Estrogenic Potencies of Phytoestrogens Commonly Found in Soybean (Glycine Max) Assessed by use of MMV-Luc Cell Line Bioassay

Authors

  • Peninah Njiraine Ngoda
  • Christopher Elliot
  • Lisa Connolly

DOI:

https://doi.org/10.47672/ajfsn.1354

Keywords:

Phytoestrogens, Gene Assays, Health Effects, Hormones, Transcriptive

Abstract

Purpose: Phytoestrogens (PE) are weakly estrogenic compounds synthesized in plants, especially in climatic adversity. The endocrine disruptive activity of PEs and some of their human metabolites are of concern as they have been associated with positive and more so negative health effects.

Methodology: The transcriptional activities (TAs) of ten PEs (apigenin, daidzein, daidzin, equol, enterodiol, formononetin, genistein, genistin, glycitein, and matairesinol) were measured as induction of the expressed luciferase activity which is correlates with biological effects. These effects were compared to transcription caused by 17β-estradiol (5 nM) hormone (standards) in vitro. This was done by the use of estrogenic receptors in the MMV-Luc cell line using a reporter gene assay tool. 

Findings: Standard curves for hormone standards were determined and the EC50 for 17β-estradiol was (4.5 x 10-11M). The order of PEs potency for the ER (ERα and ERβ)  in the MMV-Luc cell line, as determined from the EC50s (M) of their dose-response curves were in the following order: equol (4.9 x 10-7M) > formononetin (5.7 x 10-6M) > apigenin(2.4 x 10-6M) > genistein (2.1 x 10-6M ) > daidzein(3.3 x 10-5M).  Standard curves for the rest were not achieved in the MMV-Luc cell line at the tested concentrations. The % estrogenic potencies of these compounds varied from 9 x 10-3M for equol to 1.4 x 10-4M for daidzein, relative to 17β-estradiol whose percent potency was arbitrarily assigned a value of 100. This study successfully characterized the TA of ten PEs in MMV-Luc cell lines. By achieving and exceeding a full estrogenic effect in MMV-Luc cells, it was indicative that the phytoestrogen transcriptive activity was most likely through non-receptor mechanisms such as enzymatic pathways.

Recommendations: Although the potential for endocrine disruption by PE was noted in vitro, this study would recommend further in vivo tests. This is because toxicokinetics involving metabolism and bioavailability affects the potencies of the PEs. Further in vitro assays may be designed to investigate the additive effects of phytoestrogens at physiologically relevant concentrations.

Downloads

Download data is not yet available.

Author Biographies

Peninah Njiraine Ngoda

Department of Dairy and Food Science and Technology, Egerton University, Njoro, Kenya.

Christopher Elliot

Institute of Agri-Food and Land Use, Queens University Belfast, UK.    

Lisa Connolly

Institute of Agri-Food and Land Use, Queens University Belfast, UK.    

References

Amer, D. A., Kretzschmar, G., Müller, N., Stanke, N., Lindemann, D., & Vollmer, G. (2010). Activation of transgenic estrogen receptor-beta by selected phytoestrogens in a stably transduced rat serotonergic cell line. The Journal of steroid biochemistry and molecular biology, 120(4-5), 208-217. https://doi.org10.1016/j.jsbnb,2010.04.018

Aronson, K. J. (2016). Meyler's Side Effects of Drugs (Sixteenth Edition). Elsevier, pp. 755-757.https://doi.org/10.1016/B978-0-444-53717-1.00151-7

Auld, D.S., Southall, N.T., Jadhav, A., Johnson, R.L., Diller, D.J., Simeonov, A., Austin, C.P. and Inglese, J. (2008). Characterization of chemical libraries for luciferase inhibitory activity. Journal of Medicinal Chemistry, 51(8), 2372-2386. DOI: 10.1021/jm701302v

Auld, D.S., Thorne, N., Nguyen, D.T. and Inglese, J. (2008). A specific mechanism for nonspecific activation in reporter-gene assays. ACS chemical biology, 3(8), 463-470. DOI: 10.1021/cb8000793

