraglia, F. Role of medical therapy in the management of uterine adenomyosis. Fertil. Steril. 2018, 109, 398–405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Guo, S.W.; Groothuis, P.G. Is it time for a paradigm shift in drug research and development in endometriosis/adenomyosis? Hum. Reprod. Update 2018, 24, 577–598. [Google Scholar] [CrossRef] [Green Version]
  24. Brosens, I.A. Endometriosis—A disease because it is characterized by bleeding. Am. J. Obstet. Gynecol. 1997, 176, 263–267. [Google Scholar] [CrossRef]
  25. Guo, S.W. Fibrogenesis resulting from cyclic bleeding: The Holy Grail of the natural history of ectopic endometrium. Hum. Reprod. 2018, 33, 353–356. [Google Scholar] [CrossRef] [PubMed]
  26. Shaw, T.J.; Martin, P. Wound repair: A showcase for cell plasticity and migration. Curr. Opin. Cell. Biol. 2016, 42, 29–37. [Google Scholar] [CrossRef] [Green Version]
  27. Cheong, Y.; Cameron, I.T.; Critchley, H.O.D. Abnormal uterine bleeding. Br. Med. Bull. 2017, 123, 103–114, Correction in Br. Med. Bull. 2019, 131, 119. [Google Scholar] [CrossRef]
  28. Hong, E.Y.; Lin, H.Z.; Fong, Y.F. Venous Thromboembolism and Adenomyosis: A Retrospective Review. Gynecol. Minim. Invasive. Ther. 2020, 9, 64–68. [Google Scholar] [CrossRef]
  29. Kim, B.; Kim, S.H.; Kim, T. Cerebral Infarcts by Nonbacterial Thrombotic Endocarditis Associated with Adenomyosis: A Case Report. J. Stroke Cerebrovasc. Dis. 2018, 27, e50–e53. [Google Scholar] [CrossRef]
  30. Yamashiro, K.; Tanaka, R.; Nishioka, K.; Ueno, Y.; Shimura, H.; Okuma, Y.; Hattori, N.; Urabe, T. Cerebral infarcts associated with adenomyosis among middle-aged women. J. Stroke Cerebrovasc. Dis. 2012, 21, e911–e915. [Google Scholar] [CrossRef]
  31. Yin, X.; Wu, J.; Song, S.; Zhang, B.; Chen, Y. Cerebral infarcts associated with adenomyosis: A rare risk factor for stroke in middle-aged women: A case series. BMC. Neurol. 2018, 18, 213. [Google Scholar] [CrossRef] [PubMed]
  32. Zhang, J.; Xiao, X.; Luo, F.; Shi, G.; He, Y.; Yao, Y.; Xu, L. Acute disseminated intravascular coagulation developed after dilation and curettage in an adenomyosis patient: A case report. Blood. Coagul. Fibrinolysis 2013, 24, 771–773. [Google Scholar] [CrossRef] [PubMed]
  33. Aiura, R.; Nakayama, S.; Yamaga, H.; Kato, Y.; Fujishima, H. Systemic thromboembolism including multiple cerebral infarctions with middle cerebral artery occlusion caused by the progression of adenomyosis with benign gynecological tumor: A case report. BMC. Neurol. 2021, 21, 14. [Google Scholar] [CrossRef] [PubMed]
  34. Shaw, T.J.; Martin, P. Wound repair at a glance. J. Cell. Sci. 2009, 122, 3209–3213. [Google Scholar] [CrossRef] [Green Version]
  35. Rodrigues, M.; Kosaric, N.; Bonham, C.A.; Gurtner, G.C. Wound Healing: A Cellular Perspective. Physiol. Rev. 2019, 99, 665–706. [Google Scholar] [CrossRef]
  36. Nurden, A.T.; Nurden, P.; Sanchez, M.; Andia, I.; Anitua, E. Platelets and wound healing. Front. Biosci. A J. Virtual Libr. 2008, 13, 3532–3548. [Google Scholar] [CrossRef] [PubMed]
  37. Van der Meijden, P.E.J.; Heemskerk, J.W.M. Platelet biology and functions: New concepts and clinical perspectives. Nat. Rev. Cardiol. 2019, 16, 166–179. [Google Scholar] [CrossRef]
  38. Yun, S.H.; Sim, E.H.; Goh, R.Y.; Park, J.I.; Han, J.Y. Platelet Activation: The Mechanisms and Potential Biomarkers. Biomed. Res. Int. 2016, 2016, 9060143. [Google Scholar] [CrossRef] [Green Version]
  39. Gay, L.J.; Felding-Habermann, B. Contribution of platelets to tumour metastasis. Nat. Rev. Cancer. 2011, 11, 123–134. [Google Scholar] [CrossRef]
  40. Ntelis, K.; Bogdanos, D.; Dimitroulas, T.; Sakkas, L.; Daoussis, D. Platelets in Systemic Sclerosis: The Missing Link Connecting Vasculopathy, Autoimmunity, and Fibrosis? Curr. Rheumatol. Rep. 2019, 21, 15. [Google Scholar] [CrossRef]
  41. Gawaz, M.; Langer, H.; May, A.E. Platelets in inflammation and atherogenesis. J. Clin. Investig. 2005, 115, 3378–3384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Zimmerman, G.A.; Weyrich, A.S. Signal-dependent protein synthesis by activated platelets: New pathways to altered phenotype and function. Arterioscler. Thromb. Vasc. Biol. 2008, 28, s17–s24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Collins, C.E.; Rampton, D.S. Review article: Platelets in inflammatory bowel disease—Pathogenetic role and therapeutic implications. Aliment. Pharmacol. Ther. 1997, 11, 237–247. [Google Scholar] [CrossRef] [Green Version]
  44. Chapman, L.M.; Aggrey, A.A.; Field, D.J.; Srivastava, K.; Ture, S.; Yui, K.; Topham, D.J.; Baldwin, W.M., 3rd; Morrell, C.N. Platelets present antigen in the context of MHC class I. J. Immunol. 2012, 189, 916–923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Solomou, E.E.; Katsanaki, K.; Kalyvioti, E.; Gizas, V.; Perperis, A.; Babali, D.; Verigou, E.; Gogos, H.; Hahalis, G.; Davlouros, P.; et al. Platelets from patients with myocardial infarction can activate T cells. Haematologica 2021, 106, 288–290. [Google Scholar] [CrossRef] [Green Version]
  46. Tanaka, K.A.; Key, N.S.; Levy, J.H. Blood coagulation: Hemostasis and thrombin regulation. Anesth. Analg. 2009, 108, 1433–1446. [Google Scholar] [CrossRef]
  47. Lippi, G.; Salvagno, G.L.; Ippolito, L.; Franchini, M.; Favaloro, E.J. Shortened activated partial thromboplastin time: Causes and management. Blood. Coagul. Fibrinolysis 2010, 21, 459–463. [Google Scholar] [CrossRef]
  48. Korte, W.; Clarke, S.; Lefkowitz, J.B. Short activated partial thromboplastin times are related to increased thrombin generation and an increased risk for thromboembolism. Am. J. Clin. Pathol. 2000, 113, 123–127. [Google Scholar] [CrossRef]
  49. Mina, A.; Favaloro, E.J.; Mohammed, S.; Koutts, J. A laboratory evaluation into the short activated partial thromboplastin time. Blood. Coagul. Fibrinolysis 2010, 21, 152–157. [Google Scholar] [CrossRef]
  50. Mina, A.; Favaloro, E.J.; Koutts, J. Relationship between short activated partial thromboplastin times, thrombin generation, procoagulant factors and procoagulant phospholipid activity. Blood. Coagul. Fibrinolysis 2012, 23, 203–207. [Google Scholar] [CrossRef]
  51. Tripodi, A.; Chantarangkul, V.; Martinelli, I.; Bucciarelli, P.; Mannucci, P.M. A shortened activated partial thromboplastin time is associated with the risk of venous thromboembolism. Blood 2004, 104, 3631–3634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Madi, A.M.; Greci, L.S.; Nawaz, H.; Katz, D.L. The activated partial thromboplastin time in early diagnosis of myocardial infarction. Blood. Coagul. Fibrinolysis 2001, 12, 495–499. [Google Scholar] [CrossRef] [PubMed]
  53. Lippi, G.; Franchini, M.; Targher, G.; Montagnana, M.; Salvagno, G.L.; Guidi, G.C.; Favaloro, E.J. Epidemiological association between fasting plasma glucose and shortened APTT. Clin. Biochem. 2009, 42, 118–120. [Google Scholar] [CrossRef] [PubMed]
  54. Zhao, Y.; Zhang, J.; Wu, J. Diabetes mellitus is associated with shortened activated partial thromboplastin time and increased fibrinogen values. PLoS ONE 2011, 6, e16470. [Google Scholar] [CrossRef] [Green Version]
  55. Garcia-Solares, J.; Donnez, J.; Donnez, O.; Dolmans, M.M. Pathogenesis of uterine adenomyosis: Invagination or metaplasia? Fertil. Steril. 2018, 109, 371–379. [Google Scholar] [CrossRef]
  56. Vannuccini, S.; Petraglia, F. Recent advances in understanding and managing adenomyosis. F1000Research 2019, 8. [Google Scholar] [CrossRef] [Green Version]
  57. Stratopoulou, C.A.; Donnez, J.; Dolmans, M.M. Origin and Pathogenic Mechanisms of Uterine Adenomyosis: What Is Known So Far. Reprod. Sci. 2021, 28, 2087–2097. [Google Scholar] [CrossRef]
  58. Vannuccini, S.; Tosti, C.; Carmona, F.; Huang, S.J.; Chapron, C.; Guo, S.W.; Petraglia, F. Pathogenesis of adenomyosis: An update on molecular mechanisms. Reprod. Biomed. Online 2017, 35, 592–601. [Google Scholar] [CrossRef] [Green Version]
  59. Gargett, C.E. Uterine stem cells: What is the evidence? Hum. Reprod. Update 2007, 13, 87–101. [Google Scholar] [CrossRef]
  60. Ferenczy, A. Pathophysiology of adenomyosis. Hum. Reprod. Update 1998, 4, 312–322. [Google Scholar] [CrossRef]
  61. Leyendecker, G.; Wildt, L.; Mall, G. The pathophysiology of endometriosis and adenomyosis: Tissue injury and repair. Arch. Gynecol. Obstet. 2009, 280, 529–538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Leyendecker, G.; Wildt, L. A new concept of endometriosis and adenomyosis: Tissue injury and repair (TIAR). Horm. Mol. Biol. Clin. Investig. 2011, 5, 125–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Leyendecker, G.; Bilgicyildirim, A.; Inacker, M.; Stalf, T.; Huppert, P.; Mall, G.; Bottcher, B.; Wildt, L. Adenomyosis and endometriosis. Re-visiting their association and further insights into the mechanisms of auto-traumatisation. An MRI study. Arch. Gynecol. Obstet. 2015, 291, 917–932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Leyendecker, G.; Wildt, L.; Laschke, M.W.; Mall, G. Archimetrosis: The evolution of a disease and its extant presentation: Pathogenesis and pathophysiology of archimetrosis (uterine adenomyosis and endometriosis). Arch. Gynecol. Obstet. 2022, 21, 1–20. [Google Scholar] [CrossRef]
