Register      Login
Reproduction, Fertility and Development Reproduction, Fertility and Development Society
Vertebrate reproductive science and technology
RESEARCH ARTICLE

Changes in receptor location affect the ability of oxytocin to stimulate proliferative growth in prostate epithelial cells

M. L. Gould https://orcid.org/0000-0003-3733-1359 A B and H. D. Nicholson A
+ Author Affiliations
- Author Affiliations

A Anatomy Department, University of Otago, PO Box 913, Dunedin 9054, New Zealand.

B Corresponding author. Email: maree.gould@otago.ac.nz

Reproduction, Fertility and Development 31(6) 1166-1179 https://doi.org/10.1071/RD18362
Submitted: 14 September 2018  Accepted: 4 February 2019   Published: 30 April 2019

Abstract

In normal prostate cells, cell membrane receptors are located within signalling microdomains called caveolae. During cancer progression, caveolae are lost and sequestered receptors move out onto lipid rafts. The aim of this study was to investigate whether a change in the localisation of receptors out of caveolae and onto the cell membrane increased cell proliferation in vitro, and to determine whether this is related to changes in the cell signalling pathways. Normal human prostate epithelial cells (PrEC) and androgen-independent (PC3) cancer cells were cultured with 10 nM dihydrotestosterone (DHT). The effects of oxytocin (OT) and gonadal steroids on proliferation were assessed using the MTS assay. Androgen receptor (AR) and oxytocin receptor (OTR) expression was identified by immunofluorescence and quantified by western blot. OTR and lipid raft staining was determined using Pearson’s correlation coefficient. Protein–protein interactions were detected and the cell signalling pathways identified. Treatment with OT did not affect the proliferation of PrEC. In PC3 cells, OT or androgen alone increased cell proliferation, but together had no effect. In normal cells, OTR localised to the membrane and AR localised to the nucleus, whereas in malignant cells both OTR and AR were identified in the cell membrane. Colocalisation of OTR and AR increased following treatment with androgens. Significantly fewer OTR/AR protein–protein interactions were seen in PrEC. With OT treatment, several cell signalling pathways were activated. Movement of OTR out of caveolae onto lipid rafts is accompanied by activation of alternative signal transduction pathways involved in stimulating increased cell proliferation.

Additional keywords: caveolae, cell membrane, lipid rafts.


References

Adler, J., and Parmryd, I. (2010). Quantifying colocalization by correlation: the Pearson correlation coefficient is superior to the Mander’s overlap coefficient. Cytometry A 77A, 733–742.
Quantifying colocalization by correlation: the Pearson correlation coefficient is superior to the Mander’s overlap coefficient.Crossref | GoogleScholarGoogle Scholar |

Alimirah, F., Chen, J., Basrawala, Z., Xin, H., and Choubey, D. (2006). DU-145 and PC-3 human prostate cancer cell lines express androgen receptor: implications for the androgen receptor functions and regulation. FEBS Lett. 580, 2294–2300.
DU-145 and PC-3 human prostate cancer cell lines express androgen receptor: implications for the androgen receptor functions and regulation.Crossref | GoogleScholarGoogle Scholar | 16580667PubMed |

Arnold, J. T., and Isaacs, J. T. (2002). Mechanisms involved in the progression of androgen-independent prostate cancers: it is not only the cancer cell’s fault. Endocr. Relat. Cancer 9, 61–73.
Mechanisms involved in the progression of androgen-independent prostate cancers: it is not only the cancer cell’s fault.Crossref | GoogleScholarGoogle Scholar | 11914183PubMed |

Aung, C. S., Hill, M. M., Bastiani, M., Parton, R. G., and Parat, M. O. (2011). PTRF-cavin-1 expression decreases the migration of PC3 prostate cancer cells: role of matrix metalloprotease 9. Eur. J. Cell Biol. 90, 136–142.
PTRF-cavin-1 expression decreases the migration of PC3 prostate cancer cells: role of matrix metalloprotease 9.Crossref | GoogleScholarGoogle Scholar | 20732728PubMed |