Bakhtiarova, A., Taslimi, P., Elliman, S.J., Kosinski, P.A., Hubbard, B., Kavana, M. and Kemp, D.M. (2006). Resveratrol inhibits firefly luciferase. Biochemical and Biophysical Research Communications, 351(2):481-484. DOI: 10.1016/j.bbrc.2006.10.057

Boscaro, C., Carotti, M., Albiero, M., Trenti, A., Fadini, G. P., Trevisi, L. and Bolego, C. (2020). Non"genomic mechanisms in the estrogen regulation of glycolytic protein levels in endothelial cells. The FASEB Journal, 34(9), 12768-12784. DOI: 10.1096/fj.202001130R

Bovee, H.F.T., Helsdingen, R.J.R., Koks, D.P., Kuiper, A.H., Hoogenboom, P.A.L.R. and Keijer, J. (2004). Development of a rapid yeast estrogen bioassay, based on the expression of a green fluorescent protein. Gene,325,187-200, https://doi.org/10.1016/j.gene.2003.10.015.

Bovee, T.F.H., Lommerse, J.P.M., Peijnenburg, A.A.C.M., Fernandes, E.A. and Nielen, M.W.F. (2008). A new highly androgen-specific yeast biosensor, enabling optimisation of (Q) SAR model approaches. The Journal of steroid biochemistry and molecular biology, 108(1-2), 121-131. https://doi.org/10.1016/j.jsbmb.2007.05.035

Breinholt, V.M., Rasmussen, S.E., Brosen, K. and Friedberg, T.H. (2003). In vitro metabolism of genistein and tangeretin by human and murine cytochrome P450s. Pharmacology &Toxicology, 93(1), 14-22. DOI: 10.1034/j.1600-0773.2003.930102.x

Cai, K., Elliott, T.C., Phillips, H.D., Scippo, M., Muller, M. and Connolly, L. (2012) Treatment of estrogens and androgens in dairy wastewater by a constructed wetland system. Water Research,46(7), 2333-2343. https://doi.org/10.1016/j.watres.2012.01.056

Cai, X., Liu, M., Zhang, B., Zhao, S.J. and Jiang, S.-W. (2021). Phytoestrogens for the Managementof Endometriosis: Findings andIssues. Pharmaceuticals, 14, 1-22.https://doi.org/10.3390/ph14060000

Connolly, L., Cai, K., Van Der Heiden, E., Scippo, M., Muller, M., Tarbin, J. And Elliott, C. (2009). Detection of glucocorticoid bioactivity in bovine urine samples using a reporter gene assay. Analytica Chimica Acta, 637(1-2), 321-327. DOI: 10.1016/j.aca.2008.09.034

Cooper, R. (2019). Investigating the health effects of estrogenic chemicals individually and within wastewater treatment works effluents using an ERE-GFP transgenic zebrafish (Danio rerio) model.University of Exeter, PhD thesis, Devon, South West England

Devin-Leclerc, J., Meng, X., Delahaye, F., Leclerc, P., Baulieu, E.E. and Catelli, M.G. (1998). Interaction and dissociation by ligands of estrogen receptor and Hsp90: the antiestrogen RU 58668 induces a protein synthesis-dependent clustering of the receptor in the cytoplasm. Molecular endocrinology (Baltimore, Md.), 12(6), 842-854. DOI: 10.1002/bio.3091

Du, N., Song, L., Li, Y., Wang, T., Fang, Q., Ou, J. and Nandakumar, K. S. (2020). Phytoestrogens protect joints in collagen-induced arthritis by increasing IgG glycosylation and reducing osteoclast activation. International immunopharmacology, 83, 106387. DOI: 10.1016/j.intimp.2020.106387

Fan, M., Nakshatri, H. and Nephew,K.P. (2004). Inhibiting proteasomal proteolysis sustains estrogen receptor-alpha activation. Molecular endocrinology (Baltimore, Md.),18(11), 2603-2615.DOI: 10.1210/me.2004-0164