  65. Guo, S.W. The Pathogenesis of Adenomyosis vis-a-vis Endometriosis. J. Clin. Med. 2020, 9, 485. [Google Scholar] [CrossRef] [Green Version]
  66. Wang, X.; Benagiano, G.; Liu, X.; Guo, S.W. Unveiling the Pathogenesis of Adenomyosis through Animal Models. J. Clin. Med. 2022, 11, 1744. [Google Scholar] [CrossRef]
  67. Curtis, K.M.; Hillis, S.D.; Marchbanks, P.A.; Peterson, H.B. Disruption of the endometrial-myometrial border during pregnancy as a risk factor for adenomyosis. Am. J. Obstet. Gynecol. 2002, 187, 543–544. [Google Scholar] [CrossRef]
  68. Levgur, M.; Abadi, M.A.; Tucker, A. Adenomyosis: Symptoms, histology, and pregnancy terminations. Obstet. Gynecol. 2000, 95, 688–691. [Google Scholar] [CrossRef]
  69. Panganamamula, U.R.; Harmanli, O.H.; Isik-Akbay, E.F.; Grotegut, C.A.; Dandolu, V.; Gaughan, J.P. Is prior uterine surgery a risk factor for adenomyosis? Obstet. Gynecol. 2004, 104, 1034–1038. [Google Scholar] [CrossRef]
  70. Parazzini, F.; Mais, V.; Cipriani, S.; Busacca, M.; Venturini, P.; On behalf of GISE. Determinants of adenomyosis in women who underwent hysterectomy for benign gynecological conditions: Results from a prospective multicentric study in Italy. Eur. J. Obstet. Gynecol. Reprod. Biol. 2009, 143, 103–106. [Google Scholar] [CrossRef]
  71. Taran, F.A.; Weaver, A.L.; Coddington, C.C.; Stewart, E.A. Understanding adenomyosis: A case control study. Fertil. Steril. 2010, 94, 1223–1228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Hao, M.; Liu, X.; Guo, S.W. Adenomyosis in mice resulting from mechanically or thermally induced endometrial-myometrial interface disruption and its possible prevention. Reprod. Biomed. Online 2020, 41, 925–942. [Google Scholar] [CrossRef] [PubMed]
  73. Hiraoka, T.; Hirota, Y.; Aikawa, S.; Iida, R.; Ishizawa, C.; Kaku, T.; Hirata, T.; Fukui, Y.; Akaeda, S.; Matsuo, M.; et al. Constant Activation of STAT3 Contributes to the Development of Adenomyosis in Females. Endocrinology 2022, 163, bqac044. [Google Scholar] [CrossRef] [PubMed]
  74. Elsherbini, M.; Koga, K.; Hiraoka, T.; Kumasawa, K.; Maki, E.; Satake, E.; Taguchi, A.; Makabe, T.; Takeuchi, A.; Izumi, G.; et al. Establishment of a novel mouse model of adenomyosis suitable for longitudinal and quantitative analysis and perinatal outcome studies. Sci. Rep. 2022, 12, 17515. [Google Scholar] [CrossRef] [PubMed]
  75. Taylor, C.T.; McElwain, J.C. Ancient atmospheres and the evolution of oxygen sensing via the hypoxia-inducible factor in metazoans. Physiology 2010, 25, 272–279. [Google Scholar] [CrossRef] [Green Version]
  76. Semenza, G.L. Hypoxia-inducible factors in physiology and medicine. Cell 2012, 148, 399–408. [Google Scholar] [CrossRef] [Green Version]
  77. Qi, Q.; Guo, S.-W.; Liu, X. Activated Platelets Induce Hypoxia-Inducible Factor-1α Expression Likely through Transforming Growth Factor-β1 in Human Endometrial Stromal Cells. Reprod. Dev. Med. 2019, 3, 69–76. [Google Scholar] [CrossRef]
  78. Qi, Q.; Liu, X.; Zhang, Q.; Guo, S.W. Platelets induce increased estrogen production through NF-kappaB and TGF-beta1 signaling pathways in endometriotic stromal cells. Sci. Rep. 2020, 10, 1281. [Google Scholar] [CrossRef] [Green Version]
  79. Chen, Y.J.; Li, H.Y.; Huang, C.H.; Twu, N.F.; Yen, M.S.; Wang, P.H.; Chou, T.Y.; Liu, Y.N.; Chao, K.C.; Yang, M.H. Oestrogen-induced epithelial-mesenchymal transition of endometrial epithelial cells contributes to the development of adenomyosis. J. Pathol. 2010, 222, 261–270. [Google Scholar] [CrossRef]
  80. Horng, H.C.; Chang, W.H.; Yeh, C.C.; Huang, B.S.; Chang, C.P.; Chen, Y.J.; Tsui, K.H.; Wang, P.H. Estrogen Effects on Wound Healing. Int. J. Mol. Sci. 2017, 18, 2325. [Google Scholar] [CrossRef]
  81. Wilkinson, H.N.; Hardman, M.J. The role of estrogen in cutaneous ageing and repair. Maturitas 2017, 103, 60–64. [Google Scholar] [CrossRef] [PubMed]
  82. Ashcroft, G.S.; Dodsworth, J.; van Boxtel, E.; Tarnuzzer, R.W.; Horan, M.A.; Schultz, G.S.; Ferguson, M.W. Estrogen accelerates cutaneous wound healing associated with an increase in TGF-beta1 levels. Nat. Med. 1997, 3, 1209–1215. [Google Scholar] [CrossRef]
  83. Ashcroft, G.S.; Mills, S.J.; Lei, K.; Gibbons, L.; Jeong, M.J.; Taniguchi, M.; Burow, M.; Horan, M.A.; Wahl, S.M.; Nakayama, T. Estrogen modulates cutaneous wound healing by downregulating macrophage migration inhibitory factor. J. Clin. Investig. 2003, 111, 1309–1318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Hardman, M.J.; Emmerson, E.; Campbell, L.; Ashcroft, G.S. Selective estrogen receptor modulators accelerate cutaneous wound healing in ovariectomized female mice. Endocrinology 2008, 149, 551–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Pepe, G.; Braga, D.; Renzi, T.A.; Villa, A.; Bolego, C.; D’Avila, F.; Barlassina, C.; Maggi, A.; Locati, M.; Vegeto, E. Self-renewal and phenotypic conversion are the main physiological responses of macrophages to the endogenous estrogen surge. Sci. Rep. 2017, 7, 44270. [Google Scholar] [CrossRef] [Green Version]
  86. Mukai, K.; Nakajima, Y.; Urai, T.; Komatsu, E.; Nasruddin; Sugama, J.; Nakatani, T. 17beta-Estradiol administration promotes delayed cutaneous wound healing in 40-week ovariectomised female mice. Int. Wound J. 2016, 13, 636–644. [Google Scholar] [CrossRef]
  87. Holt, J.C.; Niewiarowski, S. Biochemistry of alpha granule proteins. Semin. Hematol. 1985, 22, 151–163. [Google Scholar]
  88. Gear, A.R.; Camerini, D. Platelet chemokines and chemokine receptors: Linking hemostasis, inflammation, and host defense. Microcirculation 2003, 10, 335–350. [Google Scholar] [CrossRef]
  89. Wagner, D.D.; Burger, P.C. Platelets in inflammation and thrombosis. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 2131–2137. [Google Scholar] [CrossRef]
  90. Chen, G.; Luo, X.; Wang, W.; Wang, Y.; Zhu, F.; Wang, W. Interleukin-1beta Promotes Schwann Cells De-Differentiation in Wallerian Degeneration via the c-JUN/AP-1 Pathway. Front. Cell. Neurosci. 2019, 13, 304. [Google Scholar] [CrossRef]
  91. Wang, X.; Liu, X.; Guo, S.-W. Perioperative Suppression of Schwann Cell Dedifferentiation Reduces the Risk of Adenomyosis Resulting from Endometrial–Myometrial Interface Disruption in Mice. Biomedicines 2022, 10, 1218. [Google Scholar] [CrossRef] [PubMed]
  92. Carrarelli, P.; Yen, C.F.; Funghi, L.; Arcuri, F.; Tosti, C.; Bifulco, G.; Luddi, A.; Lee, C.L.; Petraglia, F. Expression of Inflammatory and Neurogenic Mediators in Adenomyosis. Reprod. Sci. 2017, 24, 369–375. [Google Scholar] [CrossRef] [PubMed]
  93. Li, B.; Chen, M.; Liu, X.; Guo, S.W. Constitutive and tumor necrosis factor-alpha-induced activation of nuclear factor-kappaB in adenomyosis and its inhibition by andrographolide. Fertil. Steril. 2013, 100, 568–577. [Google Scholar] [CrossRef] [PubMed]
  94. Nie, J.; Lu, Y.; Liu, X.; Guo, S.W. Immunoreactivity of progesterone receptor isoform B, nuclear factor kappaB, and IkappaBalpha in adenomyosis. Fertil. Steril. 2009, 92, 886–889. [Google Scholar] [CrossRef] [PubMed]
  95. Park, H.; Kim, S.H.; Cho, Y.M.; Ihm, H.J.; Oh, Y.S.; Hong, S.H.; Chae, H.D.; Kim, C.H.; Kang, B.M. Increased expression of nuclear factor kappa-B p65 subunit in adenomyosis. Obstet. Gynecol. Sci. 2016, 59, 123–129. [Google Scholar] [CrossRef]
  96. Propst, A.M.; Quade, B.J.; Nowak, R.A.; Stewart, E.A. Granulocyte macrophage colony-stimulating factor in adenomyosis and autologous endometrium. J. Soc. Gynecol. Investig. 2002, 9, 93–97. [Google Scholar] [CrossRef]
  97. Petaja, J. Inflammation and coagulation. An overview. Thromb. Res. 2011, 127, S34–S37. [Google Scholar] [CrossRef]
  98. Lipinski, S.; Bremer, L.; Lammers, T.; Thieme, F.; Schreiber, S.; Rosenstiel, P. Coagulation and inflammation. Molecular insights and diagnostic implications. Hamostaseologie 2011, 31, 94–102, 104. [Google Scholar] [CrossRef]
  99. Gartner, L.P.; Hiatt, J.L.; Strum, J.M. BRS. Cell Biology and Histology, 8th ed.; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2018. [Google Scholar]
  100. Semple, J.W.; Italiano, J.E., Jr.; Freedman, J. Platelets and the immune continuum. Nat. Rev. Immunol. 2011, 11, 264–274. [Google Scholar] [CrossRef]
  101. Vieira-de-Abreu, A.; Campbell, R.A.; Weyrich, A.S.; Zimmerman, G.A. Platelets: Versatile effector cells in hemostasis, inflammation, and the immune continuum. Semin. Immunopathol. 2012, 34, 5–30. [Google Scholar] [CrossRef] [Green Version]
  102. Sreeramkumar, V.; Adrover, J.M.; Ballesteros, I.; Cuartero, M.I.; Rossaint, J.; Bilbao, I.; Nacher, M.; Pitaval, C.; Radovanovic, I.; Fukui, Y.; et al. Neutrophils scan for activated platelets to initiate inflammation. Science 2014, 346, 1234–1238. [Google Scholar] [CrossRef] [PubMed]
