Differential modulation of natural killer cell cytotoxicity by 17β‑estradiol and prolactin through the NKG2D/NKG2DL axis in cervical cancer cells

  • Authors:
    • Alejandro Godoy-Pacheco
    • Mariel García-Chagollán
    • Adrián Ramírez-De-Arellano
    • Christian David Hernández-Silva
    • Julio César Villegas-Pineda
    • Inocencia Guadalupe Ramírez-López
    • José Sergio Zepeda-Nuño
    • Adriana Aguilar-Lemarroy
    • Ana Laura Pereira-Suárez
  • View Affiliations

  • Published online on: June 28, 2022     https://doi.org/10.3892/ol.2022.13408
  • Article Number: 288
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Natural killer (NK) cells play a crucial role in cervical cancer (CC). As estrogens and prolactin (PRL) have been reported to be involved in CC, the present study attempted to elucidate the effects of both hormones on NK cells in CC. For this purpose, NKL cells, as well as CC‑derived cell lines (HeLa, SiHa and C33A) and non‑tumorigenic keratinocytes (HaCaT cells) were stimulated with 17β‑estradiol (E2; 10 nM), PRL (200 ng/ml), or both (E2 and PRL) for 48 h. The expression of hormone receptors (estrogen receptor α and β, G protein‑coupled estrogen receptor 1 and PRL receptor) and NK cell activating receptors [natural killer group 2D (NKG2D), natural cytotoxicity triggering receptor 3, natural cytotoxicity triggering receptor 2 and natural cytotoxicity triggering receptor 1] were measured using western blot analysis and flow cytometry, respectively. In the HeLa, SiHa, C33A and HaCaT cells stimulated with the hormones, the expression of NKG2D ligands [MHC class I polypeptide‑related sequence A/B (MICA/B)] on the membrane and the soluble form of MICA was evaluated using flow cytometry and ELISA. Cytotoxicity assay was performed using GFP‑transfected K562 cells as target cells. E2 reduced NKL cell‑mediated cytotoxicity, while PRL exerted the opposite effect. NKL cells expressed different hormone receptor forms, of which PRL only induced a decrease in NKG2D expression compared to the untreated control NKL cells. PRL increased MICA/B expression in HeLa cells and E2 and PRL reversed this effect. However, in SiHa cells, the concurrent incubation with the two hormones decreased MICA/B expression. E2 and PRL, either alone or in combination, decreased soluble MICA secretion in all CC cell lines, while E2 solely increased soluble MICA secretion in SiHa cells. On the whole, the present study provides evidence that E2 and PRL mediate the mechanisms through which NK and CC cells mediate a cytotoxic response and these have an antagonistic effect on NK cell‑mediated cytotoxicity.

Introduction

Cervical cancer (CC) is one of the most common cancers among women worldwide and the second cause of cancer mortality in developing countries (1). Human papillomavirus (HPV) is the leading risk factor for CC development (2). However, different types of lesions may be observed in the cervix prior to cancer establishment, including grade 1, 2 and 3 cervical intraepithelial neoplasia, as well as invasive carcinoma (3). Apart from HPV infection, other risk factors have been reported to be involved in the transformation process from normal to malignant cells, including smoking, oral contraceptive use and steroid sex hormones, among others (46). The tumor microenvironment (TME) is crucial for the carcinogenic process, and hormones are a key factor in this context. In addition, cells that belong to the innate immune system are located in the TME, having the ability to kill tumor cells (7). 17β-estradiol (E2) and prolactin (PRL) have been reported to be present in the TME (810); however, their role on immunological mechanisms generated in the response to CC is poorly understood.

Estrogens are sex hormones that belong to the cholesterol-derived steroids group, whose three primary forms are estrone (E1), E2 and estriol (E3), of which E2 has been reported to exhibit an increased biological activity (11). The functions of E2 are mediated through the estrogen receptors (ER)α and β, and the G protein-coupled estrogen receptor 1 (GPER) (12,13). Of note, ERα, ERβ and GPER have been shown to be overexpressed in CC tissues compared with that in the premalignant lesion and normal cervical epithelium (14,15). Studies using mice have demonstrated that a temporary presence of E2 promotes CC, being ERα signaling-dependent (16,17).

PRL is a lactogenic polypeptide hormone synthesized primarily by the pituitary gland (18). Additionally, other studies have demonstrated an extrapituitary PRL production by some tissues and organs. PRL exerts its functions through its binding to the PRL receptor (PRLR) (19,20). Previous studies have demonstrated the expression of a 60 kDa PRL in CC tissues and CC-derived cells. This PRL variant may regulate various processes, including apoptosis, cytokine production and metabolism in THP-1 and CC-derived cells (14,21,22).

PRLR is a member of the class I cytokine receptor superfamily; it presents with various isoforms, one long, one intermediate, and two short isoforms, with an average weight of 85–90, 65 and 40–50 kDa respectively (23). High PRL levels have been reported in the serum of patients with CC (24). There is also evidence of the increased expression of PRLR in premalignant lesions, CC tissues and CC-derived cell lines (21). The stimulation of CC-derived cells with PRL induces the expression of anti-apoptotic gene through the signal transducer and activator of transcription (STAT)-3 (25). This evidence confirms the importance of PRL in CC pathogenesis and some relevant events in the progression of the disease.

Natural killer (NK) cells are a major component of the innate immunity against tumors and viral infections. They constitute 5 to 15% of all lymphocytes and are phenotypically defined by the expression of CD56 and the absence of CD3 (26). NK cells are equipped with a repertoire of receptors that can both stimulate (activating receptors) or prevent (inhibitory receptors) their reactivity (27). The natural cytotoxicity receptors (NCRs), including natural cytotoxicity triggering receptor 3 (NKp30), natural cytotoxicity triggering receptor 2 (NKp44) and natural cytotoxicity triggering receptor 1 (NKp46), have been reported to induce NK cell activation; however, their corresponding ligands have not yet been well defined (28). Another activating receptor is natural killer group 2D (NKG2D), a type 2 transmembrane protein, whose ligands include MHC-I chain-related protein A and B (MICA and MICB) and the UL16 binding proteins (ULBP) from 1 to 6 (29). In 2012, a previous study revealed that NKG2D receptor expression in NK cells decreased when interacting directly with CC cell lines (30). Another study revealed that the expression of NKp30 and NKp46 receptors was decreased in squamous intraepithelial lesions and CC; however, NKG2D was only decreased in CC, and was negatively associated with NK cell cytotoxic activity (31). Of note, tumors evade the immune system through the liberation of MICA and MICB from the cellular membrane to create a soluble form (32). This process has been reported to be mediated by various metalloproteinases (33). The soluble form of MICA and MICB has been found to be associated with the internalization and degradation of NKG2D and the consequent decrease in the NK cell-mediated cytotoxicity (34,35). In cancers, such as CC, which is related to a viral infection, it is crucial to understand whether the factors included in the TME, including hormones, may modify the mechanisms that favor the malignancy of the disease.

