You are on page 1of 11

Toxicology Mechanisms and Methods

ISSN: 1537-6516 (Print) 1537-6524 (Online) Journal homepage: https://www.tandfonline.com/loi/itxm20

The effects of heavy metals on human metabolism

Zhushan Fu & Shuhua Xi

To cite this article: Zhushan Fu & Shuhua Xi (2020) The effects of heavy metals
on human metabolism, Toxicology Mechanisms and Methods, 30:3, 167-176, DOI:
10.1080/15376516.2019.1701594

To link to this article: https://doi.org/10.1080/15376516.2019.1701594

Published online: 17 Dec 2019.

Submit your article to this journal

Article views: 2363

View related articles

View Crossmark data

Citing articles: 209 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=itxm20
TOXICOLOGY MECHANISMS AND METHODS
2020, VOL. 30, NO. 3, 167–176
https://doi.org/10.1080/15376516.2019.1701594

REVIEW ARTICLE

The effects of heavy metals on human metabolism


Zhushan Fu and Shuhua Xi
Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, China

ABSTRACT ARTICLE HISTORY


As technology continues to advance, heavy metals in drinking water have exceeded recommended Received 2 July 2019
limits from regulators around the world. The main source of human exposure to heavy metals is from Revised 23 November 2019
contaminated drinking water. The effects of drinking water contaminated with heavy metals, such as Accepted 25 November 2019
arsenic, lead, nickel, cadmium and mercury, have gradually caught the attention of the relevant
KEYWORDS
departments and personnel. It is well known that occupational exposure to heavy metals occurs as a Heavy metals; arsenic; lead;
result of using these metals in a variety of industrial processes in and/or a variety of materials, includ- nickel; cadmium;
ing color pigments and alloys. A series of adverse effects on human metabolism has resulted from mercury; metabolism
exposure to heavy metal-contaminated drinking water, which has been recorded from around the
world. The general mechanism of heavy metal toxicity is through the production of reactive oxygen
species, the appearance of oxidative damage, and subsequent adverse effects on health. Therefore,
water contaminated with heavy metals causes high morbidity and mortality worldwide. In order to
address concern regarding the health effects of different heavy metals, this paper reviews its sources,
distribution and effects of heavy metal on human metabolism.

HIGHLIGHTS
1. The accumulation of heavy metals such as lead, arsenic, mercury, cadmium and nickel will destroy
the main metabolic process of human body.
2. Redox reactions in biological systems are caused by carcinogenic metal ions such as nickel and
arsenic. The free radicals produced by these reactions cause oxidative damage to proteins
and DNA.
3. The accumulation of heavy metals eventually produces reactive oxygen species that can cause
oxidative stress, which may lead to the production of various diseases.

Abbreviations: ALAD: d-aminolevulinic acid dehydratase; AS: arsenic; Cd: Cadmium; CNS: central ner-
vous system; DMA: dimethylric acid; ETC: electron transport chain; G6PD: glucose-6-phosphate
dehydrogenase; HIF- 1a: hypoxia-inducible factor-1 alpha; LDH: lactate dehydrogenase; Pb: Lead; MDA:
malondialdehyde; MT: metal sulfur protein; Hg: mercury; mTOR: mammal rapamycin target; MAPK:
mitogen-activated protein kinase; Ni: nickel; OXPHOS: oxidative phosphorylation; ROS: reactive oxy-
gen species

1. Introduction zinc, copper and iron, are required by the human body (Lane
Heavy metal naturally exists in earth’s crust (Al-Samman and Morel 2000). However, if they were ingested at higher
2015) but can cause serious problems for all living things concentrations, they would have toxic effects on the human
and are toxic pollutants for the natural environment (Fu and body (Asubiojo et al. 1997). Other heavy metals, such as,
Wang 2011). Due to exponential growth in the use of various mercury and lead, have no healthy effects but are detrimen-
products and industries, human exposure to heavy metals tal to human health, when they accumulate in the human
has dramatically increased over the past 50 years (Bedrin body (Asubiojo et al. 1997).
et al. 2003). The accumulation of heavy metals in the body has been
Metal waste pollutes the surface of water and soil, result- demonstrated to have an adverse effect on human health.
ing in harmful impacts on human health. In order to ensure The main heavy metals that that is possible to cause adverse
that humans are protected from the adverse effects of heavy effects include arsenic, lead, aluminum, iron, mercury and
metals, especially heavy metals in drinking water, inter- cadmium. These metals can enter the body through various
national organizations have developed standards for the use ways, such as skin or inhalation routes, or intake of heavy
of each individual metal (Lane and Morel 2000). Heavy met- metals through contaminated drinking water and food.
als can interfere with the metabolic function of the body Heavy metals can also react with certain compounds in the
through various means. Some metals, such as manganese, body, such as oxygen and chloride, exerting their own toxic

CONTACT Shuhua Xi shxi@cmu.edu.cn Department of Environmental and Occupational Health, School of Public Health, China Medical University,
Shenyang, 110122, China
ß 2019 Informa UK Limited, trading as Taylor & Francis Group
168 Z. FU AND S. XI

effects (Rusyniak et al. 2010). Persistent exposure to heavy Therefore, this paper gives an overview of the current sta-
metals can lead to an imbalance in the body when heavy tus of heavy metals pollution, expounds the effects of accu-
metal accumulate in the body and are used as substitutes mulation of heavy metals in the body on metabolic enzymes
for essential elements. Examples of heavy metals replacing and metabolic processes, and explains possible mechanisms
essential elements of the human body include calcium of action.
replaced by lead, zinc by cadmium, and most trace elements
by aluminum (Genestra 2007). Lead metal ions can replace
other divalllions such as Ca2þ, Mg2þ, Fe2þ and unital cations
2. Heavy metals and metabolism
such as Naþ to ultimately disrupt the biological metabolism Heavy metals naturally exist in the earth’s crust (Al-Samman
of cells. The ion mechanism of lead toxicity causes significant 2015), and accumulate in different sections of plants, which
changes in various biological processes. Even at picomolar can in turn affect human health. Although zinc, manganese,
concentrations, lead can replace calcium and affect protein cobalt, copper and iron are essential for the human body,
kinase C, which regulates nerve excitability and memory stor- they can be toxic to the human body when their concentra-
age (Flora et al. 2008). Cadmium and zinc with the same oxi- tion becomes too high (Asubiojo et al. 1997).
dation state, so cadmium can replace zinc present in metal Heavy metals can interfere with the metabolic functions
sulfur protein, causing zinc metabolism disorders that inhibit of the human body in a various of ways. In addition, they
it as a free radical scavenger in cells (Flora et al. 2008). In may accumulate in important organs of the human body,
addition, the accumulation of heavy metals destroy the main such as the brain, heart, liver, and kidneys, ruining normal
metabolic process of the human body, and at the same time biological functions. However, the biggest issue is that there
lead to an imbalance of antioxidation. Similarly, the function can be no environment without heavy metals at all. Heavy
of essential enzymes and the activity of numerous hormones metals can enter the human body through various access,
are also affected (Dhir et al. 2011). Researchers have identi- such as the consumption of contaminated food and drinking
fied various carcinogenic pathways caused by heavy metal water, and through the air, which can lead to a series of
exposure. Some studies have shown that the mutagenicity effects on the human body (Al-Samman 2015). The accumu-
and carcinogenicity of heavy metals are related to their abil- lation of heavy metals in the human body has a detrimental
ity to cause oxidative stress. In this aspect, researchers have effect on human health. The main heavy metals that cause
found that redox reactions in biological systems are caused this effect include cadmium, arsenic, aluminum, mercury and
by carcinogenic metal ions, such as chromium, nickel, cobalt, iron. Due to increased industrialization of society and
and arsenic. The free radicals that are produced by these increased exposure to heavy metals worldwide, the harmful
reactions cause oxidative damage to proteins and DNA. effects on human health associated with exposure to heavy
Besides direct DNA damage, products produced by redox metals have increased over the past few years (Rusyniak
reactions have two other functions that lead to carcinogen- et al. 2010).
esis in humans. One is the activation of redox-sensitive tran- Researchers have found a variety of carcinogenic path-
scription factors, while the other function involves its role as ways caused by heavy metal exposure (Figure 1). Numerous
a mitogenic signal. Similarly, the carcinogenicity of heavy studies have shown that the carcinogenicity and mutagenic-
metal pathways may interfere with the process of DNA repair ity of heavy metals are associated with the induction of oxi-
(Genestra 2007). dative stress. In this regard, some studies have shown that