  103. Boilard, E.; Nigrovic, P.A.; Larabee, K.; Watts, G.F.; Coblyn, J.S.; Weinblatt, M.E.; Massarotti, E.M.; Remold-O’Donnell, E.; Farndale, R.W.; Ware, J.; et al. Platelets amplify inflammation in arthritis via collagen-dependent microparticle production. Science 2010, 327, 580–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Lin, Y.J.; Lai, M.D.; Lei, H.Y.; Wing, L.Y. Neutrophils and macrophages promote angiogenesis in the early stage of endometriosis in a mouse model. Endocrinology 2006, 147, 1278–1286. [Google Scholar] [CrossRef] [Green Version]
  105. Hastings, J.M.; Jackson, K.S.; Mavrogianis, P.A.; Fazleabas, A.T. The estrogen early response gene FOS is altered in a baboon model of endometriosis. Biol. Reprod. 2006, 75, 176–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Sabbatini, M.; Magnelli, V.; Reno, F. NETosis in Wound Healing: When Enough Is Enough. Cells 2021, 10, 494. [Google Scholar] [CrossRef] [PubMed]
  107. Chrysanthopoulou, A.; Mitroulis, I.; Apostolidou, E.; Arelaki, S.; Mikroulis, D.; Konstantinidis, T.; Sivridis, E.; Koffa, M.; Giatromanolaki, A.; Boumpas, D.T.; et al. Neutrophil extracellular traps promote differentiation and function of fibroblasts. J. Pathol. 2014, 233, 294–307. [Google Scholar] [CrossRef]
  108. Munros, J.; Tassies, D.; Reverter, J.C.; Martin, L.; Perez, A.; Carmona, F.; Martinez-Zamora, M.A. Circulating Neutrophil Extracellular Traps Are Elevated in Patients With Deep Infiltrating Endometriosis. Reprod. Sci. 2019, 26, 70–76. [Google Scholar] [CrossRef] [PubMed]
  109. Roh, J.S.; Sohn, D.H. Damage-Associated Molecular Patterns in Inflammatory Diseases. Immune. Netw. 2018, 18, e27. [Google Scholar] [CrossRef]
  110. Bianchi, M.E. DAMPs, PAMPs and alarmins: All we need to know about danger. J. Leukoc. Biol. 2007, 81, 1–5. [Google Scholar] [CrossRef] [Green Version]
  111. Zhang, H.Y.; Wang, A.Q.; Zhu, S.; Yu, L.; Sun, J.F.; Xu, W.; Wang, X.L. Changes of coagulation function in patients with adenomyosis. Zhonghua Fu Chan Ke Za Zhi 2022, 57, 179–189. [Google Scholar] [CrossRef]
  112. Huang, P.; Lv, C.; Zhang, C.; Feng, H.; Li, C.; Zhang, H.; Zhao, X.; Li, M. Expression and significance of T-cell immunoglobulin mucin molecule 3 and its ligand galectin-9 in patients with adenomyosis. Gynecol. Endocrinol. 2020, 36, 605–610. [Google Scholar] [CrossRef] [PubMed]
  113. Liu, F.; Liu, L.; Zheng, J. Expression of annexin A2 in adenomyosis and dysmenorrhea. Arch. Gynecol. Obstet. 2019, 300, 711–716. [Google Scholar] [CrossRef] [Green Version]
  114. Liu, L.X.; Wu, Y.G.; Zheng, J. Increased annexin A2 and decreased beta-catenin in adenomyosis contribute to adenomyosis-associated dysmenorrhea. Histol. Histopathol. 2017, 32, 1333–1340. [Google Scholar] [CrossRef] [PubMed]
  115. Zhou, S.; Yi, T.; Liu, R.; Bian, C.; Qi, X.; He, X.; Wang, K.; Li, J.; Zhao, X.; Huang, C.; et al. Proteomics identification of annexin A2 as a key mediator in the metastasis and proangiogenesis of endometrial cells in human adenomyosis. Mol. Cell. Proteom. MCP 2012, 11, M112 017988. [Google Scholar] [CrossRef] [Green Version]
  116. Khan, K.N.; Kitajima, M.; Hiraki, K.; Fujishita, A.; Nakashima, M.; Masuzaki, H. Involvement of hepatocyte growth factor-induced epithelial-mesenchymal transition in human adenomyosis. Biol. Reprod. 2015, 92, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Matsuzaki, S.; Darcha, C. Epithelial to mesenchymal transition-like and mesenchymal to epithelial transition-like processes might be involved in the pathogenesis of pelvic endometriosis. Hum. Reprod. 2012, 27, 712–721. [Google Scholar] [CrossRef] [PubMed]
  118. Marconi, G.D.; Fonticoli, L.; Rajan, T.S.; Pierdomenico, S.D.; Trubiani, O.; Pizzicannella, J.; Diomede, F. Epithelial-Mesenchymal Transition (EMT): The Type-2 EMT in Wound Healing, Tissue Regeneration and Organ Fibrosis. Cells 2021, 10, 1587. [Google Scholar] [CrossRef]
  119. Oldenborg, P.A.; Zheleznyak, A.; Fang, Y.F.; Lagenaur, C.F.; Gresham, H.D.; Lindberg, F.P. Role of CD47 as a marker of self on red blood cells. Science 2000, 288, 2051–2054. [Google Scholar] [CrossRef]
  120. Wernig, G.; Chen, S.Y.; Cui, L.; Van Neste, C.; Tsai, J.M.; Kambham, N.; Vogel, H.; Natkunam, Y.; Gilliland, D.G.; Nolan, G.; et al. Unifying mechanism for different fibrotic diseases. Proc. Natl. Acad. Sci. USA 2017, 114, 4757–4762. [Google Scholar] [CrossRef] [Green Version]
  121. Chao, M.P.; Weissman, I.L.; Majeti, R. The CD47-SIRPalpha pathway in cancer immune evasion and potential therapeutic implications. Curr. Opin. Immunol. 2012, 24, 225–232. [Google Scholar] [CrossRef] [Green Version]
  122. Stein, E.V.; Miller, T.W.; Ivins-O’Keefe, K.; Kaur, S.; Roberts, D.D. Secreted Thrombospondin-1 Regulates Macrophage Interleukin-1beta Production and Activation through CD47. Sci. Rep. 2016, 6, 19684. [Google Scholar] [CrossRef]
  123. Murphy-Ullrich, J.E.; Suto, M.J. Thrombospondin-1 regulation of latent TGF-beta activation: A therapeutic target for fibrotic disease. Matrix. Biol. 2018, 68–69, 28–43. [Google Scholar] [CrossRef] [PubMed]
  124. Liu, Y.; Li, M.; Wei, C.; Tang, L.; Sheng, Y.; Liu, Y.; Li, D.; Ding, D.; Qiu, J.; Zhu, X. TSP1-CD47-SIRPalpha signaling facilitates the development of endometriosis by mediating the survival of ectopic endometrium. Am. J. Reprod. Immunol. 2020, 83, e13236. [Google Scholar] [CrossRef] [PubMed]
  125. Li, J.; Yan, S.; Li, Q.; Huang, Y.; Ji, M.; Jiao, X.; Yuan, M.; Wang, G. Macrophage-associated immune checkpoint CD47 blocking ameliorates endometriosis. Mol. Hum. Reprod. 2022, 28, gaac010. [Google Scholar] [CrossRef]
  126. Shazand, K.; Baban, S.; Prive, C.; Malette, B.; Croteau, P.; Lagace, M.; Racine, J.B.; Hugo, P. FOXO1 and c-jun transcription factors mRNA are modulated in endometriosis. Mol. Hum. Reprod. 2004, 10, 871–877. [Google Scholar] [CrossRef] [Green Version]
  127. Beste, M.T.; Pfaffle-Doyle, N.; Prentice, E.A.; Morris, S.N.; Lauffenburger, D.A.; Isaacson, K.B.; Griffith, L.G. Molecular network analysis of endometriosis reveals a role for c-Jun-regulated macrophage activation. Sci. Transl. Med. 2014, 6, 222ra216. [Google Scholar] [CrossRef] [Green Version]
  128. Cinar, O.; Seval, Y.; Uz, Y.H.; Cakmak, H.; Ulukus, M.; Kayisli, U.A.; Arici, A. Differential regulation of Akt phosphorylation in endometriosis. Reprod. Biomed. Online 2009, 19, 864–871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Zhang, H.; Zhao, X.; Liu, S.; Li, J.; Wen, Z.; Li, M. 17betaE2 promotes cell proliferation in endometriosis by decreasing PTEN via NFkappaB-dependent pathway. Mol. Cell Endocrinol. 2010, 317, 31–43. [Google Scholar] [CrossRef]
  130. Xu, X.Y.; Zhang, J.; Qi, Y.H.; Kong, M.; Liu, S.A.; Hu, J.J. Linc-ROR promotes endometrial cell proliferation by activating the PI3K-Akt pathway. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 2218–2225. [Google Scholar] [CrossRef]
  131. Zhang, H.; Liu, J.; Zhao, X.; Liang, B.; Wen, Z.; Li, M. Loss of PP2A and PTEN immunoexpression coexists with survivin overexpression in adenomyosis. Reprod. Biol. 2014, 14, 200–205. [Google Scholar] [CrossRef]
  132. Yun, B.H.; Chon, S.J.; Choi, Y.S.; Cho, S.; Lee, B.S.; Seo, S.K. Pathophysiology of Endometriosis: Role of High Mobility Group Box-1 and Toll-Like Receptor 4 Developing Inflammation in Endometrium. PLoS ONE 2016, 11, e0148165. [Google Scholar] [CrossRef] [PubMed]
  133. Cao, Y.; Liu, X.; Guo, S.W. Plasma High Mobility Group Box 1 (HMGB1), Osteopontin (OPN), and Hyaluronic Acid (HA) as Admissible Biomarkers for Endometriosis. Sci. Rep. 2019, 9, 9272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Liu, X.; Shen, M.; Qi, Q.; Zhang, H.; Guo, S.W. Corroborating evidence for platelet-induced epithelial-mesenchymal transition and fibroblast-to-myofibroblast transdifferentiation in the development of adenomyosis. Hum. Reprod. 2016, 31, 734–749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Shen, M.; Liu, X.; Zhang, H.; Guo, S.W. Transforming growth factor beta1 signaling coincides with epithelial-mesenchymal transition and fibroblast-to-myofibroblast transdifferentiation in the development of adenomyosis in mice. Hum. Reprod. 2016, 31, 355–369. [Google Scholar]
  136. Zhang, Q.; Duan, J.; Liu, X.; Guo, S.W. Platelets drive smooth muscle metaplasia and fibrogenesis in endometriosis through epithelial-mesenchymal transition and fibroblast-to-myofibroblast transdifferentiation. Mol. Cell. Endocrinol. 2016, 428, 1–16. [Google Scholar] [CrossRef] [PubMed]
  137. Zhang, Q.; Duan, J.; Olson, M.; Fazleabas, A.; Guo, S.W. Cellular Changes Consistent With Epithelial-Mesenchymal Transition and Fibroblast-to-Myofibroblast Transdifferentiation in the Progression of Experimental Endometriosis in Baboons. Reprod. Sci. 2016, 23, 1409–1421. [Google Scholar] [CrossRef] [Green Version]
  138. Zhang, Q.; Ding, D.; Liu, X.; Guo, S.W. Activated Platelets Induce Estrogen Receptor beta Expression in Endometriotic Stromal Cells. Gynecol. Obstet. Investig. 2015, 80, 187–190. [Google Scholar] [CrossRef]
