Citation: Rachael C. Adams, Behnam Rashidieh. Can computers conceive the complexity of cancer to cure it? Using artificial intelligence technology in cancer modelling and drug discovery[J]. Mathematical Biosciences and Engineering, 2020, 17(6): 6515-6530. doi: 10.3934/mbe.2020340
[1] | Lingling Li, Mengyao Shao, Xingshi He, Shanjing Ren, Tianhai Tian . Risk of lung cancer due to external environmental factor and epidemiological data analysis. Mathematical Biosciences and Engineering, 2021, 18(5): 6079-6094. doi: 10.3934/mbe.2021304 |
[2] | Ancheng Deng, Xiaoqiang Sun . Dynamic gene regulatory network reconstruction and analysis based on clinical transcriptomic data of colorectal cancer. Mathematical Biosciences and Engineering, 2020, 17(4): 3224-3239. doi: 10.3934/mbe.2020183 |
[3] | Erin N. Bodine, K. Lars Monia . A proton therapy model using discrete difference equations with an example of treating hepatocellular carcinoma. Mathematical Biosciences and Engineering, 2017, 14(4): 881-899. doi: 10.3934/mbe.2017047 |
[4] | Donggu Lee, Sunju Oh, Sean Lawler, Yangjin Kim . Bistable dynamics of TAN-NK cells in tumor growth and control of radiotherapy-induced neutropenia in lung cancer treatment. Mathematical Biosciences and Engineering, 2025, 22(4): 744-809. doi: 10.3934/mbe.2025028 |
[5] | Cameron Meaney, Gibin G Powathil, Ala Yaromina, Ludwig J Dubois, Philippe Lambin, Mohammad Kohandel . Role of hypoxia-activated prodrugs in combination with radiation therapy: An in silico approach. Mathematical Biosciences and Engineering, 2019, 16(6): 6257-6273. doi: 10.3934/mbe.2019312 |
[6] | Maher Alwuthaynani, Raluca Eftimie, Dumitru Trucu . Inverse problem approaches for mutation laws in heterogeneous tumours with local and nonlocal dynamics. Mathematical Biosciences and Engineering, 2022, 19(4): 3720-3747. doi: 10.3934/mbe.2022171 |
[7] | Na Li, Peng Luo, Chunyang Li, Yanyan Hong, Mingjun Zhang, Zhendong Chen . Analysis of related factors of radiation pneumonia caused by precise radiotherapy of esophageal cancer based on random forest algorithm. Mathematical Biosciences and Engineering, 2021, 18(4): 4477-4490. doi: 10.3934/mbe.2021227 |
[8] | Katrine O. Bangsgaard, Morten Andersen, Vibe Skov, Lasse Kjær, Hans C. Hasselbalch, Johnny T. Ottesen . Dynamics of competing heterogeneous clones in blood cancers explains multiple observations - a mathematical modeling approach. Mathematical Biosciences and Engineering, 2020, 17(6): 7645-7670. doi: 10.3934/mbe.2020389 |
[9] | Subhadip Paul, Prasun Kumar Roy . The consequence of day-to-day stochastic dose deviation from the planned dose in fractionated radiation therapy. Mathematical Biosciences and Engineering, 2016, 13(1): 159-170. doi: 10.3934/mbe.2016.13.159 |
[10] | Eugene Kashdan, Svetlana Bunimovich-Mendrazitsky . Hybrid discrete-continuous model of invasive bladder cancer. Mathematical Biosciences and Engineering, 2013, 10(3): 729-742. doi: 10.3934/mbe.2013.10.729 |
Mercury (Hg) is considered a devastating environmental pollutant, mainly after the environmental disaster at Minamata (Japan) and several other poisoning accidents due to the use of Hg pesticides in agriculture [1]. Hg exists as elemental form, inorganic (iHg) and organic Hg (primarily methylmercury, MeHg). In the environment, Hg is released from natural and anthropogenic sources [2]. iHg enters the air from mining ore deposits, burning coal and waste, or from manufacturing plants. It enters the water or soil from natural deposits, disposal of wastes, and the use of Hg containing fungicides. iHg is maintained in the upper sedimentary layers of water-beds and is methylated and thus transformed to the highly toxic species MeHg by sulphate-reducing bacteria.
Numerous toxicological studies have gained increasing interest in order to understand the impact of Hg on aquatic communities, which are particularly vulnerable. In this regard, fish have been used to assess pollution because they represent the most diverse group of vertebrates [3] and have genetic relatedness to the higher vertebrates including mammals. It is widely known that fish are a great source of Hg in our food and their accumulation could represent a serious risk for human beings [4]. Although fish have always been perceived as a healthy and nutritive food [5], the Environmental Protection Agency (EPA) has raised public concern as it claimed that the levels of Hg in certain fish species make them unsuitable or be restricted for children and pregnant women consumption [6]. Some data about the Hg concentration in common fish species are shown in Table 1. This data are worrying us if we consider that the projections show an increase in the demand for seafood products to year 2030.
Specie | Mercury content (ppm) | Safety |
Anchovies | 0.017 ± 0.015 | Eco-good |
Atlantic cod | 0.095 ± 0.080 | Eco-bad |
Bass (saltwater, black, striped, rockfish) | 0.167 ± 0.194 | Eco-bad |
Bass Chilean | 0.354 ± 0.199 | Eco-bad |
Carp | 0.140 ± 0.099 | Eco-bad |
Catfish | 0.049 ± 0.084 | Eco-good |
Croaker Atlantic (Atlantic) | 0.069 ± 0.049 | Eco-good |
Croaker White (Pacific) | 0.287 ± 0.069 | Eco-bad |
Grouper (all species) | 0.448 ± 0.287 | Eco-bad |
Mullet | 0.05 ± 0.078 | Eco-bad |
Salmon, wild (Alaska) | 0.014 ± 0.041 | Eco-good |
Sardines, Pacific (US) | 0.016 ± 0.007 | Eco-good |
Shark | 0.979 ± 0.626 | Eco-bad |
Swordfish | 0.976 ± 0.510 | Eco-bad |
Tilapia | 0.013 ± 0.023 | Eco-good |
Trout, rainbow (farmed, freshwater) | 0.072 ± 0.143 | Eco-good |
Tuna species | 0.415 ± 0.308 | Eco-bad |
In this regard, aquaculture is one of the most important food manufacturing industries in the coming decades trying to compensate the human consumption demand. Moreover, farmers have to know and control the impact of the environmental contaminants in the species produced for humans [7]. In this specific field, very little is known about the effects of Hg exposure in fish, and the comparison of any results is very difficult and sometimes contradictory because different investigators have used a variety of administration methods as well as concentrations of Hg. Moreover, the development of prominent trends in toxicity testing based on in vitro tests and especially in vitro mechanistic assays has gain considerable attention in recent years.
