Open Access

Inhibition of TP signaling promotes endometriosis growth and neovascularization

  • Authors:
    • Akiko Furue
    • Kyoko Hattori
    • Kanako Hosono
    • Mina Tanabe
    • Erina Sato
    • Masako Honda
    • Kazuki Sekiguchi
    • Yoshiya Ito
    • Masataka Majima
    • Shuh Narumiya
    • Kazuyoshi Kato
    • Hideki Amano
  • View Affiliations

  • Published online on: August 30, 2023     https://doi.org/10.3892/mmr.2023.13079
  • Article Number: 192
  • Copyright: © Furue et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Endometriosis is highly dependent on angiogenesis and lymphangiogenesis. Prostaglandin E2, an arachidonic acid metabolite, has been shown to promote the formation of new blood and lymphatic vessels. However, the role of another arachidonic acid metabolite, thromboxane A2 (TXA2) in angiogenesis and lymphangiogenesis during endometriosis remains largely unexplored. Using a murine model of ectopic endometrial transplantation, fragments from the endometrium of WT donor mice were transplanted into the peritoneal walls of recipient WT mice (WT→WT), resulting in an increase in both the area and density of blood and lymphatic vessels. Upon transplantation of endometrial tissue from thromboxane prostanoid (TP) receptor (TXA2 receptor)‑deficient (TP‑/‑) mice into TP‑/‑ mice (TP‑/‑→TP‑/‑), an increase in implant growth, angiogenesis, and lymphangiogenesis were observed along with upregulation of pro‑angiogenic and lymphangiogenic factors, including vascular endothelial growth factors (VEGFs). Similar results were obtained using a thromboxane synthase (TXS) inhibitor in WT→WT mice. Furthermore, TP‑/‑→TP‑/‑ mice had a higher number of F4/80+ cells than that of WT→WT mice, with increased expression of genes related to the anti‑inflammatory macrophage phenotype in endometrial lesions. In cultured bone marrow (BM)‑derived macrophages, the levels of VEGF‑A, VEGF‑C, and VEGF‑D decreased in a TP‑dependent manner. Furthermore, TP signaling affected the polarization of cultured BM‑derived macrophages to the anti‑inflammatory phenotype. These findings imply that inhibition of TP signaling promotes endometrial implant growth and neovascularization.

Introduction

Endometriosis is a serious gynecological pathology characterized by the ectopic implantation of endometrial stroma and epithelium outside the uterus (1). This disease affects ~10% of women of reproductive age and leads to numerous consequences. Despite its prevalence, patients often experience severe dysmenorrhea (painful menstruation), chronic pelvic pain, and infertility, and current therapeutic regimens frequently fail to alleviate these symptoms (2,3). Furthermore, its exact pathophysiology remains unknown.

Endometriosis is highly dependent on angiogenesis, which involves the formation of new blood vessels from pre-existing ones (4,5). In women with endometriosis, the potent pro-angiogenic stimulating factor vascular endothelial growth factor (VEGF)-A is elevated in peritoneal fluids and endometriosis lesions (4). In a model of ectopic endometriosis established by the transplantation of endometrial fragments from donor mice into the peritoneal wall of host mice, the implant area exhibited enhanced angiogenesis, as evidenced by the upregulation of pro-angiogenic factors, including VEGF-A, and endothelial cell markers, including cluster of differentiation 31 (CD31) and VEGF receptor 2 (VEGFR2), in the implant (6). Inhibition of VEGF-A suppressed the growth of endometriosis and vascular density in endometrial implants in experimental models (5,7).

Recent studies have revealed that lymphangiogenesis, the formation of new lymphatic vessels, is enhanced in endometriotic lesions (8,9). In humans, endometriosis is associated with an increased expression of pro-lymphangiogenic stimulating factors and lymphatic endothelial cell markers (4). In an ectopic endometrial transplantation model, implant growth and lymphangiogenesis were associated with increased mRNA expression of the two pro-lymphangiogenic factors VEGF-C and VEGF-D (8,9). VEGFR3, a receptor for VEGF-C and VEGF-D, kinase inhibitor reduced implant growth and lymphangiogenesis, implying that lymphangiogenesis is involved in endometriosis progression (8). However, the mechanisms underlying angiogenesis and lymphangiogenesis in endometriosis remain unclear.

Prostanoids, including prostaglandin (PG) and thromboxane (TX), are arachidonic acid metabolites. PGs have been linked to the pathogenesis of endometriosis (7,10), particularly via the cyclooxygenase-2 (COX-2)/PGE2 pathway. COX-2 participates in the onset and progression of endometriosis (11), and enhanced expression of COX-2 has been observed in endometriotic lesions (12). In addition, VEGF-A induced by microsomal PGE synthase-1 (mPGES-1), an essential enzyme in PGE2 synthesis, promoted endometriosis and angiogenesis of endometriotic lesions in mice (7). In terms of PGE2-mediated lymphangiogenesis, COX-2-derived PGE2 was found to enhance lymphangiogenesis through upregulation of pro-lymphangiogenic stimulating factors including VEGF-C and VEGF-D, in chronic inflammation (13,14), and during the healing process of gastric ulcers (15) and wounds (16).

In addition to PGE2, TXA2 is another representative prostanoid produced by COX and TX synthase (TXS), which exerts its activity via the TXA2 receptor, also known as the thromboxane prostanoid (TP) receptor (17). TP plays a key role in the aggregation of platelets and in the constriction of vascular smooth muscle cells, thus contributing to cardiovascular and cerebrovascular diseases, particularly ischemic stroke, which is associated with high expression of TXA2 (18). In mice, TP signaling facilitated blood flow recovery from hind limb ischemia via angiogenesis in ischemic tissues (19). Furthermore, TP signaling in macrophages promoted lymphangiogenesis in the diaphragm during endotoxin-induced peritonitis (20). The above findings imply that TP signaling is involved in angiogenesis and lymphangiogenesis in endometriotic lesions and promotes endometriotic growth. Thus, the present study aimed to examine the effect of inhibition of TP signaling on the development of endometriosis and the formation of new blood and lymphatic vessels.

