Skip to main content
Erschienen in: Reproductive Biology and Endocrinology 1/2018

Open Access 01.12.2018 | Review

Villainous role of estrogen in macrophage-nerve interaction in endometriosis

verfasst von: Yanchun Liang, Hongyu Xie, Jinjie Wu, Duo Liu, Shuzhong Yao

Erschienen in: Reproductive Biology and Endocrinology | Ausgabe 1/2018

Abstract

Endometriosis is a complex and heterogeneous disorder with unknown etiology. Dysregulation of macrophages and innervation are important factors influencing the pathogenesis of endometriosis-associated pain. It is known to be an estrogen-dependent disease, estrogen can promote secretion of chemokines from peripheral nerves, enhancing the recruitment and polarization of macrophages in endometriotic tissue. Macrophages have a role in the expression of multiple nerve growth factors (NGF), which mediates the imbalance of neurogenesis in an estrogen-dependent manner. Under the influence of estrogen, co-existence of macrophages and nerves induces an innovative neuro-immune communication. Persistent stimulation by inflammatory cytokines from macrophages on nociceptors of peripheral nerves aggravates neuroinflammation through the release of inflammatory neurotransmitters. This neuro-immune interaction regulated by estrogen sensitizes peripheral nerves, leading to neuropathic pain in endometriosis. The aim of this review is to highlight the significance of estrogen in the interaction between macrophages and nerve fibers, and to suggest a potentially valuable therapeutic target for endometriosis-associated pain.

Introduction

Endometriosis is a common gynecological disorder, which is defined as the presence of the endometrial-like tissue outside the uterine cavity [1]. This chronic inflammatory disease affects 6–10% of women during reproductive age [2], and 30 to 50% are accompanied with pelvic pain and is associated with infertility [3, 4]. Though extensive research has proposed many theories for the pathogenesis of endometriosis, the mechanism of endometriosis-associated pain still remains elusive. As an estrogen-dependent disease, local levels of estrogen in the menstrual blood and peritoneal fluids are higher in patients with endometriosis in comparison to healthy women [5]. Estrogen exhibits a pivotal role in the maintenance of endometriosis-associated pain.
Growing evidences support that the presence of endometriosis-associated nerve fibers is related to the severity of dysmenorrhea [6]. Studies have confirmed the presence and high expression of sensory, sympathetic and parasympathetic nerve fibers in peritoneal lesion in comparison to the normal peritoneum [7]. In addition to the aberrant distribution of nerves, impairment of immune microenvironment in the peritoneal cavity induces inflammation which can also mediate endometriosis-associated pain [8]. Endometriosis-associated inflammation is able to stimulate and sensitize peripheral nerves. Researchers found that activation of (DRG) mast cells by estrogen can trigger the release of NGF and also sensitize dorsal root ganglion cells [9]. This neuroimmune interaction mediated by estrogen is correlated with the severity of endometriosis-associated dysmenorrhea. Among the dysregulated immune components, macrophages are one of the most numerous immune cells in endometriotic lesions [10]. The concentration and proportion of macrophage are significantly increased in peritoneal fluid of women with endometriosis [11, 12], indicating their involvement in the development of pain. A recent study exhibits the critical role of estrogen in mediating the communication between macrophage and nerve fibers in endometriosis [13]. However, as an estrogen-dependent disease, the exact mechanism of estrogen regulating the cross talk between macrophages and nerve fibers in endometriosis is ambiguous.
The aim of this review is to highlight the potential role of estrogen in maintaining the cross talk between macrophages and nerve fibers in endometriosis. Theoretical elaboration of the underlying mechanism provides a new insight into the pathogenesis of endometriosis and a potential therapeutic targets for treatment.

The role of estrogen and their receptors in endometriosis

Estrogen has a central role in endometriosis. It can facilitate disease progression mainly through binding different estrogen receptors (ER) [14]. The ERs can be classically divided into two subtypes: ERα and ERβ, both act as nuclear transcription factors [15]. ERα is mainly responsible for the modulation of cell growth related genes [16], whereas ERβ plays an important role in cell cycle progression and apoptosis. Further studies reveal another subtype of estrogen receptor, membrane-association G-protein coupled estrogen receptor (GPER) in the cell menmbrane, which is able to mediate the rapid non-genomic effects of estrogen [17]. GPER is reported to be capable of activating several downstream molecules, such as ERK1/2, PI3K, PKC and calcium ion channels [17]. These studies suggest that different signaling pathways are activated by different kinds of ER, exhibiting their distinct function. As a result, the action of estrogen on endometriosis is closely related to the types of receptors and their distribution.

Distribution of estrogen receptors in endometriosis

Different estrogen receptors have abnormal distribution in the ectopic endometrium of women with endometriosis [18]. A higher ERβ and a lower ERα expression profile in endometriotic lesions have been proposed as a major background of estrogen action in endometriosis [19]. Several studies have shown significantly elevated ERβ levels and decreased ERα levels in endometriotic stromal cells and tissues compared to the eutopic endometrium [20]. ERβ-induced the formation of ASK-1/STRAP complex can prevent the activation of tumor necrosis factor α (TNFα)-mediated apoptosis in endometriotic tissues [21]. Increased expression of Ras-like and estrogen-regulated growth inhibitor (RERG) has also been identified after the activation of ERβ [22]. ERβ binds to the promoter regulatory region of RERG, inducing gene expression of RERG, and then enhancing the proliferation of endometriotic cells. Moreover, multiple molecules in endometriosis are able to stimulate the expression of ERβ, such as insulin-like growth factors 1 (IGF1) [23], activated platelets [24], serum and glucocorticoid-regulated kinase (SGK1) [25]. However, cross-talk between ERα and interleukin (IL) 6 pathways is recently shown to promote the early initiation of endometriosis [26]. IL-6 mediates the recruitment of monocytes which can differentiate into macrophages expressing ERα. Estrogen-ERα binding in turn regulates the IL-6 promoter through activation of NF-κB and CEBPβ. Further study characterizes the heterogeneous expression of ERs in different types of endometriosis. Ovarian lesions show the lowest expression of ERα and the highest expression of ERβ, whereas the fallopian tube lesions show a high expression of both receptors [27]. The largest ERβ to ERα ratio is observed in ovarian lesions compared with peritoneal, fallopian tube, and extra pelvic lesions. Moreover, aberrant expression of ERα and ERβ are both correlated to the production of proinflammatory cytokines in endometriosis [28]. The combinational interaction of ERβ with caspase 1 and NLR family pyrin domain-containing 3 (NALP3) activates caspase 1 and subsequently elevates the level of IL-1β in ectopic lesions, enhancing the inflammation in the endometriotic microenvironment [21]. In addition, the expression of GPER is also significantly increased in ectopic endometrium compared to eutopic endometrium [29], which specifically promotes the proliferation of endometrial fragment. Although abnormal expression of different ERs has been identified in endometriotic lesion, their functions involved in the pathogenesis of endometriosis are far from clarified.

Characteristics of estrogen receptors in macrophages of endometriosis

As discussed above, activation of the estrogen receptors can induce inflammatory events. In the endometriotic microenvironment, multiple immune cells, such as macrophages, mast cells, neutrophils, and mature dendritic cells are mobilized by the retrograded endometrial cells. Among them, macrophages are the primary contributor of pro-inflammatory cytokines and chemokines [30]. Both ERα and ERβ are significantly overexpressed in macrophages of endometriosis [28]. Treatment with 17β-estradiol in human macrophages in vitro induces an increase of ERα expression in macrophages. Furthermore, the estrogen-ER interaction can up-regulate the production of IL-6 and TNFα from lipopolysaccharide (LPS)-activated macrophages [31]. Specifically, macrophages with overexpression of ERβ in endometriotic lesions have been shown to exhibit enhanced phagocytic activity, and secrete inflammatory cytokines including IL1β, TNFα and IL6 [21, 32]. Deficiency of ERα, but not ERβ, increased TNFα production by mouse peritoneal macrophages in response to bacterial stimuli, indicating a prominent role of ERα in mediating the anti-inflammatory effect of estrogen. However, an opposite finding shows the ability of selective ERβ activation to inhibit the expression of inflammatory mediators. Some researchers even argue that ERα is only detected in a subset of macrophages in both peritoneal cavity and endometriotic biopsy specimens, which is different from previous studies [13]. Activation of ERα by estrogen is able to inhibit NF-κB-dependent inflammation by promoting synthesis of the negative regulator IκBα [33]. And this discrepancy about the ERs on macrophages is due to the inherent differences between peritoneal fluid macrophages and tissue macrophages. Moreover, the expression of GPER on macrophage in endometriosis is also significantly increased compared with that of healthy tissue, suggesting an alternative way that the immune response induced by macrophages is modulated by estrogen [34]. Treatment with the selective GPER agonist G-1 is able to inhibit LPS-induced TNFα production in human macrophages. Activation of GPER down-regulates NF-κB promoter activation through increase phosphorylation of JNK, resulting in the inhibition of LPS-induced IL-6 secretion [35]. GPER can also affects function macrophages by decreasing the expression of TLR4. These data indicate that the pro- or anti-inflammatory role of estrogen on macrophages is closely related to different types of estrogen receptors. Activation of ERβ has the potential to arouse pro-inflammatory response, while both ERα and GPER play significant roles in anti-inflammation.

Estrogen receptors in the nervous system

In addition to macrophage, ERs are also expressed on the nervous system. Whether in the myometrium of women with or without adenomyosis, the expression of ERβ in sympathetic nerves is much more than sensory nerves [36, 37]. Activation of ERβ in sympathetic nerve could promote the down-regulation of cyclin E, IGF1, or cytokines [38], leading to a prominent reduction of the sympathetic innervation. ERα and ERβ are also expressed in the uterine-related dorsal root ganglion (DRG) neurons [39, 40], suggesting an important plasticity regulation and developmental function in uterine sensory nerves. Although ERα and ERβ are detected in DRG, the impacts of ER agonists on neuron in endometriosis have been clarified to depend on the subtypes of ERs. Activation of ERβ in DRG can down-regulate the expression of nerve repellent factor, SLIT3. Conversely, activation of ERα upregulates SLIT3 expression in DRGs, highlighting the potentially opposite impacts of different ERs in neuron [41]. Acute activation of membrane-associated ERα in peripheral sensory neurons by estrogen is confirmed to enhance allodynia elicited by the inflammatory mediator bradykinin [42]. Moreover, GPER is also identified in peripheral sensory neurons and the expression of GPER is increased after peripheral nerve injury [43]. However, the exact mechanism of GPER in neurogenesis and peripheral sensitization remains unclear.

