Integrating modern approaches to pathogenetic concepts of malignant transformation of endometriosis

  • Authors:
    • Hiroshi Kobayashi
    • Yuki Yamada
    • Naoki Kawahara
    • Kenji Ogawa
    • Chiharu Yoshimoto
  • View Affiliations

  • Published online on: December 21, 2018     https://doi.org/10.3892/or.2018.6946
  • Pages: 1729-1738
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In the present study, we summarize the role of the shared and independent (epi)genetic background between endometrioid carcinoma (EC) and clear cell carcinoma (CCC), two histological subtypes of endometriosis-associated ovarian cancer (EAOC). Using the PubMed database, we conducted a literature review of various studies related to the malignant transformation of endometriosis. Both endometriosis and EAOC face potential environmental hazards, including hemoglobin (Hb), heme and free iron, which induces DNA damage and mutations. Although EC is distinguished from CCC due to different morphologies, both represent common environmental profiles and maintain the similar (epi)genomic abnormalities with multiple overlaps and share similar molecular signatures. By contrast, EAOC also has disease-specific gene signatures corresponding with each histological subtype: Estrogen receptor promotes EC cell proliferation (‘go’) and hepatocyte nuclear factor-1β (HNF-1β) induces CCC cell cycle arrest (‘stop’) under oxidative stress conditions. This model underscores a subtype-dependent ‘go or stop’ dichotomy, possibly through better ability to adapt in a changing environment. It was found that cyst fluid Hb and iron concentrations were significantly lower in EAOC when compared to benign ovarian endometrioma (OE), supporting the hypothesis that the redox imbalance plays a key role in the pathogenesis of EAOC. There are at least two phases of iron carcinogenesis and tumor progression: The initial wave of iron-induced oxidative stress and DNA mutations would be followed by the second big wave of subsequent synthesis of the antioxidants, which diminishes cellular oxidative stress capacity, increases apoptosis resistance and promotes tumor initiation and progression. Special emphasis is given to novel pathophysiological concepts of malignant transformation of endometriosis.

Introduction

Endometriosis is defined as the presence of the ectopic implantation of endometrial glands and stroma at extra-uterine sites. Endometriosis is a common, chronic inflammatory disease that affects ~10% of women of reproductive age (1,2). The common symptoms of this disease are dysmenorrhea, dyspareunia, chronic pelvic pain and infertility (1,2). Epidemiologically, endometriosis has been reported to increase the risk of certain types of malignancies, particularly for ovarian endometrioid carcinoma (EC), clear cell carcinoma (CCC), low-grade serous carcinoma and seromucinous neoplasms (36). CCC and EC of the ovary are the two most common types of ovarian cancer, which arise from endometriosis (46). Endometriosis is found in approximately 20% of EC and CCC cases, presents adjacent to the tumor, and has direct topological continuity with the carcinoma (3). Patients with endometriosis-associated ovarian cancer (EAOC) belong to the relatively younger-aged population, and have early-stage and low histological grade tumors compared with non-EAOC patients (7). EAOC tumors frequently occur in perimenopausal and early postmenopausal women. Ovarian cancer is known to develop in approximately 1% of women with endometriosis (4). Endometriosis may be related to an increased risk of EAOC; however, the underlying mechanism remains largely unknown. Over the past decade, a dramatic shift has occurred in our understanding of the pathophysiology of EAOC.

The aim of the present study was to provide an overview of the current pathophysiological concepts of the malignant transformation of endometriosis. We summarize recent knowledge about the role of the shared and independent (epi)genetic background between EC and CCC, and the current hypotheses regarding the pathophysiology of the malignant processes.

Data collection methods

A computerized literature search was conducted to identify relevant studies reported in the English language. We collected a comprehensive literature search from the PubMed and Embase databases up to April, 2018, combining the keywords ‘endometriosis’, ‘endometriosis-associated ovarian cancer’, ‘endometrioid carcinoma’, ‘clear cell carcinoma’, ‘pathogenesis’, ‘carcinogenesis’, ‘oxidative stress’, ‘hemoglobin’, ‘iron’, ‘inflammation’, ‘endothelial cells’, ‘extracellular matrix’ and ‘microenvironment’. A variety of combinations of these terms were used, depending on which database was searched. Furthermore, the references of each article were searched to identify potentially relevant studies. Publications of original studies and review articles were included, while those documenting opinions, points of view or anecdotes were discarded.

Results

The mechanisms underlying the malignant transformation of endometriosis

The gene expression profile-based clustering divided ovarian cancer into two groups, type I and type II, which is generally based on the potential clinical and translational value of the dualistic model of ovarian carcinogenesis (8,9). Type I ovarian cancer consists of patients with low-grade serous, mucinous, EC, CCC and slow-growing tumors, while type II is composed of patients with rapidly-growing high-grade serous carcinoma (HGSC) and highly aggressive malignancies (8,9). EAOC belongs to the type I category and consists of two major subtypes originating from EC and CCC, which exhibits different pathological and clinical features, characterized by unique morphologies and responses to treatment (6).

EC may occur during an estrogenic mode of action due to the observed induction of estrogen receptor (ESR) isoforms (1014). Estrogen is considered to be involved in ovarian cancer progression (15). The Wnt/β-catenin signaling pathway regulated by estrogen is highly activated in EC and inhibits oxidative stress-induced cell apoptosis (1618). By contrast, estrogens are known to produce reactive oxygen species (ROS) and are implicated in cellular carcinogenesis, as chronic oxidative stress promotes cell growth, survival and the tumorigenic potential of breast cancer cells (19). In the present study, we provide an update on the recent advances in the understanding of the reduction-oxidation (redox)-related molecular signaling and imbalance in the cellular redox state in malignant transformation of endometriosis (8,2027). The pathogenesis of the malignant transformation of endometriosis remains obscure; however, the results of several studies support the hypothesis that the redox imbalance, inflammatory/immune response, cell cycle regulation and hormone activity are the deregulated functions and act in a dynamic epigenetic network (20,22,23,24).

Redox imbalance: Possible unexpected results

Repeated episodes of hemorrhage occur in endometriosis throughout menstruation (23). Red blood cells accumulate in ovarian endometrioma (OE) and in the pelvic cavity through retrograde menstruation. The destruction of red blood cells leads to the release of Hb, heme and free iron (22,28). While Hb provides life-sustaining oxygen delivery, extracellular free Hb produces toxic heme degradation products and is a source of ROS due to inherent peroxidase activity (22,28,29). These findings are consistent with and are supported by in vitro experimental data (30) and in vivo clinical data (31). Yamaguchi et al presented, for the first time, that the iron-induced persistent oxidative stress within the endometriotic cyst leads to dynamic changes in the oxidative environment, which may play a crucial role in the process of endometriosis carcinogenesis (30). The authors reported that patients with endometriotic cysts had significantly higher cyst fluid concentration of free iron (100.9 mmol/l = 5,635 mg/l), compared to those with non-endometriotic cysts (0.075 mmol/l = 4.19 mg/l) (30). Free iron concentrations in CCC (4.27 mmol/l = 238 mg/l) were 20-fold lower than those in endometriotic cysts (30). Since free iron has a propensity to induce oxidative stress, DNA damage, protein modification and lipid peroxidation, we hypothesized that patients with EAOC would have much higher levels of iron-related compounds compared with those with benign OE. Therefore, Yoshimoto et al extensively investigated cyst fluid levels of iron-related compounds in benign OE and EAOC (31). The median ± SD concentrations of total iron, heme iron and free iron for OE and EAOC cysts were 244.4±204.9 mg/l vs. 14.2±36.6 mg/l (total iron), 303.9±324.4 mg/l vs. 27.6±53.4 mg/l (heme iron), and 13.5±16.2 mg/l vs. 3.9±2.7 mg/l (free iron), respectively (31). The concentrations of total iron, heme and free iron in EAOC were 17-, 11- and 3-fold lower than those in OE, respectively (31). There are no significant differences in cyst fluid concentrations of iron-related compounds between patients with CCC and those with EC. Several assays for the measurement of iron-related compounds are available: In a previous study, there was a significant difference in the cyst fluid iron levels between the two methods (30,31), which may be due to the different chelate colorimetric assay methods and differences in their analytical performances. Notwithstanding these limitations, patients with EAOC had much lower levels of iron-related compounds compared with those with benign OE.

