Endocrine Disruptor—A threat to the animal world

  • Diptatanu Das Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, Nadia, West Bengal, India
  • Shantanu Das Department of Zoology, Rammohan College, 102/1 Raja Rammohan Sarani, Kolkata-700009, West Bengal, India
Keywords: Ecology, endocrine disruptor, human health, phytoestrogen, xenoestrogen

Abstract

Various types of naturally occurring and artificially made chemicals cause disruption of endocrine processes among animals. They mimic biochemically with hormones and interfere with the normal signaling and activity of the endocrine system, causing enormous changes at the cellular level of animals from lower to higher organisms, including human being. These modified regulations of cellular activities as a result of endocrine disruptors have severe implications at the organismal level. Types and adverse effects of these natural and synthetic agents, especially estrogenic compounds causing biological threats have been discussed in details in this review.

References

Adeel, M., Songa, X., Wang, Y., Francis, D. and Yang, Y. (2017). Environmental impact of estrogens on human, animal and plant life: A critical review. Environ. Int. 99: 107-119.

Adlercreutz, H., van der Wildt, J., Kinzel, J., Attalla, H., Wähäla, K., Mäkelä, T., Hase, T. and Fotsis, T. (1995). Lignan and isoflavonoid conjugates in human urine. J. Steroid. Biochem. Mol. Biol. 52 (1): 97-103.

Baker, L., Meldrum, K. K., Wang, M., Sankula, R., Vanam, R., Raiesdana, A., Tsai, B., Hile, K., Brown, J. W. and Meldrum, D. R. (2003).The role of estrogen in cardiovascular disease. J. Surg. Res. 115 (2): 325-344.

Bingham, S.A., Atkinson, C., Liggins, J., Bluck, L. and Coward, A. (1998). Phyto-oestrogens: where are we now? Brit. J. Nutri. 79: 393-406.

Bless, E. P., Yang, J., Acharya, K. D., Nettles, S. A., Vassoler, F. M., Byrnes, E. and Tetel, M. J. (2016). Adult Neurogenesis in the Female Mouse Hypothalamus: Estradiol and High Fat Diet Alter the Generation of Newborn Neurons Expressing Estrogen Receptor α. eNeuro. 3 (4).

Bukowska, Spiller, Wolke, Lendeckel, Weinert, Hoffmann, Bornfleth, Kutschka, Gardemann, Isermann, Goette, A., L., C., U., S., J., P., I., A., B. and A. (2017). Protective regulation of the ACE2/ACE gene expression by estrogen in human atrial tissue from elderly men. Exp. Biol. Med. 242 (14): 1412-1423.

Buteau-Lozano, H., Velasco, G., Cristofari, M., Balaguer, P. and Perrot-Applanat, M. (2008). Xenoestrogens modulate vascular endothelial growth factor secretion in breast cancer cells through an estrogen receptor-dependent mechanism. J. Endocrinol. 196 (2): 399-412.

Carmeliet, P. (2005). VEGF as a Key Mediator of Angiogenesis in Cancer. Oncology. 69(3): 4-10.

Carson, R. (1962). In: Silent spring, Fawcett Publication inc., Greenwich Conn., USA. Cheng, A.S.L., Culhane, A. C., Chan, M.W.Y., Venkataramu, C.R., Ehrich, M., Nasir, A., Rodriguez, B. A. T., Liu, J., Yan, P. S., Quackenbush, J., Nephew, K. P., Yeatman, T. J. and Huang, T. H. M. (2008). Epithelial Progeny of Estrogen-Exposed Breast Progenitor Cells Display a Cancer-like Methylome. Cancer. Res. 68 (6): 1786-1796.

Chia, S. E. (2000). Endocrine disruptors and male reproductive function-a short review. Inter. J. Andro. 23: 45-46.

Clark, L.B., Rosen, R.T., Hartman, T.G., Louis, J. B. and Suffet, I. H. (1992). Determination of alkylphenol ethoxylates and their acetic acid derivatives in drinking water by particle beam liquid chromato-graphy /mass specto-metry. Inter. J. Environ. Analyt. Chem. 47: 167-180.

Colborn, T. and Clement, C. (1992). Chemically-induced Alterations in Sexual and Functional Develop-ment: The Wildlife / Human Connection. In: Environmental Health Perspective, Princeton Scientific Publishing Company. Princeton.

