Next Article in Journal
Distribution and Comparative Analysis of Genomic Microsatellites in Nine Species of Family Sillaginidae
Previous Article in Journal
Modeling Dress-Out Traits Based on Morphological Traits in the Siberian Prawn, Exopalaemon modestus (Heller, 1862)
Previous Article in Special Issue
Fish Alone Are Not Enough: Zoobenthos Improves Water Quality Assessment in Impacted Rivers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Review on Toxicity Effect of Emerging Contaminants on Trans-/Multi-Generational Fish

1
College of Chemistry and Environment, Guangdong Ocean University, Zhanjiang 524088, China
2
Zhanjiang Key Laboratory of Pollutant Control and Ecological Restoration for Coastal Marine Aquaculture, Guangdong Ocean University, Zhanjiang 524088, China
3
Research Center for Coastal Environmental Protection and Ecological Resilience, Guangdong Ocean University, Zhanjiang 524088, China
*
Authors to whom correspondence should be addressed.
Fishes 2025, 10(11), 535; https://doi.org/10.3390/fishes10110535
Submission received: 25 August 2025 / Revised: 17 October 2025 / Accepted: 18 October 2025 / Published: 22 October 2025
(This article belongs to the Special Issue Toxicology of Anthropogenic Pollutants on Fish)

Abstract

In recent years, toxicological studies on fish exposed to emerging contaminants (ECs) have been relatively in-depth. However, research on trans-/multi-generational exposure of fish to ECs remains scarce. Limited data indicate that when parental generations (P) are subjected to ECs stress, it can threaten the normal transmission of functions in offspring, such as growth and development, reproduction, physiology, endocrine, neural, and behavioral functions. Even after the exposure is terminated, these negative impacts may persist. Under the long-term presence of ECs, the health of fish offspring may affect the survival of entire populations and the stability of ecosystems. Therefore, this review summarizes studies on trans-/multi-generational effects of ECs on fish and analyzes these research results. Based on the materials collected, more research on trans-/multi-generational ECs effects on fish is urgently needed, especially regarding the F3 generation, combined toxicity, and trans-/multi-generational epigenetic effects. This will enable a comprehensive assessment of the health and ecological risks posed by ECs at environmental concentrations to fish.
Key Contribution: This review summarizes the trans-/multi-generational toxic effects and mechanisms of emerging contaminants (ECs) on fish. When parental generations are subjected to stress, it can threaten the normal transmission of functions in offspring, such as growth and development, reproduction, physiology, and endocrine, neural and behavioral functions. Even after the exposure is terminated, these negative impacts still persist. The long-term presence of ECs may affect the survival of fish populations.

Graphical Abstract

1. Introduction

With the rapid globalization of the economy and accelerated urbanization, the demand for synthetic materials in industry and agriculture has increased exponentially. This trend, coupled with relatively relaxed environmental policies, has led to widespread pollution across various ecosystems in the world. In particular, emerging contaminants (ECs) pose critical threats to the health of coastal, estuarine, and marine ecosystems, as well as human beings. ECs are anthropogenic pollutants recently detected in the environment or newly recognized for their adverse effects, and they remain unregulated or inadequately addressed by current laws and standards (Figure 1) [1]. Moreover, environmentally relevant concentrations of ECs can have detrimental impacts on the biota and human health [2]. This category includes Pharmaceutical and Personal Care Products (PPCPs), Micro/Nano Particles, Endocrine Disrupting Compounds (EDCs), Industrial substances, and other synthetic chemicals of emerging concern (Figure 1). Many ECs exhibit concerning properties including bioaccumulation, environmental persistence, reproductive toxicity, and genotoxicity, often at trace concentrations [3,4,5]. These properties not only impact exposed individual organisms but also transfer health risks across generations via food chain dynamics, potentially triggering cascading ecological and public health crises.
ECs are widely distributed in ecosystems. Some of these contaminants are bioaccumulative and can persist for a long time, meaning their toxic effects may continuously interfere with the growth and reproduction of organisms over an extended period [6,7]. Studies have shown that even after exposure ceases, the residual toxicity of EDCs may still trigger long-term physiological changes in organisms, impairing the health and genetic stability of offspring [8,9,10]. What is more challenging is that ECs have complex and diverse chemical compositions, and these substances exhibit multi-dimensional toxic mechanisms of action, making monitoring and governance efforts extremely difficult [11,12]. Therefore, in-depth research on the characteristics of ECs, their toxic effects on organisms, and their mechanisms of action holds urgent scientific value and practical significance.
A critical limitation in current ecotoxicological assessment is the disconnect between laboratory studies and environmental reality [13]. Most toxicological experiments employ concentrations far exceeding environmental levels and focus on short-term exposures, potentially underestimating the true long-term risks of ECs at environmentally relevant concentrations [14,15]. As a result, current regulatory frameworks, which typically assess single compounds, may underestimate the toxic impacts of environmentally relevant concentrations of ECs on aquatic organisms and humans [16]. Persistent trans-/multi-generational pollutant exposure effectively addresses these problems.
Transgenerational and multigenerational exposure studies address this gap by examining continuous, long-term effects across multiple generations at environmentally relevant concentrations (Figure 2) [14,17,18]. It allows extrapolation to humans, helping identify potential hazards of such pollutants to human health. However, transgenerational studies remain severely limited due to their complexity, time-consuming (often >6 months), and numerous methodological challenges. The accumulation of technical variables and potential experimental disruptions over such extended periods increases the risk of confounding factors, resulting in a severe lack of reliable multigenerational data in this field.
ECs due to their environmental persistence and bioaccumulative properties, can accumulate in fish over time even under low concentration exposure. This accumulation eventually reaches toxic levels. Over time, they cause persistent damage to fish physiology, growth, reproduction, and behavior. These effects may also be transgenerationally transmissible. Therefore, this review (1) synthesizes all available transgenerational toxicity data for major EC categories, (2) analyzes mechanisms underlying transgenerational inheritance, (3) identifies critical data gaps, and (4) discusses emerging methodologies that may accelerate research.

2. The Toxic Action of Typical ECs

We will briefly interpret the toxic effects and action mechanisms of emerging pollutants on marine organisms in recent years and discuss them from the aspects of multigenerational, transgenerational, and epigenetic inheritance. While ECs encompass a broad range of substances (Figure 1), transgenerational studies have primarily concentrated on specific categories. The following sections examine those ECs with robust trans-/multi-generational datasets in fish, including micro/nano plastics (MNPs), PPCPs and EDCs. Additionally, it is critical to distinguish between multigenerational and truly transgenerational effects. The F1 generation represents offspring directly exposed to contaminants during their development (either as embryos or as primordial germ cells in the exposed mother). The F2 generation may also experience direct exposure as primordial germ cells during F1 fetal development. Therefore, effects observed in F1 and F2 do not constitute true transgenerational inheritance, as these generations were potentially exposed during critical developmental windows. Only effects observed in the F3 generation and beyond, which were never directly exposed at any developmental stage, represent genuine transgenerational epigenetic inheritance [19,20]. Throughout this review, we use ‘multigenerational’ for studies examining F1–F2 generations and ‘transgenerational’ only for studies extending to F3 or later generations.

2.1. Micro/Nano Plastics (MNPs)

Over recent decades, the growing production and use of plastics have led to increasing accumulation of microplastics and nanoplastics (MNPs) in natural environments, creating a severe environmental problem. In aquatic environments, the concentration of MNPs can range from several micrograms to several milligrams per liter, and they are extremely widely distributed [21,22]. The ocean is widely recognized as a major sink for these pollutants [23,24]. When ingested by marine organisms, MNPs cause significant harm to metabolism, growth, development, and reproduction, with effects potentially cascading to population-level impacts [25,26,27,28].
Early microplastic research revealed that MNP-induced mortality in marine organisms primarily resulted from physical damage. Specifically, excessive ingestion of MNPs caused blockages in the esophagus, stomach, and intestines, leading to death. This mortality was not attributed to the chemical properties of MNPs [29,30,31,32]. However, advances in understanding have revealed additional toxicity mechanisms beyond physical obstruction. Microplastics (MPs) and nanoplastics (NPs) can accumulate in fish gills and other internal tissues [33]. Due to their small size, they can also cross the gastrointestinal barrier and ultimately enter fish brains via the blood–brain barrier [34,35]. Furthermore, recent studies have found that MNPs easily interact with organic pollutants, heavy metals, pharmaceuticals, and other substances due to their large specific surface area and strong lipophilicity, acting as carriers that enhanced their toxic effects on marine organisms [28].
Microplastics demonstrate multigenerational effects in fish, though true transgenerational (F3+) data remain scarce. Limited data indicate that when parental zebrafish are exposed to nanoplastics, their offspring embryos show reduced hatching rates and increased malformation rates. This effect can persist into the F2 generation. Notably, microplastics often enhance the toxicity of other pollutants through carrier effects. For example, one study found that zebrafish offspring exhibited transgenerational lipid metabolism disorders after parental combined exposure to bisphenol A (BPA) and NPs [36], suggesting that microplastics may act as vectors amplifying co-contaminant toxicity across generations. However, the mechanisms underlying this enhancement remain unclear, as studies have not determined whether microplastics increase bioavailability, alter metabolic pathways, or act through independent additive toxicity.
Furthermore, studies have found that microplastics at environmental concentrations can affect the growth, lipid storage, and external coloration of adult fish. At low-concentrations (100 particles·L−1), environmentally derived microplastics exert further endocrine-disrupting effects on parental fish and their offspring, resulting in delayed onset of spawning, reduced egg survival rate, and a higher malformation rate in the offspring [37].
Methylation changes provide potential mechanisms for these multigenerational effects. Wade et al. reported a microplastic exposure experiment on two generations of fathead minnows. The results showed that methylation was more significant in parental males than in parental females and F1 offspring. This sex-specific difference is unexplained and warrants investigation. Meanwhile, in an experiment where only the maternal generation was exposed (and offspring unexposed), researchers found that changes in Differentially Methylated Loci (DMLs) and Differentially Methylated Regions (DMRs) persisted in offspring. This led to cross-generational effects that interfered with the offspring’s gene transcription and molecular processes [38]. However, the study examined only F1 offspring, so whether these methylation changes represent true transgenerational inheritance (persisting to F3+) or transient F1 effects remains unknown.
Environmental context critically modulates microplastic toxicity, as demonstrated in marine medaka multigenerational studies. Microplastics can also reduce the growth performance of F1-generation marine medaka. Under high salinity conditions, MPs increase the MDA content and decrease the antioxidant capacity of F2 larvae. Cortisol level of F1-generation larvae decreases while that of F2-generation larvae increases. In contrast, under low salinity conditions, both the MDA content and antioxidant capacity of F1 and F2 larvae increase. The mRNA expression levels of genes in the NF-E2-related factor 2 (Nrf2) pathway are dysregulated. This phenomenon is associated with the transcriptional regulation of genes in the hypothalamic-pituitary-interrenal (HPI) axis [39]. These opposing patterns suggest that environmental context critically modulates microplastic effects, but the lack of mechanistic investigation prevents determining whether salinity alters microplastic bioavailability, particle aggregation, or physiological stress responses.
The field faces critical data gaps: (1) few of studies extend beyond F2, preventing confirmation of true transgenerational inheritance; (2) mechanistic endpoints (specific gene targets, pathway analysis) are rarely measured alongside phenotypic effects, limiting causal interpretation; (3) dose–response relationships are poorly characterized, with most studies testing single concentrations that may not reflect environmental exposure.

