Anda di halaman 1dari 8

Environment International 106 (2017) 127–134

Contents lists available at ScienceDirect

Environment International
journal homepage: www.elsevier.com/locate/envint

Associations between toxic and essential trace elements in maternal blood MARK
and fetal congenital heart defects
Yanqiu Oua,1, Michael S. Bloomb,1, Zhiqiang Niea,1, Fengzhen Hanc, Jinzhuang Maia, Jimei Chend,
Shao Linb, Xiaoqing Liua,⁎,2, Jian Zhuangd,⁎,2
a
Department of Epidemiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong General
Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
b
Departments of Environmental Health Sciences and Epidemiology and Biostatistics, University at Albany, State University of New York, Rensselaer, NY, USA
c
Department of Obstetrics, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
d
Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong General
Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China

A R T I C L E I N F O A B S T R A C T

Keywords: Prenatal exposure to toxic trace elements, including heavy metals, is an important public health concern. Few
Heart anomalies studies have assessed if individual and multiple trace elements simultaneously affect cardiac development. The
Teratogen current study evaluated the association between maternal blood lead (Pb), cadmium (Cd), chromium (Cr),
Trace elements copper (Cu), mercury (Hg), and selenium (Se) levels and congenital heart defects (CHDs) in offspring. This
Risk factor
hospital-based case-control study included 112 case and 107 control infants. Maternal peripheral blood draw
Environment
was made during gestational weeks 17–40 and used to determine trace element levels by inductively coupled
plasma mass spectrometry. Multivariable logistic regression was used to assess associations and interactions
between individual and multiple trace elements and fetal CHDs, adjusted for maternal age, parity, education,
newborn gender, migrant, folic acid or multivitamin intake, cigarette smoking, maternal prepregnancy body
mass index, and time of sample collection. Control participants had medians of 2.61 μg/dL Pb, 1.76 μg/L Cd,
3.57 μg/L Cr, 896.56 μg/L Cu, 4.17 μg/L Hg, and 186.47 μg/L Se in blood. In a model including all measured
trace elements and adjusted for confounders, high levels of maternal Pb (OR = 12.09, 95% CI: 2.81, 51.97) and
Se (OR = 0.25, 95% CI: 0.08, 0.77) were harmful and protective predictors of CHDs, respectively, with positive
and negative interactions suggested for Cd with Pb and Se with Pb, respectively. Similar associations were
detected for subgroups of CHDs, including conotruncal defects, septal defects, and right ventricle outflow tract
obstruction. Our results suggest that even under the current standard for protecting human health (10 μg/dL), Pb
exposure poses an important health threat. These data can be used for developing interventions and identifying
high-risk pregnancies.

1. Introduction of epidemiological literature has shown that a multitude of noninher-


ited risk factors may increase the risk of CHDs, including maternal
Congenital heart defects (CHDs), structural abnormalities of the illnesses like pregestational diabetes, maternal medication exposures,
cardiovascular system characterized by anatomical departures of the environmental exposures, and paternal exposures, while periconcep-
heart and great vessels from normal, are the most common birth defects tional multivitamin or folic acid intake may reduce the risk (Donofrio
in newborns, affecting 6 to 12 per 1000 live births worldwide (Donofrio et al., 2014). Environmental pollutants, including toxic trace elements
et al., 2014). CHDs remain the leading cause of morbidity, mortality, such as heavy metals, may play a contributing role (Di Renzo et al.,
and disability in neonates and children, associated with lifelong 2015).
physical and mental comorbidities, and involving substantial financial Heavy metal pollution in China and trace element deficiencies
expenses (Donofrio et al., 2014). Over the past decade, a growing body constitute an important public health concern. Lead (Pb) exposure is


Corresponding authors at: Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong General Hospital,
Guangdong Academy of Medical Sciences, 96 Dongchuan Road, Guangzhou 510080, China.
E-mail addresses: drxqliu@163.com (X. Liu), drzhuangjian5413@163.com (J. Zhuang).
1
Yanqiu Ou, Michael S. Bloom and Zhiqiang Nie contributed equally to this paper.
2
Jian Zhuang and Xiaoqing Liu contributed equally to this paper.

http://dx.doi.org/10.1016/j.envint.2017.05.017
Received 18 December 2016; Received in revised form 19 May 2017; Accepted 19 May 2017
Available online 20 June 2017
0160-4120/ © 2017 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/BY-NC-ND/4.0/).
Y. Ou et al. Environment International 106 (2017) 127–134

