International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Despite significant improvements in colorectal cancer (CRC) treatment, especially in countries with long-standing screening programs (1), CRC is still the third most frequently diagnosed oncologic disease, ranking second among causes of cancer-related mortality (2,3). A further rise in CRC incidence is predicted. The burden of CRC is attributed to increasing early-onset CRC, ageing of the population and a lifestyle shift toward a Western diet combined with low physical activity (3,4).
Interestingly, the age-standardized incidence of CRC is consistently higher in males compared to females, including CRC cases with early onset (5). CRC is more often diagnosed in the proximal colon in women, whereas distal anatomical CRC sites are detected more often in males (6–8). The most pronounced difference is observed in the incidence of rectal cancer, which is in 75% of cases diagnosed in males (2,3,9).
Among the possible reasons for this phenomenon are an unfavourable diet, obesity, alcohol consumption, smoking and low physical activity (10). These factors influence CRC progression differently depending on the sex of the patient and tumour localisation (11–13).
Although sex hormones have been implicated in the generation of differences in CRC prevalence between males and females, their precise role has not been completely elucidated (7,8,14). In particular, it is assumed that the most potent oestrogen, 17β-oestradiol (E2), influences CRC progression (8,15,16).
E2 signalling is mediated predominantly by two types of nuclear receptors, ESR1 and ESR2, and the G protein-coupled membrane receptor Gper1 (previous symbols GPER, GPR30) (15). Among the ligands of ESR1, ESR2 and Gper1 are, in addition to oestrogens, tamoxifen, raloxifene, bisphenols, dioxins, phthalates, quercetin, genistein, resveratrol and others (17,18). The ligands mentioned above can usually activate both nuclear and membrane-bound receptors, although their affinity can differ (19). Gper1 has a lower affinity for E2 than ESR1 and ESR2 (20). Direct binding of E2 to Gper1 was even questioned recently (21,22). However, there is substantial in vivo and in vitro evidence that Gper1 is involved in E2-mediated regulation (23). Recently, the binding of E2 to Gper1 was confirmed, and aldosterone was also proposed as a Gper1 agonist (24).
Nuclear E2 receptors exert most of their effects via transcriptional gene regulation. ESR1 and ESR2 activate thousands of genes overlapping by 30% via a specific DNA sequence called oestrogen response element (ERE) (25).
Compared to nuclear E2 receptors, Gper1 affects intracellular processes much faster thanks to the prompt activation of several intracellular signalling pathways. Signalling mediated by Gper1 includes an increase in cAMP synthesis and consequent activation of protein kinase A, Src-like nonreceptor tyrosine kinase (Src) and sphingosine kinase (SphK). Src and SphK can contribute to the activation of epidermal growth factor receptors (EGFR). Gper1 signalling can also lead to activation of protein kinase C and calcium mobilisation (17). Among Gper1-driven regulatory pathways also belongs Gper1/HIFα activation of NOTCH and consequent induction of VEGF transcription (26) and some others (18). Tissue-specific distribution and/or changes in the tissue-specific distribution of E2 receptors in disease can influence E2 signalling and facilitate its oncostatic or tumour suppressor effects (17,18,25).
Epidemiological studies of patients using hormone response therapy (HRT) implicated protective role of E2 in respect to CRC incidence. It was shown that oestrogen/progestin HRT is negatively associated with the risk of CRC (27). Similarly, the incidence of CRC in postmenopausal women exposed to E2 HRT was lower compared to women of the same age without HRT (28). Other studies later strengthened the evidence of E2′s protective role regarding CRC incidence (15,16,29,30). It is of interest that patients with high levels of endogenous circulating E2 (31) and those exposed to E2 therapy whilst diagnosed with CRC demonstrated a worse disease prognosis (32). On the other hand, Mori et al (33) did not confirm the association of high circulating E2 levels with increased incidence and/or prognosis of CRC in postmenopausal women.
Most of the beneficial effects of E2 for patients are attributed to ESR2 (15,34–37), which is far more abundant in the gastrointestinal tract compared to ESR1 (38–40). The beneficial E2 effect mediated via ESR2 receptors was proven with the use of several in vivo experimental models. Apcmin/+mice bearing a mutation in the Apc gene are prone to developing multiple intestinal tumours. Ovariectomy (OVX) significantly increased the number of adenomas detected in the gut compared to control, and this effect was reversed by E2 administration. E2 treatment was also accompanied by an increase in ESR2 expression in the intestine (41). ESR2 deficiency in Apcmin/+ mice was associated with a higher adenoma number compared to control (42). The protective role of E2 executed via ESR2 was also demonstrated in OVX mice where tumorigenesis was induced by azoxymethane (AOM) (43), by combined treatment of AOM and dextran sulphate sodium (44,45) and in immunodeficient mice implanted with SW480 cells overexpressing ESR2 (46).
Interestingly, the expression of Gper1 mRNA exceeds that of ESR2 in healthy gut tissue (40,47). Similarly, it is the predominant E2 receptor in several colorectal cell lines (48). Despite that, the role of the membrane receptor Gper1 in E2-mediated effects on cancer progression has not been completely elucidated.
The tumour suppressor as well as the oncogenic role of Gper1 has been demonstrated for several types of tissues. Perhaps the most promising results were achieved in melanoma treatment. However, in most cancer types, including CRC, the role of Gper1 remains inconclusive (17,49,50).
It was shown that G-1, an agonist of Gper1, inhibits the upregulation of JUN oncogene expression in HT29 cells (48). G-1 administered at a concentration of 1 µM significantly inhibited the proliferation of HCT-116 and SW480 cells, increased the number of cells in the G2/M phase and stimulated apoptosis. The decrease in cell viability induced by G-1 was prevented by the administration of the ROS scavenger NAC. Growth of tumour xenografts in nude mice was inhibited by G-1 administration (51).
On the other hand, Gper1 silencing prevented chromosomal instability induced by diethylstilbestrol and the occurrence of supernumerary centrosomes in HCT116 and control CCD 841 CoN cell lines. In these cell lines, Gper1 activation led to the presence of multipolar mitotic spindles, an increased number of cells with lagging chromosomes and aneuploidy. Interestingly, manipulation of Gper1 levels did not influence the rate of proliferation and cell cycle distribution under in vitro conditions (52). The oncogenic role of Gper1 was confirmed in HT-29 cells as Gper1 silencing prevented an E2-induced decrease in ATM. Under normoxic conditions, Gper1 activation caused down regulation of VEGF while the opposite effect was observed in a hypoxic microenvironment and prevented by Gper1 silencing. Under hypoxic conditions, Gper1 mediated an E2-induced increase in HT-29 and DLD1 cell migration (53). E2 and its agonist G-1, via Gper1, induced the expression of fatty acid synthase (FASN), which can promote CRC progression. FASN upregulation is executed by the epidermal growth factor receptor EGFR/ERK/AP1 pathway (54). Oncogenic properties of Gper1 were demonstrated in the CRC cell lines COLO205 and SW480 as the Gper1 antagonist G15 was able to reverse the effect of the E2-related endocrine disruptor nonylphenol, which induced proliferation, the expression of cyclin D1, c-myc and ERK1/2. Administration of G15 also inhibited the growth of colon carcinoma xenografts in mice (55). Disagreement in the results of performed studies can be partly related to different concentrations of G-1 used in experiments. In addition to experimental designs, the biological context seems to be of special importance in effects mediated by Gper1 (17,34,48,52,53).
In human studies, the results are also inconclusive. Up- as well as down-regulated Gper1 expression in CRC tissue was indicated (17), e.g. a strong trend toward increased Gper1 levels in CRC cancer tissue compared to adjacent tissue was reported by Gilligan et al (56) while down-regulated expression of Gper1 was reported by Liu et al (51). High expression of Gper1 was associated with better survival compared to low expression (51); by contrast, low expression of Gper1 was associated with better survival compared to high expression (56). High expression of Gper1 was also associated with worse survival by Bustos et al (53) and Abancens et al (48) but only in females in stages 3–4 of CRC. Worse survival of CRC patients exhibiting high expression of Gper1 is indicated by the Human Protein Atlas (47).