In the aquatic environment, Hg speciation, uptake, bioavailability, and toxicity are dependent upon environmental parameters including hydrophobicity, pH, salinity, hardness (Ca2+ and Mg2+ concentration) and interaction of metals with biotic and abiotic ligands [8]. In teleost fish species, bioavailability of Hg depend not only on total chemical concentration in the environment but also on how readily the fish can absorb these different Hg forms at the gill, across the skin, and within the digestive tract and on how chemical speciation affects distribution throughout the organism [9]. Thus, the metal absorption and distribution occurs as follows: first, Hg crosses the epithelium; second, it incorporates into blood including binding to plasma proteins, transport via the systemic circulation or freely dissolved to various tissues; and finally, it is transported from blood into tissues. Gills, digestive system, and, to a lesser extent, the skin, are the major sites of metal uptake in fish [8].
Hg uptake can be passive or energy-dependent, depending on the Hg species [10]. Concretely, in plasma, MeHg binds reversibly to cysteine amino acid and therefore to sulphur-containing molecules such as glutathione (GSH). The cysteine-bound form is of particular interest because it is transported by an L-neutral amino acid transporter system into the cells of sensitive tissues such as brain [11]. In the gastrointestinal tract, ingested MeHg is efficiently absorbed and its distribution to the blood is complete within approximately 30 h, and the blood level accounts for about 7% of the ingested dose. The brain is the primary target site for MeHg and approximately 10% is retained in the brain with the remainder transported to the liver and kidney where it is excreted through bile and urine [12]. In rainbow trout (Oncorhynchus mykiss), however, 90% of whole-blood MeHg is bound to the beta-chain of hemoglobin in red blood cells [13]. In addition, MeHg readily binds to metallothioneins (MTs) and metalloproteins with cysteine residues displacing Zn2+ [10,14]. The primary mechanism of MeHg as well as its specificity has yet to be identified. MeHg-cysteine conjugates have shown increased cellular efflux, presumably due to the generation and involvement of glutathione.
Regarding its bioaccumulation, concentrations of MeHg are magnified within the food chain, reaching concentrations in fish 10, 000-to 100, 000-fold greater than in the surrounding water [2]. The primary target tissues for Hg are the central nervous system (CNS) [15] and the kidney, triggering loss of appetite, brain lesions, cataracts, abnormal motor coordination, and behavioural changes, alterations that lead to the fish to have impaired growth, reproduction, and development.
Several studies have shown that Hg produce an imbalance between the reactive oxygen species (ROS) production and its clearance by the antioxidant system in the known oxidative stress response. Thus, in fish, it has been described the production of ROS after Hg exposure in vivo [16,17,18,19,20,21] or in vitro [22,23,24,25,26]. Indeed, Hg reacts with the thiol groups of GSH, which can induce GSH depletion and oxidative stress [19,20,21,27,28]. Thus, some studies have found alterations in the antioxidant system caused by Hg exposure including glutathione reductase (GR) and glutathione peroxidase (GPx) activities in zebra seabream (Diplodus cervinus) [29], modifications in superoxide dismutase (SOD), catalase (CAT), glutathione S-transferase (GST), and GPx activities in trahira species (Hoplias malabaricus) [30]. Very recently, MeHg exposure increased the ROS levels and decreased the antioxidant potential of gilthead seabream (Sparus aurata) serum while increased the SOD, CAT and GR activities in the liver [18]. In addition, significant alterations in the expression of the antioxidant enzyme genes sod, cat, gst, gpx, and gr have been observed in the freshwater fish Yamú (Brycon amazonicus) after Hg exposure leading to oxidation of lipids and proteins [31]. In a set of studies conducted on wild golden grey mullet (Liza aurata) it has been demonstrated the depletion on reduced GSH, GPx, SOD and lipid peroxidation (LPO) and increment on total GSH, GST and CAT activities towards other metabolites indicating the potential of using metabolomics to determine and probe the Hg exposure and oxidative stress relations [19,20,21]. In vitro studies including fish cell lines [SAF-1, derived from gilthead seabream, and DLB-1, derived from European sea bass (Dicentrarchus labrax)] or primary fish cell cultures (leucocytes or erythrocytes) showed an increase in ROS levels after Hg exposure; however, an up-or down-regulation in the expression of some antioxidant genes (sod, cat, gr or prx) was observed [24,25,26,27,32,33,34].
MTs also play a protective role in response to Hg exposure. The mRNA expression of two mt genes was noted in the liver of common carp (Cyprinus carpio) from Hg contaminated river [35]. However, no significant correlations between total Hg content and MT levels were described in different fish tissues from a Hg contaminated area [36]. Thus, in in vitro studies, mt gene expression was enhanced after Hg exposure in SAF-1 [26] and DLB-1 [24] cell lines as well as in gilthead seabream or European sea bass peripheral blood leucocytes (PBLs) [33] and erythrocytes [34]; however, a strong down-regulation was observed in leucocytes derived from the head-kidney, the main hematopoietic tissue, from the same species [25,32]. Indeed, Hg bind easily to MTs [37,38] but an excess of metal could provoke a MT dysfunction leading to an increase of ROS levels as we have observed in some fish or fish cell lines before.
Furthermore, the ROS imbalance leads to cell death by apoptosis. Hg forms induce apoptosis by inhibiting mitochondrial function [39] and releasing cytochrome C from the mitochondria to the cytosol [40]. Moreover, p38 mitogen-activated protein kinase is activated by mercury resulting in apoptosis [41]. In addition, mitochondrial membrane permeability is regulated through a family of anti-apoptotic (Bcl-2, Bcl-xL, etc.) and pro-apoptotic (Bad, Bax, Bak, Bid, etc.) proto-oncogenes [42]. This cell death mechanism has been demonstrated in fish exposed to Hg [43,44,45] as well as in in vitro fish systems including cell lines [24,26] and primary cultures of head-kidney leucocytes [25,32], PBLs [33] and erythrocytes [34].
Although the immunotoxicological effects of mercurial compounds have been well studied in mammals [46,47] far less is known concerning the effects in fish to date [7,48]. Moreover, due to the complexity and multifaceted of the immune system, recent articles show the difficulty to assess the immunotoxicological effects in fish and the challenge to select appropriate exposure and effect parameters out of the many immune parameters which could be measured [49]. Because of the immunotoxicological studies of Hg in fish have focused on almost exclusively on immunosuppressive effects, some aspects have been ignored. For instance, a range of scientific studies have attempted to investigate the role of Hg in mammalian autoimmunity [50] or hypersensibility [51,52].
MeHg exposure by dietary intake [53], injection [54] or waterborne [18] can trigger alterations of the fish immune system. Upon immune mediators, little is known about their regulation of cytokines in fish. The freshwater fish snakehead (Channa punctatus) exposed to 0.3 mg/L HgCl2showed an up-regulation of pro-inflammatory cytokines such as tumour necrosis factor-α (tnfα) and interleukin-6 (il6) after 7 days of exposure [1]. In vitro models have shown a down-regulation of pro-inflammatory il1b gene expression after 2 or 24 h of MeHg exposure in gilthead seabream head-kidney leucocytes, while alterations were not observed in European sea bass leucocytes [25,32]. On the other hand, effects on soluble humoral factors of fish have been widely studied after Hg exposure. For example, lysozyme activity was enhanced in goldfish (Carassius auratus) [55] or rainbow trout [56] after exposure to different HgCl2 concentrations but decreased in plaice (Pleuronectes platessa) [57]. Other humoral activities such as serum complement or peroxidase activities or total IgM levels were increased or impaired after MeHg exposure in diverse fish species [18,54,56]. Interestingly, waterborne MeHg significantly increased the immune responses in the gilthead seabream skin mucus including the microbicidal activity [18]. Upon the cellular innate immune response, MeHg increased the phagocytosis in gilthead seabream head-kidney leucocytes exposed in vivo by waterborne [18] and in vitro [25] whilst in European sea bass leucocytes the phagocyte functions (phagocytosis and respiratory burst) by in vitro exposure to Hg was reduced [22,32]. In addition, in the European sea bass leucocytes, in vitro treatment with HgCl2 induced apoptosis in head-kidney macrophages as well as reduced the ROS production and the benefits of macrophage-activating factors (MAF) [22,58]. Thus, the presence of factors such as the serum levels of corticosteroids and catecholamines that do not operate in vitro could explain the differences observed between in vitro and in vivo studies [59]. However, the mechanisms by which metals alter the phagocyte functions are still poorly understood. Strikingly, few studies have dealt with the leucocyte death by Hg. Thus, an increase in the transcription of genes related to apoptosis was shown after MeHg exposure to gilthead seabream or European sea bass blood leucocytes (PBLs) [33]. However, Hg exposure promoted both apoptosis and necrosis cell death in gilthead seabream or European sea bass head-kidney leucocytes [25,32].
Although exposure to some metals often disturbs normal metabolic processes in fish, including irritation of respiratory epithelium, changes in ventilation frequency, or inefficient oxygen delivery to tissues [60] considerably less is known regarding the effect of Hg on the respiratory system. Previous studies have shown that an exposure to dissolved Hg disrupts gill epithelium, potentially affecting gas exchange and permeability of cell membranes to cations [61,62]. Such disruptions may result in compensatory changes in ventilation frequency, increased energy demands, or altered gas exchange efficiency, possibly resulting in the increase in metabolic rate [63]. However, gill damage and a subsequent increase of metabolic rate did not occur in the mosquitofish (Gambusia holbrooki) fed with HgCl2 probably due to the Hg accumulation via intestinal absorption from dietary sources [64]. More recently, HgCl2 via food or diet decreases the plasticity of the cardiorespiratory responses reducing the survival chances of Yamú and trahira under hypoxic conditions frequently observed in theirs wild [65].
Upon histopathology studies, the gills is the organ to better study due to their function in the respiration process and since this organ is continuously and directly exposed to the external environment [62,66,67,68,69,70]. Moreover, a report shows an increase of the chloride cells (CCs) in the European sea bass gills after exposure to Hg [71], which is in concordance with another study using mosquitofish [62]. The CAT activity remained unchanged while GPx and GR activities showed a significant decrease in trahira gills after exposure to HgCl2 [65]. In wild grey mullet from Hg-contaminated areas, gill GPx and SOD activities were depleted while GST and CAT activities were increased indicating a massive GSH oxidation [20,21]. In in vitro conditions, gill cell suspensions exposed to HgCl2 showed high rate of DNA breaks (single and double stranded) measured as the comet assay in common carp and rainbow trout [72,73].
Similarly, Hg accumulation in the heart is thought to contribute to cardiomyopathy. The mechanism by which Hg produces toxic effects on the cardiovascular system is not fully elucidated, but this mechanism is believed to involve an increase in oxidative stress [74]. In a recent study, trahira specimens exposed to HgCl2 showed an anticipated bradycardia and lower heart rate probably due to damage in cardiac myocytes [65], which is in agreement with other studies in trahira [75,76] or in other tropical species [77].
Numerous studies suggest that the inhibitory effects of Hg on fish reproduction occur at multiple sites within the hypothalamic–pituitary–gonadal (HPG) axis [78]. Moreover, in most cases, studies have been carrying out in in vivo conditions due to the missing information of hormone release in in vitro assays. Thus, recent studies have reported a significantly lesser transcripts of gonadotropin-releasing hormone (GnRH) (gnrh2 and gnrh3) in the brain of both male and female zebrafish (Danio rerio) after Hg exposure, suggesting that Hg could modulate hypothalamic production of GnRHs in fish and consequently disrupted production of gonadotropin hormones such as follicle stimulating hormone (FSH) and luteinizing hormone (LH) [81,82]. Furthermore, an alteration in expression of genes commonly associated with endocrine disruption and a decrease in the production of vitellogenin as a result of dietary MeHg exposure in fathead minnows (Pimephales promelas) was observed [83]. Similarly, by using cDNA microarray analysis in the cutthroat trout (Salmo clarkii), expression levels of gnrh1 and gnrh2 were down-regulated in the liver from those specimens containing high Hg levels compared to those with low Hg concentrations [84]. Moreover, down regulation of genes involved in early stages of the spermatogenesis (fshβ, fshr, lhr or lhβ) or genes involved in the synthesis of steroid hormones (cyp17 and cyp11b) were observed in male zebrafish testis after exposure to 30 µg Hg/L [81] which results are concomitant with other studies in catfish (Clarias batrachus) [85]. Likewise, as in male, an impaired RNA transcription of genes involved in promoting follicular growth (lhβ and lhr) was observed in the ovary of zebrafish [81].
On the other hand, severe histological lesions in the gonad have been observed in fish after Hg exposure in vivo. In testis, exposure to HgCl2 caused the thickening of the tubule walls and disorderly arranged spermatozoa in zebrafish [81] as well as in medaka (Oryzias latipes) after MeHg exposure [86], probably resulting in inhibition of spermatogenesis. Moreover, a disorganization of seminiferous lobules, proliferation of interstitial tissue, congestion of blood vessels, reduction of germ cells and sperm aggregation was induced after HgCl2 injection in the tropical fish Gymnotus carapo [87]. In ovary, degenerative changes such as atresia (follicular degeneration) were found after HgCl2 exposure in zebrafish [81] in agreement with other studies [88,89,90].
In recent years, research of reproductive effects such as spawning success, spawning behaviour, fertilization success and fecundity after Hg has highlighted their importance in fish [91]. A significant reduction of the spawning success and an increase time to spawn was observed in female fathead minnows after MeHg feeding [82]. Most recent studies have shown the maternal transfer as a significant route of exposure of MeHg diet for larval and juvenile fish. The cellular mechanisms by which MeHg moves through the body is readily complexed with cysteine S-conjugates. This structure mimics that of methionine and can therefore be transferred across cell membranes to developing oocytes via methionine transporters [82]. It was originally thought that MeHg partitioned from stores in female tissues into developing oocytes [93]. However, recent research has shown that the diet of the maternal adult during oogenesis, rather than adult body burden is the principal source of Hg in eggs [94]. A study [93] employed the use of stable MeHg isotopes to investigate the sources of Hg transferred to eggs. Adult sheepshead minnows (Cyprinodon variegatus) were exposed to MeHg-spiked diets. The diets administered during the pre-oogenesis stage contained different MeHg isotopes than the diet administered during oogenesis, allowing us to characterize the proportion of Hg in eggs derived from maternal body burden versus maternal diet during oogenesis. The results indicate that a constant percentage of maternal body burdens were transferred to eggs across all treatments; however, the majority of total Hg found in eggs was from recent maternal dietary exposure. In another study, males of fathead minnow fed with MeHg had significantly higher gonad concentrations of MeHg than females, which may be attributed to losses of contaminants due to maternal transfer of dietary MeHg to eggs [95] and is in concordance with other studies [93,96]. Moreover, resulting embryos from the MeHg low-diet treatment displayed altered embryonic movement patterns (hyperactivity) and decreased time to hatch while embryos from the MeHg high-diet treatment had delayed hatching and increased mortality compared with the other treatments [95].
Notwithstanding that Hg neurotoxicity has been well reported in both human and mammalian models [15,97,98,99], information regarding its threat to fish nervous system and underlying mechanisms is still scarce. Both HgCl2 and MeHg are able to easily cross the blood-brain-barrier (BBB), reaching the fish brain where it exerts toxicity [100,101,102]. Moreover, both forms of Hg share the same toxic chemical entity [103] and, thus, neurotoxicity may depend mainly on the external bioavailability. Only a few neurotoxicological endpoints have been employed to evaluate the biological effects of Hg in fish, both in laboratory experiments and under field exposures. For instance, high concentrations of MeHg were accumulated in the medaka brain after exposure to graded sublethal concentrations [86] or in European sea bass brain [104]. Changes in oxidative stress profiles of the Atlantic salmon (Salmo salar) [105] and European sea bass brain [101] or alterations in the protein expression in the marine medaka brain [106] have been reported after HgCl2exposure. Adult zebrafish exposed to dietary HgCl2showed induced mt production at gene level [107]; however, no changes in the expression of genes involved in antioxidant defences, metal chelation, active efflux of organic compounds, mitochondrial metabolism, DNA repair, and apoptosis were observed [108]. Moreover, histopathological examinations of fish brain was performed, revealing a widespread neuronal degradation [105] or for the first time a deficit in the number of the cells of the white seabream (Diplodus sargus) brain as an effect of Hg deposition [102]. Curiously, as in humans, the long-term effects of Hg were also disclosed by numerous alterations on motor and mood/anxiety-like behaviour after 28 days of recovery [102]. Concerning in vitro studies, only one report has dealt with Hg toxicity on the cell line DLB-1, derived from European sea bass brain. In particular, MeHg reduced the viability of the DLB-1 cells, failed to increase ROS, reduced the cat gene expression and increased the mta transcription. Furthermore, DLB-1 cells exposed to Hg elicited a rapid cell death by apoptosis [24].
Although an effective barrier to iHg, the intestinal wall is permeable to Hg, due to the high lipid solubility of the compound [109]. Thus, the gastrointestinal tract represents a major route of entry for a wide variety of toxicants present in the diet or in the water that the fish inhabit [110,111,112]; nonetheless there is little information relating to the protective mechanisms adopted by the intestine epithelial surfaces of the fish against Hg uptake [113]. Light microscopy based investigations have demonstrated that there are alterations in the gut of snakehead and Stinging catfish (Heteropneustes fossilis) following Hg intoxication [110,112] but surprisingly no histopathological changes were described in the arctic charr (Salvelinus alpinus) following exposure to dietary HgCl2 and MeHg [113]. In contrast, perturbations including the notable presence of vacuoles within the cytoplasm along with various inclusions, myeloid bodies and modifications to the endoplasmic reticulum and mitochondria on the intestinal epithelium of European sea bass exposed to Hg have been shown [71]. Moreover, the presence of MeHg in the intestine epithelial cells of trahira and, at a minor extent, in the extracellular matrix represents the main tissue targets [30]. Although epithelial cells from intestinal mucosa represent a biological barrier that selects the entrance of essential nutrients as well as contaminants, it should be remarked the fact that MeHg absorption can occur by passive diffusion through neutral amino acids carrier proteins [114] and accumulate in the epithelial cells or toward the connective tissue and transported via bloodstream to other target organs.
Unlike mammals, Hg can also be depurated by the kidney, liver, and, possibly, the gills of fish [116]. Small amounts of MeHg were detected in the urine of treated juvenile white sturgeon (Acipenser transmontanus) and high concentrations of MeHg were also found in the kidneys and gills [116]. Thus, the high concentration of Hg in the kidneys and gills may reflect a transient state before MeHg is eliminated from these organs. Moreover, liver registered the highest elimination percentages (up to 64% in the liver, 20% in the brain, and 3% in the muscle) in European sea bass exposed to MeHg during 28 days [104]. Surprisingly, it was verified that the concentration of MeHg in gilthead seabream liver was higher, approximately two-fold [18], than the amount detected in the muscle as it happened in other fish species such as goldfish [118], zebrafish [108] or European sea bass [117]. Furthermore, Hg can cause liver damage as demonstrated by some studies in gilthead seabream [18], the arctic charr [113] or in trahira [27].
The kidney of teleost receives a large portion of the cardiac output because of their extensive portal system. The role of kidney on Hg elimination depends on the mercurial form, preferably iHg form by urine [119]. Previous studies showed that after a 4-week exposure to dietary MeHg, the kidney of sturgeon species showed prominent degeneration of the renal tubules [120]. Similar changes were reported in guppy (Poecilia reticulata) [121] and Indian catfish (Clarias batrachus) [122] exposed to waterborne MeHg. The accumulation of MeHg in the renal tubes of trahira kidney has also demonstrated [30].
Due to the affinity to conjugate reversibly to cysteine amino acid, Hg is transported by the blood to the rest of the organs [123]. However, there is little available information in the literature related to hematological responses in fish chronically exposed to Hg. Thereby, some reports have focused on the effect of Hg on fish blood cells, namely erythrocytes. Because of it is thought to compete with iron for binding to haemoglobin, which can result in impaired hemoglobin formation, Hg exposure resulted in anemia in two fish species (Channa gachua and Pleuronectes platessa) [124,125]. However, dietary MeHg increased significantly the values of haematocrit after exposure in trahira [54] suggesting an increase in the blood oxygen capacity, in contrast to the not significantly changes observed in Nile tilapia (Oreochromis niloticus) exposed to sublethal concentrations of Hg [126]. [127] observed a decrease in membrane fluidity, change of internal viscosity, and internal protein conformation and hemolysis in erythrocytes of common carp subjected to Hg. Moreover, a differential sensitivity of fish species towards the induction of erythrocyte micronuclei (MN) and other nuclear abnormalities has been reported after intraperitoneal injection of Hg [128,129] or in olden grey mullet (Liza aurata) along an environmental Hg contamination gradient [130]. On the other hand, in vitro toxicological tests using human erythrocytes are gaining traction as alternatives to in vitro tests, however few studies are available in fish. For example, gilthead seabream or European sea bass erythrocytes exposed to Hg exhibited cytotoxicity and alteration in the gene expression profile of genes involved in oxidative stress, cellular protection and apoptosis death [34].
Although Hg is ubiquitous in the environment, it is considered one of the most toxic elements or substances on the planet that continues to be dumped into our waterways and soil, spilled into our atmosphere, and consumed in our food and water. Research indicates that Hg exposure can induce a variety of adverse effects in fish at physiologic, histologic, bio-chemical, enzymatic, and genetic levels. Certain fish species, however, appear to show more sensitivity to Hg toxicity than others. Hence, Hg-induced toxicological pathology in fish is influenced by such factors as species, age, environmental conditions, exposure time, and exposure concentration. The exact causes of fish death are multiple and depend mainly on time-concentration combinations. In-depth toxicodynamics and toxicokinetics studies are necessary to establish an exact cause-effect relation. The scientific data discussed in this review provide a basis for understanding the potential impact, as well as for advancing our knowledge of the ecotoxicology and risk assessment of Hg.
This work was partly supported by Fundación Séneca (Grupo de Excelencia de la Región de Murcia 19883/GERM/15).
All authors declare no conflict of interest in this paper.
[1] | R. Mahumad, K. Alam, J. Dunn, J. Gow, Emerging cancer incidence, mortality, hospitalisation and associated burden among Australian cancer patients, 1982-2014: An incidence-based approach in terms of trends, determinants and inequality, BMJ Open, 5 (2019). |
[2] | M. Breitenbach, J. Hoffmann, Editorial: Cancer models, Front. Oncol., 8 (2018), 401-401. |
[3] | L. Ogilvie, A. Kovachev, C. Wierling, B. Lange, H. Lehrach, Models of models: A translational Route for cancer treatment and drug development, Front. Oncol., 7 (2017). |
[4] | K. Mak, M. Pichika, Artificial intelligence in drug development: Present status and future prospects, Drug Discovery Today, 24 (2019), 773-780. |
