Next Article in Journal
Prevalence and Molecular Characterization of Bovine Parainfluenza Virus Type 3 in Cattle Herds in China
Next Article in Special Issue
Transcriptomic Response of the Liver Tissue in Trachinotus ovatus to Acute Heat Stress
Previous Article in Journal
A Review of Three Decades of Research Dedicated to Making Equine Bones Stronger: Implications for Horses and Humans
Previous Article in Special Issue
Responses of Micropterus salmoides under Ammonia Stress and the Effects of a Potential Ammonia Antidote
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Elucidating the Effects of the Lipids Regulators Fibrates and Statins on the Health Status of Finfish Species: A Review

1
Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
2
Department of Physiology, Institute of Bioscience, Universidade de São Paulo, São Paulo 05508-090, Brazil
3
Faculty of Sciences of the University of Lisbon—FCUL, Campo Grande, 1749-016 Lisboa, Portugal
4
Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Animals 2023, 13(5), 792; https://doi.org/10.3390/ani13050792
Submission received: 10 January 2023 / Revised: 15 February 2023 / Accepted: 17 February 2023 / Published: 22 February 2023
(This article belongs to the Special Issue The Effects of Pollution and Other Stressors on Fish Health)

Abstract

:

Simple Summary

Pharmaceuticals used to treat abnormal cholesterol levels in the blood are known as lipid regulators (fibrates and statins), and their use is in constant growth. Treatment plants are usually unable to efficiently remove these compounds from wastewater, where their degradation rate is considerably slow, making them an emerging concern for aquatic systems. The present work reviews previously published research concerning the effects of these pharmaceuticals on several finfish species worldwide. Results suggest that both short- and long-term exposure to lipid regulators may have negative effects on fish health, affecting their metabolism and immune system and causing reproductive and developmental disorders. However, the information on these compounds in the available literature is still limited, and additional research is needed to fully understand the threat that their presence in aquatic systems may pose to the production of finfish by the aquaculture industry.

Abstract

The most documented fibrates are gemfibrozil, clofibrate and bezafibrate, while for statins, the majority of the published literature focuses on atorvastatin and simvastatin. The present work reviews previously published research concerning the effects of these hypocholesterolaemic pharmaceuticals on fish, with a particular focus on commercially important species, commonly produced by the European aquaculture industry, specifically in recirculated aquaculture systems (RAS). Overall, results suggest that both acute and chronic exposures to lipid-lowering compounds may have adverse effects on fish, disrupting their capacity to excrete exogenous substances, as well as both lipid metabolism and homeostasis, causing severe ontogenetic and endocrinological abnormalities, leading to hampered reproductive success (e.g., gametogenesis, fecundity), and skeletal or muscular malformations, having serious repercussions on fish health and welfare. Nonetheless, the available literature focusing on the effects of statins or fibrates on commonly farmed fish is still limited, and further research is required to understand the implications of this matter on aquaculture production, global food security and, ultimately, human health.

1. Introduction

The daily use of man-made products such as pharmaceuticals, cleaning agents and plastics is in constant and accelerating growth [1], and many of these contaminants have been detected in a variety of environments [2,3]. This uncontrolled anthropogenic activity has triggered several potential risks for aquatic environments [4,5,6] since several contaminants have been quantified in considerable amounts both in surface and ground waters, as well as within organisms [7,8]. Conventional wastewater treatment processes are still unable to efficiently remove all contaminants from influents, and a substantial share of these pollutants, known as contaminants of emerging concern (CECs), remain in the effluent due to their high persistence and low degradation rates [1,9,10,11,12,13]. The input of many compounds classified as personal care products (PPCP), flame-retardants, endocrine-disrupting chemicals, polyfluoroalkyl substances and pharmaceuticals from wastewater treatment plants are not monitored in water, potentially subjecting different organisms to chronic exposures to various chemicals [14,15,16]. Improvements in analytical techniques allow for the detection of an increasing number of these compounds in a variety of bodies of water (e.g., stagnant and running water; surface and ground waters; salt, fresh and brackish water; and wastewater treatment plant influents, effluents and sludge) [17,18,19,20], as well as in food items [16,21]. Pharmaceuticals play a major role in life quality and welfare, but the fast increase in their use has led to significant pressure on natural ecosystems [20,22,23,24]. The over- and misuse of pharmaceuticals, combined with their persistence in the environment, and their proven ability to bioaccumulate on both organisms and sediments [25], make these compounds, and their respective bioactive metabolites, potential chemical stressors to aquatic and cultured fish, and, consequently, a possible threat to humans.
Common aquaculture practices face a number of challenges (e.g., disease outbreaks, feed components shortages, droughts, unexpected flow of contaminants and xenobiotics) and present multiple threats to natural environments, such as eutrophication, habitat destruction and organisms escaping from aquaculture facilities into the wild [26,27,28]. To overcome the effects of those stressors on farmed fish, over the past few decades, recirculated aquaculture systems (RAS) have caught the attention of seafood farmers all over the world and have been increasingly developed at an international scale [27]. RAS are based on the concept of reusing or recirculating, over 90% of the production water, depending on the specific design, therefore minimising water exchange with the surrounding environments compared to traditional aquaculture practices [29,30]. The recirculated water is repeatedly treated with biological, mechanical, and often UV filters in order to maintain appropriate water quality [30,31]. It is now widely accepted that the implementation of RAS provides a wide range of benefits and that this technology may be increasingly adopted to raise all fish species. Firstly, the considerable reduction in water use allows for RAS to be installed in a variety of environments, including areas where water is scarce [31]. Second, being a virtually closed system, RAS enable the farmers to hold full control of water parameters that could naturally fluctuate in a body of water, such as oxygen levels, the concentration of nitrogenous compounds, temperature, and, eventually, the presence of pathogens [30,32,33].
In addition, RAS are usually installed indoors, therefore being sheltered from external factors such as climatic events and the potential predation of cultured organisms from wild animals [27]. Last, but not least, RAS significantly reduce the impact of aquaculture farms on neighbouring natural ecosystems. Indeed, by isolating the facility from the surrounding environments, the probabilities of eutrophication, escapees with potentially deleterious effects on wild populations, pathogen transfer or contamination outflow is minimised [27,30]. The low rates of water exchange, which directly translate to a considerable reduction in water usage, prevent surrounding rivers or groundwater to be heavily polluted or depleted as a consequence of this activity [28,33], and minimise the risk of pathogens entering the systems, translating to a decreased use of antibiotics [31,34].
These factors, in conjunction with the versatility of RAS regarding the species and life stages suitable for culture in such systems [27,31], as well as the different possible stocking and production densities [30], make RAS an appropriate approach to deal with the growing food demand while minimising the environmental impact of the food production industry [27,30,35]. Nonetheless, the array of technicalities incurred by RAS entails farmers developing further skills and expertise and must therefore undertake specific training in order to properly manage the entire system [30]. Furthermore, the different components of RAS, including power supply and sensory systems, are particularly expensive, and consequently, it has been observed that these constraints may restrict the development of these aquaculture practices to areas with greater financial power, such as European countries, North America or some Asian countries [27,36,37,38]. Therefore, further technological advances are required to make RAS truly cost-effective, and to enable its expansion and implementation all over the world [27]. In Europe, RAS farms are principally used to culture aquatic invertebrates (e.g., molluscs and crustaceans), and although the vast majority of farmed fish are salmonids (i.e., Salmo trutta, Oncorhynchus mykiss), other fish such as Anguilla anguilla (European eel) are also widely produced [37].
As pharmaceuticals are now recognised as ubiquitous in aquatic systems [39], and with treatment plants proving unable to remove these from influents [40], it can be assumed that these contaminants are also present in RAS and will affect cultured fish, and consequently aquaculture production. Here we will focus on the effects of lipid regulators, a group of pharmaceuticals engineered to treat dyslipidaemias in humans, acting as lipid-lowering agents, and as primary prevention methods for cardiovascular diseases [39,41].
Lipid regulators are usually manufactured from a variety of compounds such as atorvastatin, bezafibrate, clofibrate, ezetimibe, fenofibrate, fluvastatin, gemfibrozil, lovastatin, mevastatin, pravastatin, rosuvastatin or simvastatin. The first drug of its kind introduced to the market was clofibrate, followed by bezafibrate, fenofibrate and gemfibrozil [42]. Due to the rising need for anti-cholesterol treatments, with earlier-stage treatments and higher dosages [43], lipid regulators are widely prescribed all around the world, and consequently, comprise some of the most reported pharmaceuticals in drinking and wastewater [42,44,45]. The prescription and consumption of lipid regulators multiplied between 2000 and 2017 in countries members of the OECD, with the United Kingdom, Denmark and Belgium displaying the highest consumption rates [42]. Lipid regulators can be sub-classified into statins and fibrates [39], and, even if both are ubiquitous in water bodies, fibrates are often found at higher concentrations [42].
The present work aims to review the currently available literature on the presence of lipid-regulating agents (i.e., fibrates and statins) in the aquatic environment, and their effect on fish, including those commonly farmed in European RAS.

1.1. Fibrates

Fibrates decrease levels of fatty acids, triglycerides and low-density lipoproteins, mainly by stimulating the peroxisomal, and partly the mitochondrial, β-oxidation pathways, lowering blood cholesterol levels [39]. On the other hand, fibrates act as agonists of the peroxisome proliferator-activated receptors alpha (PPAR-α), expressed in the liver, heart and skeletal muscle [46]. In the literature, the most commonly investigated fibrates are gemfibrozil, bezafibrate and clofibric acid, with the latter being the most commonly detected in drinking water and food [42]. The most common lipid regulators in water bodies, namely, atorvastatin, bezafibrate, clofibrate, gemfibrozil and simvastatin, have been described to have similar effects in aquatic organisms, particularly vertebrates, as they do in humans, including the induction of changes in lipid levels [42]. Hence, marine and freshwater organisms are vulnerable to the potentially deleterious effects that the presence of such contaminants might incur, making this an issue of major ecological concern. It is known that exposure to lipid regulators generally present in water (e.g., wastewater treatment effluents and drinking water) can have a range of consequences on fish, affecting physiology, metabolism, reproduction, behaviour and immunity [44,47,48,49]. For instance, previous studies have shown that exposure to gemfibrozil may lead to decreased levels of cholesterol in plasma [48], and trigger the organism’s antioxidant response, as well as tamper with a fish’s capacity to efficiently swim in counter-current [47]. Moreover, gemfibrozil, like many other compounds, can bioconcentrate in different tissues in fish [50], potentially having deleterious effects on human health through the ingestion of contaminated food. Gemfibrozil, is resistant to photodegradation, has a long half-life in water [51] and is not completely removed by wastewater treatment plants (WWTP) [40]. In Spain, for instance, removal rates of this pollutant from WWTP ranged between 10–75% [52]. Its persistence in water allows us to find concentrations up to 4760 ng/L in effluents [53], and up to 758 ng/L in coastal waters [54]. Moreover, values ranging between 6.69 and 10.34 ng/L were recorded in Portuguese surface waters [55] and up to 0.8 and 1.7 ng/L in Italian tap and surface waters, respectively [56]. In comparison, in Spain, values of up to 70.27 and 77.8 ng/L were detected in different sites of the Llobregat river [57,58].
Bezafibrate was detected in wastewater treatment facilities effluents in Spain at concentrations ranging between 2 and 132 ng/L, and in estuarine environments at concentrations ranging between 2 and 67 ng/L [59]. In comparison, Portuguese surface waters were contaminated with values of bezafibrate ranging from 11.86 to 15.52 ng/L [55]. Moreover, bezafibrate, gemfibrozil and clofibric acid, an active metabolite of clofibrate, have been detected in groundwater in Barcelona, being found in concentrations of maximum 25.8, 751 and 7.57 ng/L, respectively [18]. Nonetheless, the most predominantly detected fibrate in the aquatic environment is clofibric acid [60,61].

1.2. Statins

Statins lower cholesterol levels through the suppression of its biosynthesis as a result of competitive inhibition of 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMG-CoA reductase), which is responsible for cholesterol biosynthesis in the liver [39]. Statins currently available on the market include atorvastatin, cerivastatin, fluvastatin, lovastatin, pitavastatin, pravastatin, rosuvastatin and simvastatin [62,63]. However, data on the toxicity of statins on different organisms are very limited and are generally restricted to atorvastatin and simvastatin, the two most common statins [63,64]. Few studies have focused on the environmental concentrations of statins despite the evident increase in their consumption. Statins appear to have more stable removal efficiencies compared to fibrates, but their presence has already been reported in both untreated and treated sewage water samples at values ranging from 4 to 117 ng/L and 1 to 59 ng/L, respectively [65]. Atorvastatin, for example, was found in low amounts, although its metabolites, namely p-hydroxy atorvastatin, and o-hydroxy atorvastatin, were found in greater amounts in wastewater. A similar trend occurs with simvastatin, where this statin was not directly detected in surface water and sediments unlike its specific metabolite (i.e., simvastatin hydroxy carboxylic acid) and was found up to 108 ng/L in Norwegian cities [66].
The inefficiency of wastewater treatment plants when it comes to the removal of these pharmaceuticals is translated by an inevitable exposure of both wild and cultured aquatic animals to these contaminants of emerging concern, even in closed systems, such as recirculated aquaculture systems (RAS), where they might accumulate in water. In both wild and laboratory-reared fishes, the consequences of exposure to fibrates and statins have been studied to varying extents, depending on compound and species, indicating the potentially deleterious effects of these contaminants on the overall health and welfare of fish (Figure 1). Here, we aim to compile existing knowledge on the effects of lipid-regulating agents, their environmental distribution, as well as their effects on fish, since these pharmaceuticals are likely to influence their lipid metabolism and nutritional quality, with a special interest in organisms that are highly relevant to aquaculture, and, therefore, directly linked to global food security and human health. We will also focus specifically on the most documented drugs, namely gemfibrozil, clofibric acid (a metabolite of clofibrate) and bezafibrate, representing the fibrates, and atorvastatin and simvastatin representing the statins.