As there is evidence suggesting that manipulation of E2 signalling could be effective in clinical use, several agonists and antagonists of nuclear and Gper1 receptors have been developed, and some of them have been clinically tested (18,25,57). The most promising results were obtained in the clinical study NCT04130516 evaluating the effects of Gper1 agonist LNS8801 that proved beneficial effects of Gper1 signalling in patients diagnosed with cutaneous (58) and uveal (59) melanoma. However, it was implied that G-1 can cause effects independent of Gper1 signalling (60–65). Interestingly, several clinically approved antagonists of nuclear E2 receptors also act as Gper1 agonists (18).
CRC is among the most frequently diagnosed oncologic diseases worldwide. To facilitate novel CRC treatment strategies, we focused on Gper1 functioning in CRC tumours to define the conditions under which Gper1 upregulation could be beneficial for patients.
Despite ongoing clinical trials aimed at the manipulation of Gper1 signalling in solid cancers, the involvement of Gper1 in the generation of sex-dependent CRC incidence is largely unknown. Similarly, the biological context that determines the effect of Gper1 has not been described sufficiently. Therefore, the present study is aimed to analyse the abundance of Gper1 in healthy and tumour tissues of males and females diagnosed with CRC according to disease stage and survival. Mutation in p53 has been identified in more than 50% of CRC patients and at a higher frequency in males compared to females (66). Therefore, using the CRC cell lines LoVo and DLD1, which differ in p53 functionality (67), we tested the hypothesis that the effects of Gper1 depend on the presence of wild-type p53.
Tumour and adjacent tissues were obtained in cooperation with the First Surgery Department, University Hospital, Comenius University, Bratislava. The study included patients who had to undergo surgery for CRC treatment (for details see Table SI) and agreed to sign an informed consent. Patients were not subjected to any CRC treatment before surgery and during hospitalization were exposed to a standard hospital practice with lights on from 6:00 a.m. to 9:00 p.m. Surgery was performed in the morning hours (before noon). Adjacent tissues were collected ≥10 cm proximally and ≥2 cm distally from the tumour. After tissue excision, samples were promptly frozen in liquid nitrogen and stored at −70°C until analysis. The experimental protocol was approved by the Ethics Committee of Comenius University in Bratislava (ECH 19001).
To extract mRNA from tissues and cells, RNAzol RT (Molecular Research Centre) was used as described previously (40). One microgram of RNA template from tissues and 0.02 µg from cells was used to synthesize cDNA with the ImProm-II Reverse Transcription System and random hexamers (Promega), according to the manufacturer's instructions.
To analyse gene expression, the QuantiTect SYBR Green kit (Qiagen, Germany) was used. Real-time PCR was performed with the StepOnePlus™ Real-Time PCR System (Applied Biosystems). The PCR conditions were activation of hot start polymerase at 95°C for 15 min followed by 40 cycles at 94°C for 15 sec, 53°C for 30 sec and 72°C for 30 sec. As a last step of PCR, samples were subjected to melting curve analysis. The sequences of primers for the amplification of particular genes are provided in Table SII. The expression of nuclear u6 mRNA was used for normalisation.
The human colorectal carcinoma cell lines DLD1 and LoVo obtained from the American Type Culture Collection (ATCC) were used to reveal how gper1 silencing influences cell migration and metabolism. DLD1 cells were cultured in RPMI 1640 GlutaMax medium (Gibco; Thermo Fisher Scientific, Inc.), and LoVo cells were incubated in F-12K medium (Bioconcept). Both cell lines were supplemented with 2% or 10% foetal bovine serum (FBS, Biosera) depending on the experiment; 2% FBS supplementation was used for the scratch assay while 10% FBS was used for the MTS test. The culture medium also contained penicillin (50 U/ml) (Gibco), streptomycin (50 µl/ml) (Gibco, USA) and ampicillin (50 µg/ml) (Oasis-lab). All experiments were performed in a biological Celculture® Incubator CCL-050B-8 (Esco Medical) with a humidified atmosphere containing 5% CO2 at 37°C. The cells were cultured in 96-well plates coated with 1% sterile gelatine.
To test the effect of gper1 silencing, DharmaFECT 1 reagent was used to transfect cells with siGENOME Human Gper1siRNA-SMART pool or siGENOME non-targeting siRNA Pool #2 (Horizon) at a concentration of 100 nM.
The effect of siGper1 on wound healing was determined by scratch assay performed when the cell culture reached a confluence of 80–90% with the use of a 10-ul sterile tip. Images were taken immediately after scratch and later as indicated in the figure legends with the use of an inverted fluorescence microscope NIB-100F (Nanjing Jiangnan Novel Optics Co., Ltd.) and BEL Capture 3.2 software (BEL Engineering s.r.l.). Wound closure was evaluated with ImageJ software (68).
The rate of metabolism was measured by MTS test according to the manufacturer's instructions (CellTiter 96 AQueous Cell Proliferation Assay, Promega) employing a modified tetrazolium compound to produce water-soluble formazan. The absorbance was measured at 490 nm using a UV spectrophotometer (Epoch, Agilent Technologies, Inc.).
To evaluate Gper1 mRNA expression in human samples, the cohort was split into three groups according to TNM classification. Group 1 included distant metastasis-free patients without nodal involvement (T1-4N0M0), group 2 consisted of patients with nodal involvement and without distant metastases (T3-4N1-2M0) and the 3rd group was composed of patients with distant metastases (T3-4N0-2M1). Gene expression between three groups was compared by ANOVA followed by Tukey's post hoc test.
To compare E2 receptor expression between two groups, a Student's paired t-test was used. ANOVA followed by Tukey's post hoc multiple comparisons test was used to compare the expression of three types of oestrogen receptors (ESR1, ESR2 and Gper1) in human tissues and cells.
To analyse Gper1 mRNA expression with respect to overall survival, a Kaplan-Meier survival curve and a log-rank test were performed. Values were split according to the median; high expression > median, low expression ≤ median. The starting point for the log-rank test was the day of the surgery. The association of Gper1 and VEGFA mRNA expression was analysed by correlation analysis.
Data are provided as mean ± standard error of the mean (SEM) in relative units (r.u.). The threshold for significance for all statistical tests was set at P<0.05. Statistical analyses were performed using GraphPad Prism 6 (GraphPad Software, Inc.).
E2 receptors show robust differences in expression between colorectal tumours and adjacent tissues. In tumour tissue, the expression of gper1 was higher compared to adjacent tissue, whereas the opposite pattern was observed in levels of nuclear E2 receptors. The expression of esr1 and esr2 was significantly lower in cancer tissue compared to adjacent tissues (Fig. 1A). The expression of gper1 was higher in cancer tissue compared to adjacent distal and proximal tissues (Fig. 1B). Comparison of the absolute level of expression based on the threshold value of the PCR revealed that gper1 is far most abundant in cancer as well as in the adjacent tissues compared to esr1 and esr2 mRNA levels (Fig. 1B).
The expression of gper1 was higher in cancer tissue compared to adjacent tissue in males, but in females this pattern was observed only as a non-significant trend (Fig. 2). The expression of esr1 was higher in the adjacent compared to cancer tissue and this difference, again, achieved a level of significance only in males (Fig. 2). The most robust difference in expression between cancer and adjacent tissue was observed in the expression of esr2. Unlike other receptors, this pattern was present in both sexes (Fig. 2).
In the whole cohort an increase in gper1 expression in cancer tissue compared to adjacent tissue was observed in patients without nodal involvement and distal metastases (Fig. 3A). However, when the cohort was split into male and female patients, this difference was observed only in males (Fig. 3B and C). gper1 expression did not differ between cancer and adjacent tissues in more advanced stages of disease (Fig. 3).
When the cohort was split according to nodal involvement only (without considering TNM staging as a whole) the sex-dependent difference in gper1 expression became even more pronounced, and there was a highly significant increase in gper1 expression in cancer tissue compared to adjacent tissue in males without nodal involvement (Fig. 4A). Similarly, male patients without distant metastases displayed a pronounced increase in gper1 expression in cancer tissue compared to adjacent tissue that was not observed in females (Fig. 4B).