Colborn, T., vomSaal, F. S. and Soto, A. M. (1993). Developmental effects of endocrine-disrupting chemicals in wildlife and humans. In: Environmental Health Perspective. 101: 378-384.

Costet, N., Pelé, F., Comets, E., Rouget, F., Monfort, C., Bodeau-Livinec, F., Linganiza, E.M., Bataille, H., Kadhel, P. and Multigner, L. (2015). Perinatal exposure to chlordecone and infant growth. Environ. Res. 142: 123-134.

Crackower, M. A., Sarao, R., Oudit, G.Y., Yagil, C., Kozieradzki, I., Scanga, S.E., Oliveira-dos-Santos, A.J., Costa, J. da., Zhang, L., Scholey, Y. P. and Ferrario, C.M. (2002). Angiotensin-converting enzyme 2 is an essential regulator of heart function. Nature. 417: 822-828.

Crews, D. and McLachlan, J. A. (2006). Epigenetics, Evolution, Endocrine Disruption, Health, and Disease. Endocrinology. 147 (6): s4-s10.

Crews, D., Willingham, E. and Skipper, J. K. (2000). Endocrine Disruptors: Present Issues, Future Directions. Quart. Rev. Biol. 75 (3): 243-260.

Cullinan, M.P., Palmer, J.E., Carle, A.D., West, M.J., Westerman, B. and Seymour, G.J. (2015). The influence of a triclosan toothpaste on adverse events in patients with cardiovascular disease over 5-years. Sci. Total. Env. 508: 546-552.

Dahlman-Wright, K., Cavailles, V., Fuqua, S. A., Jordan, V. C., Katzenellenbogen, J. A., Korach, K. S., Maggi, A., Muramatsu, M., Parker, M. G. and Gustafsson, J. A. (2006). International Union of Pharmacology. LXIV. Estrogen Receptors. Pharmacol. Rev. 58 (4): 773-781.

Das, S. and Ray, A.K. (2014). Possible alteration of female specific protein by estradiol-17β in silkworm, Bombyx mori L. Proc. Zool. Soc. 67: 38-42.

Das, S. (2016). Vertebrate hormones in insects: the role of estrogen in silkworm- a review. Turk. J. Zool. 40: 297-302.

Das, S. and Ray, A.K. (2007). Possible involvement of estradiol-17β on steroid metabolizing enzymes in Bombyx mori L. Proc. Zool. Soc.60: 11-17.

De, J. (2007). Search for biological responses of estradiol-17β in pupal life of silkworm, Bombyx mori L. (race Nistari) Ph. D. Jadavpur University, Kolkata, India.

Folmar, L. C., Denslow, N. D., Rao, V., Chow, M., Crain, D. A., Enblom, J., Marcino, J. and Guillette, Jr L. J. (1996). Vitellogenin induction and reduced serum testosterone concentrations in feral male carp (Cyprinuscarpio) captured near a major metropolitan sewage treatment plant. Environ. Heal. Pers. 104(10): 1096-1101.

Fowler, C.D., Liu, Y. and Wang, Z. (2008). Estrogen and adult neurogenesis in the amygdala and hypothalamus. Brain. Res. Rev. 57 (2): 342-351.

Gaylor, D.W., Sheehan, D.M., Young, J. F. and Mattison, D.R. (1988). The threshold dose questions in teratogenesis. Teratology. 38 : 389-391.

Goodman, J. E. and Peterson, M. K. (2014). Bisphenol A. In: Wexler P, editor. Encyclopedia of Toxicology (Third Edition), Academic Press. Pp. 514-518.

Gore, A. C., Crews, D., Doan, L. L., Merrill, M. L., Patisaul, H. and Zota, A. (2014). Introduction to EDCs. A Guide for Public Interest Organizations and Policy-Makers, Strategic Approach to International Chemicals Management (SAICM), IPEN and The Endocrine Society.

Green, S. and Simpkins, J.W. (2000). Neuroprotective effects of estrogens: potential mechanisms of action. Inter. J. Devel. Neuro. 18 (4-5): 347-358.

Green, S., Walter, P., Kumar, V., Krust, A., Bornert, J. M., Argos, P. and Chambon, P. (1986). Human oestrogen receptor cDNA: sequence, expression and homology to v-erb-A. Nature. 320: 134-139.

Greene, G.L., Gilna, P., Waterfield, M., Baker, A. and Hort, Y. (1986). Sequence and expression of human estrogen receptor complementary DNA. Science. 231: 1150-1154.

