You are on page 1of 6

Environmental Toxicology and Pharmacology 67 (2019) 102–107

Contents lists available at ScienceDirect

Environmental Toxicology and Pharmacology


journal homepage: www.elsevier.com/locate/etap

Cadmium: Toxic effects on placental and embryonic development T



Hui-Xia Geng, Lai Wang
Institute of Chronic Disease Risks Assessment, School of Nursing and Health Sciences, Henan University, Kaifeng, 475004, Henan Province, PR China

A R T I C LE I N FO A B S T R A C T

Keywords: Cadmium is a non-essential trace metal that has strong teratogenic and mutagenic effects in living organisms.
Cadmium The content is more highly enriched in women than in men and can enter the embryo through the placenta and
Pregnancy destroy the placenta's morphological structure, resulting in fetal growth restriction. In this report, we review
Placenta published data linking pregnancy exposure to cadmium to placenta and fetal growth and development toxicity
Embryonic development
and summarize the related mechanisms. An understanding of how cadmium exposure contributes to placental
and fetal development is necessary for the development of prevention and control strategies for fetal develop-
ment defects caused by cadmium exposure during pregnancy.

1. Introduction interfere with the normal process of fetal development, while long-
term, it leads to a variety of systemic diseases in adults. During
Cadmium is a ubiquitous heavy metal with multiple organ toxicity breastfeeding, cadmium can be secreted into the milk, which aggregates
that is constantly released into the environment through human eco- in the offspring’s body, and destroys the learning and memory ability of
nomic activities, causing great harm to human health. Cadmium enters the offspring (Dharmadasa and Kim, 2017; Halder et al., 2016). Thus,
the human body mainly through occupational exposure, diet, exposure to cadmium during pregnancy not only causes damage to
breathing, smoking, or drinking water. In recent years, diet has been women themselves, but also causes placental and fetal development
verified as the main source of human exposure to cadmium pollution. disorders and may cause long-term functional impairment of the off-
An investigation in southern Jiangsu Province, China, showed that spring (Jacobo-Estrada et al., 2017).
approximately 14.89% of cadmium in the soil was transferred into rice
grains, and up to 3.19% could be transferred from rice grains to the 2. Toxicity of cadmium in placental development
human body through rice consumption (Li et al., 2017a). However, the
survey of pregnant women also showed that the blood cadmium con- 2.1. Cadmium placenta enrichment and toxicity
centration was related to diet, and this correlation with the offspring of
serum cadmium concentration was consistent (Moynihan et al., 2017). The placenta develops between the endometrium and the embry-
Cadmium causes damage to multiple organs and tissues, such as the ophoric membrane after female pregnancy. The placenta is the main
kidneys, liver, lungs, bones and brain, and leads to cells carcinogenesis location for maternal and fetal material and energy exchange. The
in these organs. The United States Environmental Protection Agency placenta can synthesize and secrete some hormones, filter out harmful
(USEPA) classifies cadmium as a probable human carcinogen (Group substances in the blood, and provide nutrition and functional support
B1). In recent years, a number of studies have shown that female body for the development of the embryo. Therefore, during pregnancy, when
cadmium enrichment is higher than that in men, and cadmium can the normal placenta formation process is destroyed or placental func-
cross the placenta, affecting the placental development and function, tion-related gene expression levels are changed, placental function may
and into the fetus, gathering in multiple organs and systems, resulting be damaged, which ultimately affects the normal development of the
in fetal body-related gene expression disorder. Its short-term effect is to fetus. Cadmium can pass through the placenta during pregnancy and

Abbreviations: Cd, cadmium; ABC, adenosine triphosphate (ATP)-binding cassette; ABCG2, ATP-binding cassette transporter G2; ABCB4, ATP-binding cassette
transporter B4; LA-ICP-MS, laser ablation inductively coupled plasma mass spectrometry; TGF-β, transforming growth factor-β; PCFT, proton-coupled folate
transporter; PCNA, proliferating cell nuclear antigen; OXPHOS, oxidative phosphorylation; CDkn-1c, cyclin dependent kinase inhibitor-1c; Peg-10, paternally ex-
pressed gene -10; PCDH, protocadherin; NR3C1, glucocorticoid receptor; IL, interleukin; FGR, fetal growth restriction; IUGR, intrauterine growth restriction; DOHaD,
developmental origins of health and disease

Corresponding author.
E-mail address: wanglai@henu.edu.cn (L. Wang).

https://doi.org/10.1016/j.etap.2019.02.006
Received 4 August 2018; Received in revised form 12 February 2019; Accepted 14 February 2019
Available online 15 February 2019
1382-6689/ © 2019 Elsevier B.V. All rights reserved.
H.-X. Geng and L. Wang Environmental Toxicology and Pharmacology 67 (2019) 102–107