Materials and methods

Animals

Female TP-deficient (TP−/−) mice (8 weeks old) were generated as previously described (21). Female C57BL/6 WT mice (8 weeks old) were obtained from CLEA Japan, Inc. All experimental studies were approved by the Dean of Kitasato University School of Medicine after review by the Institutional Animal Care and Use Committee (approval no. 2022-060). The experiments were performed in accordance with the guidelines of the Science Council of Japan for animal experiments. In the present study, 75 mice were subjected to endometrial transplantation (n=39), pharmacological intervention (n=12), and cell culture (n=24).

Experimental model of endometriosis

An endometrial transplantation model was established as previously described (9). Briefly, mice were anesthetized by intraperitoneal (i.p.) injection of mixed anesthetic agents containing 4.0 mg/kg of midazolam (cat. no. 614243022; Sandoz; Novartis), 0.75 mg/kg medetomidine hydrochloride (cat. no. 14111; Nippon Zenyaku Kogyo), and 5.0 mg/kg butorphanol (cat. no. 219711; Meiji Seika Pharma). The anesthetic mixture of medetomidine, midazolam, and butorphanol has been recently used as a substitute for ketamine or pentobarbital sodium (22). The combination of anesthetics was approved by the Dean of Kitasato University School of Medicine after the review by the Institutional Animal Care and Use Committee. To rule out endogenous estrogen effects and menstrual cycle influences in mice, the bilateral ovaries were removed through paravertebral incisions. The effects of medetomidine were reversed by an i.p. injection of 0.75 mg/kg atipamezole (Nippon Zenyaku Kogyo). All donor and recipient mice were subcutaneously administered estradiol valerate (100 mg/kg) (Pelanin Depot; cat. no. 4987224136400; Mochida Seiyaku) weekly from the day of ovariectomy (6). Uterine tissues from donor mice were harvested 7 days after ovariectomy. A circular uterine fragment with a diameter of 3 mm was implanted into each side of the peritoneal wall of recipient mice and secured with 7-0 polypropylene sutures (cat. no. C0024501; B-Brown Ace Scrap, Inc.). Recipient WT or TP−/− mice received an implant from donor WT or TP−/− mice (Henceforth referred to as WT−/−→WT−/− and TP−/−→TP−/−). The day of implantation was defined as day 0. Six animals received daily oral administration of ozagrel (30 mg/kg; cat. no. 87449; Kissei Pharmaceutical Co., Ltd.), a selective TXS inhibitor (20). Humane endpoints during the experiment were defined as food-water intake difficulties, weight loss ≥20% of the body weight, and reduced activities and movement.

On day 14 after endometrial transplantation, the animals were euthanized using 5% isoflurane (cat. no. 4987114133403; Pfizer) for 5 min followed by cervical dislocation, and the death was confirmed by a lack of heartbeat and respiration. Then, each side of the implanted tissue was excised. Day 0 samples were collected immediately after implantation. The excised implants were digitally photographed, and the implant area (mm2) was determined using ImageJ version 1.53e (National Institutes of Health). The results are presented as the implant area per mm2. One implant sample was prepared for reverse transcription-quantitative PCR (RT-qPCR), and the other for histological analysis.

Immunofluorescence analysis

Fixed endometrial samples were embedded in Tissue-Tek O.C.T. Compound (Sakura Finetek USA, Inc.), frozen at −80°C, and cut into 8-µm thick sections. The sections were incubated overnight at 4°C with one of the following primary antibodies: Rabbit anti-mouse TP (1:100; cat. no. APR-069; Almone Labs, Jerusalem, Israel), rabbit anti-mouse TXS (1:100; cat. no. bs-4019R; Bioss Antibodies Inc, Woburn, MA, USA), rat anti-mouse CD31 monoclonal antibody (1:200; cat. no. 553370; BD Biosciences), rabbit anti-mouse lymphatic vessel endothelial hyaluronan receptor 1 (LYVE-1) (1:100; cat. no. ab14917; Abcam), goat anti-mouse LYVE-1 (1:100; cat. no. AF2125; R&D Systems Inc.), goat anti-mouse VEGF-A (1:100; cat. no. AF-493-NA; R&D Systems Inc.), rabbit anti-VEGF-C (1:100; cat. no. ab9546; Abcam), goat anti-VEGF-D (1:50; cat. no. sc6313; Santa Cruz Biotechnology, Inc.), rabbit anti-mouse CD41, a marker for platelets (1:100; cat. no. MCA2245GA; Bio-Rad Laboratories, Inc.), and rat anti-mouse F4/80, a marker for macrophages (1:100; cat. no. MCA497G; Bio-Rad Laboratories, Inc.). The sections were incubated for 1 h at room temperature with one of the following species-appropriate secondary antibodies: Alexa Fluor 488-conjugated donkey anti-rabbit IgG (cat. no. A21206), Alexa Fluor 488-conjugated donkey anti-goat IgG (cat. no. A11055), Alexa Fluor 594-conjugated donkey anti-rat IgG (cat. no. A21209) or Alexa Fluor 594-conjugated donkey anti-goat IgG (cat. no. A11058) (1:200; Molecular Probes; Thermo Fisher Scientific, Inc.). Images of stained sections were captured using a fluorescence microscope (Biozero BZ-700; Keyence Corporation). The number of F4/80+ cells was counted in five fields of view (×400 magnification) of the endometrial tissue.

Determination of vessel density

Microvessel density (MVD) and lymphatic vessel density (LVD) within endometrial tissue implants have been used to assess angiogenesis and lymphangiogenesis, respectively (9). The number of CD31+ blood vessels and LYVE-1+ lymphatic vessels in four fields of view were counted using a fluorescence microscope (Biozero BZ-700; Keyence Corporation) (×200 magnification). The results are presented as the number of CD31+ blood vessels or LYVE-1+ lymphatic vessels per square millimeter (MVD/mm2 or LVD/mm2). In addition, the area covered by blood and lymphatic vessels was determined using ImageJ and presented as the percentage of the total area observed [microvessel area (MVA)% or lymphatic vessel area (LVA)%].

