Next Article in Journal
Phage Therapy Enhances Survival, Immune Response, and Metabolic Resilience in Pacific White Shrimp (Litopenaeus vannamei) Challenged with Vibrio parahaemolyticus
Previous Article in Journal
YOLO-DFAM-Based Onboard Intelligent Sorting System for Portunus trituberculatus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bisphenols: Endocrine Disruptors and Their Impact on Fish: A Review

1
Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Hudcova 296/70, 621 00 Brno, Czech Republic
2
Department of Animal Protection and Welfare and Veterinary Public Health, University of Veterinary Sciences Brno, Palackeho tr. 1946/1, 612 42 Brno, Czech Republic
*
Author to whom correspondence should be addressed.
Fishes 2025, 10(8), 365; https://doi.org/10.3390/fishes10080365
Submission received: 17 June 2025 / Revised: 10 July 2025 / Accepted: 23 July 2025 / Published: 29 July 2025
(This article belongs to the Section Environment and Climate Change)

Abstract

Bisphenols (BPs), particularly bisphenol A (BPA) and its structural analogues, are synthetic compounds widely used in plastics and industrial materials. These substances are also recognised as endocrine-disrupting chemicals (EDCs) due to their ability to interfere with hormonal systems, which has significant implications for aquatic organisms. This review summarises the occurrence, environmental distribution, and toxicity of BPs in fish, with a focus on estrogenic, androgenic, thyroid, and glucocorticoid disruptions. Studies consistently show that exposure to BPs leads to altered gene expression, developmental abnormalities, impaired reproduction, and disrupted hormonal signalling in various fish species. Although BPA alternatives like bisphenol S, bisphenol F, or bisphenol AF were introduced as safer options, emerging evidence suggests they may pose equal or greater risks. Regulatory measures are evolving, particularly within the European Union, but legislation remains limited for many bisphenol analogues. This review emphasises the need for comprehensive environmental monitoring, stricter regulatory frameworks, and the development of genuinely safer alternatives to minimise the ecological and health impacts of BPs in aquatic systems.
Key Contribution: This review summarises current knowledge on the occurrence, toxicity, and endocrine-disrupting effects of bisphenols in fish, emphasising the comparable risks of BPA analogues and the need for stricter regulation and safer alternatives.

Graphical Abstract

1. Introduction

In recent decades, a significant and growing threat to fishes and other aquatic organisms has emerged due to the increasing global human population and the subsequent release of a wide variety of chemical wastes into freshwater ecosystems [1].
One particularly concerning group of chemicals are endocrine-disrupting compounds (EDCs), which can interfere with the normal functioning of the endocrine system in organisms. These chemicals, whether of natural or anthropogenic origin, are exogenous substances that interfere with the endocrine system, potentially mimicking or blocking hormones or altering the secretion, mechanisms of action, or rates of metabolism of endogenous hormones, thereby leading to adverse health outcomes [2]. In particular, they can lead to various types of disruption in organisms, such as estrogenic, androgenic, and thyroid disruption, among others. These compounds can be found in a wide array of sources, including industrial chemicals primarily used in the production of various plastics and epoxy resins, as well as pesticides and phthalates that have the potential to leach into food and water supplies [3,4]. Furthermore, even some pharmaceuticals and substances in personal care products can also act as EDCs and negatively impact the health and welfare of fish [5,6]. Bisphenols (BPs) are one such class of pollutants that have garnered increased attention due to their presence in the environment and their effects, as they have been linked to various adverse health effects in fish, including impaired reproduction, growth, and development [3,4].
BPs, a class of synthetic organic compounds, have gained significant attention recently because of their widespread presence in aquatic environments [7]. They can cause a wide range of physiological, behavioural, and developmental changes [8]. This review will explore in detail the endocrine effects of BPs on fish health and fitness.

2. Characteristics of Bisphenols and Their Occurrence in the Aquatic Environment

BPs are widely utilised in the production of a diverse array of products, including plastics, water pipes, toys, medical equipment, electronics, food cans, and various household applications [9,10]. As the name implies, BPs contain two phenol groups and serve as fundamental building blocks for polymers [11]. When exposed to acidic conditions or high temperatures, the ester bonds within the polycarbonate polymer can undergo breakdown, leading to the release of BPs into contained foods or beverages [11].
Among the most significant BPs, bisphenol A (BPA) stands out due to its widespread use and confirmed adverse effects, particularly its potential as an EDC. BPA has been extensively detected in various environmental compartments, leading to its regulation or ban in certain products [8,12]. As a result, efforts have been made to find substitutes for BPA. However, concerns exist that these alternatives may exhibit similar toxicological profiles. Some of the most common structural analogues include bisphenol S (BPS), bisphenol F (BPF), and bisphenol B (BPB), with others numbering in the tens [13]—a summary of the most commonly used BPA and particular bisphenol analogues, which are further mentioned, is given in Table 1.
Bisphenol analogues, used as substitutes for BPA, have significant environmental impacts. They are frequently detected in aquatic environments and can affect marine species by disrupting the neuroendocrine system, causing oxidative stress, and leading to developmental alterations [10,14]. These analogues, such as BPF and BPS, exhibit endocrine-disrupting effects similar to or greater than BPA, impacting estrogen and androgen receptor signalling in aquatic organisms [15]. Additionally, bisphenol analogues can influence soil health by affecting soil microbiomes and enzyme activity, which are crucial for plant growth and development [16]. The persistence and fate of these compounds in the environment remain areas of concern, as they can lead to long-term ecological effects [17].
Their environmental prevalence and presence in aquatic systems depend on their concentrations, which can vary significantly across different regions and ecosystems [11]. BPs have been detected in surface waters, sediments, and aquatic organism (biota) samples collected from various locations worldwide, indicating their widespread environmental distribution. Concentrations of BPs in the aquatic environment typically range from a few nanograms per litre to several micrograms per litre in surface waters, from a few nanograms per gram to hundreds of nanograms per gram in sediments, and from a few nanograms per gram to tens of nanograms per gram in biota, depending on the specific location and source of contamination [9,18].
The release of BPs into the environment can occur through various pathways, such as the breakdown of plastic products, industrial waste, and wastewater effluents [9,19]. They occur in nature not just due to their presence in wastewater but also in wastewater treatment plants, agricultural fertilisers, and thermal paper, resulting in widespread environmental contamination. This broad exposure from various sources underlines the necessity for continued research into their health effects and sources of intoxication [20]. The characteristics of BPs in the aquatic environment are of particular concern, as they can persist and accumulate in water bodies, sediments, and biota, thereby potentially impacting aquatic ecosystems [21]. Table 2 gives an overview of the occurrence of the most important representatives of BPs in various matrices of the aquatic ecosystem.
Table 2. Overview of the leading representatives of bisphenols and their occurrence in the aquatic environment (d.w. = dry weight; n/a = no data available; n.d. = not detected; w.w. = wet weight).
Table 2. Overview of the leading representatives of bisphenols and their occurrence in the aquatic environment (d.w. = dry weight; n/a = no data available; n.d. = not detected; w.w. = wet weight).
Bisphenol
Type
ContinentWaterFish (ng/g w.w.)Sediment (ng/g d.w.)
Bisphenol A (BPA)Asia5.26–76.6 ng/L [22,23];
mean 23 ng/L [24];
<810 pg/L [25]
Coilia mystus: ~4–6,
Pseudorasbora parva: ~12–14,
Cyprinus carpio: ~11–13,
Silurus asotus: ~11–14,
[26]
0.56–5.22 [22];
mean 13.0 [24];
<0.6 [25];
0.18–2010 [26]
North America<90.0 ng/Ln/a<25,300 [18]
Bisphenol S (BPS)Asia0.07–5.2 ng/L [22,23];
2.2 ng/L [24]
Coilia mystus: ~2–3,
Pseudorasbora parva: ~1–2,
Cyprinus carpio: ~1–2,
Silurus asotus: ~1–2,
[26]
n.d. −0.19 [22];
0.69 [24]
Bisphenol F (BPF)Asian.d.–12.6 ng/L [23]Coilia mystus: ~2–3, Pseudorasbora parva: ~1–2, Cyprinus carpio: ~1–2,
Silurus asotus: ~1–2,
[26]
1.6 [24]
Bisphenol AF (BPAF)Asia0.44–10.8 ng/L [22,23];
0.9–246 ng/L [26]
Coilia mystus: ~21,
Pseudorasbora parva: ~13–17, Cyprinus carpio: ~3–4,
Silurus asotus: ~8,
[26]
0.08–0.66 [22];
0.18–2010 [26]
Bisphenol B (BPB)Asian.d.–14.3 ng/L [23]Coilia mystus: ~2,
Pseudorasbora parva: ~1–4, Cyprinus carpio: ~1–2,
Silurus asotus: ~3,
[26]
n/a
Bisphenol E (BPE)Asian.d.–6.2 ng/L [23]Coilia mystus: ~1–2, Pseudorasbora parva: ~1–2, Cyprinus carpio: ~1–2,
Silurus asotus: ~1–2,
[26]
n/a
Bisphenol Z (BPZ)Asian/aCoilia mystus: ~2–4,
Pseudorasbora parva: ~2–3,
Cyprinus carpio: ~1–3,
Silurus asotus: ~1–3,
[26]
n/a
Tetrabromo
bisphenol A (TBBPA)
Asia2.3 ng/L [24]; <810 pg/L [25]n/a<0.6 [25]

3. Regulatory Framework and Restriction of Bisphenol Usage

In the face of accumulating evidence on the adverse effects of BPA, regulatory agencies worldwide have taken steps to limit its usage in consumer products, particularly those intended for children and infants. These actions represent a crucial step towards mitigating the potential harm that BPA exposure can cause, particularly in vulnerable populations such as developing fetuses and infants [27,28]. However, as new research continues to uncover the far-reaching implications of BPA exposure, regulatory agencies must remain vigilant in monitoring its presence in consumer products and adjust their guidelines accordingly to safeguard public health [28].
The regulatory landscape surrounding BPA has evolved in response to mounting scientific evidence of its endocrine-disrupting effects and potential health implications [29]. Globally, regulations for controlling EDCs vary. The European Union (EU) has introduced regulations such as REACH, which focuses on a hazard-based approach, while the United States Environmental Protection Agency (EPA) implements a risk-based Endocrine Disruptor Screening Program (EDSP) [30,31]. In the USA, BPA has been banned in baby bottles since 2012 [32] and was banned in packaging for infants a year later [33], but it can be used in other food contact materials. Both approaches aim to identify and regulate harmful EDCs, although challenges remain in testing methods and distinguishing endocrine-specific effects [34]. BPA is also banned in baby bottles and other infant products in China [35], Canada [36], as well as South Korea [37] and other countries. Other analogues are not often covered yet, although monitoring is increasing.
The European Union has not imposed a complete ban on BPA but has progressively restricted its use due to its endocrine-disrupting properties and reproductive toxicity. Several legislative measures were initiated in 2011, when the EU banned the use of BPA in the manufacture of polycarbonate infant feeding bottles [38]. This decision was based on concerns about infants’ vulnerability to BPA exposure. Later in 2016, BPA was added to the Candidate List of Substances of Very High Concern (SVHC) under the REACH Regulation (EC) No 1907/2006 [39], due to its classification as a substance toxic for reproduction (category 1B). In 2017, the European Chemicals Agency (ECHA) further identified BPA as an endocrine disruptor for human health under REACH. Consequently, BPA was also classified as an endocrine disruptor for the environment in 2018. Commission Regulation (EU) 2016/2235 then prohibited the use of BPA in thermal paper (such as cash register receipts), with the restriction entering into force on 2 January 2020 [40].
The European Commission has also banned BPA in food contact materials due to its harmful health effects, based on EFSA’s latest assessment from 2023. The ban covers adhesives, rubbers, plastics, coatings, and other materials. Products made with other BPs or bisphenol derivatives must not contain residual BPA. Exceptions exist for polysulfone filtration membranes and liquid epoxy-based coatings in large-capacity food containers (>1000 L), provided that BPA migration into food is undetectable and the materials are properly cleaned before use. The regulation also prohibits other hazardous BPs affecting the reproductive and endocrine systems. The tolerable daily intake (TDI) for BPA has been drastically lowered to 0.2 ng/kg body weight, making even minimal migration unsafe. The current specific migration limit (SML) of 0.05 mg/kg is being phased out. The European Union Reference Laboratory (EURL) will develop methods to verify the absence of BPA, with a detection limit of 1 μg/kg, unless otherwise specified in Commission Regulation (EU) 2024/3190 [41]. Unfortunately, other analogues do not have unified legislation regulation due to the definitions in the regulation, as they do not fit the definition of “bisphenol derivatives” in the regulation [41], as shown in Figure 1.
The introduction of structural analogues as substitutes for BPA has further complicated the regulatory landscape [8,12]. As these alternative BPs have been found to exhibit similar endocrine-disrupting properties, regulatory agencies must extend their oversight to these compounds as well, ensuring that the public is protected from the potential health risks associated with exposure to the broader class of BPs [19,27]. In 2021, the European Chemicals Agency (ECHA) added BPB to the Candidate List of Substances of Very High Concern (SVHC) under REACH as well as BPS. For now, with BPA, they are the only BPs included due to their endocrine-disrupting properties and the rest are monitored and analysed. Last on the list is TBBPA for its carcinogenic potential, also included in 2023. But none of the BPs is currently included in REACH Regulation (EC) No 1907/2006; Annex XIV: List of substances subject to authorisation, as some of them are just candidates for Annex XIV [39]. Researchers and industry professionals alike have turned their attention to non-bisphenol materials, including plant-based polymers and other bio-based compounds, to identify viable replacements that minimise or eliminate the risks associated with BPs [42].

