Next Article in Journal
Plant Resistance to Fungal Pathogens: Bibliometric Analysis and Visualization
Previous Article in Journal
Bioaccumulation and Biomagnification of Hexabromocyclododecane in Marine Biota from China: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Toxic Effects of Cadmium on Fish

Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Toxics 2022, 10(10), 622; https://doi.org/10.3390/toxics10100622
Submission received: 31 August 2022 / Revised: 6 October 2022 / Accepted: 17 October 2022 / Published: 19 October 2022
(This article belongs to the Section Ecotoxicology)

Abstract

:
Large amounts of enriched cadmium (Cd) in the environment seriously threatens the healthy and sustainable development of the aquaculture industry and greatly restricts the development of the food processing industry. Studying the distribution and toxic effects of Cd in fish, as well as the possible toxic effects of Cd on the human body, is very significant. A large number of studies have shown that the accumulation and distribution of Cd in fish are biologically specific, cause tissue differences, and seriously damage the integrity of tissue structure and function, the antioxidant defense system, the reproductive regulation system, and the immune system. The physiological, biochemical, enzyme, molecular, and gene expression levels change with different concentrations and times of Cd exposure, and these changes are closely related to the target sites of Cd action and tissues in fish. Therefore, the toxic effects of Cd on fish occur with multiple tissues, systems, and levels.

Graphical Abstract

1. Introduction

Cadmium (Cd), a toxic and nonessential transition metal, is a major threat to common organisms in aquatic ecosystems and has been widely recognized as a major pollutant in waters worldwide, and Cd travels up the food chain, eventually threatening human health, thus, Cd poses health risks to both humans and animals [1,2,3,4]. Cd causes a wide range of toxicological effects, including stomach cramps, vomiting, diarrhea, hematuria, kidney failure, and other symptoms. The Joint Expert Committee on Food Additives (JECFA) has stipulated that the allowable weekly intake limit of Cd for humans is 7 μg/kg, and exceeding the limit harms human health [1,5]. Epidemiological data suggest that environmental Cd exposure may be associated with various types of cancer and is also an important factor in osteoporosis [6,7]. Moreover, the liver and kidney are extremely sensitive to Cd toxicity, probably due to the ability of these tissues to synthesize metallothionein (MT), which is a Cd-induced protein that protects cells by tightly binding toxic Cd ions, thereby resulting in oxidative stress, this may be a cause of several liver and kidney diseases [8].
Cd has several characteristics, including high environmental toxicity, easy accumulation, and poor degradation. For organisms, Cd displays toxic effects on many organs, and Cd pollution is not easily found, however, Cd is easily enriched and can transmit its toxic effects through the food chain [9]. Cd pollution in the environment is caused by the development and utilization of chemical fertilizers in agriculture, metals and minerals in industry, and the use of some industrial products containing Cd, which may be discharged into water with wastewater. Even if the concentration of Cd is sometimes low, it can accumulate in algae and sediment, be absorbed by fish, shellfish, shrimp, and crabs in water, be concentrated through the food chain, and eventually enter the human body [1,10,11,12].
As fish are among the most important sources of food protein for human beings, it is highly likely that Cd can be enriched in fish and transferred into the human body through the food chain. Cd accumulation causes damage to the cardiovascular, immune, reproductive, and nervous systems in the body [13]. At the same time, it disrupts the cell cycle, proliferation, differentiation, and DNA replication and repair at the cellular level and then affects the apoptosis pathway [14,15]. Fish are an essential group of organisms in the aquatic ecosystem that are at the top of the food chain and are very sensitive to Cd pollution. If fish are exposed to Cd pollution for a long time, their different tissues will absorb and accumulate Cd, thus affecting the structure and function of the gills, liver, and gonad. Furthermore, Cd affects their physiological metabolism and reproductive and immune systems and eventually leads to metabolic disorders, physical disorders, or even death [16,17,18]. Recently, studies have shown that Cd can induce various epigenetic changes, thus, changes occur in the chemical modification of DNA, histones, and chromatin, but not the sequence of DNA nucleotides, including DNA methyltransferase, histone acetyltransferase, histone deacetylase, histone methyltransferase, and microRNA. As a result, fish incur a greater risk of disease and development of various types of cancer [18,19,20,21]. Therefore, using these indicators to indicate the impact that Cd pollutants have on the environment is an effective way to evaluate the safety of the ecosystem.
The toxic effects of Cd exposure in different fish are not the same. Adult male and female Japanese medaka were exposed to the non-lethal consequences of Cd, with 0–10 ppb concentration for 7 weeks, and the results displayed that gonadal steroid release was significantly decreased, while female plasma estradiol levels altered more significantly. Meanwhile, physiological response to Cd exposure along the hypothalamus–pituitary–gonadal axis, including the hepatic estrogen receptor, plasma vitellogenin, plasma steroids and gonadal–somatic indices, were more sensitive than the reproductive and developmental endpoints, and the reproductive and physiological endpoints for adult medaka pairs exposed to Cd for 7 weeks are shown in Table 1 [22].
When Oreochromis aureus were exposed to 0.5–10 ppm Cd, their food consumption showed a dramatic decrease, which would take 21 days to return to normal after fish were returned to Cd-free water, however, Cd was still in tissues for 34 days, and the accumulation was highest in the kidney after the fish were placed in Cd-free water [23]. Juvenile parrotfish, Oplegnathus fasciatus, were exposed to Cd mixed with a fish diet, and the result displayed that tissue Cd concentrations increased with the Cd content of the diets, and Cd accumulated the most in the liver, followed by the gill and then muscle [24]. Because zebrafish (Danio rerio) share 87% genetic homology with humans, their embryos, larvae, and adult fish are employed as standard models for many toxicity tests, and a summary of the toxic effects of cadmium in zebrafish is shown as Table 2 [25].

2. Detection Methods and Accumulation Effects of Cadmium in Fish

2.1. Cadmium Accumulation Induces Cytotoxicity in Fish

Studies have shown that Cd can enter cells through calcium ion channels. Cd and calcium (Ca) are both divalent metal ions. The radius of Cd2+ (0.097 nm) is similar to that of Ca2+ (0.099 nm), and Cd2+ has a higher affinity for anion binding sites in calcium ion channels than that of Ca2+, thus, Cd2+ can directly inhibit the active transport of Ca2+ for cells by competing with Ca2+. After Cd2+ enters cells, the physiological processes related to Ca2+ can be affected [46,47]. For example, Cd2+ can inhibit the outflow of Ca2+ by competing with Ca2+ for the binding site of Ca2+-ATPase, resulting in an increase in the intracellular Ca2+ concentration [48]. At the same time, Cd2+ can function as a substitute for Ca2+ to bind calmodulin (CaM), activate CaM-dependent kinases or directly activate its related enzymes, interfere with intracellular Ca2+-related signaling, and cause cytotoxicity [49,50]. In addition, Cd2+ can replace Ca2+ to bind to microtubules, microfilaments, and actin, destroy tight junctions and gap junctions between cells, damage cytoskeleton integrity, and ultimately affect the basic functions of cells [46,51,52]. Cd can induce cell apoptosis in two ways. First, Cd can bind to GSH rich in sulfhydryl groups on the mitochondrial membrane, causing proton (H+) influx, Ca2+ overload, and oxidative phosphoric acid uncoupling, which further leads to a decrease in the mitochondrial membrane potential, leads to the activation of Ca2+/Mg2+-dependent endonuclease (DNase I), and finally induces cell apoptosis [53,54]. Second, Cd can also lead to the release of cytochrome c and other apoptosis-related factors and increase the activity of caspases by inducing the opening of the mitochondrial permeability transition pore, which eventually leads to apoptosis [55,56]. In addition, Cd plays an important role in the induction of apoptosis by upregulating the expression of a series of proto-oncogenes [57]. The lethal effects of cadmium on different fish species are shown as Table 3.

2.2. Test Method for Cadmium Accumulation in Fish

Usually, there are multiple methods to detect the content of Cd in living organisms, and each method exhibits its own advantages and disadvantages. In the natural state, Cd mainly exists in the ionic state, but after entering the fish, most Cd ions quickly combine with metallothionein to form organic complexes, and a small part forms coordination compounds with nucleosides, proteins, and other substances in the body through ionic bonds [71,72]. At present, the methods used for detecting Cd in fish are usually atomic absorption spectroscopy (AAS) and inductively coupled plasma (ICP), the former includes graphite furnace atomic absorption spectrometry (GFAAS) and flame atomic absorption spectrometry (FAAS), and the latter includes inductively coupled plasma‒mass spectrometry (ICP‒MS) and inductively coupled plasma optical emission spectrometry (ICP‒OES) [73,74]. GFAAS has been widely used in the determination of Cd, due to its high sensitivity, low detection limit, and small sample amount, however, this method also has certain limitations, such as being susceptible to the influence of ash temperature, atomization temperature, matrix modifier condition, and the content of Cd in the sample. The FAAS method, which exhibits a high sensitivity, strong anti-interference ability, high precision, and good selectivity, is suitable for detecting the content of trace heavy metals, including Cd. The ICP‒MS method is widely used in determining the content of metal elements due to its advantages, including a strong anti-interference ability, low detection limit, high accuracy, good precision, convenient detection process, and accurate detection of low metal content. ICP‒OES has several advantages, including a wide linear range and a rapid and convenient detection process, but it is easily interfered with by spectrum and matrix effects [73,74,75].

2.3. The Accumulation of Cadmium in Fish Was Influenced by Feeding Habits and Routes

The degree of Cd accumulation in fish has a strong relationship with the fish feeding type. In the aquatic food chain, a step-by-step amplification effect, with increasing trophic levels, is observed for Cd accumulation. Considering the different cumulative effects of Cd on different feeding fish, the average accumulation coefficient of Cd in fish tissues can be generally arranged as omnivorous > carnivorous > herbivorous. The average accumulation coefficient of Cd, from largest to smallest, in the tissues of fish that live in different water layers is generally the bottom fish, middle and lower fish, and middle and upper fish, which may be due to a variety of factors, such as the position of the fish in the food chain and their metabolic capacity [76,77,78]. For instance, herbivorous fish mostly live in the upper and middle layers of water and mainly feed on phytoplankton or aquatic plants. The accumulation of Cd is mainly derived from intestinal food digestion. Omnivorous and carnivorous fish mostly inhabit the middle and lower layers of water and feed on small fish, aquatic insects, and shrimp, and they are exposed to Cd in the water environment and sediments, thus, the Cd accumulation in omnivorous and carnivorous fish is higher because they may ingest large amounts of Cd-containing sediments during feeding in the lower water [76,78].
The feeding route is another important factor that affects the accumulation and distribution of Cd in fish [79]. Cd is absorbed with water or foods and transported through the blood circulation to migrate and distribute in the different tissues and organs of fish. After being absorbed with water and transported through the blood circulation, Cd is mainly enriched in detoxifying organs, such as the kidney and liver, but when ingested with foods, Cd is mainly enriched in intestinal tissues, due to digestion and absorption in the intestine [80]. However, along with migration and distribution in fish, the accumulation of Cd that is both absorbed with water and foods displays gradual decreases in different organs or tissues. When absorbed with water, Cd ions are directly absorbed by gills in a dissolved state, and the accumulation of Cd in different tissues is generally in the following descending order: kidney, liver, intestine, gill, and muscle [81,82]. While absorbed with foods, they are absorbed by the digestive system, and the Cd accumulation in different tissues is generally in the following order, from high to low: intestine, kidney, liver, gill, and muscle [81,82,83]. The accumulation and distribution of Cd in fish were slightly different with different exposure methods. In order to assess health risk of some heavy metals, including Cd, from canned tuna and fish in Tijuana, Mexico, Rodriguez-Mendivil et al. diaplayed the range, mean concentrations, and incidence of occurrence in four species of fish for Cd in the analyzed samples in Table 4 [84].