RT-qPCR analysis

Total RNA was extracted from endometriotic tissues and homogenized in TRIzol® reagent (cat. no. 15596018; Thermo Fisher Scientific, Inc.). Total RNA (1 µg) was transcribed into single-stranded cDNA using ReverTra Ace qPCR RT Kit (cat. no. FSQ-201; TOYOBO Co., Ltd.) according to the manufacturer's protocol. qPCR was performed using TB Green Premix Ex Taq II (Tli RNase H Plus; cat. no. RR820A; Takara Bio, Inc.). PCR amplification was performed under the following thermocycling conditions: Pre-denaturation at 95°C for 10 sec, followed by a two-step PCR program consisting of 40 cycles at 95°C for 3 sec and 60°C for 20 sec. mRNA expression levels were calculated using the comparative threshold cycle method (2-∆∆Cq) (23) and normalized to GAPDH expression in each sample. The sequences of the primers used are listed in Table I.

Table I.

Sequences of the primers used for PCR.

Table I.

Sequences of the primers used for PCR.

GeneForward primer sequence (5′-3′)Reverse primer sequence (5′-3′)
TP CCTCCTGCTCAACACCGTTAG CTGAACCATCATCTCCACCTC
TXS GGATTCTGCCCAATAAGAACC GAAGTCTCTCCGCCTCTCTTC
VEGF-A ACGACAGAAGGAGAGCAGAAG ATGTCCACCAGGGTCTCAATC
VEGF-C TCTGTGTCCAGCGTAGATGAG GTCCCCTGTCCTGGTATTGAG
VEGF-D CCTATTGACATGCTGTGGGAT GTGGGTTCCTGGAGGTAAGAG
CD31 CAGAGCCAGCAGTATGAGGAC GCAACTATTAAGGTGGCGATG
VEGFR2 CTGCCTACCTCACCTGTTTCC CGGCTCTTTCGCTTACTGTTC
VEGFR3 GGAAGGCTCTGAAGATAAAGG ACAGAAGATGAGCAGGAGGAG
LYVE-1 GCTCTCCTCTTCTTTGGTGCT TGACGTCATCAGCCTTCTCTT
Prox1 GTTCTTTTACACCCGCTACCC ACTCACGGAAATTGCTGAACC
CCL2 CGGAACCAAATGAGATCAGAA TTGTGGAAAAGGTAGTGGATG
CCR2 TTACCTCAGTTCATCCACGGC CAAGGCTCACCATCATCGTAG
TNFα TCTTCTCATTCCTGCTTGTGG GATCTGAGTGTGAGGGTCTGG
IL-1β TACATCAGCACCTCACAAGCA CCAGCCCATACTTTAGGAAGA
IL-6 CAAAGCCAGAGTCCTTCAGAG TAGGAGAGCATTGGAAATTGG
MR TTTGTCCATTGCACTTTGAGG TGCCAGGTTAAAGCAGACTTG
Fizz1 CAAGGAACTTCTTGCCAATCCAG CCAAGATCCACAGGCAAAGCCA
IL-10 CGGAAATGATCCAGTTTTACC TGAGGGTCTTCAGCTTCTCAC
GAPDH ACATCAAGAAGGTGGTGAAGC AAGGTGGAAGAGTGGGAGTTG

[i] TP, thromboxane prostanoid; TXS, thromboxane synthase; VEGF, vascular endothelial growth factor; CD, cluster of differentiation; VEGFR, vascular endothelial growth factor receptor; LYVE-1, lymphatic vessel endothelial hyaluronan receptor 1; Prox1, prospero-related homeobox 1; CCL2, C-C motif chemokine 2; CCR2, C-C motif chemokine receptor 2; TNF-α, tumor necrosis factor-α; IL-1β, interleukin 1β; MR, mannose receptor; GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

Cell preparation and culture

Bone marrow (BM) cells were isolated from the femur and tibia of 8-week-old female WT and TP−/− mice. BM cells were cultured in 6-well plates (1.0×106 cells/well) and maintained in RPMI-1640 medium (cat. no. 11875-093; Gibco; Thermo Scientific) supplemented with 10% fetal calf serum and 20 ng/ml macrophage colony-stimulating factor (cat. no. 576406; BioLegend, Inc.), as previously described (9). On day 7, the cells stimulated with the TP receptor agonist U46619 (100 nM; catalog no. 16450; Cayman Chemical) were either left untreated or treated with lipopolysaccharide (10 ng/ml; cat. no. L3012; MilliporeSigma) in RPMI 1640 medium for 3 h. Cultured BM-derived macrophages were harvested and homogenized in TRIzol® reagent (cat. no. 15596018; Thermo Fisher Scientific, Inc.), and mRNA levels were determined using RT-qPCR.

Statistical analysis

All results are presented as the mean and SD. All statistical analyses were performed using GraphPad Prism version 9 (GraphPad Software, Inc.). Data between two groups were compared using an unpaired two-tailed Student's t-test, whereas comparisons between multiple groups were conducted using a one-way ANOVA followed by a Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Inhibition of TP signaling in the host enhances the development of endometriosis lesions

To investigate the involvement of TP signaling in implant growth, WT or TP−/− endometrial tissues were implanted into either WT or TP−/− recipient mice (Fig. 1). As previous data have shown that the area of endometrial implants reached a maximum on day 14 (6,9), the endometrial implant area was assessed on day 14. When TP−/− endometrial fragments were implanted into TP−/− mice (TP−/−→TP−/− mice), implant growth was greater than that in WT→WT mice (Fig. 1A). Similarly, implant growth was enhanced in the WT→TP−/− mice; however, no statistically significant difference in implant growth was observed between TP−/−→WT and WT→WT mice. These findings suggested that TP signaling acquired from recipients suppressed the development of transplanted endometrial fragments. Accordingly, the growth of implants as well as blood and lymphatic vessels in WT→WT and TP−/−→TP−/− mice was assessed in the subsequent experiments.

On day 14, the mRNA expression levels of TXS and TP in WT→WT mouse implants increased. However, TXS mRNA expression levels in TP−/−→TP−/− mouse implants did not differ from those in the WT→WT mouse implants. In addition, TP mRNA expression levels were reduced in TP−/−→TP−/− mouse implants (Fig. 1B). On day 14, double immunofluorescence analysis demonstrated that TXS and TP expression in endometrial implants from WT→WT mice co-localized with those of F4/80 in stromal tissues, indicating macrophages as a source of TXS and TP in the implants (Fig. 1C).