4. Toxicity of Bisphenols to Fish

BPs have been reported to induce a variety of effects in fish, including altered gene expression and the activity of key enzymes involved in the metabolism and production of endogenous hormones, physiological changes, and behavioural modifications [6,43,44]. In addition, BPs have also been implicated in the induction of oxidative stress [9,45,46]. The resulting oxidative stress has been linked to a range of adverse health effects, including inflammation, cell dysfunction, and tissue damage. Prolonged exposure to bisphenol-induced oxidative stress can have far-reaching health consequences, potentially contributing to the development of chronic diseases such as cancer, neurological disorders, and cardiovascular problems [27,47]. Evidence shows that exposure to BPs, even at relatively low concentrations, can lead to significant physiological, behavioural, and fitness-related changes in fish, such as cognitive disorders [48]. The change can then occur in local tissue, organs, or by mutations [49]. Apart from hormonal disruption, BPs can cause various problems in organisms.
Recent studies have revealed that alternative BPs can also induce toxic, reproductive, and neuroendocrine effects similar to those observed with BPA [10,19]. Specifically, these studies have shown that BPS and BPF can disrupt hormone signalling and lead to adverse health outcomes, including developmental and reproductive issues, in a manner comparable to the well-documented effects of BPA [13]. Numerous studies have investigated the toxic effects of BPs, with both animal and human studies demonstrating the harmful impacts of these chemicals [9]. One of the most well-documented effects of BPs is their estrogenic activity, which can disrupt the normal function of the endocrine system and lead to feminisation of male fish, altered sexual development, and impaired reproductive success [4,21]. However, the endocrine-disrupting effects of BPs extend beyond estrogen signalling, encompassing the disruption of androgen [3,50,51,52], thyroid [12,53,54,55,56,57], and glucocorticoid hormone pathways [58,59,60,61,62]. Different concrete disrupting effects are discussed in separate sections.

4.1. Estrogenic Activity and Disruption

Estrogenic disruption is a significant environmental concern, particularly due to the widespread presence of EDCs such as BPs [63,64]. BPs disrupt endocrine function primarily by mimicking the structure and function of natural estrogens, binding to estrogen receptors (ERs), and activating the estrogen signalling pathway (see Table 3 and Table 4 for an overview of significant findings on the estrogenic effects of selected BPs in fish). This interference disrupts normal hormonal balance, leading to consequences such as feminisation, impaired reproductive success, and changes in population dynamics [65]. Studies have demonstrated that even low concentrations of BPs can interfere with the reproductive axis, leading to impaired gonadal development, altered spawning behaviour, and reduced embryo viability [3,66]. Studies have shown that exposing male fish to estrogen-mimicking BPs can induce the production of vitellogenin (VTG) and lead to the development of intersex characteristics, thereby directly impacting the reproductive success and fitness of these animals [66,67].
Table 3. Overview of significant findings on the estrogenic effects of selected bisphenols in fish (↑ = increase; ↓ = decrease; dpf = days post-fertilisation; ER = estrogen receptor; hpf = hours post-fertilisation; VTG = vitellogenin).
Table 3. Overview of significant findings on the estrogenic effects of selected bisphenols in fish (↑ = increase; ↓ = decrease; dpf = days post-fertilisation; ER = estrogen receptor; hpf = hours post-fertilisation; VTG = vitellogenin).
Bisphenol TypeFish SpeciesExposure DurationConcentration Effects ObservedReferences
BPADanio rerio15 days0.01, 0.1, 1 mg/L↑ egg production, ↓ fertilisation rate, altered reproductive gene expression, ↓ DNA methylation in ovaries, impaired reproductive processes[68]
30 days1, 10, 100 µg/L↑ atretic follicles, altered estrogen receptor expression, impaired ovarian function, reduced reproductive fitness[3]
Danio rerio, embryo–larvae96 hpf–6 dpf1–200 mg/L↑ VTG, induced estrogenic response in heart, liver, muscle, and fins (ER-dependent)[50,69]
Danio rerio,
male
6 weeks100 and 2000 µg/Learly reproductive feminisation, female-like lipid metabolism, gonad damage, feminisation[70]
Danio rerio, larvae5–14 dpfup to 2500 µg/Lestrogenic response in the heart, altered heart rhythm, ↓ heart rate[71]
Pimephales promelas43, 71, 164 days1–1, 280 µg/L↑ VTG in males (≥160 μg/L), gonadal growth inhibition (≥640 μg/L), ↓ egg production[72]
4 days16–1, 280 µg/L↓ gonadal growth, altered sex cell types, ↓ egg production at high concentrations[69]
Oryzias melastigma70 days200 µg/Lfollicular atresia, irregular oocytes, empty follicles, ↓ eggs laid, ↓ fertilisation rate[73]
Carassius auratus7–90 days0.2 and 20 µg/L↑ VTG (liver, 60 days), ↑ aromatase and ERs, estrogenic effects[74]
Table 4. Overview of significant findings on the estrogenic effects of selected bisphenols in fish (↑ = increase; ↓ = decrease; dpf = days post-fertilisation; ER = estrogen receptor; hpf = hours post-fertilisation; VTG = vitellogenin).
Table 4. Overview of significant findings on the estrogenic effects of selected bisphenols in fish (↑ = increase; ↓ = decrease; dpf = days post-fertilisation; ER = estrogen receptor; hpf = hours post-fertilisation; VTG = vitellogenin).
Bisphenol TypeFish SpeciesExposure DurationConcentrationEffects ObservedReferences
BPFDanio rerio96 h (embryo–larva)1–100 µg/Linduced estrogenic response, ↑ ER and aromatase gene activity, ↑ VTG, altered reproductive neuroendocrine genes[19,50,69]
long-term (duration not specified)1–100 µg/L↑ expression of reproductive neuroendocrine genes (kiss1, gnrh3, lhβ, fshβ), ↑ VTG, ER and aromatase activity[19]
96 hpf–6 dpf1–200 mg/L↑ VTG in heart, liver, muscle, and fins[50,69]
21 days0.1 and 1 mg/L↑ VTG (males) and estrogenic effects[75]
Oryzias melastigma70 days200 µg/Lfollicular atresia, irregular oocytes, empty follicles, ↓ eggs laid, ↓ fertilisation rate[73]
BPAFDanio rerio4 hpf–120 dpf5, 25, 125 µg/l↑ 17β-estradiol in females, ↓ egg fertilisation[61]
96 h (embryo–larva)1–200 mg/Lmost potent estrogenic response, ↑ ER and aromatase gene activity, ↑ VTG[50,69]
96 hpf100 and 200 µg/Ldelayed gonadal migration, ↓ germ cell progenitors, altered hormone receptor expression[76]
96 hpf–6 dpf1–200 mg/Lmost potent ↑ VTG in heart, liver, muscle, fins[50,69]
Oryzias melastigma70 days200 µg/Lmost pronounced ↓ eggs laid, follicular atresia, irregular oocytes, empty follicles, ↓ fertilisation[73]
BPBDanio rerio21 days0.1 and 1 mg/L↓ egg production, estrogenic-like activity similar to or greater than BPA[77]
96 h (embryo–larva)1–100 µg/L↑ ER and aromatase gene activity, altered reproductive neuroendocrine genes[78]
BPSDanio rerio4 hpf–120 dpf 1 and 100 µg/Lfemale-dominant sex ratio, impaired reproductive capacity[61]
75 days0.1, 1, 10, 100 μg/L↑ female to male sex ratio, ↑ estradiol and VTG (males, females), ↓ egg production[79]
Danio rerio, male96 hpf–6 dpf1–200 mg/L↑ VTG at high concentrations, induced estrogenic response, less potent than BPA[50,69]
21 days8, 40, 200 μg/mL↑ VTG, ↑ aromatase and estradiol[52]
Research further indicates that BPs can bind to specific estrogen receptors, particularly estrogen receptor alpha (ERα), thereby either activating or inhibiting estrogenic pathways. For instance, BPA disrupts steroidogenesis in Leydig cells, leading to an imbalance in sex hormones, which is critical for reproductive health [80]. This disruption manifests as altered hormone levels, specifically increased estradiol and decreased testosterone, with profound effects on reproductive function. Furthermore, exposure to BPA has been associated with changes in the expression of genes related to estrogen signalling, such as hoxa10, which is crucial for uterine development and function [81]. Moreover, the estrogenic effects of BPs have been observed in various animal models. For example, exposure to BPS has been linked to reproductive impairments and hormonal imbalances in zebrafish (Danio rerio), highlighting the potential for these compounds to disrupt endocrine functions across species [79].
The assessment of estrogenic disruption can be conducted at multiple biological levels, each providing critical insights into endocrine interference. Gene expression analyses, particularly the measurement of VTG levels, serve as a reliable biomarker for estrogenic activity. VTG is a precursor protein synthesised in response to estrogen, making it a key indicator of endocrine disruption in male fish. Histological examinations of gonadal tissues can reveal alterations in sex differentiation and reproductive structures. Additionally, assessments of egg quality, hatching success, and behavioural studies (e.g., changes in mating and spawning behaviour) help elucidate the broader ecological consequences of estrogenic disruption [82,83,84].
BPA has been extensively studied and is known to induce VTG production in male zebrafish (Danio rerio), resulting in altered reproductive behaviours and developmental anomalies, including reduced hatching success [55,69]. Similarly, BPS exhibits estrogenic activity, leading to delayed hatching and increased malformation rates in embryos. While BPF is generally less potent than BPA, it still demonstrates significant estrogenic effects, impacting reproductive health and egg quality [82]. BPAF, on the other hand, is more potent than BPA, causing severe reproductive and developmental disruptions, including significant effects on egg quality and hatching success [78].
Furthermore, studies indicate that other BPs, such as bisphenol B (BPB) and bisphenol C (BPC), also possess estrogenic activity, although research on their specific effects remains limited [15,85]. The presence of these compounds in aquatic environments poses a significant risk to biodiversity and ecosystem health, reinforcing the need for comprehensive evaluations of their endocrine-disrupting potential.
The estrogenic activity of BPs has been well-documented in numerous studies involving various fish species. Even at low concentrations, exposure to BPs has been shown to induce the production of VTG, a female-specific egg yolk precursor protein, in male fish, a clear indication of feminisation and endocrine disruption. Furthermore, bisphenol exposure has been linked to altered sexual development, including the development of intersex characteristics, reduced spawning behaviour, and impaired reproductive success in both male and female fish [4,21].
Diagnosing estrogenic disruption caused by BPA and its analogues involves a comprehensive approach that integrates biochemical assays, in vivo studies, and molecular analyses. One primary method for assessing estrogenic activity is through cell-based assays that measure the binding affinity to estrogen receptors (ERs). Research has demonstrated that BPA and its analogues can activate both estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ) in vitro, indicating their potential to disrupt normal hormonal signalling [79,86]. Techniques such as luciferase reporter assays are effective for quantifying the transcriptional activation of ERs by these compounds, providing clear evidence of their estrogenic potential [87].
In addition to biochemical assays, animal models—particularly rodents and zebrafish (Danio rerio)—are crucial for understanding the physiological effects of bisphenol exposure. The uterotrophic assay, which measures changes in uterine weight in response to estrogenic compounds, has long been a standard method for evaluating estrogenicity in vivo. Furthermore, studies involving zebrafish embryos exposed to bisphenols have shown alterations in primordial germ cell migration and other developmental anomalies, which can serve as indicators of estrogenic disruption [50]. These in vivo models allow researchers to observe the systemic effects of bisphenols and their impact on reproductive health.
Finally, understanding the molecular mechanisms by which bisphenols exert their estrogenic effects is essential for accurate diagnosis. Research indicates that bisphenols can alter the expression of genes related to estrogen signalling pathways, including the upregulation of VTG in fish models [7]. Moreover, bisphenols may induce epigenetic changes that affect hormone receptor expression and function, contributing to long-term disruptions in endocrine signalling [83,88].
Additionally, the impact of BPs on development and behaviour, particularly in model organisms like zebrafish (Danio rerio), serves as a critical indicator. Research has shown that exposure to BPs can result in alterations in brain development and reproductive behaviours, which are linked to disrupted estrogen signalling [69,89]. These developmental effects may serve as biomarkers for estrogenic disruption. A growing body of research highlights that not only bisphenol BPA but also its analogues—including BPS, BPF, BPAF, and others—exhibit estrogenic properties and can lead to reproductive impairments in fish.