Frizzell, C., Ndossi, D., Verhaegen, S., Dahl, E., Eriksen, G., Sørlie, M., Ropstad, E., Muller, M., Elliott, C.T. and Connolly, L. (2011). Endocrine disrupting effects of zearalenone, alpha- and beta-zearalenol at the level of nuclear receptor binding and steroidogenesis. Toxicology letters, 206(2), 210 217. DOI: 10.1016/j.toxlet.2011.07.015

Fujii, S., Ohta, T., Ehama, R., Irikida, M., Nomura, S., Shoyama, Y. and Uto, T. (2023). Development of an indirect competitive enzyme-linked immunosorbent assay for formononetin and its application in a cell-based assay using MC3T3-E1 cells. Food Chemistry, 403, 134339.DOI: 10.1016/j.foodchem.2022.134339

Fujitani, T., Fujii, Y., Lyu, Z., Harada Sassa, M., and Harada, K. H. (2021). Urinary equol levels are positively associated with urinary estradiol excretion in women. Scientific reports, 11(1), 1-10.DOI: 10.1038/s41598-021-98872-2

Griekspoor, A., Zwart, W., Neefjes, J. and Michalides, R. (2007). Visualizing the action of steroid hormone receptors in living cells. Nuclear receptor signaling,5:e003. DOI: 10.1621/nrs.05003

Gronemeyer, H., Gustafsson, J.A. and Laudet, V. (2004). Principles for modulation of the nuclear receptor superfamily. Nature reviews. Drug discovery, 3(11), 950-964. https://doi.org/10.1038/nrd1551

Hall, J. M., Powell, H. R., Rajic, L. and Korach, K. S. (2019). The Role of Dietary Phytoestrogens and the Nuclear Receptor PPAR γ in Adipogenesis: An in vitro study. Environmental health perspectives, 127(3), 037007. DOI: 10.1289/EHP3444

Harris, D.M., Besselink, E., Henning, S.M., Go, V.L. and Heber, D. (2005). Phytoestrogens induce differential estrogen receptor alpha- or Beta-mediated responses in transfected breast cancer cells. Experimental biology and medicine (Maywood, N.J.), 230(8), 558-568. DOI: 10.1177/153537020523000807

Hayashi, K., Yamaguchi, H., Amaoka, H., Takahara, T., Kunisa, S., Tamai, N. and Tanaka, H. (2021). Equol-producing status affects exercise training-induced improvement in arterial compliance in postmenopausal women. Journal of Applied Physiology, 130(3), 827-835.DOI: 10.1152/japplphysiol.00651.2020

Hegazy, M.E., El-Hamd H Mohamed, A., El-Halawany, A.M., Djemgou, P.C., Shahat, A.A. and Pare, P.W. (2011). Estrogenic activity of chemical constituents from Tephrosia candida. Journal of natural products, 74(5), 937-942. DOI: 10.1021/np100378d

Hod, R., Maniam, S., Mohd, Nor. N.H. (2021). A Systematic Review of the Effects of Equol (Soy Metabolite) on Breast Cancer. Molecules; 26 (4):1105. DOI: 10.3390/molecules26041105

Hwang, S. T., Yang, M. H., Baek, S. H., Um, J. Y. and Ahn, K. S. (2020). Genistin attenuates cellular growth and promotes apoptotic cell death in breast cancer cells through modulation of the ERalpha signalling pathway. Life Sciences, 263, 118594.DOI: 10.1016/j.lfs.2020.118594

Jefferson, W.N., Patisaul, H.B. and Williams, C.J. (2012). Reproductive consequences of developmental phytoestrogen exposure. Reproduction;143(3), 247-60. Doi: 10.1530/REP-11-0369

Jin, J., Wu, P., Zhang, X., Li, D., Wong, W. L., Lu, Y. J., Sun, N. and Zhang, K. (2020). Understanding the interaction of estrogenic ligands with estrogen receptors: a survey of the functional and binding kinetic studies. Journal of Environmental Science and Health, Part C, 38(2), 142-168.https://doi=10.1080/26896583.2020.1761204