  139. Merlo, S.; Frasca, G.; Canonico, P.L.; Sortino, M.A. Differential involvement of estrogen receptor alpha and estrogen receptor beta in the healing promoting effect of estrogen in human keratinocytes. J. Endocrinol. 2009, 200, 189–197. [Google Scholar] [CrossRef] [Green Version]
  140. Campbell, L.; Emmerson, E.; Davies, F.; Gilliver, S.C.; Krust, A.; Chambon, P.; Ashcroft, G.S.; Hardman, M.J. Estrogen promotes cutaneous wound healing via estrogen receptor beta independent of its antiinflammatory activities. J. Exp. Med. 2010, 207, 1825–1833. [Google Scholar] [CrossRef] [Green Version]
  141. Mehasseb, M.K.; Panchal, R.; Taylor, A.H.; Brown, L.; Bell, S.C.; Habiba, M. Estrogen and progesterone receptor isoform distribution through the menstrual cycle in uteri with and without adenomyosis. Fertil. Steril. 2011, 95, 2228–2235.e2221. [Google Scholar] [CrossRef]
  142. Brandenberger, A.W.; Lebovic, D.I.; Tee, M.K.; Ryan, I.P.; Tseng, J.F.; Jaffe, R.B.; Taylor, R.N. Oestrogen receptor (ER)-alpha and ER-beta isoforms in normal endometrial and endometriosis-derived stromal cells. Mol. Hum. Reprod. 1999, 5, 651–655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Fujimoto, J.; Hirose, R.; Sakaguchi, H.; Tamaya, T. Expression of oestrogen receptor-alpha and -beta in ovarian endometriomata. Mol. Hum. Reprod. 1999, 5, 742–747. [Google Scholar] [CrossRef] [PubMed]
  144. Nurden, A.T. Platelets, inflammation and tissue regeneration. Thromb. Haemost. 2011, 105, S13–S33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Savagner, P. Epithelial-mesenchymal transitions: From cell plasticity to concept elasticity. Curr. Top. Dev. Biol. 2015, 112, 273–300. [Google Scholar] [CrossRef] [PubMed]
  146. Hinz, B. The role of myofibroblasts in wound healing. Curr. Res. Transl. Med. 2016, 64, 171–177. [Google Scholar] [CrossRef]
  147. Henderson, N.C.; Rieder, F.; Wynn, T.A. Fibrosis: From mechanisms to medicines. Nature 2020, 587, 555–566. [Google Scholar] [CrossRef]
  148. Mukai, K.; Urai, T.; Asano, K.; Nakajima, Y.; Nakatani, T. Evaluation of Effects of Topical Estradiol Benzoate Application on Cutaneous Wound Healing in Ovariectomized Female Mice. PLoS ONE 2016, 11, e0163560. [Google Scholar] [CrossRef] [Green Version]
  149. Bulun, S.E.; Lin, Z.; Imir, G.; Amin, S.; Demura, M.; Yilmaz, B.; Martin, R.; Utsunomiya, H.; Thung, S.; Gurates, B.; et al. Regulation of aromatase expression in estrogen-responsive breast and uterine disease: From bench to treatment. Pharmacol. Rev. 2005, 57, 359–383. [Google Scholar] [CrossRef] [Green Version]
  150. Kitawaki, J.; Noguchi, T.; Amatsu, T.; Maeda, K.; Tsukamoto, K.; Yamamoto, T.; Fushiki, S.; Osawa, Y.; Honjo, H. Expression of aromatase cytochrome P450 protein and messenger ribonucleic acid in human endometriotic and adenomyotic tissues but not in normal endometrium. Biol. Reprod. 1997, 57, 514–519. [Google Scholar] [CrossRef] [Green Version]
  151. Ding, D.; Liu, X.; Duan, J.; Guo, S.W. Platelets are an unindicted culprit in the development of endometriosis: Clinical and experimental evidence. Hum. Reprod. 2015, 30, 812–832. [Google Scholar] [CrossRef] [Green Version]
  152. Bulun, S.E. Endometriosis. N. Engl. J. Med. 2009, 360, 268–279. [Google Scholar] [CrossRef] [PubMed]
  153. Han, S.J.; Jung, S.Y.; Wu, S.P.; Hawkins, S.M.; Park, M.J.; Kyo, S.; Qin, J.; Lydon, J.P.; Tsai, S.Y.; Tsai, M.J.; et al. Estrogen Receptor beta Modulates Apoptosis Complexes and the Inflammasome to Drive the Pathogenesis of Endometriosis. Cell 2015, 163, 960–974. [Google Scholar] [CrossRef] [PubMed]
  154. Sztachelska, M.; Ponikwicka-Tyszko, D.; Martinez-Rodrigo, L.; Bernaczyk, P.; Palak, E.; Polchlopek, W.; Bielawski, T.; Wolczynski, S. Functional Implications of Estrogen and Progesterone Receptors Expression in Adenomyosis, Potential Targets for Endocrinological Therapy. J. Clin. Med. 2022, 11, 4407. [Google Scholar] [CrossRef] [PubMed]
  155. Giudice, L.C.; Kao, L.C. Endometriosis. Lancet 2004, 364, 1789–1799. [Google Scholar] [CrossRef] [PubMed]
  156. Zhai, J.; Vannuccini, S.; Petraglia, F.; Giudice, L.C. Adenomyosis: Mechanisms and Pathogenesis. Semin. Reprod. Med. 2020, 38, 129–143. [Google Scholar] [CrossRef] [PubMed]
  157. Prunotto, M.; Budd, D.C.; Gabbiani, G.; Meier, M.; Formentini, I.; Hartmann, G.; Pomposiello, S.; Moll, S. Epithelial-mesenchymal crosstalk alteration in kidney fibrosis. J. Pathol. 2012, 228, 131–147. [Google Scholar] [CrossRef]
  158. Yang, Y.M.; Yang, W.X. Epithelial-to-mesenchymal transition in the development of endometriosis. Oncotarget 2017, 8, 41679–41689. [Google Scholar] [CrossRef] [Green Version]
  159. Konrad, L.; Dietze, R.; Riaz, M.A.; Scheiner-Bobis, G.; Behnke, J.; Horne, F.; Hoerscher, A.; Reising, C.; Meinhold-Heerlein, I. Epithelial-Mesenchymal Transition in Endometriosis-When Does It Happen? J. Clin. Med. 2020, 9, 1915. [Google Scholar] [CrossRef]
  160. Guo, S.W. Cracking the enigma of adenomyosis: An update on its pathogenesis and pathophysiology. Reproduction 2022, 164, R101–R121. [Google Scholar] [CrossRef]
  161. Darby, I.; Skalli, O.; Gabbiani, G. Alpha-smooth muscle actin is transiently expressed by myofibroblasts during experimental wound healing. Lab. Investig. 1990, 63, 21–29. [Google Scholar]
  162. Desmouliere, A.; Geinoz, A.; Gabbiani, F.; Gabbiani, G. Transforming growth factor-beta 1 induces alpha-smooth muscle actin expression in granulation tissue myofibroblasts and in quiescent and growing cultured fibroblasts. J. Cell. Biol. 1993, 122, 103–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Liu, X.; Yan, D.; Guo, S.W. Sensory nerve-derived neuropeptides accelerate the development and fibrogenesis of endometriosis. Hum. Reprod. 2019, 34, 452–468. [Google Scholar] [CrossRef] [PubMed]
  164. Yan, D.; Liu, X.; Guo, S.W. Neuropeptides Substance P and Calcitonin Gene Related Peptide Accelerate the Development and Fibrogenesis of Endometriosis. Sci. Rep. 2019, 9, 2698. [Google Scholar] [CrossRef] [PubMed]
  165. Guo, L.; Mi, J.W.; Zhang, H.C.; Gao, J.; Zhang, S.; Li, L.X.; Wu, M.Y.; Wang, J.M.; Huang, H. Endothelial-mesenchymal transition as a novel mechanism for generating myofibroblasts during wound healing and scarring. J. Cosmet. Dermatol. 2022. [Google Scholar] [CrossRef] [PubMed]
  166. Yan, D.; Liu, X.; Xu, H.; Guo, S.W. Platelets induce endothelial-mesenchymal transition and subsequent fibrogenesis in endometriosis. Reprod. Biomed. Online 2020, 41, 500–517. [Google Scholar] [CrossRef]
  167. Darby, I.A.; Hewitson, T.D. Fibroblast differentiation in wound healing and fibrosis. Int. Rev. Cytol. 2007, 257, 143–179. [Google Scholar] [CrossRef]
  168. Duan, J.; Liu, X.; Wang, H.; Guo, S.W. The M2a macrophage subset may be critically involved in the fibrogenesis of endometriosis in mice. Reprod. Biomed. Online 2018, 37, 254–268. [Google Scholar] [CrossRef]
  169. Xiao, F.; Liu, X.; Guo, S.W. Platelets and Regulatory T Cells May Induce a Type 2 Immunity That Is Conducive to the Progression and Fibrogenesis of Endometriosis. Front. Immunol. 2020, 11, 610963. [Google Scholar] [CrossRef]
  170. Xiao, F.; Liu, X.; Guo, S.W. Interleukin-33 Derived from Endometriotic Lesions Promotes Fibrogenesis through Inducing the Production of Profibrotic Cytokines by Regulatory T Cells. Biomedicines 2022, 10, 2893. [Google Scholar] [CrossRef]
  171. Matsuzaki, S.; Darcha, C. Involvement of the Wnt/beta-catenin signaling pathway in the cellular and molecular mechanisms of fibrosis in endometriosis. PLoS ONE 2013, 8, e76808. [Google Scholar] [CrossRef]
  172. Marcellin, L.; Santulli, P.; Chouzenoux, S.; Cerles, O.; Nicco, C.; Dousset, B.; Pallardy, M.; Kerdine-Romer, S.; Just, P.A.; Chapron, C.; et al. Alteration of Nrf2 and Glutamate Cysteine Ligase expression contribute to lesions growth and fibrogenesis in ectopic endometriosis. Free. Radic. Biol. Med. 2017, 110, 1–10. [Google Scholar] [CrossRef]
  173. Cai, X.; Shen, M.; Liu, X.; Guo, S.W. Reduced Expression of Eukaryotic Translation Initiation Factor 3 Subunit e and Its Possible Involvement in the Epithelial-Mesenchymal Transition in Endometriosis. Reprod. Sci. 2018, 25, 102–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Cai, X.; Shen, M.; Liu, X.; Nie, J. The Possible Role of Eukaryotic Translation Initiation Factor 3 Subunit e (eIF3e) in the Epithelial-Mesenchymal Transition in Adenomyosis. Reprod. Sci. 2019, 26, 377–385. [Google Scholar] [CrossRef]
  175. Wang, S.; Li, B.; Duan, H.; Wang, Y.; Shen, X.; Dong, Q. Abnormal expression of connective tissue growth factor and its correlation with fibrogenesis in adenomyosis. Reprod. Biomed. Online 2021, 42, 651–660. [Google Scholar] [CrossRef] [PubMed]
  176. Mehasseb, M.K.; Bell, S.C.; Pringle, J.H.; Habiba, M.A. Uterine adenomyosis is associated with ultrastructural features of altered contractility in the inner myometrium. Fertil. Steril. 2010, 93, 2130–2136. [Google Scholar] [CrossRef] [PubMed]
  177. Streuli, I.; Santulli, P.; Chouzenoux, S.; Chapron, C.; Batteux, F. Activation of the MAPK/ERK Cell-Signaling Pathway in Uterine Smooth Muscle Cells of Women With Adenomyosis. Reprod. Sci. 2015, 22, 1549–1560. [Google Scholar] [CrossRef]