Estrogen mediates the interaction of macrophages and nerve fibers

As different estrogen receptors are both expressed on the macrophages and nerve fibers, estrogen is believed to be able to mediate the activity of macrophages and the characteristics of nerve fibers. Therefore, estrogen is critical to regulate the interaction between macrophages and nerve fibers. This includes the recruitment of macrophages, aberrant neurogenesis as well as the abnormal inflammation in endometriosis [13].

Recruitment of macrophages by nerve fibers in an estrogen-dependent manner

There have been multiple studies demonstrating a significantly high level of macrophages in endometriotic milieu [44, 45]. Among different mechanisms involved in the recruitment of macrophages, nerve fibers exhibit an extraordinary ability to recruit macrophages to the site of lesion. Many factors have been confirmed to mediate the recruitment of macrophages toward the nerve fibers, including leukemia inhibitory factor (LIF) [46], IL1α, IL1β [47] and pancreatitis- associated protein 3 (PAP3) [48]. The role of estrogen during the process of nerve fiber-induced macrophage recruitment has been recently clarified [13]. Following peripheral axon injury, the neuron-derived exosomal miR-21-5p is phagocytosed by macrophages, which can mediate the macrophages infiltrations towards peripheral nerves [49]. Analysis has proven that exosomal miR-21-5p release is mediated by the estrogen signaling pathway [50]. Estrogen can also stimulate the secretion of colony-stimulating factor 1 (CSF1) and C-C motif ligand 2 (CCL2) from peripheral nerves, which enhances macrophages migration towards the endometriotic lesion [13].
Macrophages are special immune cells with high plasticity and different functional profiles. These immune cells can be divided into two subtypes as classically activated (M1) and alternatively activated (M2) macrophages based on the distinct pathways of activation [51]. The function of M1 macrophages is to produce pro-inflammatory cytokines which is induced by Th1 cytokines, such as Interferon γ (IFNγ) and TNFα. M2 macrophages, which are anti-inflammatory, are polarized by Th2 cytokines, such as IL-4 and IL-13 via activation of STAT6 through IL-4 receptor alpha [52]. Interestingly, the macrophages in endometriosis are predominantly of M2 phenotype [53]. Further evidence confirms that abnormal endometriotic milieu can induce the polarization of M2 macrophages [54]. Estrogen-dependent regulation of CCL2 in peritoneal cavity is confirmed to polarize macrophages from M1 toward M2 phenotype via CCL2/CCR pathway [55]. 17β-estradiol and TCDD (2, 3, 7, 8-tetrachlorodibenzo-p-dioxin) also have a synergistic effect on inducing M2 macrophages polarization through activation of the STAT3 and P38-MAPK pathway [56]. Moreover, ERα signaling is recently clarified to preferentially polarize macrophages from a range of sources to an alternative phenotype [33]. Estrogen is able to inhibit NFκB-dependent inflammation by promoting synthesis of the negative regulator IκBα [57]. However, an opposed result shows that estrogen inhibits alternative activation polarization in a tumor-associated macrophage via the inhibition of JAK1-STAT6 pathway [58]. Although the role of estrogen in polarization of macrophages remains controversial, studies discussed above indicate the capacity of estrogen in mediating the recruitment of macrophages toward nerve fibers accompanied with M2 polarization of macrophages (Fig. 1).

Neurogenesis is regulated by macrophages in an estrogen-dependent manner

Aberrant of innervation in endometriotic lesions is also demonstrated to be involved in the pathogenesis of endometriosis-associated pain [59]. Pain sensation of endometriosis is correlated with neural hypertrophy and increased neural density in the endometriotic lesion [60, 61]. Further studies discovered that there exists an imbalanced distribution of sensory, sympathetic and parasympathetic nerve fibers in endometriosis [7]. As a result, abnormal neurogenesis is important in endometriosis. Since the coexistence of macrophages and nerve fibers as well as their close correlation, macrophages can affect the neurogenesis in endometriosis. In addition, the secretion of multiple NGF from macrophages can be induced by estrogen, indicating a pivotal role of estrogen in mediating the above process [62].

Neurotrophins

Neurotrophins (NTs) belong to one of the trophic factor family. NTs play an important role in mediating the survival, growth and programmed cell death of nerves [63]. Members of the NTs increasingly expressed in endometriosis include NGF, brain-derived nerve growth factor (BDNF) and neurotrophin 3 (NT3) [64]. Previous studies reported that estrogen could increase NGF mRNA and NGF protein in murine uterus and salivary gland [65]. It has been demonstrated that macrophages are one of the important sources of NGF. 17β-estradiol can induce c-Fos expression and shift the composition of NGF promoter, AP-1 from c-Jun homodimers to c-Fos/c-Jun heterodimers in macrophages [62]. This transformation enhances the transcriptional activity of AP-1, leading to the increased expression of NGF in macrophages. In comparison with eutopic endometrium, estradiol also has the ability to promote the secretion of NT3 and BDNF in peritoneal endometriosis [13]. Moreover, estrogen can also up-regulate the expression of the receptor of NT3, tyrosine kinase receptor B (TrkB) and the receptor of BNDF, p75NTR on sympathetic neuron, which promote neuronal survival and sympathetic axonal growth in endometriotic milieu [66]. These evidence suggest a significant impact of estrogen on macrophages to potentiate neurogenesis in endometriosis via the dysregulation of neurotrophins.

Semaphorins

Semaphorins are a group of evolutionarily highly conserved surface or locally secreted nerve repellent factors that can regulate the axons migration, axonal growth and guidance [67]. Semaphorin 3A and its receptors (NRP1 and Plexin A1) have been clarified to decrease the innervation of sympathetic nerves in endometriosis [68]. Macrophages are reported to be one of the sources of semaphorin 3A [69]. Semaphorin 3F and 3C are also demonstrated to be released from macrophages in endometriosis [70]. Their receptors (NRP1 and NRP2) are positive in noradrenergic nerve fibers, which is associated with reduced sympathetic innervation [70]. As an estrogen-dependent disease, upregulation of estrogen is found to result in an increased expression of semaphorin 3F and lead to a sympathetic denervation in the rat uterus [71]. Some investigations further reveal a constant secretion of hepatocyte growth factor (HGF) from peritoneal macrophages in response to estrogen in endometriosis [72, 73]. Elevated levels of HGF from M2 macrophages can induce the expression and secretion of semaphorin 3A from myoblast [74]. It is reasonable to speculate the role of estrogen in regulating the secretion of semaphorins from macrophages, leading to the aberrant innervation in endometriosis.

Vascular epithelial growth factor

Besides stimulating vascular growth and increasing vascular permeability, vascular epithelial growth factor (VEGF) can also be a neurotrophic factor to stimulate axonal outgrowth. The receptor of VEGF, flk1, is up-regulated in peripheral ganglia in response to axonal injury [75]. High expression of flk1 have been confirmed to mediate the pathogenesis of endometriosis [76]. Further activation of flk1 results in the survival of nerve cells and stimulation of axonal outgrowth. In addition to acting locally at the level of the axons, VEGF has the ability to be transported retrogradely and acts at the level of the neuronal cell bodies [75]. Moreover, NRP1 is a co-receptor of VEGFR2. Elevated concentration of VEGF can activate VEGF-NRP1-VEGFR2 signaling, providing chemoattractive cues for sympathetic nerve outgrowth [77]. Intriguingly, activated macrophages are a major source of VEGF in endometriosis [78]. Most importantly, the secretion of VEGF from macrophages is regulated directly by estrogen. mRNAs encoding for estrogen and progesterone have been presented in peritoneal macrophages [72]. Macrophage-derived prostaglandin E2 elevates the level of estrogen in endometriosis, which subsequently activates macrophages to release VEGF [79]. Estradiol can also upregulate the expression of flk1 in uterus, mediating the progression of disease [80]. Therefore, in response to abnormal level of estrogen, macrophages are capable to release VEGF promoting aberrant neurogenesis in endometriotic environment (Fig. 2).

The role of estrogen in inflammation and pain in endometriosis

Endometriosis is considered to be a chronic inflammatory disease and multiple molecules are demonstrated to contribute to this inflammatory milieu [81]. Since the neuroimmune communication has been identified in endometriosis, the inflammation resulting from the interplay between macrophage and nerve attracts more and more attention for the mechanism of endometriosis-associated pain [82]. According to the role of estrogen in nerve-macrophage interaction discussed above, dysregulation of estrogen exhibits a villainous role to modulate local inflammation in endometriosis and thus resulting in endometriosis-associated pain.

Inflammation-induced peripheral nerve sensitization in an estrogen-dependent manner

Peripheral nerve sensitization is a process involving reduced threshold of ion channels, increased membrane excitability, and increased nociceptors expression. Inflammatory cytokines in endometriotic milieu can influence these aspects by binding to their corresponding nociceptors on peripheral nerves. Macrophages are thought to be the major source of inflammatory mediators, which can subsequently sensitize peripheral nerve fibers. The mRNA concentration of transient receptor potential vanilloid 1 (TRPV1) and transient receptor potential cation channel, subfamily A, member 1 (TRPA1) are significantly higher in women with endometriosis [83]. TNFα, IL1β and monocyte chemotactic protein 1 (MCP1) from macrophages can influence the membrane potential of sensory nerve via TRPV-1-dependent mechanism [8385]. The NGF from macrophages can stimulate the activation of TRPV1 receptors through the activation of NGF receptors on neurons. The voltage-gated sodium channels, encoded by SCNA genes, are also stimulated by MCP1 through a CCR/Gβγ-dependent mechanism [86]. P2X (purinergic X) receptors, are another increased nociceptor in endometriosis which can be activated by extracellular ATP [87].As mentioned above, aberrant expression of ERα and ERβ in macrophages of endometriosis is correlated with the function of inflammatory cytokines, indicating the active role of estrogen in the secretion of inflammatory cytokines from macrophages [31]. The expression of those nociceptors discussed above is also up-regulated by estrogen in endometriotic cell-derived sensory neurons. These alternations by estrogen may be related to the presence of specificity protein 1 (SP1) transcription factor binding sites in the promoter regions of nociceptive genes [87]. In short, estrogen not only mediates the secretion of inflammatory cytokines from macrophages to activate nociceptors, but also increases the expression of nociceptors on peripheral nerves, which eventually exacerbates the local inflammation to induce peripheral hyper-sensitization.