Jobling, S., (1998).Review of suggested testing methods for endocrine-disrupting chemicals. Pure Applied Chemistry,70(9), 1805-1827.DOI: 10.1351/pac199870091805

Kalita, J.C. and Milligan, S.R. (2010). In vitro estrogenic potency of PE-glycosides and some plant flavonoids. Indian Journal of Science and Technology,12(3), 1142-1147. DOI: 10.17485/ijst/2010/v3i12.10

Kang, S.C., Lee, C.M., Choung, E.S., Bak, J.P., Bae, J.J., Yoo, H.S., Kwak, J.H. and Zee, O.P. (2008). Anti-proliferative effects of estrogen receptor-modulating compounds isolated from Rheum palmatum. Archives of Pharmaceutical Research, 31(6), 722-726. DOI: 10.1007/s12272-001-1218-1

Kładna, A., Berczyński, P., Kruk, I., Piechowska, T., & Aboul"Enein, H. Y. (2016). Studies on the antioxidant properties of some phytoestrogens. Luminescence,31(6),1201-1206. DOI: 10.1002/bio.3091

Ku, Y. S., Ng, M. S., Cheng, S. S., Luk, C. Y., Ludidi, N., Chung, G. and Lam, H. M. (2022). Soybean secondary metabolites and flavours: The art of compromise among climate, natural enemies, and human culture. Advances in botanical research,102, 295-347.https://doi.org/10.1016/bs.abr.2022.03.001

Kuang, W., Hu, W., Ren, H., Shao, Y. and Liu, B. (2021). Plant-derived coumestrol phytochemical targets human skin carcinoma cells by inducing mitochondrial-mediated apoptosis, cell cycle arrest, inhibition of cell migration and invasion and modulation of mTOR/PI3K/AKT signalling pathway. Saudi Journal of Biological Sciences, 28(5), 2739-2746.DOI: 10.1016/j.sjbs.2021.03.043

Kulling, S.E., Honig, D.M. and Metzler, M. (2001). Oxidative metabolism of the soy isoflavones daidzein and genistein in humans in vitro and in vivo. Journal of Agricultural and Food Chemistry, 49(6), 3024-3033. https://doi.org/10.1021/jf0012695

Lampe, J.W., Karr, S.C., Hutchins, A.M. and Slavin, J.L. (1998). Urinary equol excretion with a soy challenge: Influence of Habitual Diet. Proceedings of the Society for Experimental Biology and Medicine; 217(3):335-339. Doi:10.3181/00379727-217-44241

Latif, S., Weston, P. A., Barrow, R. A., Gurusinghe, S., Piltz, J. W. and Weston, L. A. (2020). Metabolic profiling provides unique insights into the accumulation and biosynthesis of key secondary metabolites in annual pasture legumes of Mediterranean origin. Metabolites, 10(7), 267.DOI: 10.3390/metabo10070267

Lehraiki, A., Chamaillard, C., Krust, A., Habert, R. and Levacher, C. (2011). Genistein impairs early testosterone production in fetal mouse testis via estrogen receptor alpha. Toxicology in vitro, 25(8), 1542-1547. https://doi.org/10.1016/j.tiv.2011.05.017

Lehraiki, A., Messiaen, S., Berges, R., Canivenc-Lavier, M.C., Auger, J., Habert, R. and Levacher, C. (2011). Antagonistic effects of gestational dietary exposure to low-dose vinclozolin and genistein on rat fetal germ cell development. Reproductive toxicology (Elmsford, N.Y.), 31(4), 424-430. DOI: 10.1016/j.reprotox.2010.12.005

Li, W. (2015). The Underlying Mechanisms for Resveratrol' Superinduction Effects. International Journal of Health Sciences; 3(1), 39-59. DOI: 10.15640/ijhs.v3n1a3