In accordance with the sex-dependent expression of gper1 in CRC patients, the association of overall survival and gper1 expression exerted a sex-dependent pattern. In the whole cohort, better survival was correlated with low gper1 expression in patients without nodal involvement (Fig. 5A) but not in patients in higher stages of disease (Fig. 5B). When the cohort was split according to sex, a log-rank test revealed that gper1 association with survival was generated by the male subcohort (Fig. 5C). In females the correlation between survival and gper1 expression did not reach significance (Fig. 5D).
In human samples from our cohort, the expression of gper1 significantly correlated with the mRNA expression of VEGFA in cancer tissue. This relationship was not observed in proximal and distal adjacent tissues (Fig. S1).
Transfection of siRNA targeting gper1 expression successfully inhibited Gper1 mRNA expression in DLD1 as well as LoVo cells compared to control (Fig. S2A and B, respectively).
The distribution of E2 receptors in CRC cell lines LoVo and DLD1 resembled that of human cancer tissue. The expression of the membrane gper1 receptor was much higher compared to mRNA levels of nuclear E2 receptors, and the expression of ESR1 mRNA was nearly undetectable (Fig. 6).
Silencing of gper1 expression significantly stimulated metabolism in LoVo cells in a time-dependent manner that implicates the tumour-suppressor capacity of Gper1. In DLD1 cells we did not observe this effect (Fig. 7).
Evaluation by the scratch test demonstrated a time-dependent decrease in wound closure in DLD1 cells with silenced Gper1 expression compared to control. Whereas 24h after Gper1 silencing there was no effect on the rate of wound closure, 48 h after transfection the width of the closed area was different between control and transfected cells with P=0.057 (Fig. 8A). Inhibition of wound healing by Gper1 silencing implicates the oncogenic potential of Gper1 in DLD1 cells.
Unlike in DLD1 cells, the administration of siRNA interfering with Gper1 expression did not influence the rate of wound closure in LoVo cells (Fig. 8B).
The expression of p53 mRNA did not differ significantly between DLD1 and LoVo cells. However, as DLD1 cells generate a mutated form of the p53 protein, expression of p53-inducible gene p21 was significantly lover in DLD1 cells compared to LoVo cells (Fig. S3).
The expression of gper1 and tp53 showed a significant correlation in males (A) but not in females (B) undergoing surgery for CRC treatment (Fig. S4) (TCGA, Colorectal Adenocarcinoma, Nature 2012).
The relative ratio of E2 receptors in CRC tissue undergoes remodelling. While the expression of nuclear E2 receptors decreases in cancer tissue compared to adjacent tissue, the opposite pattern is observed in Gper1 mRNA expression. Therefore, Gper1 is by far the most abundant E2 receptor in the CRC tumour followed by the ESR2 and very low ESR1 mRNA expression. The increase in gper1 expression in tumours is most pronounced in males in the lower stages of disease. A higher expression of Gper1 mRNA was associated with worse survival in the whole cohort and in males without metastases in nodes. This dependency was not detected in males with nodal involvement and females. We suppose that sex-dependent differences in E2 receptor expression can contribute to sex-dependent features of CRC progression.
In accordance with our data, Gilligan et al (56) showed an increase in Gper1 expression in CRC tissue compared to adjacent tissue. Several factors can induce an increase in Gper1 mRNA levels in cancer tissue. Firstly, Gper1 belongs to HIF-1 targeted genes that are expressed in response to hypoxia (69). The stimulatory effect of hypoxia on Gper1 mRNA expression was also demonstrated in the colon cancer cell line HT-29 and the rectal cancer cell line C80 (53). The oncogenic role of Gper1 was implicated as it was demonstrated that Gper1 cooperated with HIF-1α in the activation of VEGF expression in hypoxia (26,70). In line with this finding, we detected a positive correlation between Gper1 and VEGFA mRNA expression in cancer but not in the adjacent tissues.
We observed a decrease in ESR2 mRNA expression in CRC tumours compared to proximal and distal parts of the gut, which is consistent with previous studies (71–77) that reported lower ESR2 expression in CRC tissue compared to adjacent tissues at the protein and mRNA levels.
According to our results, the expression of ESR1 mRNA is lower in cancer tissue compared to adjacent tissues, and this difference is more pronounced in males than in females. Previously, a decrease was observed in ESR1 expression in colorectal cancer tissue, which was attributed to CpG island methylation (78). No significant differences between cancer and adjacent tissue have also been reported (38). According to Jiang et al (79), the expression of the dominant ESR1 isoform does not differ in its mRNA levels between tumour and matched normal tissues. However, the ER-α46 isoform that shares most of its sequence with the dominant isoform was down-regulated in cancer tissue compared to adjacent tissue. On the other hand, an increase in esr1 mRNA in cancer compared to adjacent healthy tissue has been reported lately (80). Although data referring to ESR1 expression in CRC seem to be inconclusive, there is a consensus on the decrease in nuclear ESR2 and increase in Gper1 expression in cancer compared to adjacent tissue.
The expression of all three types of receptors in one set of samples has rarely been studied. There is a strong evidence that the expression of ESR2 in the gut is more abundant compared to that of ESR1 (71,76,81). The assumption that ESR2 is the predominant form of the E2 receptor in the gut can, to some extent, be caused by later identification of the Gper1 receptor compared to nuclear E2 receptors (82). Previously, it has been shown that the expression of Gper1 mRNA is more abundant compared to that of both ESR1 and ESR2 in the rat colon (40). According to datasets available from the Human Protein Atlas, the expression of E2 receptors is Gper1>ESR2>>ESR1 (47), which is in line with Harvey and Harvey (83).
The density of E2 receptors changes in CRC tumour tissue. In CRC the abundance of E2 receptors shows the pattern Gper1>>ESR2>ESR1. The order of E2 receptor density in the healthy colon is similar, but there is an abrupt decrease in ESR2 and an increase in Gper1 expression in the tumour compared to the healthy gut. Changes in the abundance of E2 receptors in CRC are more pronounced in males than in females.
As CRC occurs to a lesser extent in females compared to males (83), and sex-dependent differences in nuclear E2 receptor expression have been revealed (72,74–76,84), female sex hormones were suggested to be involved in the regulation of CRC progression (85). Beneficial effects of E2 mediated via ESR2 receptors have been convincingly demonstrated (76,86); however, contradictory reports are available concerning the role of Gper1 in CRC progression (34,53,56,83). Similarly, little is known about sex-related differences in Gper1 expression, although they have been implied (70,83). According to our results, there are more pronounced differences in Gper1 expression in cancer tissue compared to adjacent tissue in males than in females. This difference is noticeable mainly in males without nodal metastases.
In our cohort poor survival correlated with high Gper1 mRNA expression more significantly in males without nodal involvement than in males in higher stages of disease or females. These results are in accordance with Gilligan et al (56), who reported an association of worse survival in patients with high Gper1 expression. Information about the correlation in males and females separately was not provided. Bustos et al (53) reported sex-dependent differences in relapse-free survival and Gper1 expression; however, as we do not have data allowing this type of analysis, a comparison was not possible. Our results are in accordance with the Human Protein Atlas (47), according to which there is a stronger association between Gper1 and survival in males compared to females and worse survival in patients with high Gper1 expression. The association reaches significance only in males in stage I–II and not in patients with higher stages of disease. Interestingly, when the survival of males and females together are correlated with Gper1 expression, the association does not reach significance, which also implicates a sex-specific dichotomy in regulation.
There are reports implicating Gper1 as a tumour suppressor in pancreatic cancer, melanoma and adrenocortical cancer and as a tumour promoter in glioblastoma and endometrial and ovarian cancers, whereas in the case of lung, prostate, breast and colorectal cancers, information about the regulatory impact of Gper1 is inconclusive (17,18,34,87,88). Gper1-mediated effects are dependent on the biological context. Our results imply that intracellular conditions are vital for the interpretation of Gper1 signalling, even at the level of one type of solid cancer.
Gper1 is known to induce signalisation mediated via the EGFR receptor with tyrosine kinase activity (17,89,90). A deregulated EGFR pathway has been associated with the progression of many types of cancer, including CRC (91,92). Therefore, several tyrosine kinase inhibitors and EFGR antibodies have been developed and introduced into clinical practice (92). Inhibition of EGFR signalling was shown to benefit patients diagnosed with CRC, and the EFGR monoclonal antibodies Cetuximab and Panitumumab are now routinely used to treat this type of cancer (93). In vitro studies elucidated the molecular mechanisms of cancer inhibition caused by EFGR repression in several cancer cell lines, including DLD1 (94–97). Gper1 signalling mediated via the Gs-coupled cAMP/PKA/CREB pathway has also been shown to promote cell proliferation (98). Therefore, as Gper1 is known to induce both EGFR- and cAMP-mediated regulation and is reciprocally involved in VEGFA release, presumably, an oncogenic role of Gper1 might be expected in CRC.
However, signalling mediated by Gper1 is highly complex. The same group that discovered the connection between Gper1 and EGFR two years later revealed that Gper1 inhibits EGFR signalling via the cAMP/PKA pathway by Raf-1 inactivation (89). To further elucidate the ambiguous effects of Gper1 on CRC progression, we investigated the effect of Gper1 silencing in CRC cell lines LoVo and DLD1.
According to our results, the distribution of E2 receptors in the CRC cell lines DLD1 and LoVo resembles that observed in human tissues: the highest mRNA expression is that of gper1, and the expression of ESR1 is nearly undetectable. Gper1 silencing in the DLD1 cell line, which expresses mutated tp53, inhibited the wound closure, implicating the oncogenic potential of Gper1. By contrast, the decrease in Gper1 availability caused an increase in the metabolic rate in LoVo cells that preserved p53 functionality (99).
The p53 protein, encoded by the gene tp53, is a well-known tumour suppressor that inhibits cell cycle progression and initiates DNA repair and/or apoptosis and autophagy, in response to DNA damage, hypoxia, nutrition deficiency, oxidative stress and some hormones. Cell cycle inhibition is executed by induction of p21 expression. p21 inhibits the activity of several types of cyclin/CDK complexes that release repression of retinoblastoma protein (RB). As cyclin/CDK complexes are inactivated by p21 and cannot phosphorylate RB, hypophosphorylated RB inhibits E2F-induced transcription, which is necessary for cell proliferation (100,101).
The p53 protein is frequently mutated in many types of cancer, including CRC. According to TCGA, more than 50% of CRC patients carry a mutated form of tp53. The presence of a mutated form of p53 is accompanied by worse survival than that of wild-type tp53. Interestingly, tp53 mutations are more frequent in males compared to females, which may also contribute to the higher CRC incidence in males compared to females (66).
We hypothesize that the signalling pathway downstream of p53 influences the outcome of Gper1 signalling. Previously, it was shown that Gper1 expression in the triple-negative breast cancer cell lines MDA-MB-231 and MDA-MB-468, which express a mutated form of tp53, had been induced by γ-radiation, whereas in MCF-7 cells expressing wild-type tp53, the opposite pattern was observed. Therefore, a tumour suppressor role dependent on p53 was attributed to Gper1 (102). Our results agree with this statement as Gper1 was associated with oncostatic functions in LoVo but not in DLD1 cell lines. Previously, the oncogenic potential of Gper1 has been described in the CRC cell lines HT-29, DLD1, COLO205 and SW480 (53,55) carrying mutations in the tp53 gene (66,103–106).
Although there are reports implicating a functional relationship between p53 and oestradiol, the exact mechanism has either not been completely elucidated or, more likely, comprises several ways by which E2 and p53 signalling interfere (107). These interferences can differ in a tissue-dependent manner, e.g. E2 administration decreases p53 expression in lung cancer cells by induction of methyltransferase 1 expression, which increases methylation of the tp53 promoter (108). On the other hand, a protective role of E2 in non-malignant colonocytes executed via p53-mediated regulation has been implicated (109).
The effects of E2 on p53 expression have been previously investigated mainly with respect to the effects of ERα on breast cancer progression. ERα interacts with tp53 and influences its expression. Most of studies report an increase in tp53 expression in response to ERα binding (107,110–113). On the other hand, there are also results implicating ERα-induced suppression of p53-regulated gene expression (114,115). However, in DLD1 cells, ESR1 expression is nearly undetectable; therefore, we do not suppose that a substantial increase in p53 expression can be induced via ERα. The results are inconclusive with respect to ERβ signalling and p53 expression. Although induction of p53 activity in response to ERβ activation has been reported (107,113), a stimulatory effect of ERβ interacting with the tp53 promoter on tp53 expression has not been detected (111).
In silico analysis showed a positive correlation between Gper1 and tp53 expression in CRC tissues of males (TCGA) (116). The E2 receptor ligands bisphenol A (117) and G-1 (102,118,119) have also been shown to induce tp53 expression. Silencing of Gper1 caused a decrease in tp53 expression in uveal melanoma cell lines (88). The exact mechanism of how Gper1 executes its effect on p53 transcription is unknown. A cAMP response element (CREB) was identified in the human tp53 gene (120,121) and CREB binding protein plays a key role in p53 activation (122). However, whether Gper1 executes its stimulatory effect on tp53 expression via this region remains to be elucidated.
To conclude, the expression of E2 receptors in healthy tissue follows the descending order Gper1>ESR2>>ESR1 with nearly undetectable expression of ESR1 mRNA. In CRC patients the ratio of receptors changes more in males than in females; the expression of Gper1 mRNA increases while the expression of ESR2 and ESR1 decreases, resulting in the descending order Gper1>>ESR2>ESR1. Therefore, under conditions of cancer progression, most E2 signalling is mediated via the membrane E2 receptor Gper1. High expression of Gper1 is associated with worse survival in males without nodal involvement in comparison with other subcohorts (Fig. 9).
The relative expression of E2 receptors in DLD1 and LoVo cells is similar to that observed in human CRC tumours. In LoVo cells with wild-type tp53, a tumour suppressor effect of Gper1 was observed. This effect was not detected in DLD1 cells with the mutated form of tp53. We suppose that Gper1 activates the EFGR/RAS oncogenic pathway as well as the p53 pathway. In case the p53 pathway is not functional, the oncogenic potential of Gper1 overwhelms its tumour suppressor effects. As the frequency of tp53 mutations as well as changes in Gper1 expression are more robust in males compared to females, we suppose that these effects can contribute to sex-dependent differences in CRC incidence.
The authors would like to thank Dr. Soňa Olejarova (Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovak Republic) for help with laboratory analyses.
The research was supported by projects APVV-16-0209 and APVV-20-0241 provided by The Slovak Research and Development Agency and project VEGA 1/0455/23 provided by grant agency of the Ministry of Education, Science, Research and Sport of the Slovak Republic.
The datasets are available from the corresponding author on reasonable request.
IH and RR designed and administered the human study. IH and RR confirm the authenticity of all the raw data. RR obtained samples and organized their transport to the laboratory. IH performed analysis of human samples. IH and DV performed cell culture experiments. IH performed in silico analysis, interpreted results, wrote the manuscript and prepared the figures. IH, RR and DV reviewed and revised the manuscript for the scientific content. All authors have read and approved the final manuscript.
The design of the study describing the sampling of human tissues was approved by the Ethics Committee of the Comenius University in Bratislava (approval no. ECH 19001). All patients included in the study agreed to sign an informed consent. The manuscript does not contain experiments using animals or embryonic cells.
Not applicable.
The authors declare that they have no competing interests.