Since TP is expressed in platelets, platelets were also assessed in endometrial tissues. Immunofluorescence analysis revealed that the distribution of CD41, a platelet marker, did not differ between WT→WT and TP−/−→TP−/− mice (Fig. 1D).

Angiogenesis in endometrial implants is enhanced in TP−/−→TP−/− mice

As endometrial implant growth is attributed to angiogenesis (6,9), the involvement of TP signaling in angiogenesis was investigated by counting the number of CD31+ blood vessels in the WT→WT and TP−/−→TP−/− mouse implants. Fig. 2A shows that the TP−/−→TP−/− mouse implants displayed a greater number of newly formed CD31+ blood vessels compared with WT→WT mouse implants. The MVD and MVA% in the implants of TP−/−→TP−/− mice were greater than those in the WT→WT mouse implants (Fig. 2B). Additionally, mRNA expression levels of CD31, VEGFR2, and pro-angiogenic factor VEGF-A were higher in the TP−/−→TP−/− mouse implants than those in the WT→WT mouse implants (Fig. 2C). Dual immunofluorescence staining demonstrated a lack of co-localization of TP with CD31 in implant tissues (Fig. 2D), suggesting that TP was not expressed in newly formed blood vessels. These findings suggest that inhibition of TP signaling does not directly facilitate angiogenesis in endometrial implants.

Lymphangiogenesis in endometrial implants is enhanced in TP−/−→TP−/− mice

The role of TP signaling in lymphangiogenesis was investigated by determining LVD and LVA% (Fig. 3A and B). LVD and LVA% in TP−/−→TP−/− mouse implants were higher than those in WT→WT mouse implants. The mRNA expression levels of lymphatic endothelial cell markers, including LYVE-1, VEGFR3, and Prospero-related homeobox 1 (Prox1), a marker for lymphangiogenesis (24), in TP−/−→TP−/− mouse implants were higher than those in WT→WT mouse implants (Fig. 3C). The mRNA expression levels of VEGF-C and VEGF-D were also higher in TP−/−→TP−/− mouse implants. These results indicate that inhibition of TP signaling stimulates lymphangiogenesis through upregulation of VEGF-C and VEGF-D. Accordingly, the localization of TP in lymphatic endothelial cells (ECs) was investigated (Fig. 3D). Dual immunofluorescence staining for TP and LYVE-1 revealed the absence of colocalization, suggesting that TP was not expressed in newly formed lymphatic vessels. Therefore, these results suggested that inhibition of TP signaling did not directly facilitate lymphangiogenesis in endometrial implants.

TXS inhibition promotes implant growth and the formation of new blood and lymphatic vessels

To confirm the role of TP signaling in endometriosis, the effects of ozagrel, a TXS inhibitor, on implant growth, angiogenesis, and lymphangiogenesis were investigated (Fig. 4). Ozagrel increased the implant area in WT→WT mice (Fig. 4A), enhanced angiogenesis and lymphangiogenesis in the implant, as evidenced by the increased MVD and MVA% (Fig. 4B), and increased LVD and LVA%, respectively (Fig. 4C). Furthermore, it enhanced the mRNA expression levels of pro-angiogenesis-related genes, including CD31, VEGFR2, and VEGF-A (Fig. 4B), and pro-lymphangiogenesis-related genes, including LYVE-1, VEGFR3, Prox1, VEGF-C, and VEGF-D in the implants (Fig. 4C).

Macrophages recruited in implants express pro-angiogenic or lymphangiogenic cytokines

Since macrophages contribute to angiogenesis and lymphangiogenesis during endometriosis development in mice (9), the number of F4/80+ cells within endometrial implants was counted (Fig. 5A). The number of F4/80+ cells was higher in TP−/−→TP−/− mouse implants than that in WT→WT mouse implants, indicating that F4/80+ cell accumulation is linked to angiogenesis and lymphangiogenesis in the implants. In addition, enhanced F4/80+ cell accumulation in implants was associated with upregulation of the macrophage chemoattractant C-C motif chemokine 2 (CCL2) and its receptor C-C motif chemokine 2 (CCR2) (Fig. 5B).

Macrophages play a pivotal role in synthesizing the VEGF protein family, which is responsible for angiogenesis (VEGF-A) and lymphangiogenesis (VEGF-C and VEGF-D). Accordingly, whether macrophages recruited in implants expressed VEGF-A, VEGF-C, or VEGF-D was determined (Fig. 5C). Double immunofluorescence analyses demonstrated that F4/80+ cells co-localized with VEGF-A as well as VEGF-C/D expression in WT→WT mouse implants on day 14 (Fig. 5C). These findings suggest that macrophages participate in angiogenesis and lymphangiogenesis in endometrial implants through upregulation of VEGF-A and VEGF-C/D, respectively.

The expression levels of the anti-inflammatory factors are increased in TP-/-→TP-/- mouse implants

The mRNA expression levels of tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6 in pro-inflammatory macrophages, and those of mannose receptor (MR), Fizz1, and IL-10 in anti-inflammatory macrophages were investigated (Fig. 6). The mRNA expression levels related to pro-inflammatory macrophage phenotypes did not differ substantially between the WT→WT and TP−/−→TP−/− mice, whereas those related to anti-inflammatory macrophage phenotypes were higher in TP−/−→TP−/− mice than in WT→WT mice, suggesting the role of anti-inflammatory macrophages in endometriosis growth.

TP stimulation reduces the expression of vascular growth factors and anti-inflammatory markers in cultured macrophages

Finally, whether inhibition of TP signaling in macrophages regulates pro-angiogenic or pro-lymphangiogenic factors in vitro was investigated. When cultured BM-macrophages were stimulated with U46619, a TXA2 analog, the mRNA expression levels of VEGF-A, VEGF-C, and VEGF-D were downregulated in BM-macrophages from WT mice, but were not altered in those from TP−/− mice (Fig. 7A). These findings indicated that VEGF-A, VEGF-C, and VEGF-D levels were reduced in cultured macrophages via TP signaling.