Liu, T., Li, N., Yan, Y. Q., Liu, Y., Xiong, K., Liu, Y. and Liu, Z. D. (2019). Recent advances in the anti-ageing effects of phytoestrogens on collagen, water content, and oxidative stress. Phytotherapy Research, 2019, 1-13.https://doi.org/10.1002/ptr.6538

Liua, R.,Yua, X.,Chena, X., Zhong, H., Liang, C., Xu, X., Xu, W., Cheng, Y., Wang, W., Yu, L., Wu, Y., Yan, N. and Hu, X. (2019).Individual factors define the overall effects of dietary genistein exposure on breast cancer patients. Nutrition Research, 67, 1-16. DOI: 10.1016/j.nutres.2019.03.015

Ma, Y., Sullivan, J.C.and Schreihofer D.A. (2010). Dietary genistein and equol (4', 7 isoflavandiol) reduce oxidative stress and protect rats against focal cerebral ischemia. American journal of physiology. Regulatory, integrative and comparative physiology; 299(3), R871-7.DOI: 10.1152/ajpregu.00031.2010

Margeat, E., Bourdoncle, A., Margueron, R., Poujol, N., Cavailles, V. and Royer, C. (2003). Ligands differentially modulate the protein interactions of the human estrogen receptors alpha and beta. Journal of Molecular Biology, 326(1), 77-92. DOI: 10.1016/s0022-2836(02)01355-4

Matsumura, A., Ghosh, A., Pope, G.S. and Darbre, P.D. (2005). Comparative study of oestrogenic properties of eight phytoestrogens in MCF7 human breast cancer cells. The Journal of steroid biochemistry and molecular biology, 94(5), 431-443. DOI: 10.1016/j.jsbmb.2004.12.041

Memariani, Z., Gorji, N., Moeini, R. and Farzaei, H.M. (2020). Traditional uses. In S.M. Nabavi, I. Suntar, D. Barreca, and H. Khan (Eds),Phytonutrients in Food from Traditional to Rational Usage (pp. 23-66). Woodhead Publishing Series. https://doi.org/10.1016/B978-0-12-815354-3.00004-6

Montani, C., Penza, M., Jeremic, M., Biasiotto, G., La Sala, G., De Felici, M., ... & Di Lorenzo, D. (2008). Genistein is an efficient estrogen in the whole-body throughout mouse development. Toxicological sciences, 103(1), 57-67. https://doi.org/10.1093/toxsci/kfn021

Mostrom, M. and Evans, J.T. (2018). Phytoestrogens. In R.C. Gupta (Ed), Veterinary Toxicology: Basic and Clinical Principles (pp. 817-833). Academic Press. https://doi.org/10.1016/B978-0-12-811410-0.00060-X

Mousavi, Y. and Adlercreutz, H. (1992). Enterolactone and estradiol inhibit each other's proliferative effect on MCF-7 breast cancer cells in culture. The Journal of steroid biochemistry and molecular biology, 41(3-8), 615-619. DOI: 10.1016/0960-0760(92)90393-w

Mueller, S.O., Simon, S., Chae, K., Metzler, M. and Korach, K.S. (2004). Phytoestrogens and their humanmetabolites show distinct agonistic and antagonistic properties on estrogen receptor alpha(ERalpha) and ERbeta in human cells. Toxicological Sciences; 80, 14-25.DOI: 10.1093/toxsci/kfh147

Nagel, S.C., Vom Saal, F.S. and Welshons, W.V. (1998). The effective free fraction of estradiol and xenoestrogens in human serum measured by whole-cell uptake assays: physiology of delivery modifies estrogenic activity. Proceedings of the Society for Experimental Biology and Medicine. Society for Experimental Biology and Medicine (New York, N.Y.), 217(3), 300-309. DOI: 10.3181/00379727-217-44236

Nguyen, M. and Osipo, C. (2022). Targeting breast cancer stem cells using naturally occurring phytoestrogens. International Journal of Molecular Sciences, 23(12), 6813.DOI: 10.3390/ijms23126813