2. Methodology

Articles published from 2011 onwards were selected in July–October 2022, using “lipid regulators”, “lipid-lowering agent”, gemfibrozil”, “clofibrate”, “clofibric acid”, “bezafibrate”, “fibrates”, atorvastatin”, “simvastatin” and “statins” as keywords. Moreover, the combinations of the aforementioned keywords, in conjunction with “effects in fish”, “aquaculture” and both the common and scientific names of the main species of interest for the European aquaculture industry (i.e., Salmo salar, Oncorhynchus mykiss, Anguilla anguilla and Clarias gariepinus) were also used. Due to the scarcity of recent studies focusing on the environmental presence and distribution of these pharmaceuticals in European countries, as well as their effects on the previously specified species of interest to the present work, articles related to these matters were selected, even if the publication was dated prior to 2011.

3. Effects of Lipids Regulators on Fish

An increasing number of studies focus on the potentially deleterious effects that the presence of lipid-regulating agents may have on non-target organisms. These suggest that lipid regulators can act similarly in mammals (e.g., humans) and other vertebrates, such as fish, when exposed to these compounds through contaminated waters. This factor, combined with the high use of these drugs, their incorrect discard and their low removal rates, imply a major environmental problem.

3.1. Fibrates

3.1.1. Gemfibrozil

A considerable quantity of the published literature has focused on the effects of gemfibrozil in aquatic organisms as it is now an issue of high concern (Table 1). In humans, gemfibrozil is capable of increasing serum aminotransferase, and, in isolated cases, of causing liver damage. The reduction in lipids is associated with the peroxisome proliferator-activated receptor (PPARα), involved in the gene expression of enzymes for the oxidation of fatty acids [45]. In fish, as in other animals, an increase in levels of lipoprotein lipase can occur, subsequently increasing triacyl glyceride-rich lipoproteins [51]. Danio rerio (zebrafish) exposed to gemfibrozil through diet for 30 days displayed decreased plasma levels of triglycerides and overall cholesterol levels [44]. These results corroborate the idea that gemfibrozil can operate in similar ways in fish as in humans, notably generating changes in lipid homeostasis and altering the availability of energy resources. Nonetheless, a study with fathead minnow (Pimephales promelas) showed an inconsistent effect of chronic exposure to gemfibrozil on lipid metabolism, steroidogenesis and reproduction, and no effect when employing environmentally relevant concentrations [48].
In gilthead seabream (Sparus aurata), gemfibrozil was found to affect swimming behaviour [47,90], with the ability to swim in counter-current for extended periods of time decreased by up to 65%. These behavioural effects were further associated with a significant increase in both catalase (CAT) and glutathione reductase (GR) activities in the gills of fish exposed to concentrations of the pharmaceuticals [47]. Furthermore, exposure to gemfibrozil appeared to have genotoxic effects on this species, causing nuclear anomalies [74]. In addition, exposure to gemfibrozil did not change gill total antioxidant capacity (TAC) but increased total oxidative status (TOS) and altered mitochondrial RNA of certain genes such as glutathione peroxidase 1 (GPx) and interleukin 1β in both gills and head kidney of the same species [49]. The transcription of key genes involved in lipid homeostasis was also affected by exposure to gemfibrozil, being characterized as a stress-inducing agent. Furthermore, exposure of S. aurata to 15,000 ng/L of gemfibrozil induced the activation of the inflammatory response in the liver [72]. Altogether, these studies suggest that, in S. aurata, this compound alters biochemical and gene functions involved in the immune and oxidative stress responses, having serious implications for the health and welfare of this species.
The bioaccumulation of gemfibrozil in both muscle and adipose tissue of juvenile rainbow trout (Oncorhynchus mykiss) following an 8-day waterborne exposure to 3000 ng/L of gemfibrozil has been previously demonstrated using space-resolved solid-phase microextraction (SR-SPME) [50]. Prindiville et al. [45] investigated the effects of exposure to gemfibrozil on the lipoprotein metabolism of O. mykiss. The authors injected female individuals with 100 mg/kg of gemfibrozil every 3 days over a 15-day period and reported a significant decrease in the concentration of plasma lipids, as well as in lipoprotein concentrations. Such results might translate into a hindered ability to reproduce, to efficiently undergo homeoviscous adaption to changes in water temperature, as well as tampered locomotor capacities, impeding long-distance, constant swimming behaviours [45]. Nonetheless, this is mostly speculative work and required additional research to be carried out to confirm these hypotheses. Furthermore, it was reported that exposure to pharmaceutical doses of gemfibrozil significantly reduced the concentration of n-3 fatty acids, which is likely to have direct repercussions on the nutritional quality of the fish [45,91]. Gagné et al. [92] further corroborated the presence of gemfibrozil in municipal wastewater treatment plants effluent in Canada, once again emphasizing the importance of studying the effects of emergent contaminants on the health of both wild and cultured fishes. However, the authors classified this specific drug as of little-to-no concern to fish, as they observed minimal toxicity of this pharmaceutical in rainbow trout hepatocytes. This is in accordance with previously published findings, also indicating that gemfibrozil had no significant effect on the hepatocytes of O. mykiss [93].
On the other hand, Donohue et al. [93] described a significant increase in bioindicators for oxidative stress on hepatocytes, caused by clofibric acid, the metabolite of clofibrate. Similarly, Triebskorn et al. [94] reported a moderate reaction of the liver of rainbow trout to exposure to clofibric acid, including dilation of blood vessels, and no significant reaction in the kidney of the exposed fish. However, results indicated a more severe response from the gills of O. mykiss to this pollutant, as the authors described epithelial lifting, increased cell production (hyperplasia) and growth (hypertrophy) of mucus cells. Nonetheless, these reactions are non-specific, as they tend to occur when the gills enter into contact with waterborne contaminants and are often regarded as a measure to preserve ion balance [95]. In addition, through an experiment focusing on the exposure of primary hepatocytes of rainbow trout to clofibric acid alone and combined with female sex hormone, it was demonstrated that this pharmaceutical is not a significant PPAR activator in hepatocytes in this species. In addition, neither lipid metabolism nor estrogenic activity resulted altered from this exposure [96].
The available information on the impact that exposure to lipid-lowering compounds has on the European eel (Anguilla anguilla) is extremely limited. According to Lyssimachou et al. [69], gemfibrozil is not a significant PPARα activator in this species. Nonetheless, injections of gemfibrozil significantly decreased the activity of certain pathways directly linked to cytochrome P450, which are responsible for the metabolism of xenobiotics, as well as the oxidation of fatty acids. This could indicate a tampered ability of the fish to efficiently extract energy from their food source, and to develop and function efficiently [97], having potentially serious implications for aquaculture production.
In zebrafish, chronic exposure to low concentrations of gemfibrozil can have transgenerational effects [77]. In adults, reproductive ability is reportedly altered, accompanied by histological changes in ovaries and kidneys. Furthermore, adult zebrafish chronically exposed to 500 and 10,000 ng/L of this pharmaceutical displayed a decrease in reproductive output, with atretic oocytes and altered kidney histology [67,68]. On the other hand, unexposed offspring of parents exposed to this contaminant display sex-dependent alterations in courtship activity and swimming behaviour, and abnormal sperm morphology, significantly affecting their fecundity and breeding success [70], although this seems limited to the first generation of offspring only [77].
Similarly, in a long-term exposure (i.e., 155 days) experiment with Japanese Medaka (Oryzias latipes), a significant decrease in fecundity was observed, tied with significant decreases in both testosterone levels in female fish and hatchability of F1 fish. Moreover, the expression of genes related to oestrogen receptors and vitellogenin was increased in both gonads and livers [78]. This suggests that, in some species, gemfibrozil can act as an endocrine disruptor, affecting reproduction and ontogeny. In male individuals of the same species, short-term exposure to gemfibrozil lowered levels of 17β-estradiol, 11-ketotestosterone and plasma cholesterol [78]. The authors argued that the endocrine disruption observed as a consequence of the challenge might be directly linked to the hypocholesterolaemic nature of the medicine [78], altering the reproductive success of individuals, and having varying effects depending on the exposure duration.
The inconsistency in the published results is likely linked to the dependence of the gemfibrozil’s mechanisms of toxicity to exposure duration, administration method, species and life stage, as well as organ analysed. However, it appears as if exposure to gemfibrozil may have strong effects on reproduction (e.g., fecundity, gametogenesis), excretion of exogenous substances (i.e., detoxification process), lipid metabolism and the overall homeostasis. In addition, the presence of this pollutant in aquatic systems is a potential factor affecting larval development, leading to ontogenetic and behavioural anomalies in both adults and larval stages, and, to some extent, in the subsequent generations.

3.1.2. Clofibrate and Clofibric Acid

Clofibric acid, the main metabolite of clofibrate, is a PPAR-α agonist that increases beta-oxidation and decreases triglyceride secretion in humans, but it is known to affect aquatic organisms [60,86]. Even though some of the previous literature investigated the consequences of exposure to clofibrate itself, a major part of the studies hereby considered focused on the effects of clofibric acid (Table 1).
In grass carp (Ctenopharyngodon idellal), it was observed that clofibrate decreased triacylglycerol, plasma cholesterol and overall lipid concentrations. Similarly, it increased hepatic enzymes involved in the synthesis of fatty acids, as well as lipoprotein lipase expression. Furthermore, a decrease in lipogenic enzymes, previously increased through feeding with a high carbohydrate diet, was observed [81]. This suggests that clofibrate, like gemfibrozil, can act as a hypolipidemic agent and alter the lipid metabolism of some fishes.
In zebrafish, exposure to clofibrate through ingestion leads to a modulation of genes such as fatty acid-binding proteins (fabp) and acyl-CoA oxidase 1 (acox1), a marker of PPAR-α activation in many tissues. Nonetheless, this process proved to be tissue dependent. Clofibrate is potentially unable to cross the blood–brain barrier of zebrafish, hence the lack of significant alterations in the transcription of acox1 in the brain [80]. In zebrafish larvae, the exposed group displayed shorter body lengths compared to the control group, in addition to changes in morphological characteristics and lethargic behaviour. When fed with this compound, an upregulation of peroxisomes in liver and heart mitochondria in was observed [81], suggesting that waterborne exposure to clofibrate affects both the metabolism of adults and larvae, and may cause strong alterations in the development of embryos, leading to severe deformities and behavioural changes. On the other hand, it has been reported that clofibric acid can cause behavioural and metabolic alterations in adults and larvae of D. rerio both after acute and chronic exposures but does not act as an endocrine disruptor. This drug can influence the enzymatic activities involved in counteracting oxidative stress, like superoxidase dismutase (SOD), CAT and GPx, although the level of alterations depends on the pollutant’s concentration. In addition, it has been found that this compound may cause alterations in biotransformation enzymes, as well as in lipid peroxidation levels [88]. Zebrafish embryos have a high potential for metabolizing xenobiotics, involving a variety of enzymatic activities, and generating derivate products such as clofibric acid [87]. It has been previously stated that a lifelong exposure of zebrafish to clofibric acid significantly affected the reproduction, spermatogenesis, growth and gene expression of fabp, in a transgenerational manner [86]. It was determined that the effects are dependent on the individual’s sex, and on the organ considered, and can express inter-generational variations, influencing fecundity, mortality and the occurrence of skeletal deformities.
With the purpose of investigating key enzymatic activities with involvement in reproduction, an in vitro study in common carp (Cyprinus carpio) showed that clofibrate had an inhibitory effect upon CYP17, related to a synthesis of active androgens on gonads [79]. In an experiment challenging C. carpio the results showed alterations in haematological parameters (decreased red blood cells count and increased white blood cell counts), biochemical endpoints (increased levels of glucose and proteins), ion regulation (decreased plasmatic sodium, increased Na+/K+ ATPase in gills) and on enzymatic responses (i.e., Lactate dehydrogenase, LDH; glutamic-oxaloacetic transaminase, GOT; and glutamate pyruvate transaminase, GPT) [82].
In a different study from the same research group, Cirrhinus mrigala was exposed to the same concentrations resulting in alterations in enzymatic parameters, such as GOT and GPT after short-term exposure, and changes in the ion homeostasis of gills (Na+/K+ ATPase activity) after both short- and long-term exposures to this clofibric acid [83]. On the same species, clofibric acid influenced biochemical parameters, causing an increase in plasmatic glucose and Na+, and K+, as well as a decrease in plasma protein and Cl following short-term exposures. On the other hand, long-term exposure to this contaminant lead to increased levels of plasmatic Na+ and Cl and oscillations in the levels of plasma glucose, protein and K+ [84]. Through a similar experiment, the authors also reported changes in thyroid hormones in the plasma of C. mrigala [85].
The published literature focusing on the effects of these lipid-regulating agents on S. salar is extremely scarce. Rørvik et al. [98], briefly described a significant decrease in lipid content in Atlantic salmon following ingestion of a feed complemented with clofibrate when compared to a control group.
Overall, these studies suggest, to a certain extent, inter-species variation regarding the effects of clofibric acid. However, the consequences of either chronic or acute exposure to this pharmaceutical often include strong alterations in lipid metabolism and homeostasis, ontogenetic disturbances, and effects on behaviour, haematological parameters and the detoxification process. Furthermore, and similarly to gemfibrozil, clofibric acid has been described as being, to a certain extent, an endocrine disruptor, and as having effects on fish displaying inter-generational and sex-dependent differences. Finally, it appears evident that rising environmental concentrations of this pollutant may have serious implications for the health and welfare of wild fish populations.