accumulate in the placenta, where it may impair placental function and placental development. However, proteomic analysis of placental spe-
affect fetal development (Taylor et al., 2018). During pregnancy, ex- cimens from an e-waste area showed that high cadmium inhibits fu-
posure to cadmium increases its concentration in the placenta, but its marate hydratase expression, thereby affecting oxidative phosphoryla-
detailed mechanism still requires elucidation. tion (OXPHOS) of cells, decreasing mitochondrial function, and
The adenosine triphosphate (ATP)-binding cassette (ABC) trans- affecting energy utilization, which delays placental development. This
porter receptor protein is a member of the superfamily of membrane study suggests that the toxicity of cadmium in placental development
proteins. It widely participates in the transport of various substrates, not only reduces trophoblast cell proliferation, promotes apoptosis, and
heavy metals, and other substances through the cell membrane, which inhibits migration, but also reduces the process of placental cell energy
must hydrolyze ATP to supply energy. ATP-binding cassette transporter production (Xu et al., 2016a).Studies on human chorionic and tropho-
G2 (ABCG2) and B4 (ABCB4), an important metal transporter, mediate blast cell lines, such as HTR-8/SVneo cells, JEG-3 cells, have shown that
the transport of divalent heavy metal ions across cell membranes. The cadmium causes a decrease in the activity, proliferation and migration
expression levels of ABCG2 and ABCG4 were found to be significantly of these cells. These results are consistent the toxicological effects of
lower in the placenta in Sprague-Dawley rats during pregnancy ex- cell apoptosis and placental structure destruction in mice exposure to
posure to cadmium, indicating that the expression of membrane pro- cadmium during pregnancy. However, if the selected cell lines derive
teins involved in heavy metal transport in the placenta after cadmium from the animal species in the in vivo experiment, these results will be
exposure was reduced, thereby decreasing their transport function and mutually verified, complement each other, and be more scientific,
increasing the intracellular accumulation of cadmium in the placenta of strictness and accuracy (Table 1).
cadmium-treated rats (Liu et al., 2016).The research of placenta his-
topathology of the rats with a single cadmium exposure on gestation 2.2. The mechanisms of cadmium placental developmental toxicity
day 18 showed that the trophoblasts in the labyrinth zone were swelling
at 1 h and had vacuolar degeneration at 2 and 3 h. After 6 and 24 h, Epigenetics are broadly described as heritable alterations in gene
syncytiotrophoblast-selective necrosis or apoptosis resulted in a drastic function, which do not involve variations in DNA sequence, genomic
decrease in cell number. In the trophoblastic septa, the expression of imprinting, and DNA methylation as the major form of epigenetic
matrix metalloproteinase was significantly increased, particularly in modification (Bitto et al., 2014). In recent years, the effects of cadmium
cytotrophoblasts at 24 h after exposure. The localization of cadmium in exposure on an organism’s epigenetic signatures have become one of its
the placenta was detected by the laser ablation inductively coupled toxicological mechanisms. The epigenetic mechanism of cadmium on
plasma mass spectrometry (LA-ICP-MS), which was prominent in the placental development is mainly DNA methylation changes (Vilahur
labyrinth zone and tended to increase with the progression of tropho- et al., 2015). However, cadmium exposure in early life alters DNA
blastic septa damages (Yamagishi et al., 2016). In the case of the same methylation differently in girls and boys. The methylation changes of
cadmium salt (CdCl2), although there were differences in animal genes are associated with organ development, morphology, and mi-
models, routes and length of cadmium exposure, it was observed that neralization of bone in girls, whereas changes in boys were found in cell
cadmium exposure resulted in decreased expression of metal trans- death-related genes (Kippler et al., 2013).
porter, cadmium ion accumulation, apoptosis and placental structure Studies of 13 imprinted genes associated with placenta and em-
destruction in the placenta of cadmium exposure during pregnancy in bryonic development in C57BL/6 mice indicate that the maternally
mice (Table 1). expressed gene, cyclin-dependent kinase inhibitor-1c (CDkn-1c), and
Cadmium reduces the expression of related functional genes in the paternally expressed gene, paternally expressed gene-10 (Peg-10), were
placenta, affecting the morphology and function of cells in different significantly upregulated and downregulated, respectively, in the Cd-
functional regions of the placenta, such as reducing the proliferation exposed placentas when compared to the normal placentas. The results
and migration of trophoblast cells, thereby affecting the normal pla- of bisulfate polymerase chain reaction (PCR) revealed that the methy-
cental formation and development. S100 P, a member of the calcium- lation patterns of the promoter regions of Cdkn1c and Peg10 were
binding protein family, is involved in the regulation of cell prolifera- changed, which led to the disturbance of gene expression in the Cd-
tion. Studies on human trophoblast-derived cell lines show that ex- exposed placentas (Xu et al., 2017). Protocadherin (PCDH), a single-
posure to cadmium inhibits the expression of S100 P and thus reduces stranded transmembrane glycoprotein, is a member of the cadherin
the proliferation of placental trophoblast cells (Zhou et al., 2016). miR- superfamily that is involved in the biological processes of cell-cell re-
26a is an important regulator of trophoblast cell migration. Cadmium cognition, adhesion, migration, communication, and tissue differ-
reduces the expression of miR-26a in trophoblast cells and inhibits the entiation. PCDH genes are primarily known for their functions in early
expression of the transforming growth factor-β (TGF-β) signal trans- embryonic and central nervous system development, and cause devel-
duction process, which reduces the rate of cell migration, resulting in opmental delay and other cerebral abnormalities (Brown et al., 2013;
the delaying or slowing of placenta formation and reducing the dia- Anitha et al., 2013). The expression of protocadherin alpha subfamily-
meter and weight of the placenta. Then, both mRNA and protein levels C1 (PCDHA-C1) is abundant in the placenta and in early embryogen-
of proton-coupled folate transporter (PCFT) in the placenta are down- esis, which promotes the growth and development of the placenta and
regulated, which limits the normal development of the fetus, leading to embryo. An increasing number of studies have shown that the expres-
increased neural tube defects (Brooks and Fry, 2017; Zhang et al., sion level of PCDHA-C1 in the placenta was inversely correlated with
2016). cadmium concentrations in the mother's toenail. The mechanism was
In addition to inhibiting the proliferation and migration of tropho- strongest amongst those with low levels of DNA methylation in the
blast cells, cadmium in the rat placenta can also cause decreased pro- promoter region of placental PCDHAC1, downregulating gene expres-
liferating cell nuclear antigen (PCNA) expression in placental macro- sion and affecting placenta and embryo development and growth
phages, spongiotrophoblast cells, trophoblast giant cells, and labyrinth (Everson et al., 2016).
trophoblast cells, thereby promoting apoptosis, significantly decreasing The placenta is involved in the development of the fetal hypotha-
the size of the placental layer, reducing the quality of the placenta, and lamic-pituitary-adrenal axis (HPA), regulates the expression of gluco-
delaying placental development as compared with the control group corticoid receptor (NR3C1) and downstream target genes, and controls
(Erboga and Kanter, 2016). Thus, the cadmium ions in the placenta can the endogenous stability of cortisol levels in the fetal body or easily
reduce trophoblast cell proliferation, promote its apoptosis, inhibit its leads to abnormal fetal development and the emergence of long-term
migration, and ultimately delay the placental development process. cognitive and neurobehavioral problems in children. Cohort studies
Then, the formation of placental size and quality is lower than the have shown that maternal cadmium exposure during pregnancy leads
normal developmental process, which is the toxicity of cadmium in to increased methylation of the NR3C1 promoter in the placenta,