4.2. Androgenic Activity and Disruption

In addition to their estrogenic effects, BPs have also been shown to exhibit androgenic activity, which can disrupt the normal functioning of the androgen system in fish [90]. Androgen disruption in fish can manifest through a range of physical, behavioural, and physiological symptoms. The effects of such disruption can vary among species and depend on the timing and extent of exposure. An overview of significant findings on the androgenic effects of selected BPs is given in Table 5.
Table 5. Overview of significant findings on the androgenic effects of selected bisphenols in fish (↑ = increase; ↓ = decrease; dpf = days post-fertilisation; hpf = hours post-fertilisation).
Table 5. Overview of significant findings on the androgenic effects of selected bisphenols in fish (↑ = increase; ↓ = decrease; dpf = days post-fertilisation; hpf = hours post-fertilisation).
Bisphenol TypeFish SpeciesExposure DurationConcentration Effects ObservedReferences
BPACarassius auratus7–90 days0.2 and 20 µg/L↓ sperm quality (number, motility, volume),
↓ testosterone, antiandrogenic effects
[74]
BPSDanio rerio21 days8, 40, 200 μg/mL↓ endogenous androgens (males)[52]
21 days0.5, 5, 50 μg/Lantiandrogenic effect, ↓ gonad weight (males and females)[79]
75 days0.1, 1, 10, 100 μg/L↓ testosterone (males), ↓ gonad weight, ↓ testosterone (males), ↓ sperm count[79]
4 hpf–120 dpf1 and 100 µg/Laltered steroid hormone levels[61]
Gadus morhua, cells EC50 25.0 µmol/L; tested in the range of 0.003–50 µmol/L↑ activity of androgen receptor gmAR [15]
BPFDanio rerio21 days0.1 and 1 mg/L↓ testosterone (males), antiandrogenic effects[75]
BPAFGadus morhua, cells EC50 25.0 µmol/L; tested in the range of 0.003–50 µmol/L↑ activation of the androgen receptor gmAR[15]
Danio rerio6 dpf 0.1 and 1 µmol/L↓ locomotor activity, ↑ aromatase B (estrogenic/antiandrogenic marker)[91]
4 hpf–120 dpf5, 25, 125 μg/L↓ testosterone (males), impaired parental sperm quality[92]
The androgenic activity of BPs can disrupt the endocrine system by mimicking or inhibiting natural androgens, leading to alterations in sexual development and reproduction [79,93]. For instance, studies have documented changes in male secondary sexual characteristics, aggression, and reproductive behaviours in fish exposed to BPs [55,94]. The mechanisms by which BPs disrupt androgen receptor (AR) pathways involve complex molecular interactions. These compounds can act as antagonists at the AR, potentially interfering with the receptor’s normal function, as well as altering gene expression and signalling pathways that regulate androgen-mediated processes [95,96].
One of the most notable indicators of androgen disruption in fish is, as said, the alteration in sexual characteristics and gonadal morphology. Male fish may exhibit feminisation, with reduced testis size, the presence of ovarian tissue, or the development of female secondary sexual characteristics [97,98]. For example, studies have shown that exposure to androgens and androgen mimetics can lead to the masculinisation of phenotypes in some species, including increased growth of male-specific anatomical features, while concomitantly reducing ovarian tissue or VTG expression as mentioned in the previous section [97,99,100,101].
Behaviour can also be significantly altered as a result of androgen disruption. For example, males exposed to androgenic EDCs have been reported to demonstrate decreased aggression, altered courtship behaviours, and compromised reproductive interactions. These behavioural changes can be linked to the endocrine changes induced by androgens, which affect brain function and are associated with neurogenesis [98,102]. Changes in social dynamics are particularly evident in species like zebrafish (Danio rerio), where exposure to androgenic substances can affect dominant/submissive interactions among males [98].
At the molecular level, disruptions in gene expression patterns that usually are regulated by androgens may occur. Key genes involved in reproduction, such as those encoding steroidogenic enzymes and receptors, may become upregulated or downregulated [99,103]. For example, research has demonstrated that exposure to androgenic substances can lead to significant changes in the expression of genes associated with spermatogenesis and testosterone synthesis by Leydig cells, critical for proper testicular function [104]. Moreover, alterations in the expression of aromatase, which is responsible for converting androgens to estrogens, may occur, leading to disrupted endocrine balance [101,105]. Gene expression profiling studies reveal that alterations induced by androgenic and antiandrogenic compounds significantly modulate the transcription of numerous genes, with potential downstream physiological implications, such as infertility and reduced fitness, in affected populations [100,106].
These changes could have broader ecosystem-level implications [43,107]. The ecological consequences of these androgenic disruptions can be profound. Altered sex ratios and hindered reproductive capacity in populations may lead to declines in biodiversity and changes in ecosystem dynamics [55,85]. Furthermore, the widespread environmental presence of BPs necessitates robust regulatory frameworks to monitor and limit their usage, addressing the dual challenges posed by their known estrogenic and androgenic activities [9,79].
Detection of androgenic disruption due to BPs can be achieved through various methods. In vitro assays using cultured cells, such as prostate cancer cell lines, allow for the assessment of androgenic and antiandrogenic activities of BPs by measuring changes in cellular proliferation and transcriptomic profiles [108]. In vivo models, particularly zebrafish (Danio rerio) and other fish species, are also utilised to observe phenotypic changes linked to androgen disruption, including alterations in growth patterns and reproductive behaviours [53,94]. Advanced techniques such as molecular docking and in silico modelling provide further insight into the binding affinities of BPs to androgen receptors and can inform the development of substitutes that might mitigate these effects [95,109].
The link between gene expression and androgenic disruption, particularly due to environmental contaminants like BPs, is a complex field that underscores how endocrine disruptors can alter reproductive and developmental pathways through modulation of gene regulation. Androgens, which play a crucial role in various physiological processes, influence gene expression by interacting with the androgen receptor (AR). When BPs like BPA and its analogues disrupt this signalling, they can lead to significant changes in gene expression that may alter developmental and reproductive functions across species [85,93]. Androgens exert their effects primarily through the androgen receptor, a transcription factor that, upon binding to its ligand, undergoes a conformational change, allowing it to translocate into the nucleus and bind to androgen response elements (AREs) in the promoters of target genes [110]. This binding promotes the transcription of several genes involved in crucial biological processes, including cell proliferation, differentiation, and survival [110,111]. In healthy conditions, this regulatory mechanism ensures that physiological levels of androgens stimulate the appropriate expression of genes necessary for male sexual differentiation and reproductive function [112]. However, exposure to BPs can disrupt this normal function. These compounds can act as antiandrogens, blocking the action of androgens on the receptor and thereby modulating the expression of genes that depend on AR activity. For instance, studies have shown that bisphenol exposure can lead to the repression of several genes critical for male reproductive health, as the androgen receptor’s ability to activate gene transcription is compromised by these environmental pollutants [9,94]. BPs have been implicated in altering gene expression profiles related to steroidogenesis and spermatogenesis, suggesting that the disruption can have direct repercussions on male fertility and reproductive behaviours [93,109,113]. In addition to these direct effects, androgen signalling can also intersect with other signalling pathways, such as the Wnt/β-catenin pathway, to influence gene expression. For instance, certain genes involved in these pathways are downregulated in response to hormonal disruptions caused by EDCs, further complicating the resultant gene expression landscape [114]. Disruption in AR signalling leads not only to decreased expression of androgen-responsive genes but also to the upregulation of stress response genes, which may act as compensatory mechanisms [1,114]. Such alterations can contribute to developmental anomalies and reproductive diseases via feedback mechanisms that engage various signalling cascades affected by initial endocrine disruption [85,94,113].

4.3. Thyroid Activity and Disruption

Thyroid disruption refers to the interference of normal thyroid gland function by chemical substances that affect the production, release, transport, or action of thyroid hormones in an organism. Studies have shown that BP exposure is associated with thyroid hormone levels and can induce thyroid autoimmunity, leading to thyroid dysfunction. Thyroid hormones play a critical role in regulating a wide range of physiological processes, including growth, development, and metabolism. Disruption of thyroid hormone homeostasis by BPs can lead to impaired growth, delayed development, and other physiological abnormalities in exposed fish [43]. An overview of significant findings on the thyroid effects of selected BPs is given in Table 6 and Table 7. The thyroid-disrupting potential of BPs is linked to several molecular mechanisms. BPs are known to act as antagonists of thyroid hormone receptors (TRs), interfering with the normal signalling pathways regulated by these hormones [13]. BPA, in particular, has been shown to bind to TRs, especially the TRβ isoform, potentially leading to altered gene expression and downstream biological effects [12,115,116]. Additionally, BPs can inhibit the synthesis of thyroid hormones by impairing iodine uptake and reducing the activity of thyroid peroxidase (TPO), an enzyme essential for hormone production [12,117]. These disruptions affect the thyroid axis as hormone synthesis depends on the transport of iodide into thyrocytes [47].
Experimental studies have further supported these findings, showing that BPs can suppress the stimulatory effects of thyroid hormones in a dose-dependent manner. Research has demonstrated that BPs disrupt the expression of genes involved in thyroid function, including thyrotropin (TSH) and deiodinases [118,119]. Analysis of BPA metabolites revealed that, while the 3-hydroxyl metabolite exhibits estrogenic activity, the glucuronide metabolite does not, indicating that bioactivities vary depending on the metabolic transformation [120].
Table 6. Overview of significant findings on the thyroid effects of selected bisphenols in fish (↑ = increase; ↓ = decrease; 3,5-T2 = 3,5-diiodothyronine; dpf = days post-fertilisation; hpf = hours post-fertilisation; HPG = hypothalamic-pituitary–gonadal axis; HPT = Hypothalamic-pituitary-thyroid axis; T3 = triiodothyronine; T4 = thyroxine; TTR = Transthyretin).
Table 6. Overview of significant findings on the thyroid effects of selected bisphenols in fish (↑ = increase; ↓ = decrease; 3,5-T2 = 3,5-diiodothyronine; dpf = days post-fertilisation; hpf = hours post-fertilisation; HPG = hypothalamic-pituitary–gonadal axis; HPT = Hypothalamic-pituitary-thyroid axis; T3 = triiodothyronine; T4 = thyroxine; TTR = Transthyretin).
Bisphenol TypeFish SpeciesExposure DurationConcentration Effects ObservedReferences
BPADanio rerio120 hpf0.4, 2, 10 mg/L↑ T3/T4, altered thyroid development/transport/metabolism genes, delayed hatching[56]
Up to 8 dpf0.2, 0.6, 1.3, 2.8 mg/L↓ T4, ↓ 3,5-T2, disrupted THS homeostasis, altered retinal morphology[121]
Up to 8 dpf0.25, 0.5, 1.2, 4 µg/L
TRβ antagonism, inhibition of deiodinases, and altered phase II enzyme transcripts[122]
96 hpf1 and 100 µg/L↓ heart rate, ↑ SV-BA distance, altered dio3b, thrβ, myh7 gene expression[123]
96 hpf–6 dpf 1–200 mg/Ldevelopmental deformities[50,69]
Oryzias melastigma70 days200 µg/L↓ hatching rate, altered HPG axis gene expression, epigenetic changes in offspring[73]
Pimephales promelas4 days16–1280 µg/L↓ hatchability in F1 generation[69]
BPSDanio rerio7 days (adult)100 µg/L↑ TTR protein (plasma, liver, brain), ↑ T3/T4, thyroid tissue damage, altered HPT axis gene expression[124]
75 days0.1, 1, 10, 100 μg/L↓ body length and weight, ↓ T3 and T4, ↑ liver weight, ↓ hatching rate, ↑ time to hatch[79]
96 h1 and 100 µg/LPromoted heart pumping, altered thrβ, myh7 gene expression[123]
4 hpf–120 dpf1 and 100 µg/L delayed hatching, ↓ offspring survival[61]
Table 7. Overview of significant findings on the thyroid effects of selected bisphenols in fish (↑ = increase; ↓ = decrease; 3,5-T2 = 3,5-diiodothyronine; dpf = days post-fertilisation; hpf = hours post-fertilisation; HPG = hypothalamic-pituitary–gonadal axis; HPT = Hypothalamic-pituitary–thyroid axis; T3 = triiodothyronine; T4 = thyroxine).
Table 7. Overview of significant findings on the thyroid effects of selected bisphenols in fish (↑ = increase; ↓ = decrease; 3,5-T2 = 3,5-diiodothyronine; dpf = days post-fertilisation; hpf = hours post-fertilisation; HPG = hypothalamic-pituitary–gonadal axis; HPT = Hypothalamic-pituitary–thyroid axis; T3 = triiodothyronine; T4 = thyroxine).
Bisphenol TypeFish SpeciesExposure DurationConcentration Effects ObservedReferences
BPFDanio rerio7 days (adult)10 and 100 µg/L↑ TTR protein (plasma, liver, brain), ↑ T3/T4, thyroid follicle pathology, altered HPT axis gene expression[124]
120 hpf (larvae)2 mg/L↑ T3/T4, altered thyroid development/transport/metabolism genes, delayed hatching[56]
96 h1 and 100 µg/Lpromoted heart pumping, altered thrβ, myh7 gene expression[123]
96 hpf–6 dpf 1–200 mg/Ldevelopmental deformities[50,69]
21 days0.1 and 1 mg/Ldisrupted HPG axis gene expression[75]
Oryzias melastigma70 days200 µg/L↓ hatching rate, altered HPG axis gene expression, epigenetic changes in offspring[73]
BPAFDanio rerio4 hpf–120 dpf5, 25, 125 µg/L↑ malformations and lower survival in offspring, delayed hatching, altered HPG axis and liver gene expression[61]
96 hpf–6 dpf1–200 mg/Ldevelopmental deformities[50,69]
4 hpf–120 dpf5, 25, 125 μg/L↑ malformations, ↓ survival rate in offspring, ↓ hatching at 5 μg/L BPAF[92]
Oryzias melastigma70 days200 µg/L↓ hatching rates, altered HPG axis gene expression, epigenetic changes in offspring[73]
BPBDanio rerio21 days1 mg/L↓ hatching rate and viability[77]
BPZDanio rerio120 hpf0.18 and 2.9 µg/Ldisrupted T3/T4, altered thyroid-related gene expression, delayed hatching[56]
TBBPADanio rerio5 dpf100, 200, 300, 400 µg/Linhibition of deiodinases, altered gene expression[125,126]
The impact of BPs on thyroid function is particularly concerning during critical periods of growth and development. Human studies have documented that exposure to BPs during sensitive windows, such as gestation and early childhood, can result in lasting disruptions in thyroid function, which may affect both cognitive and physical development [127,128]. In aquatic models, research on zebrafish (Danio rerio) has shown that waterborne exposure to BPS disrupts thyroid hormone signalling pathways, leading to developmental abnormalities [54]. Furthermore, population-based studies have associated exposure to endocrine-disrupting chemicals (EDCs), such as BPA, with thyroid dysfunction, underlining the need for regulatory scrutiny [129,130].
Finally, BPA exposure has been shown to have variable effects on thyroid hormone levels in both human and animal studies, including increased T4 levels and alterations in thyroid gland weight and histology. These alterations are consistent with its ability to interfere with gene expression related to hormone synthesis and receptor-mediated action [66,90]. It was shown that in zebrafish (Danio rerio) BPA, BPF, and BPS can bind TRβ and exert antagonistic activity. For example, BPF exposure altered T4, T3, and TSH levels and changed the expression of genes including Tg, Ttr, and Ugt1ab. BPS exposure decreased T4 and T3 levels and increased TSH levels. Furthermore, in zebrafish, BPS treatment increased the expression of genes, including Ttr and Ugt1ab [12].
Thyroid disruption in fish, particularly in model species such as zebrafish (Danio rerio), can be assessed through various methodological approaches. Detecting alterations in thyroid function is essential for understanding the ecological impact of these contaminants and for protecting aquatic organisms. One of the most direct approaches involves measuring thyroid hormone levels, such as triiodothyronine (T3) and thyroxine (T4), in plasma or tissue samples from exposed fish. Alterations in these hormone concentrations serve as indicators of disrupted thyroid function. For example, Liu et al. (2022) reported that long-term exposure to TDCPP significantly altered T4 levels in zebrafish (Danio rerio) [131].
Gene expression analysis also plays a critical role in detecting thyroid disruption. Techniques such as quantitative PCR (qPCR) and RNA sequencing are used to assess the expression of genes involved in thyroid hormone regulation, including dio1 and dio2, which are responsible for the conversion of T4 to its more active form, T3. Studies have demonstrated that exposure to various substances alters the expression of thyroid receptor genes in zebrafish larvae, confirming the occurrence of thyroid disruption [125].
Histological examination of thyroid follicles is another important tool. Structural changes in the thyroid gland can reveal damage induced by EDCs. Kraft et al. (2022) showed that exposure to triclosan and benzophenone-2 resulted in morphometric changes in zebrafish (Danio rerio) thyroid follicles, highlighting them as an effective indicator of thyroid disruption [132].
In addition to histology, researchers often monitor developmental abnormalities such as underdeveloped eyes or swim bladders, which may signal disrupted thyroid hormone function. These phenotypic outcomes are particularly relevant during early developmental stages when thyroid hormones play a key regulatory role. While specific references are not always cited for these broader findings, they are supported by studies like Baumann et al. (2016) [125].
In vivo testing using zebrafish embryos and larvae is widely employed to study the effects of EDCs. For instance, Tang et al. (2015) demonstrated that developmental exposure to BPAF disrupted the hypothalamic–pituitary–thyroid (HPT) axis, offering direct evidence of thyroid disruption [133].
Transgenic zebrafish (Danio rerio) models further enhance detection sensitivity. Some lines are engineered with thyroid response elements that trigger the expression of green fluorescent protein (GFP) in response to thyroid hormone activity. This visual and quantifiable response enables effective identification of disrupted thyroid signalling [134].
The development and application of adverse outcome pathway (AOP) frameworks provide a structured means to link molecular or cellular changes caused by EDCs to adverse biological outcomes. AOPs help identify key events and sensitive endpoints, thereby enhancing the ability to assess thyroid disruption systematically. Studies illustrated how AOP networks can be used to correlate environmental exposure with specific endocrine outcomes, offering valuable guidance for regulatory decision making [135,136].