  178. Wang, S.; Li, B.; Shen, X.; Duan, H.; Guo, Z.; Li, X.; Sun, F. The cannabinoid receptor CB1 affects the proliferation and apoptosis of adenomyotic human uterine smooth muscle cells of the junctional zone: A mechanism study. Reprod. Biol. Endocrinol. 2021, 19, 16. [Google Scholar] [CrossRef]
  179. Guo, S.W.; Mao, X.; Ma, Q.; Liu, X. Dysmenorrhea and its severity are associated with increased uterine contractility and overexpression of oxytocin receptor (OTR) in women with symptomatic adenomyosis. Fertil. Steril. 2013, 99, 231–240. [Google Scholar] [CrossRef]
  180. Mao, X.; Wang, Y.; Carter, A.V.; Zhen, X.; Guo, S.W. 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. Reprod. Sci. 2011, 18, 1025–1037. [Google Scholar] [CrossRef]
  181. Liu, X.; Nie, J.; Guo, S.W. Elevated immunoreactivity to tissue factor and its association with dysmenorrhea severity and the amount of menses in adenomyosis. Hum. Reprod. 2011, 26, 337–345. [Google Scholar] [CrossRef] [Green Version]
  182. Runic, R.; Schatz, F.; Wan, L.; Demopoulos, R.; Krikun, G.; Lockwood, C.J. Effects of norplant on endometrial tissue factor expression and blood vessel structure. J. Clin. Endocrinol. Metab. 2000, 85, 3853–3859. [Google Scholar] [CrossRef] [PubMed]
  183. Krikun, G.; Lockwood, C.J.; Paidas, M.J. Tissue factor and the endometrium: From physiology to pathology. Thromb. Res. 2009, 124, 393–396. [Google Scholar] [CrossRef]
  184. Yang, B.; Gu, N.; Shi, S.; Zhang, C.; Chen, L.; Ouyang, J.; Lin, Y.; Sun, F.; Xu, H. Immunoreactivity of Plasminogen Activator Inhibitor 1 and Its Correlation with Dysmenorrhea and Lesional Fibrosis in Adenomyosis. Reprod. Sci. 2021, 28, 2378–2386. [Google Scholar] [CrossRef] [PubMed]
  185. Osuga, Y.; Hirota, Y.; Yoshino, O.; Hirata, T.; Koga, K.; Taketani, Y. Proteinase-activated receptors in the endometrium and endometriosis. Front. Biosci. 2012, 4, 1201–1212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Lin, M.; Weng, H.; Wang, X.; Zhou, B.; Yu, P.; Wang, Y. The role of tissue factor and protease-activated receptor 2 in endometriosis. Am. J. Reprod. Immunol. 2012, 68, 251–257. [Google Scholar] [CrossRef]
  187. Hirota, Y.; Osuga, Y.; Hirata, T.; Yoshino, O.; Koga, K.; Harada, M.; Morimoto, C.; Nose, E.; Yano, T.; Tsutsumi, O.; et al. Possible involvement of thrombin/protease-activated receptor 1 system in the pathogenesis of endometriosis. J. Clin. Endocrinol. Metab. 2005, 90, 3673–3679. [Google Scholar] [CrossRef] [Green Version]
  188. Hirota, Y.; Osuga, Y.; Hirata, T.; Harada, M.; Morimoto, C.; Yoshino, O.; Koga, K.; Yano, T.; Tsutsumi, O.; Taketani, Y. Activation of protease-activated receptor 2 stimulates proliferation and interleukin (IL)-6 and IL-8 secretion of endometriotic stromal cells. Hum. Reprod. 2005, 20, 3547–3553. [Google Scholar] [CrossRef] [Green Version]
  189. Matsuzaki, S.; Canis, M.; Darcha, C.; Fukaya, T.; Yajima, A.; Bruhat, M.A. Increased mast cell density in peritoneal endometriosis compared with eutopic endometrium with endometriosis. Am. J. Reprod. Immunol. 1998, 40, 291–294. [Google Scholar] [CrossRef]
  190. Anaf, V.; Chapron, C.; El Nakadi, I.; De Moor, V.; Simonart, T.; Noel, J.C. Pain, mast cells, and nerves in peritoneal, ovarian, and deep infiltrating endometriosis. Fertil. Steril. 2006, 86, 1336–1343. [Google Scholar] [CrossRef]
  191. Saito, A.; Osuga, Y.; Yoshino, O.; Takamura, M.; Hirata, T.; Hirota, Y.; Koga, K.; Harada, M.; Takemura, Y.; Yano, T.; et al. TGF-beta1 induces proteinase-activated receptor 2 (PAR2) expression in endometriotic stromal cells and stimulates PAR2 activation-induced secretion of IL-6. Hum. Reprod. 2011, 26, 1892–1898. [Google Scholar] [CrossRef] [Green Version]
  192. Assoian, R.K.; Komoriya, A.; Meyers, C.A.; Miller, D.M.; Sporn, M.B. Transforming growth factor-beta in human platelets. Identification of a major storage site, purification, and characterization. J. Biol. Chem. 1983, 258, 7155–7160. [Google Scholar] [CrossRef] [PubMed]
  193. Thiery, J.P.; Acloque, H.; Huang, R.Y.; Nieto, M.A. Epithelial-mesenchymal transitions in development and disease. Cell 2009, 139, 871–890. [Google Scholar] [CrossRef]
  194. Gabbiani, G. The myofibroblast in wound healing and fibrocontractive diseases. J. Pathol. 2003, 200, 500–503. [Google Scholar] [CrossRef]
  195. Desmouliere, A.; Chaponnier, C.; Gabbiani, G. Tissue repair, contraction, and the myofibroblast. Wound. Repair. Regen. 2005, 13, 7–12. [Google Scholar] [CrossRef] [PubMed]
  196. Biernacka, A.; Dobaczewski, M.; Frangogiannis, N.G. TGF-beta signaling in fibrosis. Growth. Factors 2011, 29, 196–202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Yan, D.; Liu, X.; Xu, H.; Guo, S.W. Mesothelial Cells Participate in Endometriosis Fibrogenesis Through Platelet-Induced Mesothelial-Mesenchymal Transition. J. Clin. Endocrinol. Metab. 2020, 105, e4124–e4127. [Google Scholar] [CrossRef] [PubMed]
  198. Urata, Y.; Osuga, Y.; Izumi, G.; Takamura, M.; Koga, K.; Nagai, M.; Harada, M.; Hirata, T.; Hirota, Y.; Yoshino, O.; et al. Interleukin-1beta stimulates the secretion of thymic stromal lymphopoietin (TSLP) from endometrioma stromal cells: Possible involvement of TSLP in endometriosis. Hum. Reprod. 2012, 27, 3028–3035. [Google Scholar] [CrossRef] [Green Version]
  199. Truchetet, M.E.; Demoures, B.; Eduardo Guimaraes, J.; Bertrand, A.; Laurent, P.; Jolivel, V.; Douchet, I.; Jacquemin, C.; Khoryati, L.; Duffau, P.; et al. Platelets Induce Thymic Stromal Lymphopoietin Production by Endothelial Cells: Contribution to Fibrosis in Human Systemic Sclerosis. Arthritis Rheumatol. 2016, 68, 2784–2794. [Google Scholar] [CrossRef]
  200. Eidukaite, A.; Siaurys, A.; Tamosiunas, V. Aberrant expression of CD95 and CD69 molecules among CD56 cells in women with endometriosis. Am. J. Reprod. Immunol. 2006, 55, 276–281. [Google Scholar] [CrossRef]
  201. Guo, M.; Bafligil, C.; Tapmeier, T.; Hubbard, C.; Manek, S.; Shang, C.; Martinez, F.O.; Schmidt, N.; Obendorf, M.; Hess-Stumpp, H.; et al. Mass cytometry analysis reveals a distinct immune environment in peritoneal fluid in endometriosis: A characterisation study. BMC. Med. 2020, 18, 3. [Google Scholar] [CrossRef] [Green Version]
  202. Chegini, N.; Rossi, M.J.; Masterson, B.J. Platelet-derived growth factor (PDGF), epidermal growth factor (EGF), and EGF and PDGF beta-receptors in human endometrial tissue: Localization and in vitro action. Endocrinology 1992, 130, 2373–2385. [Google Scholar] [CrossRef] [PubMed]
  203. Overton, C.; Fernandez-Shaw, S.; Hicks, B.; Barlow, D.; Starkey, P. Peritoneal fluid cytokines and the relationship with endometriosis and pain. Hum. Reprod. 1996, 11, 380–386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Kalu, E.; Sumar, N.; Giannopoulos, T.; Patel, P.; Croucher, C.; Sherriff, E.; Bansal, A. Cytokine profiles in serum and peritoneal fluid from infertile women with and without endometriosis. J. Obstet. Gynaecol. Res. 2007, 33, 490–495. [Google Scholar] [CrossRef] [PubMed]
  205. Surrey, E.S.; Halme, J. Effect of platelet-derived growth factor on endometrial stromal cell proliferation in vitro: A model for endometriosis? Fertil. Steril. 1991, 56, 672–679. [Google Scholar] [CrossRef]
  206. Matsumoto, H.; Nasu, K.; Nishida, M.; Ito, H.; Bing, S.; Miyakawa, I. Regulation of proliferation, motility, and contractility of human endometrial stromal cells by platelet-derived growth factor. J. Clin. Endocrinol. Metab. 2005, 90, 3560–3567. [Google Scholar] [CrossRef]
  207. Gentilini, D.; Busacca, M.; Di Francesco, S.; Vignali, M.; Vigano, P.; Di Blasio, A.M. PI3K/Akt and ERK1/2 signalling pathways are involved in endometrial cell migration induced by 17beta-estradiol and growth factors. Mol. Hum. Reprod. 2007, 13, 317–322. [Google Scholar] [CrossRef]
  208. Bonner, J.C. Regulation of PDGF and its receptors in fibrotic diseases. Cytokine. Growth. Factor. Rev. 2004, 15, 255–273. [Google Scholar] [CrossRef]
  209. Abdollahi, A.; Li, M.; Ping, G.; Plathow, C.; Domhan, S.; Kiessling, F.; Lee, L.B.; McMahon, G.; Grone, H.J.; Lipson, K.E.; et al. Inhibition of platelet-derived growth factor signaling attenuates pulmonary fibrosis. J. Exp. Med. 2005, 201, 925–935. [Google Scholar] [CrossRef] [Green Version]
  210. Druce, M.; Rockall, A.; Grossman, A.B. Fibrosis and carcinoid syndrome: From causation to future therapy. Nat. Rev. Endocrinol. 2009, 5, 276–283. [Google Scholar] [CrossRef]
  211. Dees, C.; Akhmetshina, A.; Zerr, P.; Reich, N.; Palumbo, K.; Horn, A.; Jungel, A.; Beyer, C.; Kronke, G.; Zwerina, J.; et al. Platelet-derived serotonin links vascular disease and tissue fibrosis. J. Exp. Med. 2011, 208, 961–972. [Google Scholar] [CrossRef] [Green Version]
  212. Zhang, J.; Cui, R.; Feng, Y.; Gao, W.; Bi, J.; Li, Z.; Liu, C. Serotonin Exhibits Accelerated Bleomycin-Induced Pulmonary Fibrosis through TPH1 Knockout Mouse Experiments. Mediators. Inflamm. 2018, 2018, 7967868. [Google Scholar] [CrossRef] [PubMed]
