You are on page 1of 25

C H A P T E R

20

Effects of Metallic Elements on Reproduction


and Development
PIETRO APOSTOLI AND SIMONA CATALANI

ABSTRACT effects. Use of the diagnostic approach in occupational


medicine is necessary to achieve adequate evidence
Many factors can influence the reproductive or for a causal relationship between exposure and clinical
developmental outcomes of human exposure to metal- findings.
lic elements, including metallic element speciation,
dose, timing, route and duration of exposure, dose-
response relationship, bioavailability, and the distribu- 1 INTRODUCTION
tion and accumulation of metallic elements in various
organs. The reproductive and developmental toxicity asso-
Experimental and epidemiological studies have ciated with metallic elements covers a wide range of
reported the adverse effects of many metallic elements issues, including the number and sources of the metal-
on male and female reproductive function, for exam- lic elements involved; the difficulty in attributing the
ple infertility or subfertility, malformations, abortion, origin of reproductive impairment to males, females,
and developmental alterations. or both; and the different degree and quality of knowl-
Different mechanisms of action have been sug- edge regarding pathogenic mechanisms.
gested: increased reactive oxygen species or decreased The range of these effects in males includes infertil-
activity of the cellular antioxidant defense system; ity, subfertility, and reduced sperm quantity and qual-
direct damage on the testes or ovaries; interaction with ity. For females, they include various birth defects such
the embryonic structure; disruption of reproductive as intrauterine growth retardation, premature aging,
hormones; and, finally, immune suppression and neu- low birth weight, malformation, postnatal death, abor-
rotransmitter alterations. tion, developmental deficits of mental or psychomotor
The targets and effects can be different and specific function, and behavioral effects.
to each metallic element (e.g. neurotoxicity of Pb and Historically, reproductive toxicity has been demon-
methylmercury in the developing brain or the action of strated for metallic elements of widespread occupa-
Cd on the testicle) or similar for large groups of metal- tional and environmental interest, such as lead (Pb),
lic elements (reduced sperm motility caused by As, Cr, mercury (Hg), arsenic (As), and cadmium (Cd). Other
Hg, Ni, and Pb). metallic elements, such as chromium (Cr), cobalt (Co),
Knowledge of these mechanisms may be useful to selenium (Se), copper (Cu), and zinc (Zn) have been
occupational physicians by providing a better under- compared to a double-edged sword in that they are
standing of the dose-response relationship, as well essential on the one hand and toxic on the other. Fur-
as the choice of current indicators of dose and their thermore, there is now concern about other metallic

Handbook on the Toxicology of Metals 4E


http://dx.doi.org/10.1016/B978-0-444-59453-2.00020-2 399 Copyright © 2015 Elsevier B.V. All rights reserved.
400 Pietro Apostoli and Simona Catalani

elements [antimony (Sb), vanadium (V), platinum (Pt), 2  HOW TO MEASURE DOSE AND EFFECT
lithium (Li), and uranium (U)] due to their environ-
mental dispersal. Both the analytical methods adopted, as well as
In order to make a critical evaluation, it is also their quality and accessibility, are crucial for improv-
important to point out the limitations of the studies ing our knowledge of assessing the dose and effect
into the toxic effects of metallic elements on repro- relationships of metallic elements on reproduction and
duction. The majority of published studies consider development in humans.
only single metallic element exposures, whereas Assessment of the occupational or environmental
environmental and occupational conditions are usu- exposure dose to metallic elements is generally car-
ally characterized by combined exposures to several ried out using environmental or biological monitoring
toxic and essential metals, especially through food. procedures (see Chapter 8, “Biological Monitoring and
Evaluation of the possible additive, synergistic, Biomarkers”).
or antagonistic effects appears to be a key consid- Using Pb as an example, one can assess the internal
eration not only from a theoretical but also from a dose by determining the Pb concentration in the blood
practical point of view; however, instruments and (PbB), although this mainly reflects Pb uptake due to
methods used for their characterization are still recent exposure. In fact, there are other indicators that
unsatisfactory. may provide better information regarding Pb expo-
Other critical points may arise from the complex- sure: the adjusted PbB (i.e. PbB × years of exposure),
ity of the effects under study and from gender dif- erythrocyte zinc protoporphyrin (which reflects lon-
ferences. For example, clinical and epidemiological ger exposure periods), and indicators of body burden
findings on female reproductive function may be (such as Pb in the bone or urinary Pb after chelation);
influenced by age, ovarian reserves, hormonal imbal- however, these methods are not applicable to large-
ance, male cofactors, and sexually transmitted dis- scale studies.
eases. Thus, environmental or occupational factors The metal concentration in the relevant and criti-
interact with a wide variety of complex, multiple- cal tissue would be an ideal indicator of dose; in our
phase processes and it may be difficult to distinguish case, Pb concentration in seminal fluid, as a whole or
occupational or environmental risks from other risk in its fractions, theoretically correlates with testicular
factors. Finally, we must also consider the possibil- Pb, and therefore with histopathological alterations.
ity that extrapolating the results from animal stud- Bonde et al. (2002) demonstrated that Pb in blood, sem-
ies to humans is not feasible owing to structural inal plasma, and spermatozoa were highly correlated,
and functional differences between species, and the although no association between the measurement of
mechanisms of adverse effects are seldom known. long-term exposure and semen characteristics has been
For example, it appears to be difficult to transfer found (Figure 1). The study showed, however, that Pb
experimental fetal anomalies, embryo or fetal lethal- tends to accumulate in spermatozoa and it correlates
ity, or other developmental effects during organogen- with semen volume and sperm density.
esis after exposure to metallic elements ions during Hernandez-Ochoa et al. (2005) showed that
periods of pregnancy or development to effects on decreases in sperm quality; sperm concentration,
humans (Windham and Fenster, 2008). Teratogenetic motility, morphology, and viability were associated
bioassays in rodents have yielded positive results for negatively with Pb in spermatozoa, whereas semen
many metallic elements that produce fetal and early volume was negatively associated with Pb in seminal
postnatal deaths, as well as malformations such as fluid.
anencephaly, eye defects, cleft palate, and skeletal Other problems are exposure to multiple metallic
anomalies (Wells et al., 2005), but these have not been elements and interaction between toxic and essential
confirmed in humans. However, an experimental elements. Their concomitant measurement and mak-
approach is essential, especially for investigating the ing adjustments for their relative amounts might be
mechanism of action through which each metallic ele- important for understanding interactions relevant to
ments acts. toxicokinetics and toxicodynamics, and the mecha-
In this new edition, we modified the presentation nisms of toxic action on reproduction.
approach of this chapter by moving from the meth- Some studies have attempted an overall evalua-
odology of dose assessment (see Apostoli et al., 2007) tion of the relationship between several metallic ele-
to the mechanisms of action, hypothesized from ments (Pb, Cd, Cu, Se, and Zn) and the morphology,
epidemiological and experimental studies on the number, and motility of spermatozoa (Telisman et al.,
various phases of reproductive and developmental 2000, 2007). Apostoli et al. (2000) examined subjects
toxicity. not occupationally exposed to metallic elements and
20  Effects of Metallic Elements on Reproduction and Development 401

were significantly higher than in the seminal plasma


of controls than in Pb-exposed workers, whereas Se
was significantly lower. These results highlight the
potential of this approach, since the determination of
multiple elements may improve our knowledge of the
distribution of metallic elements in mixture exposure
situations.
To obtain better information, an evaluation of expo-
sure to metallic elements should also be made as close
as possible to the “target time.” For example, this may
be a few days in the case of an effect via seminal plasma
mediated by the prostate or seminal vesicles, or up to
12 weeks if spermatogenesis is affected, via the testis.
An ideal risk assessment may be reached when
information from the indicators of exposure is com-
bined with information from indicators of effect. In
this specific case, possible dose indicators include the
environmental or biological concentrations of metallic
elements, and effect indicators range from a synthetic
evaluation of the specific effect to measurements of fer-
tility or neurodevelopmental effects (for example, the
neurobehavioral test), and to the determination of hor-
mones in the blood or urine.
In addition, semen parameters can be used to indi-
cate a direct toxic effect on the male reproductive sys-
tem or to predict adverse effects on fertility or offspring
health.
To date, the semen quality outcomes utilized most
often in investigations of the effects of metallic ele-
ments on male reproduction have been sperm count,
motility, and morphology. Used together, these param-
eters provide a good general assessment of male fer-
tility and reproductive health status. Other techniques
for measuring the characteristics of sperm that predict
reproductive health are simultaneous DNA integrity
assays, which detect DNA strand breakage in single
sperm cells, fluorescence in situ hybridization (sperm-
FISH), which detects abnormalities in chromosome
number, and the sperm chromatin structure assay
(SCSA). Examples of the use of sperm DNA chroma-
tin assays to specifically identify associations between
metallic elements and sperm health include correla-
FIGURE 1  Scatter plots of lead concentrations in seminal plasma
and spermatozoa (A), spermatozoa and blood (B), and seminal plas- tions between seminal fluid Zn and SCSAs (Björndahl
ma and blood (C) in a subset of 165 industrial workers. All correla- and Kvist, 2011), and correlations between seminal con-
tions are highly significant. (Note 10 μg/dL = 100 μg/L.) Reproduced centrations of Cd, Pb, and malondialdehydes (MDAs)
with the permission of Occupational & Environmental Medicine (see as a marker of oxidative stress in sperm (Pandya et al.,
Bonde et al., 2002).
2012).
In an international study of semen quality involv-
workers exposed to Pb, using inductively coupled ing 503 industrial workers (Project Asclepios), the
plasma mass spectrometry (ICP-MS). Lead and barium effects of Pb exposure were evaluated through semen
(Ba) were found in higher amounts in the sperm cells studies, semen sampling, and initial processing (vol-
of Pb-exposed workers, whereas Se, Zn, and calcium ume, motility, and velocity); semen analysis (morphol-
(Ca) were present in significantly lower concentra- ogy, computer-aided sperm analysis, SCSA, FISH,
tions, compared to the controls. The Pb and Cd levels and germ cell genotoxicity); and finally through an
402 Pietro Apostoli and Simona Catalani

exposure assessment. The study showed that current due to Pb having a greater ability to cross the placental
occupational exposure levels in the United Kingdom, barrier for a given PbB concentration in some women.
Belgium, and Italy did not represent a high risk to Assessing the influence of genetic polymorphisms of
male fertility. Adverse effects on sperm concentration, Pb-binding proteins on the probability of suffering
for example, are unlikely at blood Pb (PbB) concentra- from miscarriage or other reproductive outcomes will
tions below 450 μg/L (Bonde, 2002). On the other hand, be very important for identifying groups particularly
a decrease in the sperm concentration and sperm count susceptible to the effects of Pb exposure during preg-
of 149 Pb-exposed workers was found at PbB concen- nancy (Lamadrid-Figueroa et al., 2007).
trations below 400 μg/L in studies by other authors
(Telisman et al., 2000).
The most commonly applied nonbiological mark- 3  GENDER DIFFERENCES
ers are time to pregnancy (TTP) or the fecundability
odds ratio (fOR), which compares numbers of progeny Gender differences in toxicology begin at the gam-
among groups and the number of pregnancy losses. ete and embryo stages. These differences may be
TTP is the number of months or menstrual cycles that under genetic and hormonal control, and affect xeno-
it takes a couple to conceive and covers the entire dis- biotic exposures, metabolism, susceptibility, risk, and
tribution of waiting times from 0 (high-level fecundity) health through development and maturation. The
to several years (low-level fecundity) (Joffe, 1989). The prominent role of testosterone and dihydrotestos-
fOR is the odds of conception in a single menstrual terone in male sex differentiation results in a vulner-
cycle (fOR = 1/TPP). A fOR of less than one indicates ability to chemicals that can influence their synthesis
that the exposed group has a lower fOR, or “sub” and action, such as androgen receptor antagonists
fecundability, than the comparison group. and androgen synthesis inhibitors. As for prenatal sex
Fertility studies should not be mistaken for simple differentiation, the process of puberty is also suscep-
survey studies since they represent an approach that is tible to environmental factors that alter the levels of
exceptionally susceptible to bias. It has become clear important factors or modify their actions on target tis-
that, to address the various types of bias, TTP studies sues. Once adulthood is reached, further differences
need to include information on at least three related in susceptibility may occur as a result of differences in
conditions: the TTP distribution, unprotected inter- the hypothalamic-pituitary-gonadal control of game-
course not leading to pregnancy, and accidental preg- togenesis. In the male, the relatively tonic secretion of
nancies (Bonde et al., 2006). follicle-stimulating hormone and luteinizing hormone
In addition to selective forces, causal inferences in from the pituitary gland maintains a constant produc-
TTP studies may be confounded by volitional factors tion of sperm, whereas in the female intricate feedback
related to sexual behavior. In occupational studies con- loops between the central nervous system (CNS) and
ducted in groups that are homogeneous with respect the ovary create a cyclical pattern of egg development
to socioeconomic and cultural factors, volitional fac- (Vahter et al., 2007a,b).
tors such as reproductive behavior, use of contracep- Finally, lifestyle factors may affect toxicity differ-
tion, and persistence of trying may not represent a ently in men and women. These may include exposure
severe problem. However, these volitional factors are situations in the working or general environment; life-
certainly relevant to comparisons over time and to style factors such as smoking, dietary factors, physical
comparisons between different regions and cultures. activity, physical constitution (size, fat), cosmetics, and
Finally, spontaneous abortion has been evaluated in fashion; and stress factors.
a number of studies involving metallic elements and With respect to the mechanisms of action involved,
reproduction health. Failure to recognize or consider there are reported to be gender-related differences in
very early fetal or even embryonic deaths in the analy- the activity of antioxidant defenses. There is substantial
sis may lead to a failure to detect important exposure evidence indicating that sexual hormones can modify
effects. In particular, chromosomal abnormalities are cell proliferation, neuroplasticity, and vulnerability to
estimated to occur in 30-40% of spontaneous abor- neural insults, including oxidative stress.
tions; therefore, this outcome could be informative in Testosterone and other androgens may exert bio-
the investigations of metallic elements that interfere logical effects on neural cells, such as neuroprotective
with the cell cycle, mitosis, or meiosis (Gerhard et al., effects against specific oxidative insults (Hammond
1998). There are published studies (Tabacova and Bala- et al., 2001; Nguyen et al., 2005). Additional protection
baeva, 1993; Borja-Aburto et al., 1999; Bellinger, 2005) against oxidative stress is provided by estrogen, which
indicating that a history of spontaneous abortion is has antioxidant effects by acting as a scavenger or by
related to the plasma:whole PbB ratio. This could be inducing the synthesis of protective molecules via
20  Effects of Metallic Elements on Reproduction and Development 403

activation of estrogen receptors (ERs) (Olivieri et al., failure. Exposure in adults may have a direct effect
2002; Kluxen et al., 2012). These hormones play a criti- on the germinal cells and, subsequently, on hormones
cal role in the gender differences reported in the patho- and sexual performance. In addition, exposure of
physiology and outcome of acute neurological injuries adult males may affect offspring by exposing the
where oxidative stress is clearly involved. mother and the fetus to contaminated semen or by the
There are also some differences between fetus gen- transmission of abnormal sperm DNA/chromatin to
ders in pregnancy outcome and developmental altera- the progeny.
tions. Epidemiological studies of human infants and
children, as well as experimental animal studies, have 4.1  Direct Mechanism of Spermatogenesis
reported more developmental defects in males than
Alterations
in females exposed to methylmercury (methyl-Hg)
(Malagutti et al., 2009; Grandjean et al., 2010; Robinson Spermatogenesis occurs in one of the most rapidly
et al., 2011). Results stratified by sex suggest that pre- proliferating tissues in the body. Damage caused by
natal Hg exposure may have a greater adverse effect exposure to some metallic elements early in the sper-
on psychomotor development in females than in males matogenic cycle (i.e. during spermatogenic cell dif-
(Llop et al., 2012). Kippler and coworkers (Kippler et al., ferentiation) may cause long-term defects in semen
2012) found evidence of sex differences in the associa- quality, whereas damage during spermiation or to
tion between maternal Cd exposure and birth size: the sperm present in the epididymis has more transient
association was apparent only in female offspring. The effects.
authors hypothesized that Cd might affect sex-specific The main effects on spermatic functionality are: (1)
programming, which begins very early in pregnancy a decrease in semen volume/concentration/count; (2)
and may contribute to the vulnerability of the develop- a decrease in sperm motility or quality of motility; (3)
ing fetus to maternal stressors (Mueller and Bale, 2008). an increase in abnormal sperm morphology, particu-
In a follow-up of the prospective Cincinnati Pb study, larly in the head of the sperm; and (4) impairment
the analysis of childhood Pb exposure and neuropsy- of the secretory function of the prostate and vesicula
chological effects in adolescence indicated a height- seminalis. The main results of relevant studies are
ened risk in males (Ris et al., 2004). This finding was shown in Table 1. However, some groups have found
confirmed by Jedrychowski et al. (2009), who assessed no influence of Pb, Cd, or Hg on sperm (Noack-Füller
the relationship between the very low levels of prena- et al., 1993; Kasperczyk et al., 2002; Bonde et al., 2002;
tal Pb exposure measured in cord blood (< 50 μg/L) Mocevic et al., 2013).
and possible gender-specific cognitive deficits during Positive effects on the motility, morphology, and
the first 3 years of life. Gender-based differences in concentration of spermatozoa have been reported
immunotoxicity induced by Pb have been observed in for Zn, Mg, Ca, and Ni. Zn deficiency is also associ-
both juvenile chickens and adult rats following low- ated with disturbances in reproductive function. Zinc
level exposure during embryonic development (Bunn is present in both spermatozoa and seminal plasma,
et al., 2001). In utero exposure to Pb resulted in more where its concentration is considerably higher than in
severe immunotoxicity in females than males. the other body fluids. Zinc also contributes to the form
Metabolism also plays a key role in the detoxifica- of sperm motility (Kaludin et al., 1983; Wong et al.,
tion and activation of xenobiotics, and a gender dif- 2001). In addition, higher percentages of motile sper-
ference in the methylation of inorganic As has been matozoa have been reported in semen with a higher
observed, with a higher rate in females compared to concentration of Mg in the seminal plasma (Wong
males (Huda and Vahter, 2011; Pilsner et al., 2012). et al., 2001). Calcium is required for many physiologi-
cal processes that regulate all living cells, including
spermatozoa. Although the influence of Ca on sperm
4  MECHANISM OF ACTION IN MALE AND motility has not been completely elucidated, the addi-
FEMALE GERMINAL CELLS tion of Ca2+ has been observed to stimulate flagellar
beating (Suarez et al., 1993).
The male and female reproductive systems have Hypothesized direct mechanisms of action through
sensitive windows for toxic exposure during develop- which metallic elements exert these effects include (1)
ment and throughout adult life. Metallic elements can an increase in reactive oxygen species (ROS) and a
have detrimental effects on spermatogenesis and ovar- decrease in activity of the cell antioxidant defense sys-
ian function and at several levels. tem; (2) direct damage to seminiferous tubules or the
Exposure in utero or during childhood and sub- testis; (3) alterations in DNA-protamine binding; and
sequent development may lead to later reproductive (4) interference with the sperm acrosome reaction.
404 Pietro Apostoli and Simona Catalani

TABLE 1  The Main Effects of the Metallic Elements on Spermatogenesis


In vitro/animal/
Metal Effects on sperm production human Reference(s)

As Low sperm motility Human Wirth and Mijal, 2010


As Decrease in sperm production Human Xu et al., 2012
Cd Immature forms of spermatozoa Human Chia et al., 1992
Cd Decrease in testis size Human Jurasović et al., 2004
Cd Negative effect on spermatozoa quality, especially on sperm morphology Human Massányi et al., 2004
Cd Correlation with pathological sperm alteration (large heads) Human Slivkova et al., 2009
Cr(VI) Decreased sperm count and percentage of motile sperm Human Li et al., 2001
Cr Increased percentage of morphologically abnormal spermatozoa Human Kumar et al., 2005
Cr Reduction of sperm count and sperm forward motility Monkey Subramanian et al., 2006
Cu Toxic effect on the seminiferous epithelium Human Skandhan, 1992
Cu Sperm copper was positively associated with sperm DNA fragmentation Human Schmid et al., 2013
Hg Low sperm motility Human Ernst and Lauritsen, 1991
Hg Decreased sperm motility and sperm count Rat Fossato da Silva et al., 2011
Ni Morphologically abnormal sperm, decreases in sperm count and motility, Human Massányi et al., 2004
destroyed the regular testicular structure
Ni Separated flagellum of spermatozoa Bull, fox Zemanová et al., 2007
Pb Low semen volume, increased sperm chromatin condensate, decreased sperm Human Hernandez-Ochoa et al., 2005
concentration and motility
Pb Negative impact on the motility and viability of spermatozoa and sperm count Human Eibensteiner et al., 2005
Pb Increased incidence of teratozoospermia, diminution of sperm cell production, Human Naha et al., 2006
low sperm vitality
Pb Increased immature sperm concentration and percentage of pathological sperm, Human Telisman et al., 2007
wide sperm and short sperm
Pb Decreased sperm motility, no effect in sperm viability Mice Oliveira et al., 2009
V Decreased sperm counts, damage to the seminiferous epithelium Rabbit Castellini et al., 2009
V Spermatogenic arrest, increased percentage of abnormal sperm Rat Chandra et al., 2010

Testicular apoptosis
↑Increased Leucocytes (extrinsic ROS) DNA fragmentation
FIGURE 2  Metallic elements as a source of
↑Spermatozoa stress response (intrinsic ROS) ROS and toxicity in semen.  Under normal condi-
Defective tions, reactive oxygen species (ROS) are produced
Metals by spermatozoa and leucocytes, and antagonized
Protaminosis
by circulating antioxidants. In the presence of exog-
↓Circulating Antioxidants Low sperm count enous factors, such as metallic elements, ROS pro-
duction is increased and antioxidant capacity lim-
Structural distortions ited, leading to oxidative stress and sperm damage.

4.1.1  Reactive Oxygen Species as Part of Infertility


large number of mitochondria and microsomes, are
Assessments
presumed to render the organ more vulnerable to
There is much evidence for the presence of ROS in oxidative stress (Lavranos et al., 2012). Spermatogo-
both normal sperm and sperm samples from infertile nial cells with gross DNA damage may be eliminated
men (Makker et al., 2009), although excessive ROS pro- naturally, while cells with more subtle defects may
duction is consistent with inflammation and, therefore, survive and are finally manifested as abnormal sperm
with a potential negative impact on sperm function. It (Du Plessis et al., 2010). Therefore, during spermato-
is established that testicular oxidative stress is com- genesis, metal-induced ROS could damage specific
monly induced under different normal and/or patho- genes or processes that influence the sperm structure.
physiological conditions, leading to male infertility In addition, metal-induced ROS may inhibit the activ-
(Figure 2). ity of DNA polymerase, an important enzyme in DNA
The effect of metallic elements on abnormalities in repair mechanisms (Angelopoulou et al., 2009).
sperm morphology and motility, and in altering viabil- Excessive ROS levels have been linked to lipid per-
ity may be attributed to an increased semen concentra- oxidation of the sperm plasma membrane, resulting
tion of ROS, especially of hydrogen peroxide (H2O2). in a loss of membrane fluidity, structure, and function
Lipid-rich membranes of the testis, together with the (Mahfouz et al., 2010).
20  Effects of Metallic Elements on Reproduction and Development 405

However, cells have devised a variety of mecha- after HgCl2 exposure may render the testes more sus-
nisms to protect themselves from the potential dam- ceptible to oxidative damage, leading to their func-
aging effects of ROS accumulation. Defense systems tional inactivation (Boujbiha et al., 2011). In the case of
include enzymes, such as peroxidases, catalases, Pb, chronic exposure results in increased lipid peroxi-
and superoxide dismutases, as well as small mole- dation. Structural abnormalities in spermatozoa were
cules, such as glutathione (GSH) and some vitamins also significantly limited in antioxidant-enriched ani-
(­Lavranos et al., 2012). A reduction in catalase activity mals, thus supporting an oxidation-based mechanism
may reflect a decreased capacity of the testicular mito- (Marchlewicz et al., 2004). However, the effects are
chondria and microsomes to eliminate the H2O2 pro- dose dependent: acute exposure increases the activ-
duced in response to Cr, Cd, Cu, and V (Chandra et al., ity of antioxidant defense enzymes, unlike chronic
2007; Wu et al., 2008). The balance of these enzyme exposure.
systems may be essential to testicular health; hence, a Finally, V-induced alterations in the male repro-
significant reduction in enzyme activity, accompanied ductive systems of humans and animals may involve
by a marked increase in H2O2 and lipid peroxidation, oxidative stress by inducing the oxidative deteriora-
may reflect the adverse effects of metal-induced ROS tion of testicular functions. In vitro and in vivo stud-
on the antioxidant system. ies demonstrate that V treatment result in a significant
Oxidative stress and the generation of ROS can also dose- and time-dependent increase in testicular lipid
be a consequence of arsenic exposure. ROS generation, peroxidation, a marked inhibition in the level of super-
as well as the binding of As to protein thiol groups, oxide dismutase and catalase activity, and decreased
can alter the function of many proteins. In rats, cotreat- sperm counts (Aragon et al., 2005; ­Castellini et al.,
ment with vitamin E provided significant protection 2009).
from the harmful effects of sodium arsenite on sperm
number, motility, viability, and morphology (Das et al.,
4.1.2  Direct Toxicity to the Seminiferous Tubules or
2009).
Testis
Although the testes express several antioxidant
enzymes, such as superoxide dismutase, catalase, and The testes are extremely sensitive to external insults,
glutathione peroxidase (GPx), to counteract oxida- including the action of some metallic elements; one
tive stress, their levels are greatly diminished upon possible explanation for this sensitivity is the unique
Cd exposure in rats (Pandya et al., 2012). Studies of morphological structure of the blood-testis barrier
substances with antioxidant activity, such as vitamin (BTB) (Siu et al., 2009).
C, vitamin E, Zn, Se, and melatonin, have also dem- Ultrastructural analyses of experimental studies in
onstrated that oxidative stress is associated with Cd- rabbits have revealed three types of damage to sperm
induced testicular damage, since these substances head membranes, depending on the metallic element
reduced and/or prevented both the oxidative stress used: acrosome breakage, with the formation of vari-
and damage caused by Cd (Acharya et al., 2008; Amara ous sizes of microvesicles (As, Cd, Hg, and Pt); a large
et al., 2008; Burukoğlu and Bayçu, 2008). round hole (As, Cd, and Cr); and numerous folds in
Chronic Cr (as potassium dichromate) exposure the acrosome membrane (V) (Castellini et al., 2009).
induces reversible oxidative stress in the seminal The best-studied metallic element is Cd, and some
plasma and sperm of monkeys by creating an imbal- of its mechanisms of action have been elucidated.
ance between ROS and the antioxidant systems, lead- In initial studies, morphological analysis showed
ing to sperm death and reduced motility of live sperm that Cd induces profound and irreversible injury to
(Subramanian et al., 2006). The significant changes mammalian testes including disruption of endothe-
seen in exposed groups relative to controls suggests lial cells of the microvessels, edema, and hemorrhage
that CrO3 exposure suppresses antioxidant enzymes using, apparently as the result of primary disruption
and ascorbic acid levels, with a concomitant increase in of the vascular system. For instance, successive stud-
the level of lipid peroxidation and H2O2, to adversely ies in vitro and in lung tissue have shown that cad-
affect testicular function (Acharya et al., 2006). herin-1/E-cadherin is one of the primary targets of Cd
Treatment with Hg was also associated with oxi- toxicity in epithelial cells: Cd interacts with the puta-
dative stress. Evidence of the induction of oxidative tive calcium-binding motif in E-cadherin, thus disrupt-
stress was shown by interference with antioxidant ing cadherin-based cell adhesion (Prozialeck, 2000).
defense and a significant dose-dependent increase in Cadmium may also disrupt Sertoli cell tight junction
testicular lipid peroxidation as a consequence of pro- barrier function by not only decreasing the synthesis
oxidant exposure. An increase in free radical formation and/or expression of proteins but also promoting pro-
associated with loss of the antioxidant defense system tein redistribution at the Sertoli-Sertoli cell interface,
406 Pietro Apostoli and Simona Catalani

perhaps via altering protein endocytosis and/or recy- groups that do not participate in the formation of disul-
cling (Wong and Cheng, 2005). fide bridges. The influence of Zn on protamine is there-
Cadmium-induced BTB alteration and cell junction fore thought to play a role in the normal function of the
disruption in the seminiferous epithelium is mediated spermatozoon (Björndahl and Kvist, 2010, 2011). How-
by the activation of specific signal transduction path- ever, in addition to the physiological presence of Zn in
ways. In detail, in the first step, Cd ions probably enter the spermatozoon, there is also clear evidence of the
Sertoli and/or germ cells via different mechanisms. presence of other metallic elements, such as Pb, Cu, or
Once inside the Sertoli and/or germ cell, Cd induces Ni (­Quintanilla-Vega et al., 2000a; Massányi et al., 2004;
the synthesis and release of cytokines, which, by inter- Hernandez-Ochoa et al., 2005). The toxicity in these cases
acting with their respective receptors, activate the may be either direct or mediated through their influence
stress-activated p38 mitogen-activated protein kinase on protamines. Since some metallic elements bind tightly
(MAPK) signaling pathway. At the same time, possibly to sulfhydryl (-SH) groups in proteins, such as enzymes,
also mediated by p38 MAPK, the production of prote- cell membrane proteins, and GSH, they might compete
ases (e.g. cathepsin L) is also induced. The net result with or replace the Zn atoms that are normally bound to
is that these mechanisms contribute to disruption of nuclear protamines. These changes could affect normal
the BTB and Sertoli-germ cell junctions, which leads to disulfide bond formation, alter DNA-protamine bind-
germ cell loss (Siu et al., 2009). However, Cd is known ing, or impair chromatin decondensation during fertil-
to be specifically localized to interstitial testicular cap- ization (Quintanilla-Vega et al., 2000b; Silbergeld et al.,
illaries (Nordberg, 1972) and it seems unlikely that 2003). The resulting reduced stability of the chromatin
changes in Sertoli cells are crucial for BTB disruption. and abnormal chromatin structure is strongly related to
A possible role for zinc transporter ZIP8 (Wang et al., reduced fertility in humans (Zhang et al., 2012).
2007) is discussed in Chapter 32, “Cadmium.”
Nickel destroys the normal testicular structure and 4.1.4  Sperm Acrosome Reaction
induces dose-dependent alterations in almost all mor-
phometric indicators. In Ni-treated animals, the height The metallic elements that specifically modulate
of the germinal epithelium and the diameter of the the initiation and duration of the acrosome reaction,
seminiferous tubules decreased, and the diameter and are Cd and Pb. Furthermore, As, Cd, Hg, and Pt cause
relative volume of the lumen increased (Massányi et al., acrosome breakage, with the formation of various
2007b). Qualitative analysis revealed dilatation of the sized microvesicles. As, Cd, and Cr cause the forma-
blood vessels in the interstitium, undulation of the basal tion of a large round hole and V causes numerous folds
membrane, and several empty spaces in the germinal to form in the acrosome membrane (Castellini et al.,
epithelium. A specific assay detected a higher frequency 2009). Maintenance of the acrosome is essential for the
of localized apoptosis in the interstitium of Ni-adminis- functional integrity of sperm, required for binding to
tered animals compared to controls (Forgacs et al., 2012). the zona pellucida and for responding to the appropri-
ate signals of the oocytes.
The negative impact of Cd on male fertility is medi-
4.1.3  Alteration to DNA-Protamine Binding ated by its chemical affinity for Ca channels in the cell
In mammalian spermatozoa nuclei, DNA is tightly membranes causing a decreased ability of sperma-
packaged with protamines, Zn-containing proteins that tozoa to undergo the acrosome reaction. Arabi and
protect sperm DNA by binding to it during spermatogen- ­Mohammadpour (2006) also discovered that Cd can
esis. Altered levels of protamines may result in increased alter the integrity of the acrosomal membranes of sper-
susceptibility to injury in spermatozoa DNA, causing matozoa in bulls and thus cause abnormal acrosome
infertility or poor outcomes in assisted reproduction. reactions. A role for Pb is suspected in the inexplicable
Due to their molecular properties, protamines not infertility of men caused by induction of a spontane-
only have electrostatic interactions with DNA but also ous premature acrosome reaction, and the Pb content
have the potential to bind metallic elements or other of seminal plasma may have a negative impact on
agents, either as part of normal physiology or through in vitro fertilization rates (Benoff et al., 2003).
their potential involvement in chromatin alterations.
One of the first observations that stimulated research
4.2  Direct Effects on the Ovary and Ovarian
into the possible influence of metallic elements on prot-
Production
amines was that Zn is highly abundant in sperm nucleus.
Subsequent studies have confirmed these observa- Evidence of the toxicity of metallic elements on the
tions, leading to a proposal that Zn can stabilize the ovaries and on ovarian production in humans is scarce
chromatin of spermatozoa through binding to thiol compared to that for sperm.
20  Effects of Metallic Elements on Reproduction and Development 407

Some experimental studies have shown that metal- reproductive toxicity may be due to the induction of
lic elements can have detrimental effects on ovarian oxidative stress or free radical generation; however,
function at several levels. They cause a direct effect arsenic may have direct ovarian effects in the presence
of ovaries or an indirect effect via the hypothalamus-­ of reduced gonadotropin levels (Chattopadhyay et al.,
pituitary-ovarian axis. With respect to metallic ele- 2003).
ments that directly target the ovary, those that Akram and colleagues (2009) measured DNA dam-
extensively destroy primordial and primary follicles age in the ovarian cells of rats treated with different
can cause premature ovarian failure. Alternatively, doses of sodium arsenite, and found a long comet-like
metallic elements that selectively damage large grow- tail reflecting a high degree of DNA damage in the
ing or antral follicles can cause reversible disruption of ovarian cells with increased doses of sodium arsenite.
cyclicity by impairing ovarian steroid production and The authors assumed that an imbalance in the antioxi-
ovulation (Table 2). dant system caused by arsenite treatment could have
In contrast to gametogenesis in males, in females, led to abnormal oocyte development, as well as caus-
oocytes are not continually replenished during repro- ing damage to oocyte DNA, which ultimately affects
ductive life. After folliculogenesis in the latter half of ovulation.
gestation, the maximal number of oocytes is fixed. Many hypothesized mechanisms of action have
Human female reproductive toxicity, in terms of been proposed on the basis of experimental studies
reduced fecundity and fertility, has been reported to on Cd toxicity; these include mechanisms involving
be association with high doses and occupational level specific targets and more general mechanisms involv-
exposure to Hg, Cd, and Pb. Associations with high ing the formation of ROS. Gestational exposure to
doses of these metals have been reported among cou- Cd affects female reproductive health as a result of
ples conceiving with assisted reproductive technolo- changes in ovarian function associated with altered
gies, as well as with unassisted conception (Mendola ovarian histoarchitecture, oxidative stress, anemia, and
et al., 2008). Some authors have identified associations delayed puberty with impaired steroid hormone levels
between trace amounts of Pb and Cd in follicular flu- (Thompson and Bannigan, 2008). ROS not only have
ids from a single follicle and oocyte fertilization. An effects on the male reproductive system but also have
inverse association has been detected between Pb in effects on the female reproductive system in terms of
follicular fluids and fertilization, although the associa- interfering with the implantation and fertilization of
tion is positive for Cd (Bloom et al., 2012); furthermore, eggs, female infertility, and the unregulated synthesis
embryo fragmentation is reduced by higher blood Hg of female sex hormones (Massányi et al., 2007a).
(Bloom et al., 2011). Other studies on antioxidants and free radicals
Arsenic-treated rats demonstrated reduced ovar- clearly suggest the onset of oxidative stress in the ova-
ian weight and prolonged estrous cyclicity; these ries of Cd-treated rats: there was a significant increase
adverse ovarian effects could be prevented by coad- in the concentration of H2O2 and lipid peroxidation,
ministration with l-ascorbate or sodium selenite. The and subnormal activity of most of the antioxidant
results of these studies suggest that arsenic-induced enzymes tested. Cd-induced production of ROS was

TABLE 2  The Main Effects of the Metallic Elements on the Ovary and Ovarian Production
In vitro/animal/
Metal Effects on the ovary and ovarian production human Reference(s)

As Ovarian DNA damage Rat Akram et al., 2009; Mondal et al., 2013
Reduced ovarian weight Rat Chattopadhyay et al., 2003
Cd Reduction of maturation of oocytes Buffalo Nandi et al., 2010
Failure to ovulate Rat, frog Piasek and Laskey, 1994; Fort et al., 2001
Suppressed oocyte maturation Sheep, human Leoni et al., 2002; Paksy et al., 1997
Decreased relative volume of growing follicles and increased stroma Rabbit Massányi et al., 2007a
Cr Reduced number of follicles, reduced number of ova, increased estrous Mouse Murthy et al., 1996
cycle duration
Cr(VI) Damaged ovarian histoarchitecture in various age groups studied, Rat Samuel et al., 2011
­delayed sexual maturation
Pb Reduction of maturation of oocytes Buffalo Nandi et al., 2010
Ovarian follicular cysts, fewer corpora lutea, folliculogenesis dysfunction Mouse Tuapeau et al., 2001
Correlation with pathological sperm alteration (flagellum ball) Human Slivkova et al., 2009
408 Pietro Apostoli and Simona Catalani

reported, and ROS may propagate the initial attack on in ovarian follicular cysts and fewer corpora lutea at
lipid membranes, causing lipid peroxidation (Samuel high Pb concentrations and folliculogenesis dysfunc-
et al., 2011; Stohs et al., 2001). Data on H2O2 and lipid tion, with fewer healthy primordial follicles and an
peroxidation show a dose-dependent increase in H2O2 increase in the number of atretic antral follicles at low
and lipid peroxidation levels. Pb levels (Taupeau et al., 2001). There are histological
Yang et al. (2012) showed that Cd exposure results changes in the follicular cells and oocytes of chroni-
in oxidative damage of hen ovarian tissue by altering cally Pb-intoxicated mice, including disruption of the
antioxidant defense enzyme systems, lipid peroxida- follicular membrane, increased pyknosis in the granu-
tion, and apoptosis, as well as inducing endocrine losa cells, and hypertrophy of the theca layer, indicat-
disturbance. After being treated with Cd for 20, 40, ing follicular atresia. Lead treatment also increased the
or 60 days, ovary and serum samples were collected number of oocytes that had resumed meiosis, consis-
and examined; results showed that the content of Cd, tent with the fact that follicular atresia is responsible for
malon­dialdehyde, and nitric oxide, as well as the activ- the resumption of meiosis and oocyte degeneration in
ity of nitric oxide synthase, increased in the ovaries several mammalian species (Avazeri et al., 2006).
and serum, while the level of GPx and the activity of
superoxide dismutase decreased in both the low-dose
and high-dose groups. The number of apoptotic cells 5  THE ROLE OF HORMONES: METALLIC
in the ovaries increased in the Cd treatment group, and ELEMENTS AS ENDOCRINE DISRUPTORS
extensive damage was observed in the ovaries.
Chromium caused a dose-dependent reduction in In 2002, the World Health Organization (WHO)
all stages of preantral follicles (primordial, primary, defined endocrine disruptors (EDs) as “exogenous
and growing) and in the number of oocytes ovulated, substances or mixtures which alter the functions of the
as well as lengthening of the estrous cycle. There was endocrine system and, consequently, cause adverse
also morphological evidence of follicular disruption. health effects in an intact organism, its progeny or
These data support the observation of fewer corpora in (sub) populations” (Damstra et al., 2002). The ED
lutea in a study of pregnant mice. Furthermore, the hypothesis is that low-level exposure to certain chemi-
data suggested that high levels of Cr might disrupt cals may contribute to endpoints such as lowering the
menstrual cyclicity, ovulation, and fertility in Cr- age at menarche, impairing semen quantity and qual-
exposed women, as well as potentially causing early ity, decreasing the male-to-female sex ratio at birth,
menopause. Murthy et al. (1996) reported a number of and increasing the rates of hypospadias and testicular
reproductive effects (reduced number of follicles at dif- cancer, infertility, spontaneous abortions, and structural
ferent stages of maturation, reduced number of ova per and functional congenital malformations (Sharpe, 2001).
mouse, increased estrous cycle duration, and histologi- The term ED-like to describe a substance that inter-
cal alterations) in the ovaries of female mice exposed to feres with the endocrine system is considered inappro-
potassium dichromate in drinking water for 20 days. priate by some authors, who have instead introduced
The severity of the reproductive effects appeared to be the term endocrine active (EA). In addition, since not
dose related. all chemicals with endocrine-disrupting properties
Nampoothiri et al. (2007) studied the effect of com- are equally hazardous, an assessment of potency was
bined exposure to Pb and Cd on granulose cells, dem- also proposed as a second step to differentiate high-
onstrating that metallic element exposure caused a risk chemicals from those of lower risk (Bars et al.,
decrease in the reduced GSH content, along with ele- 2012). Numerous chemical substances belong to this
vated lipid peroxidation, in all groups. Granulose cells category, including persistent organic pollutants
of Cd-treated and combined treatment groups demon- (POPs), pesticides, and substances used in cosmetics,
strated a maximum increase in lipid peroxides and cat- in addition to several metallic elements. Some of these
alase activity, along with a decrease in both GSH status adverse health effects are attributed to several metallic
and superoxide dismutase activity. Animals exposed to elements, while other effects are specific. Early studies
a combination of Pb and Cd exhibited an intermediate demonstrated that metallic elements can bind the ER;
effect on antioxidant status. Pb and Cd in combination the binding of certain metals to the zinc fingers of the
do not show an additive or synergistic effect, indicat- ER can alter the interaction of the receptor with DNA
ing that competition between them is due to their simi- (Predki and Sarkar, 1992). Several metallic elements
lar electronic affinities. Arsenic is known to cause free can displace or compete with estradiol binding to its
radical elevation that results in uterine cell degenera- receptor in MCF-7 breast cancer cells (Martin et al.,
tion (Wang et al., 2006). Many studies have suggested 2003; Nesatyy et al., 2005, 2006). Investigations of four
that Pb causes direct damage to the ovaries, resulting metallic elements (As, Cd, Pb, and Hg) have provided
20  Effects of Metallic Elements on Reproduction and Development 409

insight into the mechanism of their impact on mam- experimental studies in vivo and in vitro; however, we
malian reproductive systems. Exposure to both Cd are cognizant of the fact that the real effect on humans
and Hg stimulates progesterone synthesis. In addi- may be different and may be mediated by a number
tion, negative effects on spermatogenesis have been of external and internal confounding factors (Table 3).
observed in experimental studies using As, Hg, or Pb, The precise mechanism by which As alters
while both Mn and Zn stimulated spermatogenesis. ­hormone-stimulated gene regulation by ERs remains
Furthermore, alterations in the onset of puberty corre- to be determined. In an in vitro study on human
lated with exposure to Pb and Mn (Iavicoli et al., 2009). kidney proximal tubule epithelial cells, Zhang and
Some effects of EDs may be receptor mediated and coworkers (2011) showed that arsenic downregulates
may involve direct interaction with cellular targets. The ER mRNA and protein levels. Arsenic is clearly not
majority of studies carried out on EDs have revealed only an endocrine-disrupting chemical but also acts
that these substances have a genomic mechanism of via a unique mechanism quite distinct from those of
action. However, the biological effects of these com- previously characterized organic EDs, most of which
pounds cannot be wholly attributed to their interaction act as hormone mimetics and as either agonists or
with hormone receptors since some studies have also competitive antagonists. Arsenic clearly alters the
clearly indicated the presence of nongenomic mecha- ability of hormone-activated, DNA-bound steroid
nisms capable of altering, or at least influencing, the receptors to regulate gene transcription. The effects of
synthesis, transport, and availability of endogenous As on steroid receptor activity appear to be restricted
hormones (Waring and Harris, 2011). to DNA-dependent gene regulation (Bodwell et al.,
Studies into the effects of metallic elements on the 2004). Recent evidence indicates that inhibition
endocrine system have generally involved exposure occurs at the nuclear level and does not depend on a
to a single metal; however, both environmental and mechanism in which the metallic elements compete
occupational exposures are much more complex in for the ligand-binding site of the receptors (Watson
that individuals are simultaneously exposed to dif- and Yager, 2007).
ferent classes of xenobiotics, including both metal- Overexpression of various estrogen-linked genes
lic elements and organic compounds that may act as reflects the endocrine disruption effects of inorganic
EDs (e.g. POPs, DDT, and pesticides). For this reason, As at an early stage of life. In fact, similar to estradiol,
at present it is not known whether mixtures of differ- arsenite decreased the expression of ERα and increased
ent EDs may have additive, synergistic, or antagonis- the expression of the progesterone receptor. Moreover,
tic effects. EDs with the same mechanism of action are the estrogen-like effects of arsenite were inhibited by
generally assumed to behave additively, but there are an anti-oestrogen, suggesting that these responses
few examples in which this hypothesis has been tested were mediated by ERα. Arsenic also disrupted the cir-
(Iavicoli et al., 2009; Latini et al., 2003). culating levels of gonadotropins and estradiol, led to
We report the main mechanisms of action associ- the degeneration of luminal epithelial, stromal, and
ated with metallic elements, as extrapolated from myometrial cells of the rat uterus, and downregulated

TABLE 3  Mechanism of Action of Metallic Elements as Endocrine Disruptors, as Extrapolated from Experimental Studies
Metal Mechanism of action Reference(s)

Arsenic Stimulation or inhibition of nuclear transcription activity mediated by several hormone Kaltreider et al., 2001
receptors
Bond with ERs Jana et al., 2006
Down regulation of ER mRNA and protein levels Zhang et al., 2011
Cadmium Bond via ERs Garcia-Morales et al., 1994
Inhibition of transcription of the LDL receptor Jolibois et al., 1999
Inhibition of P450 (cholesterol side-chain cleavage) Kawai et al., 2002
Altered expression of aryl hydrocarbon receptor-associated genes Kluxen et al., 2012
Lead Reduction of the expression of the steroidogenic acute regulatory protein Srivastava et al., 2004
Inhibition of luteinizing hormone secretion Ronis et al., 1996; Srivastava et al., 2004
Inhibition of steroidogenic enzymes (3β and 17β-hydroxysteroid dehydrogenase) activities Pandya et al., 2012
Mercury Induction of 3β-hydroxysteroid dehydrogenase Mondal et al., 1997; Heath et al., 2012
Inhibition of the type I iodothyronine deiodinase Barregård et al., 1994
Binding and activating of ERs Zhang et al., 2008

ER, estrogen receptor; LDL, low-density lipoprotein.


410 Pietro Apostoli and Simona Catalani

downstream components of the estrogen signaling LH and FSH concentration (Doumouchtsis et al., 2009).
pathway (Chatterjee and Chatterji, 2010). These findings suggested that Pb initially causes some
Various effects of Cd on reproductive endocrinology subclinical testicular damage, followed by hypotha-
have been described, but definitive conclusions regard- lamic or pituitary disturbance following longer periods
ing its actions on target tissues vary depending on the of exposure. Leydig cells appear to be a target because
experimental model and the dosage employed. Cd was Pb exposure results in suppression of testosterone syn-
found to mimic the effect of estrogen on transcription of thesis in these cells, while Sertoli cell function appears
a reporter gene, as well of endogenous genes normally to be unaffected (Wirth and Mijal, 2010).
regulated by estrogen, including the ER receptor. Other A number of studies have reported an association
studies involving transfected cells, chimeric receptors, between Hg exposure in humans and plasma testos-
and competitive receptor binding have demonstrated terone reduction through inhibition of 3β-hydroxy-Δ5-
Cd binding to the hormone-binding domain of ERs. Cd steroid dehydrogenase activity in Leydig cells (Heath
binding was weakened by the replacement of cysteine, et al., 2012). Some authors have suggested that the
glutamic acid, aspartic acid, and histidine in the target increased cholesterol is due to the inhibition of its bio-
hormone-binding region (Henson and Chedrese, 2004). synthetic conversion to sex steroid hormones, such as
A detailed analysis of the Cd-­mediated reduction in testosterone (Iavicoli et al., 2009). Another possibility
progesterone production by cultured human tropho- is that mercury mimics the effect of estrogen on the
blast cells indicated that it is not due to cell death or testes (i.e. to both inhibit androgen production and
apoptosis. Cd also appears to modulate the expression cause accumulation of cholesterol), probably through
of aryl hydrocarbon receptor-associated genes by inter- upregulation of the high-density lipoprotein receptor,
acting with the ER (Kluxen et al., 2012). A review of the scavenger receptor class B member I (SRB1) (Tong et al.,
scientific evidence for the estrogenic effects of Cd indi- 2004). Mercury does not affect the binding of estradiol
cated that, although the in vitro and in vivo evidence to ERs. Therefore, HgCl2 exhibits an estrogen-like effect
of the estrogenic properties of Cd was persuasive, by binding to and activating ERs rather than competing
evidence from population-based human studies with estradiol binding to ERs (Zhang et al., 2008).
remains contradictory (Silva et al., 2012). After 10 years of experimental and human investi-
Lead-related reproductive toxicity is also linked to the gation, and heated debate, the relevance of EDs was
altered activity of uterine ERs and their affinity for ovar- reviewed by Sharpe and Irvine (2004). It can be sum-
ian steroids; exposure to Pb does not seem to pose a risk marized as follows:
  
with respect to progesterone secretion (Paksy et al., 2001).
(1) Few definitive data link human reproductive
The mechanism of action responsible for the inhibi-
disorders or cancer to exposure to environ-
tion of estradiol synthesis was probably a reduction in
mental synthetic chemicals; this may reflect the
ovarian expression of the StAR gene (encoding mito-
difficulty in obtaining such data or a genuine
chondrial steroidogenic acute regulatory protein). The
absence of effects;
administration of pregnant mare serum gonadotropin
(2) Synthetic chemicals are pervasive in the
restored StAr expression, indicating that metallic ele-
environment, but our understanding of their
ments do not exert a direct effect on the ovary response
potential to cause harm is limited;
to gonadotropins but might instead act on the hypo-
(3) The reproductive effects of environmen-
thalamus-hypophysis axis, thus altering the luteiniz-
tal chemicals in (aquatic) wildlife are well
ing hormone (LH) release required for both StAR and
­established; they may act as sentinels for
estradiol synthesis (Srivastava et al., 2004).
human effects, especially for the fetus; and
Experimental data have demonstrated that, in rats
(4) Recent discoveries raise the possibility that
exposed to Pb, the activities of steroidogenic enzymes
common environmental chemicals have effects
(3β- and 17β-hydroxysteroid dehydrogenase) also
on endogenous hormones.
decreased significantly, leading to altered testosterone   

production (Pandya et al., 2012). These conclusions raise some questions for toxicolo-
Nevertheless, some studies have reported direct gists working on the effects of toxic metallics on repro-
spermatoxicity without hormonal disruption, whereas ductive and developmental systems.
others report no effect of Pb on reproductive hormones
or spermatogenesis. In short-term Pb-exposed individ-
uals, high LH and follicle-stimulating hormone (FSH) 6 CONCEPTION
levels are usually associated with normal testosterone
concentrations; in contrast, in long-term Pb-exposed Metallic elements can influence the phase of con-
individuals, low testosterone levels do not induce high ception and implantation, and can have effects on
20  Effects of Metallic Elements on Reproduction and Development 411

couple fertility. In experimental studies on fertility, effects, mainly in males, of exposure to a specific chem-
metallic elements are generally administered orally for ical or a group of chemicals on fertility or reproductive
an extended period of time before, during, and after function rather than focusing on the broader spectrum
mating, in most cases not focusing on the specific stage of chemical exposures and their effects on TTP in both
or mechanism of action involved. men and women.
Some studies have shown that Cd inhibits gap junc- Snijder and colleagues (2012) carried out a system-
tion intercellular communication and gap junction atic review of the scientific literature up to December
protein/connexin phosphorylation, both of which 2010 regarding occupational exposure to chemical
are essential processes in the progression from the substances and TTP. The authors included nine stud-
eight-cell stage through to compaction (Thompson ies reporting TTP in relation to exposure to metallic
and Bannigan, 2008). Cadmium-suppressed synthe- elements, of which two studies were carried out in
sis of hyaluronic acid in the oocyte-cumulus complex women (Sallmen et al., 1995; Wulff et al., 1999), and
(OOC) matrix was linked to the inability of the infun- seven studies were carried out in men (Bonde, 1990;
dibular cilia to successfully introduce the oocyte into Hjollund et al., 1998; Apostoli et al., 2000; Sallmen
the fallopian tube (Vrsanska et al., 2003). Cell adhesion et al., 2000; Joffe, 2003; Shiau et al., 2004; Greene et al.,
is an integral and important part of OCC pick-up and, 2010). In the two studies on women, the fOR ranged
although the precise sequence and pattern of expres- from 0.80 to 0.93 for Pb exposure and from 0.82 to
sion of cell adhesion molecules during the process has 0.91 for exposure to a mixture of metallic elements.
yet to be determined, the involvement of gap junctions In men, four studies reported that high Pb exposure
in cumulus expansion has already been identified and (measured as PbB) reduced fecundability (Apostoli
the involvement of other junction types is possible. et al., 2000; Sallmen et al., 2000; Joffe, 2003; Shiau et al.,
Cadmium-induced misexpression of cell adhesion 2004). Figure 3 depicts the exposure-response relation-
molecules, including various connections, and also ship between different levels of PbB values and fORs,
the cellular delocalization of molecules, such as tight showing a clear trend of increasing PbB levels with
junction protein ZO-1/zonula occludens protein 1 decreasing fORs.
and cadherin-2/N-cadherin has been associated with Between 2005 and 2009, another study was carried
abnormalities in cell adhesion in the gap, occludens, out to evaluate the influence of metallic elements on
and adherens junctions (Fang et al., 2001). Trophoblast couple fecundity (Buck Louis et al., 2012). This study
formation and invasion have also been identified as was included in the Longitudinal Investigation of Fer-
targets for Cd toxicity. Modification of the effect of tility and the Environment (LIFE) Study, a prospective
Cd on the cell cycle by zaldaride, an inhibitor of the cohort study with a preconception enrolment of 501
intracellular calcium-binding protein calmodulin, couples designed specifically to assess persistent envi-
indicated that calmodulin has a role in mediating Cd- ronmental chemicals and human fecundity. Cadmium,
induced toxicity in the trophoblast (Thompson and Pb, and Hg were observed to be significantly associ-
Bannigan, 2008). ated with a reduction in couple fecundity: exposed
Nampoothiri and Gupta (2008) reported altera- couples with a higher degree of exposure took a lon-
tions in alkaline phosphatase activity and cathepsin ger time to conceive a human chorionic gonadotropin
D, two implantation enzymes, after coexposure to Pb (hCG)-confirmed pregnancy than couples with lower
and Cd; however, no change in reproductive perfor- blood concentrations. When assessing partner expo-
mance was observed. The decreased activity could be sure separately, female Cd exposure was associated
due to Pb and Cd competition with the magnesium ion with an approximate 22% reduction and male Pb expo-
to bind at its active site. In spite of changes in lyso- sure with an approximate 15% reduction in the odds
somal enzymes, implantation sites did not seem to be of conception per standard deviation increase in blood
affected. concentration. The authors did not observe any effect
In addition to the mechanistic studies described for Hg. Interestingly, their observed association of a
above, many studies have calculated couple fecundity longer TTP for male PbB concentrations below the cur-
through the assessment of the fOR or TTP related to rent occupational Pb exposure limit (> 400 μg/L) was
metallic elements concentrations. purported to be associated with a decrease in semen
A number of groups have reviewed the association quality (Apostoli et al., 1998).
between the exposure to specific chemicals or groups Finally, another prospective cohort study with a
of chemicals and TTP, and support the notion that preconception enrolment of women studied the asso-
environmental exposure may be hazardous for human ciation between some metallic elements (As, Cd, Pb,
fertility (Sallmen, 2001; Jensen et al., 2006; Jurewicz Mg, Ni, Se, and Zn) and TTP (Bloom et al., 2011). In this
et al., 2007). However, these reviews all focused on the study, no adverse influence was attributed to metallic
412 Pietro Apostoli and Simona Catalani

2,5
<20 20–29 30–39 >40
µg/dL µg/dL µg/dL µg/dL

2
Fecundability ratio

1,5

0,5

0
Shiau 2004 - Blood lead level <20 µg/dL

Apostoli 2000 - Blood lead level 20–29 µg/dL


Joffe 2003 - Blood lead level 20–29 µg/dL
Shiau 2004 - Blood lead level 20–29 µg/dL

Apostoli 2000 - Blood lead level 30–39 µg/dL


Joffe 2003 - Blood lead level 30–39 µg/dL
Shiau 2004 - Blood lead level 30–39 µg/dL

Apostoli 2000 - Blood lead level >40 µg/dL


Joffe 2003 - Blood lead level >40 µg/dL
Shiau 2004 - Blood lead level >40 µg/dL

Sallmen 2000 - 0.5–0.9 µmol/l


Sallmen 2000 - 1.0–1.4 µmol/l
Sallmen 2000 - 1.5–1.8 µmol/l
Sallmen 2000 - >1.9 µmol/l

Sallmen 1995a - <0.5 µmol/l


Sallmen 1995a - 0.5–0.9 µmol/l
Sallmen 1995a - >1.0 µmol/l
FIGURE 3  Forest plot summarizing studies into dose-response relationships between blood lead levels and the fecundability odds ratio
(1 μg/dL = 10 μg/L). Reproduced with permission from Human Reproduction Update (see Snijder et al., 2012).

elements, such as Pb, Cd, or As. Only Mg and Zn blood levels of this metallic element are related to maternal
concentrations were suggestive of a possible signifi- but not to cord blood levels (Esteban-Vasallo et al.,
cant beneficial influence on TTP. 2012). In terms of toxicokinetics, a correlation between
Cd placental levels and the expression of placental
metallothionein has been described (Sonawane et al.,
7  OTHER EFFECTS ON PREGNANCY 1975; Kippler and Hoque, 2010).
The role of the placenta as a barrier against Hg
The effects of metallic elements on female repro- is not completely clear. However, cellular uptake of
duction may derive from their action at several stages, this metal appears to be related to its chemical struc-
beginning with the fetus, and may include manifesta- ture. It has been observed that Hg vapor and methyl-
tions, such as intrauterine growth retardation, sponta- Hg easily pass the placenta by passive transport and
neous abortion, malformation, and birth defects. amino acid carriers, respectively (Ask et al., 2002);
Spontaneous abortion may result from a number in contrast, inorganic Hg is more commonly accu-
of causes, such as genetic alterations in the gametes, mulated in the placenta, thus limiting the amount
failure of implantation processes, or hormonal imbal- reaching the fetus (Yoshida et al., 2011). Lead was
ances. It has therefore been difficult to distinguish the found to easily cross the placental barrier by means
occupational and environmental causes of spontane- of passive diffusion (Goyer, 1990); similarly, a posi-
ous abortion or congenital malformations from those tive correlation was observed in the majority of the
of other factors. studies between placental and cord blood levels
Metallic elements can cross the placenta by various (Esteban-Vasallo et al., 2012). Similarly, in the majority
mechanisms. Maternofetal and fetomaternal diffu- of studies, a positive correlation has been observed
sional transfer depend on the thickness of the sepa- between placental and cord blood levels. Concerning
rating layers. Available studies have shown that Cd toxicodynamics, Pb may alter the calcium-mediated
accumulates in the placental tissues; indeed, placental cellular processes in syncytiotrophoblasts, which
20  Effects of Metallic Elements on Reproduction and Development 413

form the epithelial covering of the embryonic pla- abortion after adjusting for covariates. Consistent with
cental villi; according to Goyer (1990), Pb seems to their findings, Faikoğlu and colleagues (2006) showed
precipitate, along with calcium, in the microvilli that the Pb blood concentration measured at a gesta-
around the trophoblast (Al-Saleh et al., 2011). The Pb tional age of ≤ 20 weeks was not significantly higher in
storage observed in syncytiotrophoblast cells seems aborting pregnancies than in nonaborting pregnancies.
to be related to reduced cytochrome oxidase activity In 2010, Vigeh and coworkers showed that mean
(Reichrtova et al., 1998). PbB concentrations did not differ significantly between
It is still not known by which mechanisms placenta spontaneous abortion cases and ongoing pregnan-
cells efflux Hg, Pb, and Cd. There is experimental cies (351 ± 142 and 383 ± 199 μg/L, respectively) (Vigeh
evidence supporting the idea that the metals prefer- et al., 2010). Although these differences may have
ably bind to GSH, forming a complex (reviewed by a methodological explanation, PbB levels change
Gundacker and Hengstschläger, 2012). In addition, a throughout pregnancy and increase significantly dur-
number of other protein carriers have been identified ing the third trimester. Therefore, a comparison should
in rat placentae (i.e. organic anion-transporting poly- be carried out at the same gestational age for all par-
peptides, organic anion transporters, organic cation ticipants (Hertz-Picciotto, 2000; Gulson et al., 2004). In
transporters, and zinc transporters), which might also addition, it is necessary to consider that in many stud-
be involved in metallic element transport in the human ies blood samples were collected at different gesta-
placenta (Thévenod, 2010). tional ages for both patients and controls, i.e. full-term
Ahmed and coworkers (2011) assessed the effect delivery versus spontaneous abortion, and this aspect
of As on the placenta and reported that As expo- makes it difficult to standardize the findings of differ-
sure during pregnancy appears to enhance placental ent studies.
inflammatory responses (in part by increasing oxida- The mechanism inducing spontaneous abortion is
tive stress), reduce placental T cells, and alter cord not clear; in addition to preconceptional chromosome
blood cytokines. Lead interferes with calcium uptake damage in the egg cell or in testis (see above), or a
of the syncytiotrophoblast (Lafond et al., 2004). Mer- direct teratogenic effect on the fetus, interference with
cury toxicity in the placenta includes disturbances to the maternal/fetal hormone environment or develop-
amino acid transfer, placental oxygen consumption, mental toxicity to the embryo/fetus is possible. Vascu-
enzyme activity, hormonal secretion, and membrane lar effects on the placenta are also plausible; elevated
fluidity (Urbach et al., 1992). blood pressure during pregnancy represents increased
With respect to spontaneous abortion, the best-stud- risks to both mother and fetus (reduced birth weight,
ied metallic elements are As, Hg, and Pb. The role of Pb in abruption placentae, and perinatal mortality).
spontaneous abortion is controversial. Two studies (one The majority of epidemiological studies in As-­
demonstrating mean PbB levels of 71 μg/L in the study contaminated zones have suggested an increase in
group and 52 μg/L in the controls, and the other find- spontaneous abortions/stillbirths or preterm births.
ing maternal PbB levels of approximately 100 μg/L) have Abortion after exposure to As has been reported in the
shown spontaneous abortion to be associated with mod- USA, Hungary, North Chile, and the Indo-Bangladesh
erately elevated PbB levels (Tabacova and ­Balabaeva, region (Cherry et al., 2008). These studies attributed the
1993; Bellinger, 2005). Similarly, a Mexican study revealed effect of As on pregnancy as being related to (1) ­oxidant/
that each elevation of 50 μg/L in PbB increased the risk antioxidant homeostasis in the embryo; (2) early and
of spontaneous abortions 1.8 times; they reported mean late developmental stage, apoptosis, and inflamma-
PbB levels of 120 μg/L for the study group and 101 μg/L tion; and (3) estrogen-mediated gene expression.
for controls (Borja-Aburto et al., 1999). Speculation about how As exposure complicates
Lamadrid-Figueroa and coworkers (Lamadrid- pregnancy is based on data showing that As is trans-
Figueroa et al., 2007) reported that women with a large ferred to the developing embryo through the abundant
plasma:whole PbB ratio may have a higher incidence of placental vasculature, which suggests that As toxicity
spontaneous abortion at a mean PbB level of 62 μg/L, causes spontaneous mammalian abortion by virtue
but failed to demonstrate significant correlations of aberrant placental vasculogenesis and placental
between Pb concentrations and spontaneous abortion, insufficiency.
either in maternal plasma or in whole blood. However, There is evidence for the possibility that placental
no significant relationship was discovered between “translucency” to As might facilitate abnormal devel-
early pregnancy PbB levels and spontaneous abortion opment during vascular invasion of the uterus and
in 351 apparently healthy pregnant women. Multiple thereby “program” the embryo for future loss during
logistic regression analysis also showed no statistically very early gestation (He et al., 2007). Interruption of
significant association of PbB levels with spontaneous hormone signaling pathways, including the steroid
414 Pietro Apostoli and Simona Catalani

hormones estrogen and progesterone, represents an 2006). The association between Cd and reduced birth
alternative mechanism by which As may interfere weights in girls (an estimated decrease of 45 g for every
with gestation in rodents and humans (Sakurai and 1-μg/L increase in maternal urinary Cd) corresponds
Himeno, 2006). to about one-quarter of the estimated effect of smoking
To evaluate prenatal exposure to As in the general during pregnancy on birth weight (∼200 g lower birth
population and its effects on birth size, Guan et al. weight for mothers who smoke than for nonsmoking
(2012) carried out a cross-sectional study in China and mothers) (Rogers, 2009; Kippler et al., 2012).
showed that maternal As concentration was negatively
associated with birth weight, height, and chest circum-
ference, and that fetal As concentration was negatively 8  DEVELOPMENTAL EFFECTS
associated with head circumference.
It was shown that exposure to Cd during gesta- 8.1  Developmental Effects of Prenatal Exposure
tion was associated with reduced birth weight and an
The effects arising from prenatal exposure to metal-
increased number of preterm births. A possible expla-
lic elements have been discussed mainly in terms of
nation for these effects may be the influence of Cd on
development, particularly the effects of the metal-
the synthesis of some hormones, such as hCG, which
lic elements on morphological development, which
plays a vital role in the maintenance and progression of
might result in birth defects as well as in more subtle
pregnancy or placental Cd accumulation (Nishijo et al.,
effects on the developing organism, such as changes in
2002). Another proposed mechanism for the effect of
the growth or maturation of the CNS.
Cd on birth weight is the induction of metallothionein
Some of the effects observed in experimental stud-
in the placenta and the subsequent chelation of essen-
ies are highly specific to the metallic element, and the
tial trace elements, such as Zn and Cu (­Chmielnicka
effect itself may be the only one recorded for the organ-
and Sowa, 1996).
ism. The nature of the exposure, such as an acute single
One study reported a significant inverse associa-
dose, is unlikely to reflect the usual occupational or
tion of the cord blood Cd level with newborn head
environmental exposure of human beings.
circumference and, consistently, with height, weight,
Several studies have confirmed that prenatal expo-
and head circumference up to 3 years of age (Lin et al.,
sure to Pb impairs behavioral and cognitive develop-
2011). However, the effect on fetal growth may be at
ment of children, even at low levels (Lanphear et al.,
least partly related to the accumulation of Cd in the
2005; Plusquellec et al., 2007; Jedrychowski et al., 2009).
placenta because it was associated with the impaired
Mercury is an environmental pollutant that can have
transport of nutrients to the fetus. There are several pos-
neurotoxic effects on the human CNS, particularly
sible mechanisms by which maternal Cd exposure may
during fetal development. Developmental neurotoxic-
directly or indirectly affect size at birth. Some authors
ity was first reported in a Swedish case report in 1952
have shown that pregnant women in rural Bangladesh
and from a serious outbreak in Minamata, Japan, a few
had elevated Cd concentrations in their placentae, and
years later. The exposed patients manifested neurologi-
that these Cd concentrations were inversely associated
cal signs, and some patients exposed in utero were born
with Zn in the cord blood, suggesting that Cd may
with so-called congenital Minamata disease (this is well
disturb Zn transfer to the fetus (Kippler and Hoque,
described with references in Chapter 46, “Mercury”).
2010). Previously, an interaction between Cd and Zn
The passage of metallic elements through the pla-
in the placenta, leading to decreased Zn transfer to
centa (discussed in the previous paragraph) is a crucial
the fetus, had been shown only in vitro and in women
point in the mechanism of its developmental effects.
with increased Cd exposure due to smoking. Since
The modes of action involved mainly include oxida-
insufficient Zn transfer to the fetus is likely to have
tive stress, a direct effect through interaction with
consequences for pregnancy outcomes, especially in
the embryonic structure, and epigenetic effects (DNA
terms of intrauterine growth retardation and preterm
hypomethylation, interaction with enzymes critical
delivery, this has been suggested as one of the main
for fetal development and programming), endocrine
mechanism of Cd-related reduction in birth weight.
effects, immune suppression, and neurotransmitter
Since the effect of Cd exposure on size and weight
alteration.
at birth is greater in females, another possible expla-
nation may be its endocrine-disrupting properties. Cd
8.1.1  Reactive Oxygen Species
seems to affect corticosteroid 11-beta-dehydrogenase
isozyme 2/11-beta-hydroxysteroid dehydrogenase Developmental pathologies may result from the
type 2 (11β-HSD2), a key regulator of maternal glu- endogenous or the xenobiotic-enhanced formation
cocorticoids in the fetoplacental barrier (Yang et al., of ROS, which cause oxidative damage to cellular
20  Effects of Metallic Elements on Reproduction and Development 415

macromolecules and/or alter signal transduction. (Ríos et al., 2009; Zhang et al., 2010). The results indi-
Development may also be adversely affected by the cated that As affects the neurobehavioral parameters
reversible reaction of ROS with transduction proteins, of the developed, as well as the developing, brain.
thereby altering embryonic or fetal signal transduction There is increasing evidence that maternal Cd expo-
pathways. sure causes relatively specific forelimb ectrodactyly
Teratogenicity probably depends, to a large extent, (the deficiency or absence of one or more central dig-
upon a balance between the pathogenic pathways of its) in mouse strains when administered during early
xenobiotic bioactivation, oxidative macromolecular limb development (Robinson et al., 2009, 2011). Cd-
damage, and signal transduction, on one hand, and, on induced fetal malformations might be associated with
the other, the protective pathways of maternal elimi- impairment of placental development and a reduction
nation, embryonic detoxification of xenobiotic reactive in placental transport capacity. Maternal Cd exposure
intermediates and ROS, and embryonic pathways for during pregnancy induces placental endoplasmic retic-
the detection and repair of oxidative DNA damage. ulum stress and an unfolded protein response, which
A potential indirect mechanism by which maternal may contribute to Cd-induced impairment in placen-
and other extraembryonic pathways can modulate tal and fetal development. ROS, possibly originating in
ROS-mediated teratogenesis is via the production of trophoblast giant cells, mediate Cd-evoked placental
diffusible factors that can traverse the placental and endoplasmic reticulum stress and an unfolded protein
cellular membranes into the embryo and alter embry- response (Wang et al., 2012).
onic determinants of oxidative macromolecular dam- Some neurotoxic effects of mercurial compounds
age or signal transduction. This is exemplified by the on the different stages of development can also be
peroxynitrite pathway, in which nitric oxide synthases attributed to increased ROS. Methylmercury, a potent
(NOSs) produce relatively stable and readily diffus- neurotoxicant, can easily pass through the blood-
ible nitric oxide (NO) (Marnett et al., 2003). Once in the brain barrier and accumulate in brain regions, causing
embryo, NO can alter embryonic signal transduction severe irreversible damage. However, the neurotoxic
pathways, or react with superoxide to directly produce effects and action mechanisms of methylmercury are
embryopathic hydroxyl radicals and reactive nitrogen still unclear, especially in low-dose and long-term
species or peroxynitrite, the latter being capable of ini- exposures. Furthermore, the alteration of lipid per-
tiating the hydroxylation and nitration of embryonic oxidation, Na+/K+-ATPase activities, and nitric oxide
proteins and DNA. in brain tissue has contributed to observed neurobe-
As regards to metallic elements in particular, there havioral dysfunction and hearing impairment (Huang
are several recent findings. Arsenic is a known neuro- et al., 2008, 2011).
toxicant and is capable of traversing the placental bar-
rier and reaching the conceptus in cases of maternal
8.1.2  Epigenetic Effects
exposure and also of easily crossing the blood-brain
barrier to modify the CNS metabolism and function Since the DNA sequence is static, genetic suscepti-
at low doses (Xi et al., 2010). An increased level of bility from DNA sequence variations cannot explain
lipid peroxidation and consequent damage to the cell the mechanisms by which prenatal or early child-
membrane due to As and its compounds have been hood metallic elements exposure impact cognition and
reported by numerous investigators. Oxidative stress behavior later in life. One possible mechanistic path-
has been related to damage to cellular lipids, but there way for this phenomenon, which has yet to be fully
are also indications that As, due to its interference in explored in humans, is epigenetics. Epigenetics is the
methylation reactions and coupling with thiol groups, study of heritable changes in gene expression that occur
may induce independent effects on proteins and without changes in DNA sequence. Such changes can
DNA (Zhong and Mass, 2001). Xi et al. (2010) found have effects as important as those exerted by mutations
increased oxidative stress in the brain after prenatal but, unlike mutations, they are reversible and respon-
exposure, resulting in lowered GPx. sive to environmental influences. DNA methylation is
Human fetal brain tissue explants maintained in the best studied of the epigenetic processes that regu-
culture in the presence of As have shown the charac- late gene silencing. In general, increased methylation
teristics of cell division, cellular growth, and the devel- is inversely associated with gene expression (Wright
opment of a neuronal network that oppose the signs and Baccarelli, 2007). The growing interest in epigen-
of cellular damage, loss of ground matrix, weakening etic markers is a result of their potential to explain the
of the synaptosomes, and cell loss. Arsenic toxicity fetal origins of a disease, or even simply to explain
appears to act through interference in the tissue homeo- the latency between exposure to toxic substances and
stasis of cell growth and development, and apoptosis subsequent disease phenotypes. Several studies have
416 Pietro Apostoli and Simona Catalani

established an association between DNA methylation 8.1.3  Immune Suppression


and environmental metallic elements, including Ni,
Some metallic elements may have effects on the
Cd, Pb, and, particularly, As (Dolinoy et al., 2007). Oxi-
immune system through prenatal exposure and altera-
dative stress may be a unifying process responsible for
tions to thymic function.
these findings across different metallic elements. Oxi-
Prenatal As exposure is associated with increased
dative DNA damage can interfere with the ability of
infant morbidity and reduced thymus size, indicating
methyltransferases to interact with DNA, thus result-
arsenic-related developmental immunotoxicity. The
ing in the generalized hypomethylation of cytosine
child thymic index for thymic function was evaluated
residues (Valinluck et al., 2004).
at birth in a Bangladeshi cohort: prenatal As exposure
In addition, Takiguchi et al. (2003) showed that
was associated with reduced thymic function, possi-
Cd inhibits DNA methyltransferases in a manner
bly via the induction of oxidative stress and apoptosis,
that is noncompetitive with respect to the DNA sub-
suggesting subsequent immunosuppression in child-
strate. This finding is suggestive of interference in the
hood (Raqib et al., 2009; Ahmed et al., 2012).
enzyme-DNA interaction, possibly through an interac-
Cadmium also dysregulated two signaling path-
tion of Cd with the methyltransferase DNA-binding
ways in the thymus, resulting in altered thymocyte
domain.
development (Hanson et al., 2010).
Most of the information regarding the mechanism
Hanson and colleagues (2012) suggested that even
of action was obtained from experimental studies on
very low levels of Cd exposure during gestation can
As exposure. Arsenic mainly causes the induction of
result in long-term detrimental effects on the immune
hypomethylation, possibly through inhibition of DNA
system of the offspring, and that these effects are, to
methyltransferases (Cui et al., 2006), but hypermeth-
some extent, sex specific. The analysis of cytokine pro-
ylation of the TP53 and p16 tumor suppressor gene
duction using stimulated splenocytes demonstrated
promoters in arsenic-exposed people has also been
that prenatal Cd exposure decreased interleukin-2
reported (Chanda et al., 2006). Since DNA methyla-
(IL-2) and IL-4 production by cells in female offspring
tion is an important mechanism of fetal programming,
at 2 weeks of age. Prenatal Hg exposure exerts organ-
As-induced changes in DNA methylation may have
specific effects on cell number, proliferation, and cyto-
severe consequences for the development of health
kine production in preweaned mouse pups. At adult-
defects both before and after birth.
hood, the effects of prenatal Hg exposure could still
Kile and coworkers (2012) carried out the first epide-
be observed, but they differed in females and males.
miological study to note an association between As and
In adult females, an inhibitory effect was observed on
DNA methylation in newborns, and showed that expo-
cytokine production by thymocytes, lymph node cells,
sure to higher levels of As is positively associated with
and splenocytes; in males, a stimulatory effect was
general DNA methylation and, to a lesser extent, with
observed (Silva et al., 2005). Furthermore, gestational
methylation sites within the promoter region of p16.
exposure to Hg leads to persistent changes in the T-cell
Arsenic exerts its toxic effects on the developing
phenotype. A significant reduction in splenic cells
fetus by inhibiting enzymes such as thioredoxin reduc-
in mercury-exposed females, but not in males, was
tase, methyltransferase, and enzymes of the DNA
observed; moreover, the production of IL-4 and inter-
repair mechanism, and disrupting the homeostasis
feron (IFN) from mercury-exposed male and female
of neurotransmitters and hormone secretion.
mice was significantly increased, while IL-2 and IL-10
Moreover, numerous studies have reported altera-
levels were unaffected (Pilones et al., 2009).
tions in the expression pattern of genes involved in
reproductive cell formation. Flora and Mehta (2009)
8.1.4  Effects on Neurotransmitters
reported altered expression of gene markers of the
development and differentiation of all germ layers Metallic elements, such as Al, As, Hg, and Pb may
during embryogenesis. They studied the expression of affect chemical synaptic transmission in the brain and
approximately 47 genes, including markers of embry- the peripheral nervous system, beginning from the
onic cell development, differentiation, apoptosis, gestational period.
and cell cycle in human embryonic stem cell-derived Perinatal As exposure may disrupt the regulatory
embryoid bodies. Arsenic treatment significantly interaction between the hypothalamic-pituitary-adrenal
downregulated the majority of genes studied. These axis and the serotonergic system in dorsal hippocampal
developmental effects were also validated in vivo, formation in a manner that predisposes affected off-
where a significant reduction in litter size along with spring to depressive-like behavior (­Martinez et al., 2008).
an increased incidence of visceral and skeletal defects Arsenic also affected neurotransmitter metabolism by
was observed following As exposure. increasing acetylcholinesterase activity and glutamate
20  Effects of Metallic Elements on Reproduction and Development 417

decarboxylase mRNA expression, and decreasing glu- epigenetic modulation of the neuronal gene expression
tamine synthetase activity and ABAT mRNA (encoding required for learning and memory.
4-aminobutyrate aminotransferase) expression (Xie et al., Early life Pb exposure decreases learning, attention,
2007). Maternal Pb exposure can have consequences on and IQ, and contributes to hyperactivity, impulsive-
the dopaminergic-nitrergic interaction in the nigrostria- ness, and aggression (Schettler, 2001; Grandjean and
tal pathway (Nowak et al., 2008) and on monoaminer- Landrigan, 2006). Preschool Pb exposure has been
gic neuronal function at an adult stage of life, generally associated, using an ecological correlation, with com-
accentuating the behavioral effects of direct and indirect plex outcomes as varied as poor school achievement,
monoaminergic agonists (Szczerbak et al., 2007). unwed pregnancy, and violent crime (Nevin, 2009;
­Carpenter and Nevin, 2010).
Lanphear and coworkers (2005) collected data from
8.2  Developmental Effects of Neonatal
1333 subjects, where the IQ score was the primary
Exposure
outcome measure. After adjustment for covariates,
In addition to evidence for the effects of prenatal expo- decreases in the estimated IQ point associated with an
sure to metallic elements, child health outcomes (cogni- increase in PbB from 24-100 μg/L, 100-200 μg/L, and
tive function, behavior problems, and motor and sensory 200-300 μg/L were 3.9, 1.9, and 1.1, respectively. This
function) related to postnatal/neonatal exposure have data suggests that the dose-response curve is steeper at
also been reported in several epidemiological studies. lower levels of exposure than at higher levels (Figure 4).
The neurotoxic mechanisms of action in children PbB levels of 100 μg/L, as proposed by the U.S. Cen-
are comparable to those in adults; however, a child’s ters for Disease Control and Prevention (CDC, 1991),
brain is more susceptible and sensitive to neurotoxins. were considered worrisome and were, for a long time,
The neurotoxicity of metallic elements is specifically accepted as the guideline value. Since then, there has
addressed in Chapter 15 “Neurotoxicology of Metals.” been a significant worldwide decrease in the PbB lev-
Exposure to As through drinking water has been els in children, and the current geometric mean values
associated with impaired cognitive function in school- in European countries range from 20 to 30 μg/L.
aged children. Using multivariable-adjusted regres- The European Food Safety Authority (EFSA, 2010)
sion analyses checking for all potential confounders concluded that an increase in the PbB level by 12 μg/L
and patients lost to follow-up, the authors found the could decrease the IQ score by one point. It seems that
verbal intelligence quotient (VIQ) and full scale intelli- this value can be used as a “unit risk” for calculating
gence quotient (FSIQ) to be negatively associated with a possible decrease in IQ and, consequently, influenc-
the log of the urinary concentration of As (U-As) in ing low-level exposure to Pb (< 100 μg/L) on the health
girls. In boys, the U-As showed a consistently low and and socioeconomic status of the exposed population.
nonsignificant association with all IQ measurements. The Scientific Committee on Neurotoxicology and
An effect size calculation indicated that 100 μg/L U-As Psychophysiology and the Scientific Committee on the
was associated with a decrease of 1-3 points in both Toxicology of Metals of the International Commission
VIQ and FSIQ in girls (Hamadani et al., 2011).
Recently, an epidemiological study of developmental
neurotoxicology has increased the focus on coexposure 105
to As and Mn (Wasserman et al., 2011). To investigate
the possible synergistic impact of simultaneous expo-
100
sures, a sample of 299 children (8-11 years old) was
recruited and stratified according to As (above and
below 10 μg/L) and Mn (above and below 500 μg/L) 95
IQ

concentrations in household water wells. Both As


and Mn in whole blood were significantly negatively
90
related to most Wechsler Intelligence Scale for Children
IV subscale scores. With further adjustment for sociode-
mographic features and for ferritin, levels of Mn in the 85
blood remained significantly associated with reduced 0 10 20 30 40
“Perceptual Reasoning” and “Working Memory” Concurrent blood lead (µg/dL)
scores; as expected, the association with As was nega-
FIGURE 4  Linear models of concurrent blood lead levels and in-
tive and was significant for “Verbal Comprehension.” telligence quotient (IQ) adjusted for some covariates (maternal edu-
The neurodevelopmental toxicity of As is mediated cation, maternal IQ, and birth weight). Reproduced with permission
by an epigenetic mechanism: As interferes with the from Environmental Health Perspectives (Lanphear et al., 2005).
418 Pietro Apostoli and Simona Catalani

on Occupational Health declared that the action level and abstention from fish consumption in the local pop-
for children should be immediately reduced to a PbB ulations (JECFA, 2003; Grandjean and Herz, 2011).
of 50 μg/L (Landrigan et al., 2007). In 1997, amid concerns about a possible link between
In 2012, the CDC defined a reference level of 50 μg/L vaccines containing mercury-based chemical thiomer-
to identify children with elevated blood lead levels sal and autism, the U.S. Food and Drug Administra-
(Betts, 2012). tion was called upon to review the risks of all food and
In recent years, the mechanism of action of Pb neu- drugs that contain mercury. In 1999, despite the lack of
rotoxicity in the developmental brain has been eluci- scientific evidence, the CDC issued a recommendation
dated. Lead targets the ionotropic glutamate receptor/ to manufacturers that “thiomersal-containing vaccines
N-methyl-d-aspartate receptor (NMDAR) and alters should be removed as soon as possible” (Siva, 2012).
those physiological processes that are NMDAR depen- More than a decade later, despite several independent
dent, including hippocampus long-term potentiation. studies and consultations that did not find any asso-
Learning a behavioral task depends greatly on normal ciation between thiomersal vaccines and autism, the
hippocampus function; thus, during its developmental debate continues (Schultz et al., 2010; Siva, 2012).
period, the brain is highly vulnerable to the presence Several mechanisms have been proposed through
of Pb. Lead exposure induces alterations in NR1 and which methylmercury might damage the developing
NR2 (encoding NMDAR subunits) mRNA expression, brain. Among them are methylmercury-induced alter-
essentially in the hippocampus region, indicating the ations in microtubules, oxidative damage to neurons,
regional selectivity of Pb. impairment of neuronal and glial calcium homeostasis,
In addition, Pb disrupts the normal development of and potentiation of glutamatergic neurotransmission
the brain, causing a reduction in cellular development (Castoldi et al., 2008). The effect of disrupting microtu-
that can be seen at the dendritic, axonal, and synaptic bules and consequently mitosis, migration, and cortical
levels in different brain regions. Lead exposure also organization of neurons is especially serious prenatally,
disrupts the aminergic system in the cortex, cerebel- although these processes continue postnatally (Rodier,
lum, and hippocampus, thus possibly contributing 2004; Clarkson, 1987). Chemically, methylmercury has
to cognitive and behavioral impairment, especially in a strong affinity for the sulfhydryl groups present in
rats exposed to Pb during the developmental period proteins and GSH. When methylmercury forms com-
(Neal and Guilarte, 2010; Neal et al., 2010, 2011; Xu plexes with these compounds, it can adversely affect
et al., 2009). anabolic processes and protein synthesis. By inactivat-
Methylmercury is recognized as another important ing sulfhydryl enzymes, methylmercury can interfere
developmental neurotoxicant, although this insight with cellular metabolism and function. Methylmer-
developed slowly over many decades. Nonetheless, cury is also known to catalyze the formation of excess
exposure limits for this environmental chemical were ROS, and the regional distribution of this activity par-
based solely on adult toxicity for 50 years after the allels the sites of known neuropathological changes.
first report on developmental neurotoxicity. Meth- Methylmercury rapidly binds to reduced GSH, which
ylmercury poisoning in children shows more widely is present in most cells in millimolar concentrations
distributed damage on the brain; numerous findings (Farina et al., 2011). Such binding may serve to protect
strongly support the notion that early developmental intracellular proteins. There are also adverse effects of
exposure causes much more serious disease in chil- methylmercury on the synthesis of fetal DNA in astro-
dren than in individuals exposed as adults. Current cytes and on the growth cones of neurons.
evidence is affected by uncertainty, most importantly
by the imprecision of exposure assessment in epide-
miological studies. Detailed calculations suggest that 9  CONCLUDING REMARKS AND THE
the relative degree of imprecision may be as high as NEED FOR FUTURE RESEARCH
50% or greater, thereby substantially biasing the results
toward the null. In addition, as methylmercury expo- At present, for only a few metal ions such as Pb,
sure usually originates from fish and seafood, which As, Cd, and Hg extensive evaluations of their poten-
also contain essential nutrients, so-called negative con- tial effect on reproduction and development have been
founding may occur (Grandjean and Herz, 2011). done and their relative mode of action is known. How-
In the most recent regulatory assessments of meth- ever, in the general and occupational environments,
ylmercury neurotoxicity, the experts decided to take many other metallic elements (Cr, Ni, Platinum, Pal-
the benefits of fish consumption into account, thereby ladium, Rhodium, V, and Sb) are present, and there is
allowing greater methylmercury exposure in order to evidence that they can interfere with reproduction or
avoid causing a decrease in fish intake or undue panic development.
20  Effects of Metallic Elements on Reproduction and Development 419

A better knowledge of the mechanisms of action (­Catalani et al., 2013), and further research is needed to
involved and of the different target sites is crucial for define potential risks because exposure to V has been
understanding this complex and heterogeneous phe- associated with decreased fertility, embryolethality,
nomenon; this represents one of the main goals of fetotoxicity, and teratogenicity (Domingo, 1996, 2002).
future research. Furthermore, some experimental studies have evalu-
An appropriate use of the diagnostic approach ated the effect of rare earth elements such as scandium,
in occupational medicine is necessary to reach the yttrium, lanthanum (La), cerium (Ce), neodymium,
­“Bradford Hill Criteria” and obtain adequate evidence and gadolinium. The offspring of Ce-exposed, but
for a causal relationship between an incidence and a not of La-exposed, sperm displayed a significant con-
consequence. centration-related increase in developmental defects
An in-depth characterization of doses is another (Oral et al., 2010). Cerium also caused significant life-
critical requirement; this requires knowledge of ele- span shortening, accompanied by oxidative damage,
mental speciation, the identification of more adequate and extended the developmental time of Drosophila
matrices, nearest to the critical organs or tissues, and melanogaster (Wu et al., 2012). Many radiology depart-
determination of the dose-response relationship and ments have discouraged the use of gadolinium-based
the threshold for action. contrast media in pregnant or lactating women due to
For some metals, another aspect is new routes of expo- a lack of knowledge regarding the risks for the fetus
sure. For example, the release of Co and Cr by metallic and an unwillingness to expose neonates to unnec-
prostheses constitutes a new risk. Metal-on-metal bear- essary drugs. However, documentation and reports
ings have frequently been used by orthopedic surgeons, regarding the possible adverse effects of gadolinium-
especially in young and active patients (Cui et al., 2005), based contrast media on the fetus are sorely lacking
and hip surgery can be done during child-bearing age ­(Sundgren and Leander, 2011). After the introduction
(Stea et al., 2007). However, the implantation of metal- of automobile catalytic converters, platinum, palla-
on-metal hip devices can increase metal ion serum lev- dium, and rhodium have been emitted with exhaust
els (Hartmann et al., 2013). This has led to a concern that fumes, and increasing levels have been found in dif-
disseminated metal ions, such as Cr and Co (the main ferent environmental matrices such as road dusts, soils
constituents of the alloy used), could affect pregnan- along heavily frequented roads, and the sediments
cies in women owing to transplacental transfer. Current of urban rivers. Compared with other heavy metals,
research into the transplacental passage of metal ions the biological availability of platinum, palladium, and
suggests that 29% of Cr and 60% of Co ions cross the rhodium in some experimental studies on road dusts
placenta in mothers with metal-on-metal hip resurfacing ranged between those of Cd and Pb. Morphological
(Ziaee et al., 2007). A few studies and case reports have and functional effects on rat ovaries and testicular
explored the question of pregnancy and childbirth after tissue in rat (Ciftci et al., 2011), embryo lethality, and
total hip arthroplasty (THA) (Stea et al., 2007; Boot et al., teratogenic effects (Sharani et al., 2011) have been dem-
2003; Sierra et al., 2005). These reports primarily found onstrated after exposure to Pt complexes.
that pregnancy and vaginal delivery are safe after THA, We have already stated that current experimental
although the majority of women with this type of sur- and human studies that deal with exposure to a single
gery delivered by elective cesarean. These reports did metallic element contrast with the real environmental
not address the teratogenic or fetotoxic risk of the materi- and occupational exposure, which are generally char-
als used. The available data are reassuring because they acterized by many metal ions and other organic sub-
show that pregnancy and childbirth are not affected by stances. Therefore, identifying the possible synergic,
the presence of a hip device (Sierra et al., 2005) and that additive, multiplicative, or competitive effects of metal
pregnancy-related complications in women with THA coexposure remains an intriguing yet difficult objec-
do not differ from those in healthy women (Meldrum tive for future research.
et al., 2003). Fritzsche and coworkers (2012) described
a 41-year-old patient with bilateral metal-on-metal hip
arthroplasties, a recurrent pseudotumor, and extremely References
high metal blood levels (Cr 39 μg/L, Co 138 μg/L) at 12 Acharya, U.R., Mishra, M., Tripathy, R.R., et al., 2006. Reprod.
weeks of gestation. At 38 weeks of gestation, a healthy Toxicol. 22, 87–91.
male infant was delivered with elevated Cr and Co cord Acharya, U.R., Mishra, M., Patro, J., et al., 2008. Reprod. Toxicol. 25,
84–88.
blood levels; however, the infant’s development appeared
Ahmed, S., Mahabbat-e Khoda, S., Rekha, R.S., et al., 2011. Environ.
to be uneventful and no signs of toxicity were obvious. Health Perspect. 119, 259–264.
The release of V has also been reported in patients Ahmed, S., Ahsan, K.B., Kippler, M., et al., 2012. Toxicol. Sci. 129,
implanted with a titanium alloy hip prosthesis 305–314.
420 Pietro Apostoli and Simona Catalani

Akram, Z., Jalali, S., Shami, S.A., et al., 2009. Toxicol. Lett. 190, CDC (Centers for Disease Control and Prevention), 1991. Preventing
81–85. Lead Poisoning in Young Children. 1991. Available at U.S. Depart-
Al-Saleh, I., Shinwari, N., Mashhour, A., et al., 2011. Int. J. Hyg. ment of Health and Human Services, Atlanta (accessed 31.01.14).
Envir. Heal. 214, 79–101. http://www.cdc.gov/nceh/lead/publications/books/
Amara, S., Abdelmelek, H., Garrel, C., et al., 2008. J. Reprod. Dev. plpyc/contents.htm.
54, 129–134. Chanda, S., Dasgupta, U.B., Guhamazumder, D., et al., 2006. Toxicol.
Angelopoulou, R., Lavranos, G., Manolakou, P., et al., 2009. Reprod. Sci. 89, 431–437.
Toxicol. 28, 167–171. Chandra, A.K., Ghosh, R., Chatterjee, A., et al., 2007. J. Inorg.
Apostoli, P., Bellini, A., Porru, S., et al., 2000. Am. J. Ind. Med. 38, Biochem. 101, 944–956.
310–315. Chandra, A.K., Ghosh, R., Chatterjee, A., et al., 2010. Toxicol. Mech.
Apostoli, P., Telisman, S., Sager, P., 2007. Chapter 12 Reproductive Methods 20, 306–315.
and Developmental Toxicity of Metals. In: Nordberg, G.F., et al. Chatterjee, A., Chatterji, U., 2010. Reprod. Biol. Endocrinol. 8, 80.
(Eds.), Handbook on the Toxicology of Metals, third ed. pp. Chattopadhyay, S., Pal Ghosh, S., Ghosh, D., et al., 2003. Toxicol. Sci.
213–249. 75, 412–422.
Apostoli, P., Kiss, P., Porru, S., et al., 1998. Occup. Environ. Med. 55, Cherry, N., Shaikh, K., McDonald, C., et al., 2008. Bull. World Health
364–374. Organ. 86, 172–177.
Arabi, M., Mohammadpour, A.A., 2006. Vet. Res. Commun. 30, Chia, S.E., Ong, C.N., Lee, S.T., et al., 1992. Arch. Androl. 29, 177–183.
943–951. Chmielnicka, J., Sowa, B., 1996. Ecotoxicol. Environ. Saf. 35, 277–281.
Aragon, M.A., Ayala, M.E., Fortoul, T.I., et al., 2005. Reprod. Toxicol. Ciftci, O., Beytur, A., Cakir, O., et al., 2011. Basic Clin. Pharmacol.
20, 127–134. Toxicol. 109, 328–333.
Ask, K., Akesson, A., Berglund, M., et al., 2002. Environ. Health Clarkson, T.W., 1987. Environ. Health Perspect. 75, 59–64.
Perspect. 110, 523–526. Cui, W., Ma, C.X., Tang, Y., et al., 2005. Birth Defects Res. A Clin. Mol.
Avazeri, N., Denys, A., Lefèvre, B., 2006. Biochimie. 88, 1823 Teratol. 73, 876–880.
–1829. Cui, X., Wakai, T., Shirai, Y., et al., 2006. Hum. Pathol. 37, 298–311.
Barregård, L., Lindstedt, G., Schütz, A., et al., 1994. Occup. Environ. Damstra, T., Barlow, S., Bergamn, A., et al., 2002. In: global assess-
Med. 51, 536–540. ment of the state of the science of endocrine disruptions. WHO,
Bars, R., Fegert, I., Gross, M., et al., 2012. Regul. Toxicol. Pharmacol. Geneva (accessed 17.01.14). http://www.who.int/ipcs/publicati
64, 143–154. ons/new_issues/endocrine_disruptors/en/.
Bellinger, D.C., 2005. Birth Defects Res. A Clin. Mol. Teratol. 73, Das, J., Ghosh, J., Manna, P., et al., 2009. Toxicol. Lett. 187, 201–210.
409–420. Dolinoy, D.C., Weidman, J.R., Jirtle, R.L., 2007. Reprod. Toxicol. 23,
Benoff, S., Hurley, I.R., Millan, C., et al., 2003. Fertil. Steril. 80, 297–307.
517–525. Domingo, J.L., 1996. Reprod. Toxicol. 10, 175–181.
Betts, K.S., 2012. Environ. Health Perspect. 120. 268. Domingo, J.L., 2002. Biol. Trace Elem. Res. 88, 97–112.
Björndahl, L., Kvist, U., 2010. Mol. Hum. Reprod. 16, 23–29. Doumouchtsis, K.K., Doumouchtsis, S.K., Doumouchtsis, E.K., et al.,
Björndahl, L., Kvist, U., 2011. Syst. Biol. Reprod. Med. 57, 86–92. 2009. J. Endocrinol. Invest. 32, 175–183.
Bloom, M.S., Louis, G.M., Sundaram, R., et al., 2011. Reprod. Toxicol. Du Plessis, S.S., McAllister, D.A., Luu, A., et al., 2010. Andrologia
31, 158–163. 42, 206–210.
Bloom, M.S., Kim, K., Kruger, P.C., et al., 2012. J. Assist. Reprod. EFSA (European Food Safety Authority), 2010. Panel on Contami-
Genet. 29, 1369–1379. nants in the Food Chain (CONTAM). Scientific Opinion on Pb in
Bodwell, J.E., Kingsley, L.A., Hamilton, J.W., 2004. Chem. Res. Food 8, 1570.
Toxicol. 17, 1064–1076. Eibensteiner, L., Del Carpio Sanz, A., Frumkin, H., et al., 2005. Int. J.
Bonde, J.P., 1990. Dan. Med. Bull. 37, 105–108. Occup. Environ. Health 11, 161–166.
Bonde, J.P., 2002. G. Ital. Med. Lav. Ergon. 24, 112–117. Ernst, E., Lauritsen, J.G., 1991. Pharmacol. Toxicol. 68, 440–444.
Bonde, J.P., Joffe, M., Apostoli, P., et al., 2002. Occup. Environ. Med. Esteban-Vasallo, M.D., Aragonés, N., Pollan, M., et al., 2012. Environ.
59, 234–242. Health Perspect. 120, 1369–1377.
Bonde, J.P., Joffe, M., Sallmén, M., et al., 2006. Epidemiol. 17, Faikoğlu, R., Savan, K., Utku, C., et al., 2006. J. Environ. Sci. Health A
347–349. Tox. Hazard Subst. Environ. Eng. 41, 501–506.
Boot, C.L., Heyligers, I.C., Heins, K.F., 2003. Orthopedics 26, Fang, M.Z., Mar, W.C., Cho, M.H., 2001. Toxicol. 161, 117–127.
813–814. Farina, M., Aschner, M., Rocha, J.B., 2011. Toxicol. Appl. Pharmacol.
Borja-Aburto, V.H., Hertz-Picciotto, I., Rojas Lopez, et al., 1999. Am. 256, 405–417.
J. Epidemiol. 150, 590–597. Flora, S.J., Mehta, A., 2009. Biochem. Pharmacol. 78, 1340–1349.
Boujbiha, M.A., Hamden, K., Guermazi, F., et al., 2011. Biol. Trace Forgacs, Z., Massányi, P., Lukac, N., et al., 2012. J. Environ. Sci.
Elem. Res. 142, 598–610. Health. A Tox. Hazard. Subst. Environ. Eng. 47, 1249–1260.
Buck Louis, G.M., Sundaram, R., Schisterman, E.F., et al., 2012. Fort, D.J., Stover, E.L., Bantle, J.A., et al., 2001. J. Appl. Toxicol. 21,
Chemosphere 87, 1201–1207. 41–52.
Bunn, T.L., Parsons, P.J., Kao, E., et al., 2001. J. Toxicol. Environ. Fossato da Silva, D.A., Teixeira, C.T., Scarano, W.R., et al., 2011.
Health A. 64, 223–240. Reprod. Toxicol. 31, 431–439.
Burukoğlu, D., Bayçu, C., 2008. Bull. Environ. Contam. Toxicol. 81, Fritzsche, J., Borisch, C., Schaefer, C., 2012. Clin. Orthop. Relat. Res.
521–524. 470, 2325–2331.
Carpenter, D.O., Nevin, R., 2010. Physiol. Behav. 99, 260–268. Garcia-Morales, P., Saceda, M., Kenney, N., et al., 1994. J. Biol. Chem.
Castellini, C., Mourvakia, E., Sartin, B., et al., 2009. Reprod. Toxicol. 269, 16896–16901.
27, 46–54. Gerhard, I., Monga, B., Waldbrenner, A., et al., 1998. J. Toxicol. Environ.
Castoldi, A.F., Onishchenko, N., Johansson, C., et al., 2008. Regul. Health A. 54, 593–611.
Toxicol. Pharmacol. 51, 215–229. Goyer, R.A., 1990. Environ. Health Perspect. 89, 101–105.
Catalani, S., Stea, S., Beraudi, A., 2013. Clin. Toxicol. 51, 550–556. Grandjean, P., Herz, K.T., 2011. Mt. Sinai J. Med. 78, 107–118.
20  Effects of Metallic Elements on Reproduction and Development 421

Grandjean, P., Landrigan, P.J., 2006. Lancet 368, 2167–2178. Kippler, M., Tofail, F., Gardner, R., et al., 2012. Environ. Health
Grandjean, P., Satoh, H., Murata, K., et al., 2010. Environ. Health Per- Perspect. 120, 284–289.
spect. 118, 1137–1145. Kluxen, F.M., Höfer, N., Kretzschmar, G., et al., 2012. Arch. Toxicol.
Greene, L.E., Riederer, A.M., Marcus, et al., 2010. Int. J. Occup. Envi- 86, 591–601.
ron. Health 16, 60–68. Kumar, S., Sathwara, N.G., Gautam, A.K., et al., 2005. J. Occup.
Guan, H., Piao, F., Zhang, X., et al., 2012. Biol. Trace Elem. Res. 149, Health 47, 424–430.
10–15. Lafond, J. Hamel, A., Takser, L., et al., 2004. J. Toxicol. Environ.
Gulson, B.L., Mizon, K.J., Palmer, J.M., et al., 2004. Environ. Health Health. A. 67, 1069–1079.
Perspect. 112, 1499–1507. Lamadrid-Figueroa, H., Téllez-Rojo, M.M., Hernández-Avila, M.,
Gundacker, C., Hengstschläger, M., 2012. Wien. Med. Wochenschr. et al., 2007. BMC Pregnancy Childbirth. 27 (7), 22.
162, 201–206. Landrigan, P., Nordberg, R., Lucchini, R., et al., 2007. Am. J. Ind.
Hamadani, J.D., Tofail, F., Nermell, B., 2011. Int. J. Epidemiol. 40, Med. 50, 709–711.
1593–1604. Lanphear, B.P., Hornung, R., Khoury, J., et al., 2005. Environ. Health
Hammond, J., Le, Q., Goodyer, C., et al., 2001. J. Neurochem. 77, Perspect. 113, 894–899.
1319–1326. Latini, G., De Felice, C., Presta, G., et al., 2003. Biol. Neonate. 83,
Hanson, M.L., Brundage, K.M., Schafer, R., et al., 2010. Toxicol. Appl. 22–24.
Pharmacol. 242, 136–145. Lavranos, G., Balla, M., Tzortzopoulou, A., et al., 2012. Reprod.
Hanson, M.L., Holásková, I., Elliott, M., et al., 2012. Toxicol. Appl. Toxicol. 34, 298–307.
Pharmacol. 261, 196–203. Leoni, G., Bogliolo, L., Deiana, G., et al., 2002. Reprod. Toxicol. 16,
Hartmann, A., Hannemann, F., Lützner, J., et al., 2013. PLoS One 8, 371–377.
70359. Li, H., Chen, Q., Li, S., et al., 2001. Ann. Occup. Hyg. 45, 505–511.
He, W., Greenwell, R.J., Brooks, D.M., et al., 2007. Toxicol. Sci. 99, Lin, C.M., Doyle, P., Wang, D., et al., 2011. Occup. Environ. Med. 68,
244–253. 641–646.
Heath, J.C., Abdelmageed, Y., Braden, T.D., et al., 2012. J. Biomed. Llop, S., Guxens, M., Murcia, M., et al., 2012. Am. J. Epidemiol. 175,
Biotechnol. 2012, 815186. 451–465.
Henson, M.C., Chedrese, J., 2004. Exp. Biol. Med. 229, 383–392. Mahfouz, R.Z., Du Plessis, S.S., Aziz, N., et al., 2010. Fertil. Steril. 93,
Hernandez-Ochoa, I., Garcıa-Vargas, G., Lopez-Carrillo, L., et al., 814–821.
2005. Reprod. Toxicol. 20, 221–228. Makker, K., Agarwal, A., Sharma, R., 2009. Indian J. Med. Res. 129,
Hertz-Picciotto, I., 2000. Am. J. Ind. Med. 38, 300–309. 357–367.
Hjollund, N.H., Bonde, J.P., Jensen, T.K., 1998. Reprod. Toxicol. 12, Malagutti, K.S., Da Silva, A.P., Braga, H.C., et al., 2009. Environ. Toxi-
29–37. col. Pharmacol. 27, 293–297.
Huang, C.F., Hsu, C.J., Liu, S.H., et al., 2008. Toxicol. Lett. 176, 188–197. Marchlewicz, M., Michalska, T., Wiszniewska, B., 2004. Chemo-
Huang, C.F., Liu, S.H., Hsu, C.J., et al., 2011. Toxicol. Lett. 201, sphere 57, 1553–1562.
196–204. Marnett, L.J., Riggins, J.N., West, J.D., 2003. J. Clin. Invest. 111,
Huda, S.N., Vahter, M., 2011. Int. J. Epidemiol. 40, 1593–1604. 583–593.
Iavicoli, I., Fontana, L., Bergamaschi, A., 2009. J. Toxicol. Environ. Martin, M.B., Reiter, R., Pham, T., et al., 2003. Endocrinology 144,
Health B Crit. Rev. 12, 206–223. 2425–2436.
Jana, K., Jana, S., Samanta, P.K., et al., 2006. Reprod. Biol. Endocrinol. Martinez, E.J., Kolb, B.L., Bell, A., et al., 2008. Neurotoxicol. 29,
16, 4–9. 647–655.
JECFA (Joint FAO/WHO Expert Committee on Food Additives), Massányi, P., Trandžík, J., Naď, P., et al., 2004. J. Environ. Sci. Health
2003. Summary and conclusions. Sixty-first meeting of the Joint Part A 39, 3005–3014.
FAO/WHO Expert Committee on Food Additives held in Rome, Massányi, P., Lukác, N., Uhrín, V., et al., 2007a. Acta. Biol. Hung. 58,
10–19 June 2003. Report 2003 10–19 June 2003. 287–299.
Jedrychowski, W., Perera, F., Jankowski, J., et al., 2009. Early Hum. Massányi, P., Lukai, N., Zemanovu, J., et al., 2007b. Acta. Vet. Brno.
Dev. 85, 503–510. 76, 223–229.
Jensen, T.K., Bonde, J.P., Joffe, M., 2006. Occup. Med. 56, 544–553. Meldrum, R., Feinberg, J.R., Capello, W.N., et al., 2003. J. Arthroplasty
Joffe, M., 1989. J. Occup. Med. 31, 974–979. 18, 879–885.
Joffe, M., 2003. Am. J. Epidemiol. 157, 89–93. Mendola, P., Messer, L.C., Rappazzo, K., 2008. Fertil. Steril. 89,
Jolibois Jr, L.S., Shi, W., George, W.J., et al., 1999. Reprod. Toxicol. e81–e94.
13, 215–221. Mocevic, E., Specht, I.O., Marott, J.L., et al., 2013. Asian J. Androl.
Jurasović, J., Cvitković, P., Pizent, A., et al., 2004. Biomet. 17, 735 15, 97–104.
–743. Mondal, S., Mukhopadhyay, B., Bhattacharya, S., 1997. Biometals 10,
Jurewicz, J., Kouimintzis, D., Burdorf, A., et al., 2007. J. Public Health 285–290.
15, 265–277. Mondal, S., Mukherjee, S., Chaudhuri, K., et al., 2013. Pharm. Biol.
Kaltreider, R.C., Davis, A.M., Lariviere, J.P., et al., 2001. Environ. 51, 1363–1371.
Health Perspect. 109, 245–251. Mueller, B.R., Bale, T.L., 2008. J. Neurosci. 28, 9055–9065.
Kaludin, I., Georgiev, G.T., Marinov, M.F., 1983. Vet. Med. Nauki. 20, Murthy, R.C., Junaid, M., Saxena, D.K., 1996. Toxicol. Lett. 89,
91–96. 147–154.
Kasperczyk, A., Kasperczyk, S., Dziwisz, M., et al., 2002. Ginekol. Naha, N., Chowdhury, A.R., 2006. J. UOEH. 28, 157–171.
P01. 73, 449–453. Nampoothiri, L.P., Gupta, S., 2008. J. Biochem. Mol. Toxicol. 22,
Kawai, M., Swan, K.F., Green, A.E., et al., 2002. Biol. Reprod. 67, 337–344.
178–183. Nampoothiri, L.P., Agarwal, A., Gupta, S., et al., 2007. Arch. Toxicol.
Kile, M.L., Baccarelli, A., Hoffman, E., et al., 2012. Environ. Health 81, 145–150.
Perspect. 120, 1061–1066. Nandi, S., Gupta, P.S., Selvaraju, S., et al., 2010. Arch. Environ.
Kippler, M., Hoque, A.M.W., 2010. Toxicol. Lett. 192, 162–168. Contam. Toxicol. 58, 194–204.
422 Pietro Apostoli and Simona Catalani

Neal, A.P., Guilarte, T.R., 2010. Mol. Neurobiol. 42, 151–160. Schultz, S.T., 2010. Acta Neurobiol. Exp. (Wars) 70, 187–195.
Neal, A.P., Worley, P.F., Guilarte, T.R., 2011. Neurotoxicol. 32, Sharani, P.V., Lianwu, Y., Gong, Z., et al., 2011. Nanotoxicol. 5, 43–54.
281–289. Sharpe, R.M., 2001. Toxicol. Lett. 120, 221–232.
Neal, A.P., Stansfield, K.H., Worley, P.F., et al., 2010. Toxicol. Sci. 116, Sharpe, R.M., Irvine, D.S., 2004. BMJ 328, 447–451.
249–263. Shiau, C.Y., Wang, J.D., Chen, P.C., 2004. Occup. Environ. Med. 61,
Nesatyy, V.J., Rutishauser, B.V., Eggen, R.I.L., et al., 2005. Analyst 915–923.
130, 1087–1097. Sierra, R.J., Trousdale, R.T., Cabanela, M.E., 2005. J. Bone Joint Surg.
Nesatyy, V.J., Ammann, A.A., Rutishauser, B.V., et al., 2006. Environ. Br. 87, 21–24.
Sci. Technol. 40, 1358–1363. Silbergeld, E.K., Quintanilla-Vega, B., Gandley, R.E., 2003. Adv. Exp.
Nevin, R., 2009. Environ. Res. 109, 301–310. Med. Biol. 518, 37–48.
Nguyen, T.V., Yao, M., Pike, C.J., 2005. J. Neurochem. 94, 1639–1651. Silva, I.A., El Nabawi, M., Hoover, D., et al., 2005. Dev. Comp.
Nishijo, M., Nakagawa, H., Honda, R., et al., 2002. Occup. Environ. Immunol. 29, 171–183.
Med. 59, 394–396. Silva, N., Peiris-John, R., Wickremasinghe, R., et al., 2012. J. Appl.
Noack-Füller, G., De Beer, C., Seibert, H., 1993. Andrologia 25, 7–12. Toxicol. 32, 318–332.
Nordberg, G.F., 1972. Environ. Physiol. Biochem. 2, 7–36. Siu, E.R., Mruk, D.D., Porto, C.S., et al., 2009. Toxicol. Appl. Pharmacol.
Nowak, P., Szczerbak, G., Nitka, D., et al., 2008. Toxicol. 246, 83–89. 238, 240–249.
Oliveira, H., Spanò, M., Santos, C., et al., 2009. Cell. Biol. Toxicol. 25, Siva, N., 2012. Lancet 379, 2328.
341–353. Skandhan, K.P., 1992. Rev. Fr. Gynecol. Obstet. 87, 594–598.
Olivieri, G., Novakovic, M., Savaskan, E., et al., 2002. Neuroscience Slivkova, J., Popelkova, M., Massanyi, P., 2009. J. Environ. Sci. Health
113, 849–855. A Tox. Hazard Subst. Environ. Eng. 44, 370–375.
Oral, R., Bustamante, P., Warnau, M., et al., 2010. Chemosphere 81, Snijder, C.A., Velde, E., Roeleveld, N., et al., 2012. Hum. Reprod.
194–198. Update 18, 284–300.
Paksy, K., Rajczy, K., Forgacs, Z., et al., 1997. J. Appl. Toxicol. 17, Sonawane, B.R., Nordberg, M., Nordberg, G.F., et al., 1975. Environ.
321–327. Health Perspect. 12, 97–102.
Paksy, K., Gáti, I., Náray, M., et al., 2001. J. Toxicol. Environ. Health Srivastava, V., Dearth, R.K., Hiney, J.K., et al., 2004. Toxicol. Sci. 77,
A. 62, 359–366. 35–40.
Pandya, C., Pillai, P., Nampoothiri, L.P., et al., 2012. Andrologia 44, Stea, S., Bordini, B., De Clerico, M., et al., 2007. J. Womens Health
813–822. (Larchmt) 16, 1300–1304.
Piasek, M., Laskey, J.W., 1994. Reprod. Toxicol. 8, 495–507. Stohs, S.J., Bagchi, D., Hassoun, E., et al., 2001. J. Environ. Pathol.
Pilones, K., Tatum, A., Gavalchin, J., 2009. J. Immunotoxicol. 6, Toxicol. Oncol. 20, 77–88.
161–170. Suarez, S.S., Varosi, S.M., Dai, X., 1993. Proc. Natl. Acad. Sci. USA.
Pilsner, J.R., Hall, M.N., Liu, X., et al., 2012. PLoS One 7, e37147. 90, 4660–4664.
Plusquellec, P., Muckle, G., Dewailly, E., et al., 2007. Neurotoxicol. Subramanian, S., Rajendiran, G., Sekhar, P., et al., 2006. Toxicol. Appl.
Teratol. 29, 527–537. Pharmacol. 215, 237–249.
Predki, P.F., Sarkar, B., 1992. J. Biol. Chem. 267, 5842–5846. Sundgren, P.C., Leander, P., 2011. J. Magn. Reson. Imaging. 34,
Prozialeck, W.C., 2000. Toxicol. Appl. Pharmacol. 164, 231–249. 750–757.
Quintanilla-Vega, B., Hoover, D., Bal, W., et al., 2000a. Am. J. Ind. Szczerbak, G., Nowak, P., Kostrzewa, R.M., et al., 2007. Neurochem.
Med. 38, 324–329. Res. 32, 1791–1798.
Quintanilla-Vega, B., Hoover, D.J., Bal, W., et al., 2000b. Chem. Res. Tabacova, S., Balabaeva, L., 1993. Environ. Health Perspect. 2, 27–31.
Toxicol. 13, 594–600. Takiguchi, M., Achanzar, W.E., Qu, W., et al., 2003. Exp. Cell. Res.
Raqib, R., Ahmed, S., Sultana, R., et al., 2009. Toxicol. Lett. 185, 286, 355–365.
197–202. Taupeau, C., Poupon, J., Nomé, F., et al., 2001. Reprod. Toxicol. 15,
Reichrtova, E., Dorociak, F., Palkovicova, L., 1998. Hum. Exp. 385–391.
Toxicol. 17, 176–181. Telisman, S., Colak, B., Pizent, A., et al., 2007. Environ. Res. 105,
Ríos, R., Zarazúa, S., Santoyo, M.E., et al., 2009. Toxicol. 261, 68–75. 256–266.
Ris, M.D., Dietrich, K.N., Succop, P.A., et al., 2004. J. Int. Neuropsy- Telisman, S., Cvitković, P., Jurasović, J., et al., 2000. Environ. Health
chol. Soc. 10, 261–270. Perspect. 108, 45–53.
Robinson, J.F., Yu, X., Hong, S., et al., 2009. Toxicol. Sci. 107, 206–219. Thévenod, F., 2010. BioMetals 23, 857–875.
Robinson, J.F., Yu, X., Moreira, E.G., et al., 2011. Toxicol. Appl. Phar- Thompson, J., Bannigan, J., 2008. Reprod. Toxicol. 25, 304–315.
macol. 250, 117–129. Tong, M.H., Christenson, L.K., Song, W.C., 2004. Endocrinol. 145,
Rodier, P.M., 2004. Pediatrics 113, 1076–1083. 2487–2497.
Rogers, J.M., 2009. Reprod. Toxicol. 28, 152–160. Urbach, J., Boadi, W., Brandes, J.M., et al., 1992. Reprod. Toxicol. 6,
Ronis, M.J., Badger, T.M., Shema, S.J., et al., 1996. Toxicol. Appl. 69–75. 1992.
Pharmacol. 136, 361–371. Vahter, M., Akesson, A., Lidén, C., et al., 2007a. Environ. Res. 104,
Sakurai, T., Himeno, S., 2006. Environ. Sci. 13, 101–106. 85–95.
Sallmen, M., 2001. Int. J. Occup. Med. Environ. Health 14, 219–222. Vahter, M., Gochfeld, M., Casati, B., et al., 2007b. Environ. Res. 104,
Sallmen, M., Anttila, A., Lindbohm, M.L., et al., 1995. J. Occup. 70–84.
Environ. Med. 37, 931–934. Valinluck, V., Tsai, H.H., Rogstad, D.K., et al., 2004. Nucleic Acids
Sallmen, M., Lindbohm, M.L., Anttila, A., et al., 2000. Epidemiol. 11, Res. 32, 4100–4108.
141–147. Vigeh, M., Yokoyama, K., Kitamura, F., et al., 2010. Women Health
Samuel, J.B., Stanley, J.A., Princess, R.A., et al., 2011. J. Med. Toxicol. 50, 756–766.
7, 195–204. Vrsanska, S., Nagyova, E., Mlynarcikova, A., et al., 2003. Physiol.
Schettler, T.T., 2001. Environ. Health Perspect. 6, 813–816. Res. 52, 383–387.
Schmid, T.E., Grant, P.G., Marchetti, F., et al., 2013. Humanit. Rep. Wang, S.D., Holladay, D.C., Ahmed, S.A., et al., 2006. Int. J. Toxicol.
28, 274–282. 25, 319–331.
20  Effects of Metallic Elements on Reproduction and Development 423

Wang, S.N., Schneider, N., Dragin, K., et al., 2007. Am. J. Physiol. Xi, S., Guo, L., Qi, R., et al., 2010. J. Biochem. Mol. Toxicol. 24,
Cell. Physiol. 292, 1523–1535. 368–378.
Wang, Z., Wang, H., Xu, Z.M., et al., 2012. Toxicol. Appl. Pharmacol. Xie, Y., Liu, J., Benbrahim-Tallaa, L., et al., 2007. Toxicol. 236, 7–15.
259, 236–247. Xu, J., Yan, C.H., Yang, B., et al., 2009. Toxicol. Lett. 191, 223–230.
Waring, R.H., Harris, R.M., 2011. Maturitas 68, 111–115. Xu, W., Bao, H., Liu, F., et al., 2012. Environ Health 11, 46.
Wasserman, G.A., Liu, X., Parvez, F., et al., 2011. Neurotoxicol. 32, Yang, K., Julan, L., Rubio, F., et al., 2006. Am. J. Physiol. Endocrinol.
450–457. Metab. 290, 135–142.
Watson, W.H., Yager, J.D., 2007. Toxicol. Sci. 98, 1–4. Yang, S., Zhang, Z., He, J., et al., 2012. Biol. Trace Elem. Res. 148, 53–60.
Wells, P.G., Bhuller, Y., Chen, et al., 2005. Toxicol. Appl. Pharmacol. Yoshida, M., Suzuki, M., Satoh, M., et al., 2011. J. Toxicol. Sci. 36,
207, S354–S366. 73–80.
Windham, G., Fenster, L., 2008. Fertil. Steril. 89, 111–116. Zemanová, J., Lukác, N., Massányi, P., et al., 2007. J. Vet. Med. A
Wirth, J.J., Mijal, R.S., 2010. Syst. Biol. Reprod. Med. 56, 147–167. Physiol. Pathol. Clin. Med. 54, 281–286.
Wong, C.H., Cheng, C.Y., 2005. Curr. Top. Dev. Biol. 71, 263–296. Zhang, C., Liu, C., Li, D., et al., 2010. J. Cell. Physiol. 222, 444–455.
Wong, W.Y., Flik, G., Groenen, P.M., et al., 2001. Reprod. Toxicol. 15, Zhang, L., Wang, L., Zhang, X., et al., 2012. Int. J. Androl. 35, 752–757.
131–136. Zhang, X., Wang, Y., Zhao, Y., et al., 2008. Biometals 21, 143–150.
Wright, R.O., Baccarelli, A., 2007. J. Nutr. 137, 2809–2813. Zhang, Y., Gao, D., Bolivar, V.J., et al., 2011. Toxicol. Sci. 119, 270–280.
Wu, B., Zhang, D., Wang, D., et al., 2012. Ecotoxicol. 21, 2068–2077. Zhong, C.X., Mass, M.J., 2001. Toxicol. Lett. 122, 223–234.
Wu, C., Wang, L., Liu, C., et al., 2008. Fish Physiol. Biochem. 34, Ziaee, H., Daniel, J., Datta, A.K., et al., 2007. J. Bone Joint. Surg. Br.
43–51. 89, 301–305.
Wulff, M., Hogberg, U., Stenlund, H., 1999. J. Occup. Environ. Med.
41, 678–685.

You might also like