widespread. The public may be exposed to Pb through contaminated 2. Materials and methods
food, water, house dust, and through industrial activities such as metal
recycling and the battery industry (Barbosa et al., 2005). In Guangdong, 2.1. Study design and subjects
daily exposure also exists through consumption of cultured fish, rice
paddy soil, and atmospheric pollution (Liang et al., 2016; Yang et al., This is a hospital-based case-control study conducted from
2004; Zeng et al., 2016). Maternal exposure to Pb during pregnancy has December 2012 to September 2013 at one provincial general hospital
been associated with a higher probability for congenital abnormalities in China. Pregnant women receiving routine prenatal care in the
among offspring (Liu et al., 2015). Exposure to other trace elements hospital obstetric department mostly reside in the Pearl River Delta
were also reported to increase the likelihood for congenital abnormal- Region of Guangdong Province. Both pregnant women treated by the
ities, such as cadmium (Cd) and chromium (Cr), that are associated hospital's obstetrics department as well as mothers treated at other
with modern industrial processes, and are absorbed in significant hospitals in the Pearl River Delta Region are referred to the Department
quantities from cigarette smoke (Thompson and Bannigan, 2008; Gil of Fetal Echocardiography in our institute if a CHD is suspected
et al., 2011). Exposure of experimental animals to oral or parenteral Cd following an obstetric ultrasound examination. In 2001, the cardiovas-
causes a wide range of embryo abnormalities, conditional on the stage cular institute reported 92% sensitivity and 95% specificity for
of exposure and the administered dose (Thompson and Bannigan, echocardiogram detection of fetal CHDs (Pan et al., 2001). Consecutive
2008). Selenium (Se) is an essential element, required for anti-oxidant singleton fetuses diagnosed with defined structural cardiac defects were
enzyme activities, and deficiency can occur with inadequate dietary recruited to the study at the time of the prenatal diagnosis, between 17
intake. Detectable traces of Se in drinking water were associated with a and 40 weeks. Among 1379 fetuses examined with fetal echocardio-
lower frequency of any CHD than was observed among children graphy, 170 of them were diagnosed with CHDs after review by two
exposed to drinking water without detectable Se levels (Zierler et al., physicians specialty-trained in echocardiography. We excluded fetuses
1988). Other findings further indicated that maternal Se deficiency with: 1) only mitral or tricuspid valvular lesions or enlarged atrial
during pregnancy might contribute to neural tube defects (NTDs) in septal defects (n = 6); 2) chromosomal malformations or syndromes, or
offspring (Cengiz et al., 2004). CHDs co-existing with ex-cardiac birth defects (n = 7); 3) self-reported
Like Cd, chromium (Cr) exposure mostly results from modern family history of CHDs (n = 4); 4) diabetes (n = 8); 5) multiple
industrial process and cigarette smoking (Gil et al., 2011). Human data gestations (n = 8); and 6) prenatal CHD diagnosis not confirmed after
showed a suggestive but not necessarily causal association between higher delivery (n = 3). Of 134 eligible case 112 (84%) agreed to participate
parental Cr exposure and increased CHDs, even at levels approaching but in our study. Each CHD case was matched by gestational age
not exceeding the 0.05 mg/L action level for protecting human health ( ± 3 months) and maternal age ( ± 5 years) to a control mother
(Goldberg et al., 1990). Copper (Cu) is an essential element required in without diabetes or a family history of CHDs, who was undergoing
trace concentrations for normal physiology. Yet, the relation between routine prenatal consultation for a normal singleton fetus randomly
maternal plasma Cu concentrations and birth defects in offspring is selected from the obstetrics clinic, on the same day. Of 119 eligible
unclear as few data are available to address the risk. A small case-control controls, 107 agreed to participate in our study; a participation rate of
study reported higher serum Cu among the mothers of elective termina- 90%, which was not differ from that of cases.
tions with NTDs, relative to matched live births without a defect (Cengiz All live births with a prenatal CHD diagnosis received B-mode
et al., 2004). Seafood consumption is the most important source of echocardiographic examination to confirm the diagnosis after delivery.
mercury (Hg) exposure among non-occupationally exposed populations Live births without a prenatal CHD diagnosis were routinely evaluated
(Wells et al., 2016). A suggestive association was found between higher before discharge to determine whether further investigation for a
placental Hg and higher NTD risk in offspring from a Chinese population potential CHD was needed. At least two senior pediatric cardiologists
(Jin et al., 2013). confirmed each CHD case diagnosis after delivery, using computed
The aforementioned elements cross the placenta and are thus tomography, cardiac catheterization, surgery, or autopsy (i.e., for cases
transferred from mother to a developing fetus (Caserta et al., 2013). of stillbirth and elective termination) as clinically necessary. CHD cases
Possible biological mechanisms underlying their teratogenic effects were coded based on the International Classification of Diseases version
include oxidative damage to the DNA and modification of epigenetic 10 (ICD-10) with codes Q20-Q28.
patterns (Hansen, 2006; Valinluck et al., 2004; Pilsner et al., 2009;
Ercal et al., 2001). Moreover, complex antagonistic interactions be- 2.2. Ethical approval
tween Se and toxic trace elements have been reported in previous
studies. For example, Se may exert an antagonistic effect on Pb induced All procedures performed in studies involving human participants
expression of inflammatory factors and heat shock proteins genes were in accordance with the ethical standards of the Guangdong
(Zheng et al., 2016), the cataractogenic effects of Hg may be offset by General Hospital Human subjects committee (No. 2011120H) and with
Se (Lemire et al., 2010), and Se has been identified as a potential the 1964 Helsinki declaration and its later amendments or comparable
countermeasure against Cd inducted toxicity (Zwolak and Zaporowska, ethical standards. Oral informed consent was obtained from the
2012). mothers of each fetus prior to study enrollment.
To date, biomarker based observational studies of gestational trace
element exposures and human CHDs remain sparse. There are even 2.3. Classification of CHD case subtypes
fewer data available to assess the simultaneous effects of multiple trace
elements, and their interactions with respect to CHDs. Given the Cases were classified into five cardiac phenotypes after modification
widespread nature of the exposures and tantalizing results from the based on prior reports (Leirgul et al., 2014), including: (i) conotruncal
few studies published to date, additional investigation is necessary. The defects (d-transposition of the great arteries [TGA], tetralogy of Fallot
current study was designed to address the existing data gap with an [ToF], double outlet right ventricle [DORV], conoventricular ventricle
investigation of the individual and combined effects of Pb, Cr, Cd, Cu, septum defect [VSD], truncus arteriosus, and interrupted aortic arch
Hg, and Se, on the occurrence of CHDs among mother-infant pairs in [IAA] type B or type C); (ii) left ventricle outflow tract obstructions
southeastern China. [LVOTO] (coarctation of aorta [CoA], aortic valve stenosis [vAS], and
hypoplastic left heart syndrome [HLHS]); (iii) right ventricle outflow
tract obstructions [RVOTO] (hypoplastic right heart syndrome [HRHS],
tricuspid atresia, Ebstein anomaly, pulmonary atresia [PA] or valvular
PA, and pulmonary valve stenosis [vPS] with or without septal defects);

128
Y. Ou et al. Environment International 106 (2017) 127–134

(iv) septal defects (VSD, atrial septum defect [ASD], and atrioventri- materials, Fig. S1) based on the literature (Wells et al., 2016; Aoki et al.,
cular septal defect [AVSD]); and (v) ‘other’ defects were classified into a 2016; Hinwood et al., 2013; Liu et al., 2013). We retained maternal age,
single group due to small sample sizes, including heterotaxia, pulmon- parity, maternal education, newborn gender, migrant status, folic acid
ary venous return (APVR), other complex heart defects (e.g., single or multivitamin intake, cigarette smoking, maternal prepregnancy body
ventricle, congenital corrected transposition of the great arteries), and mass index (maternal BMI), and gestational week at blood sample
other specific heart defects (e.g., isolated valvular malformations not collection as confounders in the regression models. Each was operatio-
classified as LVOTO or RVOTO and venous malformations). nalized as a categorical variable except for maternal BMI and gesta-
tional week at sample collection, which were left as continuous. We
2.4. Information collection adjusted all models for the same set of confounders to allow for
interpretation of results in a uniform context. To examine associations
Information about maternal demographic factors, pregnancy his- with a mixed exposure, we also constructed multi-element models,
tory, family history of CHDs, perinatal diseases (i.e. diabetes), medica- simultaneously including all measured maternal trace elements as
tion or supplement use, and fetal diagnosis was captured from the predictors. In sensitivity analyses, we repeated all models using
clinical record. Case and control mothers completed a face-to-face continuous trace elements as predictors to assess the robustness of
interviewer administered structured, standardized questionnaire to our findings, and we also limited the analysis to a subsample of cases
collect information on maternal education, exposure to trace elements and the controls residing in the Pearl River Delta Region to assess the
or metals in the workplace, migrant status, life behavior and environ- impact of selection bias.
mental factors, prepregnancy body weight and height, and also We used three approaches to explore modifying effects among trace
provided a blood sample. Two investigators double-entered and elements and to assess interactions: 1) use of categorical variables; 2)
examined the data for errors and accuracy prior to statistical analysis. interaction on the multiplicative scale; and 3) interaction on the
additive scale. For the categorical approach, levels of Pb, Cd and Se
2.5. Blood samples collection, preparation, and analysis were re-categorized as ‘low’ (1st and 2nd tertiles) and ‘high’ (3rd
tertile). We analyzed effects as low Pb with high Se, low Pb with low Se,
Blood samples were collected from mothers with gestational ages high Pb with high Se, and high Pb with low Se. Likewise, we assessed
from 17 to 40 weeks. Maternal peripheral venous blood was collected analogous effects between Pb and Cd, and Cd and Se. Pairwise
using trace-element-free syringes and transferred into a heparin coated interactions were modeled on the multiplicative scale by introducing
anticoagulant tube mixed by reversing the tube. All samples were cross-product terms into regression models and assessing significance of
immediately frozen at − 80 °C and stored in the hospital central the coefficients using the Wald statistic. For the additive scale, we
laboratory for 3–15 months before shipment to KingMed Diagnostics assessed interaction contrast ratios (ICR). The computations for ICR
Company (Guangzhou, China) for trace elements analysis. Prior to was: ICR = OR11-OR10-OR01 + 1, where OR11 denotes co-exposure
analysis, all samples were thawed, mixed thoroughly by vortex, and (e.g. high Pb and low Se, high Pb and high Cd, or high Cd and low
subsequently diluted with 0.1% Triton X-100 and 0.1% HNO3 at a ratio Se), OR10 or OR01 denotes the presence of one exposure and the absence
of 1:19. The diluted samples were stored for 5 min at 4 °C for analysis. of the other (Knol et al., 2011). An ICR above 1 indicates a super-
Whole blood Pb, Cr, Cd, Cu, total Hg, and Se concentrations were additive effect, whereas an ICR below 1 indicates a sub-additive effect.
determined by inductively coupled plasma mass spectrometry (ICP-MS) Confidence intervals (95%) for ICRs were computed by the multivariate
using the Agilent 7700 × ICP-MS system (Agilent Technologies, Santa delta method as ICR ± 1.96 √ var(ICR) (Nie et al., 2010).
Clara, CA, USA). Specimens were analyzed using external calibration Participants missing data for education (n = 6) were excluded from
and an internal standardization procedure with monitoring for isotopes: multivariable models, leaving n = 213 analyzed. Data were analyzed
206
Pb, 207Pb, 208Pb, 114Cd, 111Cd, 52Cr, 65Cu, 202Hg, 77Se, and 82Se. using SPSS (v.23.0; SPSS Inc., IBM, Chicago, IL USA) and SAS (v.9.3;
Seronorm™ Trace Elements Whole Blood L-2, a certified reference SAS Institute, Inc., Raleigh, NC USA). We used PASS v.13 (NCSS, LLC
material, was used to ensure quality control. Limits of detection Kaysville, UT USA) to identify, a priori, n = 94 cases needed to achieve
(LOD), in whole blood, were 0.39 μg/dL (Pb), 4.8 μg/L (Cr), 3.7 μg/L 80% power for detecting different proportions of Pb exposure by case-
(Cd), 26.4 μg/L (Cu), 2.8 μg/L (Hg), and 73.1 μg/L (Se). No trace control status at α = 0.05 in single-pollutant models. Statistical tests
elements were measured below the LOD. were two sided with a significance level of P < 0.05.