Bale, T. L., and Dorsa, D. M. (1995). Regulation of oxytocin receptor messenger ribonucleic acid in the ventromedial hypothalamus by testosterone and its metabolites. Endocrinology 136, 5135–5138.
Regulation of oxytocin receptor messenger ribonucleic acid in the ventromedial hypothalamus by testosterone and its metabolites.Crossref | GoogleScholarGoogle Scholar | 7588251PubMed |

Bélanger, B., Bélanger, A., Labrie, F., Dupont, A., Cusan, L., and Monfette, G. (1989). Comparison of residual C-19 steroids in plasma and prostatic tissue of human, rat and guinea pig after castration: unique importance of extratesticular androgens in men. J. Steroid Biochem. 32, 695–698.
Comparison of residual C-19 steroids in plasma and prostatic tissue of human, rat and guinea pig after castration: unique importance of extratesticular androgens in men.Crossref | GoogleScholarGoogle Scholar | 2525654PubMed |

Bennett, N., Hooper, J. D., Lee, C. S., and Gobe, G. C. (2009). Androgen receptor and caveolin-1 in prostate cancer. IUBMB Life 61, 961–970.
Androgen receptor and caveolin-1 in prostate cancer.Crossref | GoogleScholarGoogle Scholar | 19787702PubMed |

Berthois, Y., Katzenellenbogen, J. A., and Katzenellenbogen, B. S. (1986). Phenol red in tissue culture media is a weak estrogen: implications concerning the study of estrogen-responsive cells in culture. Proc. Natl Acad. Sci. USA 83, 2496–2500.
Phenol red in tissue culture media is a weak estrogen: implications concerning the study of estrogen-responsive cells in culture.Crossref | GoogleScholarGoogle Scholar | 3458212PubMed |

Bookstein, R., MacGrogan, D., Hilsenbeck, S. G., Sharkey, F., and Allred, D. C. (1993). p53 is mutated in a subset of advanced-stage prostate cancers. Cancer Res. 53, 3369–3373.
| 8324747PubMed |

Blank, N., Schiller, M., Krienke, S., Wabnitz, G., Ho, A. D., and Lorenz, H.-M. (2007). Cholera toxin binds to lipid rafts but has a limited specificity for ganglioside GM1. Immunol. Cell Biol. 85, 378–382.
Cholera toxin binds to lipid rafts but has a limited specificity for ganglioside GM1.Crossref | GoogleScholarGoogle Scholar | 17325693PubMed |

Farina-Lipari, E., Lipari, D., Bellafiore, M., Anzalone, R., Cappello, F., and Valentino, B. (2009). Presence of atrial natriuretic factor in normal and hyperplastic human prostate and its relationship with oxytocin localisation. Eur. J. Histochem. 47, 133–138.
Presence of atrial natriuretic factor in normal and hyperplastic human prostate and its relationship with oxytocin localisation.Crossref | GoogleScholarGoogle Scholar |

Foradori, C. D., Weiser, M. J., and Handa, R. J. (2008). Non-genomic actions of androgens. Front. Neuroendocrinol. 29, 169–181.
Non-genomic actions of androgens.Crossref | GoogleScholarGoogle Scholar | 18093638PubMed |

Frayne, J., and Nicholson, H. D. (1998). Localization of oxytocin receptors in the human and macaque monkey male reproductive tracts: evidence for a physiological role of oxytocin in the male. Mol. Hum. Reprod. 4, 527–532.
Localization of oxytocin receptors in the human and macaque monkey male reproductive tracts: evidence for a physiological role of oxytocin in the male.Crossref | GoogleScholarGoogle Scholar | 9665335PubMed |

French, A. P., Mills, S., Swarup, R., Bennett, M. J., and Pridmore, T. P. (2008). Colocalization of fluorescent markers in confocal microscope images of plant cells. Nat. Protoc. 3, 619–628.
Colocalization of fluorescent markers in confocal microscope images of plant cells.Crossref | GoogleScholarGoogle Scholar | 18388944PubMed |

Gioeli, D., Wunderlich, W., Sebolt-Leopold, J., Bekiranov, S., Wulfkuhle, J. D., Petricoin, E. F., Conaway, M., and Weber, M. J. (2011). Compensatory pathways induced by MEK inhibition are effective drug targets for combination therapy against castration-resistant prostate cancer. Mol. Cancer Ther. 10, 1581–1590.
Compensatory pathways induced by MEK inhibition are effective drug targets for combination therapy against castration-resistant prostate cancer.Crossref | GoogleScholarGoogle Scholar | 21712477PubMed |