Hemoglobin, heme and free iron in endometriotic cysts can lead to distortion in the homeostatic redox balance, the so-called redox imbalance (22). Total iron is composed of heme iron and nonheme iron (free catalytic form of iron, Fe2+). Free iron is labile and catalyzes the Fenton chemical reaction, resulting in the generation of hydroxyl radical (OH) as follows: Fe2+ + H2O2 Ú Fe3+ + OH + OH. The iron-dependent Fenton reaction has been shown to lead to genomic alterations, including a Cdkn2a/2b deletion and a Met amplification, during carcinogenesis in an animal model (32). Yoshimoto et al found, for the first time, that the great majority of iron in the cyst fluid is considered to be heme iron, but not free iron (31). Hemoglobin and heme iron are oxidized from the oxyhemoglobin (oxyHb-Fe2+) to the methemoglobin (metHb-Fe3+) with generation of the superoxide anion (O2) as an autoxidation as follows: Hb-Fe2+ (oxyHb) + O2 Ú Hb-Fe3+ (metHb) + O2. Since heme iron is abundant in the cyst fluid of benign OE, autoxidation, rather than the Fenton reaction, may be the main process accomplishing the oxidative reaction.

It would be of interest to determine the origin and biological function of metHb that is abundantly expressed in benign OE. Peritoneal concentrations of nitric oxide (NO) metabolites (nitrite and nitrate) in patients with endometriosis have been shown to be significantly higher than those in patients with non-endometriosis (33). Inducible NO synthase (iNOS) is an enzyme that catalyzes the production of NO from L-arginine. The mRNA level of iNOS has also been shown to be higher in the endometriosis group than in the non-endometriosis group (34). Thus, the serum NO level is elevated in the endometriosis group as compared to the control group due to the induction of iNOS. NO can oxidize oxyHb to metHb as follows (35): HNO + 2[HbO2]2+ Ú 2[Hb]3+ + NO3 + HO2. Therefore, metHb is known as an oxidative stress marker and causes production of free radicals to induce oxidative stress by reacting with peroxides (hydrogen peroxide, lipid hydroperoxides) (29). MetHb is downregulated in EAOC when compared to benign OE, which supports the hypothesis that paradoxically, a shift in the balance between oxidants and antioxidants in EAOC is in favor of antioxidants (24). Glutathione, one of the most abundant endogenous antioxidant, is responsible for the conversion of metHb to oxyHb (36). It plays a role in repairing damage induced by oxidative stress in cancer cells and stops the process of cancer cachexia (36). Glutathione constitutes the survival advantage for cancer cells and is required for cancer initiation and progression (37,38). To date, a number of studies have demonstrated that the oxidant-antioxidant imbalance plays a critical role in the initiation and progression of multistage carcinogenesis of endometriosis (23,30,3941). These studies support the redox imbalance hypothesis that there are at least two stages of iron carcinogenesis and tumor progression: The initial wave of iron-induced oxidative stress would be followed by the second big wave of subsequent synthesis of the antioxidants, which diminishes cellular oxidative stress capacity, increases apoptosis resistance and promotes tumor initiation and progression.

Similar epigenetic modifications: Gene-environment interactions

The practical and theoretical implications was discussed with regard to the current knowledge of epigenetic modifications in benign OE and EAOC. An excess of heme iron and non-heme iron is toxic to cell and tissue components. Hackett et al reported the quantification of total iron from the hematoma folllowing an intracerebral hemorrhage in an animal model (42). Neuronal cell death was observed at a concentration of ~1.0 µg/cm2 (42). The iron concentration of the brain tissue at the periphery of the hematoma has also been shown to be ~400 mg/l in two human subjects (43). The iron concentration in OE (244.4 mg/l) is almost similar to intracerebral hemorrhage (400 mg/l), suggesting that endometriotic cells may face the crisis of death. Under prolonged stressful conditions, endometriotic cells must cope with various internal and external ROS for survival. The choice between cell survival and death depends on the net result of ROS production and their elimination by antioxidative enzymes (44). High levels of ROS promote DNA damage and cell death, although perhaps surprisingly, low levels of ROS are known to be associated with the development of tumors and then the process of carcinogenesis (2224,31). Thus, an imbalance in the cellular redox state may play an important role in the mechanism of its long-term carcinogenic effect; gene-environment interactions may modify an individuals susceptibility to this type of cancer.

High ROS levels damage the mitochondrial DNA and promote its mutation, which affects the epigenetic control mechanisms of nuclear DNA, by decreasing the activity of some methyltransferases, thus causing DNA hypomethylation (45). ROS-induced oxidative stress is associated not only with global/genome-wide DNA hypomethylation, but also with tumor suppressor gene promoter-specific aberrant hypermethylation via the upregulation of the expression of DNA methyltransferases (46). CpG clusters are susceptible to oxidative DNA damage to cytosine in the Fenton reaction, which is the main cause of cytosine-to-thymine transition mutations (47,48). Therefore, the dynamics of DNA methylation at CpG clusters can drive an increased likelihood of genetic mutations. Epigenetic mechanisms and then genetic mutations are considered to contribute to the necessary plasticity of endometriotic cells. Recent studies have attempted to link genetic modifications with epigenetic or environmental risk factors for EAOC (23,49). These results shed light onto the mechanisms underlying the associations of environmental stimuli and redox imbalance with risk of developing EAOC.

Defective CpG methylation affects several genes involved in endometriosis malignant transformation, such as Runt-related transcription factor 3 (RUNX3), human mutL homolog 1 (hMLH1), E-cadherin (CDH1), Ras-association domain family of gene 2 (RASSF2), p16, AT-rich interactive domain-containing protein 1A (ARID1A) and phosphatase and tensin homolog deleted on chromosome 10 (PTEN) by promoter hypermethylation (50). By contrast, steroidogenic factor-1 (SF-1), a transcriptional factor essential for estrogen biosynthesis, has been shown to be hypomethylated and aberrantly expressed (51). Furthermore, oxidative stress (exogenous H2O2) downregulates ARID1A mRNA and protein expression (39,52,53). Oxidative stress recruits DNA methyltransferase to chromatin (54), and also modifies the expression of CpG demethylases, such as ten-eleven translocation (TET) and jumonji (JMJ) genes (49). These genes may be involved in the development of endometriosis and its malignant transformation. The epigenetic switch occurs even in benign endometriosis (49).

Similar genetic abnormalities

Furthermore, endometriosis and EAOC harbor not only multiple somatic gene mutations, but also epigenetic modifications. Herein, we provide overview of the possible pathogenesis of malignant transformation of endometriosis that have exhibited distinct tumor morphological and phenotypical features, but have suggested similar (epi)genetic abnormalities. EC is distinguished from CCC due to different morphologies, but both represent common environmental profiles (53) and maintain the similar genomic abnormalities with multiple overlaps and share similar molecular signatures (54). Recent microarray, targeted sequencing and whole genome studies have identified that somatic mutations of AT-rich interaction domain 1A (ARID1A), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), PTEN, KRAS proto-oncogene, GTPase (KRAS), catenin beta 1 (CTNNB1) and mutL homolog 1 (MLH1) were commonly found across EAOC (5561). EAOC and adjacent endometriotic lesions exhibited common multiple cancer driver gene mutations, suggesting that they can share extensive genetic similarity, a common genomic origin and a common lineage (6265).

We hypothesized that endometriotic cells would acquire (epi)genetic modifications required for survival among the harshest and poorest environments. The cells selected for oxidative resistance enable clonal expansion/differentiation and could survive (66). Indeed, endometriosis is considered to be monoclonal in origin and neoplastic in nature (67). Surprisingly, mutations of classical cancer driver genes have been observed in 4% of a histopathologically benign OE and 26% of deep infiltrating endometriosis in cancer driver genes, including ARID1A, KRAS, PIK3CA and PPP2R1A (64,65). The existence of somatic driver mutations occurring in the epithelial glandular cells but not the stromal cells of the same endometriosis lesion (64) implies that endometriotic epithelial cells might incur an advantage through selective survival or proliferation and that the stromal cells are resistant to environmental hazards.