Neurogenic inflammation mediated by estrogen in endometriosis

Sustained peripheral sensitization by the inflammatory mediators from macrophages elicits the release of neuropeptides with inflammatory and nociceptive function, which induces a vicious circle to deteriorate the progression of endometriosis-associated pain. TRPV1-positive nerves can induce a neurogenic inflammation through the release of substance P (SP) and calcitonin gene-related peptide (CGRP) [88]. Further research demonstrates that estrogen can modulate corneal sensitivity by up-regulation of SP [89].The release of CGRP is also regulated by 17β-estradiol in the visceral pain sensitivity, implicating the role of estrogen in regulating of inflammatory neuropeptides [90]. The function of SP has been reported to stimulate the secretion of chemokine (C-X-C motif) ligand 8 (CXCL8) and IL-1β from macrophages, which exhibits a classically pro-inflammatory profile [91]. Furthermore, activation of neurokinin 1 receptor (NK1R), the receptor of SP, also depends upon estrogen in uterine [92]. However, activation of NK1R on macrophage has a special effect on M2 polarization through activation of P13K/Akt/mTOR signaling pathway, which may be beneficial for the repair of nerve injury [93]. The function of CGRP also exhibit compromised feature to promote M2 polarization of murine peritoneal macrophages via calmodulin, PKC and PKA pathway, leading to an attenuation of inflammation and tissue damage [94]. Although neuropeptides released from peripheral nerves generate a positive feedback to amplify the inflammation between nerves and macrophages, their function on macrophages polarization suggest that more evidence is necessary to clarify the effect of estrogen on these neuropeptides in endometriosis (Fig. 3).

Implication of estrogen in regulating the interaction of nerves and macrophages in endometriosis

The role of estrogen in regulation of nerve-macrophage communication has been emphasized in the recruitment of macrophages, neurogenesis and peripheral sensitization. Targeting estrogen levels, the receptors on macrophages and nerve fibers may be a potential approach to prevent the progression of endometriosis. GnRH antagonists are popular drugs which down-regulates the release of follicle stimulating hormone (FSH) and luteinizing hormone (LH) by blocking the GnRH receptor in the pituitary cells, resulting in the suppression of ovulation [95]. Danazol is widely used and promotes endometrial atrophy by increasing free testosterone levels leading to a hypoestrogenic state [96]. A recent study developed two ER ligands, chloroindazole (CLI) and oxabicyloheptene sulfonate (OBHS), with CLI exhibiting ERβ-dependent activity and OBHS displaying ERα-preferential binding selectivity. Both ligands are optimized to have strong anti-inflammatory activity. OBHS and CLI can reduce innervation and neuron activation though down-regulation of COX2 and NGF [97]. The infiltration of macrophages is also completely prevented by the treatment with OBHS and CLI. Selective estrogen receptor modulators (SERMs) are drugs with both agonistic and antagonistic activity at the ER. It is providing new treatment strategies for endometriosis [98]. Administration of raloxifene is effective in the treatment of chronic pelvic pain in women with endometriosis [97]. The mechanism of raloxifene action relies on its anti-inflammatory properties, exemplified by a decrease in M1 monocytes, macrophage density, and the NFκB response associated with a decrease in NO, IL-1β and IL-6 production [99]. A study has shown that there is an earlier return of pelvic pain in raloxifene group compared to the placebo group, hindering further development of raloxifene [100]. For recurrence of chronic pain, accurate targeting of the ER in macrophages and nerve fibers has potential value in decreasing the occurrence of peripheral sensitization and progression of endometriosis.

Conclusion

Dysregulation of estrogen in endometriotic milieu not only mediates the recruitment of macrophages in response to the chemokines from peripheral nerves, but also promotes neurogenesis within the microenvironment by acting on macrophages. The villainous communication between macrophages and nerve fibers has been demonstrated to be enhanced by the aberrant level of estrogen, providing a hypothesis in endometriosis-associated pain (Fig. 4). However, the role of estrogen and its receptors in the progression of endometriosis still require to be clarified. ER in macrophage polarization and different nerve fibers are still controversial. Details of the recruitment and polarization of macrophages in response to estrogen need to be further investigation. Although estrogen can alter the sensitization of peripheral nerves, evidence about the molecular mechanism of estrogen on the interaction of nerves and macrophages is not fully comprehensive. In conclusion, a better understanding of estrogen in the interaction of nerves and macrophages inspires a novel insight of endometriosis-associated pain and provides a new strategy for diagnosis and a potentially valuable target for the treatment of endometriosis-associated pain.

Acknowledgements

No application.