2.6. Statistical analysis 3. Results

Variables were first characterized by their measures of center and 3.1. Basic characteristics of the study participants
variability. For categorical factors, percentage was used to describe the
distributions and Chi-square or Fisher exact tests were used to compare Cases were categorized (not mutually exclusive) as: n = 42 con-
differences between groups as appropriate. Skewed continuous vari- otruncal defects; n = 62 septal defects; n = 16 LVOTOs; n = 32
ables were presented as median (interquartile range [IQR] RVOTOs; and n = 11 ‘other’ defects. Maternal basic characteristics
[25th ~ 75th percentiles]), and compared by the Wilcoxon-Mann- are presented in Table 1 compared by case-control status. Maternal
Whitney U test. The distribution of all of the trace elements were also parity, education level, migrant status, folic acid or multivitamin
shown in graphs by cases and controls. We also assessed the inter- intake, and maternal smoking were significantly different between
correlations among measured trace elements. the case and control groups. No mothers self-reported daily or occupa-
Because no information was available a priori to indicate a possible tional-contact with trace elements or metals.
dose threshold, trace element concentrations were divided into low,
middle and high levels according to tertiles of control distributions 3.2. Trace elements in maternal blood samples
prior to analysis (Supplementary materials, Table S1). The associations
between individual maternal trace elements as predictors and CHDs as Median (IQR) Pb [3.81 (3.13–4.84) vs. 2.61 (2.02–3.40) μg/dL,
outcomes, including total CHDs and CHD subtypes, were assessed as P < 0.001] and Cd [2.25 (1.72–3.24) vs. 1.76 (1.35–2.55) μg/L,
unadjusted and confounder-adjusted odds ratios (aOR) and their 95% P = 0.001] levels in cases were significantly higher than in controls,
confidence intervals (95% CI) using separate logistic regression models. respectively, while median (IQR) Cu [838.38 (778.90–936.98) vs.
We incorporated factors associated with exposure to trace elements and 896.56 (817.62–979.44) μg/L, P = 0.02], Hg [3.63 (2.73–4.82) vs.
the risk for CHDs, into a directed acyclic graph (DAG) (Supplementary 4.17 (3.17–5.58) μg/L, P = 0.03], and Se [Se = 172.90