CCC-specific (epi)genetic profile

Despite a similar genetic profile, EAOC tumors present a different biological profile (68). Several studies have provided new insight into the signaling pathway of genes differentially expressed between EC and CCC. The interpretation of differentially expressed genes has verified the dysregulated biological functions related to glucose utilization, cell cycle regulation and hormone metabolism, that plays important role in the development of EAOC (60,6870). Transcription factor hepatocyte nuclear factor-1β (HNF1B) was identified as a biomarker of ovarian CCC histology, but not EC, with the hypomethylation of the HNF1B promoter influencing the characteristic biology (60,68,7075). Endometriosis is composed of two subgroups: HNF-1β-positive and -negative cells (71). The expression patterns have been shown to be similar in a contiguous transition from endometriotic cells to atypical cells to CCC (60). HNF-1β-positive endometriotic cells may represent a prototypical lineage of CCC cells. Much interest has been focused on HNF-1β, which is commonly upregulated in endometriosis and CCC. HNF-1β is associated with the normal development of the liver, pancreas, gut, lungs and kidneys, and its mutations represents the frequent occurrence of familial forms of type 2 diabetes (68). The exact biological function of the HNF-1β gene in CCC has been widely reported: This gene plays key roles in glycogen synthesis, anti-oxidative defense, anti-apoptosis, resistance to anticancer agents and cell cycle regulators at G2/M transition (73,76,77).

First, in the elegant review, Mandai et al provided new insight into the biological impact of CCC in a tumor microenvironment via the upregulation of HNF-1β expression (68). HNF-1β upregulates glucose uptake and glycolysis to give rise to an increased yield of lactate in CCC, which is known as the Warburg metabolic phenotype (68,78). The Warburg effect benefits cancer cells to avoid excess ROS generation and thus gains increased survival advantage in iron-rich stressful environments such as endometriosis. The genes involved in glucose homeostasis, including dipeptidyl peptidase 4 (DPP4) (79) aldolase B (ALDOB) (80), glucose transporter-1 (GLUT-1) gene and several key enzymes in the glycolytic process (78), are downstream targets of HNF1B. HNF-1β thus may be a key regulator of glycolysis, gluconeogenesis and glucose homeostasis.

Second, HNF-1β actually reduces and protects cancer cells from oxidative stress by markedly changing antioxidant activity (78,81,82). HNF-1β may repair damage caused by oxidative stress and can promote survival by upregulating antioxidant proteins via binding with antioxidant response element (37,68,81,83,84). This gene upregulates the synthesis of glutathione (GSH), a powerful antioxidant (85). HNF-1β also triggers ROS resistance in CCC cells via rBAT, a cystine transporter (68). Thus, HNF-1β reduces oxidative stress and confers ROS resistance and a survival advantage in CCC cells (37,68).

Finally, it would also be of interest to determine the mechanisms underlying the protective effects of HNF-1β on cells against any cytotoxicity and genotoxicity caused by ROS when CCC cells were exposed to a stressful environment. DNA damage occurs continually through various intrinsic and extrinsic stressors such as ROS, ultraviolet radiation, smoking and errors during replication (86). In endometriosis, environmental hazards, including hemoglobin, heme and iron, induce lesions in genomic DNA. The cellular DNA damage response (DDR) comprises the coordinated actions of DNA repair and checkpoint systems that regulate a spectrum of processes before replication, where cell cycle arrest enables DNA repair to occur (86). The DDR also promotes cell death when the damage is beyond repair. If excessive damage exists, the DDR activates cell death and eliminates the damaged cells by apoptosis. Two key regulators of the DDR cell cycle checkpoints include ataxia telangiectasia mutated (ATM) and ataxia telangiectasia mutated and rad3-related (ATR) (86). ATR responds to a broad spectrum of DNA damage, including replication-associated DNA damage, while ATM is activated by DNA double-strand breaks (86). A number of studies have vigorously investigated the association between redox imbalance and cell cycle signaling pathways in CCC (73,77,83,8789), while no studies have focused on the influence of ROS on the pathogenesis of EC, at least to the best of our knowledge. Shigetomi et al investigated the role of HNF-1β in regulation of the cell cycle arrest in response to DNA damage in the CCC cell line, TOV21G (87). Flow cytometric analysis of cell cycle profiles indicated that HNF-1β inhibited cell cycle progression (87). Fig. 1 illustrates the typical flow cytometry histograms of the results. HNF-1β-expressing TOV21G cells exhibited a marked increase in the proportion of cells in the G2/M phase following exposure to a genotoxic agent, bleomycin, for 24 h (62.1 vs. 42.3%) (87). The knockdown of endogenous HNF-1β attenuated G2/M phase cell cycle arrest and stimulated cell death (28.0 vs. 18.2%) (87). It would also be of interest to determine which genes and their signaling pathways enhance and accelerate cell cycle arrest at G2/M phase. Shigetomi et al (87) and Ito et al (89) explored the activated and interconnected signaling network of HNF-1β to identify novel downstream targets (Fig. 2). HNF-1β promotes TOV21G cell survival through Chk1 phosphorylation (87,89). As previously demonstrated, the inactivation of HNF1B with siRNA suppressed Claspin protein expression, but failed to inhibit Claspin mRNA expression (89). Claspin transmits a replication stress signal from ATR to Chk1 and functions as an adaptor protein required for Chk1 activation. In CCC cells, HNF-1β, but not ATR, are essential for the upregulation of Claspin protein expression, suggesting that this gene functions as a Claspin protein post-translational modification. Ito et al vigorously identified potential modifiers of Claspin protein relevant to HNF-1β biology (89). To date, >450 unique protein modifications have been identified, including phosphorylation, acetylation, ubiquitination and SUMOylation through post-translational modification (90). Phosphorylation is one of the most common and reversible intracellular post-translational modifications of serine and threonine residues (91). Acetylation is a modification of the lysine residues (92). Ubiquitination is a widely studied method of post-translational protein modification (4). Claspin is reportedly regulated by ubiquitin-dependent proteasomal degradation, whereas the ubiquitin-specific processing protease (USP) 28- and USP29-mediated deubiquitination inhibits its degradation (93). Martín et al reported that USP29 controls the stability of Claspin by deubiquitination (94). However, HNF-1β did not stimulate the upregulation of USP29 protein in CCC cells. Ito et al identified, for the first time, a novel regulator of Claspin, USP28, as a direct downstream target of HNF-1 (89). USP28 interacts with Claspin and is able to deubiquitinate it. With these results, USP28 is identified as a novel player in the HNF-1β-Chk1 pathway and the control of DNA replication, via the HNF-1β-USP28-Claspin-Chk1-CDC25C pathway (89). This pathway contributes to a loss of G2/M checkpoint control, which accumulates genomic and chromosomal instability and then paves the way for further major genetic changes.

Although the impact of HNF-1β on the cell cycle arrest at G2/M phase under oxidative stress conditions is recognized in CCC, the role of HNF-1β in oxidative stress-induced endometriosis carcinogenesis remains poorly defined. When endometriotic cells are exposed to genotoxic oxidative stressors such as hemoglobin, heme or free iron, the HNF-1β gene may also be epigenetically hypomethylated and is demonstrated as a positive modulator of cell survival, through the HNF-1β signaling pathway. This hypothesis needs to be verified in future studies.

EC-specific (epi)genetic profile

Endometriosis is an estrogen-dependent disease. The enzyme aromatase P450 is expressed aberrantly in endometriosis and catalyzes the final step of estrogen production and upregulates the expression of prostaglandin E2 (PGE2) and macrophage migration inhibitory factor (MIF), which, in turn, induces the expression of aromatase within endometriotic lesions (55). The effects of estrogen on stromal cell PGE2 production may be mediated in a feed-forward manner. MIF is a cytokine marker of M2 polarization of macrophage which facilitates the onset and progression of endometriosis. Such an interplay with a positive feedback cycle is involved in cell proliferation and apoptotic resistance of endometriosis, and then its malignant transformation.