[5] | J. Vamathevan, D. Clark, P. Czodrowski, I. Dunham, E. Ferran, G. Lee, et al., Applications of machine learning in drug discovery and development, Nat. Rev. Drug Discovery, 18 (2018), 463-477. |
[6] | O. Wolkenhauer, Why model? Front. Phys., 5 (2014). |
[7] | A. Levine, C. Schlosser, J. Grewal., R. Coope, S. Jones, S. Yip, Rise of the machines: Advances in deep learning for cancer diagnosis, Trends Cancer, 5 (2019), 157-169. |
[8] | K. Vougas, T. Sakellaropolous, A. Kotsina, G. R. P. Foukas, A. Ntargaras, F. Koinis, et al., Machine learning and data mining frameworks for predicting drug response in cancer: An overview and a novel in silico screening process based on association rule mining. Pharmacol. Ther., 203 (2019), 107395. |
[9] | G. V. Sherbet, W. L. Woo, S. Dlay, Application of artificial intelligence-based technology in cancer management: A commentary on the deployment of artificial neural networks, Anticancer Res., 38 (2018), 6607-6613. |
[10] | R. M. Thomas, T. Van Dyke, G. Merlino, C. P. Day, Concepts in cancer modeling: A brief history, Cancer Res., 76 (2016), 5921-5925. |
[11] | P. Kumari, A. Nath, R. Chaube, Identification of human drug targets using machine-learning algorithms, Comput. Biol. Med., 56 (2014), 175-181. |
[12] | J. Metzcar, Y. Wang, R. Heiland, P. Macklin, A Review of cell-based computational modeling in cancer biology, JCO Clin. Cancer Inform., 3 (2019), 1-13. |
[13] | D. Hanahan, R. A. Weinberg, The hallmarks of cancer, Cell, 100 (2000), 57-70. |
[14] | A. Ghaffarizadeh, S. H. Friedman, P. Macklin, BioFVM: An efficient, parallelized diffusive transport solver for 3-D biological simulations, Bioinformatics, 32 (2016), 1256-1258. |
[15] | A. Ghaffarizadeh, R. Heiland, S. H. Friedman, S. M. Mumethaler, P. Macklin, PhysiCell: An open source physics-based cell simulator for 3-D multicellular systems, PLOS Comput. Biol., 14 (2018), e1005991. |
[16] | E. Bonabeau, Agent-based modeling: Methods and techniques for simulating human systems, Proc. Natl. Acad. Sci. USA, 99 (2002), 7280-7287. |
[17] | P. Van Liedekerke, M. M. Palm, N. Jagiella, D. Drasdo, Simulating tissue mechanics with agent-based models: Concepts, perspectives and some novel results, Comput. Particle Mech., 2 (2015), 401-444. |
[18] | R. C. Kennedy, G. E. Ropella, C. A. Hunt, A cell-centered, agent-based framework that enables flexible environment granularities, Theor. Biol. Med. Model. 13 (2016). |
[19] | J. Poleszczuk, P. Macklin, H. Enderling, Agent-based modeling of cancer stem cell driven solid tumor growth, Methods Mol. Biol., 1516 (2016), 335-346. |
[20] | Y. Cai, S. Xu, J. Wu, Q. Long, Coupled modelling of tumour angiogenesis, tumour growth and blood perfusion, J. Theor. Biol., 279 (2011), 90-101. |
[21] | A. Shirinifard, J. S. Gens, B. L. Zaitlen, N. J. Poplawski, M. Swat, J. A. Glazier, 3D multi-cell simulation of tumor growth and angiogenesis, PLOS One, 4 (2009), e7190. |
[22] | B. Chopard, R. Ouared, A. Deustch, H. Hatzikirou, D. Wolf-Gladrow, Lattice-gas cellular automaton models for biology: From fluids to cells, Acta Biotheor., 58 (2010), 329-340. |
[23] | H. Hatzikirou, D. Basanta, M. Simon, K. Schaller, A. Deustch, 'Go or Grow': The key to the emergence of invasion in tumour progression?, Math. Med. Biol.: A J. IMA, 29 (2010), 49-65. |
[24] | H. N. Weerasinghe, P. M. Burragem, K. Burrage, D. V. Nicolau, Mathematical models of cancer cell plasticity, J. Oncol., 2019. |
[25] | M. S. Alber, M. A. Kiskowski, J. A. Glazier, Y. Jiang, On cellular automaton approaches to modeling biological cells, in Mathematical Systems Theory in Biology, Communications, Computation, and Finance, Springer, New York, (2003), 1-39. |
[26] | A. Szabó, R. M. Merks, Cellular potts modeling of tumor growth, tumor invasion, and tumor evolution, Front. Oncol., 3 (2013). |
[27] | N. Guisoni, K. I. Mazzitello, L. Diambra, Modeling active cell movement with the potts model, Front. Phys., 6 (2018). |
[28] | E. G. Rens, L. Edelstein-Keshet, From energy to cellular forces in the cellular potts model: An algorithmic approach, PLOS Comput. Biol., 15 (2019), e1007459. |
[29] | K. A. Rejniak, A. R. A. Anderson, Hybrid models of tumor growth, Wiley Interdiscip. Rev.: Syst. Biol. Med., 3 (2011), 115-125. |
[30] | J. Jeon, V. Quaranta, P. T. Cummings, An off-lattice hybrid discrete-continuum model of tumor growth and invasion, Biophys. J., 98 (2010), 37-47. |
[31] | S. Koride, A. J. Loza, S. X. Sun, Epithelial vertex models with active biochemical regulation of contractility can explain organized collective cell motility, APL Bioeng., 2 (2018). |
[32] | S. Alt, P. Ganguly, G. Salbreux, Vertex models: from cell mechanics to tissue morphogenesis, Philos. Tran. R. Soc. London. Ser. B, Biol. Sci., 372 (2017). |
[33] | K. R. Foster, R. Koprowski, J. D. Skufca, Machine learning, medical diagnosis, and biomedical engineering research-commentary, Biomed. Eng. Online, 13 (2014), 94. |
[34] | A. M. Shirin, S. S. Dlay, W. L. Woo, G. V. Sherbet, Cross validation evaluation for breast cancer prediction using multilayer perceptron neural networks, Am. J. Eng. Appl. Sci., 4 (2012). |
[35] | T. Dietterich, Overfitting and undercomputing in machine learning, ACM Comput. Surveys, 27 (1995), 326-327. |
[36] | T. L. Whiteside, The tumor microenvironment and its role in promoting tumor growth, Oncogene, 27 (2018), 5904-5912. |
[37] | D. Hanahan, R. A. Weinberg, Hallmarks of cancer: the next generation, Cell, 144 (2011), 646-674. |