Both estrogen and PRL receptors have been identified in human cell lines and murine NK cells. However, the expression of GPER in these cells remains unclear (36,37). There is evidence to indicate that estrogens have been linked to a decrease in NK cell cytotoxicity using human and murine models, while PRL exert opposite effects in human NK cell lines (NK-92 and YT cell lines) (37). In addition, the effects of these hormones may affect the regulation of proteins that belong to cytotoxicity processes, including activating receptors and their ligands (3743).

Riera-Leal et al (14), observed the effects of E2 and PRL on CC-cell line metabolism and concluded that the two hormones increased cell metabolism, with PRL to a lesser extent than E2. However, PRL appears to exert a more prominent effect over E2 when simultaneously applied.

In the CC TME, E2 and PRL are present. Thus, the present study aimed to investigate the effects of the E2 and PRL stimuli, concurrently or separately applied on NKL cells and CC-derived cell lines, as well as to evaluate the expression of different molecules related to NK cell-mediated cytotoxicity, including NCR, NKG2D and MICA/B.

Materials and methods

Cell culture and hormone stimuli

The HeLa, SiHa, C33A, MCF7 (all from ATCC) and HaCaT (CLS Cell Lines Service GmbH) cell lines were cultured in DMEM medium, supplemented with 10% fetal bovine serum (FBS), 1% penicillin G (10,000 U/ml), and streptomycin (10,000 µg/ml) (Gibco; Thermo Fisher Scientific, Inc.). Similarly, the NKL (kindly donated by Dr Adriana Aguilar Lemarroy) and K562 (ATCC) cell lines were grown in supplemented RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc.). All cell lines were incubated at 37°C and 5% CO2 until 80% of confluence was obtained. NKL, HeLa, SiHa, C33A and HaCaT cell cultures were stimulated for 48 h with PRL (200 ng/ml) isolated from HeLa cell supernatant, E2 (10 nM; Sigma Aldrich; Merck KGaA), or both (E2 and PRL). HeLa, SiHa, C33A, HaCaT and K562 cell lines were authenticated by Multiplexion GmbH, using the multiplex human cell line authentication test.

Isolation and purification of the 60 kDa-weighted PRL

The isolation of PRL from the HeLa cell supernatant was performed using magnetic beads (Protein G Microbeads MultiMACS; Miltenyi Biotec GmbH) following the manufacturer's protocol. The 60 kDa PRL was purified employing the 50 kDa molecular cut-off filters (Amicon® Ultra 0.5 ml centrifugal filters; cat. no. UFC505024; MilliporeSigma). The procedure for the filtration was performed as follows: 14,000 × g for 30 min (filtration phase); 1,000 × g for 2 min (recovery phase) at 4°C. Once purified, the correct identification of the 60 kDa PRL was determined using a 12% polyacrylamide gel for electrophoresis at 95V for 90 min. Subsequently, silver nitrate (cat. no. 209139; MilliporeSigma) staining was performed for 20 min at room temperature to visualize the 60-kDa band belonging to PRL. Finally, quantification of the purified protein was performed utilizing Thermo Scientific NanoDrop 2000c Spectrophotometer (NanoDrop Technologies; Thermo Fisher Scientific, Inc.).

NK cell cytotoxicity assay

The cytotoxicity of NK cells against the Green Fluorescent Protein (GFP)-transfected K562 cell line (kindly donated from Dr Adriana Aguilar Lemarroy) was evaluated in a propidium iodide (PI) flow cytometry assay. K562 cells were seeded with a constant number (150,000) with different effector (NK cells) to target cell ratios [effector:target (E:T) 1:1, 5:1 and 10:1]. The target cells were incubated alone to measure untreated control cell death. Co-cultures between NKL and GFP-transfected K562 cells (GFP-K562) (lymphoblasts derived from chronic myeloid leukemia), characterized by its absence or decrease of MHC-I molecules, in complex medium were performed for 4 h at 37°C and 5% CO2. The cells were washed twice with 1% PBS and incubated in the same buffer with PI (cat. no. P4170; MilliporeSigma) for 20 min at room temperature in darkness. The reading was performed using Attune® NxT acoustic focus cytometer with the FACS Diva v3.1.2 software (BD Biosciences). The cytotoxic activity was expressed as the % of specific lysis by using the following formula:

Degranulation assay

CD107a was used as a marker of NKL degranulation upon target recognition. A total of 30,000 NKL cells (effector) were co-cultured with 30,000 K562 cells (target) cells at an E:T ratio of 1:1 in 96-well plates for 4 h. At the start of the incubation period, a 1:400 dilution of anti-CD107a-PE (cat. no. 555801; BD Biosciences) was added to each well. Monensin (BioLegend, Inc.) was used as a protein transport blocker an added for 1 h into the co-culture. To identify viable NKL cells from the target cells, a 1:50 dilution of CD45 antibody (BioLegend, Inc.; cat. no. 304027) and Zombie NIR dye (BioLegend, Inc.; cat. no. 423105) were used. The reading was performed using Attune® NxT acoustic focus cytometer with the FACS Diva Software v3.1.2 (BD Biosciences).

Western blot analysis

Total proteins were extracted from NKL and MCF7 cell lines (obtained from ATCC) using RIPA lysis and extraction buffer (cat. no. 89900; Thermo Fisher Scientific, Inc.), and the coomasie plus (Bradford) assay (cat. no. 23238; Thermo Fisher Scientific, Inc.) was used for protein quantification. A total of 50 µg protein was mixed with loading buffer and then denatured at 95°C for 5 min. Electrophoresis was performed on 10% polyacrylamide gels at 110 V for 60 min, and subsequently, a PVDF-membrane electrical transference (Bio-Rad Laboratories, Inc.) was performed for 90 min at 240 V. The membranes were incubated overnight at 4°C with a blocking solution of 1X PBS and 5% blotting-grade blocker (cat. no. 1706404; Bio-Rad Laboratories, Inc.). The dilution of the primary antibodies used was 1:500 for ERα, ERβ and PRLR (cat. nos. sc-8002, sc-373853, sc-20992, respectively; Santa Cruz Biotechnology, Inc.) and GPER (cat. no. ab39742; Abcam) and 1:10,000 for β-actin (cat. no. sc-47778; Santa Cruz Biotechnology, Inc.) in blocking solution consisting of 1X PBS and 5% blotting-grade blocker, and incubated overnight at 4°C. The membranes were washed five times for 7 min with PBS and Tween-20 (cat. no. P1379; MilliporeSigma) and incubated for 90 min with a dilution of 1:10,000 anti-mouse or anti-rabbit secondary antibodies (cat. nos. sc-2005, sc-2357; Santa Cruz Biotechnology, Inc.) at room temperature. Subsequently, the membranes were washed 6 times for 10 min. Luminol and horseradish peroxidase reagents (Immobilion; Merck KGaA) were used to perform the chemiluminescence process. β-actin expression was used as an internal control. The Microchemi 6.0 (DNR Bio-Imaging Systems Ltd.) was used to visualize the membranes and GelQuant software V1.7.8 (BiochemLabSolutions) was utilized for densitometric measurement.