4.4. Glucocorticoid Activity and Disruption

BPs have attracted increasing attention due to their potential to interfere with glucocorticoid hormones, most notably cortisol. Cortisol plays a central role in stress responses, energy metabolism, immune regulation, and other vital physiological processes in vertebrates, including fish [9,137]. In aquatic organisms, exposure to BPs has been linked to altered cortisol levels and disrupted enzyme activity related to glucocorticoid metabolism. These effects can impair the function of the hypothalamic–pituitary–adrenal (HPA) or hypothalamic–pituitary–interrenal (HPI) axis, ultimately compromising the fish’s ability to regulate stress [138,139]. Significant findings on the glucocorticoid disruption changes of BPs are shown in Table 8.
One primary mechanism of endocrine disruption involves the binding of BPs to glucocorticoid receptors (GRs). BPA, in particular, can mimic the action of endogenous glucocorticoids by binding to GRs, thereby disrupting hormonal homeostasis and impairing normal stress regulation. Prolonged or excessive exposure to BPA has been associated with physiological imbalances, including altered cortisol and glucose levels, which can negatively affect metabolic regulation and overall fish health and fitness [3,120].
BPs exert their disruptive effects primarily by interfering with hormone signalling pathways. Through their interaction with nuclear hormone receptors such as GRs, BPs can inhibit or modify the normal transcriptional activity driven by natural glucocorticoids. This antagonistic or agonistic activity alters gene expression and can compromise stress responses and immune function [129].
In addition to receptor-level interactions, BPs can interfere with the biosynthesis of glucocorticoids. These chemicals regulate the expression and function of genes and enzymes involved in the production of steroid hormones. EDCs, including BPs, have been shown to inhibit key enzymes required for cortisol synthesis, leading to dysregulated hormone levels and impaired stress responses [117,119].
BPs also affect the regulation of the HPI axis in fish, disrupting hormonal feedback mechanisms and altering levels of corticotropin-releasing hormone (CRH) and adrenocorticotropic hormone (ACTH). These hormones are essential for proper cortisol regulation, and their dysregulation may contribute to chronic stress conditions and impaired physiological homeostasis [119].
Table 8. Overview of significant findings on the glucocorticoid disruption of selected bisphenols in fish (↑ = increase; ↓ = decrease; hpf = hours post-fertilisation; HPI = hypothalamic–pituitary–interrenal).
Table 8. Overview of significant findings on the glucocorticoid disruption of selected bisphenols in fish (↑ = increase; ↓ = decrease; hpf = hours post-fertilisation; HPI = hypothalamic–pituitary–interrenal).
Bisphenol TypeFish SpeciesExposure DurationConcentration Effects ObservedReferences
BPADanio rerio96 h1500 µg/L↑ anxiety-like behaviour, neurotoxic effects[140]
30 days1, 10, 100 µg/L↑ oxidative stress[3]
Danio rerio,
male
6 weeks100 and 2000 µg/L↑ fat deposition, ↑ body weight, ↑ lipid synthesis, inflammation, antioxidant response[70]
Cyprinus carpio, juvenile30 days0.1, 1, 10, 100, 1000 µg/L(≥0.1 μg/L) ↓ immune response, ↑ oxidative stress[141]
Ctenopharyngodon idella, ovary cells48 h (in vitro)30 μmol/L↑ oxidative stress, altered DNA methylation[142]
Aristichthys nobilis60 days0.1, 1, 10 µg/L↑ oxidative stress, behavioural changes, and physiological disturbances[58]
BPSDanio rerio120 days (embryo to adult)1, 10, 100 µg/L ↑ whole-body cortisol, altered HPI axis gene expression, ↑ anxiety-like behaviour[62]
75 days1, 10, 30 µg/Limpaired anxiety/fear responses, altered antioxidant gene expression[143]
Danio rerio, adult female120 days1, 10, 30 µg/L1 μg/L improved cognitive behaviours,
10/30 μg/L impaired cognitive behaviours, altered glutamatergic signalling
[144]
Cyprinus carpio, juvenile60 days1, 10, 100 µg/L↑ oxidative stress, chronic inflammatory stress in the liver[141]
BPAFDanio rerio, embryo/adult120 hpf5, 50, 500 µg/Laltered anxiety-like/aggressive behaviour,
↑ oxidative stress
[92]
Changes in glucocorticoid signalling caused by bisphenol exposure can directly affect glucose metabolism. Glucocorticoids are crucial regulators of gluconeogenesis and glucose mobilisation. Chronic or excessive glucocorticoid activity, particularly when triggered by EDCs like BPA, can lead to metabolic disturbances such as insulin resistance and disrupted energy balance. These effects are especially harmful during critical periods of development [145].
Experimental studies support the hypothesis that BPs act as GR agonists. For instance, Kouche et al. (2025) demonstrated that BPA significantly activated GRs in cell lines lacking endogenous receptors. The study observed increased transcription of GR-regulated genes, highlighting the capacity of BPA to modulate gene expression pathways related to metabolism and stress responses [146]. Macíková et al. (2014) similarly documented changes in corticosteroid signalling pathways in fish exposed to bisphenols, indicating disruptions to cortisol production and downstream physiological effects, such as altered osmoregulation and immune responses [147].
The disruption of glucocorticoid signalling by bisphenols can produce a range of behavioural and physiological symptoms. Behaviourally, affected fish may show signs of heightened anxiety, increased aggression, reduced foraging, or diminished social interactions—symptoms indicative of impaired stress regulation [126,148]. On a physiological level, bisphenol-induced disruption of glucocorticoid function can lead to immunosuppression, stunted growth, and increased vulnerability to pathogens. Chronic exposure to BPs can result in prolonged activation of the stress axis, reproductive impairments, and developmental abnormalities [115,149]. Endocrine disruption may also manifest through altered gonadal development and reduced fecundity, as glucocorticoid balance is closely linked to the regulation of reproductive hormones. For instance, Wu et al. (2016) found that zebrafish (Danio rerio) exposed to BPA displayed shifts in reproductive hormone levels, suggesting compromised reproductive fitness [117].
The consequences of glucocorticoid disruption extend beyond individual organisms. Disrupted stress and endocrine function can increase mortality, decrease reproductive output, and alter population dynamics. Given the ecological importance of fish as predators, prey, and contributors to nutrient cycling, such disruptions may cascade through aquatic food webs, compromising biodiversity and ecosystem resilience [129,150].
Detecting glucocorticoid alterations in fish, particularly through the measurement of cortisol levels, plays a crucial role in assessing their physiological stress responses. Cortisol is a primary biomarker of stress, and several methods have been developed to measure its concentration in fish. These methods vary in invasiveness, sensitivity, and suitability for different experimental setups or life stages of the fish. Plasma cortisol measurement is one of the most common techniques, providing direct insight into circulating levels of cortisol in the blood. It typically involves blood sampling, followed by analytical techniques like enzyme-linked immunosorbent assay (ELISA) or radioimmunoassay (RIA) for quantification. Although this method is reliable, it is invasive and can induce stress in the fish itself, which may interfere with the results. Despite this limitation, plasma sampling remains a gold standard for establishing baseline cortisol levels. Research by Xiao et al. (2021) highlighted the significance of plasma cortisol as a reliable physiological index of stress, demonstrating its association with alterations in fish metabolism and behaviour [151].
A non-invasive alternative to blood sampling is faecal cortisol analysis. This method involves collecting faecal samples, which can be analysed using techniques such as liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). It allows researchers to monitor chronic stress levels without directly handling the fish. This approach has been validated as effective, as it preserves the welfare of the animals while providing valuable insights into stress responses over time. Meling et al. (2022) demonstrated the efficacy of this method in assessing stress in fish after their transfer to seawater, proving its usefulness in both field and laboratory settings [152].
Cortisol levels can also be measured in fish skin mucus. This method has gained attention due to the close correlation between cortisol in plasma and mucus. Since cortisol diffuses from the bloodstream into the mucus, it can be sampled non-invasively to assess stress, making it a less invasive alternative to blood sampling. The method has been demonstrated to accurately reflect cortisol levels, providing a reliable means for stress monitoring without harming the fish. According to Carbajal et al. (2019), the correlation between plasma and mucus cortisol was strong, supporting the method’s applicability for stress evaluations in both controlled and natural environments [153].
Another approach involves whole-body cortisol quantification. In this method, the entire body of the fish is homogenised, and cortisol is extracted and analysed using techniques such as ELISA. This method is particularly useful for smaller fish species or larvae, providing a comprehensive measurement of an individual’s stress response. However, it is destructive, making it unsuitable for studies that require repeated sampling of the same individual. Yeh et al. (2013) applied this method to larval zebrafish, demonstrating its potential for assessing stress in early life stages [154].
Measuring cortisol in the water surrounding fish is a non-invasive technique that provides insight into their stress levels. As cortisol is released into the surrounding environment, its concentration in water can serve as an indicator of stress without direct interaction with the fish. Waterborne cortisol levels can be measured using advanced techniques, such as chromatography. While this method offers a less intrusive way of monitoring stress, factors such as water volume, flow rate, and fish density can influence the results. Cao et al. (2017) showed that cortisol concentrations in water correlated with fish biomass and stress levels, making this method particularly useful for monitoring fish welfare in aquaculture systems [155].
Advances in sensor technology have led to the development of carbon nanotube-based biosensors that enable real-time, highly sensitive detection of plasma cortisol levels in fish. These sensors represent a promising innovation for rapid cortisol measurement, offering the advantage of being minimally invasive and providing researchers with efficient monitoring tools. While these sensors are still in the early stages of development for widespread field use, they hold significant potential for enhancing stress monitoring in aquatic environments. Wu et al. (2015) demonstrated the utility of carbon nanotube-enhanced immunosensors for cortisol detection, illustrating their potential for high-throughput and real-time stress monitoring [156].
Available sources indicate that bisphenols (such as BPA, BPS, and BPAF) can independently, but often concurrently, induce both oxidative stress and disruption of glucocorticoid systems [46,60,143]. Oxidative stress refers to a state in which there is an imbalance in an organism between the production of reactive oxygen species (ROS)—unstable oxygen-containing molecules—and the organism’s ability to neutralise them using antioxidants [46,142]. When ROS production exceeds antioxidant defences, cellular and tissue damage may occur. Reactive oxygen species include, among others, superoxide anion (O2), hydroxyl radical (OH), hydrogen peroxide (H2O2), and singlet oxygen (O2) [142].
Although the sources do not explicitly state a direct causal link between glucocorticoid disruption and oxidative stress (or vice versa) as the primary mechanism of bisphenol toxicity, it is essential to note that both are significant adverse effects of bisphenol exposure. These contribute to overall toxicity and disruption of the endocrine system within an organism. Glucocorticoids play a crucial role in regulating metabolism and immune function [154]. Disruption of these systems can affect overall homeostasis and the organism’s resilience to stress. Oxidative stress, in turn, directly impacts cellular physiology and can lead to cellular and tissue damage [142].
In the broader context of body physiology, stress responses (mediated by glucocorticoids) and oxidative status are interconnected. Stress can trigger ROS production, and oxidative stress can, in turn, influence hormonal signalling and exacerbate the stress response. However, the specific detailed mechanisms of this mutual interaction in the context of bisphenol action are not explicitly described as a primary effect in the provided excerpts.