3. Toxic Effects of Cadmium Accumulation on Fish

3.1. Cadmium Toxicity Damages Fish Tissue Structure

When fish are exposed to Cd for a long time, their organs or tissues become pathologically damaged, which can even lead to inflammation or necrosis [83]. The physiological functions of gills include regulating osmotic pressure, gas exchange, ion transport, acid-base balance, and excretion of metabolites, which play a key role in physiological, biochemical, and metabolic activities [85,86]. Gills are the first targets of Cd in water, through which fish continuously absorb Cd ions during respiration, and the absorbed Cd ions first accumulate in the surface cells of the gill tissue and are then transported through the blood to various organs or tissues. Therefore, Cd first has a toxic effect on the gill tissue structure of fish, leading to inflammation, apoptosis, necrosis, and so on [87,88]. Under Cd ion stress conditions in water, the structure of cells in gills changes, as well as the activities of ion transport and ATPase, thus, the ability of gills to regulate osmotic pressure and ion transport is affected. With the increase in concentration and time of Cd exposure, the problems, regarding inflammation, apoptosis, and necrosis, of gill tissue cells became more serious, and the lesions of the tissue structure became more obvious [89,90]. Evidence has shown that Lates calcarifer was exposed to 20.12 mg/L Cd for 48 h, resulting in aneurysm lesions in gill tissue. With the extension of exposure time, obvious pathological lesions, including hyperplasia, occurred in gill tissue at 72 h and 96 h [91]. The pathological changes in gill tissue caused by Cd mainly included cell proliferation, gill lamellae bending, aneurysm and cell shedding, and oxidative damage, such as vacuolization and swelling of mitochondria, and with the increase in Cd exposure dose and time, the problems became more obvious [88]. On the one hand, gill tissue lesions impact physiological function. On the other hand, the lesions also act a defense mechanism against Cd stress, as the distance that Cd must travel, the blood circulation is increased through the change in gill tissue structure, and to some extent, Cd can be prevented from entering other tissues in fish [92].
The liver, as the main detoxifying organ of fish, mainly protects the body from damage, turning heavy metals into less toxic substances, which are excreted from the body (Figure 1) [93]. Usually, Cd ions in the water environment first enter the fish body through gill respiration, then accumulate in the liver through the blood circulation system, and finally redistribute in various tissues. If fish are exposed to water that contains Cd ions for a long time, the Cd content in the liver exceeds its detoxification capacity, which affects the structural and functional integrity of fish liver tissue to different degrees [94,95]. With the increase in Cd concentration, the degree of irreversible damage to the structure of fish liver cells also increases significantly, which mainly includes endoplasmic reticulum expansion, nuclear deformation, nuclear membrane mitochondrial swelling, distortion, granular cytoplasm, and cell edema and cavitation. When the liver tissue structure is destroyed, the detoxification ability of the liver is significantly reduced, which has an irreversible impact on growth, reproduction, and survival [17,95,96].

3.2. Cadmium Toxicity Damages Fish Reproduction, Development and Endocrine System

Cd exposure seriously damages the reproductive system of fish. When exposed to Cd for a long time, the cellular structure and function of ovarian tissue or sperm tissue is significantly damaged, causing the reproductive ability of fish to reduce, the development of hatched embryos to be incomplete, and the growth and development of juvenile fish to be significantly retarded (Figure 1) [97,98]. Cd not only destroys the structural and functional integrity of fish gonads, but also affects the reproductive regulation system of fish by affecting the expression and secretion of hormones [99,100]. On the one hand, Cd can affect the expression and secretion of endocrine hormones, mainly by affecting the synthesis and secretion of hormones related to the hypothalamic–pituitary–gonadal (HPG) axis, and Cd also directly inhibits the expression of estrogen receptors [22,101]. For example, under Cd exposure, the expression of estrogen receptors in the gonads of Oncorhynchus mykiss was significantly inhibited, which affected the downstream pathways involved in estrogen receptors, resulting in disordered yolk production, abnormal egg development, and finally, reproductive abortion [102,103]. In addition, after Cd ion exposure, the pituitary gland secreted gonadotropin abnormally, resulting in the secretion of estrogen disorder and, finally, reducing the reproductive ability of fish. In addition to the direct inhibition of estrogen receptor expression, Cd can also bind to estrogen receptors in the form of estrogen analogs, thereby reducing the concentration of estrogen receptors in the gonads [31,102,104]. On the other hand, Cd can also affect the content of sex hormones in fish serum by inhibiting the synthesis of sterols, which has been demonstrated in mullet and guppies, in which the offspring numbers and survival rates were decreased under Cd stress conditions [103,105]. In addition, Cd can damage the genesis and maturation of germ cells, and high concentrations of Cd can reduce sperm motility and sperm number [106]. Zebrafish is a standard model animal for toxicity assessment, and studies showed that, after zebrafish were exposed to Cd solution, the gonad somatic index (GSI) of the ovary and seminal gland decreased significantly [18]. Cd mainly causes developmental abnormalities in zebrafish embryos, and the toxicity results of Cd in zebrafish are summarized in Table 2.

3.3. Cadmium Toxicity Damages the Immune System of Fish

The toxic effect of Cd on the fish immune system mainly occurs on the nonspecific immune system, and this affect mainly results in the inhibition of the number of white blood cells, the proliferation of lymphocytes, the phagocytosis rate of neutrophils, and the activity of macrophages [107,108]. The hemolymph system is an important target of Cd toxicity, and Cd exposure affects the organizational structural integrity of the fish hemolymph system and causes dysfunction, thereby resulting in an immunosuppressive effect [109,110]. Both in vitro and in vivo toxicity tests on Ictalurus melas showed that the proliferation of lymphocytes was significantly inhibited when the concentration of Cd used for exposure was higher than 2 μmol/L [111]. It has been found that exposure to high concentrations of Cd can inhibit the activities of lysozyme, alkaline phosphatase, and acid phosphatase in the liver of yellow catfish [112]. In addition, Cd can induce the proliferation of neutrophils in zebrafish larvae, and the secreted neutrophils will gradually accumulate in the inflammatory site of liver tissue (Figure 1) [113].
The other pathways that inhibit the nonspecific immune system, due to Cd exposure, inhibit the synthesis of essential trace elements [114]. Chronic exposure to environmental cadmium affects growth and survival, cellular stress, and glucose metabolism in juvenile channel catfish (Ictalurus punctatus) [115]. Because both zinc and copper are essential trace elements for organism immunity, if the zinc content is insufficient in an organism, the coefficient of the thymus, spleen, lymph node, and other organs will be reduced, and the cellular immune function will be reduced. Furthermore, if the copper content in the organism is deficient, humoral and cellular nonspecific immune functions will be reduced [114,116,117,118].

3.4. Cadmium Toxicity Damages the Energy Metabolism System of Fish

Energy metabolism plays an essential role in the growth and survival of organisms and is the basis of various life processes, such as proliferation and development, immune stress, and osmotic pressure regulation [19,119]. Cd can change ATPase activity and affect the function of ion channels. Studies have found that the activities of Ca2+-ATPase are inhibited as a result of the decrease in Ca2+ concentration in blood, which may be because Cd competitively binds to calcium channels in the gills after Cd exposure [120,121]. Researchers found that the activities of Na+/K+-ATPase in gills could be inhibited when the concentration of Cd in water was up to 1 mg/L, and the reason was that a large number of free radicals oxidize the amino acid and destroy the structure of ATPase, thus showing the inhibitory effects of Cd on ATPase [122]. Cd can not only affect the energy metabolism process in fish, but also cause damage to mitochondria, the main energy metabolism site. Regarding the effects of Cd on the mitochondria in Silurus meridionalis, it was shown that in Silurus meridionalis exposed to 500 μg/L Cd, the mitochondrial structure and function of the liver was significantly damaged and the activity of mitochondria enzymes was decreased, which results in a decrease in the energy of the liver metabolism [95]. Additional evidence verified that Cd could significantly reduce the level of ATP in the hemolymph of Ostreidae and induce energy metabolism disorder [123]. In addition, Cd exposure significantly affects the respiratory control rate and oxidative phosphorylation efficiency of mitochondria in Ostreidae and Oncorhynchus mykiss and interferes with the expression of key mitochondrial genes [53,124]. For example, Cd can induce the production of differentially expressed metabolites, such as succinate and glycogen, and proteins, including isocitrate dehydrogenase, pyruvate carboxylase, and glyceraldehyde-3-phosphate, that are related to energy metabolism in the gill tissues of Tegillarca granosa (Figure 1) [125].

3.5. Cadmium Toxicity Affects Nervous System Development in Fish

The neurotoxicity caused by Cd stress seriously threatens the normal life activities of fish because Cd directly inhibits the biological activities of enzymes that contain sulfhydryl by binding with sulfhydryl groups. At the same time, Cd leads to a decrease in the content of a series of metabolic intermediates, which causes serious damage to the nervous system of fish [126,127,128]. Cd can enter the brain tissue of fish and affect cerebellar functions, causing the fish to lose balance during swimming, while the effects on the brain are mainly neuronal damage. Cd exposure inhibited the formation of neural crest cells during the early development of fish, resulting in motor neuron damage and behavioral abnormalities [27,29,129]. Research has shown that Cd induces damage to nerve cells in the lateral line system of Dicentrarchus labrax and affects its escape and swimming behavior [130]. Cd also significantly inhibited acetylcholinesterase (AChE) activity in the brain tissue of zebrafish and Lepisosteus oculatus and induced neurological disorders [131,132]. Similarly, the behavior of juvenile zebrafish was severely inhibited under Cd exposure conditions [133]. In addition, Cd accumulates in the olfactory epithelium and bulb of fish, which reduces the expression of the genes related to olfactory sensory neurons, thereby damaging the olfactory sense of fish and damaging their anti-predator behavior (Figure 1) [134].

3.6. Cadmium Toxicity Leads to Changes in Blood Plasma Parameters

A study on Nile tilapia showed that albumin concentrations and serum total protein were decreased significantly (p < 0.001), while serum creatinine, glutamic-pyruvic transaminase (GPT), and aspartate aminotransferase (AST) significantly (p < 0.001) increased with increasing cadmium level in fish diets, and the urea-N concentration was insignificantly affected [135]. The results of the study on carp showed that cadmium poisoning would cause renal tubule reabsorption dysfunction and a large amount of calcium and phosphorus excretion, along with urine, and result in a decrease in serum calcium and phosphorus. In addition, cadmium can damage renal tubules, inhibit the activation of vitamin D, and also show a series of negative effects on calcium and phosphorus metabolism [92,136,137]. Usually, aminotransferases are most abundant in the liver and kidney, as well as in the heart muscle and bones, with very little in the serum, however, when these tissues, especially the liver and kidneys, are damaged or diseased, the enzyme enters the bloodstream and increases the activity of transaminase in the serum. Therefore, the increase and decrease of glutamic oxalacetic transaminase (GOT) and GPT activities in the serum may reflect the poisoning or pathological changes [138,139]. A study on the effects of cadmium exposure to Sparus aurata suggested that GOT activity decreased in liver cytoplasm, but increased in serum (Figure 1) [140].