Gould, M. L., Williams, G., and Nicholson, H. D. (2010). Changes in caveolae, caveolin, and polymerase 1 and transcript release factor (PTRF) expression in prostate cancer progression. Prostate 70, 1609–1621.
Changes in caveolae, caveolin, and polymerase 1 and transcript release factor (PTRF) expression in prostate cancer progression.Crossref | GoogleScholarGoogle Scholar | 20564315PubMed |

Guzzi, F., Zanchetta, D., Cassoni, P., Guzzi, V., Francolini, M., Parenti, M., and Chini, B. (2002). Localization of the human oxytocin receptor in caveolin-1 enriched domains turns the receptor-mediated inhibition of cell growth into a proliferative response. Oncogene 21, 1658–1667.
Localization of the human oxytocin receptor in caveolin-1 enriched domains turns the receptor-mediated inhibition of cell growth into a proliferative response.Crossref | GoogleScholarGoogle Scholar | 11896597PubMed |

Han, S., Brenner, J. C., Sabolch, A., Jackson, W., Speers, C., Wilder-Romans, K., Knudsen, K. E., Lawrence, T. S., Chinnaiyan, A. M., and Feng, F. Y. (2013). Targeted radiosensitization of ETS fusion-positive prostate cancer through PARP1 inhibition. Neoplasia 15, 1207–1217.
Targeted radiosensitization of ETS fusion-positive prostate cancer through PARP1 inhibition.Crossref | GoogleScholarGoogle Scholar | 24204199PubMed |

Harris, W. P., Mostaghel, E. A., Nelson, P. S., and Montgomery, B. (2009). Androgen deprivation therapy: progress in understanding mechanisms of resistance and optimizing androgen depletion. Nat. Clin. Pract. Urol. 6, 76–85.
Androgen deprivation therapy: progress in understanding mechanisms of resistance and optimizing androgen depletion.Crossref | GoogleScholarGoogle Scholar | 19198621PubMed |

Hatzoglou, A., Kampa, M., Kogia, C., Charalampopoulos, I., Theodoropoulos, P. A., Anezinis, P., Dambaki, C., Papakonstanti, E. A., Stathopoulos, E. N., Stournaras, C., Gravanis, A., and Castanas, E. (2005). Membrane androgen receptor activation induces apoptotic regression of human prostate cancer cells in vitro and in vivo. J. Clin. Endocrinol. Metab. 90, 893–903.
Membrane androgen receptor activation induces apoptotic regression of human prostate cancer cells in vitro and in vivo.Crossref | GoogleScholarGoogle Scholar | 15585562PubMed |

Hill, M. M., Bastiani, M., Luetterforst, R., Kirkham, M., Kirkham, A., Nixon, S. J., Walser, P., Abankwa, D., Oorschot, V. M., Martin, S., Hancock, J. F., and Parton, R. G. (2008). PTRF-Cavin, a conserved cytoplasmic protein required for caveola formation and function. Cell 132, 113–124.
PTRF-Cavin, a conserved cytoplasmic protein required for caveola formation and function.Crossref | GoogleScholarGoogle Scholar | 18191225PubMed |

Kaighn, M. E., Narayan, K. S., Ohnuki, Y., Lechner, J. F., and Jones, L. W. (1979). Establishment and characterization of a human prostatic carcinoma cell line (PC-3). Invest. Urol. 17, 16–23.
| 447482PubMed |

King, K. J., Nicholson, H. D., and Assinder, S. J. (2006). Effect of increasing ratio of estrogen : androgen on proliferation of normal human prostate stromal and epithelial cells, and the malignant cell line LNCaP. Prostate 66, 105–114.
Effect of increasing ratio of estrogen : androgen on proliferation of normal human prostate stromal and epithelial cells, and the malignant cell line LNCaP.Crossref | GoogleScholarGoogle Scholar | 16114065PubMed |