There are two types of estrogen receptors (ESRs), ESR1 (also known as ERα) and ESR2 (ERβ). The ESR expression level has been shown to be higher in EC and high-grade serous than in CCC and mucinous carcinoma (95). Among EAOC, ESR positivity has been shown to be significantly higher in EC (91%), but lower in CCC (8%) (60). ESR gene expression is modulated by a number of factors, such as DNA methylation of the promoter region, histone deacetylation, chromatin remodeling, or heme and iron binding (58). The interrelation between the ESR expression and these factors is complex, as genetic characteristics and environmental factors can mutually impact upon each other. The significant up- and downregulation of ESR has been shown to be associated with marked epigenomic alterations: ESR2 is the predominant ESR in endometriosis due to the hypomethylation of promoter CpG islands, whereas ESR1 levels are lower in endometriosis (69). EC shares estrogen-dependent oncogenic pathways and signaling network. A hyperestrogenic state or the upregulation of ESR expression may be shared in common with benign and malignant endometriosis, which may denote that endometriosis has carcinogenic potential. Furthermore, G-protein-coupled estrogen receptor-30 (GPR30) is the novel estrogen-responsive receptor G protein-coupled estrogen receptor 1, GPER (96). GPR30 expression is higher in EAOC than in benign OE (96). The upregulation of ESR expression is associated with a better clinical outcome in ovarian cancer (95) and CCC (14), suggesting the role of ESR in tumor initiation or the early development of primary EC, but not in EC progression.

Taken together, EAOC is a heterogeneous disease, with at least two intrinsic subtypes, EC and CCC. Although the role of DNA methylation in EAOC development is not yet fully understood, its profiling defines cancer subclasses differing in clinicopathologic characteristics, molecular profiles and survival. Genes with promoter hypermethylation and hypomethylation are consistent in cancer function and characteristics of concordant methylation. The promoter hypomethylated ESR gene is reversely correlated with the promoter hypermethylated HNF1B gene in EC (58,60,97). A low expression of ESR (95) and high expression of HNF-1β (71) are identified as potential biomarkers for CCC.

Other crucial aspects

We discuss the potential involvement of microenvironment in endometriosis and its malignant transformation. The dysfunctional regulation of immune and inflammatory microenvironment, extracellular matrix remodeling, or new blood vessel formation is a crucial aspect of pathogenesis of endometriosis and its malignant transformation. Ovarian cancer is initially associated with pelvic inflammatory disease, such as endometriosis, demonstrating a similarity between the processes of inflammation and carcinogenesis. Endometriotic cells adapt to survive in the unique microenvironment conditions with high levels of iron, inflammatory cytokines and chemokines (98). Microenvironment-cell interplay may modulate the major signaling pathways associated with cell cycle regulation, growth factor signaling, immune and inflammatory pathways, and the extracellular matrix remodeling, which results in phenotype transformation (99). Researchers have focused on the function of matrix metalloproteinase (MMPs) (100), lysyl oxidases (LOXs) and nuclear factor κ-light-chain-enhancer of activated B (NF-κB) in the pathophysiology of inflammation and EAOC (101). Several studies have identified the NF-κB-dependent multiple oncogenic pathways in endometriosis (102) and highlight its malignant transformation (103). MMP-2 promotes angiogenesis during endometriosis progression via the cyclooxygenase (COX)-2/PGE2/pAKT axis (104). The upregulation of lysyl oxidase (LOX) expression is involved in extracellular membrane degradation, invasive and metastatic potential of endometriosis (105). The disruption of epithelial-stromal communication networks elicits a feed-forward loop involving endometriosis to drive inflammation, which may be relevant in diseases such as EAOC.

In addition, chemokines are key players in the activation and are recruitment of immune cells at sites of inflammation. The CXCR4/CXCL12 axis is functional in endometriosis and plays a role in a number of diverse cellular functions, including immune surveillance, inflammation response, tissue homeostasis, and tumor growth and metastasis (106). CXCR4 expression is upregulated by vascular endothelial growth factor (VEGF), and plays an important role in the malignant transformation of endometriosis (107). Furthermore, the microvascular endothelium of ectopic endometrial tissue originates from circulating endothelial progenitor cells mobilized from the bone marrow, which is also controlled by the CXCL12/CXCR4 axis (108). The neovascularization of endometriotic lesions is not only driven by angiogenesis, but also vasculogenesis from circulating endothelial progenitor cells (108). Thus, angiogenesis and vasculogenesis play an integral part in the establishment and growth of endometriotic lesions and malignant transformation (108,109). Therefore, these changes in the microenvironment are necessary to accumulate enough epigenetic, genetic and pathological alterations for malignant transformation of endometriosis (110).

Discussion

In the present study, we provide a literature review of various lines of evidence supporting the concept of an altered redox environmental model for malignant transformation of endometriosis. Fig. 3 summarizes the current knowledge about the role of the shared and independent (epi)genetic background between EAOC tumors, and their interaction with environmental stimuli. We initially updated the epigenetic, genetic and environmental backgrounds of EAOC and surveyed the examples of environmentally induced epigenetic changes. Despite the differences in morphology between EC and CCC, they share remarkable (epi)genetic similarities and enrichment for driver somatic mutations affecting ARID1A, PTEN and KRAS genes (5561). The hemoglobin, heme and free iron accumulated during endometriosis development are a prerequisite to modification of genomic DNA for prompt cellular responses to oxidative stress (58). An excess of heme iron and nonheme iron participates in the Fenton reaction generating the toxic hydroxyl radical. Autoxidation of oxyHb to metHb always occurs due to abundant heme iron in the contents of benign OE. Autoxidation, rather than the Fenton reaction, might be the main process accomplishing the oxidative reaction in endometriosis (31). Redox biology is considered to alter (epi)genetic events. Environmentally-induced epigenetic alterations may result in a change of the adaptive gene function, leading to phenotypic plasticity. Endometriosis is predisposed to develop into EAOC through the progressive accumulation of epigenetic alterations (3,4,6,9) during obvious redox imbalance (20,23). However, there is only limited knowledge of the mechanisms through which environmental factors affect gene function.

Figure 3.

The concept of an altered redox environmental model for malignant transformation of endometriosis. EAOC consists of different histological subtypes mainly originating from EC and CCC. First, repeated episodes of hemorrhage occur in endometriosis throughout menstruation. Extracellular free hemoglobin produces toxic heme degradation products and is a source of ROS. Hemoglobin, heme and free iron in endometriotic cysts cause redox imbalance. Second, there is a link between environmental stimuli and (epi)genetic modifications. EC is distinguished from CCC due to different morphologies, but both represent common environmental profiles and maintain the similar genomic abnormalities with multiple overlaps and share similar molecular signatures, including ARID1A, PIK3CA, PTEN, or KRAS. Finally, ESR and HNF-1β proteins are mutually exclusive in EAOC. HNF-1β-positive and ESR-negative endometriotic cells may represent a prototypical lineage of CCC cells. The positive ESR expression and negative HNF-1β expression is a frequent finding in EC. EAOC tumors had enrichment of cancer-specific gene signatures corresponding with each histological subtype: ESR induces EC cell proliferation (‘go’) and HNF-1β induces CCC cell cycle arrest (‘stop’) for a survival mechanism in response to several stresses. EAOC, endometriosis-associated ovarian cancer; EC, endometrioid carcinoma; CCC, clear cell carcinoma; ARID1A, AT-rich interactive domain-containing protein 1A; PIK3CA, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha; PTEN, phosphatase and tensin homolog deleted on chromosome 10; HNF-1β, hepatocyte nuclear factor-1β.

In addition, a previous approach identified different genetic backgrounds between EAOC tumors (60). By comparing the gene expression profile, at least two differentially expressed genes were identified in EC and CCC. A positive ESR expression and negative HNF-1β expression is a frequent finding in EC, but not in CCC (58,60). EC may develop in the setting of estrogen-driven pathway (111). On the other hand, HNF-1β-dependent ovarian cancer arising from endometriosis is substantially more associated with CCC than with EC (11,60,77,83,87,89). Immunohistochemical data have indicated that atypical endometriosis is a precursor lesion molecularly similar to adjacent invasive cancer (60). Pre-malignant endometriotic cells exposed to mixture of genotoxic oxidative stressors inhibit cell proliferation and promote cell cycle arrest at G2/M phase for DNA damage repair through the HNF-1β/USP28/Clasipin/Chk1 pathway (89). Therefore, HNF-1β induces cell cycle arrest, DNA damage and genomic instability, thereby promoting erroneous DNA repair and can predispose to CCC. EAOC tumors had enrichment of cancer-specific gene signatures corresponding with each histological subtype: ESR induces EC cell proliferation (‘go’) and HNF-1β induces CCC cell cycle arrest (‘stop’). This model underscores a subtype-dependent dichotomy between ‘go’ and ‘stop’ in EAOC, through potentially better ability to adapt in a changing environment (Fig. 3).