4. Mechanism of the Toxic Effects of Cadmium Exposure on Fish

4.1. Cadmium Toxicity Leads to Oxidative Damage in Fish

When fish are stressed due to Cd in the water environment, a large number of reactive oxygen species (ROS) and reactive nitrogen species (RNS) radicals, which are beyond their own scavenging capacity, will accumulate in the fishs’ bodies, thereby resulting in a series of oxidative damage reactions [42,108]. The toxic effect of Cd on fish is mainly reflected in the oxidative stress caused by ROS, and the excessive accumulation of ROS will lead to structural changes in biological macromolecules, such as proteins and DNA, disorder of DNA replication and repair, and eventually pathological changes [141]. At the same time, ROS can cause unsaturated fatty acids on the cell membrane lipid peroxidation reaction to produce malondialdehyde (MDA) and cause cell membrane structural damage, in addition, MDA itself is a kind of toxic substance [94,142]. Combined, all of these factors will further affect the function of enzymes related to regulating cell proliferation and differentiation and apoptosis on the membrane, finally causing a series of functional disorders and damage to the body [94,143,144]. The increase in intracellular ROS caused by Cd mainly occurs through the following pathways: (I) Cd affects cellular respiration by inhibiting the respiratory transmission chain in the mitochondria, resulting in an increase in byproduct ROS content [145]; (II) Cd can increase ROS content by increasing the activity of nitric oxide synthase and the expression level of the related genes in the body [146]; (III) Cd can also remove Ca, Cu, Fe, and other elements from the binding proteins in the cell, thereby causing an increase in calcium, copper, iron, and other elements in the cytoplasm, resulting in oxidative stress and an increase in intracellular ROS content [147,148]; (IV) Cd reduces the antioxidant capacity of cells by reducing the activity of intracellular antioxidant enzymes (Figure 2) [143,149].

4.2. Cadmium Toxicity Affected the Expression of Stress Genes in Fish

Usually, when fish are subjected to various stresses in a water environment, a series of stress-related genes in the body are significantly inhibited or induced to be expressed, so that the toxic effect of stress can be alleviated through the body’s self-regulation [150]. As a stress defense mechanism, multiple types of genes are induced to be expressed under Cd stress, such as the oxidative stress-related genes GR, CYP1A, MT, and HSPs, antioxidant enzyme genes, such as CAT, SOD, and GSH-Px, and apoptosis-related genes, such as Bax, Bcl-2, Caspase-3, Caspase-8, and Caspase-9 [143,151]. The expression levels of antioxidant enzyme genes in the liver, spleen, and muscle of Takifugu obscurus exposed to Cd in a water environment were detected, and the results showed that the expression levels of MN-SOD, Cu/Zn-SOD, and CAT genes in the liver and spleen were significantly increased. These levels in muscle tissue were inhibited at the early stage and significantly increased at the middle and late stages of Cd stress, indicating that the muscle produced a large amount of antioxidant enzymes to protect the fish body in the middle and late stages of Cd stress through body self-regulation [152]. Therefore, the fish body can regulate the generation of a large number of antioxidant enzymes to reduce the oxidative damage caused by ROS. Moreover, Cd also induces the abnormal expression of stress response protein genes, such as heat shock protein and metallic globulin, as determined in studies on the liver in Cyprinus carpio and Labeo rohita, in which the expression levels of CYP1A, HSP47, HSP60, HSP70, HSP90, and MT-B genes were significantly increased and then inhibited with prolonged Cd exposure time [153,154]. Furthermore, the expression levels of anti-stress genes in different tissues of Cyprinus carpio were significantly different after Cd exposure for 96 h. For example, the expression level of HSP60 in the liver was significantly induced, but the expression levels of HSP70 and HSP90 were not significantly changed and inhibited. The expression level of HSP70 in the gills first significantly increased and then decreased, and the HSP90 expression level was significantly inhibited at all times (Figure 3) [153,155,156].

4.3. Cadmium Toxicity Inhibits Multiple Enzyme Activities

Cd can reduce the activities of many enzymes in fish, especially antioxidant enzymes that contain zinc and sulfhydryl, and Cd can bind to glutathione and metallothionein, which are two important thiol proteins that play an antioxidant role, thus, the ability of ROS elimination in the body is reduced and oxidative damage eventually occurs [157,158]. Cd binds to the sulfhydryl groups of superoxide dismutase (SOD) and glutathione reductase (GR), forms a Se-Cd complex with selenium (Se) in glutathione peroxidase (GPx), or replaces Zn in SOD to form Cd-SOD, thus, the activity of these enzymes is reduced or lost [159,160]. Studies have shown that the activities of SOD, catalase (CAT), and glutathione peroxidase (GPx) display an increasing trend in fish exposed to Cd at low concentrations, but usually show a decreasing trend at high concentrations, and the dose‒response curve of Cd generally shows a parabolic form, which has been verified in the liver and gills of Charybdis japonica [161]. Furthermore, excessive ROS will further lead to lipid peroxidation, mediate DNA damage, and promote the activation of poly-ADP-ribotransferase, thus reducing intracellular ATP levels. In addition, Cd inhibits the repair of DNA damage and the activity of DNA polymerase β, thereby affecting the immune function of the fish (Figure 3) [162,163].

5. Conclusions and Future Perspectives

This review summarizes the biological accumulation and toxicological effects of Cd in fish, including the research progress on the toxic effects of Cd on tissue structure and functional integrity, physiological metabolism, reproduction, immune system, and blood plasma parameters. We focus on the toxicological effects and mechanism of Cd in fish and anticipate future research directions to discover and reduce Cd pollution in the aquatic environment in a timely manner, achieve the purpose of protecting the aquatic ecological environment and aquatic life, and provide scientific evidence for humans to develop safe aquatic products.
At present, research on Cd toxicity is very active. Although many achievements have been made, from the molecular and metabolic perspectives, we believe that further research needs to be performed, as follows: (I) analyze the effects of low-dose and long-period Cd stress on fish and establish a database of toxicological effects of fish over time to provide a scientific basis for formulating water environmental sanitation standards; (II) strengthen the study on the combined toxicological effects of Cd and other environmental pollutants in the water environment, improve the database of toxicological indicators of the water environment and fish collected in the field, and comprehensively evaluate the ecotoxicological effects of Cd in the water environment; (III) and it is of great significance to strengthen the research on the toxicological effects and mechanisms of different forms of Cd on fish, to evaluate the ecological risk of Cd completely, to set environmental quality standards for Cd, and to repair and control Cd pollution in the water environment. Anyway, the mechanisms of the toxic effects of cadmium, combined with other heavy metals, on fish needs to be further studied. In recent years, the environmental hazards of microplastics are gaining researchers’ attention, and microplastics enhance the movement of cadmium through the food chain, which should be taken seriously.

Author Contributions

Investigation, data curation, writing—original draft, Y.L. (Yinai Liu) and Q.C.; writing—review and editing, Y.L. (Yinai Liu), Q.C., L.J. and R.P.; software, supervision, validation, Y.L. (Yaoqi Li) and L.B.; conceptualization, L.J. and R.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Natural Science Foundation of Zhejiang Province (grant No. LQ20C020003).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare that there are no conflicts of interest.

Abbreviations

Cdcadmium
MTmetallothionein
AASatomic absorption spectroscopy
ICPinductively coupled plasma
GFAASgraphite furnace atomic absorption spectrometry
FAASflame atomic absorption spectrometry
ICP‒MSinductively coupled plasma‒mass spectrometry
ICP‒OESinductively coupled plasma optical emission spectrometry
HPGhypothalamic–pituitary–gonadal axis
GSIgonad somatic index
AChEacetylcholinesterase
CaMcalmodulin
ROSreactive oxygen species
RNSreactive nitrogen species
MDAmalondialdehyde
GRglutathione reductase
SODsuperoxide dismutase
CATcatalase
GPxglutathione peroxidase

References

  1. Satarug, S.; Garrett, S.H.; Sens, M.A.; Sens, D.A. Cadmium, environmental exposure, and health outcomes. Environ. Health Perspect. 2010, 118, 182–190. [Google Scholar] [CrossRef] [PubMed]
  2. Jaiswal, A.; Verma, A.; Jaiswal, P. Detrimental Effects of Heavy Metals in Soil, Plants, and Aquatic Ecosystems and in Humans. J. Environ. Pathol. Toxicol. Oncol. Off. Organ Int. Soc. Environ. Toxicol. Cancer 2018, 37, 183–197. [Google Scholar] [CrossRef] [PubMed]
  3. Okereafor, U.; Makhatha, M.; Mekuto, L.; Uche-Okereafor, N.; Sebola, T.; Mavumengwana, V. Toxic Metal Implications on Agricultural Soils, Plants, Animals, Aquatic life and Human Health. Int. J. Environ. Res. Public Health 2020, 17, E2204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Silva, N.; Peiris-John, R.; Wickremasinghe, R.; Senanayake, H.; Sathiakumar, N. Cadmium a metalloestrogen: Are we convinced? J. Appl. Toxicol. 2012, 32, 318–332. [Google Scholar] [CrossRef] [PubMed]
  5. Bernhoft, R.A. Cadmium toxicity and treatment. Sci. World J. 2013, 2013, 394652. [Google Scholar] [CrossRef] [PubMed]
  6. Mezynska, M.; Brzóska, M.M. Environmental exposure to cadmium-a risk for health of the general population in industrialized countries and preventive strategies. Environ. Sci. Pollut. Res. Int. 2018, 25, 3211–3232. [Google Scholar] [CrossRef]
  7. Ma, Y.; Ran, D.; Shi, X.; Zhao, H.; Liu, Z. Cadmium toxicity: A role in bone cell function and teeth development. Sci. Total Environ. 2021, 769, 144646. [Google Scholar] [CrossRef] [PubMed]
  8. Matović, V.; Buha, A.; Ðukić-Ćosić, D.; Bulat, Z. Insight into the oxidative stress induced by lead and/or cadmium in blood, liver and kidneys. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2015, 78, 130–140. [Google Scholar] [CrossRef] [PubMed]
  9. Satarug, S. Dietary Cadmium Intake and Its Effects on Kidneys. Toxics 2018, 6, E15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Wang, M.; Chen, Z.; Song, W.; Hong, D.; Huang, L.; Li, Y. A review on Cadmium Exposure in the Population and Intervention Strategies Against Cadmium Toxicity. Bull. Environ. Contam. Toxicol. 2021, 106, 65–74. [Google Scholar] [CrossRef]
  11. Garner, R.; Levallois, P. Cadmium levels and sources of exposure among Canadian adults. Health Rep. 2016, 27, 10–18. [Google Scholar]
  12. Schaefer, H.R.; Dennis, S.; Fitzpatrick, S. Cadmium: Mitigation strategies to reduce dietary exposure. J. Food Sci. 2020, 85, 260–267. [Google Scholar] [CrossRef] [Green Version]
  13. Mielcarek, K.; Nowakowski, P.; Puścion-Jakubik, A.; Gromkowska-Kępka, K.J.; Soroczyńska, J.; Markiewicz-Żukowska, R.; Naliwajko, S.K.; Grabia, M.; Bielecka, J.; Żmudzińska, A.; et al. Arsenic, cadmium, lead and mercury content and health risk assessment of consuming freshwater fish with elements of chemometric analysis. Food Chem. 2022, 379, 132167. [Google Scholar] [CrossRef] [PubMed]
  14. Liu, Y.; Wang, P.; Wang, Y.; Zhu, Z.; Lao, F.; Liu, X.; Cong, W.; Chen, C.; Gao, Y.; Liu, Y. The influence on cell cycle and cell division by various cadmium-containing quantum dots. Small 2013, 9, 2440–2451. [Google Scholar] [CrossRef]
  15. Liao, Y.; Zheng, H.; Wu, L.; He, L.; Wang, Y.; Ou, Y.; Yang, H.; Peng, S.; Chen, F.; Wang, X.; et al. Cadmium cytotoxicity and possible mechanisms in human trophoblast HTR-8/SVneo cells. Environ. Toxicol. 2021, 36, 1111–1124. [Google Scholar] [CrossRef] [PubMed]
  16. Ferro, J.P.; Ferrari, L.; Eissa, B.L. Acute toxicity of cadmium to freshwater fishes and its relationship with body size and respiratory strategy. Comp. Biochem. Physiol. Toxicol. Pharmacol. CBP 2021, 248, 109109. [Google Scholar] [CrossRef] [PubMed]
  17. Noor, Z.; Khan, S.A.; Noor, M. Assessment of cadmium toxicity and its possible effects on goldfish (Carassius auratus), employing microscopy and biochemical techniques. Microsc. Res. Tech. 2020, 83, 1441–1449. [Google Scholar] [CrossRef] [PubMed]
  18. Zheng, J.-L.; Peng, L.-B.; Xia, L.-P.; Li, J.; Zhu, Q.-L. Effects of continuous and intermittent cadmium exposure on HPGL axis, GH/IGF axis and circadian rhythm signaling and their consequences on reproduction in female zebrafish: Biomarkers independent of exposure regimes. Chemosphere 2021, 282, 130879. [Google Scholar] [CrossRef] [PubMed]
  19. Cai, Y.; Yin, Y.; Li, Y.; Guan, L.; Zhang, P.; Qin, Y.; Wang, Y.; Li, Y. Cadmium exposure affects growth performance, energy metabolism, and neuropeptide expression in Carassius auratus gibelio. Fish Physiol. Biochem. 2020, 46, 187–197. [Google Scholar] [CrossRef] [PubMed]
  20. Ibrahim, A.T.A.; Banaee, M.; Sureda, A. Genotoxicity, oxidative stress, and biochemical biomarkers of exposure to green synthesized cadmium nanoparticles in Oreochromis niloticus (L.). Comp. Biochem. Physiol. Toxicol. Pharmacol. CBP 2021, 242, 108942. [Google Scholar] [CrossRef] [PubMed]
  21. Zhu, J.; Huang, Z.; Yang, F.; Zhu, M.; Cao, J.; Chen, J.; Lin, Y.; Guo, S.; Li, J.; Liu, Z. Cadmium disturbs epigenetic modification and induces DNA damage in mouse preimplantation embryos. Ecotoxicol. Environ. Saf. 2021, 219, 112306. [Google Scholar] [CrossRef] [PubMed]
  22. Tilton, S.C.; Foran, C.M.; Benson, W.H. Effects of cadmium on the reproductive axis of Japanese medaka (Oryzias latipes). Comp. Biochem. Physiol. Toxicol. Pharmacol. CBP 2003, 136, 265–276. [Google Scholar] [CrossRef] [PubMed]
  23. Woo, P.T.K.; Sin, Y.M.; Wong, M.K. The effects of short-term acute cadmium exposure on blue tilapia, Oreochromis aureus. Environ. Biol. Fishes 1993, 37, 67–74. [Google Scholar] [CrossRef]
  24. Okorie, O.E.; Bae, J.Y.; Lee, J.-H.; Lee, S.; Park, G.-H.; Mohseni, M.; Bai, S.C. Effects of Different Dietary Cadmium Levels on Growth and Tissue Cadmium Content in Juvenile Parrotfish, Oplegnathus fasciatus. Asian-Australas. J. Anim. Sci. 2014, 27, 62–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Min, E.K.; Lee, A.N.; Lee, J.-Y.; Shim, I.; Kim, P.; Kim, T.-Y.; Kim, K.-T.; Lee, S. Advantages of omics technology for evaluating cadmium toxicity in zebrafish. Toxicol. Res. 2021, 37, 395–403. [Google Scholar] [CrossRef]
  26. Kim, J.-Y.; Kim, S.-J.; Bae, M.A.; Kim, J.-R.; Cho, K.-H. Cadmium exposure exacerbates severe hyperlipidemia and fatty liver changes in zebrafish via impairment of high-density lipoproteins functionality. Toxicol. Vitr. 2018, 47, 249–258. [Google Scholar] [CrossRef]
  27. Monaco, A.; Grimaldi, M.C.; Ferrandino, I. Neuroglial alterations in the zebrafish brain exposed to cadmium chloride. J. Appl. Toxicol. 2016, 36, 1629–1638. [Google Scholar] [CrossRef]
  28. Senger, M.R.; Rosemberg, D.B.; Rico, E.P.; Arizi, M.d.B.; Dias, R.D.; Bogo, M.R.; Bonan, C.D. In vitro effect of zinc and cadmium on acetylcholinesterase and ectonucleotidase activities in zebrafish (Danio rerio) brain. Toxicol. Vitr. 2006, 20, 954–958. [Google Scholar] [CrossRef]
  29. Chow, E.S.H.; Hui, M.N.Y.; Lin, C.C.; Cheng, S.H. Cadmium inhibits neurogenesis in zebrafish embryonic brain development. Aquat. Toxicol. 2008, 87, 157–169. [Google Scholar] [CrossRef]
  30. Tu, H.; Fan, C.; Chen, X.; Liu, J.; Wang, B.; Huang, Z.; Zhang, Y.; Meng, X.; Zou, F. Effects of cadmium, manganese, and lead on locomotor activity and neurexin 2a expression in zebrafish. Environ. Toxicol. Chem. 2017, 36, 2147–2154. [Google Scholar] [CrossRef]
  31. Chouchene, L.; Pellegrini, E.; Gueguen, M.-M.; Hinfray, N.; Brion, F.; Piccini, B.; Kah, O.; Saïd, K.; Messaoudi, I.; Pakdel, F. Inhibitory effect of cadmium on estrogen signaling in zebrafish brain and protection by zinc. J. Appl. Toxicol. 2016, 36, 863–871. [Google Scholar] [CrossRef] [PubMed]
  32. Hen Chow, E.S.; Cheng, S.H. Cadmium Affects Muscle Type Development and Axon Growth in Zebrafish Embryonic Somitogenesis. Toxicol. Sci. 2003, 73, 149–159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Zhang, T.; Zhou, X.-Y.; Ma, X.-F.; Liu, J.-X. Mechanisms of cadmium-caused eye hypoplasia and hypopigmentation in zebrafish embryos. Aquat. Toxicol. 2015, 167, 68–76. [Google Scholar] [CrossRef] [PubMed]
  34. Fraysse, B.; Mons, R.; Garric, J. Development of a zebrafish 4-day embryo-larval bioassay to assess toxicity of chemicals. Ecotoxicol. Environ. Saf. 2006, 63, 253–267. [Google Scholar] [CrossRef] [PubMed]
  35. White, R.J.; Collins, J.E.; Sealy, I.M.; Wali, N.; Dooley, C.M.; Digby, Z.; Stemple, D.L.; Murphy, D.N.; Billis, K.; Hourlier, T.; et al. A high-resolution mRNA expression time course of embryonic development in zebrafish. ELife 2017, 6, e30860. [Google Scholar] [CrossRef]
  36. Xiao, B.; Chen, T.-M.; Zhong, Y. Possible molecular mechanism underlying cadmium-induced circadian rhythms disruption in zebrafish. Biochem. Biophys. Res. Commun. 2016, 481, 201–205. [Google Scholar] [CrossRef] [PubMed]
  37. Matz, C.J.; Krone, P.H. Cell Death, Stress-Responsive Transgene Activation, and Deficits in the Olfactory System of Larval Zebrafish Following Cadmium Exposure. Environ. Sci. Technol. 2007, 41, 5143–5148. [Google Scholar] [CrossRef]
  38. Veneman, W.J.; Spaink, H.P.; Brun, N.R.; Bosker, T.; Vijver, M.G. Pathway analysis of systemic transcriptome responses to injected polystyrene particles in zebrafish larvae. Aquat. Toxicol. 2017, 190, 112–120. [Google Scholar] [CrossRef]
  39. Pan, Y.-X.; Luo, Z.; Zhuo, M.-Q.; Wei, C.-C.; Chen, G.-H.; Song, Y.-F. Oxidative stress and mitochondrial dysfunction mediated Cd-induced hepatic lipid accumulation in zebrafish Danio rerio. Aquat. Toxicol. 2018, 199, 12–20. [Google Scholar] [CrossRef]