Figure 1. Toxic effect mechanism of heavy metals.


TOXICOLOGY MECHANISMS AND METHODS 169

redox reactions in biological systems are carried out by car-


cinogenic metal ions, such as nickel, chromium, cobalt and
arsenic. Therefore, the free radicals produced by these reac-
tions cause oxidative damage to proteins and DNA. Despite
the direct damage of DNA, species produced by these redox
reactions also have two other important functions that cause
cancer in humans. One is the activation of redox-sensitive
transcription factors, while the other functions involves its
role as a mitotic signal (Genestra 2007). Similarly, another
pathway of heavy metal carcinogenesis is thought to be a
process that interferes with DNA repair (Rehman et al. 2018).
The heavy metals stored destroy the body’s main meta-
bolic processes and cause an imbalance of antioxidants.
Similarly, the activity of various hormones and the function
of indispensable enzymes are also affected (Jayawardena
et al. 2017). With changes in carbohydrate, protein and lipid Figure 2. Lead affects the main mechanisms of metabolism.
metabolism, the body’s susceptibility to infections is apt to
increase. Ultimately, all of these mechanisms alter the syn- as O2þ, H2O2 and hydroperoxides, Studies have shown that
thesis and application of neurotransmitters in the body, lead can raise reactive oxygen levels and Ca2þ within cells,
which in turn modify a range of functions of the central ner- which in turn leads to a decrease in mitochondrial potential
vous system (CNS) (Qayyum et al. 2012). Therefore, regard- and apoptosis through the release of cytochrome c (Moreira
less of the molecular pathways that are affected by heavy et al. 2001).
metals, they eventually produce reactive oxygen species that While the second mechanism is achieved by the con-
can cause oxidative stress, a process that can lead to differ- sumption of antioxidants of cells (Ercal et al. 2001). Previous
ent types of cancers, impaired kidney function, neurological studies indicate that lead can inhibit mitochondrial oxidative
diseases and other endocrine diseases (Qayyum et al. 2012). phosphorylation, thereby changing the d-aminolevulinic acid
dehydratase (ALAD) of cells. Lead also causes oxidative stress
and destroys the oxidant/antioxidant balance of cells
2.1. The effects of lead on metabolism (Kasperczyk et al. 2012). Oxidative stress caused by lead is
Lead(Pb) is a common environmental and industrial contam- the main mechanism of its toxicity. In addition to changes in
inant (Qayyum et al. 2012; Rahman and Sultana 2006) which antioxidant enzymes, it is directly involved in free radical-
is found in the form of elements, both inorganic and organic, mediated reactions (Knowles and Donaldson 1990), enzyme
and in trace amounts in food, water and soil (Patocka and activity and changes in the concentration of antioxidant mol-
Cerny 2003). Most of the environmental pollution caused by ecules, such as glutathione (Mudipalli 2007). As an antioxi-
lead is the result of the use of lead paint on the outer side dant present in cells, Glutathione protects them from free
of buildings (Godwin 2001). At the same time, more than radicals. Under normal conditions, the reduced form (GSH) of
900 occupations, including refining, mining, smelting, weld- Glutathione accounts for 90% of the total Glutathione and its
ing, and painting, making stained glass and ceramic glass, oxidized accounts for 10%. However, in the case of oxidative
are exposed to lead, resulting in lead poisoning (Patocka and stress, GSSG concentration exceeds GSH concentration
Cerny 2003). Inorganic lead compounds and elemental lead, (Samadi et al. 2001).
such as lead dust, can be absorbed through the digestive Permpongpaiboon et al. (2011) and Kasperczyk et al.
system and the respiratory system (Patocka and Cerny 2003). (2005), found that significantly elevated malondialdehyde
The presence of lead determines its toxicity in the environ- (MDA) levels were observed in workers exposed to lead
ment. Organic lead compounds based on covalent bonds (other signs of lipid oxidative damage were also observed)
have different toxicological effects from that of inorganic (Kasperczyk et al. 2005; Permpongpaiboon et al. 2011). The
lead salts (such as lead acetate). Organic lead compounds effects of lead acetate on human red blood cell metabolism
can be absorbed through the skin and then enter the brain, in vitro, and the results are consistent with the results of
giving rise to a toxin in the central nervous system (Marshall Kasperczyk et al. (2005), Xia et al. (2018). In the study, the
et al. 2007). Subsequently, lead is absorbed into the plasma, function of the pentose phosphate pathway was found to
after the extracellular fluid are in a state of and lead accumu- have been reversed as a result of decreased Glucose-6-phos-
lates in soft tissues and hard tissues. Finally, lead is mainly phate dehydrogenase (G6PD) activity (Kasperczyk et al.
excreted through the urinary system and the digestive sys- 2005). However, Garier-Orchan et al. reported conflict-
tem (Patocka and Cerny 2003). ing outcome.
The main mechanisms of lead affecting metabolism are Paglia et al. (1975) investigated the activity of enzymes in
shown in Figure 2. Due to the release of free radicals, the human red blood cells exposed to lead in vitro and obtained
damage caused by lead to living systems involve two separ- the same results as those of Antonowicz et al. (1990), (Paglia
ate mechanisms (Ercal et al. 2001). The first mechanism is et al. 1975; Rosenberg et al. 2002), who studied lead expos-
the direct production of reactive oxygen species (ROS), such ure in workers. The results of this study found that the lead
170 Z. FU AND S. XI