Funding

This work was supported by: (1) Natural Science Foundation of Guangdong Province, China (Grant No. 2016A030310151); (2) Student Innovation Training Program of Sun Yat-sen University (Grant No. 201701109); (3) Special Funds for the Cultivation of Guangdong College Students’ Scientific and Technological Innovation (“Climbing Program” Special Funds, pdjh2017b0015); (4) National Natural Science Foundation of China (Youth Science fund) (Grant No. 81701416).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Mehedintu C, Plotogea MN, Ionescu S, Antonovici M. Endometriosis still a challenge. J Med Life. 2014;7(3):349–57.PubMedPubMedCentral Mehedintu C, Plotogea MN, Ionescu S, Antonovici M. Endometriosis still a challenge. J Med Life. 2014;7(3):349–57.PubMedPubMedCentral
2.
Zurück zum Zitat Taylor HS, Giudice LC, Lessey BA, Abrao MS, Kotarski J, Archer DF, Diamond MP, Surrey E, Johnson NP, Watts NB, et al. Treatment of endometriosis-associated pain with Elagolix, an Oral GnRH Antagonist. N Engl J Med. 2017;377(1):28–40.CrossRefPubMed Taylor HS, Giudice LC, Lessey BA, Abrao MS, Kotarski J, Archer DF, Diamond MP, Surrey E, Johnson NP, Watts NB, et al. Treatment of endometriosis-associated pain with Elagolix, an Oral GnRH Antagonist. N Engl J Med. 2017;377(1):28–40.CrossRefPubMed
3.
Zurück zum Zitat Dunselman GA, Vermeulen N, Becker C, Calhaz-Jorge C, D'Hooghe T, De Bie B, Heikinheimo O, Horne AW, Kiesel L, Nap A, et al. ESHRE guideline: management of women with endometriosis. Hum Reprod. 2014;29(3):400–12.CrossRefPubMed Dunselman GA, Vermeulen N, Becker C, Calhaz-Jorge C, D'Hooghe T, De Bie B, Heikinheimo O, Horne AW, Kiesel L, Nap A, et al. ESHRE guideline: management of women with endometriosis. Hum Reprod. 2014;29(3):400–12.CrossRefPubMed
4.
Zurück zum Zitat Johnson NP, Hummelshoj L, Adamson GD, Keckstein J, Taylor HS, Abrao MS, Bush D, Kiesel L, Tamimi R, Sharpe-Timms KL, et al. World endometriosis society consensus on the classification of endometriosis. Hum Reprod. 2017;32(2):315–24.CrossRefPubMed Johnson NP, Hummelshoj L, Adamson GD, Keckstein J, Taylor HS, Abrao MS, Bush D, Kiesel L, Tamimi R, Sharpe-Timms KL, et al. World endometriosis society consensus on the classification of endometriosis. Hum Reprod. 2017;32(2):315–24.CrossRefPubMed
5.
Zurück zum Zitat Chen P, Wang DB, Liang YM. Evaluation of estrogen in endometriosis patients: regulation of GATA-3 in endometrial cells and effects on Th2 cytokines. J Obstet Gynaecol Res. 2016;42(6):669–77.CrossRefPubMed Chen P, Wang DB, Liang YM. Evaluation of estrogen in endometriosis patients: regulation of GATA-3 in endometrial cells and effects on Th2 cytokines. J Obstet Gynaecol Res. 2016;42(6):669–77.CrossRefPubMed
6.
Zurück zum Zitat McKinnon B, Bersinger NA, Wotzkow C, Mueller MD. Endometriosis-associated nerve fibers, peritoneal fluid cytokine concentrations, and pain in endometriotic lesions from different locations. Fertil Steril. 2012;97(2):373–80.CrossRefPubMed McKinnon B, Bersinger NA, Wotzkow C, Mueller MD. Endometriosis-associated nerve fibers, peritoneal fluid cytokine concentrations, and pain in endometriotic lesions from different locations. Fertil Steril. 2012;97(2):373–80.CrossRefPubMed
7.
Zurück zum Zitat Tokushige N, Markham R, Russell P, Fraser IS. Nerve fibres in peritoneal endometriosis. Hum Reprod. 2006;21(11):3001–7.CrossRefPubMed Tokushige N, Markham R, Russell P, Fraser IS. Nerve fibres in peritoneal endometriosis. Hum Reprod. 2006;21(11):3001–7.CrossRefPubMed
8.
Zurück zum Zitat Laux-Biehlmann A, D'Hooghe T, Zollner TM. Menstruation pulls the trigger for inflammation and pain in endometriosis. Trends Pharmacol Sci. 2015;36(5):270–6.CrossRefPubMed Laux-Biehlmann A, D'Hooghe T, Zollner TM. Menstruation pulls the trigger for inflammation and pain in endometriosis. Trends Pharmacol Sci. 2015;36(5):270–6.CrossRefPubMed
9.
Zurück zum Zitat Zhu TH, Ding SJ, Li TT, Zhu LB, Huang XF, Zhang XM. Estrogen is an important mediator of mast cell activation in ovarian endometriomas. Reprod. 2018;155(1):73–83.CrossRef Zhu TH, Ding SJ, Li TT, Zhu LB, Huang XF, Zhang XM. Estrogen is an important mediator of mast cell activation in ovarian endometriomas. Reprod. 2018;155(1):73–83.CrossRef
10.
Zurück zum Zitat Tran LV, Tokushige N, Berbic M, Markham R, Fraser IS. Macrophages and nerve fibres in peritoneal endometriosis. Hum Reprod. 2009;24(4):835–41.CrossRefPubMed Tran LV, Tokushige N, Berbic M, Markham R, Fraser IS. Macrophages and nerve fibres in peritoneal endometriosis. Hum Reprod. 2009;24(4):835–41.CrossRefPubMed
12.
Zurück zum Zitat Scheerer C, Bauer P, Chiantera V, Sehouli J, Kaufmann A, Mechsner S. Characterization of endometriosis-associated immune cell infiltrates (EMaICI). Arch Gynecol Obstet. 2016;294(3):657–64.CrossRefPubMed Scheerer C, Bauer P, Chiantera V, Sehouli J, Kaufmann A, Mechsner S. Characterization of endometriosis-associated immune cell infiltrates (EMaICI). Arch Gynecol Obstet. 2016;294(3):657–64.CrossRefPubMed
13.
Zurück zum Zitat Greaves E, Temp J, Esnal-Zufiurre A, Mechsner S, Horne AW, Saunders PT. Estradiol is a critical mediator of macrophage-nerve cross talk in peritoneal endometriosis. Am J Pathol. 2015;185(8):2286–97.CrossRefPubMedPubMedCentral Greaves E, Temp J, Esnal-Zufiurre A, Mechsner S, Horne AW, Saunders PT. Estradiol is a critical mediator of macrophage-nerve cross talk in peritoneal endometriosis. Am J Pathol. 2015;185(8):2286–97.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Galvankar M, Singh N, Modi D. Estrogen is essential but not sufficient to induce endometriosis. J Biosci. 2017;42(2):251–63.CrossRefPubMed Galvankar M, Singh N, Modi D. Estrogen is essential but not sufficient to induce endometriosis. J Biosci. 2017;42(2):251–63.CrossRefPubMed
15.
Zurück zum Zitat Jia M, Dahlman-Wright K, Gustafsson JA. Estrogen receptor alpha and beta in health and disease. Best Pract Res Clin Endocrinol Metab. 2015;29(4):557–68.CrossRefPubMed Jia M, Dahlman-Wright K, Gustafsson JA. Estrogen receptor alpha and beta in health and disease. Best Pract Res Clin Endocrinol Metab. 2015;29(4):557–68.CrossRefPubMed
16.
Zurück zum Zitat Paterni I, Granchi C, Katzenellenbogen JA, Minutolo F. Estrogen receptors alpha (ERalpha) and beta (ERbeta): subtype-selective ligands and clinical potential. Steroids. 2014;90:13–29.CrossRefPubMed Paterni I, Granchi C, Katzenellenbogen JA, Minutolo F. Estrogen receptors alpha (ERalpha) and beta (ERbeta): subtype-selective ligands and clinical potential. Steroids. 2014;90:13–29.CrossRefPubMed
17.
Zurück zum Zitat Prossnitz ER, Barton M. Estrogen biology: new insights into GPER function and clinical opportunities. Mol Cell Endocrinol. 2014;389(1–2):71–83.CrossRefPubMedPubMedCentral Prossnitz ER, Barton M. Estrogen biology: new insights into GPER function and clinical opportunities. Mol Cell Endocrinol. 2014;389(1–2):71–83.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Shao R, Cao S, Wang X, Feng Y, Billig H. The elusive and controversial roles of estrogen and progesterone receptors in human endometriosis. Am J Transl Res. 2014;6(2):104–13.PubMedPubMedCentral Shao R, Cao S, Wang X, Feng Y, Billig H. The elusive and controversial roles of estrogen and progesterone receptors in human endometriosis. Am J Transl Res. 2014;6(2):104–13.PubMedPubMedCentral
19.
Zurück zum Zitat Izawa M, Taniguchi F, Harada T. Molecular background of estrogen receptor gene expression in Endometriotic cells. Reprod Sci. 2016;23(7):871–6.CrossRefPubMed Izawa M, Taniguchi F, Harada T. Molecular background of estrogen receptor gene expression in Endometriotic cells. Reprod Sci. 2016;23(7):871–6.CrossRefPubMed
20.
Zurück zum Zitat Lin K, Zhan H, Ma J, Xu K, Wu R, Zhou C, Lin J. Silencing of SRA1 regulates ER expression and attenuates the growth of stromal cells in ovarian endometriosis. Reprod Sci. 2017;24(6):836–43.CrossRefPubMed Lin K, Zhan H, Ma J, Xu K, Wu R, Zhou C, Lin J. Silencing of SRA1 regulates ER expression and attenuates the growth of stromal cells in ovarian endometriosis. Reprod Sci. 2017;24(6):836–43.CrossRefPubMed
21.
Zurück zum Zitat Han SJ, Jung SY, Wu SP, Hawkins SM, Park MJ, Kyo S, Qin J, Lydon JP, Tsai SY, Tsai MJ, et al. Estrogen receptor beta modulates apoptosis complexes and the Inflammasome to drive the pathogenesis of endometriosis. Cell. 2015;163(4):960–74.CrossRefPubMedPubMedCentral Han SJ, Jung SY, Wu SP, Hawkins SM, Park MJ, Kyo S, Qin J, Lydon JP, Tsai SY, Tsai MJ, et al. Estrogen receptor beta modulates apoptosis complexes and the Inflammasome to drive the pathogenesis of endometriosis. Cell. 2015;163(4):960–74.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Monsivais D, Dyson MT, Yin P, Coon JS, Navarro A, Feng G, Malpani SS, Ono M, Ercan CM, Wei JJ, et al. ERbeta- and prostaglandin E2-regulated pathways integrate cell proliferation via Ras-like and estrogen-regulated growth inhibitor in endometriosis. Mol Endocrinol. 2014;28(8):1304–15.CrossRefPubMedPubMedCentral Monsivais D, Dyson MT, Yin P, Coon JS, Navarro A, Feng G, Malpani SS, Ono M, Ercan CM, Wei JJ, et al. ERbeta- and prostaglandin E2-regulated pathways integrate cell proliferation via Ras-like and estrogen-regulated growth inhibitor in endometriosis. Mol Endocrinol. 2014;28(8):1304–15.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Zhou Y, Zeng C, Li X, Wu PL, Yin L, Yu XL, Zhou YF, Xue Q. IGF-I stimulates ERbeta and aromatase expression via IGF1R/PI3K/AKT-mediated transcriptional activation in endometriosis. J Mol Med (Berl). 2016;94(8):887–97.CrossRef Zhou Y, Zeng C, Li X, Wu PL, Yin L, Yu XL, Zhou YF, Xue Q. IGF-I stimulates ERbeta and aromatase expression via IGF1R/PI3K/AKT-mediated transcriptional activation in endometriosis. J Mol Med (Berl). 2016;94(8):887–97.CrossRef