Grossman, A.W., Churchill, J.D., McKinney, B.C., Kodish, I.M., Otte, S.L. and Greenough, W.T. (2003). Experience effects on brain development: possible contributions to psychopathology. J. Child. Psycho. Psychi. Alli. Dis. 44 (1): 33-63.

Hoffmann, M., Kleine-Weber, H., Schroeder, S., Krüger, N., Herrler, T., Erichsen, S., Schiergens, T.S., Herrler, G., Wu, N.H., Nitsche, A., Müller, M.A., Drosten, C. and Pöhlmann, S. (2020). SARS-CoV-2cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 181: 271-280.

Hsu, P. Y., Deatherage, D. E., Rodriguez, B. A.T., Liyanarachchi, S., Weng, Y. I., Zuo, T., Liu, J., Cheng, A.S.L. and Huang, T. H. M. (2009). Xenoestrogen-Induced Epigenetic Repression of microRNA-9-3 in Breast Epithelial Cells. Cancer Res. 69 (14): 5936-5945.

Huang, L., Cheng, Y., Huang, K., Zhou, Y., Ma, Y.and Zhang, M. (2018). Ameliorative effect of Sedum sarmentosum Bunge extract on Tilapia fatty liver via the PPAR and P53 signaling pathway. Scientific Reports. 8: 8456.

Huang, T.H.M. and Esteller, M. (2010). Chromatin Remodelling in Mammary Gland Differentiation and Breast Tumorigenesis. Cold Spring Harbor Persp. Biol. 2 (9): a004515.

Huber, W.W., Grasl-Kraupp, B. and Schulte-Hermann, R. (1996). Hepatocarcinogenic potential of di(2-ethylhexyl) phthalate in rodents and its implications on human risk. Critical. Rev. Toxicol. 26: 365-481.

Hutz, R. J., Carvan, M. J., Larson, J. K., Liu, Q., Stelzer, R. V., King-Heiden, T. C., Baldridge, M. G., Shahnoor, N. and Julien, K. (2014). Familiar and novel reproductive endocrine disruptors: xenoestrogens, dioxins and nanoparticles. Cur. Tren. Endocrinol. 7: 111-122.

Imai, Y.Kuba, K.Rao, S.Huan, Y.Guo, F.Guan, B.Yang, P.Sarao, R.Wada, T.Leong, P. H.Crackower, M. A.Fukamizu, A.Hui, C. C.Hein, L., Uhlig, S.Slutsky, A. S., Jiang, C. and Penninger, J. M. (2005). Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature. 436 (7047): 112-116.

Janesick, A. and Blumberg, B. (2011). Mini review: PPAR gamma as the target of obesogens. J. Steroid. Biochem. Mol. Biol. 127: 4-8.

Jensen, E. V. and Jordan, V. C. (2003). The estrogen receptor: a model for molecular medicine. Clin. Can. Res.9: 1980-1989.

Keshan, B. and Ray, A.K. (2001). The presence of estradiol-17β and its specific binding sites in posterior silk gland of Bombyx mori. Gen. Com. Endocrinol.123: 23-30.

Kirchner, S., Kieu, T., Chow, C., Casey, S. and Blumberg, B. (2010). Prenatal exposure to the environmental obesogen tributyltin predisposes multipotent stem cells to become adipocytes. Mol. Endocrinol. 24: 526-539.

Kuhl, A. J., Manning, S. and Brouwer, M. (2005). Brain aromatase in Japanese medaka (Oryziaslatipes): Molecular characterization and role in xenoestrogen-induced sex reversal. J. Steroid. Biochem. Mol. Biol. 96 (1): 67-77.

Kuiper, G. G., Enmark, E., Pelto-Huikko, M., Nilsson, S. and Gustafsson, J. A. (1996). Cloning of a novel receptor expressed in rat prostate and ovary. Proc. Nat. Acad. Sci. 93 (12): 5925-5930.

Kyle, D., Rebecca, L., Hoagland, S. and Siegfried, B.D. (1996). Effects of organic toxic substances. In: Stevenson, R.J., Bothwell, M. L., Lowe, R.L. editors. Algal Ecology, Academic Press.

Lazarov, O. and Hollands, C. (2016). Hippocampal neurogenesis: Learning to remember. Prog. Neurobiol. 138-140: 1-18.

Lemaire, G., Mnif, W. and Mauvais, P. (2006). Activation of alpha- and beta-estrogen receptors by persistent pesticides in reporter cell lines. Life. Sci. 79:1160-1169.