103
H.-X. Geng and L. Wang

Table 1
Summary table of the difference of cadmium exposure.
Model Cell lines Type of cadmium salt Routes of exposure Length of Effects of exposure References
exposure

Placenta Sprague-Dawley rat Unclear CdCl2 Gavage 20 Days Down-regulated ABCG2 and ABCB4 transporters (Liu et al., 2016)
Wistar Hannover rat Unclear CdCl2 Subcutaneous injection 18 Days Histopathological changes (Yamagishi et al., 2016)
HTR-8/SVneo CdCl2 Medium 24 h Decreased S100 P expression and cell proliferation (Zhou et al., 2016)
cells
JEG-3 cells CdCl2 Medium 48 h Inhibited trophoblast cell migration (Brooks and Fry, 2017)
ICR mice Unclear CdCl2 Intraperitoneal injection 10 Days Downregulated placental folate transporters (Zhang et al., 2016)
Wistar rat Unclear CdCl2 Subcutaneous injection 21 Days Inhibited trophoblast cell proliferation (Erboga and Kanter, 2016)
Maternal placenta Unclear E-waste exposure Unclear Unclear Downregulated fumarate hydratase (Xu et al., 2016a)
C57BL/6 mice Unclear CdCl2 Drinking water 6,9 and 12 Days Dysregulation of DNAM and expression of imprinted genes (Xu et al., 2017)
Maternal and newborn Unclear Unclear Unclear Unclear Downregulated DNAM and expression of PCDHAC1 (Everson et al., 2016)
toenails
Maternal and newborn Unclear Unclear Unclear Unclear Increased DNAM of glucocorticoid receptor (Appleton et al., 2017)
toenails
CD-1 mice JEG-3 cells CdCl2, Intraperitoneal injection 8 and 24 h Induced inflammatory cytokines expression (Hu et al., 2018)
Maternal placenta Unclear Unclear Unclear Unclear Increased DNAM of inflammatory signaling genes (Everson et al., 2018)
Embryo Zebrafish embryo Unclear CdCl2 medium 9h Downregulated XPC and p-regulated UV-DDB2 gene (Ling et al., 2017)

104
expression
Zebrafish embryo Unclear CdCl2 Medium 4h Downregulated expression of both RBBP6 and CRYL1 genes (Scudiero et al., 2017)
Sprague-Dawley rat Unclear CdCl2 Intraperitoneal injection 8 Days Blocked fetal Leydig cell development (Li et al., 2017b)
Neonate Unclear E-waste recycling Unclear Reduced female neonate development (Zhang et al., 2017)
area
B6D2 Sperm and embryo Unclear Cadmium acetate Medium < 84 h Reduced sperm motility, fertilization rate and blastocyst (Zhao et al., 2017)
formation rate in vitro
Maternal toenails Unclear Unclear Unclear Unclear Resulted in intrauterine growth restriction (Everson et al., 2017)
Maternal serum Unclear Unclear Unclear Unclear Resulted in SGA infants (Wang et al., 2016a)
Maternal urine Unclear Unclear Unclear Unclear Delayed fetal growth (Cheng et al., 2017)
Maternal serum Unclear Unclear Unclear Unclear Reduced female neonatal development (Taylor and Golding, 2016)
Maternal urine Unclear Unclear Unclear Unclear Reduced female neonatal development (Romano et al., 2016)
57BL/6 mice Unclear CdCl2 Drinking water 6, 9, and 12 Days Upregulated the DNMT3B/3 L expressions, while (Xu et al., 2016b)
downregulating GLUT3
Wistar rat Unclear CdCl2 Drinking water 20 Days Disrupted zinc transplacental handover (Mikolić et al., 2015)
CD-1 mice Unclear CdCl2 Intraperitoneal injection, Drinking 9 and 18 Days Downregulated Znt1 and Znt2 expressions (Wang et al., 2016b)
water