Ni, Y.D., Hong, W.J., Zhou, Y.C., Grossmann, R. and Zhao, R.Q. (2010). Dual effects of daidzein on chicken hepatic vitellogenin II expression and estrogen receptor-mediated transactivation in vitro. Steroids, 75(3), 245-251. DOI: 10.1016/j.steroids.2009.12.009

Norris, J.D., Paige, L.A., Christensen, D.J., Chang, C.Y., Huacani, M.R., Fan, D., Hamilton, P.T., Fowlkes, D.M. and Mcdonnell, D.P. (1999). Peptide antagonists of the human estrogen receptor. Science (New York, N.Y.),285(5428), 744-746. DOI: 10.1126/science.285.5428.744

Omidvari, M., Flematti, G. R., You, M. P., Abbaszadeh"Dahaji, P. and Barbetti, M. J. (2022a). Phoma black stem severity and phytoestrogen production in annual Medicago spp. is primarily determined by the interaction of cultivar and pathogen isolated. Plant Pathology, 71(4), 860-872. DOI: 10.1111/ppa.13533

Omidvari, M., Flematti, R.G., You, P.M., Abbaszadeh-Dahaji, P. and Barbetti, J.M. (2022b).Sequential infections by 32 isolates of Phoma medicaginisincrease the production ofphytoestrogens in Medicago polymorphavar. brevispina.Crop & Pasture Science,1367-1384. https://doi.org/10.1071/CP22098

Onoda, A., Ueno, T., Uchiyama, S., Hayashi, S., Kato, K. and Wake, N. (2011). Effects of S-equol and natural S-eguol supplement (SE5-OH) on the growth of MCF-7 in vitro and as tumours implanted into ovariectomized athymic mice. Food and chemical toxicology: an international journal published by British Industrial Biological Research Association, 49(9), 2279-2284. DOI: 10.1016/j.fct.2011.06.027

Palanza, P., Nagel, S. C., Parmigiani, S., & vom Saal, F. S. (2016). Perinatal exposure to endocrine disruptors: sex, timing and behavioural endpoints. Current Opinion in Behavioral Sciences, 7, 69-75.https://doi.org/10.1016/j.cobeha.2015.11.017

Pickering, A.D. and Sumpter, J.P. (2003). Comprehending endocrine disrupters in aquatic environments. Environmental science & technology, 37(17), 331A-336A.

Pohjanvirta, R. and Nasri, A. (2022). The Potent Phytoestrogen8-Prenylnaringenin: A Friend or aFoe? Int. J. Mol. Sci., (23, 3168), 1-30.https://doi.org/10.3390/ijms23063168

Razandi, M., Pedram, A., Merchenthaler, I., Greene, G.L. and Levin, E.R. (2004). Plasma membrane estrogen receptors exist and function as dimers. Molecular endocrinology (Baltimore, Md.), 18(12), 2854-2865. DOI: 10.1210/me.2004-0115

Reiter, E., Gerster, P. and Jungbauer, A. (2011). Red clover and soy isoflavones--an in vitro safety assessment. Gynaecological endocrinology: the official journal of the International Society of Gynecological Endocrinology, 27(12), 1037-1042.DOI: 10.3109/09513590.2011.588743

Resende, F.A., de Oliveira, A.P.S., de Camargo, M.S., Vilegas, W. and Varanda, E.A. (2013) Evaluation of Estrogenic Potential of Flavonoids Using a Recombinant Yeast

Strain and MCF7/BUS Cell Proliferation Assay. PLoS ONE, 8(10), 1-7. DOI: 10.1371/ journal.pone.0074881

Richards, J.S. (2001). New signalling pathways for hormones and cyclic adenosine 3', 5'-monophosphate action in endocrine cells. Molecular endocrinology (Baltimore, Md.), 15(2), 209-218. DOI: 10.1210/mend.15.2.0606

Rietjens, I.M.C.M., Louisse, J. and Beekmann, K. (2017). The potential health effects of dietary phytoestrogens. British Journal of Pharmacology;174(11):1263-1280. DOI: 10.1111/bph.13622