  213. Chaturvedi, S.; Misra, D.P.; Prasad, N.; Rastogi, K.; Singh, H.; Rai, M.K.; Agarwal, V. 5-HT2 and 5-HT2B antagonists attenuate pro-fibrotic phenotype in human adult dermal fibroblasts by blocking TGF-beta1 induced non-canonical signaling pathways including STAT3: Implications for fibrotic diseases like scleroderma. Int. J. Rheum. Dis. 2018, 21, 2128–2138. [Google Scholar] [CrossRef] [PubMed]
  214. Hutcheson, J.D.; Ryzhova, L.M.; Setola, V.; Merryman, W.D. 5-HT(2B) antagonism arrests non-canonical TGF-beta1-induced valvular myofibroblast differentiation. J. Mol. Cell. Cardiol. 2012, 53, 707–714. [Google Scholar] [CrossRef] [PubMed]
  215. Lofdahl, A.; Rydell-Tormanen, K.; Muller, C.; Martina Holst, C.; Thiman, L.; Ekstrom, G.; Wenglen, C.; Larsson-Callerfelt, A.K.; Westergren-Thorsson, G. 5-HT2B receptor antagonists attenuate myofibroblast differentiation and subsequent fibrotic responses in vitro and in vivo. Physiol. Rep. 2016, 4, e12873. [Google Scholar] [CrossRef]
  216. Mosele, S.; Stratopoulou, C.A.; Camboni, A.; Donnez, J.; Dolmans, M.M. Investigation of the role of platelets in the aetiopathogenesis of adenomyosis. Reprod. Biomed. Online 2021, 42, 826–834. [Google Scholar] [CrossRef]
  217. Liu, X.; Zhang, Q.; Guo, S.W. Histological and Immunohistochemical Characterization of the Similarity and Difference Between Ovarian Endometriomas and Deep Infiltrating Endometriosis. Reprod. Sci. 2018, 25, 329–340. [Google Scholar] [CrossRef]
  218. Lin, q.; Li, T.; Ding, S.; Yu, Q.; Zhang, X. Anemia-Associated Platelets and Plasma Prothrombin Time Increase in Patients with Adenomyosis. J. Clin. Med. 2022, 11, 4382. [Google Scholar] [CrossRef]
  219. Guo, S.W.; Du, Y.; Liu, X. Endometriosis-Derived Stromal Cells Secrete Thrombin and Thromboxane A2, Inducing Platelet Activation. Reprod. Sci. 2016, 23, 1044–1052. [Google Scholar] [CrossRef]
  220. Caligiuri, M.A. Human natural killer cells. Blood 2008, 112, 461–469. [Google Scholar] [CrossRef]
  221. Smyth, M.J.; Hayakawa, Y.; Takeda, K.; Yagita, H. New aspects of natural-killer-cell surveillance and therapy of cancer. Nat. Rev. Cancer. 2002, 2, 850–861. [Google Scholar] [CrossRef]
  222. Lanier, L.L. Natural killer cell receptor signaling. Curr. Opin. Immunol. 2003, 15, 308–314. [Google Scholar] [CrossRef] [PubMed]
  223. Cheent, K.; Khakoo, S.I. Natural killer cells: Integrating diversity with function. Immunology 2009, 126, 449–457. [Google Scholar] [CrossRef] [PubMed]
  224. Bourdon, M.; Santulli, P.; Jeljeli, M.; Vannuccini, S.; Marcellin, L.; Doridot, L.; Petraglia, F.; Batteux, F.; Chapron, C. Immunological changes associated with adenomyosis: A systematic review. Hum. Reprod. Update 2021, 27, 108–129. [Google Scholar] [CrossRef]
  225. Du, Y.; Liu, X.; Guo, S.W. Platelets impair natural killer cell reactivity and function in endometriosis through multiple mechanisms. Hum. Reprod. 2017, 32, 794–810. [Google Scholar] [CrossRef] [Green Version]
  226. Guo, S.W.; Du, Y.; Liu, X. Platelet-derived TGF-beta1 mediates the down-modulation of NKG2D expression and may be responsible for impaired natural killer (NK) cytotoxicity in women with endometriosis. Hum. Reprod. 2016, 31, 1462–1474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Critchley, H.O.; Kelly, R.W.; Baird, D.T.; Brenner, R.M. Regulation of human endometrial function: Mechanisms relevant to uterine bleeding. Reprod. Biol. Endocrinol. 2006, 4, S5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Munro, M.G.; Critchley, H.O.D.; Fraser, I.S.; Committee, F.M.D. The two FIGO systems for normal and abnormal uterine bleeding symptoms and classification of causes of abnormal uterine bleeding in the reproductive years: 2018 revisions. Int. J. Gynaecol. Obstet. 2018, 143, 393–408. [Google Scholar] [CrossRef] [Green Version]
  229. Zhu, B.; Chen, Y.; Shen, X.; Liu, X.; Guo, S.W. Anti-platelet therapy holds promises in treating adenomyosis: Experimental evidence. Reprod. Biol. Endocrinol. 2016, 14, 66. [Google Scholar] [CrossRef] [Green Version]
  230. Guo, S.W.; Ding, D.; Geng, J.G.; Wang, L.; Liu, X. P-selectin as a potential therapeutic target for endometriosis. Fertil. Steril. 2015, 103, 990–1000.e1008. [Google Scholar] [CrossRef]
  231. Guo, S.W.; Ding, D.; Liu, X. Anti-platelet therapy is efficacious in treating endometriosis induced in mouse. Reprod. Biomed. Online 2016, 33, 484–499. [Google Scholar] [CrossRef] [Green Version]
  232. Sheth, S.S.; Ray, S.S. Severe adenomyosis and CA125. J. Obstet. Gynaecol. 2014, 34, 79–81. [Google Scholar] [CrossRef] [PubMed]
  233. Jovin, T.G.; Boosupalli, V.; Zivkovic, S.A.; Wechsler, L.R.; Gebel, J.M. High titers of CA-125 may be associated with recurrent ischemic strokes in patients with cancer. Neurology 2005, 64, 1944–1945. [Google Scholar] [CrossRef]
  234. Tas, F.; Kilic, L.; Bilgin, E.; Keskin, S.; Sen, F.; Ciftci, R.; Yildiz, I.; Yasasever, V. Clinical and prognostic significance of coagulation assays in advanced epithelial ovarian cancer. Int. J. Gynecol. Cancer 2013, 23, 276–281. [Google Scholar] [CrossRef] [PubMed]
  235. Critchley, H.O.D.; Maybin, J.A.; Armstrong, G.M.; Williams, A.R.W. Physiology of the Endometrium and Regulation of Menstruation. Physiol. Rev. 2020, 100, 1149–1179. [Google Scholar] [CrossRef]
  236. Winter, W.E.; Flax, S.D.; Harris, N.S. Coagulation Testing in the Core Laboratory. Lab. Med. 2017, 48, 295–313. [Google Scholar] [CrossRef] [PubMed]
  237. Zwaginga, J.J.; Koomans, H.A.; Sixma, J.J.; Rabelink, T.J. Thrombus formation and platelet-vessel wall interaction in the nephrotic syndrome under flow conditions. J. Clin. Investig. 1994, 93, 204–211. [Google Scholar] [CrossRef] [Green Version]
  238. Tuktamyshov, R.; Zhdanov, R. The method of in vivo evaluation of hemostasis: Spatial thrombodynamics. Hematology 2015, 20, 584–586. [Google Scholar] [CrossRef]
  239. Bassus, S.; Herkert, O.; Kronemann, N.; Gorlach, A.; Bremerich, D.; Kirchmaier, C.M.; Busse, R.; Schini-Kerth, V.B. Thrombin causes vascular endothelial growth factor expression in vascular smooth muscle cells: Role of reactive oxygen species. Arterioscler. Thromb. Vasc. Biol. 2001, 21, 1550–1555. [Google Scholar] [CrossRef] [Green Version]
  240. Huang, Y.Q.; Li, J.J.; Hu, L.; Lee, M.; Karpatkin, S. Thrombin induces increased expression and secretion of VEGF from human FS4 fibroblasts, DU145 prostate cells and CHRF megakaryocytes. Thromb. Haemost. 2001, 86, 1094–1098. [Google Scholar]
  241. Lockwood, C.J.; Krikun, G.; Koo, A.B.; Kadner, S.; Schatz, F. Differential effects of thrombin and hypoxia on endometrial stromal and glandular epithelial cell vascular endothelial growth factor expression. J. Clin. Endocrinol. Metab. 2002, 87, 4280–4286. [Google Scholar] [CrossRef] [Green Version]
  242. Furukawa, Y.; Kawano, Y.; Fukuda, J.; Matsumoto, H.; Narahara, H. The production of vascular endothelial growth factor and metalloproteinase via protease-activated receptor in human endometrial stromal cells. Fertil. Steril. 2009, 91, 535–541. [Google Scholar] [CrossRef] [PubMed]
  243. Babayev, S.N.; Kanchwala, M.; Xing, C.; Akgul, Y.; Carr, B.R.; Word, R.A. Thrombin Alters Human Endometrial Stromal Cell Differentiation During Decidualization. Reprod. Sci. 2019, 26, 278–288. [Google Scholar] [CrossRef] [PubMed]
  244. Ding, D.; Liu, X.; Guo, S.W. Further Evidence for Hypercoagulability in Women With Ovarian Endometriomas. Reprod. Sci. 2018, 25, 1540–1548. [Google Scholar] [CrossRef] [PubMed]
  245. Liu, X.; Ding, D.; Ren, Y.; Guo, S.W. Transvaginal Elastosonography as an Imaging Technique for Diagnosing Adenomyosis. Reprod. Sci. 2018, 25, 498–514. [Google Scholar] [CrossRef]
  246. Nie, J.; Zhao, C.; Lagana, A.S.; Liu, X.; Guo, S.W. Identification of lesional attributes of dysmenorrhea severity and the serum antimullerian hormone levels in women with ovarian endometriomas. Fertil. Steril. 2022, 118, 191–202. [Google Scholar] [CrossRef]
  247. Nishimura, F.; Mogami, H.; Moriuchi, K.; Chigusa, Y.; Mandai, M.; Kondoh, E. Mechanisms of thrombin-Induced myometrial contractions: Potential targets of progesterone. PLoS ONE 2020, 15, e0231944. [Google Scholar] [CrossRef]
  248. O’Sullivan, C.J.; Allen, N.M.; O’Loughlin, A.J.; Friel, A.M.; Morrison, J.J. Thrombin and PAR1-activating peptide: Effects on human uterine contractility in vitro. Am. J. Obstet. Gynecol. 2004, 190, 1098–1105. [Google Scholar] [CrossRef]
  249. Elovitz, M.A.; Ascher-Landsberg, J.; Saunders, T.; Phillippe, M. The mechanisms underlying the stimulatory effects of thrombin on myometrial smooth muscle. Am. J. Obstet. Gynecol. 2000, 183, 674–681. [Google Scholar] [CrossRef]
  250. Elovitz, M.A.; Saunders, T.; Ascher-Landsberg, J.; Phillippe, M. Effects of thrombin on myometrial contractions in vitro and in vivo. Am. J. Obstet. Gynecol. 2000, 183, 799–804. [Google Scholar] [CrossRef]
  251. Cruz, M.A.; Gonzalez, C.; Acevedo, C.G.; Sepulveda, W.H.; Rudolph, M.I. Effects of histamine and serotonin on the contractility of isolated pregnant and nonpregnant human myometrium. Gynecol. Obstet. Investig. 1989, 28, 1–4. [Google Scholar] [CrossRef]