Professor Iveta Herichová, ORCID: 0000-0002-0475-0461.
|
Cardoso R, Guo F, Heisser T, Hackl M, Ihle P, De Schutter H, Van Damme N, Valerianova Z, Atanasov T, Májek O, et al: Colorectal cancer incidence, mortality, and stage distribution in European countries in the colorectal cancer screening era: An international population-based study. Lancet Oncol. 22:1002–1013. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2021. CA Cancer J Clin. 71:7–33. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Xi Y and Xu P: Global colorectal cancer burden in 2020 and projections to 2040. Transl Oncol. 14:1011742021. View Article : Google Scholar : PubMed/NCBI | |
|
Murphy N, Ward HA, Jenab M, Rothwell JA, Boutron-Ruault MC, Carbonnel F, Kvaskoff M, Kaaks R, Kühn T, Boeing H, et al: Heterogeneity of colorectal cancer risk factors by anatomical subsite in 10 european countries: Amultinational cohort study. Clin Gastroenterol Hepatol. 17:1323–1331.e6. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Gausman V, Dornblaser D, Anand S, Hayes RB, O'Connell K, Du M and Liang PS: Risk factors associated with early-onset colorectal cancer. Clin Gastroenterol Hepatol. 18:2752–2759.e2. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Jacobs ET, Thompson PA and Martínez ME: Diet, gender, and colorectal neoplasia. J Clin Gastroenterol. 41:731–746. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Quirt JS, Nanji S, Wei X, Flemming JA and Booth CM: Is there a sex effect in colon cancer? Disease characteristics, management, and outcomes in routine clinical practice. Curr Oncol. 24:e15–e23. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Lopes-Ramos CM, Quackenbush J and DeMeo DL: Genome-wide sex and gender differences in cancer. Front Oncol. 10:5977882020. View Article : Google Scholar : PubMed/NCBI | |
|
Høydahl Ø, Edna TH, Xanthoulis A, Lydersen S and Endreseth BH: Long-term trends in colorectal cancer: Incidence, localization, and presentation. BMC Cancer. 20:10772020. View Article : Google Scholar : PubMed/NCBI | |
|
Johnson CM, Wei C, Ensor JE, Smolenski DJ, Amos CI, Levin B and Berry DA: Meta-analyses of colorectal cancer risk factors. Cancer Causes Control. 24:1207–1222. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Murphy N, Ward HA, Jenab M, Rothwell JA, Boutron-Ruault MC, Carbonnel F, Kvaskoff M, Kaaks R, Kühn T, Boeing H, et al: Heterogeneity of colorectal cancer risk factors by anatomical subsite in 10 European countries: A multinational cohort study. Clin Gastroenterol Hepatol. 17:1323–1331.e6. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Murphy N, Moreno V, Hughes DJ, Vodicka L, Vodicka P, Aglago EK, Gunter MJ and Jenab M: Lifestyle and dietary environmental factors in colorectal cancer susceptibility. Mol Aspects Med. 69:2–9. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wele P, Wu X and Shi H: Sex-dependent differences in colorectal cancer: With a focus on obesity. Cells. 11:36882022. View Article : Google Scholar : PubMed/NCBI | |
|
Barzi A, Lenz AM, Labonte MJ and Lenz HJ: Molecular pathways: Estrogen pathway in colorectal cancer. Clin Cancer Res. 19:5842–5848. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Nie X, Xie R and Tuo B: Effects of estrogen on the gastrointestinal tract. Dig Dis Sci. 63:583–596. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Labadie JD, Harrison TA, Banbury B, Amtay EL, Bernd S, Brenner H, Buchanan DD, Campbell PT, Cao Y, Chan AT, et al: Postmenopausal hormone therapy and colorectal cancer risk by molecularly defined subtypes and tumor location. JNCI Cancer Spectr. 4:pkaa0422020. View Article : Google Scholar : PubMed/NCBI | |
|
Qiu YA, Xiong J and Yu T: Role of G Protein-coupled estrogen receptor in digestive system carcinomas: A minireview. Onco Targets Ther. 14:2611–2622. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Prossnitz ER and Barton M: The G protein-coupled oestrogen receptor GPER in health and disease: An update. Nat Rev Endocrinol. 19:407–424. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Heldring N, Pike A, Andersson S, Matthews J, Cheng G, Hartman J, Tujague M, Ström A, Treuter E, Warner M and Gustafsson JA: Estrogen receptors: How do they signal and what are their targets. Physiol Rev. 87:905–931. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Fuentes N and Silveyra P: Estrogen receptor signaling mechanisms. Adv Protein Chem Struct Biol. 116:135–170. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Otto C, Rohde-Schulz B, Schwarz G, Fuchs I, Klewer M, Brittain D, Langer G, Bader B, Prelle K, Nubbemeyer R and Fritzemeier KH: G protein-coupled receptor 30 localizes to the endoplasmic reticulum and is not activated by estradiol. Endocrinology. 149:4846–4856. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Ahmadian Elmi M, Motamed N and Picard D: Proteomic analyses of the G Protein-coupled estrogen receptor GPER1 reveal constitutive links to endoplasmic reticulum, glycosylation, trafficking, and calcium signaling. Cells. 12:25712023. View Article : Google Scholar : PubMed/NCBI | |
|
Mauvais-Jarvis F, Lange CA and Levin ER: Membrane-initiated estrogen, androgen, and progesterone receptor signaling in health and disease. Endocr Rev. 43:720–742. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ding Q, Chorazyczewski J, Gros R, Motulsky HJ, Limbird LE and Feldman RD: Correlation of functional and radioligand binding characteristics of GPER ligands confirming aldosterone as a GPER agonist. Pharmacol Res Perspect. 10:e009952022. View Article : Google Scholar : PubMed/NCBI | |
|
Leitman DC, Paruthiyil S, Vivar OI, Saunier EF, Herber CB, Cohen I, Tagliaferri M and Speed TP: Regulation of specific target genes and biological responses by estrogen receptor subtype agonists. Curr Opin Pharmacol. 10:629–636. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
De Francesco EM, Lappano R, Santolla MF, Marsico S, Caruso A and Maggiolini M: HIF-1α/GPER signaling mediates the expression of VEGF induced by hypoxia in breast cancer associated fibroblasts (CAFs). Breast Cancer Res. 15:R642013. View Article : Google Scholar : PubMed/NCBI | |
|
Rennert G, Rennert HS, Pinchev M, Lavie O and Gruber SB: Use of hormone replacement therapy and the risk of colorectal cancer. J Clin Oncol. 27:4542–4547. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Johnson JR, Lacey JV Jr, Lazovich D, Geller MA, Schairer C, Schatzkin A and Flood A: Menopausal hormone therapy and risk of colorectal cancer. Cancer Epidemiol Biomarkers Prev. 18:196–203. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Symer MM, Wong NZ, Abelson JS, Milsom JW and Yeo HL: Hormone replacement therapy and colorectal cancer incidence and mortality in the prostate, lung, colorectal, and ovarian cancer screening trial. Clin Colorectal Cancer. 17:e281–e288. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Jang YC, Huang HL and Leung CY: Association of hormone replacement therapy with mortality in colorectal cancer survivor: A systematic review and meta-analysis. BMC Cancer. 19:11992019. View Article : Google Scholar : PubMed/NCBI | |
|
Hang D, He X, Kværner AS, Chan AT, Wu K, Ogino S, Hu Z, Shen H, Giovannucci EL and Song M: Plasma sex hormones and risk of conventional and serrated precursors of colorectal cancer in postmenopausal women. BMC Med. 19:182021. View Article : Google Scholar : PubMed/NCBI | |
|
Foster PA: Oestrogen and colorectal cancer: Mechanisms and controversies. Int J Colorectal Dis. 28:737–749. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Mori N, Keski-Rahkonen P, Gicquiau A, Rinaldi S, Dimou N, Harlid S, Harbs J, Van Guelpen B, Aune D, Cross AJ, et al: Endogenous circulating sex hormone concentrations and colon cancer risk in postmenopausal women: A prospective study and meta-analysis. JNCI Cancer Spectr. 5:pkab0842021. View Article : Google Scholar : PubMed/NCBI | |
|
Das PK, Saha J, Pillai S, Lam AK, Gopalan V and Islam F: Implications of estrogen and its receptors in colorectal carcinoma. Cancer Med. 12:4367–4379. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Mal R, Magner A, David J, Datta J, Vallabhaneni M, Kassem M, Manouchehri J, Willingham N, Stover D, Vandeusen J, et al: Estrogen receptor beta (ERβ): A ligand activated tumor suppressor. Front Oncol. 10:5873862020. View Article : Google Scholar : PubMed/NCBI | |
|
Mahbub AA, Aslam A, Elzubier ME, El-Boshy M, Abdelghany AH, Ahmad J, Idris S, Almaimani R, Alsaegh A, El-Readi MZ, et al: Enhanced anti-cancer effects of oestrogen and progesterone co-therapy against colorectal cancer in males. Front Endocrinol (Lausanne). 13:9418342022. View Article : Google Scholar : PubMed/NCBI | |
|
Refaat B, Aslam A, Idris S, Almalki AH, Alkhaldi MY, Asiri HA, Almaimani RA, Mujalli A, Minshawi F, Alamri SA, et al: Profiling estrogen, progesterone, and androgen receptors in colorectal cancer in relation to gender, menopausal status, clinical stage, and tumour sidedness. Front Endocrinol (Lausanne). 14:11872592023. View Article : Google Scholar : PubMed/NCBI | |
|
Campbell-Thompson M, Lynch IJ and Bhardwaj B: Expression of estrogen receptor (ER) subtypes and ERbeta isoforms in colon cancer. Cancer Res. 61:632–640. 2001.PubMed/NCBI | |
|
Maingi JW, Tang S, Liu S, Ngenya W and Bao E: Targeting estrogen receptors in colorectal cancer. Mol Biol Rep. 47:4087–4091. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Herichová I, Jendrisková S, Pidíková P, Kršková L, Olexová L, Morová M, Stebelová K and Štefánik P: Effect of 17β-estradiol on the daily pattern of ACE2, ADAM17, TMPRSS2 and estradiol receptor transcription in the lungs and colon of male rats. PLoS One. 17:e02706092022. View Article : Google Scholar : PubMed/NCBI | |
|
Weyant MJ, Carothers AM, Mahmoud NN, Bradlow HL, Remotti H, Bilinski RT and Bertagnolli MM: Reciprocal expression of ERalpha and ERbeta is associated with estrogen-mediated modulation of intestinal tumorigenesis. Cancer Res. 61:2547–2551. 2001.PubMed/NCBI | |
|
Giroux V, Lemay F, Bernatchez G, Robitaille Y and Carrier JC: Estrogen receptor beta deficiency enhances small intestinal tumorigenesis in ApcMin/+ mice. Int J Cancer. 123:303–311. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Weige CC, Allred KF and Allred CD: Estradiol alters cell growth in nonmalignant colonocytes and reduces the formation of preneoplastic lesions in the colon. Cancer Res. 69:9118–9124. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Song CH, Kim N, Lee SM, Nam RH, Choi SI, Kang SR, Shin E, Lee DH, Lee HN and Surh YJ: Effects of 17β-estradiol on colorectal cancer development after azoxymethane/dextran sulfate sodium treatment of ovariectomized mice. Biochem Pharmacol. 164:139–151. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Son HJ, Sohn SH, Kim N, Lee HN, Lee SM, Nam RH, Park JH, Song CH, Shin E, Na HY, et al: Effect of estradiol in an Azoxymethane/Dextran sulfate Sodium-treated mouse model of colorectal cancer: Implication for sex difference in colorectal cancer development. Cancer Res Treat. 51:632–648. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Hartman J, Edvardsson K, Lindberg K, Zhao C, Williams C, Ström A and Gustafsson JA: Tumor repressive functions of estrogen receptor beta in SW480 colon cancer cells. Cancer Res. 69:6100–6106. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson Å, Kampf C, Sjöstedt E, Asplund A, et al: Proteomics. Tissue-based map of the human proteome. Science. 347:12604192015. View Article : Google Scholar : PubMed/NCBI | |
|
Abancens M, Harvey BJ and McBryan J: GPER agonist G1 prevents Wnt-induced JUN upregulation in HT29 colorectal cancer cells. Int J Mol Sci. 23:125812022. View Article : Google Scholar : PubMed/NCBI | |
|
Muller C, Chaney MF, Cohen JV, Garyantes T, Lin JJ, LoRusso P, Mita AC, Mita MM, Natale C, Orloff MM, et al: Phase 1b study of the novel first-in-class G protein-coupled estrogen receptor (GPER) agonist, LNS8801, in combination with pembrolizumab in patients with immune checkpoint inhibitor (ICI)-relapsed and refractory solid malignancies and dose escalation update. J Clinical Oncol. 40 (16_suppl):S2574. 2022. View Article : Google Scholar | |
|
Hall KA and Filardo EJ: The G Protein-coupled estrogen receptor (GPER): A critical therapeutic target for cancer. Cells. 12:24602023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Q, Chen Z, Jiang G, Zhou Y, Yang X, Huang H, Liu H, Du J and Wang H: Epigenetic down regulation of G protein-coupled estrogen receptor (GPER) functions as a tumor suppressor in colorectal cancer. Mol Cancer. 16:872017. View Article : Google Scholar : PubMed/NCBI | |
|
Bühler M, Fahrländer J, Sauter A, Becker M, Wistorf E, Steinfath M and Stolz A: GPER1 links estrogens to centrosome amplification and chromosomal instability in human colon cells. Life Sci Alliance. 6:e2022014992022. View Article : Google Scholar : PubMed/NCBI | |
|
Bustos V, Nolan ÁM, Nijhuis A, Harvey H, Parker A, Poulsom R, McBryan J, Thomas W, Silver A and Harvey BJ: GPER mediates differential effects of estrogen on colon cancer cell proliferation and migration under normoxic and hypoxic conditions. Oncotarget. 8:84258–84275. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Santolla MF, Lappano R, De Marco P, Pupo M, Vivacqua A, Sisci D, Abonante S, Iacopetta D, Cappello AR, Dolce V, et al: G protein-coupled estrogen receptor mediates the up-regulation of fatty acid synthase induced by 17β-estradiol in cancer cells and cancer-associated fibroblasts. J Biol Chem. 287:43234–43245. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Xie M, Liang JL, Huang HD, Wang MJ, Zhang T and Yang XF: Low doses of nonylphenol promote growth of colon cancer cells through activation of ERK1/2 via G Protein-coupled receptor 30. Cancer Res Treat. 51:1620–1631. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Gilligan LC, Rahman HP, Hewitt AM, Sitch AJ, Gondal A, Arvaniti A, Taylor AE, Read ML, Morton DG and Foster PA: Estrogen activation by steroid sulfatase increases colorectal cancer proliferation via GPER. J Clin Endocrinol Metab. 102:4435–4447. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Rouhimoghadam M, Lu AS, Salem AK and Filardo EJ: Therapeutic perspectives on the modulation of G-protein coupled estrogen receptor, GPER, Function. Front Endocrinol (Lausanne). 11:5912172020. View Article : Google Scholar : PubMed/NCBI | |
|
Rodon J, Chaney M, Cohen J, Garyantes TK, Ishizuka J, Lin JJ, Lorusso P, Mita A, Mita M, Muller C, et al: The effect of LNS8801 in combination with pembrolizumab in patients with treatment-refractory cutaneous melanoma. J Immuno Ther Cancer. 11 (Suppl 1):A6272023. | |
|
Shoushtari AN, Chaney MF, Cohen JV, Garyantes T, Lin JJ, Ishizuka JJ, Mita AC, Mita MM, Muller C, Natale C, et al: The effect of LNS8801 alone and in combination with pembrolizumab in patients with metastatic uveal melanoma. J Clin Oncol. 41 (16_Suppl):S9543. 2023. View Article : Google Scholar | |
|
Holm A, Grände PO, Ludueña RF, Olde B, Prasad V, Leeb-Lundberg LM and Nilsson BO: The G protein-coupled oestrogen receptor 1 agonist G-1 disrupts endothelial cell microtubule structure in a receptor-independent manner. Mol Cell Biochem. 366:239–249. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Wang C, Lv X, Jiang C and Davis JS: The putative G-protein coupled estrogen receptor agonist G-1 suppresses proliferation of ovarian and breast cancer cells in a GPER-independent manner. Am J Transl Res. 4:390–402. 2012.PubMed/NCBI | |
|
Gui Y, Shi Z, Wang Z, Li JJ, Xu C, Tian R, Song X, Walsh MP, Li D, Gao J, et al: The GPER agonist G-1 induces mitotic arrest and apoptosis in human vascular smooth muscle cells independent of GPER. J Cell Physiol. 230:885–895. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Mori T, Ito F, Matsushima H, Takaoka O, Tanaka Y, Koshiba A, Kusuki I and Kitawaki J: G protein-coupled estrogen receptor 1 agonist G-1 induces cell cycle arrest in the mitotic phase, leading to apoptosis in endometriosis. Fertil Steril. 103:1228–1235.e1. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Lv X, He C, Huang C, Hua G, Wang Z, Remmenga SW, Rodabough KJ, Karpf AR, Dong J, Davis JS, et al: G-1 Inhibits breast cancer cell growth via targeting Colchicine-binding site of tubulin to interfere with microtubule assembly. Mol Cancer Ther. 16:1080–1091. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Torres-López L, Olivas-Aguirre M, Villatoro-Gómez K and Dobrovinskaya O: The G-protein-coupled estrogen receptor agonist G-1 inhibits proliferation and causes apoptosis in leukemia cell lines of T lineage. Front Cell Dev Biol. 10:8114792022. View Article : Google Scholar : PubMed/NCBI | |
|
Haupt S, Caramia F, Herschtal A, Soussi T, Lozano G, Chen H, Liang H, Speed TP and Haupt Y: Identification of cancer sex-disparity in the functional integrity of p53 and its X chromosome network. Nat Commun. 10:53852019. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y and Bodmer WF: Analysis of P53 mutations and their expression in 56 colorectal cancer cell lines. Proc Natl Acad Sci USA. 103:976–981. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Schneider CA, Rasband WS and Eliceiri KW: NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 9:671–675. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Recchia AG, De Francesco EM, Vivacqua A, Sisci D, Panno ML, Andò S and Maggiolini M: The G protein-coupled receptor 30 is up-regulated by hypoxia-inducible factor-1alpha (HIF-1alpha) in breast cancer cells and cardiomyocytes. J Biol Chem. 286:10773–10782. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Jacenik D, Beswick EJ, Krajewska WM and Prossnitz ER: G protein-coupled estrogen receptor in colon function, immune regulation and carcinogenesis. World J Gastroenterol. 25:4092–4104. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Foley EF, Jazaeri AA, Shupnik MA, Jazaeri O and Rice LW: Selective loss of estrogen receptor beta in malignant human colon. Cancer Res. 60:245–248. 2000.PubMed/NCBI | |
|
Jassam N, Bell SM, Speirs V and Quirke P: Loss of expression of oestrogen receptor beta in colon cancer and its association with Dukes' staging. Oncol Rep. 14:17–21. 2005.PubMed/NCBI | |
|
Mostafaie N, Kállay E, Sauerzapf E, Bonner E, Kriwanek S, Cross HS, Huber KR and Krugluger W: Correlated downregulation of estrogen receptor beta and the circadian clock gene Per1 in human colorectal cancer. Mol Carcinog. 48:642–647. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Herichova I, Reis R, Hasakova K, Vician M and Zeman M: Sex-dependent regulation of estrogen receptor beta in human colorectal cancer tissue and its relationship with clock genes and VEGF-A expression. Physiol Res. 68 (Suppl 3):S297–S305. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Hasakova K, Vician M, Reis R, Zeman M and Herichova I: Sex-dependent correlation between survival and expression of genes related to the circadian oscillator in patients with colorectal cancer. Chronobiol Int. 35:1423–1434. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Williams C, DiLeo A, Niv Y and Gustafsson JÅ: Estrogen receptor beta as target for colorectal cancer prevention. Cancer Lett. 372:48–56. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Ya G, Wang H, Ma Y, Hu A, Ma Y, Hu J and Yu Y: Serum miR-129 functions as a biomarker for colorectal cancer by targeting estrogen receptor (ER) β. Pharmazie. 72:107–112. 2017.PubMed/NCBI | |
|
Issa JP, Ottaviano YL, Celano P, Hamilton SR, Davidson NE and Baylin SB: Methylation of the oestrogen receptor CpG island links ageing and neoplasia in human colon. Nat Genet. 7:536–540. 1994. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang H, Teng R, Wang Q, Zhang X, Wang H, Wang Z, Cao J and Teng L: Transcriptional analysis of estrogen receptor alpha variant mRNAs in colorectal cancers and their matched normal colorectal tissues. J Steroid Biochem Mol Biol. 112:20–24. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Topi G, Ghatak S, Satapathy SR, Ehrnström R, Lydrup ML and Sjölander A: Combined estrogen alpha and beta receptor expression has a prognostic significance for colorectal cancer patients. Front Med (Lausanne). 9:7396202022. View Article : Google Scholar : PubMed/NCBI | |
|
Kennelly R, Kavanagh DO, Hogan AM and Winter DC: Oestrogen and the colon: Potential mechanisms for cancer prevention. Lancet Oncol. 94:385–391. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Barton M, Filardo EJ, Lolait SJ, Thomas P, Maggiolini M and Prossnitz ER: Twenty years of the G protein-coupled estrogen receptor GPER: Historical and personal perspectives. J Steroid Biochem Mol Biol. 176:4–15. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Harvey BJ and Harvey HM: Sex differences in colon cancer: Genomic and nongenomic signalling of oestrogen. Genes (Basel). 14:22252023. View Article : Google Scholar : PubMed/NCBI | |
|
Nüssler NC, Reinbacher K, Shanny N, Schirmeier A, Glanemann M, Neuhaus P, Nussler AK and Kirschner M: Sex-specific differences in the expression levels of estrogen receptor subtypes in colorectal cancer. Gend Med. 5:209–217. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Deli T, Orosz M and Jakab A: Hormone replacement therapy in cancer survivors-review of the literature. Pathol Oncol Res. 26:63–78. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Topi G, Satapathy SR, Dash P, Fred Mehrabi S, Ehrnström R, Olsson R, Lydrup ML and Sjölander A: Tumour-suppressive effect of oestrogen receptor β in colorectal cancer patients, colon cancer cells, and a zebrafish model. J Pathol. 251:297–309. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Tirado-Garibay AC, Falcón-Ruiz EA, Ochoa-Zarzosa A and López-Meza JE: GPER: An estrogen receptor key in metastasis and tumoral microenvironments. Int J Mol Sc. 24:149932023. View Article : Google Scholar : PubMed/NCBI | |
|
Ambrosini G, Natale CA, Musi E, Garyantes T and Schwartz GK: The GPER agonist LNS8801 induces mitotic arrest and apoptosis in uveal melanoma cells. Cancer Res Commun. 3:540–547. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Filardo EJ, Quinn JA, Bland KI and Frackelton AR Jr: Estrogen-induced activation of Erk-1 and Erk-2 requires the G protein-coupled receptor homolog, GPR30, and occurs via trans-activation of the epidermal growth factor receptor through release of HB-EGF. Mol Endocrinol. 14:1649–1660. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Hsu LH, Chu NM, Lin YF and Kao SH: G-Protein coupled estrogen receptor in breast cancer. Int J Mol Sci. 20:3062019. View Article : Google Scholar : PubMed/NCBI | |
|
Rubin Grandis J, Melhem MF, Gooding WE, Day R, Holst VA, Wagener MM, Drenning SD and Tweardy DJ: Levels of TGF-alpha and EGFR protein in head and neck squamous cell carcinoma and patient survival. J Natl Cancer Inst. 90:824–832. 1998. View Article : Google Scholar : PubMed/NCBI | |
|
Uribe ML, Marrocco I and Yarden Y: EGFR in cancer: Signaling mechanisms, drugs, and acquired resistance. Cancers (Basel). 13:27482021. View Article : Google Scholar : PubMed/NCBI | |
|
Janani B, Vijayakumar M, Priya K, Kim JH, Prabakaran DS, Shahid M, Al-Ghamdi S, Alsaidan M, Othman Bahakim N, Hassan Abdelzaher M and Ramesh T: EGFR-based targeted therapy for colorectal cancer-promises and challenges. Vaccines (Basel). 10:4992022. View Article : Google Scholar : PubMed/NCBI | |
|
Giannopoulou E, Antonacopoulou A, Floratou K, Papavassiliou AG and Kalofonos HP: Dual targeting of EGFR and HER-2 in colon cancer cell lines. Cancer Chemother Pharmacol. 63:973–981. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Yuan HH, Han Y, Bian WX, Liu L and Bai YX: The effect of monoclonal antibody cetuximab (C225) in combination with tyrosine kinase inhibitor gefitinib (ZD1839) on colon cancer cell lines. Pathology. 44:547–551. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Palumbo I, Piattoni S, Valentini V, Marini V, Contavalli P, Calzuola M, Vecchio FM, Cecchini D, Falzetti F and Aristei C: Gefitinib enhances the effects of combined radiotherapy and 5-fluorouracil in a colorectal cancer cell line. Int J Colorectal Dis. 29:31–41. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Chen X, Liu Y, Yang HW, Zhou S, Cheng C, Zheng MW, Zhong L, Fu XY, Pan YL, Ma S, et al: SKLB-287, a novel oral multikinase inhibitor of EGFR and VEGFR2, exhibits potent antitumor activity in LoVo colorectal tumor model. Neoplasma. 61:514–22. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Chuang SC, Chen CH, Chou YS, Ho ML and Chang JK: G Protein-coupled estrogen receptor mediates cell proliferation through the cAMP/PKA/CREB pathway in murine bone marrow mesenchymal stem cells. Int J Mol Sci. 21:64902020. View Article : Google Scholar : PubMed/NCBI | |
|
Berg KCG, Eide PW, Eilertsen IA, Johannessen B, Bruun J, Danielsen SA, Bjørnslett M, Meza-Zepeda LA, Eknæs M, Lind GE, et al: Multi-omics of 34 colorectal cancer cell lines-a resource for biomedical studies. Mol Cancer. 16:1162017. View Article : Google Scholar : PubMed/NCBI | |
|
Moulder DE, Hatoum D, Tay E, Lin Y and McGowan EM: The roles of p53 in mitochondrial dynamics and cancer metabolism: The pendulum between survival and death in breast cancer? Cancers (Basel). 10:1892018. View Article : Google Scholar : PubMed/NCBI | |
|
Engeland K: Cell cycle regulation: P53-p21-RB signaling. Cell Death Differ. 29:946–960. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Weißenborn C, Ignatov T, Ochel HJ, Costa SD, Zenclussen AC, Ignatova Z and Ignatov A: GPER functions as a tumor suppressor in triple-negative breast cancer cells. J Cancer Res Clin Oncol. 140:713–23. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Rochette PJ, Bastien N, Lavoie J, Guérin SL and Drouin R: SW480, a p53 double-mutant cell line retains proficiency for some p53 functions. J Mol Biol. 352:44–57. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Berglind H, Pawitan Y, Kato S, Ishioka C and Soussi T: Analysis of p53 mutation status in human cancer cell lines: A paradigm for cell line cross-contamination. Cancer Biol Ther. 7:699–708. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Hassin O, Nataraj NB, Shreberk-Shaked M, Aylon Y, Yaeger R, Fontemaggi G, Mukherjee S, Maddalena M, Avioz A, Iancu O, et al: Different hotspot p53 mutants exert distinct phenotypes and predict outcome of colorectal cancer patients. Nat Commun. 13:28002022. View Article : Google Scholar : PubMed/NCBI | |
|
Leroy B, Girard L, Hollestelle A, Minna JD, Gazdar AF and Soussi T: Analysis of TP53 mutation status in human cancer cell lines: A reassessment. Hum Mutat. 35:756–765. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Berger C, Qian Y and Chen X: The p53-estrogen receptor loop in cancer. Curr Mol Med. 13:1229–1240. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Chen YC, Young MJ, Chang HP, Liu CY, Lee CC, Tseng YL, Wang YC, Chang WC and Hung JJ: Estradiol-mediated inhibition of DNMT1 decreases p53 expression to induce M2-macrophage polarization in lung cancer progression. Oncogenesis. 11:252022. View Article : Google Scholar : PubMed/NCBI | |
|
Weige CC, Allred KF, Armstrong CM and Allred CD: P53 mediates estradiol induced activation of apoptosis and DNA repair in non-malignant colonocytes. J Steroid Biochem Mol Biol. 128:113–120. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Qin C, Nguyen T, Stewart J, Samudio I, Burghardt R and Safe S: Estrogen up-regulation of p53 gene expression in MCF-7 breast cancer cells is mediated by calmodulin kinase IV-dependent activation of a nuclear factor kappaB/CCAAT-binding transcription factor-1 complex. Mol Endocrinol. 16:1793–809. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Berger CE, Qian Y, Liu G, Chen H and Chen X: p53, a target of estrogen receptor (ER) α, modulates DNA damage-induced growth suppression in ER-positive breast cancer cells. J Biol Chem. 287:30117–30127. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Swetzig WM, Wang J and Das GM: Estrogen receptor alpha (ERα/ESR1) mediates the p53-independent overexpression of MDM4/MDMX and MDM2 in human breast cancer. Oncotarget. 7:16049–16069. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Mancini F, Giorgini L, Teveroni E, Pontecorvi A and Moretti F: Role of sex in the therapeutic targeting of p53 circuitry. Front Oncol. 11:6989462021. View Article : Google Scholar : PubMed/NCBI | |
|
Konduri SD, Medisetty R, Liu W, Kaipparettu BA, Srivastava P, Brauch H, Fritz P, Swetzig WM, Gardner AE, Khan SA, et al: Mechanisms of estrogen receptor antagonism toward p53 and its implications in breast cancer therapeutic response and stem cell regulation. Proc Natl Acad Sci USA. 107:15081–6. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Lu W and Katzenellenbogen BS: Estrogen receptor-β modulation of the ERα-p53 loop regulating gene expression, proliferation, and apoptosis in breast cancer. Horm Cancer. 8:230–242. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al: The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2:401–404. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Bilancio A, Bontempo P, Di Donato M, Conte M, Giovannelli P, Altucci L, Migliaccio A and Castoria G: Bisphenol A induces cell cycle arrest in primary and prostate cancer cells through EGFR/ERK/p53 signaling pathway activation. Oncotarget. 8:115620–115631. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Wei W, Chen ZJ, Zhang KS, Yang XL, Wu YM, Chen XH, Huang HB, Liu HL, Cai SH, Du J, et al: The activation of G protein-coupled receptor 30 (GPR30) inhibits proliferation of estrogen receptor-negative breast cancer cells in vitro and in vivo. Cell Death Dis. 5:e14282014. View Article : Google Scholar : PubMed/NCBI | |
|
Morelli E, Hunter ZR, Fulciniti M, Gullà A, Perrotta ID, Zuccalà V, Federico C, Juli G, Manzoni M, Ronchetti D, et al: Therapeutic activation of G protein-coupled estrogen receptor 1 in Waldenström Macroglobulinemia. Exp Hematol Onco. 11:542022. View Article : Google Scholar | |
|
Giebler HA, Lemasson I and Nyborg JK: p53 recruitment of CREB binding protein mediated through phosphorylated CREB: A novel pathway of tumor suppressor regulation. Mol Cell Biol. 20:4849–4858. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Okoshi R, Ando K, Suenaga Y, Sang M, Kubo N, Kizaki H, Nakagawara A and Ozaki T: Transcriptional regulation of tumor suppressor p53 by cAMP-responsive element-binding protein/AMP-activated protein kinase complex in response to glucose deprivation. Genes Cells. 14:1429–1440. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Lee CW, Ferreon JC, Ferreon AC, Arai M and Wright PE: Graded enhancement of p53 binding to CREB-binding protein (CBP) by multisite phosphorylation. Proc Natl Acad Sci USA. 107:19290–19295. 2010. View Article : Google Scholar : PubMed/NCBI |