Next Article in Journal
Rapid Maxillary Expansion Treatment in Patients with Cleft Lip and Palate: A Survey on Clinical Experience in the European Cleft Centers
Previous Article in Journal
Pelvic Floor Muscle Training versus Functional Magnetic Stimulation for Stress Urinary Incontinence in Women: A Randomized Controlled Trial
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Endocrine Disruptors in Food, Estrobolome and Breast Cancer

1
Center for Integrative Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
2
Breast Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
3
Department of Ecological and Biological Sciences (DEB), Tuscia University, Largo dell’Università snc, 01100 Viterbo, Italy
4
Women’s Health Department, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(9), 3158; https://doi.org/10.3390/jcm12093158
Submission received: 9 March 2023 / Revised: 17 April 2023 / Accepted: 26 April 2023 / Published: 27 April 2023
(This article belongs to the Section Infectious Diseases)

Abstract

:
The microbiota is now recognized as one of the major players in human health and diseases, including cancer. Regarding breast cancer (BC), a clear link between microbiota and oncogenesis still needs to be confirmed. Yet, part of the bacterial gene mass inside the gut, constituting the so called “estrobolome”, influences sexual hormonal balance and, since the increased exposure to estrogens is associated with an increased risk, may impact on the onset, progression, and treatment of hormonal dependent cancers (which account for more than 70% of all BCs). The hormonal dependent BCs are also affected by environmental and dietary endocrine disruptors and phytoestrogens which interact with microbiota in a bidirectional way: on the one side disruptors can alter the composition and functions of the estrobolome, ad on the other the gut microbiota influences the metabolism of endocrine active food components. This review highlights the current evidence about the complex interplay between endocrine disruptors, phytoestrogens, microbiome, and BC, within the frames of a new “oncobiotic” perspective.

1. Introduction

Breast cancer (BC) is currently one of the most prevalent cancers, with an estimated number of 2.3 million new cases worldwide [1]. It represents the fifth most common cause of cancer-related deaths [2].
BC incidence is expected to increase further, particularly in low- income countries, due to the westernization of lifestyles (e.g., lack of physical activity and poor diet), and improved cancer detection [3]. Current projections indicate that by 2030, the number of new cases diagnosed will reach 2.7 million annually [4].
The World Health Organization (WHO) distinguishes at least 18 different histological BC types among a wide spectrum of tumors featuring different morphologies, molecular characteristics, and clinical behaviors [5]. Invasive BC can be categorized into molecular subtypes based on mRNA gene expression levels independently of histological subtypes. In 2000, Perou et al. identified four molecular subtypes from microarray gene expression data: Luminal, HER2-enriched, Basal-like, and Normal Breast-like [6]; further studies allowed to divide the Luminal group into two subgroups (Luminal A and B) [7,8,9,10,11].
Luminal A tumors are characterized by the presence of estrogen-receptor (ER) and/or progesterone-receptor (PR) and absence of HER2. This subtype [12,13] is associated to a low expression of genes related to cell proliferation and shows a better prognosis, compared to Luminal B tumors, which are ER positive but may be PR negative and/or HER2 positive.
Overall, 80% and 65% of patients are diagnosed with BC positive for estrogen receptor (ER) and progesterone receptor (PR), respectively [9].
A new classification has recently been proposed for HER 2 tumors with a score of 1+ or 2+ without amplification by the ISH method (in situ hybridization); these are nicknamed HER 2 low breast cancer and account for more than half of all breast cancer cases.
On the basis of the latest studies, it has been seen that this subcategory of tumors could benefit from new anti-HER 2 drugs. However, we are far from being able to define HER 2 low tumors as a separate clinical entity with its prognosis and specific features [14].
Validation of techniques to identify HER2 heterogeneity in order to effectively treat tumors with non-uniform HER2 expression is needed [15].
BC is a multifactorial disease, and several genetic and environmental aspects are recognized as risk factors for its onset and progression [16]. Among them, age, and modifiable factors such as obesity, type II diabetes, sedentary habits, alcohol, radiation, hormonal replacement therapy, and periodontal disease have direct implications on gut microbiota composition, so that recent studies have highlighted the association between microbial alterations and those risk factors for BC, through metabolic and immunitary pathways, hormonal balance, and cancer microenvironment [17,18,19].
Regarding the sexual hormonal balance, estrogens, and endocrine active compounds play a role in shaping the gut microbiome, potentially impacting the clinical management of hormone-dependent cancers [20].

2. Endocrine Disruptors, Phytoestrogens and Breast Cancer

An Endocrine Disruptor (ED) is defined by the U.S. Environmental Protection Agency (EPA) as “an exogenous agent that interferes with synthesis, secretion, transport, metabolism, binding action, or elimination of natural blood-borne hormones that are present in the body and are responsible for homeostasis, reproduction, and developmental process” [21].
Both estrogens and EDs, binding to estrogen receptors, elicit downstream gene activation and trigger intracellular signalling cascades [22] in a variety of tissues, thus affecting reproductive health and hormonal dependent cancers risk [23,24,25].
Endocrine disruptors are a group of highly heterogeneous molecules, grossly divided into synthetic and natural compounds (phytoestrogens).

2.1. Synthetic Endocrine Disruptors

The synthetic chemicals with endocrine activities have multiple uses, such as industrial solvents/lubricants (polychlorinated biphenyls (PCBs), polybrominated biphenyls (PBBs)), plastics (bisphenol A (BPA)), plasticizers (phthalates), pesticides (methoxychlor, chlorpyrifos, dichlorodiphenyltrichloroethane (DDT)), fungicides (vinclozolin), pharmaceutical agents (diethylstilbestrol (DES)) and heavy metals such as cadmium [25,26].
The most common pathways of exposure to EDs are by inhalation, food intake, transplacental and skin contact [25,27,28]. By these means, EDs enter the food chain and accumulate in animal tissues up to humans mainly in adipose tissue, since most of EDs are highly lipophilic [29,30,31].
The mechanisms of action of EDs include a variety of possible pathways involved in endocrine and reproductive systems: via nuclear receptors, nonnuclear steroid hormone receptors (e.g., membrane estrogen receptors (ERs)), nonsteroid receptors (e.g., neurotransmitter receptors such as serotonin, dopamine, norepinephrine), orphan receptors [e.g., aryl hydrocarbon receptor (AhR)], enzymatic pathways involved in steroid biosynthesis and/or metabolism [25].
Another mechanism is the aromatase up-regulation (e.g., phenolic EDs) and increased estradiol biosynthesis, which is linked to ER-positive breast cancer cell proliferation in vitro [32].
Furthermore, an epigenetic action, such as DNA methylation and/or acetylation and histone modifications, may be involved in mechanisms related to endocrine disruption [33,34,35].
The exposure to EDs has been related to multiple diseases, such as diabetes, metabolic syndrome, obesity, cardiovascular and neurological disorders [29,30,31,32,33,34,35,36,37]. Some EDs such as bisphenol A (BPA), dichlorodiphenyltrichloroethane (DDT) and polychlorinated biphenyls (PCBs) are also associated with infertility and cancer [29,30,31,32,33,34,35,36,37,38,39,40].
According to the International Agency for Research on Cancer (IARC) classification, some of the EDs (BPA, DDT and PCBs) have key characteristics of human carcinogens, since they can alter cell proliferation, cell death or nutrient supply; are genotoxic; have immunosuppressive activity; induce epigenetic alterations, oxidative stress and chronic inflammation [39]. In addition, BPA by interacting with the estrogen receptor-α (ERα), induces cell proliferation and reduces apoptosis rate, affecting the prognosis of BC patients [40,41,42].
A growing number of studies have investigated the correlations between EDs and BC onset and progression [43]. Breast tissue is particularly susceptible to carcinogenic effects during the third trimester of the first pregnancy, and prolonged exposure to low levels of EDs [44,45,46] can raise the risk of developing cancer in the following years [47,48].
Some pesticides, including DDT, dichloro-diphenyl-dichloroethylene (DDE), aldrin, and lindane, have been linked in pre- and post-menopausal women to a higher risk of BC [49,50], either estrogen receptor-positive (-hexachlorocyclohexane and Pentachlorothioanisole) [51] or HER2-positive tumors (DDT) [52,53,54]. Among the heavy metals, cadmium was positively associated with BC [55,56].
Interestingly, women with an altered body composition and an excess of fat mass have shown a greater likelihood of BC after exposure to PCB [57], due to the lipophilic nature of these molecules.
Some EDs, such as Bisphenol S (BPS), are also involved in enhancing the progression and the metastatic spread of BC cells, by inducing tumor proliferation and epithelial-mesenchymal transition [58,59]. The Interplay between endocrine disruptors and microbiota with potential drivers of BC are summarized in Table 1.