Flow cytometry

For the NKL cell lines with and without hormonal stimulation, the cell density was adjusted to 2×105 cells in total. The cells were washed with 1X PBS and centrifuged at 1,800 × g for 10 min at 4°C. Subsequently, cells were incubated with anti-NKG2D, anti-NKp30, anti-NKp44 and anti-NKp46 antibodies at 1:100 dilution (cat. nos. 130-123-948, 130-121-995, 130-120-623 and 130-126-054, respectively; Miltenyi Biotec GmbH) at 4°C for 30 min in the dark. The cells were washed again and centrifuged at 1,800 × g for 5 min at 4°C. The cells were then fixed with 1 ml 0.05% PBS-formaldehyde solution. Following the same procedure, CC-derived and HaCaT cell lines were labeled against anti-MICA/B antibodies (cat. no. 130-100-889; Miltenyi Biotec GmbH) for analysis using flow cytometry. The percentages and mean fluorescence intensity (MFI) were determined with appropriate protocols and controls to electronically compensate the overlapping signals using the Attune® NxT Software v3.1.2 acoustic focus cytometer (Invitrogen; Thermo Fisher Scientific, Inc.).

Soluble MICA quantification in cell culture supernatants

Soluble MICA levels were analyzed using the Human MICA ELISA kit (cat. no. RAB0358-1KT; MilliporeSigma) in the supernatant of HeLa, SiHa, C33A and HaCaT cell lines stimulated with E2 and PRL, according to the manufacturer´s instructions. The results were obtained from two independent experiments using the appropriate absorbance values (450 nm).

Statistical analysis

Data capture was performed using the statistical program GraphPad 8.0.2 (GraphPad Software, Inc.). Statistical analysis to compare the expression patterns of hormone receptors, activating receptors, ligands and differences in the cytotoxicity activity were carried out, using the ANOVA test followed by the Tukey's post hoc test for multiple comparisons. P<0.05 was considered to indicate a statistically significant difference.

Results

Antagonistic effects between E2 and PRL on NK cell-mediated cytotoxicity

To evaluate the effect of hormones on NK cell-mediated cytotoxicity, NKL cells were stimulated with E2 (10 nM) or PRL (200 ng/ml) either alone or in combination for 48 h and subsequently co-cultured with GFP-K562 cells at various E:T ratios (1:1, 5:1 and 10:1) for 4 h. The identification of dead target cells was characterized as GFP+PI+ by flow cytometric analysis (Fig. 1A).

Comparing the effect of hormones against untreated control cells (without stimulation), it was demonstrated that stimulation with E2 tends to decrease the lysis of GFP-K562 cells; however, PRL stimulation tended to increase cytotoxicity against GFP-K562 cells. Notably, at a 10:1 ratio, stimulation with PRL exerted a positive effect on the cytotoxicity on NKL cells, contrary to E2, which exerts an antagonistic effect as compared to PRL (P<0.05). Notably, the combined effect of the two hormones exerted similar effects as those observed with PRL alone, as regards NKL cell-mediated cytotoxicity (P<0.05) (Fig. 1B). It was revealed that the hormones located within the TME may have the ability to regulate the cytotoxicity of NK cells with antagonistic outcomes.

To confirm the cytotoxicity assay results, a degranulation assay in NKL cells was performed, and it was observed that CD107a expression tended to decrease in NKL cells stimulated with E2 compared to the untreated control cells. It was also demonstrated that PRL stimulation was able to induce CD107a expression compared to the unstimulated cells, which is consistent with the antagonistic effects shown by the cytotoxicity assay. The E2 + PRL stimulation did not cause a marked change in degranulation marker expression in NKL cells (Fig. 1C).

Expression of hormonal receptors in NKL cells

Once it was observed that hormones can regulate NKL cell-mediated cytotoxicity, the expression of the estrogen receptors (ERα, ERβ and GPER) and the PRL receptor were characterized using western blotting with protein extracts from NKL cells.

The expression of the four hormonal receptors is depicted in Fig. 2. The expression of ERα is denoted by the presence of a single 46-kDa band. The bands indicating the expression of ERβ are 32, 45 and 56 kDa. Of note, the 45-kDa band was thicker in the two cell lines. To the best of our knowledge, this is the first study to demonstrate the presence of GPER in NK cells. GPER was expressed as 42- and 100-kDa bands. Notably, the highest GPER expression was the 100-kDa band, which has been related to glycosylation of this receptor. Finally, PRLR was observed in several bands of approximately 44, 50, 65 and 90 kDa in agreement with the various isoforms of the receptor. Of note, the expression of PRLR was higher in the 50-kDa band, which corresponds to a short isoform of this receptor. When compared with that of the MCF7 cell line, which was used as a positive control, the expression pattern of ERα and ERβ was similar to that observed in the NKL cells. In the MCF7 cells, a higher expression level of the normal form of GPER was observed compared with that in the NKL cells. Finally, it was observed that MCF7 cells expressed the long isoforms of PRLR in a greater proportion in comparison to the NKL cells. The presence of all hormone receptors and their isoforms contribute to a more detailed understanding of the hormonal effects on NK cells.

Expression of NCR and NKG2D in NKL cells stimulated with E2 and PRL

To evaluate the effect of hormone stimuli on activating receptors including NKp30, NKp44, NKp46 and NKG2D expression on NKL cell surface, cells were stimulated with E2 and PRL for 48 h and flow cytometric analysis was performed (Fig. 3). In the untreated control cells (without stimulation), the percentage of NKG2D and NKp30 receptors (100 and 96.2%, respectively) was higher than that for NKp46 (14.6%). As was expected, the percentage of positivity for the NKp44 receptor was zero, as previously reported (44). The histograms demonstrated that the percentages of positive NKL cells for the different receptors were not markedly altered due to hormonal stimuli (Fig. 3A, C, E and G). However, PRL stimuli induced a significant decrease in the expression of NKG2D compared to the untreated control cells or E2 stimuli (P<0.05; Fig. 3B). The expression of NKp30, NKp44 and NKp46 receptors in NKL cells was not significantly altered by E2 and PRL stimuli (Fig. 3D, F, H). The downregulation of NKG2D due to PRL stimulation demonstrated that it may be able to modulate signaling pathways involved in NK cell cytotoxicity.

Modulation in the expression of NKG2D ligands in CC-derived cells by hormonal stimuli

Since CC cells induce the expression of stress ligands, including MICA and MICB (NKG2D receptor ligands in NK cells), the present study then evaluated the effects of E2 and PRL on MICA/B expression in CC-derived cell lines (HeLa, SiHa and C33A) and a non-tumorigenic immortalized keratinocyte cell line (HaCaT). As previously described in the literature, it was observed that, in the untreated control cells, the HPV-18 and HPV-16 positive cell lines (HeLa and SiHa, respectively) presented a higher percentage of cells expressing MICA/B (99.7 and 99.8%, respectively) (45), contrary to HPV-negative cells (C33A and HaCaT, 8.35 and 2.09%, respectively; Fig. 4A). Stimulation of HeLa cells with PRL increased the expression of MICA/B compared to the untreated control cells (P<0.05); however, the simultaneous stimulation with E2 and PRL reversed this effect (P<0.05). By contrast, in SiHa cells, the concurrent stimulation with E2 and PRL decreased MICA/B expression compared to the untreated control cells and E2 stimulus (P<0.05; Fig. 4A). Hormonal stimuli did not induce changes in MICA/B expression in the C33A or HaCaT cells.