2.2. Pharmaceutical and Personal Care Products (PPCPs)

While microplastics represent physical pollutants with associated chemical toxicity, pharmaceuticals and personal care products constitute a distinct class of emerging contaminants designed for biological activity, raising different concerns for multigenerational fish toxicity. PPCPs represent a diverse group of chemicals widely applied in human healthcare and personal care, including antibiotics, analgesics, hormones, and preservatives. Global PPCP usage has increased annually, driven by population growth and improved living standards worldwide. A significant fraction of incompletely degraded PPCPs enters aquatic ecosystems through pathways such as domestic sewage, hospital effluents, and livestock breeding wastewater [40,41]. PPCPs have been detected in surface waters globally at concentrations ranging from nanograms per liter (ng·L−1) to micrograms per liter (μg·L−1) [42,43]. Their environmental persistence varies based on chemical structure, aqueous solubility, biodegradability, and ambient conditions. Regional differences in public health policies further contribute to variations in PPCP composition and concentrations [41,44]. Traditional wastewater treatment systems cannot achieve complete PPCP removal, resulting in their progressive accumulation in water columns, sediments, and aquatic biota [45,46,47].
PPCPs can disrupt fish endocrine systems, impairing key physiological processes including growth, development, reproduction, and behavior [48,49]. Endocrine disrupting compounds in PPCPs induce specific adverse effects such as sex reversal, reproductive organ malformations, and reduced fecundity in fish [50]. Antibiotic-containing PPCPs interfere with fish immune function and associated microbial communities, increasing susceptibility to pathogens while promoting the emergence and dissemination of antibiotic resistance genes [51,52]. Certain PPCPs also cause structural and functional damage to critical organs including the nervous system, cardiovascular system, and liver, compromising overall physiological homeostasis in fish [53].
Recent years have witnessed growing scholarly attention to long-term and multi-generational fish exposure studies on PPCPs [54,55]. Existing research confirms that PPCPs exposure induces multi-generational toxic effects in offspring, though mechanistic understanding remains limited. For instance, parental zebrafish exposed to low concentrations of carbamazepine produce offspring with significant alterations in growth, development, and behavioral effects that persist across generations [55,56]. Similarly, parental zebrafish exposed to antibiotic mixtures (15 commonly detected compounds) at nominal concentrations of 1 and 100 μg·L−1 exhibit transgenerational transfer of these antibiotics, resulting in cardiotoxicity in offspring [57]. However, these studies do not specify whether effects extend to F3 generation, leaving uncertain whether observed impacts represent true transgenerational inheritance or transient multigenerational responses to direct exposure during germ cell development.
Notably, current research on PPCP-induced trans-/multi-generational toxicity remains limited, with most studies focusing on single-compound exposures. The toxic effects and underlying mechanisms of multi-compound PPCP exposures in complex environmental settings require further systematic investigation, including F3+ generation assessment to confirm true transgenerational inheritance, mechanistic endpoints (methylation profiling, histone modifications, miRNA expression) paired with phenotypic measurements, environmentally realistic mixture compositions and concentrations, comparative studies across fish species to identify susceptibility determinants and recovery experiments to determine whether effects are reversible or permanent.

2.3. Endocrine Disrupting Chemicals (EDCs)

The health hazards of EDCs to organisms have been widely recognized. Meanwhile, EDCs have been detected in multiple environmental media, including water, soil, aerosols, the atmosphere, and living organisms [58]. Due to their high hazards (such as potent toxicity, teratogenicity, carcinogenicity, mutagenicity, etc.), high environmental persistence, and bioaccumulation, EDCs are regarded as a typical type of ECs [59]. In recent years, studies on the effects of EDCs on trans-/multi-generational fish have been increasing [60]. Therefore, understanding the trans-/multi-generational toxic effects and mechanisms of EDCs is of great significance for a comprehensively assessing the ecological risks of endocrine disruption.
EDCs can cause disorders in the reproductive, developmental, and endocrine systems of fish [61,62]. In trans-/multi-generational studies, it was found that when female viviparous eelpout (Zoarces viviparus) were exposed to octylphenol (OP) at concentrations of 25 and 100 μg·L−1 for 17 days or 35 days, the expression levels of estrogen receptors and vitellogenin in F1-generation embryos increased significantly. Meanwhile, intersex phenomena occurred during the process of gonadal differentiation [63]. Similar studies have also found that nonylphenol (NP) at concentrations of 20 and 200 μg·L−1 can inhibited the growth of zebrafish and caused gonadal damage [6]. Meanwhile, gnrh2 and gnrh3 were upregulated, and fshβ and lhβ were downregulated in the hypothalamic-pituitary-gonadal axis (HPG). For female zebrafish, the expression of fshβ and lhβ was upregulated in the parental (P) generation and F1 generation but showed a downward trend in the F2 generation. In male zebrafish, the expression of the cyp19a1a gene in the gonads was downregulated, while the expression of fshr, lhr, and esr genes in females showed a decreasing trend. Compared with the P generation, the F2 generation exhibited stronger tolerance to high concentrations of NP (20 and 200 μg·L−1), but higher sensitivity to low-concentration NP (2 μg·L−1) [6].
EDCs can cause malformations and induce the development of cancer in trans-/multi-generational fish. In inland silverside (Menidia beryllina), early-life exposure to 3–10 ng·L−1 EDCs (bifenthrin, levonorgestrel, ethinylestradiol, trenbolone) caused significant trans-/multi-generational (F0–F2) DNA methylation differences (via reduced representation bisulfite sequencing, RRBS) in promoters/genes, enriched key pathways, inherited EDC-responsive genes (EDCRG) methylation, showing low EDCs alter epigenetics across unexposed generations [64]. These results illustrate the potential reasons why fish may develop teratogenicity and carcinogenicity upon exposure to EDCs. This represents the most mechanistically rigorous EDC multigenerational study to date, directly linking exposure to heritable epigenetic marks. However, even this study examined only F0–F2 generations, leaving uncertain whether methylation patterns persist to F3 (true transgenerational inheritance) or dissipate after removal of direct germ cell exposure.
Major knowledge gaps persist: (1) No EDC studies extend to F3+ to confirm transgenerational versus multigenerational effects; (2) the mechanistic basis for generation-specific gene expression reversals (upregulation in F1, downregulation in F2) remains unexplored; (3) non-monotonic dose–response patterns across generations are documented but unexplained, requiring investigation of whether different concentration ranges trigger distinct molecular pathways; (4) sex-specific differences in multigenerational susceptibility are observed but not mechanistically characterized—do males and females show differential epigenetic reprogramming, EDC metabolism, or germline vulnerability?

2.4. Mixture Effects

Real-world aquatic organisms face simultaneous exposure to complex contaminant mixtures, yet multigenerational toxicity research has focused almost exclusively on single-compound exposures (Table 1). The limited mixture studies reveal critical knowledge gaps regarding synergistic and antagonistic interactions across generations.
Evidence for mixture enhancement of multigenerational effects is emerging but mechanistically unexplored. Nanoplastics combined with BPA produced F1–F2 lipid metabolism disorders in zebrafish [40], suggesting microplastics may act as vectors amplifying co-contaminant transgenerational toxicity. Similarly, environmental microplastics (100 particles·L−1) enhanced endocrine-disrupting effects in exposed parents and offspring beyond what would be expected from microplastics alone [65]. The antibiotic mixture study [61] documented direct transgenerational transfer of 15 pharmaceutical compounds from exposed parents to embryos, causing F1 cardiotoxicity. In inland silverside, early-life exposure to an EDC mixture (bifenthrin, levonorgestrel, ethinylestradiol, trenbolone at 3–10 ng·L−1) produced DNA methylation changes persisting through F0–F2 generations [64].
However, neither study formally tested whether effects were synergistic (greater than additive), antagonistic (less than additive), or simply additive, limiting interpretation of interaction mechanisms. Most of studies examined only two chemical classes simultaneously, far fewer than the dozens of contaminants co-occurring in real environments. Whether low-dose environmental mixtures produce different multigenerational toxicity patterns than the high-dose single compounds dominating current research remains unknown. Most critically, no studies extend beyond F2 generation, leaving uncertain whether mixture exposures cause true transgenerational (F3+) effects or only multigenerational responses. The near-absence of mixture multigenerational data represents a critical gap given that wastewater effluents contain 50+ pharmaceutical compounds, urban runoff combines pesticides and microplastics, and agricultural areas expose fish to complex pesticide cocktails.
Table 1. The endpoints and effects of ECs on fish across trans-/multi-generational exposures.
Table 1. The endpoints and effects of ECs on fish across trans-/multi-generational exposures.
ECsFish SpeciesExposure DoseExposure DurationGenerationsEndpoint in OffspringReference
Single ECs on trans-/multi-generational fish
Microplasticsfathead minnows
(Pimephales promelas)
100, 2000 particles·L−1 (150–500 μm)P: 178 dpf
F1: 12 dpf
P, F1P: Affect the growth, lipid storage, and external coloration.
F1: In the low-concentration treatment (100 particles·L−1), microplastics can delayed onset of spawning, reduced egg survival rate, and a higher malformation rate in the offspring.
[37]
fathead minnows
(Pimephales promelas)
100, 2000 particles·L−1 (150–500 μm)P: 178 dpf
F1: 12 dpf
P, F1F1: Methylation was more significant in parental males than in parental females and F1 offspring.[38]
CarbamazepineZebrafish
(Danio rerio)
10 μg/LP: 6 weeksP, F1, F2, F3, F4P: Reproductive output, courtship and aggressive behaviors, 11-ketotestosterone (11 KT), and sperm morphology decreased.
F1-F4 (only paternal fish exposed): 11 KT, reproductive output, altered courtship, aggression, and sperm morphology decreased.
[55]
OPviviparous eelpout (Zoarces viviparus) 25, 100 μg·L−1P: 17 and 35 dpfP, F1F1: the mortality of embryos increased, and the body length and weight decreased; the expression levels of estrogen receptors and vitellogenin of F1 embryos were increased; abnormal gonadal differentiation occurred.[63]
PFOSZebrafish
(Danio rerio)
5, 50, 250 μg·L−1P: 150 dpf
F1: 7 dpf
P, F1F1: The mortality rate of F1 larvae was 100% at 250 μg·L−1; the locomotor activity of F1 larvae were abnormally enhanced in 5 μg·L−1.[66]
BPArare minnow (Gobiocypris rarus)15, 225 μg·L−1P: 21 dpfP, F1P: induced poor quality of the embryos.
F1: The fertilization, hatching, and spontaneous coiling rates decreased; the malformation rate increased; ossification in craniofacial cartilage were delayed; embryonic heart rate increased.
[60]
NPZebrafish
(Danio rerio)
2, 20, 200 μg·L−1P: 32 dpf
F1: 120 dpf
F2: 140 dpf
F3: 7 dpf (recovery)
P, F1, F2, F3P: the expression of fshβ and lhβ were upregulated;
F1: inhibit the growth of zebrafish and cause gonadal damage. the expression of fshβ and lhβ were upregulated. fshr, lhr, and esr were decreased.
F2: inhibit the growth of zebrafish and cause gonadal damage. exhibits stronger tolerance to high concentrations of NP (20 and 200 μg·L−1), but higher sensitivity to low-concentration NP (2 μg·L−1)
F3: Fertility ratio and hatching ratio were decreased.
[6]
Combined ECs on trans-/multi-generational fish
Microplastics and EE2fathead minnows
(Pimephales promelas)
MPs, MP + EE2 (10 ng·L−1),
MP + EE2 (50 ng·L−1)
P: 44 dpf
F1: 14 or 21 dpf
P, F1F1: reduced embryos locomotor activity; stronger swimming ability.[65]
Microplastics
and salinity
marine medaka (Oryzias melastigma)180 μg·L−1 polystyrene
( 5.0 μm), 5‰ and 25‰ salinity
F1: 25 dph
F2: 120 dph (recovery)
F0 (unexposed), F1, F2 (recovery)F1: high salinity treatment increases in MDA content, TAC, CAT activity and cortisol levels. Additionally, also upregulated the mRNA levels of antioxidant genes (nrf2, keap1a, keap1b, sod1, cat, mt2) and HPI axis genes (crhr1, crhr2, pomca, mc2r, cyp11c1). Reduced the growth performance.[39]
Nanoplastics and bisphenolZebrafish
(Danio rerio)
BPA 20 μg·L−1,
NPs 1 mg·L−1,
NPs 1 mg·L−1 + BPA 20 μg·L−1 (N + B)
F0: 28 dpf
F1: 4 months (recovery)
F0, F1F0: N + B groups induced the gut damage and microbiota dysbiosis; Differentially expressed metabolites were mainly enriched in purine metabolism, mTOR, FoxO pathways, and all enriched in mTOR in control/NPs/N + B and control/BPA/N + B groups. The N + B group had the most such metabolites; nanoparticle + BPA co-exposure exacerbated F0 zebrafish liver metabolic disturbance, with mTOR possibly the mechanism.
F1: Dead egg rate was increased at NPs, and N + B groups; the TC and TG level increased at N + B groups.
[36]
Bifenthrin,
Levonorgestrel, ethinylestradiolt, renbolone
inland silverside
(Menidia beryllina)
bifenthrin (3.02 ng·L−1),
EE2 (6.79 ng·L−1), levonorgestrel ( 9.27 ng·L−1),
trenbolone ( 9.60 ng·L−1).
F0: 8–21 dph
F1: unexposed
F2: unexposed
F0, F1, F2F0, F1, F2: Through RRBS, which were attributed to strict inheritance of DNA methylation alterations and dysregulation of epigenetic control mechanisms. differential methylation was more frequent in gene bodies than in promoter regions across all treatments and generations[64]
AntibioticCAM, CIP, CTC, EYR, ENR, LCM, NOR, OFL, OTC, ROX, SDZ, SMX, SMZ, TC, trimethoprim0, 1, and 100 μg·L−1 F0: 150 dpf
F1: eggs
F0, F1F1: All antibiotics were detected in F1 eggs in 1 and 100 μg·L−1 treatment. Cardiotoxicity, and the apoptosis of cardiac cells. let-7a-5p regulated the cardiac hypertrophy signaling pathway.[57]