2.2. Phytoestrogens

Due to their chemical structures and/or activities similar to 17-estradiol (E2) [38,67,68], some plant-derived polyphenolic non-steroidal substances, defined phytoestrogens, are classified as endocrine disruptors, with both potentially favorable (reduced risk of osteoporosis, heart disease, and menopausal symptoms) and harmful health consequences [69,70].
In epidemiological studies, Asian populations who consume on average much more soy products than Western populations, have lower rates of hormone-dependent breast and endometrial cancers [71] and a lower incidence of menopausal symptoms and osteoporosis. Soy is the main dietary source of isoflavones. Isoflavones have a chemical structure similar to the human hormone oestrogen. However, they bind to the body’s oestrogen receptors differently, and function differently. Activation of some receptors seems to promote cell growth, but isoflavones more often bind to oestrogen receptors with other effects, potentially acting as a tumour suppressor [71].
Different kinds of oestrogen receptors are present in different parts of the body. Activation of some receptors seems to promote cell growth. But studies suggest that isoflavones more often bind to oestrogen receptors with other effects, potentially acting as a tumour suppressor. Nevertheless, in Asian immigrants living in Western nations, whose diet includes more proteins and lipids and less fibers and soy, the risks for hormone-dependent cancers reach the same levels as the western population [72].
The main groups of phytoestrogens are lignans, coumestans, stilbenes and isoflavones.
Lignans, as components of plant cell walls, are found in many fiber-rich foods such as seeds (flax, pumpkin, sunflower, and sesame), whole grains (such as rye, oat, and barley), bran (such as wheat, oat, and rye), beans, fruits (especially berries), and cruciferous vegetables such as broccoli and cabbage [73].
The richest dietary source of plant lignans is flaxseed (Linum usatissimum), and crushing or milling flaxseed can increase lignan bioavailability [74].
Compared to isoflavones and lignans, coumestans are less prevalent in the human diet. Coumestans are primarily found in legume shoots and sprouts, primarily in clover and alfalfa, though small amounts have also been found in spinach and brussel sprouts [75]. Coumestrol is also found in trace levels in a variety of legumes, including split peas, pinto beans, lima beans, and soybean sprouts [75].
The most prevalent and studied stilbene, resveratrol, may be found in a number of plants and acts as a phytoalexin to ward off fungus infections. The skin of grapes (Vitis vinifera), red wine, and other highly pigmented fruit juices are the most recognized sources of resveratrol. Resveratrol is also present in pistachios, notably the papery skin surrounding the nut, and peanuts (Arachis). While flavonoids and resveratrol both have vascular effects that are frequently addressed, only the trans isomers of resveratrol have been found to have some phytoestrogenic effects [76].
Isoflavones are present in berries, wine, grains, and nuts, but are most abundant in soybeans, soy products, and other legumes [67,68].
Phytoestrogens, particularly isoflavones, exhibit both agonistic and antagonistic effects on ERβ and ERα receptors, depending on their concentration and affinity for various estrogen receptors [77]. This mechanism explains why phytoestrogens have a dual impact in ER-positive breast cancer cells, stimulating growth at low doses while inhibiting development at higher concentrations [78]. Coumestrol, genistein, and equol have a stronger affinity for ERβ [79,80].
Overall, phytoestrogens and their analogs inhibit cell cycle progression across different breast carcinomas by reducing mRNA or protein expression levels of cyclin (D1, E) and CDK (1, 2, 4, 6) and enhancing their inhibitors (p21, p27, p57) and tumor suppressor genes (APC, ATM, PTEN, SERPINB5) [73]. Even isoflavones, lignans, and resveratrol analogs influence cell cycle regulator expression, impacting different kinds of BC cell lines in vitro [81].
They also suppress the expression of oncogenic cyclin D1, as well as raise the levels of a variety of cyclin-dependent kinase inhibitors (p21, p27, and p57). Phytoestrogens, analogues, and derivatives may potentially influence BC behaviour, by interfering with estrogen production and metabolism as well as showing antiangiogenic, antimetastatic, and epigenetic effects. Furthermore, these bioactive molecules have the potential to reverse multi-drug resistance [81]. The benefits of phytoestrogens on human health, and particularly in BC patients, may also depend on their metabolism affected by the host’s microbiota present in the small and large intestine. For instance, genistein, equol, enterolignans, urolithins and other metabolites with higher binding affinity for estrogen receptors are more likely to yield beneficial effects.
Despite several research, the topic of whether phytoestrogens are useful or hurtful to people with BC remains unanswered: The answers are challenging and may vary with age, health state, and even gut microbial composition [82] (Table 2).

3. Estrobolome

The gut microbiota regulates the levels and bioavailability of estrogens, steroid hormones, and cytokines [94], all of which have a role in the development, progression, and outcome of the majority of BCs [95,96,97,98]. In addition to steroid hormones, BC may be influenced by adipose tissue hormones such as leptin and insulin, which are also regulated by intestinal microbiota.
Two main pathways have been identified through which microbiome influences the sexual hormonal balance. In the deconjugation-independent pathway, some phytoestrogens contained in food, such as plant lignans, are metabolized by specific intestinal bacteria into bioactive compounds. In the deconjugation-dependent pathway, several genera such as Collinsella, Edwardsiella, Alistipes, Bacteroides, Bifidobacterium, Citrobacter, Clostridium, Dermabacter, Escherichia, Faecalibacterium, Lactobacillus, Marvinbryantia, Propionibacterium, Roseburia, Tannerella, constituting the so called “estrobolome”, by the means of hydrolytic enzymes such as β-glucuronidases and β-glucosidases, can deconjugate estrogens excreted by the liver into the intestinal lumen as well as endocrine active food components, increasing their reabsorption through the entero-hepatic circulation [22,83,84,85]. Progesterone and testosterone bioavailability can also be affected by the sulfatase activity of certain gut microrganisms which convert circulating steroids into active hormones [85,86,87].
In premenopausal women following a “Western diet”, estrogen levels were found to be three times higher in feces and 15% to 20% lower in serum compared to a population of vegetarians eating a high fiber, moderate fat diet [99]. In another research, Asian immigrants showed 30% lower systemic estrogen levels compared to a similar population of American women consuming a diet higher in fats [100], probably due to the estrobolome composition, even though other variables such as lifestyle and oral supplements may also play a role [101]. Changes in the estrobolome composition induced by diet, physical activity, antibiotics and chemotherapeutics affect the systemic levels of estrogen and its metabolites through the entero-hepatic circulation [102] and this mechanism has been related to cancer progression in hormonal dependent BC patients and survivors [86,103,104].
Several studies showed that cancerogenesis can also be promoted by enhanced local exposures of breast tissue to hormonal triggers, both from estrogen and progesterone metabolites: an abundance of β-glucuronidase signalling has been found in nipple aspirate fluid of BC survivors [105], while BC tissue shows higher concentrations of estrogen metabolites compared to normal breast tissue [100,106]. Among the possible mechanisms leading to an increased production of progesterone metabolites in tumour microenvironment, Bacillus cereus seem to play a role in promoting cancer cells proliferation [107,108,109,110]. Among the gram-negative family of Sphingomonadaceae, Sphyngomonas yanoikuyae, relatively enriched in paired normal breast tissue as compared to cancer tissue [111], has shown the ability to digest monocyclic compounds and degrade estrogens within the breast tissue [112], which could interfere with cancerogenesis through the local estrogen bioavailability.

4. Interplay between Human Microbiota, Endocrine Disruptors, and Phytoestrogens

The complex relationship between microbiota and endocrine active compounds derived from diet act in a bidirectional way: enteric commensals can metabolize EDs into biologically active or inactive forms, while EDs may selectively induce the growth of specific bacterial populations.
The biotransformation of lignans is an intriguing example of how deeply the microbiota affect the metabolism of some xenobiotics: for instance, anhydrosecoisolariciresinol is converted by the gram-positive Clostridium methoxybenzovorans [60,61], the secoisolariciresinol diglucoside by B. pseudocatenulatum WC 401 and other Bifidobacterium strains through deglucosylation [62]. Among prenylflavonoids, a subgroup of chalcones and flavanones, the most significant are xanthohumol (XN) and desmethyxanthohumol (DMX) derived from hops, which are widely used in beer industry [113]: XN’s metabolite 8-prenylnaringenin (8-PN), produced in the gut by the commensal [114], is one of the most potent phytoestrogens [88], with a noticeable affinity for the ERα receptor [115,116].
These dietary-induced interactions between gut microbiota and hormonal balance may lead to a dysbiosis, thus affecting human health and diseases [117].

5. Role of the Endocrine Disruptors on Microbiota Composition

Several studies underline the association between EDs exposure and metabolic disorders, diabetes, obesity, and some neurobehavioral disorders [118], which have been related to gut dysbiosis, suggesting a role of gut microbiome and its products (post-biotics) as mediators of the effects induced by EDs in human metabolism [65].
Both the exposure to EDs and their bioactive metabolites may disrupt the microbiota composition and lead to dysbiosis [66], but also alter the microbiome functions and metabolic activities [119]. According to data from animal models, changes in the gut microbiota may have an im-pact on the host’s hepatic enzyme levels in addition to the levels of microbial enzymes [120].
Several EDs have been proved to promote dysbiosis or avoid bacterial growth both in vitro and in vivo [65], suggesting a significant influence on gut colonization with a consequence on host health. Furthermore, a “leaky gut” wall facilitates circulating EDs to flow into the intestinal milieu directly and, interacting with the enteric nervous system, could impact the composition and functions of the gut microbiota [121,122,123].
Clavel et al. showed that the isoflavone daidzein and its metabolites modulate the composition of gut microbiota in postmenopausal women after two months supplementation, finding an association between the equol production and the increase of the F. prausnitzii and Lactobacillus-Enterococcus groups [124]. In a long-term study exploring the effects of isoflavones supplementation on the faecal microbiota of healthy menopausal women, a significant change of microbial populations was recorded, but without any difference between equol-producers and non-producers [63].
In another study a 4 weeks supplementation with a pomegranate extract, ellagitannin and its metabolites reveal changes in the composition of gut microbiota (Actinobacter, Firmicutes, and Verrucomicrobia) on healthy subjects [125].
Luo et al. investigated the in vivo anti-obesity effect of flaxseed gums (FG) in obese rats and found the FG diet decreased the relative abundance of Clostridiales and increased the Clostridium, Sutterella, Veillonella, Burkholderiales and Enterobacteriaceae family in their gut microbiota [126]. The supplementation with syringaresinol, a plant lignan, increases the Firmicutes/Bacteroidetes ratio in an aging mouse model [64].
The resveratrol mechanisms of action are largely attributed to the modulation of gut microbiota and its metabolites. An in vitro study demonstrated a different conversion of trans-resveratrol into dihydroresveratrol, 3,4′–dihydroxybibenzyl, also known as lunularin, and 3,4′-dihydroxy-trans-stilbene, depending on the bacterial diversity of each individual’s faecal samples [89]. Chen et al. (2016) observed that modulation of gut microbiota induced by resveratrol reduced the levels of trimethylamine-N-oxide (TMAO) by inhibiting microbial trimethylamine (TMA) production and increased hepatic bile acid (BA) de novo synthesis [90]. An increase in Bacteroides/Firmicutes ratio was also observed in vivo after resveratrol supplementation in animal studies along with other effects, such as anti-diabetic effect [91], improved carbohydrate metabolism [92] and glucose homeostasis [93]. Giuliani et al., using an advanced gastrointestinal stimulator, showed that an extract containing a combination of t-resveratrol and ε-vinifrin induced changes in microbial functions and composition together with a strong decrease in the levels of SCFA and NH4+ [127].

