Hostname: page-component-788cddb947-jbkpb Total loading time: 0 Render date: 2024-10-19T17:26:18.565Z Has data issue: false hasContentIssue false

Oestrogens, adipose tissues and environmental exposures influence obesity and diabetes across the lifecycle

Published online by Cambridge University Press:  02 February 2024

Olgert Bardhi
Affiliation:
Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
Pallavi Dubey
Affiliation:
Department of Obstetrics and Gynecology, Paul L Foster School of Medicine, El Paso, TX, USA
Biff Franklin Palmer
Affiliation:
Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical, Center, Dallas, TX, USA
Deborah J. Clegg*
Affiliation:
Vice President for Research, Texas Tech Health Sciences Center, El Paso, TX, USA
*
*Corresponding author: Deborah J. Clegg, email: dclegg@ttuhsc.edu
Rights & Permissions [Opens in a new window]

Abstract

Endogenous oestrogens regulate essential functions to include menstrual cycles, energy balance, adipose tissue distribution, pancreatic β-cell function, insulin sensitivity and lipid homeostasis. Oestrogens are a family of hormones which include oestradiol (E2), oestrone (E1) and oestriol (E3). Oestrogens function by binding and activating oestrogen receptors (ERs). Phytoestrogens are plant-derived compounds which exhibit oestrogenic-like activity and can bind to ERs. Phytoestrogens exert potential oestrogenic-like benefits; however, their effects are context-dependent and require cautious consideration regarding generalised health benefits. Xenoestrogens are synthetic compounds which have been determined to disrupt endocrine function through binding to ERs. Xenoestrogens enter the body through various routes and given their chemical structure they can accumulate, posing long-term health risks. Xenoestrogens interfere with endogenous oestrogens and their functions contributing to conditions like cancer, infertility, and metabolic disorders. Understanding the interplay between endogenous and exogenous oestrogens is critical in order to determine their potential health consequences and requires further investigation. This manuscript provides a summary of the role endogenous oestrogens have in regulating metabolic functions. Additionally, we discuss the impact phytoestrogens and synthetic xenoestrogens have on biological systems across various life stages. We highlight their mechanisms of action, potential benefits, risks and discuss the need for further research to bridge gaps in understanding and mitigate exposure-related health risks.

Type
Conference on ‘Understanding the Role of Sex and Gender in Nutrition Research’
Copyright
© Texas Tech University Health Science Center El Paso, 2024. Published by Cambridge University Press on behalf of The Nutrition Society

Overview

Functions of endogenous oestrogens

Sex steroids, including testosterone and oestrogens, are present in both males and females, and their biological functions and production undergoes changes across the life cycle. We will focus our article on the role of oestrogens in mediating metabolic functions. In females, certain oestrogens surge during puberty and pregnancy but decline during menopause. Oestrogens are pivotal for growth, development, sexual differentiation and reproduction. Oestrogens have diverse roles throughout the body, and they regulate functions to include menstrual cycles(Reference Reed and Carr1), energy balance(Reference Mauvais-Jarvis, Clegg and Hevener2), adipose tissue distribution and well-being(Reference Bracht, Vieira-Potter and De Souza Santos3,Reference Kuryłowicz4) , pancreatic β-cell function and viability(Reference Mauvais-Jarvis5), skeletal muscle insulin sensitivity(Reference Yan, Yang and Zhou6), liver lipid balance(Reference Palmisano, Zhu and Stafford7) and macrophage function and polarisation(Reference Bolego, Cignarella and Staels8). Importantly, any disruption to the function of oestrogens can potentially lead to conditions such as obesity(Reference Lizcano and Guzmán9), adipose tissue inflammation(Reference Kuryłowicz4), atherosclerosis(Reference Meng, Li and Chao10), changes in adipose tissue distribution and function(Reference Kuryłowicz4), pancreatic β-cell dysfunction(Reference Liu and Mauvais-Jarvis11,Reference Santos, Batista and Camargo12) , fatty liver(Reference Della Torre13), impaired peripheral tissue glucose regulation(Reference De Paoli, Zakharia and Werstuck14) and systemic inflammation(Reference Straub15). The goal of this review is to explore how natural oestrogens affect metabolic function and discuss how plant-based and artificial oestrogens also impact metabolism across the life cycle. We conclude by emphasising the need for further research to fill knowledge gaps.

Endogenous oestrogens

Endogenous oestrogens – a ‘family’ of oestrogens

Oestrogens are a cohort of sex steroid hormones derived from cholesterol. Among the endogenous physiological oestrogens, three primary forms exist: oestrone (E1), oestradiol (E2) and oestriol (E3). E2 holds prominence as the primary oestrogenic compound circulating during a woman’s reproductive years, and it remains dominant until menopause. Following menopause, E1 gains significance as a key oestrogenic compound, while E3 assumes a major role during pregnancy, even though it’s considered the least potent oestrogen(Reference Cui, Shen and Li16).

The synthesis of oestrogens in ovarian granulosa cells hinges on the activation of an enzyme named aromatase (CYP19A1), which belongs to the cytochrome P450 superfamily. This enzyme facilitates the aromatisation process, wherein androgens, particularly testosterone and androstenedione, are transformed into oestradiol and oestrone. Notably, studies on animals lacking this enzyme have revealed an inability to produce oestrogens(Reference Jones, Thorburn and Britt17,Reference Dalla, Antoniou and Papadopoulou-Daifoti18) . In women during their reproductive years, the ovaries, corpus luteum and placenta serve as primary sites for oestrogen production. Additionally, smaller amounts of oestrogens are synthesised by non-gonadal organs such as the liver, heart, skin, brain, adipose tissues, intestines and adrenal glands(Reference Barakat, Oakley and Kim19). In prepubescent females and males, oestrogens are produced in non-gonadal locations, where their functional effects are typically exerted locally through paracrine and/or intracrine mechanisms(Reference Cui, Shen and Li16).

Following the onset of menopause, adipose tissue stromal cells and preadipocytes exhibit high expression levels of aromatase, 17b-hydroxysteroid dehydrogenases (17bHSD) and CYP1B1. Among these, 17bHSD plays a role in converting weak androgens and/or oestrogens into their more potent counterparts to include the conversion of androstenedione into testosterone and oestrone into oestradiol(Reference Hilborn, Stål and Jansson20). Due to the substantial mass of adipose tissue, its contribution to whole-body steroid metabolism is notably significant.

E2 levels in humans and rodents

In pre-menopausal individuals, the levels of E2, or oestradiol, exhibit a range of 15–400 pg/ml, a variance largely contingent on the phase of the menstrual cycle. Conversely, E2 levels plummet to less than 10 pg/ml during menopause. Rodents have been used to study and learn more about the physiological functions of oestrogens. Rodents undergo a reproductive cycle, termed the oestrous cycle, divided into four distinct phases—proestrus, oestrus, metestrus and diestrus—spanning about 4–5 d. Throughout this cycle, the concentration of circulating E2 within rodents displays significant fluctuations. Among these phases, proestrus showcases the highest E2 concentration, roughly ranging between 30–60 pg/ml. Even during diestrus and metestrus, E2 remains detectable with the use of sensitive instrumentation, albeit at levels not surpassing 5 pg/ml. Remarkably, during the oestrus stage, E2 has been found to be undetectable(Reference Frick21Reference Wall, Desai and Khant Aung24).

Oestrogens modulate homeostasis through binding and activation of oestrogen receptors

Oestrogens exert their effects through the mediation of oestrogen receptors (ERs), which comprise nuclear receptors, surface membrane receptors, and are found in various forms such as ERα (ESR1), ERβ (ESR2), G protein-coupled receptors (GRP30 and Gq-mER) and ER-X. Nuclear ERs function as transcription factors, overseeing the modulation of specific gene transcription. On the other hand, surface membrane ERs, once activated by oestrogens, instigate rapid intracellular signalling pathways(Reference Cui, Shen and Li16). Beyond their interaction with ERs, oestrogens can also regulate enzymatic activities and engage with non-steroid-hormone-nuclear receptors, consequently initiating signalling pathways that are independent of ERs(Reference Cui, Shen and Li16).

Effects of endogenous oestrogens on obesity, diabetes and the metabolic syndrome

Many women exhibit features of the metabolic syndrome (abdominal adiposity, insulin resistance and dyslipidemia) with the onset of oestrogen deficiency at menopause. There are direct effects of oestrogen deficiency on body fat distribution, insulin action, effect on arterial wall and fibrinolysis that may contribute to an increased prevalence of the metabolic syndrome in post-menopausal women compared to pre-menopausal women(Reference Park, Zhu, Palaniappan and Heshka25). Fabre et al., postulated that membrane-initiated ERα extra-nuclear signalling contributes to female, but not to male, protection against high-fat-diet-induced obesity and associated metabolic disorders in mouse(Reference Fabre, Tramunt and Montagner26).

Exogenous oestrogens

Exogenous oestrogens obtained through the diet

Oestrogens can also be introduced into the body from external sources, either through the consumption of certain foods or exposure to various environmental factors. These exogenous oestrogen exposures are capable of exhibiting oestrogen-like activities, which involve binding to ERs and other nuclear hormone receptors. There are two distinct categories of exogenous oestrogens: xenoestrogens, which are synthetic oestrogens, and phytoestrogens, which are phytochemicals derived naturally from plants. Phytoestrogens possess the potential to exert both beneficial effects and function as endocrine disruptors(Reference Wang, Ha and Yoshitake27). The roles and effects of phytoestrogens will be explored in greater detail further below. Xenoestrogens also bind to and activate ERs and currently data suggests these exert endocrine disruption and may have long-term negative effects as will also be discussed below.

Phytoestrogens

Phytoestrogens, compounds resembling oestrogens and sourced from plants, are commonly found in soy products, grains, peas, beans, as well as in specific fruits and vegetables(Reference Domínguez-López, Yago-Aragón and Salas-Huetos28,Reference Desmawati and Sulastri29) . Phytoestrogens can be classified into three main categories based on their chemical structures: (1) Flavonoids, such as genistein, daidzein, glycitein and biochanin, and they are frequently present in soy products and are often marketed as dietary supplements; (2) Coumestans, represented by compounds like coumestrol, wedelolactone and plicadin, can be found in foods like broccoli and sprouts, with their oestrogenic activity significantly surpassing that of isoflavones; (3) Lignans, originate from plant cell walls and are polyphenolic components present in plants, seeds, whole grains and certain vegetables. This category also encompasses compounds like enterodiol, enterolactone, pinoresinol, matairesinol and sesamin, some of which are produced by intestinal bacteria and exert relatively mild oestrogenic effects(Reference Domínguez-López, Yago-Aragón and Salas-Huetos28Reference Viggiani, Polimeno and Di Leo30).

Phytoestrogens have the ability to bind to and activate oestrogen receptors

Phytoestrogens exhibit the ability to bind to and activate ERs. Interestingly, data suggest phytoestrogens demonstrate a preference for oestrogen receptor beta activation (ERβ), as evidenced by the ratios of binding affinity of these compounds relative to binding to ERα v. ERβ: Genistein β/α = 20, Daidzein β/α = 7, S-Equol β/α = 32, Coumestrol β/α = 7, Naringenin β/α = 11, Apigenin β/α = 20(Reference Minutolo, Macchia and Katzenellenbogen31,Reference Paterni, Granchi and Katzenellenbogen32) . Moreover, phytoestrogens are known to also bind to serotonergic receptors and insulin-like growth factor receptors exerting biological effects which are impacted by the duration and age for which exposure has occurred. Phytoestrogens possess the capability to enhance the binding of free radicals and can directly or indirectly influence the activation of tyrosine kinases, cyclic adenosine monophosphate pathways, phosphatidylinositol-3 kinase (PI3K), DNA methylation, as well as histone and RNA expression(Reference Domínguez-López, Yago-Aragón and Salas-Huetos28).

Detrimental or beneficial phytoestrogen function(s)?

The discussion surrounding whether phytoestrogens yield positive or negative effects is ongoing. Crucially, it is important to recognise that the timing, type, duration and level of exposure plays pivotal roles in determining whether these oestrogenic compounds offer advantages or pose risks. Moreover, the context matters significantly as to whether phytoestrogens are therapeutic for conditions like alleviating menopausal symptoms, reducing osteoporosis, or potentially exerting anti-tumorigenic properties. When low endogenous levels of oestrogens exist, there are data suggesting phytoestrogenic benefits; however, the potential for off-target or side effects post-consumption/exposure remains less understood(Reference Patisaul and Jefferson33). Moreover, when phytoestrogens are consumed in combination with drugs, phytoestrogens can interfere with the drug efficacy and function, implying that additional data and caution may be necessary to ascertain if phytoestrogen intake affects the safety of other medications. It is worth noting that the beneficial effects of phytoestrogens could differ in pre-menopausal women with higher endogenous oestrogen levels(Reference Patisaul and Jefferson33) when compared to post-menopausal women. Additionally, the impact of phytoestrogens on men, who naturally have lower circulating levels of oestrogens, is also less understood.

The timing of phytoestrogen exposure emerges as a critical factor influencing their potential positive or negative effects. Exposure can occur during foetal development, adulthood, in the presence or absence of endogenous oestrogens, and in both females and males. However, establishing an association between dietary phytoestrogens and endocrine biomarkers remains inconclusive, partly due to variations in the type and concentration of compounds, the bioavailability of phytoestrogens, and whether they affect circulating v. urinary excretion of metabolites(Reference Patisaul and Jefferson33,Reference Patisaul34) .

Xenoestrogens

Xenoestrogens are compounds which mimic oestrogenic functions and are not produced within the body. Xenoestrogens are synthetic in origin and include chemicals used as solvents/lubricants. Xenoestrogens, like phytoestrogens, are structurally and functionally similar to oestrogens and they bind to the ERs producing biological effects some of which might be beneficial; however, in most cases they have been shown to be detrimental. In fact, xenoestrogens and their byproducts such as plastics like bisphenol A (BPA), plasticisers (phthalates), pesticides (DDT), pharmaceutical agents, are considered to be environmental hazards due to their hormone-disruptive effects(Reference Wang, Ha and Yoshitake27) and xenoestrogens are classified as endocrine disrupting chemicals (EDCs) because they: ‘interfere with the synthesis, secretion, transport, binding, action, or elimination of natural hormones in the body that are responsible for development, behaviour, fertility and maintenance of homeostasis(Reference Crisp, Clegg and Cooper35).

Xenoestrogens enter the body through various routes, including ingestion and absorption from foods, exposure to dust and water, inhalation of airborne gases and particles, and direct contact with the skin(Reference Sonnenschein and Soto36). Moreover, they can be transmitted to developing foetuses through maternal transfer and to infants via breast milk or formula prepared with water containing xenoestrogens. Once inside the body, xenoestrogens engage with endogenous oestrogens and oestrogen receptors (both membrane and nuclear receptors), leading to disruption of oestrogenic signalling(Reference Mueller and Kim37Reference Soto, Chung and Sonnenschein39).

Xenoestrogens, akin to other endocrine disrupting substances (EDCs), can interfere with the synthesis, secretion, transport, metabolism, binding, action, or elimination of oestrogens(Reference Diamanti-Kandarakis, Bourguignon and Giudice40,Reference Zoeller, Brown and Doan41) . Their relatively low water solubility and high lipid solubility contribute to a lengthy environmental half-life. Additionally, due to their chemical makeup, xenoestrogens are widely distributed in the environment and tend to accumulate in wastewater, consequently impacting marine animal reproduction(Reference Delfosse, Grimaldi and le Maire42).

Among the most hazardous classes of xenoestrogens is bisphenol-A (BPA). BPA is extensively used in the production of plastics, thermal paper, can linings and dental sealants(Reference Gonsioroski, Mourikes and Flaws43). Exposure to BPA has been associated with various hormone-related cancers(Reference Gao, Yang and Li44). Moreover, data suggest that BPA can interfere with neuronal development, leading to physiological and behavioural effects(Reference Rebuli, Cao and Sluzas45). Another perilous group is halogenated bisphenols A (H-BPAs), including tetrachlorobisphenol A (TCBPA). These compounds are employed in manufacturing and present in different environmental contexts, demonstrating greater toxicity than BPA(Reference Song, Liang and Liang46). Diethylstilbestrol (DES), another xenoestrogen, was administered to women from the 1940s to the 1970s to lower the risk of miscarriages(Reference Sonnenschein and Soto36,Reference Brotons, Olea-Serrano and Villalobos47Reference Das and Thomas49) ; tragically, it resulted in birth defects and has since been banned.

BPA enhances the pathogenesis processes of type 2 diabetes mellitus which includes insulin resistance, impaired glucagon secretion and pancreatic β-cell dysfunction(Reference Soundararajan, Prabu and Mohan50Reference Chen, Zhong and Wu52). BPA accelerates the cellular senescence and apoptosis by increasing the metabolic stress of high glucose(Reference Jiang, Ding and Huang53). BPA exposure is also associated with promoting disturbances in lipid metabolism and insulin resistance(Reference Nadal54). Many studies in the past were able to identify a possible link between the BPA-induced organ damage mechanisms and pathogenesis of diabetic complications(Reference Moreno-Gómez-Toledano, Arenas and Muñoz-Moreno55,Reference Bulzomi and Marino56) .

Xenoestrogen’s ‘estrogen-like’ activity

Xenoestrogens not only engage with and activate ERs and oestrogen-related receptors, but they also interact with androgen receptors and retinoid receptors. Moreover, xenoestrogens can directly or indirectly bind to non-steroid receptors, including neurotransmitter receptors like serotonin, dopamine and norepinephrine receptors. They can also interact with orphan receptors, such as the aryl hydrocarbon receptor (AhR), triggering enzymatic pathways that influence steroid biosynthesis and metabolism(Reference Bulzomi and Marino56). Many xenoestrogens possess a phenolic structure resembling that of E2, allowing them to interact with ERs as either agonists or antagonists(Reference Bolli and Marino57). Their binding affinities for ERs span from sub-nanomolar to micromolar values, and once bound, they can function as full agonists or partial agonists/antagonists(Reference Delfosse, Grimaldi and le Maire42). Xenoestrogens can also serve as hormone precursors, thereby impacting steroid metabolism and functioning as steroid-sensitive substrates(Reference Wang, Chen and Chen58). However, the relationship between BPA and metabolic outcomes is complicated by a non-monotonic dose response curve. Various epidemiological studies assumed a linear relationship between BPA exposure and diabetes risk but lately it has been postulated by the regulatory agencies that BPA can mitigate adverse effects at much lower doses than the calculated safe dose(Reference Song, Chou and Baecker59,Reference Gore, Chappell and Fenton60) .

Xenoestrogens may negatively influence various health conditions, including breast and prostate cancer, endometriosis, infertility, diabetes, metabolic syndrome, early puberty and obesity. Xenoestrogens can interfere with the development of the female reproductive tract by competitively inhibiting endogenous oestrogens and selectively binding to ERs. Specific substances, such as nine PCBs, three pesticides, a furan, and two phthalates, have been linked to gradual damage to the follicular pool and associated with earlier onset of menopause(Reference Vabre, Gatimel and Moreau61). Xenoestrogens can also contribute to the formation of ovarian cysts, leading to a condition known as polycystic ovary syndrome(Reference Götz, Thieme and Dörner62). Adolescents residing in areas with significant industrial development and PCB exposure experienced earlier menarche and thelarche compared to their unexposed peers(Reference Grindler, Allsworth and Macones63). A National Health and Nutrition Examination study showed higher BPA exposure is associated with obesity in the adult population of the United States(Reference Carwile and Michels64). Riffee et al., explored the relationship between bisphenol exposure and lipid profile parameters and exercise-induced glucose uptake mechanisms and indicated that BPA and bisphenol S are highly correlated with oxidative stress generation and impaired lipid metabolism(Reference Riffee, Wade and Sine65).

Foetal development

Xenoestrogens have been demonstrated to provide detrimental effects on foetal development, with evidence indicating negative impacts on neuronal and endocrine function(Reference Paterni, Granchi and Minutolo66). These compounds have also been associated with cancer-promoting mutations within cells during foetal development(Reference Palmlund67). Furthermore, exposure to xenoestrogens during pregnancy has been linked to alterations in the genetic sequence within the placenta, thereby adversely affecting foetal growth and overall development(Reference Vilahur, Bustamante and Byun68,Reference García-Arevalo, Alonso-Magdalena and Rebelo Dos Santos69) . A study by Garcia-Arvelo et al., exposed mice to BPA subcutaneously and fed a high-fat diet and the authors found the male offspring had fasting hyperglycaemia, glucose intolerance and high levels of NEFA in the plasma compared with the controls(Reference García-Arevalo, Alonso-Magdalena and Rebelo Dos Santos69). An epigenomic-wide analysis of cord blood DNA methylation indicated that there may be sex specific epigenomic responses to BPA exposure in offspring as well(Reference Miura, Araki and Minatoya70).

The term used to define the timing for which an individual is exposed to xenoestrogens is referred to as the ‘term window’(Reference Diamanti-Kandarakis, Bourguignon and Giudice40). Determining the ‘term window’ is extremely challenging and yet may be critical in determining if and when the impact of the exposure is manifested and to what harm the organism may experience due to the exposure. The ‘term window’ is the period of developmental susceptibility during which the developing organism can be altered by environmental factors resulting in structural, functional and/or cellular changes(Reference Diamanti-Kandarakis, Bourguignon and Giudice40).

Limitations and gaps in knowledge

Two recent Scientific Statements from The Endocrine Society have highlighted the urgent necessity to delve into the fundamental mechanisms of action xenoestrogens exert and the subsequent physiological impacts of endocrine disruptors they impart(Reference Diamanti-Kandarakis, Bourguignon and Giudice40,Reference Zoeller, Brown and Doan41) . It cannot be understated that there is of utmost importance to conduct foundational in vitro molecular investigations to uncover the pathways through which xenoestrogens exert their influence on endocrine tissues(Reference Diamanti-Kandarakis, Bourguignon and Giudice40,Reference Zoeller, Brown and Doan41) .

How to minimise exposure

Given the documented adverse effects of xenoestrogen exposure, one effective strategy to minimise risk is to reduce the use of cosmetic products that contain parabens and phthalates. Additionally, using glass or ceramic containers for storing food, as opposed to plastic, is recommended due to the heightened presence of xenoestrogens in plastics. It’s advisable to avoid using plastic containers in the microwave to prevent potential leaching of BPA into microwaved products. Additionally, refraining from heating plastics, even in direct sunlight, is recommended to mitigate exposure(Reference Yang, Yaniger and Jordan71,Reference Bittner, Yang and Stoner72) .

Moreover, adjusting dietary consumption of foods which contain high levels of xenoestrogens can aid in decreasing exposure. Specifically, restricting the intake of canned fish which can contain elevated levels of mercury and PCBs, would be prudent(Reference Darnerud, Eriksen and Jóhannesson73,Reference Hites, Foran and Schwager74) . Health professionals and nutrition experts advocate for consumption of fresh fish which is high in n-3 long-chain PUFA(Reference Yokoyama, Origasa and Matsuzaki75,Reference Troesch, Eggersdorfer and Laviano76) . Additionally, various studies indicate that consuming fish might protect against the effects of methyl mercury(Reference Hibbeln, Davis and Steer77Reference Myers, Davidson and Cox79). For instance, research conducted in the Seychelles Child Development Study suggests a positive correlation between fish consumption and cognition(Reference Davidson, Myers and Cox78,Reference Myers, Davidson and Cox79) . Moreover, opting for pesticide-free fruits and vegetables is advisable whenever possible. If this is not feasible, then ensuring that fruit and vegetables are washed thoroughly prior to consumption could minimise exposure(Reference Mukherjee, Pandya and Baxi80Reference Kuiper, Lemmen and Carlsson82).

Inability to conduct research on xenoestrogens

Research on xenoestrogens is constrained by ethical considerations because conducting studies to explore the harmful effects of these substances on humans can pose moral challenges. Many studies available in the literature are observational or conducted on animals, thereby limiting the direct applicability of their findings to humans. The scarcity of randomised controlled trials in the realm of xenoestrogen exploration is notable. As previously indicated, potential detrimental effects of xenoestrogens occur as they accumulate within organisms and are contingent on factors such as dosage and exposure duration, which may not be discernible within shorter timeframes. A deeper understanding necessitates further experiments involving the examination of the impact of xenoestrogens both in their environmental context and in isolation.

Conclusion

Oestrogens exert many critical functions throughout the body. Endogenous oestrogens, particularly oestradiol, have vital roles in growth, development and reproduction. Additionally, oestrogens function beyond reproduction and include regulating energy balance, adipose tissue distribution and insulin sensitivity. Because of the vital biological function oestrogens have, exposure to exogenous oestrogens, like phytoestrogens from plants and synthetic xenoestrogens, can have profound effects which may be both beneficial and harmful.

Phytoestrogens, found in foods, can bind to oestrogen receptors, potentially offering benefits for various conditions. However, their effects depend on timing and exposure levels, raising questions about safety and interactions with medications. Xenoestrogens on the other hand, are synthetic compounds derived from various sources and they mimic oestrogens and can disrupt endocrine function, affecting health outcomes, including cancer and reproductive disorders. Avoiding certain products containing xenoestrogens and making dietary choices can help reduce exposure risks.

Clearly understanding the impact xenoestrogens have on biological systems is critical yet research on xenoestrogens faces ethical and complexity challenges, limiting our understanding. Many studies are observational or conducted in animal models which do not completely replicate human physiological function therefore requiring further investigation to bridge gaps between animal and human responses. Variability in doses and exposure durations underscores the need for more extensive experiments to comprehend their impacts accurately. Understanding the interplay between endogenous and exogenous oestrogens is crucial for health. Ongoing research guided by ethical considerations is essential to unravel complexities, inform strategies to minimise exposures and mitigate potential health risks.

Acknowledgements

None.

Financial support

None.

Authorship

Dr. D.J.C. is responsible for Conceptualization of the article. Dr’s D.J.C., O.B., P.D., and B.F.P. equally contributed to the writing of the manuscript.

Conflict of interest

No conflicts of interest for any of the authors.

References

Reed, BG & Carr, BR (2018) The Normal Menstrual Cycle and the Control of Ovulation. Endotext. South Dartmouth, MA: MDText.com, Inc.Google Scholar
Mauvais-Jarvis, F, Clegg, DJ & Hevener, AL (2013) The role of estrogens in control of energy balance and glucose homeostasis. Endocr Rev 34, 309338. doi: 10.1210/er.2012-1055.CrossRefGoogle ScholarPubMed
Bracht, JR, Vieira-Potter, VJ, De Souza Santos, R et al. (2020) The role of estrogens in the adipose tissue milieu. Ann N Y Acad Sci 1461, 127143. doi: 10.1111/nyas.14281.CrossRefGoogle ScholarPubMed
Kuryłowicz, A (2023) Estrogens in adipose tissue physiology and obesity-related dysfunction. Biomedicines 11, 690. doi: 10.3390/biomedicines11030690.CrossRefGoogle ScholarPubMed
Mauvais-Jarvis, F (2016) Role of sex steroids in β cell function, growth, and survival. Trends Endocrinol Metab 27, 844855. doi: 10.1016/j.tem.2016.08.008.CrossRefGoogle ScholarPubMed
Yan, H, Yang, W, Zhou, F et al. (2019) Estrogen improves insulin sensitivity and suppresses gluconeogenesis via the transcription factor foxo1. Diabetes 68, 291304. doi: 10.2337/db18-0638.CrossRefGoogle ScholarPubMed
Palmisano, BT, Zhu, L & Stafford, JM (2017) Role of estrogens in the regulation of liver lipid metabolism. Adv Exp Med Biol 1043, 227256. doi: 10.1007/978-3-319-70178-3_12 CrossRefGoogle ScholarPubMed
Bolego, C, Cignarella, A, Staels, B et al. (2013) Macrophage function and polarization in cardiovascular disease: a role of estrogen signaling? Arterioscler Thromb Vasc Biol 33, 11271134. doi: 10.1161/ATVBAHA.113.301328.CrossRefGoogle ScholarPubMed
Lizcano, F & Guzmán, G (2014) Estrogen deficiency and the origin of obesity during menopause. Biomed Res Int 2014, 757461. doi: 10.1155/2014/757461.CrossRefGoogle ScholarPubMed
Meng, Q, Li, J, Chao, Y et al. (2020) β-estradiol adjusts intestinal function via ERβ and GPR30 mediated PI3K/AKT signaling activation to alleviate postmenopausal dyslipidemia. Biochem Pharmacol 180, 114134. doi: 10.1016/j.bcp.2020.114134.CrossRefGoogle ScholarPubMed
Liu, S & Mauvais-Jarvis, F (2010) Minireview: estrogenic protection of beta-cell failure in metabolic diseases. Endocrinology 151, 859864. doi: 10.1210/en.2009-1107. (published correction appears in Endocrinology. 2010; 151, 4597).CrossRefGoogle Scholar
Santos, RS, Batista, TM, Camargo, RL et al. (2016) Lacking of estradiol reduces insulin exocytosis from pancreatic β-cells and increases hepatic insulin degradation. Steroids 114, 1624. doi: 10.1016/j.steroids.2016.05.002.CrossRefGoogle ScholarPubMed
Della Torre, S (2020) Non-alcoholic fatty liver disease as a canonical example of metabolic inflammatory-based liver disease showing a sex-specific prevalence: relevance of estrogen signaling. Front Endocrinol (Lausanne) 11, 572490. doi: 10.3389/fendo.2020.572490.CrossRefGoogle ScholarPubMed
De Paoli, M, Zakharia, A & Werstuck, GH (2021) The role of estrogen in insulin resistance: a review of clinical and preclinical data. Am J Pathol 191, 14901498. doi: 10.1016/j.ajpath.2021.05.011.CrossRefGoogle ScholarPubMed
Straub, RH (2007) The complex role of estrogens in inflammation. Endocr Rev 28, 521574. doi: 10.1210/er.2007-0001.CrossRefGoogle ScholarPubMed
Cui, J, Shen, Y & Li, R (2013) Estrogen synthesis and signaling pathways during aging: from periphery to brain. Trends Mol Med 19, 197209. doi: 10.1016/j.molmed.2012.12.007.CrossRefGoogle ScholarPubMed
Jones, ME, Thorburn, AW, Britt, KL et al. (2000) Aromatase-deficient (ArKO) mice have a phenotype of increased adiposity. Proc Natl Acad Sci U S A 97, 1273512740. doi: 10.1073/pnas.97.23.12735.CrossRefGoogle ScholarPubMed
Dalla, C, Antoniou, K, Papadopoulou-Daifoti, Z et al. (2004) Oestrogen-deficient female aromatase knockout (ArKO) mice exhibit depressive-like symptomatology. Eur J Neurosci 20, 217228. doi: 10.1111/j.1460-9568.2004.03443.x.CrossRefGoogle ScholarPubMed
Barakat, R, Oakley, O, Kim, H et al. (2016) Extra-gonadal sites of estrogen biosynthesis and function. BMB Rep 49, 488496. doi: 10.5483/bmbrep.2016.49.9.141.CrossRefGoogle ScholarPubMed
Hilborn, E, Stål, O & Jansson, A (2017) Estrogen and androgen-converting enzymes 17β-hydroxysteroid dehydrogenase and their involvement in cancer: with a special focus on 17β-hydroxysteroid dehydrogenase type 1, 2, and breast cancer. Oncotarget 8, 3055230562. doi: 10.18632/oncotarget.15547.CrossRefGoogle Scholar
Frick, KM (2009) Estrogens and age-related memory decline in rodents: what have we learned and where do we go from here? Horm Behav 55, 223. doi: 10.1016/j.yhbeh.2008.08.015.CrossRefGoogle Scholar
Zenclussen, ML, Casalis, PA, Jensen, F et al. (2014) Hormonal fluctuations during the estrous cycle modulate heme oxygenase-1 expression in the uterus. Front Endocrinol (Lausanne) 5, 32. doi: 10.3389/fendo.2014.00032.CrossRefGoogle ScholarPubMed
Caligioni, CS (2009) Assessing reproductive status/stages in mice. Curr Protoc Neurosci Appendix 4, Appendix-4I. doi: 10.1002/0471142301.nsa04is48.CrossRefGoogle Scholar
Wall, eg Desai, R, Khant Aung, Z et al. (2023) Unexpected plasma gonadal steroid and prolactin levels across the mouse estrous cycle. Endocrinology 164, bqad070. doi: 10.1210/endocr/bqad070.CrossRefGoogle ScholarPubMed
Park, YW, Zhu, S, Palaniappan, L, Heshka, S et al. (2003) The metabolic syndrome: prevalence and associated risk factor findings in the US population from the Third National Health and Nutrition Examination Survey, 1988–1994. Arch Intern Med 163, 427436. doi: 10.1001/archinte.163.4.427 CrossRefGoogle ScholarPubMed
Fabre, A, Tramunt, B, Montagner, A et al. (2023) Membrane estrogen receptor-α contributes to female protection against high-fat diet-induced metabolic disorders. Front Endocrinol (Lausanne) 14, 1215947. doi: 10.3389/fendo.2023.1215947 CrossRefGoogle ScholarPubMed
Wang, X, Ha, D, Yoshitake, R et al. (2021) Exploring the biological activity and mechanism of xenoestrogens and phytoestrogens in cancers: emerging methods and concepts. Int J Mol Sci 22, 8798. doi: 10.3390/ijms22168798.CrossRefGoogle ScholarPubMed
Domínguez-López, I, Yago-Aragón, M, Salas-Huetos, A et al. (2020) Effects of dietary phytoestrogens on hormones throughout a human lifespan: a review. Nutrients 12, 2456. doi: 10.3390/nu12082456.CrossRefGoogle ScholarPubMed
Desmawati, D & Sulastri, D (2019) Phytoestrogens and their health effect. Open Access Maced J Med Sci 7, 495499. doi: 10.3889/oamjms.2019.044.CrossRefGoogle ScholarPubMed
Viggiani, MT, Polimeno, L, Di Leo, A et al. (2019) Phytoestrogens: dietary intake, bioavailability, and protective mechanisms against colorectal neoproliferative lesions. Nutrients 11, 1709. doi: 10.3390/nu11081709.CrossRefGoogle ScholarPubMed
Minutolo, F, Macchia, M, Katzenellenbogen, BS et al. (2011) Estrogen receptor β ligands: recent advances and biomedical applications. Med Res Rev 31, 364442. doi: 10.1002/med.20186.CrossRefGoogle ScholarPubMed
Paterni, I, Granchi, C, Katzenellenbogen, JA et al. (2014) Estrogen receptors alpha (ERα) and beta (ERβ): subtype-selective ligands and clinical potential. Steroids 90, 1329. doi: 10.1016/j.steroids.2014.06.012.CrossRefGoogle ScholarPubMed
Patisaul, HB & Jefferson, W (2010) The pros and cons of phytoestrogens. Front Neuroendocrinol 31, 400419. doi: 10.1016/j.yfrne.2010.03.003.CrossRefGoogle ScholarPubMed
Patisaul, HB (2017) Endocrine disruption by dietary phyto-oestrogens: impact on dimorphic sexual systems and behaviours. Proc Nutr Soc 76, 130144. doi: 10.1017/S0029665116000677.CrossRefGoogle ScholarPubMed
Crisp, TM, Clegg, ED, Cooper, RL et al. (1998) Environmental endocrine disruption: an effects assessment and analysis. Environ Health Perspect 106, 1156. doi: 10.1289/ehp.98106s111.CrossRefGoogle Scholar
Sonnenschein, C & Soto, AM (1998) An updated review of environmental estrogen and androgen mimics and antagonists. J Steroid Biochem Mol Biol 65, 143150. doi: 10.1016/s0960-0760(98)00027-2.CrossRefGoogle Scholar
Mueller, G & Kim, UH (1978) Displacement of estradiol from estrogen receptors by simple alkyl phenols. Endocrinol 102, 14291435.CrossRefGoogle ScholarPubMed
McLachlan, JA (1993) Functional toxicology: a new approach to detect biologically active xenobiotics. Environ Health Perspect 101, 386387. doi: 10.1289/ehp.93101386.CrossRefGoogle ScholarPubMed
Soto, AM, Chung, KL & Sonnenschein, C (1994) The pesticides endosulfan, toxaphene, and dieldrin have estrogenic effects on human estrogen-sensitive cells. Environ Health Perspect 102, 380383. doi: 10.1289/ehp.94102380.CrossRefGoogle ScholarPubMed
Diamanti-Kandarakis, E, Bourguignon, JP, Giudice, LC et al. (2009) Endocrine-disrupting chemicals: an endocrine society scientific statement. Endocr Rev 30, 293342. doi: 10.1210/er.2009-0002.CrossRefGoogle ScholarPubMed
Zoeller, RT, Brown, TR, Doan, LL et al. (2012) Endocrine-disrupting chemicals and public health protection: a statement of principles from The Endocrine Society. Endocrinology 153, 40974110. doi: 10.1210/en.2012-1422.CrossRefGoogle Scholar
Delfosse, V, Grimaldi, M, le Maire, A et al. (2014) Nuclear receptor profiling of bisphenol-A and its halogenated analogues. Vitam Horm 94, 229251. doi: 10.1016/B978-0-12-800095-3.00009-2.CrossRefGoogle ScholarPubMed
Gonsioroski, A, Mourikes, VE & Flaws, JA (2020) Endocrine disruptors in water and their effects on the reproductive system. Int J Mol Sci 21, 1929. doi: 10.3390/ijms21061929 CrossRefGoogle ScholarPubMed
Gao, H, Yang, BJ, Li, N et al. (2015) Bisphenol A and hormone-associated cancers: current progress and perspectives. Med (Baltimore) 94, e211. doi: 10.1097/MD.0000000000000211.CrossRefGoogle Scholar
Rebuli, ME, Cao, J, Sluzas, E et al. (2014) Investigation of the effects of subchronic low dose oral exposure to bisphenol A (BPA) and ethinyl estradiol (EE) on estrogen receptor expression in the juvenile and adult female rat hypothalamus. Toxicol Sci 140, 190203. doi: 10.1093/toxsci/kfu074.CrossRefGoogle ScholarPubMed
Song, M, Liang, D, Liang, Y et al. (2014) Assessing developmental toxicity and estrogenic activity of halogenated bisphenol A on zebrafish (Danio rerio). Chemosphere 112, 275281. doi: 10.1016/j.chemosphere.2014.04.084.CrossRefGoogle ScholarPubMed
Brotons, JA, Olea-Serrano, MF, Villalobos, M et al. (1995) Xenoestrogens released from lacquer coatings in food cans. Environ Health Perspect 103, 608612. doi: 10.1289/ehp.95103608.CrossRefGoogle ScholarPubMed
Steinmetz, R, Mitchner, NA, Grant, A et al. (1998) The xenoestrogen bisphenol A induces growth, differentiation, and c-fos gene expression in the female reproductive tract. Endocrinology 139, 27412747. doi: 10.1210/endo.139.6.6027.CrossRefGoogle ScholarPubMed
Das, S & Thomas, P (1999) Pesticides interfere with the nongenomic action of a progestogen on meiotic maturation by binding to its plasma membrane receptor on fish oocytes. Endocrinology 140, 19531956. doi: 10.1210/endo.140.4.6781.CrossRefGoogle ScholarPubMed
Soundararajan, A, Prabu, P, Mohan, V et al. (2019) Novel insights of elevated systemic levels of bisphenol-A (BPA) linked to poor glycemic control, accelerated cellular senescence and insulin resistance in patients with type 2 diabetes. Mol Cell Biochem 458, 171183. doi: 10.1007/s11010-019-03540-9 CrossRefGoogle ScholarPubMed
Gong, H, Zhang, X, Cheng, B et al. (2013) Bisphenol A accelerates toxic amyloid formation of human islet amyloid polypeptide: a possible link between bisphenol A exposure and type 2 diabetes. PLoS One 8, e54198. doi: 10.1371/journal.pone.0054198 CrossRefGoogle ScholarPubMed
Chen, J, Zhong, L, Wu, J et al. (2018) A murine pancreatic islet cell-based screening for diabetogenic environmental chemicals. J Vis Exp 136, 57327. doi: 10.3791/57327 Google Scholar
Jiang, W, Ding, K, Huang, W et al. (2023) Potential effects of bisphenol A on diabetes mellitus and its chronic complications: a narrative review. Heliyon 9, e16340. doi: 10.1016/j.heliyon.2023.e16340 CrossRefGoogle ScholarPubMed
Nadal, A (2013) Obesity: fat from plastics? Linking bisphenol A exposure and obesity. Nat Rev Endocrinol 9, 910. doi: 10.1038/nrendo.2012.205 CrossRefGoogle ScholarPubMed
Moreno-Gómez-Toledano, R, Arenas, MI, Muñoz-Moreno, C et al. (2022) Comparison of the renal effects of bisphenol A in mice with and without experimental diabetes. Role of sexual dimorphism. Biochim Biophys Acta Mol Basis Dis 1868, 166296. doi: 10.1016/j.bbadis.2021.166296 CrossRefGoogle ScholarPubMed
Bulzomi, P & Marino, M (2011) Environmental endocrine disruptors: does a sex-related susceptibility exist? Front Biosci (Landmark Ed) 16, 24782498. doi: 10.2741/3867.CrossRefGoogle ScholarPubMed
Bolli, A & Marino, M (2011) Current and future development of estrogen receptor ligands: applications in estrogen-related cancers. Recent Pat Endocr Metab Immune Drug Discov 5, 210229. doi: 10.2174/187221411797265881.CrossRefGoogle ScholarPubMed
Wang, LH, Chen, LR & Chen, KH (2021) In vitro and vivo identification, metabolism and action of xenoestrogens: an overview. Int J Mol Sci 22, 4013. doi: 10.3390/ijms22084013.CrossRefGoogle ScholarPubMed
Song, Y, Chou, EL, Baecker, A et al. (2016) Endocrine-disrupting chemicals, risk of type 2 diabetes, and diabetes-related metabolic traits: a systematic review and meta-analysis. J Diabetes 8, 516532. doi: 10.1111/1753-0407.12325 CrossRefGoogle ScholarPubMed
Gore, AC, Chappell, VA, Fenton, SE et al. (2015) Executive summary to EDC-2: the endocrine society’s second scientific statement on endocrine-disrupting chemicals. Endocr Rev 36, 593602. doi: 10.1210/er.2015-1093 CrossRefGoogle ScholarPubMed
Vabre, P, Gatimel, N, Moreau, J et al. (2017) Environmental pollutants, a possible etiology for premature ovarian insufficiency: a narrative review of animal and human data. Environ Health 16, 37. doi: 10.1186/s12940-017-0242-4 CrossRefGoogle ScholarPubMed
Götz, F, Thieme, S & Dörner, G (2001) Female infertility--effect of perinatal xenoestrogen exposure on reproductive functions in animals and humans. Folia Histochem Cytobiol 2, 4043.Google Scholar
Grindler, NM, Allsworth, JE, Macones, GA et al. (2011) Persistent organic pollutants and early menopause in U.S. women. PLoS One. Bolli A, Marino M. Current and future development of estrogen receptor ligands: applications in estrogen-related cancers. Recent Pat Endocr Metab Immune Drug Discov 5, 210229. doi: 10.2174/187221411797265881.Google Scholar
Carwile, JL & Michels, KB (2011) Urinary bisphenol A and obesity: NHANES 2003–2006. Environ Res 111, 825830. doi: 10.1016/j.envres.2011.05.014 CrossRefGoogle Scholar
Riffee, JNJ, Wade, M, Sine, T et al. (2023) The link of environmental estrogens exposure to oxidative stress and their association with insulin- and exercise-induced glucose uptake. Obes Med 42, 100503. doi: 10.1016/j.obmed.2023.100503.CrossRefGoogle Scholar
Paterni, I, Granchi, C & Minutolo, F (2017) Risks and benefits related to alimentary exposure to xenoestrogens. Crit Rev Food Sci Nutr 57, 33843404. doi: 10.1080/10408398.2015.1126547.CrossRefGoogle ScholarPubMed
Palmlund, I (1996) Exposure to a xenoestrogen before birth: the diethylstilbestrol experience. J Psychosom Obstet Gynaecol 17, 7184. doi: 10.3109/01674829609025667.CrossRefGoogle ScholarPubMed
Vilahur, N, Bustamante, M, Byun, HM et al. (2014) Prenatal exposure to mixtures of xenoestrogens and repetitive element DNA methylation changes in human placenta. Environ Int 71, 8187. doi: 10.1016/j.envint.2014.06.006.CrossRefGoogle ScholarPubMed
García-Arevalo, M, Alonso-Magdalena, P, Rebelo Dos Santos, J et al. (2014) Exposure to bisphenol-A during pregnancy partially mimics the effects of a high-fat diet altering glucose homeostasis and gene expression in adult male mice. PLoS One 9, e100214. doi: 10.1371/journal.pone.0100214 CrossRefGoogle ScholarPubMed
Miura, R, Araki, A, Minatoya, M et al. (2019) An epigenome-wide analysis of cord blood DNA methylation reveals sex-specific effect of exposure to bisphenol A. Sci Rep 9, 12369. doi: 10.1038/s41598-019-48916-5 CrossRefGoogle ScholarPubMed
Yang, CZ, Yaniger, SI, Jordan, VC et al. (2011) Most plastic products release estrogenic chemicals: a potential health problem that can be solved. Environ Health Perspect 119, 989996. doi: 10.1289/ehp.1003220.CrossRefGoogle ScholarPubMed
Bittner, GD, Yang, CZ & Stoner, MA (2014) Estrogenic chemicals often leach from BPA-free plastic products that are replacements for BPA-containing polycarbonate products. Environ Health 13, 41. doi: 10.1186/1476-069X-13-41.CrossRefGoogle ScholarPubMed
Darnerud, PO, Eriksen, GS, Jóhannesson, T et al. (2001) Polybrominated diphenyl ethers: occurrence, dietary exposure, and toxicology. Environ Health Perspect 109, 4968. doi: 10.1289/ehp.01109s149 CrossRefGoogle Scholar
Hites, RA, Foran, JA, Schwager, SJ et al. (2004) Global assessment of polybrominated diphenyl ethers in farmed and wild salmon. Environ Sci Technol 38, 49454949. doi: 10.1021/es049548m CrossRefGoogle ScholarPubMed
Yokoyama, M, Origasa, H, Matsuzaki, M et al. (2007) Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): a randomised open-label, blinded endpoint analysis. Lancet 369, 10901098. doi: 10.1016/S0140-6736(07)60527-3. (published correction appears in Lancet. 2007 Jul 21 370(9583):220).CrossRefGoogle Scholar
Troesch, B, Eggersdorfer, M, Laviano, A et al. (2020) Expert opinion on benefits of long-chain n-3 fatty acids (DHA and EPA) in aging and clinical nutrition. Nutrients 12, 2555. doi: 10.3390/nu12092555 CrossRefGoogle ScholarPubMed
Hibbeln, JR, Davis, JM, Steer, C et al. (2007) Maternal seafood consumption in pregnancy and neurodevelopmental outcomes in childhood (ALSPAC study): an observational cohort study. Lancet 369, 578585. doi: 10.1016/S0140-6736(07)60277-3 CrossRefGoogle ScholarPubMed
Davidson, PW, Myers, GJ, Cox, C et al. (1995) Longitudinal neurodevelopmental study of Seychellois children following in utero exposure to methylmercury from maternal fish ingestion: outcomes at 19 and 29 months. Neurotoxicology 16, 677688.Google Scholar
Myers, GJ, Davidson, PW, Cox, C et al. (2003) Prenatal methylmercury exposure from ocean fish consumption in the Seychelles child development study. Lancet 361, 16861692. doi: 10.1016/S0140-6736(03)13371-5 CrossRefGoogle ScholarPubMed
Mukherjee, R, Pandya, P, Baxi, D et al. (2021) Endocrine disruptors-‘food’ for thought. Proc Zool Soc 74, 432442. doi: 10.1007/s12595-021-00414-1 CrossRefGoogle ScholarPubMed
Lee, A, Bensaada, S, Lamothe, V et al. (2022) Endocrine disruptors on and in fruits and vegetables: estimation of the potential exposure of the French population. Food Chem 373, 131513. doi: 10.1016/j.foodchem.2021.131513 CrossRefGoogle ScholarPubMed
Kuiper, GG, Lemmen, JG, Carlsson, B et al. (1998) Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor beta. Endocrinology 139, 42524263. doi: 10.1210/endo.139.10.6216.CrossRefGoogle ScholarPubMed