  252. Martinez-Mir, M.I.; Estan, L.; Morales-Olivas, F.J.; Rubio, E. Effect of histamine and histamine analogues on human isolated myometrial strips. Br. J. Pharmacol. 1992, 107, 528–531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Cordeaux, Y.; Missfelder-Lobos, H.; Charnock-Jones, D.S.; Smith, G.C. Stimulation of contractions in human myometrium by serotonin is unmasked by smooth muscle relaxants. Reprod. Sci. 2008, 15, 727–734. [Google Scholar] [CrossRef] [PubMed]
  254. Li, H.; Diao, M.; Ni, J.; Li, P.; Zhu, Q.; Wan, Y.L.; Ma, Y.S.; Lin, X.M. Stimulation of contractions in pregnant human myometrium is associated with 5-HT3 receptors. Int. J. Obstet. Anesth. 2016, 28, 28–33. [Google Scholar] [CrossRef] [PubMed]
  255. Tsuda, M.; Tozaki-Saitoh, H.; Inoue, K. Platelet-activating factor and pain. Biol. Pharm. Bull. 2011, 34, 1159–1162. [Google Scholar] [CrossRef]
  256. Simoni, J.; Simoni, G.; Lox, C.D.; McGunegle, D.E.; Feola, M. Cytokines and PAF release from human monocytes and macrophages: Effect of hemoglobin and contaminants. Artif. Cells. Blood. Substit. Immobil. Biotechnol. 1994, 22, 525–534. [Google Scholar] [CrossRef]
  257. Hemmings, R.; Miron, P.; Falcone, T.; Bourque, J.; Lepage, N.; Langlais, J. Platelet-activating factor acetylhydrolase activity in peritoneal fluids of women with endometriosis. Obstet. Gynecol. 1993, 81, 276–279. [Google Scholar]
  258. Hellman, K.M.; Yu, P.Y.; Oladosu, F.A.; Segel, C.; Han, A.; Prasad, P.V.; Jilling, T.; Tu, F.F. The Effects of Platelet-Activating Factor on Uterine Contractility, Perfusion, Hypoxia, and Pain in Mice. Reprod. Sci. 2018, 25, 384–394. [Google Scholar] [CrossRef]
  259. Nie, J.; Liu, X.; Guo, S.W. Immunoreactivity of oxytocin receptor and transient receptor potential vanilloid type 1 and its correlation with dysmenorrhea in adenomyosis. Am. J. Obstet. Gynecol. 2010, 202, 346.e1–346.e8. [Google Scholar] [CrossRef]
  260. Chen, Y.; Zhu, B.; Zhang, H.; Ding, D.; Liu, X.; Guo, S.W. Possible Loss of GABAergic Inhibition in Mice With Induced Adenomyosis and Treatment With Epigallocatechin-3-Gallate Attenuates the Loss With Improved Hyperalgesia. Reprod. Sci. 2014, 21, 869–882. [Google Scholar] [CrossRef] [Green Version]
  261. Zhang, X.; Lu, B.; Huang, X.; Xu, H.; Zhou, C.; Lin, J. Innervation of endometrium and myometrium in women with painful adenomyosis and uterine fibroids. Fertil. Steril. 2010, 94, 730–737. [Google Scholar] [CrossRef]
  262. Lertvikool, S.; Sukprasert, M.; Pansrikaew, P.; Rattanasiri, S.; Weerakiet, S. Comparative study of nerve fiber density between adenomyosis patients with moderate to severe pain and mild pain. J. Med. Assoc. Thai. 2014, 97, 791–797. [Google Scholar] [PubMed]
  263. Zhang, X.; Lu, B.; Huang, X.; Xu, H.; Zhou, C.; Lin, J. Endometrial nerve fibers in women with endometriosis, adenomyosis, and uterine fibroids. Fertil. Steril. 2009, 92, 1799–1801. [Google Scholar] [CrossRef] [PubMed]
  264. Xu, X.; Cai, X.; Liu, X.; Guo, S.W. Possible involvement of neuropeptide and neurotransmitter receptors in Adenomyosis. Reprod. Biol. Endocrinol. 2021, 19, 25. [Google Scholar] [CrossRef]
  265. Guo, S.W.; Zhang, Q.; Liu, X. Social psychogenic stress promotes the development of endometriosis in mouse. Reprod. Biomed. Online 2017, 34, 225–239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Long, Q.; Liu, X.; Qi, Q.; Guo, S.W. Chronic stress accelerates the development of endometriosis in mouse through adrenergic receptor beta2. Hum. Reprod. 2016, 31, 2506–2519. [Google Scholar] [CrossRef]
  267. Kishi, Y.; Suginami, H.; Kuramori, R.; Yabuta, M.; Suginami, R.; Taniguchi, F. Four subtypes of adenomyosis assessed by magnetic resonance imaging and their specification. Am. J. Obstet. Gynecol. 2012, 207, 114.e1–114.e7. [Google Scholar] [CrossRef]
  268. Bourdon, M.; Oliveira, J.; Marcellin, L.; Santulli, P.; Bordonne, C.; Maitrot Mantelet, L.; Millischer, A.E.; Plu Bureau, G.; Chapron, C. Adenomyosis of the inner and outer myometrium are associated with different clinical profiles. Hum. Reprod. 2021, 36, 349–357. [Google Scholar] [CrossRef] [PubMed]
  269. Reed, G.L. Platelet secretory mechanisms. Semin. Thromb. Hemost. 2004, 30, 441–450. [Google Scholar] [CrossRef]
  270. Yang, B.; Wang, L.; Wan, X.; Li, Y.; Yu, X.; Qin, Y.; Luo, Y.; Wang, F.; Huang, O. Elevated plasma levels of lysophosphatidic acid and aberrant expression of lysophosphatidic acid receptors in adenomyosis. BMC Womens Health 2017, 17, 118. [Google Scholar] [CrossRef] [Green Version]
  271. Dietze, R.; Starzinski-Powitz, A.; Scheiner-Bobis, G.; Tinneberg, H.R.; Meinhold-Heerlein, I.; Konrad, L. Lysophosphatidic acid triggers cathepsin B-mediated invasiveness of human endometriotic cells. Biochim. Biophys. Acta. Mol. Cell. Biol. Lipids 2018, 1863, 1369–1377. [Google Scholar] [CrossRef]
  272. Wu, Y.; Wang, H.; Li, Y.; Li, Y.; Liang, Y.; Zhong, G.; Zhang, Q. Estrogen-increased SGK1 Promotes Endometrial Stromal Cell Invasion in Adenomyosis by Regulating with LPAR2. Reprod. Sci. 2022, 29, 3026–3038. [Google Scholar] [CrossRef] [PubMed]
  273. Kowalczyk-Zieba, I.; Woclawek-Potocka, I.; Wasniewski, T.; Boruszewska, D.; Grycmacher, K.; Sinderewicz, E.; Staszkiewicz, J.; Wolczynski, S. LPAR2 and LPAR4 are the Main Receptors Responsible for LPA Actions in Ovarian Endometriotic Cysts. Reprod. Sci. 2019, 26, 139–150. [Google Scholar] [CrossRef] [PubMed]
  274. Waldegger, S.; Klingel, K.; Barth, P.; Sauter, M.; Rfer, M.L.; Kandolf, R.; Lang, F. h-sgk serine-threonine protein kinase gene as transcriptional target of transforming growth factor beta in human intestine. Gastroenterology 1999, 116, 1081–1088. [Google Scholar] [CrossRef] [PubMed]
  275. Walker, B.; Schmid, E.; Russo, A.; Schmidt, E.M.; Burk, O.; Munzer, P.; Velic, A.; Macek, B.; Schaller, M.; Schwab, M.; et al. Impact of the serum- and glucocorticoid-inducible kinase 1 on platelet dense granule biogenesis and secretion. J. Thromb. Haemost. 2015, 13, 1325–1334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  276. Boucharaba, A.; Serre, C.M.; Gres, S.; Saulnier-Blache, J.S.; Bordet, J.C.; Guglielmi, J.; Clezardin, P.; Peyruchaud, O. Platelet-derived lysophosphatidic acid supports the progression of osteolytic bone metastases in breast cancer. J. Clin. Investig. 2004, 114, 1714–1725. [Google Scholar] [CrossRef]
  277. Vannuzzi, V.; Bernacchioni, C.; Muccilli, A.; Castiglione, F.; Nozzoli, F.; Vannuccini, S.; Capezzuoli, T.; Ceccaroni, M.; Bruni, P.; Donati, C.; et al. Sphingosine 1-phosphate pathway is dysregulated in adenomyosis. Reprod. Biomed. Online 2022, 45, 15–18. [Google Scholar] [CrossRef]
  278. Yoshino, O.; Yamada-Nomoto, K.; Kano, K.; Ono, Y.; Kobayashi, M.; Ito, M.; Yoneda, S.; Nakashima, A.; Shima, T.; Onda, T.; et al. Sphingosine 1 Phosphate (S1P) Increased IL-6 Expression and Cell Growth in Endometriotic Cells. Reprod. Sci. 2019, 26, 1460–1467. [Google Scholar] [CrossRef]
  279. Ono, Y.; Kawakita, T.; Yoshino, O.; Sato, E.; Kano, K.; Ohba, M.; Okuno, T.; Ito, M.; Koga, K.; Honda, M.; et al. Sphingosine 1-Phosphate (S1P) in the Peritoneal Fluid Skews M2 Macrophage and Contributes to the Development of Endometriosis. Biomedicines 2021, 9, 1519. [Google Scholar] [CrossRef]
  280. Bernacchioni, C.; Capezzuoli, T.; Vannuzzi, V.; Malentacchi, F.; Castiglione, F.; Cencetti, F.; Ceccaroni, M.; Donati, C.; Bruni, P.; Petraglia, F. Sphingosine 1-phosphate receptors are dysregulated in endometriosis: Possible implication in transforming growth factor beta-induced fibrosis. Fertil. Steril. 2021, 115, 501–511. [Google Scholar] [CrossRef]
  281. Takeya, H.; Gabazza, E.C.; Aoki, S.; Ueno, H.; Suzuki, K. Synergistic effect of sphingosine 1-phosphate on thrombin-induced tissue factor expression in endothelial cells. Blood 2003, 102, 1693–1700. [Google Scholar] [CrossRef]
  282. Huang, Q.; Liu, X.; Guo, S.W. Changing prostaglandin E2 (PGE2) signaling during lesional progression and exacerbation of endometriosis by inhibition of PGE2 receptor EP2 and EP4. Reprod. Med. Biol. 2022, 21, e12426. [Google Scholar] [CrossRef] [PubMed]
  283. Huang, Q.; Liu, X.; Guo, S.W. Higher fibrotic content of endometriotic lesions is associated with diminished prostaglandin E2 signaling. Reprod. Med. Biol. 2022, 21, e12423. [Google Scholar] [CrossRef] [PubMed]
  284. Ding, D.; Cai, X.; Zheng, H.; Guo, S.W.; Liu, X. Scutellarin Suppresses Platelet Aggregation and Stalls Lesional Progression in Mouse With Induced Endometriosis. Reprod. Sci. 2019, 26, 1417–1428. [Google Scholar] [CrossRef] [PubMed]
  285. Zheng, Y.; Liu, X.; Guo, S.W. Therapeutic potential of andrographolide for treating endometriosis. Hum. Reprod. 2012, 27, 1300–1313. [Google Scholar] [CrossRef] [Green Version]
  286. Luo, M.; Cai, X.; Yan, D.; Liu, X.; Guo, S.W. Sodium tanshinone IIA sulfonate restrains fibrogenesis through induction of senescence in mice with induced deep endometriosis. Reprod. Biomed. Online 2020, 41, 373–384. [Google Scholar] [CrossRef]