6. Different Metabolic Pathways of Endocrine Disruptors Depending on Gut Microbiota

Gut microbiota are crucial in the conversion of EDs and phytoestrogens, such as isoflavones, ellagitannins, and lignans, into compounds with biological activity (equol, urolithins, and enterolignans, respectively) [61,128].
The enzymatic degradation of plant lignans, such as secoisolariciresinol, into phytoestrogens enterodiol and enterolactone by various gut bacteria, such as Eggerthella lenta and Peptostreptococcus productus, provides a model for the deconjugation-independent process [124]. Enterodiol and enterolactone may serve as selective estrogen modulators with anticancer properties [19,129] and a favorable prognostic impact in postmenopausal BC patients [130].
Van de Wiele et al. [131] reported that colonic microbiota can metabolize polyaromatic hydrocarbons into 1-hydroxy pyrene and 7-hydroxybenzo[a]pyrene, biologically active estrogen metabolites.
A recent review of Velmurugan et al. [66] focused on the role of gut microbiota in glucose dysregulation, glucose intolerance and insulin resistance induced by several classes of EDs from plastics, pesticides, synthetic fertilizers, electronic waste and food additives. They included bisphenols, dioxins, phthalates, organochlorines, organophosphates, fungicides, polychlorinated biphenyls and polychlorinated dibenzofurans, and other waste pollutants.
On the other hand, hyperglycemia induces changes of microbiota composition, favoring the growth of non-commensal germs, at the expense of beneficial phyla such as Bacilli (e.g., Lactobacillus), Bacteroidetes, Proteobacteria and Actinobacteria [66]. Lactobacilli can reduce pesticide toxicity and protect against EDs-induced oxidative stress by limiting contaminant absorption in the gut, strengthening tight junctions in the intestinal barrier, and activating host immunity [132]. Exposure to EDs, such as polychlorinated biphenyls, may impair intestinal permeability by suppressing the expression of tight junction proteins [133,134].
Gut dysbiosis is linked with many disorders such as obesity, diabetes, endocrine and immunological diseases [117,135,136,137,138,139,140], which have been proven as risk factors for BC in both pre- and post-menopausal women [141,142].
Furthermore, all major classes of EDs (bisphenols, phthalates, polychlorinated biphenyls, organochlorine pesticides, dioxins, and parabens) may increase the risk of obesity, developing insulin resistance and diabetes [143] by enhancing adipogenesis via hormone regulation of food intake, appetite, and disruption of pancreatic β-cell function [144,145,146,147,148,149]. Even the fungicide tributyltin, which has been shown to reduce gut microbial richness and microbiome composition in mice [150], stimulates adipogenesis by interacting with nuclear PPAR γ and its heteromeric companion retinoid X receptor.
The interplay between EDs and human microbiota affects BC risk and clinical management not only through the sexual hormonal balance, but also through the innate as well as the acquired immunity [132,151,152,153,154], but these fundamental pathways are beyond the topic of the present review.
Beside this, a plethora of studies show that the gut microbiome affects the side effects, the toxicity and the outcomes of anticancer treatments such as radiation therapy, chemotherapy, immunotherapy and hormone therapy.
On the other hand, anticancer agents such as letrozole, an aromatase inhibitor, are associated with a time-dependent reduction of phylogenetic richness in the gut microbiota and a significant decrease in overall species [107].
Based on these results, the gut microbiota could become a key part of a microbiota-host-cancer triad as a new paradigm in order to better predict patients’ response to therapies and build a more tailored approach to cancer patients.