Additionally, the mRNA expression levels of pro- and anti-inflammatory factors in macrophages were determined in vitro. U46619 stimulation had no effect on the expression levels of pro-inflammatory factors, including TNFα, IL-1β, and IL-6, in WT and TP−/− mouse macrophages. In contrast, U46619 reduced the expression levels of anti-inflammatory factors, such as MR, Fizz1, and IL-10, in WT mouse macrophages, but had no effect on TP−/− mouse macrophages (Fig. 7B).

Discussion

Endometriosis is associated with both angiogenesis and lymphangiogenesis. Recent studies have demonstrated that TP signaling enhances pro-angiogenic activities under pathological conditions, including inflammation and tumor growth (19,25,26). The present study was the first to demonstrate that TP signaling facilitated lymphangiogenesis in inflamed diaphragms in mice (20). Additionally, using an endometrial transplantation model, the present study revealed that inhibiting endogenous TP signaling contributed to the enhancement of angiogenesis and lymphangiogenesis in endometrial lesions in addition to endometriotic growth. TXS inhibitors increased endometrial growth and formation of new blood and lymphatic vessels. Anti-inflammatory macrophages lacking TP signaling in endometrial lesions enhanced the levels of angiogenesis and lymphangiogenesis-related cytokines. These findings suggest that activation of TP signaling mitigates endometrial growth and neovascularization.

Although the underlying mechanisms of endometriosis are unknown, angiogenesis and lymphangiogenesis in endometrial lesions contribute to its development. The pro-angiogenic factor VEGF-A is upregulated in patients and mice with endometriosis. Inhibition of VEGF-A and VEGFR1 suppresses endometriosis growth and the formation of new blood vessels in endometrial lesions (6). In addition, the pro-lymphangiogenic factors VEGF-C and VEGF-D were increased during the development of endometriosis in humans and mice (9), and VEGFR3 inhibition suppressed endometriosis growth and the formation of new lymphatic vessels in endometrial lesions (8). Endometriosis was associated with overexpression of pro-angiogenic and pro-lymphangiogenic genes (27). Consistent with this, our findings revealed that the promotion of angiogenesis and lymphangiogenesis in endometrial tissue was associated with endometriotic growth.

Endometrial stromal cells have been implicated in angiogenesis and lymphangiogenesis. Macrophage accumulation in endometrial lesions was shown to be associated with neovascularization (6,8,9). BM-derived macrophages were found to be involved in endometriosis and angiogenesis (6). By contrast, large peritoneal macrophages were implicated in endometriosis (28). The accumulation of macrophages in endometrial tissues warrants further investigation. Furthermore, the specific macrophage phenotype is critical for endometriosis development (28). The results of the present study suggested that the anti-inflammatory macrophage phenotype (alternatively activated macrophages) contributed to endometriosis progression. In a murine endometriosis model, anti-inflammatory macrophages promoted the growth of endometriotic lesions, whereas pro-inflammatory macrophages protected against the disease (29). Endometriotic lesions were reduced when the anti-inflammatory macrophage phenotype expressing CD206 was deleted (30). In addition, the macrophage phenotype changes as endometriosis progresses. Pro-inflammatory macrophages appeared in the peritoneal cavity within the first 14 days after endometrial tissue implantation, and the macrophage phenotype switched to the anti-inflammatory phenotype 14 days after endometriosis induction (31). However, the oversimplification of macrophage polarization makes it difficult to understand the macrophage phenotype complex in tissues. Recent technological advances in single-cell transcript analyses have revealed five distinct subpopulations of macrophages in human endometrial lesions, suggesting the heterogeneity of macrophages during endometriosis development (32). Further studies are required to determine the macrophage subpopulations involved in the progression of endometriosis.

COX-2 is involved in the development of endometriosis (11). In patients with endometriosis, its expression is upregulated in the endometrial tissue (12). COX-2 inhibitors reduced endometrial growth, vascular density, and VEGF-A expression in rodent endometrial implants (7,33,34). Additionally, mPGES-1 deficient mice showed attenuated endometriosis growth and angiogenesis through the downregulation of VEGF-A in endometrial implants (7). These findings indicate that COX-2/mPGES-1-derived PGE2 contributes to the development of endometriosis by enhancing angiogenesis. However, the role of TXA2 in endometriosis pathology remains to be elucidated. The findings of the present study revealed that inhibition of TP signaling or TXS stimulated endometrial growth, angiogenesis, and lymphangiogenesis in endometrial lesions.

Under pathological conditions, TP signaling has both pro- and anti-angiogenic properties (35). In addition, TP signaling promotes tumor growth and angiogenesis. TXA2 stimulates the secretion of VEGF-A from tumor cells (26). Platelet-derived TXA2 promotes angiogenesis and blood flow recovery after hindlimb ischemia (19) and gastric ulcer (15). In contrast, TP activation of endothelial cells (ECs) reduces cell migration and proliferation (36). The activation of TP signaling inhibits the pro-angiogenic capacity of human umbilical vein endothelial cells (HUVECs). Deletion of TP signaling in ECs enhances VEGF-induced angiogenesis in vivo. In addition, TP activation reduces VEGFR2 expression in HUVECs, whereas TP knockdown increases its expression (37,38). The present study showed that ablation of TP signaling enhanced angiogenesis via the production of VEGF-A from macrophages accumulated in the stroma of endometrial implants. In cultured BM-macrophages, VEGF-A expression was reduced via TP signaling. As TP was not expressed in endometrial implant microvessels, activating TP signaling did not directly induce pro-angiogenic functions in the endothelium but indirectly induced angiogenesis by releasing pro-angiogenic factors from macrophages accumulated in endometrial implants.

COX-2 inhibition prolonged inflammation-induced lymphedema in mouse tails and ears (13,14), and COX-2-derived PGE2 was responsible for lymphangiogenesis (39). In terms of endogenous TXA2, recent studies have shown that TP signaling in macrophages facilitated lymphangiogenesis in the diaphragm during endotoxin-induced peritonitis (20). In contrast, ablation of TP signaling enhanced lymphangiogenesis in this murine model of endometriosis, and decreased VEGF-C and VEGF-D levels via TP signaling in cultured BM-derived macrophages. TP expression was observed in endometrial stromal macrophages rather than in lymphatic vessels. These findings imply that TP signaling in macrophages suppresses lymphangiogenesis during endometriosis. However, studies on the role of TP signaling in lymphangiogenesis have yielded contradictory results, possibly because of the differences in the experimental systems used and the sex of the mice. Recruitment of pro-inflammatory macrophages to endometrial implants can also impair lymphangiogenesis, as pro-inflammatory macrophages can disrupt the formation of new lymphatic vessels at the site of injury (40).