2.5. Population-Level Implications of Multigenerational Effects

Individual-level multi-/trans-generational effects have potential population consequences, though empirical population-level data remain scarce. Reduced F2 fecundity and altered sex ratios observed in multiple studies could theoretically decrease population growth rates if exposure is sustained and widespread. For example, ethinylestradiol exposure causing skewed sex ratios across generations could reduce effective breeding population size, while decreased hatching rates in F2 offspring would lower recruitment [67]. However, whether these individual-level effects translate to population decline in wild settings remains uncertain for several reasons:
First, laboratory studies use genetically uniform populations lacking the genetic diversity that might buffer wild populations against transgenerational effects through varied susceptibility or adaptive capacity. Second, density-dependent compensation mechanisms may offset reduced fecundity, if fewer offspring survive to reproduction, remaining individuals may experience reduced competition and higher per-capita reproductive success. Third, recovery potential after exposure cessation is unknown, if epigenetic changes are reversible, populations might recover once contamination is removed. Fourth, most studies examine isolated single-species effects rather than community-level interactions where predators, competitors, and prey might also be affected.
Critical data gaps preventing population risk assessment include: (1) no field studies documenting population decline attributable to transgenerational contaminant effects; (2) lack of population modeling integrating multigenerational data with demographic parameters; (3) unknown threshold exposure durations or concentrations causing irreversible population effects versus recoverable impacts; (4) absence of data on whether natural selection can lead to rapid adaptation, potentially mitigating population-level consequences.
Among the studies reviewed above, the majority examine multigenerational effects in F1-F2 generations. True transgenerational effects (F3+) have been documented for only a limited number of compounds. For example, NP showed persistent effects in F3 zebrafish [6], while benzo[a]pyrene (BaP) demonstrated F3 impacts in marine medaka [8]. The scarcity of F3+ data reflects the practical challenges of conducting multi-year studies spanning three or more generations, representing a significant knowledge gap in understanding true transgenerational inheritance of emerging contaminant effects in fish.

3. Mechanisms of the Trans-/Multi-Generations Toxicity of ECs on Fishes

To date, most studies on aquatic organisms have focused on the effects of a single toxic compound on one or more generations and their offspring. These results may have cognitive deficiencies and inadequacies in understanding the toxic effects and mechanisms of endocrine-disrupting chemicals (EDCs) on aquatic organisms [68]. Based on the current research findings, the following three types of toxicological mechanisms have been summarized (Figure 3).

3.1. Trans-Generational Transmission, Bioaccumulation and Oxidative Damage

ECs can accumulate in fish across generations to induce toxic effects, a common toxicological mechanism. ECs have been detected in various fish species across different trophic levels [69], and this long-term accumulation may reach teratogenic and lethal levels. When ECs enter organisms for metabolism, the parent compounds are often the most toxic throughout the biological metabolic process, also referred to as the ultimate toxicant [70]. As the concentration of ECs in fish increases across generations, more reactive oxygen species (ROS) are generated [71]. ROS can disrupt intracellular redox balance, causing oxidative damage to lipids, proteins, and nucleic acids. Organisms initially respond to ROS accumulation through compensatory upregulation of antioxidant defense enzymes (SOD, CAT, GPx, GR). Zhou et al. demonstrated that F0 zebrafish exposed to low concentrations (10 μg·L−1 PS-NPs) successfully mounted adaptive responses with significantly increased SOD and CAT activities [72]. However, this compensatory capacity is concentration- and time-dependent. Li et al. documented a biphasic pattern in rainbow trout where antioxidant enzymes initially increased at 7 days but became exhausted and decreased below control levels at 21–42 days under prolonged carbamazepine exposure, accompanied by elevated oxidative damage [73]. Most critically, this compensatory failure is inherited across generations. Pitt et al. showed that F1 zebrafish offspring, despite never being directly exposed to nanoplastics, exhibited depleted glutathione reductase and could not mount the GPx upregulation observed in their parents, indicating inherited impairment of compensatory mechanisms [74]. Zhang et al. further demonstrated progressive deterioration in F2 marine medaka, where oxidative stress worsened compared to F1, with increased MDA and decreased antioxidant capacity under high salinity [39]. When compensatory antioxidant systems fail or become exhausted in this manner, unopposed ROS accumulation proceeds to activate downstream damage pathways.
Excessive ROS can activate the nuclear factor κB (NF-κB) signaling pathway in acute and chronic exposures. NF-κB is a key regulator of immune and inflammatory responses. Once activated, it migrates into the nucleus and upregulates the expression of a series of pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) [75]. Continuous activation of this pathway, induced by ECs-generated ROS, may lead to chronic inflammation in fish. Long-term inflammation can disrupt normal cellular functions, trigger cell death, and ultimately result in fish mortality or the emergence of teratogenic phenotypes [76,77]. While ROS can activate NF-κB signaling in directly exposed organisms [75,76,77], whether this pathway contributes to transgenerational effects in F2/F3 offspring remains undemonstrated, as multigenerational studies have not measured NF-κB activation, inflammatory cytokines, or downstream inflammation markers across unexposed generations. The persistent transgenerational phenotypes observed may result from inherited compensatory antioxidant failure, epigenetic inheritance of oxidative damage, continued inflammation signaling, or a combination of these mechanisms.

3.2. Endocrine Interference and Signaling Pathways Disorder

After entering the bloodstream, ECs migrate to target organs or tissues. During this process, they transform into multiple intermediate contaminants. These contaminants, including PPCPs and pesticides, may disrupt fish endocrine systems. For example, some PPCPs, such as bisphenol A (BPA) and 17α-ethinylestradiol (EE2), can mimic or antagonize natural hormones [78,79]. BPA binds to estrogen receptors in fish, disrupting the normal regulation of the hypothalamic-pituitary-gonadal (HPG) axis [80]. This interference may cause abnormal sexual development, such as feminization in male fish. Manifestations include reduced sperm production, abnormal gonadal morphology, and altered secondary sexual characteristics [81]. Additionally, exposure to per- and polyfluoroalkyl substances (PFAS) can imbalance estrogen receptor (ER) and androgen receptor (AR) expression in adult zebrafish and reduce sperm quality [82].
Beyond endocrine disruption, ECs may interfere with fish metabolic pathways. For instance, triclosan inhibits fatty acid synthase (FAS) activity, leading to hepatic fat accumulation in zebrafish. This likely occurs via interactions with short-chain fatty acids and miR-101a [83,84]. Pesticides can inhibit key enzymes in fish metabolism. Some organophosphorus pesticides target acetylcholinesterase, an enzyme critical for neural function and involved in certain metabolic regulatory processes [85]. Disrupted metabolic pathways cause energy imbalance and abnormal nutrient metabolism. These effects ultimately impact fish growth, development, and survival. In severe cases, they may result in lethal or teratogenic outcomes, such as growth retardation, developmental malformations, and increased mortality [86].
Studies show that different EC types target specific tissues and organs. For example, persistent organic pollutants (POPs) and EDCs primarily act by interfering with fish endocrine systems. They bind to hormone receptors, disrupt normal hormonal signal transduction, and thus affect various fish physiological functions [87,88,89]. Antibiotics may alter the normal fish microbial community, disrupt physiological balance, and induce oxidative stress through certain metabolic pathways [90]. When organisms are exposed to ECs over multiple generations or transgenerationally, these tissues and organs sustain continuous damage. Particularly during fetal, neonatal, and embryonic stages, when detoxification mechanisms are underdeveloped, ECs more easily affect offspring via germ cells, the placental barrier, etc. This increases multi-generational or transgenerational toxicity. Such long-term accumulation may reach teratogenic and lethal levels.

3.3. The Transgenerational Inheritance of Epigenetic Modifications

Emerging contaminants can establish heritable epigenetic changes that persist across unexposed offspring generations, representing a critical mechanism of transgenerational toxicity. Understanding which epigenetic modifications are causal versus correlative, when contaminant exposure establishes inheritable marks, and why some effects persist to F3 is essential for risk assessment.

3.3.1. Epigenetic Modifications

Not all contaminant-induced epigenetic changes transmit transgenerationally or directly cause phenotypes. Three contaminant studies illustrate this distinction. Promoter methylation shows strongest causal links. PFOA exposure in rainbow trout induced promoter methylation of lipid metabolism genes (fabp, pparα), directly reducing transcript levels and disrupting lipid homeostasis in both exposed fish and their offspring [91]. Cadmium exposure increased promoter methylation of detoxification genes in zebrafish embryos, causally silencing transcription [92,93,94]. This establishes the causal pathway that contaminant exposure induces promoter methylation, leading to transcriptional silencing, thus exhibiting phenotypic outcome. Distal DMRs may be correlative. MEHP phthalate exposure (30 µM, 0–6 dpf) in zebrafish caused 410 DMRs, with 44% overlapping conserved non-genic elements. However, locus-specific validation across 103 CpG sites at 10 loci revealed only 20% showed consistent F2 transmission [95], indicating genome-wide discovery overestimates true transgenerational inheritance by ~5-fold. Many distal DMRs represent bystander marks without direct functional consequences. Absence of methylation-phenotype correlation indicates alternative mechanisms. Benzo[a]pyrene exposure caused skeletal malformations persisting to F3, yet no DNA methylation correlation was detected [96]. This suggests histone modifications or ncRNAs may drive transgenerational effects for some contaminants independently of DNA methylation, which is a critical consideration when interpreting negative methylation results.

3.3.2. Critical Developmental Windows

Contaminant exposure timing determines whether effects transmit transgenerationally. Gametogenesis exposure establishes most stable marks. Adult marine medaka exposed to hypoxia (1.4 mg/L dissolved oxygen) during spermatogenesis established sperm methylome changes with 409 differentially methylated gene promoters transmitted to unexposed F2 offspring [97]. Marks incorporated during active gametogenesis bypass embryonic reprogramming, explaining why adult exposures can cause F2 effects [98,99]. Early embryonic exposure during sex determination creates delayed-onset effects. BPA/EE2 exposure during medaka days 0–7 (encompassing germ cell differentiation at days 5–7) showed no apparent F0 or F1 abnormalities, yet F2 exhibited reduced fertilization and F3 showed decreased embryo survival [54]. The mechanism underlying is that contaminants disrupting epigenetic programming during germ cell specification establish marks that manifest only when those cells complete gametogenesis in subsequent generations, explaining why F0/F1 can appear normal while F2/F3 show effects. Species-specific reprogramming influences contaminant sensitivity. Zebrafish maintain 80–85% methylation throughout PGC development with no global erasure [98], making them highly permissive for transgenerational transmission. Medaka shows bi-phasic demethylation yet retains 25% of non-promoter CpG islands hypermethylated [99]. Contaminant exposures during low-methylation windows (15 dpf nadir in medaka) may establish more persistent marks than exposures outside these windows.