  40. Zheng, J.-L.; Yuan, S.-S.; Wu, C.-W.; Li, W.-Y. Chronic waterborne zinc and cadmium exposures induced different responses towards oxidative stress in the liver of zebrafish. Aquat. Toxicol. 2016, 177, 261–268. [Google Scholar] [CrossRef]
  41. Zheng, J.-L.; Yuan, S.-S.; Wu, C.-W.; Lv, Z.-M. Acute exposure to waterborne cadmium induced oxidative stress and immunotoxicity in the brain, ovary and liver of zebrafish (Danio rerio). Aquat. Toxicol. 2016, 180, 36–44. [Google Scholar] [CrossRef] [PubMed]
  42. Zheng, J.-L.; Yuan, S.-S.; Wu, C.-W.; Lv, Z.-M.; Zhu, A.-Y. Circadian time-dependent antioxidant and inflammatory responses to acute cadmium exposure in the brain of zebrafish. Aquat. Toxicol. 2017, 182, 113–119. [Google Scholar] [CrossRef] [PubMed]
  43. Avallone, B.; Agnisola, C.; Cerciello, R.; Panzuto, R.; Simoniello, P.; Cretì, P.; Motta, C.M. Structural and functional changes in the zebrafish (Danio rerio) skeletal muscle after cadmium exposure. Cell Biol. Toxicol. 2015, 31, 273–283. [Google Scholar] [CrossRef]
  44. Wu, S.M.; Tsai, P.J.; Chou, M.Y.; Wang, W.-D. Effects of Maternal Cadmium Exposure on Female Reproductive Functions, Gamete Quality, and Offspring Development in Zebrafish (Danio rerio). Arch. Environ. Contam. Toxicol. 2013, 65, 521–536. [Google Scholar] [CrossRef]
  45. Avallone, B.; Crispino, R.; Cerciello, R.; Simoniello, P.; Panzuto, R.; Maria Motta, C. Cadmium effects on the retina of adult Danio rerio. Comptes Rendus Biol. 2015, 338, 40–47. [Google Scholar] [CrossRef]
  46. Choong, G.; Liu, Y.; Templeton, D.M. Interplay of calcium and cadmium in mediating cadmium toxicity. Chem. -Biol. Interact. 2014, 211, 54–65. [Google Scholar] [CrossRef]
  47. Zhou, X.; Hao, W.; Shi, H.; Hou, Y.; Xu, Q. Calcium homeostasis disruption-a bridge connecting cadmium-induced apoptosis, autophagy and tumorigenesis. Oncol. Res. Treat. 2015, 38, 311–315. [Google Scholar] [CrossRef] [PubMed]
  48. Da Costa, R.; Botana, D.; Piñero, S.; Proverbio, F.; Marín, R. Cadmium inhibits motility, activities of plasma membrane Ca2+-ATPase and axonemal dynein-ATPase of human spermatozoa. Andrologia 2016, 48, 464–469. [Google Scholar] [CrossRef]
  49. Chmielowska-Bąk, J.; Izbiańska, K.; Deckert, J. The toxic Doppelganger: On the ionic and molecular mimicry of cadmium. Acta Biochim. Pol. 2013, 60, 369–374. [Google Scholar] [CrossRef] [Green Version]
  50. Suzuki, Y.; Chao, S.H.; Zysk, J.R.; Cheung, W.Y. Stimulation of calmodulin by cadmium ion. Arch. Toxicol. 1985, 57, 205–211. [Google Scholar] [CrossRef]
  51. Templeton, D.M.; Liu, Y. Effects of cadmium on the actin cytoskeleton in renal mesangial cells. Can. J. Physiol. Pharmacol. 2013, 91, 1–7. [Google Scholar] [CrossRef] [PubMed]
  52. Ge, Y.; Song, X.; Chen, L.; Hu, D.; Hua, L.; Cui, Y.; Liu, J.; An, Z.; Yin, Z.; Ning, H. Cadmium induces actin cytoskeleton alterations and dysfunction in Neuro-2a cells. Environ. Toxicol. 2019, 34, 469–475. [Google Scholar] [CrossRef] [PubMed]
  53. Adiele, R.C.; Stevens, D.; Kamunde, C. Reciprocal enhancement of uptake and toxicity of cadmium and calcium in rainbow trout (Oncorhynchus mykiss) liver mitochondria. Aquat. Toxicol. 2010, 96, 319–327. [Google Scholar] [CrossRef] [PubMed]
  54. Nigam, D.; Shukla, G.S.; Agarwal, A.K. Glutathione depletion and oxidative damage in mitochondria following exposure to cadmium in rat liver and kidney. Toxicol. Lett. 1999, 106, 151–157. [Google Scholar] [CrossRef]
  55. Mao, W.P.; Zhang, N.N.; Zhou, F.Y.; Li, W.X.; Liu, H.Y.; Feng, J.; Zhou, L.; Wei, C.J.; Pan, Y.B.; He, Z.J. Cadmium directly induced mitochondrial dysfunction of human embryonic kidney cells. Hum. Exp. Toxicol. 2011, 30, 920–929. [Google Scholar] [CrossRef] [PubMed]
  56. Gobe, G.; Crane, D. Mitochondria, reactive oxygen species and cadmium toxicity in the kidney. Toxicol. Lett. 2010, 198, 49–55. [Google Scholar] [CrossRef] [PubMed]
  57. Li, G.-Y.; Kim, M.; Kim, J.-H.; Lee, M.-O.; Chung, J.-H.; Lee, B.-H. Gene expression profiling in human lung fibroblast following cadmium exposure. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2008, 46, 1131–1137. [Google Scholar] [CrossRef] [PubMed]
  58. Liu, W.; Qiu, H.; Yan, Y.; Xie, X. Acute Cd Toxicity, Metal Accumulation, and Ion Loss in Southern Catfish (Silurus meridionalis Chen). Toxics 2021, 9, 202. [Google Scholar] [CrossRef] [PubMed]
  59. Renieri, E.A.; Sfakianakis, D.G.; Alegakis, A.A.; Safenkova, I.V.; Buha, A.; Matović, V.; Tzardi, M.; Dzantiev, B.B.; Divanach, P.; Kentouri, M.; et al. Nonlinear responses to waterborne cadmium exposure in zebrafish. An in vivo study. Environ. Res. 2017, 157, 173–181. [Google Scholar] [CrossRef] [PubMed]
  60. Ismail, A.; Yusof, S. Effect of mercury and cadmium on early life stages of Java medaka (Oryzias javanicus): A potential tropical test fish. Mar. Pollut. Bull. 2011, 63, 347–349. [Google Scholar] [CrossRef]
  61. Reyes-Becerril, M.; Angulo, C.; Sanchez, V.; Cuesta, A.; Cruz, A. Methylmercury, cadmium and arsenic (III)-induced toxicity, oxidative stress and apoptosis in Pacific red snapper leukocytes. Aquat. Toxicol. 2019, 213, 105223. [Google Scholar] [CrossRef]
  62. Jurgelėnė, Ž.; Stankevičiūtė, M.; Kazlauskienė, N.; Baršienė, J.; Jokšas, K.; Markuckas, A. Toxicological Potential of Cadmium Impact on Rainbow Trout (Oncorhynchus mykiss) in Early Development. Bull. Environ. Contam. Toxicol. 2019, 103, 544–550. [Google Scholar] [CrossRef] [PubMed]
  63. Shuhaimi-Othman, M.; Yakub, N.; Ramle, N.-A.; Abas, A. Comparative toxicity of eight metals on freshwater fish. Toxicol. Ind. Health 2015, 31, 773–782. [Google Scholar] [CrossRef] [PubMed]
  64. Robohm, R.A. Paradoxical effects of cadmium exposure on antibacterial antibody responses in two fish species: Inhibition in cunners (Tautogolabrus adspersus) and enhancement in striped bass (Morone saxatilis). Vet. Immunol. Immunopathol. 1986, 12, 251–262. [Google Scholar] [CrossRef]
  65. Garcia-Santos, S.; Fontaínhas-Fernandes, A.; Wilson, J.M. Cadmium tolerance in the Nile tilapia (Oreochromis niloticus) following acute exposure: Assessment of some ionoregulatory parameters. Environ. Toxicol. 2006, 21, 33–46. [Google Scholar] [CrossRef]
  66. Delahaut, V.; Rašković, B.; Salvado, M.S.; Bervoets, L.; Blust, R.; De Boeck, G. Toxicity and bioaccumulation of Cadmium, Copper and Zinc in a direct comparison at equitoxic concentrations in common carp (Cyprinus carpio) juveniles. PLoS ONE 2020, 15, e0220485. [Google Scholar] [CrossRef] [Green Version]
  67. Annune, P.A.; Ebele, S.O.; Oladimeji, A.A. Acute toxicity of cadmium to juveniles of Clarias gariepinus (Teugels) and Oreochromis niloticus (Trewavas). J. Environ. Sci. Health Part A Environ. Sci. Eng. Toxicol. 1994, 29, 1357–1365. [Google Scholar] [CrossRef]
  68. Dutta, T.K.; Kaviraj, A. Acute toxicity of cadmium to fish Labeo rohita and copepod Diaptomus forbesi pre-exposed to CaO and KMnO4. Chemosphere 2001, 42, 955–958. [Google Scholar] [CrossRef]
  69. Roy, S.; Karmakar, D.; Pal, S. Acute Toxicity Bioassay and Determination of LC50 of Cadmium Chloride in Trichogaster (Colisa) fasciata. Appl. Biochem. Biotechnol. 2022, 194, 3890–3900. [Google Scholar] [CrossRef] [PubMed]
  70. Zhang, H.; Cao, H.; Meng, Y.; Jin, G.; Zhu, M. The toxicity of cadmium (Cd2+) towards embryos and pro-larva of soldatov’s catfish (Silurus soldatovi). Ecotoxicol. Environ. Saf. 2012, 80, 258–265. [Google Scholar] [CrossRef]
  71. Andersen, O. Chelation of cadmium. Environ. Health Perspect. 1984, 54, 249–266. [Google Scholar] [CrossRef] [PubMed]
  72. Sandbichler, A.M.; Höckner, M. Cadmium Protection Strategies—A Hidden Trade-Off? Int. J. Mol. Sci. 2016, 17, 139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Pyrzynska, K.; Kilian, K. On-line sorption-based systems for determination of cadmium with atomic spectrometry detectors. Water Res. 2007, 41, 2839–2851. [Google Scholar] [CrossRef] [PubMed]
  74. Ferreira, S.L.C.; de Andrade, J.B.; Maria das Graças, A.K.; Pereira, M.D.G.; Lemos, V.A.; dos Santos, W.N.L.; Rodrigues, F.D.M.; Souza, A.S.; Ferreira, H.S.; da Silva, E.G.P. Review of procedures involving separation and preconcentration for the determination of cadmium using spectrometric techniques. J. Hazard. Mater. 2007, 145, 358–367. [Google Scholar] [CrossRef] [PubMed]
  75. Lemos, V.A.; de Carvalho, A.L. Determination of cadmium and lead in human biological samples by spectrometric techniques: A review. Environ. Monit. Assess. 2010, 171, 255–265. [Google Scholar] [CrossRef] [PubMed]
  76. Tao, Y.; Yuan, Z.; Xiaona, H.; Wei, M. Distribution and bioaccumulation of heavy metals in aquatic organisms of different trophic levels and potential health risk assessment from Taihu lake, China. Ecotoxicol. Environ. Saf. 2012, 81, 55–64. [Google Scholar] [CrossRef]
  77. Weber, P.; Behr, E.R.; Knorr, C.D.L.; Vendruscolo, D.S.; Flores, E.M.M.; Dressler, V.L.; Baldisserotto, B. Metals in the water, sediment, and tissues of two fish species from different trophic levels in a subtropical Brazilian river. Microchem. J. 2013, 106, 61–66. [Google Scholar] [CrossRef]
  78. Rejomon, G.; Nair, M.; Joseph, T. Trace metal dynamics in fishes from the southwest coast of India. Environ. Monit. Assess. 2010, 167, 243–255. [Google Scholar] [CrossRef] [PubMed]