Among the escape mechanisms of tumor cells towards immunological recognition is the release or secretion of soluble forms of activating ligands. In line with this, it has been discovered that MIC molecules can be released into the extracellular matrix and thereby promote an immune escape strategy for tumor cells (32). For this reason, in the present study, the levels of soluble MICA (sMICA) in CC-derived and HaCaT cell line supernatants, stimulated for 48 h with E2, PRL or both was evaluated (Fig. 4B). In comparison to the untreated control cells, stimulation with E2 or PRL alone, and E2 and PRL in combination, decreased the liberation of MICA into the supernatant of all CC-derived cell lines (P<0.05), apart from the SiHa cells, where E2 stimulation resulted in increased sMICA levels (P<0.05). This effect was abrogated by stimulation with PRL alone, and with E2 and PRL in combination (P<0.01) in SiHa cells. Notably, the hormone stimuli had no effect on MICA secretion in the HaCaT cell supernatant. Both the membrane and soluble form of the MICA ligand are regulated by E2 and PRL in CC-derived cell lines.

Discussion

CC represents one of the main health issues in women. Of note, 604,000 new cases worldwide were estimated in 2020. The main risk factor associated with CC is HPV infection, which is present in >99% of patients with CC (1). However, it has been revealed that HPV infection alone is not sufficient for CC to manifest (16). In this sense, the hormonal role constitutes an important factor for the carcinogenesis of this type of tumor. Hormones, including 17β-estradiol and PRL are related to the genesis, persistence and development of CC (14,16,21,46), since in addition to being present in the TME of this cancer type, they can contribute to anti-apoptotic, proliferative, invasive, survival effects and metabolic adaptation of CC cells (10,15,21,22,25,47). In addition, they can regulate the expression of HPV oncogenes (48). The functionality of these hormones within the TME may also depend on a bilateral regulation between the two hormones, since there are studies demonstrating the possible regulation of PRLR by E2, as well as the regulation of estrogen receptors exerted by PRL effects (4951).

In the TME there are also cells of the innate immune system, including NK cells, which have the potential to kill cells transformed and infected by HPV (52). As regards CC, studies have revealed that there is a poor infiltration of NK cells, and therefore this may be associated with a decrease in their cytotoxic activity against tumor cells (53,54). In both in vitro models and patients, it has been observed that CC cells are capable of regulating NK cell cytotoxicity given that tumor-infiltrating NK cells decrease the expression of perforins, activating receptors and IFN-γ, and on the other hand increasing the expression of inhibition receptors (54,55). Likewise, the expression of activating receptors, such as NKG2D, and the expression of cell stress ligands with CC have been related (30,31,45,56). The present study demonstrated that E2 decreased the cytotoxicity of NKL cells, as well as CD107a expression, which is consistent with the findings in the studies by Hao et al (57,58) underlining that various concentrations of E2 may have a negative effect on proliferative capacity, IFN-γ expression and the cytotoxic effects of the NK cells extracted from mouse spleens against the YAC-1 target cells. A possible explanation for this phenomenon is the indirect decrease in granzyme B levels, due to the effect of E2, where it has been demonstrated that estrogen induces the expression of inhibitory proteinase 9, a potent inhibitor of granzyme B (38,58).

Subsequently, when confirming the effect of E2 on the cytotoxicity of NKL cells, the present study analyzed the possible isoforms of the estrogen receptors that these cells express, with the aim of determining the pathway through which E2 may exert such an effect. As regards ERα, it has been revealed that it presents with three main isoforms, known as ERα66, ERα46 and ERα36, named for their characteristic molecular weights (59). NKL cells express the 46 kDa isoform of ERα, which has also been previously detected in lymphocytes with CD3+ CD8+ and CD3CD56+ phenotypes obtained from peripheral blood (60). This isoform is characterized by the lack of the first 173 amino acids of the amino terminal AF-1 domain and has been associated with an inhibitory role on tumor cell growth, as in breast cancer cells ERα46 may inhibit the estrogenic effects of ERα66, inducing in turn cell proliferation and cell cycle progression. It has been suggested that these effects occur due to a functional competition between both isoforms (59,61,62). In relation to ERβ, in other human cancer models it has been demonstrated that this receptor presents with various isoforms, known as ERβ1, β2, β3, β4 and β5 whose molecular weights range from 50 to 59 kDa (63). NKL cells strongly express ERβ, represented as a 45-kDa band and a weaker expression of a 56-kDa band. Similarly, in peripheral blood lymphocytes the expression of ERβ with weights lower than 56 kDa has been detected (60). Furthermore, in breast cancer cells the presence of ERβ isoforms with molecular weights of around 44 kDa has also been observed. This may be attributed to the fact that exons 5 and 6 of the ERβ mRNA are eliminated, thereby generating a protein of lower molecular weight (60,64). To date, the possible role of these ERβ isoforms with molecular weights <50 kDa is unknown; therefore, further studies are warranted to achieve a better understanding at the functional level of these variants.

Another receptor through which E2 has been reported to exert its effects is the G protein-coupled estrogen receptor, GPER, which has been reported to be associated with non-genomic pathways through kinase-dependent signaling for rapid gene regulation (65). Recent findings have revealed that GPER is overexpressed in biopsies of patients with CC and its agonistic activation increases mitochondrial permeability, as well as apoptosis, as well decreases the proliferation of CC cells (15). NKL cells express a weak band of 42 kDa and a strong band of around 100 kDa. Currently, no evidence has been reported concerning the presence of GPER in these cells; however, the high weight of GPER has been related to glycosylated forms and/or dimerization of this receptor (66,67). It has been demonstrated that GPER glycosylation may occur mainly in an asparagine residue known as Asn44 and this post-translational modification has been associated with its location in the plasma membrane, where GPER can regulate the rapid non-genomic response of estrogens (68,69).

By contrast, in the present study it was observed that stimulation with PRL induced an increase in NKL cell-mediated cytotoxicity and CD107a expression. This is in line with previous studies by Sun et al (37,70), where NK cells extracted from mice treated with PRL and also from cell lines including NK-92 were used. This increase in NK cell-mediated cytotoxicity may be attributed to the fact that PRL, in conjunction with IL-2 and IL-15, may increase the expression of IFN-γ, perforins and Fas-L (37). Considering that PRL has been reported to exert its effects through its receptor, it was decided to visualize the possible isoforms by which PRL could exert this effect on cytotoxicity. PRLR is expressed in a number of isoforms, including a long (between 80 and 90 kDa), an intermediate (65 kDa) and 2 short isoforms (between 40 and 55 kDa) (23). The variant with the highest expression in NKL cells was the short isoform corresponding to the 50-kDa band, characterized by the lack of the Box 2 region, which is crucial for the interaction with proteins containing an SH2 domain, including STAT proteins, leading to a negative regulation on the effects triggered by the long isoform of the PRLR (18,23). Further more detailed studies are required to determine whether the different PRLR isoforms may have a functional effect on NK cells. Notably, the concurrent stimulation of E2 and PRL also increased the cytotoxicity of NKL cells, as observed with PRL alone. It was observed that PRL may overcome the effects of E2 and as previously mentioned, this may be attributed to both hormones having a bilateral regulation (4951). This is in line with Riera-Leal et al (14), who observed in a context of CC cell metabolism, that PRL may have a greater impact over the estrogenic effects induced by E2.

Subsequently, the present study aimed to evaluate whether the differences in the cytotoxicity of NKL cells by hormones may be attributed to changes in the expression of activation receptors, including NCR and NKG2D. The data obtained did not indicate that these effects were related to the NCR, since it was observed that the hormones did not modify the expression of NKp30, NKp44 and NKp46. By contrast, it was observed that PRL may decrease the expression of NKG2D in NKL cells. This is in line with a previous study by Ma et al (71) in 2010, where it was demonstrated that the expression of NKG2D may decrease in T lymphocytes from patients with prolactinoma.

When the change in the expression of NKG2D by these hormones was observed, it was decided to evaluate MICA and MICB ligands of this receptor, which are expressed in the membrane, as well as in soluble forms. MICA and MICB are known as stress proteins and these ligands have been reported to be elevated in CC patient biopsies and to be also overexpressed in CC-derived cell lines, including SiHa, HeLa, CALO and INBL (45,54,71). The results of the present study are consistent with the findings from the study by del Toro-Arreola et al (45), with MICA/B being expressed mainly in HaCaT, C33A, HeLa and SiHa cells. Furthermore, sMICA was detected in cell supernatants at relatively similar levels. Of note, it was demonstrated that PRL may increase MICA/B expression on the HeLa cell surface, while decreasing sMICA release in the supernatant of all CC-derived cells. In the context of the interaction that exists in the CC microenvironment, the increase in cytotoxicity which was observed under the effect of PRL may be explained by the increase in MICA/B in the membrane, which can bind to NKG2D; this is also supported by the decrease in sMICA. To the best of our knowledge, there no studies available to date that relate the effect of PRL with the release of MIC molecules. However, it has been revealed that metalloprotease 9, which has the ability to cleave MICA, decreases its expression due to the effects of PRL, possibly explaining the aforementioned result (72,73). By contrast, in the present study, E2 decreased sMICA in HeLa and C33A cells, whereas an opposite effect was observed in SiHa cells, possibly indicating that the effects of E2 vary depending on the cell type. Although an effect of E2 on MICA/B surface expression, when stimulating the cells with both hormones was not detected, it was observed that sMICA expression decreased in all CC-derived cell lines. This may indicate that the joint effect of the hormones may be related to the increase in the cytotoxicity of NKL cells and also supporting the regulatory effect of one hormone on the other.

In conclusion, the results of the present study suggested that E2 and PRL, which are overexpressed in the CC microenvironment, may antagonistically regulate the cytotoxicity of NK cells. Furthermore, NKL cells express different variants of the hormone receptors by which their effects may be exerted. By contrast, hormones regulate the expression of molecules, including the NKG2D receptor, MICA/B ligands and their soluble forms, which may be involved in the cytotoxicity of NK cells. This knowledge revealed an overview that may help in understanding further the mechanism by which these hormones may contribute to the development of CC. It would be of interest to evaluate the possible molecules involved in pathways triggered by E2 and PRL on NK cell-mediated cytotoxicity in future studies, using next-generation RNA sequencing, ultimately aiming to identify novel therapeutic targets involved in CC.

Acknowledgements

Not applicable.

Funding

This study was supported by the Sectorial Research Fund for Education, SEP-CONACYT (grant no. A1-S-51207), and the Jalisco Scientific Development Fund (FODECIJAL) to Attend State Problems 2019 (Project #8168). The study was supported by a CONACYT fellowship (#885574).

Availability of data and materials

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

Authors' contributions

AGP and CDHS performed the experiments. MGC assisted in the flow cytometry data analysis and in the interpretation of the results. ARdA, JCVP, IGRL, AAL and MGC contributed to the statistical analysis and the critical review of the manuscript. JSZN participated in the ELISA. AGP and MGC confirm the authenticity of all the raw data. ALPS designed the study and wrote the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Wójcik L, Samulak D, Makowska M, Romanowicz H, Kojs Z, Smolarz B and Michalska MM: The role of human papillomavirus in cervical cancer. Int J Cancer Clin Res. 6:1252019.

3 

Van hede D, Langers I, Delvenne P and Jacobs N: Origin and immunoescape of uterine cervical cancer. Presse Med. 43:e413–e421. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Zhang S, Xu H, Zhang L and Qiao Y: Cervical cancer: Epidemiology, risk factors and screening. Chin J Cancer Res. 32:720–728. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Momenimovahed Z and Salehiniya H: Incidence, mortality and risk factors of cervical cancer in the world. Biomed Res Ther. 4:1795–1811. 2017. View Article : Google Scholar

6 

Chung SH, Franceschi S and Lambert PF: Estrogen and ERalpha: Culprits in cervical cancer? Trends Endocrinol Metab. 21:504–511. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Barros MR Jr, de Melo CML, Barros MLCMGR, de Cássia Pereira de Lima R, de Freitas AC and Venuti A: Activities of stromal and immune cells in HPV-related cancers. J Exp Clin Cancer Res. 37:1372018. View Article : Google Scholar : PubMed/NCBI

8 

Ding L, Liu C, Zhou Q, Feng M and Wang J: Association of estradiol and HPV/HPV16 infection with the occurrence of cervical squamous cell carcinoma. Oncol Lett. 17:3548–3554. 2019.

9 

Adurthi S, Kumar MM, Vinodkumar HS, Mukherjee G, Krishnamurthy H, Acharya KK, Bafna UD, Uma DK, Abhishekh B, Krishna S, et al: Oestrogen Receptor-α binds the FOXP3 promoter and modulates regulatory T-cell function in human cervical cancer. Sci Rep. 7:172892017. View Article : Google Scholar : PubMed/NCBI

10 

Lopez-Pulido EI, Muñoz-Valle JF, Del Toro-Arreola S, Jave-Suárez LF, Bueno-Topete MR, Estrada-Chávez C and Pereira-Suárez AL: High expression of prolactin receptor is associated with cell survival in cervical cancer cells. Cancer Cell Int. 13:1032013. View Article : Google Scholar

11 

Gruber CJ, Tschugguel W, Schneeberger C and Huber JC: Production and actions of estrogens. N Engl J Med. 346:340–352. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Shanle EK and Xu W: Endocrine disrupting chemicals targeting estrogen receptor signaling: Identification and mechanisms of action. Chem Res Toxicol. 24:6–19. 2011. View Article : Google Scholar