3.3.3. Methodological Approaches

Genome-wide methylation profiling identifies contaminant-induced DMRs but requires validation. WGBS provides single-base resolution but assesses only ~50% of fish genomes [100]. The methylmercury study used MeDIP-Seq on F2 sperm, revealing increased epimutations compared to F0 and generation-specific DMR patterns [101]. RRBS enables population-scale studies with lower DNA input, the endocrine disruptor study used RRBS to identify 6–11% of genes differentially methylated in F0-F2 silverside following bifenthrin, EE2, and trenbolone exposure [64]. Teleost genome duplications complicate alignment (54–75% versus >85% in mammals), and zebrafish RRBS shows only 1.7-fold CpG enrichment versus 3–4-fold in mammals [100]. Locus-specific validation is essential. The MEHP phthalate study demonstrated that exposed zebrafish exhibited 410 genome-wide DMRs, but only 20% validated at F2 [95]. Bisulfite PCR amplicon sequencing of specific candidate loci provides higher confidence than genome-wide discovery alone. Integration with transcriptomics and functional validation establishes causation. The hypoxia study integrated MeDIP-Seq, RNA-seq, and proteomics, revealing that EHMT2 promoter hypomethylation lead to an increased expression, followed by elevating H3K9me2, thus resulting in silencing of reproductive genes across F0-F2 [97]. The dnmt1 mutant study provided strongest causal proof, where knockdown of hypermethylated genes (runx3, rptor) recapitulated transmitted phenotypes [102]. Fish-specific methodological limitations include: (1) F2/F3 larvae tissue amounts often insufficient for WGBS (1–5 µg DNA required); (2) ChIP-seq antibody cross-reactivity issues; (3) lack of comprehensive histone modification data across F0-F3 in any contaminant study. No microplastic, pharmaceutical, or EDC studies have measured complete histone modification profiles transgenerationally.

3.3.4. Transmit Contaminant-Induced Marks

Fish exhibit fundamentally permissive transgenerational epigenetic inheritance due to absent germline erasure. Unlike mammals undergoing massive PGC demethylation (71% to 7–14%), zebrafish PGCs maintain 84% methylation throughout specification [98]. This creates vulnerability that contaminant-induced methylation changes face no global erasure checkpoint and can persist across generations. Paternal template dominance explains male-biased transmission in many contaminant studies. The maternal genome reprograms to match paternal methylation patterns during early cleavage, establishing sperm methylome as the embryonic template [98]. Contaminant exposures affecting male germline therefore have high transgenerational transmission probability. Limited active demethylation creates default methylation maintenance.

3.3.5. Mechanisms Enabling Contaminant Effects to Persist Through F3

The F3 generation represents the first truly unexposed generation (F1 was exposed as embryos, F2 as germline within F1). Four mechanisms enable contaminant-induced marks to persist through F3. First, imprint-like resistance to demethylation. Contaminant-induced repressive histone marks (H3K9me2, H3K27me3) [97] shield CpG sites from TET enzyme access, creating local environments resistant to demethylation. The marine medaka salinity/microplastics study showed F2 oxidative stress worsened compared to F1 [39], suggesting stable epigenetic inheritance rather than gradual erasure. Second, genetic-epigenetic coupling. Cadmium exposure selected for SNP-methylation coupling in cep19: G allele frequency increased from 0.17 to 0.34, with G/G genotype showing 83.48% methylation versus 7.08% in A/A [94]. Once established, genetic variants maintain methylation patterns across generations independently of continued contaminant exposure. Third, epigenetic hub amplification. Contaminant-induced methylation of chromatin-modifying enzymes (e.g., EHMT2 in hypoxia study) drives genome-wide histone changes affecting hundreds of downstream targets [97]. Reversing hub effects requires coordinated demethylation across multiple loci, which is a substantial barrier. Fourth, environmental stress impairs erasure. Contaminant co-exposure with environmental stress (salinity, temperature, hypoxia) suppresses TET expression [103] or enhances DNMT activity [102], stabilizing induced marks. The marine medaka microplastics study showed salinity-dependent F2 effects [72], demonstrating environmental context modulates epigenetic persistence.

4. Prospects and Challenges of ML and AI for Toxicity of ECs to Multi-Generational Fish

The transgenerational effects of emerging contaminants reviewed above, from microplastic-induced reproductive dysfunction to EDC-mediated epigenetic inheritance in fish, present a fundamental challenge: how can we predict and mechanistically understand such effects for the thousands of untested chemicals entering aquatic ecosystems? Machine learning and AI technologies offer a complementary approach, with demonstrated success in predicting toxicity endpoints and elucidating mechanisms [104,105,106,107,108]. While traditional trans-/multi-generational fish toxicity research depends on multi-year in vivo experiments observing mortality, growth inhibition, and reproductive impairment across generations [6,109,110], ML approaches can potentially predict F2/F3 outcomes from F0 data, identify mechanistic biomarkers, and reveal structure-toxicity relationships.
Machine learning approaches have demonstrated substantial success in predicting various toxicity endpoints in fish [106,107,111], evolving from simple structure-activity relationships to sophisticated deep learning architectures [112]. Gustavsson et al. developed transformer-based deep learning models that significantly advanced chemical toxicity prediction in aquatic organisms, achieving high accuracy across fish, algae, and aquatic invertebrates while substantially outperforming traditional QSAR methods [113]. Similarly, Fan et al. successfully applied support vector machines to predict reproductive toxicity of endocrine disrupting chemicals in aquatic species, demonstrating that ML algorithms could capture complex relationships between chemical structure and reproductive endpoints [114].
While no published studies yet apply ML to fish transgenerational toxicity, mammalian research provides a computational roadmap. Fish transgenerational inheritance, like mammals, operates primarily through DNA methylation changes [105], suggesting mammalian ML approaches can transfer to aquatic systems. Liu et al. developed comprehensive machine learning models for rat multigenerational reproductive toxicity, analyzing 275 chemicals using seven different algorithms, achieving balanced accuracies of 58–65% [115]. Beyond reproductive endpoints, ML has proven capable of predicting the underlying epigenetic mechanisms. Haque et al. achieved 100% accuracy in predicting genome-wide locations of transgenerational epimutations using Active Learning and Imbalanced Class Learning [116], while Beck et al. employed deep learning to identify exposure-specific transgenerational DNA methylation patterns [117].
The absence of ML applications to transgenerational toxicity in fish stems from disciplinary isolation rather than technical limitations. The experimental data on microplastics, PPCPs, and EDCs generate the type of data ML requires, such as exposure conditions, epigenetic measurements, and multi-generational phenotypes (Table 1). However, these datasets remain scattered, incompletely characterized for ML purposes (missing chemical descriptors, inconsistent endpoints, limited metadata), and siloed within communities lacking computational expertise. The biological foundation for ML application is strong which fish transgenerational inheritance operates through DNA methylation mechanisms [98,99], the same pathway where mammalian ML models achieved 95–100% prediction accuracy [116].
Several challenges must be addressed using existing experimental data. Data scarcity presents the primary obstacle. Approximately 30–50 chemicals tested across zebrafish, medaka, and other species represent a small fraction of datasets typically used in successful ML applications [104,105,106]. Mechanistic complexity compounds the difficulty, as transgenerational effects manifest through multiple epigenetic pathways (as discussed in Section 3.3) and affect diverse endpoints across generations, requiring models integrating multiple biological levels. Species-specific variations in susceptibility add another layer, differential responses between zebrafish and medaka for similar exposures demonstrate that phylogenetic differences complicate broadly applicable models [36,37,38,39,47]. Finally, regulatory acceptance requires transparent, explainable AI approaches that can mechanistically justify predictions of effects in unexposed generations.
Despite these limitations, ML offers specific capabilities for mechanistic understanding. The diverse contaminants reviewed share no obvious structural similarities, yet some produce transgenerational effects while others do not. ML could identify cryptic structural features or physicochemical properties that predict transgenerational toxicity across chemical classes. Some chemicals cause immediate F0 effects that diminish by F2 [6,72], while others show minimal F0 impact but severe F2/F3 consequences [39,54]. ML could identify which molecular features predict transient versus persistent effects. By integrating methylation data with chemical structures and phenotypic outcomes, ML could predict which genomic regions are susceptible to heritable modifications, explaining mechanistically why certain contaminants cause transgenerational inheritance while structurally similar chemicals do not.

5. Concluding Remarks and Future Perspectives

Previous studies indicate that ECs induce multiple effects in trans-/multi-generational fish, including decreased fertilization and hatching rates, increased teratogenicity and carcinogenicity, abnormal development of tissues (e.g., bones and heart), neurotoxicity, and endocrine disruption. While these effects persist through several generations, evidence for population-level or ecosystem impacts remains hypothetical. The documented trans-/multi-generational effects suggest potential risks to population dynamics, though field validation and longer-term studies are needed to confirm ecological significance.
Based on critical knowledge gaps identified in this review, we propose the following prioritized research agenda. First, current studies mainly focus on short-term exposure to high pollutant doses, which far exceed environmentally relevant levels (ng·L−1 to low μg·L−1 range), limiting our understanding of biological changes under long-term stress. Therefore, future research should prioritize to conducted on the toxic effects and mechanisms of environmentally relevant concentrations on fish. Second, studies on ECs exposure to trans-/multi-generational fish, especially those spanning more than three generations, remain very limited. Most reviewed studies examine F1-F2 generations, which, while valuable for understanding multigenerational impacts, cannot definitively establish transgenerational epigenetic inheritance. Existing results show that the damage caused by ROS and endocrine disruption is limited to a single generation. Future research priorities should include expanding F3+ assessments to verify which emerging contaminants cause genuine transgenerational inheritance versus transient multigenerational effects. Third, Combined EC exposures in fish remain mechanistically uncharacterized, with no studies formally distinguishing synergistic from antagonistic interactions or extending mixture effects beyond F2 generations (Section 2.4). Finally, the number of ECs shows an annual increasing trend, and existing experimental toxicology cannot fully analyze the toxic effects and mechanisms of ECs on fish. Therefore, efforts can be initiated from the field of ML and AI to further strengthen investment in this area.

Author Contributions

Conceptualization, D.S. and M.D.; software, D.S.; validation, D.S. and M.D.; investigation, Y.H. and S.C.; resources, Y.H.; writing—original draft preparation, D.S.; writing—review and editing, M.D.; visualization, D.S.; supervision, D.S.; project administration, D.S.; funding acquisition, D.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Basic and Applied Basic Research Foundation of Guangdong Province (2022A1515010460) and the Youth Innovative Talents Project of Zhanjiang (2021E05025) and supported by the program for scientific research start-up funds of Guangdong Ocean University (060302122009), Guangdong Province College Students Innovation and Entrepreneurship Training Program Project Funding (S202210566070), and Funding for the Innovation and Entrepreneurship Training Program of Guangdong Ocean University (CXXL2022177).

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.