3.1.3. Bezafibrate

Despite being one of the first lipid regulators to be introduced in the pharmaceutical market, there are not many studies investigating the effects of bezafibrate on aquatic organisms when compared to other fibrates, such as gemfibrozil and clofibrate, or its metabolite, clofibric acid. To the best of the authors’ knowledge, the only article available that investigates the effects of this compound reported that exposure to bezafibrate through diet can act like an endocrine disruptor in male zebrafish. This drug can lower plasma cholesterol, decrease 11-ketotestosterone (11-KT), and induce changes at the histological and molecular level in the testis. In addition, the expression of a variety of genes was affected, being either up- or down-regulated depending on exposure time (Table 1). Therefore, bezafibrate affects the gonadal steroidogenesis and spermatogenesis of this organism, consequently having strong implications on the reproduction of the species and affecting it at the population level [89].

3.1.4. Overall Effects of Fibrates in Fishes

It is clear from the literature reviewed that the effects of fibrates in fishes depend strongly upon the still unresolved influence of species-specific physiologies and lifecycles, developmental stage, dosage differences across studies and the amount of time exposed to environmentally relevant concentrations of fibrates. To date, gemfibrozil seems to be the focus of most studies, but overall, the main deleterious effects of fibrates on fish physiology seem to be restricted primarily to alterations in cell and tissue lipid metabolism. This, in turn, may affect the timing and functionality of the reproductive axis and vitellogenesis and may result in abnormalities during the larval development, affecting normal swimming, and perhaps foraging, performance. From the data reviewed, it is not clear if these effects impair the immune responses in adults, but the few reports that indicate alterations in the expression of genes related to inflammation processes suggest that fibrates do not damage long-term immune reactivity. The antioxidant response associated with exposure to fibrates seems, in this sense, mostly related to aerobic metabolic outbursts as a byproduct of lipid oxidative alterations rather than to energy-consuming inflammatory responses. However, this may not be the case in cultured fishes, even in more advanced RAS systems, fed with lipid-enriched formulations, prone to elicit oxidative stress and inflammatory responses [99].
It seems safe to assume that a short-life antioxidant response may dysregulate the metabolic budget during development but not the lifelong (adaptive) performance of adults. However, it is particularly worrying that some of the effects on lipid metabolism can be transgenerational as mentioned in zebrafish studies [77]. Although chronic exposures are hard to reproduce in non-model, free-living species, in fishes, the long-term impact of stressors (including pharmaceuticals, endocrine disruptors and persistent changes in physical variables such as temperature), at the population level seems to rely, partly, on the transgenerational persistence of stress-related disruptive effects in the thyroidal-, estrogenic/androgenic- and growth-related axis [100,101,102,103] all of them highly sensitive to alterations in the metabolism of lipids. Moreover, sex plasticity in fishes depends on the concerted influences of lipid-, temperature-, seasonality- and stress-responsive gene pathways regulating gonadal maturation and fate [104,105,106]. Pharmaceutical-related alterations in the availability and amount of lipid substrates may result in an impairment of fertility output and biases sex ratios.
Of special concern are the changes of Na+/K+ ATPase transporters and activity resulting from exposure to fibrates in Cyprinus carpio and Cirrhinus mrigala [82,83]. In salmonids, these changes have still not been described, but if present, they may interfere with the osmoregulatory adjustments (smoltification) required for the migratory phase of diadromous fishes, in which branchial mitochondrial-rich ionocytes and the management of hepatic lipid stores determine the onset of smoltification [107], a complex process still understudied. In Atlantic salmon, prior to sea migration, lipid metabolism becomes less plastic to changes in lipid diets as demonstrated by the variations in the expression of genes linked to lipid uptake in the gut and hepatic lipogenesis and lipid transport [108]. These shifts in the metabolism of lipids are life-stage dependent, and constrict the growth rate that, in turn, acts as a trigger to undergo, or not, smoltification. In this sense, it would be worth studying the effects that exposure to fibrates during phases of major osmoregulatory and morphological changes may have on the obligate or facultative development of smoltification in migrant salmonids, a process that requires behavioural changes and modifications of swimming performance that may be influenced by the presence of gemfibrozil in the brain circuitry (see Figure 1).

3.2. Statins

Statins inhibit an enzyme called 3-hydroxy-3-methylglutaryl CoA reductase (HMGCR) that converts 3-hydroxy-3-methylglutaryl-CoA (HMG CoA) into the cholesterol precursor mevalonate. Thus, statins decrease cholesterol synthesis as they compete with HMG CoA for the active site of the enzyme, altering its conformation and inhibiting its function [109].
Although waterborne statins are expected to be bioavailable to fish due to their high input rate and persistence, there are indications that they will not bioaccumulate in organisms [25,50]. In addition to uptake via passive diffusion through membranes, statins may be taken up and excreted by membrane transporters. The activity and expression of membrane transporters in the gills of aquatic organisms may therefore influence their bioavailability by affecting their influx and/or efflux rates [110]. Therefore, the effects of this contaminant on an aquatic organism may vary greatly between species, or life stages within the same species (Table 2), but might be less severe than those observed for fibrates.

3.2.1. Atorvastatin

The prescription rate of statins continues to increase, mainly with the high sale of atorvastatin, often under the brand name “Lipitor” [44,124], representing a pioneering treatment for hypercholesterolemia [125] but increasing the concentration of this pollutant in aquatic systems.
Exposure to atorvastatin in rainbow trout was reported to alter genes involved in membrane transport, oxidative stress response, apoptosis and metabolism in gills at low concentrations. However, no effect was observed on genes involved in cholesterol biosynthesis or peroxisomal proliferation [111]. In contrast, an experiment focusing on primary rainbow trout hepatocytes showed that this environmental pollutant may alter lipid synthesis and reduce cholesterol biosynthesis [113].
In zebrafish, exposure to this emergent contaminant was directly linked to alterations in cholesterol metabolism, lipid regulation, and steroid production. Specifically, an observed reduction in cholesterol was associated with decreased levels of cortisol and sex steroids. Moreover, a reduction in triglyceride was associated with changes in mRNA levels involved with lipid regulation [44]. Additionally, exposure to this compound resulted in signs of skeletal muscle breakdown, indicating rhabdomyolysis, and strongly tampering with the welfare of male fish. Interestingly, this effect on skeletal muscle seemed to be gender-specific, as different response between male and female was observed [44].
Besides the effects observed on adult individuals, a strong, dose-dependent, myotoxic effect of this drug was reported in zebrafish larvae [112]. In addition, exposure to atorvastatin significantly reduced the response to tactile stimuli, and caused a decrease in enzyme activity, like citrate synthase (CS) and cytochrome oxidase (COX). This suggests involvement in metabolism in larvae, thus, affecting the early development of the fish, and, ultimately, success in adults. [112].
The effects of atorvastatin seem somewhat similar to those described for fibrates, with important implications for an individual’s development, and lipid, as well as overall metabolism, also acting as an endocrine disruptor.

3.2.2. Simvastatin

As with the other lipid-regulating agents mentioned before, exposure to simvastatin has been reported to have a variety of consequences in fish. For instance, exposure of fewer than 7 days was able to cause significant alterations in the antioxidant system of mosquitofish (Gambusia affinis) and related enzymatic activity, such as nuclear factor erythroid 2–related factor 2 (Nrf2) and mitogen-activated protein kinases (MAPK) [122]. Furthermore, the presence of this contaminant in aquatic environments is reportedly responsible for strong histological changes in hepatic cells of the same species [119,122]. Similarly, histological changes were reported to occur in Mugilogobius abei following short-term exposure to this statin. Furthermore, increased expression of genes related to xenobiotic resistance and detoxification (pregnane X receptor, PXR), including cytochromes P450 (i.e., CYP1A, CYP3A), has been shown to be somewhat induced by short-term exposure to this pollutant, concomitantly with alterations in enzymatic activity [120].
In zebrafish, it has been reported that chronic exposure to simvastatin at environmentally relevant concentrations, may have, in certain parameters, both dose-dependent, and sex-dependent consequences [118,121]. Indeed, following a 90-day waterborne exposure to this contaminant, the authors observed significant differences in the expression of genes responsible for energy metabolism (e.g., fatty acids synthesis and metabolism, glucose metabolism) in the brain [121]. Further analyses performed during the same experiment indicated significant alterations in the mevalonate pathway (e.g., directly involved in the synthesis of cholesterol), as well as a sex-dependent variation in body weight and body length at the highest exposure concentrations. However, a significant increase in reproductive success (fertilisation %) was observed at 200 ng/L, although this was followed by an increase in developmental anomalies. In addition, chronic exposure to simvastatin resulted in significant changes in cholesterol and triglyceride levels, although this response was not dose dependent. Moreover, the authors described a significant decrease in the heartbeat rate of embryos following parental exposure to 8 ng/L of this statin [118].
This agrees with what was previously described by Campos et al. [115], who also reported significantly slower heartbeat in zebrafish larvae as a consequence of exposure to simvastatin. Additionally, the authors reported decreased movement and significant structural alterations (e.g., changes in septum shape and size of somites, essential for locomotion in fish). The authors further speculate that exposure to this lipid-regulator may directly affect DNA replication, apoptosis, and muscle function [115]. Zebrafish embryos exposed to high concentrations of simvastatin experienced important mortality, and sever morphological anomalies, compared to low concentrations, which induced barely any mortality, and no morphological alterations [114]. In addition, it has been observed that exposure to this compound caused strong developmental issues, leading to abnormalities in the septum, somites and myofibrils and most likely tampering with the individual’s ability to swim, further corroborating previous findings [114].
On the other hand, Cunha et al. [117] described no significant effects of simvastatin exposure on cholesterol levels of D. rerio larvae, although the experimental methods differed with regard to the developmental stages challenged, and the concentrations tested, potentially indicating dose-dependent differences in the effects of simvastatin. Furthermore, the authors reported that exposure to this lipid-lowering agent may cause either up- or down-regulation of nuclear receptors (i.e., PPAR; PXR; constitutive androstane receptor, CAR; retinoid X receptors, RXR), as well as in aryl hydrocarbon receptor (AhR) in zebrafish, depending on the dose of simvastatin, and the exposure duration [117]. In that regard, the authors had previously reported that waterborne exposure to simvastatin could cause severe developmental abnormalities in zebrafish embryos, particularly tail deformities [116].
Moreover, the authors detected significant upregulation of ethoxyresorufin-O-deethylase (EROD), glutathione S-transferases (GST) and CAT as well as different ATP-binding cassette (ABC) transporters and cytochrome P450, indicating the induction of the cell detoxification process, and the related antioxidant procedure. Furthermore, the authors demonstrated the potential for simvastatin to interact with other chemical environmental pollutants, possibly having synergistic effects [116]. In contrast, Rebelo et al. [123] observed a significant decrease in SOD, GST and GPx, as well as thiobarbituric acid reactive substances (TBARS), in zebrafish embryos and juveniles following both acute and chronic exposures to simvastatin, indicating a reduction in antioxidant defences. In addition, significant behavioural changes were detected throughout these experiments, but no alterations in gonad development or sex determination were noted.

3.2.3. Overall Effects of Statins in Fishes

The caveats mentioned above for fibrates are equally relevant for the effects of statins in the physiology and behaviour of fishes, with an amendment: statins are even less studied and in fewer species. Due to the pleiotropic nature of the effects of statins [126], some unrelated to their ability to reduce cholesterol levels, it is hard to say if the intensity and scope of statins’ effects in fish are comparable to those described for fibrates beyond changes in lipid metabolism, abnormal development in larvae and oxidative stress. Either way, even conflicting results seem to indicate no major effects on long-term inflammatory outcomes. However, unexpected systemic alterations related to interferences with metabolic and mitochondrial oxidative pathways should not be ruled out. In mammals, statins have been linked to dose-dependent alterations in angiogenesis [127], and in this sense it would be worth further analysing their putative myotoxic effects during cardiovascular development in fishes [118], widening the focus beyond the classical zebrafish ecotoxicology resources [128] to cover non-model species.