129
Y. Ou et al. Environment International 106 (2017) 127–134

Table 1 3.3. Maternal blood trace elements level and fetal CHDs
Descriptive characteristics of the study sample.
Associations between the trace elements and CHDs were first
Variable Case (n = 112) Control P-valueb
n (%) or median (n = 107) investigated using single-element regression models. The unadjusted
(IQR)a n (%) or median effect estimates are shown in the Supplementary materials (Table S3).
(IQR)a Multivariable models were adjusted for maternal age, parity, education,
newborn gender, migrant status, folic acid or multivitamin intake,
Maternal age (years)
≤ 25 31 (27.7) 20 (18.7) 0.47 cigarette smoking, maternal pre-pregnancy BMI, and gestational week
26–30 50 (44.6) 52 (48.6) at the time of sample collection. As shown in Table 3, compared with
31–35 26 (23.2) 29 (27.1) the low level of Pb, mothers in the middle and high levels had higher
≥ 36 5 (4.5) 6 (5.6) odds for total CHDs, with ORs of 6.95 (95% CI: 1.69, 28.62) and 14.46
Parity (95% CI: 3.59, 58.17), respectively. On the other hand, mothers with
Multiparous 42 (37.5) 15 (14.0) < 0.001 high level Se bore the lowest odds for total CHDs [OR = 0.35, 95% CI:
Primiparous 70 (62.5) 92 (86.0)
0.15, 0.81] vs. the low level. The middle level of Cr was also protective
Newborn gender for total CHDs [OR = 0.22, 95% CI: 0.08, 0.57], yet the high level was
Male 48 (42.9) 58 (54.2) 0.24
not associated. The association of the high level of Pb and Se was also
Female 54 (48.2) 41 (38.3)
Unknown 10 (8.9) 8 (7.5) present in subtypes of conotruncal defects, septal defects, and RVOTO.
However, the association of the middle level of Cr was not statistically
Educationc
≤ 12 years 65 (58.0) 18 (17.8) < 0.001
significant in CHD subtypes. For Cu, protective effects were statistically
> 12 years 47 (42.0) 83 (82.2) significant only for LVOTO defects (Supplementary materials, Table
S4).
Migrantd
Yes 23 (20.5) 11 (10.3) 0.04 To further elaborate on the associations detected between trace
No 89 (79.5) 96 (89.7) elements and CHDs, multi-element models were employed to simulta-
Folic acid or multivitamin intake neously incorporate all measured trace elements as predictors of CHDs,
No 14 (12.5) 5 (4.7) 0.04 adjusted for confounders. As shown in Table 4, with the multi-element
Yes 98 (87.5) 102 (95.3) models, associations for the high levels of Pb and Se maintained
Maternal occupation as manual worker statistical significance for total CHDs (Pb: OR = 12.09, 95% CI:2.81,
Yes 10 (8.9) 4 (3.7) 0.12 51.97; Se: OR = 0.25, 95% CI: 0.08, 0.77), conotruncal defect, septal
No 102 (91.1) 103 (96.3) defect, and RVOTO defects. Associations for the middle level of Hg was
Maternal smoking also statistical significance with RVOTO defects (OR = 5.20, 95% CI:
Yes 15 (13.4) 1 (0.9) < 0.001 1.03, 26.18), but not in total CHDs or other subtypes. Other elements
No 97 (86.6) 106 (99.1)
including Cd, Cr and Cu did not show any statistically significant
Living in a newly renovated home association. In a sensitivity analysis using trace elements as continuous
Yes 8 (7.1) 6 (5.6) 0.64
predictors (Supplementary materials, Table S5), higher blood Pb was
No 104 (92.9) 101 (94.4)
Maternal prepregnancy BMI 19.8 (18.4–21.3) 20.3 (19.0–20.7) 0.29
also associated with a higher odds for total CHDs, conotruncal defect,
(kg/m2) septal defect, and RVOTO defects. However, protective effects were
Gestational week at sample 27.0 (25.0–29.0) 28.0 (24.0–28.0) 0.26 significant only for RVOTO defects with higher continuous Se expo-
collection (weeks) sures. We also conducted a sensitivity analysis limited to cases (n = 96)
a
and controls (n = 93) residing in the nearby Pearl River Delta Region,
For categorical variables, percentage was used to describe the distribution; for
abnormal distributed continuous variables, median (IQR) was used to describe the
with similar results as for the overall analysis (Table S6).
distribution.
b
P-value was calculated with Chi-square or Fisher exact tests. 3.4. Interaction between Pb and Cd, and Se
c
n = 6 missing observations.
d
Migrant means the people were not permeant residents in the local region, and did
We tested pairwise interactions among maternal blood Pb, Cd, and
not have local medical insurance.
Se to assess for synergic or antagonistic effects on the occurrence of
CHDs. Maternal blood Pb, Cd, and Se levels were first dichotomized as
Table 2
Median (interquartile range) for blood trace element levels measured in cases and ‘low’ (the 1st and 2nd tertiles) and ‘high’ (the 3rd tertile), and then
controls (n =219 )a. cross-product terms entered into logistic regression models, adjusted for
confounders (Table 5). There was no significant interaction on the
Trace elements Case median (IQR) Control median (IQR) P-value multiplicative or additive scales for Pb and Se, Pb and Cd, or for Cd and
Pb (μg/dL) 3.81 (3.13–4.84) 2.61 (2.02–3.40) < 0.001 Se. The only significant results came from the use of categorical
Cd (μg/L) 2.25 (1.72–3.24) 1.76 (1.35–2.55) 0.001 variables. The positive associations for Pb or Cd and total CHDs was
Cr (μg/L) 3.63 (2.09–4.10) 3.57 (3.27–3.99) 0.16 stronger among low Se mothers than among high Se mother. Likewise,
Cu (μg/L) 838.38 (778.90–936.98) 896.56 (817.62–979.44) 0.02 having high Cd in addition to high Pb appeared to elicit a stronger
Hg (μg/L) 3.63 (2.73–4.82) 4.17 (3.17–5.58) 0.03
Se (μg/L) 172.90 (153.87–192.23) 186.47 (172.45–207.34) < 0.001
association on fetal cardiac development than high Pb alone, although
ICR confidence intervals included the null (Table 5).
Pb, lead; Cd, cadmium; Cr, chromium; Cu, copper; Hg, mercury; Se, selenium.
a
Wilcoxon-Mann-Whitney U test was used to compare the difference between groups. 4. Discussion

(153.87–192.23) vs. 186.47 (172.45–207.34) μg/L, P < 0.001] levels To the best of our knowledge, this is the first study to identify
were significantly lower in cases than in controls, respectively (Table 2 associations between CHDs in offspring and maternal exposure to
and Supplementary materials, Fig. S2). As shown Table S2 in the multiple trace elements during pregnancy as measured in blood.
Supplementary materials, we detected modest to moderate correlations Specifically, we observed that maternal blood Pb and Se were harmful
between Pb and Cd (r = 0.29, P < 0.001), Pb and Cu (r = - 0.14, and protective predictors of total CHDs respectively. The findings were
P = 0.04), Cd and Cu (r = − 0.23, P < 0.001), Cr and Cu (− 0.27, consistent for CHD subtypes, including conotruncal defects, RVOTO,
P < 0.001), and Hg and Se (r = 0.57, P < 0.001). and septal defects. Finally, our results suggest that mothers with low Se

130
Y. Ou et al. Environment International 106 (2017) 127–134

Table 3
Adjusted ORs (95% CI) for associations between maternal blood trace element levels and total congenital heart defects using multivariable single-element logistic regression modelsa.

Trace element Low level Middle level High level

n cases/controls n cases/controls OR (95%CI) n cases/controls OR (95%CI)

Pb 2/36 24/35 6.95 (1.69, 28.62) 86/36 14.46 (3.59, 58.17)


Cd 21/36 30/36 1.23 (0.54, 2.82) 61/35 1.49 (0.65, 3.40)
Cr 50/37 13/35 0.22 (0.08, 0.57) 49/35 1.07 (0.50, 2.30)
Cu 55/36 28/35 0.48 (0.21, 1.10) 29/36 0.63 (0.29, 1.39)
Hg 48/36 40/36 0.82 (0.38, 1.77) 24/35 0.52 (0.22, 1.22)
Se 63/36 29/36 0.58 (0.27, 1.26) 20/35 0.35 (0.15, 0.81)

Pb, lead; Cd, cadmium; Cr, chromium; Cu, copper; Hg, mercury; Se, selenium; OR: odds ratio; CI, confidence interval.
a
Low level serves as the reference group. Models were adjusted for maternal age (≤ 25; 26–30; 31–35 or ≥ 36 years old), parity (multiparous/primiparous), education (≤ 12 years/
> 12 years), newborn gender (male/female), migrant (yes/no), folic acid or multivitamin intake (yes/no), cigarette smoking (yes/no), maternal pre-pregnancy body mass index (kg/m2),
and time of sample collection (gestational week).

Table 4
Adjusted ORs (95% CI) for associations between maternal blood trace element levels and congenital heart defects using multivariable multi-element logistic regression modelsa.

Total defects (n = 112) Conotruncal defects (n = 42) Septal defects (n = 62) RVOTO (n = 32)

Middle level High level Middle level High level Middle level High level Middle level High level

Pb 5.89 (1.38, 25.20) 12.09 (2.81, 51.97) 12.89 (1.02, 162.94) 21.86 (1.70, 280.77) 4.18 (0.70, 24.90) 15.28 (2.63, 88.84) 1.10 (0.15, 8.28) 7.06 (1.06, 47.27)
Cd 1.28 (0.51, 3.24) 1.26 (0.48, 3.31) 0.56 (0.14, 2.14) 1.09 (0.29, 4.03) 2.33 (0.73, 7.49) 1.77 (0.54, 5.82) 0.53 (0.11, 2.49) 0.97 (0.21, 4.50)
Cr 0.24 (0.08, 1.69) 0.84 (0.36, 1.96) 0.44 (0.10, 1.89) 1.34 (0.40, 4.50) 0.19 (0.05, 1.74) 0.86 (0.31, 2.39) 0.30 (0.05, 1.67) 0.66 (0.15, 2.80)
Cu 0.57 (0.22, 1.47) 0.77 (0.31, 1.89) 0.50 (0.12, 2.15) 0.58 (0.16, 2.17) 0.81 (0.25, 2.63) 1.14 (0.38, 3.46) 0.74 (0.16, 3.38) 1.06 (0.21, 5.50)
Hg 1.36 (0.54, 3.48) 1.42 (0.45, 4.45) 1.62 (0.49, 5.37) 0.84 (0.17, 4.25) 1.15 (0.37, 3.53) 1.35 (0.37, 4.89) 5.20 (1.03, 26.18) 1.99 (0.26, 15.29)
Se 0.55 (0.22, 1.38) 0.25 (0.08, 0.77) 0.53 (0.15, 1.86) 0.13 (0.03, 0.68) 0.34 (0.11, 1.07) 0.20 (0.06, 0.71) 0.27 (0.06, 1.27) 0.10 (0.01, 0.72)

Pb, lead; Cr, chromium; Cd, cadmium; Cu, copper; Hg, mercury; Se, selenium; RVOTO, right ventricle outflow tract obstructions; OR: odds ratio.
The results for left ventricle outflow tract obstructions (LVOTO) was not shown because of limited space. There was no statistical significance for LVOTO.
a
Low level serves as the reference group. Models were adjusted for maternal age (≤ 25; 26–30; 31–35 or ≥ 36 years old), parity (multiparous/primiparous), education (≤ 12 years/
> 12 years), newborn gender (male/female), migrant (yes/no), folic acid or multivitamin intake (yes/no), cigarette smoking (yes/no), maternal prepregnancy body mass index (kg/m2),
and time of sample collection (gestational week).

or high Cd in addition to high Pb had inflated odds for CHDs relative to occupational exposure assessments. A widely validated biomarker that
mothers with elevated blood Pb only. These data add to the growing integrates all sources of exposure and reflects body burden over
concern for developmental toxicity with maternal Pb exposures at levels extended intervals in the context of ongoing exposures (Hu et al.,
lower than the Chinese reference level for child health equal to 10 μg/ 2007), is likely to offer a stronger design for assessing associations with
dL (Li et al., 2015). congenital abnormalities. To our knowledge, only two previous studies
Trace element levels measured in our study population were evaluated the effect of lead on birth defects using maternal exposure
generally higher than reported by previous studies of pregnant women. biomarkers. In Fallujah, Iraq, adult tooth enamel Pb was 1.4 times
The median value of blood Pb among controls (2.61 μg/dL) far higher in the parents of children with birth defects than in the parents
exceeded that reported for a representative sample of pregnant U.S. of controls (Al-Sabbak et al., 2012). A hospital-based Chinese study of
women in 2003–2004 (0.60 μg/dL); in fact, our lowest exposure n = 316 cases and n = 348 controls reported a 2–4 times higher odds
category (< 2.15 μg/dL) corresponded approximately to the 95th% for carrying a CHD baby (including total CHDs, septal defects,
tile in that study (1.8 μg/dL) (Woodruff et al., 2011). Our mean Pb level contruncal defect, RVOTO, and LVOTO) in association with higher
(2.90 μg/dL) was closer to that reported for 178 Spanish women with maternal hair Pb (Liu et al., 2015). In the current study, we detected
occupational exposure to heavy metals (2.77 μg/dL) (Gil et al., 2011). similar effects using maternal blood Pb as a biomarker of exposure,
The median levels of blood Cd (1.76 μg/L) and Hg (4.17 μg/L) among although our sample size was smaller and effect estimates imprecise. In
controls in our study also exceeded those reported for pregnant contrast to the prior Chinese study, our results do not indicate an
Canadian women (0.20 μg/L for Cd and 0.56 μg/L for Hg) (Arbuckle association between maternal Pb exposure and LVOTO, possibly due to
et al., 2016). For blood Se, the median level (186.47 μg/L) in our different CHD subtypes considered, as well as differences in the
control participants was higher than that reported for 230 primiparous covariates included in regression models. Still, our overall data are
women (1.31 μmol/L, equal to 103.44 μg/L) participating in a British consistent with the results of previous studies using biomarkers of
preeclampsia prevention trial (Rayman et al., 2015). exposure to assess associations between maternal Pb and fetal CHDs.
Lead readily crosses the placenta, transferred from the mother to the
4.1. Associations between maternal blood Pb and CHDs developing fetus. Yet, the biological mechanisms underlying Pb′s
teratogenic effects are poorly elucidated. Excessive maternal Pb can
Experimental neural crest cell ablation induces conotruncal anoma- reduce total antioxidant capacity, which is regarded as a common
lies in mice (Porras and Brown, 2008) and high Pb concentrations mechanism of teratogenesis (Hansen, 2006), and specifically for CHDs
altered rat neural crest cell proliferation and differentiation in vitro (Hobbs et al., 2005). Oxidative damage to DNA could also impact
(Huang and Schneider, 2004), so it is plausible that high Pb levels could methylation patterns, possibly leading to inherited changes in gene
affect conotruncal anomalies. Previous studies also described associa- expression (Valinluck et al., 2004); in a prior study, prenatal Pb
tions between maternal or parental lead exposure and heart defects in exposure was inversely associated with genomic DNA methylation in
offspring, including defects in APVR (Jackson et al., 2004), PA (Correa- cord blood (Pilsner et al., 2009). In addition, blood Pb influenced
Villasenor et al., 1993), and single ventricle (Steinberger et al., 2002). homocysteine level, which is also associated with reduced DNA
However, these prior investigations were limited by self-reported or methylation (Pilsner et al., 2009). While it is tempting to speculate

131
Y. Ou et al. Environment International 106 (2017) 127–134

Table 5 4.4. Interactions between trace elements


Interaction effects among blood Pb, Cd, and Se on the odds for total congenital heart
defects using multivariable logistic regression (n = 219)a.
To our knowledge, there have been no previous studies of trace
Exposure OR (95% CI) P-value element interactions on CHDs. Antagonism between Se, Pb (Zheng
et al., 2016), and Cd (Zwolak and Zaporowska, 2012) has been well
Pb/Se category documented, and therefore Se status may have modified associations
Low Pb and high Se Reference –
between Pb and CHDs in our study. In addition, it has been proposed
Low Pb and low Se 2.03 (0.60, 6.89) 0.26
High Pb and high Se 3.47 (0.88, 13.65) 0.07 that co-exposure to Pb and Cd might exert a synergistic detrimental
High Pb and low Se 9.63 (2.83, 32.74) < 0.001 effect on reproductive capabilities (Gollenberg et al., 2010). Pb and Cd
Cross-Product term for interaction 1.37 (0.29, 6.46) 0.69 may act through a similar oxidative-stress related biologic mechanism
ICR for interaction on the additive scale 5.13 (− 2.32, 12.57) 0.18 (Ercal et al., 2001). Although not statistically significant, our results
Pb/Cd category suggested that co-exposure to higher Pb and Cd, or higher Pb and lower
Low Pb and low Cd Reference – Se, or higher Cd and lower Se, potentiated the odds for CHDs, although
Low Pb and high Cd 1.61 (0.57, 4.54) 0.37
possibly due to random chance. However, this null result may have
High Pb and low Cd 4.32 (1.86, 10.04) < 0.001
High Pb and high Cd 5.21 (2.25, 12.07) < 0.001 been due to insufficient power because of the relatively small sample
Cross-Product term for interaction 0.75 (0.19, 2.89) 0.67 size. Still, given the paucity of research on the joint effects of toxic
ICR for interaction on the additive scale 0.28 (− 4.10, 4.65) 0.90 elements for fetal cardiac development, further studies are warranted to
Cd/Se category more definitively interpret these findings.
Low Cd and high Se Reference –
Low Cd and low Se 1.55 (0.62, 3.88) 0.35 4.5. Strengths and limitations
High Cd and high Se 0.98 (0.28, 3.49) 0.98
High Cd and low Se 3.47 (1.29, 9.30) 0.01
Cross-Product term for interaction 2.29 (0.53, 9.88) 0.27 Our study offers several strengths, which lend validity to our results.
ICR for interaction on the additive scale 1.94 (− 0.50, 4.38) 0.12 We used a biomarker to capture maternal exposures during pregnancy,
and clinical records to capture demographic, pregnancy, and birth data,
Low Pb: 1st and 2nd tertiles of blood Pb level; high Pb: 3rd tertile of blood Pb level. which are not vulnerable to recall bias associated with differential
Low Se: 1st and 2nd tertiles of blood Se level; high Se: 3rd tertile of blood Se level.
reporting between case and control mothers. For each case, we matched
Low Cd: 1st and 2nd tertiles of blood Cd level; high Cd: 3rd tertile of blood Cd level.
OR: Odds ratio; CI, confidence interval; ICR: interaction contrast ratio.
controls on the same day as the diagnosis and from the same obstetrics
a
Adjusted for maternal age (≤25; 26–30; 31–35 or ≥ 36 years old), parity (multi- clinic to ensure representativeness of the sampling frame. We also
parous/primiparous), education (≤ 12 years/ > 12 years), newborn gender (male/fe- assessed for associations with additional trace elements measured in
male), migrant (yes/no), folic acid or multivitamin intake (yes/no), cigarette smoking maternal blood specimens, and adjusted for a panel of potentially
(yes/no), maternal prepregnancy body mass index (kg/m2), and time of sample collection
confounding variables.
(gestational week).
However, the results of this study are also limited by several
important factors. Firstly, our higher-tier hospital may experience
on a role for epigenetic changes in mediating Pb-CHD associations, to
differential referral patterns for residents of Guangdong Province living
our knowledge, no data have been reported to describe associations
outside of the Pearl River Delta, in which cases are referred from a large
between changing epigenetic patterns and CHDs. Additional research is
area, whereas controls mostly reside nearby; this might lead to a
warranted to assess whether prenatal Pb exposure influences cardiac
selection bias. However, we found similar results in a sensitivity
gene-specific DNA methylation profiles, and to characterize the impact
analysis limited to study participants (both cases and controls) residing
of different methylation profiles on cardiac development.
in the nearby Pearl River Delta area and so we believe any impact to
have been minimal. Secondly, our sample size was relatively small. In
4.2. Associations between Se and CHDs the subgroup analysis for Pb and conotruncal defects and LVOTO
defects, no cases were available for low level exposure, potentially
There is a paucity of evidence showing the contribution of Se to limiting statistical power to detect modest associations and interactions,
CHDs. However, several studies underscore the importance of Se to and producing the extremely wide confidence intervals that we
NTDs. In a prospective cohort study, serum Se was significantly lower in observed. However, we did find significant differences and consistent
mothers of infants with congenital malformations compared to the effect trends for Pb and Se, even with the limited sample size. Thirdly,
mothers of infants without the malformation (Hammouda et al., 2013). blood was collected from participants in mid- and late-pregnancy, after
Another study indicated that maternal Se deficiency during pregnancy the critical period for cardiac development during the 3rd-8th week of
might contribute to NTDs (Cengiz et al., 2004). Compelling evidence gestation (Sadler, 2004), which may have misclassified exposure for
suggests that although toxic at high doses, Se is essential at trace some participants. However, Pb measurements reflect not only recent
concentrations for antioxidant enzyme activities and normal fetal exposure, but also provide an indication for historic exposures as a
development (Martin et al., 2004). Changes in epigenetic patterning result of Pb mobilization from bone (Gulson et al., 2003). In subjects
were also associated with Se exposure in human studies and with without excessive Pb exposure, 45–75% of the blood Pb may reflect
animal models (Pilsner et al., 2011; Davis et al., 2000). bone stores (Barbosa et al., 2005). Arbuckle recently reported decreases
in blood Pb without changes in blood Cd over pregnancy (Arbuckle
et al., 2016). Mistry reported relatively stable Se levels across preg-
4.3. Associations between other trace elements and CHDs nancy, unless women are depleted (Mistry et al., 2012). So, exposure
misclassification due to mistimed specimen collection is likely to be
Although some association were found between Cr, Cu, or Hg and non-differential with respect to CHDs and bias effects estimates towards
total or certain subtypes of CHDs in the single-element or multi-element the null hypothesis. Furthermore, we matched on gestational age and so
multivariable models, these seemed to be chance findings given mobilized bone Pb is unlikely to have contributed differentially
inconsistencies throughout the analysis. Null associations for these between cases and control. Still, any exposure misclassification due to
elements with CHDs are consistent with several previous studies, which mistimed blood specimen collection is likely to have been similar
reported no associations between these trace elements and CHDs in between CHD cases and controls, which may have undermined our
animals or humans (Hansen, 2006; Ercal et al., 2001; Al-Sabbak et al., ability to detect modest associations for other trace elements with null
2012). results. Finally, we did not capture data for additional potential

132
Y. Ou et al. Environment International 106 (2017) 127–134

confounders, such as diet, and so we are unable to assess the effect on 2011. Biomonitorization of cadmium, chromium, manganese, nickel and lead in
whole blood, urine, axillary hair and saliva in an occupationally exposed population.
associations between trace elements and CHDs. Sci. Total Environ. 409 (6), 1172–1180. http://dx.doi.org/10.1016/j.scitotenv.2010.
11.033.
5. Conclusions Goldberg, S.J., Lebowitz, M.D., Graver, E.J., Hicks, S., 1990. An association of human
congenital cardiac malformations and drinking water contaminants. J. Am. Coll.
Cardiol. 16 (1), 155–164.
Environmental Pb contamination remains a public health concern in Gollenberg, A.L., Hediger, M.L., Lee, P.A., Himes, J.H., Louis, G.M., 2010. Association
China. Blood Pb levels in our study were mostly lower than the Chinese between lead and cadmium and reproductive hormones in peripubertal U.S. girls.
Environ. Health Perspect. 118 (12), 1782–1787. http://dx.doi.org/10.1289/ehp.
reference level for child health equal to 10 μg/dL (Li et al., 2015). Still, 1001943.
we found strong effects for CHDs with higher maternal blood Pb at Gulson, B.L., Mizon, K.J., Korsch, M.J., Palmer, J.M., Donnelly, J.B., 2003. Mobilization
these levels of exposure; an effect that may have been enhanced by of lead from human bone tissue during pregnancy and lactation–a summary of long-
term research. Sci. Total Environ. 303 (1–2), 79–104.
concurrent Cd exposure and mitigated by concurrent Se exposure. Our
Hammouda, S.A., Abd Al-Halim, O.A., Mohamadin, A.M., 2013. Serum levels of some
study results should prove useful for developing interventions to reduce micronutrients and congenital malformations: a prospective cohort study in healthy
exposures among women of childbearing age residing in Pb-polluted saudi-arabian first-trimester pregnant women. Int. J. Vitam. Nutr. Res. 83 (6),
and Se-deficient areas of Guangdong Province, and to help to identify 346–354. http://dx.doi.org/10.1024/0300-9831/a000176.
Hansen, J.M., 2006. Oxidative stress as a mechanism of teratogenesis. Birth Defects Res.
high risk pregnancies for referral to fetal echocardiography and C. Embryo Today 78 (4), 293–307. http://dx.doi.org/10.1002/bdrc.20085.
intensive prenatal monitoring. Hinwood, A.L., Callan, A.C., Ramalingam, M., Boyce, M., Heyworth, J., McCafferty, P.,
et al., 2013. Cadmium, lead and mercury exposure in non smoking pregnant women.
Environ. Res. 126, 118–124. http://dx.doi.org/10.1016/j.envres.2013.07.005.
Funding Hobbs, C.A., Cleves, M.A., Zhao, W., Melnyk, S., James, S.J., 2005. Congenital heart
defects and maternal biomarkers of oxidative stress. Am. J. Clin. Nutr. 82 (3),
This work was supported by Guangdong Science and Technology 598–604.
Hu, H., Shih, R., Rothenberg, S., Schwartz, B.S., 2007. The epidemiology of lead toxicity
Department, China [grant number 2014A050503048]; the National in adults: measuring dose and consideration of other methodologic issues. Environ.
Natural Science Foundation of China [grant number U1401255]. Health Perspect. 115 (3), 455–462.
Huang, F., Schneider, J.S., 2004. Effects of lead exposure on proliferation and
differentiation of neural stem cells derived from different regions of embryonic rat
Conflict of interest
brain. Neurotoxicology 25, 1001–1012.
Jackson, L.W., Correa-Villasenor, A., Lees, P.S., Dominici, F., Stewart, P.A., Breysse, P.N.,
The authors declare that they have no competing interests. et al., 2004. Parental lead exposure and total anomalous pulmonary venous return.
Birth Defects Res. A Clin. Mol. Teratol. 70 (4), 185–193. http://dx.doi.org/10.1002/
bdra.20014.
Appendix A. Supplementary data Jin, L., Zhang, L., Li, Z., Liu, J.M., Ye, R., Ren, A., 2013. Placental concentrations of
mercury, lead, cadmium, and arsenic and the risk of neural tube defects in a Chinese
Supplementary data to this article can be found online at http://dx. population. Reprod. Toxicol. 35, 25–31. http://dx.doi.org/10.1016/j.reprotox.2012.
10.015.
doi.org/10.1016/j.envint.2017.05.017. Knol, M.J., VanderWeele, T.J., Groenwold, R.H., Klungel, O.H., Rovers, M.M., Grobbee,
D.E., 2011. Estimating measures of interaction on an additive scale for preventive
References exposures. Eur. J. Epidemiol. 26, 433–438. http://dx.doi.org/10.1007/s10654-011-
9554-9.
Leirgul, E., Fomina, T., Brodwall, K., Greve, G., Holmstrom, H., Vollset, S.E., et al., 2014.
Al-Sabbak, M., Sadik Ali, S., Savabi, O., Savabi, G., Dastgiri, S., Savabieasfahani, M., Birth prevalence of congenital heart defects in Norway 1994–2009 — a nationwide
2012. Metal contamination and the epidemic of congenital birth defects in Iraqi study. Am. Heart J. 168 (6), 956–964. http://dx.doi.org/10.1016/j.ahj.2014.07.030.
cities. Bull. Environ. Contam. Toxicol. 89 (5), 937–944. http://dx.doi.org/10.1007/ Lemire, M., Fillion, M., Frenette, B., Mayer, A., Philibert, A., Passos, C.J., et al., 2010.
s00128-012-0817-2. Selenium and mercury in the Brazilian Amazon: opposing influences on age-related
Aoki, Y., Brody, D.J., Flegal, K.M., Fakhouri, T.H., Axelrad, D.A., Parker, J.D., 2016. cataracts. Environ. Health Perspect. 118 (11), 1584–1589. http://dx.doi.org/10.
Blood lead and other metal biomarkers as risk factors for cardiovascular disease 1289/ehp.0901284.
mortality. Medicine (Baltimore) 95 (1), e2223. http://dx.doi.org/10.1097/MD. Li, M.M., Cao, J., Gao, Z.Y., Shen, X.M., Yan, C.H., 2015. The trend of lead poisoning rate
0000000000002223. in Chinese population aged 0–18 years old: a meta-analysis. BMC Public Health 15,
Arbuckle, T.E., Liang, C.L., Morisset, A.S., Fisher, M., Weiler, H., Cirtiu, C.M., et al., 2016. 756. http://dx.doi.org/10.1186/s12889-015-2103-9.
Maternal and fetal exposure to cadmium, lead, manganesium and mercury: the Liang, P., Wu, S.C., Zhang, J., Cao, Y., Yu, S., Wong, M.H., 2016. The effects of
MIREC study. Chemosphere 163, 270–282. http://dx.doi.org/10.1016/j. mariculture on heavy metal distribution in sediments and cultured fish around the
chemosphere.2016.08.023. Pearl River Delta region, south China. Chemosphere 148, 171–177. http://dx.doi.
Barbosa Jr., Fernando, Tanus-Santos, José Eduardo, Gerlach, Raquel Fernanda, Parsons, org/10.1016/j.chemosphere.2015.10.110.
Patrick J., 2005. A critical review of biomarkers used for monitoring human exposure Liu, Z., Li, X., Li, N., Li, S., Deng, K., Lin, Y., et al., 2013. Association between maternal
to lead: advantages, limitations, and future needs. Environ. Health Perspect. 113 exposure to housing renovation and offspring with congenital heart disease: a multi-
(12), 1669–1674. hospital case-control study. Environ. Health 12, 25. http://dx.doi.org/10.1186/1476-
Caserta, D., Graziano, A., Lo Monte, G., Bordi, G., Moscarini, M., 2013. Heavy metals and 069X-12-25.
placental fetal-maternal barrier: a mini-review on the major concerns. Eur. Rev. Med. Liu, Z., Yu, Y., Li, X., Wu, A., Mu, M., Li, N., et al., 2015. Maternal lead exposure and risk
Pharmacol. Sci. 17, 2198–2206. of congenital heart defects occurrence in offspring. Reprod. Toxicol. 51, 1–6. http://
Cengiz, B., Soylemez, F., Ozturk, E., Cavdar, A.O., 2004. Serum zinc, selenium, copper, dx.doi.org/10.1016/j.reprotox.2014.11.002.
and lead levels in women with second-trimester induced abortion resulting from Martin, I., Gibert, M.J., Pintos, C., Noguera, A., Besalduch, A., Obrador, A., 2004.
neural tube defects: a preliminary study. Biol. Trace Elem. Res. 97 (3), 225–235. Oxidative stress in mothers who have conceived fetus with neural tube defects: the
http://dx.doi.org/10.1385/BTER:97:3:225. role of aminothiols and selenium. Clin. Nutr. 23 (4), 507–514. http://dx.doi.org/10.
Correa-Villasenor, A., Ferencz, C., Loffredo, C., Magee, C., 1993. Paternal exposures and 1016/j.clnu.2003.09.010.
cardiovascular malformations. The Baltimore-Washington infant study group. J. Mistry, H., Broughton Pipkin, F., Redman, C.W.G., Poston, L., 2012. Selenium in
Expo. Anal. Environ. Epidemiol. 3 (Suppl. 1), 173–185. reproductive health. Am. J. Obstet. Gynecol. 206, 21–30. http://dx.doi.org/10.1016/
Davis, C.D., Uthus, E.O., Finley, J.W., 2000. Dietary selenium and arsenic affect DNA j.ajog.2011.07.034.
methylation in vitro in Caco-2 cells and in vivo in rat liver and colon. J. Nutr. 130 Nie, L., Chu, H.T., Li, F., Cole, S.R., 2010. Relative excess risk due to interaction:
(12), 2903–2909. resampling-based confidence intervals. Epidemiology 21 (4), 552–556. http://dx.doi.
Di Renzo, G.C., Conry, J.A., Blake, J., DeFrancesco, M.S., DeNicola, N., Martin Jr., J.N., org/10.1097/EDE.0b013e3181e09b0b.
et al., 2015. International Federation of Gynecology and Obstetrics opinion on Pan, W., Wu, G.P., Li, Y.F., Qian, M.Y., Wang, H.S., Li, H., et al., 2001. The experience of
reproductive health impacts of exposure to toxic environmental chemicals. Int. J. diagnosing congenital heart disease by fetal echocardiography on 900 fetuses. South
Gynaecol. Obstet. 131 (3), 219–225. http://dx.doi.org/10.1016/j.ijgo.2015.09.002. China J. Cardiovasc. Dis. 7 (3), 180–182 (Article in Chinese).
Donofrio, M.T., Moon-Grady, A.J., Hornberger, L.K., Copel, J.A., Sklansky, M.S., Pilsner, J.R., Hu, H., Ettinger, A., Sanchez, B.N., Wright, R.O., Cantonwine, D., et al.,
Abuhamad, A., et al., 2014. Diagnosis and treatment of fetal cardiac disease: a 2009. Influence of prenatal lead exposure on genomic methylation of cord blood
scientific statement from the American Heart Association. Circulation 129 (21), DNA. Environ. Health Perspect. 117 (9), 1466–1471. http://dx.doi.org/10.1289/ehp.
2183–2242. http://dx.doi.org/10.1161/01.cir.0000437597.44550.5d. 0800497.
Ercal, N., Gurer-Orhan, H., Aykin-Burns, N., 2001. Toxic metals and oxidative stress part Pilsner, J.R., Hall, M.N., Liu, X., Ahsan, H., Ilievski, V., Slavkovich, V., et al., 2011.
I: mechanisms involved in metal-induced oxidative damage. Curr. Top. Med. Chem. 1 Associations of plasma selenium with arsenic and genomic methylation of leukocyte
(6), 529–539. DNA in Bangladesh. Environ. Health Perspect. 119 (1), 113–118. http://dx.doi.org/
Gil, F., Hernandez, A.F., Marquez, C., Femia, P., Olmedo, P., Lopez-Guarnido, O., et al., 10.1289/ehp.1001937.