Abbreviations

The following abbreviations are used in this manuscript:
ANNsartificial neural networks
ARandrogen receptor
ARGsantibiotic resistance genes
BaPbenzo[a]pyrene
BPAbisphenol A
CAMclarithromycin
CATcatalase
Cdcadmium
CIPciprofloxacin
CTCchlortetracycline
DFdeep forest
DMLsdifferentially Methylated Loci
DMRsdifferentially methylated regions
ECsemerging contaminants
EDCRGEDC-responsive genes
EDCsendocrine-disrupting chemicals
EE217α-ethinylestradiol
ENRenrofloxacin
ERestrogen receptor
EYRerythromycin
FASfatty acid synthase
FoxOfactor forkhead box
GATgraph attention network
GPxglutathione peroxidase
GRglutathione reductase
HPGhypothalamic-pituitary-gonadal
HPIhypothalamic-pituitary-interrenal
IL-1βinterleukin-1β
IL-6interleukin-6
LCMlincomycin
MDAMalondialdehyde
MeHgmethylmercury
MEHPmono-(2-ethylhexyl) phthalate
MGEsmobile genetic elements
miRNAmicroRNA
MLMachine Learning
MNPsmicroplastics and nanoplastics
MPsmicroplastics
mRNAmessenger RNA
mTORmechanistic target of rapamycin
NF-κBnuclear factor κB
NORnorfloxacin
NPnonylphenol
NPsnanoplastics
Nrf2NF-E2-related factor 2
OFLofloxacin
OPoctylphenol
OTCoxytetracycline
PBMTpersistence, bioaccumulation, mobility, and toxicity
PFASpolyfluoroalkyl substances
PFOAperfluorooctanoic acid
PFOSperfluorooctane sulfonate
PMTpersistent, mobile, and toxic
POPsPersistent Organic Pollutants
PPCPspharmaceutical and personal care products
QSARquantitative structure-activity relationship
ROSreactive oxygen species
ROXroxithromycin
RRBSreduced representation bisulfite sequencing
SDZsulfadiazine
SMXsulfamethoxazole
SMZsulfamethazine
TCtetracycline
TLtransfer learning
TMPtrimethoprim
TNF-αtumor necrosis factor-α
vPvMvery persistent and very mobile