4. Concluding Remarks

The manufacturing and use of lipid-regulating agents are constantly growing, and many of these compounds (e.g., statins and fibrates) are ubiquitous in aquatic systems. However, limited information is available on the effects of lipid-regulating agents on the most common species of fish in European RAS, with most research having focused on O. mykiss. Nonetheless, the published literature offers a worrying insight into the implications of having aquaculture fishes exposed to such hypocholesterolaemic agents. Indeed, it has been reported that fish might be continuously exposed to these contaminants, and that, although not all these compounds are particularly likely to bioaccumulate, prolonged contact may potentially lead to modified swimming, feeding and social behaviour in these organisms. Furthermore, it has been suggested that lipid-lowering agents may tamper with fish’s ability to reproduce and develop properly, causing deformities if exposed during early life stages, and therefore, greatly affecting the welfare of farmed animals. In addition, preliminary findings indicate that the nutritional quality of cultured fish may be significantly decreased by these contaminants of emerging concern, which may have serious implications for food security, as fish represents a considerable proportion of protein sources worldwide.
The low-level activation of inflammatory responses in fishes exposed to fibrates is intriguing and deserves more attention, considering the mutual influence between lipid and immune tissues [129]. Some regulators of key metabolic pathways, such as the mTOR anabolic signalling pathway, participate not only in the hepatic balance between lipolysis and lipogenesis, but also regulate the onset of inflammatory responses [130]. These pathways, together with the lipid tissue-dependent regulation of local immune responses in fishes [131] enforce the crosstalk between visceral adipose masses and immune cell trafficking and antigen presentation in fishes, similar to what has been observed in mammals.
More chronic studies are needed to ascertain the effects of lipid-regulating agents at the population level in free-living fishes, including those that, such as salmonids, have experienced several rounds of genome duplication, gained a duplicated set of genes related to lipid regulation and management, and, thus, may be more resilient to pharmaceutical-derived metabolic alterations [132]. Seasonal trends in lipid allocation for vitellogenesis and gonadal maturation, coupled with transgenerational alterations due to chronic exposure to environmental stressors have been described in three-spined sticklebacks (Gasterosteus aculeatus) in an attempt to dissect the complexity of reproductive allocation of resources during the breeding season [133]. These, and other species with context-dependent lifecycles deserve more studies concerning the synergistic or antagonistic effects of lipid regulators, acting alone or coupled with other stressors, on normal and maladaptive physiology of fishes. In this sense, the activation of the hypothalamic-pituitary-interrenal (HPI) stress axis in fishes responds not only to environmental cues but also to the presence of pharmaceuticals, xenobiotics and other contaminants that may bias the effect of HPI byproducts on the sex plasticity pathways [134], compromising the maintenance of healthy populations.
All these factors suggest that, if the environmental concentrations of statins and fibrates continue to rise, the effects on both wild and cultured organisms will intensify. Therefore, it appears that both the health and welfare of fishes in aquaculture systems will be at risk. Nonetheless, the effects of lipid-regulating agents seem to be somewhat dependent on a variety of factors (e.g., dose, exposure time, sex, and life stage). Thus, although the available literature allows, to a certain degree, for extrapolation of results on farmed species, additional research, with a particular focus on commercially important species (e.g., cultured in RAS), should be carried out to fully understand the implications for aquaculture production and, ultimately, global food security.