7. Conclusions

Endocrine disruptors and phytoestrogens interact with the human microbiota both at the intestinal and the breast tissue levels, affecting estrogens’ balance, bioavailability, and functions. This complex interplay results in a modification of BC cells behaviours, at least for hormonal dependent tumors, which account for more than 70% of cases globally.
A better understanding of this interplay, as well as the chance of modulating the exposure to EDs and targeting the microbiome composition (via dietary interventions and probiotics) could pave the way to a new oncobiotic approach in order to improve the clinical management of BC patients.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data are available upon request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Sharma, R. Breast Cancer Incidence, Mortality and Mortality-to-Incidence Ratio (MIR) Are Associated with Human Development, 1990–2016: Evidence from Global Burden of Disease Study 2016. Breast Cancer 2019, 26, 428–445. [Google Scholar] [CrossRef] [PubMed]
  3. Łukasiewicz, S.; Czeczelewski, M.; Forma, A.; Baj, J.; Sitarz, R.; Stanisławek, A. Breast Cancer-Epidemiology, Risk Factors, Classification, Prognostic Markers, and Current Treatment Strategies—An Updated Review. Cancers 2021, 13, 4287. [Google Scholar] [CrossRef] [PubMed]
  4. Bray, F.; Jemal, A.; Grey, N.; Ferlay, J.; Forman, D. Global Cancer Transitions According to the Human Development Index (2008–2030): A Population-Based Study. Lancet Oncol. 2012, 13, 790–801. [Google Scholar] [CrossRef] [PubMed]
  5. Eble, J.N.; Tavassoli, F.A.; Devilee, P. (Eds.) Pathology and Genetics of Tumours of the Breast and Female Genital Organs; IARC: Lyon, France, 2003. [Google Scholar]
  6. Perou, C.M.; Sørlie, T.; Eisen, M.B.; van de Rijn, M.; Jeffrey, S.S.; Rees, C.A.; Pollack, J.R.; Ross, D.T.; Johnsen, H.; Akslen, L.A.; et al. Molecular Portraits of Human Breast Tumours. Nature 2000, 406, 747–752. [Google Scholar] [CrossRef]
  7. Sørlie, T.; Perou, C.M.; Tibshirani, R.; Aas, T.; Geisler, S.; Johnsen, H.; Hastie, T.; Eisen, M.B.; van de Rijn, M.; Jeffrey, S.S.; et al. Gene Expression Patterns of Breast Carcinomas Distinguish Tumor Subclasses with Clinical Implications. Proc. Natl. Acad. Sci. USA 2001, 98, 10869–10874. [Google Scholar] [CrossRef]
  8. Prat, A.; Perou, C.M. Deconstructing the Molecular Portraits of Breast Cancer. Mol. Oncol. 2011, 5, 5–23. [Google Scholar] [CrossRef] [PubMed]
  9. Howlader, N.; Altekruse, S.F.; Li, C.I.; Chen, V.W.; Clarke, C.A.; Ries, L.A.G.; Cronin, K.A. US Incidence of Breast Cancer Subtypes Defined by Joint Hormone Receptor and HER2 Status. J. Natl. Cancer Inst. 2014, 106, dju055. [Google Scholar] [CrossRef] [PubMed]
  10. Weigelt, B.; Geyer, F.C.; Reis-Filho, J.S. Histological Types of Breast Cancer: How Special Are They? Mol. Oncol. 2010, 4, 192–208. [Google Scholar] [CrossRef]
  11. Makki, J. Diversity of Breast Carcinoma: Histological Subtypes and Clinical Relevance. Clin. Med. Insights Pathol. 2015, 8, 23–31. [Google Scholar] [CrossRef] [PubMed]
  12. Weigelt, B.; Baehner, F.L.; Reis-Filho, J.S. The Contribution of Gene Expression Profiling to Breast Cancer Classification, Prognostication and Prediction: A Retrospective of the Last Decade. J. Pathol. 2010, 220, 263–280. [Google Scholar] [CrossRef]
  13. Prat, A.; Cheang, M.C.U.; Martín, M.; Parker, J.S.; Carrasco, E.; Caballero, R.; Tyldesley, S.; Gelmon, K.; Bernard, P.S.; Nielsen, T.O.; et al. Prognostic Significance of Progesterone Receptor-Positive Tumor Cells within Immunohistochemically Defined Luminal A Breast Cancer. J. Clin. Oncol. 2013, 31, 203–209. [Google Scholar] [CrossRef]
  14. Tarantino, P.; Hamilton, E.; Tolaney, S.M.; Cortes, J.; Morganti, S.; Ferraro, E.; Marra, A.; Viale, G.; Trapani, D.; Cardoso, F.; et al. HER2-Low Breast Cancer: Pathological and Clinical Landscape. J. Clin. Oncol. 2020, 38, 1951–1962. [Google Scholar] [CrossRef] [PubMed]
  15. Hamilton, E.; Shastry, M.; Shiller, S.M.; Ren, R. Targeting HER2 heterogeneity in breast cancer. Cancer Treat. Rev. 2021, 100, 102286. [Google Scholar] [CrossRef]
  16. Loibl, S.; Poortmans, P.; Morrow, M.; Denkert, C.; Curigliano, G. Breast Cancer. Lancet 2021, 397, 1750–1769. [Google Scholar] [CrossRef]
  17. Kovács, T.; Mikó, E.; Ujlaki, G.; Sári, Z.; Bai, P. The Microbiome as a Component of the Tumor Microenvironment. Adv. Exp. Med. Biol. 2020, 1225, 137–153. [Google Scholar] [CrossRef] [PubMed]
  18. Costantini, L.; Magno, S.; Albanese, D.; Donati, C.; Molinari, R.; Filippone, A.; Masetti, R.; Merendino, N. Characterization of Human Breast Tissue Microbiota from Core Needle Biopsies through the Analysis of Multi Hypervariable 16S-RRNA Gene Regions. Sci. Rep. 2018, 8, 16893. [Google Scholar] [CrossRef]
  19. Eslami-S, Z.; Majidzadeh-A, K.; Halvaei, S.; Babapirali, F.; Esmaeili, R. Microbiome and Breast Cancer: New Role for an Ancient Population. Front. Oncol. 2020, 10, 120. [Google Scholar] [CrossRef]
  20. Kwa, M.; Plottel, C.S.; Blaser, M.J.; Adams, S. The Intestinal Microbiome and Estrogen Receptor-Positive Female Breast Cancer. J. Natl. Cancer Inst. 2016, 108, djw029. [Google Scholar] [CrossRef] [PubMed]
  21. Kavlock, R.J.; Daston, G.P.; DeRosa, C.; Fenner-Crisp, P.; Gray, L.E.; Kaattari, S.; Lucier, G.; Luster, M.; Mac, M.J.; Maczka, C.; et al. Research Needs for the Risk Assessment of Health and Environmental Effects of Endocrine Disruptors: A Report of the U.S. EPA-Sponsored Workshop. Environ. Health Perspect. 1996, 104 (Suppl. 4), 715–740. [Google Scholar] [CrossRef]
  22. Rietjens, I.M.C.M.; Louisse, J.; Beekmann, K. The Potential Health Effects of Dietary Phytoestrogens. Br. J. Pharmacol. 2017, 174, 1263–1280. [Google Scholar] [CrossRef]
  23. Muhleisen, A.L.; Herbst-Kralovetz, M.M. Menopause and the Vaginal Microbiome. Maturitas 2016, 91, 42–50. [Google Scholar] [CrossRef] [PubMed]
  24. Toran-Allerand, C.D.; Miranda, R.C.; Bentham, W.D.; Sohrabji, F.; Brown, T.J.; Hochberg, R.B.; MacLusky, N.J. Estrogen Receptors Colocalize with Low-Affinity Nerve Growth Factor Receptors in Cholinergic Neurons of the Basal Forebrain. Proc. Natl. Acad. Sci. USA 1992, 89, 4668–4672. [Google Scholar] [CrossRef]
  25. Kabir, E.R.; Rahman, M.S.; Rahman, I. A Review on Endocrine Disruptors and Their Possible Impacts on Human Health. Environ. Toxicol. Pharmacol. 2015, 40, 241–258. [Google Scholar] [CrossRef] [PubMed]
  26. Monneret, C. What Is an Endocrine Disruptor? C. R. Biol. 2017, 340, 403–405. [Google Scholar] [CrossRef] [PubMed]
  27. Gore, A.C.; Chappell, V.A.; Fenton, S.E.; Flaws, J.A.; Nadal, A.; Prins, G.S.; Toppari, J.; Zoeller, R.T. EDC-2: The Endocrine Society’s Second Scientific Statement on Endocrine-Disrupting Chemicals. Endocr. Rev. 2015, 36, E1–E150. [Google Scholar] [CrossRef]
  28. Cohn, B.A.; La Merrill, M.A.; Krigbaum, N.Y.; Wang, M.; Park, J.-S.; Petreas, M.; Yeh, G.; Hovey, R.C.; Zimmermann, L.; Cirillo, P.M. In Utero Exposure to Poly- and Perfluoroalkyl Substances (PFASs) and Subsequent Breast Cancer. Reprod. Toxicol. 2020, 92, 112–119. [Google Scholar] [CrossRef]
  29. Heindel, J.J.; Newbold, R.; Schug, T.T. Endocrine Disruptors and Obesity. Nat. Rev. Endocrinol. 2015, 11, 653–661. [Google Scholar] [CrossRef]
  30. Sargis, R.M. Metabolic Disruption in Context: Clinical Avenues for Synergistic Perturbations in Energy Homeostasis by Endocrine Disrupting Chemicals. Endocr. Disruptors 2015, 3, e1080788. [Google Scholar] [CrossRef]
  31. Barouki, R. Endocrine Disruptors: Revisiting Concepts and Dogma in Toxicology. C. R. Biol. 2017, 340, 410–413. [Google Scholar] [CrossRef]
  32. Williams, G.P.; Darbre, P.D. Low-Dose Environmental Endocrine Disruptors, Increase Aromatase Activity, Estradiol Biosynthesis and Cell Proliferation in Human Breast Cells. Mol. Cell. Endocrinol. 2019, 486, 55–64. [Google Scholar] [CrossRef] [PubMed]
  33. Zama, A.M.; Uzumcu, M. Epigenetic Effects of Endocrine-Disrupting Chemicals on Female Reproduction: An Ovarian Perspective. Front. Neuroendocrinol. 2010, 31, 420–439. [Google Scholar] [CrossRef]