exposure group had significantly higher than the average (Korrick 2004). More than 140 million people worldwide con-
PBB levels of lead exposed workers, as reported in the study sume arsenic-contaminated drinking water that exceeds the
by Kasperczyk et al. (2005). The difference in these results World Health Organization’s 10 ppb limit (Lenczewski 2009),
may be due to lead exposure time, lead concentration and with groundwater in Bangladesh and West Bengal having
intracellular oxidative stress intensity. the highest levels of arsenic (Rahman 2002). In this part of
Wang et al. (2010) studied the damage of cadmium in the world, 35 million to 77 million people are exposed to
renal cortex by combined exposure of lead and cadmium. arsenic, concentrations of which in drinking water that range
The conclusion that low concentrations of lead acetate can from less than 10 ppb to more than 4 ppm, resulting in the
improve the respiratory rate is consistent with that of death of about 1/5 people due to exposure to arsenic.
Kasperczyk et al. (2005). This is the indirect result of the malic Therefore, arsenic pollution is of global concern (Rahman
acid-aspartic acid shuttle in cells, which increases pyruvate 2002; Argos et al. 2010).
oxidative decarboxylation and Nicotinamide adenine Arsenic metabolites exist in both organic and inorganic
dinucleotide (NADH) transportation from the cytoplasm to forms, and both types can be presented in trivalent or penta-
mitochondria, which may lead to a decrease in Lactate priced oxidation states. Sodium arsenate or arsenic dioxide
dehydrogenase (LDH) activity. However, there was no signifi- in triaval form reacts with many biosites with sulfur groups,
cant change in FBA activity, indicating that lead has a limited and as a result, arsenic compounds inhibit many enzymes
effect on the glycolysis pathway. The in vitro study by Yun (Challenger 1947; Sharma et al. 2015). Methyl oxide and
and Hoyer (2000) on rat brains indicate that lead can inhibit glutathione concosering are thought to be the main path-
the glycolysis enzyme and mitochondrial respiration (Yun ways due to metabolism (Hayakawa et al. 2005). The toxic
and Hoyer 2000). It interferes with many body functions effect mechanism of arsenic is shown in Figure 3. Urine
inside the body, affecting most organs. The organic form of removes most of the arsenic ingested in inorganic form, and
ingestion is absorbed by the liver and is therefore metabo- the first step after methylation can bioconvert a small
lized, compared to inorganic lead that is not metabolized by amount of as to convert inorganic AS (III) methylation into
the liver (Banfalvi 2011). dimethylric acid (DMA).These processes usually detoxify
organic arsenic, but some organic arsenic metabolites may
contribute to arsenic toxicity (Bernstam and Nriagu 2000).
2.2. The effects of arsenic on metabolism
Apart from affecting the skin (Yu et al. 2006) and leading
Human exposure to arsenic (AS) may be caused by exposure to bladder cancer (Marshall et al. 2007), arsenic also affects
through drinking water contaminated by industrial waste or the liver (Liaw et al. 2008), kidneys (Yuan et al. 2010) and
agricultural chemical waste (Hughes et al. 2011). Another lungs (Smith et al. 2006). Studies by Ditzel et al. (2016) and
source of water pollution may be smoke or arsenic fog. Shi et al. (2014) found that arsenic is associated with cancer
Poisoning may be caused by eating pesticide-contaminated (Shi et al. 2014; Ditzel et al. 2016), metabolic syndrome and
foods, arsenic-rich foods, or foods that grow in fertile soils other metabolism-related diseases and symptoms (Arteel
(Antonowicz et al. 1990). Incredibly, drinking milk is also one et al. 2008; Ditzel et al. 2016). However, even after decades
of the sources of arsenic poisoning, since adulterated milk of research, the exact mechanism by which arsenic causes
contains a mixture of arsenic-containing water. In addition to disease is still unclear. The production of reactive oxygen
these sources, AS access to the human body also includes species (ROS) (Shi et al. 2004), altered DNA methylation
inhalation and skin contact. Additionally, another major (Zhao et al. 1997), DNA damage (Wang et al. 2001) and
source of arsenic poisoning is the use of acids and crude enzyme inhibition are considered to play a role (Kitchin and
metals. Inorganic arsenic has acute and chronic toxicity Wallace 2008). Arsenite can enter the mitochondria through

Figure 3. Mechanism of arsenic poisoning.


TOXICOLOGY MECHANISMS AND METHODS 171

hydroglycerin, where it binds to and suppresses many turn leads to renal cell damage and death (Ohta and Cherian
enzymes involved in energy production, including pyruvate, 1991; Jomova and Valko 2011). Cadmium produces ROS and
succinic acid, isocitrate and a-ketoglutarate dehydrogenase induces oxidative stress by replacing copper and iron, caus-
(Braeckman et al. 2009; Higashi et al. 1965). The electron ing fenton reactions, reducing the number of copper and
transport chain (ETC) complexes II and IV indicate that mito- iron ions participating in oxidative stress response or increas-
chondrial dysfunction caused by arsenic may be present in ing the number of uncombined free radicals (Watjen and
arsenic-related pathological processes (Naranmandura Beyersmann 2004). Recent studies have shown that oxidative
et al. 2011). stress is a potential cause of neuronal toxicity, at the same
The results of Zhao et al. (2013) show that chronic, low time, oxidative stress is also considered to be one of the
dose (75 ppb) arsenite exposure induces the Warburg effect related mechanisms of genotoxicity. Oxidative stress caused
(Zhao et al. 2013), which is defined as the conversion from by cadmium exposure can damage (Valko et al. 2006) and
mitochondrial oxidative phosphorylation (OXPHOS) to aer- destroy the normal function of organelles and produce DNA
obic glycolysis, which is also a common sign of cancer devel- mutations that lead to changes in gene expression and
opment. Importantly, the observed glycolytic transformation finally, inducing apoptosis (Ognjanovic et al. 2010). In add-
is accompanied by a decrease in Krebs circulating activity, an ition, cadmium can impair the repair function of DNA dam-
increase in aneuploidy and loss of anchorage-dependent age, which can lead to the accumulation of DNA damage,
growth, all of which are dependent on the stability of the inducing genetic mutations and thus cancer (Giaginis et al.
transcription factor, hypoxia-inducible factor-1 alpha (HIF- 1a) 2006; Zhou et al. 2012). Some DNA damage repair enzymes
(Zhao et al. 2014). These results reveal that the destruction have zinc finger structure, cadmium can replace the activity
of mitochondrial energy metabolism is one of the important of zinc inhibitory enzymes on this enzyme, thereby inhibiting
mechanisms of arsenic carcinogenesis. Robey et al. (2015) DNA damage repair (Zhou et al. 2012).
reviewed the possibility of carcinogenesis through environ- Reactive oxygen plays a leading role in cadmium cancer-
mental exposure by changing mitochondrial metabolism causing process because reactive oxygen causes apoptosis,
(Robey et al. 2015), but there are few examples for these abnormal gene expression and inhibits DNA damage repair.
mechanisms. High arsenic levels were observed to inhibit the Although studies generally indicate that reactive oxygen lev-
activity of hexalysinkinse (Zhang et al. 2015), Furthermore, els in normal cells are lower than in tumor cells, there are
arsenite exposure result in severe mitochondrial dysfunction, also studies showing that reactive oxygen levels in normal
reduced ATP levels (Luz et al. 2016). cells are higher than in tumor cells (Zhou et al. 2012).
Studies have shown that two neuronal cell types: PC 12 and
SHSY5Y cells increase ROS production after cadmium expos-
2.3. The effects of cadmium on metabolism ure (Chen et al. 2008). The two signaling pathways produced
Cadmium (Cd) is a type of heavy metal that exists naturally by ROS leading to apoptosis are the mTOR (mammal rapa-
in ores and is usually used as a stabilizer for different prod- mycin target) pathway and the mitogen-activated protein
ucts, such as color pigments, alloys and other related prod- kinases(MAPKs)ignaling pathway (Chen et al. 2008, 2011).
ucts (Jarup et al. 1998). Occupational exposure to cadmium Studies have also shown that cadmium alters the activity
includes, smoke inhalation, the nickel-cadmium battery of many carbohydrate-related metabolic enzymes. Cadmium
industry, electroplating and paint pigments (Pang et al. has the potential to limit the glycozy process in the liver and
2016). Oral exposure to cadmium is the consumption of con- muscles by reducing the activity of fructose kinase phos-
taminated food and/or water. This pathway is the most phate (Almeida et al. 2001). Cadmium also increases the
important pathway for nonsmokers and those with no occu- activity of several other enzymes involved in amino acid
pational exposure to cadmium (Pang et al. 2016). decomposition metabolism, such as amino acid oxidase, glu-
Studies have found that cadmium exposure has an inhibi- tamate dehydrogenase, etc (Cicik and Engin 2005). Similar to
tory effect on the main antioxidant enzymes: glutathione arsenic, Maria et al. found that cadmium also inhibits hexaly-
reductase and superoxide dismutase (Stohs et al. 2001; Valko sinkinase (Ramirez-Bajo et al. 2014), the results of Li et al.’s
et al. 2006). Increased lipid peroxidation has been observed research show that cadmium promotes glycolysis at the
in some brain regions after cadmium exposure, such as the beginning of the experiment. Long-term exposure to cad-
cerebellum and cortex (Mendez-Armenta et al. 2003). mium, but inhibited glycolysis. Therefore, it is concluded that
Through various experimental studies, it has been found that by increasing cadmium concentration, glycolysis can be
cadmium has a disturbing effect on the levels of enzymes inhibited earlier (Zhou et al. 2012) (Figure 4).
that maintain the redox reaction. Due to this disorder, the
occurrence of lipid peroxidation eventually induces damage
2.4. The effects of nickel on metabolism
to the brain microvasculature (Shukla et al. 1996). Cadmium
binds mainly to metal sulfur protein (MT) in the body to Nickel(Ni) is the 24th most abundant element on earth, and
form Cd-MT, which is reabsorbed in the small tube of the 3% of the earth’s crust is composed of nickel. Nickel is widely
kidney, degrades in the lysozyme of the near-curved tube, distributed in drinking water around the world (Coogan et al.
isolates and releases free cadmium, thus interfering with the 1989). Nickel is considered to be important for various func-
metabolite of the chowtoral epithelial cell mitochondrial tions of the human body. However, increased exposure may
energy metabolism and oxidation phosphorylation, which in cause human poisoning (Diagomanolin et al. 2004; Haber
172 Z. FU AND S. XI