13 

Mizukami Y: In vivo functions of GPR30/GPER-1, a membrane receptor for estrogen: From discovery to functions in vivo. Endocr J. 57:101–107. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Riera-Leal A, Ramírez De Arellano A, Ramírez-López IG, Lopez-Pulido EI, Dávila Rodríguez JR, Macías-Barragan JG, Ortiz-Lazareno PC, Jave-Suárez LF, Artaza-Irigaray C, Del Toro Arreola S, et al: Effects of 60 kDa prolactin and estradiol on metabolism and cell survival in cervical cancer: Co-expression of their hormonal receptors during cancer progression. Oncol Rep. 40:3781–3793. 2018.PubMed/NCBI

15 

Hernandez-Silva CD, Riera-Leal A, Ortiz-Lazareno PC, Jave-Suárez LF, Ramírez De Arellano A, Lopez-Pulido EI, Macías-Barragan JG, Montoya-Buelna M, Dávila-Rodríguez JR, Chabay P, et al: GPER overexpression in cervical cancer versus premalignant lesions: Its activation induces different forms of cell death. Anticancer Agents Med Chem. 19:783–791. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Brake T and Lambert PF: Estrogen contributes to the onset, persistence, and malignant progression of cervical cancer in a human papillomavirus-transgenic mouse model. Proc Natl Acad Sci USA. 102:2490–2495. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Chung SH, Wiedmeyer K, Shai A, Korach KS and Lambert PF: Requirement for estrogen receptor alpha in a mouse model for human papillomavirus-associated cervical cancer. Cancer Res. 68:9928–9934. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Bernard V, Young J, Chanson P and Binart N: New insights in prolactin: Pathological implications. Nat Rev Endocrinol. 11:265–275. 2015. View Article : Google Scholar

19 

Marano RJ and Ben-Jonathan N: Minireview: Extrapituitary prolactin: An update on the distribution, regulation, and functions. Mol Endocrinol. 28:622–633. 2014. View Article : Google Scholar

20 

Brooks CL: Molecular mechanisms of prolactin and its receptor. Endocr Rev. 33:504–525. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Ascencio-Cedillo R, López-Pulido EI, Muñoz-Valle JF, Villegas-Sepúlveda N, Del Toro-Arreola S, Estrada-Chávez C, Daneri-Navarro A, Franco-Topete R, Pérez-Montiel D, García-Carrancá A and Pereira-Suárez AL: Prolactin and prolactin receptor expression in cervical intraepithelial neoplasia and cancer. Pathol Oncol Res. 21:241–246. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Ramírez De Arellano A, Riera Leal A, Lopez-Pulido EI, González-Lucano LR, Macías Barragan J, Del Toro Arreola S, García-Chagollan M, Palafox-Sánchez CA, Muñoz-Valle JF and Pereira-Suárez AL: A 60 kDa prolactin variant secreted by cervical cancer cells modulates apoptosis and cytokine production. Oncol Rep. 39:1253–1260. 2018.

23 

Abramicheva PA and Smirnova OV: Prolactin receptor isoforms as the basis of tissue-specific action of prolactin in the norm and pathology. Biochemistry (Mosc). 84:329–345. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Hsu CT, Yu MH, Lee CY, Jong HL and Yeh MY: Ectopic production of prolactin in uterine cervical carcinoma. Gynecol Oncol. 44:166–171. 1992. View Article : Google Scholar

25 

Ramírez de Arellano A, Lopez-Pulido EI, Martínez-Neri PA, Estrada Chávez C, González Lucano R, Fafutis-Morris M, Aguilar-Lemarroy A, Muñoz-Valle JF and Pereira-Suárez AL: STAT3 activation is required for the antiapoptotic effects of prolactin in cervical cancer cells. Cancer Cell Int. 15:832015. View Article : Google Scholar

26 

Cooper MA, Fehniger TA and Caligiuri MA: The biology of human natural killer-cell subsets. Trends Immunol. 22:633–640. 2001. View Article : Google Scholar

27 

Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, Yokoyama WM and Ugolini S: Innate or Adaptive Immunity? The example of natural killer cells. Science. 331:44–49. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Brandt CS, Baratin M, Yi EC, Kennedy J, Gao Z, Fox B, Haldeman B, Ostrander CD, Kaifu T, Chabannon C, et al: The B7 family member B7-H6 is a tumor cell ligand for the activating natural killer cell receptor NKp30 in humans. J Exp Med. 206:1495–1503. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Srivastava RM, Savithri B and Khar A: Activating and inhibitory receptors and their role in natural killer cell function. Indian J Biochem Biophys. 40:291–299. 2003.PubMed/NCBI

30 

Jimenez-Perez MI, Jave-Suarez LF, Ortiz-Lazareno PC, Bravo-Cuellar A, Gonzalez-Ramella O, Aguilar-Lemarroy A, Hernandez-Flores G, Pereira-Suarez AL, Daneri-Navarro A and del Toro-Arreola S: Cervical cancer cell lines expressing NKG2D-ligands are able to down-modulate the NKG2D receptor on NKL cells with functional implications. BMC Immunol. 13:72012. View Article : Google Scholar

31 

Garcia-Iglesias T, Del Toro-Arreola A, Albarran-Somoza B, Del Toro-Arreola S, Sanchez-Hernandez PE, Ramirez-Dueñas MG, Balderas-Peña LM, Bravo-Cuellar A, Ortiz-Lazareno PC and Daneri-Navarro A: Low NKp30, NKp46 and NKG2D expression and reduced cytotoxic activity on NK cells in cervical cancer and precursor lesions. BMC Cancer. 9:1862009. View Article : Google Scholar : PubMed/NCBI

32 

Duan S, Guo W, Xu Z, He Y, Liang C, Mo Y, Wang Y, Xiong F, Guo C, Li Y, et al: Natural killer group 2D receptor and its ligands in cancer immune escape. Mol Cancer. 18:292019. View Article : Google Scholar : PubMed/NCBI

33 

López-Soto A, Huergo-Zapico L, Acebes-Huerta A, Villa-Alvarez M and Gonzalez S: NKG2D signaling in cancer immunosurveillance: NKG2D signaling. Int J Cancer. 136:1741–1750. 2015. View Article : Google Scholar

34 

Baragaño Raneros A, Suarez-Álvarez B and López-Larrea C: Secretory pathways generating immunosuppressive NKG2D ligands: New targets for therapeutic intervention. Oncoimmunology. 3:e284972014. View Article : Google Scholar

35 

Chitadze G, Bhat J, Lettau M, Janssen O and Kabelitz D: Generation of soluble NKG2D ligands: Proteolytic cleavage, exosome secretion and functional implications. Scand J Immunol. 78:120–129. 2013. View Article : Google Scholar

36 

Curran EM, Berghaus LJ, Vernetti NJ, Saporita AJ, Lubahn DB and Estes DM: Natural killer cells express estrogen receptor-alpha and estrogen receptor-beta and can respond to estrogen via a non-estrogen receptor-alpha-mediated pathway. Cell Immunol. 214:12–20. 2001. View Article : Google Scholar