Rowe, I. J. and Baber, R. J. (2021). The effects of phytoestrogens on postmenopausal health. Climacteric, 24(1), 57-63.DOI: 10.1080/13697137.2020.1863356

Ruff, M., Gangloff, M., Wurtz, J.M. and Moras, D. (2000). Estrogen receptor transcription and transactivation: Structure-function relationship in DNA- and ligand-binding domains of estrogen receptors. Breast cancer research: BCR, 2(5), 353-359. DOI: 10.1186/bcr80

Russo, M., Russo, G. L., Daglia, M., Kasi, P. D., Ravi, S., Nabavi, S. F. and Nabavi, S. M. (2016). Understanding genistein in cancer: The "good" and the "bad" effects: A review. Food Chemistry, 196, 589-600.DOI: 10.1016/j.foodchem.2015.09.085

Schaufele, F., Chang, C.Y., Liu, W., Baxter, J.D., Nordeen, S.K., Wan, Y., Day, R.N. and Mcdonnell, D.P. (2000). Temporally distinct and ligand-specific recruitment of nuclear-receptor-interacting peptides and cofactors to subnuclear domains containing the estrogen receptor. Molecular endocrinology (Baltimore, Md.), 14(12), 2024-2039.DOI: 10.1210/mend.14.12.0572

Sharma, A., Jyoti, N. D. and Saxena, S. (2014). Health benefits of phytoestrogens. International Journalof Advanced Research, 2(4), 1024-1030.

Sim, K. S., Park, S., Seo, H., Lee, S. H., Lee, H. S., Park, Y. and Kim, J. H. (2022). Comparative study of estrogenic activities of phytoestrogens using OECD in vitro and in vivo testing methods. Toxicology and Applied Pharmacology, 434, 115815. DOI: 10.1016/j.taap.2021.115815

Skledar, D. G., Tvrdý, V., Kenda, M., Zega, A., Pour, M., Horký, P. and MaÅ¡iÄ, L. P. (2020). Applicability of the OECD 455 in-vitro assay for determination of hERa agonistic activity of isoflavonoids. Toxicology and Applied Pharmacology, 386, 114831. DOI: 10.1016/j.taap.2019.114831

Sotoca, A.M., Bovee, T.F.H., Brand, W., Velikova, N., Boeren, S., Murk, A.J., Vervoort, J. and Rietjens, I.M.C.M. (2010). Superinduction of estrogen receptor-mediated gene expression in luciferase-based reporter gene assays is mediated by a post-transcriptional mechanism. Journal of Steroid Biochemistry and Molecular Biology, 122(4), 204-211. DOI: 10.1016/j.jsbmb.2010.06.010

Sun, M., Zhou, T., Zhou, L., Chen, Q., Yu, Y., Yang, H., Zhong, K., Zhang, X., Xu, F., Cai, S., Yu, A., Zhang, H., Xiao, R., Xiao, D. and Chui, D. (2012). Formononetin protects neurons against hypoxia-induced cytotoxicity through the upregulation of ADAM10 and sAbetaPPalpha. Journal of Alzheimer's disease: JAD, 28(4), 795-808. DOI: 10.3233/JAD-2011-110506

Suo, F., Zhou, X., Setroikromo, R. and Quax, W. J. (2022). Receptor Specificity Engineering ,;/l,m,mjof TNF Superfamily Ligands. Pharmaceutics, 14(1), 181.DOI: 10.3390/pharmaceutics14010181

Sweeney, M. F., Hasan, N., Soto, A. M. and Sonnenschein, C. (2016). Environmental endocrine disruptors: Effects on the human male reproductive system. Reviews in Endocrine and Metabolic Disorders,16(4):341-57.DOI: 10.1007/s11154-016-9337-4