24.
Zurück zum Zitat Zhang Q, Ding D, Liu X, Guo SW. Activated platelets induce estrogen receptor beta expression in Endometriotic stromal cells. Gynecol Obstet Investig. 2015;80(3):187–92.CrossRef Zhang Q, Ding D, Liu X, Guo SW. Activated platelets induce estrogen receptor beta expression in Endometriotic stromal cells. Gynecol Obstet Investig. 2015;80(3):187–92.CrossRef
25.
Zurück zum Zitat Monsivais D, Dyson MT, Yin P, Navarro A, Coon JT, Pavone ME, Bulun SE. Estrogen receptor beta regulates endometriotic cell survival through serum and glucocorticoid-regulated kinase activation. Fertil Steril. 2016;105(5):1266–73.CrossRefPubMedPubMedCentral Monsivais D, Dyson MT, Yin P, Navarro A, Coon JT, Pavone ME, Bulun SE. Estrogen receptor beta regulates endometriotic cell survival through serum and glucocorticoid-regulated kinase activation. Fertil Steril. 2016;105(5):1266–73.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Burns KA, Thomas SY, Hamilton KJ, Young SL, Cook DN, Korach KS. Early endometriosis in females is directed by immune-mediated estrogen receptor alpha and IL-6 cross-talk. Endocrinology. 2018;159(1):103–18.CrossRefPubMed Burns KA, Thomas SY, Hamilton KJ, Young SL, Cook DN, Korach KS. Early endometriosis in females is directed by immune-mediated estrogen receptor alpha and IL-6 cross-talk. Endocrinology. 2018;159(1):103–18.CrossRefPubMed
27.
Zurück zum Zitat Colon-Caraballo M, Garcia M, Mendoza A, Flores I. Human endometriosis tissue microarray reveals site-specific expression of estrogen receptors, progesterone receptor, and Ki67. Appl Immunohistochem Mol Morphol. 2018. Colon-Caraballo M, Garcia M, Mendoza A, Flores I. Human endometriosis tissue microarray reveals site-specific expression of estrogen receptors, progesterone receptor, and Ki67. Appl Immunohistochem Mol Morphol. 2018.
28.
Zurück zum Zitat Montagna P, Capellino S, Villaggio B, Remorgida V, Ragni N, Cutolo M, Ferrero S. Peritoneal fluid macrophages in endometriosis: correlation between the expression of estrogen receptors and inflammation. Fertil Steril. 2008;90(1):156–64.CrossRefPubMed Montagna P, Capellino S, Villaggio B, Remorgida V, Ragni N, Cutolo M, Ferrero S. Peritoneal fluid macrophages in endometriosis: correlation between the expression of estrogen receptors and inflammation. Fertil Steril. 2008;90(1):156–64.CrossRefPubMed
29.
Zurück zum Zitat Heublein S, Lenhard M, Vrekoussis T, Schoepfer J, Kuhn C, Friese K, Makrigiannakis A, Mayr D, Jeschke U. The G-protein-coupled estrogen receptor (GPER) is expressed in normal human ovaries and is upregulated in ovarian endometriosis and pelvic inflammatory disease involving the ovary. Reprod Sci. 2012;19(11):1197–204.CrossRefPubMed Heublein S, Lenhard M, Vrekoussis T, Schoepfer J, Kuhn C, Friese K, Makrigiannakis A, Mayr D, Jeschke U. The G-protein-coupled estrogen receptor (GPER) is expressed in normal human ovaries and is upregulated in ovarian endometriosis and pelvic inflammatory disease involving the ovary. Reprod Sci. 2012;19(11):1197–204.CrossRefPubMed
30.
Zurück zum Zitat Ahn SH, Monsanto SP, Miller C, Singh SS, Thomas R, Tayade C. Pathophysiology and immune dysfunction in endometriosis. Biomed Res Int. 2015;2015:795976.PubMedPubMedCentral Ahn SH, Monsanto SP, Miller C, Singh SS, Thomas R, Tayade C. Pathophysiology and immune dysfunction in endometriosis. Biomed Res Int. 2015;2015:795976.PubMedPubMedCentral
31.
Zurück zum Zitat Khan KN, Kitajima M, Inoue T, Fujishita A, Nakashima M, Masuzaki H. 17beta-estradiol and lipopolysaccharide additively promote pelvic inflammation and growth of endometriosis. Reprod Sci. 2015;22(5):585–94.CrossRefPubMedPubMedCentral Khan KN, Kitajima M, Inoue T, Fujishita A, Nakashima M, Masuzaki H. 17beta-estradiol and lipopolysaccharide additively promote pelvic inflammation and growth of endometriosis. Reprod Sci. 2015;22(5):585–94.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Cutolo M, Villaggio B, Bisso A, Sulli A, Coviello D, Dayer JM. Presence of estrogen receptors in human myeloid monocytic cells (THP-1 cell line). Eur Cytokine Netw. 2001;12(2):368–72.PubMed Cutolo M, Villaggio B, Bisso A, Sulli A, Coviello D, Dayer JM. Presence of estrogen receptors in human myeloid monocytic cells (THP-1 cell line). Eur Cytokine Netw. 2001;12(2):368–72.PubMed
33.
Zurück zum Zitat Campbell L, Emmerson E, Williams H, Saville CR, Krust A, Chambon P, Mace KA, Hardman MJ. Estrogen receptor-alpha promotes alternative macrophage activation during cutaneous repair. J Invest Dermatol. 2014;134(9):2447–57.CrossRefPubMed Campbell L, Emmerson E, Williams H, Saville CR, Krust A, Chambon P, Mace KA, Hardman MJ. Estrogen receptor-alpha promotes alternative macrophage activation during cutaneous repair. J Invest Dermatol. 2014;134(9):2447–57.CrossRefPubMed
34.
Zurück zum Zitat Heublein S, Vrekoussis T, Kuhn C, Friese K, Makrigiannakis A, Mayr D, Lenhard M, Jeschke U. Inducers of G-protein coupled estrogen receptor (GPER) in endometriosis: potential implications for macrophages and follicle maturation. J Reprod Immunol. 2013;97(1):95–103.CrossRefPubMed Heublein S, Vrekoussis T, Kuhn C, Friese K, Makrigiannakis A, Mayr D, Lenhard M, Jeschke U. Inducers of G-protein coupled estrogen receptor (GPER) in endometriosis: potential implications for macrophages and follicle maturation. J Reprod Immunol. 2013;97(1):95–103.CrossRefPubMed
35.
Zurück zum Zitat Okamoto M, Suzuki T, Mizukami Y, Ikeda T. The membrane-type estrogen receptor G-protein-coupled estrogen receptor suppresses lipopolysaccharide-induced interleukin 6 via inhibition of nuclear factor-kappa B pathway in murine macrophage cells. Anim Sci J. 2017;88(11):1870–9.CrossRefPubMed Okamoto M, Suzuki T, Mizukami Y, Ikeda T. The membrane-type estrogen receptor G-protein-coupled estrogen receptor suppresses lipopolysaccharide-induced interleukin 6 via inhibition of nuclear factor-kappa B pathway in murine macrophage cells. Anim Sci J. 2017;88(11):1870–9.CrossRefPubMed
36.
Zurück zum Zitat Barcena DAM, Oldeweme J, Arnold J, Schneider A, Mechsner S. Remodeling of estrogen-dependent sympathetic nerve fibers seems to be disturbed in adenomyosis. Fertil Steril. 2013;100(3):801–9.CrossRef Barcena DAM, Oldeweme J, Arnold J, Schneider A, Mechsner S. Remodeling of estrogen-dependent sympathetic nerve fibers seems to be disturbed in adenomyosis. Fertil Steril. 2013;100(3):801–9.CrossRef
37.
Zurück zum Zitat Zoubina EV, Smith PG. Distributions of estrogen receptors alpha and beta in sympathetic neurons of female rats: enriched expression by uterine innervation. J Neurobiol. 2002;52(1):14–23.CrossRefPubMed Zoubina EV, Smith PG. Distributions of estrogen receptors alpha and beta in sympathetic neurons of female rats: enriched expression by uterine innervation. J Neurobiol. 2002;52(1):14–23.CrossRefPubMed
38.
Zurück zum Zitat Martineti V, Picariello L, Tognarini I, Carbonell SS, Gozzini A, Azzari C, Mavilia C, Tanini A, Falchetti A, Fiorelli G, et al. ERbeta is a potent inhibitor of cell proliferation in the HCT8 human colon cancer cell line through regulation of cell cycle components. Endocr Relat Cancer. 2005;12(2):455–69.CrossRefPubMed Martineti V, Picariello L, Tognarini I, Carbonell SS, Gozzini A, Azzari C, Mavilia C, Tanini A, Falchetti A, Fiorelli G, et al. ERbeta is a potent inhibitor of cell proliferation in the HCT8 human colon cancer cell line through regulation of cell cycle components. Endocr Relat Cancer. 2005;12(2):455–69.CrossRefPubMed
39.
Zurück zum Zitat Patrone C, Andersson S, Korhonen L, Lindholm D. Estrogen receptor-dependent regulation of sensory neuron survival in developing dorsal root ganglion. Proc Natl Acad Sci U S A. 1999;96(19):10905–10.CrossRefPubMedPubMedCentral Patrone C, Andersson S, Korhonen L, Lindholm D. Estrogen receptor-dependent regulation of sensory neuron survival in developing dorsal root ganglion. Proc Natl Acad Sci U S A. 1999;96(19):10905–10.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Monica BM, Smith PG. Estrogen and female reproductive tract innervation: cellular and molecular mechanisms of autonomic neuroplasticity. Auton Neurosci. 2015;187:1–17.CrossRef Monica BM, Smith PG. Estrogen and female reproductive tract innervation: cellular and molecular mechanisms of autonomic neuroplasticity. Auton Neurosci. 2015;187:1–17.CrossRef
41.
Zurück zum Zitat Greaves E, Collins F, Esnal-Zufiaurre A, Giakoumelou S, Horne AW, Saunders PT. Estrogen receptor (ER) agonists differentially regulate neuroangiogenesis in peritoneal endometriosis via the repellent factor SLIT3. Endocrinology. 2014;155(10):4015–26.CrossRefPubMed Greaves E, Collins F, Esnal-Zufiaurre A, Giakoumelou S, Horne AW, Saunders PT. Estrogen receptor (ER) agonists differentially regulate neuroangiogenesis in peritoneal endometriosis via the repellent factor SLIT3. Endocrinology. 2014;155(10):4015–26.CrossRefPubMed
42.
Zurück zum Zitat Rowan MP, Berg KA, Roberts JL, Hargreaves KM, Clarke WP. Activation of estrogen receptor alpha enhances bradykinin signaling in peripheral sensory neurons of female rats. J Pharmacol Exp Ther. 2014;349(3):526–32.CrossRefPubMedPubMedCentral Rowan MP, Berg KA, Roberts JL, Hargreaves KM, Clarke WP. Activation of estrogen receptor alpha enhances bradykinin signaling in peripheral sensory neurons of female rats. J Pharmacol Exp Ther. 2014;349(3):526–32.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Altun I, Kurutas EB. G protein-coupled estrogen receptor levels after peripheral nerve injury in an experimental rat model. World Neurosurg. 2015;84(6):1903–6.CrossRefPubMed Altun I, Kurutas EB. G protein-coupled estrogen receptor levels after peripheral nerve injury in an experimental rat model. World Neurosurg. 2015;84(6):1903–6.CrossRefPubMed
44.
Zurück zum Zitat Wickstrom K, Stavreus-Evers A, Vercauteren O, Olovsson M, Edelstam G. Effect of lignocaine on IL-6, IL-8, and MCP-1 in peritoneal macrophages and Endometriotic stromal cells. Reprod Sci. 2017;24(3):382–92.CrossRefPubMed Wickstrom K, Stavreus-Evers A, Vercauteren O, Olovsson M, Edelstam G. Effect of lignocaine on IL-6, IL-8, and MCP-1 in peritoneal macrophages and Endometriotic stromal cells. Reprod Sci. 2017;24(3):382–92.CrossRefPubMed
45.
Zurück zum Zitat Hutter S, Heublein S, Knabl J, Andergassen U, Vrekoussis T, Makrigiannakis A, Friese K, Mayr D, Jeschke U. Macrophages: are they involved in endometriosis, abortion and preeclampsia and how? J Nippon Med Sch. 2013;80(2):97–103.CrossRefPubMed Hutter S, Heublein S, Knabl J, Andergassen U, Vrekoussis T, Makrigiannakis A, Friese K, Mayr D, Jeschke U. Macrophages: are they involved in endometriosis, abortion and preeclampsia and how? J Nippon Med Sch. 2013;80(2):97–103.CrossRefPubMed
46.
Zurück zum Zitat Tofaris GK, Patterson PH, Jessen KR, Mirsky R. Denervated Schwann cells attract macrophages by secretion of leukemia inhibitory factor (LIF) and monocyte chemoattractant protein-1 in a process regulated by interleukin-6 and LIF. J Neurosci. 2002;22(15):6696–703.CrossRefPubMed Tofaris GK, Patterson PH, Jessen KR, Mirsky R. Denervated Schwann cells attract macrophages by secretion of leukemia inhibitory factor (LIF) and monocyte chemoattractant protein-1 in a process regulated by interleukin-6 and LIF. J Neurosci. 2002;22(15):6696–703.CrossRefPubMed
47.
Zurück zum Zitat Shamash S, Reichert F, Rotshenker S. The cytokine network of Wallerian degeneration: tumor necrosis factor-alpha, interleukin-1alpha, and interleukin-1beta. J Neurosci. 2002;22(8):3052–60.CrossRefPubMed Shamash S, Reichert F, Rotshenker S. The cytokine network of Wallerian degeneration: tumor necrosis factor-alpha, interleukin-1alpha, and interleukin-1beta. J Neurosci. 2002;22(8):3052–60.CrossRefPubMed
48.
Zurück zum Zitat Namikawa K, Okamoto T, Suzuki A, Konishi H, Kiyama H. Pancreatitis-associated protein-III is a novel macrophage chemoattractant implicated in nerve regeneration. J Neurosci. 2006;26(28):7460–7.CrossRefPubMed Namikawa K, Okamoto T, Suzuki A, Konishi H, Kiyama H. Pancreatitis-associated protein-III is a novel macrophage chemoattractant implicated in nerve regeneration. J Neurosci. 2006;26(28):7460–7.CrossRefPubMed
49.
Zurück zum Zitat Simeoli R, Montague K, Jones HR, Castaldi L, Chambers D, Kelleher JH, Vacca V, Pitcher T, Grist J, Al-Ahdal H, et al. Exosomal cargo including microRNA regulates sensory neuron to macrophage communication after nerve trauma. Nat Commun. 2017;8(1):1778.CrossRefPubMedPubMedCentral Simeoli R, Montague K, Jones HR, Castaldi L, Chambers D, Kelleher JH, Vacca V, Pitcher T, Grist J, Al-Ahdal H, et al. Exosomal cargo including microRNA regulates sensory neuron to macrophage communication after nerve trauma. Nat Commun. 2017;8(1):1778.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Peng Z, Pan L, Niu Z, Li W, Dang X, Wan L, Zhang R, Yang S. Identification of microRNAs as potential biomarkers for lung adenocarcinoma using integrating genomics analysis. Oncotarget. 2017;8(38):64143–56.CrossRefPubMedPubMedCentral Peng Z, Pan L, Niu Z, Li W, Dang X, Wan L, Zhang R, Yang S. Identification of microRNAs as potential biomarkers for lung adenocarcinoma using integrating genomics analysis. Oncotarget. 2017;8(38):64143–56.CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Rhee I. Diverse macrophages polarization in tumor microenvironment. Arch Pharm Res. 2016;39(11):1588–96.CrossRefPubMed Rhee I. Diverse macrophages polarization in tumor microenvironment. Arch Pharm Res. 2016;39(11):1588–96.CrossRefPubMed
52.
Zurück zum Zitat Moghaddam AS, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, Seifi B, Mohammadi A, Afshari JT, Sahebkar A. Macrophage plasticity, polarization and function in health and disease. J Cell Physiol. 2018;233(9):6425–40.CrossRef Moghaddam AS, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, Seifi B, Mohammadi A, Afshari JT, Sahebkar A. Macrophage plasticity, polarization and function in health and disease. J Cell Physiol. 2018;233(9):6425–40.CrossRef
53.
Zurück zum Zitat Jensen AL, Collins J, Shipman EP, Wira CR, Guyre PM, Pioli PA. A subset of human uterine endometrial macrophages is alternatively activated. Am J Reprod Immunol. 2012;68(5):374–86.CrossRefPubMedPubMedCentral Jensen AL, Collins J, Shipman EP, Wira CR, Guyre PM, Pioli PA. A subset of human uterine endometrial macrophages is alternatively activated. Am J Reprod Immunol. 2012;68(5):374–86.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Wang Y, Fu Y, Xue S, Ai A, Chen H, Lyu Q, Kuang Y. The M2 polarization of macrophage induced by fractalkine in the endometriotic milieu enhances invasiveness of endometrial stromal cells. Int J Clin Exp Pathol. 2014;7(1):194–203.PubMed Wang Y, Fu Y, Xue S, Ai A, Chen H, Lyu Q, Kuang Y. The M2 polarization of macrophage induced by fractalkine in the endometriotic milieu enhances invasiveness of endometrial stromal cells. Int J Clin Exp Pathol. 2014;7(1):194–203.PubMed
55.
Zurück zum Zitat Kwon MJ, Shin HY, Cui Y, Kim H, Thi AH, Choi JY, Kim EY, Hwang DH, Kim BG. CCL2 mediates neuron-macrophage interactions to drive Proregenerative macrophage activation following preconditioning injury. J Neurosci. 2015;35(48):15934–47.CrossRefPubMed Kwon MJ, Shin HY, Cui Y, Kim H, Thi AH, Choi JY, Kim EY, Hwang DH, Kim BG. CCL2 mediates neuron-macrophage interactions to drive Proregenerative macrophage activation following preconditioning injury. J Neurosci. 2015;35(48):15934–47.CrossRefPubMed
56.
Zurück zum Zitat Wang Y, Chen H, Wang N, Guo H, Fu Y, Xue S, Ai A, Lyu Q, Kuang Y. Combined 17beta-estradiol with TCDD promotes M2 polarization of macrophages in the Endometriotic milieu with aid of the interaction between endometrial stromal cells and macrophages. PLoS One. 2015;10(5):e125559. Wang Y, Chen H, Wang N, Guo H, Fu Y, Xue S, Ai A, Lyu Q, Kuang Y. Combined 17beta-estradiol with TCDD promotes M2 polarization of macrophages in the Endometriotic milieu with aid of the interaction between endometrial stromal cells and macrophages. PLoS One. 2015;10(5):e125559.
57.
Zurück zum Zitat Xing D, Oparil S, Yu H, Gong K, Feng W, Black J, Chen YF, Nozell S. Estrogen modulates NFkappaB signaling by enhancing IkappaBalpha levels and blocking p65 binding at the promoters of inflammatory genes via estrogen receptor-beta. PLoS One. 2012;7(6):e36890.CrossRefPubMedPubMedCentral Xing D, Oparil S, Yu H, Gong K, Feng W, Black J, Chen YF, Nozell S. Estrogen modulates NFkappaB signaling by enhancing IkappaBalpha levels and blocking p65 binding at the promoters of inflammatory genes via estrogen receptor-beta. PLoS One. 2012;7(6):e36890.CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Yang W, Lu Y, Xu Y, Xu L, Zheng W, Wu Y, Li L, Shen P. Estrogen represses hepatocellular carcinoma (HCC) growth via inhibiting alternative activation of tumor-associated macrophages (TAMs). J Biol Chem. 2012;287(48):40140–9.CrossRefPubMedPubMedCentral Yang W, Lu Y, Xu Y, Xu L, Zheng W, Wu Y, Li L, Shen P. Estrogen represses hepatocellular carcinoma (HCC) growth via inhibiting alternative activation of tumor-associated macrophages (TAMs). J Biol Chem. 2012;287(48):40140–9.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Liang Y, Yao S. Potential role of estrogen in maintaining the imbalanced sympathetic and sensory innervation in endometriosis. Mol Cell Endocrinol. 2016;424:42–9.CrossRefPubMed Liang Y, Yao S. Potential role of estrogen in maintaining the imbalanced sympathetic and sensory innervation in endometriosis. Mol Cell Endocrinol. 2016;424:42–9.CrossRefPubMed