  287. Nagira, K.; Taniguchi, F.; Nakamura, K.; Tokita, Y.; Tsuchiya, N.; Khine, Y.M.; Harada, T. Tokishakuyakusan, a Kampo medicine, attenuates endometriosis-like lesions and hyperalgesia in murine with endometriosis-like symptoms. Am. J. Reprod. Immunol. 2019, 82, e13182. [Google Scholar] [CrossRef]
  288. Lee, H.W.; Jun, J.H.; Kil, K.J.; Ko, B.S.; Lee, C.H.; Lee, M.S. Herbal medicine (Danggui Shaoyao San) for treating primary dysmenorrhea: A systematic review and meta-analysis of randomized controlled trials. Maturitas 2016, 85, 19–26. [Google Scholar] [CrossRef]
  289. Chen, Y.; Zhu, B.; Zhang, H.; Liu, X.; Guo, S.W. Epigallocatechin-3-gallate reduces myometrial infiltration, uterine hyperactivity, and stress levels and alleviates generalized hyperalgesia in mice induced with adenomyosis. Reprod. Sci. 2013, 20, 1478–1491. [Google Scholar] [CrossRef] [Green Version]
  290. Zhu, B.; Chen, Y.; Zhang, H.; Liu, X.; Guo, S.W. Resveratrol Reduces Myometrial Infiltration, Uterine Hyperactivity, and Stress Levels and Alleviates Generalized Hyperalgesia in Mice With Induced Adenomyosis. Reprod. Sci. 2015, 22, 1336–1349. [Google Scholar] [CrossRef]
  291. Nie, J.; Liu, X. Quercetin alleviates generalized hyperalgesia in mice with induced adenomyosis. Mol. Med. Rep. 2017, 16, 5370–5376. [Google Scholar] [CrossRef] [Green Version]
  292. Nie, J.; Liu, X. Leonurine Attenuates Hyperalgesia in Mice with Induced Adenomyosis. Med. Sci. Monit. 2017, 23, 1701–1706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  293. Zhu, B.; Chen, Y.; Guo, M.; Zhang, C.; Huang, L.; Pan, Q.; Lin, T.; Lu, Y.; Shen, X.; Zhang, H. Berberine attenuates hyperalgesia in mice with adenomyosis. Arch. Gynecol. Obstet. 2022, 3016, 115–125. [Google Scholar] [CrossRef]
  294. Liu, X.; Guo, S.W. Valproic acid alleviates generalized hyperalgesia in mice with induced adenomyosis. J. Obstet. Gynaecol. Res. 2011, 37, 696–708. [Google Scholar] [CrossRef] [PubMed]
  295. Wan, L.; Zou, Y.; Wan, L.H.; Wang, L.Q.; Huang, M.Z.; Wu, J.; Zhu, Y.B.; Huang, O.P. Tanshinone IIA inhibits the proliferation, migration and invasion of ectopic endometrial stromal cells of adenomyosis via 14-3-3zeta downregulation. Arch. Gynecol. Obstet. 2015, 292, 1301–1309. [Google Scholar] [CrossRef] [PubMed]
  296. Wu, Y.; Guo, S.W. Histone deacetylase inhibitors trichostatin A and valproic acid induce cell cycle arrest and p21 expression in immortalized human endometrial stromal cells. Eur. J. Obstet. Gynecol. Reprod. Biol. 2008, 137, 198–203. [Google Scholar] [CrossRef]
  297. Li, Y.D.; Ye, B.Q.; Zheng, S.X.; Wang, J.T.; Wang, J.G.; Chen, M.; Liu, J.G.; Pei, X.H.; Wang, L.J.; Lin, Z.X.; et al. NF-kappaB transcription factor p50 critically regulates tissue factor in deep vein thrombosis. J. Biol. Chem. 2009, 284, 4473–4483. [Google Scholar] [CrossRef] [Green Version]
  298. Paul, M.; Hemshekhar, M.; Kemparaju, K.; Girish, K.S. Berberine mitigates high glucose-potentiated platelet aggregation and apoptosis by modulating aldose reductase and NADPH oxidase activity. Free. Radic. Biol. Med. 2019, 130, 196–205. [Google Scholar] [CrossRef]
  299. Bertelli, A.A.; Giovannini, L.; Giannessi, D.; Migliori, M.; Bernini, W.; Fregoni, M.; Bertelli, A. Antiplatelet activity of synthetic and natural resveratrol in red wine. Int. J. Tissue React. 1995, 17, 1–3. [Google Scholar]
  300. Beretz, A.; Cazenave, J.P.; Anton, R. Inhibition of aggregation and secretion of human platelets by quercetin and other flavonoids: Structure-activity relationships. Agents. Actions. 1982, 12, 382–387. [Google Scholar] [CrossRef]
  301. Zhang, R.H.; Liu, Z.K.; Yang, D.S.; Zhang, X.J.; Sun, H.D.; Xiao, W.L. Phytochemistry and pharmacology of the genus Leonurus: The herb to benefit the mothers and more. Phytochemistry 2018, 147, 167–183. [Google Scholar] [CrossRef]
  302. Maione, F.; De Feo, V.; Caiazzo, E.; De Martino, L.; Cicala, C.; Mascolo, N. Tanshinone IIA, a major component of Salvia milthorriza Bunge, inhibits platelet activation via Erk-2 signaling pathway. J. Ethnopharmacol. 2014, 155, 1236–1242. [Google Scholar] [CrossRef]
  303. Wang, H.; Zhong, L.; Mi, S.; Song, N.; Zhang, W.; Zhong, M. Tanshinone IIA prevents platelet activation and down-regulates CD36 and MKK4/JNK2 signaling pathway. BMC. Cardiovasc. Disord. 2020, 20, 81. [Google Scholar] [CrossRef]
  304. Davidson, D.C.; Hirschman, M.P.; Spinelli, S.L.; Morrell, C.N.; Schifitto, G.; Phipps, R.P.; Maggirwar, S.B. Antiplatelet activity of valproic acid contributes to decreased soluble CD40 ligand production in HIV type 1-infected individuals. J. Immunol. 2011, 186, 584–591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  305. Fraser, I.S.; Burridge, J. Danazol treatment and platelet function. Med. J. Aust. 1980, 1, 313–314. [Google Scholar] [CrossRef] [PubMed]
  306. Liu, M.; Liu, X.; Zhang, Y.; Guo, S.W. Valproic acid and progestin inhibit lesion growth and reduce hyperalgesia in experimentally induced endometriosis in rats. Reprod. Sci. 2012, 19, 360–373. [Google Scholar] [CrossRef] [PubMed]
  307. Liu, X.; Guo, S.W. A pilot study on the off-label use of valproic acid to treat adenomyosis. Fertil. Steril. 2008, 89, 246–250. [Google Scholar] [CrossRef]
  308. Liu, X.; Yu, S.; Guo, S.-W. A pilot study on the use of andrographolide to treat symptomatic adenomyosis. Gynecol. Minim. Invasive. Ther. 2014, 3, 119–126. [Google Scholar] [CrossRef] [Green Version]
  309. Igarashi, M.; Abe, Y.; Fukuda, M.; Ando, A.; Miyasaka, M.; Yoshida, M.; Shawki, O.A. Novel conservative medical therapy for uterine adenomyosis with a danazol-loaded intrauterine device. Fertil. Steril. 2000, 74, 412–413. [Google Scholar] [CrossRef]
  310. Guo, S.W. An overview of the current status of clinical trials on endometriosis: Issues and concerns. Fertil. Steril. 2014, 101, 183–190.e4. [Google Scholar] [CrossRef]
  311. Ludwig, N.; Hilger, A.; Zarbock, A.; Rossaint, J. Platelets at the Crossroads of Pro-Inflammatory and Resolution Pathways during Inflammation. Cells 2022, 11, 1957. [Google Scholar] [CrossRef]
Figure 1. Schematic illustration of the roles of platelets in the progression of adenomyotic lesions. The arrows indicate the crosstalk between adenomyotic lesions and other cells in the lesional microenvironment. 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), and sensory nerve fibers can accelerate the development of adenomyotic lesions. Type 2 immunity also promotes fibrogenesis of adenomyotic lesions. Histamine, 5-HT, PAF, TXA2 and thrombin derived from activated platelets may also enhance uterine contractility. The upward arrows indicate upregulation/overexpression or increase. The directional arrows mean “lead to” or “result in”. Abbreviations used: 5-HT = 5-hydroxytryptamine, serotonin; E2 = 17β-estradiol; 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; PAF = platelet activating factor; PAR2 = protease activated receptor 2; PGE2 = prostaglandin E2; S1P = sphingosine 1-phosphate; S1P3= sphingosine 1-phosphate receptor 3; PDGF = platelet-derived growth factor; TGF-β1 = transforming growth factor β1; TXA2 = thromboxane A2; TSLP = thymic stromal lymphopoietin; TSP1 = thrombospondin-1; TF = tissue factor.
Figure 1. Schematic illustration of the roles of platelets in the progression of adenomyotic lesions. The arrows indicate the crosstalk between adenomyotic lesions and other cells in the lesional microenvironment. 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), and sensory nerve fibers can accelerate the development of adenomyotic lesions. Type 2 immunity also promotes fibrogenesis of adenomyotic lesions. Histamine, 5-HT, PAF, TXA2 and thrombin derived from activated platelets may also enhance uterine contractility. The upward arrows indicate upregulation/overexpression or increase. The directional arrows mean “lead to” or “result in”. Abbreviations used: 5-HT = 5-hydroxytryptamine, serotonin; E2 = 17β-estradiol; 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; PAF = platelet activating factor; PAR2 = protease activated receptor 2; PGE2 = prostaglandin E2; S1P = sphingosine 1-phosphate; S1P3= sphingosine 1-phosphate receptor 3; PDGF = platelet-derived growth factor; TGF-β1 = transforming growth factor β1; TXA2 = thromboxane A2; TSLP = thymic stromal lymphopoietin; TSP1 = thrombospondin-1; TF = tissue factor.
Jcm 12 00842 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Guo, S.-W. The Role of Platelets in the Pathogenesis and Pathophysiology of Adenomyosis. J. Clin. Med. 2023, 12, 842. https://doi.org/10.3390/jcm12030842

AMA Style

Guo S-W. The Role of Platelets in the Pathogenesis and Pathophysiology of Adenomyosis. Journal of Clinical Medicine. 2023; 12(3):842. https://doi.org/10.3390/jcm12030842

Chicago/Turabian Style

Guo, Sun-Wei. 2023. "The Role of Platelets in the Pathogenesis and Pathophysiology of Adenomyosis" Journal of Clinical Medicine 12, no. 3: 842. https://doi.org/10.3390/jcm12030842

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop