You are on page 1of 37

Journal of Toxicology and Environmental Health, Part B

Critical Reviews

ISSN: 1093-7404 (Print) 1521-6950 (Online) Journal homepage: http://www.tandfonline.com/loi/uteb20

Biomarkers of mercury toxicity: Past, present, and


future trends

Vasco Branco, Sam Caito, Marcelo Farina, João Teixeira da Rocha, Michael
Aschner & Cristina Carvalho

To cite this article: Vasco Branco, Sam Caito, Marcelo Farina, João Teixeira da Rocha,
Michael Aschner & Cristina Carvalho (2017): Biomarkers of mercury toxicity: Past,
present, and future trends, Journal of Toxicology and Environmental Health, Part B, DOI:
10.1080/10937404.2017.1289834

To link to this article: http://dx.doi.org/10.1080/10937404.2017.1289834

Published online: 05 Apr 2017.

Submit your article to this journal

View related articles

View Crossmark data

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


http://www.tandfonline.com/action/journalInformation?journalCode=uteb20

Download by: [Iowa State University] Date: 05 April 2017, At: 13:48
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B
http://dx.doi.org/10.1080/10937404.2017.1289834

Biomarkers of mercury toxicity: Past, present, and future trends


Vasco Brancoa, Sam Caitob, Marcelo Farinac, João Teixeira da Rochad, Michael Aschnerb, and Cristina Carvalhoa
a
Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal; bDepartment of
Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA; cDepartamento de Bioquímica, Centro de Ciências
Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil; dDepartamento Bioquímica e Biologia Molecular, Universidade
Federal de Santa Maria, RS, Brazil

ABSTRACT
Mercury (Hg) toxicity continues to represent a global health concern. Given that human populations
are mostly exposed to low chronic levels of mercurial compounds (methylmercury through fish,
mercury vapor from dental amalgams, and ethylmercury from vaccines), the need for more sensitive
and refined tools to assess the effects and/or susceptibility to adverse metal-mediated health risks
remains. Traditional biomarkers, such as hair or blood Hg levels, are practical and provide a reliable
measure of exposure, but given intra-population variability, it is difficult to establish accurate cause–
effect relationships. It is therefore important to identify and validate biomarkers that are predictive of
early adverse effects prior to adverse health outcomes becoming irreversible. This review describes
the predominant biomarkers used by toxicologists and epidemiologists to evaluate exposure, effect
and susceptibility to Hg compounds, weighing on their advantages and disadvantages. Most impor-
tantly, and in light of recent findings on the molecular mechanisms underlying Hg-mediated toxicity,
potential novel biomarkers that might be predictive of toxic effect are presented, and the applicability
of these parameters in risk assessment is examined.

Mercury toxicity (ASSGM) mainly in developing countries (Armah


et al. 2016; Bose-O’Reilly et al. 2016; Castilhos et al.
Mercury (Hg) is a ubiquitous environmental pollu-
2015; Gibb and O’Leary 2014; Kristensen, Thomsen,
tant to which humans are exposed to in varying
and Mikkelsen 2014), and the number of humans
amounts and chemical forms (Clarkson and Magos
exposed to this element is still high (Gibb and
2006). The three main forms of Hg that are a cause of
O’Leary 2014).
concern to nonoccupationally exposed human popu-
All Hg forms generate Hg2+. In the case of Hg0,
lations include methylmercury (MeHg), mercury
oxidation is facilitated by catalase (CAT), a process
vapor (Hg0), and ethylmercury (EtHg) (Clarkson,
first described in red blood cells by Halbach and
Magos, and Myers 2003). Ingestion of fish containing
Clarkson (1978). Demethylathion of MeHg to Hg2+
MeHg is by far the most common route of exposure
is also known to occur (Mottet et al. 1997; Vahter
to Hg, inhalation of Hg0 occurs due to its release
et al. 1995; Watanabe 2002). Several hypotheses sug-
from dental amalgams, whereas exposure to EtHg
gest the involvement of selenium (Se) and free radi-
results from the use of Thimerosal (Ethyl (2-mercap-
cal attack in this process, but the precise mechanism
tobenzoato-(2-)-O,S) mercurate(1-) sodium)-con-
has yet to be determined (Yang et al. 2008).
taining vaccines (TCV) (Dórea, Farina, and Rocha
The main adverse health effects of Hg include
2013). In addition, given that Hg compounds are still
neurotoxicity, teratogenicity, nephrotoxicity, and
in use in many human activities, occupational expo-
immunotoxicity (Counter et al. 2002; Ratcliffe,
sure is also of relevance to metal-induced toxicity
Swanson, and Fischer 1996; Sweet and Zelikoff
and an important driving force for the discovery of
2001). The magnitude of these toxic effects may
new putative biomarkers. It is well known that Hg0 is
vary considerably, depending upon the life-stage,
employed in artisanal small-scale gold mining

CONTACT Cristina Carvalho cristina.carvalho@ff.ulisboa.pt Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de
Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal.
Color versions of one or more of the figures in the article can be found online at www.tandfonline.com/uteb.
© 2017 Taylor & Francis Group, LLC
2 V. BRANCO ET AL.

dose, and duration of exposure (Clarkson and from a clinical perspective (Figure 1A) (Benford
Magos 2006), and symptoms may intensify, et al. 2000; Wallace, Kormos, and Pleil 2016).
become irreversible, and eventually lead to death Ideally, biomarkers should be noninvasive, sensi-
(WHO 2003). Even though the adverse health tive, and specific for the xenobiotic that is being
effects attributed to Hg compounds are well assessed (Bernard 2008; Grandjean et al. 1994;
known, the molecular basis underlying the devel- Paustenbach 2001; Tan et al. 2012). Biomarkers
opment of toxicity is still not fully understood. play an essential role in risk assessment by redu-
Given the adverse effects of Hg compounds, it is cing overall uncertainty (Maier, Savage, and Haber
imperative to assess exposure and detect early 2004).
effects predictive of toxicity, that is, an early stage Classically, biomarkers have been divided into
of an adverse effect, especially in vulnerable popu- biomarkers of exposure, effect, or susceptibility
lations. This is of extreme importance for risk (National Research Council (NRC) 1987), although
assessment, particularly when dealing with expo- the boundaries between these are difficult to estab-
sure levels at the lower end of the dose-response lish (Grandjean et al. 1994; WHO 1993) (Figure 1A).
curve, since biomarkers of early effects exhibit Currently, the use of biomarkers has broadened,
greater sensitivity when compared to classical encompassing employment of pharmacogenomics,
assessment of clinical symptoms, functional tests, and other biomarkers in medicine as shown in
or morbidity (Bernard 2008; Ratcliffe, Swanson, Figure 1B.
and Fischer 1996). Exposure biomarkers may be markers of internal
Biomarkers are indicator-signaling events in bio- dose, that is, the direct measurement of the com-
logical systems or samples (body fluids, cells, or pound, its metabolites or conjugates in a biological
tissues) (NRC 1987). The usefulness of biomarkers sample such as urine, blood, hair, and markers of
in identifying a postulated adverse health effect is biologically effective dose (BED) when they evaluate
intrinsically related to their role (Benford et al. 2000) interaction between xenobiotics and macromole-
within the mode of action of the toxicant. cules, such as proteins or DNA (World Health
The objective of this review is to describe the Organization (WHO) 1993). Although markers of
principal biomarkers used in the evaluation of Hg- BED may not be crucial for the development of
induced toxicity weighing their predictiveness and/ toxicity as they are not necessarily linked to the
or insufficiencies. In addition, recent discoveries on events that trigger the neurotoxicity nephrotoxicity,
the mechanisms underlying Hg-mediated toxicity or other biological system, these markers share a
are presented and we examine whether these find- common etiology (National Research Council
ings, for which there may be none or limited epide- (NRC) 1987; Timbrell 1998; Woods 1995).
miological data available, may potentially prove Biomarkers of effect represent alterations includ-
useful as novel biomarkers of metal-induced toxicity. ing biochemical, physiological, or behavioral that
can be measured in an organism and attributed to
exposure to a xenobiotic. Hence, biomarkers of effect
Biomarker applications are directly linked to toxicity (this needs biomarker
Biomarkers have been defined as measures in cel- validation). Ideally, these biomarkers should be pre-
lular or biochemical components or processes, dictive of effect, as these measures should identify
structures, or functions (including neurobeha- initial alterations or adverse effects prior to becom-
vioral) that can be measured in a biological system ing irreversible or considered pathological evidence
(Gil and Pla 2001; Kendall et al. 2001; Paustenbach (Benford et al. 2000; Bernard 2008; Gil and Pla 2001;
2001; Silbergeld and Davis 1994; Tan et al. 2012; World Health Organization (WHO) 1993).
Wallace, Kormos, and Pleil 2016). These altera- Biomarkers of susceptibility reflect characteristics
tions may occur at any stage along the causal that increase individual sensitivity to a given com-
pathway, which varies from minimal molecular pound by either increasing its internal dose or by
changes resulting from exposure to the toxic com- lowering the threshold toxic dose, for example, as a
pound until establishment of overt toxicity, when result of genetic polymorphisms (Schmidt 2006)
evident endpoints of toxicity become apparent (Figure 1).
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B 3

Figure 1. Applications of biomarkers in different areas of toxicology and medicine. Biomarkers can be used to evaluate exposure and
predict the toxicity (A) of a xenobiotic by measuring the internal dose, the dose interacting with molecular targets or molecular
effects that reflect early changes or that represent an established toxic response. If toxicity persists, disease ensues (B), and it may be
diagnosed by standard clinical markers. New approaches make use of biomarkers for the development of new medicines, and
moreover, the identification of biomarkers that follow disease progression will become a key element for personalized medicine

Mercury biomarkers compounds. Consequently, it is important that the


biological indicator used as a biomarker of exposure
Biomarkers of exposure
translates the Hg burden in the organism especially
Monitoring of exposure levels in the human popula- at the target organ(s). In addition, the appropriate
tion, in particular in groups at risk, such as pregnant biomarker of exposure depends upon the type of
women, newborns, children and occupationally exposure (acute vs. chronic) (Clarkson, Vyas, and
exposed workers, has been the basis of Hg-associated Ballatori 2007).
risk assessment. Mercury enters the human body by
various routes and in different chemical forms, and
Biomarkers of exposure: Internal dose
each of this has its own distinctive toxicological
features. Thus, not all biological indicators have the The most common biomarkers of exposure to mer-
same reliability as biomarkers of exposure for all Hg curials are internal dose markers that encompass
4 V. BRANCO ET AL.

Toxicity Susceptibility
Overt
Altered Toxicity
structure or
function

Early
biological
Biologically effect
effective
dose
Internal
dose

Exposure
Dose

Figure 2. Pathway leading from exposure to a xenobiotic to toxic manifestations, and the relative position of exposure and effect biomarkers

measurements of total Hg levels in hair, urine, and 2000). Normal levels of Hg in hair range between 1
blood (Berglund et al. 2005), in the absence of and 2 μg/g, but individuals who consume large
speciation analysis. Other biomarkers, such as the amounts of fish may have hair Hg levels in excess
levels of Hg in placental cord-blood, feces, breast- of 10 μg/g (WHO/UNEP 2008) with the lowest
milk, or nails, are seldom used although in parti- observed adverse effect level (LOAEL) for neuro-
cular situations they have been proven quite useful toxic effects (paresthesia) in adults set at 50 μg/g
(Bose-O’Reilly et al. 2010; Cooke 2014; LaKind et al. (Clarkson and Magos 2006).
2005; Ohno et al. 2007). Despite their common use Another advantage of quantifying Hg levels in
in Hg-induced toxicity evaluation (Maier, Savage, hair is that it allows for retrospective studies by
and Haber 2004), it should be stressed that, due to evaluation of Hg in different sections of the hair
the inter-individual human variability, the levels of strand (Mottet et al. 1997). Additionally, levels of
Hg in hair, blood, or urine do not necessarily reflect Hg in maternal hair correlate well with the blood
the presence or absence of toxicity (Bose-O’Reilly Hg levels in the fetus, which makes it a useful bio-
et al. 2010). marker of pre-natal exposure for risk assessment
purposes (Cooke Ohno. 2014; World Health
Mercury levels in the hair Organization/United Nations Environment
MeHg binds sulfhydryl (SH-) groups of keratin Programme (WHO/UNEP) 2008). The no observed
and is integrated in the hair (Mottet et al. 1997) adverse effect level (NOAEL) for developmental
constituting more than 80% of the hair metal effects is estimated to be 10 μg Hg/g in maternal
burden (Berglund et al. 2005). Hair follicles are hair (Clarkson and Magos 2006).
thought to accumulate the same transportable spe- During pregnancy, maternal hair Hg levels may
cies that reach the brain (Clarkson, Vyas, and decrease up to 20%, indicating placental transfer of
Ballatori 2007). In fact, hair Hg levels correlate Hg to the fetus (Barbosa, Silva, and Dórea 1998). In
well with levels in the brain and whole blood at a fact, placental transfer of MeHg is far more impor-
ratio of 250:5:1, respectively (Clarkson and Magos tant than transfer by breast-milk (Barbosa, Silva,
2006) (Figure 3). However, the ratio of Hg levels and Dórea 1998; Björnberg et al. 2005) in determin-
between blood and brain, and blood and hair may ing the relationship between maternal and newborn
vary according to individual’s characteristics such hair Hg levels, in fish eating populations (Barbosa,
as age, gender, and genetics (Bartell et al. 2000; Silva, and Dórea 1998; Díez et al. 2009; Kim, Jeon,
Doi and Tagawa 1983). and Paek 2008, 2015; Marques et al. 2013; Oliveira
The concentration of Hg in hair is an appro- et al. 2010; Ou et al. 2015; Savabieasfahani, Hoseiny,
priate biomarker to address exposure to MeHg in and Goodarzi 2012; Sikorski, Paszkowski, and
a noninvasive manner (Berglund et al. 2005; Satoh Szprengier-Juszkiewicz 1986). Infant hair Hg
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B 5

Mercury
Brain levels in
(5)* hair (250)*

MeHg Mercury
intake Brain Hair
Vapour
5 250
Ethylmercury
exposure BBB Hair follicle
MeHg-Cys

Hg levels in 1 RBC 2
Plasma 0.1
Total Blood
Developing
fetus Kidney

Mercury
levels in
Urine

Figure 3. Main pathways of exposure to mercury compounds, exposure biomarkers and relative concentrations of MeHg in hair,
brain, and blood. Humans are exposed to different mercury compounds by different routes, such as MeHg (fish consumption), Hg°
(dental amalgams), and EtHg (TCV). The major target organs include the brain, kidney, and the developing fetus, and exposure levels
are normally assessed by measuring mercury levels in hair, blood, and urine. In the case of MeHg, levels in hair and blood correlate
well with the values found in the brain in the proportion 250:5:1. RBC – red blood cells; BBB – blood brain barrier. (adapted from
Clarkson, Vyas, and Ballatori 2007)

concentration is a reliable biomarker for in utero et al. 1997), as the Hg2+ found in hair is likely the
MeHg exposure. Notably, breast-fed infants receiv- result of MeHg demethylation within hair follicles
ing Thimerosal-containing vaccines (TCV) show (Berglund et al. 2005) or external deposition
asymmetric changes in Hg hair levels relatively to (Bose-O’Reilly et al. 2008). Moreover, in pregnant
their mother, indicating that hair Hg levels also women living in areas where ASSGM is practiced,
reflect exposure to EtHg in addition to MeHg Hg levels in hair did not accurately reflect concen-
(Marques et al. 2007). Speciation analysis demon- trations found in other biological media such as
strated that EtHg levels in hair are positively corre- breast-milk, which might underestimate infant
lated with the number of TCV inoculations and exposure (Bose-O’Reilly et al. 2008). Barbosa
negatively correlated with the time elapsed since et al. (1995) compared populations from the
the last vaccination (Dórea et al. 2011). This addi- Brazilian Amazon that were exposed to Hg by
tional burden of EtHg from TCV further adds to different routes (ASSGM vs. fish consumption)
the pre- and post-natal Hg burden (Dórea, and showed that in general, levels in hair were a
Marques, and Isejima 2012). However, it should more reliable descriptor for exposure to Hg from
be stressed that several epidemiological studies fish (mainly MeHg) than for Hg vapor from
(Andrews et al. 2004; Fombonne et al. 2006; ASSGM activities. However, in utero exposure to
Heron and Golding 2004; Hviid et al. 2003; Hg0 from maternal dental amalgams was claimed
Madsen et al. 2003; Stehr-Green et al. 2003; to contribute to increased levels of Hg in baby hair
Taylor, Swerdfeger, and Eslick 2014; Uno et al. (Lindow et al. 2003). To overcome the possibility
2015; Verstraeten et al. 2003) failed to detect any of mixed sources of exposure (dietary MeHg vs.
positive association between TCVs and impaired occupational Hg0), the measurement of Hg stable
neurodevelopment. isotopic signatures in hair was applied as a useful
Hair Hg levels are normally not considered an technique to distinguish between different sources
appropriate biomarker of exposure to Hg0 (Mottet of Hg in the organism (Laffont et al. 2011).
6 V. BRANCO ET AL.

Mercury levels in urine evenly distributed between RBCs and plasma


The concentration of Hg in urine (U-Hg) is the most (Berglund et al. 2005; Clarkson, Vyas, and Ballatori
common biomarker of exposure to Hg0 both in occu- 2007).
pational exposures and in dental amalgams (Magos Plasma Hg accounts for a smaller fraction of whole
1997). Its utilization in monitoring workers exposed blood Hg that may range from 5 to 35%, depending
to Hg dates back to the first half of the 20th century upon the characteristics of the population such as age
(Buckell et al. 1946; Neal 1938). Urinary mercury is and diet (Carneiro, Grotto, and Barbosa 2014). Hg2+ is
derived from Hg that accumulates in kidney cells after normally the dominant species, although large varia-
acute (Bose-O’Reilly et al. 2010) or chronic exposure tions exist between individuals concerning the ratio
to Hg0 (Cherian and Clarkson 1976), and conse- between Hg2+ and MeHg in plasma (Carneiro, Grotto,
quently, in the mid- to long term, this reflects the and Barbosa 2014). Since Hg kinetics in blood is
Hg2+ body burden (Clarkson and Magos 2006; relatively fast, this biomarker has the limitation of
Zalups 2000). Neurotoxic effects attributed to Hg0 only being useful for a short time after an acute
are evident in subjects with U-Hg levels exceeding exposure, or in the case of continuous (chronic) expo-
35 μg/g creatinine (SCOEL 2007). However, several sure (Satoh 2000).
investigators indicated the occurrence of neurobeha- Concerning pre-natal development, Hg cord-
vioral effects in the 20–30 μg of Hg /g creatinine range blood levels may be employed to quantify fetal
or lower (Echeverria et al. 1998; Meyer-Baron, exposure to MeHg (Aylward et al. 2014; Cooke
Schaeper, and Seeber 2002). 2014). However, since fetal blood levels are
Experiments with rats showed that even in chronic known to be 1.5- to twofold higher than maternal
exposures the urinary excretion of MeHg is low blood concentrations, the latter is the preferred
(Zalups, Barfuss, and Kostyniak 1992), since the biomarker for assessing pre-natal exposure
MeHg-Cys conjugate is slowly excreted in urine (World Health Organization/United Nations
(Yasutake, Hirayma, and Inoue 1989). In addition, Environment Programme (WHO/UNEP) 2008).
in human populations exposed to different sources
of Hg, urine levels were noted to be a less sensitive
biomarker for MeHg exposure through fish consump-
Biomarkers of exposure: Biologically effective
tion in comparison with Hg0 derived from ASSGM
dose
activities (Barbosa et al. 1995). Accordingly, U-Hg
levels are not considered a useful biomarker for asses- As referred to above, biomarkers of biologically
sing MeHg exposure (Berglund et al. 2005). effective dose (BED) indicate the interaction of
the xenobiotic with macromolecules such as pro-
Mercury levels in blood teins including albumin or hemoglobin in blood or
The blood is responsible for the distribution of all nucleic acids, but these interactions are mainly a
forms of Hg to target organs (Clarkson, Vyas, and consequence of the absorption distribution meta-
Ballatori 2007). Thus, the concentration of Hg in bolism excretion (ADME) processes and not
blood (B-Hg) is considered a reliable biomarker of necessarily related to adverse effects development.
exposure. In fish eating populations, metal levels in Due to the electrophilic nature of Hg, once
whole blood are usually interpreted as reflecting expo- absorbed into the organism, it binds nucleophilic
sure to MeHg (Berglund et al. 2005; Bose-O’Reilly groups, such as thiols (SH) and selenols (SeH) in
et al. 2010). B-Hg levels in individuals with no con- cysteine (Cys) and selenocysteine (Sec), respec-
sumption of highly contaminated fish are normally tively. Consequently, peptides and proteins con-
between 5 and 10 μg/L (World Health Organization/ taining these moieties are targeted by mercurials
United Nations Environment Programme (WHO/ (Carvalho et al. 2008a; Farina et al. 2009; Rooney
UNEP) 2008), while in populations with limited fish 2007; Silva De Paula et al. 2016).
consumption, B-Hg normally does not exceed 2μg/L Conjugation of Hg compounds with cysteine-
(NRC 2000). In blood, approximately 90% of MeHg is containing compounds, such as GSH, primarily
found in red blood cells (RBC) bound to hemoglobin facilitates detoxification (Bridges and Zalups
(Figure 3), while inorganic Hg (Hg0 and Hg2+) is 2010; Clarkson and Magos 2006); however, it
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B 7

may also trigger toxicity upon metabolism. Aschner, and Rocha 2011). However, it is difficult
Simmon-Willis et al (2002) showed that by bind- to understand how the equimolar interaction
ing to the thiol in Cys, MeHg gains access to the between Hg and GSH might lead to GSH depletion
central nervous system (CNS), since the MeHg- and, consequently, oxidative stress. In fact, GSH is
Cys complex mimics the amino acid methionine present in some mammalian cells at concentrations
and thus is transported across membranes by neu- as high as 10 millimolar (Cooper and Kristal 1997),
tral amino acid transporters. Similarly, Hg2+ is and Hg compounds (particularly, MeHg) were
known to bind two Cys or GSH molecules forming reported to induce oxidative damage in cultured
complexes similar to cystine (Cys-Cys) and glu- cells when present at concentrations as low as 1
tathione disulfide (GSSG). Formation of these micromolar (Farina et al. 2009). In addition, experi-
types of conjugates has been involved in Hg2+ mental studies based on in vivo approaches noted
and MeHg uptake and excretion from the CNS, reduced cerebellar (Franco et al. 2006), cerebral
kidneys, liver, intestine, placenta (Bridges and (Stringari et al. 2008), and blood (Silva De Paula
Zalups 2010). In addition, inhibition of Cys- and et al. 2016) GSH levels in MeHg-exposed animals
Sec-containing enzymes produced by covalent when Hg levels in these organs were in the low
binding of mercurials (discussed below) may lead micromolar range. Thus, it is noteworthy that
to increased reactive oxygen species (ROS) genera- decreased GSH levels following Hg exposure do
tion and oxidative stress. not necessarily indicate direct interaction between
Taken together, Hg compounds have several Hg and thiol groups, but may represent the conse-
potential binding targets, and it becomes challenging quence of direct oxidative effects of reactive species
to determine which of these interactions are critical (i.e., hydrogen peroxide) whose levels are increased
for the development of toxicity. The BED biomar- after Hg exposure (Franco et al. 2007).
kers endpoints which have been thoroughly studied A comparison of blood GSH levels in populations
in relation to Hg toxicity are presented; however, from the Amazon showed an association with hair Hg
these have not been directly linked to the main path- concentrations (Pinheiro et al. 2008). In fact, hair Hg
ways of toxicity that culminate in cell death. Some of levels above 10 μg/g typically led to higher GSH (20–
the important BED biomarkers for Hg exposure and 60%) quantities than following lower exposure (hair
their potential value are discussed below. Hg levels below <4 μg/g on average), even tough the
correlation is scattered (Pinheiro et al. 2008).
Depletion of intracellular glutathione Similarly, elevated GSH levels in blood (+45%) were
Glutathione is the most abundant nonprotein cellu- associated with seasonal variation in hair Hg concen-
lar thiol, present at millimolar concentrations in trations in sports fishermen (1.4 μg/g vs. 2.8 μg/g)
several organs including liver, kidneys, and brain (Bélanger et al. 2008).
(Meister 1988; Pastore et al. 2003). Conjugation In miners, previously exposed to Hg0, a 10%
with GSH is an important step in the toxicokinetics increase in erythrocyte GSH levels was noted (Kobal
and systemic transport of mercurials, and several in et al. 2008). Nonetheless, in older, retired miners levels
vitro and in vivo studies (Bo, Miller, and Woods of GSH were 20% lower than in controls, suggesting
1993; Franco et al. 2007; Fujiyama, Hirayama, and the existence of age-related factors influencing overall
Yasutake 1994; Piccoli et al. 2012) demonstrated GSH blood levels (Kobal et al. 2008). However, many
GSH depletion following exposure to MeHg and other compounds, such as paraquat (Djukic et al.
Hg2+. However, for the case of Hg2+, the binding to 2012), arsenic (Jain et al. 2012), benzo(a)pyrene
free Cys is more important than GSH, because Hg- (Romero et al. 1997), and acetaminophen
Cys complexes are more efficiently transported than (McMurtry, Snodgrass, and Mitchell 1978) may also
Hg-GSH complexes in tubular cells (Zalups and decrease GSH levels. Consequently, in a multi-con-
Barfuss 2002a). taminant or therapeutic context, this endpoint does
An excessive Hg burden may result in marked not permit distinction of Hg-related toxicity from that
depletion of reduced GSH, with serious conse- produced by other compounds. Thus, the usefulness
quences to the cell antioxidant capacity (Farina, of GSH as a BED is quite limited.
8 V. BRANCO ET AL.

Impairment of heme-biosynthesis and involved and promoting porphyrin accumulation


coproporphyrin accumulation (Woods et al. 2009). These accumulated prophyr-
Porphyrin metabolism has been proposed as an ins are then excreted, and their rise in urine can
indicator of metal exposure and toxicity (Woods be utilized as a biomarker of exposure to metals.
1995). The synthesis of the prosthetic group heme Mercury targets the enzymes uroporphyrinogen
is an important physiological process that occurs decarboxylase (UROD), and especially copropor-
in all tissues and involves several product inter- phyrinogen oxidase (CPOX), thus leading to
mediates and enzymatic processes, from the accumulation and excretion of 4 and 5 carboxyl
initial substrate (Succinil CoA) to the final heme porphyrines (Woods 1995; Woods et al. 2005).
group (Figure 4) (Woods et al. 2009). In the Specifically, in the case of Hg, there is also excre-
normal unfolding of the heme biosynthesis pro- tion of an atypical “precoproporphyrin” known as
cess, overproduction of intermediate porphyrino- keto-isocoproporphyrin (KICP) (Woods, Bowers,
gens is followed by their oxidation to porphyrins, and Davis 1991), which was proposed as a bio-
which are then excreted in the urine (Woods et al. marker of exposure for both MeHg and Hg0
1993). Several metals, including lead (Pb), arsenic (Marks 1985; Woods 1995). Moreover, such
(As), and mercury (MeHg and Hg2+) (Fowler and alterations occur prior to the onset of target tissue
Mahaffey 1978; Pingree et al. 2001; Sakai 2000; injury, suggesting that porphyrines may be used
Woods and Fowler 1977, 1978), interfere in a as biomarkers predictive of early toxic effects
dose-dependent manner with the normal course (Woods 1995; Woods et al. 2009). Animal studies
of the heme biosynthetic pathway by inhibiting (Pingree et al. 2001) have supported the useful-
the activity of several of the specific enzymes ness of coproporphyrines in predicting

3,4
Porphobilinogen Uroporphynogen
2 5

Heptacarboxyporphynogen
δ-Aminolevulinic Acid
5
1

Succinyl CoA Hexacarboxyporphynogen


Mitochondria 5

Pentacarboxyporphynogen
5
Cytosol
Coproporphynogen

6
8 7
Heme Protoporphyrin IX Protoporphynogen IX

Figure 4. The Heme biosynthetic pathway. The enzymes involved in heme biosynthesis include: 1-δ-aminolevulinic acid (ALA)
synthetase; 2-ALA dehydratase; 3-uroporphyrinogen I synthetase; 4-uroporphyrinogen III cosynthetase; 5-uroporphyrinogen dec-
arboxylase; 6-coproporphyrinogen oxidase; 7-protoporphyrinogen oxidase; 8-ferrochelatase. Mercury targets the enzymes uropor-
phyrinogen decarboxylase (UROD; 5), and especially coproporphyrinogen oxidase (CPOX; 6), thus leading to the accumulation and
excretion of 4 and 5 carboxylporphyrinogens which afterward are oxidized to porphyrins and excreted in urine (adapted from
Pingree et al. 2001)
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B 9

A
+HgCH (MeHg)
3 Selenium
from dietary
sources
+HgCH (MeHg)
3
—SeHgCH3
dysfunctional
protein
—SeH
HSe- HSeHgCH3
functional protein AMP + PPi

ATP (4)

Ser PSer Sec

Ser + ATP ATP H2SePO3-

(1) (2) (3)


AMP + PPi ADP 2Pi

tRNA(Ser)Sec Ser-tRNA(Ser)Sec PSer-tRNA(Ser)Sec Sec-tRNA(Ser)Sec


B

Figure 5. Potential mechanisms by which MeHg decreases selenoproteins’ homeostasis. (A) MeHg directly interacts with thE selenol
group of selenocysteine. Due to its high electrophilicity, MeHg directly interacts with the selenol group (–SeH) of selenoccysteine
(red arrow). Because this group is classically responsible for catalytic activity in several selenoproteins, loss of protein function is
observed. (B) MeHg may impair the biosynthesis of selenocysteine. Selenocysteine (Sec) is crucial for the proper synthesis and
functioning of selenoproteins. The biosynthesis of Sec occurs on its tRNA, and the pathway begins with the attachment of serine to
Sec tRNA[Ser]Sec (event 1; catalyzed by seryl-tRNAsynthetase) in the presence of ATP. Phosphoseryl-tRNA kinase (event 2)
phosphorylates the serine moiety to form an intermediate, phosphoseryl-tRNA[Ser]Sec (PSer-tRNA[Ser]Sec). Sec synthase (event 3)
catalyzes the formation of Sec-tRNA[Ser]Sec, which is subsequently incorporated into selenoproteins during protein synthesis. This
metabolic step requires the presence of selenophosphate (H2SePO3–), whose precursor is selenide (HSe−) (event 4). MeHg (CH3Hg+)
directly interacts with selenide (red arrow), leading to a “selenium-deficient-like” condition, which lead to inappropriate selenopro-
tein synthesis (Usuki, Yamashita, and Fujimura 2011). For additional details on Sec biosynthesis, see (Hatfield et al. 2014).
Abbreviations: Pi, inorganic phosphate; PPi, inorganic pyrophosphate

nephrotoxic effects. Further, a relationship 2005). Therefore, MTs contain several thiol
between urinary porphyrin changes and altera- groups, which may be complexed by heavy metal
tions in neurobehavioral function resulting from ions (up to 7 molecules). Hg0 and MeHg are
prolonged Hg exposure was described by known to induce MT synthesis after conversion
Echeverria et al. (1994). However, alterations in to Hg2+ in tissues, especially in the kidney and
porphyrin excretion do not necessarily represent liver (Gerson and Shaikh 1982; Piotrowski et al.
a step in the casual pathway between exposure 1974; Yasutake and Nakamura 2011), and binding
and effect, and thus, its value as a predictive to its SH groups concurrently enhances urinary
biomarker of neurotoxic effects has yet to be excretion of Cu2+ and Zn2+ (Chmielnicka,
established. Brzeźnicka, and Sniady 1986; Liu, Nordberg, and
Jin 1992). However, other metals, both essential
Induction of metallothioneins (Zn2+ and Cu2+) and nonessential (Ag2+; Cd2+),
Metallothioneins (MTs) are low-molecular weight also induce MT synthesis (Zalups 2000); thus, it
proteins (6–7 kDa) rich in Cys residues (30% of becomes exceedingly difficult to discern between
amino acid residues of MT are cysteine), and with- Hg-induced alterations in the presence of other
out aromatic amino acids (Eroglu, Atli, and Canli metals with this biomarker.
10 V. BRANCO ET AL.

Oxidative stress indicators (ROS and NOS) predictive of early toxic manifestations (Santos et al.
Mercurials are known to enhance production of 2007). Of note, several of the aforementioned beha-
reactive oxygen/nitrogen species (ROS/RNS), such vioral impairments, which are detected in Hg-exposed
as hydrogen peroxide (H2O2), the superoxide humans, were also observed in rodent-based models
anion (O2-), and nitrous oxide (NO−) (Farina, following in vivo exposures (Dietrich et al. 2005; Haut
Aschner, and Rocha 2011; Grotto et al. 2010; et al. 1999; Moreira et al. 2012; Watanabe et al., 1999b;
Lund, Miller, and Woods 1991, 1993). Increased Zimmermann et al. 2014). Notably, these behavioral
ROS production enhances lipid peroxidation changes were also noted after exposures to xenobiotics
(LPO), which has been widely used as an indicator other that Hg compounds, thus not enabling for spe-
of Hg-mediated oxidative stress (Huang et al. cific use as biomarkers of Hg-associated effects.
2011; Kong et al. 2012; Mahboob et al. 2001; Nonetheless, neurobehavioral alteration may be
Suda and Takahashi 1992). However, many com- employed as tools for quantifying the extent of toxicity
pounds in the environment increase ROS produc- (mainly neurotoxicity) in subjects knowingly exposed
tion including pesticides, cadmium, and PAHs to to Hg compounds.
name a few. Thus, analogous to GSH, these bio-
chemical changes are not ideal markers of selective Renal dysfunction
Hg-induced toxicity in a multi-contaminant sce- The kidneys are the main organs of Hg accumula-
nario due to the lack of specificity. tion following exposure to Hg0 and Hg2+, and all
mercuric forms produce nephrotoxicity (Bridges
and Zalups 2010; Zalups 2000). The primary site
Biomarkers of effect
of Hg deposition in the kidney is the proximal
Neurobehavioral alterations tubule with segments S2 and S3 being affected
In order to correlate exposure to mercurials with the first (Bridges and Zalups 2010). Unless exposure
development of neurotoxic effects, many studies to Hg compounds occurs acutely, in which case
relied on clinical observations of subtle alterations kidneys accumulate the metal rapidly (Zalups and
in neuromotor, neuropsychological, and neurophy- Barfuss 2002b) alterations in renal function often
siological functions (Bose-O’Reilly et al. 2010). These remain clinically undetected (Bernard and
include assessment of various endpoints such as Lauwerys 1989). Thus, the need arises for reliable
language skills, attention deficit, memory, hand–eye biomarkers capable of detecting changes in kidney
coordination, reaction time, IQ tests, and neurologi- function prior to their irreversible damage.
cal and cardiac function (Bose-O’Reilly et al. 2010; Early toxicity to the proximal tubule is identified by
Cordier et al. 2002; Dahl et al. 1996, 1999; Grandjean measuring the activity in urinary enzyme activities
et al. 1997; Lebel et al. 1998, 2003; Myers et al. 1997). from the luminal brush border of the proximal tubule,
In toddlers, frequently assessed endpoints include such as γ-glutamiltransferase (γ -GT) (Diamond and
the age-related achievement of developmental mile- Zalups 1998). As toxicity and cell death progress,
stones such as sitting, standing up, walking, and intracellular enzymes such as lactate dehydrogenase
talking (Dórea, Marques, and Abreu 2014; (LDH), N-acetyl-β-D-glucosaminidase (NAG), and
Grandjean, Weihe, and White 1995; Myers et al. β-galactosidase are found in urine (Bernard and
1997). Lauwerys 1989). Levels of α1, β2 microglobulins, and
The strength of association between Hg exposure retinol-binding protein (RBP) also represent sensitive
and neurobehavioral alterations varies according to markers of tubular dysfunction (Franko, Budihna,
the endpoint analyzed (Wigle et al. 2008), which cor- and Dodic-Fikfak 2005). Kidney injury molecule-1
relates with the fact that inherent to this type of end- (Kim-1) has also been associated with Hg-mediated
points is the occurrence of confounding factors that nephrotoxicity in animal studies (Wang et al. 2015;
make interpretation of results difficult (Grandjean, Zalups, Joshee, and Bridges 2014; Zhang et al. 2008;
Weihe, and White 1995). In addition, these alterations Zhou et al. 2008) and is claimed to allow detection of
may represent either reversible effects or already early tubular damage (Zhou et al. 2008). Neutrophile
established manifestations of toxicity; thus, for risk gelatinase-associated lipocalin (Ngal) was utilized as a
prevention, it is imperative to identify biomarkers predictor of early metal nephrotoxicity (Zalups,
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B 11

Joshee, and Bridges 2014). Altered glomerular kidney correlation was noted between Hg and α1 and β2-
function is often assessed by measuring urinary levels microglobulin, RBP, or albumin levels. In addition, in
of high-molecular proteins, such as albumin, IgG, and a study analyzing renal function markers in a popula-
transferrin (Bernard and Lauwerys 1989) or by evalu- tion with low exposure to different metals, including
ating glomerular filtration rate (eGFR) by measuring Hg (median urine levels = 1.4 μg Hg/g creatinine), no
plasma creatinine or blood urea nitrogen (Zalups, marked correlation between urinary Hg with eGFR
Joshee, and Bridges 2014). and Kim-1 (Callan et al. 2015) was noted. Assessment
Several studies in humans used different combi- of the relationship between Hg exposure (blood Hg)
nations of these biomarkers to establish a connec- and renal function in the US population (aged over
tion between nephrotoxic effects and Hg exposure. 40 years) by Lin et al. (2014) showed no significant
In chloroalkali workers exposed to Hg (U-Hg association between exposure (<10 μg Hg/L) and
range: 1.1–124 μg/g creatinine), a significant albuminuria. Nevertheless, Hg blood levels correlated
increase in NAG activity was observed as well as negatively with eGFR. A prospective cohort examin-
in α1-microglobulin excretion (U-Hg > 35 μg/g ing premenopausal women with low-level exposure to
creatinine) (Jarosínska et al. 2008).The same metals reported that a twofold elevation in Hg was
study reported elevated albumin levels in urine, associated with increased protein levels in serum,
indicating altered glomerular function. Similarly, whereas alkaline phosphatase was reduced (Pollack
workers exposed to Hg0 with U-Hg above 50 μg et al. 2015). The significance of these biomarker
Hg/g creatinine showed signs of glomerular pro- responses to low exposure levels findings is unclear;
teinuria as evidenced by enhanced excretion of however, it is evident that the abundance of markers
albumin, IgG, and transferrin (Bernard and of renal function allowing for identification of early
Lauwerys 1989). In miners, previously exposed to nephrotoxicity effects contrasts with the absence of
Hg (average U-Hg: 68.24 μg/L), glomerular and measurable biochemical endpoints predictive of early
tubular dysfunction was noted with elevated excre- neurotoxic effects.
tion of albumin, IgG, and α1-microglobulin, sug-
gesting that nephrotoxic effects remain long after
Indicators of susceptibility
cessation of exposure (Franko, Budihna, and
Dodic-Fikfak 2005). Common indicators of susceptibility to Hg include life
In nonoccupationally exposed subjects, where stage, socioeconomic and geographic situation, fish
excretion of U-Hg is considerably lower, the response consumption habits, and occupation (World Health
observed in renal function markers is unclear. In Organization/United Nations Environment
subjects living near chloroalkali plants, no significant Programme (WHO/UNEP) 2008). The in utero devel-
association between U-Hg (median 1.2 μg Hg/g crea- oping fetus and first stages of infancy are the life stages
tinine) and renal markers (α1-microglobulin, NAG, most sensitive to mercurials (Clarkson and Magos
albumin) was observed (Jarosínska et al. 2008). In 2006), and thus, these population groups constitute
contrast, in individuals residing in the vicinity of a the core target of Hg risk assessment (Carvalho et al.
ASSGM area in China (average 1.24 μg Hg/L urine), 2008b, 2004; Grandjean et al. 1992, 1997; Myers et al.
concentrations of β2 microglobulin were increased 2003; Nunes, Cavaco, and Carvalho 2014a, 2014b).
relative to control subjects (Tian et al. 2009). Further, the constraints imposed by geographical
The effect of Hg0 release from dental amalgams on and socioeconomic context (Burger et al. 1999;
kidney function has also been addressed in several Carvalho et al. 2008b) of a population, such as food
studies. NAG activity in urine demonstrated signifi- habits (high fish consumption) or occupation
cant positive correlation with Hg0 release from dental (ASSGM workers) might alter vulnerability to Hg-
amalgam fillings in children from Saudi Arabia (aver- mediated toxicity and have been used in epidemiolo-
age 2.75 μg Hg/g creatinine) (Al-Saleh, Al-Sedairi, and gical studies as indicators of susceptibility (Bose-
Elkhatib 2012). However, in a similar study in US O’Reilly 2008; Carvalho et al. 2008b; Cordier et al.
children, no marked effect of Hg0 was found with 2002; Grandjean et al., 2004).
NAG activity in urine (Barregard, Trachtenberg, and Research linking genetic polymorphisms to sus-
McKinlay 2008). In both studies, no significant ceptibility to mercuric compounds has brought a
12 V. BRANCO ET AL.

new perspective into identifying vulnerable popula- biomarkers is particularly important to enable predic-
tion subgroups. These genetic factors are subse- tion of adverse effects to overcome insufficiencies in
quently discussed. risk assessment analysis, particularly inter-individual
variations in sensitivity to Hg compounds.
Assessment of Hg-induced nephrotoxicity relies
New perspectives: Mechanistic-based on biomarkers present in urine that reflect directly
biomarkers early target organ (kidney) toxicity. The equivalent
in neurotoxicity evaluation would be to analyze
New biomarkers of effect
biomarkers in the cerebrospinal fluid, which is
As previously mentioned, biomonitoring of Hg not feasible for routine analysis. Thus, the major
exposure is predominantly based upon quantita- challenge in Hg-mediated toxicity is to develop
tive analyses of Hg in tissues and biological fluids biomarkers that are predictive of neurotoxic
such as urine, nails, blood, and hair. In addition, events, similar to those existing for nephrotoxicity.
there are specific proteins (enzymes, transporters
and transcriptional factors) and signaling path- Glutamine/glutamate changes
ways that have been linked to exposure to Hg Mercuric compounds affect multiple neurotransmit-
compounds. Albeit, there is no consensus whether ter systems. Neurotoxic effects associated with MeHg
these proteins and pathways might be adequate to were shown to involve the glutamate/glutamine cycle.
quantify the extent of toxicity in Hg-exposed sub- Glutamate is the predominant excitatory neurotrans-
jects, some of these “endpoints” may potentially be mitter in the CNS, but in excess is a neurotoxin. For
useful as biomarkers. this reason, glutamate is actively removed from the
Conjugation of mercury compounds with cysteine synapse. While both neurons and astrocytes take up
(Cys)-containing compounds and Se (e.g., SeCys-con- glutamate, the majority is removed by astrocytes.
taining proteins) may lead to toxicity. Our knowledge Excitatory amino acid carrier 1 (EAAT3), excitatory
regarding metal speciation in relation to binding to amino acid transporter 4 and 5 (EAAT4 and EAAT5),
both low-molecular mass molecules containing sele- glutamate/aspartate transporter (GLAST), and gluta-
nol and thiol groups or high-molecular mass proteins mate transporter 1 (GLT1) are implicated in the
is limited. The identification of single- or, preferen- removal of glutamate from the synapse by astrocytes
tially, multi-targets of Hg that might represent early (GLT and GLAST) and neurons (EAAT3, EAAT4,
markers of effect to low levels of mercurial exposure is and EAAT5). MeHg preferentially accumulates in
highly desirable, yet a difficult task to achieve. astrocytes and increases extracellular glutamate levels
“Proteomic profiling” has recently emerged as a strat- by reducing glutamate uptake by GLT and GLAST
egy capable of identifying early targets of Hg that may (Aschner et al. 1993; Brookes 1992; Qu et al. 2003). In
potentially be employed as new biomarkers (De mice exposed to MeHg through milk, there was nearly
Oliveira Souza et al. 2016) albeit this toxicoproteomic 50% reduction in glutamate uptake in cerebellar slices
approach remains incipient (Merrick 2006). Thus, compared to controls (Manfroi et al. 2004). Increased
future studies need to be conducted in experimental levels of synaptic glutamate resulted in N-methyl-D-
models of intoxication with mercurials encompassing aspartate (NMDA) receptor-dependent excitotoxicity.
Hg speciation in plasma, blood, and target tissues. Park et al. (1996) found that blockade of the NMDA
This requires advances in high throughput accurate receptor with antagonists prevented death in cultured
mass and time (AMT) proteomic analytical techni- cerebral neurons. In addition, MeHg primes neurons
ques, mass spectroscopy techniques (MS), particularly for excitotoxic death by enhancing expression of
inductively coupled plasma-MS (ICP-MS), MALDI NMDA receptor NR-2B subunit in the hippocampus
and ESI-MS to identify proteins covalently bound to of exposed rats (Baraldi et al. 2002).
Hg. It is noteworthy that micro-liquid chromatogra- Glutamate released from neurons is normally
phy (μLC)-ICP-MS, ESI-MS, and MALDI-MS tech- taken up by astrocytes, where it is converted into
niques were utilized to quantify the labeling of glutamine to be transported back to the neuronal
ovalbumin by para-hydroxy-mercurybenzoate terminal. In addition to inhibiting uptake of gluta-
(Kutscher et al. 2008). The development of new mate, Hg compounds disrupt glutamine synthesis
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B 13

and transport. All Hg compounds were reported to et al. 1998; El-Fawal et al. 1996; Gao et al. 2006).
decrease glutamine synthetase (GS) activity and Increased GFAP following MeHg exposure was also
inhibit glutamine transport in cultured astrocytes observed in zebrafish (Cambier et al. 2012), suggest-
(Allen, Mutkus, and Aschner 2001a; Yin et al. ing its use as a biomarker for analyzing wild-life
2007) and in rat brain (De Oliveira Souza et al. specimens. It is noteworthy that exposure to orga-
2016). Further, MeHg inhibits GS activity in brains nophosphates or neoniocotinoid also results in
of rats, mice, and sheep, where elevated blood levels enhanced GFAP staining in humans rats (Abou-
of MeHg correlated with reduced GS activity Donia et al. 2008, 2013). In summary, the use of
(Kwon and Park 2003). Both MeHg and HgCl2 GFAP as a marker of Hg exposure in humans lacks
were found to inhibit the glutamine/amino acid sensitivity, selectivity, or predictability as a biological
transporter (ASCT2), which shuttles glutamine indicator of Hg exposure per se.
into neurons from astrocytes (Oppedisano,
Galluccio, and Indiveri 2010). Changes in the activity of enzymatic systems
In summary, the use of glutamatergic transmis- As previously mentioned, many critical enzymes
sion as a marker of Hg exposure in humans is including those involved in antioxidant defense rely
complicated due to lack of sensitivity, selectivity, or on the presence of reactive thiol and/or selenol
predictability as a biological indicator of Hg expo- groups in the active site. Binding of mercurials to
sure per se. The presence of NMDA-like receptors these groups impairs their catalytic activity and leads
and high levels of glutamate in platelets (1998; to toxicity. Enzymes containing reactive thiol and/or
Franconi et al. 1996; Kalev-Zylinska et al. 2014) selenol groups are potential targets for Hg com-
indicates that NMDA-mediated responses do not pounds and thus may constitute useful biomarkers.
necessarily reflect exposure to Hg, as these receptors Notably, selenols (SeH) have generally higher
are modified by a myriad of factors including nucleophilicity than thiols and are present in mam-
genetic, epigenetic metabolic, and external stressors malian cells as selenocysteine (Sec) in the active site of
including other metals as well as age and gender. selenoproteins catalyzing redox reactions (Hatfield
et al. 2014; Lu and Holmgren 2009). The selenol
Glial fibrillary acidic protein group is generally more reactive than thiols (lower
Injury to the CNS by multiple stimuli, including Hg pKa) toward electrophilic forms of Hg (Sugiura et al.
compounds, results in reactive gliosis, a condition 1976). In addition, abundance of selenoproteins is
where there is hypertrophy and activation of glial lower. Accordingly, selenoproteins represent impor-
cells, particularly astrocytes. Glial fibrillary acidic tant (and relatively “more specific”) targets for Hg-
protein (GFAP) is one of many intermediate fila- mediated toxicity. Among the enzymes known to be
ment proteins that are upregulated in glial cells dur- altered by exposure to Hg compounds, one needs to
ing reactive gliosis. Exposure to noncytotoxic consider ROS scavenging enzymatic systems, such as
concentrations of MeHg increased the expression superoxide dismutase (SOD), catalase (CAT)and glu-
of GFAP in both primary rat astrocytes and micro- tathione peroxidase (GPx), and redox active proteins
glia in vitro, which correlated with decreased gluta- from the thioredoxin (Trx) system.
mate and serotonin reuptake by astrocytes and
enhanced IL-6 release from microglia (Dave et al. Catalase and superoxide dismutase
1994; Eskes et al. 2002). Interestingly, Jebbett et al. Changes in CAT and SOD activities were proposed
(2013) found that MeHg increased ciliary neuro- as biomarkers of Hg-induced oxidative stress.
trophic factor (CNTF)-induced expression of However, reported findings are inconsistent regard-
STAT3-target genes, including GFAP, in neural pro- ing the effect of mercurials on the activity of these
genitor cells enabling glial differentiation. enzymes in vivo. In workers exposed to Hg0, Perrin-
Data suggest that developmental exposure to Nadif et al. (1996) observed a rise in CAT activity in
MeHg may affect proper glial differentiation in plasma similar to the observations by Kobal et al.
vivo. In rat pups exposed to MeHg in utero, elevated (2004, 2008) comparing miners with past exposure
levels of GFAP were detected in the corpus callosum, to Hg (U-Hg: 2.1 μg/L) and control subjects (U-Hg:
brain stem, cerebellum, and hippocampus (Barone 1.4 μg/L). On the other hand, Pinheiro et al. (2008)
14 V. BRANCO ET AL.

found a negative association between blood CAT activity was a consequence of direct inhibitory
activity and Hg exposure as seen in hair levels in effects, which were probably related to Hg-selenol
women living in the Amazon. Similarly, Carneiro interactions. Interestingly, these events were
et al. (2014) observed a negative association responsible for enhanced susceptibility to perox-
between plasma MeHg and CAT activity. These ides, enhanced lipid peroxidation, and neuronal
contradictory results are also inherent in animal death (Farina et al. 2009). In agreement, another
studies. In rats, Nath et al. (1996) and more recently study established that GPx also serves as a crucial
Silva De Paula et al. (2016) noted no significant molecule involved with MeHg-induced neurotoxi-
changes in CAT activity following exposure to Hg city (Franco et al. 2009).
compounds, whereas Kong et al. (2012) reported a Zemolin et al. (2012) showed that the protein levels
decrease in activity levels during the embryonic of GPx4, an important isoform of GPx family whose
development of a fish model. activity is crucial to detoxify lipid peroxides in cell
The lack of a clear pattern was also demonstrated membranes, were decreased in encephalic structures
for SOD activity levels following exposure to Hg of MeHg-exposed mice. Thus, in addition to direct
(Hussain et al. 1997; Perrin-Nadif et al. 1996). A interaction between MeHg and the selenol group of
numerical decrease in erythrocyte SOD activity (5%) GPx, the fall in GPx expression also contributes to loss
was observed in workers exposed to Hg0 (U-Hg: of activity. In agreement, cultured cells exposed to
77.44 ± 48.15 μg/L) (Zabiński et al. 2000). In Hg0 MeHg demonstrated a “selenium-deficient-like” con-
exposed dental personnel (B-Hg: 7.74 ± 1.03 μg/L vs. dition, which affects GPx1 synthesis through a post-
4.79 ± 0.84 μg/L in control subjects), Samir and Aref transcriptional action (Usuki, Yamashita, and
(2011) reported that erythrocyte SOD activity was Fujimura 2011) (Figure 5). Collectively, these effects
reduced by 40% and that this decrease was inversely indicate that the selenoenzyme GPx is an important
correlated with the number of working years. Kobal molecular target of MeHg-induced neurotoxicity.
et al. (2004) compared RBC SOD activity between Nevertheless, studies in humans are less clear. In den-
former Hg miners (U-Hg: 2.1 μg/L) and control sub- tal workers exposed to Hg0 (B-Hg: 7.74 ± 1.03 μg/L vs.
jects (U-Hg: 1.4 μg/L) and noted no significant 4.79 ± 0.84 μg/L in control subjects), a negative corre-
difference. lation was found between Hg exposure and GPx activ-
In fish exposed to MeHg, no marked effect on ity (40% reduction) in erythrocytes (Samir and Aref
SOD activity was found in liver and brain (Branco 2011). However, for higher exposure levels (average
et al. 2011), whereas in brain of rats exposed to B-Hg: 79.1 μg/L), Jayaprakash (2009) reported a
MeHg, a significant increase in activity was reduction in only 30% in RBC GPx activity.
observed (De Oliveira Souza et al. 2016). In the Interestingly, Kobal et al. (2008) found no marked
blood of rats chronically exposed to MeHg, SOD differences in RBC GPx activity between former Hg
activity remained unchanged relative to controls miners (average U-Hg: 2.53 μg/g creatinine) and con-
(Silva De Paula et al. 2016) trol subjects (average U-Hg: 1.31 μg/g creatinine),
even though Se excretion was higher in miners as
Glutathione peroxidase evidenced by lower plasma and higher urinary Se
Selenols are present in the active site of selenopro- (Kobal et al. 2004). In contrast, in Chinese workers,
teins catalyzing redox reactions (Hatfield et al. Hg0 exposure (U-Hg: 86.8 ± 65.2 vs 1.25 ± 1.5 μg/L)
2014; Lu and Holmgren 2009) and are preferential was associated with higher (+13%) plasma activity of
targets for Hg-mediated toxicity. An experimental GPx (Chen et al. 2006). In addition, in sports fisher-
in vitro study with primary cultures of cerebellar men consuming Hg via dietary fish, a positive effect of
neurons showed that the activity of the selenoen- exposure was seen in the activity of blood GPx
zyme glutathione peroxidase (isoform 1; GPx1), (+10%), albeit with only numerical rise in blood Se
was decreased by low-dose MeHg (Farina et al. (Bélanger et al. 2008), whereas in a fish-eating popula-
2009), prior to changes in several biochemical tion from the Brazilian Amazon, a negative association
parameters that are normally affected by high- between plasma MeHg and GPx activity was detected
dose MeHg exposures. The diminished enzyme (Carneiro, Grotto, and Barbosa 2014).
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B 15

Thioredoxin and thioredoxin reductase showing a rapid inhibition of TrxR by mercurials.


Carvalho et al. (2008a, 2011) showed in vitro that Interestingly, the activity of glutathione reductase
mercurials target the enzymes of the thioredoxin (GR) was not affected by mercurials in HeLa cells,
system—thioredoxin reductase (TrxR) and thiore- reinforcing the fact that the Sec residue in the
doxin (Trx)—binding to their active site thus C-terminus of TrxR is a target for Hg (Carvalho
decreasing activity. The thioredoxin system is et al. 2008a). Experiments in a fish model (Branco
found in all tissues and involved in multiple func- et al. 2011, 2012a) further corroborated the impor-
tions associated with cellular maintenance and sur- tance of targeting the Trx system and in particular
vival including protein repair, regulation of the cell TrxR as a key step in mediating Hg toxicity. This fact
cycle, and cell signaling (Lillig and Holmgren 2007). was further supported by the findings that the activ-
Since the Trx system is upstream of several biochem- ities of GR and GPx1 were less sensitive to Hg com-
ical pathways, its inhibition by mercurials represents pounds (Branco et al. 2011, 2012a). Experiments with
a key step in mediation of Hg-induced toxicity rodents also demonstrated that TrxR activity is mod-
(Carvalho et al. 2008a). ified by MeHg administration; however, the mangni-
Several compounds are known to produce inhibi- tude of the effects varied depending upon the schedule
tion of both enzymes of the Trx system, with TrxR of intoxication, the species, developmental period,
being particularly prone to interaction with electro- animal gender, and subcellular fraction considered
philic agents due to the presence of selenol (seleno- (Dalla Corte et al. 2013; Ruszkiewicz et al. 2016;
late at physiological pH) in the open C-terminus of Wagner et al. 2010). Trx was also reported to be
its active site (Arnér 2009). As depicted in Table 1, affected by mercurials in cells and animal models
many of the compounds known to target TrxR but not as severely as TrxR (Branco et al. 2011,
include soft metals, such as gold and platinum (e.g., 2012a; Carvalho et al. 2008a; Ruszkiewicz et al. 2016).
auranofin and cisplatin) (Mustacich and Powis 2000; Results in human THP1 monocytic cells exposed
Rigobello et al. 2004; Witte et al. 2005) cadmium, to Hg2+ suggested that Hg induced transcription of
arsenic and zinc (Lu, Chew, and Holmgren 2007; the enzymes of the Trx system, via an increase in
Rigobello et al. 2004). nuclear factor (erythroid-derived 2)-like 2(Nrf2)
It is noteworthy that Hg compounds, especially levels (Wataha et al. 2008). Recently, it was shown
Hg2+, exhibit an inhibitory capacity against purified that induction of cytosolic thioredoxin reductase
TrxR1 which is remarkable among metallic com- (TrxR1) via Nrf-2 is faster during exposure of liver
pounds (Table 1). Further, the inhibitory effect of cells to Hg2+—leading to elevated mRNA and
mercurial compounds is particularly fast, with com- enzyme expression—than to MeHg, which may
plete blockade of TrxR activity after only 5-min explain the higher cytotoxicity of this compound
incubation (Carvalho et al. 2008a). These results (Branco et al. 2014). Most importantly, mitochon-
clearly show that TrxR is highly susceptible to inhi- drial thioredoxin reductase (TrxR2) is not regulated
bition by mercurials (Carvalho et al. 2008a, 2011). In by Nrf-2, and its activity is markedly affected by both
addition, it is significant that Hg2+ displays greater Hg2+ and MeHg, suggesting its importance as a
potency as an inhibitor of TrxR, suggesting that target for Hg-associated toxicity (Branco et al.
MeHg upon demethylation produces an even greater 2014) (Figure 6). The targeting of the mitochondria
toxic outcome (Carvalho et al. 2008a). Trx activity and the effects of Hg in the overall function of this
was also found to decrease upon exposure to Hg in organelle may therefore constitute a promising field
vitro, with both Hg2+ and MeHg targeting the dithiol of research in the discovery of additional novel
at the active site of Trx leading to loss of activity, biomarkers.
albeit with higher IC50 than the one observed for Overall, results to date indicate that both TrxR
TrxR (Carvalho et al. 2008a). and Trx are important targets for mercurials, and
Mercurials also affect TrxR in HeLa and HEK293 their inhibition is likely upstream of the develop-
cells in a time- and concentration-dependent manner ment of Hg-initiated toxic outcomes (Branco et al.
(Carvalho et al. 2008a). Inhibition of TrxR was 2012b). Accordingly, evaluation of TrxR and Trx
detected after only 7-hr exposure (Carvalho et al. in accessible matrices, such as blood and plasma,
2008a), correlating well with observations in vitro may provide a useful tool for the prediction of Hg-
16 V. BRANCO ET AL.

Table 1. Comparison of IC50 values for the inhibition of purified TrxR by different metals
Compound [TrxR] (nM) IC50 (µM) incubation time (min) Reference
Mercury HgCl2 50 0.0072 5 Carvalho et al. 2008a
MeHgCl 50 0.0197 5
Gold (I) Auranofin 2 0.020 10 Rigobello et al. 2004
TEPAu 2 0.065 10
Aurothiomalate 2 0.280 10
Gold (III) Aupy 2 1.42 10
Audien 2 0.42 10
Aubipy 2 0.28 10
Aubipyxil 2 0.21 10
Tin Tributyltin 2 76.1 10 Rigobello et al. 2004
Cadmium Cadmium acetate 2 23.5 10
Zinc ZincAcetate 2 19.5 10
Zincpyrithione 2 11.8 10
Platinum Cisplatin 300 ~200 20 Witte et al. 2005
Oxaliplatin 300 ~100 20
Silver Ag2SO4 25 ~0.025/5 5 Srivastava, Singh, and Self 2012
Arsenic As2O3 50 0.25/30 30 Lu, Chew, and Holmgren 2007

Mercury Compounds

1A
1B

TrxR1

2
TrxR2 7

Nrf-2 Keap-1

3
Mitochondria
TrxR1mRNA Keap-1

6
4

Cytosol

Nrf-2 5
TXNRD1 gene ARE

Nucleus

Figure 6. Interaction between mercury compounds and TrxR1 and TrxR2 at the cytosol and mitochondria. Mercury compounds enter
liver cells and target TrxR1 and 2 at the cytosol and mitochondria, respectively (1A and 1B), leading to a decrease in activity. Loss of
TrxR1 promotes Nrf-2 release from Keap-1 (2 and 3) and its subsequent translocation to the nucleus of the cell (4) where it binds the
ARE element in the promoter region of the TXNRD1 gene (5). This process is faster upon exposure to Hg2+ than to MeHg (see Branco
et al. 2014 for detailed explanation). Transcription of the gene is enhanced and TrxR1 mRNA builds up in the cytosol (6), and TrxR1 is
de novo synthetized (7)

induced adverse effects, but to our best knowledge, et al. 2006; Hagmar et al. 1998); however, the status
this possibility has not been addressed in human of Se may be critical in limiting the toxicity of Hg
subjects. (Farina, Aschner, and Rocha 2011; Khan and Wang
Only few studies evaluated changes in the activity 2009; Watanabe 2002). Consequently, the use of
of selenoenzymes in humans exposed to Hg (Chen selenoenzymes and/or selenoproteins as biomarkers
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B 17

of effect for Hg might require concomitant Se status toxicogenomics studies (Hwang et al. 2011a;
determination. Robinson et al. 2011; Theunissen et al. 2011;
Yadetie et al. 2013). Most of these studies were
Pentose phosphate pathway dehydrogenases performed using cell lines; however, their applica-
NADPH is mostly generated by the thiol-contain- tion to in vivo exposures is relevant as demon-
ing enzymes glucose-6-phosphate dehydrogenase strated by studies investigating the roles of the
(G6PDH) and 6-phosphogluconate dehydrogenase transcription factors nuclear factor (erythroid-
(6PGDH) of the pentose phosphate pathway derived 2)-like 2(Nrf2) and nuclear factor kappa-
(PPP). NADPH functions as one of the main light-chain enhancer of activated B cells (NF-κB)
intracellular reducing agents and an essential as discussed below.
cofactor required for the normal function of anti-
oxidant cycles, such as GSH and Trx systems Nrf2
(Horecker 2002). Targeting of these enzymes by Nrf2 is a basic leucine zipper transcription factor
Hg compounds was reported for several mercur- that responds to oxidative stress. Under basal con-
ials in HepG2 and SH-SY5Y cells with inhibition ditions, Nrf2 resides in the cytoplasm bound by
intensity of Hg2+>MeHg≈EtHg>Thimerosal (Rod- kelch-like ECH-associated protein1 (Keap1).
rigues et al. 2015). EtHg and MeHg produced Keap1 is associated with Cullin 3, an E3 ubiquitin
equivalent and significant inhibition of G6PDH ligase, which targets Nrf2 for proteasomal degra-
activity and to a lesser extent 6PGDH, especially dation (Baird and Dinkova-Kostova 2011).
in neuroblastoma cells. In MeHg-incubated fibro- Oxidative stress in general, including exposure to
blasts, Amoli et al. (2011) noted that a decrease in MeHg, enhances Nrf2 to be released from Keap1
the activity of the oxidative branch of PPP impairs by disruption of cysteine residues on Keap1 or
ribose synthesis, which is essential for nucleotide through oxidant-dependent kinase signaling.
production, causing DNA damage. Interestingly, Unbound Nrf2 translocates into the nucleus,
Zabiński et al. (2000) observed that in workers where it can bind to antioxidant response element
exposed to Hg0 (U-Hg: 77.44 ± 48.15 μg Hg/L), (ARE) promoters as a heterodimer with Maf pro-
G6PDH activity in erythrocytes was reduced by teins (Baird and Dinkova-Kostova 2011). Nrf2
25% relative to nonexposed controls. induces the expression of several cytoprotective
During the last decade, evidence has been accu- proteins, including NAD(P)H quinine oxidoreduc-
mulating concerning the role of selenoenzymes, tase (NQO1), glutamate-cysteine ligase catalytic
namely TrxR and GPx, as targets for Hg com- subunit and modifier subunit (GCLC and
pounds. The importance of these enzymes as well GCLM), and heme oxygenase-1 (HO-1) (Baird
as of Trx and GSH for cellular function and home- and Dinkova-Kostova 2011). Nrf-2 also enhances
ostasis stresses the importance of a more compre- transcription of the TXNRD1 gene, increasing
hensive understanding of their role in Hg- TrxR1 mRNA build up in the cytosol and TrxR1
associated toxicity. It is worthwhile noting that it de novo synthesis. In HepG2 cells, Branco et al.
is possible to measure in plasma the activity of (2014) demonstrated that this process is faster
these enzymes, and thus, it is necessary to deter- upon exposure to Hg+2 than to MeHg (Figure 6).
mine whether activity of selenoenzymes in blood In addition to redox regulation, Nrf2 is also regu-
reflects Hg exposure and whether enzymic activity lated by protein kinase pathways (Sherratt et al. 2004).
is affected in brain. In the case of the thioredoxin Nrf2 interacts with p38 mitogen-activated protein
system (TrxR and Trx), since their inhibition is an kinase (Alam et al. 2000; Yu et al. 2000), ER-resident
early event in Hg-mediated toxicity, this system kinase PERK (Kobayashi et al. 2006), and a Src family
may serve as a potential predictive biomarker of tyrosine kinase, Fyn (Jain and Jaiswal 2006). The
toxic effect. PI3K/Akt pathway controls Nrf2’s function upstream
of Fyn (Niture, Khatri, and Jaiswal 2014). Activation
Transcription factors of Nrf2 by MeHg was noted in cell lines and primary
Altered gene transcription following MeHg expo- cells allowing for upregulation of NQO1 and HO-1
sure has been documented in many recent (Ni et al. 2010; Toyama et al. 2010, 2011b; Wang et al.
18 V. BRANCO ET AL.

2009). Further, MeHg induced Nrf2 activity in ultimately phosphorylate IKK, leading to its acti-
rodents and fish (Deng et al. 2014; Toyama et al. vation. Many of these kinases (IKK, PI3K, AKT,
2011a; Yadetie et al. 2013). Interestingly, levels of MEKK-1) are redox sensitive (Pantano et al. 2006),
Cullin 3 were decreased following MeHg exposure in enabling activation by oxidative stressors, such as
SH-SY5Y cells (Toyama et al. 2011b), suggesting that ROS and oxidized lipids. Therefore, both lipid
there is coordinated control to prevent degradation of peroxidation end products and ROS derived from
Nrf2. While oxidative stress derived from MeHg MeHg exposure possess the potential to activate
exposure might activate Nrf2, direct Nrf2 activation NF-κB.
was observed following MeHg binding to recombi- Exposure to MeHg leads to increased markers of
nant Keap1 and by MeHg-GSH adducts (Toyama inflammation mediated by NF-κB gene transcrip-
et al. 2007; Yoshida, Abiko, and Kumagai 2014). tion. MeHg exposure enhanced NF-κB activation as
Kinase regulation of MeHg-induced Nrf2 activation measured by elevated levels of the RelA/p65 subunit
has yet to be demonstrated, although microarray stu- and DNA bound NF-κB, in both cerebrum and
dies of Atlantic cod found upregulation of MAP cerebellum of rats and mice (Dong et al. 2001;
kinase MEK4 by MeHg, which potentially may acti- Hwang et al. 2011). This leads to increased expres-
vate Nrf2 by JNK activation (Yadetie et al. 2013). Nrf2 sion of chemokines CCL2 and CCL4 and anti-apop-
was shown to be protective in a Caenorhabditis elegans tosis protein Bcl2a1b (Hwang et al. 2011; Lee et al.
(C. elegans) model of dopaminergic neurodegenera- 2012). In vitro MeHg elevated both CCL2 and IL-6
tion induced by MeHg, where reduction in SKN-1, the release from glial cells (Chang 2007; Kim et al. 2012).
Nrf2 homologue, increases the vulnerability for neu- Even though there is sufficient evidence reporting
rodegeneration following either an acute or a chronic a relationship between Hg exposure and changes in
exposure to this compound (Martinez-Finley et al. Nrf2 and NF-κB pathways, most of the available data
2013; Vanduyn et al. 2010). Knockdown of Nrf2 in are derived from experimental (mainly in vitro) stu-
worms displays a elevated levels of ROS and increased dies. Further, these responses lack specificity and are
measures of dopaminergic cell dysfunction highly pleiotropic which diminishes their value as
(Martinez-Finley et al. 2013). Activation of Nrf2 by biomarkers of Hg-induced toxicity. Thus, it is diffi-
MeHg represents a protective response; however, cult to predict the usefulness of these transcription
MeHg often alters gene expression resulting in neu- factors for employment as human biomarkers. In
rotoxic effects. summary, due to the numerous stimuli activating
Nrf2 and NF-kB, these are not specific biomarkers
NF-κB for Hg toxicity.
NF-κB is a transcription factor that controls the
regulation of inflammatory cytokines (IL-6 and IL-
8) and chemokines (CCL2 and CCL4), as well as Immunological outcomes
anti-apoptotic proteins (Bcl2, TRAF1 and TRAF2). Immunotoxicity due to mercurial compounds has
NF-κB consists of a heterodimer of a class I NF-κB been described in a variety of animal species,
protein (p50 or p52) and a class II NF-κB protein including humans (Sweet and Zelikoff 2001).
(p65/RelA, RelB, or c-Rel) (Hoesel and Schmid Immunotoxic effects arise at Hg concentrations
2013). Combinations of the subunits vary by cell that are below those reported to damage the CNS
type and development stage. NF-κB is bound in and kidneys (Vas and Monestier 2008) and, con-
the cytoplasm by the inhibitor IκB where it is sequently, may be considered as a critical effect for
sequestered from activating transcription. When the development of toxicity. The mercuric ion
stimulated, IκB kinase (IKK) phosphorylates IκB, exerts a general stimulatory effect on the immune
targeting IκB for proteasomal degradation, leaving system, and several studies in rodents (Fournie
NF-κB free to translocate to the nucleus and initi- et al. 2001; Nielsen and Hultman 2002; Pollard
ate gene transcription. There are several stimuli and Hultman 1997) demonstrated the ability of
that result in activation of NF-κB. Binding of Hg2+ to promote autoimmunity, which is primar-
cytokines or bacterial or viral antigens to cell sur- ily mediated via NF-kB signaling (Pollard and
face receptors activates kinase cascades, which Kono 2013).
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B 19

In vitro results suggest that MeHg exerts a similar Other biological responses to mercury
effect on the immune system as Hg2+ with induction
Protein adducts
of pro-inflammatory cytokines. However, in vivo,
the immune response to MeHg appears to be more Protein adducts may occur in response to Hg
complex, since it acts initially as an immunosuppres- exposure by two mechanisms. Mercury, given its
sor, but as exposure continues, and it is converted to electrophilicity, directly interacts with protein thiol
Hg2+, a stimulating effect on the immune system and selenol groups leading to a loss in residues
occurs inducing production of autoantibodies and nucleophilic potential. This may result in
increasing serum pro-inflammatory cytokine levels decreased protein activity. As indicated previously,
(Havarinasab and Hultman 2005). MeHg binds the selenol group in GPx1 contribut-
Data in cells indicate that EtHg appears to yield an ing to ROS generation (Farina et al. 2009). MeHg
atypical pattern of pro- and anti-inflammatory cyto- was also found to directly modify cysteine groups
kines (Gardner, Nyland, and Silbergeld 2010b). of delta-aminolevulinate dehydratase (ALAD), an
Dendritic cells exposed to increasing levels of enzyme important for heme biosynthesis (Rocha
Thimerosal showed a reduction in secretion of pro- et al. 1993). ALAD inhibition was reduced in both
inflammatory cytokines associated with GSH deple- brain and liver of suckling rats exposed to MeHg;
tion. Consequently, Th2 activation is enhanced, while however, the enzyme was not inhibited in kidney.
Th1-dependent inflammatory response is reduced There is potential to use ALAD inhibition as a
(Agrawal et al. 2007). In contrast, in H2 mice exposed biomarker for MeHg exposure, as ALAD activity
to Thimerosal, the immune response was skewed assays are commonly performed in blood (Fujita
toward Th1 activation similarly to a typical Hg2+ 2001). However, it is unknown whether there is
exposure (Havarinasab et al., 2007) and resulted in enzymatic inhibition of ALAD in this tissue by
production of antinucleolar antibodies toward a 34- mercury. It should be stressed that measurement
kDa nucleolar protein fibrillarin (Havarinasab et al. of this enzyme is affected by other metals and not
2005). This autoimmune response is significantly a specific indicator of Hg-mediated toxicity.
more potent when mice are exposed to EtHg (as In addition, oxidative stress derived from Hg
Thimerosal) compared to MeHg, which might be exposure may lead to modification of proteins.
attributed to a faster conversion to Hg2+ MeHg induces inhibition of glutamate, cystine
(Havarinasab et al. 2004). and cysteine uptake, thus adversely affecting intra-
Occupational exposure to Hg0 has also been asso- cellular GSH content, and redox status in astro-
ciated with a rise in serum levels of both antinuclear cytes (Alen et al. 2001b, 2001c; Aschner et al. 1993;
(ANA) and antinucleolar (ANoA) autoantibodies Shanker et al. 2001; Shanker and Aschner 2001,
and pro-inflammatory cytokines (IFN-γ, TNF-α 2003). MeHg also stimulates cytosolic phospholi-
and IL-1β) (Gardner et al. 2010a; Motts et al. 2014). pase A2 (cPLA2) resulting in arachidonic acid
Accordingly, in sensitized individuals, exposure to (AA) release from astrocytes, the latter directly
low levels of Hg0 or MeHg from fish deregulates the inhibiting glutamate transporters and glutamate
immune response, altering serum levels of pro- uptake into astrocytes (Aschner 2000; Shanker,
inflammatory cytokines (e.g., IL-1β and TNF-α), Syversen, and Aschner 2003). ROS interact with
and anti-inflammatory cytokines such as IL-4 and cysteine thiol groups on proteins. Nucleophilic Sec
IL-17 (Nyland et al. 2011). and Cys residues readily react with electrophilic
In summary, immune responses to mercurial ROS to form sulfenic (-SOH) or selenenic acids
compounds have not been thoroughly investi- (-SeOH) which are short lived, either reacting with
gated, and knowledge of molecular mechanisms other cysteine residues to form disulfide bonds or
is still elusive. However, a limitation exists in that further oxidized to sulfinic (-SO2H) and sulfonic
these responses occur with numerous compounds acids (-SO3H) by potent oxidants such as super-
and thus could not be reasonably employed as oxide anion (Janssen-Heininger et al. 2008). Both
biomarkers of Hg-mediated toxicity. Nevertheless, sulfinic and sulfonic acid formations are irreversi-
potential for discovery and validation of new bio- ble, and their intracellular accumulation may lead
markers is possible. to altered protein function, protein degradation,
20 V. BRANCO ET AL.

and even cell death. RNS formed from MeHg- amount of lipid peroxides did not change. In addi-
induced activation of nNOS produces nitrosylation tion, LPO has been used for biomonitoring wild-
of amino acids, such as tyrosine (Caito et al. 2014; life exposure to MeHg (Berntssen, Aatland, and
Miyamoto et al. 2001). NMDA, one such protein Handy 2003; Hoffman, Spalding, and Frederick
nitrosylated by MeHg, altered synaptic activity of 2005). In these samples, LPO is typically measured
cortical neurons (Miyamoto et al. 2001). by levels of individual LPO species, such as mal-
Surprisingly, there are no data in humans con- ondialdehyde, or by thiobarbituric acid reactive
cerning this type of interactions with Hg com- species (TBARS) assay (Huang et al. 2011;
pounds. However, these interactions lack Watanabe et al. 1999a). However, the limitation
specificity and thus diminish their utility as bio- to the utilization of LPO as a biomarker for Hg
markers solely for Hg-associated toxicity. exposure lies in the fact that a vast amount of
chemicals act via oxidative stress and consequent
Lipid peroxidation and F(2)-isoprostanes LPO that this measure could not be reasonably
Lipid peroxidation (LPO) is the process by which used to address the issue of mercury-related
reactive species may damage lipid membranes. toxicity.
Mercury-induced lipid peroxidation is mainly F(2)-isoprostanes (F2-IsoPs) are specific LPO
dependent upon the ability of this metal to products nonenzymatically formed from peroxida-
increase, directly or indirectly, the levels of H2O2 tion of arachidonic acid by ROS. Due to their
and superoxide anion. Although the induction of chemical stability and prevalence in all human
lipid peroxidation by H2O2 or superoxide is rela- tissues and biological fluids, F2-IsoPs are regarded
tively low, both molecules may contribute to the as the gold standard for quantification of oxidative
formation of the hydroxyl radical by the Fenton damage in mammals (Morrow et al. 1990).
reaction, allowing for initiation of LPO. Indeed, Exposure to MeHg in vivo and in vitro showed
the chelator deferoxamine is protective against increased levels of F2-IsoPs (Farina et al. 2009; Jin
MeHg-induced lipid peroxidation in rat brain et al. 2012; Stringari et al. 2008; Wormser et al.
(LeBel, Ali, and Bondy 1992), by preventing the 2012). There are few studies that examined the use
Fenton reaction secondary to ROS and biological of F2-IsoPs in human tissues. In a study of pre-
metals, such as iron. Organs such as the brain menopausal women in New York State, no signif-
contain large amounts of polyunsaturated fatty icant relationship was found between MeHg blood
acids, which are highly susceptible to LPO levels (mean of 1.1 μg/l) and F2-IsoPs (Pollack
(Grintal et al. 2009), which seems to occur not et al. 2012). Similarly, a study of Canadian Inuits
only in the cell membrane susceptible to peroxida- noted an inverse correlation between blood Se
tion, but also in intracellular organelles. Damage levels and F2-IsoPs, but no correlation with Hg.
to both lysosomal and the mitochondrial mem- Interestingly, in the same study, Alkazemi et al.
branes by Hg compounds might lead to apoptotic (2013) found a correlation between blood Hg
cell death (Dare et al. 2001; Franco et al. 2010). levels and isofuran (IsoFs) levels. IsoFs are also
Evidence for Hg compounds eliciting LPO has formed by LPO from arachidonic acid under con-
been extensively reported in both cultured cells ditions of elevated oxygen tension that occur dur-
and rodent models (Huang et al. 2011; Lin, ing mitochondrial dysfunction (Fessel and Roberts
Huang, and Huang 1996; Martinez et al. 2014; 2005). Although more research is needed to verify
Silva De Paula et al. 2016; Vendrell et al. 2007; IsoFs as biomarkers for Hg exposure, this para-
Watanabe et al. 1999a; Yin et al. 2007). Several meter would unfortunately lack specificity.
studies demonstrated a link between enhanced
LPO and Hg exposure in humans. Chen et al. Paraoxonase
(2006) analyzed serum samples from miners Paraoxonase-1 (PON1) is an enzyme present on
exposed to Hg0 and found a positive correlation high-density lipoproteins that prevents oxidation of
between MDA levels and exposure. Further, Kobal blood lipids and their deposition in vascular
et al. (2004) found higher urinary levels of MDA endothelium. PON1 is potentially useful as a clinical
in miners exposed to Hg0 even though the total biomarker of cardiovascular risk, as well as a critical
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B 21

enzyme in detoxification of certain organophosphate xenobiotic conjugation reactions, have a well-docu-


oxons. MeHg and other metals are known to inhibit mented link to individual variability in excretion of
PON1 activity in vitro. MeHg was associated with mercuric compounds (Barcelos et al. 2013; Custodio
lowered serum PON1 activity in a fish-eating popu- et al. 2004, 2005; Goodrich et al. 2011; Gundacker
lation, and this maybe related to the MeHg-induced et al. 2009; Lee et al. 2010; Parajuli et al. 2016;
cardiovascular risk (Ginsberg et al. 2014). Schläwicke-Engström et al. 2008; Woods et al.
2014a).
Polymorphisms of genes encoding for ATP-bind-
New biomarkers of susceptibility to mercury
ing cassette (ABC) membrane transporters have
As research on the interaction between genes and been linked to elevated uptake of MeHg across the
environment evolves, it has become increasingly placenta and increased cord-blood Hg levels (Llop
apparent that toxicity and disease risk are the et al. 2014) and altered urinary excretion of Hg in
result of complex interactions between genetic ASSGM exposed populations (Engström et al. 2013).
and environmental (epigenetic) factors that deter- Other membrane transporters such as organic anion
mine a high degree of variability in individual transporters (OATs) and L-amino acid transporters
response to a given exposure (Basu, Goodrich, (LATs) are known to be involved in Hg uptake
and Head 2014). through membranes (Bridges and Zalups 2005;
Zalups 1998), and SNPs in their genes have also
Polymorphisms been related to changes in Hg excretion and accu-
A field of research that has grown in recent mulation (Engström et al. 2013). Similarly, SNPs in
years is the study in human populations of the genes encoding for metallothioneins (MTs), espe-
relationship between Hg exposure biomarker cially the isoforms MT1, MT2 and MT4, have been
levels and genetic polymorphisms, namely sin- associated with alterations in concentrations of
gle-nucleotide polymorphisms (SNPs) in genes exposure biomarkers, namely Hg levels in urine
linked to xenobiotic ADME. The influence of and hair (Gundacker et al. 2009; Wang et al. 2012),
these particular genes on Hg accumulation may and neurobehavioral endpoints (Woods et al. 2013).
vary considerably depending upon the level of The precise significance of these SNPs for MT func-
exposure, the Hg species to which the popula- tion has not been clarified, but it is reasonable to
tion is exposed and also the degree to which the postulate that SNPs might lead to structural changes
SNP is inherent in the population or if the trait in MTs that consequently alter protein ability to bind
is poligenic (Barcelos et al. 2013; Llop, Ballester, metals (Wang et al. 2012).
and Broberg 2015). Further, false-positive find- A polymorphism in the gene encoding for inositol
ings cannot be totally ruled out (Schläwicke- 1,4,5-triphosphate kinase C (ITPKC) has been
Engström et al. 2008). related to the development of Kawasaki syndrome
Among the several SNPs related to Hg exposure, (KS) in susceptible children, following exposure to
those concerning GSH synthesis and metabolism are EtHg after TCV immunization (Yeter and Deth
the best studied. Several investigators (Barcelos et al. 2012). This SNP results in increased Ca release
2013; Custodio et al. 2004, 2005; Goodrich et al. from cells, which is at the basis for observed auto-
2011; Gundacker et al. 2009; Parajuli et al. 2016. immunity and coronary arterial lesions in KS (Yeter
Schläwicke-Engström et al. 2008) showed that Hg and Deth 2012).
accumulation was altered by SNPs in genes encoding In a study on the effect of Hg0 exposure via dental
for glutamyl cysteine ligase (GCL) and glutathione amalgams in children, it was found that a genetic
synthetase (GS), enzymes involved in GSH synthesis. variant of coproporphyrinogen oxidase (CPOX),
Depending upon the resulting phenotype (increase CPOX4, was associated with enhanced susceptibility
or decreased expression), such SNPs may lead to to Hg, as evidenced by lower performance in neuro-
different levels of GSH, affecting Hg retention/excre- behavioral testing (Woods et al. 2012, 2014a).
tion. Similarly, SNPs in genes controlling the synth- Similar outcomes were noted in subjects with SNPs
esis of different classes of glutathione S-transferases in catechol-O-methyltransferase (COMT) (Woods
(GST), which are responsible for mediating GSH- et al. 2014a, 2014b). Notably, both effects of
22 V. BRANCO ET AL.

CPOX4 and COMT SNPs were only evident in boys more vulnerable populations or its subsets, which
and not girls, suggesting gender-related differences could then be the focus of risk management/commu-
in susceptibility. Specific polymorphisms of COMT nication strategies.
(Val158Met), brain-derived neurotrophic factor
(BDNF), and serotonin transporter gene promoter Epigenetic markers
region (5-HTTLPR) have been associated with ele- The effect of Hg compounds on factors affecting
vated Hg levels and moodiness in dental workers gene expression other than direct modification of
exposed to Hg0 (Heyer et al. 2004, 2008, 2009; DNA sequences is the most recently emerging field
Parajuli et al. 2016; Woods et al. 2014a). of research in this metal toxicology. Although
It is of interest in that SNPs in the gene encoding there are a few studies concerning the influence
for selenoprotein P1 (SelP1) were linked to changes in of Hg exposure on epigenetic markers in wildlife
Hg2+ excretion in urine and MeHg incorporation in and lab animals (Basu, Goodrich, and Head 2014),
hair (Goodrich et al. 2011). SelP1 contains multiple data from human studies are scarce.
Sec residues and is secreted from liver to plasma and Hypermethylation of CpG islands, an epigenetic
thought to be involved in Se distribution throughout trait associated with gene silencing, was described
the body (Burk and Hill 2005; Papp et al. 2007) as well by Cardenas et al. (2015) in umbilical cord blood
as known to bind Hg (Suzuki and Ogra 2001). Thus, of infants exposed in utero to Hg. In addition, both
changes in SelP1expression or Sec content resulting GSTM1 (Hanna et al. 2012) and SEPP1 genes
from SNPs may exert an important impact in Hg (Goodrich et al. 2013) were shown to be
distribution, excretion and affect the pool of Se avail- hypermethylated.
able for metabolism. In this sense, it would be of Sanders et al. (2015) examined cervical swabs
interest to extend this analysis to the effects of other from pregnant women and found that the expres-
critical molecular targets for mercurials. In addition to sion of several miRNA was negatively associated
SelP1, Parajuli et al. (2016) found a positive associa- with Hg exposure, but the significance of such
tion between an SNP in the gene encoding for TrxR2 findings is not totally clear.
and internal dose makers for Hg. Given the relevance
of this enzyme for Hg toxicity, this association is
Remarks and perspectives
worth further detailed analysis.
Several other SNPs have been related to alterations The available knowledge concerning Hg toxicity
in levels of Hg exposure biomarkers even though and biomarkers suggests that:
correlations are often absent or nonexistent. Among -Among the possibilities for putative biomar-
others, these include apolipoprotein E (Ng et al., 2013; kers in Hg toxicity, promising endpoints include
Woods et al. 2014a), eNos (De Marco, Braga, and selenoprotein (such as TrxR and GPx isoforms)
Barbosa 2011), paraoxonase 1 progesterone receptor, activities in noninvasive biological samples. The
transferrin (Julvez et al. 2013), kidney and brain latter may serve in the future as potential early
expressed protein (KIBRA), and glutamate receptors indicators of cellular damage, that is, predictive
(Woods et al. 2014a). of damage and eventual disease.
One of the main limitations of SNPs analysis is -Although transcription factors such as Nrf2 and
related to sample size, which needs to be sufficient to NF-kappa B, as well as proteins such as GFAP and
incorporate a significant number of the less frequent glutamate transporters, are considered as important
alleles in the population, in order to enable an ade- molecules affected by and modulating MeHg-induced
quate statistical analysis and understanding of the neurotoxicity, their use as biomarkers is likely not
actual relevance of the polymorphism to Hg toxicity. practical because of the lack of specificity with respect
It should be noted that prospective birth-cohort stu- to solely Hg, as well as difficulty of properly assessing
dies with appropriate confounding controls aimed at samples from the CNS.
addressing the link between SNPs and neurotoxicity -Immunotoxicity, including autoimmune effects
in children remain to be done (Llop, Ballester, and have been established at metal concentrations well
Broberg 2015). Nevertheless, this is a promising field below the adverse threshold for CNS and kidney
of research and may serve a great value at identifying toxicity, and this issue seems to have been overlooked
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B 23

as a critical effect; however, the responses noted again following in utero exoposure. Journal Toxicogical
are not practical due to lack of specificity. Environment Health, A 71:119–30. doi:10.1080/
15287390701613140.
- Protein adducts with mercuric species are
Agrawal, A., P. Kaushal, S. Agrawal, S. Gollapudi, and S.
presently amenable to analysis by sophisticated Gupta. 2007. Thimerosal induces Th2 responses via influ-
methods based on mass spectrometry, and this encing cytokine secretion by human dendritic cells.
undoubtedly will provide a future focus for the Journal of Leukocyte Biology 81:474–82. doi:10.1189/
discovery of protein targets with critical function jlb.0706467.
(s) as predictive biomarkers of Hg-induced toxi- Alam, J., C. Wicks, D. Stewart, P. Gong, C. Touchard, S.
Otterbein, A. M. Choi, M. E. Burow, and J. Tou. 2000.
city. Finally, new developments in genomics and
Mechanism of heme oxygenase-1 gene activation by cad-
SNPs identification of target proteins and epige- mium in MCF-7 mammary epithelial cells. Role of p38
netic markers may lead to individualized predic- kinase and Nrf2 transcription factor. The Journal of
tion of Hg-induced toxicity; Biological Chemistry 275:27694–702.
Taken together, a critical need exists for validation Alkazemi, D., G. M. Egeland, L. J. Roberts 2nd, H. M. Chan, and
of reliable and specific biomarkers that represent the S. Kubow. 2013. New insights regarding tissue Se and Hg
interactions on oxidative stress from plasma IsoP and IsoF
earliest signs of Hg exposure in an attempt to reduce
measures in the Canadian Inuit population. The Journal of
or eliminate exposure and protect the most vulnerable Lipid Research 54:1972–79. doi:10.1194/jlr.M033068.
populations from this metal. New insights into the Allen, J. W., L. A. Mutkus, and M. Aschner. 2001a. Mercuric
mechanistic pathways affected by mercurial com- chloride, but not methylmercury, inhibits glutamine synthe-
pounds might further stimulate discovery in this fast tase activity in primary cultures of cortical astrocytes. Brain
evolving field. research 891:148–57. doi:10.1016/S0006-8993(00)03185-1.
Allen, J. W., L. A. Mutkus, and M. Aschner. 2001b.
Methylmercury-mediated inhibition of 3H-d-aspartate
Acknowledgments transport in cultured astrocytes is reversed by the antiox-
idant catalase. Brain research 902:92–100. doi:10.1016/
The authors thank the reviewers of this manuscript whose S0006-8993(01)02375-7.
comments and suggestions contributed to significantly Allen, J. W., G. Shanker, and M. Aschner. 2001c.
improve the original submission. Methylmercury inhibits the in vitro uptake of the glu-
tathione precursor, cystine, in astrocytes, but not in neu-
rons. Brain research 894:131–40. doi:10.1016/S0006-8993
Funding (01)01988-6.
Cristina Carvalho and Vasco Branco are supported by Fundação Al-Saleh, I., A. Al-Sedairi, and R. Elkhatib. 2012. Effect of
para a Ciência e Tecnologia, Portugal (www.fct.pt) through mercury (Hg) dental amalgam fillings on renal and oxida-
MERTOX project (PTDC/QUI-BIQ/117281/2010) and the Post tive stress biomarkers in children. Science of the Total
Doc fellowship (SFRH/BPD/85219/2012). The work was funded, Environment 431:188–96. doi:10.1016/j.
in part, by iMed.ULisboa through project UID/DTP/04138/2013 scitotenv.2012.05.036.
also from FCT. MA was supported in part by grants from the Amoli, J. S., A. Barin, M. Ebrahimi-Rad, and P. Sadighara.
NIH, R01 ES10563 and R01 ES07331, and R01 ES020852. MF was 2011. Cell damage through pentose phosphate pathway in
supported in part by grants from the Brazilian Agencies CNPq, fetus fibroblast cells exposed to methyl mercury. Journal
CAPES and FAPESC. Applications Toxicogical 31:685–89. doi:10.1002/jat.1628.
Andrews, N., E. Miller, A. Grant, J. Stowe, V. Osborne, and B.
Taylor. 2004. Thimerosal exposure in infants and develop-
References mental disorders: A retrospective cohort study in the
United Kingdom does not support a causal association.
Abou-Donia, M. B., M. M. Abou-Donia, E. M. ElMasry, J. A. Pediatrics 114:584–91. doi:10.1542/peds.2003-1177-L.
Monro, and M. F. A. Mulder. 2013. Autoantibodies to Armah, F. A., S. A. Boamah, R. Quansah, S. Obiri, and I.
nervous system specific proteins are elevated in sera of Luginaah. 2016. Unsafe occupational health behaviors:
flight crew members: Biomarkers for nervous sytem injury. Understanding mercury-related environmental health
Journal Toxicogical Environment Health, A 76:363–80. risks to artisanal gold miners in Ghana. Frontiers
doi:10.1080/15287394.2013.765369. Environment Sciences 4:29. doi:10.3389/
Abou-Donia, M. B., L. B. Goldstein, S. Bullman, T. Tu, W. A. fenvs.2016.00029.
Khan, A. M. Dechkovskaia, and A. A. Abdel-Rahman. Arnér, E. S. J. 2009. Focus on mammalian thioredoxin reduc-
2008. Imidacloprid induces neurobehavioral deficits and tases - important selenoproteins with versatile functions.
increases protein expression of glial fibrillary acidic protein Biochim Biophysics Acta 1790:495–526. doi:10.1016/j.
in the motor cortex and hippocampus in offspring rats bbagen.2009.01.014.
24 V. BRANCO ET AL.

Aschner, M. 2000. Astrocytic swelling, phospholipase A2, Bélanger, M.-C., M.-E. Mirault, E. Dewailly, M. Plante, L.
glutathione and glutamate: Interactions in methylmer- Berthiaume, M. Noel, and P. Julien. 2008. Seasonal mer-
cury-induced neurotoxicity. Cellular Molecular Biologic cury and oxidant-antioxidant status of James Bay sport
46:843–54. fishermen. Metabolism 57:630–36. doi:10.1016/j.
Aschner, M., Y.-L. Du, M. Gannon, and H. K. Kimelberg. 1993. metabol.2007.12.006.
Methylmercury-induced alterations in excitatory amino acid Benford, D. J., A. B. Hanley, K. Bottrill, S. Oehlshlager, M.
transport in rat primary astrocyte cultures. Brain research Balls, F. Branca, J. J. Castegnaro, J. Descotes, K.
602:181–86. doi:10.1016/0006-8993(93)90680-L. Hemminiki, D. Lindsay, and B. Schilter. 2000.
Aylward, L. L., S. M. Hays, C. R. Kirman, S. A. Marchitti, J. F. Biomarkers as predictive tools in toxicity testing. ATLA -
Kenneke, C. English, D. R. Mattison, and R. A. Becker. Alternative Laboratory Animal 28:119–31.
2014. Relationships of chemical concentrations in maternal Berglund, M., B. Lind, K. A. Björnberg, B. Palm, Ö.
and cord blood: A review of available data. Journal Einarsson, and M. Vahter. 2005. Inter-individual varia-
Toxicogical Environment Health, B 17:175–203. tions of human mercury exposure biomarkers: A cross-
doi:10.1080/10937404.2014.884956. sectional assessment. Environment Health 4:20.
Baird, L., and A. T. Dinkova-Kostova. 2011. The cytoprotec- doi:10.1186/1476-069X-4-20.
tive role of the Keap1-Nrf2 pathway. Archives of Toxicology Bernard, A. 2008. Biomarkers of metal toxicity in population
85:241–72. doi:10.1007/s00204-011-0674-5. studies: Research potential and interpretation issues.
Baraldi, M., P. Zanoli, F. Tascedda, J. M. Blom, and N. Journal Toxicogical Environment Health A 71:1259–65.
Brunello. 2002. Cognitive deficits and changes in gene doi:10.1080/15287390802211885.
expression of NMDA receptors after prenatal methylmer- Bernard, A., and R. Lauwerys. 1989. Epidemiological applica-
cury exposure. Environment Health Persp 110 (Suppl tion of early markers of nephrotoxicity. Toxicology Letters
5):855–58. doi:10.1289/ehp.02110s5855. 46:293–306. doi:10.1016/0378-4274(89)90137-9.
Barbosa, A. C., A. A. Boischio, G. A. East, I. Ferrari, A. Berntssen, M. H., A. Aatland, and R. D. Handy. 2003.
Gonçalves, P. R. M. Silva, and T. M. E. Cruz. 1995. Chronic dietary mercury exposure causes oxidative stress,
Mercury contamination in the Brazilian Amazon. brain lesions, and altered behavior in Atlantic salmon
Environmental and occupational aspects. Water, Air, and (Salmo salar) parr. Aquatic Toxicology 65:55–72.
Soil Pollution 80:109–21. doi:10.1007/BF01189660. doi:10.1016/S0166-445X(03)00104-8.
Barbosa, A. C., S. R. L. Silva, and J. G. Dórea. 1998. Concentration Björnberg, K. A., M. Vahter, B. Berglund, B. Niklasson, M.
of mercury in hair of indigenous mothers and infant from the Blennow, and G. Sandborgh-Englund. 2005. Transport of
Amazon basin. Archives Environment Contamin Toxicogical methylmercury and inorganic mercury to the fetus and
34:100–05. doi:10.1007/s002449900291. breast-fed infant. Environmental Health Perspectives
Barcelos, G. R., D. Grotto, K. C. De Marco, J. Valentini, A. 113:1381–85. doi:10.1289/ehp.7856.
Lengert, A. A. De Oliveira, S. C. Garcia, G. Ú. Braga, K. Bo, L., D. M. Miller, and J. S. Woods. 1993. Studies on Hg(II)-
Schläwicke- Engström, I. M. Cólus, K. Broberg, and F. induced H2O2 formation and oxidative stress in vivo and in
Barbosa Jr. 2013. Polymorphisms in glutathione-related vitro in rat kidney mitochondria. Biochemical Pharmacology
genes modify mercury concentrations and antioxidant sta- 45:2017–24. doi:10.1016/0006-2952(93)90012-L.
tus in subjects environmentally exposed to methylmercury. Bose-O’Reilly, S., B. Lettmeier, G. Roider, U. Sieberta, and G.
The Science of the Total Environment 463-464:319–25. Drasch. 2008. Mercury in breast milk – a health hazard for
doi:10.1016/j.scitotenv.2013.06.029. infants in gold mining areas? International Journal Hygiene
Barone, S. Jr., N. Haykal-Coates, D. K. Parran, and H. A. Environment Health 211:615–23. doi:10.1016/j.ijheh.2007.
Tilson. 1998. Gestational exposure to methylmercury alters 09.015.
the developmental pattern of Trk-like immunoreactivity in Bose-O’Reilly, S., K. M. McCarty, N. Steckling, and B.
the rat brain and results in cortical dysmorphology. Lettmeier. 2010. Mercury exposure and children’s health.
Developments Brain Researcher 109:13–31. doi:10.1016/ Current Problems Pediatric Adolescent Health Care 40:186–
S0165-3806(98)00038-8. 215. doi:10.1016/j.cppeds.2010.07.002.
Barregard, L., F. Trachtenberg, and S. McKinlay. 2008. Renal Bose-O’Reilly, S., R. Schierl, D. Nowak, U. Siebert, J. F. William,
effects of dental amalgam in children: The New England F. T. Owi, and Y. I. Ir. 2016. A preliminary study on health
children’s amalgam trial. Environment Health Persp effects in villagers exposed to mercury in a small-scale arti-
116:394–99. doi:10.1289/ehp.10504. sanal gold mining area in Indonesia. Environmental Research
Bartell, S. M., R. A. Ponce, R. N. Sanga, and E. M. Faustman. 149:274–81. doi:10.1016/j.envres.2016.04.007.
2000. Human variability in mercury toxicokinetics and Branco, V., J. Canário, A. Holmgren, and C. Carvalho. 2011.
steady state biomarker ratios. Environmental Research Inhibition of the thioredoxin system in the brain and liver
84:127–32. doi:10.1006/enrs.2000.4104. of zebra-seabreams exposed to waterborne methylmercury.
Basu, N., J. M. Goodrich, and J. Head. 2014. Ecogenetics of Toxicology and Applied Pharmacology 251:95–103.
mercury: From genetic polymorphisms and epigenetics to doi:10.1016/j.taap.2010.12.005.
risk assessment and decision making. Environment Toxiol Branco, V., J. Canário, J. Lu, A. Holmgren, and C. Carvalho.
Chemical 33:1248–58. doi:10.1002/etc.2375. 2012a. Mercury and selenium interaction in vivo: Effects
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B 25

on thioredoxin reductase and glutathione peroxidase. Free to mercury and arsenic in utero. Epigenetics 10:508–15.
Radical Biologic Medica 52:781–93. doi:10.1016/j. doi:10.1080/15592294.2015.1046026.
freeradbiomed.2011.12.002. Carneiro, M. F., D. Grotto, and F. Barbosa Jr. 2014. Inorganic
Branco, V., A. Godinho-Santos, J. Gonçalves, J. Lu, A. and methylmercury levels in plasma are differentially asso-
Holmgren, and C. Carvalho. 2014. Mitochondrial thiore- ciated with age, gender, and oxidative stress markers in a
doxin reductase inhibition, selenium status and Nrf-2 acti- population exposed to mercury through fish consumption.
vation are determinant factors modulating the toxicity of Journal Toxicogical Environment Health A 77:69–79.
mercury compounds. Free Radical Biologic Medica 73:95– doi:10.1080/15287394.2014.865584.
105. doi:10.1016/j.freeradbiomed.2014.04.030. Carvalho, C., J. Lu, X. Zhang, E. Arnér, and A. Holmgren.
Branco, V., P. Ramos, J. Canário, J. Lu, A. Holmgren, and C. 2011. Effects of selenite and chelating agents on mamma-
Carvalho. 2012b. Biomarkers of adverse response to mer- lian thioredoxin reductase inhibited by mercury:
cury: Histopathology vs. thioredoxin reductase activity. Implications for treatment of mercury poisoning. FASEB
Journal of Biomedicine & Biotechnology 2012:359879. Journal 25:370–81. doi:10.1096/fj.10-157594.
doi:10.1155/2012/359879. Carvalho, C. M. L., E. Chew, S. I. Hashemy, J. Lu, and A.
Bridges, C. C., and R. K. Zalups. 2005. Molecular and ionic Holmgren. 2008a. Inhibition of human thioredoxin sys-
mimicry and the transport of toxic metals. Toxicology and tem: A molecular mechanism of mercury toxicity. The
Applied Pharmacology 204:274–308. doi:10.1016/j. Journal of Biological Chemistry 283:11913–23.
taap.2004.09.007. doi:10.1074/jbc.M710133200.
Bridges, C. C., and R. K. Zalups. 2010. Transport of inorganic Carvalho, C. M. L., A. I. N. M. Matos, M. L. Mateus, A. P. M.
mercury and methylmercury in target tissues and organs. Santos, and M. C. C. Batoréu. 2008b. High-fish consump-
Journal Toxicogical Environment Health B 13:385–410. tion and risk prevention: Exposure assessment to methyl-
doi:10.1080/10937401003673750. mercury in Portugal. Journal Toxicogical Environment
Brookes, N. 1992. In vitro evidence for the role of glutamate Health A 71:1279–88. doi:10.1080/15287390801989036.
in the CNS toxicity of mercury. Toxicology 76:245–56. Castilhos, Z., S. Rodrigues-Filho, R. Cesar, A. P. Rodrigues, R.
doi:10.1016/0300-483X(92)90193-I. Villas-Bôas, I. De Jesus, M. Lima, K. Faial, A. Miranda, E.
Buckell, M., D. Hunter, R. Milton, and K. M. A. Perry. 1946. Brabo, and C. Beinhoff. 2015. Human exposure and risk
Chronic mercury poisoning. British Journal Industrial assessment associated with mercury contamination in arti-
Medica 3:55–63. sanal gold mining areas in the Brazilian Amazon.
Burger, J., W. L. Stephens, C. S. Boring Jr., M. Kuklinski, J. Environment Sciences Pollution Researcher International
W. Gibbons, and M. Gochfeld. 1999. Factors in exposure 22:11255–64. doi:10.1007/s11356-015-4340-y.
assessment: Ethnic and socioeconomic differences in fish- Chang, J. Y. 2007. Methylmercury causes glial IL-6 release.
ing and consumption of fish caught along the Savannah Neuroscience Letters 416:217–20. doi:10.1016/j.
River. Risk Analysis 19:427–38. doi:10.1111/j.1539- neulet.2007.01.076.
6924.1999.tb00418.x. Chen, C., H. Yu, J. Zhao, B. Li, L. Qu, S. Liu, P. Zhang, and Z.
Burk, R. F., and K. E. Hill. 2005. Selenoprotein P: An extra- Chai. 2006. The roles of serum selenium and selenopro-
cellular protein with unique physical characteristics and a teins on mercury toxicity in environmental and occupa-
role in selenium homeostasis. Annual Review of Nutrition tional exposure. Environment Health Persp 114:297–301.
25:215–35. doi:10.1146/annurev.nutr.24.012003.132120. doi:10.1289/ehp.7861.
Caito, S., H. Zeng, J. L. Aschner, and M. Aschner. 2014. Cherian, M. G., and T. W. Clarkson. 1976. Biochemical
Methylmercury alters the activities of Hsp90 client pro- changes in rat kidney on exposure to elemental mercury
teins, prostaglandin E synthase/p23 (PGES/23) and nNOS. vapor: Effect on biosynthesis of metallothionein.
Plos One 9:e98161. doi:10.1371/journal.pone.0 Chemistry-Biology Interaction 13:109–20. doi:10.1016/
098161. 0009-2797(76)90093-4.
Callan, A. C., A. Devine, L. Qi, J. C. Ng, and A. L. Hinwood. Chmielnicka, J., E. Brzeźnicka, and A. Sniady. 1986. Kidney
2015. Investigation of the relationship between low envir- concentrations and urinary excretion of mercury, zinc and
onmental exposure to metals and bone mineral density, copper following the administration of mercuric chloride
bone resorption and renal function. International Journal and sodium selenite to rats. Archives of Toxicology 59:16–
Hygiene Environment Health 218:444–51. doi:10.1016/j. 20. doi:10.1007/BF00263951.
ijheh.2015.03.010. Clarkson, T. W., and L. Magos. 2006. The toxicology of
Cambier, S., P. Gonzalez, N. Mesmer-Dudons, D. Brethes, M. mercury and its chemical compounds. Critical Reviews in
Fujimura, and J. P. Bourdineaud. 2012. Effects of dietary Toxicology 36:609–62. doi:10.1080/10408440600845619.
methylmercury on the zebrafish brain: Histological, mito- Clarkson, T. W., L. Magos, and G. J. Myers. 2003. Human
chondrial, and gene transcription analyses. Biometals exposure to mercury: The three modern dilemmas. Journal
25:165–80. doi:10.1007/s10534-011-9494-6. Trace Elements Experiments Medica 16:321–43.
Cardenas, A., D. C. Koestler, E. A. Houseman, B. P. Jackson, M. doi:10.1002/(ISSN)1520-670X.
L. Kile, M. R. Karagas, and C. J. Marsit. 2015. Differential Clarkson, T. W., J. B. Vyas, and N. Ballatori. 2007.
DNA methylation in umbilical cord blood of infants exposed Mechanisms of mercury disposition in the body.
26 V. BRANCO ET AL.

American Journal of Industrial Medicine 50:757–64. (ethylmercury). Journal Toxicogical Environment Health
doi:10.1002/(ISSN)1097-0274. A 79:502–12. doi:10.1080/15287394.2016.1182003.
Cooke, G. M. 2014. Biomonitoring of human fetal exposure Deng, Y., Z. Xu, B. Xu, W. Liu, Y. Wei, Y. Li, S. Feng, and T.
to environmental chemicals in early pregnancy. Journal Yang. 2014. Exploring cross-talk between oxidative
Toxicogical Environment Health B 17:205–24. damage and excitotoxicity and the effects of riluzole in
doi:10.1080/10937404.2014.898167. the rat cortex after exposure to methylmercury.
Cooper, A. J., and B. S. Kristal. 1997. Multiple roles of Neurotoxicol Researcher 26:40–51. doi:10.1007/s12640-
glutathione in the central nervous system. Biological 013-9448-6.
Chemistry 378:793–802. Diamond, G. L., and R. K. Zalups. 1998. Understanding renal
Cordier, S., M. Garel, L. Mandereau, H. Morcel, P. Doineau, toxicity of heavy metals. Toxicogical Pathologists 26:92–
and S. Gosme-Seguret. 2002. Neurodevelopmental investi- 103. doi:10.1177/019262339802600111.
gations among methylmercury exposed children in French Dietrich, M. O., C. E. Mantese, G. Dos Anjos, D. O. Souza,
Guiana. Environmental Research 89:1–11. doi:10.1006/ and M. Farina. 2005. Motor impairment induced by oral
enrs.2002.4349. exposure to methylmercury in adult mice. Environmental
Counter, S. A., L. H. Buchanan, F. Ortega, and G. Laurell. Toxicology and Pharmacology 19:169–75. doi:10.1016/j.
2002. Elevated blood mercury and neuro-otological obser- etap.2004.07.004.
vations in children of the Ecuadorian gold mines. Journal Díez, S., S. Delgado, I. Aguilera, J. Astray, B. Pérez-Gómez,
of Toxicology and Environmental Health. Part A 65:149–63. M. Torrent, J. Sunyer, and J. M. Bayona. 2009. Prenatal
doi:10.1080/152873902753396785. and early childhood exposure to mercury and methylmer-
Custodio, H. M., K. Broberg, M. Wennberg, J. H. Jansson, B. cury in Spain, a high-fish-consumer country. Archives of
Vessby, G. Hallmans, B. Stegmayr, and S. Skerfving. 2004. Environmental Contamination and Toxicology 56:615–22.
Polymorphisms in glutathione-related genes affect methyl- doi:10.1007/s00244-008-9213-7.
mercury retention. Archives of Environmental Health Djukic, M. M., M. D. Jovanovic, M. Ninkovic, I. Stevanovic,
59:588–95. doi:10.1080/00039890409603438. K. Ilic, M. Curcic, and J. Vekic. 2012. Protective role of
Custodio, H. M., R. Harari, L. Gerhardsson, S. Skerfving, and glutathione reductase in paraquat induced neurotoxicity.
K. Broberg. 2005. Genetic influences on the retention of Chemistry-Biology Interaction 199:74–86. doi:10.1016/j.
inorganic mercury. Archives Environment Occupational cbi.2012.05.008.
Health 60:17–23. doi:10.3200/AEOH.60.1.17-23. Doi, R., and M. Tagawa. 1983. A study on the biochemical
Dahl, R., R. F. White, P. Weihe, N. Sørensen, R. Letz, H. K. and biological behavior of methylmercury. Toxicology and
Hudnell, D. A. Otto, and P. Grandjean. 1996. Feasibility Applied Pharmacology 69:407–16. doi:10.1016/0041-008X
and validity of three computer-assisted neurobehavioral (83)90264-8.
tests in 7-year-old children. Neurotoxicology Teratol Dong, L., Z. Li, X. Bi, and L. Ling. 2001. Effects of methyl
18:413–19. doi:10.1016/0892-0362(96)00031-1. mercury chloride on nuclear factor-kappa B DNA binding
Dalla Corte, C. L., C. Wagner, J. H. Sudati, B. Comparsi, G. activities of nuclear protein extracts from developing rat
O. Leite, A. Busanello, F. A. Soares, M. Aschner, and J. B. cerebra and cerebella. Wei Sheng Yan Jiu 30:7–9.
Rocha. 2013. Effects of diphenyl diselenide on methylmer- Dórea, J. G., V. L. Bezerra, V. Fajon, and M. Horvat. 2011.
cury toxicity in rats. Biomedical Researcher International Speciation of methyl- and ethyl-mercury in hair of
2013:983821. doi:10.1155/2013/983821. breastfed infants acutely exposed to Thimerosal-containing
Dare, E., W. Li, B. Zhivotovsky, X. Yuan, and S. Ceccatelli. vaccines. Clinica Chimica Acta 412:1563–66. doi:10.1016/j.
2001. Methylmercury and H2O2 provoke lysosomal cca.2011.05.003.
damage in human astrocytoma D384 cells followed by Dórea, J. G., M. Farina, and J. B. T. Rocha. 2013. Toxicity of
apoptosis. Free Radical Biologic Medica 30:1347–56. ethylmercury (and Thimerosal): A comparison with
doi:10.1016/S0891-5849(01)00526-3. methylmercury. Journal Applications Toxicogical 33:700–
Dave, V., K. J. Mullaney, S. Goderie, H. K. Kimelberg, and M. 11. doi:10.1002/jat.2855.
Aschner. 1994. Astrocytes as mediators of methylmercury Dórea, J. G., R. C. Marques, and L. Abreu. 2014. Milestone
neurotoxicity: Effects on D-aspartate and serotonin uptake. achievement and neurodevelopment of rural Amazonian
Developmental Neuroscience 16:222–31. doi:10.1159/ toddlers (12 to 24 Months) with different methylmercury
000112110. and ethylmercury exposure. Journal Toxicogical
De Marco, K. C., G. U. Braga, and F. Barbosa Jr. 2011. Environment Health A 77:1–13. doi:10.1080/
Determination of the effects of eNOS gene polymorphisms 15287394.2014.861335.
(T-786C and Glu298Asp) on nitric oxide levels in a Dórea, J. G., R. C. Marques, and C. Isejima. 2012. Antenatal
methylmercury-exposed population. Journal Toxicogical and postnatal exposure to methyl- and ethylmercury.
Environment Health A 74:1323–33. doi:10.1080/ Journal of Biomedicine & Biotechnology 2012:9.
15287394.2011.600665. doi:10.1155/2012/132876.
De Oliveira Souza, V. C., K. C. De Marco, H. J. Laure, J. C. Echeverria, D., H. V. Aposhian, J. S. Woods, N. J. Heyer, M.
Rosa, and F. Barbosa Jr. 2016. A brain proteome profile in M. Aposhian, A. C. Bittner Jr, R. K. Mahurin, and M.
rats exposed to methylmercury or thimerosal Cianciola. 1998. Neurobehavioral effects from exposure
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B 27

to dental amalgam Hg0: New distinctions between recent Franco, J. L., H. C. Braga, J. Stringari, F. C. Missau, T.
exposure and Hg body burden. FASEB Journal 12:971–80. Posser, B. G. Mendes, R. B. Leal, A. R. Santos, A. L.
Echeverria, D., N. J. Heyer, M. D. Martin, C. A. Naleway, J. S. Dafre, M. G. Pizzolatti, and M. Farina. 2007. Mercurial-
Woods, and A. C. Bittner. 1994. Behavioral effects of low- induced hydrogen peroxide generation in mouse brain
level exposure to elemental mercury among dentists. mitochondria: Protective effects of quercetin. Chemical
Neurotoxicology Teratol 17:161–68. doi:10.1016/0892-0362 Research in Toxicology 20:1919–26. doi:10.1021/
(94)00049-J. tx7002323.
El-Fawal, H. A., Z. Gong, A. R. Little, and H. L. Evans. 1996. Franco, J. L., T. Posser, P. R. Dunkley, P. W. Dickson, J. J.
Exposure to methylmercury results in serum autoantibo- Mattos, R. Martins, A. C. Bainy, M. R. Marques, A. L.
dies to neurotypic and gliotypic proteins. Neurotoxicology Dafre, and M. Farina. 2009. Methylmercury neurotoxicity
17:531–39. is associated with the inhibition of the antioxidant enzyme
Engström, K., S. Ameer, L. Bernaudat, G. Drasch, J. Baeuml, glutathione peroxidase. Free Radical Biologic Medica
S. Skerfving, S. Bose-O’Reilly, and K. Broberg. 2013. 47:449–57. doi:10.1016/j.freeradbiomed.2009.05.013.
Polymorphisms in genes encoding potential mercury Franco, J. L., T. Posser, F. Missau, M. G. Pizzolatti, A. R. Dos
transporters and urine mercury concentrations in popula- Santos, D. O. Souza, M. Aschner, J. B. T. Rocha, A. L.
tions exposed to mercury vapor from gold mining. Dafre, and M. Farina. 2010. Structure-activity relationship
Environment Health Persp 121:85–91. of flavonoids derived from medicinal plants in preventing
Eroglu, K., G. Atli, and M. Canli. 2005. Effects of metal (Cd, methylmercury-induced mitochondrial dysfunction.
Cu, Zn) interactions on the profiles of metallothionein-like Environmental Toxicology and Pharmacology 30:272–78.
proteins in the Nile fish Oreochromis niloticus. Bulletin doi:10.1016/j.etap.2010.07.003.
Environment Contamination Toxicogical 75:390–99. Franco, J. L., A. Teixeira, F. C. Meotti, C. M. Ribas, J.
doi:10.1007/s00128-005-0766-0. Stringari, S. C. Garcia Pomblum, A. M. Moro, D. Bohrer,
Eskes, C., P. Honegger, L. Juillerat-Jeanneret, and F. Monnet- A. V. Bairros, A. L. Dafre, A. R. Santos, and M. Farina.
Tschudi. 2002. Microglial reaction induced by noncyto- 2006. Cerebellar thiol status and motor deficit after lacta-
toxic methylmercury treatment leads to neuroprotection tional exposure to methylmercury. Environmental Research
via interactions with astrocytes and IL-6 release. Glia 102:22–28. doi:10.1016/j.envres.2006.02.003.
37:43–52. doi:10.1002/(ISSN)1098-1136. Franconi, F., M. Miceli, L. Alberti, G. Seghieri, M. G. De
Farina, M., M. Aschner, and J. B. T. Rocha. 2011. Oxidative Montis, and A. Tagliamonte. 1998. Further insights into
stress in MeHg-induced neurotoxicity. Toxicology and the anti-aggregating activity of NMDA in human platelets.
Applied Pharmacology 256:405–17. doi:10.1016/j. British Journal of Pharmacology 124:35–40. doi:10.1038/sj.
taap.2011.05.001. bjp.0701790.
Farina, M., F. Campos, I. Vendrell, J. Berenguer, M. Barzi, S. Franconi, F., M. Miceli, M. G. De Montis, E. L. Crisafi, F.
Pons, and C. Sunol. 2009. Probucol increases glutathione Bennardini, and A. Tagliamonte. 1996. NMDA receptors
peroxidase-1 activity and displays long-lasting protection play an anti-aggregating role in human platelets. Thromb
against methylmercury toxicity in cerebellar granule cells. Haemost 76:84–87.
Toxicological Sciences 112:416–26. doi:10.1093/toxsci/kfp219. Franko, A., M. V. Budihna, and M. Dodic-Fikfak. 2005. Long.
Fessel, J. P., and L. J. Roberts II. 2005. Isofurans: Novel term effects of elemental mercury on renal function in
products of lipid peroxidation that define the occurrence miners of the Idrija mercury mine. Annals Occupational
of oxidant injury in settings of elevated oxygen tension. Hygiene 49:521–27. doi:10.1093/annhyg/mei022.
Antioxidants & Redox Signaling 7:202–09. doi:10.1089/ Fujita, H. 2001. Measurement of delta-aminolevulinate dehy-
ars.2005.7.202. dratase activity. In Current protocols in toxicology, eds L. G.
Fombonne, E., R. Zakarian, A. Bennett, L. Meng, and D. Costa, E. Hodgson, D. A. Lawrence, D. J. Reed, and W. F.
McLean-Heywood. 2006. Pervasive developmental disor- GreenLee, 8.6:8.61-8.66.11. New Jersey, USA: John Wiley
ders in Montreal, Quebec, Canada: Prevalence and links & Sons Inc.
with immunizations. Pediatrics 118:e139–150. doi:10.1542/ Fujiyama, J., K. Hirayama, and A. Yasutake. 1994.
peds.2005-2993. Mechanism of methylmercury efflux from cultured astro-
Fournie, G. J., M. Mas, B. Cautain, M. Savignac, J. F. Subra, L. cytes. Biochemical Pharmacology 47:1525–30. doi:10.1016/
Pelletier, A. Saoudi, D. Lagrange, M. Calise, and P. Druet. 0006-2952(94)90527-4.
2001. Induction of autoimmunity through bystander Gao, Y., C. H. Yan, X. D. Yu, and S. H. Wu. 2006. Effects of
effects. Lessons from immunological disorders induced perinatal exposure to methylmercury on the structure of
by heavy metals. Journal of Autoimmunity 16:319–26. hippocampus and cerebellum in young rats. Wei Sheng
doi:10.1006/jaut.2000.0482. Yan Jiu 35:402–05.
Fowler, B. A., and K. R. Mahaffey. 1978. Interactions among Gardner, R. M., J. F. Nyland, and E. K. Silbergeld. 2010b.
lead, cadmium, and arsenic in relation to porphyrin excre- Differential immunotoxic effects of inorganic and organic
tion patterns. Environment Health Persp 25:87–90. mercury species in vitro. Toxicology Letters 198:182–90.
doi:10.1289/ehp.782587. doi:10.1016/j.toxlet.2010.06.015.
28 V. BRANCO ET AL.

Gardner, R. M., J. F. Nyland, I. A. Silva, A. M. Ventura, J. M. with prenatal exposure to methylmercury. Neurotoxicology
De Souza, and E. K. Silbergeld. 2010a. Mercury exposure, Teratol 19:417–28. doi:10.1016/S0892-0362(97)00097-4.
serum antinuclear/antinucleolar antibodies, and serum Grintal, B., G. Champeil-Potokar, M. Lavialle, S. Vancassel, S.
cytokine levels in mining populations in amazonian Breton, and I. Denis. 2009. Inhibition of astroglial gluta-
Brazil: A cross-sectional study. Environmental Research mate transport by polyunsaturated fatty acids: Evidence for
110:345–54. doi:10.1016/j.envres.2010.02.001. a signaling role of docosahexaenoic acid. Neurochemistry
Gerson, R. J., and Z. A. Shaikh. 1982. Uptake and binding of International 54:535–43. doi:10.1016/j.neuint.2009.02.018.
cadmium and mercury to metallothionein in rat hepato- Grotto, D., J. Valentini, M. Fillion, C. J. S. Passos, S. C.
cyte primary cultures. The Biochemical Journal 208:465–72. Garcia, D. Mergler, and F. Barbosa Jr. 2010. Mercury
doi:10.1042/bj2080465. exposure and oxidative stress in communities of the
Gibb, H., and K. G. O’Leary. 2014. Mercury exposure and Brazilian Amazon. The Science of the Total Environment
health impacts among individuals in the artisanal and 408:806–11. doi:10.1016/j.scitotenv.2009.10.053.
small-scale gold mining community: A comprehensive Gundacker, C., K. J. Wittmann, M. Kukuckova, G.
review. Environment Health Persp 122:667. Komarnicki, I. Hikkel, and M. Gencik. 2009. Genetic back-
Gil, F., and A. Pla. 2001. Biomarkers as biological indicators ground of lead and mercury metabolism in a group of
of xenobiotic exposure. Journal Applications Toxicogical medical students in Austria. Environmental Research
21:245–55. doi:10.1002/(ISSN)1099-1263. 109:786–96. doi:10.1016/j.envres.2009.05.003.
Ginsberg, G., B. Sonawane, R. Nath, and P. Lewandowski. Hagmar, L., M. Persson-Moschos, B. Åkesson, and A. Schütz.
2014. Methylmercury-induced inhibition of paraoxonase-1 1998. Plasma levels of selenium, selenoprotein P and glu-
(PON-1)—Implications for cardiovascular risk. Journal tathione peroxidase and their correlations to fish intake and
Toxicogical Environment Health A 77:1004–23. serum levels of thyrotropin and thyroid hormones: A study
doi:10.1080/15287394.2014.919837. on Latvian fish consumers. European Journal Clinical
Goodrich, J. M., N. Basu, A. Franzblau, and D. C. Dolinoy. Nutritional 52:796–800. doi:10.1038/sj.ejcn.1600649.
2013. Mercury biomarkers and DNA methylation among Halbach, S., and T. W. Clarkson. 1978. Enzymatic oxidation
Michigan dental professionals. Environmental and of mercury vapour by erythrocytes. Biochimica Et
Molecular Mutagenesis 54:195–203. doi:10.1002/em.v54.3. Biophysica Acta 523:522–31. doi:10.1016/0005-2744(78)
Goodrich, J. M., Y. Wang, B. Gillespie, R. Werner, A. 90055-4.
Franzblau, and N. Basu. 2011. Glutathione enzyme and Hanna, C. W., M. S. Bloom, W. P. Robinson, D. Kim, P. J.
selenoprotein polymorphisms associate with mercury bio- Parsons, F. S. Vom Saal, J. A. Taylor, A. J. Steuerwald, and
marker levels in Michigan dental professionals. Toxicology V. Y. Fujimoto. 2012. DNA methylation changes in whole
and Applied Pharmacology 257:301–08. doi:10.1016/j. blood is associated with exposure to the environmental
taap.2011.09.014. contaminants, mercury, lead, cadmium and bisphenol A,
Grandjean, P., S. S. Brown, P. Reavey, and D. S. Young. 1994. in women undergoing ovarian stimulation for IVF.
Biomarkers of chemical exposure: State of the art. Clinical Human Reprod 27:1401–10. doi:10.1093/humrep/des038.
Chemistry 40:1360–62. Hatfield, D. L., P. A. Tsuji, B. A. Carlson, and V. N.
Grandjean, P., E. Budtz-Jørgensen, R. F. White, P. J. Gladyshev. 2014. Selenium and selenocysteine: Roles in
Jørgensen, P. Weihe, F. Debes, and N. Keiding. 1999. cancer, health, and development. Trends in Biochemical
Methylmercury exposure biomarkers as indicators of neu- Sciences 39:112–20. doi:10.1016/j.tibs.2013.12.007.
rotoxicity in children aged 7 years. American Journal of Haut, M. W., L. A. Morrow, D. Pool, T. S. Callahan, J. S.
Epidemiology 150:301–05. doi:10.1093/oxfordjournals.aje. Haut, and M. D. Franzen. 1999. Neurobehavioral effects of
a010002. acute exposure to inorganic mercury vapor. Appl
Grandjean, P., K. Murata, E. Budtz-Jorgensen, and P. Weihe. Neuropsychol 6:193–200. doi:10.1207/s15324826an0604_1.
2004. Cardiac autonomic activity in methylmercury neu- Havarinasab, S., E. Björn, J. Ekstrand, and P. Hultman. 2007.
rotoxicity: 14-years follow-up of a Faroese birth cohort. Dose and Hg species determine the T-helper cell activation
The Journal of Pediatrics 144:189–176. doi:10.1016/j. in murine autoimmunity. Toxicology 229:23–32.
jpeds.2003.10.058. doi:10.1016/j.tox.2006.09.006.
Grandjean, P., P. Weihe, P. J. Jorgensen, T. Clarkson, E. Havarinasab, S., B. Häggqvist, E. Björn, K. M. Pollard, and P.
Cernichiari, and T. Videro. 1992. Impact of maternal sea- Hultman. 2005. Immunosuppressive and autoimmune
food diet on fetal exposure to mercury, selenium and lead. effects of Thimerosal in mice. Toxicology and Applied
Archives of Environmental Health 47:185–95. doi:10.1080/ Pharmacology 204:109–21. doi:10.1016/j.taap.2004.08.019.
00039896.1992.9938348. Havarinasab, S., and P. Hultman. 2005. Organic mercury
Grandjean, P., P. Weihe, and R. F. White. 1995. Milestone compounds and autoimmunity. Autoimmun Reviews
development in infants exposed to methylmercury from 4:270–75. doi:10.1016/j.autrev.2004.12.001.
human milk. Neurotoxicology 16:27–33. Havarinasab, S., L. Lambertson, J. Qvarnström, and P.
Grandjean, P., P. Weihe, R. F. White, F. Debes, S. Araki, K. Hultman. 2004. Dose-response study of Thimerosal-induce
Yokoyama, K. Murata, N. Sørensen, R. Dahl, and P. J. murine systemic autoimmunity. Toxicology and Applied
Jørgensen. 1997. Cognitive deficit in 7-year-old children Pharmacology 194:169–79. doi:10.1016/j.taap.2003.09.006.
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B 29

Heron, J., and J. Golding. 2004. Thimerosal exposure in of Biological Chemistry 281:12132–42. doi:10.1074/jbc.
infants and developmental disorders: A prospective cohort M511198200.
study in the United Kingdom does not support a causal Janssen-Heininger, Y. M., B. T. Mossman, N. H. Heintz, H. J.
association. Pediatrics 114:577–83. doi:10.1542/peds.2003- Forman, B. Kalyanaraman, T. Finkel, J. S. Stamler, S. G.
1176-L. Rhee, and A. Van Der Vliet. 2008. Redox-based regulation
Heyer, N. J., D. Echeverria, A. C. Bittner Jr., F. M. Farin, C. of signal transduction: Principles, pitfalls, and promises.
C. Garabedian, and J. S. Woods. 2004. Chronic low-level Free Radical Biologic Medica 45:1–17. doi:10.1016/j.
mercury exposure, BDNF polymorphism, and associations freeradbiomed.2008.03.011.
with self-reported symptoms and mood. Toxicological Jarosínska, D., M. Horvat, G. Sällsten, B. Mazzolai, B. Dąbkowska,
Sciences 81:354–63. doi:10.1093/toxsci/kfh220. A. Prokopowicz, M. Biesiada, and L. Barregård. 2008. Urinary
Heyer, N. J., D. Echeverria, F. M. Farin, and J. S. Woods. mercury and biomarkers of early renal dysfunction in envir-
2008. The association between serotonin transporter gene onmentally and occupationally exposed adults: A three-coun-
promoter polymorphism (5-HTTLPR), self-reported try study. Environmental Research 108:224–32. doi:10.1016/j.
symptoms, and dental mercury exposure. Journal envres.2008.06.005.
Toxicogical Environment Health A 71:1318–26. Jayaprakash, K. 2009. Mercury vapor inhalation and its effect
doi:10.1080/15287390802240850. on glutathione peroxidase in goldsmiths exposed occupa-
Heyer, N. J., D. Echeverria, M. D. Martin, F. M. Farin, and J. tionally. Toxicogical Industrial Health 25:463–65.
S. Woods. 2009. Catechol O-Methyltransferase (COMT) doi:10.1177/0748233709106769.
VAL158MET functional polymorphism, dental mercury Jebbett, N. J., J. W. Hamilton, M. D. Rand, and F. Eckenstein.
exposure, and self-reported symptoms and mood. Journal 2013. Low level methylmercury enhances CNTF-evoked
Toxicogical Environment Health A 72:599–609. STAT3 signaling and glial differentiation in cultured cor-
doi:10.1080/15287390802706405. tical progenitor cells. Neurotoxicology 38:91–100.
Hoesel, B., and J. A. Schmid. 2013. The complexity of NF- doi:10.1016/j.neuro.2013.06.008.
kappaB signaling in inflammation and cancer. Molecular Jin, X., N. Hidiroglou, E. Lok, M. Taylor, K. Kapal, N. Ross,
Cancer 12:86. doi:10.1186/1476-4598-12-86. K. Sarafin, A. Lau, A. De Souza, H. M. Chan, and E.
Hoffman, D. J., M. G. Spalding, and P. C. Frederick. 2005. Mehta. 2012. Dietary selenium (Se) and vitamin E (V(E))
Subchronic effects of methylmercury on plasma and organ supplementation modulated methylmercury-mediated
biochemistries in great egret nestlings. Environmental changes in markers of cardiovascular diseases in rats.
Toxicology and Chemistry 24:3078–84. doi:10.1897/04- Cardiovascular Toxicology 12:10–24. doi:10.1007/s12012-
570.1. 011-9134-y.
Horecker, B. L. 2002. The pentose phosphate pathway. Julvez, J., G. D. Smith, J. Golding, S. Ring, B. S. Pourcain, J. R.
Journal Biologic Chemical 277:47965–71. doi:10.1074/jbc. Gonzalez, and P. Grandjean. 2013. Prenatal methylmer-
X200007200. cury exposure and genetic predisposition to cognitive def-
Huang, C. F., S. H. Liu, C. J. Hsu, and S. Y. Lin-Shiau. 2011. icit at age 8 years. Epidemiology 24:643–50. doi:10.1097/
Neurotoxicological effects of low-dose methylmercury and EDE.0b013e31829d5c93.
mercuric chloride in developing offspring mice. Toxicology Kalev-Zylinska, M. L., T. N. Green, M. C. Morel-Kopp, P. P.
Letters 201:196–204. doi:10.1016/j.toxlet.2010.12.016. Sun, Y. E. Park, A. Lasham, M. J. During, and C. M. Ward.
Hussain, S., D. A. Rodgers, H. M. Duhart, and S. F. Ali. 1997. 2014. N-methyl-d-aspartate receptors amplify activation
Mercuric chloride-induced reactive oxygen species and its and aggregation of human platelets. Thrombosis Research
effect on antioxidant enzymes in different regions of rat 133:837–47. doi:10.1016/j.thromres.2014.02.011.
brain. Journal Environment Sciences Health, Particle B Kendall, R. J., T. A. Anderson, R. J. Baker, C. M. Bens, J. A.
32:395–409. doi:10.1080/03601239709373094. Carr, G. P. C. Chiodo, R. L. Dickerson, K. R. Dixon, L. T.
Hviid, A., M. Stellfeld, J. Wohlfahrt, and M. Melbye. 2003. Frame, M. J. Hooper, C. F. Martin, S. T. McMurry, R.
Association between thimerosal-containing vaccine and aut- Patino, E. E. Smith, and C. W. Theodorakis. 2001.
ism. Jam Medica Association 290:1763–66. doi:10.1001/ Ecotoxicology. In Casarett and Doull’s toxicology. The
jama.290.13.1763. basic science of poisons. Chapter 29, ed. C. D. Klaassen,
Hwang, G. W., J. Y. Lee, K. Ryoke, F. Matsuyama, J. M. Kim, 6th ed., 1013–45. New York: McGraw-Hill.
T. Takahashi, and A. Naganuma. 2011. Gene expression Khan, M. A. K., and F. Wang. 2009. Mercury-selenium com-
profiling using DNA microarray analysis of the cerebellum pounds and their toxicological significance: Toward a
of mice treated with methylmercury. Journal Toxicogical molecular understanding of the mercury-selenium antag-
Sciences 36:389–91. doi:10.2131/jts.36.389. onism. Environmental Toxicology and Chemistry 28:1567–
Jain, A., G. J. Flora, R. Bhargava, and S. J. Flora. 2012. 77. doi:10.1897/08-375.1.
Influence of age on arsenic-induced oxidative stress in Kim, M. S., T. Takahashi, J. Y. Lee, G. W. Hwang, and A.
rat. Biologic Trace Elements Researcher 149:382–90. Naganuma. 2012. Methylmercury induces CCL2 expres-
doi:10.1007/s12011-012-9432-7. sion through activation of NF-kappaB in human 1321N1
Jain, A. K., and A. K. Jaiswal. 2006. Phosphorylation of astrocytes. Journal Toxicogical Sciences 37:1275–78.
tyrosine 568 controls nuclear export of Nrf2. The Journal doi:10.2131/jts.37.1275.
30 V. BRANCO ET AL.

Kim, S. A., C. K. Jeon, and D. M. Paek. 2008. Hair mercury Lebel, J., D. Mergler, F. Branches, M. Lucotte, M. Amorim,
concentrations of children and mothers in Korea: Implication and F. Larribe. 1998. Neurotoxic effects of low-level methyl
for exposure and evaluation. The Science of the Total mercury contamination in the Amazonian Basin.
Environment 402:36–42. doi:10.1016/j.scitotenv.2008.04.010. Environmental Research 79:20–32. doi:10.1006/
Kobal, A. B., M. Horvat, M. Prezelj, A. S. Briski, M. Krsnik, T. enrs.1998.3846.
Dizdarevic, D. Mazej, I. Falnoga, V. Stibilj, N. Arneric, D. Lee, B. E., Y. C. Hong, H. Park, M. Ha, B. S. Koo, N. Chang,
Kobal, and J. Osredkar. 2004. The impact of long-term past Y. M. Roh, B. N. Kim, Y. J. Kim, B. M. Kim, S. J. Jo, and E.
exposure to elemental mercury on antioxidative capacity H. Ha. 2010. Interaction between GSTM1/GSTT1 poly-
and lipid peroxidation in mercury miners. Journal Trace morphism and blood mercury on birth weight. Environ
Elements Medica Biologic 17:262–74. doi:10.1016/S0946- Health Persp 118:437–43. doi:10.1289/0900731.
672X(04)80028-2. Lee, J. Y., G. W. Hwang, M. S. Kim, T. Takahashi, and A.
Kobal, A. B., M. Prezelj, M. Horvat, M. Krsnik, D. Gibicar, Naganuma. 2012. Methylmercury induces a brain-specific
and J. Osredkar. 2008. Glutathione level after long-term increase in chemokine CCL4 expression in mice. Journal
occupational elemental mercury exposure. Environmental Toxicogical Sciences 37:1279–82. doi:10.2131/jts.37.1279.
Research 107:115–23. doi:10.1016/j.envres.2007.07.001. Lillig, C. H., and A. Holmgren. 2007. Thioredoxin and related
Kobayashi, A., M. I. Kang, Y. Watai, K. I. Tong, T. Shibata, K. molecules – from biology to health and disease.
Uchida, and M. Yamamoto. 2006. Oxidative and electro- Antioxidants Redox Signalling 9:25–47. doi:10.1089/
philic stresses activate Nrf2 through inhibition of ubiqui- ars.2007.9.25.
tination activity of Keap1. Molecular and Cellular Biology Lin, T. H., Y. L. Huang, and S. F. Huang. 1996. Lipid perox-
26:221–29. doi:10.1128/MCB.26.1.221-229.2006. idation in liver of rats administrated with methyl mercuric
Kong, X., S. Wang, H. Jiang, G. Nie, and X. Li. 2012. chloride. Biologic Trace Elements Researcher 54:33–41.
Responses of acid/alkaline phosphatase, lysozyme, and cat- doi:10.1007/BF02785318.
alase activities and lipid peroxidation to mercury exposure Lin, Y. S., G. Ginsberg, J. W. Lin, and B. Sonawane. 2014.
during the embryonic development of goldfish Carassius Mercury exposure and omega-3 fatty acid intake in rela-
auratus. Aquatic Toxicology 120/121:119–25. doi:10.1016/j. tion to renal function in the US population. International
aquatox.2012.05.005. Journal Hygiene Environment Health 217:465–72.
Kristensen, A. K. B., J. F. Thomsen, and S. Mikkelsen. 2014. A doi:10.1016/j.ijheh.2013.09.004.
review of mercury exposure among artisanal small-scale Lindow, S. W., R. Knight, J. Batty, and S. J. Haswell. 2003.
gold miners in developing countries. International Maternal and neonatal hair mercury concentrations: The
Archives of Occupational and Environmental Health effect of dental amalgam. British Journal of Obstetrics and
87:579–90. doi:10.1007/s00420-013-0902-9. Gynaecology 110:287–91. doi:10.1046/j.1471-
Kutscher, D. J., M. E. Del Castillo Busto, N. Zinn, A. Sanz-Medel, 0528.2003.02257.x.
and J. Bettmer. 2008. Protein labelling with mercury tags: Liu, X., G. F. Nordberg, and T. Jin. 1992. Increased urinary
Fundamental studies on ovalbumin derivatised with p-hydro- excretion of zinc and copper by mercuric chloride injec-
xymercuribenzoic acid (pHMB). Journal Analysis Atom tion in rats. Biometals 5:17–22. doi:10.1007/BF01079693.
Spectrometry 23:1359–64. doi:10.1039/b806118a. Llop, S., F. Ballester, and K. Broberg. 2015. Effect of gene-
Kwon, O. S., and Y. J. Park. 2003. In vitro and in vivo dose- merury interactions on mercury toxicokinetics and neuro-
dependent inhibition of methylmercury on glutamine toxicity. Current Environment Heath Reports 2:179–94.
synthetase in the brain of different species. doi:10.1007/s40572-015-0047-y.
Environmental Toxicology and Pharmacology 14:17–24. Llop, S., K. Engström, F. Ballester, E. Franforte, A.
doi:10.1016/S1382-6689(03)00006-1. Alhamdow, F. Pisa, J. S. Tratnik, D. Mazej, M. Murcia,
Laffont, L., J. E. Sonke, L. Maurice, S. L. Monrroy, J. M. Rebagliato, M. Bustamante, J. Sunyer, A. Sofianou-
Chincheros, D. Amouroux, and P. Behra. 2011. Hg specia- Katsoulis, A. Prasouli, E. Antonopoulou, I. Antoniadou,
tion and stable isotope signatures in human hair as a tracer S. Nakou, F. Barbone, M. Horvat, and K. Broberg. 2014.
for dietary and occupational exposure to mercury. Polymorphisms in ABC transporter genes and concentra-
Environmental Science & Technology 45:9910–16. tions of mercury in newborns–evidence from two
doi:10.1021/es202353m. Mediterranean birth cohorts. Plos One 9:e97172.
LaKind, J. S., R. L. Brent, M. L. Dourson, S. Kacew, G. Koren, doi:10.1371/journal.pone.0097172.
B. Sonawane, A. J. Tarzian, and K. Uhl. 2005. Human milk Lu, J., E. H. Chew, and A. Holmgren. 2007. Targeting thior-
biomonitoring data: Interpretation and risk assessment edoxin reductase is a basis for cancer therapy by arsenic
issues. Journal Toxicogical Environment Health A trioxide. Proceedings National Academic Sciences USA
68:1713–69. doi:10.1080/15287390500225724. 104:12288–93. doi:10.1073/pnas.0701549104.
LeBel, C. P., S. F. Ali, and S. C. Bondy. 1992. Deferoxamine Lu, J., and A. Holmgren. 2009. Selenoproteins. The Journal of
inhibits methyl mercury-induced increases in reactive oxy- Biological Chemistry 284:723–27. doi:10.1074/jbc.
gen species formation in rat brain. Toxicology and Applied R800045200.
Pharmacology 112:161–65. doi:10.1016/0041-008X(92) Lund, B., D. M. Miller, and J. S. Woods. 1991. Mercury-
90292-Z. induced H2O2 production and lipid peroxidation in vitro
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B 31

in rat kidney and mitochondria. Biochemical Pharmacology Martinez-Finley, E. J., S. Caito, J. C. Slaughter, and M. Aschner.
42:S181–187. doi:10.1016/0006-2952(91)90408-W. 2013. The role of skn-1 in methylmercury-induced latent
Lund, B., D. M. Miller, and J. S. Woods. 1993. Studies on Hg(II)- dopaminergic neurodegeneration. Neurochemical Research
induced H2O2 formation and oxidative stress in vivo and in 38:2650–60. doi:10.1007/s11064-013-1183-0.
vitro in rat kidney mitochondria. Biochemical Pharmacology McMurtry, R. J., W. R. Snodgrass, and J. R. Mitchell. 1978. Renal
45:2017–24. doi:10.1016/0006-2952(93)90012-L. necrosis, glutathione depletion, and covalent binding after
Madsen, K. M., M. B. Lauritsen, C. B. Pedersen, P. Thorsen, acetaminophen. Toxicology and Applied Pharmacology 46:87–
A.-M. Plesner, P. H. Andersen, and P. B. Mortensen. 2003. 100. doi:10.1016/0041-008X(78)90139-4.
Thimerosal and the occurrence of autism: Negative ecolo- Meister, A. 1988. Glutathione metabolism and its selective
gical evidence from Danish population-based data. modification. The Journal of Biological Chemistry
Pediatrics 112:604–06. doi:10.1542/peds.112.3.604. 263:17205–08.
Magos, L. 1997. Physiology and toxicology of mercury. In Merrick, B. A. 2006. Toxicoproteomics in liver injury and
Metal Ions in biological systems, volume 34: Mercury and its inflammation. Annals N Y Academic Sciences 1076:707–17.
effects on environment and biology, eds. A. Sigel, and H. doi:10.1196/annals.1371.017.
Sigel, 321–70. New York: Marcel Dekker Inc. Meyer-Baron, M., M. Schaeper, and A. Seeber. 2002. A meta-
Mahboob, M., K. F. Shireen, A. Atkinson, and A. T. Khan. analysis for neurobehavioural results due to occupational
2001. Lipid peroxidation and antioxidant enzyme activity mercury exposure. Archives of Toxicology 76:127–36.
in different organs of mice exposed to low level of mer- doi:10.1007/s00204-002-0327-9.
cury. Journal Environment Sciences Health, Particle B Miyamoto, K., H. Nakanishi, S. Moriguchi, N. Fukuyama,
36:687–97. doi:10.1081/PFC-100106195. K. Eto, J. Wakamiya, K. Murao, K. Arimura, and M.
Maier, A., R. E. Savage Jr., and L. T. Haber. 2004. Assessing Osame. 2001. Involvement of enhanced sensitivity of
biomarker use in risk assessment–a survey of practitioners. N-methyl-D-aspartate receptors in vulnerability of
Assessing biomarker use in risk assessment - a survey of developing cortical neurons to methylmercury neuro-
practitioners. Journal Toxicogical Environment Health A toxicity. Brain Research 901:252–58. doi:10.1016/S0006-
67:687–95. doi:10.1080/15287390490428161. 8993(01)02281-8.
Manfroi, C. B., F. D. Schwalm, V. Cereser, F. Abreu, A. Moreira, E. L., J. De Oliveira, M. F. Dutra, D. B. Santos, C. A.
Oliveira, L. Bizarro, J. B. T. Rocha, M. E. Frizzo, D. O. Goncalves, E. M. Goldfeder, A. F. De Bem, R. D. Prediger,
Souza, and M. Farina. 2004. Maternal milk as methylmer- M. Aschner, and M. Farina. 2012. Does methylmercury-
cury source for suckling mice: Neurotoxic effects involved induced hypercholesterolemia play a causal role in its
with the cerebellar glutamatergic system. Toxicological neurotoxicity and cardiovascular disease? Toxicological
Sciences 81:172–78. doi:10.1093/toxsci/kfh201. Sciences 130:373–82. doi:10.1093/toxsci/kfs252.
Marks, G. S. 1985. Exposure to toxic agents: The heme Morrow, J. D., K. E. Hill, R. F. Burk, T. M. Nammour, K. F.
biosynthetic pathway and hemoproteins as indicator. Badr, and L. J. Roberts 2nd. 1990. A series of prostaglandin
Critical Reviews in Toxicology 15:151–79. doi:10.3109/ F2-like compounds are produced in vivo in humans by a
10408448509029323. non-cyclooxygenase, free radical-catalyzed mechanism.
Marques, R. C., J. V. Bernardi, L. Abreu, and J. G. Dórea. Proceedings National Academic Sciences USA 87:9383–87.
2015. Neurodevelopment outcomes in children exposed to doi:10.1073/pnas.87.23.9383.
organic mercury from multiple sources in a tin-ore mine Mottet, N. K., M. E. Vahter, J. S. Charleston, and L. T.
environment in Brazil. A Rch Environment Contamination Friberg. 1997. Metabolism of methylmercury in the brain
Toxicogical 68:432–41. doi:10.1007/s00244-014-0103-x. and its toxicological significance. In Metal Ions in biologi-
Marques, R. C., J. V. Bernardi, J. G. Dórea, R. S. Leão, and O. cal systems, volume 34: mercury and its effects on environ-
Malm. 2013. Mercury transfer during pregnancy and ment and biology, eds. A. Sigel, and H. Sigel, 371–403. New
breastfeeding: Hair mercury concentrations as biomarker. York: Marcel Dekker Inc.
Biologic Trace Elements Researcher 154:326–32. Motts, J. A., D. L. Shirley, E. K. Silbergeld, and J. F. Nyland.
doi:10.1007/s12011-013-9743-3. 2014. Novel biomarkers of mercury-induced autoimmune
Marques, R. C., J. G. Dórea, M. F. Fonseca, W. R. Bastos, and dysfunction: A cross-sectional study in Amazonian Brazil.
O. Malm. 2007. Hair mercury in breast-fed infants exposed Environmental Research 132:12–18. doi:10.1016/j.
to Thimerosal-preserved vaccines. European Journal of envres.2014.03.024.
Pediatrics 166:935–41. doi:10.1007/s00431-006-0362-2. Mustacich, D., and G. Powis. 2000. Thioredoxin reductase.
Martinez, C. S., A. G. Escobar, J. G. Torres, D. S. Brum, F. W. The Biochemical Journal 346:1–8. doi:10.1042/bj3460001.
Santos, M. J. Alonso, M. Salaices, D. V. Vassallo, F. M. Myers, G., P. W. Davidson, C. F. Shamlaye, C. D. Axtell, E.
Pecanha, F. G. Leivas, and G. A. Wiggers. 2014. Chronic Cernichiari, O. Choisy, A. Choi, C. Cox, and T. W.
exposure to low doses of mercury impairs sperm quality Clarkson. 1997. Effects of prenatal methylmercury expo-
and induces oxidative stress in rats. Journal Toxicogical sure from a high fish diet on developmental milestones in
Environment Health A 77:143–54. doi:10.1080/ the Seychelles child development study. Neurotoxicology
15287394.2014.867202. 18:819–30.
32 V. BRANCO ET AL.

Myers, G. J., P. W. Davidson, C. Cox, C. Shamlaye, D. (Amazon): Impact on hair mercury. Annals of Human
Palumbo, E. Cernichiari, J. Sloane-Reeves, G. E. Wilding, Biology 37:629–42. doi:10.3109/03014460903525177.
J. Kost, L.-S. Haung, and T. W. Clarkson. 2003. Prenatal Oppedisano, F., M. Galluccio, and C. Indiveri. 2010.
methylmercury exposure from ocean fish consumption in Inactivation by Hg2+ and methylmercury of the gluta-
the Seychelles Child Development Study. Lancet 361:1686– mine/amino acid transporter (ASCT2) reconstituted in
92. doi:10.1016/S0140-6736(03)13371-5. liposomes: Prediction of the involvement of a CXXC
Nath, K. A., A. J. Croatt, S. Likely, T. W. Behrens, and D. motif by homology modelling. Biochemical Pharmacology
Warden. 1996. Renal oxidant injury and oxidant response 80:1266–73. doi:10.1016/j.bcp.2010.06.032.
induced by mercury. Kidney International 50:1032–43. Ou, L., C. Chen, L. Chen, H. Wang, T. Yang, H. Xie, Y. Tong,
doi:10.1038/ki.1996.406. D. Hu, W. Zhang, and X. Wang. 2015. Low-level prenatal
National Research Council (NRC). 1987. Biological markers mercury exposure in north China: An exploratory study of
in environmental health research. Environment Health anthropometric effects. Environmental Science &
Persp 74:3–9. Technology 49:6899–908. doi:10.1021/es5055868.
National Research Council (NRC). 2000. Toxicological effects of Pantano, C., N. L. Reynaert, A. Van Der Vliet, and Y. M.
Methylmercury. Washington, DC: National Academy Press. Janssen-Heininger. 2006. Redox-sensitive kinases of the
Neal, P. A. 1938. Mercury poisoning from the public health nuclear factor-kappaB signaling pathway. Antioxidants &
viewpoint. American Journal Public Health 28:907–15. Redox Signaling 8:1791–806. doi:10.1089/ars.2006.8.1791.
doi:10.2105/AJPH.28.8.907. Papp, L. V., J. Lu, A. Holmgren, and K. K. Khanna. 2007.
Ng, S., C. C. Lin, Y. H. Hwang, W. S. Hsieh, H. F. Liao, and From selenium to selenoproteins: Synthesis, identity and
P. C. Chen. 2013. Mercury, APOE, and children’s neuro- their role in human health. Antioxidants & Redox Signaling
development. Neurotoxicology 37:85–92. doi:10.1016/j. 9:775–806. doi:10.1089/ars.2007.1528.
neuro.2013.03.012. Parajuli, R. P., J. M. Goodrich, H. N. Chou, S. E. Gruninger,
Ni, M., X. Li, Z. Yin, H. Jiang, M. Sidoryk-Wegrzynowicz, D. D. C. Dolinoy, A. Franzblau, and N. Basu. 2016. Genetic
Milatovic, J. Cai, and M. Aschner. 2010. Methylmercury polymorphisms are associated with hair, blood, and urine
induces acute oxidative stress, altering Nrf2 protein level in mercury levels in the American Dental Association (ADA)
primary microglial cells. Toxicological Sciences 116:590– study participants. Environmental Research 149:247–58.
603. doi:10.1093/toxsci/kfq126. doi:10.1016/j.envres.2015.11.032.
Nielsen, J. B., and P. Hultman. 2002. Mercury-induced auto- Park, S. T., K. T. Lim, Y. T. Chung, and S. U. Kim. 1996.
immunity in mice. Environment Health Persp 110:877–81. Methylmercury-induced neurotoxicity in cerebral neuron
doi:10.1289/ehp.02110s5877. culture is blocked by antioxidants and NMDA receptor
Niture, S. K., R. Khatri, and A. K. Jaiswal. 2014. Regulation of antagonists. Neurotoxicology 17:37–45.
Nrf2-an update. Free Radical Biologic Medica 66:36–44. Pastore, A., G. Federici, E. Bertini, and F. Piemonte. 2003.
doi:10.1016/j.freeradbiomed.2013.02.008. Analysis of glutathione: Implication in redox and detox-
Nunes, E., A. Cavaco, and C. Carvalho. 2014a. Children’s ification. Clinica Chimica Acta 333:19–39. doi:10.1016/
health risk and benefits of fish consumption: Risk indices S0009-8981(03)00200-6.
based on a diet diary follow-up of two weeks. Journal Paustenbach, D. J. 2001. The practice of exposure assessment.
Toxicogical Environment Health A 77:103–14. In Principles and methods of toxicology. Chapter 9, ed. A.
doi:10.1080/15287394.2014.866926. W. Hayes, 387–448. Philadelphia: Taylor & Francis.
Nunes, E., A. Cavaco, and C. Carvalho. 2014b. Exposure Perrin-Nadif, R., M. Dusch, C. Koch, P. Schmitt, and J. M.
assessment of pregnant Portuguese women to methylmer- Mur. 1996. Catalase and superoxide dismutase activities as
cury through the ingestion of fish: Cross-sectional survey biomarkers of oxidative stress in workers exposed to mer-
and biomarker validation. Journal Toxicogical Environment cury vapors. Journal Toxicogical Environment Health
Health A 77:133–42. doi:10.1080/15287394.2014.867200. 7:107–19. doi:10.1080/009841096161366.
Nyland, J. F., M. Fillion, F. Barbosa, D. L. Shirley, C. Chine, Piccoli, C., A. D’Aprile, R. Scrima, L. Ambrosi, R. Zefferino,
M. Lemire, D. Mergler, and E. K. Silbergeld. 2011. and N. Capitanio. 2012. Subcytotoxic mercury chloride
Biomarkers of methylmercury exposure immunotoxicity inhibits gap junction intercellular communication by a
among fish consumers in Amazonian Brazil. Environment redox- and phosphorylation-mediated mechanism. Free
Health Persp 119:1733–38. doi:10.1289/ehp.1103741. Radical Biologic Medica 52:916–27. doi:10.1016/j.
Ohno, T., M. Sakamoto, T. Kurosawa, M. Dakeishia, T. Iwata, freeradbiomed.2011.12.018.
and K. Murataa. 2007. Total mercury levels in hair, toenail, Pingree, S. D., P. L. Simmonds, K. T. Rummel, and J. S. Woods.
and urine among women free from occupational exposure 2001. Quantitative evaluation of urinary porphyrins as a mea-
and their relations to renal tubular function. Environmental sure of kidney mercury content and mercury body burden
Research 103:191–97. doi:10.1016/j.envres.2006.06.009. during prolonged methylmercury exposure in rats.
Oliveira, R. C., J. G. Dórea, J. V. Bernardi, W. R. Bastos, R. Toxicological Sciences 61:234–40. doi:10.1093/toxsci/61.2.234.
Almeida, and A. G. Manzatto. 2010. Fish consumption by Pinheiro, M. C., B. M. Macchi, J. L. Vieira, T. Oikawa, W. W.
traditional subsistence villagers of the Rio Madeira Amoras, G. A. Guimarães, C. A. Costa, M. E. Crespo-
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B 33

López, A. M. Herculano, L. C. Silveira, and J. L. M. Do Rodrigues, J., V. Branco, J. Lu, A. Holmgren, and C.
Nascimento. 2008. Mercury exposure and antioxidant Carvalho. 2015. Toxicological effects of thiomersal and
defenses in women: A comparative study in the Amazon. ethylmercury: Inhibition of the thioredoxin system and
Environmental Research 107:53–59. doi:10.1016/j. NADP+-dependent dehydrogenases of the pentose phos-
envres.2007.08.007. phate pathway. Toxicology and Applied Pharmacology
Piotrowski, J. K., B. Trojanowska, J. M. Wiśniewska-Knypl, 286:216–23. doi:10.1016/j.taap.2015.05.002.
and W. Bolanowska. 1974. Mercury binding in the kidney Romero, D. L., B. J. Mounho, F. T. Lauer, J. L. Born, and S.
and liver of rats repeatedly exposed to mercuric chloride: W. Burchiel. 1997. Depletion of glutathione by benzo(a)
Induction of metallothionein by mercury and cadmium. pyrene metabolites, ionomycin, thapsigargin, and phorbol
Toxicology and Applied Pharmacology 27:11–19. myristate in human peripheral blood mononuclear cells.
doi:10.1016/0041-008X(74)90169-0. Toxicology and Applied Pharmacology 144:62–69.
Pollack, A. Z., S. L. Mumford, P. Mendola, N. J. Perkins, Y. doi:10.1006/taap.1997.8113.
Rotman, J. Wactawski-Wende, and E. F. Schisterman. Rooney, J. P. K. 2007. The role of thiols, dithiols, nutritional
2015. Kidney biomarkers associated with blood lead, mer- factors and interacting ligands in the toxicology of mer-
cury, and cadmium in premenopausal women: A prospec- cury. Toxicology 234:145–56. doi:10.1016/j.tox.2007.02.016.
tive cohort study. Journal Toxicogical Environment Health Ruszkiewicz, J. A., A. B. Bowman, M. Farina, J. B. Rocha, and
A 78:119–31. doi:10.1080/15287394.2014.944680. M. Aschner. 2016. Sex-and structure-specific differences in
Pollack, A. Z., E. F. Schisterman, L. R. Goldman, S. L. antioxidant responses to methylmercury during early
Mumford, N. J. Perkins, M. S. Bloom, C. B. Rudra, R. W. development. NeuroToxicology 56:118–26. doi:10.1016/j.
Browne, and J. Wactawski-Wende. 2012. Relation of blood neuro.2016.07.009.
cadmium, lead, and mercury levels to biomarkers of lipid Sakai, T. 2000. Biomarkers of lead exposure. Industrial Health
peroxidation in premenopausal women. American Journal 38:127–42. doi:10.2486/indhealth.38.127.
of Epidemiology 175:645–52. doi:10.1093/aje/kwr375. Samir, A. M., and W. M. Aref. 2011. Impact of occupational
Pollard, K. M., and P. Hultman. 1997. Effects of mercury in the exposure to elemental mercury on some antioxidative
immune system. In Metal Ions in biological systems, volume enzymes among dental staff. Toxicogical Industrial Health
34: Mercury and its effects on environment and biology, eds. A. 27:779–86. doi:10.1177/0748233710397420.
Sigel, and H. Sigel, 421–60. New York: Marcel Dekker Inc. Sanders, A. P., H. H. Burris, A. C. Just, V. Motta, C.
Pollard, K. M., and D. H. Kono. 2013. Requirements for Amarasiriwardena, K. Svensson, E. Oken, M. Solano-
innate immune pathways in environmentally induced Gonzalez, A. Mercado-Garcia, I. Pantic, J. Schwartz, M.
autoimmunity. BMC Medicine 11:100. doi:10.1186/1741- M. Tellez-Rojo, A. A. Baccarelli, and R. O. Wright. 2015.
7015-11-100. Altered miRNA expression in the cervix during pregnancy
Qu, H., T. Syversen, M. Aschner, and U. Sonnewald. 2003. associated with lead and mercury exposure. Epigenomics
Effect of methylmercury on glutamate metabolism in cer- 7:885–96. doi:10.2217/epi.15.54.
ebellar astrocytes in culture. Neurochemistry International Santos, A. P. M., M. L. Mateus, C. M. L. Carvalho, and M. C.
43:411–16. doi:10.1016/S0197-0186(03)00029-9. C. Batoréu. 2007. Biomarkers of exposure and effect as
Ratcliffe, H. E., G. M. Swanson, and L. J. Fischer. 1996. indicators of the interference of selenomethionine on
Human exposure to mercury: A critical assessment of the methylmercury toxicity. Toxicology Letters 169:121–28.
evidence of adverse health effects. Journal Toxicogical doi:10.1016/j.toxlet.2006.12.007.
Environment Health 49:221–70. doi:10.1080/ Satoh, H. 2000. Occupational and environmental toxicology
00984108.1996.11667600. of mercury and its compounds. Industrial Health 38:153–
Rigobello, M. P., G. Scutari, A. Folda, and A. Bindoli. 2004. 64. doi:10.2486/indhealth.38.153.
Mitochondrial thioredoxin reductase inhibition by gold(I) Savabieasfahani, M., M. Hoseiny, and S. Goodarzi. 2012.
compounds and concurrent stimulation of permeability Toxic and essential trace metals in first baby haircuts and
transition and release of cytochrome c. Biochemical mother hair from Imam Hossein Hospital Tehran, Iran.
Pharmacology 67:689–96. doi:10.1016/j.bcp.2003.09.038. Bulletin Environment Contamination Toxicogical 88:140–
Robinson, J. F., P. T. Theunissen, D. A. Van Dartel, J. L. 44. doi:10.1007/s00128-011-0487-5.
Pennings, E. M. Faustman, and A. H. Piersma. 2011. Schläwicke-Engström, K., U. Strömberg, T. Lundh, I.
Comparison of MeHg-induced toxicogenomic responses Johansson, B. Vessby, G. Hallmans, S. Skerfving, and K.
across in vivo and in vitro models used in developmental Broberg. 2008. Genetic variation in glutathione-related
toxicology. Reproductive Toxicology 32:180–88. genes and body burden of methylmercury. Environment
doi:10.1016/j.reprotox.2011.05.011. Health Persp 116:734–39. doi:10.1289/ehp.10804.
Rocha, J. B., A. J. Freitas, M. B. Marques, M. E. Pereira, T. Schmidt, C. W. 2006. Signs of the times: Biomarkers in
Emanuelli, and D. O. Souza. 1993. Effects of methylmer- perspective. Environment Health Persp 114:A700–705.
cury exposure during the second stage of rapid postnatal doi:10.1289/ehp.114-a700.
brain growth on negative geotaxis and on delta-aminole- Scientific Committee on Occupational Exposure Limits
vulinate dehydratase of suckling rats. Brazilian Journal of (SCOEL). 2007. Recommendation from the Scientific
Medical and Biological Research 26:1077–83. Committee on Occupational Exposure Limits for
34 V. BRANCO ET AL.

elemental mercury and inorganic divalent mercury com- Sugiura, Y., Y. Hojo, Y. Tamai, and H. Tanaka. 1976.
pounds. European Commission, SCOEL/SUM/84. ec. Selenium protection against mercury toxicity. Binding of
europa.eu/social/BlobServlet?docId=3852&langId=en methylmercury by the selenohydryl-containing ligand.
Shanker, G., J. W. Allen, L. A. Mutkus, and M. Aschner. Journal of the American Chemical Society 98:2339–41.
2001. Methylmercury inhibits cysteine uptake in cultured Suzuki, K. T., and Y. Ogra. 2001. Metabolism of selenium and its
primary astrocytes, but not in neurons. Brain Research interaction with mercury: Mechanisms by a speciation study.
914:159–65. doi:10.1016/S0006-8993(01)02791-3. Phosphorus, Sulfur Silicon Related Elements 171:135–69.
Shanker, G., and M. Aschner. 2001. Identification and char- Sweet, L. I., and J. T. Zelikoff. 2001. Toxicology and immunotox-
acterization of uptake systems for cystine and cysteine in icology of mercury: A comparative review in fish and humans.
cultured astrocytes and neurons: Evidence for methylmer- Journal Toxicogical Environment Health B 4:161–205.
cury-targeted disruption of astrocytic transport. Journal of Tan, Y.-M., J. Sobus, D. Chang, R. Tornero-Velez, M.
Neuroscience Research 66:998–1002. doi:10.1002/jnr.10066. Goldsmith, J. Pleil, and C. Daty. 2012. Reconstructing
Shanker, G., T. Syversen, and M. Aschner. 2003. Astrocyte- human exposures using biomarkers and other “clues’.
mediated methylmercury neurotoxicity. Biologic Trace Journal Toxicogical Environment Health B 15:22–38.
Elements Researcher 95:1–10. doi:10.1385/BTER:95:1:1. Taylor, L. E., A. L. Swerdfeger, and G. D. Eslick. 2014.
Sherratt, P. J., H. C. Huang, T. Nguyen, and C. B. Pickett. 2004. Vaccines are not associated with autism: An evidence-
Role of protein phosphorylation in the regulation of NF-E2- based meta-analysis of case-control and cohort studies.
related factor 2 activity. Meth Enzymol 378:286–301. Vaccine 32:3623–29.
Sikorski, R., T. Paszkowski, and T. Szprengier-Juszkiewicz. Theunissen, P. T., J. L. Pennings, J. F. Robinson, S. M.
1986. Mercury in neonatal scalp hair. The Science of the Claessen, J. C. Kleinjans, and A. H. Piersma. 2011. Time-
Total Environment 57:105–10. doi:10.1016/0048-9697(86) response evaluation by transcriptomics of methylmercury
90015-X. effects on neural differentiation of murine embryonic stem
Silbergeld, E. K., and D. L. Davis. 1994. Role of biomarkers in cells. Toxicological Sciences 122:437–47.
identifying and understanding environmentally induced Tian, L., H. F. Guo, A. Gao, X. T. Lu, and Q. Y. Li. 2009. Effects of
disease. Clinical Chemistry 40:1363–67. mercury released from glold extraction by amalgamation on
Silva De Paula, E., M. F. Carneiro, D. Grotto, L. C. renal function and environment in Shanxi, China. Bulletin
Hernandes, L. M. Antunes, and F. Barbosa Jr. 2016. Environment Contamination Toxicogical 83:71–74.
Protective effects of niacin against methylmercury-induced Timbrell, J. A. 1998. Biomarkers in toxicology. Toxicology
genotoxicity and alterations in antioxidant status in rats. 129:1–12.
Journal Toxicogical Environment Health, A 79:174–83. Toyama, T., Y. Shinkai, D. Sumi, and Y. Kumagai. 2010.
doi:10.1080/15287394.2015.1137264. Carbon monoxide derived from heme oxygenase-2 med-
Simmons-Willis, T. A., A. S. Koh, T. W. Clarkson, and N. iates reduction of methylmercury toxicity in SH-SY5Y
Ballatori. 2002. Transport of a neurotoxicant by molecular cells. Toxicology and Applied Pharmacology 249:86–90.
mimicry: The methylmercury–L-cysteine complex is a sub- Toyama, T., Y. Shinkai, A. Yasutake, K. Uchida, M.
strate for human L-type large neutral amino acid trans- Yamamoto, and Y. Kumagai. 2011a. Isothiocyanates
porter (LAT) 1 and LAT2. The Biochemical Journal reduce mercury accumulation via an Nrf2-dependent
367:239–46. doi:10.1042/bj20020841. mechanism during exposure of mice to methylmercury.
Srivastava, M., S. Singh, and W. T. Self. 2012. Exposure to Environment Health Persp 119:1117–22.
silver nanoparticles inhibits selenoprotein synthesis and Toyama, T., D. Sumi, Y. Shinkai, A. Yasutake, K. Taguchi, K.
the activity of thioredoxin reductase. Environment Health I. Tong, M. Yamamoto, and Y. Kumagai. 2007.
Persp 120:56–61. doi:10.1289/ehp.1103928. Cytoprotective role of Nrf2/Keap1 system in methylmer-
Stehr-Green, P., P. Tullb, M. Stellfeld, P. Mortenson, and D. cury toxicity. Biochemical and Biophysical Research
Simpson. 2003. Autism and thimerosal-containing vac- Communications 363:645–50.
cines: Lack of consistent evidence for an association. Toyama, T., E. Yoshida, Y. Shinkai, and Y. Kumagai. 2011b.
American Journal Prevention Medica 25:101–06. DNA microarray analysis of human neuroblastoma SH-
doi:10.1016/S0749-3797(03)00113-2. SY5Y cells exposed to methylmercury. Journal Toxicogical
Stringari, J., A. K. Nunes, J. L. Franco, D. Bohrer, S. C. Sciences 36:843–45.
Garcia, A. L. Dafre, D. Milatovic, D. O. Souza, J. B. Uno, Y., T. Uchiyama, M. Kurosawa, B. Aleksic, and N. Ozaki.
Rocha, M. M. Aschner, and M. Farina. 2008. Prenatal 2015. Early exposure to the combined measles–mumps–
methylmercury exposure hampers glutathione antioxidant rubella vaccine and thimerosal-containing vaccines and risk
system ontogenesis and causes long-lasting oxidative stress of autism spectrum disorder. Vaccine 33:2511–16.
in the mouse brain. Toxicology and Applied Pharmacology Usuki, F., A. Yamashita, and M. Fujimura. 2011. Post-tran-
227:147–54. doi:10.1016/j.taap.2007.10.010. scriptional defects of antioxidant selenoenzymes cause oxi-
Suda, I., and H. Takahashi. 1992. Degradation of methyl and dative stress under methylmercury exposure. The Journal
ethyl mercury into inorganic mercury by other reactive of Biological Chemistry 286:6641–49.
oxygen species besides hydroxyl radical. Archives of Vahter, M. E., N. K. Mottet, L. T. Friberg, S. B. Lind, J. S.
Toxicology 66:34–39. Charleston, and T. M. Burbacher. 1995. Demethylation of
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART B 35

methylmercury in different brain sites of Macaca fascicularis environmental chemical contaminants. Journal Toxicogical
monkeys during long-term subclinical methylmercury expo- Environment Health B 11:373–517.
sure. Toxicology and Applied Pharmacology 134:273–84. Witte, A. B., K. Anestål, E. Jerremalm, H. Ehrsson, and E. S.
Vanduyn, N., R. Settivari, G. Wong, and R. Nass. 2010. SKN- Arnér. 2005. Inhibition of thioredoxin reductase but not of
1/Nrf2 inhibits dopamine neuron degeneration in a glutathione reductase by the major classes of alkylating
Caenorhabditis elegans model of methylmercury toxicity. and platinum-containing anticancer compounds. Free
Toxicological Sciences 118:613–24. Radical Biologic Medica 39:696–703.
Vas, J., and M. Monestier. 2008. Immunology of mercury. Woods, J. S. 1995. Porphyrin metabolism as indicator of
Annals N Y Academic Sciences 1143:240–67. metal exposure and toxicity. In Handbook of experimental
Vendrell, I., M. Carrascal, M. T. Vilaro, J. Abian, E. pharmacology, vol. 105 – toxicology of metal—biochemical
Rodriguez-Farre, and C. Sunol. 2007. Cell viability and aspects, eds. R. A. Goyer, and M. G. Cherian, 19–52. Berlin,
proteomic analysis in cultured neurons exposed to methyl- Germany: Springer-Verlag.
mercury. Human & Experimental Toxicology 26:263–72. Woods, J. S., M. A. Bowers, and H. A. Davis. 1991. Urinary
Verstraeten, T., R. L. Davis, F. DeStefano, T. A. Lieu, P. H. porphyrin profiles as biomarkers of trace metal exposure
Rhodes, S. B. Black, H. Shinefield, and R. T. Chen. 2003. and toxicity: Studies on urinary porphryin excretion pat-
Safety of thimerosal-containing vaccines: A two-phased terns in rats during prolonged exposure to methylmercury.
study of computerized health maintenance organization Toxicogical Applpharmacol 110:464–76.
databases. Pediatrics 112:1039–48. Woods, J. S., D. Echeverria, N. J. Heyer, P. L. Simmonds, J.
Wagner, C., J. H. Sudati, C. W. Nogueira, and J. B. Rocha. Wilkerson, and F. M. Farin. 2005. The association between
2010. In vivo and in vitro inhibition of mice thioredoxin genetic polymorphisms of coproporphyrinogen oxidase and
reductase by methylmercury. Biometals 23:1171–77. an atypical porphyrinogenic response to mercury exposure in
Wallace, M. A. G., T. M. Kormos, and J. Pleil. 2016. Blood- humans. Toxicogical Applpharmacol 206:113–20.
bornebiomarkers and bioindicators for linking exposure to Woods, J. S., and B. A. Fowler. 1977. Renal porphyrinuria
health effects in environmental health science. Journal during chronic methyl mercury exposure. The Journal of
Toxicogical Environment Health B 19:380–409. Laboratory and Clinical Medicine 90:266–72.
Wang, L., H. Jiang, Z. Yin, M. Aschner, and J. Cai. 2009. Woods, J. S., and B. A. Fowler. 1978. Altered regulation
Methylmercury toxicity and Nrf2-dependent detoxification of mammalian hepatic heme biosynthesis and urinary
in astrocytes. Toxicological Sciences 107:135–43. porphyrin excretion during prolonged exposure to
Wang, Y., J. M. Goodrich, B. Gillespie, R. Werner, N. Basu, sodium arsenate. Toxicology and Applied
and A. Franzblau. 2012. An investigation of modifying Pharmacology 43:361–71.
effects of metallothionein single-nucleotide polymorph- Woods, J. S., N. Heyer, J. E. Russo, M. D. Martin, P. B.
isms on the association between mercury exposure and Pillai, and F. M. Farin. 2013. Modification of neurobe-
biomarker levels. Environment Health Persp 120:530–34. havioral effects of mercury by genetic polymorphisms of
Wang, Y., D. Wang, J. Wu, B. Wang, L. Wang, X. Gao, H. metallothionein in children. Neurotoxicology Teratol
Huang, and H. Ma. 2015. Cinnabar induces renal inflam- 39:36–44.
mation and fibrogenesis in rats. Biomed Researcher Woods, J. S., N. J. Heyer, D. Echeverria, J. E. Russo, M. D.
International 2015:ID280958. Martin, M. F. Bernardo, H. S. Luis, L. Vaz, and F. M. Farin.
Wataha, J. C., J. B. Lewis, V. V. McCloud, M. Shaw, Y. Omata, P. 2012. Modification of neurobehavioral effects of mercury
E. Lockwood, R. L. W. Messer, and J. M. Hansen. 2008. Effect by a genetic polymorphism of coproporphyrinogen oxi-
of mercury (II) on Nrf2, thioredoxin reductase-1 and thior- dase in children. Neurotoxicology Teratol 34:513–21.
edoxin-1 in human monocytes. Dent Materials 24:765–72. Woods, J. S., N. J. Heyer, J. E. Russo, M. D. Martin, and F. M.
Watanabe, C. 2002. Modification of mercury toxicity by Farin. 2014a. Genetic polymorphisms affecting susceptibil-
selenium: Practical importance? The Tohoku Journal of ity to mercury neurotoxicity in children: Summary find-
Experimental Medicine 196:71–77. ings from the Casa Pia children’s amalgam clinical trial.
Watanabe, C., Y. Kasanuma, Y. Dejima, and H. Satoh. 1999a. Neurotoxicology 44:288–302.
The effect of prenatal methylmercury exposure on the Woods, J. S., N. J. Heyer, J. E. Russo, M. D. Martin, P. B.
GSH level and lipid peroxidation in the fetal brain and Pillai, T. K. Bammler, and F. M. Farin. 2014b. Genetic
placenta of mice. The Tohoku Journal of Experimental polymorphisms of catechol-O-methyltransferase modify
Medicine 187:121–26. the neurobehavioral effects of mercury in children.
Watanabe, C., K. Yin, Y. Kasanuma, and H. Satoh. 1999b. In Journal Toxicogical Environment Health A 77:293–312.
utero exposure to methylmercury and Se deficiency con- Woods, J. S., M. D. Martin, B. G. Leroux, T. A. DeRouen, M.
verge on the neurobehavioral outcome in mice. F. Bernardo, H. S. Luis, J. G. Leitão, P. L. Simmonds, D.
Neurotoxicology Teratol 21:83–88. Echeverria, and T. C. Rue. 2009. Urinary porphyrin excre-
Wigle, D. T., T. E. Arbuckle, M. C. Turner, A. Bérubé, Q. Yang, S. tion in children with mercury amalgam treatment:
Liu, and D. Krewski. 2008. Epidemiologic evidence of relation- Findings from the Casa Pia children’s dental amalgam
ships between reproductive and child health outcomes and trial. Journal Toxicogical Environment Health A 72:891–96.
36 V. BRANCO ET AL.

Woods, J. S., M. D. Martin, C. A. Naleway, and D. Echeverria. Yu, R., S. Mandlekar, W. Lei, W. E. Fahl, T. H. Tan, and A.
1993. Urinary porphyrin profiles as a biomarker of mer- N. Kong. 2000. p38 mitogen-activated protein kinase nega-
cury exposure: Studies on dentists with occupational expo- tively regulates the induction of phase II drug-metaboliz-
sure to mercury vapor. Journal Toxicogical Environment ing enzymes that detoxify carcinogens. The Journal of
Health 40:235–46. Biological Chemistry 275:2322–27.
World Health Organization (WHO). 1993. Biomarkers and risk Zabiński, Z., Z. Dabrowski, P. Moszczyński, and J. Rutowski.
assessment: Concepts and principles. International programme 2000. The activity of erythrocyte enzymes and basic indices
on chemical safety, environmental health criteria 155, Geneva, of peripheral blood erythrocytes from workers chronically
82. World Health Organization, Geneva, Switzerland. http:// exposed to mercury vapours. Toxicogical Industrial Health
www.inchem.org/documents/ehc/ehc/ehc155.htm 16:58–64.
World Health Organization (WHO). 2003. Elemental Zalups, R. K. 1998. Basolateral uptake of mercuric conjugates
Mercury and Inorganic Mercury Compounds: Human of N-acetylcysteine and cysteine in the kidney involves the
Health Aspects. Sixty-first report of the Joint FAO/WHO organic anion transport system. Journal Toxicogical
Expert Committee on Food Additives, WHO Technical Environment Health A 55:13–29.
Report Series 922, Geneva, 176. World Health Zalups, R. K. 2000. Molecular interactions with mercury in
Organization, Geneva, Switzerland. http://www.who.int/ the kidney. Pharmacological Reviews 52:113–43.
ipcs/publications/cicad/en/cicad50.pdf Zalups, R. K., and D. W. Barfuss. 2002a. Renal organic anion
World Health Organization/United Nations Environment transport system: A mechanism for the basolateral uptake
Programme (WHO/UNEP). 2008. Guidance for identifying of mercury-thiol conjugates along the pars recta of the
populations at risk from mercury exposure, 170. World proximal tubule. Toxicology and Applied Pharmacology
Health Organization, Geneva, Switzerland: Inter- 182:234–43.
Organization Programme for the Sound Management of Zalups, R. K., and D. W. Barfuss. 2002b. Simultaneous coex-
Chemicals. posure to inorganic mercury and cadmium: A study of the
Wormser, U., B. Brodsky, D. Milatovic, Y. Finkelstein, M. renal and hepatic disposition of mercury and cadmium.
Farina, J. B. Rocha, and M. Aschner. 2012. Protective effect Journal Toxicogical Environment Health A 65:1471–90.
of a novel peptide against methylmercury-induced toxicity Zalups, R. K., D. W. Barfuss, and P. J. Kostyniak. 1992.
in rat primary astrocytes. Neurotoxicology 33:763–68. Altered intrarenal accumulation of mercury in unine-
Yadetie, F., O. A. Karlsen, A. Lanzen, K. Berg, P. Olsvik, C. phrectomized rats treated with methylmercury chloride.
Hogstrand, and A. Goksoyr. 2013. Global transcriptome Toxicology and Applied Pharmacology 115:174–82.
analysis of Atlantic cod (Gadus morhua) liver after in vivo Zalups, R. K., L. Joshee, and C. C. Bridges. 2014. Novel Hg2
+
methylmercury exposure suggests effects on energy meta- -induced Nephropathy in rats and mice lacking Mrp2:
bolism pathways. Aquatic Toxicology 126:314–25. Evidence of axial heterogeneity in the handling of Hg2+
Yang, D., C. Yu-Wei, J. M. Gunn, and N. Belzille. 2008. along the proximal tubule. Toxicological Sciences 142:250–60.
Selenium and mercury in organisms: Interactions and Zemolin, A. P., D. F. Meinerz, M. T. De Paula, D. O.
mechanisms. Environment Reviews 16:71–92. Mariano, J. B. Rocha, A. B. Pereira, T. Posser, and J. L.
Yasutake, A., K. Hirayma, and M. Inoue. 1989. Mechanism of Franco. 2012. Evidences for a role of glutathione perox-
urinary excretion of methylmercury in mice. Archives of idase 4 (GPx4) in methylmercury induced neurotoxicity in
Toxicology 63:479–83. vivo. Toxicology 302:60–67.
Yasutake, A., and M. Nakamura. 2011. Induction by mercury Zhang, J., R. P. Brown, M. Shaw, V. S. Vaidya, Y. Zhou, P.
compounds of metallothioneins in mouse tissues: Espandiari, N. Sadrieh, M. Stratmeyer, J. Keenan, C. G.
Inorganic mercury accumulation is not a dominant factor Kilty, J. V. Bonventre, and P. L. Goering. 2008.
for metallothionein induction in the liver. Journal Immunolocalization of Kim-1, RPA-1, and RPA-2 in
Toxicogical Sciences 36:365–72. kidney of gentamicin-, mercury-, or chromium- treated
Yeter, D., and R. Deth. 2012. ITPKC susceptibility in rats: Relationship to renal distributions of iNOS and
Kawasaki syndrome as a sensitizing factor for autoimmu- nitrotyrosine. Toxicogical Pathologists 36:397–409.
nity and coronary arterial wall relaxation induced by Zhou, Y., V. S. Vaidya, R. P. Brown, J. Zhang, B. A.
Thimerosal’s effects on calcium signaling via IP3. Rosenzweig, K. L. Thompson, T. J. Miller, J. V.
Autoimmun Reviews 11:903–08. Bonventre, and P. L. Goering. 2008. Comparison of kidney
Yin, Z., D. Milatovic, J. L. Aschner, T. Syversen, J. B. Rocha, injury molecule-1 and other nephrotoxicity biomarkers in
D. O. Souza, M. Sidoryk, J. Albrecht, and M. Aschner. urine and kidney following acute exposure to gentamicin,
2007. Methylmercury induces oxidative injury, alterations mercury and chromium. Toxicology Science 101:159–70.
in permeability and glutamine transport in cultured astro- Zimmermann, L. T., D. B. Dos Santos, D. Colle, A. A. Dos
cytes. Brain Research 1131:1–10. Santos, M. A. Hort, S. C. Garcia, L. P. Bressan, D. Bohrer,
Yoshida, E., Y. Abiko, and Y. Kumagai. 2014. Glutathione and M. Farina. 2014. Methionine stimulates motor impair-
adduct of methylmercury activates the Keap1-Nrf2 path- ment and cerebellar mercury deposition in methylmer-
way in SH-SY5Y Cells. Chemical Research in Toxicology cury-exposed mice. Journal Toxicogical Environment
27:1780–86. Health A 77:46–56.

You might also like