133
Y. Ou et al. Environment International 106 (2017) 127–134

Porras, D., Brown, C.B., 2008. Temporal-spatial ablation of neural crest in the mouse sex. Environ. Health Perspect. 124 (3), 373–379. http://dx.doi.org/10.1289/ehp.
results in cardiovascular defects. Dev. Dyn. 237, 153–162. 1408596.
Rayman, M.P., Bath, S.C., Westaway, J., Williams, P., Mao, J.Y., Vanderlelie, J.J., et al., Woodruff, T.J., Zota, A.R., Schwartz, J.M., 2011. Environmental chemicals in pregnant
2015. Selenium status in UK pregnant women and its relationship with hypertensive women in the United States: NHANES 2003–2004. Environ. Health Perspect. 119 (6),
conditions of pregnancy. Br. J. Nutr. 113, 249–258. http://dx.doi.org/10.1017/ 878–885. http://dx.doi.org/10.1289/ehp.1002727.
S000711451400364X. Yang, Q.W., Shu, W.S., Qiu, J.W., Wang, H.B., Lan, C.Y., 2004. Lead in paddy soils and
Sadler, T.W., 2004. Langman's Medical Embryology, 9th ed. Lippincott rice plants and its potential health risk around Lechang lead/zinc mine, Guangdong,
Williams & Wilkins, Baltimore. China. Environ. Int. 30 (7), 883–889. http://dx.doi.org/10.1016/j.envint.2004.02.
Steinberger, E.K., Ferencz, C., Loffredo, C.A., 2002. Infants with single ventricle: a 002.
population-based epidemiological study. Teratology 65 (3), 106–115. http://dx.doi. Zeng, X., Xu, X., Zheng, X., Reponen, T., Chen, A., Huo, X., 2016. Heavy metals in PM2.5
org/10.1002/tera.10017. and in blood, and children's respiratory symptoms and asthma from an e-waste
Thompson, J., Bannigan, J., 2008. Cadmium: toxic effects on the reproductive system and recycling area. Environ. Pollut. 210, 346–353. http://dx.doi.org/10.1016/j.envpol.
the embryo. Reprod. Toxicol. 25 (3), 304–315. http://dx.doi.org/10.1016/j.reprotox. 2016.01.025.
2008.02.001. Zheng, S., Song, H., Gao, H., Liu, C., Zhang, Z., Fu, J., 2016. The antagonistic effect of
Valinluck, V., Tsai, H.H., Rogstad, D.K., Burdzy, A., Bird, A., Sowers, L.C., 2004. Oxidative selenium on lead-induced inflammatory factors and heat shock protein mRNA level in
damage to methyl-CpG sequences inhibits the binding of the methyl-CpG binding chicken cartilage tissue. Biol. Trace Elem. Res. 173 (1), 177–184. http://dx.doi.org/
domain (MBD) of methyl-CpG binding protein 2 (MeCP2). Nucleic Acids Res. 32 (14), 10.1007/s12011-016-0630-6.
4100–4108. http://dx.doi.org/10.1093/nar/gkh739. Zierler, S., Theodore, M., Cohen, A., Rothman, K.J., 1988. Chemical quality of maternal
Wells, E.M., Herbstman, J.B., Lin, Y.H., Jarrett, J., Verdon, C.P., Ward, C., et al., 2016. drinking water and congenital heart disease. Int. J. Epidemiol. 17 (3), 589–594.
Cord blood methylmercury and fetal growth outcomes in Baltimore newborns: Zwolak, I., Zaporowska, H., 2012. Selenium interactions and toxicity: a review. Cell Biol.
potential confounding and effect modification by omega-3 fatty acids, selenium, and Toxicol. 28 (1), 31–46. http://dx.doi.org/10.1007/s10565-011-9203-9.

134

Anda mungkin juga menyukai