[38] | A. M. Newman, C. B. Steen, C. L. Liu, A. J. Gentles, A. A. Chaudhuri, F. Scherer, et al., Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat. Biotech., 37 (2019), 773-782. |
[39] | B. Chen, M. S. Khodadoust, C. L. Liu, A. M. Newman, A. A. Alizadeh, Profiling tumor infiltrating immune cells with CIBERSORT, Methods Mol. Biol., 1711 (2018), 243-259. |
[40] | K. Menden, M. Marouf, S. Oller, A. Dalmia, D. S. Magruder, K. Kloiber, et al., Deep learning-based cell composition analysis from tissue expression profiles, Sci. Adv., 6 (20100), eaba2619. |
[41] | X. Sun, B. Hu, Mathematical modeling and computational prediction of cancer drug resistance, Brief Bioinf., 19 (2018), 1382-1399. |
[42] | J. Cosgrove, J. Butler, K. Alden, M. Read, V. Kumar, L. Cucurull-Sanchez, et al., Agent-based modeling in systems pharmacology, CPT: Pharmacometrics Syst. Pharmacol., 4 (2015), 615-629. |
[43] | R. L. Dedrick, D. S. Zaharko, R. A. Bender, W. A. Bleyer, R. J. Lutz, Pharmacokinetic considerations on resistance to anticancer drugs, Cancer Chemother Rep., 59 (1975), 795-804. |
[44] | H. B. Frieboes, M. E. Edgerton, J. P. Fruehauf, F. R. A. J. Rose, L. K. Worrall, R. A. Gatenby, et al., Prediction of drug response in breast cancer using integrative experimental/computational modeling, Cancer Res., 69 (2009, 4484-4492. |
[45] | B. G. Birkhead, E. M. Rankin, S. Gallivan, L. Dones, R. D. Rubens, A mathematical model of the development of drug resistance to cancer chemotherapy, Eur. J. Cancer Clin. Oncol., 23 (1987), 1421-1427. |
[46] | A. Ghaffarizadeh, S. H. Friedman, P. Macklin, BioFVM: an efficient, parallelized diffusive transport solver for 3-D biological simulations, Bioinformatics, 32 (2015), 1256-1258. |
[47] | E. F. Juarez, R. Lau, S. H. Friedman, A. Ghaffarizadeh, E. Jonckheere, D. B. Agus, et al., Quantifying differences in cell line population dynamics using CellPD, BMC Syst. Biol., 10 (2016). |
[48] | S. H. Friedman, A. R. A. Anderson, D. M. Bortz, A. G. Fletcher, H. B. Frieboes, A. Ghaffarizadeh, et al., MultiCellDS: a standard and a community for sharing multicellular data, preprint, bioRxiv 090696. |
[49] | H. C. Tang, Y. C. Chen, Insight into molecular dynamics simulation of BRAF(V600E) and potent novel inhibitors for malignant melanoma, Int. J. Nanomedicine, 10 (2015), 3131-3146. |
[50] | K. J. Mahasa, R. Ouifki, A. Eladdadi, L. de Pillis, Mathematical model of tumor-immune surveillance, J. Theor. Biol., 404 (2016), 312-330. |
[51] | T. Jackson, A. Radunskaya, Applications of Dynamical Systems in Biology and Medicine, 2015. |
[52] | A. Dhawan, T. A. Graham, A. G. Fletcher, A computational modeling approach for deriving biomarkers to predict cancer risk in premalignant disease, Cancer Prev. Res., 9 (2016), 283-295. |
[53] | X. Yang, Y. Wang, R. Byrne, G. Schneider, S. Yang, Concepts of artificial intelligence for computer-assisted drug discovery, Chem. Rev., 119 (2019), 10520-10594. |
[54] | W. Kolch, D. Fey, Personalized computational models as biomarkers, J. Pers. Med., 7 (2017), 9. |
[55] | C. L. Fischer, A. M. Bates, E. A. Lanzel, J. M. Guthmiller, G. K. Johnson, N. K. Singh, et al., Computational models accurately predict multi-cell biomarker profiles in inflammation and cancer, Sci. Rep., 9 (2019), 10877. |
[56] | F. J. Esteva, G. N. Hortobagyi, Prognostic molecular markers in early breast cancer, Breast Cancer Res., 6 (2004), 109-118. |
[57] | S. Mojarad, B. Venturini, P. Fulgenzi, R. Papaleo, M. Brisigotti, F. Monti, et al., Prediction of nodal metastasis and prognosis of breast cancer by ANN-based assessment of tumour size and p53, Ki-67 and steroid receptor expression, Anticancer Res., 33 (2013), 3925-3933. |
[58] | M. P. Menden, F. Iorio, M. Garnett, U. McDermott, C. H. Benes, P. J. Ballester, et al., Machine learning prediction of cancer cell sensitivity to drugs based on genomic and chemical properties, Plos One, 8 (2013), e61318. |
[59] | A. Bravo, J. Pinero, N. Queralt-Rosinach, M. Rautschka, L. I. Furlong, Extraction of relations between genes and diseases from text and large-scale data analysis: implications for translational research, BMC Bioinfor., 16 (2015). |
[60] | J. Kim, J-j. Kim, H. Lee, An analysis of disease-gene relationship from medline abstracts by DigSee, Sci. Rep., 7 (2017), 40154. |
[61] | P. Mamoshina, M. Volosnikova, I. V. Ozerov, E. Putin, E. Skibina, F. Cortese, et al., Machine learning on human muscle transcriptomic data for biomarker discovery and tissue-specific drug target identification, Front. Genet., 9 (2018). |
[62] | T. Zhu, S, Cao, P. C. Su, R. Patel, D. Shah, H. B. Chokshi, et al., Hit identification and optimization in virtual screening: Practical recommendations based on a critical literature analysis, J. Med. Chem., 56 (2013), 6560-6572. |
[63] | G. Klopman, S. K. Chakravarti, H, Zhu, J. M. Ivanov, R. D. Saiakhov, ESP: A method to predict toxicity and pharmacological properties of chemicals using multiple MCASE databases, J. Chem. Infor. Comput. Sci., 44 (2004), 704-715. |
[64] | I. W. Mak, N. Evaniew, M. Ghert, Lost in translation: animal models and clinical trials in cancer treatment, Am. J. Trans. Res., 6 (2014), 114-118. |
[65] | B. Ramsundar, B. Liu, Z. Wu, A. Verra, M. Tudor, R. P. Sherridan, et al., Is multitask deep learning practical for pharma?, J. Chem. Infor. Model., 57 (2017), 2068-2076. |