References

  1. Morin-Crini, N.; Lichtfouse, E.; Liu, G.; Balaram, V.; Ribeiro, A.R.L.; Lu, Z.; Stock, F.; Carmona, E.; Teixeira, M.R.; Picos-Corrales, L.A.; et al. Worldwide cases of water pollution by emerging contaminants: A review. Environ. Chem. Lett. 2022, 20, 2311–2338. [Google Scholar] [CrossRef]
  2. Puri, M.; Gandhi, K.; Kumar, M.S. Emerging environmental contaminants: A global perspective on policies and regulations. J. Environ. Manag. 2023, 332, 117344. [Google Scholar] [CrossRef]
  3. Liu, J.L.; Wong, M.H. Pharmaceuticals and personal care products (PPCPs): A review on environmental contamination in China. Environ. Int. 2013, 59, 208–224. [Google Scholar] [CrossRef] [PubMed]
  4. Rissman, E.F.; Adli, M. Minireview: Transgenerational epigenetic inheritance: Focus on endocrine disrupting compounds. Endocrinology 2014, 155, 2770–2780. [Google Scholar] [CrossRef] [PubMed]
  5. Li, X.; Shen, X.; Jiang, W.; Xi, Y.; Li, S. Comprehensive review of emerging contaminants: Detection technologies, environmental impact, and management strategies. Ecotoxicol. Environ. Saf. 2024, 278, 116420. [Google Scholar] [CrossRef] [PubMed]
  6. Sun, D.; Chen, Q.; Zhu, B.; Zhao, H.; Duan, S. Multigenerational reproduction and developmental toxicity, and HPG axis gene expression study on environmentally-relevant concentrations of nonylphenol in zebrafish. Sci. Total Environ. 2021, 764, 144259. [Google Scholar] [CrossRef]
  7. Junaid, M.; Siddiqui, J.A.; Liu, S.; Lan, R.; Abbas, Z.; Chen, G.; Wang, J. Adverse multigeneration combined impacts of micro(nano)plastics and emerging pollutants in the aquatic environment. Sci. Total Environ. 2023, 882, 163679. [Google Scholar] [CrossRef]
  8. Sun, D.; Chen, Q.; Zhu, B.; Lan, Y.; Duan, S. Long-Term Exposure to Benzo[a]Pyrene Affects Sexual Differentiation and Embryos Toxicity in Three Generations of Marine Medaka (Oryzias Melastigma). Int. J. Environ. Res. Public Health 2020, 17, 970. [Google Scholar] [CrossRef]
  9. Chakraborty, S.; Dissanayake, M.; Godwin, J.; Wang, X.; Bhandari, R.K. Ancestral BPA exposure caused defects in the liver of medaka for four generations. Sci. Total Environ. 2023, 856, 159067. [Google Scholar] [CrossRef]
  10. Chakraborty, S.; Anand, S.; Coe, S.; Reh, B.; Bhandari, R.K. The PCOS-NAFLD Multidisease Phenotype Occurred in Medaka Fish Four Generations after the Removal of Bisphenol A Exposure. Environ. Sci. Technol. 2023, 57, 12602–12619. [Google Scholar] [CrossRef]
  11. Deviller, G.; Lundy, L.; Fatta-Kassinos, D. Recommendations to derive quality standards for chemical pollutants in reclaimed water intended for reuse in agricultural irrigation. Chemosphere 2020, 240, 124911. [Google Scholar] [CrossRef]
  12. Richardson, S.D.; Manasfi, T. Water Analysis: Emerging Contaminants and Current Issues. Anal. Chem. 2024, 96, 8184–8219. [Google Scholar] [CrossRef] [PubMed]
  13. Kappenberg, F.; Duda, J.C.; Schurmeyer, L.; Gul, O.; Brecklinghaus, T.; Hengstler, J.G.; Schorning, K.; Rahnenfuhrer, J. Guidance for statistical design and analysis of toxicological dose-response experiments, based on a comprehensive literature review. Arch. Toxicol. 2023, 97, 2741–2761. [Google Scholar] [CrossRef] [PubMed]
  14. Kaur, J.; Khatri, M.; Puri, S. Toxicological evaluation of metal oxide nanoparticles and mixed exposures at low doses using zebra fish and THP1 cell line. Environ. Toxicol. 2019, 34, 375–387. [Google Scholar] [CrossRef]
  15. Ribeiro, M.J.; Scott-Fordsmand, J.J.; Amorim, M.J.B. Multigenerational exposure to cobalt (CoCl(2)) and WCCo nanoparticles in Enchytraeus crypticus. Nanotoxicology 2019, 13, 751–760. [Google Scholar] [CrossRef]
  16. Carvalho, R.N.; Arukwe, A.; Ait-Aissa, S.; Bado-Nilles, A.; Balzamo, S.; Baun, A.; Belkin, S.; Blaha, L.; Brion, F.; Conti, D.; et al. Mixtures of chemical pollutants at European legislation safety concentrations: How safe are they? Toxicol. Sci. 2014, 141, 218–233. [Google Scholar] [CrossRef]
  17. Li, H.; Zeng, L.; Wang, C.; Shi, C.; Li, Y.; Peng, Y.; Chen, H.; Zhang, J.; Cheng, B.; Chen, C.; et al. Review of the toxicity and potential molecular mechanisms of parental or successive exposure to environmental pollutants in the model organism Caenorhabditis elegans. Environ. Pollut. 2022, 311, 119927. [Google Scholar] [CrossRef]
  18. Carvalho, L.; Oya-Silva, L.F.; Perussolo, M.C.; Oliveira Guaita, G.; Moreira Brito, J.C.; Evans, A.A.; Prodocimo, M.M.; Cestari, M.M.; Braga, T.T.; Silva de Assis, H.C. Experimentally exposed toxic effects of long-term exposure to environmentally relevant concentrations of CIP in males and females of the silver catfish Rhamdia quelen. Chemosphere 2023, 336, 139216. [Google Scholar] [CrossRef]
  19. Xin, F.; Susiarjo, M.; Bartolomei, M.S. Multigenerational and transgenerational effects of endocrine disrupting chemicals: A role for altered epigenetic regulation? Semin. Cell Dev. Biol. 2015, 43, 66–75. [Google Scholar] [CrossRef]
  20. DeCourten, B.M.; Forbes, J.P.; Roark, H.K.; Burns, N.P.; Major, K.M.; White, J.W.; Li, J.; Mehinto, A.C.; Connon, R.E.; Brander, S.M. Multigenerational and Transgenerational Effects of Environmentally Relevant Concentrations of Endocrine Disruptors in an Estuarine Fish Model. Environ. Sci. Technol. 2020, 54, 13849–13860. [Google Scholar] [CrossRef]
  21. Bergmann, M.; Allen, S.; Krumpen, T.; Allen, D. High Levels of Microplastics in the Arctic Sea Ice Alga Melosira arctica, a Vector to Ice-Associated and Benthic Food Webs. Environ. Sci. Technol. 2023, 57, 6799–6807. [Google Scholar] [CrossRef] [PubMed]
  22. Jolaosho, T.L.; Rasaq, M.F.; Omotoye, E.V.; Araomo, O.V.; Adekoya, O.S.; Abolaji, O.Y.; Hungbo, J.J. Microplastics in freshwater and marine ecosystems: Occurrence, characterization, sources, distribution dynamics, fate, transport processes, potential mitigation strategies, and policy interventions. Ecotoxicol. Environ. Saf. 2025, 294, 118036. [Google Scholar] [CrossRef] [PubMed]
  23. Rochman, C.M.; Hoellein, T. The global odyssey of plastic pollution. Science 2020, 368, 1184–1185. [Google Scholar] [CrossRef] [PubMed]
  24. Jambeck, J.R.; Geyer, R.; Wilcox, C.; Siegler, T.R.; Perryman, M.; Andrady, A.; Narayan, R.; Law, K.L. Marine pollution. Plastic waste inputs from land into the ocean. Science 2015, 347, 768–771. [Google Scholar] [CrossRef]
  25. Reboa, A.; Cutroneo, L.; Consani, S.; Geneselli, I.; Petrillo, M.; Besio, G.; Capello, M. Mugilidae fish as bioindicator for monitoring plastic pollution: Comparison between a commercial port and a fishpond (north-western Mediterranean Sea). Mar. Pollut. Bull. 2022, 177, 113531. [Google Scholar] [CrossRef]
  26. Kang, H.M.; Byeon, E.; Jeong, H.; Kim, M.S.; Chen, Q.; Lee, J.S. Different effects of nano- and microplastics on oxidative status and gut microbiota in the marine medaka Oryzias melastigma. J. Hazard. Mater. 2021, 405, 124207. [Google Scholar] [CrossRef]
  27. Kim, L.; Cui, R.; Il Kwak, J.; An, Y.J. Trophic transfer of nanoplastics through a microalgae-crustacean-small yellow croaker food chain: Inhibition of digestive enzyme activity in fish. J. Hazard. Mater. 2022, 440, 129715. [Google Scholar] [CrossRef]
  28. Bai, Z.; He, Y.; Hu, G.; Cheng, L.; Wang, M. Microplastics at an environmentally relevant dose enhance mercury toxicity in a marine copepod under multigenerational exposure: Multi-omics perspective. J. Hazard. Mater. 2024, 478, 135529. [Google Scholar] [CrossRef]
  29. Tanaka, K.; Takada, H. Microplastic fragments and microbeads in digestive tracts of planktivorous fish from urban coastal waters. Sci. Rep. 2016, 6, 34351. [Google Scholar] [CrossRef]
  30. Hamer, J.; Gutow, L.; Kohler, A.; Saborowski, R. Fate of microplastics in the marine isopod Idotea emarginata. Environ. Sci. Technol. 2014, 48, 13451–13458. [Google Scholar] [CrossRef]
  31. Wright, S.L.; Thompson, R.C.; Galloway, T.S. The physical impacts of microplastics on marine organisms: A review. Environ. Pollut. 2013, 178, 483–492. [Google Scholar] [CrossRef]
  32. Cole, M.; Lindeque, P.; Fileman, E.; Halsband, C.; Galloway, T.S. The impact of polystyrene microplastics on feeding, function and fecundity in the marine copepod Calanus helgolandicus. Environ. Sci. Technol. 2015, 49, 1130–1137. [Google Scholar] [CrossRef]
  33. Yang, H.; Xiong, H.; Mi, K.; Xue, W.; Wei, W.; Zhang, Y. Toxicity comparison of nano-sized and micron-sized microplastics to Goldfish Carassius auratus Larvae. J. Hazard. Mater. 2020, 388, 122058. [Google Scholar] [CrossRef]
  34. Sokmen, T.O.; Sulukan, E.; Turkoglu, M.; Baran, A.; Ozkaraca, M.; Ceyhun, S.B. Polystyrene nanoplastics (20 nm) are able to bioaccumulate and cause oxidative DNA damages in the brain tissue of zebrafish embryo (Danio rerio). Neurotoxicology 2020, 77, 51–59. [Google Scholar] [CrossRef]
  35. Ding, J.; Huang, Y.; Liu, S.; Zhang, S.; Zou, H.; Wang, Z.; Zhu, W.; Geng, J. Toxicological effects of nano- and micro-polystyrene plastics on red tilapia: Are larger plastic particles more harmless? J. Hazard. Mater. 2020, 396, 122693. [Google Scholar] [CrossRef]
  36. Liu, Z.; Li, L.; Sun, B.; Ding, Y.; Lv, Y.; Wu, Q.; Zhao, S.; Zhang, X.; Shen, T. Transgenerational effects of Nanoplastics and bisphenol A on Zebrafish lipid metabolism: Disruption of the gut Microbiota-liver axis via mTOR pathway. Aquat. Toxicol. 2025, 284, 107401. [Google Scholar] [CrossRef] [PubMed]
  37. Bucci, K.; Bayoumi, M.; Stevack, K.; Watson-Leung, T.; Rochman, C.M. Microplastics may induce food dilution and endocrine disrupting effects in fathead minnows (Pimephales promelas), and decrease offspring quality. Environ. Pollut. 2024, 345, 123551. [Google Scholar] [CrossRef] [PubMed]
  38. Wade, M.J.; Bucci, K.; Rochman, C.M.; Meek, M.H. Microplastic exposure is associated with epigenomic effects in the model organism Pimephales promelas (fathead minnow). J. Hered. 2025, 116, 113–125. [Google Scholar] [CrossRef] [PubMed]
  39. Zhang, X.; Chen, X.; Gao, L.; Zhang, H.T.; Li, J.; Ye, Y.; Zhu, Q.L.; Zheng, J.L.; Yan, X. Transgenerational effects of microplastics on Nrf2 signaling, GH/IGF, and HPI axis in marine medaka Oryzias melastigma under different salinities. Sci. Total Environ. 2024, 906, 167170. [Google Scholar] [CrossRef]
  40. Molnarova, L.; Halesova, T.; Tomesova, D.; Vaclavikova, M.; Bosakova, Z. Monitoring Pharmaceuticals and Personal Care Products in Healthcare Effluent Wastewater Samples and the Effectiveness of Drug Removal in Wastewater Treatment Plants Using the UHPLC-MS/MS Method. Molecules 2024, 29, 1480. [Google Scholar] [CrossRef]
  41. Lesser, L.E.; Mora, A.; Moreau, C.; Mahlknecht, J.; Hernandez-Antonio, A.; Ramirez, A.I.; Barrios-Pina, H. Survey of 218 organic contaminants in groundwater derived from the world’s largest untreated wastewater irrigation system: Mezquital Valley, Mexico. Chemosphere 2018, 198, 510–521. [Google Scholar] [CrossRef]
  42. K’Oreje, K.O.; Kandie, F.J.; Vergeynst, L.; Abira, M.A.; Van Langenhove, H.; Okoth, M.; Demeestere, K. Occurrence, fate and removal of pharmaceuticals, personal care products and pesticides in wastewater stabilization ponds and receiving rivers in the Nzoia Basin, Kenya. Sci. Total Environ. 2018, 637–638, 336–348. [Google Scholar] [CrossRef] [PubMed]
  43. Kasprzyk-Hordern, B.; Dinsdale, R.M.; Guwy, A.J. The occurrence of pharmaceuticals, personal care products, endocrine disruptors and illicit drugs in surface water in South Wales, UK. Water Res. 2008, 42, 3498–3518. [Google Scholar] [CrossRef]
  44. Perez-Alvarez, I.; Islas-Flores, H.; Gomez-Olivan, L.M.; Barcelo, D.; Lopez De Alda, M.; Perez Solsona, S.; Sanchez-Aceves, L.; SanJuan-Reyes, N.; Galar-Martinez, M. Determination of metals and pharmaceutical compounds released in hospital wastewater from Toluca, Mexico, and evaluation of their toxic impact. Environ. Pollut. 2018, 240, 330–341. [Google Scholar] [CrossRef] [PubMed]
  45. Kim, S.D.; Cho, J.; Kim, I.S.; Vanderford, B.J.; Snyder, S.A. Occurrence and removal of pharmaceuticals and endocrine disruptors in South Korean surface, drinking, and waste waters. Water Res. 2007, 41, 1013–1021. [Google Scholar] [CrossRef] [PubMed]
  46. Shen, M.; Yu, B.; Hu, Y.; Liu, Z.; Zhao, K.; Li, C.; Li, M.; Lyu, C.; Lu, H.; Zhong, S.; et al. Occurrence and Health Risk Assessment of Sulfonamide Antibiotics in Different Freshwater Fish in Northeast China. Toxics 2023, 11, 835. [Google Scholar] [CrossRef]
  47. Ye, C.; Shi, J.; Zhang, X.; Qin, L.; Jiang, Z.; Wang, J.; Li, Y.; Liu, B. Occurrence and bioaccumulation of sulfonamide antibiotics in different fish species from Hangbu-Fengle River, Southeast China. Environ. Sci. Pollut. Res. Int. 2021, 28, 44111–44123. [Google Scholar] [CrossRef]
  48. Fent, K.; Weston, A.A.; Caminada, D. Ecotoxicology of human pharmaceuticals. Aquat. Toxicol. 2006, 76, 122–159. [Google Scholar] [CrossRef]
  49. Ogunwole, G.A.; Saliu, J.K.; Osuala, F.I.; Odunjo, F.O. Chronic levels of ibuprofen induces haematoxic and histopathology damage in the gills, liver, and kidney of the African sharptooth catfish (Clarias gariepinus). Environ. Sci. Pollut. Res. Int. 2021, 28, 25603–25613. [Google Scholar] [CrossRef]