  79. Ayyat, M.S.; Mahmoud, H.K.; El-Hais, A.E.-A.M.; Abd El-Latif, K.M. The role of some feed additives in fish fed on diets contaminated with cadmium. Environ. Sci. Pollut. Res. Int. 2017, 24, 23636–23645. [Google Scholar] [CrossRef]
  80. Junejo, S.H.; Baig, J.A.; Kazi, T.G.; Afridi, H.I. Cadmium and Lead Hazardous Impact Assessment of Pond Fish Species. Biol. Trace Elem. Res. 2019, 191, 502–511. [Google Scholar] [CrossRef] [PubMed]
  81. Klinck, J.S.; Green, W.W.; Mirza, R.S.; Nadella, S.R.; Chowdhury, M.J.; Wood, C.M.; Pyle, G.G. Branchial cadmium and copper binding and intestinal cadmium uptake in wild yellow perch (Perca flavescens) from clean and metal-contaminated lakes. Aquat. Toxicol. 2007, 84, 198–207. [Google Scholar] [CrossRef] [PubMed]
  82. Franklin, N.M.; Glover, C.N.; Nicol, J.A.; Wood, C.M. Calcium/cadmium interactions at uptake surfaces in rainbow trout: Waterborne versus dietary routes of exposure. Environ. Toxicol. Chem. 2005, 24, 2954–2964. [Google Scholar] [CrossRef] [PubMed]
  83. Li, J.; Yan, Y.; Xie, X. Tissue-Specific Antioxidative Responses and Cadmium Accumulation in Silurus meridionalis Under Chronic Waterborne Cadmium Exposure. Bull. Environ. Contam. Toxicol. 2018, 100, 485–491. [Google Scholar] [CrossRef] [PubMed]
  84. Rodriguez-Mendivil, D.D.; Garcia-Flores, E.; Temores-Pena, J.; Wakida, F.T. Health Risk Assessment of Some Heavy Metals from Canned Tuna and Fish in Tijuana, Mexico. Health Scope 2019, 8, e78956. [Google Scholar] [CrossRef] [Green Version]
  85. Olson, K.R. Vasculature of the fish gill: Anatomical correlates of physiological functions. J. Electron. Microsc. Tech. 1991, 19, 389–405. [Google Scholar] [CrossRef] [PubMed]
  86. 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]
  87. Manera, M.; Giari, L.; Depasquale, J.A.; Dezfuli, B.S. European Sea bass gill pathology after exposure to cadmium and terbuthylazine: Expert versus fractal analysis. J. Microsc. 2016, 261, 291–299. [Google Scholar] [CrossRef] [PubMed]
  88. Manera, M.; Giari, L.; De Pasquale, J.A.; Sayyaf Dezfuli, B. Local connected fractal dimension analysis in gill of fish experimentally exposed to toxicants. Aquat. Toxicol. 2016, 175, 12–19. [Google Scholar] [CrossRef]
  89. Garcia-Santos, S.; Vargas-Chacoff, L.; Ruiz-Jarabo, I.; Varela, J.L.; Mancera, J.M.; Fontaínhas-Fernandes, A.; Wilson, J.M. Metabolic and osmoregulatory changes and cell proliferation in gilthead sea bream (Sparus aurata) exposed to cadmium. Ecotoxicol. Environ. Saf. 2011, 74, 270–278. [Google Scholar] [CrossRef] [PubMed]
  90. Da Silva, A.O.F.; Martinez, C.B.R. Acute effects of cadmium on osmoregulation of the freshwater teleost Prochilodus lineatus: Enzymes activity and plasma ions. Aquat. Toxicol. 2014, 156, 161–168. [Google Scholar] [CrossRef]
  91. Thophon, S.; Kruatrachue, M.; Upatham, E.S.; Pokethitiyook, P.; Sahaphong, S.; Jaritkhuan, S. Histopathological alterations of white seabass, Lates calcarifer, in acute and subchronic cadmium exposure. Environ. Pollut. 2003, 121, 307–320. [Google Scholar] [CrossRef]
  92. Yeşilbudak, B.; Erdem, C. Cadmium accumulation in gill, liver, kidney and muscle tissues of common carp, Cyprinus carpio, and Nile tilapia, Oreochromis niloticus. Bull. Environ. Contam. Toxicol. 2014, 92, 546–550. [Google Scholar] [CrossRef] [PubMed]
  93. Franco-Fuentes, E.; Moity, N.; Ramírez-González, J.; Andrade-Vera, S.; González-Weller, D.; Hardisson, A.; Paz, S.; Rubio, C.; Gutiérrez, A.J. Metal and metalloids concentration in Galapagos fish liver and gonad tissues. Mar. Pollut. Bull. 2021, 173, 112953. [Google Scholar] [CrossRef] [PubMed]
  94. Dai, Z.; Cheng, J.; Bao, L.; Zhu, X.; Li, H.; Chen, X.; Zhang, Y.; Zhang, J.; Chu, W.; Pan, Y.; et al. Exposure to waterborne cadmium induce oxidative stress, autophagy and mitochondrial dysfunction in the liver of Procypris merus. Ecotoxicol. Environ. Saf. 2020, 204, 111051. [Google Scholar] [CrossRef] [PubMed]
  95. Li, J.; Xie, X. Inconsistent responses of liver mitochondria metabolism and standard metabolism in Silurus meridionalis when exposed to waterborne cadmium. Comp. Biochem. Physiol. Toxicol. Pharmacol. CBP 2018, 214, 17–22. [Google Scholar] [CrossRef] [PubMed]
  96. Jabeen, G.; Ishaq, S.; Arshad, M.; Fatima, S.; Kanwal, Z.; Ali, F. Investigation on Immune-Related Protein (Heat Shock Proteins and Metallothionein) Gene Expression Changes and Liver Histopathology in Cadmium-Stressed Fish. BioMed Res. Int. 2022, 2022, 2075791. [Google Scholar] [CrossRef]
  97. Pinto, G.L.; da Silva Castro, J.; Val, A.L. Copper and cadmium impair sperm performance, fertilization and hatching of oocytes from Amazonian fish Colossoma macropomum. Chemosphere 2021, 266, 128957. [Google Scholar] [CrossRef]
  98. Sierra-Marquez, L.; Espinosa-Araujo, J.; Atencio-Garcia, V.; Olivero-Verbel, J. Effects of cadmium exposure on sperm and larvae of the neotropical fish Prochilodus magdalenae. Comp. Biochem. Physiol. Toxicol. Pharmacol. CBP 2019, 225, 108577. [Google Scholar] [CrossRef] [PubMed]
  99. Liu, P.; Zhao, Y.; Wang, S.; Xing, H.; Dong, W.-F. Effect of combined exposure to silica nanoparticles and cadmium chloride on female zebrafish ovaries. Environ. Toxicol. Pharmacol. 2021, 87, 103720. [Google Scholar] [CrossRef] [PubMed]
  100. Luo, Y.; Shan, D.; Zhong, H.; Zhou, Y.; Chen, W.; Cao, J.; Guo, Z.; Xiao, J.; He, F.; He, Y.; et al. Subchronic effects of cadmium on the gonads, expressions of steroid hormones and sex-related genes in tilapia Oreochromis niloticus. Ecotoxicology 2015, 24, 2213–2223. [Google Scholar] [CrossRef]
  101. Tian, J.; Hu, J.; He, W.; Zhou, L.; Huang, Y. Parental exposure to cadmium chloride causes developmental toxicity and thyroid endocrine disruption in zebrafish offspring. Comp. Biochem. Physiol. Toxicol. Pharmacol. CBP 2020, 234, 108782. [Google Scholar] [CrossRef]
  102. Guével, R.L.; Petit, F.G.; Goff, P.L.; Métivier, R.; Valotaire, Y.; Pakdel, F. Inhibition of rainbow trout (Oncorhynchus mykiss) estrogen receptor activity by cadmium. Biol. Reprod. 2000, 63, 259–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Amutha, C.; Subramanian, P. Cadmium alters the reproductive endocrine disruption and enhancement of growth in the early and adult stages of Oreochromis mossambicus. Fish Physiol. Biochem. 2013, 39, 351–361. [Google Scholar] [CrossRef] [PubMed]
  104. Nesatyy, V.J.; Ammann, A.A.; Rutishauser, B.V.; Suter, M.J.F. Effect of cadmium on the interaction of 17beta-estradiol with the rainbow trout estrogen receptor. Environ. Sci. Technol. 2006, 40, 1358–1363. [Google Scholar] [CrossRef]
  105. Sellin, M.K.; Kolok, A.S. Cd exposures in fathead minnows: Effects on adult spawning success and reproductive physiology. Arch. Environ. Contam. Toxicol. 2006, 51, 594–599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Vergilio, C.S.; Moreira, R.V.; Carvalho, C.E.V.; Melo, E.J.T. Evolution of cadmium effects in the testis and sperm of the tropical fish Gymnotus carapo. Tissue Cell 2015, 47, 132–139. [Google Scholar] [CrossRef] [PubMed]
  107. Choudhury, C.; Mazumder, R.; Biswas, R.; Sengupta, M. Cadmium exposure induces inflammation through the canonical NF-κΒ pathway in monocytes/macrophages of Channa punctatus Bloch. Fish Shellfish. Immunol. 2021, 110, 116–126. [Google Scholar] [CrossRef]
  108. Jiaxin, S.; Shengchen, W.; Yirong, C.; Shuting, W.; Shu, L. Cadmium exposure induces apoptosis, inflammation and immunosuppression through CYPs activation and antioxidant dysfunction in common carp neutrophils. Fish Shellfish. Immunol. 2020, 99, 284–290. [Google Scholar] [CrossRef]
  109. Naz, S.; Hussain, R.; Ullah, Q.; Chatha, A.M.M.; Shaheen, A.; Khan, R.U. Toxic effect of some heavy metals on hematology and histopathology of major carp (Catla catla). Environ. Sci. Pollut. Res. Int. 2021, 28, 6533–6539. [Google Scholar] [CrossRef]
  110. Gill, T.S.; Pant, J.C. Erythrocytic and leukocytic responses to cadmium poisoning in a freshwater fish, Puntius conchonius ham. Environ. Res. 1985, 36, 327–337. [Google Scholar] [CrossRef]
  111. Albergoni, V.; Viola, A. Effects of cadmium on lymphocyte proliferation and macrophage activation in catfish, Ictalurus melas. Fish Shellfish. Immunol. 1995, 5, 301–311. [Google Scholar] [CrossRef]
  112. Tan, X.-Y.; Luo, Z.; Zhang, G.-Y.; Liu, X.-J.; Jiang, M. Effect of dietary cadmium level on the growth, body composition and several hepatic enzymatic activities of juvenile yellow catfish, Pelteobagrus fulvidraco. Aquac. Res. 2010, 41, 1022–1029. [Google Scholar] [CrossRef]
  113. Xu, H.; Zhang, X.; Li, H.; Li, C.; Huo, X.-J.; Hou, L.-P.; Gong, Z. Immune response induced by major environmental pollutants through altering neutrophils in zebrafish larvae. Aquat. Toxicol. 2018, 201, 99–108. [Google Scholar] [CrossRef] [PubMed]
  114. Zhao, L.; Xia, Z.; Wang, F. Zebrafish in the sea of mineral (iron, zinc, and copper) metabolism. Front. Pharmacol. 2014, 5, 33. [Google Scholar] [CrossRef] [Green Version]
  115. Paul, J.S.; Small, B.C. Chronic exposure to environmental cadmium affects growth and survival, cellular stress, and glucose metabolism in juvenile channel catfish (Ictalurus punctatus). Aquat. Toxicol. 2021, 230, 105705. [Google Scholar] [CrossRef]
  116. Altarelli, M.; Ben-Hamouda, N.; Schneider, A.; Berger, M.M. Copper Deficiency: Causes, Manifestations, and Treatment. Nutr. Clin. Pract. Off. Publ. Am. Soc. Parenter. Enter. Nutr. 2019, 34, 504–513. [Google Scholar] [CrossRef]