Li, X., Gao, Y., Guo, L.H. and Jiang, G. (2013). Structure-dependent activities of hydroxylated polybrominated diphenyl ethers on human estrogen receptor. Toxicology. 309 (2): 15-22.

Li, Y., Burns, K.A., Arao, Y., Luh, C.J. andKorach, K.S. (2012). Differential estrogenic actions of endocrine-disrupting chemicals bisphenol A, bisphenol AF, and zearalenone through estrogen receptor alpha and beta in vitro. Environ. Heal. Persp. 120: 1029-1035.

Liggins, J., Bluck, L. J., Runswick, S., Atkinson, C., Coward, W. A. and Bingham, S. A. (2000). Daidzein and genistein contents of vegetables. Brit. J. Nutri. 84: 717-25.

Manavathi, B. and Kumar, R. (2006). Steering estrogen signals from the plasma membrane to the nucleus: two sides of the coin. J. Cell. Physiol. 207: 594-604.

Moore, S. C., Matthews, C. E., Shu, X. O., Yu, K., Gail, M. H., Xu, X., Ji, B. T., Chow, W. R., Cai, Q., Li, H., Yang, G., Ruggieri, D., Boyd-Morin, J., Rothman, N., Hoover, R. N., Gao, Y. T., Zheng, W. and Ziegler, R. G. (2016). Endogenous Estrogens, Estrogen Metabolites and Breast Cancer Risk in Postmenopausal Chinese Women. J. Nat. Can. Inst. 108 (10): djw103.

Moral, R., Santucci-Pereira, J., Wang, R., Russo, I.H., Lamartiniere, C. A. and Russo, J. (2011). In utero exposure to butyl benzyl phthalate induces modifications in the morphology and the gene expression profile of the mammary gland: an experimental study in rats. Environ. Health. 10 (1): 5.

Murphy, E. and Kelly, D.P. (2011). EstrogenSignaling and Cardiovascular Disease. Circu. Res.109 (6): 687-696.

Nelles, J. L., Hu, W. Y. and Prins, G. S. (2011). Estrogen action and prostate cancer. Exp. Rev. Endocrinol. Metab. 6 (3): 437-451.

Obiorah, I., Sengupta, S. and Curpan, R. (2014). Defining the conformation of the estrogen receptor complex that controls estrogen-induced apoptosis in breast cancer. Mol. Pharmacol. 85 (5): 789-799.

Ohnishi, E., Ogiso, M., Wakabayashi, K., Fujimoto, Y. and Ikekawa, N. (1985). Identification of estradiol in ovaries of the silkworm, Bombyx mori. Gen. Com. Endocrinol. 60: 35-38.

Paterni, I., Granchi, C. and Minutolo, F. (2017). Risks and benefits related to alimentary exposure to xenoestrogens. Crit. Rev. Food. Sci. Nutri. 57 (16): 3384-3404.

PenzaMontaniRomaniVignoliniPampaloniTaniniBrandiNadalOttobriniParoliniBignottiCalzaMaggiGrigolatoDi Lorenzo, M., C., , A.,, P., B., A., M. L., Alonso, M. P., A., L., O., E., S., A., P.G. and D. (2006). Genistein affects adipose tissue deposition in a dose-dependent and gender-specific manner. Endocrinology. 147: 5740-5751.

Penza, M., Montani, C., Romani, A., Vignolini, P., Pampaloni, B., Tanini, A., Brandi, M. L., Alonso, M.P., Nadal, A., Ottobrini, L., Parolini, O., Bignotti, E., Calza, S., Maggi, A., Grigolato, P.G. and Di Lorenzo, D. (2006). Genistein affects adipose tissue deposition in a dose-dependent and gender-specific manner. Endocrinology. 147: 5740-5751.

Purdom, C. E., Hardiman, P. A., Bye, V. V. J., Eno, N. C., Tyler, C. R., Sumpter, J. P. (1994). Estrogenic Effects of Effluents from Sewage Treatment Works. Chem. Ecol. 8 (4): 275-285.

Rathee, M., Malik, P. and Singh, J. (2012). Bisphenol A in dental sealants and its estrogen like effect. Ind. J. Endocrinol. Meta. 16 (3): 339-342.

Roeder, R. A., Garber, M. J. and Schelling, G.T. (1998). Assessment of dioxins in foods from animal origins. J. Ani. Sci. 76 (1): 142-151.

Rose, J., Holbech, H., Lindholst, C., Norum, U., Povlsen, A., Korsgaard, B. and Bjerregaard, P. (2002). Vitellogenin induction by 17beta-estradiol and 17alpha-ethinylestradiol in male zebra fish (Daniorerio). Com. Biochem. Physiol. Part 3Toxicol. Pharmacol. 131 (4): 531-539.

Roy, S. (2007). Subcellular action of estradiol-17β in fifth instar larvae of silkworm, Bombyx mori L. (race Nistari). Ph.D., Jadavpur University, Kolkata, India.

Roy, D., Palangat, M., Chen, C.W., Thomas, R. T., Colerangle, J. C., Atkinson, A. and Yan, Z. J. (1997). Biochemical and molecular changes at the cellular levels in response to exposure of environmental estrogen-like chemicals. J. Toxicol. Environ. Heal. 49: 101-129.

Sahab-Negah, S., Hajali, V., Moradi, H. R. and Gorji, A. (2020). The Impact of Estradiol on Neurogenesis and Cognitive Functions in Alzheimer’s disease. Cell. Mol. Neurobiol. 40 (3): 283-299.

Setchell, K.D., Zimmer-Nechemias, L., Cai, J. andHeubi, J.E. (1998). Isoflavone content of infant formulas and the metabolic fate of these phytoestrogens in early life. Am. J. Clin. Nutr. 68: 1453S-1461S.

Shang, J., Wan, Y., Luo, C., Ye, G., Geng, O., Auerbach, A. and Li, F. (2020). Cell entry mechanisms of SARS-CoV-2. Proc. Nat. Acad. Sci. 117 (21): 11727-11734.

Shanle, E. K. and Xu, W. (2010). Endocrine disrupting chemicals targeting estrogen receptor signaling: identification and mechanisms of action. Chem. Res. Toxicol. 24: 6-19.

Shen, G., Li, Y., Yang, C., Xing, R., Zhang, H., Chen, E., Han, C., Liu, H., Zhang, W. and Xia, Q. (2015) Vertebrate estrogen regulates the development of female characteristics in silkworm, Bombyx mori. Gen. Comp. Endocrinol. 210: 30–37.

Sheppard, P.A.S., Choleris, E. and Galea, L.A.M. (2019). Structural plasticity of the hippocampus in response to estrogens in female rodents. Mol. Brain. 12: 22. Singer, P. L. (1949). Occupational oligospermia. J. Am. Med. Ass. 140: 1249.

Singleton, D.W. and Khan, S. A. (2003). Xenoestrogen exposure and mechanisms of endocrine disruption. Front. Bio. 8: 110-118.

Soto, A.M., Lin, T.M., Justicia, H., Silvia, R.M. andSonnenschein, C. (1992). An "in culture" bioassay to assess the estrogenicity of xenobiotics (E-SCREEN). In: Colborn T, Clement C, editors. Chemically Induced Alterations in Sexual and Functional Development: The Wildlife/Human Connection. Princeton, NJ: Princeton Scientific Publishing Co., Inc. (Mehlman, M.A., ed.) Adv. Mod. Environ. Toxicol. 21: 295-309.

Sotoca, A. M., Gelpke, M. D. and Boeren, S. (2011). Quantitative proteomics and transcriptomics addressing the estrogen receptor subtype-mediated effects in T47D breast cancer cells exposed to the phytoestrogen genistein. Mol. Cell. Prot. 10 (1): M110.002170.

Spencer, J. L., Waters, E. M., Romeo, R. D., Wood, G. E., Milner, T. A. and McEwen, B. S. (2008). Uncovering the mechanisms of estrogen effects on hippocampal function. Fron. Neuro. 29 (2): 219-237.

Staples, C. A., Peterson, D. R., Parkerton, T.F. and Adams, W. J. (1997). The environmental fate of phthalate esters: a literature review. Chemosphere. 35 (4): 667-749.

Stelzig, K. E., Canepa-Escaro, F., Schiliro, M., Berdnikovs, S. and Prakash, Y.S. (2020). Estrogen regulates the expression of SARS-CoV-2 receptor ACE2 in differentiated airway epithelial cells. Am. J. Physiol.-Lung Cell. Mol. Physiol.318: 6, L1280-L1281.

Stoker, T. E., Gibson, E. K. and Zorrilla, L. M. (2010). Triclosan exposure modulates estrogen dependent responses in the female Wistar rat. Toxicol. Sci. 117 (1): 45- 53.