5. Conclusions

The widespread presence of BPs, particularly in plastics and consumer products, represents a growing concern for both environmental and public health. These compounds, while useful for their versatility in plastics and other industrial applications, have proven to be potent endocrine disruptors that can cause adverse effects on multiple physiological systems. These compounds, especially BPA and its structural analogues, have been shown to act as potent endocrine disruptors, interfering with hormonal systems across multiple axes—including estrogenic, androgenic, thyroidal, and glucocorticoid pathways. In fish, such disruptions lead to reproductive impairments, developmental anomalies, behavioural alterations, and physiological dysfunctions, posing serious threats to individual fitness and population stability. Despite increasing awareness of the risks posed by BPA, its widespread substitution with structurally similar analogues such as BPS, BPF, and BPAF has introduced additional complexity. Emerging evidence suggests these alternatives may exhibit equal or even greater endocrine-disrupting potency, highlighting the inadequacy of simple chemical substitution without thorough toxicological evaluation.
In conclusion, the presence of BPs in our modern world represents a significant health and environmental concern, requiring a comprehensive and multifaceted response. Their widespread use and complex toxicological profiles make effective regulation challenging, necessitating ongoing research, collaboration, and public education. The transition to alternative compounds must consider the potential for similar or even more potent endocrine-disrupting properties, and risk assessment frameworks should account for the potential synergistic effects of multiple disruptors.
It is vital to emphasise the intricacies involved in understanding BPs’ behaviour and distribution in the environment. Understanding the environmental fate, bioaccumulation, and biomagnification of BPs in aquatic ecosystems is essential for assessing long-term ecological risks. Their persistence in sediments, water, and biota enables continuous exposure of fish and other organisms, exacerbating endocrine disruption across trophic levels. Regulatory measures must consider these factors to minimise exposure of fish and other aquatic organisms. Finally, even after regulatory measures have been implemented, ongoing monitoring and evaluation of BPs and their substitutes are crucial to ensure their efficacy and adapt to new information.
While the transition to alternative compounds has begun, this process is fraught with difficulties, as these substitutes may possess similar or even more potent endocrine-disrupting properties. This highlights the importance of ongoing research and interdisciplinary collaboration in fully understanding the toxicological profiles of these compounds and informing effective risk management strategies. Overall, the regulatory landscape for BPs is dynamic and evolving, necessitating a continued focus on developing robust policies that safeguard public health and environmental sustainability.
In addressing the health and environmental concerns posed by BPs, it is crucial to take a multifaceted approach that combines risk assessment and regulation with public awareness and education. Effective communication of the potential risks associated with these compounds, as well as practical steps individuals can take to limit their exposure, is crucial in fostering a more informed and proactive society.
Given the potential for BPs to leach into aquatic environments, leading to adverse effects on fish and other aquatic organisms, regulating these compounds also requires a deeper understanding of their environmental fate and persistence. Strategies that minimise exposure to marine life and mitigate environmental impacts must be prioritised alongside the protection of human health.

Author Contributions

Writing—original draft preparation, N.P.; editing, funding acquisition, supervision, J.B. All authors have read and agreed to the published version of the manuscript.