  34. Giulivo, M.; Lopez de Alda, M.; Capri, E.; Barceló, D. Human Exposure to Endocrine Disrupting Compounds: Their Role in Reproductive Systems, Metabolic Syndrome and Breast Cancer. A Review. Environ. Res. 2016, 151, 251–264. [Google Scholar] [CrossRef] [PubMed]
  35. Burks, H.; Pashos, N.; Martin, E.; Mclachlan, J.; Bunnell, B.; Burow, M. Endocrine Disruptors and the Tumor Microenvironment: A New Paradigm in Breast Cancer Biology. Mol. Cell. Endocrinol. 2017, 457, 13–19. [Google Scholar] [CrossRef]
  36. Quagliariello, V.; Rossetti, S.; Cavaliere, C.; Di Palo, R.; Lamantia, E.; Castaldo, L.; Nocerino, F.; Ametrano, G.; Cappuccio, F.; Malzone, G.; et al. Metabolic Syndrome, Endocrine Disruptors and Prostate Cancer Associations: Biochemical and Pathophysiological Evidences. Oncotarget 2017, 8, 30606–30616. [Google Scholar] [CrossRef]
  37. Rodgers, K.M.; Udesky, J.O.; Rudel, R.A.; Brody, J.G. Environmental Chemicals and Breast Cancer: An Updated Review of Epidemiological Literature Informed by Biological Mechanisms. Environ. Res. 2018, 160, 152–182. [Google Scholar] [CrossRef]
  38. Diamanti-Kandarakis, E.; Bourguignon, J.-P.; Giudice, L.C.; Hauser, R.; Prins, G.S.; Soto, A.M.; Zoeller, R.T.; Gore, A.C. Endocrine-Disrupting Chemicals: An Endocrine Society Scientific Statement. Endocr. Rev. 2009, 30, 293–342. [Google Scholar] [CrossRef]
  39. Calaf, G.M.; Ponce-Cusi, R.; Aguayo, F.; Muñoz, J.P.; Bleak, T.C. Endocrine Disruptors from the Environment Affecting Breast Cancer. Oncol. Lett. 2020, 20, 19–32. [Google Scholar] [CrossRef] [PubMed]
  40. Sengupta, S.; Obiorah, I.; Maximov, P.Y.; Curpan, R.; Jordan, V.C. Molecular Mechanism of Action of Bisphenol and Bisphenol A Mediated by Oestrogen Receptor Alpha in Growth and Apoptosis of Breast Cancer Cells. Br. J. Pharmacol. 2013, 169, 167–178. [Google Scholar] [CrossRef] [PubMed]
  41. Mlynarcikova, A.; Macho, L.; Fickova, M. Bisphenol A Alone or in Combination with Estradiol Modulates Cell Cycle- and Apoptosis-Related Proteins and Genes in MCF7 Cells. Endocr. Regul. 2013, 47, 189–199. [Google Scholar] [CrossRef]
  42. Katchy, A.; Pinto, C.; Jonsson, P.; Nguyen-Vu, T.; Pandelova, M.; Riu, A.; Schramm, K.-W.; Samarov, D.; Gustafsson, J.-Å.; Bondesson, M.; et al. Coexposure to Phytoestrogens and Bisphenol a Mimics Estrogenic Effects in an Additive Manner. Toxicol. Sci. 2014, 138, 21–35. [Google Scholar] [CrossRef] [PubMed]
  43. Rocha, P.R.S.; Oliveira, V.D.; Vasques, C.I.; Dos Reis, P.E.D.; Amato, A.A. Exposure to Endocrine Disruptors and Risk of Breast Cancer: A Systematic Review. Crit. Rev. Oncol. Hematol. 2021, 161, 103330. [Google Scholar] [CrossRef]
  44. Javed, A.; Lteif, A. Development of the Human Breast. Semin. Plast. Surg. 2013, 27, 5–12. [Google Scholar] [CrossRef] [PubMed]
  45. Gulledge, C.C.; Burow, M.E.; McLachlan, J.A. Endocrine Disruption in Sexual Differentiation and Puberty. What Do Pseudohermaphroditic Polar Bears Have to Do with the Practice of Pediatrics? Pediatr. Clin. N. Am. 2001, 48, 1223–1240. [Google Scholar] [CrossRef]
  46. Paulose, T.; Speroni, L.; Sonnenschein, C.; Soto, A.M. Estrogens in the Wrong Place at the Wrong Time: Fetal BPA Exposure and Mammary Cancer. Reprod. Toxicol. 2015, 54, 58–65. [Google Scholar] [CrossRef] [PubMed]
  47. Palmer, J.R.; Wise, L.A.; Hatch, E.E.; Troisi, R.; Titus-Ernstoff, L.; Strohsnitter, W.; Kaufman, R.; Herbst, A.L.; Noller, K.L.; Hyer, M.; et al. Prenatal Diethylstilbestrol Exposure and Risk of Breast Cancer. Cancer Epidemiol. Biomarkers Prev. 2006, 15, 1509–1514. [Google Scholar] [CrossRef] [PubMed]
  48. Thompson, W.D.; Janerich, D.T. Maternal Age at Birth and Risk of Breast Cancer in Daughters. Epidemiology 1990, 1, 101–106. [Google Scholar] [CrossRef]
  49. Arrebola, J.P.; Belhassen, H.; Artacho-Cordón, F.; Ghali, R.; Ghorbel, H.; Boussen, H.; Perez-Carrascosa, F.M.; Expósito, J.; Hedhili, A.; Olea, N. Risk of Female Breast Cancer and Serum Concentrations of Organochlorine Pesticides and Polychlorinated Biphenyls: A Case-Control Study in Tunisia. Sci. Total Environ. 2015, 520, 106–113. [Google Scholar] [CrossRef]
  50. Boada, L.D.; Zumbado, M.; Henríquez-Hernández, L.A.; Almeida-González, M.; Alvarez-León, E.E.; Serra-Majem, L.; Luzardo, O.P. Complex Organochlorine Pesticide Mixtures as Determinant Factor for Breast Cancer Risk: A Population-Based Case-Control Study in the Canary Islands (Spain). Environ. Health 2012, 11, 28. [Google Scholar] [CrossRef] [PubMed]
  51. Yang, J.-Z.; Wang, Z.-X.; Ma, L.-H.; Shen, X.-B.; Sun, Y.; Hu, D.-W.; Sun, L.-X. The Organochlorine Pesticides Residues in the Invasive Ductal Breast Cancer Patients. Environ. Toxicol. Pharmacol. 2015, 40, 698–703. [Google Scholar] [CrossRef] [PubMed]
  52. Cohn, B.A.; La Merrill, M.; Krigbaum, N.Y.; Yeh, G.; Park, J.-S.; Zimmermann, L.; Cirillo, P.M. DDT Exposure In Utero and Breast Cancer. J. Clin. Endocrinol. Metab. 2015, 100, 2865–2872. [Google Scholar] [CrossRef]
  53. Aronson, K.J.; Miller, A.B.; Woolcott, C.G.; Sterns, E.E.; McCready, D.R.; Lickley, L.A.; Fish, E.B.; Hiraki, G.Y.; Holloway, C.; Ross, T.; et al. Breast Adipose Tissue Concentrations of Polychlorinated Biphenyls and Other Organochlorines and Breast Cancer Risk. Cancer Epidemiol. Biomarkers Prev. 2000, 9, 55–63. [Google Scholar] [PubMed]
  54. Recio-Vega, R.; Velazco-Rodriguez, V.; Ocampo-Gómez, G.; Hernandez-Gonzalez, S.; Ruiz-Flores, P.; Lopez-Marquez, F. Serum Levels of Polychlorinated Biphenyls in Mexican Women and Breast Cancer Risk. J. Appl. Toxicol. 2011, 31, 270–278. [Google Scholar] [CrossRef]
  55. Eriksen, K.T.; McElroy, J.A.; Harrington, J.M.; Levine, K.E.; Pedersen, C.; Sørensen, M.; Tjønneland, A.; Meliker, J.R.; Raaschou-Nielsen, O. Urinary Cadmium and Breast Cancer: A Prospective Danish Cohort Study. J. Natl. Cancer Inst. 2017, 109, djw204. [Google Scholar] [CrossRef]
  56. Nagata, C.; Nagao, Y.; Nakamura, K.; Wada, K.; Tamai, Y.; Tsuji, M.; Yamamoto, S.; Kashiki, Y. Cadmium Exposure and the Risk of Breast Cancer in Japanese Women. Breast. Cancer Res. Treat. 2013, 138, 235–239. [Google Scholar] [CrossRef] [PubMed]
  57. Stellman, S.D.; Djordjevic, M.V.; Britton, J.A.; Muscat, J.E.; Citron, M.L.; Kemeny, M.; Busch, E.; Gong, L. Breast Cancer Risk in Relation to Adipose Concentrations of Organochlorine Pesticides and Polychlorinated Biphenyls in Long Island, New York. Cancer Epidemiol. Biomarkers Prev. 2000, 9, 1241–1249. [Google Scholar]
  58. Zhang, X.-L.; Liu, N.; Weng, S.-F.; Wang, H.-S. Bisphenol A Increases the Migration and Invasion of Triple-Negative Breast Cancer Cells via Oestrogen-Related Receptor Gamma. Basic Clin. Pharmacol. Toxicol. 2016, 119, 389–395. [Google Scholar] [CrossRef] [PubMed]
  59. Jenkins, S.; Wang, J.; Eltoum, I.; Desmond, R.; Lamartiniere, C.A. Chronic Oral Exposure to Bisphenol A Results in a Nonmonotonic Dose Response in Mammary Carcinogenesis and Metastasis in MMTV-ErbB2 Mice. Environ. Health Perspect. 2011, 119, 1604–1609. [Google Scholar] [CrossRef] [PubMed]
  60. Struijs, K.; Vincken, J.-P.; Gruppen, H. Bacterial Conversion of Secoisolariciresinol and Anhydrosecoisolariciresinol. J. Appl. Microbiol. 2009, 107, 308–317. [Google Scholar] [CrossRef] [PubMed]
  61. Gaya, P.; Medina, M.; Sánchez-Jiménez, A.; Landete, J.M. Phytoestrogen Metabolism by Adult Human Gut Microbiota. Molecules 2016, 21, 1034. [Google Scholar] [CrossRef] [PubMed]
  62. Roncaglia, L.; Amaretti, A.; Raimondi, S.; Leonardi, A.; Rossi, M. Role of Bifidobacteria in the Activation of the Lignan Secoisolariciresinol Diglucoside. Appl. Microbiol. Biotechnol. 2011, 92, 159–168. [Google Scholar] [CrossRef] [PubMed]
  63. Guadamuro, L.; Delgado, S.; Redruello, B.; Flórez, A.B.; Suárez, A.; Martínez-Camblor, P.; Mayo, B. Equol Status and Changes in Fecal Microbiota in Menopausal Women Receiving Long-Term Treatment for Menopause Symptoms with a Soy-Isoflavone Concentrate. Front. Microbiol. 2015, 6, 777. [Google Scholar] [CrossRef] [PubMed]
  64. Cho, S.-Y.; Kim, J.; Lee, J.H.; Sim, J.H.; Cho, D.-H.; Bae, I.-H.; Lee, H.; Seol, M.A.; Shin, H.M.; Kim, T.-J.; et al. Modulation of Gut Microbiota and Delayed Immunosenescence as a Result of Syringaresinol Consumption in Middle-Aged Mice. Sci. Rep. 2016, 6, 39026. [Google Scholar] [CrossRef] [PubMed]
  65. Gálvez-Ontiveros, Y.; Páez, S.; Monteagudo, C.; Rivas, A. Endocrine Disruptors in Food: Impact on Gut Microbiota and Metabolic Diseases. Nutrients 2020, 12, 1158. [Google Scholar] [CrossRef] [PubMed]
  66. Velmurugan, G.; Ramprasath, T.; Gilles, M.; Swaminathan, K.; Ramasamy, S. Gut Microbiota, Endocrine-Disrupting Chemicals, and the Diabetes Epidemic. Trends Endocrinol. Metab. 2017, 28, 612–625. [Google Scholar] [CrossRef] [PubMed]
  67. Dixon, R.A. Phytoestrogens. Annu. Rev. Plant Biol. 2004, 55, 225–261. [Google Scholar] [CrossRef]
  68. Michel, T.; Halabalaki, M.; Skaltsounis, A.-L. New Concepts, Experimental Approaches, and Dereplication Strategies for the Discovery of Novel Phytoestrogens from Natural Sources. Planta Med. 2013, 79, 514–532. [Google Scholar] [CrossRef]
  69. Lissin, L.W.; Cooke, J.P. Phytoestrogens and Cardiovascular Health. J. Am. Coll. Cardiol. 2000, 35, 1403–1410. [Google Scholar] [CrossRef]
  70. Turner, J.V.; Agatonovic-Kustrin, S.; Glass, B.D. Molecular Aspects of Phytoestrogen Selective Binding at Estrogen Receptors. J. Pharm. Sci. 2007, 96, 1879–1885. [Google Scholar] [CrossRef]
  71. Zhang, G.Q.; Chen, J.L.; Liu, Q.; Zhang, Y.; Zeng, H.; Zhao, Y. Soy intake is associated with lower endometrial cancer risk: A systematic review and meta-analysis of observational studies. Medicine 2015, 94, e2281. [Google Scholar] [CrossRef]
  72. Messina, M. Soy and health update: Evaluation of the clinical and epidemiologic literature. Nutrients 2016, 8, 754. [Google Scholar] [CrossRef] [PubMed]
  73. Meagher, L.P.; Bentz, E.K. Assessment of data on the lignan content of foods. J. Food Compos. Anal. 2000, 13, 935–947. [Google Scholar] [CrossRef]
  74. Kuijsten, A.; Arts, I.C.; van’t Veer, P.; Hollman, P.C. The relative bioavailability of enterolignans in humans is enhanced by milling and crushing of flaxseed. J. Nutr. 2005, 135, 2812–2816. [Google Scholar] [CrossRef] [PubMed]
  75. Poluzzi, E.; Piccinni, C.; Raschi, E.; Rampa, A.; Recanatini, M.; De Ponti, F. Phytoestrogens in postmenopause: The state of the art from a chemical, pharmacological and regulatory perspective. Curr. Med. Chem. 2014, 21, 417–436. [Google Scholar] [CrossRef] [PubMed]
  76. Bagchi, D.; Das, D.K.; Tosaki, A.; Bagchi, M.; Kothari, S.C. Benefits of resveratrol in women’s health. Drugs Exp. Clin. Res. 2001, 27, 233–248. [Google Scholar] [PubMed]
  77. Fitzpatrick, L.A. Phytoestrogens—Mechanism of Action and Effect on Bone Markers and Bone Mineral Density. Endocrinol. Metab. Clin. North Am. 2003, 32, 233–252. [Google Scholar] [CrossRef]
  78. Lecomte, S.; Demay, F.; Ferrière, F.; Pakdel, F. Phytochemicals Targeting Estrogen Receptors: Beneficial Rather Than Adverse Effects? Int. J. Mol. Sci. 2017, 18, 1381. [Google Scholar] [CrossRef]
  79. Soto, A.M.; Sonnenschein, C.; Chung, K.L.; Fernandez, M.F.; Olea, N.; Serrano, F.O. The E-SCREEN Assay as a Tool to Identify Estrogens: An Update on Estrogenic Environmental Pollutants. Environ. Health Perspect. 1995, 103 (Suppl. 7), 113–122. [Google Scholar] [CrossRef]
  80. Mueller, S.O.; Simon, S.; Chae, K.; Metzler, M.; Korach, K.S. Phytoestrogens and Their Human Metabolites Show Distinct Agonistic and Antagonistic Properties on Estrogen Receptor Alpha (ERalpha) and ERbeta in Human Cells. Toxicol. Sci. 2004, 80, 14–25. [Google Scholar] [CrossRef]
  81. Basu, P.; Maier, C. Phytoestrogens and Breast Cancer: In Vitro Anticancer Activities of Isoflavones, Lignans, Coumestans, Stilbenes and Their Analogs and Derivatives. Biomed Pharmacother. 2018, 107, 1648–1666. [Google Scholar] [CrossRef]
  82. Stojanov, S.; Kreft, S. Gut Microbiota and the Metabolism of Phytoestrogens. Rev. Bras. Farmacogn. 2020, 30, 145–154. [Google Scholar] [CrossRef]
  83. Arrieta, M.-C.; Stiemsma, L.T.; Amenyogbe, N.; Brown, E.M.; Finlay, B. The Intestinal Microbiome in Early Life: Health and Disease. Front. Immunol. 2014, 5, 427. [Google Scholar] [CrossRef] [PubMed]
  84. Selber-Hnatiw, S.; Sultana, T.; Tse, W.; Abdollahi, N.; Abdullah, S.; Al Rahbani, J.; Alazar, D.; Alrumhein, N.J.; Aprikian, S.; Arshad, R.; et al. Metabolic Networks of the Human Gut Microbiota. Microbiology 2020, 166, 96–119. [Google Scholar] [CrossRef]
  85. Flores, R.; Shi, J.; Fuhrman, B.; Xu, X.; Veenstra, T.D.; Gail, M.H.; Gajer, P.; Ravel, J.; Goedert, J.J. Fecal Microbial Determinants of Fecal and Systemic Estrogens and Estrogen Metabolites: A Cross-Sectional Study. J. Transl. Med. 2012, 10, 253. [Google Scholar] [CrossRef] [PubMed]
  86. Fuhrman, B.J.; Feigelson, H.S.; Flores, R.; Gail, M.H.; Xu, X.; Ravel, J.; Goedert, J.J. Associations of the Fecal Microbiome with Urinary Estrogens and Estrogen Metabolites in Postmenopausal Women. J. Clin. Endocrinol. Metab. 2014, 99, 4632–4640. [Google Scholar] [CrossRef] [PubMed]
  87. Goedert, J.J.; Jones, G.; Hua, X.; Xu, X.; Yu, G.; Flores, R.; Falk, R.T.; Gail, M.H.; Shi, J.; Ravel, J.; et al. Investigation of the Association between the Fecal Microbiota and Breast Cancer in Postmenopausal Women: A Population-Based Case-Control Pilot Study. J. Natl. Cancer Inst. 2015, 107, djv147. [Google Scholar] [CrossRef]
  88. Milligan, S.R.; Kalita, J.C.; Heyerick, A.; Rong, H.; De Cooman, L.; De Keukeleire, D. Identification of a Potent Phytoestrogen in Hops (Humulus Lupulus L.) and Beer. J. Clin. Endocrinol. Metab. 1999, 84, 2249–2252. [Google Scholar] [CrossRef]
  89. Bode, L.M.; Bunzel, D.; Huch, M.; Cho, G.-S.; Ruhland, D.; Bunzel, M.; Bub, A.; Franz, C.M.A.P.; Kulling, S.E. In Vivo and in Vitro Metabolism of Trans-Resveratrol by Human Gut Microbiota. Am. J. Clin. Nutr. 2013, 97, 295–309. [Google Scholar] [CrossRef]
  90. Chen, M.; Yi, L.; Zhang, Y.; Zhou, X.; Ran, L.; Yang, J.; Zhu, J.; Zhang, Q.; Mi, M. Resveratrol Attenuates Trimethylamine-N-Oxide (TMAO)-Induced Atherosclerosis by Regulating TMAO Synthesis and Bile Acid Metabolism via Remodeling of the Gut Microbiota. MBio 2016, 7, e02210-15. [Google Scholar] [CrossRef]
  91. Sung, M.M.; Kim, T.T.; Denou, E.; Soltys, C.-L.M.; Hamza, S.M.; Byrne, N.J.; Masson, G.; Park, H.; Wishart, D.S.; Madsen, K.L.; et al. Improved Glucose Homeostasis in Obese Mice Treated with Resveratrol Is Associated with Alterations in the Gut Microbiome. Diabetes 2017, 66, 418–425. [Google Scholar] [CrossRef]
  92. Sung, M.M.; Byrne, N.J.; Robertson, I.M.; Kim, T.T.; Samokhvalov, V.; Levasseur, J.; Soltys, C.-L.; Fung, D.; Tyreman, N.; Denou, E.; et al. Resveratrol Improves Exercise Performance and Skeletal Muscle Oxidative Capacity in Heart Failure. Am. J. Physiol. Heart Circ. Physiol. 2017, 312, H842–H853. [Google Scholar] [CrossRef]
  93. Kim, T.T.; Parajuli, N.; Sung, M.M.; Bairwa, S.C.; Levasseur, J.; Soltys, C.-L.M.; Wishart, D.S.; Madsen, K.; Schertzer, J.D.; Dyck, J.R.B. Fecal Transplant from Resveratrol-Fed Donors Improves Glycaemia and Cardiovascular Features of the Metabolic Syndrome in Mice. Am. J. Physiol. Endocrinol. Metab. 2018, 315, E511–E519. [Google Scholar] [CrossRef] [PubMed]
  94. Baker, J.M.; Al-Nakkash, L.; Herbst-Kralovetz, M.M. Estrogen-Gut Microbiome Axis: Physiological and Clinical Implications. Maturitas 2017, 103, 45–53. [Google Scholar] [CrossRef] [PubMed]
  95. Integrative HMP (iHMP) Research Network Consortium. The Integrative Human Microbiome Project. Nature 2019, 569, 641–648. [Google Scholar] [CrossRef]
  96. Cavuoto, P.; Fenech, M.F. A Review of Methionine Dependency and the Role of Methionine Restriction in Cancer Growth Control and Life-Span Extension. Cancer Treat. Rev. 2012, 38, 726–736. [Google Scholar] [CrossRef]
  97. Hoffman, R.M. Development of Recombinant Methioninase to Target the General Cancer-Specific Metabolic Defect of Methionine Dependence: A 40-Year Odyssey. Expert Opin. Biol. Ther. 2015, 15, 21–31. [Google Scholar] [CrossRef] [PubMed]
  98. Filippone, A.; Magno, S. Clinical Connections Between the Microbiota and Breast Cancer (Onset, Progression and Management). In Comprehensive Gut Microbiota; Glibetic, M., Ed.; Elsevier: Amsterdam, The Netherlands, 2022; Volume 1, pp. 35–45. [Google Scholar] [CrossRef]