60.
Zurück zum Zitat Morotti M, Vincent K, Brawn J, Zondervan KT, Becker CM. Peripheral changes in endometriosis-associated pain. Hum Reprod Update. 2014;20(5):717–36.CrossRefPubMedPubMedCentral Morotti M, Vincent K, Brawn J, Zondervan KT, Becker CM. Peripheral changes in endometriosis-associated pain. Hum Reprod Update. 2014;20(5):717–36.CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Kanda N, Watanabe S. 17Beta-estradiol enhances the production of nerve growth factor in THP-1-derived macrophages or peripheral blood monocyte-derived macrophages. J Invest Dermatol. 2003;121(4):771–80.CrossRefPubMed Kanda N, Watanabe S. 17Beta-estradiol enhances the production of nerve growth factor in THP-1-derived macrophages or peripheral blood monocyte-derived macrophages. J Invest Dermatol. 2003;121(4):771–80.CrossRefPubMed
63.
Zurück zum Zitat Khan N, Smith MT. Neurotrophins and neuropathic pain: role in pathobiology. MOLECULES. 2015;20(6):10657–88.CrossRefPubMed Khan N, Smith MT. Neurotrophins and neuropathic pain: role in pathobiology. MOLECULES. 2015;20(6):10657–88.CrossRefPubMed
64.
Zurück zum Zitat Ceni C, Unsain N, Zeinieh MP, Barker PA. Neurotrophins in the regulation of cellular survival and death. Handb Exp Pharmacol. 2014;220:193–221.CrossRefPubMed Ceni C, Unsain N, Zeinieh MP, Barker PA. Neurotrophins in the regulation of cellular survival and death. Handb Exp Pharmacol. 2014;220:193–221.CrossRefPubMed
65.
Zurück zum Zitat Bjorling DE, Beckman M, Clayton MK, Wang ZY. Modulation of nerve growth factor in peripheral organs by estrogen and progesterone. Neuroscience. 2002;110(1):155–67.CrossRefPubMed Bjorling DE, Beckman M, Clayton MK, Wang ZY. Modulation of nerve growth factor in peripheral organs by estrogen and progesterone. Neuroscience. 2002;110(1):155–67.CrossRefPubMed
66.
Zurück zum Zitat Kohn J, Aloyz RS, Toma JG, Haak-Frendscho M, Miller FD. Functionally antagonistic interactions between the TrkA and p75 neurotrophin receptors regulate sympathetic neuron growth and target innervation. J Neurosci. 1999;19(13):5393–408.CrossRefPubMed Kohn J, Aloyz RS, Toma JG, Haak-Frendscho M, Miller FD. Functionally antagonistic interactions between the TrkA and p75 neurotrophin receptors regulate sympathetic neuron growth and target innervation. J Neurosci. 1999;19(13):5393–408.CrossRefPubMed
67.
Zurück zum Zitat Koncina E, Roth L, Gonthier B, Bagnard D. Role of semaphorins during axon growth and guidance. Adv Exp Med Biol. 2007;621:50–64.CrossRefPubMed Koncina E, Roth L, Gonthier B, Bagnard D. Role of semaphorins during axon growth and guidance. Adv Exp Med Biol. 2007;621:50–64.CrossRefPubMed
68.
Zurück zum Zitat Liang Y, Wang W, Huang J, Tan H, Liu T, Shang C, Liu D, Guo L, Yao S. Potential role of Semaphorin 3A and its receptors in regulating aberrant sympathetic innervation in peritoneal and deep infiltrating endometriosis. PLoS One. 2015;10(12):e146027. Liang Y, Wang W, Huang J, Tan H, Liu T, Shang C, Liu D, Guo L, Yao S. Potential role of Semaphorin 3A and its receptors in regulating aberrant sympathetic innervation in peritoneal and deep infiltrating endometriosis. PLoS One. 2015;10(12):e146027.
69.
Zurück zum Zitat Sawano S, Suzuki T, Do MK, Ohtsubo H, Mizunoya W, Ikeuchi Y, Tatsumi R. Supplementary immunocytochemistry of hepatocyte growth factor production in activated macrophages early in muscle regeneration. Anim Sci J. 2014;85(12):994–1000.CrossRefPubMed Sawano S, Suzuki T, Do MK, Ohtsubo H, Mizunoya W, Ikeuchi Y, Tatsumi R. Supplementary immunocytochemistry of hepatocyte growth factor production in activated macrophages early in muscle regeneration. Anim Sci J. 2014;85(12):994–1000.CrossRefPubMed
70.
Zurück zum Zitat Scheerer C, Frangini S, Chiantera V, Mechsner S. Reduced sympathetic innervation in endometriosis is associated to Semaphorin 3C and 3F expression. Mol Neurobiol. 2017;54(7):5131–41.CrossRefPubMed Scheerer C, Frangini S, Chiantera V, Mechsner S. Reduced sympathetic innervation in endometriosis is associated to Semaphorin 3C and 3F expression. Mol Neurobiol. 2017;54(7):5131–41.CrossRefPubMed
71.
Zurück zum Zitat Richeri A, Chalar C, Martinez G, Greif G, Bianchimano P, Brauer MM. Estrogen up-regulation of semaphorin 3F correlates with sympathetic denervation of the rat uterus. Auton Neurosci. 2011;164(1–2):43–50.CrossRefPubMed Richeri A, Chalar C, Martinez G, Greif G, Bianchimano P, Brauer MM. Estrogen up-regulation of semaphorin 3F correlates with sympathetic denervation of the rat uterus. Auton Neurosci. 2011;164(1–2):43–50.CrossRefPubMed
72.
Zurück zum Zitat Khan KN, Masuzaki H, Fujishita A, Kitajima M, Sekine I, Matsuyama T, Ishimaru T. Estrogen and progesterone receptor expression in macrophages and regulation of hepatocyte growth factor by ovarian steroids in women with endometriosis. Hum Reprod. 2005;20(7):2004–13.CrossRefPubMed Khan KN, Masuzaki H, Fujishita A, Kitajima M, Sekine I, Matsuyama T, Ishimaru T. Estrogen and progesterone receptor expression in macrophages and regulation of hepatocyte growth factor by ovarian steroids in women with endometriosis. Hum Reprod. 2005;20(7):2004–13.CrossRefPubMed
73.
Zurück zum Zitat Khan KN, Kitajima M, Hiraki K, Fujishita A, Sekine I, Ishimaru T, Masuzaki H. Immunopathogenesis of pelvic endometriosis: role of hepatocyte growth factor, macrophages and ovarian steroids. Am J Reprod Immunol. 2008;60(5):383–404.CrossRefPubMed Khan KN, Kitajima M, Hiraki K, Fujishita A, Sekine I, Ishimaru T, Masuzaki H. Immunopathogenesis of pelvic endometriosis: role of hepatocyte growth factor, macrophages and ovarian steroids. Am J Reprod Immunol. 2008;60(5):383–404.CrossRefPubMed
74.
Zurück zum Zitat Sakaguchi S, Shono J, Suzuki T, Sawano S, Anderson JE, Do MK, Ohtsubo H, Mizunoya W, Sato Y, Nakamura M, et al. Implication of anti-inflammatory macrophages in regenerative moto-neuritogenesis: promotion of myoblast migration and neural chemorepellent semaphorin 3A expression in injured muscle. Int J Biochem Cell Biol. 2014;54:272–85.CrossRefPubMed Sakaguchi S, Shono J, Suzuki T, Sawano S, Anderson JE, Do MK, Ohtsubo H, Mizunoya W, Sato Y, Nakamura M, et al. Implication of anti-inflammatory macrophages in regenerative moto-neuritogenesis: promotion of myoblast migration and neural chemorepellent semaphorin 3A expression in injured muscle. Int J Biochem Cell Biol. 2014;54:272–85.CrossRefPubMed
75.
Zurück zum Zitat Sondell M, Sundler F, Kanje M. Vascular endothelial growth factor is a neurotrophic factor which stimulates axonal outgrowth through the flk-1 receptor. Eur J Neurosci. 2000;12(12):4243–54.CrossRefPubMed Sondell M, Sundler F, Kanje M. Vascular endothelial growth factor is a neurotrophic factor which stimulates axonal outgrowth through the flk-1 receptor. Eur J Neurosci. 2000;12(12):4243–54.CrossRefPubMed
76.
Zurück zum Zitat Steinthorsdottir V, Thorleifsson G, Aradottir K, Feenstra B, Sigurdsson A, Stefansdottir L, Kristinsdottir AM, Zink F, Halldorsson GH, Munk NN, et al. Common variants upstream of KDR encoding VEGFR2 and in TTC39B associate with endometriosis. Nat Commun. 2016;7:12350.CrossRefPubMedPubMedCentral Steinthorsdottir V, Thorleifsson G, Aradottir K, Feenstra B, Sigurdsson A, Stefansdottir L, Kristinsdottir AM, Zink F, Halldorsson GH, Munk NN, et al. Common variants upstream of KDR encoding VEGFR2 and in TTC39B associate with endometriosis. Nat Commun. 2016;7:12350.CrossRefPubMedPubMedCentral
77.
78.
Zurück zum Zitat Cattin AL, Burden JJ, Van Emmenis L, Mackenzie FE, Hoving JJ, Garcia CN, Guo Y, McLaughlin M, Rosenberg LH, Quereda V, et al. Macrophage-induced blood vessels guide Schwann cell-mediated regeneration of peripheral nerves. Cell. 2015;162(5):1127–39.CrossRefPubMedPubMedCentral Cattin AL, Burden JJ, Van Emmenis L, Mackenzie FE, Hoving JJ, Garcia CN, Guo Y, McLaughlin M, Rosenberg LH, Quereda V, et al. Macrophage-induced blood vessels guide Schwann cell-mediated regeneration of peripheral nerves. Cell. 2015;162(5):1127–39.CrossRefPubMedPubMedCentral
79.
Zurück zum Zitat Sacco K, Portelli M, Pollacco J, Schembri-Wismayer P, Calleja-Agius J. The role of prostaglandin E2 in endometriosis. Gynecol Endocrinol. 2012;28(2):134–8.CrossRefPubMed Sacco K, Portelli M, Pollacco J, Schembri-Wismayer P, Calleja-Agius J. The role of prostaglandin E2 in endometriosis. Gynecol Endocrinol. 2012;28(2):134–8.CrossRefPubMed
80.
Zurück zum Zitat Le AW, Shan L, Wang ZH, Dai XY, Xiao TH, Zuo R. Effects of icariin on the expression of ER, VEGF, and KDR in the endometrial cells of thin endometrium. Genet Mol Res. 2015;14(3):11250–8.CrossRefPubMed Le AW, Shan L, Wang ZH, Dai XY, Xiao TH, Zuo R. Effects of icariin on the expression of ER, VEGF, and KDR in the endometrial cells of thin endometrium. Genet Mol Res. 2015;14(3):11250–8.CrossRefPubMed