Author Contributions

Conceptualization, M.B., J.L. and M.T.; methodology, M.B. and J.L.; validation, L.T. and M.T.; investigation, M.B. and J.L.; writing—original draft preparation, M.B. and J.L.; writing—review and editing, all authors; visualization, J.C.B. and C.G.; supervision, M.T. and L.T.; project administration, M.T. and L.T.; funding acquisition, M.T., L.T., M.B. and J.L. declare equal contributions to all aspects of the present work. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the European Commission’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie (MSCA) grant agreement No 956481 through the RASOPTA Ph.D. fellowship. M.T. was supported through “Plan Nacional de Investigación” with reference PID2020-113221RB-I00 and a Ramon y Cajal contract with reference RYC2019-026841-I. L.T. was supported by the “Agencia Estatal de Investigación” with a “Plan Nacional de Investigación” with reference PID2020-117557RB-C21.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable. No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Thomaidis, N.S.; Asimakopoulos, A.G.; Bletsou, A.A. Emerging contaminants: A tutorial mini-review. Glob. Nest. J. 2012, 14, 72–79. [Google Scholar]
  2. Chinnaiyan, P.; Thampi, S.G.; Kumar, M.; Mini, K.M. Pharmaceutical products as emerging contaminant in water: Relevance for developing nations and identification of critical compounds for Indian environment. Environ. Monit. Assess. 2018, 190, 288. [Google Scholar] [CrossRef]
  3. Patel, M.; Kumar, R.; Kishor, K.; Mlsna, T.; Pittman, C.U.; Mohan, D. Pharmaceuticals of Emerging Concern in Aquatic Systems: Chemistry, Occurrence, Effects, and Removal Methods. Chem. Rev. 2019, 119, 3510–3673. [Google Scholar] [CrossRef] [Green Version]
  4. Gavrilescu, M.; Demnerova, K.; Aamand, J.; Agathos, S.; Fava, F. Emerging pollutants in the environment: Present and future challenges in biomonitoring, ecological risks and bioremediation. New Biotechnol. 2015, 32, 147–156. [Google Scholar] [CrossRef]
  5. Shi, Q.; Xiong, Y.; Kaur, P.; Sy, N.D.; Gan, J. Contaminants of emerging concerns in recycled water: Fate and risks in agroecosystems. Sci. Total Environ. 2022, 814, 152527. [Google Scholar] [CrossRef]
  6. Wang, J.; Wang, S. Removal of pharmaceuticals and personal care products (PPCPs) from wastewater: A review. J. Environ. Manag. 2016, 182, 620–640. [Google Scholar] [CrossRef]
  7. Bu, Q.; Wang, B.; Huang, J.; Deng, S.; Yu, G. Pharmaceuticals and personal care products in the aquatic environment in China: A review. J. Hazard. Mater. 2013, 262, 189–211. [Google Scholar] [CrossRef]
  8. Wilkinson, J.L.; Swinden, J.; Hooda, P.S.; Barker, J.; Barton, S. Markers of anthropogenic contamination: A validated method for quantification of pharmaceuticals, illicit drug metabolites, perfluorinated compounds, and plasticisers in sewage treatment effluent and rain runoff. Chemosphere 2016, 159, 638–646. [Google Scholar] [CrossRef] [Green Version]
  9. Hwang, J.-H.; Church, J.; Lee, S.-L.; Park, J.; Lee, W.H. Use of Microalgae for Advanced Wastewater Treatment and Sustainable Bioenergy Generation. Environ. Eng. Sci. 2016, 33, 11. [Google Scholar] [CrossRef] [Green Version]
  10. Jelic, A.; Gros, M.; Ginebreda, A.; Cespedes-Sánchez, R.; Ventura, F.; Petrovic, M.; Barceló, D. Occurrence, partition and removal of pharmaceuticals in sewage water and sludge during wastewater treatment. Water Res. 2011, 45, 1165–1176. [Google Scholar] [CrossRef]
  11. Luo, Y.; Guo, W.; Ngo, H.H.; Nghiem, L.D.; Hai, F.I.; Zhang, J.; Liang, S.; Wang, X.C. A review on the occurrence of micropollutants in the aquatic environment and their fate and removal during wastewater treatment. Sci. Total Environ. 2014, 473–474, 619–641. [Google Scholar] [CrossRef]
  12. Rathi, B.S.; Kumar, P.S.K.; Show, P.-L. A review on effective removal of emerging contaminants from aquatic systems: Current trends and scope for further research. J. Hazard. Mater. 2021, 409, 124413. [Google Scholar] [CrossRef]
  13. Rodriguez-Narvaez, O.M.; Peralta-Hernandez, J.M.; Goonetilleke, A.; Bandala, E.R. Treatment technologies for emerging contaminants in water: A review. J. Chem. Eng. 2017, 323, 361–380. [Google Scholar] [CrossRef] [Green Version]
  14. Fatta-Kassinos, D.; Hapeshi, E.; Achilleos, A.; Meric, S.; Gros, M.; Petrovic, M.; Barcelo, D. Existence of Pharmaceutical Compounds in Tertiary Treated Urban Wastewater that is Utilized for Reuse Applications. Water Resour Manag. 2011, 25, 1183–1193. [Google Scholar] [CrossRef]
  15. Salimi, M.; Esrafili, A.; Gholami, M.; Jafari, A.J.; Kalantary, R.R.; Farzadkia, M.; Kermani, M.; Sobhi, H.R. Contaminants of emerging concern: A review of new approach in AOP technologies. Environ. Monit. Assess. 2017, 189, 414. [Google Scholar] [CrossRef]
  16. Wilkinson, J.; Hooda, P.S.; Barker, J.; Barton, S.; Swinden, J. Occurrence, Fate and Transformation of Emerging Contaminants in Water: An Overarching Review of the Field. Environ. Pollut. 2017, 231, 954–970. [Google Scholar] [CrossRef] [Green Version]
  17. Fernandes, J.P.; Almeida, C.M.R.; Salgado, M.A.; Carvalho, M.F.; Mucha, A.P. Pharmaceutical Compounds in Aquatic Environments- Occurrence, Fate and Bioremediation Prospective. Toxics 2021, 9, 257. [Google Scholar] [CrossRef]
  18. López-Serna, R.; Jurado, A.; Vázquez-Suñé, E.; Carrera, J.; Petrovic, M.; Barceló, D. Occurrence of 95 pharmaceuticals and transformation products in urban groundwaters underlying the metropolis of Barcelona, Spain. Environ. Pollut. 2013, 174, 305–315. [Google Scholar] [CrossRef]
  19. Ribeiro, A.R.; Sures, B.; Schmidt, T.C. Cephalosporin Antibiotics in the Aquatic Environment: A Critical Review of Occurrence, Fate, Ecotoxicity and Removal Technologies. Environ. Pollut. 2018, 241, 1153–1166. [Google Scholar] [CrossRef]
  20. Rivera-Utrilla, J.; Sanchez-Polo, M.; Ferro-García, M.A.; PradosJoya, G.; Ocampo-Perez, R. Pharmaceuticals as Emerging Contaminants and Their Removal from Water. A Review. Chemosphere 2013, 93, 1268–1287. [Google Scholar] [CrossRef]
  21. Starling, M.C.V.; Amorim, C.C.; Leão, M.M.D. Occurrence, control and fate of contaminants of emerging concern in environmental compartments in Brazil. J. Hazard. Mater. 2019, 372, 17–36. [Google Scholar] [CrossRef]
  22. Bottoni, P.; Caroli, S. Presence of residues and metabolites of pharmaceuticals in environmental compartments, food commodities and workplaces: A review spanning the three-year period 2014–2016. Microchem. J. 2018, 136, 2–24. [Google Scholar] [CrossRef]
  23. Li, W.C. Occurrence, sources, and fate of pharmaceuticals in aquatic environment and soil. Environ. Pollut. 2014, 187, 193–201. [Google Scholar] [CrossRef] [PubMed]
  24. Verlicchi, P.; Aukidy, M.A.; Galletti, A.; Petrovic, M.; Barceló, D. Hospital effluent: Investigation of the concentrations and distribution of pharmaceuticals and environmental risk assessment. Sci. Total Environ. 2012, 430, 109–118. [Google Scholar] [CrossRef]
  25. Hernando, M.D.; Agueera, A.; Fernandez-Alba, A.R. LC-MS. Analysis and environmental risk of lipid regulators. Anal. Bioanal. Chem. 2007, 387, 1269–1285. [Google Scholar] [CrossRef]
  26. Ahmed, N.; Thompson, S.; Glaser, M. Global aquaculture productivity, environmental sustainability, and climate change adaptability. Environ. Manag. 2019, 63, 159–172. [Google Scholar] [CrossRef]
  27. Ahmed, N.; Turchini, G.M. Recirculating aquaculture systems (RAS): Environmental solution and climate change adaptation. J. Clean. Prod. 2021, 297, 126604. [Google Scholar] [CrossRef]
  28. Hall, S.J. Blue frontiers: Managing the environmental costs of aquaculture. WorldFish Center 2011. Available online: https://www.worldfishcenter.org/publication/blue-frontiers-managing-environmental-costs-aquaculture (accessed on 14 October 2022).
  29. Badiola, M.; Mendiola, D.; Bostock, J. Recirculating Aquaculture Systems (RAS) analysis: Main issues on management and future challenges. Aquac. Eng. 2012, 51, 26–35. [Google Scholar] [CrossRef] [Green Version]
  30. Bregnballe, J. A guide to recirculation aquaculture. An introduction to the new environmentally friendly and highly productive closed fish farming systems. FAO Eurofish Rep 2015, 100, 120. [Google Scholar]
  31. Murray, F.; Bostock, J.; Fletcher, D. Review of recirculation aquaculture system technologies and their commercial application. In Highlands and Islands Enterprise; University of Stirling Aquaculture: Stirling, UK, 2014. [Google Scholar]
  32. Malone, R. Recirculating Aquaculture Tank Production Systems; USDA, Southern Regional Aquaculture Center: Stoneville, MS, USA, 2013; Volume 12. [Google Scholar]
  33. Summerfelt, S.T.; Vinci, B.J. Better management practices for recirculating aquaculture systems. Environ. Best Manage. Pract. Aquacult. 2008, 10, 389–426. [Google Scholar] [CrossRef]
  34. Yanong, R.P.; Erlacher-Reid, C. Biosecurity in aquaculture, part 1: An overview. SRAC Publ. 2012, 4707, 522. [Google Scholar]
  35. Merino, G.; Barange, M.; Blanchard, J.L.; Harle, J.; Holmes, R.; Allen, I.; Allison, E.H.; Badjeck, M.C.; Dulvy, N.K.; Holt, J.; et al. Can marine fisheries and aquaculture meet fish demand from a growing human population in a changing climate? Glob. Environ. Chang. 2012, 22, 795–806. [Google Scholar] [CrossRef]
  36. Badiola, M.; Basurko, O.C.; Piedrahita, R.; Hundley, P.; Mendiola, D. Energy use in recirculating aquaculture systems (RAS): A review. Aquac. Eng. 2018, 81, 57–70. [Google Scholar] [CrossRef]
  37. European Commission. Directorate-General for Maritime Affairs and Fisheries. In The EU Fish Market: 2020 Edition; Publications Office of the European Union: Luxembourg, 2020. [Google Scholar] [CrossRef]
  38. Shen, Y.; Ma, K.; Yue, G.H. Status, challenges and trends of aquaculture in Singapore. Aquaculture 2021, 533, 736210. [Google Scholar] [CrossRef]
  39. Pahan, K. Lipid-lowering drugs. Cell. Mol. Life Sci. 2006, 63, 1165–1178. [Google Scholar] [CrossRef]
  40. Fang, Y.U.; Karnjanapiboonwong, A.; Chase, D.A.; Wang, J.; Morse, A.N.; Anderson, T.A. Occurrence, fate, and persistence of gemfibrozil in water and soil. Environ. Toxicol. Chem. 2012, 31, 550–555. [Google Scholar] [CrossRef]
  41. Ido, A.; Hiromori, Y.; Meng, L.; Usuda, H.; Nagase, H.; Yang, M.; Hu, J.; Nakanishi, T. Occurrence of fibrates and their metabolites in source and drinking water in Shanghai and Zhejiang, China. Sci. Rep. 2017, 7, 45931. [Google Scholar] [CrossRef] [Green Version]
  42. Zhang, K.; Zhao, Y.; Fent, K. Cardiovascular drugs and lipid regulating agents in surface waters at global scale: Occurrence, ecotoxicity and risk assessment. Sci. Total Environ. 2020, 729, 138770. [Google Scholar] [CrossRef]
  43. Mc Namara, K.; Alzubaidi, H.; Jackson, J.K. Cardiovascular disease as a leading cause of death: How are pharmacists getting involved? Integr. Pharm. Res. Pract. 2019, 8, 1–11. [Google Scholar] [CrossRef] [Green Version]
  44. Al-Habsi, A.A.; Massarsky, A.; Moon, T.W. Exposure to gemfibrozil and atorvastatin affects cholesterol metabolism and steroid production in zebrafish (Danio rerio). Comp. Biochem. Physiol. B Biochem. Mol. Biol. 2016, 199, 87–96. [Google Scholar] [CrossRef]
  45. Prindiville, J.S.; Mennigen, J.A.; Zamora, J.M.; Moon, T.W.; Weber, J.M. The fibrate drug gemfibrozil disrupts lipoprotein metabolism in rainbow trout. Toxicol. Appl. Pharmacol. 2011, 251, 201–208. [Google Scholar] [CrossRef]
  46. Staels, B.; Maes, M.; Zambon, A. Fibrates and future PPARα agonists in the treatment of cardiovascular disease. Nat. Clin. Pract. Cardiovasc. Med. 2008, 5, 542–553. [Google Scholar] [CrossRef]
  47. Barreto, A.; Luis, L.G.; Paíga, P.; Santos, L.H.M.L.M.; Delerue-Matos, C.; Soares, A.M.V.M.; Hylland, K.; Loureiro, S.; Oliveira, M. A multibiomarker approach highlights effects induced by the human pharmaceutical gemfibrozil to gilthead seabream Sparus aurata. Aquat. Toxicol. 2018, 200, 266–274. [Google Scholar] [CrossRef] [Green Version]
  48. Skolness, S.Y.; Durhan, E.J.; Jensen, K.M.; Kahl, M.D.; Makynen, E.A.; Villeneuve, D.L.; Ankley, G.T. Effects of gemfibrozil on lipid metabolism, steroidogenesis, and reproduction in the fathead minnow (Pimephales promelas). Environ. Toxicol. Chem. 2012, 3, 2615–2624. [Google Scholar] [CrossRef]
  49. Oliveira, M.; Franco, L.; Balasch, J.C.; Fierro-Castro, C.; Tvarijonaviciute, A.; Soares, A.M.V.M.; Tort, L.; Teles, M. Tools to assess effects of human pharmaceuticals in fish: A case study with gemfibrozil. Ecol. Indic. 2018, 95, 1100–1107. [Google Scholar] [CrossRef]
  50. Zhang, X.; Oakes, K.D.; Cui, S.; Bragg, L.; Servos, M.R.; Pawliszyn, J. Tissue-specific in vivo bioconcentration of pharmaceuticals in rainbow trout (Oncorhynchus mykiss) using space-resolved solid-phase microextraction. Environ. Sci. Technol. 2010, 44, 3417–3422. [Google Scholar] [CrossRef]
  51. Araujo, L.; Villa, N.; Camargo, N.; Bustos, M.; García, T.; Prieto, A.J. Persistence of gemfibrozil, naproxen and mefenamic acid in natural waters. Environ. Chem. Lett. 2011, 9, 13–18. [Google Scholar] [CrossRef]
  52. Gracia-Lor, E.; Sancho, J.V.; Serrano, R.; Hernández, F. Occurrence and removal of pharmaceuticals in wastewater treatment plants at the Spanish Mediterranean area of Valencia. Chemosphere 2012, 87, 453–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Andreozzi, R.; Raffaele, M.; Nicklas, P. Pharmaceuticals in STP effluents and their solar photodegradation in aquatic environment. Chemosphere 2003, 50, 1319–1330. [Google Scholar] [CrossRef] [PubMed]
  54. Gaw, S.; Thomas, K.V.; Hutchinson, T.H. Sources, impacts and trends of pharmaceuticals in the marine and coastal environment. Philos. Trans. R. Soc. B Biol. Sci. 2014, 369, 20130572. [Google Scholar] [CrossRef] [Green Version]
  55. Pereira, A.M.P.T.; Silva, L.J.G.; Laranjeiro, C.S.M.; Meisel, L.M.; Lino, C.M.; Pena, A. Human pharmaceuticals in Portuguese rivers: The impact of water scarcity in the environmental risk. Sci. Total Environ. 2017, 609, 1182–1191. [Google Scholar] [CrossRef]
  56. Loos, R.; Wollgast, J.; Huber, T.; Hanke, G. Polar herbicides, pharmaceutical products, perfluorooctanesulfonate (PFOS), perfluorooctanoate (PFOA), and nonylphenol and its carboxylates and ethoxylates in surface and tap waters around Lake Maggiore in Northern Italy. Anal. Bioanal. Chem. 2007, 387, 1469–1478. [Google Scholar] [CrossRef]
  57. Muñoz, I.; Lopez-Doval, J.C.; Ricart, M.; Villagrasa, M.; Brix, R.; Geiszinger, A.; Ginebreda, A.; Guaschm, H.; Alda, J.L.; Romani, A.M.; et al. Bridging levels of pharmaceuticals in river water with biological community structure in the Llobregat river basin (Northeast Spain). Environ. Toxicol. Chem. 2009, 28, 2706–2714. [Google Scholar] [CrossRef] [Green Version]
  58. Osorio, V.; Larrañaga, A.; Aceña, J.; Pérez, S.; Barceló, D. Concentration and risk of pharmaceuticals in freshwater systems are related to the population density and the livestock units in Iberian Rivers. Sci. Total Environ. 2016, 540, 267–277. [Google Scholar] [CrossRef] [Green Version]
  59. Mijangos, L.; Ziarrusta, H.; Ros, O.; Kortazar, L.; Fernández, L.A.; Olivares, M.; Zuloaga, O.; Prieto, A.; Etxebarria, N. Occurrence of emerging pollutants in estuaries of the Basque Country: Analysis of sources and distribution, and assessment of the environmental risk. Water Res. 2018, 147, 152–163. [Google Scholar] [CrossRef]
  60. Corcoran, J.; Winter, M.J.; Lange, A.; Cumming, R.; Owen, S.F.; Tyler, C.R. Effects of the lipid regulating drug clofibric acid on PPARα-regulated gene transcript levels in common carp (Cyprinus carpio) at pharmacological and environmental exposure levels. Aquat. Toxicol. 2015, 161, 127–137. [Google Scholar] [CrossRef] [Green Version]
  61. Fent, K.; Weston, A.A.; Caminada, D. Ecotoxicology of Human Pharmaceuticals. Aquat. Toxicol. 2006, 76, 122–159. [Google Scholar] [CrossRef]
  62. Miao, D.; Peng, J.; Wang, M.; Shao, S.; Wang, L.; Gao, S. Removal of atorvastatin in water mediated by CuFe2O4 activated peroxymonosulfate. Chem. Eng. J. 2018, 346, 110. [Google Scholar] [CrossRef]
  63. Nováková, L.; Šatínský, D.; Solich, P. HPLC Methods for the determination of simvastatin and atorvastatin. TRAC 2018, 27, 4. [Google Scholar] [CrossRef]
  64. Santos, L.H.M.L.M.; Araújo, A.N.; Fachini, A.; Pena, A.; Delerue-Matos, C.; Montenegroa, M.C.B.S.M. Ecotoxicological aspects related to the presence of pharmaceuticals in the aquatic environment. J. Hazard. Mater. 2010, 175, 45–49. [Google Scholar] [CrossRef] [Green Version]
  65. Miao, X.S.; Metcalfe, C.D. Determination of cholesterol-lowering statin drugs in aqueous samples using liquid chromatography–electrospray ionization tandem mass spectrometry. J. Chromatogr. A 2003, 998, 133–141. [Google Scholar] [CrossRef]
  66. Langford, K.; Thomas, K.V. Input of selected human pharmaceutical metabolites into the Norwegian aquatic environment. J. Environ. Monit. 2011, 13, 416–421. [Google Scholar] [CrossRef]
  67. Galus, M.; Jeyaranjaan, J.; Smith, E.; Li, H.; Metcalfe, C.; Wilson, J.Y. Chronic effects of exposure to a pharmaceutical mixture and municipal wastewater in zebrafish. Aquat.Toxicol. 2013, 132–133, 212–222. [Google Scholar] [CrossRef]
  68. Galus, M.; Kirischiana, N.; Higgins, S.; Purdya, J.; Chowa, J.; Rangaranjana, S.; Li, H.; Metcalfe, C.; Wilsona, J.Y. Chronic, low concentration exposure to pharmaceuticals impacts multiple organ systems in zebrafish. Aquat. Toxicol. 2013, 132–133, 200–211. [Google Scholar] [CrossRef]
  69. Lyssimachou, A.; Thibaut, R.; Gisbert, E.; Porte, C. Gemfibrozil modulates cytochrome P450 and peroxisome proliferation-inducible enzymes in the liver of the yellow European eel (Anguilla anguilla). Environ. Sci. Pollut. Res. Int. 2014, 21, 862–871. [Google Scholar] [CrossRef]
  70. Galus, M.; Rangarajan, S.; Lai, A.; Shaya, L.; Balshine, S.; Wilson, J.Y. Effects of chronic, parental pharmaceutical exposure on zebrafish (Danio rerio) offspring. Aquat. Toxicol. 2014, 151, 124–134. [Google Scholar] [CrossRef]
  71. Solé, M.; Fortuny, A.; Mañanós, E. Effects of selected xenobiotics on hepatic and plasmatic biomarkers in juveniles of Solea senegalensis. Environ. Res. 2014, 135, 227–235. [Google Scholar] [CrossRef]
  72. Teles, M.; Fierro-Castro, C.; Na-Phatthalung, P.; Tvarijonaviciute, A.; Soares, A.M.V.M.; Tort, L.; Oliveira, M. Evaluation of gemfibrozil effects on a marine fish (Sparus aurata) combining gene expression with conventional endocrine and biochemical endpoints. J. Hazard. Mater. 2016, 318, 600–607. [Google Scholar] [CrossRef]
  73. Henriques, J.F.; Almeida, A.R.; Andrade, T.; Koba, O.; Golovko, O.; Soares, A.M.; Oliveira, M.; Domingues, I. Effects of the lipid regulator drug gemfibrozil: A toxicological and behavioral perspective. Aquat. Toxicol. 2016, 170, 355–364. [Google Scholar] [CrossRef]
  74. Barreto, A.; Luisa, L.G.; Soares, A.M.V.M.; Paíga, P.; Santos, L.H.M.L.M.; Delerue-Matos, C.; Hylland, K.; Loureiro, S.; Oliveira, M. Genotoxicity of gemfibrozil in the gilthead seabream (Sparus aurata). Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2017, 821, 36–42. [Google Scholar] [CrossRef]
  75. Fraz, S.; Lee, A.H.; Wilson, J.Y. Gemfibrozil and carbamazepine decrease steroid production in zebrafish testes (Danio rerio). Aquat. Toxicol. 2018, 198, 1–9. [Google Scholar] [CrossRef]
  76. 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] [Green Version]
  77. Fraz, S.; Lee, A.H.; Pollard, S.; Srinivasan, K.; Vermani, A.; Wilson, J.Y. Parental gemfibrozil exposure impacts zebrafish F1 offspring, but not subsequent generations. Aquat. Toxicol. 2019, 212, 194–204. [Google Scholar] [CrossRef]
  78. Lee, G.; Lee, S.; Ha, N.; Kho, Y.; Park, K.; Kim, P.; Ahn, B.; Kim, S.; Choi, K. Effects of gemfibrozil on sex hormones and reproduction related performances of Oryzias latipes following long-term (155 d) and short-term (21 d) exposure. Ecotoxicol. Environ. Saf. 2019, 173, 174–181. [Google Scholar] [CrossRef]
  79. Fernandes, D.; Schnell, S.; Porte, S. Can pharmaceuticals interfere with the synthesis of active androgens in male fish? An in vitro study. Mar. Pollut. Bull. 2011, 62, 2250–2253. [Google Scholar] [CrossRef]
  80. Venkatachalam, A.B.; Santosh, P.L.; Eileen, M.D.W.; Jonathan, M.W. Tissue-specific differential induction of duplicated fatty acid-binding protein genes by the peroxisome proliferator, clofibrate, in zebrafish (Danio rerio). BMC Evol. 2012, 12, 112. [Google Scholar] [CrossRef] [Green Version]
  81. Guo, X.; Liang, X.F.; Fang, L.; Yuan, X.; Zhou, Y.; He, S.; Shen, D. Effects of lipid-lowering pharmaceutical clofibrate on lipid and lipoprotein metabolism of grass carp (Ctenopharyngodon idellal Val.) fed with the high non-protein energy diets. Fish Physiol. Biochem. 2015, 41, 331–343. [Google Scholar] [CrossRef]
  82. Saravanan, M.; Karthika, S.; Malarvizhi, A.; Ramesh, M. Ecotoxicological impacts of clofibric acid and diclofenac in common carp (Cyprinus carpio) fingerlings: Hematological, biochemical, ionoregulatory and enzymological responses. J. Hazard. Mater. 2011, 195, 188–194. [Google Scholar] [CrossRef]
  83. Saravanan, M.; Ramesh, M.; Petkam, R. Alteration in certain enzymological parameters of an Indian major carp, Cirrhinus mrigala exposed to short- and longterm exposure of clofibric acid and diclofenac. Fish Physiol. Biochem. 2013, 39, 1431–1440. [Google Scholar] [CrossRef]
  84. Saravanan, M.; Ramesh, M. Short and long-term effects of clofibric acid and diclofenac on certain biochemical and ionoregulatory responses in an Indian major carp, Cirrhinus mrigala. Chemosphere 2013, 93, 388–396. [Google Scholar] [CrossRef]
  85. Saravanana, M.; Hur, J.H.; Arul, N.; Ramesha, M. Toxicological effects of clofibric acid and diclofenac on plasma thyroid hormones of an Indian major carp, Cirrhinus mrigala during short and long-term exposures. Environ. Toxicol. Pharmacol. 2014, 38, 948–958. [Google Scholar] [CrossRef]
  86. Coimbra, A.M.; Peixoto, M.J.; Coelho, I.; Lacerda, R.; Carvalho, A.P.; Gesto, M.; Lyssimachou, A.; Lima, D.; Soares, J.; André, A.; et al. Chronic effects of clofibric acid in zebrafish (Danio rerio): A multigenerational study. Aquat. Toxicol. 2015, 160, 76–86. [Google Scholar] [CrossRef]
  87. Brox, S.; Seiwert, B.; Haase, N.; Küster, E.; Reemtsma, T. Metabolism of clofibric acid in zebrafish embryos (Danio rerio) as determined by liquid chromatography–high resolution–mass spectrometry. CBP Part C 2016, 185–186, 20–28. [Google Scholar] [CrossRef]
  88. Rebelo, D.; Correia, A.T.; Nunes, B. Acute and chronic effects of environmental realistic concentrations of clofibric acid in Danio rerio: Behaviour, oxidative stress, biotransformation and lipid peroxidation endpoints. Environ. Toxicol. Pharmacol. 2020, 80, 103468. [Google Scholar] [CrossRef]
  89. Velasco-Santamaríaa, M.Y.; Korsgaardb, B.; Madsenb, S.S.; Bjerregaard, P. Bezafibrate, a lipid-lowering pharmaceutical, as a potential endocrine disruptor in male zebrafish (Danio rerio). Aquat. Toxicol. 2011, 105, 107–118. [Google Scholar] [CrossRef]
  90. Barreto, A.; Luis, L.G.; Pinto, E.; Almeida, A.; Paíga, P.; Santos, L.H.M.L.M.; Delerue-Matos, C.; Trindade, T.; Soares, A.M.V.M.; Hylland, K.; et al. Effects and bioaccumulation of gold nanoparticles in the gilthead seabream (Sparus aurata)- Single and combined exposures with gemfibrozil. Chemosphere 2019, 215, 248–260. [Google Scholar] [CrossRef]
  91. Steffens, W.; Wirth, M. Freshwater fish-an important source of n-3 polyunsaturated fatty acids: A review. Fish Aquatic Sci. 2005, 13, 5–16. [Google Scholar]
  92. Gagné, F.; Blaise, C.; André, C. Occurrence of pharmaceutical products in a municipal effluent and toxicity to rainbow trout (Oncorhynchus mykiss) hepatocytes. Ecotoxicol. Environ. Saf. 2006, 64, 329–336. [Google Scholar] [CrossRef]
  93. Donohue, M.; Baldwin, L.A.; Leonard, D.A.; Kostecki, P.T.; Calabrese, E.J. Effect of hypolipidemic drugs gemfibrozil, ciprofibrate, and clofibric acid on peroxisomal β-oxidation in primary cultures of rainbow trout hepatocytes. Ecotoxicol. Environ. Saf. 1993, 26, 127–132. [Google Scholar] [CrossRef]
  94. Triebskorn, R.; Casper, H.; Scheil, V.; Schwaiger, J. Ultrastructural effects of pharmaceuticals (carbamazepine, clofibric acid, metoprolol, diclofenac) in rainbow trout (Oncorhynchus mykiss) and common carp (Cyprinus carpio). Anal. Bioanal. Chem. 2007, 387, 1405–1416. [Google Scholar] [CrossRef]
  95. Cerqueira, C.C.; Fernandes, M.N. Gill tissue recovery after copper exposure and blood parameter responses in the tropical fish Prochilodus scrofa. Ecotoxicol. Environ. Saf. 2002, 52, 83–91. [Google Scholar] [CrossRef]
  96. Sovadinová, I.; Liedtke, A.; Schirmer, A. Effects of clofibric acid alone and in combination with 17b-estradiol on mRNA abundance in primary hepatocytes isolated from rainbow trout. Toxicol. Vitr. 2014, 28, 1106–1116. [Google Scholar] [CrossRef]
  97. Olivares-Rubio, H.F.; Vega-López, A. Fatty acid metabolism in fish species as a biomarker for environmental monitoring. Environ. Pollut. 2016, 218, 297–312. [Google Scholar] [CrossRef]
  98. Rørvik, K.A.; Alne, H.; Gaarder, M.; Ruyter, B.; Måseide, N.P.; Jakobsen, J.V.; Berge, R.K.; Sigholt, T.; Thomassen, M.S. Does the capacity for energy utilization affect the survival of post-smolt Atlantic salmon, Salmo salar L.; during natural outbreaks of infectious pancreatic necrosis? J. Fish Dis. 2007, 30, 399–409. [Google Scholar] [CrossRef]
  99. Song, F.; Qin, Y.; Geng, H.; He, C.; Yang, P.; Wang, W.; Chen, Y. Transcriptome analysis reveals the effects of dietary lipid level on growth performance and immune response in golden pompano (Trachinotus ovatus). Aquaculture 2023, 563, 738959. [Google Scholar] [CrossRef]
  100. Hamid, N.; Junaid, M.; Pei, D.S. Combined toxicity of endocrine-disrupting chemicals: A review. Ecotoxicol. Environ. Saf. 2021, 215, 112136. [Google Scholar] [CrossRef]
  101. Petitjean, Q.; Jean, S.; Gandar, A.; Côte, J.; Laffaille, P.; Jacquin, L. Stress responses in fish: From molecular to evolutionary processes. Sci. Total Environ. 2019, 684, 371–380. [Google Scholar] [CrossRef]
  102. Penney, C.M.; Tabh, J.K.R.; Wilson, C.C.; Burness, G. Within-Generation and Transgenerational Plasticity of a Temperate Salmonid in Response to Thermal Acclimation and Acute Temperature Stress. Physiol. Biochem. Zool. 2022, 95, 484–499. [Google Scholar] [CrossRef]
  103. Vera-Chang, M.N.; Moon, T.W.; Trudeau, V.L. Cortisol disruption and transgenerational alteration in the expression of stress-related genes in zebrafish larvae following fluoxetine exposure. Toxicol. Appl. Pharmacol. 2019, 382, 114742. [Google Scholar] [CrossRef]
  104. Fukada, H.; Yabuki, H.; Miura, C.; Miura, T.; Kato, K. Regulation of lipid metabolism by water temperature and photoperiod in yellowtail Seriola quinqueradiata. Fish. Sci. 2023. [Google Scholar] [CrossRef]
  105. Geffroy, B.; Wedekind, C. Effects of global warming on sex ratios in fishes. J. Fish. Biol. 2020, 97, 596–606. [Google Scholar] [CrossRef]
  106. Li, D.L.; Huang, Y.J.; Gao, S.; Chen, L.Q.; Zhang, M.L.; Du, Z.Y. Sex-specific alterations of lipid metabolism in zebrafish exposed to polychlorinated biphenyls. Chemosphere 2019, 221, 768–777. [Google Scholar] [CrossRef]
  107. McCormick, S.D.; O’Dea, M.F.; Moeckel, A.M.; Björnsson, B.T. Endocrine and physiological changes in Atlantic salmon smolts following hatchery release. Aquaculture 2003, 222, 45–57. [Google Scholar] [CrossRef]
  108. Gillard, G.; Harvey, T.N.; Gjuvsland, A.; Jin, Y.; Thomassen, M.; Lien, S.; Leaver, M.; Torgersen, J.S.; Hvidsten, T.R.; Vik, J.O.; et al. Life-stage-associated remodelling of lipid metabolism regulation in Atlantic salmon. Mol Ecol. 2018, 27, 1200–1213. [Google Scholar] [CrossRef] [Green Version]
  109. Santos, M.M.; Ruivo, R.; Lopes-Marques, M.; Torres, T.; de los Santos, C.B.; Castro, L.F.C.; Neuparth, T. Statins: An undesirable class of aquatic contaminants? Aquat. Toxicol. 2016, 174, 1–9. [Google Scholar] [CrossRef]
  110. Yamasaki, D.; Nakamura, T.; Okamura, N.; Kokudai, M.; Inui, N.; Takeuchi, K.; Watanabe, H.; Hirai, M.; Okumura, K.; Sakaeda, T. Effects of acid and lactone forms of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors on the induction of MDR1 expression and function in LS180 cells. Eur. J. Pharm. Sci. 2009, 37, 126–132. [Google Scholar] [CrossRef]
  111. Ellesat, K.S.; Holth, T.F.; Wojewodzic, M.W.; Hylland, K. Atorvastatin up-regulate toxicologically relevant genes in rainbow trout gills. Ecotoxicology 2012, 21, 1841–1856. [Google Scholar] [CrossRef]
  112. Pasha, R.; Moon, T.W. Coenzyme Q10 protects against statin-induced myotoxicity in zebrafish larvae (Danio rerio). Environ. Toxicol. Pharmacol. 2017, 52, 150–160. [Google Scholar] [CrossRef]
  113. Al-Habsi, A.A.; Massarsky, A.; Moon, T.W. Atorvastatin alters gene expression and cholesterol synthesis in primary rainbow trout (Oncorhynchus mykiss) hepatocytes. Comp. Biochem. Physiol. B Biochem. Mol. Biol. 2018, 224, 262–269. [Google Scholar] [CrossRef]
  114. Campos, L.M.; Rios, E.A.; Midlej, V.; Atella, G.C.; Herculano-Houzel, S.; Benchimol, M.; Mermelstein, C.; Costa, M.L. Structural analysis of alterations in zebrafish muscle differentiation induced by simvastatin and their recovery with cholesterol. J. Histochem. Cytochem. 2015, 63, 427–437. [Google Scholar] [CrossRef] [Green Version]
  115. Campos, L.M.; Rios, E.A.; Guapyassu, L.; Midlej, V.; Atella, G.C.; Herculano-Houzel, S.; Benchimol, M.; Mermelstein, C.; Costa, M.L. Alterations in zebrafish development induced by simvastatin: Comprehensive morphological and physiological study, focusing on muscle. Exp. Biol. Med. 2016, 241, 1950–1960. [Google Scholar] [CrossRef] [Green Version]
  116. Cunha, V.; Santos, M.M.; Moradas-Ferreira, P.; Ferreira, M. Simvastatin effects on detoxification mechanisms in Danio rerio embryos. Environ. Sci. Pollut. Res. Int. 2016, 23, 10615–10629. [Google Scholar] [CrossRef]
  117. Cunha, V.; Santos, M.M.; Moradas-Ferreira, P.; Castro, L.F.C.; Ferreira, M. Simvastatin modulates gene expression of key receptors in zebrafish embryos. J. Toxicol. Environ. Health Part A 2017, 80, 465–476. [Google Scholar] [CrossRef] [PubMed]
  118. Barros, S.; Montes, R.; Quintana, J.B.; Rodil, R.; André, A.; Capitão, A.; Soares, J.; Santos, M.M.; Neuparth, T. Chronic environmentally relevant levels of simvastatin disrupt embryonic development, biochemical and molecular responses in zebrafish (Danio rerio). Aquat. Toxicol. 2018, 201, 47–57. [Google Scholar] [CrossRef]
  119. Bao, S.; Nie, X.; Liu, Y.; Wang, C.; Liu, S. Response of PXR signaling pathway to simvastatin exposure in mosquitofish (Gambusia affinis) and its histological changes. Ecotoxicol. Environ. Saf. 2018, 154, 228–236. [Google Scholar] [CrossRef]
  120. Wang, C.; Ku, P.; Nie, X.; Bao, S.; Wang, Z.; Li, K. Effects of simvastatin on the PXR signaling pathway and the liver histology in Mugilogobius abei. Sci. Total Environ. 2019, 651, 399–409. [Google Scholar] [CrossRef]
  121. Barros, S.; Coimbra, A.M.; Alves, N.; Pinheiro, M.; Quintana, J.B.; Santos, M.M.; Neuparth, T. Chronic exposure to environmentally relevant levels of simvastatin disrupts zebrafish brain gene signaling involved in energy metabolism. J. Toxicol. Environ. Health Part A 2020, 83, 113–125. [Google Scholar] [CrossRef]
  122. Bao, S.; Lin, J.; Xie, M.; Wang, C.; Nie, X. Simvastatin affects Nrf2/MAPK signaling pathway and hepatic histological structure change in Gambusia affinis. Chemosphere 2021, 269, 128725. [Google Scholar] [CrossRef]
  123. Rebelo, D.; Correia, A.T.; Nunes, B. Acute and chronic effects of environmental realistic concentrations of simvastatin in Danio rerio: Evidences of oxidative alterations and endocrine disruptive activity. Environ. Toxicol. Pharmacol. 2021, 81, 103522. [Google Scholar] [CrossRef]
  124. Cavallucci, S. Top 200: What’s topping the charts in prescription drugs this year. Pharm. Pract. 2007, 27, 25–32. [Google Scholar]
  125. Jackevicius, C.A.; Tu, J.V.; Krumholz, H.M. Statins: Is it safe and effective to use generic “equivalents”? Can. J. Cardiol. 2013, 29, 408–410. [Google Scholar] [CrossRef]
  126. Zahedipour, F.; Butler, A.E.; Rizzo, M.; Sahebkar, A. Statins and angiogenesis in non-cardiovascular diseases. Drug Discov. Today. 2022, 27, 103320. [Google Scholar] [CrossRef]
  127. Thompson, P.D.; Panza, G.; Zaleski, A.; Taylor, B. Statin-associated side effects. J. Am. Coll. Cardiol. 2016, 67, 2395–2410. [Google Scholar] [CrossRef]
  128. Dubińska-Magiera, M.; Migocka-Patrzałek, M.; Lewandowski, D.; Daczewska, M.; Jagla, K. Zebrafish as a Model for the Study of Lipid-Lowering Drug-Induced Myopathies. Int. J. Mol. Sci. 2021, 22, 5654. [Google Scholar] [CrossRef]
  129. Veenstra, K.A.; Wangkahart, E.; Wang, T.; Tubbs, L.; Ben Arous, J.; Secombes, C.J. Rainbow trout (Oncorhynchus mykiss) adipose tissue undergoes major changes in immune gene expression following bacterial infection or stimulation with pro-inflammatory molecules. Dev. Comp. Immunol. 2018, 81, 83–94. [Google Scholar] [CrossRef]
  130. Xu, J.; Yao, X.; Li, X.; Xie, S.; Chi, S.; Zhang, S.; Cao, J.; Tan, B. Farnesoid X receptor regulates PI3K/AKT/mTOR signaling pathway, lipid metabolism, and immune response in hybrid grouper. Fish Physiol. Biochem. 2022, 48, 1521–1538. [Google Scholar] [CrossRef]
  131. Veenstra, K.A.; Alnabulsi, A.; Tubbs, L.; Ben Arous, J.; Secombes, C.J. Immunohistochemical examination of immune cells in adipose tissue of rainbow trout (Oncorhynchus mykiss) following intraperitoneal vaccination. Fish Shellfish Immunol. 2019, 87, 559–564. [Google Scholar] [CrossRef]
  132. Valenzuela-Muñoz, V.; Gallardo-Escárate, C.; Benavente, B.P.; Valenzuela-Miranda, D.; Núñez-Acuña, G.; Escobar-Sepulveda, H.; Váldes, J.A. Whole-Genome Transcript Expression Profiling Reveals Novel Insights into Transposon Genes and Non-Coding RNAs during Atlantic Salmon Seawater Adaptation. Biology 2021, 11, 1. [Google Scholar] [CrossRef]
  133. Magierecka, A.; Aristeidou, A.; Papaevripidou, M.; Gibson, J.K.; Sloman, K.A.; Metcalfe, N.B. Timing of reproduction modifies transgenerational effects of chronic exposure to stressors in an annual vertebrate. Proc. Biol. Sci. 2022, 289, 20221462. [Google Scholar] [CrossRef]
  134. Hattori, R.S.; Castañeda-Cortés, D.C.; Arias Padilla, L.F.; Strobl-Mazzulla, P.H.; Fernandino, J.I. Activation of stress response axis as a key process in environment-induced sex plasticity in fish. Cell. Mol. Life Sci. 2020, 77, 4223–4236. [Google Scholar] [CrossRef]
Figure 1. Putative effects of fibrates and statins in fish. Scheme of the effects of acute or chronic exposure to lipid regulators (see text for details) on major organs and tissues of fish.
Figure 1. Putative effects of fibrates and statins in fish. Scheme of the effects of acute or chronic exposure to lipid regulators (see text for details) on major organs and tissues of fish.
Animals 13 00792 g001
Table 1. The published literature on fibrates (gemfibrozil, clofibrate, clofibric acid and bezafibrate) and their main effects on fish.
Table 1. The published literature on fibrates (gemfibrozil, clofibrate, clofibric acid and bezafibrate) and their main effects on fish.
SpeciesDrugExposure PeriodConcentrationExposure RouteSample TypeMain EffectsReference (2011–2022)
Oncorhynchus mykissGEM15 d100 mg/kgInjection (IP)Plasma, liver, adipose tissue, red muscle and white muscle↓ lipoprotein concentration ↓ n-3 fatty acids[45]
Pimephales mykissGEM2, 8 and 21 d1, 5, 15,
600, 1500 µg/L
WaterborneBlood, liver, gonads, brain and pituitary↓ plasma cholesterol at 600 µg/L in both sexes
↑ plasma Chol at 1500 µg/L in males
pparα after 8 d at 600 µg/L in males
lpl after 2 d at 600 µg/L in males
ldlr, fasn, apoeb, apoa1 in males
fasn, apoeb in females
apoa1 at 2 d in females
apoa1 at 21 d in females
[48]
Danio rerioGEM6 w0.5 and 10 μg/LWaterborne Plasma, gonads and whole body↓ embryo viability
↓ survival in exposed embryos from non-exposed parents
↑ yolk-sac oedema
↑ skeletal deformities
↑ histopathological score
↑ regressive changes in kidney tubule
[67]
D. rerioGEM6 w0.5 and 10 μg/LWaterborne Plasma, gonads and whole body↓ embryo production
↑ embryo mortality after direct exposure
↑ histopathological score
↑ regressive changes in kidney tubule
↑ regressive changes in liver
↑ irregular oocytes in females
[68]
Anguilla anguillaGEM24 and 96 h0.1, 1, 10, 2, 20, 200 μg/g BWInjection (IP)Blood and liver↓ body weight
↓ EROD activity (CYP1A-like)
↓ BFCOD activity (CYP3A-like)
↓ CYP2K-like activity
↑ Hepatic AOX and CAT
[69]
D. rerioGEM6 w0, 10 g/LWaterborneEmbryos↓ breeding success
↓ courtship time
↑ courtship frequency
↓ sperm size
↑ sperm speed
[70]
Solea senegalensisGEM2 d1 mg/kg BWInjection (IP)Blood and liver↑CYP450-related activities
↑ UDPGT
↓ hepatic CAT, GPx and GST.
[71]
D. rerioGEM30 d16 µg/g BWDietLiver and brain↓ Chol
↓ TG, and E
↓ T in females
↑ PPARα in females
↑ PPARϒ
↑ liver SREBP-2, CYP3A65, atrogin1
[44]
Sparus aurataGEM96 h0, 1.5, 15, 150, 1500 and 15,000 μg/LWaterborne Liver and blood↑ APOA1, LPL
↑ IL-1β, TNF-α, and CASP3 genes at 15 µg/L.
↑ cortisol
[72]
D. rerioGEM120 and 144 h0, 1.5, 3 and 6 mg WaterborneEmbryos↓ embryo and larvae survival
↑ yolk-sac deformities and oedema
↓ locomotion
[73]
S. aurataGEM96 h1.5, 15, 150, 1500 and 15,000 μg/LWaterborne Blood↑ DNA damage
↑cortisol at 1.5 μg L−1
[74]
D. rerioGEM42 d/67 d0 and 10 μg/LWaterborneBlood, gonads, and whole bodies↓ reproductive output
↓ whole body, plasma and testicular 11-KT
[75]
D. rerioGEM6 w0 or 10 µg/LCell cultureGonads↓ sexual differentiation in F1 offspring depending on which parent was exposed[76]
S. aurataGEM96 h0, 1.5, 15, 150, 1500 and 15,000 μg/LWaterborne Gills and head kidney↑ TOS, GPx1, and IL1β in gills and head kidney[49]
S. aurataGEM96 h1.5, 15, 150, 1500 and 15,000 μg/L WaterborneLiver, gills, brain and muscle↓ locomotion capacity.
↑ CAT, GR, GPx
↑ TBARS
↓ LPO
↑ activity of enzymes of antioxidant defence activity (15–15,000 μg L−1).
[47]
D. rerioGEM6 w10 μg/LWaterSperm, whole fish, blood and organs↓ embryo production
↓ 11-KT
↓ courtship duration, ↓ sperm size
↑ sperm speed
effects in unexposed F1, but not subsequent generations
[77]
Oryzias latipesGEM155 d (embryos) and 1 d (adults)0, 0.04, 0.4, 3.7 and 40 mg/LWaterborne Blood, liver and gonads↓ T at long-term exposure
↓ Hatching rate in F1
↑ ER and VTG
↓ E2 and 11-KT at short-term exposure
↓ Chol in males
[78]
Cyprinus carpioGEM, CLO and CA-1 mMIn vitroGonadsGEM and CLO: ↓ CYP11b
CA: ↑ CYP11b
CLO: ↓ CYP17
[79]
D. rerioCLO4 w0, 0.25, 0.50, 0.75 and 1.00% of the dietDietLiver, intestine, muscle, brain and heart↑ FABP1, FABP7, FABP10, FABP11
↑ acox1
↑ peroxisomes (liver) and mitochondria (heart)
[80]
Ctenopharyngodon idellalCLO4 w0, 1.25 g/kgDietLiver, muscle and mesenteric fat↓ TG, cholesterol and total lipid concentration
↑ hepatic ACO and LPL
↓ in FAS and ACC
[81]
C. carpioCA96 h1, 10 and 100 μg/LWaterborne Blood↓ RBC, plasma Na+, K+ and GOT
↑ WBC, glucose, protein, LDH enzyme and gill Na+/K+ ATPase
[82]
Cirrhinus mrigalaCA96 h and 35 d1, 10 and 100 μg/LWaterborne Blood↓ plasma GOT, GPT and gill Na+/K+ ATPase at short-term exposure
↑ gill Na+/K+ ATPase at long-term exposure
[83]
C. mrigalaCA96 h and 35 d1, 10 and 100 μg/LWaterborne Blood↑ plasmatic glucose, Na+ and K+ level at short-term exposure
↓ plasma protein and Cl levels
↑ plasma Na+ and Cl levels at long-term exposure
[84]
C. mrigalaCA96 h and 35 d1, 10 and 100 μg/LWaterborne Blood↓ TSH
↓ T4 level at 1 and 100 g L−l at 96 h
↓ T4 at long-term exposure
↓ T3
[85]
D. rerioCA5 to 140 d1 and 10 mg/gDietMuscle, gonads and liver↓, growth of F1 generation, TG in muscle and fecundity at high doses
↑ embryo’s abnormalities
↑ PPARα, PPARβ and ACO transcript of F1
↑ weight of F2 generation (control diet) compared to F1 at low dose
[86]
C. carpioCA4 and 10 d20 mg/L and 4 μg/LWaterborne Blood↑ PPAR
↑Acox1
↑ CYP4 and lpl, apoa1
↓ CYP27A
↑ CYP2K, CYP3A, GSTP, MDR1, MRP2
[60]
D. rerioCA3, 6, 24, 48, 72 and 96 h50 mg/LWaterborne Embryosembryos present high metabolic potential to CA, with 18 transformation products detected[87]
D. rerioCA120 h and 60 d10.35, 20.7, 41.4, 82.8, and 165.6 μg/LWaterborne Eggs, embryos and adults↑ SOD, GPx and LPO at high doses.
↑ CAT and GST at low dose
↓ CAT and GST at high dose
↓LPO at low dose
↓ total distance travelled and swimming time
[88]
D. rerioBEZ48 h, 7 and 21 d1.7, 33 and 70 mg/gDietBlood and gonads↓ Chol
↓ 11-KT after 21 d
↓ in PPARβ and PPARϒ after 48 h
↑ PPARϒ PPARβ, StAR and CYP17A1 at 70 mg/g after 21 days.
↓ CYP19A1a
↑ cystic spermatocytes
[89]
Abbreviations: 11-ketotestosterone (11-KT); Acetyl coenzyme A carboxylase (ACC); Acyl-CoA oxidase activity (ACO, Acox); Apolipoprotein AI (APOA1); Bezafibrate (BEZ); Chloride (Cl); Clofibrate (CLO); Clofibric acid (CA); Days (d); Estrogen receptor (ER); Gemfibrozil (GEM); Glutamate oxaloacetate transaminase (GOT); Glutathione peroxidase 1 (GPx1); Glutathione S-transferase P (GSTP); Hours (h); Interleukin 1β (IL1β); Intraperitoneal Injection (IP); Lactate dehydrogenase (LDH); Lipoprotein lipase (LPL); Potassium (K+); Red blood cell (RBC); Sodium (Na+); Testosterone (T); Thyroid stimulating hormone (TSH); Thyroxine (T4); Triiodothyronine (T3); Vitellogenin (VTG); Weeks (w); White blood cell (WBC); Peroxisomal proliferation-associated receptor (PPAR); Peroxisomal proliferation-associated receptor α (PPARα); Peroxisomal proliferation-associated receptor ϒ (PPARϒ); Peroxisomal proliferation-associated receptor β (PPARβ) Glutamate oxaloacetate transaminase (GOT); Glutathione peroxidase (GPx); Farnesyl diphosphate synthase (FDPS); Superoxide dismutase (SOD); Sulphotransferase (sult2b); Glucose transporters (glut1b); Catalase (CAT); UDP-glucuronosyl transferase (UDPGT); Tumor necrosis factor α (TNFα); Steroidogenic Acute Regulatory Protein (StAR); Total oxidant status (TOS); Lactate dehydrogenase (LDH); Glutamate-pyruvate transaminase (GPT); Alternative oxidase (AOX); Cytochrome P450 (CYP450); Cytochrome P450, family 11, subfamily B (CYP11B); Cytochrome P450, family 17, subfamily A, polypeptide 1 (CYP17A1); Cytochrome P450, 1A (CYP1A); Cytochrome P450, 3A (CYP3A); Cytochrome P450, 2K (CYP2K); Cytochrome P450, family 3, subfamily A, polypeptide 65 (CYP3A65); Cytochrome P450, family 4 (CYP4); Cytochrome P450, family 27, subfamily A (CYP27A); Cytochrome P450, family 19, subfamily A, polypeptide 1a (CYP19A1a); 17β-estradiol (E2); Estradiol (E); Triglycerides (TG); sterol regulatory element-binding protein 2 (SREBP2); Caspase 3 (CASP3); Glutathione reductase (GR); Multidrug Resistance Mutation 1 (MDR1); Multidrug Resistance-Associated Protein 2 (MRP2); and Cholesterol (Chol).
Table 2. The published literature on statins (atorvastatin and simvastatin) and their main effects on fish.
Table 2. The published literature on statins (atorvastatin and simvastatin) and their main effects on fish.
SpeciesDrugExposure PeriodConcentrationExposure RouteSamples TypeMain EffectsReference (2012–2022)
Oncorhynchus mykissATV7 d200 ng/L and 10 μg/LWaterborne Gills and liver↑ SOD, MT, BAX, P-gp, MRP1 and SULT2b in gill at low concentration[111]
Danio rerioATV30 d16 µg/g BWDietLiver and brain↓ cortisol, Chol, T, E, and atrogin1
↑ TG and CYP3A65 in males
↓ TG in females
↑ PPARα in females
↑ PPARϒ, SREBP1, SREBP2, HMGCR1 and HMGCR2
[44]
D. rerioATV120 h0.045, 0.5, 1 mg/LWaterborne Larvae↑ mortality
↓ spontaneous movement and response to physical stimuli
↓ COX, CS and LDH
↑ atrogen-1 at 0.045 mg/L and 1 mg/L
↓ atrogen-1 at 0.5 mg/L
↑ MURF
pgc-lα at 0.045 mg/L
pgc-lα at 0.5 mg/L and 1 mg/L
[112]
O. mykissATV3 or 6 h45 ng mL−1 and 80 nMIn vitroHepatocytes↑ HMGCR1, LDLR, PPARα, PPARϒ, and SREBP1 (lipid metabolism)
↑ CYP3A27 (xenobiotic metabolism)
↓ Chol
[113]
D. rerioSIM18 or 13 h0.3, 3 and 6 nM, 0.375, 0.5 and 0.75 μMWaterborne Embryos↑ mortality
↓ movement and heart rate
uneven distribution of septa
↓ myofibril size
↓ somite size
[114]
D. rerioSIM18 or 13 h0.3, 3 and 6 nM, 0.375, 0.5 and 0.75 μMWaterborne Embryos↑ mortality
↓ chol
↓ yolk extension, tail length, somite length, septa angle
↓ swimming capacity
↑ pericardial oedema
↓ heartbeat
↓ total cell number
[115]
D. rerioSIM80 h5 and 50 μg/LWaterborne Embryos↑ mortality, tail abnormalities, developmental delays, pericardium oedema
↓ ABC proteins
↑ EROD and GST
↓ Cu/ZnSOD and CAT
↑ accumulation
↑ ABCB4, ABCC1, ABCC2, ABCG2A, CYP3A65, CYP1A1 and CAT at 50μg/l
↓ GST, SOD and CAT at 5 μg/l
[116]
D. rerioSIM2 and 80 h5 or 50 μg/LWaterborne Embryos↑ mortality
↑ PPARα, PPARβ, PPARϒ, PXR, AhR, RAR-α, RAR-β, RAG-α after 2 h exposure
↓ rxraa, rxrab, rxrbb, rxrga, rxrgb after 2 h exposure
↑ RAR-αb, and AhR. After 80 h exposure
↓ PXR and rxrgb after 80 h exposure
[117]
D. rerioSIM90 d8, 40, 200 and 1000 ng/LWaterborne Liver↓ body weight
↑ fertilization rate
↑ abnormalities (short tail, pericardium oedema, yolk-sac and eye deformities)
↓ heartbeat after parental exposure
↓ Chol and TG
↓ HMGCRA
↓ CYP51 in males
[118]
Gambusia affinisSIM24, 72 and 168 h0.5, 5, 50 and 500 μg/LWaterborne Liver↑ PXR, CYP3A, P-gp, MRP2, UGT
↓ PXR and MRP2 at high doses
↑ ERND (72 h)
↓ ERND (24)
↑ hepatocyte abnormalities at 5 μg/L1 (168 h)
[119]
Mugilogobius abeiSIM72 h0.5, 5, 50 and 500 μg/LWaterborne Liver↑ P-gp, CYP1A, CYP3A, α-GST and PXR genes
↑ miR-148a at 50 μg/L
↑ miR-34c at 0.5μg/L (24 h)
↓ miR-34b and miR-34c at 0.5μg/L, 5μg/L, and 500μg/L (72 h)
↓ miR-34a at 500μg/L
↑ miR-27b at low doses
↓ miR-27b at high doses
↓ miR-27a
↑ ERND, T-SOD, CAT, GPX, MDA
↓ GSH
↓ hepatic vacuole diameter
[120]
D. rerioSIM90 d8, 40, 200 and 1000 ng/LWaterborne Brain↑ glut1b and COX4I1 in females
↓ glut1b and COX4I1 in males
↓ GAPDH
↓ ACADM and COX5aa in females
↑ ACADM in males
[121]
G. affinisSIM3 or 7 d0.5, 5, 50 and 500 µg/LWaterborne Liver↑ Nrf2
↑ SOD2, CAT and GST
↑ GSTA (168 h)
↓ GCLC and GSTA
↓ NQO1
↓ GSH
↑ GSH at 5 µg/L (24 h)
↓ MDA
↑ MDA (24 h)
↓ ERK
↓ JNK at low doses
↑ JNK at high doses
↑ histological changes in hepatic tissue (endoplasmic reticulum and mitochondria anomalies)
[122]
D. rerioSIM120 h and 60 d92.45, 184.9, 369.8, 739.6 and 1479.2 ng/LWaterborne Embryos ↑ and ↓ locomotion (erratic vs. purposeful swimming, distance travelled and time) depending on dose and day/night phase
↑ Cu-ZnSOD
↓ GPx, GST, TBARS
[123]
Abbreviations: Atorvastatin (ATV); Simvastatin (SIM); Superoxide Dismutase (SOD); Metallothionein (MT), bcl-2 associated X protein (BAX), P-glycoprotein (P-gp), multidrug resistance protein 1 (MRP1), Sulfotransferase Family 2B (SULT2b); cholesterol (Chol), testosterone (T); estrogen (E); triglycerides (TG); cytochrome P450, family 3, subfamily A, polypeptide 65 (CYP3A65); cytochrome P450, family 3, subfamily A (CYP3A); cytochrome P450, family 3, subfamily A, polypeptide 27 (CYP3A27); cytochrome P450, family 1, subfamily A, polypeptide 1 (CYP1A); cytochrome P450, family 1, subfamily A, polypeptide 1 (CYP1A1); cytochrome P450, family 51 (CYP51); Peroxisome proliferator-activated receptor alpha (PPARα); Peroxisome proliferator-activated receptor Beta (PPARβ) peroxisome proliferator-activated receptor gamma (PPARϒ); sterol regulatory element-binding protein 1 (SREBP1); sterol regulatory element-binding protein 2 (SREBP2); 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase 1 (HMGCR1); 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMGCRA); 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase 2 (HMGCR2); cytochrome oxidase (COX); citrate synthase (CS); lactate dehydrogenase (LDH); muscle RING-finer (MURF); peroxisome proliferator-activated receptor gamma coactivator 1α (pgc-1α); low-density lipoprotein receptor (LDLR); ATP-binding cassette transporter (ABC); ATP-binding cassette, subfamily B, member 4 (ABCB4); ATP-binding cassette, subfamily C, member 1 (ABCC1); ATP-binding cassette, subfamily C, member 2 (ABCC2); ATP-binding cassette, subfamily G, member 2A (ABCG2A); ethoxyresorufin-O-deethylase (EROD); glutathione S-transferases (GST); Copper (Cu); Zinc (Zn); catalase (CAT); pregnane X receptor (PXR); aryl hydrocarbon receptor (AhR); retinoic acid receptor alpha (RAR-α); retinoic acid receptor Beta (RAR-β); retinoic acid receptor alpha b (RAR-αb); recombination activating gene alpha (RAG-α); retinoid x receptor alpha a (rxrαa); retinoid x receptor alpha b (rxrαb); retinoid x receptor beta b (rxrβb); retinoid x receptor gamma a (rxrga); retinoid x receptor gamma b (rxrgb); multidrug resistance-associated protein 2 (MRP2); uridine 5′-diphospho-glucuronosyltransferase (UGT); Erythromycin-N-Demethylase (ERND); microRNA 148a (miR-148a); microRNA 148a (miR-148a); microRNA 34a (miR-34a); microRNA 34c (miR-34c); microRNA 34b (miR-34b); microRNA 27a (miR-27a); microRNA 27b (miR-27b); malondialdehyde (MDA); glutathione (GSH); Glutathione peroxidase (GPx); glucose transporter 1b (glut1b); cytochrome c oxidase subunits 4I1 (COX4I1); cytochrome c oxidase subunits 5aa (COX5aa); glyceraldehyde 3-phosphate dehydrogenase (GAPDH); medium-chain acyl-CoA dehydrogenase (ACADM); NF-E2-related factor 2 protein (Nrf2); superoxide dismutase 2 (SOD2); Alpha-glutathione S-transferase (GSTA); glutamate cysteine ligase catalytic subunit (GCLC); NAD(P)H quinone dehydrogenase 1 (NQO1); extracellular regulated protein kinase (ERK); c-Jun N-terminal kinase (JNK); and Thiobarbituric acid reactive substances (TBARS).
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

Blonç, M.; Lima, J.; Balasch, J.C.; Tort, L.; Gravato, C.; Teles, M. Elucidating the Effects of the Lipids Regulators Fibrates and Statins on the Health Status of Finfish Species: A Review. Animals 2023, 13, 792. https://doi.org/10.3390/ani13050792

AMA Style

Blonç M, Lima J, Balasch JC, Tort L, Gravato C, Teles M. Elucidating the Effects of the Lipids Regulators Fibrates and Statins on the Health Status of Finfish Species: A Review. Animals. 2023; 13(5):792. https://doi.org/10.3390/ani13050792

Chicago/Turabian Style

Blonç, Manuel, Jennifer Lima, Joan Carles Balasch, Lluis Tort, Carlos Gravato, and Mariana Teles. 2023. "Elucidating the Effects of the Lipids Regulators Fibrates and Statins on the Health Status of Finfish Species: A Review" Animals 13, no. 5: 792. https://doi.org/10.3390/ani13050792

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

Article Metrics

Back to TopTop