Stimulation of macrophages with PGE2 has been reported to change their phenotype from a pro-inflammatory to an anti-inflammatory state (16,41); however, little is known regarding the effect of TP signaling on macrophage polarization. The results of the current study suggested that ablation of TP signaling increased anti-inflammatory macrophage phenotype-related gene expression during endometriosis, including upregulation of CCL2 expression. Increased CCL2 expression has been observed in the endometrium of women with endometriosis (42) and endometrial stromal tissues from women as well as in mice with endometriosis (43), suggesting endometrial gland and stromal cells in the implant tissues as a source of CCL2. In addition, the recruitment of anti-inflammatory macrophages was associated with the upregulation of CCL2 expression in endometrial lesions, and the CCR2 inhibitor RS102895 reduced endometrial growth in endometriotic mice (43). These data suggest that the accumulation of anti-inflammatory macrophages in endometrial lesions via the CCL2/CCR2 pathway promotes endometriosis development. Further studies are required to elucidate the role of the CCL2/CCR2 axis in the recruitment of anti-inflammatory macrophages to endometrial implant tissues from TP−/− mice. The application of a TP agonist (U46619) reduced anti-inflammatory macrophage phenotype-related gene expression in cultured BM-derived macrophages, suggesting that TP signaling switches the macrophage phenotype, resulting in anti-inflammatory macrophages. In contrast, a previous study found that TP stimulation increased pro-inflammatory macrophage phenotype-related gene expression in isolated peritoneal thioglycolate-induced macrophages from male mice (44). The discrepancies in the results could be attributed to differences in sex and origin of macrophages and stimulation prior to TP agonist application.

In conclusion, the results of the present study showed that inhibition of TP signaling promoted endometriosis development by enhancing neovascularization in endometrial lesions. Inhibition of TP signaling did not act on blood and lymphatic ECs, but rather on the accumulation of anti-inflammatory macrophages into the endometrial lesions. TP receptor agonist decreased anti-inflammatory macrophage phenotype-related and neovascularization-related gene expression in cultured macrophages. Therefore, the promotion of endometriosis and neovascularization are likely driven by the inhibition of TP signaling in anti-inflammatory macrophages. However, further studies are necessary to confirm this possibility. Given that selective TP receptor activation with an agonist may reduce endometrial growth, targeting TP signaling may be a viable option for treating endometriosis; however, further research is needed.

Acknowledgments

The authors would like to thank Ms. Michiko Ogino and Ms. Kyoko Yoshikawa (both Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Japan) for their technical assistance.

Funding

This work was supported by grants from the Japanese Ministry of Education, Culture, Sports, Science and Technology (MEXT) (grant nos. 21K16821 and 21K09502), the Integrative Research Program of the Graduate School of Medical Science at Kitasato University (grant no. 2022-B26) and the Parents' Association Grant of the Kitasato University School of Medicine.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

Authors' contributions

AF conceived and designed the study, performed the experiments and wrote the manuscript. KHa, ES, MT and KHo performed the experiments. MH, KS and YI performed the data analysis and interpretation, and provided technical support. MM and SN were involved in the breeding process of the TP−/− mice and helped to design the animal experiments. KK and HA designed the study, analyzed the data and revised the manuscript. AF, YI and HA confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The animal experiments were approved by the Animal Experimentation and Ethics Committee of Kitasato University School of Medicine (approval no. 2022-060).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

BM

bone marrow

CCL2

C-C motif chemokine 2

CCR2

C-C motif chemokine receptor 2

COX

cyclooxygenase

IL

interleukin

i.p.

intraperitoneal

LVA

lymphatic vessel area

LVD

lymphatic vessel density

LYVE-1

lymphatic vessel endothelial hyaluronan receptor 1

mPGES-1

microsomal prostaglandin E synthase-1

MR

mannose receptor

MVA

microvessel area

MVD

microvessel density

PG

prostaglandin

Prox1

prospero-related homeobox 1

TP

thromboxane prostanoid

TNF-α

tumor necrosis factor-α

TXA2

thromboxane A2

TXS

thromboxane synthase

VEGF

vascular endothelial growth factor

VEGFR

VEGF receptor

References

1 

Saunders PTK and Horne AW: Endometriosis: Etiology, pathobiology, and therapeutic prospects. Cell. 184:2807–2824. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Zondervan KT, Becker CM and Missmer SA: Endometriosis. N Engl J Med. 382:1244–1256. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Taylor HS, Kotlyar AM and Flores VA: Endometriosis is a chronic systemic disease: Clinical challenges and novel innovations. Lancet. 397:839–852. 2021. View Article : Google Scholar : PubMed/NCBI

4 

Hey-Cunningham AJ, Peters KM, Zevallos HB, Berbic M, Markham R and Fraser IS: Angiogenesis, lymphangiogenesis and neurogenesis in endometriosis. Front Biosci (Elite Ed). 5:1033–1056. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Chung MS and Han SJ: Endometriosis-associated angiogenesis and anti-angiogenic therapy for endometriosis. Front Glob Womens Health. 3:8563162022. View Article : Google Scholar : PubMed/NCBI

6 

Sekiguchi K, Ito Y, Hattori K, Inoue T, Hosono K, Honda M, Numao A, Amano H, Shibuya M, Unno N and Majima M: VEGF receptor 1-expressing macrophages recruited from bone marrow enhances angiogenesis in endometrial tissues. Sci Rep. 9:70372019. View Article : Google Scholar : PubMed/NCBI