37 

Sun R, Li AL, Wei HM and Tian ZG: Expression of prolactin receptor and response to prolactin stimulation of human NK cell lines. Cell Res. 14:67–73. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Jiang X, Ellison SJ, Alarid ET and Shapiro DJ: Interplay between the levels of estrogen and estrogen receptor controls the level of the granzyme inhibitor, proteinase inhibitor 9 and susceptibility to immune surveillance by natural killer cells. Oncogene. 26:4106–4114. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Jiang X, Orr BA, Kranz DM and Shapiro DJ: Estrogen induction of the granzyme B inhibitor, proteinase inhibitor 9, protects cells against apoptosis mediated by cytotoxic T lymphocytes and natural killer cells. Endocrinology. 147:1419–1426. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Mavoungou E, Bouyou-Akotet MK and Kremsner PG: Effects of prolactin and cortisol on natural killer (NK) cell surface expression and function of human natural cytotoxicity receptors (NKp46, NKp44 and NKp30). Clin Exp Immunol. 139:287–296. 2005. View Article : Google Scholar

41 

Basu S, Pioli PA, Conejo-Garcia J, Wira CR and Sentman CL: Estradiol regulates MICA expression in human endometrial cells. Clin Immunol. 129:325–332. 2008. View Article : Google Scholar

42 

Ren J, Nie Y, Lv M, Shen S, Tang R, Xu Y, Hou Y, Zhao S and Wang T: Estrogen upregulates MICA/B expression in human non-small cell lung cancer through the regulation of ADAM17. Cell Mol Immunol. 12:768–776. 2015. View Article : Google Scholar

43 

Wolfson B, Padget MR, Schlom J and Hodge JW: Exploiting off-target effects of estrogen deprivation to sensitize estrogen receptor negative breast cancer to immune killing. J Immunother Cancer. 9:e0022582021. View Article : Google Scholar : PubMed/NCBI

44 

Gunesch JT, Angelo LS, Mahapatra S, Deering RP, Kowalko JE, Sleiman P, Tobias JW, Monaco-Shawver L, Orange JS and Mace EM: Genome-wide analyses and functional profiling of human NK cell lines. Mol Immunol. 115:64–75. 2019. View Article : Google Scholar

45 

del Toro-Arreola S, Arreygue-Garcia N, Aguilar-Lemarroy A, Cid-Arregui A, Jimenez-Perez M, Haramati J, Barros-Nuñez P, Gonzalez-Ramella O, Del Toro-Arreola A, Ortiz-Lazareno P, et al: MHC class I-related chain A and B ligands are differentially expressed in human cervical cancer cell lines. Cancer Cell Int. 11:152011. View Article : Google Scholar

46 

Huang Y, Li J, Xiang L, Han D, Shen X and Wu X: 17β-Oestradiol activates proteolysis and increases invasion through phosphatidylinositol 3-kinase pathway in human cervical cancer cells. Eur J Obstet Gynecol Reprod Biol. 165:307–312. 2012. View Article : Google Scholar

47 

Riera Leal A, Ortiz-Lazareno PC, Jave-Suárez LF, Ramírez De Arellano A, Aguilar-Lemarroy A, Ortiz-García YM, Barrón-Gallardo CA, Solís-Martínez R, Luquin De Anda S, Muñoz-Valle JF and Pereira-Suárez AL: 17β-estradiol-induced mitochondrial dysfunction and Warburg effect in cervical cancer cells allow cell survival under metabolic stress. Int J Oncol. 56:33–46. 2020.

48 

Ramírez-López IG, Ramírez de Arellano A, Jave-Suárez LF, Hernández-Silva CD, García-Chagollan M, Hernández-Bello J, Lopez-Pulido EI, Macias-Barragan J, Montoya-Buelna M, Muñoz-Valle JF and Pereira-Suárez AL: Interaction between 17β-estradiol, prolactin and human papillomavirus induce E6/E7 transcript and modulate the expression and localization of hormonal receptors. Cancer Cell Int. 19:2272019. View Article : Google Scholar

49 

Leondires MP, Hu ZZ, Dong J, Tsai-Morris CH and Dufau ML: Estradiol stimulates expression of two human prolactin receptor isoforms with alternative exons-1 in T47D breast cancer cells. J Steroid Biochem Mol Biol. 82:263–268. 2002. View Article : Google Scholar

50 

Adamson AD, Friedrichsen S, Semprini S, Harper CV, Mullins JJ, White MR and Davis JR: Human prolactin gene promoter regulation by estrogen: Convergence with tumor necrosis factor-alpha signaling. Endocrinology. 149:687–694. 2008. View Article : Google Scholar : PubMed/NCBI

51 

González L, Zambrano A, Lazaro-Trueba I, Lopéz E, González JJA, Martín-Pérez J and Aranda A: Activation of the unliganded estrogen receptor by prolactin in breast cancer cells. Oncogene. 28:1298–1308. 2009. View Article : Google Scholar

52 

Sasagawa T, Takagi H and Makinoda S: Immune responses against human papillomavirus (HPV) infection and evasion of host defense in cervical cancer. J Infect Chemother. 18:807–815. 2012. View Article : Google Scholar : PubMed/NCBI

53 

Garzetti G, Ciavattini A, Muzzioli M, Goteri G, Mannello B, Romanini C and Fabris N: Natural killer cell activity in patients with invasive cervical carcinoma: Importance of a longitudinal evaluation in follow-up. Gynecol Obstet Invest. 40:133–138. 1995. View Article : Google Scholar

54 

Textor S, Dürst M, Jansen L, Accardi R, Tommasino M, Trunk MJ, Porgador A, Watzl C, Gissmann L and Cerwenka A: Activating NK cell receptor ligands are differentially expressed during progression to cervical cancer. Int J Cancer. 123:2343–2353. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Chang WC, Li CH, Chu LH, Huang PS, Sheu BC and Huang SC: Regulatory T cells suppress natural killer cell immunity in patients with human cervical carcinoma. Int J Gynecol Cancer. 26:156–162. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Arreygue-Garcia NA, Daneri-Navarro A, del Toro-Arreola A, Cid-Arregui A, Gonzalez-Ramella O, Jave-Suarez LF, Aguilar-Lemarroy A, Troyo-Sanroman R, Bravo-Cuellar A, Delgado-Rizo V, et al: Augmented serum level of major histocompatibility complex class I-related chain A (MICA) protein and reduced NKG2D expression on NK and T cells in patients with cervical cancer and precursor lesions. BMC Cancer. 8:162008. View Article : Google Scholar : PubMed/NCBI

57 

Hao S, Zhao J, Zhou J, Zhao S, Hu Y and Hou Y: Modulation of 17beta-estradiol on the number and cytotoxicity of NK cells in vivo related to MCM and activating receptors. Int Immunopharmacol. 7:1765–1775. 2007. View Article : Google Scholar

58 

Hao S, Li P, Zhao J, Hu Y and Hou Y: 17beta-estradiol suppresses cytotoxicity and proliferative capacity of murine splenic NK1.1+ cells. Cell Mol Immunol. 5:357–364. 2008. View Article : Google Scholar