Takeuchi, S., Takahashi, T., Sawada, Y., Iida, M., Matsuda, T. and Kojima, H. (2009). Comparative study on the nuclear hormone receptor activity of various phytochemicals and their metabolites by reporter gene assays using Chinese hamster ovary cells. Biological & pharmaceutical bulletin, 32(2), 195-202. DOI: 10.1248/bpb.32.195

Tanwar, A. K., Dhiman, N., Kumar, A. and Jaitak, V. (2021). Engagement of phytoestrogens in breast cancer suppression: Structural classification and mechanistic approach. European Journal of Medicinal Chemistry, 213, 113037. DOI: 10.1016/j.ejmech.2020.113037

Thangavel, P., Puga-Olguín, A., Rodríguez-Landa, J. F. and Zepeda, R. C. (2019). Genistein as potential therapeutic candidate for menopausal symptoms and other related diseases. Molecules, 24(21), 3892.DOI: 10.3390/molecules24213892

Thompson, J.F., Geoghegan, K.F., Lloyd, D.B., Lanzetti, A.J., Magyar, R.A., Anderson, S.M. and Branchini, B.R. (1997). Mutation of a protease-sensitive region in firefly luciferase alters light emission properties. The Journal of biological chemistry, 272(30), 18766-18771. DOI: 10.1074/jbc.272.30.18766

Van Duursen, M.B., Nijmeijer, S.M., De Morree, E.S., De Jong, P.C. and Van Den Berg, M. (2011). Genistein induces breast cancer-associated aromatase and stimulates estrogen-dependent tumour cell growth in in vitro breast cancer models. Toxicology, 289(2-3), 67-73. DOI: 10.1016/j.tox.2011.07.005

Vasantha Padmanabhan, V., Jacob Moeller, J. and Muraly Puttabyatappa, M. (2021). Impact of gestational exposure to endocrine disrupting chemicals on pregnancy and birth outcomes. In L. N. Vandenberg, and J.L. Turgeon (Eds), Advances in Pharmacology (pp. 279-346).Academic Press.https://doi.org/10.1016/bs.apha.2021.04.004

Wang, L. H., Chen, L. R. and Chen, K. H. (2021). In vitro and vivo identification, metabolism and action of xenoestrogens: An overview. International Journal of Molecular Sciences, 22(8), 4013.DOI: 10.3390/ijms22084013

Willemsen, P., Scippo, M.L., Kausel, G., Figueroa, J., Maghuin-Rogister, G., Martial, J.A. and Muller, M. (2004). Use of reporter cell lines for detection of endocrine-disrupter activity. Analytical and bioanalytical chemistry, 378(3), 655-663. DOI: 10.1007/s00216-003-2217-2

Woods, H. F. (2003). Committee on Toxicity of Chemicals in Food, Consumer Products and the Environment. In I. Hughes (Ed), Phytoestrogens and Health (pp.103-120). Crown, London.

Wu, J., Li, J., Liu, Y., Liao, X., Wu, D., Chen, Y. and Wen, L. (2021). Tannic acid repair of zearalenone-induced damage by regulating the death receptor and mitochondrial apoptosis signalling pathway in mice. Environmental Pollution, 287, 117557.DOI: 10.1016/j.envpol.2021.117557

Wu, Z. Y., Sang, L. X. and Chang, B. (2020). Isoflavones and inflammatory bowel disease. World Journal of Clinical Cases, 8(11), 2081.DOI: 10.12998/wjcc.v8.i11.2081

Yang, J., Wen, L., Jiang, Y. and Yang, B. (2019). Natural estrogen receptor modulators and their heterologous biosynthesis. Trends in Endocrinology & Metabolism, 30(1), 66-76.DOI: 10.1016/j.tem.2018.11.002

Downloads

Published

2023-02-17

How to Cite

Ngoda, P. N. ., Elliot, C. ., & Connolly, L. . (2023). Estrogenic Potencies of Phytoestrogens Commonly Found in Soybean (Glycine Max) Assessed by use of MMV-Luc Cell Line Bioassay. American Journal of Food Sciences and Nutrition, 5(1), 1–22. https://doi.org/10.47672/ajfsn.1354