CdCl2: Cadmium chloride; S100P: Ca2+-binding S100 protein family; DNAM: DNA methylation; XPC: xeroderma pigmentosum C; UV-DDB2: UV-damaged DNA-binding protein 2; RBBP6: retinoblastoma binding protein
6; CRYL1 :crystallin-lambda 1; B6D2:C57BL/6 × DBA/2; SGA: small for gestational age; DNMT: DNA methyltransferase; GLUT3: glucose transporters 3; Znt: zinc transporters.
Environmental Toxicology and Pharmacology 67 (2019) 102–107
H.-X. Geng and L. Wang Environmental Toxicology and Pharmacology 67 (2019) 102–107

decreased NR3C1 expression and function, and fetal development at excision repair was downregulated. This study showed that cadmium
abnormal cortisol concentrations. Taken together, these results indicate exposure easily leads to increased intra-embryonic oxidative status, and
that prenatal exposure to cadmium may affect the offspring's NR3C1 at lower concentrations, showed increased DNA repair-related gene
activity, which may help explain cognitive and neurodevelopmental expression, indicating that at low concentrations, it can cause em-
abnormalities and other mechanisms of long-term toxicological effects bryonic genome damage and increase the DNA repair process.
in life (Appleton et al., 2017). However, at high concentrations, it results in decreased expression le-
These are various inflammatory gene expression throughout mid-to- vels of nucleotide excision repair genes, which affects the DNA repair
late gestation in rat placenta and warrant further the placenta and fetus process, resulting in embryonic development abnormalities (Ling et al.,
development (Vaswani et al., 2018).Nevertheless, pro-inflammatory 2017).
cytokine expression disorders may be associated with placental dys- Retinoblastoma binding protein 6 (RBBP6) is associated with in-
function and impaired fetal growth, which lead to preeclampsia and creased protein degradation and cell proliferation, and crystallin-
fetal growth restriction (Cotechini et al., 2014; Liang et al., 2018 Jun; lambda 1 (CRYL1) is a lens protein with redox activity. Some studies
Zenerino et al., 2017). A growing body of evidence suggests that cad- have also shown that exposure to cadmium upregulates the expression
mium has pro-inflammatory activities. Several inflammatory cytokines, of both RBBP6 and CRYL1 genes in a dose-dependent manner at the
tumor necrosis factor-α (TNF-α), interleukin (IL)-8, and IL-6, were gastrula stage, the early phases of zebrafish development. Eventually,
upregulated in the placentas of mice with CdCl2 (3.0 mg/kg) on ge- the retinal morphology in adult zebrafish retinas are altered due to
stational day 15 (GD15) (intraperitoneally injected) compared with the exposure to cadmium during pregnancy (Scudiero et al., 2017). Em-
control mice. Additional experiments showed that gestational Cd ex- bryonic interstitial Leydig cells are the main source of production and
posure activated Akt signaling in the mouse placenta, and LY294002, a secretion of male hormones, which are the critical factors for testis
specific inhibitor of PI3K, blocked CdCl2-evoked Akt phosphorylation descent and the development of the male reproductive tract. Adult 64-
and inhibited CdCl2-induced inflammatory cytokine expression (Hu day-old Sprague-Dawley rats received a single intraperitoneal injection
et al., 2018). An epigenome-wide association study (EWAS) between of 0, 0.25, 0.5, and 1.0 mg/kg cadmium on gestational day 12. On ge-
cadmium concentrations and DNA methylation (DNAM) from human stational day 20 (GD), the results from this study indicated that cad-
placentae showed that higher cadmium was associated with increased mium dose-dependently reduced testosterone production of fetal testis
DNAM levels in inflammatory signaling genes (TNFAIP2, ACOT7, and and downregulated protein expression levels of Leydig (LHCGR,
RORA), which disturbed inflammatory processes, and impaired pla- SCARB1, STAR, CYP11A1, HSD3B1, and CYP17A1) and Sertoli cells
cental function and development (Everson et al., 2018). (HSD17B3, DHH, and FSHR). Thereby, maternal exposure to cadmium
Cadmium exposure during pregnancy leads to changes in the me- during the pregnancy reduced the formation of testosterone in fetal
thylation pattern of imprinted genes, protocadherin, glucocorticoid rats, lowered fetal Leydig cell numbers, and interfered with normal
receptors and genes governing placental development, which affects the male fetal development (Li et al., 2017b).
development of the placenta and exerts a toxicological effect
(Table 1).Although these studies have different cadmium exposure 3.2. Cadmium causes fetal growth restriction
animal models, types of cadmium salts, routes and length of cadmium
exposure, the changes in the methylation patterns of related genes have In addition to disrupting the expression of certain genes in the fetus,
all been observed, especially in the study of human placenta, which cadmium that enters the embryo through the placenta affects normal
provide a scientific basis for prevention and treatment of placental or fetal development, resulting in intrauterine growth restriction (IUGR)
fetal developmental defects due to cadmium exposure during preg- or fetal growth restriction (FGR) to lower fetal development below
nancy. However, there are many types of genes involved in placental normal months of age and reduce birth anthropometry, such as birth
development. If it can prove that the changes of methylation profile of weight, birth height, head circumference, and Apgar scores in the
placental development genes caused by cadmium exposure during newborn (Zhang et al., 2017). Mouse fertilized egg in vitro experiments
pregnancy, it can reveal the extensiveness and consistency of cadmium confirmed that cadmium exposure inhibits early embryonic develop-
placental developmental toxicity. ment and dramatically decreased the blastocyst formation rate, in-
dicating that cadmium exposure during pregnancy leads to abnormal-
3. Embryonic developmental toxicity of cadmium ities in embryonic development, but also shows that cadmium exposure
during pregnancy is more hazardous to fetal development than to the
3.1. Cadmium causes abnormal expression of related genes in embryos mother (Zhao et al., 2017).
A population-based survey of the associations between maternal
In addition to affecting the formation and development of the pla- toenail cadmium and fetal development showed that Cd concentrations
centa during pregnancy, cadmium exposure during pregnancy can also in maternal toenails increase intrauterine growth restriction (IUGR) and
be enriched in the embryo, reducing the expression of related genes in small for gestational age (SGA) births by 1.95 and 1.46 times, respec-
the fetus, leading to abnormal embryonic development and congenital tively, which demonstrates that maternal exposure to Cd during preg-
disorders of the structure and function of some tissues and organs in the nancy reduces embryo development and results in fetal growth re-
offspring. Zebrafish are the best model organism for studying the en- striction or limited functional development (Everson et al., 2017). A
vironmental toxicity in vertebrates due to its advantages of small size, prospective cohort study based on a population of 3254 pregnancies
convenient breeding, strong fertility, transparent embryonic body, and also showed similar results, such as maternal serum Cd level in preg-
nearly 90% homology with human genes. The expression profile of nancy, with an approximately 10.6% risk of small for gestational age
nucleotide excision repair genes (NER) was altered when the zebrafish (SGA) infants in the high cadmium group (≥1.06 μg/L), significantly
embryos were exposed to cadmium. Exposure of embryos at 1 h post- higher than 7.5% among subjects in the low cadmium group
fertilization to 3–5 μM of cadmium induced a 2- to 3-fold increase of (< 1.06 μg/L). However, the study also pointed out that exposure to
oxidative stress. The expression of xeroderma pigmentosum C (XPC) cadmium during pregnancy leads to fetal growth restriction. In addi-
was downregulated, while the expression of DNA-damage-binding tion, the risk of SGA may be related to the time point of cadmium ex-
protein 2 (UV-DDB2) was upregulated by real-time reverse transcrip- posure. In the middle gestational stage, SGA elevates the risk in contrast
tion (RT)-PCR in mildly-stressed embryos. With the increase of cad- to the early gestational stage (Wang et al., 2016a). However, a cohort
mium concentration, oxidative stress in embryos was further increased study of maternal urinary Cd levels showed that increases in cadmium
with the increase of 8-oxoguanine DNA glycosylase (OGG1) gene ex- concentrations in maternal blood in the first trimester of pregnancy are
pression. However, the expression of genes related to nucleotide related to fetal growth restriction, suggesting that Cd exposure time

105
H.-X. Geng and L. Wang Environmental Toxicology and Pharmacology 67 (2019) 102–107

during pregnancy is a crucial factor for fetal growth restriction. How- placental metal ions and Zn2+ in Wistar rats and reduces the main-
ever, this correlation is variable due to differences in the number of tenance of fetal nutrition and viability (Mikolić et al., 2015). Studies on
people surveyed. It also shows that the mechanism of fetal growth re- cadmium-caused placenta and fetal metal ion transport disorder have
striction caused by exposure to cadmium during pregnancy would shown that maternal exposure to cadmium during pregnancy increased
change with the development of the embryo (Cheng et al., 2017). In a placental zinc ion concentrations and reduced the concentration within
large prospective pregnancy cohort, prenatal exposure to cadmium re- the embryo. Moreover, placental Znt1 and Znt2, two zinc transporters,
duced neonatal anthropometry (birthweight, birth length, head cir- were downregulated in cadmium -exposed mice. These results suggest
cumference, and ponderal index) had a sex-specific effect, in which that maternal Cd exposure during pregnancy reduces Zn transport
embryotoxicity was associated with girls but not boys. These results across the placenta, resulting in the enrichment of zinc ions in the
indicate that the effects of prenatal cadmium exposure on fetal growth placenta and the reduction in the fetus body, inducing fetal growth
restriction are gender-specific, and also reflect the complexity and di- restriction due to the lack of zinc ions (Wang et al., 2016b).
versity of embryonic developmental toxicity of cadmium (Taylor and In summary, the mechanism for cadmium-induced embryo growth
Golding, 2016; Romano et al., 2016). restriction may be related to the reduction of nutrient supply. These
It can be seen from the above discussion that cadmium exposure studies in rats and mice indicate that decreased expression of glucose
during pregnancy causes the expression levels of multiple genes in transporter and Znt1/2 may be one of its mechanisms. However, the
zebrafish and mouse embryos to be disordered, leading to abnormal animal models used in these studies are all rodents, and the mechanism
embryonic development, especially fetal growth restriction. However, by which cadmium-induced embryo growth restriction in primate em-
population-based investigations show that cadmium-induced fetal bryos is consistent with these results and further research is needed.
growth restriction during pregnancy exposure is connected with ex- Secondly, in the process of cadmium-induced embryo growth restric-
posure dose, time point of cadmium exposure and fetal sex. The dif- tion, the alteration of the methylation patterns of genes in placenta or
ference of these studies between different species indicate differences in embryo is also one of its mechanisms (Table 1).
embryonic developmental toxicity after the exposure to Cd during
pregnancy (Table 1). Therefore, a large number of studies on animal
4. Conclusions
models and population-based are needed to refine cadmium dose, time
points, length of exposure and other factors to further clarify the dif-
The developmental origins of health and disease (DOHaD) empha-
ferences and complexities of embryonic developmental toxicity caused
size the impact of the uterine environment on childhood and lifelong
by cadmium exposure during pregnancy.
health, suggesting that destruction of embryonic development in early
life and the disturbance of endogenous biological function may lead to
3.3. The mechanism of embryonic developmental toxicity of cadmium
premature death of the fetus and a variety of systemic diseases in
adulthood, such as hypertension, obesity, and diabetes. The low doses
3.3.1. Reduced nutrient supply
of environmental pollutant exposure in early embryo development is a
The detailed mechanism by which cadmium causes fetal growth
causative factor of DOHaD outcomes, which have different types of
restriction remains to be elucidated, but cadmium exposure leads to a
epigenetic change in the genome (Hoffman et al., 2017). In this paper,
decrease in the expression of carrier proteins associated with nutrient
we review the effects of cadmium exposure on placental and embryonic
transport in the placenta. Reducing the nutritional supply of embryos
development during pregnancy. Although there are differences in an-
may be involved in this process. In the early stage of embryonic de-
imal models, cell lines, routes and length of exposure, type of cadmium
velopment, there is a large amount of cell proliferation and growth. The
salt and so on in each study, all studies have shown cadmium exposure
process requires the consumption of large amounts of glucose and other
during pregnancy. It causes abnormal development in placentas and
energy substances, and the lack of energy molecules may interfere with
embryos, indicating that the placental and embryonic toxicity of cad-
cell division and growth processes, delaying embryonic development.
mium coexist between different species, and the toxicological me-
In the placenta, a large number of nutrient transporters, such as
chanisms are similar, such as abnormal changes in gene methylation
glucose transporters (GLUTs), can provide a variety of nutrients needed
(Table 1). In summary, exposure to cadmium during pregnancy affects
during fetal development. Some studies have shown that
not only placental formation and function, but also changes in the ex-
the expression of DNA methyltransferase (DNMT) 3B was sig-
pression of many genes in the embryo, limiting fetal growth and de-
nificantly upregulated, and GLUT3 was significantly downregulated in
velopment and long-term harmful effects on the function of certain
cadmium-exposed mice when compared to the normal placentas. Data
organs and tissues in offspring, and these processes are associated with
from bisulfite PCR demonstrated the hypermethylation of the promoter
cadmium exposure, leading to aberrant methylation of genes within the
region of GLUT3. Therefore, maternal exposure to Cd during pregnancy
placenta and embryo. Changes in the epigenetic modification patterns
downregulates the expression of GLUT3, reduces placenta glucose
induced by cadmium, such as aberrant methylation, have been linked to
transport, decreases embryonic nutrient and energy supply, affects the
their ability to easily bind to thiols. Cadmium-thiol interactions may
growth and development of the fetus, and leads to fetal growth re-
lead to depletion of the methyl donor S-adenosyl-methionine, resulting
striction (Xu et al., 2016b). Further studies have shown that cadmium-
in methylome alterations, and subsequently altered DNA methyl-
induced fetal growth restriction is related to placental levels of cad-
transferase activity, altering the critical developmental process in early
mium. At low doses, cadmium has no significant association with
embryonic stages, causing blockages in fetal development, or changes
neonatal indices, such as birth weight, head circumference, and body
in certain functions of the body (Ruiter et al., 2016). Reduced exposure
length, while at high doses, cadmium causes fetal growth restriction
to cadmium during pregnancy inhibited the abnormal changes of me-
(Guo et al., 2016).
thylation patterns in the promoter region of the genes in placentas or
embryos. This has important theoretical and practical significance in
3.3.2. Zinc ion (Zn2+) disorder
reducing placental and embryonic developmental toxicity of cadmium,
Zn2+ is a trace metal element necessary for organisms and plays a
maintaining the normal intrauterine growth environment of the fetus,
key role in many proteins involved in enzymatic reactions, signal
and elucidating the developmental origins of health and disease.
transduction, and DNA repair in vivo, especially in the early stages of
embryo development. Appropriate Zn2+ concentrations determine the
differentiation of nerves, the formation of neuroblasts, and the growth Declarations of interest
and development of embryos (Chowanadisai et al., 2013). Oral cad-
mium exposure during pregnancy leads to disturbed translocation of None.

106
H.-X. Geng and L. Wang Environmental Toxicology and Pharmacology 67 (2019) 102–107

Transparency document Epigenetics 8 (May (5)), 494–503.


Li, T., Chang, Q., Yuan, X., et al., 2017a. Cadmium transfer from contaminated soils to the
human body through rice consumption in southern Jiangsu Province, China. Environ.
The Transparency document associated with this article can be Sci. Process Impacts 19 (6), 843–850.
found in the online version. Li, X., Liu, J., Wu, S., et al., 2017b. In utero single low-dose exposure of cadmium induces
rat fetal Leydig cell dysfunction. Chemosphere 194, 57–66.
Liang, T., Jinglong, X., Shusheng, D., Aiyou, W., 2018 Jun. Maternal obesity stimulates
Acknowledgments lipotoxicity and up-regulates inflammatory signaling pathways in the full-term swine
placenta. Anim. Sci. J.(June 26). https://doi.org/10.1111/asj.13064.
This work was supported by grants from the Science and Ling, L.B., Chang, Y., Liu, C.W., et al., 2017. Oxidative stress intensity-related effects of
cadmium (Cd) and paraquat (PQ) on UV-damaged-DNA binding and excision repair
Technology Projects of Henan province (172102310001), Henan activities in zebrafish (Danio rerio) embryos. Chemosphere 167, 10–18.
Province Foundation for University Key Teacher (16A330001) Liu, L., Zhou, L., Hu, S., et al., 2016. Down-regulation of ABCG2 and ABCB4 transporters
in the placenta of rats exposed to cadmium. Oncotarget 7 (25), 38154–38163.
Mikolić, A., Piasek, M., Sulimanec Grgec, A., et al., 2015. Oral cadmium exposure during
References
rat pregnancy: assessment of transplacental micronutrient transport and ster-
oidogenesis at term. J. Appl. Toxicol. 35 (5), 508–519.
Anitha, A., Thanseem, I., Nakamura, K., et al., 2013. Protocadherin α (PCDHA) as a novel Moynihan, M., Peterson, K.E., Cantoral, A., et al., 2017. Dietary predictors of urinary
susceptibility gene for autism. J. Psychiatry Neurosci. 38 (3), 192–198. cadmium among pregnant women and children. Sci. Total Environ. 575, 1255–1262.
Appleton, A.A., Jackson, B.P., Karagas, M., et al., 2017. Prenatal exposure to neurotoxic Romano, M.E., Enquobahrie, D.A., Simpson, C., et al., 2016. Maternal body burden of
metals is associated with increased placental glucocorticoid receptor DNA methyla- cadmium and offspring size at birth. Environ. Res. 147, 461–468.
tion. Epigenetics 12 (8), 607–615. Ruiter, S., Sippel, J., Bouwmeester, M.C., et al., 2016. Programmed effects in neurobe-
Bitto, A., Pizzino, G., Irrera, N., Galfo, F., Squadrito, F., 2014. Epigenetic modifications havior and antioxidative physiology in zebrafish embryonically exposed to cadmium:
due to heavy metals exposure in children living in polluted areas. Curr. Genomics 15 observations and hypothesized adverse outcome pathway framework. Int. J. Mol. Sci.
(December (6)), 464–468. 17 (11) pii: E1830.
Brooks, S.A., Fry, R.C., 2017. Cadmium inhibits placental trophoblast cell migration via Scudiero, R., Esposito, M.G., Simoniello, P., et al., 2017. Retinoblastoma binding protein
miRNA regulation of the transforming growth factor beta (TGF-β) pathway. Food 6 and crystallin lambda 1 are cadmium-responsive genes in zebrafish embryos and
Chem. Toxicol. 109 (Pt 1), 721–726. adults retinae. C. R. Biol. 340 (4), 197–203.
Brown, N., Burgess, T., Forbes, R., et al., 2013. 5q31.3 Microdeletion syndrome: clinical Taylor, C.M., Golding, J., 2016. EmondAM.Moderate Prenatal Cadmium Exposure and
and molecular characterization of two further cases. Am. J. Med. Genet. A 161A (10), Adverse Birth Outcomes: a Role for Sex-Specific Differences? Paediatr. Perinat.
2604–2608. Epidemiol. 30 (6), 603–611.
Cheng, L., Zhang, B., Zheng, T., et al., 2017. Critical windows of prenatal exposure to Taylor, C.M., Emond, A.M., Lingam, R., Golding, J.P., 2018. Renatal lead, cadmium and
cadmium and size at birth. Int. J. Environ. Res. Public Health 14 (1) pii: E58. mercury exposure and associations with motor skills at age 7 years in a UK ob-
Chowanadisai, W., Graham, D.M., Keen, C.L., et al., 2013. Neurulation and neurite ex- servational birth cohort. Environ. Int. 117, 40–47.
tension require the zinc transporter ZIP12 (slc39a12). Proc. Natl. Acad. Sci. U. S. A. Vaswani, K., Dekker Nitert, M., Chan, H.W., Almughlliq F, B., 2018. Mid-to-Late gesta-
110 (24), 9903–9908. tional changes in inflammatory gene expression in the rat placenta. Reprod. Sci. 25
Cotechini, T., Komisarenko, M., Sperou, A., Macdonald-Goodfellow, S., Adams, M.A., (February (2)), 222–229.
Graham, C.H., 2014. Inflammation in rat pregnancy inhibits spiral artery remodeling Vilahur, N., Vahter, M., Broberg, K., 2015. The epigenetic effects of prenatal cadmium