In conclusion, a special emphasis is given to current pathophysiological concepts of malignant transformation of endometriosis, including redox imbalance, environmental stimuli-induced (epi)genetic modifications and mutually exclusive expression of ESR and HNF-1β genes for a survival mechanism in response to several stresses.

Acknowledgements

Not applicable.

Funding

The present study was supported by JSPS KAKENHI (grant no. JP16K11150) and Tohoku Bureau of Economy, Trade and Industry (Tohoku 1607028).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors contributions

YY, NK and KO collected the data regarding the epigenetic and genetic abnormalities and the underlying mechanism of endometriosis transformation using the PubMed database. NK, KO and CY performed the literature search and supervised the study. HK and CY made substantial contributions to the conception of the study. HK contributed to the study design and interpretation of the included research studies. The final version of the manuscript has been read and approved by all authors.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare no potential competing interests with respect to the research, authorship and publication of this article.

References

1 

Bulletti C, Coccia ME, Battistoni S and Borini A: Endometriosis and infertility. J Assist Reprod Genet. 27:441–447. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Giudice LC and Kao LC: Endometriosis. Lancet. 364:1789–1799. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Brinton LA, Sakoda LC, Sherman ME, Frederiksen K, Kjaer SK, Graubard BI, Olsen JH and Mellemkjaer L: Relationship of benign gynecologic diseases to subsequent risk of ovarian and uterine tumors. Cancer Epidemiol Biomarkers Prev. 14:2929–2935. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Kobayashi H, Sumimoto K, Kitanaka T, Yamada Y, Sado T, Sakata M, Yoshida S, Kawaguchi R, Kanayama S, Shigetomi H, et al: Ovarian endometrioma--risks factors of ovarian cancer development. Eur J Obstet Gynecol Reprod Biol. 138:187–193. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Kim HS, Kim TH, Chung HH and Song YS: Risk and prognosis of ovarian cancer in women with endometriosis: A meta-analysis. Br J Cancer. 110:1878–1890. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Wilbur MA, Shih IM, Segars JH and Fader AN: Cancer implications for patients with endometriosis. Semin Reprod Med. 35:110–116. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Munksgaard PS and Blaakaer J: The association between endometriosis and gynecological cancers and breast cancer: A review of epidemiological data. Gynecol Oncol. 123:157–163. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Kurman RJ and Shih IeM: Pathogenesis of ovarian cancer: Lessons from morphology and molecular biology and their clinical implications. Int J Gynecol Pathol. 27:151–160. 2008.PubMed/NCBI

9 

Kurman RJ and Shih IeM: The origin and pathogenesis of epithelial ovarian cancer: A proposed unifying theory. Am J Surg Pathol. 34:433–443. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Wu RC, Veras E, Lin J, Gerry E, Bahadirli-Talbott A, Baras A, Ayhan A, Shih IM and Wang TL: Elucidating the pathogenesis of synchronous and metachronous tumors in a woman with endometrioid carcinomas using a whole-exome sequencing approach. Cold Spring Harb Mol Case Stud. 3:a0016932017. View Article : Google Scholar : PubMed/NCBI

11 

Kajihara H, Yamada Y, Shigetomi H, Higashiura Y and Kobayashi H: The dichotomy in the histogenesis of endometriosis-associated ovarian cancer: Clear cell-type versus endometrioid-type adenocarcinoma. Int J Gynecol Pathol. 31:304–312. 2012. View Article : Google Scholar : PubMed/NCBI

12 

van der Horst PH, van der Zee M, Heijmans-Antonissen C, Jia Y, DeMayo FJ, Lydon JP, van Deurzen CH, Ewing PC, Burger CW and Blok LJ: A mouse model for endometrioid ovarian cancer arising from the distal oviduct. Int J Cancer. 135:1028–1037. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Wu R, Zhai Y, Kuick R, Karnezis AN, Garcia P, Naseem A, Hu TC, Fearon ER and Cho KR: Impact of oviductal versus ovarian epithelial cell of origin on ovarian endometrioid carcinoma phenotype in the mouse. J Pathol. 240:341–351. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Rambau P, Kelemen LE, Steed H, Quan ML, Ghatage P and Köbel M: Association of hormone receptor expression with survival in ovarian endometrioid carcinoma: Biological validation and clinical implications. Int J Mol Sci. 18:E5152017. View Article : Google Scholar : PubMed/NCBI

15 

Lukanova A and Kaaks R: Endogenous hormones and ovarian cancer: Epidemiology and current hypotheses. Cancer Epidemiol Biomarkers Prev. 14:98–107. 2005.PubMed/NCBI

16 

Wu R, Hendrix-Lucas N, Kuick R, Zhai Y, Schwartz DR, Akyol A, Hanash S, Misek DE, Katabuchi H, Williams BO, et al: Mouse model of human ovarian endometrioid adenocarcinoma based on somatic defects in the Wnt/beta-catenin and PI3K/Pten signaling pathways. Cancer Cell. 11:321–333. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Tanwar PS, Zhang L, Kaneko-Tarui T, Curley MD, Taketo MM, Rani P, Roberts DJ and Teixeira JM: Mammalian target of rapamycin is a therapeutic target for murine ovarian endometrioid adenocarcinomas with dysregulated Wnt/β-catenin and PTEN. PLoS One. 6:e207152011. View Article : Google Scholar : PubMed/NCBI

18 

Tao GZ, Lehwald N, Jang KY, Baek J, Xu B, Omary MB and Sylvester KG: Wnt/β-catenin signaling protects mouse liver against oxidative stress-induced apoptosis through the inhibition of forkhead transcription factor FoxO3. J Biol Chem. 288:17214–17224. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Mahalingaiah PK and Singh KP: Chronic oxidative stress increases growth and tumorigenic potential of MCF-7 breast cancer cells. PLoS One. 9:e873712014. View Article : Google Scholar : PubMed/NCBI

20 

Mandai M, Matsumura N, Baba T, Yamaguchi K, Hamanishi J and Konishi I: Ovarian clear cell carcinoma as a stress-responsive cancer: Influence of the microenvironment on the carcinogenesis and cancer phenotype. Cancer Lett. 310:129–133. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Suryawanshi S, Huang X, Elishaev E, Budiu RA, Zhang L, Kim S, Donnellan N, Mantia-Smaldone G, Ma T, Tseng G, et al: Complement pathway is frequently altered in endometriosis and endometriosis-associated ovarian cancer. Clin Cancer Res. 20:6163–6174. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Iwabuchi T, Yoshimoto C, Shigetomi H and Kobayashi H: Oxidative stress and antioxidant defense in endometriosis and its malignant transformation. Oxid Med Cell Longev 2015. 8485952015.

23 

Kobayashi H: Potential scenarios leading to ovarian cancer arising from endometriosis. Redox Rep. 21:119–126. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Iwabuchi T, Yoshimoto C, Shigetomi H and Kobayashi H: Cyst fluid hemoglobin species in endometriosis and its malignant transformation: The role of metallobiology. Oncol Lett. 11:3384–3388. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Bandini S, Macagno M, Hysi A, Lanzardo S, Conti L, Bello A, Riccardo F, Ruiu R, Merighi IF, Forni G, et al: The non-inflammatory role of C1q during Her2/neu-driven mammary carcinogenesis. Oncoimmunology. 5:e12536532016. View Article : Google Scholar : PubMed/NCBI

26 

McKinnon BD, Kocbek V, Nirgianakis K, Bersinger NA and Mueller MD: Kinase signalling pathways in endometriosis: Potential targets for non-hormonal therapeutics. Hum Reprod Update. 22:382–403. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Jung M, Weigert A, Mertens C, Rehwald C and Brüne B: Iron handling in tumor-associated macrophages: Is there a new role for lipocalin-2? Front Immunol. 8:11712017. View Article : Google Scholar : PubMed/NCBI

28 

Ngô C, Chéreau C, Nicco C, Weill B, Chapron C and Batteux F: Reactive oxygen species controls endometriosis progression. Am J Pathol. 175:225–234. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Alayash AI: Oxygen therapeutics: Can we tame haemoglobin? Nat Rev Drug Discov. 3:152–159. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Yamaguchi K, Mandai M, Toyokuni S, Hamanishi J, Higuchi T, Takakura K and Fujii S: Contents of endometriotic cysts, especially the high concentration of free iron, are a possible cause of carcinogenesis in the cysts through the iron-induced persistent oxidative stress. Clin Cancer Res. 14:32–40. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Yoshimoto C, Iwabuchi T, Shigetomi H and Kobayashi H: Cyst fluid iron-related compounds as useful markers to distinguish malignant transformation from benign endometriotic cysts. Cancer Biomark. 15:493–499. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Akatsuka S, Yamashita Y, Ohara H, Liu YT, Izumiya M, Abe K, Ochiai M, Jiang L, Nagai H, Okazaki Y, et al: Fenton reaction induced cancer in wild type rats recapitulates genomic alterations observed in human cancer. PLoS One. 7:e434032012. View Article : Google Scholar : PubMed/NCBI

33 

Kianpour M, Nematbakhsh M and Ahmadi SM: Asymmetric dimethylarginine (ADMA), nitric oxide metabolite, and estradiol levels in serum and peritoneal fluid in women with endometriosis. Iran J Nurs Midwifery Res. 20:484–489. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Yeo SG, Won YS, Lee HY, Kim YI, Lee JW and Park DC: Increased expression of pattern recognition receptors and nitric oxide synthase in patients with endometriosis. Int J Med Sci. 10:1199–1208. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Bellavia L, DuMond JF, Perlegas A, Bruce King S and Kim-Shapiro DB: Nitroxyl accelerates the oxidation of oxyhemoglobin by nitrite. Nitric Oxide. 31:38–47. 2013. View Article : Google Scholar : PubMed/NCBI

36 

den Boer PJ, Bleeker WK, Rigter G, Agterberg J, Stekkinger P, Kannegieter LM, de Nijs IM and Bakker JC: Intravascular reduction of methemoglobin in plasma of the rat in vivo. Biomater Artif Cells Immobilization Biotechnol. 20:647–650. 1992. View Article : Google Scholar : PubMed/NCBI

37 

Amano Y, Mandai M, Yamaguchi K, Matsumura N, Kharma B, Baba T, Abiko K, Hamanishi J, Yoshioka Y and Konishi I: Metabolic alterations caused by HNF1β expression in ovarian clear cell carcinoma contribute to cell survival. Oncotarget. 6:26002–26017. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Harris IS, Treloar AE, Inoue S, Sasaki M, Gorrini C, Lee KC, Yung KY, Brenner D, Knobbe-Thomsen CB, Cox MA, et al: Glutathione and thioredoxin antioxidant pathways synergize to drive cancer initiation and progression. Cancer Cell. 27:211–222. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Winarto H, Tan MI, Sadikin M and Wanandi SI: ARID1A Expression is down-regulated by oxidative stress in endometriosis and endometriosis-associated ovarian cancer. Transl Oncogenomics. February 24–2017.doi. 1177272716689818. View Article : Google Scholar : PubMed/NCBI

40 

Yan S, Sorrell M and Berman Z: Functional interplay between ATM/ATR-mediated DNA damage response and DNA repair pathways in oxidative stress. Cell Mol Life Sci. 71:3951–3967. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Toyokuni S: Iron and thiols as two major players in carcinogenesis: Friends or foes? Front Pharmacol. 5:2002014. View Article : Google Scholar : PubMed/NCBI

42 

Hackett MJ, DeSouza M, Caine S, Bewer B, Nichol H, Paterson PG and Colbourne F: A new method to image heme-Fe, total Fe, and aggregated protein levels after intracerebral hemorrhage. ACS Chem Neurosci. 6:761–770. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Chaudhary N, Pandey AS, Merchak K, Gemmete JJ, Chenevert T and Xi G: Perihematomal cerebral tissue iron quantification on MRI following intracerebral hemorrhage in two human subjects: Proof of principle. Acta Neurochir Suppl (Wien). 121:179–183. 2016. View Article : Google Scholar

44 

Nagano O, Okazaki S and Saya H: Redox regulation in stem-like cancer cells by CD44 variant isoforms. Oncogene. 32:5191–5198. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Szumiel I: Ionizing radiation-induced oxidative stress, epigenetic changes and genomic instability: The pivotal role of mitochondria. Int J Radiat Biol. 91:1–12. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Wu Q and Ni X: ROS-mediated DNA methylation pattern alterations in carcinogenesis. Curr Drug Targets. 16:13–19. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Bandaru B, Gopal J and Bhagwat AS: Overproduction of DNA cytosine methyltransferases causes methylation and C --> T mutations at non-canonical sites. J Biol Chem. 271:7851–7859. 1996. View Article : Google Scholar : PubMed/NCBI

48 

Samson-Thibault F, Madugundu GS, Gao S, Cadet J and Wagner JR: Profiling cytosine oxidation in DNA by LC-MS/MS. Chem Res Toxicol. 25:1902–1911. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Ito F, Yamada Y, Shigemitsu A, Akinishi M, Kaniwa H, Miyake R, Yamanaka S and Kobayashi H: Role of oxidative stress in epigenetic modification in endometriosis. Reprod Sci. 24:1493–1502. 2017. View Article : Google Scholar : PubMed/NCBI

50 

He J, Chang W, Feng C, Cui M and Xu T: Endometriosis malignant transformation: Epigenetics as a probable mechanism in ovarian tumorigenesis. Int J Genomics 2018. 14653482018.

51 

Bulun SE, Monsivais D, Kakinuma T, Furukawa Y, Bernardi L, Pavone ME and Dyson M: Molecular biology of endometriosis: From aromatase to genomic abnormalities. Semin Reprod Med. 33:220–224. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Xie H, Chen P, Huang HW, Liu LP and Zhao F: Reactive oxygen species downregulate ARID1A expression via its promoter methylation during the pathogenesis of endometriosis. Eur Rev Med Pharmacol Sci. 21:4509–4515. 2017.PubMed/NCBI

53 

Kolin DL, Dinulescu DM and Crum CP: Origin of clear cell carcinoma: nature or nurture? J Pathol. 244:131–134. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Chang CM, Yang YP, Chuang JH, Chuang CM, Lin TW, Wang PH, Yu MH and Chang CC: Discovering the deregulated molecular functions involved in malignant transformation of endometriosis to endometriosis-associated ovarian carcinoma using a data-driven, function-based analysis. Int J Mol Sci. 18:23452017.doi: 10.3390/ijms18112345. View Article : Google Scholar :

55 

Bulun SE: Endometriosis. N Engl J Med. 360:268–279. 2009. View Article : Google Scholar : PubMed/NCBI

56 

Ayhan A, Mao TL, Seckin T, Wu CH, Guan B, Ogawa H, Futagami M, Mizukami H, Yokoyama Y, Kurman RJ, et al: Loss of ARID1A expression is an early molecular event in tumor progression from ovarian endometriotic cyst to clear cell and endometrioid carcinoma. Int J Gynecol Cancer. 22:1310–1315. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Veillat V, Sengers V, Metz CN, Roger T, Leboeuf M, Mailloux J and Akoum A: Macrophage migration inhibitory factor is involved in a positive feedback loop increasing aromatase expression in endometriosis. Am J Pathol. 181:917–927. 2012. View Article : Google Scholar : PubMed/NCBI

58 

Tanase Y, Yamada Y, Shigetomi H, Kajihara H, Oonogi A, Yoshizawa Y, Furukawa N, Haruta S, Yoshida S, Sado T, et al: Modulation of estrogenic action in clear cell carcinoma of the ovary (Review). Exp Ther Med. 3:18–24. 2012. View Article : Google Scholar : PubMed/NCBI

59 

Worley MJ Jr, Welch WR, Berkowitz RS and Ng SW: Endometriosis-associated ovarian cancer: A review of pathogenesis. Int J Mol Sci. 14:5367–5379. 2013. View Article : Google Scholar : PubMed/NCBI

60 

Lai CR, Hsu CY, Chen YJ, Yen MS, Chao KC and Li AF: Ovarian cancers arising from endometriosis: A microenvironmental biomarker study including ER, HNF1β, p53, PTEN, BAF250a, and COX-2. J Chin Med Assoc. 76:629–634. 2013. View Article : Google Scholar : PubMed/NCBI