81.
Zurück zum Zitat Czyzyk A, Podfigurna A, Szeliga A, Meczekalski B. Update on endometriosis pathogenesis. Minerva Ginecol. 2017;69(5):447–61.PubMed Czyzyk A, Podfigurna A, Szeliga A, Meczekalski B. Update on endometriosis pathogenesis. Minerva Ginecol. 2017;69(5):447–61.PubMed
82.
Zurück zum Zitat McKinnon BD, Bertschi D, Bersinger NA, Mueller MD. Inflammation and nerve fiber interaction in endometriotic pain. Trends Endocrinol Metab. 2015;26(1):1–10.CrossRefPubMed McKinnon BD, Bertschi D, Bersinger NA, Mueller MD. Inflammation and nerve fiber interaction in endometriotic pain. Trends Endocrinol Metab. 2015;26(1):1–10.CrossRefPubMed
83.
Zurück zum Zitat Bohonyi N, Pohoczky K, Szalontai B, Perkecz A, Kovacs K, Kajtar B, Orban L, Varga T, Szegedi S, Bodis J, et al. Local upregulation of transient receptor potential ankyrin 1 and transient receptor potential vanilloid 1 ion channels in rectosigmoid deep infiltrating endometriosis. Mol Pain. 2017;13:2071457956.CrossRef Bohonyi N, Pohoczky K, Szalontai B, Perkecz A, Kovacs K, Kajtar B, Orban L, Varga T, Szegedi S, Bodis J, et al. Local upregulation of transient receptor potential ankyrin 1 and transient receptor potential vanilloid 1 ion channels in rectosigmoid deep infiltrating endometriosis. Mol Pain. 2017;13:2071457956.CrossRef
84.
Zurück zum Zitat Jin X, Gereau RT. Acute p38-mediated modulation of tetrodotoxin-resistant sodium channels in mouse sensory neurons by tumor necrosis factor-alpha. J Neurosci. 2006;26(1):246–55.CrossRefPubMed Jin X, Gereau RT. Acute p38-mediated modulation of tetrodotoxin-resistant sodium channels in mouse sensory neurons by tumor necrosis factor-alpha. J Neurosci. 2006;26(1):246–55.CrossRefPubMed
85.
Zurück zum Zitat Jung H, Toth PT, White FA, Miller RJ. Monocyte chemoattractant protein-1 functions as a neuromodulator in dorsal root ganglia neurons. J Neurochem. 2008;104(1):254–63.PubMed Jung H, Toth PT, White FA, Miller RJ. Monocyte chemoattractant protein-1 functions as a neuromodulator in dorsal root ganglia neurons. J Neurochem. 2008;104(1):254–63.PubMed
86.
Zurück zum Zitat Belkouch M, Dansereau MA, Reaux-Le GA, Van Steenwinckel J, Beaudet N, Chraibi A, Melik-Parsadaniantz S, Sarret P. The chemokine CCL2 increases Nav1.8 sodium channel activity in primary sensory neurons through a Gbetagamma-dependent mechanism. J Neurosci. 2011;31(50):18381–90.CrossRefPubMed Belkouch M, Dansereau MA, Reaux-Le GA, Van Steenwinckel J, Beaudet N, Chraibi A, Melik-Parsadaniantz S, Sarret P. The chemokine CCL2 increases Nav1.8 sodium channel activity in primary sensory neurons through a Gbetagamma-dependent mechanism. J Neurosci. 2011;31(50):18381–90.CrossRefPubMed
87.
Zurück zum Zitat Greaves E, Grieve K, Horne AW, Saunders PT. Elevated peritoneal expression and estrogen regulation of nociceptive ion channels in endometriosis. J Clin Endocrinol Metab. 2014;99(9):E1738–43.CrossRefPubMedPubMedCentral Greaves E, Grieve K, Horne AW, Saunders PT. Elevated peritoneal expression and estrogen regulation of nociceptive ion channels in endometriosis. J Clin Endocrinol Metab. 2014;99(9):E1738–43.CrossRefPubMedPubMedCentral
88.
Zurück zum Zitat Lian YL, Cheng MJ, Zhang XX, Wang L. Elevated expression of transient receptor potential vanilloid type 1 in dorsal root ganglia of rats with endometriosis. Mol Med Rep. 2017;16(2):1920–6.CrossRefPubMedPubMedCentral Lian YL, Cheng MJ, Zhang XX, Wang L. Elevated expression of transient receptor potential vanilloid type 1 in dorsal root ganglia of rats with endometriosis. Mol Med Rep. 2017;16(2):1920–6.CrossRefPubMedPubMedCentral
89.
Zurück zum Zitat Kumar V, et al. Estrogen Modulates Corneal Nociception and Maintains Corneal Homeostasis in Rat Eye. Cornea. 2018;37(4):508–14.CrossRefPubMed Kumar V, et al. Estrogen Modulates Corneal Nociception and Maintains Corneal Homeostasis in Rat Eye. Cornea. 2018;37(4):508–14.CrossRefPubMed
90.
Zurück zum Zitat Pota V, Quagliariello V, Armenia E, Aurilio C, Passavanti MB, Sansone P, Iannotti M, Catauro M, Coaccioli S, Barbarisi M, et al. CGRP and visceral pain: the role of sex hormones in in vitro experiment. J Cell Biochem. 2017;118(3):510–7.CrossRefPubMed Pota V, Quagliariello V, Armenia E, Aurilio C, Passavanti MB, Sansone P, Iannotti M, Catauro M, Coaccioli S, Barbarisi M, et al. CGRP and visceral pain: the role of sex hormones in in vitro experiment. J Cell Biochem. 2017;118(3):510–7.CrossRefPubMed
91.
Zurück zum Zitat Sipka A, Langner K, Seyfert HM, Schuberth HJ. Substance P alters the in vitro LPS responsiveness of bovine monocytes and blood-derived macrophages. Vet Immunol Immunopathol. 2010;136(3–4):219–26.CrossRefPubMed Sipka A, Langner K, Seyfert HM, Schuberth HJ. Substance P alters the in vitro LPS responsiveness of bovine monocytes and blood-derived macrophages. Vet Immunol Immunopathol. 2010;136(3–4):219–26.CrossRefPubMed
92.
Zurück zum Zitat Simeonidis S, Castagliuolo I, Pan A, Liu J, Wang CC, Mykoniatis A, Pasha A, Valenick L, Sougioultzis S, Zhao D, et al. Regulation of the NK-1 receptor gene expression in human macrophage cells via an NF-kappa B site on its promoter. Proc Natl Acad Sci U S A. 2003;100(5):2957–62.CrossRefPubMedPubMedCentral Simeonidis S, Castagliuolo I, Pan A, Liu J, Wang CC, Mykoniatis A, Pasha A, Valenick L, Sougioultzis S, Zhao D, et al. Regulation of the NK-1 receptor gene expression in human macrophage cells via an NF-kappa B site on its promoter. Proc Natl Acad Sci U S A. 2003;100(5):2957–62.CrossRefPubMedPubMedCentral
93.
Zurück zum Zitat Lim JE, Chung E, Son Y. A neuropeptide, substance-P, directly induces tissue-repairing M2 like macrophages by activating the PI3K/Akt/mTOR pathway even in the presence of IFNgamma. Sci Rep. 2017;7(1):9417.CrossRefPubMedPubMedCentral Lim JE, Chung E, Son Y. A neuropeptide, substance-P, directly induces tissue-repairing M2 like macrophages by activating the PI3K/Akt/mTOR pathway even in the presence of IFNgamma. Sci Rep. 2017;7(1):9417.CrossRefPubMedPubMedCentral
94.
Zurück zum Zitat Duan JX, Zhou Y, Zhou AY, Guan XX, Liu T, Yang HH, Xie H, Chen P. Calcitonin gene-related peptide exerts anti-inflammatory property through regulating murine macrophages polarization in vitro. Mol Immunol. 2017;91:105–13.CrossRefPubMed Duan JX, Zhou Y, Zhou AY, Guan XX, Liu T, Yang HH, Xie H, Chen P. Calcitonin gene-related peptide exerts anti-inflammatory property through regulating murine macrophages polarization in vitro. Mol Immunol. 2017;91:105–13.CrossRefPubMed
95.
96.
Zurück zum Zitat Goenka L, George M, Sen M. A peek into the drug development scenario of endometriosis - a systematic review. Biomed Pharmacother. 2017;90:575–85.CrossRefPubMed Goenka L, George M, Sen M. A peek into the drug development scenario of endometriosis - a systematic review. Biomed Pharmacother. 2017;90:575–85.CrossRefPubMed
97.
Zurück zum Zitat Zhao Y, Gong P, Chen Y, Nwachukwu JC, Srinivasan S, Ko C, Bagchi MK, Taylor RN, Korach KS, Nettles KW, et al. Dual suppression of estrogenic and inflammatory activities for targeting of endometriosis. Sci Transl Med. 2015;7(271):271r–9r.CrossRef Zhao Y, Gong P, Chen Y, Nwachukwu JC, Srinivasan S, Ko C, Bagchi MK, Taylor RN, Korach KS, Nettles KW, et al. Dual suppression of estrogenic and inflammatory activities for targeting of endometriosis. Sci Transl Med. 2015;7(271):271r–9r.CrossRef
98.
99.
Zurück zum Zitat Poirier AA, Cote M, Bourque M, Morissette M, Di Paolo T, Soulet D. Neuroprotective and immunomodulatory effects of raloxifene in the myenteric plexus of a mouse model of Parkinson's disease. Neurobiol Aging. 2016;48:61–71.CrossRefPubMed Poirier AA, Cote M, Bourque M, Morissette M, Di Paolo T, Soulet D. Neuroprotective and immunomodulatory effects of raloxifene in the myenteric plexus of a mouse model of Parkinson's disease. Neurobiol Aging. 2016;48:61–71.CrossRefPubMed
100.
Zurück zum Zitat Naqvi H, Sakr S, Presti T, Krikun G, Komm B, Taylor HS. Treatment with bazedoxifene and conjugated estrogens results in regression of endometriosis in a murine model. Biol Reprod. 2014;90(6):121.CrossRefPubMedPubMedCentral Naqvi H, Sakr S, Presti T, Krikun G, Komm B, Taylor HS. Treatment with bazedoxifene and conjugated estrogens results in regression of endometriosis in a murine model. Biol Reprod. 2014;90(6):121.CrossRefPubMedPubMedCentral
Metadaten
Titel
Villainous role of estrogen in macrophage-nerve interaction in endometriosis
verfasst von
Yanchun Liang
Hongyu Xie
Jinjie Wu
Duo Liu
Shuzhong Yao
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Reproductive Biology and Endocrinology / Ausgabe 1/2018
Elektronische ISSN: 1477-7827
DOI
https://doi.org/10.1186/s12958-018-0441-z

Weitere Artikel der Ausgabe 1/2018

Reproductive Biology and Endocrinology 1/2018 Zur Ausgabe

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.