  99. Adlercreutz, H. Western Diet and Western Diseases: Some Hormonal and Biochemical Mechanisms and Associations. Scand. J. Clin. Lab. Invest. Suppl. 1990, 201, 3–23. [Google Scholar] [CrossRef] [PubMed]
  100. Goldin, B.R.; Adlercreutz, H.; Gorbach, S.L.; Warram, J.H.; Dwyer, J.T.; Swenson, L.; Woods, M.N. Estrogen Excretion Patterns and Plasma Levels in Vegetarian and Omnivorous Women. N. Engl. J. Med. 1982, 307, 1542–1547. [Google Scholar] [CrossRef] [PubMed]
  101. Saarinen, N.M.; Wärri, A.; Airio, M.; Smeds, A.; Mäkelä, S. Role of Dietary Lignans in the Reduction of Breast Cancer Risk. Mol. Nutr. Food Res. 2007, 51, 857–866. [Google Scholar] [CrossRef]
  102. Selber-Hnatiw, S.; Rukundo, B.; Ahmadi, M.; Akoubi, H.; Al-Bizri, H.; Aliu, A.F.; Ambeaghen, T.U.; Avetisyan, L.; Bahar, I.; Baird, A.; et al. Human Gut Microbiota: Toward an Ecology of Disease. Front. Microbiol. 2017, 8, 1265. [Google Scholar] [CrossRef]
  103. Adlercreutz, H.; Martin, F.; Pulkkinen, M.; Dencker, H.; Rimér, U.; Sjöberg, N.O.; Tikkanen, M.J. Intestinal Metabolism of Estrogens. J. Clin. Endocrinol. Metab. 1976, 43, 497–505. [Google Scholar] [CrossRef]
  104. Yaghjyan, L.; Colditz, G.A. Estrogens in the Breast Tissue: A Systematic Review. Cancer Causes Control. 2011, 22, 529–540. [Google Scholar] [CrossRef]
  105. Chan, A.A.; Bashir, M.; Rivas, M.N.; Duvall, K.; Sieling, P.A.; Pieber, T.R.; Vaishampayan, P.A.; Love, S.M.; Lee, D.J. Characterization of the Microbiome of Nipple Aspirate Fluid of Breast Cancer Survivors. Sci. Rep. 2016, 6, 28061. [Google Scholar] [CrossRef] [PubMed]
  106. Gorbach, S.L.; Goldin, B.R. Diet and the Excretion and Enterohepatic Cycling of Estrogens. Prev. Med. 1987, 16, 525–531. [Google Scholar] [CrossRef] [PubMed]
  107. Alpuim Costa, D.; Nobre, J.G.; Batista, M.V.; Ribeiro, C.; Calle, C.; Cortes, A.; Marhold, M.; Negreiros, I.; Borralho, P.; Brito, M.; et al. Human Microbiota and Breast Cancer—Is There Any Relevant Link?—A Literature Review and New Horizons Toward Personalised Medicine. Front. Microbiol. 2021, 12, 584332. [Google Scholar] [CrossRef]
  108. Wiebe, J.P.; Muzia, D.; Hu, J.; Szwajcer, D.; Hill, S.A.; Seachrist, J.L. The 4-Pregnene and 5α-Pregnane Progesterone Metabolites Formed in Nontumorous and Tumorous Breast Tissue Have Opposite Effects on Breast Cell Proliferation and Adhesion1. Cancer Res. 2000, 60, 936–943. [Google Scholar]
  109. Urbaniak, C.; Gloor, G.B.; Brackstone, M.; Scott, L.; Tangney, M.; Reid, G. The Microbiota of Breast Tissue and Its Association with Breast Cancer. Appl. Environ. Microbiol. 2016, 82, 5039–5048. [Google Scholar] [CrossRef]
  110. Chadha, J.; Nandi, D.; Atri, Y.; Nag, A. Significance of Human Microbiome in Breast Cancer: Tale of an Invisible and an Invincible. Semin. Cancer Biol. 2021, 70, 112–127. [Google Scholar] [CrossRef] [PubMed]
  111. Xuan, C.; Shamonki, J.M.; Chung, A.; Dinome, M.L.; Chung, M.; Sieling, P.A.; Lee, D.J. Microbial Dysbiosis Is Associated with Human Breast Cancer. PLoS ONE 2014, 9, e83744. [Google Scholar] [CrossRef]
  112. Lawani-Luwaji, E.U.; Alade, T. Sphingomonadaceae: Protective against Breast Cancer? Bull. Natl. Res. Cent. 2020, 44, 191. [Google Scholar] [CrossRef]
  113. Karabin, M.; Hudcova, T.; Jelinek, L.; Dostalek, P. Biotransformations and Biological Activities of Hop Flavonoids. Biotechnol. Adv. 2015, 33 Pt 2, 1063–1090. [Google Scholar] [CrossRef] [PubMed]
  114. Possemiers, S.; Heyerick, A.; Robbens, V.; De Keukeleire, D.; Verstraete, W. Activation of Proestrogens from Hops (Humulus lupulus L.) by Intestinal Microbiota; Conversion of Isoxanthohumol into 8-Prenylnaringenin. J. Agric. Food Chem. 2005, 53, 6281–6288. [Google Scholar] [CrossRef] [PubMed]
  115. Overk, C.R.; Yao, P.; Chadwick, L.R.; Nikolic, D.; Sun, Y.; Cuendet, M.A.; Deng, Y.; Hedayat, A.S.; Pauli, G.F.; Farnsworth, N.R.; et al. Comparison of the in Vitro Estrogenic Activities of Compounds from Hops (Humulus lupulus) and Red Clover (Trifolium pratense). J. Agric. Food Chem. 2005, 53, 6246–6253. [Google Scholar] [CrossRef]
  116. Schaefer, O.; Hümpel, M.; Fritzemeier, K.-H.; Bohlmann, R.; Schleuning, W.-D. 8-Prenyl Naringenin Is a Potent ERalpha Selective Phytoestrogen Present in Hops and Beer. J. Steroid Biochem. Mol. Biol. 2003, 84, 359–360. [Google Scholar] [CrossRef] [PubMed]
  117. Rosenfeld, C.S. Gut Dysbiosis in Animals Due to Environmental Chemical Exposures. Front. Cell Infect. Microbiol. 2017, 7, 396. [Google Scholar] [CrossRef]
  118. Andújar, N.; Gálvez-Ontiveros, Y.; Zafra-Gómez, A.; Rodrigo, L.; Álvarez-Cubero, M.J.; Aguilera, M.; Monteagudo, C.; Rivas, A.A. Bisphenol A Analogues in Food and Their Hormonal and Obesogenic Effects: A Review. Nutrients 2019, 11, 2136. [Google Scholar] [CrossRef]
  119. Claus, S.P.; Guillou, H.; Ellero-Simatos, S. Erratum: The Gut Microbiota: A Major Player in the Toxicity of Environmental Pollutants? NPJ Biofilms Microbiomes 2017, 3, 17001. [Google Scholar] [CrossRef]
  120. Snedeker, S.M.; Hay, A.G. Do Interactions between Gut Ecology and Environmental Chemicals Contribute to Obesity and Diabetes? Environ. Health Perspect. 2012, 120, 332–339. [Google Scholar] [CrossRef]
  121. Cresci, G.A.; Bawden, E. Gut Microbiome: What We Do and Don’t Know. Nutr. Clin. Pract. 2015, 30, 734–746. [Google Scholar] [CrossRef] [PubMed]
  122. Heintz-Buschart, A.; Wilmes, P. Human Gut Microbiome: Function Matters. Trends Microbiol. 2018, 26, 563–574. [Google Scholar] [CrossRef] [PubMed]
  123. Salvucci, E. The Human-Microbiome Superorganism and Its Modulation to Restore Health. Int. J. Food Sci. Nutr. 2019, 70, 781–795. [Google Scholar] [CrossRef]
  124. Clavel, T.; Henderson, G.; Alpert, C.-A.; Philippe, C.; Rigottier-Gois, L.; Doré, J.; Blaut, M. Intestinal Bacterial Communities That Produce Active Estrogen-like Compounds Enterodiol and Enterolactone in Humans. Appl. Environ. Microbiol. 2005, 71, 6077–6085. [Google Scholar] [CrossRef]
  125. Li, Z.; Henning, S.M.; Lee, R.-P.; Lu, Q.-Y.; Summanen, P.H.; Thames, G.; Corbett, K.; Downes, J.; Tseng, C.-H.; Finegold, S.M.; et al. Pomegranate Extract Induces Ellagitannin Metabolite Formation and Changes Stool Microbiota in Healthy Volunteers. Food Funct. 2015, 6, 2487–2495. [Google Scholar] [CrossRef] [PubMed]
  126. Luo, J.; Li, Y.; Mai, Y.; Gao, L.; Ou, S.; Wang, Y.; Liu, L.; Peng, X. Flaxseed Gum Reduces Body Weight by Regulating Gut Microbiota. J. Funct. Foods 2018, 47, 136–142. [Google Scholar] [CrossRef]
  127. Giuliani, C.; Marzorati, M.; Innocenti, M.; Vilchez-Vargas, R.; Vital, M.; Pieper, D.H.; Van de Wiele, T.; Mulinacci, N. Dietary Supplement Based on Stilbenes: A Focus on Gut Microbial Metabolism by the in Vitro Simulator M-SHIME®. Food Funct. 2016, 7, 4564–4575. [Google Scholar] [CrossRef] [PubMed]
  128. Landete, J.M.; Gaya, P.; Rodríguez, E.; Langa, S.; Peirotén, Á.; Medina, M.; Arqués, J.L. Probiotic Bacteria for Healthier Aging: Immunomodulation and Metabolism of Phytoestrogens. Biomed. Res. Int. 2017, 2017, 5939818. [Google Scholar] [CrossRef]
  129. McCann, S.E.; Thompson, L.U.; Nie, J.; Dorn, J.; Trevisan, M.; Shields, P.G.; Ambrosone, C.B.; Edge, S.B.; Li, H.-F.; Kasprzak, C.; et al. Dietary Lignan Intakes in Relation to Survival among Women with Breast Cancer: The Western New York Exposures and Breast Cancer (WEB) Study. Breast Cancer Res. Treat. 2010, 122, 229–235. [Google Scholar] [CrossRef]
  130. Org, E.; Mehrabian, M.; Parks, B.W.; Shipkova, P.; Liu, X.; Drake, T.A.; Lusis, A.J. Sex Differences and Hormonal Effects on Gut Microbiota Composition in Mice. Gut Microbes 2016, 7, 313–322. [Google Scholar] [CrossRef]
  131. Van de Wiele, T.; Vanhaecke, L.; Boeckaert, C.; Peru, K.; Headley, J.; Verstraete, W.; Siciliano, S. Human Colon Microbiota Transform Polycyclic Aromatic Hydrocarbons to Estrogenic Metabolites. Environ. Health Perspect. 2005, 113, 6–10. [Google Scholar] [CrossRef]