61 

Takeda T, Banno K, Okawa R, Yanokura M, Iijima M, Irie-Kunitomi H, Nakamura K, Iida M, Adachi M, Umene K, et al: ARID1A gene mutation in ovarian and endometrial cancers (Review). Oncol Rep. 35:607–613. 2016. View Article : Google Scholar : PubMed/NCBI

62 

Bissell MJ: Modelling molecular mechanisms of breast cancer and invasion: Lessons from the normal gland. Biochem Soc Trans. 35:18–22. 2007. View Article : Google Scholar : PubMed/NCBI

63 

Kobayashi H, Kajiwara H, Kanayama S, Yamada Y, Furukawa N, Noguchi T, Haruta S, Yoshida S, Sakata M, Sado T, et al: Molecular pathogenesis of endometriosis-associated clear cell carcinoma of the ovary (Review). Oncol Rep. 22:233–240. 2009.PubMed/NCBI

64 

Anglesio MS, Papadopoulos N, Ayhan A, Nazeran TM, Noë M, Horlings HM, Lum A, Jones S, Senz J, Seckin T, et al: Cancer-associated mutations in endometriosis without cancer. N Engl J Med. 376:1835–1848. 2017. View Article : Google Scholar : PubMed/NCBI

65 

Zou Y, Zhou JY, Guo JB, Wang LQ, Luo Y, Zhang ZY, Liu FY, Tan J, Wang F and Huang OP: The presence of KRAS, PPP2R1A and ARID1A mutations in 101 Chinese samples with ovarian endometriosis. Mutat Res. 809:1–5. 2018. View Article : Google Scholar : PubMed/NCBI

66 

Jamnongkan W, Thanee M, Yongvanit P, Loilome W, Thanan R, Kimawaha P, Boonmars T, Silakit R, Namwat N and Techasen A: Antifibrotic effect of xanthohumol in combination with praziquantel is associated with altered redox status and reduced iron accumulation during liver fluke-associated cholangiocarcinogenesis. PeerJ. 6:e42812018. View Article : Google Scholar : PubMed/NCBI

67 

Jimbo H, Hitomi Y, Yoshikawa H, Yano T, Momoeda M, Sakamoto A, Tsutsumi O, Taketani Y and Esumi H: Evidence for monoclonal expansion of epithelial cells in ovarian endometrial cysts. Am J Pathol. 150:1173–1178. 1997.PubMed/NCBI

68 

Mandai M, Amano Y, Yamaguchi K, Matsumura N, Baba T and Konishi I: Ovarian clear cell carcinoma meets metabolism; HNF-1β confers survival benefits through the Warburg effect and ROS reduction. Oncotarget. 6:30704–30714. 2015. View Article : Google Scholar : PubMed/NCBI

69 

Bukulmez O, Hardy DB, Carr BR, Word RA and Mendelson CR: Inflammatory status influences aromatase and steroid receptor expression in endometriosis. Endocrinology. 149:1190–1204. 2008. View Article : Google Scholar : PubMed/NCBI

70 

Kato N, Tamura G and Motoyama T: Hypomethylation of hepatocyte nuclear factor-1beta (HNF-1beta) CpG island in clear cell carcinoma of the ovary. Virchows Arch. 452:175–180. 2008. View Article : Google Scholar : PubMed/NCBI

71 

Kato N, Sasou S and Motoyama T: Expression of hepatocyte nuclear factor-1beta (HNF-1beta) in clear cell tumors and endometriosis of the ovary. Mod Pathol. 19:83–89. 2006. View Article : Google Scholar : PubMed/NCBI

72 

Takano M, Kikuchi Y, Kudoh K, Goto T, Furuya K, Kikuchi R, Kita T, Fujiwara K, Shiozawa T and Aoki D: Weekly administration of temsirolimus for heavily pretreated patients with clear cell carcinoma of the ovary: A report of six cases. Int J Clin Oncol. 16:605–609. 2011. View Article : Google Scholar : PubMed/NCBI

73 

Shigetomi H, Tsunemi T, Haruta S, Kajihara H, Yoshizawa Y, Tanase Y, Furukawa N, Yoshida S, Sado T and Kobayash H: Molecular mechanisms linking endometriosis under oxidative stress with ovarian tumorigenesis and therapeutic modalities. Cancer Invest. 30:473–480. 2012. View Article : Google Scholar : PubMed/NCBI

74 

Kao YC, Lin MC, Lin WC, Jeng YM and Mao TL: Utility of hepatocyte nuclear factor-1β as a diagnostic marker in ovarian carcinomas with clear cells. Histopathology. 61:760–768. 2012. View Article : Google Scholar : PubMed/NCBI

75 

Ye S, Yang J, You Y, Cao D, Huang H, Wu M, Chen J, Lang J and Shen K: Clinicopathologic significance of HNF-1β, AIRD1A, and PIK3CA expression in ovarian clear cell carcinoma: A tissue microarray study of 130 cases. Medicine (Baltimore). 95:e30032016. View Article : Google Scholar : PubMed/NCBI

76 

Barbacci E, Chalkiadaki A, Masdeu C, Haumaitre C, Lokmane L, Loirat C, Cloarec S, Talianidis I, Bellanne-Chantelot C and Cereghini S: HNF1beta/TCF2 mutations impair transactivation potential through altered co-regulator recruitment. Hum Mol Genet. 13:3139–3149. 2004. View Article : Google Scholar : PubMed/NCBI

77 

Kobayashi H, Yamada Y, Kanayama S, Furukawa N, Noguchi T, Haruta S, Yoshida S, Sakata M, Sado T and Oi H: The role of hepatocyte nuclear factor-1beta in the pathogenesis of clear cell carcinoma of the ovary. Int J Gynecol Cancer. 19:471–479. 2009. View Article : Google Scholar : PubMed/NCBI

78 

Okamoto T, Mandai M, Matsumura N, Yamaguchi K, Kondoh H, Amano Y, Baba T, Hamanishi J, Abiko K, Kosaka K, et al: Hepatocyte nuclear factor-1β (HNF-1β) promotes glucose uptake and glycolytic activity in ovarian clear cell carcinoma. Mol Carcinog. 54:35–49. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Senkel S, Lucas B, Klein-Hitpass L and Ryffel GU: Identification of target genes of the transcription factor HNF1beta and HNF1alpha in a human embryonic kidney cell line. Biochim Biophys Acta 1731. 179–190. 2005.

80 

Gregori C, Porteu A, Mitchell C, Kahn A and Pichard AL: In vivo functional characterization of the aldolase B gene enhancer. J Biol Chem. 277:28618–28623. 2002. View Article : Google Scholar : PubMed/NCBI

81 

Yamaguchi K, Mandai M, Oura T, Matsumura N, Hamanishi J, Baba T, Matsui S, Murphy SK and Konishi I: Identification of an ovarian clear cell carcinoma gene signature that reflects inherent disease biology and the carcinogenic processes. Oncogene. 29:1741–1752. 2010. View Article : Google Scholar : PubMed/NCBI

82 

Shigetomi H, Higashiura Y, Kajihara H and Kobayashi H: A potential link of oxidative stress and cell cycle regulation for development of endometriosis. Gynecol Endocrinol. 28:897–902. 2012. View Article : Google Scholar : PubMed/NCBI

83 

Akasaka J, Uekuri C, Shigetomi H, Koike M and Kobayashi H: Hepatocyte nuclear factor (HNF)-1β and its physiological importance in endometriosis. Biomed Rep. 1:13–17. 2013. View Article : Google Scholar : PubMed/NCBI

84 

Wang X, Wu H, Yu W, Liu J, Peng J, Liao N, Zhang J, Zhang X and Hai C: Hepatocyte nuclear factor 1b is a novel negative regulator of white adipocyte differentiation. Cell Death Differ. 24:1588–1597. 2017. View Article : Google Scholar : PubMed/NCBI

85 

Lopes-Coelho F, Gouveia-Fernandes S, Gonçalves LG, Nunes C, Faustino I, Silva F, Félix A, Pereira SA and Serpa J: HNF1β drives glutathione (GSH) synthesis underlying intrinsic carboplatin resistance of ovarian clear cell carcinoma (OCCC). Tumour Biol. 37:4813–4829. 2016. View Article : Google Scholar : PubMed/NCBI

86 

Sundar R, Brown J, Ingles Russo A and Yap TA: Targeting ATR in cancer medicine. Curr Probl Cancer. 41:302–315. 2017. View Article : Google Scholar : PubMed/NCBI

87 

Shigetomi H, Sudo T, Shimada K, Uekuri C, Tsuji Y, Kanayama S, Naruse K, Yamada Y, Konishi N and Kobayashi H: Inhibition of cell death and induction of G2 arrest accumulation in human ovarian clear cells by HNF-1β transcription factor: Chemosensitivity is regulated by checkpoint kinase CHK1. Int J Gynecol Cancer. 24:838–843. 2014. View Article : Google Scholar : PubMed/NCBI

88 

Koshiyama M, Matsumura N and Konishi I: Recent concepts of ovarian carcinogenesis: Type I and type II. BioMed Res Int 2014. 9342612014.

89 

Ito F, Yoshimoto C, Yamada Y, Sudo T and Kobayashi H: The HNF-1β-USP28-Claspin pathway upregulates DNA damage-induced Chk1 activation in ovarian clear cell carcinoma. Oncotarget. 9:17512–17522. 2018. View Article : Google Scholar : PubMed/NCBI

90 

Venne AS, Kollipara L and Zahedi RP: The next level of complexity: Crosstalk of posttranslational modifications. Proteomics. 14:513–524. 2014. View Article : Google Scholar : PubMed/NCBI

91 

Manning G, Whyte DB, Martinez R, Hunter T and Sudarsanam S: The protein kinase complement of the human genome. Science. 298:1912–1934. 2002. View Article : Google Scholar : PubMed/NCBI

92 

Terman JR and Kashina A: Post-translational modification and regulation of actin. Curr Opin Cell Biol. 25:30–38. 2013. View Article : Google Scholar : PubMed/NCBI

93 

Zhang D, Zaugg K, Mak TW and Elledge SJ: A role for the deubiquitinating enzyme USP28 in control of the DNA-damage response. Cell. 126:529–542. 2006. View Article : Google Scholar : PubMed/NCBI

94 

Martín Y, Cabrera E, Amoedo H, Hernández-Pérez S, Domínguez-Kelly R and Freire R: USP29 controls the stability of checkpoint adaptor Claspin by deubiquitination. Oncogene. 34:1058–1063. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Chen S, Dai X, Gao Y, Shen F, Ding J and Chen Q: The positivity of estrogen receptor and progesterone receptor may not be associated with metastasis and recurrence in epithelial ovarian cancer. Sci Rep. 7:169222017. View Article : Google Scholar : PubMed/NCBI

96 

Long L, Cao Y and Tang LD: Transmembrane estrogen receptor GPR30 is more frequently expressed in malignant than benign ovarian endometriotic cysts and correlates with MMP-9 expression. Int J Gynecol Cancer. 22:539–545. 2012. View Article : Google Scholar : PubMed/NCBI

97 

Yamaguchi K, Huang Z, Matsumura N, Mandai M, Okamoto T, Baba T, Konishi I, Berchuck A and Murphy SK: Epigenetic determinants of ovarian clear cell carcinoma biology. Int J Cancer. 135:585–597. 2014. View Article : Google Scholar : PubMed/NCBI

98 

Wendel JRH, Wang X and Hawkins SM: The endometriotic tumor microenvironment in ovarian cancer. Cancers (Basel). 10:E2612018. View Article : Google Scholar : PubMed/NCBI

99 

Kobayashi H, Sugimoto H, Onishi S and Nakano K: Novel biomarker candidates for the diagnosis of ovarian clear cell carcinoma. Oncol Lett. 10:612–618. 2015. View Article : Google Scholar : PubMed/NCBI

100 

Liu H, Wang J, Wang H, Tang N, Li Y, Zhang Y and Hao T: Correlation between matrix metalloproteinase-9 and endometriosis. Int J Clin Exp Pathol. 8:13399–13404. 2015.PubMed/NCBI

101 

Kisielewski R, Tołwińska A, Mazurek A and Laudański P: Inflammation and ovarian cancer--current views. Ginekol Pol. 84:293–297. 2013. View Article : Google Scholar : PubMed/NCBI

102 

Kaponis A, Iwabe T, Taniguchi F, Ito M, Deura I, Decavalas G, Terakawa N and Harada T: The role of NF-kappaB in endometriosis. Front Biosci (Schol Ed). 4:1213–1234. 2012.PubMed/NCBI

103 

Suzuki E, Kajita S, Takahashi H, Matsumoto T, Tsuruta T and Saegusa M: Transcriptional upregulation of HNF-1β by NF-κB in ovarian clear cell carcinoma modulates susceptibility to apoptosis through alteration in bcl-2 expression. Lab Invest. 95:962–972. 2015. View Article : Google Scholar : PubMed/NCBI

104 

Jana S, Chatterjee K, Ray AK, DasMahapatra P and Swarnakar S: Regulation of matrix metalloproteinase-2 activity by COX-2-PGE2-pAKT axis promotes angiogenesis in endometriosis. PLoS One. 11:e01635402016. View Article : Google Scholar : PubMed/NCBI

105 

Ruiz LA, Báez-Vega PM, Ruiz A, Peterse DP, Monteiro JB, Bracero N, Beauchamp P, Fazleabas AT and Flores I: Dysregulation of lysyl oxidase expression in lesions and endometrium of women with endometriosis. Reprod Sci. 22:1496–1508. 2015. View Article : Google Scholar : PubMed/NCBI

106 

Ruiz A, Ruiz L, Colón-Caraballo M, Torres-Collazo BJ, Monteiro JB, Bayona M, Fazleabas AT and Flores I: Pharmacological blockage of the CXCR4-CXCL12 axis in endometriosis leads to contrasting effects in proliferation, migration, and invasion. Biol Reprod. 98:4–14. 2018. View Article : Google Scholar : PubMed/NCBI

107 

Del Carmen MG, Smith Sehdev AE, Fader AN, Zahurak ML, Richardson M, Fruehauf JP, Montz FJ and Bristow RE: Endometriosis-associated ovarian carcinoma: Differential expression of vascular endothelial growth factor and estrogen/progesterone receptors. Cancer. 98:1658–1663. 2003. View Article : Google Scholar : PubMed/NCBI

108 

Laschke MW and Menger MD: Basic mechanisms of vascularization in endometriosis and their clinical implications. Hum Reprod Update. 24:207–224. 2018. View Article : Google Scholar

109 

Becker CM, Beaudry P, Funakoshi T, Benny O, Zaslavsky A, Zurakowski D, Folkman J, D'Amato RJ and Ryeom S: Circulating endothelial progenitor cells are up-regulated in a mouse model of endometriosis. Am J Pathol. 178:1782–1791. 2011. View Article : Google Scholar : PubMed/NCBI

110 

Wei JJ, William J and Bulun S: Endometriosis and ovarian cancer: A review of clinical, pathologic, and molecular aspects. Int J Gynecol Pathol. 30:553–568. 2011. View Article : Google Scholar : PubMed/NCBI

111 

Mandai M, Yamaguchi K, Matsumura N, Baba T and Konishi I: Ovarian cancer in endometriosis: Molecular biology, pathology, and clinical management. Int J Clin Oncol. 14:383–391. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2019
Volume 41 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kobayashi H, Yamada Y, Kawahara N, Ogawa K and Yoshimoto C: Integrating modern approaches to pathogenetic concepts of malignant transformation of endometriosis. Oncol Rep 41: 1729-1738, 2019
APA
Kobayashi, H., Yamada, Y., Kawahara, N., Ogawa, K., & Yoshimoto, C. (2019). Integrating modern approaches to pathogenetic concepts of malignant transformation of endometriosis. Oncology Reports, 41, 1729-1738. https://doi.org/10.3892/or.2018.6946
MLA
Kobayashi, H., Yamada, Y., Kawahara, N., Ogawa, K., Yoshimoto, C."Integrating modern approaches to pathogenetic concepts of malignant transformation of endometriosis". Oncology Reports 41.3 (2019): 1729-1738.
Chicago
Kobayashi, H., Yamada, Y., Kawahara, N., Ogawa, K., Yoshimoto, C."Integrating modern approaches to pathogenetic concepts of malignant transformation of endometriosis". Oncology Reports 41, no. 3 (2019): 1729-1738. https://doi.org/10.3892/or.2018.6946