59 

Arnal JF, Lenfant F, Metivier R, Flouriot G, Henrion D, Adlanmerini M, Fontaine C, Gourdy P, Chambon P, Katzenellenbogen B and Katzenellenbogen J: Membrane and nuclear estrogen receptor alpha actions: from tissue specificity to medical implications. Physiol Rev. 97:1045–1087. 2017. View Article : Google Scholar : PubMed/NCBI

60 

Pierdominici M, Maselli A, Colasanti T, Giammarioli AM, Delunardo F, Vacirca D, Sanchez M, Giovannetti A, Malorni W and Ortona E: Estrogen receptor profiles in human peripheral blood lymphocytes. Immunol Lett. 132:79–85. 2010. View Article : Google Scholar

61 

Penot G, Le Péron C, Mérot Y, Grimaud-Fanouillère E, Ferrière F, Boujrad N, Kah O, Saligaut C, Ducouret B, Métivier R and Flouriot G: The human estrogen receptor-alpha isoform hERalpha46 antagonizes the proliferative influence of hERalpha66 in MCF7 breast cancer cells. Endocrinology. 146:5474–5484. 2005. View Article : Google Scholar : PubMed/NCBI

62 

Miller MM, McMullen PD, Andersen ME and Clewell RA: Multiple receptors shape the estrogen response pathway and are critical considerations for the future of in vitro-based risk assessment efforts. Crit Rev Toxicol. 47:570–586. 2017. View Article : Google Scholar

63 

Leung YK, Mak P, Hassan S and Ho SM: Estrogen receptor (ER)-beta isoforms: A key to understanding ER-beta signaling. Proc Natl Acad Sci USA. 103:13162–13167. 2006. View Article : Google Scholar : PubMed/NCBI

64 

Saunders PT, Millar MR, Williams K, Macpherson S, Bayne C, O'Sullivan C, Anderson TJ, Groome NP and Miller WR: Expression of oestrogen receptor beta (ERbeta1) protein in human breast cancer biopsies. Br J Cancer. 86:250–256. 2002. View Article : Google Scholar : PubMed/NCBI

65 

Gaudet HM, Cheng SB, Christensen EM and Filardo EJ: The G-protein coupled estrogen receptor, GPER: The inside and inside-out story. Mol Cell Endocrinol. 418:207–219. 2015. View Article : Google Scholar

66 

Sandén C, Broselid S, Cornmark L, Andersson K, Daszkiewicz-Nilsson J, Mårtensson UE, Olde B and Leeb-Lundberg LM: G protein-coupled estrogen receptor 1/G protein-coupled receptor 30 localizes in the plasma membrane and traffics intracellularly on cytokeratin intermediate filaments. Mol Pharmacol. 79:400–410. 2011. View Article : Google Scholar

67 

Jala VR, Radde BN, Haribabu B and Klinge CM: Enhanced expression of G-protein coupled estrogen receptor (GPER/GPR30) in lung cancer. BMC Cancer. 12:6242012. View Article : Google Scholar : PubMed/NCBI

68 

Pupo M, Bodmer A, Berto M, Maggiolini M, Dietrich PY and Picard D: A genetic polymorphism repurposes the G-protein coupled and membrane-associated estrogen receptor GPER to a transcription factor-like molecule promoting paracrine signaling between stroma and breast carcinoma cells. Oncotarget. 8:46728–46744. 2017. View Article : Google Scholar

69 

Gonzalez de Valdivia E, Sandén C, Kahn R, Olde B and Leeb-Lundberg LMF: Human G protein-coupled receptor 30 is N-glycosylated and N-terminal domain asparagine 44 is required for receptor structure and activity. Biosci Rep. 39:BSR201824362019. View Article : Google Scholar : PubMed/NCBI

70 

Sun R, Wei H, Zhang J, Li A, Zhang W and Tian ZG: Recombinant human prolactin improves antitumor effects of murine natural killer cells in vitro and in vivo. Neuroimmunomodulation. 10:169–176. 2002. View Article : Google Scholar : PubMed/NCBI

71 

Ma L, Li G, Su Y, He Q, Zhang C and Zhang J: The soluble major histocompatibility complex class I-related chain A protein reduced NKG2D expression on natural killer and T cells from patients with prolactinoma and non-secreting pituitary adenoma. J Clin Neurosci. 17:241–247. 2010. View Article : Google Scholar

72 

Zaga-Clavellina V, Parra-Covarrubias A, Ramirez-Peredo J, Vega-Sanchez R and Vadillo-Ortega F: The potential role of prolactin as a modulator of the secretion of proinflammatory mediators in chorioamniotic membranes in term human gestation. Am J Obstet Gynecol. 211:48.e1–e6. 2014. View Article : Google Scholar

73 

Shiraishi K, Mimura K, Kua LF, Koh V, Siang LK, Nakajima S, Fujii H, Shabbir A, Yong WP, So J, et al: Inhibition of MMP activity can restore NKG2D ligand expression in gastric cancer, leading to improved NK cell susceptibility. J Gastroenterol. 51:1101–1111. 2016. View Article : Google Scholar

Related Articles

Journal Cover

August-2022
Volume 24 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Godoy-Pacheco A, García-Chagollán M, Ramírez-De-Arellano A, Hernández-Silva CD, Villegas-Pineda JC, Ramírez-López IG, Zepeda-Nuño JS, Aguilar-Lemarroy A and Pereira-Suárez AL: Differential modulation of natural killer cell cytotoxicity by 17β‑estradiol and prolactin through the NKG2D/NKG2DL axis in cervical cancer cells. Oncol Lett 24: 288, 2022
APA
Godoy-Pacheco, A., García-Chagollán, M., Ramírez-De-Arellano, A., Hernández-Silva, C.D., Villegas-Pineda, J.C., Ramírez-López, I.G. ... Pereira-Suárez, A.L. (2022). Differential modulation of natural killer cell cytotoxicity by 17β‑estradiol and prolactin through the NKG2D/NKG2DL axis in cervical cancer cells. Oncology Letters, 24, 288. https://doi.org/10.3892/ol.2022.13408
MLA
Godoy-Pacheco, A., García-Chagollán, M., Ramírez-De-Arellano, A., Hernández-Silva, C. D., Villegas-Pineda, J. C., Ramírez-López, I. G., Zepeda-Nuño, J. S., Aguilar-Lemarroy, A., Pereira-Suárez, A. L."Differential modulation of natural killer cell cytotoxicity by 17β‑estradiol and prolactin through the NKG2D/NKG2DL axis in cervical cancer cells". Oncology Letters 24.2 (2022): 288.
Chicago
Godoy-Pacheco, A., García-Chagollán, M., Ramírez-De-Arellano, A., Hernández-Silva, C. D., Villegas-Pineda, J. C., Ramírez-López, I. G., Zepeda-Nuño, J. S., Aguilar-Lemarroy, A., Pereira-Suárez, A. L."Differential modulation of natural killer cell cytotoxicity by 17β‑estradiol and prolactin through the NKG2D/NKG2DL axis in cervical cancer cells". Oncology Letters 24, no. 2 (2022): 288. https://doi.org/10.3892/ol.2022.13408