  50. Kidd, K.A.; Blanchfield, P.J.; Mills, K.H.; Palace, V.P.; Evans, R.E.; Lazorchak, J.M.; Flick, R.W. Collapse of a fish population after exposure to a synthetic estrogen. Proc. Natl. Acad. Sci. USA 2007, 104, 8897–8901. [Google Scholar] [CrossRef]
  51. Shi, F.; Huang, Y.; Yang, M.; Lu, Z.; Li, Y.; Zhan, F.; Lin, L.; Qin, Z. Antibiotic-induced alternations in gut microflora are associated with the suppression of immune-related pathways in grass carp (Ctenopharyngodon idellus). Front. Immunol. 2022, 13, 970125. [Google Scholar] [CrossRef]
  52. Au-Yeung, C.; Lam, K.L.; Choi, M.H.; Chan, K.W.; Cheung, Y.S.; Tsui, Y.L.; Mo, W.Y. Impact of Prophylactic Antibiotic Use in Ornamental Fish Tanks on Microbial Communities and Pathogen Selection in Carriage Water in Hong Kong Retail Shops. Microorganisms 2024, 12, 1184. [Google Scholar] [CrossRef]
  53. Hamid, N.; Junaid, M.; Wang, Y.; Pu, S.Y.; Jia, P.P.; Pei, D.S. Chronic exposure to PPCPs mixture at environmentally relevant concentrations (ERCs) altered carbohydrate and lipid metabolism through gut and liver toxicity in zebrafish. Environ. Pollut. 2021, 273, 116494. [Google Scholar] [CrossRef] [PubMed]
  54. Bhandari, R.K.; vom Saal, F.S.; Tillitt, D.E. Transgenerational effects from early developmental exposures to bisphenol A or 17alpha-ethinylestradiol in medaka, Oryzias latipes. Sci. Rep. 2015, 5, 9303. [Google Scholar] [CrossRef] [PubMed]
  55. Fraz, S.; Lee, A.H.; Pollard, S.; Srinivasan, K.; Vermani, A.; David, E.; Wilson, J.Y. Paternal Exposure to Carbamazepine Impacts Zebrafish Offspring Reproduction Over Multiple Generations. Environ. Sci. Technol. 2019, 53, 12734–12743. [Google Scholar] [CrossRef] [PubMed]
  56. Hammill, K.M.; Fraz, S.; Lee, A.H.; Wilson, J.Y. The effects of parental carbamazepine and gemfibrozil exposure on sexual differentiation in zebrafish (Danio rerio). Environ. Toxicol. Chem. 2018, 37, 1696–1706. [Google Scholar] [CrossRef]
  57. Xuan, R.; Qiu, W.; Zhou, Y.; Magnuson, J.T.; Luo, S.; Greer, J.B.; Xu, B.; Liu, J.; Xu, E.G.; Schlenk, D.; et al. Parental transfer of an antibiotic mixture induces cardiotoxicity in early life-stage zebrafish: A cross-generational study. Sci. Total Environ. 2022, 849, 157726. [Google Scholar] [CrossRef]
  58. Xiao, Y.; Han, D.; Currell, M.; Song, X.; Zhang, Y. Review of Endocrine Disrupting Compounds (EDCs) in China’s water environments: Implications for environmental fate, transport and health risks. Water Res. 2023, 245, 120645. [Google Scholar] [CrossRef]
  59. Yu, Y.; Wang, S.; Yu, P.; Wang, D.; Hu, B.; Zheng, P.; Zhang, M. A bibliometric analysis of emerging contaminants (ECs) (2001-2021): Evolution of hotspots and research trends. Sci. Total Environ. 2024, 907, 168116. [Google Scholar] [CrossRef]
  60. Fan, X.; Wu, L.; Hou, T.; He, J.; Wang, C.; Liu, Y.; Wang, Z. Maternal Bisphenol A exposure impaired endochondral ossification in craniofacial cartilage of rare minnow (Gobiocypris rarus) offspring. Ecotoxicol. Environ. Saf. 2018, 163, 514–520. [Google Scholar] [CrossRef]
  61. Scsukova, S.; Rollerova, E.; Bujnakova Mlynarcikova, A. Impact of endocrine disrupting chemicals on onset and development of female reproductive disorders and hormone-related cancer. Reprod. Biol. 2016, 16, 243–254. [Google Scholar] [CrossRef]
  62. Maqbool, F.; Mostafalou, S.; Bahadar, H.; Abdollahi, M. Review of endocrine disorders associated with environmental toxicants and possible involved mechanisms. Life Sci. 2016, 145, 265–273. [Google Scholar] [CrossRef] [PubMed]
  63. Rasmussen, T.H.; Andreassen, T.K.; Pedersen, S.N.; Van der Ven, L.T.; Bjerregaard, P.; Korsgaard, B. Effects of waterborne exposure of octylphenol and oestrogen on pregnant viviparous eelpout (Zoarces viviparus) and her embryos in ovario. J. Exp. Biol. 2002, 205, 3857–3876. [Google Scholar] [CrossRef] [PubMed]
  64. Major, K.M.; DeCourten, B.M.; Li, J.; Britton, M.; Settles, M.L.; Mehinto, A.C.; Connon, R.E.; Brander, S.M. Early Life Exposure to Environmentally Relevant Levels of Endocrine Disruptors Drive Multigenerational and Transgenerational Epigenetic Changes in a Fish Model. Front. Mar. Sci. 2020, 7, 2020. [Google Scholar] [CrossRef]
  65. Persinger, M.; Ward, J. Evaluation of cross-generational exposure to microplastics and co-occurring contaminants on embryonic and larval behavior in fathead minnows, Pimephales promelas. PeerJ 2025, 13, e19927. [Google Scholar] [CrossRef]
  66. Wang, M.; Chen, J.; Lin, K.; Chen, Y.; Hu, W.; Tanguay, R.L.; Huang, C.; Dong, Q. Chronic zebrafish PFOS exposure alters sex ratio and maternal related effects in F1 offspring. Environ. Toxicol. Chem. 2011, 30, 2073–2080. [Google Scholar] [CrossRef]
  67. Pierron, F.; Lorioux, S.; Heroin, D.; Daffe, G.; Etcheverria, B.; Cachot, J.; Morin, B.; Dufour, S.; Gonzalez, P. Transgenerational epigenetic sex determination: Environment experienced by female fish affects offspring sex ratio. Environ. Pollut. 2021, 277, 116864. [Google Scholar] [CrossRef]
  68. Nilsen, E.; Smalling, K.L.; Ahrens, L.; Gros, M.; Miglioranza, K.S.B.; Pico, Y.; Schoenfuss, H.L. Critical review: Grand challenges in assessing the adverse effects of contaminants of emerging concern on aquatic food webs. Environ. Toxicol. Chem. 2019, 38, 46–60. [Google Scholar] [CrossRef]
  69. Cheng, H.; Lv, C.; Li, J.; Wu, D.; Zhan, X.; Song, Y.; Zhao, N.; Jin, H. Bioaccumulation and biomagnification of emerging poly- and perfluoroalkyl substances in marine organisms. Sci. Total Environ. 2022, 851, 158117. [Google Scholar] [CrossRef]
  70. Aubert, N.; Ameller, T.; Legrand, J.J. Systemic exposure to parabens: Pharmacokinetics, tissue distribution, excretion balance and plasma metabolites of [14C]-methyl-, propyl- and butylparaben in rats after oral, topical or subcutaneous administration. Food Chem. Toxicol. 2012, 50, 445–454. [Google Scholar] [CrossRef]
  71. Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.; Mazur, M.; Telser, J. Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 2007, 39, 44–84. [Google Scholar] [CrossRef]
  72. Zhou, R.R.; Lu, G.H.; Yan, Z.H.; Jiang, R.R.; Sun, Y.; Zhang, P. Interactive transgenerational effects of polystyrene nanoplastics and ethylhexyl salicylate on zebrafish. Environ. Sci.-Nano 2021, 8, 146–159. [Google Scholar] [CrossRef]
  73. Li, Z.H.; Zlabek, V.; Velisek, J.; Grabic, R.; Machova, J.; Randak, T. Modulation of antioxidant defence system in brain of rainbow trout (Oncorhynchus mykiss) after chronic carbamazepine treatment. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2010, 151, 137–141. [Google Scholar] [CrossRef]
  74. Pitt, J.A.; Trevisan, R.; Massarsky, A.; Kozal, J.S.; Levin, E.D.; Di Giulio, R.T. Maternal transfer of nanoplastics to offspring in zebrafish (Danio rerio): A case study with nanopolystyrene. Sci. Total Environ. 2018, 643, 324–334. [Google Scholar] [CrossRef]
  75. Hayden, M.S.; Ghosh, S. Shared principles in NF-kappaB signaling. Cell 2008, 132, 344–362. [Google Scholar] [CrossRef] [PubMed]
  76. Bowden, T.J. Modulation of the immune system of fish by their environment. Fish. Shellfish. Immunol. 2008, 25, 373–383. [Google Scholar] [CrossRef] [PubMed]
  77. Milla, S.; Depiereux, S.; Kestemont, P. The effects of estrogenic and androgenic endocrine disruptors on the immune system of fish: A review. Ecotoxicology 2011, 20, 305–319. [Google Scholar] [CrossRef] [PubMed]
  78. Jobling, S.; Williams, R.; Johnson, A.; Taylor, A.; Gross-Sorokin, M.; Nolan, M.; Tyler, C.R.; van Aerle, R.; Santos, E.; Brighty, G. Predicted exposures to steroid estrogens in U.K. rivers correlate with widespread sexual disruption in wild fish populations. Environ. Health Perspect. 2006, 114 (Suppl. 1), 32–39. [Google Scholar] [CrossRef]
  79. Morshead, M.L.; Jensen, K.M.; Ankley, G.T.; Vliet, S.; LaLone, C.A.; Aller, A.V.; Watanabe, K.H.; Villeneuve, D.L. Putative adverse outcome pathway development based on physiological responses of female fathead minnows to model estrogen versus androgen receptor agonists. Aquat. Toxicol. 2023, 261, 106607. [Google Scholar] [CrossRef]
  80. Matsushima, A.; Teramoto, T.; Kakuta, Y. Crystal structure of endocrine-disrupting chemical bisphenol A and estrogen-related receptor gamma. J. Biochem. 2022, 171, 23–25. [Google Scholar] [CrossRef]
  81. Georgin, J.; Franco, D.S.P.; Manzar, M.S.; Meili, L.; El Messaoudi, N. A critical and comprehensive review of the current status of 17beta-estradiol hormone remediation through adsorption technology. Environ. Sci. Pollut. Res. Int. 2024, 31, 24679–24712. [Google Scholar] [CrossRef] [PubMed]
  82. Sciorio, R.; Greco, P.F.; Greco, E.; Tramontano, L.; Elshaer, F.M.; Fleming, S. Potential effects of environmental toxicants on sperm quality and potential risk for fertility in humans. Front. Endocrinol. 2025, 16, 1545593. [Google Scholar] [CrossRef] [PubMed]
  83. Yan, J.; Li, J.; Wang, Y.; Song, J.; Ni, A.; Fang, L.; Xi, M.; Qian, Q.; Wang, Z.; Wang, H. Deciphering the molecular mediators of triclosan-induced lipid accumulation: Intervention via short-chain fatty acids and miR-101a. Environ. Pollut. 2024, 343, 123153. [Google Scholar] [CrossRef]
  84. Liu, J.; Lu, L.; Song, H.; Liu, S.; Liu, G.; Lou, B.; Shi, W. Effects of triclosan on lipid metabolism and underlying mechanisms in the cyprinid fish Squalidus argentatus. Sci. Total Environ. 2024, 951, 175627. [Google Scholar] [CrossRef] [PubMed]
  85. Thapa, S.; Lv, M.; Xu, H. Acetylcholinesterase: A Primary Target for Drugs and Insecticides. Mini Rev. Med. Chem. 2017, 17, 1665–1676. [Google Scholar] [CrossRef]
  86. Burch, E.; Hussein, M.A.; Zaki, M.; Kamal, L.T.; Zaki, G.; Shoeib, T.; Dawood, M.; Sewilam, H.; Abdelnaser, A. Assessing the Effects of Pesticides on Aquacultured Fish and Ecosystems: A Comprehensive Environmental Health Review. Fishes 2025, 10, 223. [Google Scholar] [CrossRef]
  87. Berg, V.; Kraugerud, M.; Nourizadeh-Lillabadi, R.; Olsvik, P.A.; Skare, J.U.; Alestrom, P.; Ropstad, E.; Zimmer, K.E.; Lyche, J.L. Endocrine effects of real-life mixtures of persistent organic pollutants (POP) in experimental models and wild fish. J. Toxicol. Environ. Health A 2016, 79, 538–548. [Google Scholar] [CrossRef]
  88. Zhou, H.; Wu, H.; Liao, C.; Diao, X.; Zhen, J.; Chen, L.; Xue, Q. Toxicology mechanism of the persistent organic pollutants (POPs) in fish through AhR pathway. Toxicol. Mech. Methods 2010, 20, 279–286. [Google Scholar] [CrossRef]
  89. Tohyama, S.; Miyagawa, S.; Lange, A.; Ogino, Y.; Mizutani, T.; Tatarazako, N.; Katsu, Y.; Ihara, M.; Tanaka, H.; Ishibashi, H.; et al. Understanding the molecular basis for differences in responses of fish estrogen receptor subtypes to environmental estrogens. Environ. Sci. Technol. 2015, 49, 7439–7447. [Google Scholar] [CrossRef]
  90. Liu, C.; Zhao, L.P.; Shen, Y.Q. A systematic review of advances in intestinal microflora of fish. Fish. Physiol. Biochem. 2021, 47, 2041–2053. [Google Scholar] [CrossRef]
  91. Kim, S.; Stroski, K.M.; Killeen, G.; Smitherman, C.; Simcik, M.F.; Brooks, B.W. 8:8 Perfluoroalkyl phosphinic acid affects neurobehavioral development, thyroid disruption, and DNA methylation in developing zebrafish. Sci. Total Environ. 2020, 736, 139600. [Google Scholar] [CrossRef] [PubMed]
  92. Park, K.; Han, E.J.; Ahn, G.; Kwak, I.S. Effects of combined stressors to cadmium and high temperature on antioxidant defense, apoptotic cell death, and DNA methylation in zebrafish (Danio rerio) embryos. Sci. Total Environ. 2020, 716, 137130. [Google Scholar] [CrossRef] [PubMed]
  93. Bian, X.; Gao, Y. DNA methylation and gene expression alterations in zebrafish embryos exposed to cadmium. Environ. Sci. Pollut. Res. Int. 2021, 28, 30101–30110. [Google Scholar] [CrossRef] [PubMed]
  94. Pierron, F.; Daffe, G.; Daramy, F.; Heroin, D.; Barre, A.; Bouchez, O.; Clerendeau, C.; Romero-Ramirez, A.; Nikolski, M. Transgenerational endocrine disruptor effects of cadmium in zebrafish and contribution of standing epigenetic variation to adaptation. J. Hazard. Mater. 2023, 455, 131579. [Google Scholar] [CrossRef]
  95. Kamstra, J.H.; Sales, L.B.; Alestrom, P.; Legler, J. Differential DNA methylation at conserved non-genic elements and evidence for transgenerational inheritance following developmental exposure to mono(2-ethylhexyl) phthalate and 5-azacytidine in zebrafish. Epigenetics Chromatin 2017, 10, 20. [Google Scholar] [CrossRef]
  96. Wan, T.; Mo, J.; Au, D.W.; Qin, X.; Tam, N.Y.; Kong, R.Y.; Seemann, F. The role of DNA methylation on gene expression in the vertebrae of ancestrally benzo[a]pyrene exposed F1 and F3 male medaka. Epigenetics 2023, 18, 2222246. [Google Scholar] [CrossRef]
  97. Wang, S.Y.; Lau, K.; Lai, K.P.; Zhang, J.W.; Tse, A.C.; Li, J.W.; Tong, Y.; Chan, T.F.; Wong, C.K.; Chiu, J.M.; et al. Hypoxia causes transgenerational impairments in reproduction of fish. Nat. Commun. 2016, 7, 12114. [Google Scholar] [CrossRef]
  98. Skvortsova, K.; Tarbashevich, K.; Stehling, M.; Lister, R.; Irimia, M.; Raz, E.; Bogdanovic, O. Retention of paternal DNA methylome in the developing zebrafish germline. Nat. Commun. 2019, 10, 3054. [Google Scholar] [CrossRef]
  99. Wang, X.; Bhandari, R.K. The dynamics of DNA methylation during epigenetic reprogramming of primordial germ cells in medaka (Oryzias latipes). Epigenetics 2020, 15, 483–498. [Google Scholar] [CrossRef]
  100. Beck, D.; Ben Maamar, M.; Skinner, M.K. Genome-wide CpG density and DNA methylation analysis method (MeDIP, RRBS, and WGBS) comparisons. Epigenetics 2022, 17, 518–530. [Google Scholar] [CrossRef]
  101. Carvan, M.J., 3rd; Kalluvila, T.A.; Klingler, R.H.; Larson, J.K.; Pickens, M.; Mora-Zamorano, F.X.; Connaughton, V.P.; Sadler-Riggleman, I.; Beck, D.; Skinner, M.K. Mercury-induced epigenetic transgenerational inheritance of abnormal neurobehavior is correlated with sperm epimutations in zebrafish. PLoS ONE 2017, 12, e0176155. [Google Scholar] [CrossRef]
  102. Iwanami, N.; Lawir, D.F.; Sikora, K.; O’Meara, C.; Takeshita, K.; Schorpp, M.; Boehm, T. Transgenerational inheritance of impaired larval T cell development in zebrafish. Nat. Commun. 2020, 11, 4505. [Google Scholar] [CrossRef] [PubMed]
  103. Ge, L.; Zhang, R.P.; Wan, F.; Guo, D.Y.; Wang, P.; Xiang, L.X.; Shao, J.Z. TET2 plays an essential role in erythropoiesis by regulating lineage-specific genes via DNA oxidative demethylation in a zebrafish model. Mol. Cell Biol. 2014, 34, 989–1002. [Google Scholar] [CrossRef] [PubMed]
  104. Cavasotto, C.N.; Scardino, V. Machine Learning Toxicity Prediction: Latest Advances by Toxicity End Point. ACS Omega 2022, 7, 47536–47546. [Google Scholar] [CrossRef] [PubMed]
  105. Vo, A.H.; Van Vleet, T.R.; Gupta, R.R.; Liguori, M.J.; Rao, M.S. An Overview of Machine Learning and Big Data for Drug Toxicity Evaluation. Chem. Res. Toxicol. 2020, 33, 20–37. [Google Scholar] [CrossRef]
  106. Wu, Y.; Wang, G. Machine Learning Based Toxicity Prediction: From Chemical Structural Description to Transcriptome Analysis. Int. J. Mol. Sci. 2018, 19, 2358. [Google Scholar] [CrossRef]
  107. Cheng, Y.; Wang, Y.; Xu, Z.; Feng, C.; Dong, Z.; Fan, W.; Leung, K.M.Y.; Wu, F. Predicting the Site-Specific Toxicity of Metals to Fishes Using a New Machine Learning-Based Approach. Environ. Sci. Technol. 2025, 59, 14881–14891. [Google Scholar] [CrossRef]
  108. Sun, T.; Wei, C.; Liu, Y.; Ren, Y. Explainable machine learning models for predicting the acute toxicity of pesticides to sheepshead minnow (Cyprinodon variegatus). Sci. Total Environ. 2024, 957, 177399. [Google Scholar] [CrossRef]
  109. Costa, D.F.D.; Zanardini, M.; Sanches, E.A.; de Souza, A.R.S.; da Silva Rodrigues, M.; de Moraes, A.C.N.; Habibi, H.R.; Nobrega, R.H. Androgenic overactivation and epigenetic remodeling drive intergenerational toxicity of bisphenol S in zebrafish. Ecotoxicol. Environ. Saf. 2025, 303, 118831. [Google Scholar] [CrossRef]
  110. Zhang, B.F.; Wu, X.Z.; Wang, J.X.; Liao, Y.L.; Cai, Z.X.; Liu, Y.; Pei, D.S. Neurotoxicity of chronic nano-neodymium oxide exposure in zebrafish: Behavioral and molecular insights. J. Hazard. Mater. 2025, 495, 138879. [Google Scholar] [CrossRef]
  111. Feng, J.X.; Li, P.; Liu, Y.; Liu, L.; Li, Z.H. A latest progress in the study of fish behavior: Cross-generational effects of behavior under pollution pressure and new technologies for behavior monitoring. Environ. Sci. Pollut. Res. Int. 2024, 31, 11529–11542. [Google Scholar] [CrossRef] [PubMed]
  112. Sheffield, T.Y.; Judson, R.S. Ensemble QSAR Modeling to Predict Multispecies Fish Toxicity Lethal Concentrations and Points of Departure. Environ. Sci. Technol. 2019, 53, 12793–12802. [Google Scholar] [CrossRef] [PubMed]
  113. Gustavsson, M.; Kall, S.; Svedberg, P.; Inda-Diaz, J.S.; Molander, S.; Coria, J.; Backhaus, T.; Kristiansson, E. Transformers enable accurate prediction of acute and chronic chemical toxicity in aquatic organisms. Sci. Adv. 2024, 10, eadk6669. [Google Scholar] [CrossRef] [PubMed]
  114. Fan, J.; Huang, G.; Chi, M.; Shi, Y.; Jiang, J.; Feng, C.; Yan, Z.; Xu, Z. Prediction of chemical reproductive toxicity to aquatic species using a machine learning model: An application in an ecological risk assessment of the Yangtze River, China. Sci. Total Environ. 2021, 796, 148901. [Google Scholar] [CrossRef]
  115. Liu, J.; Guo, W.; Dong, F.; Aungst, J.; Fitzpatrick, S.; Patterson, T.A.; Hong, H. Machine learning models for rat multigeneration reproductive toxicity prediction. Front. Pharmacol. 2022, 13, 1018226. [Google Scholar] [CrossRef]
  116. Haque, M.M.; Holder, L.B.; Skinner, M.K. Genome-Wide Locations of Potential Epimutations Associated with Environmentally Induced Epigenetic Transgenerational Inheritance of Disease Using a Sequential Machine Learning Prediction Approach. PLoS ONE 2015, 10, e0142274. [Google Scholar] [CrossRef]
  117. Mavaie, P.; Holder, L.; Skinner, M. Identifying unique exposure-specific transgenerational differentially DNA methylated region epimutations in the genome using hybrid deep learning prediction models. Environ. Epigenet 2023, 9, dvad007. [Google Scholar] [CrossRef]
Figure 1. Emerging contaminants (ECs) in aquatic ecosystems and their classification into six major categories: (1) Pharmaceutical and Personal Care Products (PPCPs)—antibiotics, hormones, cosmetics, illicit drugs; (2) Endocrine Disrupting Compounds (EDCs)—bisphenols, phthalates, alkylphenols, synthetic hormones; (3) Industrial Substances—PFAS, flame retardants, surfactants, plasticizers; (4) Micro/Nano Particles—microplastics and nanoplastics (MNPs), engineered nanoparticles; (5) Biological Agents—antibiotic resistance genes, cyanobacterial toxins; (6) Non-Nutritive Sweeteners—sucralose, saccharin, acesulfame.
Figure 1. Emerging contaminants (ECs) in aquatic ecosystems and their classification into six major categories: (1) Pharmaceutical and Personal Care Products (PPCPs)—antibiotics, hormones, cosmetics, illicit drugs; (2) Endocrine Disrupting Compounds (EDCs)—bisphenols, phthalates, alkylphenols, synthetic hormones; (3) Industrial Substances—PFAS, flame retardants, surfactants, plasticizers; (4) Micro/Nano Particles—microplastics and nanoplastics (MNPs), engineered nanoparticles; (5) Biological Agents—antibiotic resistance genes, cyanobacterial toxins; (6) Non-Nutritive Sweeteners—sucralose, saccharin, acesulfame.
Fishes 10 00535 g001
Figure 2. Experimental exposure scenarios and endpoint assessment in fish transgenerational toxicity studies. (Left panel) Three primary exposure modes: (A) Multigenerational exposure—continuous exposure across F1–F3 generations with all fish directly exposed; (B) Transgenerational exposure—ancestral F1 exposure only, with F2 embryos exposed in utero/ovo, F3 representing the first truly unexposed generation, and F4 representing definitive transgenerational inheritance; (C) Single-parent exposure—either paternal or maternal F1 exposure to assess sex-specific transmission. (Middle panel) Post-exposure period showing F2 and F3 generations reared in clean water to assess persistent effects. (Right panel) Hierarchical endpoint monitoring: (A) Reproductive and developmental endpoints—fertilization, hatching, survival rates; (B) Physiological endpoints—skeletal deformities, cardiac abnormalities, impaired spermatogenesis; (C) Systemic function endpoints—neurobehavioral toxicity, endocrine disruption, immune dysfunction; (D) Molecular mechanism endpoints—multi-omics integration (genomics, transcriptomics, proteomics, metabolomics) and epigenomic analysis revealing mechanistic pathways underlying phenotypic outcomes.
Figure 2. Experimental exposure scenarios and endpoint assessment in fish transgenerational toxicity studies. (Left panel) Three primary exposure modes: (A) Multigenerational exposure—continuous exposure across F1–F3 generations with all fish directly exposed; (B) Transgenerational exposure—ancestral F1 exposure only, with F2 embryos exposed in utero/ovo, F3 representing the first truly unexposed generation, and F4 representing definitive transgenerational inheritance; (C) Single-parent exposure—either paternal or maternal F1 exposure to assess sex-specific transmission. (Middle panel) Post-exposure period showing F2 and F3 generations reared in clean water to assess persistent effects. (Right panel) Hierarchical endpoint monitoring: (A) Reproductive and developmental endpoints—fertilization, hatching, survival rates; (B) Physiological endpoints—skeletal deformities, cardiac abnormalities, impaired spermatogenesis; (C) Systemic function endpoints—neurobehavioral toxicity, endocrine disruption, immune dysfunction; (D) Molecular mechanism endpoints—multi-omics integration (genomics, transcriptomics, proteomics, metabolomics) and epigenomic analysis revealing mechanistic pathways underlying phenotypic outcomes.
Fishes 10 00535 g002
Figure 3. Molecular mechanisms underlying the trans-/multi-generational toxicity of Emerging contaminants (ECs) on fish. ECs enter fish through multiple routes and are absorbed across epithelial cells of the small intestine into systemic circulation. Once internalized, ECs trigger three primary toxicity pathways that can transmit across generations: (I) Oxidative stress pathway (left, yellow box): ECs induce excessive reactive oxygen species (ROS) generation, overwhelming antioxidant defenses and causing lipid peroxidation in cellular membranes. Oxidative damage to oocytes and sperm can transmit to offspring through compromised gamete quality and oxidative modifications to DNA/proteins in germ cells. (II) Endocrine disruption pathway (center, blue green box): ECs bind to hormone receptors (estrogen, androgen, thyroid), disrupting normal endocrine signaling in reproductive tissues. This alters gonadal development, gamete production, and hormone-regulated gene expression, with effects persisting in offspring through altered maternal/paternal provisioning and epigenetic modifications in germ cells. (III) Genotoxic pathway (right, pink box): ECs cause non-lethal DNA mutations and epigenetic alterations (DNA methylation, histone modifications) in somatic and germ cells. While lethal mutations lead to cell death (bottom left), sub-lethal genetic and epigenetic changes produce unhealthy cells (bottom right) with altered gene expression patterns. When these changes occur in germ cells, they transmit to F1–F3 generations, causing the transgenerational phenotypes reviewed in Section 2. Black dashed arrows indicate intergenerational transmission pathways; colored solid arrows show pathway progression within exposed organisms: Red symbols indicate inhibitory effects; blue symbols indicate receptor binding and activation.
Figure 3. Molecular mechanisms underlying the trans-/multi-generational toxicity of Emerging contaminants (ECs) on fish. ECs enter fish through multiple routes and are absorbed across epithelial cells of the small intestine into systemic circulation. Once internalized, ECs trigger three primary toxicity pathways that can transmit across generations: (I) Oxidative stress pathway (left, yellow box): ECs induce excessive reactive oxygen species (ROS) generation, overwhelming antioxidant defenses and causing lipid peroxidation in cellular membranes. Oxidative damage to oocytes and sperm can transmit to offspring through compromised gamete quality and oxidative modifications to DNA/proteins in germ cells. (II) Endocrine disruption pathway (center, blue green box): ECs bind to hormone receptors (estrogen, androgen, thyroid), disrupting normal endocrine signaling in reproductive tissues. This alters gonadal development, gamete production, and hormone-regulated gene expression, with effects persisting in offspring through altered maternal/paternal provisioning and epigenetic modifications in germ cells. (III) Genotoxic pathway (right, pink box): ECs cause non-lethal DNA mutations and epigenetic alterations (DNA methylation, histone modifications) in somatic and germ cells. While lethal mutations lead to cell death (bottom left), sub-lethal genetic and epigenetic changes produce unhealthy cells (bottom right) with altered gene expression patterns. When these changes occur in germ cells, they transmit to F1–F3 generations, causing the transgenerational phenotypes reviewed in Section 2. Black dashed arrows indicate intergenerational transmission pathways; colored solid arrows show pathway progression within exposed organisms: Red symbols indicate inhibitory effects; blue symbols indicate receptor binding and activation.
Fishes 10 00535 g003
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

Sun, D.; Huang, Y.; Chen, S.; Duan, M. Review on Toxicity Effect of Emerging Contaminants on Trans-/Multi-Generational Fish. Fishes 2025, 10, 535. https://doi.org/10.3390/fishes10110535

AMA Style

Sun D, Huang Y, Chen S, Duan M. Review on Toxicity Effect of Emerging Contaminants on Trans-/Multi-Generational Fish. Fishes. 2025; 10(11):535. https://doi.org/10.3390/fishes10110535

Chicago/Turabian Style

Sun, Dong, Yuna Huang, Shuyuan Chen, and Meina Duan. 2025. "Review on Toxicity Effect of Emerging Contaminants on Trans-/Multi-Generational Fish" Fishes 10, no. 11: 535. https://doi.org/10.3390/fishes10110535

APA Style

Sun, D., Huang, Y., Chen, S., & Duan, M. (2025). Review on Toxicity Effect of Emerging Contaminants on Trans-/Multi-Generational Fish. Fishes, 10(11), 535. https://doi.org/10.3390/fishes10110535

Article Metrics

Back to TopTop