  132. Feng, P.; Ye, Z.; Kakade, A.; Virk, A.K.; Li, X.; Liu, P. A Review on Gut Remediation of Selected Environmental Contaminants: Possible Roles of Probiotics and Gut Microbiota. Nutrients 2019, 11, 22. [Google Scholar] [CrossRef]
  133. Choi, Y.J.; Seelbach, M.J.; Pu, H.; Eum, S.Y.; Chen, L.; Zhang, B.; Hennig, B.; Toborek, M. Polychlorinated Biphenyls Disrupt Intestinal Integrity via NADPH Oxidase-Induced Alterations of Tight Junction Protein Expression. Environ. Health Perspect. 2010, 118, 976–981. [Google Scholar] [CrossRef] [PubMed]
  134. Blandino, G.; Inturri, R.; Lazzara, F.; Di Rosa, M.; Malaguarnera, L. Impact of Gut Microbiota on Diabetes Mellitus. Diabetes Metab. 2016, 42, 303–315. [Google Scholar] [CrossRef] [PubMed]
  135. Barlow, G.M.; Yu, A.; Mathur, R. Role of the Gut Microbiome in Obesity and Diabetes Mellitus. Nutr. Clin. Pract. 2015, 30, 787–797. [Google Scholar] [CrossRef]
  136. Maruvada, P.; Leone, V.; Kaplan, L.M.; Chang, E.B. The Human Microbiome and Obesity: Moving beyond Associations. Cell Host Microbe 2017, 22, 589–599. [Google Scholar] [CrossRef] [PubMed]
  137. Bouter, K.E.; van Raalte, D.H.; Groen, A.K.; Nieuwdorp, M. Role of the Gut Microbiome in the Pathogenesis of Obesity and Obesity-Related Metabolic Dysfunction. Gastroenterology 2017, 152, 1671–1678. [Google Scholar] [CrossRef] [PubMed]
  138. Chen, X.; Devaraj, S. Gut Microbiome in Obesity, Metabolic Syndrome, and Diabetes. Curr. Diab. Rep. 2018, 18, 129. [Google Scholar] [CrossRef]
  139. Vallianou, N.G.; Stratigou, T.; Tsagarakis, S. Microbiome and Diabetes: Where Are We Now? Diabetes Res. Clin. Pract. 2018, 146, 111–118. [Google Scholar] [CrossRef]
  140. Saad, M.J.A.; Santos, A.; Prada, P.O. Linking Gut Microbiota and Inflammation to Obesity and Insulin Resistance. Physiology 2016, 31, 283–293. [Google Scholar] [CrossRef]
  141. Dalamaga, M. Obesity, Insulin Resistance, Adipocytokines and Breast Cancer: New Biomarkers and Attractive Therapeutic Targets. World J. Exp. Med. 2013, 3, 34–42. [Google Scholar] [CrossRef]
  142. Crisóstomo, J.; Matafome, P.; Santos-Silva, D.; Gomes, A.L.; Gomes, M.; Patrício, M.; Letra, L.; Sarmento-Ribeiro, A.B.; Santos, L.; Seiça, R. Hyperresistinemia and Metabolic Dysregulation: A Risky Crosstalk in Obese Breast Cancer. Endocrine 2016, 53, 433–442. [Google Scholar] [CrossRef]
  143. Ruiz, D.; Becerra, M.; Jagai, J.S.; Ard, K.; Sargis, R.M. Disparities in Environmental Exposures to Endocrine-Disrupting Chemicals and Diabetes Risk in Vulnerable Populations. Diabetes Care 2018, 41, 193–205. [Google Scholar] [CrossRef]
  144. Chevalier, N.; Fénichel, P. Endocrine Disruptors: New Players in the Pathophysiology of Type 2 Diabetes? Diabetes Metab. 2015, 41, 107–115. [Google Scholar] [CrossRef]
  145. Casals-Casas, C.; Desvergne, B. Endocrine Disruptors: From Endocrine to Metabolic Disruption. Annu. Rev. Physiol. 2011, 73, 135–162. [Google Scholar] [CrossRef] [PubMed]
  146. Bodin, J.; Stene, L.C.; Nygaard, U.C. Can Exposure to Environmental Chemicals Increase the Risk of Diabetes Type 1 Development? Biomed. Res. Int. 2015, 2015, 208947. [Google Scholar] [CrossRef] [PubMed]
  147. Petrakis, D.; Vassilopoulou, L.; Mamoulakis, C.; Psycharakis, C.; Anifantaki, A.; Sifakis, S.; Docea, A.O.; Tsiaoussis, J.; Makrigiannakis, A.; Tsatsakis, A.M. Endocrine Disruptors Leading to Obesity and Related Diseases. Int. J. Environ. Res. Public Health 2017, 14, 1282. [Google Scholar] [CrossRef] [PubMed]
  148. Le Magueresse-Battistoni, B.; Multigner, L.; Beausoleil, C.; Rousselle, C. Effects of Bisphenol A on Metabolism and Evidences of a Mode of Action Mediated through Endocrine Disruption. Mol. Cell Endocrinol. 2018, 475, 74–91. [Google Scholar] [CrossRef]
  149. Ahn, C.; Kang, H.-S.; Lee, J.-H.; Hong, E.-J.; Jung, E.-M.; Yoo, Y.-M.; Jeung, E.-B. Bisphenol A and Octylphenol Exacerbate Type 1 Diabetes Mellitus by Disrupting Calcium Homeostasis in Mouse Pancreas. Toxicol. Lett. 2018, 295, 162–172. [Google Scholar] [CrossRef] [PubMed]
  150. Guo, H.; Yan, H.; Cheng, D.; Wei, X.; Kou, R.; Si, J. Tributyltin Exposure Induces Gut Microbiome Dysbiosis with Increased Body Weight Gain and Dyslipidemia in Mice. Environ. Toxicol. Pharmacol. 2018, 60, 202–208. [Google Scholar] [CrossRef]
  151. Bansal, A.; Henao-Mejia, J.; Simmons, R.A. Immune System: An Emerging Player in Mediating Effects of Endocrine Disruptors on Metabolic Health. Endocrinology 2018, 159, 32–45. [Google Scholar] [CrossRef] [PubMed]
  152. Nowak, K.; Jabłońska, E.; Ratajczak-Wrona, W. Immunomodulatory Effects of Synthetic Endocrine Disrupting Chemicals on the Development and Functions of Human Immune Cells. Environ. Int. 2019, 125, 350–364. [Google Scholar] [CrossRef]
  153. Coruzzi, G. Overview of Gastrointestinal Toxicology. Curr. Protoc. Toxicol. 2010, 21, 21.1. [Google Scholar] [CrossRef] [PubMed]
  154. Takiishi, T.; Fenero, C.I.M.; Câmara, N.O.S. Intestinal Barrier and Gut Microbiota: Shaping Our Immune Responses throughout Life. Tissue Barriers 2017, 5, e1373208. [Google Scholar] [CrossRef] [PubMed]
Table 1. Interplay between endocrine disruptors and microbiota with potential drivers of breast cancer.
Table 1. Interplay between endocrine disruptors and microbiota with potential drivers of breast cancer.
SourceMoleculesMicrorganismsOutcomeReferences
FoodsLignans
Isoflavones
C. methoxybenzovorans
B. pseudocatenulatum WC 401
Firmicutes
Bacteroidetes
F. prausnitzii
Lactobacillus
Enterococcus
Estrogen
Bioavailability
[60,61,62,63,64]
PlasticsBPA
BPS
Helicobacteraceae
Firmicutes
Clostridia
Lipogenesis
Gluconeogenesis
Tumor proliferation
Metastatic spread
[58,59,65,66]
PesticidesOrganophosphates
DDT
DDE
PCB
Bacteroides,
Burkholderiales
Clostridiaceae
Erysiopelotrichaceae
Coprobacillus
Lachnospiraceae
Staphylococcaceae
Gluconeogenesis
Oxidative stress
Changes in insulin
and ghrelin secretion
[49,50,65,66]
Heavy metalsArsenic
Lead
Cadmium
Bacteroides
Firmicutes
Proteobacteria
Altered gluconeogenesis
Lipogenesis
Inflammation
Body fat
[65,66]
BPA, Bisphenol A; BPS, Bisphenol S; DDT, dichloro-diphenyl-trichloroethane; DDE, Dichloro-diphenyl-dichloroethylene; PCB, polychlorinated biphenyl.
Table 2. Interplay between phytoestrogens and their metabolites with microrganism.
Table 2. Interplay between phytoestrogens and their metabolites with microrganism.
Chemical FamilyMoleculesMicrorganismsReferences
LignansAnhydrosecoisolariciresinol
Secoisolariciresinol diglucoside
Syringaresinol
C. methoxybenzovorans
B. pseudocatenulatum WC 401
Firmicutes
Bacteroidetes
[60,61,62,64]
IsoflavonesCoumestrol
Genistein
Equol
Daidzein
F. prausnitzii
Lactobacillus
Enterococcus
[63]
SteroidsEstradiol
Estrone
Collinsella, Edwardsiella, Alistipes, Bacteroides, Bifidobacterium, Citrobacter, Clostridium, Dermabacter, Escherichia, Faecalibacterium, Lactobacillus, Marvinbryantia, Propionibacterium, Roseburia, Tannerella[22,83,84,85,86,87]
PrenylflavonoidsXanthohumol
Desmethyxanthohumol
E. limosum[88]
Stilbenes Resveratrol
Trans-resveratrol
Dihydroresveratrol
3,4′–dihydroxybibenzyl,
3,4′-dihydroxy-trans-stilbene
Firmicutes
Bacteroidetes,
Actinobacteria
Verrucomicrobia,
Cyanobacteria
[89,90,91,92,93]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Filippone, A.; Rossi, C.; Rossi, M.M.; Di Micco, A.; Maggiore, C.; Forcina, L.; Natale, M.; Costantini, L.; Merendino, N.; Di Leone, A.; et al. Endocrine Disruptors in Food, Estrobolome and Breast Cancer. J. Clin. Med. 2023, 12, 3158. https://doi.org/10.3390/jcm12093158

AMA Style

Filippone A, Rossi C, Rossi MM, Di Micco A, Maggiore C, Forcina L, Natale M, Costantini L, Merendino N, Di Leone A, et al. Endocrine Disruptors in Food, Estrobolome and Breast Cancer. Journal of Clinical Medicine. 2023; 12(9):3158. https://doi.org/10.3390/jcm12093158

Chicago/Turabian Style

Filippone, Alessio, Cristina Rossi, Maria Maddalena Rossi, Annalisa Di Micco, Claudia Maggiore, Luana Forcina, Maria Natale, Lara Costantini, Nicolò Merendino, Alba Di Leone, and et al. 2023. "Endocrine Disruptors in Food, Estrobolome and Breast Cancer" Journal of Clinical Medicine 12, no. 9: 3158. https://doi.org/10.3390/jcm12093158

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop