Next Article in Journal
Review of the Accuracy of Satellite Remote Sensing Techniques in Identifying Coastal Aquaculture Facilities
Previous Article in Journal
Estimating Cognitive Ability in the Wild: Validation of a Detour Test Paradigm Using a Cichlid Fish (Neolamprologus pulcher)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Review of P-Glycoprotein Function and Regulation in Fish

by
Christina U. Johnston
and
Christopher J. Kennedy
*
Department of Biological Sciences, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
*
Author to whom correspondence should be addressed.
Fishes 2024, 9(2), 51; https://doi.org/10.3390/fishes9020051
Submission received: 21 December 2023 / Revised: 22 January 2024 / Accepted: 25 January 2024 / Published: 27 January 2024

Abstract

:
The teleost ATP Binding Cassette (ABC) transporter P-glycoprotein (P-gp) is an active transmembrane transporter that plays a pivotal role in facilitating the movement of both endogenous and xenobiotic substrates (moderately hydrophobic and amphipathic compounds) across cell membranes. P-gp exhibits substrate specificity often shared with other ABC transporters and solute carrier proteins, thereby ensuring the maintenance of chemical homeostasis within cells. These transporters are integral to chemical defense systems in fish, as they actively expel a wide range of substrates, primarily unmodified compounds, from cells. This transport process assists in preventing chemical absorption (e.g., intestine), safeguarding sensitive tissues (e.g., brain and gonads), and effectively excreting substances (e.g., liver and kidney). Upregulated P-gp export activity in aquatic animals results in the multi-xenobiotic resistance (MXR) phenotype that plays an essential protective role in survival in contaminated environments. Pollutants inhibiting P-gp are termed chemosensitizers and heighten fish sensitivity to toxic P-gp substrates. While the known intrinsic functions of P-gp in fish encompass steroid hormone and bile acid processing, relatively little attention has been given to endogenous substrates and inhibitors. Fish P-glycoprotein regulation is orchestrated by pivotal nuclear transcription factors, including pregnane X receptor (PXR) and nuclear factor erythroid 2-related factor 2 (Nrf2). This comprehensive review provides profound insights into P-gp’s significance across diverse fish species, contributing to an enhanced understanding of fish physiology, evolution, and toxicology, and provides information with potential applications, such as environmental monitoring.
Key Contribution: 1. P-gp protects sensitive tissues from its potentially harmful substrates, and helps to remove those substrates from the body; 2. More research into the endogenous physiological functions of P-gp will allow for more accurate predictions of off-target drug effects and sensitive ecotoxicology endpoints; 3. Continued research into the functions and protective effects of P-gp in fish will provide valuable information for veterinary medicine, aquaculture, and environmental monitoring.

1. Introduction

Cell membranes house a diverse array of transporters essential for maintaining cellular homeostasis and functionality. These transporters are categorized into distinct classes based on their mechanism of action and substrate specificity. Solute carrier (SLC) transporters encompass a large superfamily that facilitates the transport of nutrients, ions, and metabolites. Ion channels permit the passive flux of ions across membranes, critical for electrical signaling and osmoregulation. Symporters and antiporters, categorized as secondary active transporters, utilize electrochemical gradients to transport substrates simultaneously in the same direction or in opposite directions, respectively. Finally, vesicular transporters mediate the intracellular trafficking of molecules through vesicle formation and fusion. These transporters control the movement of molecules across membrane barriers and serve to maintain chemical homeostasis. They also export endogenous molecules from sites of production to where they are needed, such as in the transport of morphogenic signaling molecules during embryonic development [1], the movement of gut microbiome metabolites into host intestinal tissue [2,3], and the export of fragrant volatile organic compounds from flowers [4]. ATP-binding cassette (ABC) transporters utilize energy from ATP hydrolysis to efflux a wide range of substrates, including lipids, ions, and xenobiotics. These transporters maintain concentrations of endogenous molecules at appropriate physiological levels and serve to reduce xenobiotic accumulation to prevent potential toxicity as part of the cell’s chemical defense system [1].
Cells have evolved intricate defense mechanisms against the accumulation of xenobiotic compounds, and they process these xenobiotics through a sequence of four phases: (phase 0) unmodified efflux, (phase I) functionalization reactions, (phase II) conjugation reactions, and (phase III) metabolite elimination. Phases I and II are biotransformation reactions that convert hydrophobic compounds into more hydrophilic metabolites with reduced membrane permeability. In phase III, hydrophilic metabolites are actively exported from the cell by transmembrane transport proteins, including the breast cancer-resistance protein (BCRP) and multidrug resistance proteins (MRPs) [5]. In phase 0, unmodified parent compounds are actively exported from the cell by transmembrane transporters, which include the ABC protein P-glycoprotein [6,7,8].
The ATP Binding Cassette (ABC) family of proteins facilitates the translocation of an extensive array of substrates. These transporters engage in critical cellular processes such as nutrient uptake, lipid trafficking, and antigen presentation, and play a major role in limiting the toxic potential of xenobiotics. Their structure is modular, and consists of highly conserved nucleotide binding domains (NBDs), which bind and hydrolyze ATP, and structurally diverse transmembrane domains (TMDs), which anchor the protein in the membrane and bind and transport substrates [9,10]. Full transporters include four components (two NBDs and two TMDs) in one protein, while half transporters include one NBD and one TMD, and must dimerize to function [9]. Eukaryotic ABC proteins are categorized into ten different families: ABCA to ABCJ [9,11]. The ABCA and ABCC families consist entirely of full transporters, the ABCB family includes both full and half transporters, and the ABCD, ABCG, and ABCH families are all half transporters [12]. Transporters in the ABCI family are composed of four separate proteins, with each NBD and TMD encoded by a different gene [9]. The recently proposed ABCJ family consists of DNA repair enzymes. These proteins are composed of half-enzymes, with one NBD and one substrate-interacting domain each, that form dimers around one or two DNA strands [13,14]. ABCE and ABCF proteins contain only a pair of NBDs without any TMDs, and function in the assembly of sub-cellular components by NBD dimerization [12].
ABC transporters are represented in all branches of life; prokaryotes have both importers and exporters, while eukaryotic ABC transporters are mostly exporters with only a few examples of importers [9,10]. Plants have an especially broad set of ABC transporters, with more than 120 members, compared to about 60 in fish and mammals [9,11]. This diversity of plant ABC transporters appears to be related to secondary metabolism, as well as the increased importance of stress responses and detoxification for sessile organisms compared to motile organisms [9]. The physiological functions of ABC transporters are an area of active research, and they tend to fall into the broad categories of homeostasis, signaling, and detoxification.
The permeability glycoprotein (P-glycoprotein [P-gp]) is a transmembrane exporter in the ABCB family. It transports a broad range of substrates, including both endogenous and xenobiotic molecules that tend to be mildly hydrophobic [15,16]. Its expression in excretory (e.g., liver, kidney), absorptive (e.g., intestine), and barrier (e.g., blood–brain, blood–eye, blood–gonad) tissues allows it to modulate its substrates’ absorption, tissue distribution, and elimination [17,18,19]. P-gp’s main identified function is xenobiotic resistance, but its role in regulating endogenous molecules is being increasingly recognized [1,11].
The discovery of P-gp marked a significant milestone in the understanding of cellular drug resistance mechanisms. P-gp was first identified in Chinese hamster ovary cells resistant to colchicine, revealing its role in the efflux of drugs from cells [20]. A major impetus for investigating P-gp stems from its role in multidrug resistance (MDR) observed in some forms of cancer. Tumor cells that overexpress P-gp exhibit an enhanced ability to efflux chemotherapy drugs, rendering them less susceptible to the cytotoxic effects of these agents. This phenomenon, often accompanied by overexpression of other transporters and metabolic enzymes, poses a significant challenge in cancer treatment [21]. The ability of P-gp (sometimes called multidrug resistance protein 1 [MDR1]) to mediate MDR highlights its clinical relevance and the need for a deeper understanding of its underlying mechanisms.
Research efforts into P-gp have been primarily directed towards unraveling the molecular basis of MDR and exploring strategies to circumvent this resistance. The study of mammalian P-gp, in particular, has garnered attention due to its involvement in drug interactions, bioavailability, and treatment outcomes. Investigations into the structural and functional aspects of P-gp have revealed insights into its substrate specificity, drug-binding sites, and the conformational changes associated with substrate transport [22]. These studies provide a foundation for designing targeted interventions aimed at modulating P-gp activity to enhance drug efficacy.
P-gp has received relatively little attention in the context of cancer in fish, although the MDR phenotype has been observed in several fish cancers [23,24,25]. The focus of research on fish and other aquatic animals originated in the discovery that animals living in contaminated aquatic environments over-express the same set of proteins that are overexpressed in MDR cancers, resulting in the multi-xenobiotic resistance (MXR) phenotype that allows them to survive and reproduce despite continuous chemical exposure. Specifically, resistant animals display higher levels of expression and activity of both efflux transporters (especially ABCB, ABCC and ABCG transporters) and biotransformation enzymes (including cytochromes and transferases) [26,27]. The MXR phenotype was first identified in marine and freshwater mussels [28,29], and was reported in fish soon after [30].
The evolution of fish populations towards pollution tolerance or resistance underscores the intricate interplay between natural selection and environmental pressures. Anthropogenic pollution is a recent and novel stressor in aquatic systems, and in response, fish populations have exhibited adaptive changes in chemical defense that, over generations, enhances their ability to reduce the potential of adverse effects [31]. Over time, the frequency of these beneficial genetic variations can increase within a population. For example, stickleback populations exposed to pulp mill effluent over many generations show significant changes in the genotype compared to populations from uncontaminated sites [32]. Pollution-driven selection can lead to shifts in the allele frequencies of certain genes associated with stress response mechanisms. In two North Atlantic eel species, the allele frequencies of genes related to sterol regulation and the response to oxidative stress differed significantly between populations exposed to varying contaminant profiles [33]. Fish populations exposed to pollutants may experience increased selection pressure on genes that code for transporters and enzymes involved in biotransformation and detoxification [33,34,35]. This can result in the evolution of fish with improved abilities to reduce accumulation, biotransform, and eliminate contaminants.
In addition to multi-generational genetic changes, rapid responses, such as phenotypic plasticity, allow fish to adjust their traits within their lifetime in response to pollution-induced stressors. This can include changes in behavior, morphology, or physiology that improve their chances of survival in polluted habitats, including the induction of P-gp and other genes that lead to the MXR phenotype [26,27]. These plastic responses can provide an initial buffer against pollution impacts while genetic adaptations accumulate over longer timeframes. The inhibitors of MXR proteins, including P-gp, are an emerging class of environmental pollutant [36,37] called chemosensitizers, making resistant organisms sensitive to toxic MXR protein substrates by reducing toxic concentration thresholds.
While most research on P-gp’s function in fish has focused on its role in xenobiotic defense and reducing chemical accumulation, the physiological roles of P-gp in fish are also beginning to be explored. This review aims to collect and synthesize the research on P-gp in fish to date, for the purpose of identifying knowledge gaps and providing a solid background for future research.

2. P-Glycoprotein Genes in Fish Species

In humans, P-glycoprotein is encoded by the ABCB1 gene on chromosome 7 [38,39]. Human ABCB4 has a high sequence homology to ABCB1 and is located next to it on the same chromosome due to a gene duplication [40]. ABCB5 also has a high sequence homology to ABCB1 due to an earlier gene duplication, and is located farther away on the same chromosome [40]. All fish species whose P-glycoprotein genes have been investigated thus far have Abcb4 or an Abcb4-like gene [38,41,42,43]. In zebrafish, the protein encoded by Abcb4 is functionally the most similar to the mammalian P-glycoprotein encoded by Abcb1 [38]. Thus, Abcb4 appears to be the primary functional P-glycoprotein in fish. Fish Abcb4 is an ortholog to the gene that gave rise to mammalian Abcb1 and Abcb4 [15]. Multidrug transport is the ancestral function of this gene, which was retained in mammalian Abcb1 [15]. All fish P-gp genes were initially labelled Abcb1 (or a derivation thereof) after their mammalian counterpart, and this nomenclature persists in the literature [38].
Most fish species investigated to date, including Atlantic cod (Gadus morhua), Nile tilapia (Oreochromis niloticus), medaka (Oryzias latipes), and three-spined stickleback (Gasterosteus aculeatus), lack Abcb1 and Abcb5 genes [15,38]. Two pufferfish species (Takifugu rubripes and Tetraodon nigroviridis) have Abcb1 in addition to Abcb4 [38], while African coelacanth (Latimeria chalumnae), channel catfish (Ictalurus punctatus), rainbow trout (Oncorhynchus mykiss), and zebrafish (Danio rerio) have Abcb5 in addition to Abcb4 [38,41,42]. The Abcb1 gene present in pufferfish likely arose from a separate family-specific duplication of Abcb4 [15,38]. The P-gp genes that have been identified in fish species are shown in Table 1.
Fish Abcb5 is an ortholog of mammalian Abcb5 [15]. Its function is not entirely clear, but it provides some multidrug transport function in fish and mammals [15,38,44]. It also appears to have endogenous transport functions, perhaps related to biliary excretion, epithelial cell adhesion, and stem cell maturation [15,38,41].

3. P-Glycoprotein Tissue Expression and Function

P-gp tends to be expressed in tissues with excretory (e.g., liver, kidney), absorptive (e.g., intestine), and barrier (e.g., skin, blood–brain, blood–eye and blood–gonad barriers) functions in both fish and mammals [17,18,19]. P-gp activity in excretory and absorptive tissues eliminates P-gp substrates from the body through excretory fluids (urine, bile, feces), while P-gp activity in blood–tissue barriers protect sanctuary tissues from the accumulation of P-gp substrates. The P-gp mRNA and protein expressions reported in fish tissues are shown in Table 2.
Many studies into P-gp expression used samples of only a single tissue, almost exclusively the liver [45,46,47,48]. Studies of this nature confirm the presence of a P-gp gene in a species without examining tissue expression in any detail. When P-gp expression is measured in multiple tissues, expression is usually highest in the kidney, followed by the liver and intestine [19,49,50,51]. P-gp expression tends to be moderate in the brain, eye, and gonads, while gill, muscle, skin, and other tissues generally have low P-gp expression [17,19,49,51,52]. This expression pattern suggests that the primary role of P-gp in fish is the elimination of substrates from the body, and that among sanctuary tissues, the brain, eye, and gonads are the most protected by P-gp.
In fish, as well as in mammals and invertebrates, P-gp can play a role in preventing the absorption of chemicals from the intestinal tract, protecting the organism from potentially harmful substances [53]. P-gp is expressed in the apical membrane of enterocytes, where it actively pumps substrates back into the intestinal lumen, limiting their uptake into the bloodstream [11]. P-gp expression increases along the intestinal tract, allowing for increasing rates of active transport as the lumen contents become more concentrated [11,17,19]. This efflux mechanism significantly contributes to the overall reduced accumulation and subsequent elimination of potentially harmful compounds.
P-gp plays a pivotal role in the excretion of chemicals by both the liver and the kidney, contributing to the elimination of xenobiotics. In the liver, P-gp is expressed in the canalicular membrane of hepatocytes, facilitating the transport of various substances, including drugs and environmental contaminants, from the hepatocyte cytoplasm into the bile canaliculus [54]. This excretion into the bile ultimately leads to the elimination of these substances from the body via feces. Studies using fish hepatocytes have demonstrated the active export of xenobiotic compounds and its impairment by P-gp inhibitors [54,55,56,57].
In the kidney, P-gp is expressed in the proximal tubules, where it is involved in the active secretion of chemicals from the blood into the renal filtrate [58,59]. This process aids in the elimination of xenobiotics, contributing to their excretion in urine. By preventing the reabsorption of these substances from the filtrate, P-gp assists in reducing their systemic exposure and potential toxicity. Isolated renal proximal tubules from flounder (Paralichthys lethostigma, Pseudopleuronectes americanus) and killifish (Fundulus heteroclitus) were an important early model system for studying xenobiotic transport in fish [59,60]. These studies provided evidence for the P-gp-mediated transport of numerous xenobiotics, including cyclosporin A, daunomycin, rapamycin, and ivermectin, into the renal tubule lumen [58,59,61,62], as well as an understanding of the relationship between SLC importers and ABC exporters expressed on opposite sides of the same epithelial cells [58,62,63].
Table 2. Expression of P-glycoprotein mRNA or protein (Abcb1, Abcb4, Abcb5, P-glycoprotein not otherwise specified) in fish tissues.
Table 2. Expression of P-glycoprotein mRNA or protein (Abcb1, Abcb4, Abcb5, P-glycoprotein not otherwise specified) in fish tissues.
SpeciesTissueReferences
Anoplarchus insignis
(Slender cockscomb blenny)
Liver[64]
Anoplarchus purpurescens
(High cockscomb blenny)
Liver[27]
Barbus barbus
(Barbel)
Liver[47]
Carassius auratus gibelio
(Silver Prussian carp)
Liver[47]
Chelon labrosus
(Thicklip grey mullet)
Liver, brain[65]
Chondrostoma nasus
(Sneep)
Liver[47]
Cyprinodon variegatus
(Sheepshead minnow)
Liver, kidney, intestine, exocrine pancreas[66]
Cyprinus carpio
(Common carp)
Liver[47]
Danio rerio
(Zebrafish)
Liver, kidney, intestine, ovary, testis, gill, skin, brain, brain vasculature, eye, muscle, heart, embryo (72 hpf), embryonic ionocytes[52,67,68,69]
Fundulus heteroclitus
(Killifish, mummichog)
Liver, distal intestine, brain capillaries[70,71,72]
Gambusia affinis
(Western mosquitofish)
Liver[46]
Gobiocypris rarus
(Chinese rare minnow)
Liver, kidney, intestine, skin, brain, spleen[51]
Hatcheria macraei
(Patagonian catfish)
Liver, gill[45]
Ictalurus punctatus
(Channel catfish)
Liver, intestine[73]
Jenynsia multidentata
(Killifish, one-sided livebearer)
Liver, gill, brain[74]
Lepomis macrochirus
(Bluegill sunfish)
Liver[46]
Limanda limanda
(Common dab)
Liver[37]
Oncorhynchus mykiss
(Rainbow trout)
Liver, kidney, intestine, gonad, gill, skin, brain, larval abdominal viscera, larval skin, larval head, larval muscle, larval yolk sac[17,19,41,45,54,75,76,77,78]
Oreochromis niloticus
(Nile tilapia)
Liver, intestine, zygote, embryo, larva[79,80]
Poecilia reticulata
(Guppy)
Liver, kidney, intestine, gill, muscle, ovary, esophagus, pancreas, branchial blood vessels, adrenal cortical tissue, gas gland[49,81]
Salmo trutta
(Brown trout)
Liver[45]
Scophthalmus maximus
(Turbot)
Liver, kidney, intestine, gill, brain, muscle, esophagus, heart[50]
Squalius cephalus
(European chub)
Liver[47]
Thunnus albacares
(Yellowfin tuna)
Liver, gill, brain, gonad[43]
Trematomus bernacchii
(Emerald rockcod)
Liver[48]
The blood–brain barrier (BBB) in fish, much like that in mammals, plays a pivotal role in regulating the exchange of substances between the bloodstream and the brain in order to maintain an environment optimal for neural function. Central to this barrier’s function is the presence of efflux transporters, notably P-gp, which actively transport molecules out of brain endothelial cells, limiting their entry into the brain. The evidence for P-gp’s role in the fish BBB is supported by studies demonstrating its presence in brain capillary endothelial cells, where it contributes to restricting the passage of xenobiotics and potential neurotoxicants into the brain parenchyma [71]. This is particularly important as fish species are frequently exposed to aquatic contaminants that act as neurotoxicants [82,83,84].
The importance of the BBB in fish varies among species and is influenced by ecological and evolutionary factors. Evidence for a strong BBB in certain fish species is shown in studies where P-gp actively prevents the entry of lipophilic compounds. For example, in isolated killifish (Fundulus heteroclitus) and dogfish shark (Squalus acanthias) brain capillaries, the fluorescent analogues of the P-gp substrates verapamil and cyclosporin A were actively exported into the capillary lumen; this transport was inhibited by P-gp inhibitor PSC-833 (valspodar) [71]. Similarly, the exposure of rainbow trout to the P-gp substrate ivermectin revealed limited brain accumulation due to P-gp-mediated efflux, indicating a robust barrier function [85]. Conversely, studies on some fish species suggest a more permeable BBB [86]. Evidence for a weak BBB can be seen in the relatively higher brain uptake of certain molecules or drugs, possibly due to lower P-gp expression levels or less restrictive tight junctions between endothelial cells [86]. The variability in BBB properties across fish species is likely linked to their habitats and ecological niches, as well as to prior chemical exposures.
In addition to the blood–brain barrier, P-gp is expressed with other transmembrane transporters in barriers protecting sensitive tissues. In the mammalian eye, P-gp transports its substrates from the endothelium of ocular capillaries into the blood as part of the blood-retina or blood-aqueous barrier [87,88]. Fish also possess a blood-retina barrier, but the role of P-gp in this barrier has not been investigated in detail [88]. Mammalian gonads are protected by blood-testis and blood-follicle barriers, which include P-gp [89,90]. P-gp is also expressed in fish gonads [17,19,43,49,52], but the existence of blood–gonad barriers has not been investigated in fish.
Conversely, P-gp plays a relatively small role in xenobiotic transport in gills. Fish gills form an important interface between the animal and its environment, allowing for the exchange of gasses, ions, and organic molecules between the water and the blood [91]. P-pg expression is typically lower in the gills than in the liver, kidney, intestine, brain, and gonad [17,19]. In rainbow trout gills, the main xenobiotic transporter form of P-gp (Abcb4) is nearly absent, while Abcb5 is expressed only in interlamellar progenitor cells in contact with neither the water nor the blood [41]. Gills express high levels of biotransformation enzymes, similar to or sometimes higher than expression in the liver and intestine [41,80]. Perhaps due to the prevalence of biotransformed metabolites, phase III transporters are expressed at much higher levels in fish gills than P-gp [19,41,80].

4. P-Glycoprotein in the Chemical Defense System

Proteins perform a wide spectrum of chemical defense functions, and collaboratively direct the movement of endogenous and xenobiotic molecules across bodily barriers, their distribution within an organism, and their subsequent elimination. Playing a key role in this chemical defense system in fish is P-gp, which operates in tandem with other defense components. Fish tissues commonly express P-gp alongside other transporters and enzymes, including phase I functionalization enzymes (e.g., cytochrome 450 oxidases), phase II conjugating enzymes (e.g., glutathione S-transferase, sulfotransferase, UDP-glucuronosyltransferase) [75,92,93,94,95], phase III exporters (e.g., multidrug resistance proteins [MRPs], breast cancer resistance protein [BCRP]) [41,75,93,96]), and solute carrier (SLC) transporters (e.g., organic anion transporting polypeptides [OATPs], organic anion transporters [OATs], organic cation transporters [OCTs], and multidrug and toxin extrusion proteins [MATEs]) [53,97,98]. Although the ABCB (MDR, P-gp) and ABCC (MRP) families of transporters are both referred to as “multidrug resistance proteins”, they are distinct classes of transporters that perform different functions in the chemical defense system. The interrelation of these enzymes and transporters in epithelial cells is illustrated in Figure 1.
In fish, the liver, kidney, and gill express high levels of biotransformation (phase I and II) enzymes [41,80]. In these tissues, biotransformation enzyme mRNA expression is 10- to 150-fold higher than ABC transporter expression [41,80]. These tissues also have higher expressions of phase III transporters (MRPs [Abccs], BCRP [Abcg]) than P-gp, as expected for tissues that have a high concentration of biotransformed metabolites [19,41,80]. Conversely, the fish intestine displays relatively modest biotransformation enzyme expression, with nearly equivalent levels of biotransformation enzymes and ABC transporters [80]. In the proximal intestine, phase III Abcc and Abcg transporters are expressed at similar levels to P-gp, but the distal intestine expresses much more P-gp compared to phase III transporters [19,80]. This transporter expression pattern favors the transport of unmodified parent compounds, which will be the predominant form (as opposed to biotransformed metabolites) due to the lower expression of biotransformation enzymes in this tissue.
Importer proteins tend to be expressed in the same cells as exporters, usually on the opposite sides of barrier tissues. The SLC21 (OATP) and SLC22 (OAT, OCT) families of secondary active transporters in particular are commonly expressed with P-gp in fish tissues [53,97,98]. These transport proteins import both membrane-permeable (lipophilic) and non-membrane permeable (hydrophilic) molecules. Membrane-permeable molecules require import proteins for the same reasons that they require export proteins including P-gp: active transport moves these molecules across barriers much faster than passive diffusion, and allows the organism to maintain substrate concentrations that are substantially different from passive equilibrium concentrations [4,9,99]. These import proteins share a very similar set of substrates and inhibitors with P-gp [1,100,101,102]. This common substrate base is expected for an integrated system that controls the movement of compounds across barriers: each substrate molecule needs to be sequentially imported then exported in order to cross an epithelial cell, thus the importers and exporters in the same cell will transport a shared set of substrates.
The regulation of these defense mechanisms is orchestrated by various signaling pathways and transcription factors. Nuclear receptors, including the aryl hydrocarbon receptor (AhR) and pregnane X receptor (PXR), play a crucial role in coordinating the expression of biotransformation enzymes, P-gp, and MRPs. Upon binding to their ligands, these receptors initiate the transcription of target genes involved in xenobiotic metabolism and transport. For example, AhR activation in fish induces the expression of phase I enzymes [27,70], while PXR activation upregulates both biotransformation enzymes and efflux transporters, including P-gp and MRPs [103,104,105,106].
P-gp performs essential chemical defense functions in different tissues, but it is not essential for life until faced with specific chemical challenges. For example, the dog and cat genomes both include alleles coding for non-functional P-gp proteins, but affected individuals show no adverse health effects unless exposed to a toxic P-gp substrate (e.g., ivermectin) [107]. Similarly, P-gp knockout mice are viable and generally indistinguishable from the wild-type, but are much more sensitive to drugs that are P-gp substrates [108]. In fish, zebrafish embryos subjected to the morpholino knockdown of Abcb4 and Abcb5 are viable and develop normally, but accumulate higher levels of P-gp substrate molecules compared to untreated embryos [38].

5. P-Glycoprotein Structure and Transport Mechanism

P-gp is a full transporter with two transmembrane domains (TMDs) and two nucleotide binding domains (NBDs) (Figure 2). In both fish and mammals, each TMD contains six transmembrane helices [16,38,41,80]. The NBDs contain highly conserved motifs that are a hallmark of all ABC transporters: A-loop, Walker A, Walker B, and the ABC signature [9,41]. P-gp has an atomic mass of 170 kDa in both fish and mammals [16,43,72,74]. The three-dimensional structure has been determined for human and mouse P-gp [109,110], but the structure of fish P-gp has not yet been fully elucidated. There is a 64% amino acid sequence identity between zebrafish Abcb4 and human ABCB1, while zebrafish Abcb5 shares 57% of its amino acid sequence with human ABCB1 [38]. Thus, fish and mammalian P-gp are expected to share most aspects of their structure and function [38,69]. The differences in P-gp function between these animal lineages are found in the finer points of their operation, such as substrate specificities and post-translational regulation [69,111]. These structural disparities might be attributed to differences in evolutionary history, ecological niches, and physiological requirements between mammals and fish.
Substrates enter the drug-binding pocket of P-gp either from the cytoplasm or directly from the hydrophobic region of the plasma membrane [109,110]. The drug-binding pocket is surrounded by the 12 α-helices of the two TMDs, and is lined with hydrophobic, amphipathic, polar, and charged amino acid side chains [109]. Substrate molecules interact with a subset of these amino acid side chains, forming the molecule’s binding site [109]. While the amino acid composition of human and zebrafish Abcb4 and Abcb5 is similar, there are different amino acids in their TMD α-helices, leading to different substrate specificities and affinities [69].
The resting state of the P-gp molecule is the inward-facing configuration [112]. In this state, the outside ends of two TMDs are in contact with each other (closed) on the outer surface of the cell membrane. The intracellular ends of the two TMDs are separated (open) on the cytoplasmic side of the cell membrane, and the two NBDs are separated from each other in the cytoplasm [112]. When a substrate is bound in the drug-binding pocket and an ATP molecule is bound to each of the two NBDs, the transport cycle begins (Figure 3) [112]. First, the cytoplasmic ends of the TMDs come together, and the NBDs dimerize with the two ATP molecules sandwiched between them. One of the bound ATP molecules is then hydrolyzed, and the P-gp molecule transitions to an outward-facing configuration. The outer ends of the TMDs separate, changing the shape of the drug binding site, and releasing the substrate into the extracellular media. Finally, the second ATP molecule is hydrolyzed, and the two NBDs separate. The P-gp molecule transitions back to the inward-facing configuration, from which it is able to begin a new transport cycle [112].
Figure 3. The P-glycoprotein transport cycle. 1. P-gp has an inward-open conformation, with no substrate or ATP bound. 2. A substrate binds in the drug-binding pocket between the transmembrane domains (TMDs), and one ATP molecule binds to each of the two nucleotide binding domains (NBDs). The NBDs dimerize with the two ATP molecules sandwiched between them. 3. One of the ATP molecules is hydrolyzed. The P-gp molecule transitions to an outward-facing conformation and releases the substrate into the extracellular space. 4. The second ATP molecule is hydrolyzed, allowing P-gp to return to an inward-facing conformation [112].
Figure 3. The P-glycoprotein transport cycle. 1. P-gp has an inward-open conformation, with no substrate or ATP bound. 2. A substrate binds in the drug-binding pocket between the transmembrane domains (TMDs), and one ATP molecule binds to each of the two nucleotide binding domains (NBDs). The NBDs dimerize with the two ATP molecules sandwiched between them. 3. One of the ATP molecules is hydrolyzed. The P-gp molecule transitions to an outward-facing conformation and releases the substrate into the extracellular space. 4. The second ATP molecule is hydrolyzed, allowing P-gp to return to an inward-facing conformation [112].
Fishes 09 00051 g003

6. P-Glycoprotein Substrates

The substrates of fish P-gp tend to be moderately hydrophobic, amphipathic, and often have a positively charged nitrogen atom and aromatic rings, closely matching the substrates of mammalian P-gp [16]. The known substrates of fish P-glycoprotein are shown in Table 3.
In studies comparing the substrates of fish P-gp to mammalian P-gp directly, the sets of molecules that are substrates in each animal lineage are similar but not identical. In a high-throughput study of 90 known substrates of human P-gp, zebrafish abcb4-expressing cells closely matched human P-gp-expressing cells in cytotoxicity resistance assays, while zebrafish abcb5-expressing cells showed resistance to fewer substrates in these assays [69]. In a study comparing human P-gp to P-gp from the livebearer fish Poeciliopsis lucida, 16 substrates were common to both species, while 5 substrates were exclusive to fish P-gp and 7 substrates were exclusive to human P-gp [113].
The majority of research efforts have concentrated on substrates of anthropogenic origin, which are materials that have become pervasive environmental contaminants due to human activities, such as fluorescent dyes, pharmaceuticals, pesticides, and industrial chemicals. This emphasis can be attributed to the origins of P-gp’s discovery, which revolved around drug resistance in mammalian cancers and resistance to xenobiotic pollutants in aquatic organisms. Xenobiotic substrates present in the environment throughout evolutionary history likely included such chemicals as bacterial toxins (e.g., doxorubicin, microcystin-LR, lipopolysaccharides) and plant metabolites (e.g., quinidine, nerol, isoeugenol).
P-gp has played an important role in chemical homeostasis in fish predating the onset of anthropogenic pollution, but its physiological roles have received relatively little attention. Few studies have investigated endogenous substrates in fish to any great extent; only two steroid hormones (cortisol and testosterone) have been identified as substrates to date [113,114]. Mammalian P-gp actively transports a diverse array of endogenous substrates including steroid hormones (e.g., cortisol, aldosterone, testosterone), lipids (e.g., phospholipids, cholesterol), and peptides (e.g., enkephalins) [131,132,133,134,135]. By managing intracellular levels of these compounds, P-gp plays a pivotal role in hormone signaling, lipid metabolism, and neurotransmission. While research focus has been on xenobiotic efflux and multidrug resistance, understanding the intricate interplay between P-gp and endogenous substrates provides evidence that it has broader regulatory significance in several physiological pathways [1].
Studies of P-gp expression have provided preliminary evidence that fish P-gp, especially Abcb5, may be involved in processing hormones and growth factors. In zebrafish ovaries, abcb5 is highly expressed in follicles during the early pre-vitellogenic stages of egg development, but is absent during the later vitellogenic and pre-ovulatory stages, while abcb4 is expressed at low levels at all stages of egg development [69]. This expression pattern suggests a possible role for fish P-gp in transporting hormones that stimulate early oocyte maturation, including follicle-stimulating hormone (FSH), luteinizing hormone (LH), and estradiol [136,137], and that abcb5 may have a higher affinity for these substrates than abcb4 does [69]. In rainbow trout gills, abcb5 is highly expressed in immature progenitor cells that mature into epithelial pavement (respiratory) cells, but abcb5 is not expressed in mature gill epithelial cells [41,138]. This suggests that P-gp plays a role in cell maturation in fish gills [41]. In mammals, P-gp has been implicated in stem cell differentiation; the ability of stem cells to differentiate into specialized cell types involves the regulation of multiple transport mechanisms including the P-gp transport of endogenous signaling molecules, such as growth factors and hormones, crucial for directing stem cell fate [139]. Exploring the endogenous substrates of P-gp in fish holds great potential and aligns with similar inquiries into the intrinsic functions of membrane transporters in mammals and other species [1].

7. The Regulation of P-Glycoprotein Expression

Diverse factors including xenobiotic exposure, heat stress, and oxidative stress can increase P-gp expression as part of an adaptive cellular defense response [16]. In mammals, the induction of P-gp has been extensively studied, especially in the context of drug interactions and multidrug resistance. Exposure to xenobiotics activates nuclear transcription factors including the pregnane X receptor (PXR), constitutive androstane receptor (CAR), aryl hydrocarbon receptor (AhR), and nuclear factor erythroid 2-related factor 2 (Nrf2), which bind to specific enhancer sequences in the ABCB1 promoter region, orchestrating the transcriptional upregulation of P-gp [140,141].
In fish, P-gp induction has gained attention due to its role in environmental detoxification and chemical defense. Exposure to aquatic pollutants, heavy metals, and pharmaceuticals can induce P-gp expression in fish, particularly in the liver, gills, and intestines. The regulation of P-gp in fish involves molecular mechanisms similar to those in mammals, including interactions between receptors, genetic response elements, and nuclear transcription factors. A key similarity between fish and mammals is the involvement of the nuclear receptors PXR and Nrf2 [94,122]. These receptors are activated by oxidative stress (Nrf2), or upon binding to specific ligands (PXR), and induce the subsequent transcription of P-gp and other genes. In fish, PXR has been identified as a central regulator of P-gp expression [103,104]. Studies have shown that PXR activation by xenobiotics, including pharmaceuticals (e.g., paracetamol, simvastatin) and plant flavonoids (e.g., quercetin, rotenone), leads to the upregulation of P-gp expression in fish tissues [95,142,143]. Similarly, Nrf2 activation has been linked to the regulation of P-gp in both fish and mammals [94,144]. For example, in zebrafish, the organophosphate pesticide malathion induced oxidative stress in zebrafish livers, leading to an increase in P-gp expression via the Nrf2 pathway [94].
Despite these similarities, there are notable differences in P-gp regulation between fish and mammals. Although AhR has been shown to regulate the expression of cytochrome P450 in fish, it does not affect P-gp expression [27,70]. Fish also lack the constitutive androstane receptor (CAR) that induces P-gp expression in mammals [105]. Mammalian research has provided detailed insights into the role of nuclear receptors and transcription factors in P-gp expression, often involving well-defined pathways. In contrast, fish studies are still evolving, and while the key players are identified, the intricate network of interactions remains to be fully elucidated.

8. Modulators of P-Glycoprotein Expression

P-gp expression in fish tissues can be induced or inhibited by many different chemicals, including both xenobiotic and endogenous compounds. Chemicals that modulate P-gp expression include those of anthropogenic origin such as pharmaceuticals (e.g., morphine, paracetamol, simvastatin), pesticides (e.g., malathion, emamectin benzoate), and industrial chemicals (e.g., crude oil, perfluorooctane sulfonate [PFOS]), and those of naturally-derived origin, such as plant metabolites (e.g., glycyrrhizic acid, quercetin) and bacterial compounds (e.g., microcystin-LR). Endogenous chemical modulators include bile acids and steroid hormones. Compounds that induce P-gp expression in fish are listed in Table 4, and those that inhibit P-gp expression are shown in Table 5. In addition to the specific modulating substances listed, complex mixtures of toxicants in industrial and urban effluents and receiving waters induce P-gp expression, and contribute to the MXR phenotype [11,45,76]. Environmental stressors can also alter P-gp expression. For example, fasted (4 weeks) rainbow trout exhibit induced P-gp expression in intestinal epithelia [145].
Alterations in P-gp expression are intricate in terms of timing, effective dosages, and combinations of chemicals. Certain compounds can induce or inhibit P-gp expression at different time intervals, adding to the complexity of this modulation. For example, in zebrafish brains, malathion induced P-gp expression after 3 d, but inhibited its expression after 7 d [94]. Stress that results from acute exposures to toxicants can differ from long-term exposures [157], and may require different gene expression patterns in response. In addition, early inductions of coordinately regulated genes (e.g., antioxidant enzymes) may alter the condition of the tissue (e.g., oxidative stress), thereby altering later P-gp expression [94].
Some substances affect P-gp expression only when present in mixtures. The pesticides cypermethrin and chlorpyrifos significantly decreased P-gp expression in killifish gill and brain tissue in combination, but had no significant effect in these tissues when present individually [151]. Individually, titanium dioxide nanoparticles and the dioxin TCDD did not affect P-gp expression in sea bass liver, but the combination of both compounds caused a significant reduction in P-gp expression [96]. The pesticide methyl parathion induced P-gp expression in zebrafish liver only in combination with verapamil [125]. These additive or synergistic effects may indicate that each of the compounds in the mixture interacts with different aspects of the mechanisms regulating P-gp expression in fish.
The effects of P-gp expression modulators can be species-specific. For example, the synthetic steroid pregnenolone 16α-carbonitrile (PCN) induced P-gp expression in rainbow trout hepatocytes [146], inhibited P-gp expression in zebrafish livers [104], and had no effect on P-gp expression in killifish hepatoma cells [158]. Likewise, the bile salt 5α-cyprinol 27-sulfate induced P-gp expression in zebrafish hepatocytes, but similar induction effects were not observed in sea lampreys (Petromyzon marinus) [105]. Within a single species, the effects of expression modulators can vary by sex and by tissue. For example, the antimicrobial agent triclosan increased liver P-gp expression in male swordtail fish (Xiphophorus helleri), but decreased expression in females [153]. The fungicide carbendazim induced P-gp expression in killifish gill, but decreased expression in the liver [150]. Emamectin benzoate-treated rainbow trout had increased P-gp expression in the liver, but decreased P-gp expression in the intestine [152]. These differences may arise from variations in exposure profiles between tissues, sexes, and species, which would require different chemical defense strategies.
The modulation of P-gp expression is of great importance in both clinical and environmental contexts. In clinical settings, changes in P-gp expression can alter the bioavailability and pharmacokinetics of therapeutic drugs. In aquatic toxicology, P-gp expression confers resistance to environmental toxicants; thus, alterations can have major impacts on fitness and survival in contaminated environments. Further research into inducers and inhibitors of P-gp expression in fish will therefore provide valuable information relating to veterinary medicine, aquaculture, and environmental risk assessment.
Table 5. Inhibitors of P-glycoprotein (Abcb1, Abcb4, Abcb5, P-glycoprotein not otherwise specified) mRNA or protein expression in fish.
Table 5. Inhibitors of P-glycoprotein (Abcb1, Abcb4, Abcb5, P-glycoprotein not otherwise specified) mRNA or protein expression in fish.
Expression InhibitorSpeciesTissueReferences
Hormone
Pregnenolone 16α-carbonitrile
(Synthetic steroid)
Danio rerioLiver[104]
Phytochemical
GenisteinIctalurus punctatusKidney[142]
Carbon Allotrope
Fullerene (nC60)Cyprinus carpioLiver[123]
Industrial Chemicals
Metal mine tailings
(Mn, Cd, As, Cu, Cr)
Astyanax lacustrisLiver[159]
Pesticides
CarbendazimJenynsia multidentateLiver[150]
Emamectin benzoateOncorhynchus mykissIntestine[152]
Malathion (7-day exposure)Danio rerioBrain[94]
TriclosanXiphophorus helleri (females)Liver[153]
Analgesic Pharmaceutical
MorphineDanio rerioEmbryo[116]

9. Inhibitors of P-Glycoprotein Transport Activity

P-gp activity inhibitors attenuate the efflux function of the transporter, thereby interfering with its role in actively exporting substrates out of cells. Inhibitors of P-gp transport activity are being recognized as an important class of environmental pollutants that inhibit chemical defense enzymes and transporters [160]. They interfere with MXR mechanisms that contribute to chemical defense in contaminated environments [7,11]. P-gp is the most studied MXR protein, but many transporters and enzymes contribute to the MXR phenotype [15,28,29]. The inhibition of MXR proteins allows substrates to exert harmful effects at lower concentrations than they would in the absence of these inhibitors [160,161,162,163,164]. This increases the organism’s sensitivity to environmental toxicants; thus, these inhibitors are referred to as chemosensitizers. Chemosensitizers in complex mixtures increase the toxicity of the mixture [160]. Inhibitors of MXR transporters including P-gp, MRPs, BCRP, and MATEs interfere with transport activity, which leads to the increased intracellular accumulation of their substrates [7,160]. In clinical contexts, compounds that act as chemosensitizers are major contributors to drug–drug and drug–food interactions, since they can alter the pharmacokinetics of co-administered pharmaceuticals [165].
Chemosensitizers in fish are usually studied in the context of complex mixtures of environmental pollutants: most identified inhibitors of fish P-gp are anthropogenic in origin (e.g., pesticides, industrial chemicals, pharmaceuticals, personal care products). Only one endogenous P-gp inhibitor is known in fish: the bile acid taurochenodeoxycholate [100]. More P-gp inhibitors have been identified in fish than any other type of interacting chemical (inhibitors, substrates, and expression modulators). Chemosensitizers are relatively easy to detect using in vitro methods, especially dye accumulation and ATP consumption assays [113,160]. However, experiments of this nature, especially if they use a high-throughput design, usually provide a simple confirmation of the effect rather than a more thorough concentration–response relationship [160]. The known inhibitors of fish P-gp activity are shown in Table 6.
P-gp inhibitors act by one of two general mechanisms: competitive and non-competitive inhibition. Competitive inhibitors are P-gp substrates that inhibit the transport of other substrates by occupying P-gp transport capacity [160]. The most potent competitive P-gp inhibitors have high affinity in the P-gp drug-binding pocket, and high membrane permeability [173,174]. These properties allow for a futile and energetically costly cycle of outward active transport and inward diffusion [15]. Non-competitive inhibitors impede some aspect of the P-gp transport mechanism. Vanadate, for example, inhibits P-gp ATPase activity, while tariquidar blocks the movement of transmembrane helices necessary for substrate translocation [110]. Non-competitive inhibitors reduce ATP consumption by preventing P-gp from actively transporting substrates [15,113]. Many studies do not determine the inhibition mechanism of chemosensitizers; however, if a substance has been identified as both a substrate and an inhibitor, it is likely a competitive inhibitor.
In vivo P-gp inhibition can exacerbate the behavioral effects of substrate exposure, as observed with the neurotoxic P-gp substrate ivermectin (IVM). IVM exposure causes behavioral deficits in fish, including altered swimming performance, poor motor coordination, and lethargy [82,83,84]. During co-exposure to the P-gp inhibitor cyclosporin A, all of these neurotoxic IVM effects were more severe in killifish, rainbow trout, and zebrafish [82,83,84].
Substantial knowledge gaps remain with respect to the magnitude of chemosensitizer effects as environmental pollutants [160,175]. More research is also needed to understand the contribution of the inhibition of other MXR transporters in chemical defense, since nearly all studies of chemosensitizers in aquatic animals have focused on P-gp [160]. A greater diversity of chemical compounds, and combinations of compounds, should be tested for chemosensitizing effects, especially the industrial and agricultural chemicals currently in use. More detailed information regarding effect concentration thresholds is needed as well. Studies of chemosensitizer exposure in aquatic media should report measured concentrations wherever possible, rather than nominal concentrations. Many reported IC50 concentrations for chemosensitizers exceed the water solubility of those chemicals; thus, the observed chemosensitizing effects will have occurred at much lower aqueous concentrations than those reported [160]. Expanding the available information about chemosensitizer effects in these areas will greatly improve our ability to accurately account for chemosensitizers in environmental risk assessments.

10. The Energetic Costs of P-Glycoprotein Activity

P-gp transport activity is powered by ATP hydrolysis, and may be energetically costly in terms of overall energy budgets. P-gp activity increases cellular energy consumption when fish hepatocytes are challenged in vitro with P-gp substrates [55,56]. During in vivo exposure, P-gp induction alone does not increase whole-body respiration rates in rainbow trout [176]. Increased P-gp activity during substrate exposure likewise does not affect whole-animal energy consumption [176]. Only when P-gp expression is induced and the organism is simultaneously challenged with substrate exposure does P-gp activity increase the in vivo respiration rate of rainbow trout [176].
The protective function of P-gp is a high priority in fishes’ energy budgets. P-gp activity is maintained during fasting and starvation in zebrafish and rainbow trout [82,177]. Similarly, four weeks of starvation induced P-gp expression in rainbow trout intestinal epithelia [145]. Thus, the cellular-level activity of P-gp is integrated into protective effects at the organismal level, and this protection is prioritized even during periods of limited resources.

11. Conclusions

In fish, P-gp protects sensitive tissues from its potentially harmful substrates, and helps to remove those substrates from the body. Although P-gp has been studied less extensively in fish than in mammals, it seems to share similar functions in both animal lineages. P-gp expression confers resistance to toxic substrates, and this resistance can be diminished by chemosensitizing P-gp inhibitors. Fish P-gp has been studied primarily in the context of resistance to anthropogenic contamination, so its endogenous functions are only beginning to be explored.
P-gp likely has broad regulatory significance in fish, potentially transporting hormones, growth factors, and other signaling molecules, thereby influencing processes including cell maturation, reproductive development, and energy metabolism. P-gp transport helps to maintain homeostasis at all levels of biological organization, from molecules to the whole organism, and contributes to the health of populations and ecosystems [1,178]. A deeper understanding of the endogenous physiological functions of P-gp will allow for more accurate predictions of the off-target effects of drugs and the identification of sensitive endpoints in ecotoxicology research.
Environmental exposure to P-gp inhibitors and toxic P-gp substrates presents serious risks to the health of aquatic ecosystems. P-gp confers multixenobiotic resistance to fish and invertebrates living in contaminated environments, but chemosensitizing compounds inhibit this resistance. While substantial research efforts have identified a wide range of environmental contaminants that are substrates, inhibitors, and expression modulators of fish P-gp, much remains to be discovered with respect to their effect concentrations, physiological effects, and interactions with other compounds. Continued research into the endogenous functions and xenobiotic protective effects of P-gp in fish will provide valuable information for applications, including those in veterinary medicine, aquaculture, and environmental monitoring.

Author Contributions

Conceptualization, C.U.J. and C.J.K.; investigation, C.U.J.; resources, C.J.K.; writing—original draft preparation, C.U.J.; writing—review and editing, C.U.J. and C.J.K.; visualization, C.U.J. and C.J.K.; supervision, C.J.K.; project administration, C.J.K.; funding acquisition, C.J.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by a Discovery grant from the Natural Sciences and Engineering Research Council of Canada, grant number R611305 to C.J.K.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

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

Acknowledgments

We thank Vinicius Cavicchioli Azevedo (Simon Fraser University) for consultation and manuscript feedback.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Nigam, S.K. What Do Drug Transporters Really Do? Nat. Rev. Drug Discov. 2015, 14, 29–44. [Google Scholar] [CrossRef] [PubMed]
  2. Wu, W.; Jamshidi, N.; Eraly, S.A.; Liu, H.C.; Bush, K.T.; Palsson, B.O.; Nigam, S.K. Multispecific Drug Transporter Slc22a8 (Oat3) Regulates Multiple Metabolic and Signaling Pathways. Drug Metab. Dispos. 2013, 41, 1825–1834. [Google Scholar] [CrossRef] [PubMed]
  3. Wikoff, W.R.; Nagle, M.A.; Kouznetsova, V.L.; Tsigelny, I.F.; Nigam, S.K. Untargeted Metabolomics Identifies Enterobiome Metabolites and Putative Uremic Toxins as Substrates of Organic Anion Transporter 1 (Oat1). J. Proteome Res. 2011, 10, 2842–2851. [Google Scholar] [CrossRef] [PubMed]
  4. Adebesin, F.; Widhalm, J.R.; Boachon, B.; Lefèvre, F.; Pierman, B.; Lynch, J.H.; Alam, I.; Junqueira, B.; Benke, R.; Ray, S.; et al. Emission of Volatile Organic Compounds from Petunia Flowers Is Facilitated by an ABC Transporter. Science 2017, 356, 1386–1388. [Google Scholar] [CrossRef] [PubMed]
  5. Iversen, D.B.; Andersen, N.E.; Dalgård Dunvald, A.-C.; Pottegård, A.; Stage, T.B. Drug Metabolism and Drug Transport of the 100 Most Prescribed Oral Drugs. Basic Clin. Pharmacol. Toxicol. 2022, 131, 311–324. [Google Scholar] [CrossRef]
  6. Epel, D.; Luckenbach, T.; Stevenson, C.N.; Macmanus-Spencer, L.A.; Hamdoun, A.; Smital, T. Efflux Transporters: Newly Appreciated Roles in Protection against Pollutants. Environ. Sci. Technol. 2008, 42, 3914–3920. [Google Scholar] [CrossRef]
  7. Ferreira, M.; Costa, J.; Reis-Henriques, M.A. ABC Transporters in Fish Species: A Review. Front. Physiol. 2014, 5, 266. [Google Scholar] [CrossRef]
  8. Kroll, T.; Prescher, M.; Smits, S.H.J.; Schmitt, L. Structure and Function of Hepatobiliary ATP Binding Cassette Transporters. Chem. Rev. 2021, 121, 5240–5288. [Google Scholar] [CrossRef]
  9. Pierman, B.; Boutry, M.; Lefèvre, F. The ABC of ABC Transporters. In Advances in Botanical Research; Maurel, C., Ed.; Membrane Transport in Plants; Academic Press: New York, NY, USA, 2018; Volume 87, pp. 1–23. [Google Scholar]
  10. Rice, A.J.; Park, A.; Pinkett, H.W. Diversity in ABC Transporters: Type I, II and III Importers. Crit. Rev. Biochem. Mol. Biol. 2014, 49, 426–437. [Google Scholar] [CrossRef]
  11. Bieczynski, F.; Painefilú, J.C.; Venturino, A.; Luquet, C.M. Expression and Function of ABC Proteins in Fish Intestine. Front. Physiol. 2021, 12, 791834. [Google Scholar] [CrossRef]
  12. Dean, M.; Annilo, T. Evolution of the ATP-Binding Cassette (ABC) Transporter Superfamily in Vertebrates. Annu. Rev. Genom. Hum. Genet. 2005, 6, 123–142. [Google Scholar] [CrossRef] [PubMed]
  13. Figueira-Mansur, J.; Schrago, C.G.; Salles, T.S.; Alvarenga, E.S.L.; Vasconcellos, B.M.; Melo, A.C.A.; Moreira, M.F. Phylogenetic Analysis of the ATP-Binding Cassette Proteins Suggests a New ABC Protein Subfamily J in Aedes Aegypti (Diptera: Culicidae). BMC Genom. 2020, 21, 463. [Google Scholar] [CrossRef] [PubMed]
  14. Hopfner, K.-P.; Tainer, J.A. Rad50/SMC Proteins and ABC Transporters: Unifying Concepts from High-Resolution Structures. Curr. Opin. Struct. Biol. 2003, 13, 249–255. [Google Scholar] [CrossRef] [PubMed]
  15. Luckenbach, T.; Fischer, S.; Sturm, A. Current Advances on ABC Drug Transporters in Fish. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2014, 165, 28–52. [Google Scholar] [CrossRef] [PubMed]
  16. Silva, R.; Vilas-Boas, V.; Carmo, H.; Dinis-Oliveira, R.J.; Carvalho, F.; de Lourdes Bastos, M.; Remião, F. Modulation of P-Glycoprotein Efflux Pump: Induction and Activation as a Therapeutic Strategy. Pharmacol. Ther. 2015, 149, 1–123. [Google Scholar] [CrossRef] [PubMed]
  17. Love, R.C.; Osachoff, H.L.; Kennedy, C.J. Short Communication: Tissue-Specific Transcript Expression of P-Glycoprotein Isoforms Abcb1a and Abcb1b in Rainbow Trout (Oncorhynchus mykiss) Following Induction with Clotrimazole. Comp. Biochem. Physiol. B Biochem. Mol. Biol. 2021, 252, 110538. [Google Scholar] [CrossRef] [PubMed]
  18. Leslie, E.M.; Deeley, R.G.; Cole, S.P.C. Multidrug Resistance Proteins: Role of P-Glycoprotein, MRP1, MRP2, and BCRP (ABCG2) in Tissue Defense. Toxicol. Appl. Pharmacol. 2005, 204, 216–237. [Google Scholar] [CrossRef]
  19. Lončar, J.; Popović, M.; Zaja, R.; Smital, T. Gene Expression Analysis of the ABC Efflux Transporters in Rainbow Trout (Oncorhynchus mykiss). Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2010, 151, 209–215. [Google Scholar] [CrossRef]
  20. Juliano, R.L.; Ling, V. A Surface Glycoprotein Modulating Drug Permeability in Chinese Hamster Ovary Cell Mutants. Biochim. Biophys. Acta BBA—Biomembr. 1976, 455, 152–162. [Google Scholar] [CrossRef]
  21. Catalano, A.; Iacopetta, D.; Ceramella, J.; Scumaci, D.; Giuzio, F.; Saturnino, C.; Aquaro, S.; Rosano, C.; Sinicropi, M.S. Multidrug Resistance (MDR): A Widespread Phenomenon in Pharmacological Therapies. Molecules 2022, 27, 616. [Google Scholar] [CrossRef]
  22. Gottesman, M.M.; Ling, V. The Molecular Basis of Multidrug Resistance in Cancer: The Early Years of P-Glycoprotein Research. FEBS Lett. 2006, 580, 998–1009. [Google Scholar] [CrossRef] [PubMed]
  23. Koehler, A.; Alpermann, T.; Lauritzen, B.; Van Noorden, C.J.F. Clonal Xenobiotic Resistance during Pollution-Induced Toxic Injury and Hepatocellular Carcinogenesis in Liver of Female Flounder (Platichthys flesus (L.)). Acta Histochem. 2004, 106, 155–170. [Google Scholar] [CrossRef] [PubMed]
  24. Machado, S.P.; Cunha, V.; Reis-Henriques, M.A.; Ferreira, M. Histopathological Lesions, P-Glycoprotein and PCNA Expression in Zebrafish (Danio Rerio) Liver after a Single Exposure to Diethylnitrosamine. Environ. Toxicol. Pharmacol. 2014, 38, 720–732. [Google Scholar] [CrossRef] [PubMed]
  25. Vogelbein, W.K.; Fournie, J.W.; Cooper, P.S.; Van Veld, P.A. Hepatoblastomas in the Mummichog, Fundulus heteroclitus (L.), from a Creosote-Contaminated Environment: A Histologic, Ultrastructural and Immunohistochemical Study. J. Fish Dis. 1999, 22, 419–431. [Google Scholar] [CrossRef]
  26. Paetzold, C.S.; Ross, N.W.; Richards, R.C.; Jones, M.; Hellou, J.; Bard, S.M. Up-Regulation of Hepatic ABCC2, ABCG2, CYP1A1 and GST in Multixenobiotic-Resistant Killifish (Fundulus heteroclitus) from the Sydney Tar Ponds, Nova Scotia, Canada. Mar. Environ. Res. 2009, 68, 37–47. [Google Scholar] [CrossRef] [PubMed]
  27. Bard, S.M.; Woodin, B.R.; Stegeman, J.J. Expression of P-Glycoprotein and Cytochrome P450 1A in Intertidal Fish (Anoplarchus purpurescens) Exposed to Environmental Contaminants. Aquat. Toxicol. 2002, 60, 17–32. [Google Scholar] [CrossRef] [PubMed]
  28. Kurelec, B.; Pivčević, B. Evidence for a Multixenobiotic Resistance Mechanism in the Mussel Mytilus Galloprovincialis. Aquat. Toxicol. 1991, 19, 291–301. [Google Scholar] [CrossRef]
  29. Kurelec, B.; Pivčević, B. Distinct Glutathione-Dependent Enzyme Activities and a Verapamil-Sensitive Binding of Xenobiotics in a Fresh-Water Mussel Anodonta Cygnea. Biochem. Biophys. Res. Commun. 1989, 164, 934–940. [Google Scholar] [CrossRef]
  30. Kurelec, B. The Multixenobiotic Resistance Mechanism in Aquatic Organisms. Crit. Rev. Toxicol. 1992, 22, 23–43. [Google Scholar] [CrossRef]
  31. Hamilton, P.B.; Rolshausen, G.; Uren Webster, T.M.; Tyler, C.R. Adaptive Capabilities and Fitness Consequences Associated with Pollution Exposure in Fish. Philos. Trans. R. Soc. B Biol. Sci. 2017, 372, 20160042. [Google Scholar] [CrossRef]
  32. Lind, E.E.; Grahn, M. Directional Genetic Selection by Pulp Mill Effluent on Multiple Natural Populations of Three-Spined Stickleback (Gasterosteus aculeatus). Ecotoxicology 2011, 20, 503–512. [Google Scholar] [CrossRef]
  33. Laporte, M.; Pavey, S.A.; Rougeux, C.; Pierron, F.; Lauzent, M.; Budzinski, H.; Labadie, P.; Geneste, E.; Couture, P.; Baudrimont, M.; et al. RAD Sequencing Reveals Within-Generation Polygenic Selection in Response to Anthropogenic Organic and Metal Contamination in North Atlantic Eels. Mol. Ecol. 2016, 25, 219–237. [Google Scholar] [CrossRef]
  34. Williams, L.M.; Oleksiak, M.F. Ecologically and Evolutionarily Important SNPs Identified in Natural Populations. Mol. Biol. Evol. 2011, 28, 1817–1826. [Google Scholar] [CrossRef]
  35. Wirgin, I.; Roy, N.K.; Loftus, M.; Chambers, R.C.; Franks, D.G.; Hahn, M.E. Mechanistic Basis of Resistance to PCBs in Atlantic Tomcod from the Hudson River. Science 2011, 331, 1322–1325. [Google Scholar] [CrossRef] [PubMed]
  36. Kurelec, B. Reversion of the Multixenobiotic Resistance Mechanism in Gills of a Marine Mussel Mytilus Galloprovincialis by a Model Inhibitor and Environmental Modulators of P170-Glycoprotein. Aquat. Toxicol. 1995, 33, 93–103. [Google Scholar] [CrossRef]
  37. Smital, T.; Kurelec, B. The Chemosensitizers of Multixenobiotic Resistance Mechanism in Aquatic Invertebrates: A New Class of Pollutants. Mutat. Res. Mol. Mech. Mutagen. 1998, 399, 43–53. [Google Scholar] [CrossRef] [PubMed]
  38. Fischer, S.; Klüver, N.; Burkhardt-Medicke, K.; Pietsch, M.; Schmidt, A.-M.; Wellner, P.; Schirmer, K.; Luckenbach, T. Abcb4 Acts as Multixenobiotic Transporter and Active Barrier against Chemical Uptake in Zebrafish (Danio Rerio) Embryos. BMC Biol. 2013, 11, 69. [Google Scholar] [CrossRef]
  39. Dean, M.; Moitra, K.; Allikmets, R. The Human ATP-Binding Cassette (ABC) Transporter Superfamily. Hum. Mutat. 2022, 43, 1162–1182. [Google Scholar] [CrossRef]
  40. Moitra, K.; Scally, M.; McGee, K.; Lancaster, G.; Gold, B.; Dean, M. Molecular Evolutionary Analysis of ABCB5: The Ancestral Gene Is a Full Transporter with Potentially Deleterious Single Nucleotide Polymorphisms. PLoS ONE 2011, 6, e16318. [Google Scholar] [CrossRef]
  41. Kropf, C.; Fent, K.; Fischer, S.; Casanova, A.; Segner, H. ABC Transporters in Gills of Rainbow Trout (Oncorhynchus mykiss). J. Exp. Biol. 2020, 223, jeb221069. [Google Scholar] [CrossRef] [PubMed]
  42. Liu, S.; Li, Q.; Liu, Z. Genome-Wide Identification, Characterization and Phylogenetic Analysis of 50 Catfish ATP-Binding Cassette (ABC) Transporter Genes. PLoS ONE 2013, 8, e63895. [Google Scholar] [CrossRef]
  43. Nicklisch, S.C.T.; Pouv, A.K.; Rees, S.D.; McGrath, A.P.; Chang, G.; Hamdoun, A. Transporter-Interfering Chemicals Inhibit P-Glycoprotein of Yellowfin Tuna (Thunnus albacares). Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2021, 248, 109101. [Google Scholar] [CrossRef]
  44. Chartrain, M.; Riond, J.; Stennevin, A.; Vandenberghe, I.; Gomes, B.; Lamant, L.; Meyer, N.; Gairin, J.E.; Guilbaud, N.; Annereau, J.P. Melanoma Chemotherapy Leads to the Selection of ABCB5-Expressing Cells. PLoS ONE 2012, 7, e36762. [Google Scholar] [CrossRef]
  45. Assef, Y.A.; Di Prinzio, C.Y.; Horak, C.N. Differential Activities of the Multixenobiotic Resistance Mechanism in Freshwater Fishes Inhabiting Environments of Patagonia Argentina. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2019, 217, 32–40. [Google Scholar] [CrossRef]
  46. Damaré, C.L.; Kaddoumi, A.K.; Baer, K.N. Investigation of the Multixenobiotic Resistance Mechanism in the Freshwater Fishes Western Mosquitofish, Gambusia affinis, and Bluegill Sunfish, Lepomis Macrochirus. Bull. Environ. Contam. Toxicol. 2009, 83, 640–643. [Google Scholar] [CrossRef]
  47. Klobučar, R.; Žaja, R.; Franjević, D.; Brozović, A.; Smital, T. Presence of Ecotoxicologically Relevant Pgp and MRP Transcripts and Proteins in Cyprinid Fish. Arch. Ind. Hyg. Toxicol. 2010, 61, 175–182. [Google Scholar] [CrossRef] [PubMed]
  48. Zucchi, S.; Corsi, I.; Luckenbach, T.; Bard, S.M.; Regoli, F.; Focardi, S. Identification of Five Partial ABC Genes in the Liver of the Antarctic Fish Trematomus Bernacchii and Sensitivity of ABCB1 and ABCC2 to Cd Exposure. Environ. Pollut. 2010, 158, 2746–2756. [Google Scholar] [CrossRef] [PubMed]
  49. Saeed, M.E.M.; Boulos, J.C.; Machel, K.; Andabili, N.; Marouni, T.; Roth, W.; Efferth, T. Expression of the Stem Cell Marker ABCB5 in Normal and Tumor Tissues. In Vivo 2022, 36, 1651–1666. [Google Scholar] [CrossRef] [PubMed]
  50. Tutundjian, R.; Cachot, J.; Leboulenger, F.; Minier, C. Genetic and Immunological Characterisation of a Multixenobiotic Resistance System in the Turbot (Scophthalmus maximus). Comp. Biochem. Physiol. B Biochem. Mol. Biol. 2002, 132, 463–471. [Google Scholar] [CrossRef] [PubMed]
  51. Yuan, L.; Lv, B.; Zha, J.; Wang, Z. Transcriptional Expression Analysis of ABC Efflux Transporters and Xenobiotic-Metabolizing Enzymes in the Chinese Rare Minnow. Environ. Toxicol. Pharmacol. 2014, 37, 984–995. [Google Scholar] [CrossRef] [PubMed]
  52. Lu, X.; Long, Y.; Sun, R.; Zhou, B.; Lin, L.; Zhong, S.; Cui, Z. Zebrafish Abcb4 Is a Potential Efflux Transporter of Microcystin-LR. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2015, 167, 35–42. [Google Scholar] [CrossRef]
  53. Romersi, R.F.; Nicklisch, S.C.T. Interactions of Environmental Chemicals and Natural Products with ABC and SLC Transporters in the Digestive System of Aquatic Organisms. Front. Physiol. 2022, 12, 2252. [Google Scholar] [CrossRef]
  54. Sturm, A.; Ziemann, C.; Hirsch-Ernst, K.I.; Segner, H. Expression and Functional Activity of P-Glycoprotein in Cultured Hepatocytes from Oncorhynchus mykiss. Am. J. Physiol.-Regul. Integr. Comp. Physiol. 2001, 281, R1119–R1126. [Google Scholar] [CrossRef]
  55. Bains, O.S.; Kennedy, C.J. Alterations in Respiration Rate of Isolated Rainbow Trout Hepatocytes Exposed to the P-Glycoprotein Substrate Rhodamine 123. Toxicology 2005, 214, 87–98. [Google Scholar] [CrossRef] [PubMed]
  56. Hildebrand, J.L.; Bains, O.S.; Lee, D.S.H.; Kennedy, C.J. Functional and Energetic Characterization of P-Gp-Mediated Doxorubicin Transport in Rainbow Trout (Oncorhynchus mykiss) Hepatocytes. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2009, 149, 65–72. [Google Scholar] [CrossRef] [PubMed]
  57. Johnston, C.U.; Kennedy, C.J. Effects of the Chemosensitizer Verapamil on P-Glycoprotein Substrate Efflux in Rainbow Trout Hepatocytes. Comp. Biochem. Physiol. C Pharmacol. Toxicol. Endocrinol. 2024, 275, 109763. [Google Scholar] [CrossRef] [PubMed]
  58. Miller, D.S. Daunomycin Secretion by Killfish Renal Proximal Tubules. Am. J. Physiol.-Regul. Integr. Comp. Physiol. 1995, 269, R370–R379. [Google Scholar] [CrossRef] [PubMed]
  59. Schramm, U.; Fricker, G.; Wenger, R.; Miller, D.S. P-Glycoprotein-Mediated Secretion of a Fluorescent Cyclosporin Analogue by Teleost Renal Proximal Tubules. Am. J. Physiol.-Ren. Physiol. 1995, 268, F46–F52. [Google Scholar] [CrossRef]
  60. Miller, D.S. Aquatic Models for the Study of Renal Transport Function and Pollutant Toxicity. Environ. Health Perspect. 1987, 71, 59–68. [Google Scholar] [CrossRef] [PubMed]
  61. Miller, D.S.; Fricker, G.; Drewe, J. P-Glycoprotein-Mediated Transport of a Fluorescent Rapamycin Derivative in Renal Proximal Tubule. J. Pharmacol. Exp. Ther. 1997, 282, 440–444. [Google Scholar]
  62. Fricker, G.; Gutmann, H.; Droulle, A.; Drewe, J.; Miller, D.S. Epithelial Transport of Anthelmintic Ivermectin in a Novel Model of Isolated Proximal Kidney Tubules. Pharm. Res. 1999, 16, 1570–1575. [Google Scholar] [CrossRef]
  63. Miller, D.S. Sphingolipid Signaling Reduces Basal P-Glycoprotein Activity in Renal Proximal Tubule. J. Pharmacol. Exp. Ther. 2014, 348, 459–464. [Google Scholar] [CrossRef]
  64. Bard, S.M. Multixenobiotic Resistance as a Cellular Defense Mechanism in Aquatic Organisms. Aquat. Toxicol. 2000, 48, 357–389. [Google Scholar] [CrossRef]
  65. Diaz de Cerio, O.; Bilbao, E.; Cajaraville, M.P.; Cancio, I. Regulation of Xenobiotic Transporter Genes in Liver and Brain of Juvenile Thicklip Grey Mullets (Chelon labrosus) after Exposure to Prestige-like Fuel Oil and to Perfluorooctane Sulfonate. Gene 2012, 498, 50–58. [Google Scholar] [CrossRef] [PubMed]
  66. Hemmer, M.J.; Courtney, L.A.; Benson, W.H. Comparison of Three Histological Fixatives on the Immunoreactivity of Mammalian P-Glycoprotein Antibodies in the Sheepshead Minnow, Cyprinodon Variegatus. J. Exp. Zool. 1998, 281, 251–259. [Google Scholar] [CrossRef]
  67. Bieczynski, F.; Burkhardt-Medicke, K.; Luquet, C.M.; Scholz, S.; Luckenbach, T. Chemical Effects on Dye Efflux Activity in Live Zebrafish Embryos and on Zebrafish Abcb4 ATPase Activity. FEBS Lett. 2021, 595, 828–843. [Google Scholar] [CrossRef] [PubMed]
  68. Gordon, W.E.; Espinoza, J.A.; Leerberg, D.M.; Yelon, D.; Hamdoun, A. Xenobiotic Transporter Activity in Zebrafish Embryo Ionocytes. Aquat. Toxicol. 2019, 212, 88–97. [Google Scholar] [CrossRef] [PubMed]
  69. Robey, R.W.; Robinson, A.N.; Ali-Rahmani, F.; Huff, L.M.; Lusvarghi, S.; Vahedi, S.; Hotz, J.M.; Warner, A.C.; Butcher, D.; Matta, J.; et al. Characterization and Tissue Localization of Zebrafish Homologs of the Human ABCB1 Multidrug Transporter. Sci. Rep. 2021, 11, 24150. [Google Scholar] [CrossRef] [PubMed]
  70. Bard, S.M.; Bello, S.M.; Hahn, M.E.; Stegeman, J.J. Expression of P-Glycoprotein in Killifish (Fundulus heteroclitus) Exposed to Environmental Xenobiotics. Aquat. Toxicol. 2002, 59, 237–251. [Google Scholar] [CrossRef] [PubMed]
  71. Miller, D.S.; Graeff, C.; Droulle, L.; Fricker, S.; Fricker, G. Xenobiotic Efflux Pumps in Isolated Fish Brain Capillaries. Am. J. Physiol.-Regul. Integr. Comp. Physiol. 2002, 282, R191–R198. [Google Scholar] [CrossRef] [PubMed]
  72. Cooper, P.S.; Vogelbein, W.K.; Van Veld, P.A. Altered Expression of the Xenobiotic Transporter P-Glycoprotein in Liver and Liver Tumours of Mummichog Fundulus heteroclitus from a Creosote-Contaminated Environment. Biomarkers 1999, 4, 48–58. [Google Scholar] [CrossRef]
  73. Doi, A.M.; Holmes, E.; Kleinow, K.M. P-Glycoprotein in the Catfish Intestine: Inducibility by Xenobiotics and Functional Properties. Aquat. Toxicol. 2001, 55, 157–170. [Google Scholar] [CrossRef]
  74. Amé, M.V.; Baroni, M.V.; Galanti, L.N.; Bocco, J.L.; Wunderlin, D.A. Effects of Microcystin–LR on the Expression of P-Glycoprotein in Jenynsia Multidentata. Chemosphere 2009, 74, 1179–1186. [Google Scholar] [CrossRef] [PubMed]
  75. Kropf, C.; Segner, H.; Fent, K. ABC Transporters and Xenobiotic Defense Systems in Early Life Stages of Rainbow Trout (Oncorhynchus mykiss). Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2016, 185–186, 45–56. [Google Scholar] [CrossRef] [PubMed]
  76. Shúilleabháin, S.N.; Davoren, M.; Mothersill, C.; Sheehan, D.; Hartl, M.G.J.; Kilemade, M.; O’Brien, N.M.; O’Halloran, J.; Van Pelt, F.N.A.M.; Lyng, F.M. Identification of a Multixenobiotic Resistance Mechanism in Primary Cultured Epidermal Cells from Oncorhynchus mykiss and the Effects of Environmental Complex Mixtures on Its Activity. Aquat. Toxicol. 2005, 73, 115–127. [Google Scholar] [CrossRef] [PubMed]
  77. Sturm, A.; Cravedi, J.P.; Segner, H. Prochloraz and Nonylphenol Diethoxylate Inhibit an Mdr1-like Activity in Vitro, but Do Not Alter Hepatic Levels of P-Glycoprotein in Trout Exposed In Vivo. Aquat. Toxicol. 2001, 53, 215–228. [Google Scholar] [CrossRef] [PubMed]
  78. Zaja, R.; Munić, V.; Klobučar, R.S.; Ambriović-Ristov, A.; Smital, T. Cloning and Molecular Characterization of Apical Efflux Transporters (ABCB1, ABCB11 and ABCC2) in Rainbow Trout (Oncorhynchus mykiss) Hepatocytes. Aquat. Toxicol. 2008, 90, 322–332. [Google Scholar] [CrossRef] [PubMed]
  79. Costa, J.; Reis-Henriques, M.A.; Castro, L.F.C.; Ferreira, M. ABC Transporters, CYP1A and GSTα Gene Transcription Patterns in Developing Stages of the Nile Tilapia (Oreochromis niloticus). Gene 2012, 506, 317–324. [Google Scholar] [CrossRef] [PubMed]
  80. Costa, J.; Reis-Henriques, M.A.; Castro, L.F.C.; Ferreira, M. Gene Expression Analysis of ABC Efflux Transporters, CYP1A and GSTα in Nile Tilapia after Exposure to Benzo(a)Pyrene. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2012, 155, 469–482. [Google Scholar] [CrossRef]
  81. Hemmer, M.J.; Courtney, L.A.; Ortego, L.S. Immunohistochemical Detection of P-Glycoprotein in Teleost Tissues Using Mammalian Polyclonal and Monoclonal Antibodies. J. Exp. Zool. 1995, 272, 69–77. [Google Scholar] [CrossRef]
  82. Azevedo, V.C.; Kennedy, C.J. P-Glycoprotein Inhibition Affects Ivermectin-Induced Behavioural Alterations in Fed and Fasted Zebrafish (Danio Rerio). Fish Physiol. Biochem. 2022, 48, 1267–1283. [Google Scholar] [CrossRef]
  83. Bard, S.M.; Gadbois, S. Assessing Neuroprotective P-Glycoprotein Activity at the Blood-Brain Barrier in Killifish (Fundulus heteroclitus) Using Behavioural Profiles. Mar. Environ. Res. 2007, 64, 679–682. [Google Scholar] [CrossRef]
  84. Kennedy, C.J.; Tierney, K.B.; Mittelstadt, M. Inhibition of P-Glycoprotein in the Blood–Brain Barrier Alters Avermectin Neurotoxicity and Swimming Performance in Rainbow Trout. Aquat. Toxicol. 2014, 146, 176–185. [Google Scholar] [CrossRef]
  85. Azevedo, V.C.; Johnston, C.U.; Kennedy, C.J. Ivermectin Toxicokinetics in Rainbow Trout (Oncorhynchus mykiss) Following P-Glycoprotein Inhibition. Arch. Environ. Contam. Toxicol. 2024; manuscrript submitted. [Google Scholar] [CrossRef]
  86. Cserr, H.F.; Bundgaard, M. Blood-Brain Interfaces in Vertebrates: A Comparative Approach. Am. J. Physiol.-Regul. Integr. Comp. Physiol. 1984, 246, R277–R288. [Google Scholar] [CrossRef]
  87. Kajikawa, T.; Mishima, H.; Mishima, H.; Murakami, T.; Takano, M. Role of P-Glycoprotein in Distribution of Rhodamine 123 into Aqueous Humor in Rabbits. Curr. Eye Res. 1999, 18, 240–246. [Google Scholar] [CrossRef]
  88. Chen, L. Visual System: An Understudied Target of Aquatic Toxicology. Aquat. Toxicol. 2020, 225, 105542. [Google Scholar] [CrossRef] [PubMed]
  89. Guerreiro, D.D.; de Lima, L.F.; Mbemya, G.T.; Maside, C.M.; Miranda, A.M.; Tavares, K.C.S.; Alves, B.G.; Faustino, L.R.; Smitz, J.; de Figueiredo, J.R.; et al. ATP-Binding Cassette (ABC) Transporters in Caprine Preantral Follicles: Gene and Protein Expression. Cell Tissue Res. 2018, 372, 611–620. [Google Scholar] [CrossRef] [PubMed]
  90. Su, L.; Cheng, C.Y.; Mruk, D.D. Drug Transporter, P-Glycoprotein (MDR1), Is an Integrated Component of the Mammalian Blood–Testis Barrier. Int. J. Biochem. Cell Biol. 2009, 41, 2578–2587. [Google Scholar] [CrossRef]
  91. Evans, D.H.; Piermarini, P.M.; Choe, K.P. The Multifunctional Fish Gill: Dominant Site of Gas Exchange, Osmoregulation, Acid-Base Regulation, and Excretion of Nitrogenous Waste. Physiol. Rev. 2005, 85, 97–177. [Google Scholar] [CrossRef] [PubMed]
  92. 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]
  93. Cunha, V.; Rodrigues, P.; Santos, M.M.; Moradas-Ferreira, P.; Ferreira, M. Danio Rerio Embryos on Prozac—Effects on the Detoxification Mechanism and Embryo Development. Aquat. Toxicol. 2016, 178, 182–189. [Google Scholar] [CrossRef]
  94. Karmakar, S.; Sen Gupta, P.; Bhattacharya, S.; Sarkar, A.; Rahaman, A.; Mandal, D.P.; Bhattacharjee, S. Vitamin B12 Alleviates Malathion-Induced Toxicity in Zebra Fish by Regulating Cytochrome P450 and PgP Expressions. Toxicol. Mech. Methods 2022, 33, 364–377. [Google Scholar] [CrossRef]
  95. Meinan, X.; Yimeng, W.; Chao, W.; Tianli, T.; Li, J.; Peng, Y.; Xiangping, N. Response of the Sirtuin/PXR Signaling Pathway in Mugilogobius Chulae Exposed to Environmentally Relevant Concentration Paracetamol. Aquat. Toxicol. 2022, 249, 106222. [Google Scholar] [CrossRef]
  96. Vannuccini, M.L.; Grassi, G.; Leaver, M.J.; Corsi, I. Combination Effects of Nano-TiO2 and 2,3,7,8-Tetrachlorodibenzo-p-Dioxin (TCDD) on Biotransformation Gene Expression in the Liver of European Sea Bass Dicentrarchus Labrax. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2015, 176–177, 71–78. [Google Scholar] [CrossRef]
  97. Bolten, J.S.; Pratsinis, A.; Alter, C.L.; Fricker, G.; Huwyler, J. Zebrafish (Danio Rerio) Larva as an in Vivo Vertebrate Model to Study Renal Function. Am. J. Physiol.-Ren. Physiol. 2022, 322, F280–F294. [Google Scholar] [CrossRef]
  98. Muzzio, A.M.; Noyes, P.D.; Stapleton, H.M.; Lema, S.C. Tissue Distribution and Thyroid Hormone Effects on mRNA Abundance for Membrane Transporters Mct8, Mct10, and Organic Anion-Transporting Polypeptides (Oatps) in a Teleost Fish. Comp. Biochem. Physiol. A Mol. Integr. Physiol. 2014, 167, 77–89. [Google Scholar] [CrossRef]
  99. Widhalm, J.R.; Jaini, R.; Morgan, J.A.; Dudareva, N. Rethinking How Volatiles Are Released from Plant Cells. Trends Plant Sci. 2015, 20, 545–550. [Google Scholar] [CrossRef] [PubMed]
  100. Mihaljević, I.; Popović, M.; Žaja, R.; Maraković, N.; Šinko, G.; Smital, T. Interaction between the Zebrafish (Danio Rerio) Organic Cation Transporter 1 (Oct1) and Endo- and Xenobiotics. Aquat. Toxicol. 2017, 187, 18–28. [Google Scholar] [CrossRef] [PubMed]
  101. Popovic, M.; Zaja, R.; Fent, K.; Smital, T. Interaction of Environmental Contaminants with Zebrafish Organic Anion Transporting Polypeptide, Oatp1d1 (Slco1d1). Toxicol. Appl. Pharmacol. 2014, 280, 149–158. [Google Scholar] [CrossRef] [PubMed]
  102. Willi, R.A.; Fent, K. Interaction of Environmental Steroids with Organic Anion Transporting Polypeptide (Oatp1d1) in Zebrafish (Danio Rerio). Environ. Toxicol. Chem. 2018, 37, 2670–2676. [Google Scholar] [CrossRef] [PubMed]
  103. Bresolin, T.; de Freitas Rebelo, M.; Celso Dias Bainy, A. Expression of PXR, CYP3A and MDR1 Genes in Liver of Zebrafish. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2005, 140, 403–407. [Google Scholar] [CrossRef] [PubMed]
  104. Jackson, J.S.; Kennedy, C.J. Regulation of Hepatic Abcb4 and Cyp3a65 Gene Expression and Multidrug/Multixenobiotic Resistance (MDR/MXR) Functional Activity in the Model Teleost, Danio Rerio (Zebrafish). Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2017, 200, 34–41. [Google Scholar] [CrossRef] [PubMed]
  105. Reschly, E.J.; Bainy, A.C.D.; Mattos, J.J.; Hagey, L.R.; Bahary, N.; Mada, S.R.; Ou, J.; Venkataramanan, R.; Krasowski, M.D. Functional Evolution of the Vitamin D and Pregnane X Receptors. BMC Evol. Biol. 2007, 7, 222. [Google Scholar] [CrossRef] [PubMed]
  106. De Anna, J.S.; Darraz, L.A.; Painefilú, J.C.; Cárcamo, J.G.; Moura-Alves, P.; Venturino, A.; Luquet, C.M. The Insecticide Chlorpyrifos Modifies the Expression of Genes Involved in the PXR and AhR Pathways in the Rainbow Trout, Oncorhynchus mykiss. Pestic. Biochem. Physiol. 2021, 178, 104920. [Google Scholar] [CrossRef]
  107. Mealey, K.L.; Owens, J.G.; Freeman, E. Canine and Feline P-Glycoprotein Deficiency: What We Know and Where We Need to Go. J. Vet. Pharmacol. Ther. 2023, 46, 1–16. [Google Scholar] [CrossRef]
  108. Borst, P.; Schinkel, A.H. P-Glycoprotein ABCB1: A Major Player in Drug Handling by Mammals. J. Clin. Investig. 2013, 123, 4131–4133. [Google Scholar] [CrossRef]
  109. Aller, S.G.; Yu, J.; Ward, A.; Weng, Y.; Chittaboina, S.; Zhuo, R.; Harrell, P.M.; Trinh, Y.T.; Zhang, Q.; Urbatsch, I.L.; et al. Structure of P-Glycoprotein Reveals a Molecular Basis for Poly-Specific Drug Binding. Science 2009, 323, 1718–1722. [Google Scholar] [CrossRef]
  110. Nosol, K.; Romane, K.; Irobalieva, R.N.; Alam, A.; Kowal, J.; Fujita, N.; Locher, K.P. Cryo-EM Structures Reveal Distinct Mechanisms of Inhibition of the Human Multidrug Transporter ABCB1. Proc. Natl. Acad. Sci. USA 2020, 117, 26245–26253. [Google Scholar] [CrossRef]
  111. Löscher, W.; Gericke, B. Novel Intrinsic Mechanisms of Active Drug Extrusion at the Blood-Brain Barrier: Potential Targets for Enhancing Drug Delivery to the Brain? Pharmaceutics 2020, 12, 966. [Google Scholar] [CrossRef]
  112. Srikant, S.; Gaudet, R. Mechanics and Pharmacology of Substrate Selection and Transport by Eukaryotic ABC Exporters. Nat. Struct. Mol. Biol. 2019, 26, 792–801. [Google Scholar] [CrossRef]
  113. Zaja, R.; Lončar, J.; Popovic, M.; Smital, T. First Characterization of Fish P-Glycoprotein (Abcb1) Substrate Specificity Using Determinations of Its ATPase Activity and Calcein-AM Assay with PLHC-1/Dox Cell Line. Aquat. Toxicol. 2011, 103, 53–62. [Google Scholar] [CrossRef]
  114. Johnston, C.U.; Kennedy, C.J. Potency and Mechanism of P-Glycoprotein Chemosensitizers in Rainbow Trout (Oncorhynchus mykiss) Hepatocytes. Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada. 2024; manuscrript submitted. [Google Scholar]
  115. Gutmann, H.; Miller, D.S.; Droulle, A.; Drewe, J.; Fahr, A.; Fricker, G. P-Glycoprotein- and Mrp2-Mediated Octreotide Transport in Renal Proximal Tubule: Octreotide Transport. Br. J. Pharmacol. 2000, 129, 251–256. [Google Scholar] [CrossRef]
  116. Mottaz, H.; Schönenberger, R.; Fischer, S.; Eggen, R.I.L.; Schirmer, K.; Groh, K.J. Dose-Dependent Effects of Morphine on Lipopolysaccharide (LPS)-Induced Inflammation, and Involvement of Multixenobiotic Resistance (MXR) Transporters in LPS Efflux in Teleost Fish. Environ. Pollut. 2017, 221, 105–115. [Google Scholar] [CrossRef]
  117. Caminada, D.; Zaja, R.; Smital, T.; Fent, K. Human Pharmaceuticals Modulate P-Gp1 (ABCB1) Transport Activity in the Fish Cell Line PLHC-1. Aquat. Toxicol. 2008, 90, 214–222. [Google Scholar] [CrossRef]
  118. Fischer, S.; Loncar, J.; Zaja, R.; Schnell, S.; Schirmer, K.; Smital, T.; Luckenbach, T. Constitutive mRNA Expression and Protein Activity Levels of Nine ABC Efflux Transporters in Seven Permanent Cell Lines Derived from Different Tissues of Rainbow Trout (Oncorhynchus mykiss). Aquat. Toxicol. 2011, 101, 438–446. [Google Scholar] [CrossRef]
  119. Zaja, R.; Klobučar, R.S.; Smital, T. Detection and Functional Characterization of Pgp1 (ABCB1) and MRP3 (ABCC3) Efflux Transporters in the PLHC-1 Fish Hepatoma Cell Line. Aquat. Toxicol. 2007, 81, 365–376. [Google Scholar] [CrossRef]
  120. Uchea, C.; Owen, S.F.; Chipman, J.K. Functional Xenobiotic Metabolism and Efflux Transporters in Trout Hepatocyte Spheroid Cultures. Toxicol. Res. 2015, 4, 494–507. [Google Scholar] [CrossRef] [PubMed]
  121. Smital, T.; Sauerborn, R. Measurement of the Activity of Multixenobiotic Resistance Mechanism in the Common Carp Cyprinus Carpio. Mar. Environ. Res. 2002, 54, 449–453. [Google Scholar] [CrossRef] [PubMed]
  122. Hu, J.; Tian, J.; Zhang, F.; Wang, H.; Yin, J. Pxr- and Nrf2- Mediated Induction of ABC Transporters by Heavy Metal Ions in Zebrafish Embryos. Environ. Pollut. 2019, 255, 113329. [Google Scholar] [CrossRef] [PubMed]
  123. Chen, Q.; Hu, X.; Wang, R.; Yuan, J.; Yin, D. Fullerene Inhibits Benzo(a)Pyrene Efflux from Cyprinus Carpio Hepatocytes by Affecting Cell Membrane Fluidity and P-Glycoprotein Expression. Aquat. Toxicol. 2016, 174, 36–45. [Google Scholar] [CrossRef] [PubMed]
  124. Cunha, V.; Burkhardt-Medicke, K.; Wellner, P.; Santos, M.M.; Moradas-Ferreira, P.; Luckenbach, T.; Ferreira, M. Effects of Pharmaceuticals and Personal Care Products (PPCPs) on Multixenobiotic Resistance (MXR) Related Efflux Transporter Activity in Zebrafish (Danio Rerio) Embryos. Ecotoxicol. Environ. Saf. 2017, 136, 14–23. [Google Scholar] [CrossRef] [PubMed]
  125. Nornberg, B.F.; Batista, C.R.; Almeida, D.V.; Trindade, G.S.; Marins, L.F. ABCB1 and ABCC4 Efflux Transporters Are Involved in Methyl Parathion Detoxification in ZFL Cells. Toxicol. In Vitro 2015, 29, 204–210. [Google Scholar] [CrossRef] [PubMed]
  126. Masereeuw, R.; Terlouw, S.A.; Aubel, R.A.M.H.v.; Russel, F.G.M.; Miller, D.S. Endothelin B Receptor-Mediated Regulation of ATP-Driven Drug Secretion in Renal Proximal Tubule. Mol. Pharmacol. 2000, 57, 59–67. [Google Scholar] [PubMed]
  127. Bourgeois, Z.M.; Comfort, J.; Schultz, M.; Challis, J.K.; Cantin, J.; Ji, X.; Giesy, J.P.; Brinkmann, M. Predicting Hepatic Clearance of Psychotropic Drugs in Isolated Perfused Fish Livers Using a Combination of Two In Vitro Assays. Environ. Sci. Technol. 2022, 56, 15839–15847. [Google Scholar] [CrossRef] [PubMed]
  128. Stott, L.C.; Schnell, S.; Hogstrand, C.; Owen, S.F.; Bury, N.R. A Primary Fish Gill Cell Culture Model to Assess Pharmaceutical Uptake and Efflux: Evidence for Passive and Facilitated Transport. Aquat. Toxicol. 2015, 159, 127–137. [Google Scholar] [CrossRef]
  129. Gutmann, H.; Fricker, G.; Drewe, J.; Toeroek, M.; Miller, D.S. Interactions of HIV Protease Inhibitors with ATP-Dependent Drug Export Proteins. Mol. Pharmacol. 1999, 56, 383–389. [Google Scholar] [CrossRef] [PubMed]
  130. Sussman-Turner, C.; Renfro, J.L. Heat-Shock-Stimulated Transepithelial Daunomycin Secretion by Flounder Renal Proximal Tubule Primary Cultures. Am. J. Physiol.-Ren. Physiol. 1995, 268, F135–F144. [Google Scholar] [CrossRef]
  131. Zhou, S.-F. Structure, Function and Regulation of P-Glycoprotein and Its Clinical Relevance in Drug Disposition. Xenobiotica 2008, 38, 802–832. [Google Scholar] [CrossRef]
  132. Yano, K.; Seto, S.; Kamioka, H.; Mizoi, K.; Ogihara, T. Testosterone and Androstenedione Are Endogenous Substrates of P-Glycoprotein. Biochem. Biophys. Res. Commun. 2019, 520, 166–170. [Google Scholar] [CrossRef]
  133. Abulrob, A.G.; Gumbleton, M. Transport of Phosphatidylcholine in MDR3-Negative Epithelial Cell Lines via Drug-Induced MDR1 P-Glycoprotein. Biochem. Biophys. Res. Commun. 1999, 262, 121–126. [Google Scholar] [CrossRef]
  134. Wang, E.; Casciano, C.N.; Clement, R.P.; Johnson, W.W. Two Transport Binding Sites of P-Glycoprotein Are Unequal yet Contingent: Initial Rate Kinetic Analysis by ATP Hydrolysis Demonstrates Intersite Dependence. Biochim. Biophys. Acta BBA—Protein Struct. Mol. Enzymol. 2000, 1481, 63–74. [Google Scholar] [CrossRef] [PubMed]
  135. Dagenais, C.; Ducharme, J.; Pollack, G.M. Uptake and Efflux of the Peptidic Delta-Opioid Receptor Agonist [D-Penicillamine2,5]-Enkephalin at the Murine Blood–Brain Barrier by In Situ Perfusion. Neurosci. Lett. 2001, 301, 155–158. [Google Scholar] [CrossRef] [PubMed]
  136. Patiño, R.; Sullivan, C.V. Ovarian Follicle Growth, Maturation, and Ovulation in Teleost Fish. Fish Physiol. Biochem. 2002, 26, 57–70. [Google Scholar] [CrossRef]
  137. Nagahama, Y.; Yamashita, M. Regulation of Oocyte Maturation in Fish. Dev. Growth Differ. 2008, 50, S195–S219. [Google Scholar] [CrossRef]
  138. Goss, G.G.; Perry, S.F.; Wood, C.M.; Laurent, P. Mechanisms of Ion and Acid-Base Regulation at the Gills of Freshwater Fish. J. Exp. Zool. 1992, 263, 143–159. [Google Scholar] [CrossRef]
  139. Frank, N.Y.; Pendse, S.S.; Lapchak, P.H.; Margaryan, A.; Shlain, D.; Doeing, C.; Sayegh, M.H.; Frank, M.H. Regulation of Progenitor Cell Fusion by ABCB5 P-Glycoprotein, a Novel Human ATP-Binding Cassette Transporter*. J. Biol. Chem. 2003, 278, 47156–47165. [Google Scholar] [CrossRef]
  140. Mirzaei, S.; Gholami, M.H.; Hashemi, F.; Zabolian, A.; Farahani, M.V.; Hushmandi, K.; Zarrabi, A.; Goldman, A.; Ashrafizadeh, M.; Orive, G. Advances in Understanding the Role of P-Gp in Doxorubicin Resistance: Molecular Pathways, Therapeutic Strategies, and Prospects. Drug Discov. Today 2022, 27, 436–455. [Google Scholar] [CrossRef] [PubMed]
  141. Chan, G.N.Y.; Hoque, M.T.; Bendayan, R. Role of Nuclear Receptors in the Regulation of Drug Transporters in the Brain. Trends Pharmacol. Sci. 2013, 34, 361–372. [Google Scholar] [CrossRef]
  142. Wang, Z.; Liu, Y.; Ai, X.; Zhong, L.; Han, G.; Song, J.; Yang, Q.; Dong, J. Effects of 27 Natural Products on Drug Metabolism Genes in Channel Catfish (Ictalurus punctatus) Cell Line. Xenobiotica 2020, 50, 1043–1051. [Google Scholar] [CrossRef]
  143. 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]
  144. Wu, B.; Li, H.-X.; Lian, J.; Guo, Y.-J.; Tang, Y.-H.; Chang, Z.-J.; Hu, L.-F.; Zhao, G.-J.; Hong, G.-L.; Lu, Z.-Q. Nrf2 Overexpression Protects against Paraquat-induced A549 Cell Injury Primarily by Upregulating P-glycoprotein and Reducing Intracellular Paraquat Accumulation. Exp. Ther. Med. 2019, 17, 1240–1247. [Google Scholar] [CrossRef] [PubMed]
  145. Baumgarner, B.L.; Bharadwaj, A.S.; Inerowicz, D.; Goodman, A.S.; Brown, P.B. Proteomic Analysis of Rainbow Trout (Oncorhynchus mykiss) Intestinal Epithelia: Physiological Acclimation to Short-Term Starvation. Comp. Biochem. Physiol. Part D Genomics Proteomics 2013, 8, 58–64. [Google Scholar] [CrossRef] [PubMed]
  146. Wassmur, B.; Gräns, J.; Kling, P.; Celander, M.C. Interactions of Pharmaceuticals and Other Xenobiotics on Hepatic Pregnane X Receptor and Cytochrome P450 3A Signaling Pathway in Rainbow Trout (Oncorhynchus mykiss). Aquat. Toxicol. 2010, 100, 91–100. [Google Scholar] [CrossRef] [PubMed]
  147. Monserrat, J.M.; Garcia, M.L.; Ventura-Lima, J.; González, M.; Ballesteros, M.L.; Miglioranza, K.S.B.; Amé, M.V.; Wunderlin, D.A. Antioxidant, Phase II and III Responses Induced by Lipoic Acid in the Fish Jenynsia Multidentata (Anablapidae) and Its Influence on Endolsulfan Accumulation and Toxicity. Pestic. Biochem. Physiol. 2014, 108, 8–15. [Google Scholar] [CrossRef] [PubMed]
  148. Della Torre, C.; Zaja, R.; Loncar, J.; Smital, T.; Focardi, S.; Corsi, I. Interaction of ABC Transport Proteins with Toxic Metals at the Level of Gene and Transport Activity in the PLHC-1 Fish Cell Line. Chem. Biol. Interact. 2012, 198, 9–17. [Google Scholar] [CrossRef] [PubMed]
  149. Della Torre, C.; Mariottini, M.; Vannuccini, M.L.; Trisciani, A.; Marchi, D.; Corsi, I. Induction of CYP1A and ABC Transporters in European Sea Bass (Dicentrarchus Labrax) upon 2,3,7,8-TCDD Waterborne Exposure. Mar. Environ. Res. 2014, 99, 218–222. [Google Scholar] [CrossRef] [PubMed]
  150. Götte, J.Y.; Carrizo, J.C.; Panzeri, A.M.; Amé, M.V.; Menone, M.L. Sublethal Effects of Carbendazim in Jenynsia Multidentata Detected by a Battery of Molecular, Biochemical and Genetic Biomarkers. Ecotoxicol. Environ. Saf. 2020, 205, 111157. [Google Scholar] [CrossRef]
  151. Bonansea, R.I.; Marino, D.J.G.; Bertrand, L.; Wunderlin, D.A.; Amé, M.V. Tissue-Specific Bioconcentration and Biotransformation of Cypermethrin and Chlorpyrifos in a Native Fish (Jenynsia Multidentata) Exposed to These Insecticides Singly and in Mixtures. Environ. Toxicol. Chem. 2017, 36, 1764–1774. [Google Scholar] [CrossRef]
  152. Cárcamo, J.G.; Aguilar, M.N.; Barrientos, C.A.; Carreño, C.F.; Quezada, C.A.; Bustos, C.; Manríquez, R.A.; Avendaño-Herrera, R.; Yañez, A.J. Effect of Emamectin Benzoate on Transcriptional Expression of Cytochromes P450 and the Multidrug Transporters (Pgp and MRP1) in Rainbow Trout (Oncorhynchus mykiss) and the Sea Lice Caligus Rogercresseyi. Aquaculture 2011, 321, 207–215. [Google Scholar] [CrossRef]
  153. Liang, X.; Nie, X.; Ying, G.; An, T.; Li, K. Assessment of Toxic Effects of Triclosan on the Swordtail Fish (Xiphophorus helleri) by a Multi-Biomarker Approach. Chemosphere 2013, 90, 1281–1288. [Google Scholar] [CrossRef]
  154. Ren, T.; Fu, G.-H.; Liu, T.-F.; Hu, K.; Li, H.-R.; Fang, W.-H.; Yang, X.-L. Toxicity and Accumulation of Zinc Pyrithione in the Liver and Kidneys of Carassius Auratus Gibelio: Association with P-Glycoprotein Expression. Fish Physiol. Biochem. 2017, 43, 1–9. [Google Scholar] [CrossRef] [PubMed]
  155. Bao, S.; Nie, X.; Liu, Y.; Wang, C.; Li, W.; Liu, S. Diclofenac Exposure Alter the Expression of PXR and Its Downstream Target Genes in Mosquito Fish (Gambusia affinis). Sci. Total Environ. 2018, 616–617, 583–593. [Google Scholar] [CrossRef]
  156. Liang, X.; Wang, L.; Ou, R.; Nie, X.; Yang, Y.; Wang, F.; Li, K. Effects of Norfloxacin on Hepatic Genes Expression of P450 Isoforms (CYP1A and CYP3A), GST and P-Glycoprotein (P-Gp) in Swordtail Fish (Xiphophorus helleri). Ecotoxicology 2015, 24, 1566–1573. [Google Scholar] [CrossRef] [PubMed]
  157. Weltje, L.; Simpson, P.; Gross, M.; Crane, M.; Wheeler, J.R. Comparative Acute and Chronic Sensitivity of Fish and Amphibians: A Critical Review of Data. Environ. Toxicol. Chem. 2013, 32, 984–994. [Google Scholar] [CrossRef]
  158. Wassmur, B.; Gräns, J.; Norström, E.; Wallin, M.; Celander, M.C. Interactions of Pharmaceuticals and Other Xenobiotics on Key Detoxification Mechanisms and Cytoskeleton in Poeciliopsis Lucida Hepatocellular Carcinoma, PLHC-1 Cell Line. Toxicol. In Vitro 2013, 27, 111–120. [Google Scholar] [CrossRef]
  159. Macêdo, A.K.S.; Santos, K.P.E.d.; Brighenti, L.S.; Windmöller, C.C.; Barbosa, F.A.R.; Ribeiro, R.I.M.d.A.; Santos, H.B.d.; Thomé, R.G. Histological and Molecular Changes in Gill and Liver of Fish (Astyanax Lacustris Lütken, 1875) Exposed to Water from the Doce Basin after the Rupture of a Mining Tailings Dam in Mariana, MG, Brazil. Sci. Total Environ. 2020, 735, 139505. [Google Scholar] [CrossRef]
  160. Kurth, D.; Brack, W.; Luckenbach, T. Is Chemosensitisation by Environmental Pollutants Ecotoxicologically Relevant? Aquat. Toxicol. 2015, 167, 134–142. [Google Scholar] [CrossRef] [PubMed]
  161. Faria, M.; Pavlichenko, V.; Burkhardt-Medicke, K.; Soares, A.M.V.M.; Altenburger, R.; Barata, C.; Luckenbach, T. Use of a Combined Effect Model Approach for Discriminating between ABCB1- and ABCC1-Type Efflux Activities in Native Bivalve Gill Tissue. Toxicol. Appl. Pharmacol. 2016, 297, 56–67. [Google Scholar] [CrossRef]
  162. Müller, W.E.G.; Riemer, S.; Kurelec, B.; Smodlaka, N.; Puskaric, S.; Jagic, B.; Müller-Niklas, G.; Queric, N.V. Chemosensitizers of the Multixenobiotic Resistance in Amorphous Aggregates (Marine Snow): Etiology of Mass Killing on the Benthos in the Northern Adriatic? Environ. Toxicol. Pharmacol. 1998, 6, 229–238. [Google Scholar] [CrossRef]
  163. Smital, T.; Luckenbach, T.; Sauerborn, R.; Hamdoun, A.M.; Vega, R.L.; Epel, D. Emerging Contaminants—Pesticides, PPCPs, Microbial Degradation Products and Natural Substances as Inhibitors of Multixenobiotic Defense in Aquatic Organisms. Mutat. Res. Mol. Mech. Mutagen. 2004, 552, 101–117. [Google Scholar] [CrossRef]
  164. Wu, B.; Torres-Duarte, C.; Cole, B.J.; Cherr, G.N. Copper Oxide and Zinc Oxide Nanomaterials Act as Inhibitors of Multidrug Resistance Transport in Sea Urchin Embryos: Their Role as Chemosensitizers. Environ. Sci. Technol. 2015, 49, 5760–5770. [Google Scholar] [CrossRef] [PubMed]
  165. Varma, M.V.; Pang, K.S.; Isoherranen, N.; Zhao, P. Dealing with the Complex Drug–Drug Interactions: Towards Mechanistic Models. Biopharm. Drug Dispos. 2015, 36, 71–92. [Google Scholar] [CrossRef] [PubMed]
  166. Bieczynski, F.; De Anna, J.S.; Pirez, M.; Brena, B.M.; Villanueva, S.S.M.; Luquet, C.M. Cellular Transport of Microcystin-LR in Rainbow Trout (Oncorhynchus mykiss) across the Intestinal Wall: Possible Involvement of Multidrug Resistance-Associated Proteins. Aquat. Toxicol. 2014, 154, 97–106. [Google Scholar] [CrossRef] [PubMed]
  167. Alharbi, H.A.; Saunders, D.M.V.; Al-Mousa, A.; Alcorn, J.; Pereira, A.S.; Martin, J.W.; Giesy, J.P.; Wiseman, S.B. Inhibition of ABC Transport Proteins by Oil Sands Process Affected Water. Aquat. Toxicol. 2016, 170, 81–88. [Google Scholar] [CrossRef] [PubMed]
  168. Zaja, R.; Terzić, S.; Senta, I.; Lončar, J.; Popović, M.; Ahel, M.; Smital, T. Identification of P-Glycoprotein Inhibitors in Contaminated Freshwater Sediments. Environ. Sci. Technol. 2013, 47, 4813–4821. [Google Scholar] [CrossRef]
  169. Tan, X.; Yim, S.-Y.; Uppu, P.; Kleinow, K.M. Enhanced Bioaccumulation of Dietary Contaminants in Catfish with Exposure to the Waterborne Surfactant Linear Alkylbenzene Sulfonate. Aquat. Toxicol. 2010, 99, 300–308. [Google Scholar] [CrossRef]
  170. Keiter, S.; Burkhardt-Medicke, K.; Wellner, P.; Kais, B.; Färber, H.; Skutlarek, D.; Engwall, M.; Braunbeck, T.; Keiter, S.H.; Luckenbach, T. Does Perfluorooctane Sulfonate (PFOS) Act as Chemosensitizer in Zebrafish Embryos? Sci. Total Environ. 2016, 548–549, 317–324. [Google Scholar] [CrossRef]
  171. Zaja, R.; Caminada, D.; Lončar, J.; Fent, K.; Smital, T. Development and Characterization of P-Glycoprotein 1 (Pgp1, ABCB1)-Mediated Doxorubicin-Resistant PLHC-1 Hepatoma Fish Cell Line. Toxicol. Appl. Pharmacol. 2008, 227, 207–218. [Google Scholar] [CrossRef]
  172. Tutundjian, R.; Minier, C.; Le Foll, F.; Leboulenger, F. Rhodamine Exclusion Activity in Primary Cultured Turbot (Scophthalmus maximus) Hepatocytes. Mar. Environ. Res. 2002, 54, 443–447. [Google Scholar] [CrossRef] [PubMed]
  173. Doppenschmitt, S.; Spahn-Langguth, H.; Regårdh, C.G.; Langguth, P. Role of P-glycoprotein-mediated Secretion in Absorptive Drug Permeabiity: An Approach Using Passive Membrane Permeability and Affinity to P-glycoprotein††Dedicated to Prof. B. C. Lippold on the Occasion of His 60th Birthday. J. Pharm. Sci. 1999, 88, 1067–1072. [Google Scholar] [CrossRef] [PubMed]
  174. von Richter, O.; Glavinas, H.; Krajcsi, P.; Liehner, S.; Siewert, B.; Zech, K. A Novel Screening Strategy to Identify ABCB1 Substrates and Inhibitors. Naunyn. Schmiedebergs Arch. Pharmacol. 2009, 379, 11–26. [Google Scholar] [CrossRef]
  175. Chu, X.; Bleasby, K.; Evers, R. Species Differences in Drug Transporters and Implications for Translating Preclinical Findings to Humans. Expert Opin. Drug Metab. Toxicol. 2013, 9, 237–252. [Google Scholar] [CrossRef] [PubMed]
  176. Kennedy, C.J. P-Glycoprotein Induction and Its Energetic Costs in Rainbow Trout (Oncorhynchus mykiss). Fish Physiol. Biochem. 2021, 47, 265–279. [Google Scholar] [CrossRef] [PubMed]
  177. Gourley, M.E.; Kennedy, C.J. Energy Allocations to Xenobiotic Transport and Biotransformation Reactions in Rainbow Trout (Oncorhynchus mykiss) during Energy Intake Restriction. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2009, 150, 270–278. [Google Scholar] [CrossRef] [PubMed]
  178. Nigam, S.K.; Bush, K.T.; Bhatnagar, V.; Poloyac, S.M.; Momper, J.D. The Systems Biology of Drug Metabolizing Enzymes and Transporters: Relevance to Quantitative Systems Pharmacology. Clin. Pharmacol. Ther. 2020, 108, 40–53. [Google Scholar] [CrossRef] [PubMed]
Figure 1. A schematic representation of the relationships between transmembrane transporters and biotransformation enzymes in epithelial cells. The apical membrane faces the lumen of the vessel, which is in contact with blood in blood–tissue barriers (e.g., blood–brain, blood–eye, blood–gonad), and is in contact with excretory fluid (urine, bile, feces) in absorptive (e.g., intestine) and excretory (e.g., liver, kidney) tissues. Substrate entry on the basolateral membrane is represented by membrane diffusion, as well as importer proteins: the SLC21 family (organic anion transporter proteins [OATP]) and the SLC22 family (organic cation transporters [OCT], organic anion transporters [OAT]). Chemical biotransformation occurs inside the cell by phase I functionalization enzymes (represented by cytochrome monooxygenases [CYP]), and phase II conjugation enzymes (represented by glutathione-S-transferase [GST]). Glutathione conjugates are further processed through the mercapturic acid pathway, and exported as mercapturate conjugates. Substrate efflux on the apical membrane is performed in phase 0 (unmodified substrates) by the ABCB family (p-glycoprotein [P-gp]), and in phase III (biotransformed substrates) by the ABCC family (multidrug resistance proteins [MRP]) and the ABCG family (breast cancer resistance protein [BCRP]). The SLC47 family (multidrug and toxicant extrusion proteins [MATE]) exports substrates, but its specificity for the transport of unmodified or biotransformed substrates (and therefore its phase) is unknown in fish.
Figure 1. A schematic representation of the relationships between transmembrane transporters and biotransformation enzymes in epithelial cells. The apical membrane faces the lumen of the vessel, which is in contact with blood in blood–tissue barriers (e.g., blood–brain, blood–eye, blood–gonad), and is in contact with excretory fluid (urine, bile, feces) in absorptive (e.g., intestine) and excretory (e.g., liver, kidney) tissues. Substrate entry on the basolateral membrane is represented by membrane diffusion, as well as importer proteins: the SLC21 family (organic anion transporter proteins [OATP]) and the SLC22 family (organic cation transporters [OCT], organic anion transporters [OAT]). Chemical biotransformation occurs inside the cell by phase I functionalization enzymes (represented by cytochrome monooxygenases [CYP]), and phase II conjugation enzymes (represented by glutathione-S-transferase [GST]). Glutathione conjugates are further processed through the mercapturic acid pathway, and exported as mercapturate conjugates. Substrate efflux on the apical membrane is performed in phase 0 (unmodified substrates) by the ABCB family (p-glycoprotein [P-gp]), and in phase III (biotransformed substrates) by the ABCC family (multidrug resistance proteins [MRP]) and the ABCG family (breast cancer resistance protein [BCRP]). The SLC47 family (multidrug and toxicant extrusion proteins [MATE]) exports substrates, but its specificity for the transport of unmodified or biotransformed substrates (and therefore its phase) is unknown in fish.
Fishes 09 00051 g001
Figure 2. Structure of fish P-glycoprotein. The protein is composed of two transmembrane domains (TMDs) and two nucleotide-binding domains (NBDs). Each transmembrane domain contains six transmembrane helices [41,69].
Figure 2. Structure of fish P-glycoprotein. The protein is composed of two transmembrane domains (TMDs) and two nucleotide-binding domains (NBDs). Each transmembrane domain contains six transmembrane helices [41,69].
Fishes 09 00051 g002
Table 1. P-glycoprotein genes in fish species, determined by synteny analysis. + indicates that the gene is present in the species, − indicates that the gene is absent in the species.
Table 1. P-glycoprotein genes in fish species, determined by synteny analysis. + indicates that the gene is present in the species, − indicates that the gene is absent in the species.
SpeciesAbcb1Abcb4Abcb5References
Danio rerio
(Zebrafish)
++[38]
Gadus morhua
(Atlantic cod)
+[15,38]
Gasterosteus aculeatus
(Three-spined stickleback)
+[15,38]
Ictalurus punctatus
(Channel catfish)
++[15,42]
Latimeria chalumnae
(African coelacanth)
++[38,41]
Oncorhynchus mykiss
(Rainbow trout)
++[41]
Oreochromis niloticus
(Nile tilapia)
+[41]
Oryzias latipes
(Medaka)
+[15,38]
Takifugu rubripes
(Japanese pufferfish)
++[15,38]
Tetraodon nigroviridis
(Green spotted pufferfish)
++[38]
Table 3. Substrates of fish P-glycoprotein (P-gp), as determined by methods including transport inhibition by model chemosensitizers, decreased accumulation in P-gp-over expressing cells, increased cytotoxicity with model chemosensitizers, and increased ATPase activity in combination with the inhibition of model substrate accumulation.
Table 3. Substrates of fish P-glycoprotein (P-gp), as determined by methods including transport inhibition by model chemosensitizers, decreased accumulation in P-gp-over expressing cells, increased cytotoxicity with model chemosensitizers, and increased ATPase activity in combination with the inhibition of model substrate accumulation.
SubstrateSpeciesReferences
Hormones
19-methyl testosteronePoeciliopsis lucida[113]
CortisolOncorhynchus mykiss[114]
Octreotide
(synthetic somatostatin)
Fundulus heteroclitus[115]
Bacterial Toxins
LipopolysaccharidesDanio rerio[116]
Microcystin-LRDanio rerio[52]
Fluorescent Dyes
BCECF-AMDanio rerio[68]
Calcein-AMDanio rerio, Oncorhynchus mykiss, Poeciliopsis lucida[38,68,78,117,118,119]
DiOC6Danio rerio[68]
Rhodamine 123Oncorhynchus mykiss, Poeciliopsis lucida[54,78,113,114,117,119,120]
Rhodamine BHatcheria macraei, Salmo trutta, Oncorhynchus mykiss, Oncorhynchus tshawytscha, Danio rerio, Cyprinus carpio[38,45,67,121]
Metal Compounds
Arsenic trioxide (As2O3)Poeciliopsis lucida[113]
Cadmium chloride (CdCl2)Danio rerio[122]
Silver nitrate (AgNO3)Danio rerio[122]
Polycyclic Aromatic Hydrocarbons
2-aminoanthraceneLeuciscus idus melanotus[30]
Benzo[a]pyreneCyprinus carpio[123]
PhenanthreneDanio rerio[38]
Fragrance Ingredients
α-amylcinnamaldehydeDanio rerio[124]
α-hexylcinnamaldehydeDanio rerio[124]
GalaxolideDanio rerio[38]
IsoeugenolDanio rerio[124]
Musk xyleneDanio rerio[124]
NerolDanio rerio[124]
TonalideDanio rerio[38]
Pesticides
ChlorpyrifosPoeciliopsis lucida[113]
DiazinonPoeciliopsis lucida[113]
Emamectin benzoateOncorhynchus mykiss[84]
Methyl parathionDanio rerio[125]
PhosalonePoeciliopsis lucida[113]
Hydrophobic Peptide
Reversin 205Poeciliopsis lucida[113]
Immunosuppressants
Cyclosporin AFundulus heteroclitus, Squalus acanthias[71,97,126]
RapamycinFundulus heteroclitus[61]
Psychoactive Pharmaceuticals
BupropionOncorhynchus mykiss[127]
CitalopramOncorhynchus mykiss[127]
ClozapineOncorhynchus mykiss[127]
FluoxetineDanio rerio[124]
VenlafaxineOncorhynchus mykiss[127]
Analgesic Pharmaceuticals
ColchicinePoeciliopsis lucida[113]
DiclofenacDanio rerio[124]
Antiarrhythmics
AcebutololPoeciliopsis lucida[117]
DiltiazemPoeciliopsis lucida[113]
NicardipinePoeciliopsis lucida[113]
PrazosinPoeciliopsis lucida[113]
PropanololOncorhynchus mykiss, Poeciliopsis lucida[113,128]
QuinidineOncorhynchus mykiss, Poeciliopsis lucida[113,114]
VerapamilDanio rerio, Poeciliopsis lucida, Fundulus heteroclitus, Squalus acanthias, Oncorhynchus mykiss[38,71,78,113]
Antiretroviral
SaquinavirFundulus heteroclitus[129]
Lipid-Lowering Pharmaceuticals
AtorvastatinPoeciliopsis lucida[117]
PravastatinPoeciliopsis lucida[113]
SimvastatinDanio rerio[124]
Antiparasitic
IvermectinDanio rerio, Oncorhynchus mykiss, Fundulus heteroclitus[62,82,83,84]
Phosphodiesterase Inhibitor
SildenafilPoeciliopsis lucida[113]
Anticancer Pharmaceuticals
AT9283Danio rerio[69]
BisantreneDanio rerio[69]
Daunomycin (Daunorubicin)Fundulus heteroclitus, Pleuronectes americanus[58,130]
DoxorubicinOncorhynchus mykiss, Poeciliopsis lucida, Danio rerio[54,56,69,117]
EtoposideDanio rerio[69]
KW-2478Danio rerio[69]
MitoxantroneDanio rerio[69]
PaclitaxelDanio rerio[69]
RomidepsinDanio rerio[69]
Sepantronium bromide (YM-155)Danio rerio[69]
Tanespimycin (17-AAG)Danio rerio[69]
Tozasertib (VX-680)Danio rerio[69]
VinblastineDanio rerio, Ictalurus punctatus[38,69,73]
VincristineDanio rerio[38]
VinorelbineOncorhynchus mykiss[57]
Table 4. Inducers of P-glycoprotein (Abcb1, Abcb4, Abcb5, P-glycoprotein not otherwise specified) mRNA or protein expression in fish.
Table 4. Inducers of P-glycoprotein (Abcb1, Abcb4, Abcb5, P-glycoprotein not otherwise specified) mRNA or protein expression in fish.
Expression InducerSpeciesTissueReferences
Bile Acids and Salts
5α-cyprinol 27-sulfateDanio rerioLiver[105]
Lithocholic acidOncorhynchus mykissLiver[146]
Enzyme Cofactor
Lipoic acidJenynsia multidentataLiver[147]
Hormone
Pregnenolone 16α-carbonitrile
(Synthetic steroid)
Oncorhynchus mykissLiver[146]
Vitamin
Vitamin B12Danio rerioLiver[94]
Cyanotoxin
Microcystin-LRDanio rerioEmbryo[52]
Microcystin-LRJenynsia multidentataLiver, gill, brain[74]
Metal Compounds
Arsenic trioxide (As2O3)Poeciliopsis lucidaLiver[148]
Cadmium chloride (CdCl2)Trematomus bernacchiiLiver[48]
Cadmium chloride (CdCl2)Danio rerioEmbryo[122]
Silver nitrate (AgNO3)Danio rerioEmbryo[122]
Phytochemicals
Glycyrrhizic acidIctalurus punctatusKidney[142]
QuercetinIctalurus punctatusKidney[142]
Schisandrin AIctalurus punctatusKidney[142]
Schisandrin BIctalurus punctatusKidney[142]
Schisandrol AIctalurus punctatusKidney[142]
schisandrol BIctalurus punctatusKidney[142]
Industrial Chemicals
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)Dicentrarchus labraxLiver[149]
Crude oil (Exxon Valdez)Anoplarchus purpurescensLiver[27]
Heavy fuel oil (fresh)Chelon labrosusLiver[65]
Perfluorooctane sulfonate (PFOS)Chelon labrosusLiver[65]
Pesticides
CarbendazimJenynsia multidentataGill[150]
ChlorpyrifosJenynsia multidentataLiver[151]
Emamectin benzoateOncorhynchus mykissLiver[152]
MalathionDanio rerioLiver, brain[94]
RotenoneIctalurus punctausKidney[142]
TriclosanXiphophorus helleri (males)Liver[153]
Zinc pyrithioneCarassius auratus gibelioLiver, kidney[154]
Analgesic Pharmaceuticals
DiclofenacGambusia affinisLiver[155]
Paracetamol (Acetaminophen)Mugilogobius chulaeLiver[95]
Antibiotic
NorfloxacinXiphophorus HelleriLiver[156]
Antifungal Pharmaceutical
ClotrimazoleOncorhynchus mykissIntestine, brain[17]
Lipid-Lowering Pharmaceutical
SimvastatinMugilogobius abeiLiver[143]
SimvastatinGambusia affinisLiver[92]
Table 6. Inhibitors of fish P-glycoprotein activity, as determined by methods including inhibition of transport of a known P-gp substrate, and inhibition of stimulated ATPase activity.
Table 6. Inhibitors of fish P-glycoprotein activity, as determined by methods including inhibition of transport of a known P-gp substrate, and inhibition of stimulated ATPase activity.
InhibitorSpeciesReferences
Fluorescent Dye
Hoechst 3334Poeciliopsis lucida[113]
Cyanotoxin
Microcystin-LROncorhynchus mykiss[166]
Metal Compounds
Arsenic trioxide (As2O3)Poeciliopsis lucida[148]
Cadmium chloride (CdCl2)Poeciliopsis lucida[148]
Mercury (II) chloride (HgCl2)Poeciliopsis lucida[148]
Orthovanadate (VO4)Poeciliopsis lucida[113]
Potassium dichromate (K2Cr2O7)Poeciliopsis lucida[148]
Hormone
Octreotide
(Synthetic somatostatin)
Fundulus heteroclitus[115]
Bile Acid
TaurochenodeoxycholatePoeciliopsis lucida[113]
Fragrance Ingredients
GalaxolideDanio rerio[38]
α-hexylcinnamaldehydeDanio rerio[124]
IsoeugenolDanio rerio[124]
Musk ketoneDanio rerio[124]
Musk xyleneDanio rerio[124]
NerolDanio rerio[124]
TonalideDanio rerio[38,67]
Polycyclic Aromatic Hydrocarbon
PhenanthreneDanio rerio[38]
Industrial Chemicals
Oil-Sands Process-Affected Water (fresh)Oryzias latipes[167]
Polychlorinated Biphenyls (PCBs)Thunnus albacares[43]
Surfactants
Alcohol polyethoxylatesPoeciliopsis lucida[168]
C-12 linear alkylbenzene sulfonateIctalurus punctatus[169]
Nonylphenol diethoxylateOncorhynchus mykiss[77]
Perfluorooctane sulfonate (PFOS)Danio rerio[170]
Polypropylene glycolsPoeciliopsis lucida[168]
Flame Retardants
Polybrominated diphenyl ethers (BDEs)Thunnus albacares[43]
Pesticides
Azinphos-methylDanio rerio[67]
DiazinonPoeciliopsis lucida[113]
Dichlorodiphenyldichloroethane (DDD)Thunnus albacares[43]
Dichlorodiphenyldichloroethylene (DDE)Thunnus albacares, Poeciliopsis lucida[43,113]
Dichlorodiphenyltrichloroethane (DDT)Thunnus albacares[43]
DieldrinThunnus albacares[43]
2,4-dinitrophenolFundulus heteroclitus[59]
Emamectin benzoateOncorhynchus mykiss[84]
EndosulfanPoeciliopsis lucida[113]
EndrinThunnus albacares[43]
MetazachlorDanio rerio[67]
MirexThunnus albacares[43]
PhosalonePoeciliopsis lucida[113]
ProchlorazOncorhynchus mykiss[77]
TerbuthylazineDanio rerio[67]
Hydrophobic Peptide
Reversin 205Oncorhynchus mykiss, Poeciliopsis lucida[41,113,118,119]
Anticoagulant
DipyridamoleDanio rerio[67]
Antimalarial
QuinineFundulus heteroclitus[59]
Antiparasitic
IvermectinDanio rerio, Oncorhynchus mykiss, Fundulus heteroclitus[62,67,84]
Leukotriene Antagonist
MK571Danio rerio, Poeciliopsis lucida[38,100,142]
Antihistamine
CimetidineOncorhynchus mykiss[128]
Antibiotics
ErythromycinDanio rerio, Poeciliopsis lucida[97,113]
TroleandomycinPoeciliopsis lucida[158]
Analgesic Pharmaceutical
DiclofenacDanio rerio, Poeciliopsis lucida[124,158]
Antifungal Pharmaceutical
ClotrimazoleOncorhynchus mykiss[75]
Anti-Inflammatories
IndomethacinPoeciliopsis lucida[119]
SulfasalazinePoeciliopsis lucida[113]
Antihypertensives
FurosemidePoeciliopsis lucida[117]
ReserpinePoeciliopsis lucida[113]
Antihyperuricemic
ProbenecidPoeciliopsis lucida[119]
Phosphodiesterase Inhibitor
SildenafilPoeciliopsis lucida[113,117]
Antiretrovirals
RitonavirFundulus heteroclitus[129]
SaquinavirFundulus heteroclitus[129]
Lipid-Lowering Pharmaceuticals
AtorvastatinPoeciliopsis lucida[113,117]
GemfibrozilPoeciliopsis lucida[117]
PravastatinPoeciliopsis lucida[117]
SimvastatinPoeciliopsis lucida[117]
Psychoactive Pharmaceuticals
CarbamazepineDanio rerio[67]
FluoxetineDanio rerio[124]
SertralineDanio rerio[124]
TrifluoperazinePoeciliopsis lucida[113]
Immunosuppressants
Cyclosporin ADanio rerio, Oncorhynchus mykiss, Thunnus albacares, Poeciliopsis lucida, Cyprinus carpio, Fundulus heteroclitus, Pleuronectes americanus[38,41,43,58,61,67,68,77,113,114,115,117,118,119,121,128,130,148,168,171]
Cyclosporin GFundulus heteroclitus[59]
RapamycinFundulus heteroclitus[61]
Tacrolimus (FK506)Fundulus heteroclitus[61]
Antiarrhythmics
DiltiazemPoeciliopsis lucida[113]
NicardipinePoeciliopsis lucida[113]
PrazosinPoeciliopsis lucida[113]
PropranololPoeciliopsis lucida[113]
QuinidineOncorhynchus mykiss, Poeciliopsis lucida[113,114,128]
VerapamilDanio rerio, Oncorhynchus mykiss, Thunnus albacares, Hatcheria macraei, Salmo trutta, Oncorhynchus tshawytscha, Poeciliopsis lucida, Scophthalmus maximus, Cyprinus carpio, Ictalurus punctatus, Fundulus heteroclitus, Pleuronectes americanus[38,43,45,54,59,61,62,67,69,73,77,78,97,113,114,115,119,121,124,130,172]
Anti-Cancer Pharmaceuticals
DoxorubicinOncorhynchus mykiss[54]
ElacridarDanio rerio[69]
FluorouracilPoeciliopsis lucida[113]
MitoxantronePoeciliopsis lucida[113]
TamoxifenPoeciliopsis lucida[113]
Tariquidar (XR9576)Danio rerio, Oncorhynchus mykiss[56,69]
Valspodar (PSC833)Oncorhynchus mykiss, Thunnus albacares, Danio rerio, Poeciliopsis lucida, Fundulus heteroclitus, Squalus acanthias[38,43,61,62,68,69,71,113,114,115,118,171]
VinblastineDanio rerio, Oncorhynchus mykiss, Fundulus heteroclitus, Pleuronectes americanus[38,54,59,68,77,130]
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

Johnston, C.U.; Kennedy, C.J. A Review of P-Glycoprotein Function and Regulation in Fish. Fishes 2024, 9, 51. https://doi.org/10.3390/fishes9020051

AMA Style

Johnston CU, Kennedy CJ. A Review of P-Glycoprotein Function and Regulation in Fish. Fishes. 2024; 9(2):51. https://doi.org/10.3390/fishes9020051

Chicago/Turabian Style

Johnston, Christina U., and Christopher J. Kennedy. 2024. "A Review of P-Glycoprotein Function and Regulation in Fish" Fishes 9, no. 2: 51. https://doi.org/10.3390/fishes9020051

Article Metrics

Back to TopTop