Stygar, D., Masironi, B., Eriksson, H. andSahlin, L. (2020). Studies on estrogen receptor (ER) α and β responses on gene regulation in peripheral blood leukocytes in vivo using selective ER agonists. J. Endocrinol. 194 (1): 101-119.

Tapiero, H., Tew, K. D., Ba, G. N. and Mathé, G. (2002). Polyphenols: do they play a role in the prevention of human pathologies? Biomed. Pharma. 56 (4): 200-207.

Ticconi, C., Pietropolli, A. andPiccione, E. (2013). Estrogen replacement therapy and asthma. Pul. Pharma. Thera. 26 (6): 617-623.

Toppari, J., John, C. L., Christiansen, P., Giwercman, A., Grandjean, P., Guillette, L. and Skakkebæk, N. (1996). Male Reproductive Health and Environmental Xenoestrogens. Environ.Heal.Pers. 104 (4): 741-803.

Velarde, M. C. (2013). Pleiotropic actions of estrogen: a mitochondrial matter. Physiol. Gen. 45 (3): 106-109. Verdeal, K. and Ryan, S. (1979). Naturally occurring estrogens in plant foodstuffs– a review. J. Food. Pro. 42 (7): 577-583.

Wagner, K.D. and Wagner, N. (2010). Peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) acts as regulator of metabolism linked to multiple cellular functions. Pharmacol. Thera. 125 (3): 423-435.

Wang, S., Awad, K. S. and Elinoff, J. M. (2015). G Protein-coupled Receptor 40 (GPR40) and Peroxisome Proliferator-activated Receptor γ (PPARγ): An integrated two-receptor signalling pathway. J. Biol. Chem. 290 (32): 19544-19557.

Watson, C.S., Bulayeva, N.N., Wozniak, A.L. andAlyea, R.A. (2007). Xenoestrogens are potent activators of nongenomic estrogenic responses. Steroids. 72: 124-134.

Weatherly, L. M. and Gosse, J. A. (2017). Triclosan exposure, transformation and human health effects. J. Toxicol. Environ. Health. B. Crit. Rev. 20 (8): 447-469.

Wee, S. and Wang, V. (2020). China Grapples With Mystery Pneumonia-Like Illness. In: The New York Times, Published: 6 January 2020, Updated: 21 January 2020.

Wilbanks, M. S., Gust, K.A., Atwa, S., Sunesara, I., Johnson, D., Ang, C. W., Meyer, S. A. and Perkins, E. J. (2014). Validation of a genomics-based hypothetical adverse outcome pathway: 2, 4-dinitrotoluene perturbs PPAR signaling thus impairing energy metabolism and exercise endurance. Toxico. Sci. 141 (1): 44-58.

Wocławek-Potocka, I., Mannelli, C., Boruszewska, D., Kowalczyk-Zieba, I., Waśniewski, T. and Skarżyński, D. J. (2013). Diverse Effects of Phytoestrogens on the Reproductive Performance: Cow as a Model. Int. J. Endo. 2013: 1-15.

Woodwell, G.M., Wurster, C.F. and Isaacson, P.A.D. (1967). DT residues in an east coast estuary: a case of biological concentration of a persistent insecticide. Science. 156: 821-824.

Yang, C. Z., Yaniger, S. I., Jordan, V. C., Klein, D.J. and Bittner, G. D. (2011). Most plastic products release estrogenic chemicals: a potential health problem that can be solved. Environ. Heal. Pers. 119 (7): 989-996.

Ying, G. G., Kookana, R.S. and Ru, Y. J. (2002). Occurrence and fate of hormone steroids in the environment. Environ. Int. 28 (6): 545-551.

Yueh, M. F. and Tukey, R. H. (2016). Triclosan a wide spread environmental toxicant with many biological effects. Ann. Rev. Pharmacol. Toxicol. 56: 251-272.

Zhang, H., Penninger, J.M., Li, Y., Zhong, N. and Slutsky, A. S. (2020). Angiotensin-converting enzyme 2 (ACE2) as a SARS-CoV-2 receptor: molecular mechanisms and potential therapeutic target. Inten. Care. Med. 46 (4): 586-590.

Published
2021-04-30
How to Cite
Das, D., & Das, S. (2021). Endocrine Disruptor—A threat to the animal world. International Journal of Experimental Research and Review, 24, 10-23. https://doi.org/10.52756/ijerr.2021.v24.002
Section
Articles