Funding

Project was funded by the ERDF/ESF “Profish” (no. CZ.02.1.01/0.0/0.0/16_019/0000869), by the Ministry of Agriculture of the Czech Republic (RO 0523), and the University of Veterinary Sciences Brno (IGA VETUNI: 220/2024/FVHE).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Wang, H.; Li, S.; Wu, T.; Wu, Z.; Guo, J. The effect of androgen on wool follicles and keratin production in Hetian sheep. Braz. J. Biol. 2021, 81, 526–536. [Google Scholar] [CrossRef]
  2. De Coster, S.; van Larebeke, N. Endocrine-disrupting chemicals: Associated disorders and mechanisms of action. J. Environ. Public Health 2012, 2012, 713696. [Google Scholar] [CrossRef]
  3. Faheem, M.; Bhandari, R. Detrimental Effects of bisphenol compounds on physiology and reproduction in fish: A literature review. Environ. Toxicol. Pharmacol. 2021, 81, 103497. [Google Scholar] [CrossRef] [PubMed]
  4. Reid, A.J.; Carlson, A.K.; Creed, I.F.; Eliason, E.J.; Gell, P.A.; Johnson, P.T.J.; Kidd, K.A.; MacCormack, T.J.; Olden, J.D.; Ormerod, S.J.; et al. Emerging threats and persistent conservation challenges for freshwater biodiversity. Biol. Rev. 2018, 94, 849–873. [Google Scholar] [CrossRef] [PubMed]
  5. Martyniuk, C.J.; Mehinto, A.C.; Denslow, N.D. Organochlorine pesticides: Agrochemicals with potent endocrine-disrupting properties in fish. Mol. Cell. Endocrinol. 2020, 507, 110764. [Google Scholar] [CrossRef] [PubMed]
  6. Alves da Silva, A.P.; Lins de Oliveira, C.D.; Siqueira Quirino, A.M.; Morais da Silva, F.D.; de Aquino Saraiva, R.; Santos Silva-Cavalcanti, J. Endocrine disruptors in aquatic environment: Effects and consequences on the biodiversity of fish and amphibian species. Adv. Sci. Technol. 2018, 6, 35. [Google Scholar] [CrossRef]
  7. Pinto, I.P.; Estêvão, D.M.; Power, M.D. Effects of estrogens and estrogenic disrupting compounds on fish mineralized tissues. Mar. Drugs 2014, 12, 4474–4494. [Google Scholar] [CrossRef]
  8. Molina-López, A.M.; Bujalance-Reyes, F.; Ayala-Soldado, N.; Mora-Medina, R.; Lora-Benítez, A.J.; Moyano-Salvago, R. An overview of the health effects of bisphenol A from a One Health perspective. Animals 2023, 13, 2439. [Google Scholar] [CrossRef]
  9. Kim, J.; Kumar, S.; Kumar, V.; Lee, Y.; Kim, Y.; Kumar, V. Bisphenols as a legacy pollutant, and their effects on organ vulnerability. Int. J. Environ. Res. Public Health 2019, 17, 112. [Google Scholar] [CrossRef]
  10. Kim, H.; Lee, S.; Choi, J.; Soung, N.; Heo, J. Effects of bisphenol A and its alternatives, bisphenol F and tetramethyl bisphenol F on osteoclast differentiation. Molecules 2021, 26, 6100. [Google Scholar] [CrossRef]
  11. Ragavan, K.V.; Rastogi, N.K.; Thakur, M.S. Sensors and biosensors for analysis of bisphenol-A. TrAC Trends Anal. Chem. 2013, 52, 248–260. [Google Scholar] [CrossRef]
  12. Kim, M.; Park, Y. Bisphenols and thyroid hormone. Endocrinol. Metab. 2019, 34, 340. [Google Scholar] [CrossRef] [PubMed]
  13. Berto-Júnior, C.; Santos-Silva, A.P.; Ferreira, A.C.F.; Graceli, J.B.; Carvalho, D.P.; Soares, P.; Romeiro, N.C.; Miranda-Alves, L. Unraveling molecular targets of bisphenol A and S in the thyroid gland. Environ. Sci. Pollut. Res. 2018, 25, 26916–26926. [Google Scholar] [CrossRef] [PubMed]
  14. Fabrello, J.; Matozzo, V. Bisphenol analogs in aquatic environments and their effects on marine species—A review. J. Mar. Sci. Eng. 2022, 10, 1271. [Google Scholar] [CrossRef]
  15. Goksøyr, S.; Yadetie, F.; Johansen, C.; Jacobsen, R.; Lille-Langøy, R.; Goksøyr, A.; Karlsen, O. Interaction of bisphenol A and its analogs with estrogen and androgen receptor from Atlantic cod (Gadus morhua). Environ. Sci. Technol. 2024, 58, 14098–14109. [Google Scholar] [CrossRef] [PubMed]
  16. Zaborowska, M.; Wyszkowska, J.; Borowik, A.; Kucharski, J. Bisphenol A—A dangerous pollutant distorting the biological properties of soil. Int. J. Mol. Sci. 2021, 22, 12753. [Google Scholar] [CrossRef]
  17. Chen, D.; Kannan, K.; Tan, H.; Zheng, Z.; Feng, Y.; Wu, Y.; Widelka, M. Bisphenol analogues other than BPA: Environmental occurrence, human exposure, and toxicity—A review. Environ. Sci. Technol. 2016, 50, 5438–5453. [Google Scholar] [CrossRef]
  18. Liao, C.; Liu, F.; Moon, H.-B.; Yamashita, N.; Yun, S.; Kannan, K. Bisphenol analogues in sediments from industrialized areas in the nited States, Japan, and Korea: Spatial and temporal distributions. Environ. Sci. Technol. 2012, 46, 11558–11565. [Google Scholar] [CrossRef]
  19. Qiu, W.; Fang, M.; Liu, J.; Fu, C.; Zheng, C.; Chen, B.; Wang, K. In vivo actions of bisphenol F on the reproductive neuroendocrine system after long-term exposure in zebrafish. Sci. Total Environ. 2019, 665, 995–1002. [Google Scholar] [CrossRef]
  20. Meeker, J.D.; Sathyanarayana, S.; Swan, S.H. Phthalates and other additives in plastics: Human exposure and associated health outcomes. Philos. Trans. R. Soc. B 2009, 364, 2097–2113. [Google Scholar] [CrossRef]
  21. Arcand-Hoy, D.L.; Benson, H.W. Fish reproduction: An ecologically relevant indicator of endocrine disruption. Environ. Toxicol. Chem. 1998, 17, 49–57. [Google Scholar] [CrossRef]
  22. Gao, Y.; Xiao, S.; Wu, Q.; Pan, C. Bisphenol analogues in water and sediment from the Beibu Gulf, South China Sea: Occurrence, partitioning and risk assessment. Sci. Total Environ. 2022, 857, 159445. [Google Scholar] [CrossRef]
  23. Zhang, H.; Zhang, Y.; Li, J.; Yang, M. Occurrence and exposure assessment of bisphenol analogues in source water and drinking water in China. Sci. Total Environ. 2019, 655, 607–613. [Google Scholar] [CrossRef] [PubMed]
  24. Xie, J.; Zhao, N.; Zhang, Y.; Hu, H.; Zhao, M.; Jin, H. Occurrence and partitioning of bisphenol analogues, triclocarban, and triclosan in seawater and sediment from East China Sea. Chemosphere 2021, 287 Pt 2, 132218. [Google Scholar] [CrossRef] [PubMed]
  25. Xiong, J.; Li, G.; An, T.; Zhang, C.; Wei, C. Emission patterns and risk assessment of polybrominated diphenyl ethers and bromophenols in water and sediments from the Beijiang River, South China. Environ. Pollut. 2016, 219, 596–603. [Google Scholar] [CrossRef] [PubMed]
  26. Wang, Q.; Chen, M.; Shan, G.; Chen, P.; Cui, S.; Yi, S.; Zhu, L. Bioaccumulation and biomagnification of emerging bisphenol analogues in aquatic organisms from Taihu Lake, China. Sci. Total Environ. 2017, 598, 814–820. [Google Scholar] [CrossRef]
  27. Akash, H.; Sajid, M.; Rasheed, S.; Rehman, K.; Imran, M.; Assiri, M.A. Toxicological evaluation of bisphenol analogues: Preventive measures and therapeutic interventions. RSC Adv. 2023, 13, 21613–21628. [Google Scholar] [CrossRef]
  28. Braun, M.; Joe, M.; Hauser, R. Bisphenol A and children’s health. Curr. Opin. Pediatr. 2011, 23, 233–239. [Google Scholar] [CrossRef]
  29. Maamar, B.; Lesné, M.; Lethimonier, C.; Coiffec, I.; Lassurguère, J.; Lavoué, V.; Deceuninck, Y.; Antignac, J.-P.; Le Bizec, B.; Perdu, E.; et al. An investigation of the endocrine-disruptive effects of bisphenol A in human and rat fetal testes. PLoS ONE 2015, 10, e0117226. [Google Scholar] [CrossRef]
  30. Lindeman, B.; Ritz, V. Regulation and Risk Management of Endocrine Disruptors: Current Status and Future Perspectives; Royal Society of Chemistry: London, UK, 2020; pp. 495–511. [Google Scholar] [CrossRef]
  31. Wheeler, R.J.; Coady, K.K. Are all chemicals endocrine disruptors? Integr. Environ. Assess. Manag. 2016, 12, 402–403. [Google Scholar] [CrossRef]
  32. Office of the Federal Register, National Archives and Records Administration. 77 FR 41899—Indirect Food Additives: Polymers; Office of the Federal Register, National Archives and Records Administration: Washington, DC, USA, 2012. Available online: https://www.govinfo.gov/app/details/FR-2012-07-17/2012-17366 (accessed on 1 February 2025).
  33. Office of the Federal Register, National Archives and Records Administration. 78 FR 41840—Indirect Food Additives: Adhesives and Components of Coatings; Office of the Federal Register, National Archives and Records Administration: Washington, DC, USA, 2013. Available online: https://www.govinfo.gov/app/details/FR-2013-07-12/2013-16684 (accessed on 12 February 2025).
  34. Grignard, E.; Jesus, D.K.; Hubert, P. Regulatory testing for endocrine disruptors; need for validated methods and integrated approaches. Front. Toxicol. 2022, 3, 821736. [Google Scholar] [CrossRef]
  35. Ministry of Health, People’s Republic of China. Bulletin No. 15 of 2011: Safety Evaluation of Bisphenol A in Food Contact Materials; Ministry of Health, People’s Republic of China: Beijing, China, 2011. [Google Scholar]
  36. Canada Consumer Product Safety Act (S.C. 2010, c. 21); Department of Justice Canada: Ottawa, ON, Canada, 2010.
  37. Ministry of Food and Drug Safety. Standards and Specifications for Utensils, Containers and Packages (2019-2, 20190109); MFDS: Seoul, Republic of Korea, 2019. [Google Scholar]
  38. EU. Commission Implementing Regulation (EU) No 321/2011 of 1 April 2011 Amending Regulation (EU) No 10/2011 as Regards the Restriction of Use of Bisphenol A in Plastic Infant Feeding Bottles. Available online: https://eur-lex.europa.eu/eli/reg_impl/2011/321/oj/eng (accessed on 20 January 2025).
  39. EU. Regulation (EC) No 1907/2006 of the European Parliament and of the Council of 18 December 2006 Concerning the Registration, Evaluation, Authorisation and Restriction of Chemicals (REACH), Establishing a European Chemicals Agency, Amending Directive 1999/45/EC and Repealing Council Regulation (EEC) No 793/93 and Commission Regulation (EC) No 1488/94 as well as Council Directive 76/769/EEC and Commission Directives 91/155/EEC, 93/67/EEC, 93/105/EC and 2000/21/EC. Available online: https://eur-lex.europa.eu/eli/reg/2006/1907/oj/eng (accessed on 20 January 2025).
  40. EU. Commission Regulation (EU) 2016/2235 of 12 December 2016 Amending Annex XVII to Regulation (EC) No 1907/2006 of the European Parliament and of the Council Concerning the Registration, Evaluation, Authorisation and Restriction of Chemicals (REACH) as Regards Bisphenol A. Available online: https://eur-lex.europa.eu/eli/reg/2016/2235/oj/eng (accessed on 20 January 2025).
  41. EU. Commission Regulation (EU) 2024/3190 of 19 December 2024 on the Use of bisphenol A (BPA) and Other Bisphenols and Bisphenol Derivatives with Harmonised Classification for Specific Hazardous Properties in Certain Materials and Articles Intended to Come into Contact with Food, Amending Regulation (EU) No 10/2011 and Repealing Regulation (EU) 2018/213. Available online: https://eur-lex.europa.eu/eli/reg/2024/3190/oj/eng (accessed on 20 January 2025).
  42. Ji, Z.; Liu, J.; Sakkiah, S.; Guo, W.; Hong, H. BPA replacement compounds: Current status and perspectives. ACS Sustain. Chem. Eng. 2021, 9, 2433–2446. [Google Scholar] [CrossRef]
  43. Carnevali, O.; Morini, M.; Santi, A.; Volpi, N.; Neri, F.; Franchi, M.; Maggi, M. Endocrine-disrupting chemicals in aquatic environment: What are the risks for fish gametes? Fish Physiol. Biochem. 2018, 44, 1561–1576. [Google Scholar] [CrossRef] [PubMed]
  44. Crisp, M.; Clegg, E.D.; Cooper, R.L.; Wood, W.R.; Anderson, D.G.; Baetcke, K.P.; Hoffmann, J.; Morrow, M.S.; Rodier, D.J.; Schaeffer, J.E.; et al. Environmental endocrine disruption: An effects assessment and analysis. Natl. Inst. Environ. Health Sci. 1998, 106 (Suppl. S1), 11–56. [Google Scholar] [CrossRef]
  45. Gassman, N. Induction of oxidative stress by bisphenol A and its pleiotropic effects. Environ. Mol. Mutagen. 2017, 58, 60–71. [Google Scholar] [CrossRef]
  46. Kovacic, P. How safe is bisphenol A? Fundamentals of toxicity: Metabolism, electron transfer and oxidative stress. Med. Hypotheses 2010, 75, 1–4. [Google Scholar] [CrossRef]
  47. Silva, D.A.; Martins, M.; Xavier, L.L.F.; Gonçalves, C.F.L.; Santos-Silva, A.P.; Paiva-Melo, F.D.; de Freitas, M.L.; Fortunato, R.S.; Miranda-Alves, L.; Ferreira, A.C.F. Bisphenol A increases hydrogen peroxide generation by thyrocytes both in vivo and in vitro. Endocr. Connect. 2018, 7, 1196–1207. [Google Scholar] [CrossRef]
  48. Stroustrup, A.; Swan, S.H. Endocrine Disruptors. In Textbook of Children’s Environmental Health, 2nd ed.; Etzel, R.A., Landrigan, P.J., Eds.; Oxford Academic: New York, NY, USA, 2024. [Google Scholar] [CrossRef]
  49. Segovia-Mendoza, M.; Palacios-Arreola, M.I.; Monroy-Escamilla, L.M.; Soto-Piña, A.E.; Nava-Castro, K.E.; Becerril-Alarcón, Y.; Camacho-Beiza, R.; Aguirre-Quezada, D.E.; Cardoso-Peña, E.; Amador-Muñoz, O.; et al. Association of serum levels of plasticizers compounds, phthalates and bisphenols, in patients and survivors of breast cancer: A real connection? Int. J. Environ. Res. Public Health 2022, 19, 8040. [Google Scholar] [CrossRef]
  50. Moreman, J.; Lee, O.; Trznadel, M.; David, A.; Kudoh, T.; Tyler, C. Acute Toxicity, Teratogenic, and Estrogenic Effects of bisphenol A and its alternative replacements bisphenol S, bisphenol F, and bisphenol AF in zebrafish embryo-larvae. Environ. Sci. Technol. 2017, 51, 12796–12805. [Google Scholar] [CrossRef]
  51. Naderi, M.; Wong, M.; Gholami, F. Developmental exposure of zebrafish (Danio rerio) to bisphenol-S impairs subsequent reproduction potential and hormonal balance in adults. Aquat. Toxicol. 2014, 148, 195–203. [Google Scholar] [CrossRef]
  52. Park, C.; Kim, G.; On, J.; Pyo, H.; Park, J.; Cho, S. Sex-specific effects of bisphenol S with tissue-specific responsiveness in adult zebrafish: The antiandrogenic and antiestrogenic effects. Ecotoxicol. Environ. Saf. 2021, 229, 113102. [Google Scholar] [CrossRef] [PubMed]
  53. Huang, G.; Tian, X.; Fang, X.; Ji, F. Waterborne exposure to bisphenol F causes thyroid endocrine disruption in zebrafish larvae. Chemosphere 2016, 147, 188–194. [Google Scholar] [CrossRef] [PubMed]
  54. Zhang, D.; Zhou, E.; Yang, Z. Waterborne exposure to BPS causes thyroid endocrine disruption in zebrafish larvae. PLoS ONE 2017, 12, e0176927. [Google Scholar] [CrossRef] [PubMed]
  55. Frenzilli, G.; Martorell-Ribera, J.; Bernardeschi, M.; Scarcelli, V.; Jönsson, E.; Diano, N.; Moggio, M.; Guidi, P.; Sturve, J.; Asker, N. Bisphenol A and bisphenol S induce endocrine and chromosomal alterations in bown trout. Front. Endocrinol. 2021, 12, 645519. [Google Scholar] [CrossRef]