7 

Numao A, Hosono K, Suzuki T, Hayashi I, Uematsu S, Akira S, Ogino Y, Kawauchi H, Unno N and Majima M: The inducible prostaglandin E synthase mPGES-1 regulates growth of endometrial tissues and angiogenesis in a mouse implantation model. Biomed Pharmacother. 65:77–84. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Hattori K, Ito Y, Honda M, Sekiguchi K, Hosono K, Shibuya M, Unno N and Majima M: Lymphangiogenesis induced by vascular endothelial growth factor receptor 1 signaling contributes to the progression of endometriosis in mice. J Pharmacol Sci. 143:255–263. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Honda M, Ito Y, Hattori K, Hosono K, Sekiguchi K, Tsujikawa K, Unno N and Majima M: Inhibition of receptor activity-modifying protein 1 suppresses the development of endometriosis and the formation of blood and lymphatic vessels. J Cell Mol Med. 24:11984–11997. 2020. View Article : Google Scholar : PubMed/NCBI

10 

Motohashi E, Kawauchi H, Endo H, Kondo H, Kitasato H, Kuramoto H, Majima M, Unno N and Hayashi I: Regulatory expression of lipoxin A4 receptor in physiologically estrus cycle and pathologically endometriosis. Biomed Pharmacother. 59:330–338. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Lai ZZ, Yang HL, Ha SY, Chang KK, Mei J, Zhou WJ, Qiu XM, Wang XQ, Zhu R, Li DJ and Li MQ: Cyclooxygenase-2 in endometriosis. Int J Biol Sci. 15:2783–2797. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Lousse JC, Defrere S, Colette S, Van Langendonckt A and Donnez J: Expression of eicosanoid biosynthetic and catabolic enzymes in peritoneal endometriosis. Hum Reprod. 25:734–741. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Hosono K, Suzuki T, Tamaki H, Sakagami H, Hayashi I, Narumiya S, Alitalo K and Majima M: Roles of prostaglandin E2-EP3/EP4 receptor signaling in the enhancement of lymphangiogenesis during fibroblast growth factor-2-induced granulation formation. Arterioscler Thromb Vasc Biol. 31:1049–1058. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Kashiwagi S, Hosono K, Suzuki T, Takeda A, Uchinuma E and Majima M: Role of COX-2 in lymphangiogenesis and restoration of lymphatic flow in secondary lymphedema. Lab Invest. 91:1314–1325. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Ae T, Ohno T, Hattori Y, Suzuki T, Hosono K, Minamino T, Sato T, Uematsu S, Akira S, Koizumi W and Majima M: Role of microsomal prostaglandin E synthase-1 in the facilitation of angiogenesis and the healing of gastric ulcers. Am J Physiol Gastrointest Liver Physiol. 299:G1139–G1146. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Hosono K, Isonaka R, Kawakami T, Narumiya S and Majima M: Signaling of prostaglandin E receptors, EP3 and EP4 facilitates wound healing and lymphangiogenesis with enhanced recruitment of M2 macrophages in mice. PLoS One. 11:e01625322016. View Article : Google Scholar : PubMed/NCBI

17 

Narumiya S, Sugimoto Y and Ushikubi F: Prostanoid receptors: Structures, properties, and functions. Physiol Rev. 79:1193–1226. 1999. View Article : Google Scholar : PubMed/NCBI

18 

Szczuko M, Koziol I, Kotlega D, Brodowski J and Drozd A: The role of thromboxane in the course and treatment of ischemic stroke: Review. Int J Mol Sci. 22:116442021. View Article : Google Scholar : PubMed/NCBI

19 

Amano H, Ito Y, Eshima K, Kato S, Ogawa F, Hosono K, Oba K, Tamaki H, Sakagami H, Shibuya M, et al: Thromboxane A2 induces blood flow recovery via platelet adhesion to ischaemic regions. Cardiovasc Res. 107:509–521. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Matsuda H, Ito Y, Hosono K, Tsuru S, Inoue T, Nakamoto S, Kurashige C, Hirashima M, Narumiya S, Okamoto H and Majima M: Roles of thromboxane receptor signaling in enhancement of lipopolysaccharide-induced lymphangiogenesis and lymphatic drainage function in diaphragm. Arterioscler Thromb Vasc Biol. 41:1390–1407. 2021. View Article : Google Scholar : PubMed/NCBI

21 

Takayama K, Yuhki K, Ono K, Fujino T, Hara A, Yamada T, Kuriyama S, Karibe H, Okada Y, Takahata O, et al: Thromboxane A2 and prostaglandin F2alpha mediate inflammatory tachycardia. Nat Med. 11:562–566. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Navarro KL, Huss M, Smith JC, Sharp P, Marx JO and Pacharinsak C: Mouse anesthesia: The art and science. ILAR J. 62:238–273. 2021. View Article : Google Scholar : PubMed/NCBI

23 

Schmittgen TD and Livak KJ: Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 3:1101–1108. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Wigle JT and Oliver G: Prox1 function is required for the development of the murine lymphatic system. Cell. 98:769–778. 1999. View Article : Google Scholar : PubMed/NCBI

25 

Majima M, Hosono K, Ito Y and Amano H: Biologically active lipids in the regulation of lymphangiogenesis in disease states. Pharmacol Ther. 232:1080112022. View Article : Google Scholar : PubMed/NCBI

26 

Wei J, Yan W, Li X, Ding Y and Tai HH: Thromboxane receptor alpha mediates tumor growth and angiogenesis via induction of vascular endothelial growth factor expression in human lung cancer cells. Lung Cancer. 69:26–32. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Fonseca MAS, Haro M, Wright KN, Lin X, Abbasi F, Sun J, Hernandez L, Orr NL, Hong J, Choi-Kuaea Y, et al: Single-cell transcriptomic analysis of endometriosis. Nat Genet. 55:255–267. 2023. View Article : Google Scholar : PubMed/NCBI

28 

Hogg C, Horne AW and Greaves E: Endometriosis-associated macrophages: Origin, phenotype, and function. Front Endocrinol (Lausanne). 11:72020. View Article : Google Scholar : PubMed/NCBI

29 

Bacci M, Capobianco A, Monno A, Cottone L, Di Puppo F, Camisa B, Mariani M, Brignole C, Ponzoni M, Ferrari S, et al: Macrophages are alternatively activated in patients with endometriosis and required for growth and vascularization of lesions in a mouse model of disease. Am J Pathol. 175:547–556. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Ono Y, Yoshino O, Hiraoka T, Akiyama I, Sato E, Ito M, Kobayashi M, Nakashima A, Wada S, Onda T, et al: IL-33 exacerbates endometriotic lesions via polarizing peritoneal macrophages to M2 subtype. Reprod Sci. 27:869–876. 2020. View Article : Google Scholar : PubMed/NCBI

31 

Johan MZ, Ingman WV, Robertson SA and Hull ML: Macrophages infiltrating endometriosis-like lesions exhibit progressive phenotype changes in a heterologous mouse model. J Reprod Immunol. 132:1–8. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Tan Y, Flynn WF, Sivajothi S, Luo D, Bozal SB, Davé M, Luciano AA, Robson P, Luciano DE and Courtois ET: Single-cell analysis of endometriosis reveals a coordinated transcriptional programme driving immunotolerance and angiogenesis across eutopic and ectopic tissues. Nat Cell Biol. 24:1306–1318. 2022. View Article : Google Scholar : PubMed/NCBI

33 

Machado DE, Berardo PT, Landgraf RG, Fernandes PD, Palmero C, Alves LM, Abrao MS and Nasciutti LE: A selective cyclooxygenase-2 inhibitor suppresses the growth of endometriosis with an antiangiogenic effect in a rat model. Fertil Steril. 93:2674–2679. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Olivares C, Ricci A, Bilotas M, Baranao RI and Meresman G: The inhibitory effect of celecoxib and rosiglitazone on experimental endometriosis. Fertil Steril. 96:428–433. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Ashton AW, Zhang Y, Cazzolli R and Honn KV: The role and regulation of thromboxane A2 signaling in cancer-trojan horses and misdirection. Molecules. 27:62342022. View Article : Google Scholar : PubMed/NCBI

36 

Ashton AW, Yokota R, John G, Zhao S, Suadicani SO, Spray DC and Ware JA: Inhibition of endothelial cell migration, intercellular communication, and vascular tube formation by thromboxane A(2). J Biol Chem. 274:35562–35570. 1999. View Article : Google Scholar : PubMed/NCBI

37 

Tsou PS, Amin MA, Campbell PL, Zakhem G, Balogh B, Edhayan G, Ohara RA, Schiopu E, Khanna D, Koch AE and Fox DA: Activation of the thromboxane A2 receptor by 8-isoprostane inhibits the pro-angiogenic effect of vascular endothelial growth factor in scleroderma. J Invest Dermatol. 135:3153–3162. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Eckenstaler R, Ripperger A, Hauke M, Petermann M, Hemkemeyer SA, Schwedhelm E, Ergün S, Frye M, Werz O, Koeberle A, et al: A thromboxane A2 receptor-driven COX-2-dependent feedback loop that affects endothelial homeostasis and angiogenesis. Arterioscler Thromb Vasc Biol. 42:444–461. 2022. View Article : Google Scholar : PubMed/NCBI

39 

Matsuda H, Hosono K, Tsuru S, Kurashige C, Sekiguchi K, Akira S, Uematsu S, Okamoto H and Majima M: Roles of mPGES-1, an inducible prostaglandin E synthase, in enhancement of LPS-induced lymphangiogenesis in a mouse peritonitis model. Life Sci. 142:1–7. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Mishima T, Ito Y, Nishizawa N, Amano H, Tsujikawa K, Miyaji K, Watanabe M and Majima M: RAMP1 signaling improves lymphedema and promotes lymphangiogenesis in mice. J Surg Res. 219:50–60. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Nishizawa N, Ito Y, Eshima K, Ohkubo H, Kojo K, Inoue T, Raouf J, Jakobsson PJ, Uematsu S, Akira S, et al: Inhibition of microsomal prostaglandin E synthase-1 facilitates liver repair after hepatic injury in mice. J Hepatol. 69:110–120. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Jolicoeur C, Boutouil M, Drouin R, Paradis I, Lemay A and Akoum A: Increased expression of monocyte chemotactic protein-1 in the endometrium of women with endometriosis. Am J Pathol. 152:125–133. 1998.PubMed/NCBI

43 

Gou Y, Li X, Li P, Zhang H, Xu T, Wang H, Wang B, Ma X, Jiang X and Zhang Z: Estrogen receptor β upregulates CCL2 via NF-κB signaling in endometriotic stromal cells and recruits macrophages to promote the pathogenesis of endometriosis. Hum Reprod. 34:646–658. 2019. View Article : Google Scholar : PubMed/NCBI

44 

Minamino T, Ito Y, Ohkubo H, Hosono K, Suzuki T, Sato T, Ae T, Shibuya A, Sakagami H, Narumiya S, et al: Thromboxane A(2) receptor signaling promotes liver tissue repair after toxic injury through the enhancement of macrophage recruitment. Toxicol Appl Pharmacol. 259:104–114. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2023
Volume 28 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Furue A, Hattori K, Hosono K, Tanabe M, Sato E, Honda M, Sekiguchi K, Ito Y, Majima M, Narumiya S, Narumiya S, et al: Inhibition of TP signaling promotes endometriosis growth and neovascularization. Mol Med Rep 28: 192, 2023
APA
Furue, A., Hattori, K., Hosono, K., Tanabe, M., Sato, E., Honda, M. ... Amano, H. (2023). Inhibition of TP signaling promotes endometriosis growth and neovascularization. Molecular Medicine Reports, 28, 192. https://doi.org/10.3892/mmr.2023.13079
MLA
Furue, A., Hattori, K., Hosono, K., Tanabe, M., Sato, E., Honda, M., Sekiguchi, K., Ito, Y., Majima, M., Narumiya, S., Kato, K., Amano, H."Inhibition of TP signaling promotes endometriosis growth and neovascularization". Molecular Medicine Reports 28.4 (2023): 192.
Chicago
Furue, A., Hattori, K., Hosono, K., Tanabe, M., Sato, E., Honda, M., Sekiguchi, K., Ito, Y., Majima, M., Narumiya, S., Kato, K., Amano, H."Inhibition of TP signaling promotes endometriosis growth and neovascularization". Molecular Medicine Reports 28, no. 4 (2023): 192. https://doi.org/10.3892/mmr.2023.13079