[66] | M. Olivecrona, T. Blashcke, O. Engvist, H. Chen, Molecular de-novo design through deep reinforcement learning, J. Cheminfor., 9 (2017). |
[67] | T. Luechtefeld, D. Marsh, C. Rowlands, T. Hartung, Machine learning of toxicological big data enables read-across structure activity relationships (RASAR) outperforming animal test reproducibility, Toxicol. Sci., 165 (2018), 198-212. |
[68] | J. L. Perez-Gracia, M. F. Sanmamed, A. Bosch, A. Patino-Garcia, K. A. Schalper, V. Segura, et al., Strategies to design clinical studies to identify predictive biomarkers in cancer research, Cancer Treat. Rev., 53 (2017), 79-97. |
[69] | E. E. Bain, L. SHafner, D. P. Walling, A. A. Othman, C. C. Stein, J. Hinkle, et al., Use of a novel artificial intelligence platform on mobile devices to assess dosing compliance in a Phase 2 clinical trial in subjects with schizophrenia, JMIR mHealth uHealth, 5 (2017), e18. |
[70] | A. H. Beck, A. R. Sangoi, S. Leung, R. K. Marinelli, T. O. Nielsen, M. J. van de Vijver, et al., Systematic analysis of breast cancer morphology uncovers stromal features associated with survival, Sci. Trans. Med., 3 (2011). |
[71] | N. L. Mani, K. A. Schalper, C. Hatzis, O. Saglam, F. Tavassoli, M. Butler, et al., Quantitative assessment of the spatial heterogeneity of tumor-infiltrating lymphocytes in breast cancer, Breast Cancer Res., 18 (2016). |
[72] | F. Lake, Artificial intelligence in drug discovery: what is new, and what is next?, Future Drug Discovery, 1 (2019). |
[73] | K. L. Fetah, B. J. DiPArdo, E. M. Kongadzem, J. S. Tomlinson, A. Elzaghied, M. Elmusrati, et al., Cancer modeling-on-a-chip with future artificial intelligence integration, Small, 15 (2019). |
[74] | V. Assadollahi, B. Rashidieh, M. Alasvand, A. Abdolahi, J. A. Lopez, Interaction and molecular dynamics simulation study of Osimertinib (AstraZeneca 9291) anticancer drug with the EGFR kinase domain in native protein and mutated L844V and C797S, J. Cell. Biochem., 120 (2019), 13046-13055. |
[75] | S. Ghafari, M. Komeilian, M. Hashemi, S. Oushani, G. Rigi, B. Rashidieh, et al., Molecular docking based screening of Listeriolysin-O for improved inhibitors, Bioinformation, 13 (2017), 160-163. |
[76] | V. Assadollahi, B. Rashidieh, Molecular dynamics simulation of EFGR L844V mutant sensitive to AZD9291 in non-small cell lung cancer, J. Thorac. Oncol., 12 (2017), 1210. |
[77] | M. M. Ranbar, V. Assadolahi, M. Yazdani, D. Nikaein, B. Rashidieh, Virtual dual inhibition of COX-2/5-LOX enzymes based on binding properties of alpha-amyrins, the anti-inflammatory compound as a promising anti-cancer drug, EXCLI J., 15 (2016), 238-245. |
[78] | B. Rashidieh, M. Valizadeh, V. Assadollahi, M. M. Ranjbar, Molecular dynamics simulation on the low sensitivity of mutants of NEDD-8 activating enzyme for MLN4924 inhibitor as a cancer drug, Am. J. Cancer Res., 5 (2015), 3400-3406. |
[79] | B. Rashidieh, Z. Madani. M. K. Azam, S. K. Maklavani, N. R. Akbari, S. Tavakoli, et al., Molecular docking based virtual screening of compounds for inhibiting sortase A in L. monocytogenes, Bioinformation, 11 (2015), 501-505. |
[80] | B. Rashidieh, S. Etemadiafshar, G. Memari, M. Mirzaeichegeni, S. Yazdi, F. Farsimadan, et al., A molecular modeling based screening for potential inhibitors to alpha hemolysin from Staphylococcus aureus, Bioinformation, 11 (2015), 373-377. |
[81] | J. Ozik, N. Collier, R. Heiland, G. An, P. Macklin, Learning-accelerated discovery of immune-tumour interactions, Mol. Syst. Des. Eng., 4 (2019), 747-760. |
[82] | J. Ozik, N. Collier, J. M. Wozniak, C. Macal, C. Cockrell, S. F. Friedman, et al., High-throughput cancer hypothesis testing with an integrated PhysiCell-EMEWS workflow, BMC Bioinfor., 19 (2018). |
Specie | Mercury content (ppm) | Safety |
Anchovies | 0.017 ± 0.015 | Eco-good |
Atlantic cod | 0.095 ± 0.080 | Eco-bad |
Bass (saltwater, black, striped, rockfish) | 0.167 ± 0.194 | Eco-bad |
Bass Chilean | 0.354 ± 0.199 | Eco-bad |
Carp | 0.140 ± 0.099 | Eco-bad |
Catfish | 0.049 ± 0.084 | Eco-good |
Croaker Atlantic (Atlantic) | 0.069 ± 0.049 | Eco-good |
Croaker White (Pacific) | 0.287 ± 0.069 | Eco-bad |
Grouper (all species) | 0.448 ± 0.287 | Eco-bad |
Mullet | 0.05 ± 0.078 | Eco-bad |
Salmon, wild (Alaska) | 0.014 ± 0.041 | Eco-good |
Sardines, Pacific (US) | 0.016 ± 0.007 | Eco-good |
Shark | 0.979 ± 0.626 | Eco-bad |
Swordfish | 0.976 ± 0.510 | Eco-bad |
Tilapia | 0.013 ± 0.023 | Eco-good |
Trout, rainbow (farmed, freshwater) | 0.072 ± 0.143 | Eco-good |
Tuna species | 0.415 ± 0.308 | Eco-bad |
Specie | Mercury content (ppm) | Safety |
Anchovies | 0.017 ± 0.015 | Eco-good |
Atlantic cod | 0.095 ± 0.080 | Eco-bad |
Bass (saltwater, black, striped, rockfish) | 0.167 ± 0.194 | Eco-bad |
Bass Chilean | 0.354 ± 0.199 | Eco-bad |
Carp | 0.140 ± 0.099 | Eco-bad |
Catfish | 0.049 ± 0.084 | Eco-good |
Croaker Atlantic (Atlantic) | 0.069 ± 0.049 | Eco-good |
Croaker White (Pacific) | 0.287 ± 0.069 | Eco-bad |
Grouper (all species) | 0.448 ± 0.287 | Eco-bad |
Mullet | 0.05 ± 0.078 | Eco-bad |
Salmon, wild (Alaska) | 0.014 ± 0.041 | Eco-good |
Sardines, Pacific (US) | 0.016 ± 0.007 | Eco-good |
Shark | 0.979 ± 0.626 | Eco-bad |
Swordfish | 0.976 ± 0.510 | Eco-bad |
Tilapia | 0.013 ± 0.023 | Eco-good |
Trout, rainbow (farmed, freshwater) | 0.072 ± 0.143 | Eco-good |
Tuna species | 0.415 ± 0.308 | Eco-bad |