  56. Lee, S.; Kim, C.; Shin, H.; Kho, Y.; Choi, K. Comparison of thyroid hormone disruption potentials by bisphenols A, S, F, and Z in embryo-larval zebrafish. Chemosphere 2019, 221, 115–123. [Google Scholar] [CrossRef]
  57. Wei, P.; Zhao, F.; Zhang, X.; Liu, W.; Jiang, G.; Wang, H.; Ru, S. Transgenerational thyroid endocrine disruption induced by bisphenol S affects the early development of zebrafish offspring. Environ. Pollut. 2018, 243 Pt B, 800–808. [Google Scholar] [CrossRef]
  58. Akram, R.; Iqbal, R.; Hussain, R.; Jabeen, F.; Ali, M. Evaluation of oxidative stress, antioxidant enzymes and genotoxic potential of bisphenol A in fresh water bighead carp (Aristichthys nobilis) fish at low concentrations. Environ. Pollut. 2020, 268, 115896. [Google Scholar] [CrossRef]
  59. Anjali, V.; Remya, V.; Reshmi, S.; Mahim, S.; Devi, C. Impact of bisphenol S as an endocrine disruptor in a freshwater fish, Oreochromis mossambicus. J. Endocrinol. Reprod. 2021, 23, 49–63. [Google Scholar] [CrossRef]
  60. Birceanu, O.; Mai, T.; Vijayan, M. Maternal transfer of bisphenol A impacts the ontogeny of cortisol stress response in rainbow trout. Aquat. Toxicol. 2015, 168, 11–18. [Google Scholar] [CrossRef]
  61. Shi, J.; Jiao, Z.; Zheng, S.; Li, M.; Zhang, J.; Feng, Y.; Yin, J.; Shao, B. Long-term effects of bisphenol AF (BPAF) on hormonal balance and genes of hypothalamus-pituitary-gonad axis and liver of zebrafish (Danio rerio), and the impact on offspring. Chemosphere 2015, 128, 252–257. [Google Scholar] [CrossRef]
  62. Wei, P.; Zhao, F.; Zhang, X.; Ru, S. Long-term exposure of zebrafish to bisphenol S impairs stress function of hypothalamic-pituitary-interrenal axis and causes anxiety-like behavioral responses to novelty. Sci. Total Environ. 2020, 716, 137092. [Google Scholar] [CrossRef] [PubMed]
  63. Arslan, P.; Özeren, S.C.; Dikmen, B.Y. The effects of endocrine disruptors on fish. Environ. Res. Technol. 2021, 4, 145–151. [Google Scholar] [CrossRef]
  64. Wojnarowski, K.; Cholewińska, P.; Palić, D.; Bednarska, M.; Jarosz, M.; Wiśniewska, I. Estrogen receptors mediated negative effects of estrogens and xenoestrogens in teleost fishes—Review. Int. J. Mol. Sci. 2022, 23, 2605. [Google Scholar] [CrossRef] [PubMed]
  65. Wahab, M.; Tyler, R.C. Endocrine disrupting chemicals and sexual behaviors in fish—A critical review on effects and possible consequences. Crit. Rev. Toxicol. 2012, 42, 653–668. [Google Scholar] [CrossRef]
  66. Lombó, M.; Depincé, A.; Labbé, C.; Herráez, M.P. Embryonic exposure to bisphenol A impairs primordial germ cell migration without jeopardizing male breeding capacity. Biomolecules 2019, 9, 307. [Google Scholar] [CrossRef]
  67. Cai, M.; Liu, Y.; Zheng, C.; Zhao, L.; Liu, X.; Hu, M.; Zheng, X. Evaluation of the effects of low concentrations of bisphenol AF on gonadal development using the Xenopus laevis model: A finding of testicular differentiation inhibition coupled with feminization. Environ. Pollut. 2020, 260, 113980. [Google Scholar] [CrossRef]
  68. Laing, L.; Viana, J.; Dempster, E.; Trznadel, M.; Trunkfield, L.; Webster, T.; Van Aerle, R.; Paull, G.; Wilson, R.; Mill, J.; et al. Bisphenol A causes reproductive toxicity, decreases DNMT1 transcription, and reduces global DNA methylation in breeding zebrafish (Danio rerio). Epigenetics 2016, 11, 526–538. [Google Scholar] [CrossRef]
  69. Mu, X.; Huang, Y.; Li, X.; Lei, Y.; Teng, M.; Li, X.; Wang, C. Developmental effects and estrogenicity of bisphenol A alternatives in a zebrafish embryo model. Environ. Sci. Technol. 2018, 52, 3002–3010. [Google Scholar] [CrossRef]
  70. Sun, S.-X.; Wu, J.-L.; Lv, H.-B.; Zhang, H.-Y.; Zhang, J.; Limbu, S.M.; Qiao, F.; Chen, L.-Q.; Yang, Y.; Zhang, M.-L.; et al. Environmental estrogen exposure converts lipid metabolism in male fish to a female pattern mediated by AMPK and mTOR signaling pathways. J. Hazard. Mater. 2020, 394, 122537. [Google Scholar] [CrossRef]
  71. Moreman, J.; Takesono, A.; Trznadel, M.; Winter, M.; Perry, A.; Wood, M.; Rogers, N.; Kudoh, T.; Tyler, C. Estrogenic mechanisms and cardiac responses following early life exposure to bisphenol A (BPA) and its metabolite 4-methyl-2,4-bis(p-hydroxyphenyl)pent-1-ene (MBP) in zebrafish. Environ. Sci. Technol. 2018, 52, 6656–6665. [Google Scholar] [CrossRef]
  72. Sohoni, P.; Tyler, C.R.; Hurd, K.; Caunter, J.; Hetheridge, M.; Williams, T.; Woods, C.; Evans, M.; Toy, R.; Gargas, J.P.; et al. Reproductive effects of long-term exposure to bisphenol A in the fathead minnow (Pimephales promelas). Environ. Sci. Technol. 2001, 35, 2917–2925. [Google Scholar] [CrossRef]
  73. Chen, Z.; Li, X.; Gao, J.; Liu, Y.; Zhang, N.; Guo, Y.; Wang, Z.; Dong, Z. Reproductive toxic effects of chronic exposure to bisphenol A and its analogues in marine medaka (Oryzias melastigma). Aquat. Toxicol. 2024, 271, 106927. [Google Scholar] [CrossRef]
  74. Hatef, A.; Zare, A.; Alavi, S.; Habibi, H.; Linhart, O. Modulations in androgen and estrogen mediating genes and testicular response in male goldfish exposed to bisphenol A. Environ. Toxicol. Chem. 2012, 31, 2069–2077. [Google Scholar] [CrossRef] [PubMed]
  75. Yang, Q.; Yang, X.; Liu, J.; Ren, W.; Chen, Y.; Shen, S. Effects of BPF on steroid hormone homeostasis and gene expression in the hypothalamic–pituitary–gonadal axis of zebrafish. Environ. Sci. Pollut. Res. 2017, 24, 21311–21322. [Google Scholar] [CrossRef] [PubMed]
  76. Meng, X.; Su, S.; Wei, X.; Wang, S.; Guo, T.; Li, J.; Song, H.; Wang, M.; Wang, Z. Exposure to bisphenol A alternatives bisphenol AF and fluorene-9-bisphenol induces gonadal injuries in male zebrafish. Ecotoxicol. Environ. Saf. 2023, 253, 114634. [Google Scholar] [CrossRef] [PubMed]
  77. Serra, H.; Beausoleil, C.; Habert, R.; Minier, C.; Picard-Hagen, N.; Michel, C. Evidence for bisphenol B endocrine properties: Scientific and regulatory perspectives. Environ. Health Perspect. 2019, 127, 106001. [Google Scholar] [CrossRef]
  78. Qiu, W.; Liu, S.; Chen, H.; Luo, S.; Xiong, Y.; Wang, X.; Xu, B.; Zheng, C.; Wang, K. The comparative toxicities of BPA, BPB, BPS, BPF, and BPAF on the reproductive neuroendocrine system of zebrafish embryos and its mechanisms. J. Hazard. Mater. 2020, 406, 124303. [Google Scholar] [CrossRef]
  79. Rochester, J.; Bolden, A. Bisphenol S and F: A systematic review and comparison of the hormonal activity of bisphenol A substitutes. Environ. Health Perspect. 2015, 123, 643–650. [Google Scholar] [CrossRef]
  80. Lan, H.-C.; Wu, K.-Y.; Lin, I.-W.; Yang, Z.-J.; Chang, A.-A.; Hu, M.-C. Bisphenol A disrupts steroidogenesis and induces a sex hormone imbalance through c-Jun phosphorylation in Leydig cells. Chemosphere 2017, 185, 237–246. [Google Scholar] [CrossRef]
  81. Jorgensen, E.; Alderman, M.; Taylor, H. Preferential epigenetic programming of estrogen response after in utero xenoestrogen (bisphenol-A) exposure. FASEB J. 2016, 30, 3194–3201. [Google Scholar] [CrossRef]
  82. Jin, S.; Yang, F.; Xu, Y.; Dai, H.; Liu, W. Risk assessment of xenoestrogens in a typical domestic sewage-holding lake in China. Chemosphere 2013, 93, 892–898. [Google Scholar] [CrossRef]
  83. Kundakovic, M.; Gudsnuk, K.; Franks, B.; Madrid, J.; Miller, R.L.; Perera, F.P.; Champagne, F.A. Sex-specific epigenetic disruption and behavioral changes following low-dose in utero bisphenol A exposure. Proc. Natl. Acad. Sci. USA 2013, 110, 9956–9961. [Google Scholar] [CrossRef] [PubMed]
  84. Mattsson, A.; Brunström, B. Effects of selective and combined activation of estrogen receptor α and β on reproductive organ development and sexual behaviour in Japanese quail (Coturnix japonica). PLoS ONE 2017, 12, e0180548. [Google Scholar] [CrossRef] [PubMed]
  85. Chelcea, I.; Örn, S.; Hamers, T.; Koekkoek, J.; Legradi, J.; Vogs, C.; Andersson, P. Physiologically based toxicokinetic modeling of bisphenols in zebrafish (Danio rerio) accounting for variations in metabolic rates, brain distribution, and liver accumulation. Environ. Sci. Technol. 2022, 56, 10216–10228. [Google Scholar] [CrossRef] [PubMed]
  86. Delfosse, V.; Grimaldi, M.; Pons, J.L.; Boulahtouf, A.; Le Maire, A.; Cavaillès, V.; Labesse, G.; Bourguet, W.; Balaguer, P. Structural and mechanistic insights into bisphenols action provide guidelines for risk assessment and discovery of bisphenol A substitutes. Proc. Natl. Acad. Sci. USA 2012, 109, 14930–14935. [Google Scholar] [CrossRef]
  87. Mesnage, R.; Phedonos, A.; Biserni, M.; Arno, M.; Balu, S.; Corton, J.C.; Ugarte, R.; Antoniou, M.N. Evaluation of estrogen receptor alpha activation by glyphosate-based herbicide constituents. Food Chem. Toxicol. 2017, 108 Pt A, 30–42. [Google Scholar] [CrossRef]
  88. Liu, Y.H.; Huang, J.W.; Huang, Z.; Mei, Y.X.; Zhao, J.L.; Ying, G.G. Tissue-specific bioaccumulation and health risks of bisphenols in wild fish from West and North Rivers, South China. J. Environ. Expo. Assess. 2023, 2, 21. [Google Scholar] [CrossRef]
  89. Cano-Nicolau, J.; Vaillant, C.; Pellegrini, E.; Charlier, T.; Kah, O.; Coumailleau, P. Estrogenic effects of several BPA analogs in the developing zebrafish brain. Front. Neurosci. 2016, 10, 112. [Google Scholar] [CrossRef]
  90. Nakamura, D.; Yanagiba, Y.; Duan, Z.; Ito, Y.; Okamura, A.; Asaeda, N.; Tagawa, Y.; Li, C.; Taya, K.; Zhang, S.; et al. Bisphenol A may cause testosterone reduction by adversely affecting both testis and pituitary systems similar to estradiol. Toxicol. Lett. 2010, 194, 16–25. [Google Scholar] [CrossRef]
  91. Coumailleau, P.; Trempont, S.; Pellegrini, E.; Charlier, T. Impacts of bisphenol A analogues on zebrafish post-embryonic brain. J. Neuroendocrinol. 2020, 32, e12879. [Google Scholar] [CrossRef]
  92. Rao, R.; Cao, X.; Li, L.; Zhou, J.; Sun, D.; Li, B.; Guo, S.; Yuan, R.; Cui, H.; Chen, J. Bisphenol AF induces multiple behavioral and biochemical changes in zebrafish (Danio rerio) at different life stages. Aquat. Toxicol. 2022, 253, 106345. [Google Scholar] [CrossRef]
  93. Ahn, C.; Jeung, E. Endocrine-disrupting chemicals and disease endpoints. Int. J. Mol. Sci. 2023, 24, 5342. [Google Scholar] [CrossRef] [PubMed]
  94. Kinch, C.; Ibhazehiebo, K.; Jeong, J.; Habibi, H.; Kurrasch, D. Low-dose exposure to bisphenol A and replacement bisphenol S induces precocious hypothalamic neurogenesis in embryonic zebrafish. Proc. Natl. Acad. Sci. USA 2015, 112, 1475–1480. [Google Scholar] [CrossRef] [PubMed]
  95. Chen, Q.; Wang, X.; Tan, H.; Shi, W.; Zhang, X.; Wei, S.; Yu, H. Molecular initiating events of bisphenols on androgen receptor-mediated pathways provide guidelines for in silico screening and design of substitute compounds. Environ. Sci. Technol. Lett. 2019, 6, 205–210. [Google Scholar] [CrossRef]
  96. Li, X.; Wen, Z.; Wang, Y.; Mo, J.; Zhong, Y.; Ge, R. Bisphenols and Leydig cell development and function. Front. Endocrinol. 2020, 11, 447. [Google Scholar] [CrossRef] [PubMed]
  97. Werner, J.; Ouellet, J.; Cheng, C.; Ju, Y.; Law, R. Pulp and paper mill effluents induce distinct gene expression changes linked to androgenic and estrogenic responses in the fathead minnow (Pimephales promelas). Environ. Toxicol. Chem. 2009, 29, 430–439. [Google Scholar] [CrossRef]
  98. Larsen, M.; Baatrup, E. Functional behavior and reproduction in androgenic sex reversed zebrafish (Danio rerio). Environ. Toxicol. Chem. 2010, 29, 1828–1833. [Google Scholar] [CrossRef]
  99. Brander, S.; Connon, R.; He, G.; Hobbs, J.; Smalling, K.; Teh, S.; Cherr, G. From ‘omics to otoliths: Responses of an estuarine fish to endocrine disrupting compounds across biological scales. PLoS ONE 2013, 8, e74251. [Google Scholar] [CrossRef]
  100. Huang, G.; Liu, Y.; Chen, X.; Liang, Y.; Liu, S.; Yang, Y.; Ying, G. Feminization and masculinization of western mosquitofish (Gambusia affinis) observed in rivers impacted by municipal wastewaters. Sci. Rep. 2016, 6, 20884. [Google Scholar] [CrossRef]
  101. Brockmeier, E.; Jayasinghe, B.; Pine, W.; Wilkinson, K.; Denslow, N. Exposure to paper mill effluent at a site in North Central Florida elicits molecular-level changes in gene expression indicative of progesterone and androgen exposure. PLoS ONE 2014, 9, e106644. [Google Scholar] [CrossRef]
  102. Narita, Y.; Tsutiya, A.; Nakano, Y.; Ashitomi, M.; Sato, K.; Hosono, K.; Ohtani-Kaneko, R. Androgen-induced cellular proliferation, neurogenesis, and generation of GnRH3 neurons in the brain of mature female Mozambique tilapia. Sci. Rep. 2018, 8, 35303. [Google Scholar] [CrossRef] [PubMed]
  103. Chen, L.; Jiang, X.; Feng, H.; Shi, H.; Sun, L.; Tao, W.; Wang, D. Simultaneous exposure to estrogen and androgen resulted in feminization and endocrine disruption. J. Endocrinol. 2016, 228, 205–218. [Google Scholar] [CrossRef] [PubMed]
  104. Rolland, A.; Lardenois, A.; Goupil, A.; Lareyre, J.; Houlgatte, R.; Chalmel, F.; Gac, F. Profiling of androgen response in rainbow trout pubertal testis: Relevance to male gonad development and spermatogenesis. PLoS ONE 2013, 8, e53302. [Google Scholar] [CrossRef] [PubMed]
  105. Orlando, E.; Davis, W.; Guillette, L. Aromatase activity in the ovary and brain of the eastern mosquitofish (Gambusia holbrooki) exposed to paper mill effluent. Environ. Health Perspect. 2002, 110 (Suppl. S3), 429–433. [Google Scholar] [CrossRef]
  106. León, A.; Wu, P.; Hall, L.; Johnson, M.; Teh, S. Global gene expression profiling of androgen disruption in QURT strain medaka. Environ. Sci. Technol. 2007, 42, 962–969. [Google Scholar] [CrossRef]
  107. Schooley, D.J.; Geik, A.; Scarnecchia, L.D. First observations of intersex development in paddlefish Polyodon spathula. J. Fish Biol. 2020, 97, 919–925. [Google Scholar] [CrossRef]
  108. Cortés-Ramírez, S.; Salazar, A.; Sordo, M.; Ostrosky-Wegman, P.; Morales-Pacheco, M.; Cruz-Burgos, M.; Rodríguez-Dorantes, M. Transcriptome-wide analysis of low-concentration exposure to bisphenol A, S, and F in prostate cancer cells. Int. J. Mol. Sci. 2023, 24, 9462. [Google Scholar] [CrossRef]
  109. Conroy-Ben, O.; García, I.; Teske, S. In silico binding of 4,4′-bisphenols predicts in vitro estrogenic and antiandrogenic activity. Environ. Toxicol. 2018, 33, 569–578. [Google Scholar] [CrossRef]
  110. Zhang, Z.; Jin, B.; Jin, Y.; Huang, S.; Niu, X.; Mao, Z.; Xin, D. Pttg1, a novel androgen responsive gene is required for androgen-induced prostate cancer cell growth and invasion. Exp. Cell Res. 2017, 350, 1–8. [Google Scholar] [CrossRef]