leading to fetal growth restriction and features of preeclampsia. J. Exp. Med. 211 exposure. Curr. Environ. Health Rep. 2 (2), 195–203.
(January (1)), 165–179. Wang, H., Liu, L., Hu, Y.F., et al., 2016a. Maternal serum cadmium level during preg-
Dharmadasa, P., Kim, N., Thunders, M., 2017. Maternal cadmium exposure and impact on nancy and its association with small for gestational age infants: a population-based
foetal gene expression through methylation changes. Food Chem. Toxicol. 109 (Pt 1), birth cohort study. Sci. Rep. 6, 22631.
714–720. Wang, H., Wang, Y., Bo, Q.L., et al., 2016b. Maternal cadmium exposure reduces pla-
Erboga, M., Kanter, M., 2016. Effect of cadmium on trophoblast cell proliferation and cental zinc transport and induces fetal growth restriction in mice. Reprod. Toxicol.
apoptosis in different gestation periods of rat placenta. Biol. Trace Elem. Res. 169 (2), 63, 174–182.
285–293. Xu, L., Ge, J., Huo, X., et al., 2016a. Differential proteomic expression of human placenta
Everson, T.M., Armstrong, D.A., Jackson, B.P., et al., 2016. Maternal cadmium, placental and fetal development following e-waste lead and cadmium exposure in utero. Sci.
PCDHAC1, and fetal development. Reprod. Toxicol. 65, 263–271. Total Environ. 550, 1163–1170.
Everson, T.M., Kappil, M., Hao, K., et al., 2017. Maternal exposure to selenium and Xu, P., Wu, Z., Xi, Y., et al., 2016b. Epigenetic regulation of placental glucose transporters
cadmium, fetal growth, and placental expression of steroidogenic and apoptotic mediates maternal cadmium-induced fetal growth restriction. Toxicology 372,
genes. Environ. Res. 158, 233–244. 34–41.
Everson, T.M., Punshon, T., Jackson, B.P., Hao, K., Lambertini, L., 2018. Cadmium-as- Xu, P., Wu, Z., Yang, W., et al., 2017. Dysregulation of DNA methylation and expression
sociated differential methylation throughout the placental genome: epigenome-wide of imprinted genes in mouse placentas of fetal growth restriction induced by maternal
association study of two U.S. Birth cohorts. Environ. Health Perspect. 126 (January cadmium exposure. Toxicology 390, 109–116.
(1)), 017010. Yamagishi, Y., Furukawa, S., Tanaka, A., et al., 2016. Histopathological localization of
Guo, J., Wu, C., Qi, X., et al., 2016. Adverse associations between maternal and neonatal cadmium in rat placenta by LA-ICP-MS analysis. J. Toxicol. Pathol. 29 (4), 279–283.
cadmium exposure and birth outcomes. Sci. Total Environ. 16, 31934–31939. Zenerino, C., Nuzzo, A.M., Giuffrida, D., Biolcati, M., Zicari, A., Todros, T., Rolfo, A.,
Halder, S., Kar, R., Galav, V., et al., 2016. Cadmium exposure during lactation causes 2017. The HMGB1/RAGE pro-inflammatory Axis in the human placenta: modulating
learning and memory-impairment in F1 generation mice: amelioration by quercetin. effect of low molecular weight heparin. Molecules 22 (11) pii: E1997.
Drug Chem. Toxicol. 39 (3), 272–278. Zhang, G.B., Wang, H., Hu, J., et al., 2016. Cadmium-induced neural tube defects and
Hoffman, D.J., Reynolds, R.M., Hardy, D.B., 2017. Developmental origins of health and fetal growth restriction: association with disturbance of placental folate transport.
disease: current knowledge and potential mechanisms. Nutr. Rev. 75 (12), 951–970. Toxicol. Appl. Pharmacol. 306, 79–85.
Hu, J., Wang, H., Hu, Y.F., Xu, X.F., Chen, Y.H., Xia, M.Z., Zhang, C., Xu, D.X., 2018. Zhang, Y., Xu, X., Chen, A., et al., 2017. Maternal urinary cadmium levels during preg-
Cadmium induces inflammatory cytokines through activating Akt signaling in mouse nancy associated with risk of sex-dependent birth outcomes from an e-waste pollution
placenta and human trophoblast cells. Placenta 65, 7–14. site in China. Reprod. Toxicol. 75, 49–55.
Jacobo-Estrada, T., Santoyo-Sánchez, M., Thévenod, F., Barbier, O.C., 2017. Admium Zhao, L.L., Ru, Y.F., Liu, M., et al., 2017. Reproductive effects of cadmium on sperm
handling, toxicity and molecular targets involved during pregnancy: lessons from function and early embryonic development in vitro. PLoS One 12 (11), e0186727.
experimental models. Int. J. Mol. Sci. 18 (July (7)) pii: E1590. Zhou, T., Wang, H., Zhang, S., et al., 2016. S100P is a potential molecular target of
Kippler, M., Engström, K., Mlakar, S.J., Bottai, M., Ahmed, S., 2013. Sex-specific effects of cadmium-induced inhibition of human placental trophoblast cell proliferation. Exp.
early life cadmium exposure on DNA methylation and implications for birth weight. Toxicol. Pathol. 68 (10), 565–570.

107

You might also like