Kreisberg, J. I., Malik, S. N., Prihoda, T. J., Bedolla, R. G., Troyer, D. A., Kreisberg, S., and Ghosh, P. M. (2004). Phosphorylation of Akt (Ser473) is an excellent predictor of poor clinical outcome in prostate cancer. Cancer Res. 64, 5232–5236.
Phosphorylation of Akt (Ser473) is an excellent predictor of poor clinical outcome in prostate cancer.Crossref | GoogleScholarGoogle Scholar | 15289328PubMed |

Labrie, F., Belanger, A., Dupont, A., Luu-The, V., Simard, J., and Labrie, C. (1993). Science behind total androgen blockade: from gene to combination therapy. Clin. Invest. Med. 16, 475–492.
| 8013153PubMed |

Nicholson, H. D., and Whittington, K. (2007). Oxytocin and the human prostate in health and disease. Int. Rev. Cytol. 263, 253–286.
Oxytocin and the human prostate in health and disease.Crossref | GoogleScholarGoogle Scholar | 17725969PubMed |

Nicholson, H. D., Peeling, W. B., and Pickering, B. T. (1985). Oxytocin in the prostate and semen? J. Endocrinol. 104, 127.

Niu, Y., Altuwaijri, S., Lai, K. P., Wu, C. T., Ricke, W. A., Messing, E. M., Yao, J., Yeh, S., and Chang, C. (2008). Androgen receptor is a tumor suppressor and proliferator in prostate cancer. Proc. Natl Acad. Sci. USA 105, 12182–12187.
Androgen receptor is a tumor suppressor and proliferator in prostate cancer.Crossref | GoogleScholarGoogle Scholar | 18723679PubMed |

Papadopoulou, N., Charalampopoulos, I., Anagnostopoulou, V., Konstantinidis, G., Foller, M., Gravanis, A., Alevizopoulos, K., Lang, F., and Stournaras, C. (2008). Membrane androgen receptor activation triggers down-regulation of PI-3K/Akt/NF-kappaB activity and induces apoptotic responses via Bad, FasL and caspase-3 in DU145 prostate cancer cells. Mol. Cancer 7, 88.
Membrane androgen receptor activation triggers down-regulation of PI-3K/Akt/NF-kappaB activity and induces apoptotic responses via Bad, FasL and caspase-3 in DU145 prostate cancer cells.Crossref | GoogleScholarGoogle Scholar | 19055752PubMed |

Richard, P., Moos, F., and Freund-Mercier, M. J. (1991). Central effects of oxytocin. Physiol. Rev. 71, 331–370.
Central effects of oxytocin.Crossref | GoogleScholarGoogle Scholar | 1672455PubMed |

Rick, F. G., Schally, A. V., Szalontay, L., Block, N. L., Szepeshazi, K., Nadji, M., Zarandi, M., Hohla, F., Buchholz, S., and Seitz, S. (2012). Antagonists of growth hormone-releasing hormone inhibit growth of androgen-independent prostate cancer through inactivation of ERK and Akt kinases. Proc. Natl Acad. Sci. USA 109, 1655–1660.
Antagonists of growth hormone-releasing hormone inhibit growth of androgen-independent prostate cancer through inactivation of ERK and Akt kinases.Crossref | GoogleScholarGoogle Scholar | 22307626PubMed |

Ricote, M., Garcia-Tunon, I., Bethencourt, F., Fraile, B., Onsurbe, P., Paniagua, R., and Royuela, M. (2006). The p38 transduction pathway in prostatic neoplasia. J. Pathol. 208, 401–407.
The p38 transduction pathway in prostatic neoplasia.Crossref | GoogleScholarGoogle Scholar | 16369914PubMed |

Salemi, M., Galia, A., Fraggetta, F., La Corte, C., Pepe, P., La Vignera, S., Improta, G., Bosco, P., and Calogero, A. E. (2013). Poly (ADP-ribose) polymerase 1 protein expression in normal and neoplastic prostatic tissue. Eur. J. Histochem. 57, e13.
Poly (ADP-ribose) polymerase 1 protein expression in normal and neoplastic prostatic tissue.Crossref | GoogleScholarGoogle Scholar | 23807292PubMed |

Smith, A. J., Karpova, Y., D’Agostino, R., Willingham, M., and Kulik, G. (2009). Expression of the Bcl-2 protein BAD promotes prostate cancer growth. PLoS One 4, e6224.
Expression of the Bcl-2 protein BAD promotes prostate cancer growth.Crossref | GoogleScholarGoogle Scholar | 19593445PubMed |

Söderberg, O., Gullberg, M., Jarvius, M., Ridderstråle, K., Leuchowius, K.-J., Jarvius, J., Wester, K., Hydbring, P., Bahram, F., Larsson, L.-G., and Landegren, U. (2006). Direct observation of individual endogenous protein complexes in situ by proximity ligation. Nat. Methods 3, 995–1000.
Direct observation of individual endogenous protein complexes in situ by proximity ligation.Crossref | GoogleScholarGoogle Scholar | 17072308PubMed |

Tikhomirov, O., and Carpenter, G. (2004). Ligand-induced, p38-dependent apoptosis in cells expressing high levels of epidermal growth factor receptor and ErbB-2. J. Biol. Chem. 279, 12988–12996.
Ligand-induced, p38-dependent apoptosis in cells expressing high levels of epidermal growth factor receptor and ErbB-2.Crossref | GoogleScholarGoogle Scholar | 14711810PubMed |

Whittington, K., Assinder, S. J., Parkinson, T., Lapwood, K. R., and Nicholson, H. D. (2001). Function and localization of oxytocin receptors in the reproductive tissue of rams. Reproduction 122, 317–325.
Function and localization of oxytocin receptors in the reproductive tissue of rams.Crossref | GoogleScholarGoogle Scholar | 11467983PubMed |

Whittington, K., Assinder, S., Gould, M., and Nicholson, H. (2004). Oxytocin, oxytocin-associated neurophysin and the oxytocin receptor in the human prostate. Cell Tissue Res. 318, 375–382.
Oxytocin, oxytocin-associated neurophysin and the oxytocin receptor in the human prostate.Crossref | GoogleScholarGoogle Scholar | 15459766PubMed |

Whittington, K., Connors, B., King, K., Assinder, S., Hogarth, K., and Nicholson, H. (2007). The effect of oxytocin on cell proliferation in the human prostate is modulated by gonadal steroids: implications for benign prostatic hyperplasia and carcinoma of the prostate. Prostate 67, 1132–1142.
The effect of oxytocin on cell proliferation in the human prostate is modulated by gonadal steroids: implications for benign prostatic hyperplasia and carcinoma of the prostate.Crossref | GoogleScholarGoogle Scholar | 17492653PubMed |

Wu, Y., Eghbali, M., Ou, J., Lu, R., Toro, L., and Stefani, E. (2010). Quantitative determination of spatial protein–protein correlations in fluorescence confocal microscopy. Biophys. J. 98, 493–504.
Quantitative determination of spatial protein–protein correlations in fluorescence confocal microscopy.Crossref | GoogleScholarGoogle Scholar | 20141764PubMed |

Xu, H., Fu, S., Chen, Q., Gu, M., Zhou, J., Liu, C., Chen, Y., and Wang, Z. (2017a). The function of oxytocin: a potential biomarker for prostate cancer diagnosis and promoter of prostate cancer. Oncotarget 8, 31215–31226.
The function of oxytocin: a potential biomarker for prostate cancer diagnosis and promoter of prostate cancer.Crossref | GoogleScholarGoogle Scholar | 28415720PubMed |

Xu, H., Fu, S., Chen, Y., Chen, Q., Gu, M., Liu, C., Qiao, Z., Zhou, J., and Wang, Z. (2017b). Oxytocin: its role in benign prostatic hyperplasia via the ERK pathway. Clin. Sci. (Lond.) 131, 595–607.
Oxytocin: its role in benign prostatic hyperplasia via the ERK pathway.Crossref | GoogleScholarGoogle Scholar | 28130436PubMed |

Yeh, S., Lin, H. K., Kang, H. Y., Thin, T. H., Lin, M. F., and Chang, C. (1999). From HER2/Neu signal cascade to androgen receptor and its coactivators: a novel pathway by induction of androgen target genes through MAP kinase in prostate cancer cells. Proc. Natl Acad. Sci. USA 96, 5458–5463.
From HER2/Neu signal cascade to androgen receptor and its coactivators: a novel pathway by induction of androgen target genes through MAP kinase in prostate cancer cells.Crossref | GoogleScholarGoogle Scholar | 10318905PubMed |