  111. Sen, A.; Prizant, H.; Light, A.; Biswas, A.; Hayes, E.; Lee, H.; Hammes, S. Androgens regulate ovarian follicular development by increasing follicle stimulating hormone receptor and microrna-125b expression. Proc. Natl. Acad. Sci. USA 2014, 111, 3008–3013. [Google Scholar] [CrossRef]
  112. Bu, H.; Narisu, N.; Schlick, B.; Rainer, J.; Manke, T.; Schäfer, G.; Klocker, H. Putative prostate cancer risk SNP in an androgen receptor-binding site of the melanophilin gene illustrates enrichment of risk SNPs in androgen receptor target sites. Hum. Mutat. 2015, 37, 52–64. [Google Scholar] [CrossRef]
  113. Martinez, H.; Hsiao, J.; Jasavala, R.; Hinkson, I.; Eng, J.; Wright, M. Androgen-sensitive microsomal signaling networks coupled to the proliferation and differentiation of human prostate cancer cells. Genes Cancer 2011, 2, 956–978. [Google Scholar] [CrossRef] [PubMed]
  114. Tan, H.; Wu, G.; Wang, S.; Lawless, J.; Sinn, A.; Chen, D.; Zheng, Z. Prenatal exposure to atrazine induces cryptorchidism and hypospadias in F1 male mouse offspring. Birth Defects Res. 2021, 113, 469–484. [Google Scholar] [CrossRef] [PubMed]
  115. Beg, M.; Sheikh, I. Endocrine disruption: Molecular interactions of environmental bisphenol contaminants with thyroid hormone receptor and thyroxine-binding globulin. Toxicol. Ind. Health 2020, 36, 322–335. [Google Scholar] [CrossRef] [PubMed]
  116. Moriyama, K.; Tagami, T.; Akamizu, T.; Usui, T.; Saijo, M.; Kanamoto, N.; Hataya, Y.; Shimatsu, A.; Kuzuya, H.; Nakao, K. Thyroid hormone action is disrupted by bisphenol A as an antagonist. J. Clin. Endocrinol. Metab. 2002, 87, 5185–5190. [Google Scholar] [CrossRef]
  117. Wu, Y.; Beland, F.; Fang, J. Effect of triclosan, triclocarban, 2,2′,4,4′-tetrabromodiphenyl ether, and bisphenol A on the iodide uptake, thyroid peroxidase activity, and expression of genes involved in thyroid hormone synthesis. Toxicol. Vitr. 2016, 32, 310–319. [Google Scholar] [CrossRef]
  118. Andrianou, X.; Gängler, S.; Piciu, A.; Charisiadis, P.; Zira, C.; Aristidou, K.; Makris, K. Human exposures to bisphenol A, bisphenol F and chlorinated bisphenol A derivatives and thyroid function. PLoS ONE 2016, 11, e0155237. [Google Scholar] [CrossRef]
  119. Choi, J.; Choi, E.; Lee, S.; Park, B.; Lee, H.; Hong, Y.; Park, H. Relationship of urinary bisphenol A in childhood on thyroid hormone function in adolescents: A cohort study. PLoS ONE 2023, 20, e0322658. [Google Scholar] [CrossRef]
  120. Kitamura, S. Comparative study of the endocrine-disrupting activity of bisphenol A and 19 related compounds. Toxicol. Sci. 2005, 84, 249–259. [Google Scholar] [CrossRef]
  121. Volz, S.; Poulsen, R.; Hansen, M.; Holbech, H. Bisphenol A alters retinal morphology, visually guided behavior, and thyroid hormone levels in zebrafish larvae. Chemosphere 2023, 340, 140776. [Google Scholar] [CrossRef]
  122. Yang, J.; Chan, K. Evaluation of the toxic effects of brominated compounds (BDE-47, 99, 209, TBBPA) and bisphenol A (BPA) using a zebrafish liver cell line, ZFL. Aquat. Toxicol. 2015, 159, 138–147. [Google Scholar] [CrossRef]
  123. Qin, J.; Jia, W.; Ru, S.; Xiong, J.; Wang, J.; Wang, W.; Hao, L.; Zhang, X. Bisphenols induce cardiotoxicity in zebrafish embryos: Role of the thyroid hormone receptor pathway. Aquat. Toxicol. 2022, 254, 106354. [Google Scholar] [CrossRef]
  124. Zheng, Y.; Li, Y.; Zhang, Z.; Liu, M.; Cui, X.; Wang, J. Evaluation of thyroid-disrupting effects of bisphenol F and bisphenol S on zebrafish (Danio rerio) using anti-transthyretin monoclonal antibody-based immunoassays. Aquat. Toxicol. 2024, 273, 106968. [Google Scholar] [CrossRef] [PubMed]
  125. Baumann, L.; Ros, A.; Rehberger, K.; Neuhauss, S.; Segner, H. Thyroid disruption in zebrafish (Danio rerio) larvae: Different molecular response patterns lead to impaired eye development and visual functions. Aquat. Toxicol. 2016, 172, 44–55. [Google Scholar] [CrossRef] [PubMed]
  126. Baumann, L.; Segner, H.; Ros, A.; Knapen, D.; Vergauwen, L. Thyroid hormone disruptors interfere with molecular pathways of eye development and function in zebrafish. Int. J. Mol. Sci. 2019, 20, 1543. [Google Scholar] [CrossRef] [PubMed]
  127. Liu, J.; Tian, M.; Qin, H.; Chen, D.; Mzava, S.; Wang, X.; Bigambo, F. Maternal bisphenols exposure and thyroid function in children: A systematic review and meta-analysis. Front. Endocrinol. 2024, 15, 1420540. [Google Scholar] [CrossRef]
  128. Guignard, D.; Gayrard, V.; Lacroix, M.; Puel, S.; Picard-Hagen, N.; Viguié, C. Evidence for bisphenol A-induced disruption of maternal thyroid homeostasis in the pregnant ewe at low level representative of human exposure. Chemosphere 2017, 182, 458–467. [Google Scholar] [CrossRef]
  129. Gorini, F.; Bustaffa, E.; Coi, A.; Iervasi, G.; Bianchi, F. Bisphenols as environmental triggers of thyroid dysfunction: Clues and evidence. Int. J. Environ. Res. Public Health 2020, 17, 2654. [Google Scholar] [CrossRef]
  130. Lü, W.; Zhuo, S.; Wang, Z.; Qu, M.; Shi, Z.; Song, Q.; Zang, J. The joint effects of bisphenols and iodine exposure on thyroid during pregnancy. Nutrients 2023, 15, 3422. [Google Scholar] [CrossRef]
  131. Liu, X.; Lu, X.; Hong, J.; Zhang, J.; Lin, J.; Jiang, M.; Zhang, J. Effects of long-term exposure to TDCPP in zebrafish (Danio rerio)—Alternations of hormone balance and gene transcriptions along hypothalamus-pituitary axes. Anim. Models Exp. Med. 2022, 5, 239–247. [Google Scholar] [CrossRef]
  132. Kraft, M.; Gölz, L.; Rinderknecht, M.; Koegst, J.; Braunbeck, T.; Baumann, L. Developmental exposure to triclosan and benzophenone-2 causes morphological alterations in zebrafish (Danio rerio) thyroid follicles and eyes. Environ. Sci. Pollut. Res. 2022, 30, 33711–33724. [Google Scholar] [CrossRef] [PubMed]
  133. Tang, T.; Yang, Y.; Chen, Y.; Tang, W.; Wang, F.; Diao, X. Thyroid disruption in zebrafish larvae by short-term exposure to bisphenol AF. Int. J. Environ. Res. Public Health 2015, 12, 13069–13084. [Google Scholar] [CrossRef] [PubMed]
  134. Špirhanzlová, P.; Leemans, M.; Demeneix, B.; Fini, J. Following endocrine-disrupting effects on gene expression in Xenopus laevis. Cold Spring Harb. Protoc. 2019, 2019, pdb-prot098301. [Google Scholar] [CrossRef] [PubMed]
  135. Knapen, D.; Stinckens, E.; Cavallin, J.; Ankley, G.; Holbech, H.; Villeneuve, D.; Vergauwen, L. Toward an AOP network-based tiered testing strategy for the assessment of thyroid hormone disruption. Environ. Sci. Technol. 2020, 54, 8491–8499. [Google Scholar] [CrossRef]
  136. Holbech, H.; Matthiessen, P.; Hansen, M.; Schüürmann, G.; Knapen, D.; Reuver, M.; Baumann, L. ERGO: Breaking down the wall between human health and environmental testing of endocrine disrupters. Int. J. Mol. Sci. 2020, 21, 2954. [Google Scholar] [CrossRef]
  137. Franchimont, D.; Kino, T.; Galon, J.; Meduri, G.U.; Chrousos, G. Glucocorticoids and inflammation revisited: The state of the art. NIH clinical staff conference. Neuroimmunomodulation 2002, 10, 247–260. [Google Scholar] [CrossRef]
  138. Barton, B.; Iwama, K.G. Physiological changes in fish from stress in aquaculture with emphasis on the response and effects of corticosteroids. Annu. Rev. Fish Dis. 1991, 1, 3–26. [Google Scholar] [CrossRef]
  139. Zhang, Y.; Ren, X.; Li, Y.; Yao, X.; Li, C.; Qin, Z.; Guo, L. Bisphenol A alternatives bisphenol S and bisphenol F interfere with thyroid hormone signaling pathway in vitro and in vivo. Environ. Int. 2018, 237, 1072–1079. [Google Scholar] [CrossRef]
  140. Heredia-García, G.; Elizalde-Velázquez, G.; Gómez-Oliván, L.; Islas-Flores, H.; García-Medina, S.; Galar-Martínez, M.; Dublán-García, O. Realistic concentrations of bisphenol A trigger a neurotoxic response in the brain of zebrafish: Oxidative stress, behavioral impairment, acetylcholinesterase inhibition, and gene expression disruption. Chemosphere 2023, 330, 138729. [Google Scholar] [CrossRef]
  141. Qiu, W.; Chen, J.; Li, Y.; Chen, Z.; Jiang, L.; Yang, M.; Wu, M. Oxidative stress and immune disturbance after long-term exposure to bisphenol A in juvenile common carp (Cyprinus carpio). Ecotoxicol. Environ. Saf. 2016, 130, 93–102. [Google Scholar] [CrossRef]
  142. Fan, X.; Hou, T.; Jia, J.; Tang, K.; Wei, X.; Wang, Z. Discrepant dose responses of bisphenol A on oxidative stress and DNA methylation in grass carp ovary cells. Chemosphere 2020, 248, 126110. [Google Scholar] [CrossRef]
  143. Salahinejad, A.; Attaran, A.; Naderi, M.; Meuthen, D.; Niyogi, S.; Chivers, D. Chronic exposure to bisphenol S induces oxidative stress, abnormal anxiety, and fear responses in adult zebrafish (Danio rerio). Sci. Total Environ. 2020, 750, 141633. [Google Scholar] [CrossRef]
  144. Naderi, M.; Salahinejad, A.; Attaran, A.; Chivers, D.; Niyogi, S. Chronic exposure to environmentally relevant concentrations of bisphenol S differentially affects cognitive behaviors in adult female zebrafish. Environ. Pollut. 2020, 261, 114060. [Google Scholar] [CrossRef] [PubMed]
  145. Lee, R.; Harris, C.; Wang, J. Glucocorticoid receptor and adipocyte biology. Nucl. Recept. Res. 2018, 5, 101373. [Google Scholar] [CrossRef] [PubMed]
  146. Kouche, S.; Halvick, S.; Morel, C.; Duca, R.; Nieuwenhuyse, A.; Turner, J.; Meyre, D. Pollution, stress response, and obesity: A systematic review. Obes. Rev. 2025, 26, e13895. [Google Scholar] [CrossRef] [PubMed]
  147. Macíková, P.; Groh, K.; Ammann, A.; Schirmer, K.; Suter, M. Endocrine disrupting compounds affecting corticosteroid signaling pathways in Czech and Swiss waters: Potential impact on fish. Environ. Sci. Technol. 2014, 48, 12902–12911. [Google Scholar] [CrossRef]
  148. Biddie, S.; Conway-Campbell, B.; Lightman, S. Dynamic regulation of glucocorticoid signalling in health and disease. Rheumatology 2011, 51, 403–412. [Google Scholar] [CrossRef]
  149. Darbre, P. Chemical components of plastics as endocrine disruptors: Overview and commentary. Birth Defects Res. 2020, 112, 1300–1307. [Google Scholar] [CrossRef]
  150. Liang, W.; Chen, Y.; Huang, M.; Song, Z.; Li, C.; Zheng, Y.; Yi, Z. Interaction between halogenated phenols and thyroxine-binding protein: A multispectral and computational approach. ChemistrySelect 2024, 9, e202401288. [Google Scholar] [CrossRef]
  151. Xiao, X.; Wang, X.; Liu, W.-B.; Zhang, D.-D.; Li, X.-F.; Zhang, C.-N.; Chen, W.-H.; Abasubong, K.P.; Jiang, G.-Z. Corticosterone Can Be an Essential Stress Index in Channel Catfish (Ictalurus punctatus). Front. Mar. Sci. 2021, 8, 692726. [Google Scholar] [CrossRef]
  152. Meling, H.; Berge, K.; Knudsen, D.L.; Rønning, P.O.; Brede, C. Monitoring farmed fish welfare by measurement of cortisol as stress marker in fish feces by liquid chromatography coupled with tandem mass spectrometry. Molecules 2022, 27, 2481. [Google Scholar] [CrossRef]
  153. Carbajal, A.; Soler, P.; Tallo-Parra, O.; Isasa, M.; Echevarria, C.; Lopez-Bejar, M.; Vinyoles, D. Towards non-invasive methods in measuring fish welfare: The measurement of cortisol concentrations in fish skin mucus as a biomarker of habitat quality. Animals 2019, 9, 839. [Google Scholar] [CrossRef]
  154. Yeh, P.; Glöck, M.; Ryu, S. An optimized whole-body cortisol quantification method for assessing stress levels in larval zebrafish. PLoS ONE 2013, 8, e79406. [Google Scholar] [CrossRef]
  155. Cao, Y.; Tveten, A.-K.; Stene, A. Establishment of a non-invasive method for stress evaluation in farmed salmon based on direct fecal corticoid metabolites measurement. Fish Shellfish. Immunol. 2017, 68, 317–324. [Google Scholar] [CrossRef]
  156. Wu, Y.; Ohnuki, H.; Ren, H.; Endo, H. Carbon-nanotube-enhanced label-free immunosensor for highly sensitive detection of plasma cortisol level in fish. Sens. Mater. 2015, 27, 793–803. [Google Scholar] [CrossRef]
Figure 1. The chemical structure of substances for which the definitions of “bisphenol” and “bisphenol derivative” apply. (a) Bisphenol structure, (b) Bisphenol derivative structure. Notes: X refers to any bridging group separating the two phenyl rings by one single atom, but the atom can have any substituent(s). R1 to R10 refers to any substituent. At least one of the substituents is not a hydrogen atom (H).
Figure 1. The chemical structure of substances for which the definitions of “bisphenol” and “bisphenol derivative” apply. (a) Bisphenol structure, (b) Bisphenol derivative structure. Notes: X refers to any bridging group separating the two phenyl rings by one single atom, but the atom can have any substituent(s). R1 to R10 refers to any substituent. At least one of the substituents is not a hydrogen atom (H).
Fishes 10 00365 g001
Table 1. BPA and other specific bisphenol analogues, which are most commonly used and further discussed in the review, along with their chemical formula and structure.
Table 1. BPA and other specific bisphenol analogues, which are most commonly used and further discussed in the review, along with their chemical formula and structure.
Bisphenol TypeChemical FormulaStructural formula
Bisphenol A (BPA)C15H16O2Fishes 10 00365 i001
Bisphenol S (BPS)C12H10O4SFishes 10 00365 i002
Bisphenol F (BPF)C13H12O2Fishes 10 00365 i003
Bisphenol AF (BPAF)C15H10F6O2Fishes 10 00365 i004
Bisphenol B (BPB)C16H18O2Fishes 10 00365 i005
Bisphenol E (BPE)C14H14O2Fishes 10 00365 i006
Bisphenol Z (BPZ)C18H22O2Fishes 10 00365 i007
Tetrabromobisphenol A (TBBPA)C15H12Br4O2Fishes 10 00365 i008
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

Peskova, N.; Blahova, J. Bisphenols: Endocrine Disruptors and Their Impact on Fish: A Review. Fishes 2025, 10, 365. https://doi.org/10.3390/fishes10080365

AMA Style

Peskova N, Blahova J. Bisphenols: Endocrine Disruptors and Their Impact on Fish: A Review. Fishes. 2025; 10(8):365. https://doi.org/10.3390/fishes10080365

Chicago/Turabian Style

Peskova, Nikola, and Jana Blahova. 2025. "Bisphenols: Endocrine Disruptors and Their Impact on Fish: A Review" Fishes 10, no. 8: 365. https://doi.org/10.3390/fishes10080365

APA Style

Peskova, N., & Blahova, J. (2025). Bisphenols: Endocrine Disruptors and Their Impact on Fish: A Review. Fishes, 10(8), 365. https://doi.org/10.3390/fishes10080365

Article Metrics

Back to TopTop