  117. Bala, S.; Failla, M.L.; Lunney, J.K. Alterations in splenic lymphoid cell subsets and activation antigens in copper-deficient rats. J. Nutr. 1991, 121, 745–753. [Google Scholar] [CrossRef]
  118. Livingstone, C. Zinc: Physiology, deficiency, and parenteral nutrition. Nutr. Clin. Pract. Off. Publ. Am. Soc. Parenter. Enter. Nutr. 2015, 30, 371–382. [Google Scholar] [CrossRef]
  119. Speakman, J.R. Body size, energy metabolism and lifespan. J. Exp. Biol. 2005, 208, 1717–1730. [Google Scholar] [CrossRef] [Green Version]
  120. Genchi, G.; Sinicropi, M.S.; Lauria, G.; Carocci, A.; Catalano, A. The Effects of Cadmium Toxicity. Int. J. Environ. Res. Public Health 2020, 17, 3782. [Google Scholar] [CrossRef]
  121. Verbost, P.M.; Flik, G.; Lock, R.A.; Wendelaar Bonga, S.E. Cadmium inhibits plasma membrane calcium transport. J. Membr. Biol. 1988, 102, 97–104. [Google Scholar] [CrossRef]
  122. Peles, J.D.; Pistole, D.H.; Moffe, M. Influence of cadmium concentration and length of exposure on metabolic rate and gill Na+/K+ ATPase activity of golden shiners (Notemigonus crysoleucas). Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2012, 156, 24–28. [Google Scholar] [CrossRef] [PubMed]
  123. Sokolova, I.M.; Evans, S.; Hughes, F.M. Cadmium-induced apoptosis in oyster hemocytes involves disturbance of cellular energy balance but no mitochondrial permeability transition. J. Exp. Biol. 2004, 207, 3369–3380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Kurochkin, I.O.; Etzkorn, M.; Buchwalter, D.; Leamy, L.; Sokolova, I.M. Top-down control analysis of the cadmium effects on molluscan mitochondria and the mechanisms of cadmium-induced mitochondrial dysfunction. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2011, 300, R21–R31. [Google Scholar] [CrossRef] [Green Version]
  125. Bao, Y.; Liu, X.; Zhang, W.; Cao, J.; Li, W.; Li, C.; Lin, Z. Identification of a regulation network in response to cadmium toxicity using blood clam Tegillarca granosa as model. Sci. Rep. 2016, 6, 35704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Green, A.J.; Planchart, A. The neurological toxicity of heavy metals: A fish perspective. Comp. Biochem. Physiol. Toxicol. Pharmacol. CBP 2018, 208, 12–19. [Google Scholar] [CrossRef]
  127. Lee, D.-C.; Choi, Y.J.; Kim, J.-H. Toxic effects of waterborne cadmium exposure on hematological parameters, oxidative stress, neurotoxicity, and heat shock protein 70 in juvenile olive flounder, Paralichthysolivaceus. Fish Shellfish. Immunol. 2022, 122, 476–483. [Google Scholar] [CrossRef]
  128. Xu, Y.; Zhao, H.; Wang, Z.; Gao, H.; Liu, J.; Li, K.; Song, Z.; Yuan, C.; Lan, X.; Pan, C.; et al. Developmental exposure to environmental levels of cadmium induces neurotoxicity and activates microglia in zebrafish larvae: From the perspectives of neurobehavior and neuroimaging. Chemosphere 2022, 291, 132802. [Google Scholar] [CrossRef] [PubMed]
  129. Tian, J.; Hu, J.; Liu, D.; Yin, J.; Chen, M.; Zhou, L.; Yin, H. Cadmium chloride-induced transgenerational neurotoxicity in zebrafish development. Environ. Toxicol. Pharmacol. 2021, 81, 103545. [Google Scholar] [CrossRef]
  130. Faucher, K.; Fichet, D.; Miramand, P.; Lagardère, J.P. Impact of acute cadmium exposure on the trunk lateral line neuromasts and consequences on the “C-start” response behaviour of the sea bass (Dicentrarchus labrax L.; Teleostei, Moronidae). Aquat. Toxicol. 2006, 76, 278–294. [Google Scholar] [CrossRef] [Green Version]
  131. Huang, T.L.; Obih, P.O.; Jaiswal, R.; Hartley, W.R.; Thiyagarajah, A. Evaluation of liver and brain esterases in the spotted gar fish (Lepisosteus oculatus) as biomarkers of effect in the lower Mississippi River Basin. Bull. Environ. Contam. Toxicol. 1997, 58, 688–695. [Google Scholar] [CrossRef]
  132. Zhang, T.; Yang, M.; Pan, H.; Li, S.; Ren, B.; Ren, Z.; Xing, N.; Qi, L.; Ren, Q.; Xu, S.; et al. Does time difference of the acetylcholinesterase (AChE) inhibition in different tissues exist? A case study of zebra fish (Danio rerio) exposed to cadmium chloride and deltamethrin. Chemosphere 2017, 168, 908–916. [Google Scholar] [CrossRef] [PubMed]
  133. Han, J.; Liu, K.; Wang, R.; Zhang, Y.; Zhou, B. Exposure to cadmium causes inhibition of otolith development and behavioral impairment in zebrafish larvae. Aquat. Toxicol. 2019, 214, 105236. [Google Scholar] [CrossRef] [PubMed]
  134. Volz, S.N.; Hausen, J.; Nachev, M.; Ottermanns, R.; Schiwy, S.; Hollert, H. Short exposure to cadmium disrupts the olfactory system of zebrafish (Danio rerio)-Relating altered gene expression in the olfactory organ to behavioral deficits. Aquat. Toxicol. 2020, 226, 105555. [Google Scholar] [CrossRef]
  135. Nogami, E.M.; Kimura, C.C.; Rodrigues, C.; Malagutti, A.R.; Lenzi, E.; Nozaki, J. Effects of dietary cadmium and its bioconcentration in tilapia Oreochromis niloticus. Ecotoxicol. Environ. Saf. 2000, 45, 291–295. [Google Scholar] [CrossRef] [PubMed]
  136. Kondera, E.; Ługowska, K.; Sarnowski, P. High affinity of cadmium and copper to head kidney of common carp (Cyprinus carpio L.). Fish Physiol. Biochem. 2014, 40, 9–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Sövényi, J.; Szakolczai, J. Studies on the toxic and immunosuppressive effects of cadmium on the common carp. Acta Vet. Hung. 1993, 41, 415–426. [Google Scholar]
  138. Zachoval, R. Clinical meaning of elevated aminotransferases. MMW Fortschr. Der Med. 2021, 163, 51. [Google Scholar] [CrossRef]
  139. Xing, R.Y.; Whitman, W.B. Characterization of amino acid aminotransferases of Methanococcus aeolicus. J. Bacteriol. 1992, 174, 541–548. [Google Scholar] [CrossRef] [Green Version]
  140. Vaglio, A.; Landriscina, C. Changes in liver enzyme activity in the teleost Sparus aurata in response to cadmium intoxication. Ecotoxicol. Environ. Saf. 1999, 43, 111–116. [Google Scholar] [CrossRef]
  141. Zhang, Y. Cell toxicity mechanism and biomarker. Clin. Transl. Med. 2018, 7, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Souid, G.; Souayed, N.; Yaktiti, F.; Maaroufi, K. Effect of acute cadmium exposure on metal accumulation and oxidative stress biomarkers of Sparus aurata. Ecotoxicol. Environ. Saf. 2013, 89, 1–7. [Google Scholar] [CrossRef]
  143. Chen, J.; Chen, D.; Li, J.; Liu, Y.; Gu, X.; Teng, X. Cadmium-induced Oxidative Stress and Immunosuppression Mediated Mitochondrial Apoptosis via JNK-FoxO3a-PUMA pathway in Common Carp (Cyprinus carpio L.) Gills. Aquat. Toxicol. 2021, 233, 105775. [Google Scholar] [CrossRef] [PubMed]
  144. Hu, W.; Zhu, Q.-L.; Zheng, J.-L.; Wen, Z.-Y. Cadmium induced oxidative stress, endoplasmic reticulum (ER) stress and apoptosis with compensative responses towards the up-regulation of ribosome, protein processing in the ER, and protein export pathways in the liver of zebrafish. Aquat. Toxicol. 2022, 242, 106023. [Google Scholar] [CrossRef] [PubMed]
  145. Yan, L.-J.; Allen, D.C. Cadmium-Induced Kidney Injury: Oxidative Damage as a Unifying Mechanism. Biomolecules 2021, 11, 1575. [Google Scholar] [CrossRef]
  146. Wu, Z.; Zheng, R.; Liu, G.; Liu, R.; Wu, S.; Sun, C. Calcium protects bacteria against cadmium stress via reducing nitric oxide production and increasing iron acquisition. Environ. Microbiol. 2021, 23, 3541–3553. [Google Scholar] [CrossRef]
  147. Zhang, D.; Gao, J.; Zhang, K.; Liu, X.; Li, J. Effects of chronic cadmium poisoning on Zn, Cu, Fe, Ca, and metallothionein in liver and kidney of rats. Biol. Trace Elem. Res. 2012, 149, 57–63. [Google Scholar] [CrossRef]
  148. Moulis, J.-M. Cellular mechanisms of cadmium toxicity related to the homeostasis of essential metals. Biometals Int. J. Role Met. Ions Biol. Biochem. Med. 2010, 23, 877–896. [Google Scholar] [CrossRef] [PubMed]
  149. Cheng, C.-H.; Ma, H.-L.; Deng, Y.-Q.; Feng, J.; Jie, Y.-K.; Guo, Z.-X. Oxidative stress, cell cycle arrest, DNA damage and apoptosis in the mud crab (Scylla paramamosain) induced by cadmium exposure. Chemosphere 2021, 263, 128277. [Google Scholar] [CrossRef]
  150. Petitjean, Q.; Jean, S.; Gandar, A.; Côte, J.; Laffaille, P.; Jacquin, L. Stress responses in fish: From molecular to evolutionary processes. Sci. Total Environ. 2019, 684, 371–380. [Google Scholar] [CrossRef] [PubMed]
  151. Sheader, D.L.; Williams, T.D.; Lyons, B.P.; Chipman, J.K. Oxidative stress response of European flounder (Platichthys flesus) to cadmium determined by a custom cDNA microarray. Mar. Environ. Res. 2006, 62, 33–44. [Google Scholar] [CrossRef] [PubMed]
  152. Kim, J.-H.; Rhee, J.-S.; Lee, J.-S.; Dahms, H.-U.; Lee, J.; Han, K.-N.; Lee, J.-S. Effect of cadmium exposure on expression of antioxidant gene transcripts in the river pufferfish, Takifugu obscurus (Tetraodontiformes). Comp. Biochem. Physiol. Part C: Toxicol. Pharmacol. 2010, 152, 473–479. [Google Scholar] [CrossRef] [PubMed]
  153. Jiang, X.; Guan, X.; Yao, L.; Zhang, H.; Jin, X.; Han, Y. Effects of Single and Joint Subacute Exposure of Copper and Cadmium on Heat Shock Proteins in Common Carp (Cyprinus carpio). Biol. Trace Elem. Res. 2016, 169, 374–381. [Google Scholar] [CrossRef] [PubMed]
  154. Giri, S.S.; Sen, S.S.; Jun, J.W.; Sukumaran, V.; Park, S.C. Immunotoxicological effects of cadmium on Labeo rohita, with emphasis on the expression of HSP genes. Fish Shellfish. Immunol. 2016, 54, 164–171. [Google Scholar] [CrossRef]
  155. Gökalp, F.D.; Doğanlar, O.; Doğanlar, Z.B.; Güner, U. The genotoxic effects of mixture of aluminum, arsenic, cadmium, cobalt, and chromium on the gill tissue of adult zebrafish (Danio rerio, Hamilton 1822). Drug Chem. Toxicol. 2022, 45, 1158–1167. [Google Scholar] [CrossRef] [PubMed]
  156. Wang, H.; Feng, Y.; Ming, M.; Song, J.; Chen, Z.; Xiao, Z. Amelioration of Cd-induced bioaccumulation, hematological parameters, and heat shock protein-related genes by Vitamin C on common carp. Comp. Biochem. Physiol. Toxicol. Pharmacol. CBP 2022, 258, 109362. [Google Scholar] [CrossRef] [PubMed]
  157. Cirillo, T.; Cocchieri, R.A.; Fasano, E.; Lucisano, A.; Tafuri, S.; Ferrante, M.C.; Carpenè, E.; Andreani, G.; Isani, G. Cadmium Accumulation and Antioxidant Responses in Sparus aurata Exposed to Waterborne Cadmium. Arch. Environ. Contam. Toxicol. 2012, 62, 118–126. [Google Scholar] [CrossRef] [PubMed]
  158. Pereira, L.S.; Ribas, J.L.C.; Vicari, T.; Silva, S.B.; Stival, J.; Baldan, A.P.; Domingos, F.X.V.; Grassi, M.T.; Cestari, M.M.; Assis, H.C.S.d. Effects of ecologically relevant concentrations of cadmium in a freshwater fish. Ecotoxicol. Environ. Saf. 2016, 130, 29–36. [Google Scholar] [CrossRef]
  159. Huang, Y.-H.; Shih, C.-M.; Huang, C.-J.; Lin, C.-M.; Chou, C.-M.; Tsai, M.-L.; Liu, T.P.; Chiu, J.-F.; Chen, C.-T. Effects of cadmium on structure and enzymatic activity of Cu, Zn-SOD and oxidative status in neural cells. J. Cell. Biochem. 2006, 98, 577–589. [Google Scholar] [CrossRef] [PubMed]
  160. Jihen, E.H.; Imed, M.; Fatima, H.; Abdelhamid, K. Protective effects of selenium (Se) and zinc (Zn) on cadmium (Cd) toxicity in the liver of the rat: Effects on the oxidative stress. Ecotoxicol. Environ. Saf. 2009, 72, 1559–1564. [Google Scholar] [CrossRef] [PubMed]
  161. Pan, L.; Zhang, H. Metallothionein, antioxidant enzymes and DNA strand breaks as biomarkers of Cd exposure in a marine crab, Charybdis japonica. Comp. Biochem. Physiol. Toxicol. Pharmacol. CBP 2006, 144, 67–75. [Google Scholar] [CrossRef] [PubMed]
  162. Dabas, A.; Nagpure, N.S.; Kumar, R.; Kushwaha, B.; Kumar, P.; Lakra, W.S. Assessment of tissue-specific effect of cadmium on antioxidant defense system and lipid peroxidation in freshwater murrel, Channa punctatus. Fish Physiol. Biochem. 2012, 38, 469–482. [Google Scholar] [CrossRef] [PubMed]
  163. Wu, C.; Hong, F.; Liu, C.; Wang, Y.; Wu, K.; Gao, F.; Yang, F. Regulative mechanism of Ce3+ relieves DNA damage caused by Cd2+ in the kidney of silver crucian carp. Biol. Trace Elem. Res. 2006, 113, 231–245. [Google Scholar] [CrossRef]
Figure 1. Schematic diagram of toxic effects of Cd on fish organs.
Figure 1. Schematic diagram of toxic effects of Cd on fish organs.
Toxics 10 00622 g001
Figure 2. Schematic diagram of ROS metabolism and oxidative stress induced by Cd. Under normal physiological conditions, the ROS in the body are mainly converted into less toxic or harmless substances by antioxidant enzymes, such as superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx), while under the condition of Cd exposure, a large amount of ROS are generated and destroy the antioxidant system in cells.
Figure 2. Schematic diagram of ROS metabolism and oxidative stress induced by Cd. Under normal physiological conditions, the ROS in the body are mainly converted into less toxic or harmless substances by antioxidant enzymes, such as superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx), while under the condition of Cd exposure, a large amount of ROS are generated and destroy the antioxidant system in cells.
Toxics 10 00622 g002
Figure 3. Schematic diagram of Cd toxicity on molecule and cell in fish.
Figure 3. Schematic diagram of Cd toxicity on molecule and cell in fish.
Toxics 10 00622 g003
Table 1. Reproductive and physiological endpoints for adult medaka pairs exposed to Cd for 7 weeks. Adapted with permission from [22]. Copyright 2003, Elsevier Inc. All rights reserved.
Table 1. Reproductive and physiological endpoints for adult medaka pairs exposed to Cd for 7 weeks. Adapted with permission from [22]. Copyright 2003, Elsevier Inc. All rights reserved.
Control1 ppb Cd5 ppb Cd10 ppb Cd
Reproductive endpointsTotal eggs99.2 ± 32.7124.0 ± 29.5134.7 ± 29.5113.0 ± 26.1
Total eggs/day8.0 ± 2.09.2 ± 2.010.3 ± 1.48.6 ± 2.0
Fertilized eggs/day7.4 ± 1.88.9 ± 2.010.1 ± 1.38.3 ± 2.2
Hatched eggs/day6.2 ± 1.07.3 ± 0.99.4 ± 0.9*7.6 ± 2.7
Days to first hatch12.4 ± 2.511.6 ± 1.510.5 ± 1.911.8 ± 2.3
Percent survival0.92 ± 0.40.88 ± 0.190.93 ± 0.040.98 ± 0.02
Spawning frequency12.2 ± 1.513.4 ± 0.513.0 ± 1.413.2 ± 0.7
Egg size (μm)1.34 ± 0.021.35 ± 0.021.34 ± 0.031.36 ± 0.02
Physiological endpoints
(females)
Plasma VTG (IOD/positve)0.35 ± 0.190.40 ± 0.200.38 ± 0.100.40 ± 0.15
Hepatic ER (IOD/positive)0.79 ± 0.100.72 ± 0.070.74 ± 0.060.74 ± 0.07
GSI (mm2/mg)0.050 ± 0.0050.052 ± 0.0080.051 ± 0.0110.049 ± 0.003
Plasma E2 (pg/μL)29.2 ± 13.936.6 ± 4.554.3 ± 7.1 *12.4 ± 5.9 *
Plasma T (pg/μL)10.3 ± 6.86.5 ± 2.815.9 ± 3.618.4 ± 25.1
Gonadal E2 (pg)314 ± 9058 ± 63 *56 ± 50 *164 ± 108 *
Gonadal T (pg)561 ± 106261 ± 146 *167 ± 168 *121 ± 62 *
Physiological endpoints (males)Plasma VTG (IOD/positve)0.18 ± 0.020.16 ± 0.010.17 ± 0.010.19 ± 0.01
Hepatic ER (IOD/positive)0.78 ± 0.150.73 ± 0.050.76 ± 0.050.76 ± 0.08
GSI (mm2/mg)0.013 ± 0.0020.011 ± 0.0020.007 ± 0.003 *0.010 ± 0.003
Plasma E2 (pg/μL)39.1 ± 15.362.0 ± 17.165.6 ± 4.433.0 ± 10.4
Plasma T (pg/μL)7.4 ± 4.711.9 ± 2.09.7 ± 3.428.4 ± 36.8
Gonadal E2 (pg)638 ± 219BD *BD *89 ± 154 *
Gonadal T (pg)326 ± 109168 ± 98 *88 ± 57 *115 ± 57 *
Values are mean S.D. VTG, vitellogenin; ER, estrogen receptor; GSI, gonadal somatic index; E2, estradiol; T, testosterone.* Significantly different at p < 0.05 from the corresponding control value for male and female medaka. BD is below detection limit.
Table 2. Summary of toxic effects of cadmium in zebrafish. Reproduced with permission from [25]. Copyright 2021, Korean Society of Toxicology.
Table 2. Summary of toxic effects of cadmium in zebrafish. Reproduced with permission from [25]. Copyright 2021, Korean Society of Toxicology.
Toxicity Ref.
Embryos
LiverHepatic lipid accumulation[26]
NerveNeuroglia alterations[27]
Increased ATPase activity in brain[28]
Reduction of neuronal differentiation and axonogenesis[29]
Interference of neural development[30]
Anti-estrogen in brain[31]
Abnormal somite patterning[32]
Myoskeletal retinaEye hypoplasia and hypopigmentation[33]
Cardiovascular organHeart edema and increased pericardial area[34]
Activation of cell death pathway in olfactory epithelium[35]
Olfactory organDelay in hatching time[34]
OthersTail and axis malformation[34]
Larvae
NerveCircadian rhythms disruption[36]
OthersCell death and structural alterations in olfactory epithelium[37]
Adults
LiverCarcinogenesis[38]
Hepatic lipid accumulation[39]
Oxidative damage[40,41]
NerveOxidative damage[41,42]
MyoskeletalStructural disorganization, disassembly of muscular myofibrils[43]
Reproductive organPair spawning reduction and teratogenicity[44]
Ovary: oxidative damage[41]
RetinaNerve fiber thickening and vacuolating[45]
Table 3. Table of lethal effects of cadmium on different fish species.
Table 3. Table of lethal effects of cadmium on different fish species.
SpeciesConcentrationTimeEffectRef.
Silurus meridionalis6.85 mg/L96 hMedian lethal[58]
Danio rerio25 μg/L9 dMedian lethal[59]
Oryzias javanicus1.0 ppm-Embryo developmental arrest[60]
Lutjanus peru0.05 mM2 hCell viability reduction[61]
Oncorhynchus mykiss8 µg /L96 hEmbryonic mortality rate (97.5%)[62]
Rasbora sumatrana0.1 mg/L96 hMedian lethal[63]
Poeciliareticulata0.17 mg/L96 hMedian lethal[63]
Tautogolabrus adspersus26 μg/mL96 hMedian lethal[64]
Morone saxatilis20 μg/mL96 hMedian lethal[64]
Oreochromis niloticus14.8 mg/L96 hMedian lethal[65]
Cyprinus carpio0.20 ± 0.16 μM96 hMedian lethal[66]
Clarias gariepinus10.85 mg/L96 hMedian lethal[67]
Labeo rohita89.5 mg/L96 hMedian lethal[68]
Trichogaster (Colisa) fasciata49.5 mg/L96 hMedian lethal[69]
Silurus soldatovi2.74 mg/L96 hMedian lethal[70]
Table 4. Minimum, maximum, mean, and standard deviation (SD) of Cd in canned tuna samples and fresh fish samples (mg/kg ww). Adapted with permission from [84]. Copyright 2019, Health Scope.
Table 4. Minimum, maximum, mean, and standard deviation (SD) of Cd in canned tuna samples and fresh fish samples (mg/kg ww). Adapted with permission from [84]. Copyright 2019, Health Scope.
SpeciesCd
MinMaxMean ± SD
Mako shark
(Isurusoxyrinchus)
0.0010.0030.0023 ± 0.0005
Yellowfin tuna
(Thunnusalbacares)
0.0010.0020.0019 ± 0.0001
Soupfin shark
(Galeorhinusgaleus)
0.0010.0020.0018 ± 0.0002
Swordfish
(Xiphias gladius)
0.0010.0030.0022 ± 0.0004
Abbreviation: ww, wet weight.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Liu, Y.; Chen, Q.; Li, Y.; Bi, L.; Jin, L.; Peng, R. Toxic Effects of Cadmium on Fish. Toxics 2022, 10, 622. https://doi.org/10.3390/toxics10100622

AMA Style

Liu Y, Chen Q, Li Y, Bi L, Jin L, Peng R. Toxic Effects of Cadmium on Fish. Toxics. 2022; 10(10):622. https://doi.org/10.3390/toxics10100622

Chicago/Turabian Style

Liu, Yinai, Qianqian Chen, Yaoqi Li, Liuliu Bi, Libo Jin, and Renyi Peng. 2022. "Toxic Effects of Cadmium on Fish" Toxics 10, no. 10: 622. https://doi.org/10.3390/toxics10100622

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

Article Metrics

Back to TopTop