production of reactive oxygen species caused by Redox


homeostasis disruption may lead to cell death and decrease
the number of living cells (Oukarroum et al. 2017). Studies
by Martınez-Ruiz and Martınez-Jer Nimo have shown that
nickel induces the production of reactive oxygen species and
increases the activity of antioxidant enzymes through the
generation of free radicals by Fenton reaction (Miazek et al.
2015). Excess nickel can also induce the production of free
radicals and reactive oxygen species by direct electron trans-
fer, which inactivates enzymes in the antioxidant defense
systems of other photosynthetic organisms, such as
Glutathione reductase, mitochondrial (Nagajyoti et al. 2010).

2.5. The effects of mercury on metabolism


Mercury (Hg) is a common heavy metal pollutant found in
the natural environment, mainly in the following forms:
element, inorganic and organic (Li et al. 2016). These three
forms have different toxicity, among which the least toxic
form is the element (Clarkson 1997). Although mercury is
Figure 4. The role of cadmium in glycolysis. found at low levels in the environment, its toxicity, persist-
ence and bioaccumulation may still pose a health threat to
et al. 2000; Ognjanovic et al. 2010). The two main pathways humans and other organisms (Eagles-Smith et al. 2016).
of oral nickel exposure are through water and food contami- Elemental mercury is oxidized in the air into an inorganic
nated with nickel-containing compounds (Haber et al. 2000). form. In its vapor form, metallic mercury is usually absorbed
Ni has many industrial and commercial applications, render- through the respiratory tract and difficult to absorb through
ing it accessible to these types of workers. Nickel emissions
the gastrointestinal tract. Because of its soluble properties,
are naturally found from forest fires, volcanic emissions and
elemental mercury spreads highly through the cell mem-
wind dust. Other sources of nickel emitted into the air
brane and blood brain, reaching the target organ. Once
include coal combustion and waste incineration, while
enter in the bloodstream, elemental mercury is easily oxi-
another source of nickel exposure for humans is tobacco
dized into inorganic Hg and Hg in red blood cells and tissues
smoke (Yang and Ren 2010).
in the presence of hydrogen peroxide and peroxidase.
In the human body, nickel binds to mercaptan to form a
Compared with the organic form that mainly affects the cen-
nickel-mercaptan complex. Then, the complex reacts with
tral nervous system, inorganic Mercury follows non-regular
molecular oxygen to produce free radicals, which eventually
distributed, accumulating mainly in the kidneys; causing
leads to nickel toxicity (Das et al. 2006). Experimental studies
acute renal failure (Al-Saleh et al. 2012). Mercury is a major
by Das et al. have shown that nickel binds to thiol proteins,
leading to a decrease in glutathione levels, which in turn cause of autoimmune diseases and antinuclear antibodies
leads to nickel toxicity in the body (Valko et al. 2005). produced by individuals exposed to inorganic mercury
Researchers have found that physicochemical properties in (Crowe et al. 2017). Hg is also a causative agent of
the human body may change owing to nickel exposure, as a Alzheimer’s disease and Parkinson’s disease (Chin-Chan et al.
result of reduced calcium excretion in urine. In addition, 2015). Therefore, all forms of mercury have strong toxic
nitrogen retention is also reduced due to nickel exposure effects on the central nervous system and digestive system
(Kumar et al. 2019). The pathway of oxidative phosphoryl- (Crowe et al. 2017).
ation is impaired by reduced levels of nicotinamide resulting Some studies have shown that exposure to mercury
from chronic nickel exposure. M’Bemba-Meka et al. studied increases the production of free radicals, ROS and superoxide
cultured human red blood cells and found that these cells anions due to the Fenton reaction (Ehara et al. 2001; Magos
were under oxidative stress because of nickel exposure 1997; Miller et al. 1991). Wildemann et al. (2016) argue that
(2006). There is evidence that nickel plays a role in inducing the pivotal roles of mercury-induced toxicity are inhibition of
blood toxicity. Salnikow et al. conducted a study in which antioxidant defense systems, changes in the oxidant-antioxi-
nickel exposure in rats and mice was induced through inhal- dant balance, and the increase of ROS (Wildemann et al.
ation. Blood red blood cell count, hemoglobin concentration 2016). Methylmercury is a neurotoxic compound that causes
and other indicators increased. It was also found that the lipid peroxidation, mitochondrial damage, microtubule dam-
changes in all these parameters were due an increase in age, and the accumulation of neurotoxic molecules (Patrick
erythropoietin synthesis caused by nickel-exposure induced 2002). Methylmercury significantly increases the level of free
hypoxia (Denkhaus and Salnikow 2002). calcium ions in nerve cells in the cerebral cortex, disrupting
Nickel is a necessary auxiliary factor for SOD to regulate calcium stability and, in turn, affecting the physiological
cell ROS level and protect nitrogenase. The excess processes of cells, inducing oxidative stress by interfering
TOXICOLOGY MECHANISMS AND METHODS 173

with mitochondrial breathing chains, affecting DNA repair fillings should always be used. Removal of heavy metals
and leading to apoptosis (Ho et al. 2013). from water can be done through ion exchange (Tariq et al.
HgCl2 is one of the most important forms of mercury that 2008), adsorption, membrane filtration and electrodialysis
is widely used in various acess. HgCl2 compounds have been (Chen 2004). Therefore, we need to establish a defense
proven to be organotoxic and carcinogenic (Risher et al. mechanism to prevent exposure to heavy metals, which
1999). Nava et al. (2000) have both shown that HgCl2 can increases the content of reactive oxygen species (Ros) in the
destroy the function of mitochondrial intima, while Mahboob human body and eventually induces a series of oxidative
et al. (2001) found that HgCl2 can alter membrane perme- damage processes. In future, more research is needed to
ability. In cells, Hg2þ ions interact with antioxidant proteins, ensure that human health is protected and is safe from the
DNA repair enzymes and protein active sites involved in effects and hazards of heavy metals.
intracellular homeostasis (Crespo-Lopez et al. 2009). Studies
have shown that mercury ions have a high affinity for the
Author Contributions
protein in cells, leading to nonspecific inhibition and damage
of cell enzyme systrms (Clarkson and Magos 2006). However, Zhushan Fu conceived and drafted this manuscript. Shuhua Xi revised
overexpression of metal sulfur protein after mercury expos- and approved the final manuscript.
ure may acts as a protective role in the body (Chan 2011).
Ekinci et al. (2007) found that exposure to heavy metals Disclosure statement
interfere with the normal activity of metabolic enzymes. For
example, HgCl2 can inhibit only two metabolic pathways, the The authors declare that they have no known competing financial inter-
ests or personal relationships that could have appeared to influence the
glycolysis pathway and hexose-phosphate lysis pathway, in work reported in this paper.
red blood cells by downregulating the activity of labeling
enzymes. The enzyme hexokinase in the first step of glycoly-
sis, and the activity of the last enzyme of the glycolysis pro- Funding
cess, pyruvate kinase, were found to be significantly reduced. This work was supported by the National Natural Science Foundation of
Hexokinase and pyruvate kinase are known to be inactivated China (NSFC) [81972982 and 81673207].
by oxidative stress (Anastasiou et al. 2011). Since the glyco-
lytic process is the only source of ATP in red blood cells,
which lack mitochondria, these changes can lead to a drop
in energy levels. Schutt et al. (2012) found that the depletion References
of ATP makes cells more vulnerable to oxidative damage. Al-Saleh I, Al-Sedairi AA, Elkhatib R. 2012. Effect of mercury (Hg) dental
G6PD is the first enzyme to be identified to shunt hexose amalgam fillings on renal and oxidative stress biomarkers in children.
monophosphate, and the product of Hexokinase is glucose Sci Total Environ. 431:188–196.
Al-Samman T. 2015. Effect of heavy metal impurities in secondary Mg
6-phosphate. Hexokinase and substrate G6PD activity were
alloys on the microstructure and mechanical properties during
found to have decreased as a result of exposure. Studies by deformation. Mater Des. 65:983–988.
Rabbani and Thornalley (2012) show that glyoxal-i inhibits Almeida JA, Novelli ELB, Silva MD, Alves R. 2001. Environmental cad-
the formation of glyoxal-derived deleterious products, and mium exposure and metabolic responses of the Nile tilapia,
that the enhancement of its activity provides an enzyme Oreochromis niloticus. Environ Pollut. 114(2):169–175.
Anastasiou D, Poulogiannis G, Asara JM, Boxer MB, Jiang JK, Shen M,
defense mechanism for methoxyl-induced protein Bellinger G, Sasaki AT, Locasale JW, Auld DS, et al. 2011. Inhibition of
modifications. pyruvate kinase M2 by reactive oxygen species contributes to cellular
antioxidant responses. Science. 334(6060):1278–1283.
Antonowicz J, Andrzejak R, Smolik R. 1990. Influence of heavy metal mix-
3. Conclusions tures on erythrocyte metabolism. Int Arch Occup Environ Heath.
62(3):195–198.
This article discusses the various hazards associated with Argos M, Kalra T, Rathouz PJ, Chen Y, Pierce B, Parvez F, Islam T, Ahmed
heavy metal exposure on human metabolism. However, it is A, Rakibuz-Zaman M, Hasan R, et al. 2010. Arsenic exposure from
important to understand the methods by which the public drinking water, and all-cause and chronic-disease mortalities in
Bangladesh (HEALS): a prospective cohort study. Lancet. 376(9737):
come into contact with heavy metals, in order to avoid fur-
252–258.
ther contact. Based on previous research results, for many Arteel GE, Guo L, Schlierf T, Beier JI, Kaiser JP, Chen TS, Liu M, Conklin
people regulatory limits have been exceeded and they are DJ, Miller HL, von Montfort C, et al. 2008. Subhepatotoxic exposure to
at the threshold of developing major organ toxicity. arsenic enhances lipopolysaccharide-induced liver injury in mice.
Governments must therefore take important steps to protect Toxicol Appl Pharmacol. 226(2):128–139.
Asubiojo OI, Nkono NA, Ogunsua AO, Oluwole AF, Ward NI, Akanle OA,
the population from this potential hazard to people’s health
Spyrou NM. 1997. Trace elements in drinking and groundwater sam-
by advising people to avoid fish species that contain high ples in southern Nigeria. Sci Total Environ. 208(1–2):1–8.
levels of arsenic and mercury(Salnikow and Zhitkovich 2008). Banfalvi G. 2011. Cellular effects of heavy metals. Dordrecht; New York:
Therefore, contaminated water must be treated before it can Springer.
be released into the environment to ensure that safe drink- Bedrin AG, Bubnov IA, Dashuk SP, Mironov IS, Borypaev GG. 2003.
Compact spectral analyzer of heavy-metal impurities in air. J Opt
ing water is available to the public. In terms of agricultural Technol. 70(4):234–237.
safety, arsenic-containing herbicides should be replaced by Bernstam L, Nriagu J. 2000. Molecular aspects of arsenic stress. J Toxicol
safer alternatives. Unlike Amalgam fillings, dental composite Environ Health B-Crit Rev. 3:293–322.
174 Z. FU AND S. XI

Braeckman BP, Houthoofd K, Vanfleteren JR. 2009. Intermediary metabol- Genestra M. 2007. Oxyl radicals, redox-sensitive signalling cascades and
ism. WormBook. 3:1–24. antioxidants. Cell Signal. 19(9):1807–1819.
Challenger F. 1947. Biological methylation. Sci Prog. 35(139):396–416. Giaginis C, Gatzidou E, Theocharis S. 2006. DNA repair systems as targets
Chan TY. 2011. Inorganic mercury poisoning associated with skin-lighten- of cadmium toxicity. Toxicol Appl Pharmacol. 213(3):282–290.
ing cosmetic products. Clin Toxicol (Phila). 49(10):886–891. Godwin HA. 2001. The biological chemistry of lead. Curr Opin Chem Biol.
Chen G. H. 2004. Electrochemical technologies in wastewater treatment. 5(2):223–227.
Sep Purif Technol. 38(1):11–41. Haber LT, Erdreicht L, Diamond GL, Maier AM, Ratney R, Zhao Q,
Chen L, Liu L, Luo Y, Huang S. 2008. MAPK and mTOR pathways are Dourson ML. 2000. Hazard identification and dose response of inhaled
involved in cadmium-induced neuronal apoptosis. J Neurochem. nickel-soluble salts. Regul Toxicol Pharmacol. 31(2):210–230.
105(1):251–261. Hayakawa T, Kobayashi Y, Cui X, Hirano S. 2005. A new metabolic path-
Chen L, Xu B, Liu L, Luo Y, Zhou H, Chen W, Shen T, Han X, Kontos CD, way of arsenite: arsenic-glutathione complexes are substrates for
Huang S. 2011. Cadmium induction of reactive oxygen species acti- human arsenic methyltransferase Cyt19. Arch Toxicol. 79(4):183–191.
vates the mTOR pathway, leading to neuronal cell death. Free Radic Higashi T, Maruyama E, Otani T, Sakamoto Y. 1965. Studies on the isoci-
Biol Med. 50(5):624–632. trate dehydrogenase. I. Some properties of isocitrate dehydrogenase
Chin-Chan M, Navarro-Yepes J, Quintanilla-Vega B. 2015. Environmental of beef heart muscle. J Biochem. 57(6):793–798.
pollutants as risk factors for neurodegenerative disorders: Alzheimer Ho NY, Yang LX, Legradi J, Armant O, Takamiya M, Rastegar S, Strahle U.
and Parkinson diseases. Front Cell Neurosci. 9:124. 2013. Gene responses in the central nervous system of zebrafish
Cicik B, Engin K. 2005. The effects of cadmium on levels of glucose in embryos exposed to the neurotoxicant methyl mercury. Environ Sci
serum and glycogen reserves in the liver and muscle tissues of Technol. 47:3316–3325.
Cyprinus carpio (L., 1758). Turk J Vet Anim Sci. 29:113–117. Hughes MF, Beck BD, Chen Y, Lewis AS, Thomas DJ. 2011. Arsenic expos-
Clarkson TW. 1997. The toxicology of mercury. Crit Rev Clin Lab Sci. ure and toxicology: a historical perspective. Toxicol Sci. 123(2):
34(4):369–403. 305–332.
Clarkson TW, Magos L. 2006. The toxicology of mercury and its chemical Jarup L, Berglund M, Elinder CG, Nordberg G, Vahter M. 1998. Health
compounds. Crit Rev Toxicol. 36(8):609–662. effects of cadmium exposure–a review of the literature and a risk esti-
Coogan TP, Latta DM, Snow ET, Costa DM. 1989. Toxicity and carcino- mate. Scand J Work Environ Health. 24(1):1–51.
genicity of nickel compounds. Crit Rev Toxicol. 19(4):341–384. Jayawardena UA, Angunawela P, Wickramasinghe DD, Ratnasooriya WD,
Crespo-Lopez ME, Macedo GL, Pereira SI, Arrifano GP, Picanco-Diniz DL, Udagama PV. 2017. Heavy metal-induced toxicity in the Indian green
do Nascimento JL, Herculano AM. 2009. Mercury and human genotox- frog: biochemical and histopathological alterations. Environ Toxicol
icity: critical considerations and possible molecular mechanisms. Chem. 36(10):2855–2867.
Pharmacol Res. 60:212–220. Jomova K, Valko M. 2011. Advances in metal-induced oxidative stress
Crowe W, Allsopp PJ, Watson GE, Magee PJ, Strain JJ, Armstrong DJ, Ball and human disease. Toxicology. 283(2-3):65–87.
Kasperczyk S, Birkner E, Kasperczyk A, Kasperczyk J. 2005. Lipids, lipid
E, McSorley EM. 2017. Mercury as an environmental stimulus in the
peroxidation and 7-ketocholesterol in workers exposed to lead. Hum
development of autoimmunity – a systematic review. Autoimmun
Exp Toxicol. 24(6):287–295.
Rev. 16(1):72–80.
Kasperczyk A, Machnik G, Dobrakowski M, Sypniewski D, Birkner E,
Das KK, Gupta AD, Dhundasi SA, Patil AM, Das SN, Ambekar JG. 2006.
Kasperczyk S. 2012. Gene expression and activity of antioxidant
Effect of L-ascorbic acid on nickel-induced alterations in serum lipid
enzymes in the blood cells of workers who were occupationally
profiles and liver histopathology in rats. J Basic Clin Physiol
exposed to lead. Toxicology. 301(1–3):79–84.
Pharmacol. 17(1):29–44.
Kitchin KT, Wallace K. 2008. The role of protein binding of trivalent
Denkhaus E, Salnikow K. 2002. Nickel essentiality, toxicity, and carcino-
arsenicals in arsenic carcinogenesis and toxicity. J Inorg Biochem.
genicity. Crit Rev Oncol Hematol. 42(1):35–56.
102(3):532–539.
Dhir B, Sharmila P, Pardha Saradhi P, Sharma S, Kumar R, Mehta D. 2011.
Knowles SO, Donaldson WE. 1990. Dietary modification of lead toxicity:
Heavy metal induced physiological alterations in Salvinia natans.
effects on fatty acid and eicosanoid metabolism in chicks. Comp
Ecotoxicol Environ Saf. 74(6):1678–1684.
Biochem Physiol C. 95(1):99–104.
Diagomanolin V, Farhang M, Ghazi-Khansari M, Jafarzadeh N. 2004.
Korrick S. 2004. Effects of exposure to chemicals from industrial contam-
Heavy metals (Ni, Cr, Cu) in the Karoon waterway river, Iran. Toxicol ination in new Bedford, Massachusetts. Neurotoxicology. 25:673–673.
Lett. 151(1):63–68. Kumar S, Islam A, Ahmad H, Zaidi N. 2019. Graphene oxide supported
Ditzel E. J, Nguyen T, Parker P, Camenisch T. D. 2016. Effects of arsenite on amberlite resin for the analytical method development for
exposure during fetal development on energy metabolism and sus- enhanced column preconcentration/sensitive flame atomic absorption
ceptibility to diet-induced fatty liver disease in male mice. Environ spectrometric determination of toxic metal ions in environmental
Health Perspect. 124(2):201–209. samples. Ind Eng Chem Res. 58:8309–8316.
Eagles-Smith CA, Wiener JG, Eckley CS, Willacker JJ, Evers DC, Marvin- Lane TW, Morel FM. 2000. A biological function for cadmium in marine
DiPasquale M, Obrist D, Fleck JA, Aiken GR, Lepak JM, et al. 2016. diatoms. Proc Natl Acad Sci USA. 97(9):4627–4631.
Mercury in western North America: a synthesis of environmental con- Lenczewski E. 2009. Arsenic pollution: a global synthesis. Choice Curr
tamination, fluxes, bioaccumulation, and risk to fish and wildlife. Sci Rev Acad Libr. 47:326–326.
Total Environ. 568:1213–1226. Li L, Tian X, Yu X, Dong S. 2016. Effects of acute and chronic heavy
Ehara S, Ueda M, Naruko T, Haze K, Itoh A, Otsuka M, Komatsu R, metal (Cu, Cd, and Zn) exposure on sea cucumbers (Apostichopus
Matsuo T, Itabe H, Takano T, et al. 2001. Elevated levels of oxidized japonicus). Biomed Res Int. 2016:4532697.
low density lipoprotein show a positive relationship with the severity Liaw JG, Marshall Y, Yuan C, Ferreccio C, Steinmaus, Smith AH. 2008.
of acute coronary syndromes. Circulation. 103(15):1955–1960. Increased childhood liver cancer mortality and arsenic in drinking
Ekinci D, Beydemir S, Kufrevioglu O. I. 2007. In vitro inhibitory effects of water in northern Chile. Cancer Epidemiol Biomarkers Prev. 17:
some heavy metals on human erythrocyte carbonic anhydrases. J 1982–7.
Enzyme Inhib Med Chem. 22:745–750. Luz AL, Godebo TR, Bhatt DP, Ilkayeva OR, Maurer LL, Hirschey MD,
Ercal N, Aykin-Burns N, Gurer-Orhan H. 2001. Assessment of lead toxicity Meyer JN. 2016. From the cover: arsenite uncouples mitochondrial
in PC-12 cells. Free Rad Biol Med. 31:S116. respiration and induces a Warburg-like effect in Caenorhabditis ele-
Flora SJ, Mittal M, Mehta A. 2008. Heavy metal induced oxidative stress gans. Toxicol Sci. 152(2):349–362.
& its possible reversal by chelation therapy. Indian J Med Res. 128(4): M’Bemba-Meka P, Lemieux N, Chakrabarti S. K. 2006. Role of oxidative
501–523. stress, mitochondrial membrane potential, and calcium homeostasis
Fu F, Wang Q. 2011. Removal of heavy metal ions from wastewaters: a in human lymphocyte death induced by nickel carbonate hydroxide
review. J Environ Manage. 92(3):407–418. in vitro. Arch Toxicol. 80:405–420.
TOXICOLOGY MECHANISMS AND METHODS 175

Magos L. 1997. Physiology and toxicology of mercury. Met Ions Biol Syst. Rahman S, Sultana S. 2006. Chemopreventive activity of glycyrrhizin on
34:321–370. lead acetate mediated hepatic oxidative stress and its hyperprolifera-
Mahboob M, Shireen KF, Atkinson A, Khan AT. 2001. Lipid peroxidation tive activity in Wistar rats. Chem Biol Interact. 160(1):61–69.
and antioxidant enzyme activity in different organs of mice exposed Ramirez-Bajo MJ, de Atauri P, Ortega F, Westerhoff HV, Gelpi JL,
to low level of mercury. J Environ Sci Health B. 36(5):687–697. Centelles JJ, Cascante M. 2014. Effects of cadmium and mercury on
Marshall G, Ferreccio C, Yuan Y, Bates MN, Steinmaus C, Selvin S, Liaw J, the upper part of skeletal muscle glycolysis in mice. PLoS One. 9(1):
Smith AH. 2007. Fifty-year study of lung and bladder cancer mortality e80018.
in Chile related to arsenic in drinking water. J Natl Cancer Inst. 99(12): Rehman K, Fatima F, Waheed I, Akash MSH. 2018. Prevalence of expos-
920–928. ure of heavy metals and their impact on health consequences. J Cell
Mendez-Armenta M, Villeda-Hernandez J, Barroso-Moguel R, Nava-Ruiz C, Biochem. 119(1):157–184.
Jimenez-Capdeville ME, Rios C. 2003. Brain regional lipid peroxidation Risher JF, De Rosa CT, Jones DE, Murray HE. 1999. Updated toxicological
and metallothionein levels of developing rats exposed to cadmium profile for mercury. Toxicol Ind Health. 15(5):480–482.
Robey RB, Weisz J, Kuemmerle NB, Salzberg AC, Berg A, Brown DG,
and dexamethasone. Toxicol Lett. 144:151–157.
Miazek K, Iwanek W, Remacle C, Richel A, Goffin D. 2015. Effect of met- Kubik L, Palorini R, Al-Mulla F, Al-Temaimi R, et al. 2015. Metabolic
reprogramming and dysregulated metabolism: cause, consequence
als, metalloids and metallic nanoparticles on microalgae growth and
and/or enabler of environmental carcinogenesis? CARCIN. 36 (Suppl
industrial product biosynthesis: a review. IJMS. 16(10):23929–23969.
1):S203–S231.
Miller DM, Lund BO, Woods JS. 1991. Reactivity of Hg(II) with superoxide:
Rosenberg CE, Salibian A, Fink NE. 2002. An enzyme-linked immunosorb-
evidence for the catalytic dismutation of superoxide by Hg(II). J
ent assay for measuring anti-sheep red blood cells antibodies in lead-
Biochem Toxicol. 6(4):293–298.
exposed toads. J Pharmacol Toxicol Methods. 47(2):121–128.
Moreira EG, Vassilieff I, Vassilieff VS. 2001. Developmental lead exposure:
Rusyniak DE, Arroyo A, Acciani J, Froberg B, Kao L, Furbee B. 2010.
behavioral alterations in the short and long term. Neurotoxicol Heavy metal poisoning: management of intoxication and antidotes.
Teratol. 23(5):489–495. EXS. 100:365–396.
Mudipalli A. 2007. Lead hepatotoxicity & potential health effects. Indian Salnikow K, Zhitkovich A. 2008. Genetic and epigenetic mechanisms in
J Med Res. 126(6):518–527. metal carcinogenesis and cocarcinogenesis: nickel, arsenic, and chro-
Nagajyoti PC, Lee KD, Sreekanth TVM. 2010. Heavy metals, occurrence mium. Chem Res Toxicol. 21(1):28–44.
and toxicity for plants: a review. Environ Chem Lett. 8(3):199–216. Samadi A, Martinez L. A, Miranda M. A, Morera I. M. 2001. Mechanism of
Naranmandura H, Xu S, Sawata T, Hao WH, Liu H, Bu N, Ogra Y, Lou YJ, lipid peroxidation photosensitized by tiaprofenic acid: product studies
Suzuki N. 2011. Mitochondria are the main target organelle for triva- using linoleic acid and 1,4-cyclohexadienes as model substrates.
lent monomethylarsonous acid (MMA(III))-induced cytotoxicity. Chem Photochem Photobiol. 73(4):359–365.
Res Toxicol. 24(7):1094–1103. Schutt F, Aretz S, Auffarth GU, Kopitz J. 2012. Moderately reduced ATP
Nava M, Romero F, Quiroz Y, Parra G, Bonet L, Rodriguez-Iturbe B. 2000. levels promote oxidative stress and debilitate autophagic and phago-
Melatonin attenuates acute renal failure and oxidative stress induced cytic capacities in human RPE cells. Invest Ophthalmol Vis Sci. 53:
by mercuric chloride in rats. Am J Physiol Renal Physiol. 279(5): 5354–5361.
F910–F918. Sharma S, Kaul D, Singh D. 2015. Arsenic toxi-RNomics has the ability to
Ognjanovic BI, Markovic SD, Ethordevic NZ, Trbojevic IS, Stajn AS, Saicic tailor the host immune response. Exp Mol Pathol. 99(2):360–364.
ZS. 2010. Cadmium-induced lipid peroxidation and changes in anti- Shi H, Shi X, Liu KJ. 2004. Oxidative mechanism of arsenic toxicity and
oxidant defense system in the rat testes: protective role of coenzyme carcinogenesis. Mol Cell Biochem. 255(1/2):67–78.
Q(10) and vitamin E. Reprod Toxicol. 29:191–197. Shi X, Wei X, Koo I, Schmidt RH, Yin X, Kim SH, Vaughn A, McClain CJ,
Ohta H, Cherian M. G. 1991. Gastrointestinal absorption of cadmium and Arteel GE, Zhang X, et al. 2014. Metabolomic analysis of the effects of
metallothionein. Toxicol Appl Pharmacol. 107(1):63–72. chronic arsenic exposure in a mouse model of diet-induced Fatty liver
Oukarroum A, Zaidi W, Samadani M, Dewez D. 2017. Toxicity of nickel disease. J Proteome Res. 13(2):547–554.
oxide nanoparticles on a freshwater green algal strain of Chlorella vul- Shukla A, Shukla GS, Srimal RC. 1996. Cadmium-induced alterations in
garis. Biomed Res Int. 2017:1. blood-brain barrier permeability and its possible correlation with
Paglia DE, Valentine WN, Dahlgren JG. 1975. Effects of low-level lead decreased microvessel antioxidant potential in rat. Hum Exp Toxicol.
exposure on pyrimidine 5’-nucleotidase and other erythrocyte 15(5):400–405.
enzymes. Possible role of pyrimidine 5’-nucleotidase in the pathogen- Smith AH, Marshall G, Yuan Y, Ferreccio C, Liaw J, von Ehrenstein O,
esis of lead-induced anemia. J Clin Invest. 56(5):1164–1169. Steinmaus C, Bates MN, Selvin S. 2006. Increased mortality from lung
Pang Y, Jones MR, Tellez-Plaza M, Guallar E, Vaidya D, Post WS, Kaufman cancer and bronchiectasis in young adults after exposure to arsenic
in utero and in early childhood. Environ Health Perspect. 114(8):
JD, Delaney JA, Navas-Acien A. 2016. Association of geography and
1293–1296.
ambient air pollution with urine metal concentrations in six US cities:
Stohs SJ, Bagchi D, Hassoun E, Bagchi M. 2001. Oxidative mechanisms in
the multi-ethnic study of atherosclerosis. Int J Environ Res Public
the toxicity of chromium and cadmium ions. J Environ Pathol Toxicol
Health. 13:324.
Oncol. 20(2):12–88.
Patocka J, Cerny K. 2003. Inorganic lead toxicology. Acta Medica (Hradec
Tariq SR, Shah MH, Shaheen N, Jaffar M, Khalique A. 2008. Statistical
Kralove). 46(2):65–72. source identification of metals in groundwater exposed to industrial
Patrick L. 2002. Mercury toxicity and antioxidants: part 1: role of glutathi-
contamination. Environ Monit Assess. 138(1–3):159–165.
one and alpha-lipoic acid in the treatment of mercury toxicity. Altern Valko M, Morris H, Cronin MT. 2005. Metals, toxicity and oxidative stress.
Med Rev. 7(6):456–471. Curr Med Chem. 12(10):1161–1208.
Permpongpaiboon T, Nagila A, Pidetcha P, Tuangmungsakulchai K, Valko M, Rhodes CJ, Moncol J, Izakovic M, Mazur M. 2006. Free radicals,
Tantrarongroj S, Porntadavity S. 2011. Decreased paraoxonase 1 activ- metals and antioxidants in oxidative stress-induced cancer. Chem Biol
ity and increased oxidative stress in low lead-exposed workers. Hum Interact. 160(1):1–40.
Exp Toxicol. 30(9):1196–1203. Wang TS, Hsu TY, Chung CH, Wang AS, Bau DT, Jan KY. 2001. Arsenite
Qayyum S, Ara A, Usmani JA. 2012. Effect of nickel and chromium expos- induces oxidative DNA adducts and DNA-protein cross-links in mam-
ure on buccal cells of electroplaters. Toxicol Ind Health. 28(1):74–82. malian cells. Free Radic Biol Med. 31(3):321–330.
Rabbani N, Thornalley PJ. 2012. Methylglyoxal, glyoxalase 1 and the Wang L, Li J, Li J, Liu Z. 2010. Effects of lead and/or cadmium on the oxi-
dicarbonyl proteome. Amino Acids. 42(4):1133–1142. dative damage of rat kidney cortex mitochondria. Biol Trace Elem
Rahman M. 2002. Arsenic and contamination of drinking-water in Res. 137(1):69–78.
Bangladesh: a public-health perspective. J Health Popul Nutr. 20(3): Watjen W, Beyersmann D. 2004. Cadmium-induced apoptosis in C6 gli-
193–197. oma cells: influence of oxidative stress. Biometals. 17:65–78.
176 Z. FU AND S. XI

Wildemann TM, Siciliano SD, Weber LP. 2016. The mechanisms associ- Zhang HN, Yang LN, Ling JY, Czajkowsky DM, Wang JF, Zhang XW, Zhou
ated with the development of hypertension after exposure to lead, YM, Ge F, Yang MK, Xiong Q, et al. 2015. Systematic identification of
mercury species or their mixtures differs with the metal and the mix- arsenic-binding proteins reveals that hexokinase-2 is inhibited by
ture ratio. Toxicology. 339:1–8. arsenic. Proc Natl Acad Sci USA. 112(49):15084–15089.
Xia J, Lu L, Jin C, Wang S, Zhou J, Ni Y, Fu Z, Jin Y. 2018. Effects of short Zhao F, Malm SW, Hinchman AN, Li H, Beeks CG, Klimecki WT. 2014.
term lead exposure on gut microbiota and hepatic metabolism in Arsenite-induced pseudo-hypoxia results in loss of anchorage-
dependent growth in BEAS-2B pulmonary epithelial cells. PLoS One.
adult zebrafish. Comp Biochem Physiol C Toxicol Pharmacol. 209:1–8.
9(12):e114549.
Yang K, Ren YB. 2010. Nickel-free austenitic stainless steels for medical
Zhao F, Severson P, Pacheco S, Futscher BW, Klimecki WT. 2013. Arsenic
applications. Sci Technol Adv Mater. 11:014105.
exposure induces the Warburg effect in cultured human cells. Toxicol
Yu HS, Liao WT, Chai CY. 2006. Arsenic carcinogenesis in the skin. J
Appl Pharmacol. 271(1):72–77.
Biomed Sci. 13(5):657–666. Zhao CQ, Young MR, Diwan BA, Coogan TP, Waalkes MP. 1997.
Yuan Y, Marshall G, Ferreccio C, Steinmaus C, Liaw J, Bates M, Smith AH. Association of arsenic-induced malignant transformation with DNA
2010. Kidney cancer mortality: fifty-year latency patterns related to hypomethylation and aberrant gene expression. Proc Natl Acad Sci
arsenic exposure. Epidemiology. 21(1):103–108. USA. 94(20):10907–10912.
Yun SW, Hoyer S. 2000. Effects of low-level lead on glycolytic enzymes Zhou Z. H, Lei YX, Wang CX. 2012. Analysis of aberrant methylation in
and pyruvate dehydrogenase of rat brain in vitro: relevance to spor- DNA repair genes during malignant transformation of human bron-
adic Alzheimer’s disease? J Neural Transm. 107(3):355–368. chial epithelial cells induced by cadmium. Toxicol Sci. 125(2):412–417.

You might also like