Skip to main content
Erschienen in: BMC Public Health 1/2017

Open Access 01.12.2017 | Research article

Maternal blood cadmium, lead and arsenic levels, nutrient combinations, and offspring birthweight

verfasst von: Yiwen Luo, Lauren E. McCullough, Jung-Ying Tzeng, Thomas Darrah, Avner Vengosh, Rachel L. Maguire, Arnab Maity, Carmen Samuel-Hodge, Susan K. Murphy, Michelle A. Mendez, Cathrine Hoyo

Erschienen in: BMC Public Health | Ausgabe 1/2017

Abstract

Background

Cadmium (Cd), lead (Pb) and arsenic (As) are common environmental contaminants that have been associated with lower birthweight. Although some essential metals may mitigate exposure, data are inconsistent. This study sought to evaluate the relationship between toxic metals, nutrient combinations and birthweight among 275 mother-child pairs.

Methods

Non-essential metals, Cd, Pb, As, and essential metals, iron (Fe), zinc (Zn), selenium (Se), copper (Cu), calcium (Ca), magnesium (Mg), and manganese (Mn) were measured in maternal whole blood obtained during the first trimester using inductively coupled plasma mass spectrometry. Folate concentrations were measured by microbial assay. Birthweight was obtained from medical records. We used quantile regression to evaluate the association between toxic metals and nutrients due to their underlying wedge-shaped relationship. Ordinary linear regression was used to evaluate associations between birth weight and toxic metals.

Results

After multivariate adjustment, the negative association between Pb or Cd and a combination of Fe, Se, Ca and folate was robust, persistent and dose-dependent (p < 0.05). However, a combination of Zn, Cu, Mn and Mg was positively associated with Pb and Cd levels. While prenatal blood Cd and Pb were also associated with lower birthweight. Fe, Se, Ca and folate did not modify these associations.

Conclusion

Small sample size and cross-sectional design notwithstanding, the robust and persistent negative associations between some, but not all, nutrient combinations with these ubiquitous environmental contaminants suggest that only some recommended nutrient combinations may mitigate toxic metal exposure in chronically exposed populations. Larger longitudinal studies are required to confirm these findings.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12889-017-4225-8) contains supplementary material, which is available to authorized users.
Abkürzungen
As
Arsenic
BMI
Body mass index
Cd
Cadmium
Ca
Calcium
Cu
Copper
ICP-MS
Inductively coupled plasma mass spectrometry
Fe
iron
Pb
Lead
LOD
Limits of detection
LOQ
Limits of quantification
Mg
magnesium
Mn
Manganese
NAN
Negative-association nutrient
PAN
Positive-association nutrient
Se
selenium
Zn
zinc

Background

Trace metals including cadmium (Cd), lead (Pb) and arsenic (As) are common environmental contaminants. Main routes of human exposure are ingestion and inhalation of contaminated dust and food [1]. These metals readily enter the food supply [2, 3], bioaccumulate, and target multiple organs. Exposure to these trace metals is implicated in cardiometabolic diseases, including cardiovascular diseases [4, 5], diabetes [4, 6], renal disorders [7, 8] and some types of cancer [913]. Some data suggest effects of these metals are sex- or race-specific [14, 15]. In both humans and experimental model systems, fetal exposure to these trace metals is associated with lower birthweight [1623] and shorter birth length [17, 19, 2430]. While non-specific, low birthweight is a consistent risk factor for obesity and cardiometabolic disease in adulthood [3133]. Strategies to reduce or eliminate toxic metal exposure could have widespread implications for fetal health and adult chronic disease.
Dietary manipulation together with environmental management [34], are recommended strategies to mitigate exposure and its downstream health effects in chronically exposed populations. For example, iron (Fe) deficiency in humans has been linked to elevated levels of Cd in both blood and urine, independent of smoking, poverty, age, race, obesity and parity [35]. Deficiencies in essential metals including Cu [36], Fe [3740], manganese (Mn) [41] and magnesium (Mg) [4244], and zinc (Zn) [45, 46] have been associated with increased risk of low birth weight and adiposity in some, but not all investigations [47, 48]. In animals, deficiencies in folate (a one-carbon cycle nutrient critical to the generation of S-adenosyl methionine, the universal methyl group donor) reduce As methylation and excretion [49]. This animal data is bolstered by epidemiologic data showing that one-carbon cycle supplementation is associated with re-methylation of interspersed repeat elements [50] and lower As levels in populations chronically exposed to As [51].
Collectively, these data suggest that dietary repletion with essential metals may alleviate trace metal exposure and/or effects. However, associations are inconsistent and, to date, most investigations have only evaluated individual nutrients without examining their combinations or accounting for the non-linear relationship between toxic metals and nutrients [52]. These analyses overcome somes of these limitations by: [1] evaluating the cross sectional relationship between Cd, Pb and As, and eight nutrients (e.g. Fe, selenium [Se], calcium [Ca], Mg, Mn, copper [Cu], Zn and folate), both independently and in combination; and [2] using quantile regression models to account for non-linear effects. We additionally examined whether maternal blood concentrations of either toxic metals or nutrient combinations are associated with offspring weight at birth.

Methods

Study participants

Study participants were English or Spanish speaking pregnant women whom, during an 18 month period, between April 2009 and October 2011, were aged ≥18-years of age and attending a prenatal clinic serving Duke University and Durham Regional obstetric facilities [53]. Of 2548 eligible women that were approached, 1700 (66.7%) consented. Women who declined study participation were more likely to be of Asian and Native American (p < 0.001) descent, but similar to enrolled women with respect to other characteristics. Cd, As and Pb were measured in whole peripheral blood of the first 310 women enrolled and the present analyses are limited to the 275 mother-infant pairs with complete exposure data for heavy metals, nutrients and birthweight. The distributions of covariates including maternal age at delivery, race/ethnicity, pre-pregnancy body mass index (BMI), education, smoking and gestational age at blood draw and at delivery were comparable in the 275 women and the larger cohort of 1700 (p values >0.05).

Data collection

At enrollment, all participants completed a self- or interviewer-administered questionnaire soliciting information on demographic, reproductive history, lifestyle behaviors, and anthropometric characteristics. Maternal peripheral blood samples were collected at enrollment at median gestational age 12 weeks (Inter-quantile range 8 – 14 weeks) in 10 mL EDTA-treated vaccutainer tubes from which one mL of whole blood was removed for use in these studies and stored at −80 °C. Upon delivery, parturition data were abstracted from medical records.

Measurement of folate and trace metals

Maternal concentrations of folate, Cd, Pb, As, Fe, Zn, Se, Cu, Ca, Mg and Mn were measured in maternal whole blood. Folate concentrations were measured using a commercial kit, ID-Vit Folic acid (Immundiagnostic-ALPCO; Salem, NH) [54]. Metal concentrations were measured as nanograms per gram (ng/g) using solution-based inductively coupled plasma mass spectrometry (ICP-MS) methods previously described [5557]. All standards, including aliquots of the certified NIST 955c, and procedural blanks were prepared by the same process. Maternal metal concentrations were measured using a Perkin Elmer DRC II (Dynamic Reaction Cell) axial field ICP-MS at GeoMed Analtyical [5558].

Quality control

To clean sample lines and reduce memory effects, sample lines were sequentially washed with 18.2 MΩ cm resistance (by a Milli-Q water purification system, Millipore, Bedford, Mass., USA) water for 90 s and a 2% nitric acid solution for 120 s between analyses. To monitor and correct for instrumental and procedural backgrounds, procedural blanks were analyzed within each block of 10 samples. Calibration standards included aliquots of 18.2 MΩ cm resistance H2O, NIST 955c SRM, and NIST 955c SRM spiked with known quantities of each metal in a linear range from 0.025 to 10 ng/g. Standards were prepared from 1000 mg/L single element standards obtained from SCP Science, USA. Method detection limits (MDLs) were calculated according to the two-step approach using the t99SLLMV method (USEPA, 1993) at 99% CI (t = 3.71). To facilitate comparisons with prior studies, trace metal concentrations were converted from ng/g to μg/dl based on blood density of 1.035g/ml. The MDLs yielded values of 0.006, 0.005, and 0.071 μg/dL, for Cd, Pb, and As, respectively. Limits of detection (LOD) were 0.002, 0.002, and 0.022 μg/dL, for Cd, Pb and As, respectively, and limits of quantification (LOQ) (according to Long and Winefordner, 1983) were 0.0007, 0.0006, and 0.0073 μg/dL for Cd, Pb, and As, respectively. The number of samples below the LOD for Cd, Pb, and As were 2, 2, and 1, respectively.

Assessment of birthweight and covariates

Parturition data were abstracted from medical records by trained personnel after delivery. These data included birthweight (grams), gestational age at birth (weeks) and infant sex (male/female). Infant birthweight was normally distributed and analyzed as a continuous variable. The median values and range for Cd and Pb varied in strata of several covariates. These covariates were explored as potential confounders in the associations between toxic metals and nutrients, as well as birthweight. Covariates included maternal age at delivery (<30, 30–35, and >35 years), race/ethnicity (White, African American, Hispanic), pre-pregnancy BMI (<30/≥30 kg/m2), education (< high school, high school graduates/GED, and college graduates), smoking status (non-smoker/smoker), infant sex (male/female), and gestational age at birth (<37/≥37 weeks).
If we observed significant differences in toxic metal levels, we considered these covariates independently as potential confounders. Each individual covariate was retained in the model if its inclusion changed the association under investigation by 10% or more. For analysis of association between birthweight and Cd, Pb or As, a global test was performed to detect significant interaction between these metals and covariates. A priori, we considered maternal smoking and infant sex as potential effect modifiers of the relationship between toxic metals and maternal nutrients, as well as birthweight. We therefore examined associations within strata of these two variables.

Statistical analyses

Quantile regression models were used to evaluate associations between Cd, Pb and As, individually and eight nutrient concentrations (Fe, Zn, Se, Cu, Ca, Mg, Mn, and total folate) adjusting for maternal delivery age, race/ethnicity, pre-pregnancy BMI, education and smoking. We considered quantile regression because we observed a wedge-shaped relationship between maternal toxic metal and nutrient concentrations (see Additional file 1: Figure S1), which suggested that the magnitude of associations may differ by toxic metal quantile. The quantile regression [59] provided the slope of each nutrient at different quantile levels (τ) of the toxic metal (i.e., τ = 0.1 to 0.9 by increments of 0.05), adjusting for other nutrients and covariates. For each nutrient, a global test was performed to examine if the nutrient was associated with the toxic metal at any quantile level, and the significance level was Bonferroni-corrected for multiple nutrients and toxic metals, i.e., 0.05/(3 toxic metals ×8 nutrients) = 0.002.
Because nutrients are taken in mixtures and are correlated (see Additional file 2: Table S1), we also considered the aggregate effect of nutrients by computing two multi-nutrient indices: the negative-association nutrient (NAN) measure and the positive-association nutrient (PAN) measure. The NAN index was computed as the sum of the standardized value of Fe, Se, Ca, and folate, which appeared to be negatively associated with toxic metals in the joint analysis. The PAN index was computed as the sum of standardized values of Zn, Cu, Mg and Mn, which appeared to be positively associated with toxic metals in the joint analysis. Standardization before summing assured that the nutrients were pooled together on a comparable scale. For each toxic metal, quantile regressions were conducted treating the toxic metal as the response variable and the two nutrients indices and other covariates as explanatory variables. All results were also Bonferroni-corrected (0.05/(3 toxic metals ×2 nutrient indices) = 0.008) to account for multiple comparisons.
Ordinary linear regression analysis was used to evaluate the association between toxic metals and birthweight, adjusted for maternal age at delivery, race/ethnicity, education, smoking status, pre-pregnancy BMI, gestational age at birth and infant sex, as well as the association between the two nutrient indices (NAN and PAN) and birthweight. Due to the potential non-linear relationship between birthweight and toxic metals (i.e., Additional file 3: Figure S2) and between birthweight and nutrient indices (i.e., Additional file 4: Figure S3), we categorized the toxic metal values and nutrient indices into low (i.e., concentration below the 33rd percentile), moderate (between the 33rd and 67th percentiles) and high (i.e., above the 67th percentile). In addition, because the exposures of Cd and Pb in the NEST cohort are geographically clustered with co-exposure to Cd and Pb [1] (but not among other toxic metals), we modeled the co-exposure effect of Cd and Pb by including the interaction terms between Cd and Pb. We also considered the interactions between toxic metals and nutrient indices to assess if the metal effects on birthweight can be modified by nutrients. Cigarette smoking is a major source of numerous toxic metals including Cd and Pb, and sex-specific effects have been hypothesized previously. Therefore, associations between Cd, Pb or As in relation to birthweight were evaluated in all participants and in strata of prenatal exposure to infant sex and cigarette smoke adjusted by other covariates [30, 60] (a priori p = 0.05). Due to the small number of smokers (n = 39), we did not estimate associations in this stratum.

Results

The distributions of individual toxic metals in strata of potential covariates are shown in Table 1. While there was some variation in the range of As by covariate, the median value remained comparable across covariates.
Table 1
Characteristics of the 275 study participants
Category
N
Cadmium (μg/dL) quantile: median [IQR*]
Lead (μg/dL) quantile: median [IQR*]
Arsenic (μg/dL) quantile: median [IQR*]
Maternal age at delivery
≤30
178
0.025 [0.012, 0.06]
0.345 [0.153, 0.811]
0.043 [0.038, 0.051]
(30, 35]
66
0.02 [0.01, 0.039]
0.299 [0.152, 0.726]
0.044 [0.039, 0.051]
>35
31
0.023 [0.009, 0.033]
0.517 [0.225, 1.407]
0.044 [0.04, 0.048]
Pre pregnancy BMI
≤30
201
0.023 [0.011, 0.053]
0.358 [0.161, 0.83]
0.044 [0.039, 0.052]
>30
74
0.025 [0.01, 0.042]
0.321 [0.153, 0.844]
0.044 [0.037, 0.049]
Education
College graduate
89
0.014 [0.009, 0.028]
0.275 [0.12, 0.718]
0.043 [0.038, 0.05]
High school graduate/GED
97
0.029 [0.015, 0.061]
0.268 [0.17, 0.663]
0.043 [0.038, 0.051]
<high school
89
0.026 [0.012, 0.098]
0.535 [0.235, 1.113]
0.045 [0.041, 0.053]
Smoking status
Non-smoker
235
0.019 [0.01, 0.035]
0.341 [0.15, 0.779]
0.044 [0.038, 0.051]
Smoker
40
0.051 [0.035, 0.1]
0.404 [0.177, 1.304]
0.045 [0.04, 0.056]
Race/Ethnicity
Black
104
0.031 [0.015, 0.059]
0.339 [0.162, 0.761]
0.043 [0.037, 0.051]
Hispanic
89
0.02 [0.01, 0.09]
0.517 [0.227, 1.105]
0.044 [0.041, 0.051]
White
82
0.016 [0.009, 0.029]
0.242 [0.126, 0.716]
0.045 [0.038, 0.051]
Infant sex
Male
132
0.019 [0.01, 0.041]
0.367 [0.145, 0.864]
0.044 [0.039, 0.051]
Female
143
0.026 [0.012, 0.053]
0.337 [0.178, 0.809]
0.044 [0.038, 0.051]
Gestational age at delivery
<37
25
0.023 [0.009, 0.035]
0.451 [0.151, 0.954]
0.047 [0.043, 0.05]
> = 37
250
0.024 [0.012, 0.047]
0.341 [0.158, 0.817]
0.043 [0.038, 0.051]
*IQR: interquartile range

Association between toxic metals and individual nutrients

Quantile regression coefficient estimations (τ), along with their 95% simultaneous confidence bands [59] adjusted for other nutrients and maternal age, race/ethnicity, pre-pregnancy BMI, education, and smoking, are shown in Fig. 1. We found significant negative associations between Cd and Fe and folate concentrations, and positive associations with Cu and Mn. With the exception of Mn and folate, most regression coefficients were significant in the middle range of Cd. Similarly, negative associations were also found between Pb concentrations and Fe, Ca and folate, while associations with Zn, Cu and Mn were positive. In general, regression coefficients were significant in the upper quantiles of Pb for Fe, Cu and Ca. In contrast, coefficients for folate and Mn were significant in a limited range. For As, we found significant negative associations with Se and folate at only one quantile point, and significant positive associations with Zn and Cu. Coefficients of Se remained relatively constant across quantiles of As, while Zn, Cu and folate had slope estimates further away from zero in upper quantiles of As exposure.

Association between toxic metals and nutrient mixtures

The last column of Fig. 1 shows quantile regression for the sum of standardized negatively and positively associated indices, NAN and PAN, respectively, in relation to Cd, Pb and As. Cd levels were significantly associated with both nutrition indices (p-values <0.0002). In contrast to individual nutrients, where associations were observed for some range of quantiles (i.e. for Fe, τ = 0.55 , 0.7 ~ 0.85), associations with the nutrient index remained significant for most of the quantiles. Similarly, Pb levels were significantly associated with both NAN and PAN indices (p-values <0.0002), and the associations remained consistent for all quantiles of Pb. We also observed that As levels remained significantly negatively associated with the NAN index only in a limited range (τ = 0.1 ~ 0.15 , 0.4 ~ 0.75) and were no longer significant at the highest quintiles.

Association between toxic metals, nutrient mixtures and birthweight

Overall, medium level of As exposure were also associated with lower birthweight in all subjects (Table 2A), male infants (Table 2B) and non-smokers (Table 2D). In addition, high level of Cd was found associated with lower birthweight in male infants (Table 2B). In male-only analyses, we also observed significant interaction effects between Cd and PAN as well as between As and PAN. These interaction effects suggested that higher level of PAN might reduce the magnitude of birthweight loss in male infants compared to the baseline levels (i.e.,. low Cd and low PAN and low As and low PAN, respectively).
Table 2
Regression coefficients for the associations between toxic metal concentrations and birth weight in males, females and non-smokers
 
A. All data (n = 272)
B. Male (n = 130)
C. Female (n = 142)
D. Non-smokers (n = 233)
Caharacteristic
Estimate (Std. Error)
P-value
Estimate (Std. Error)
P-value
Estimate (Std. Error)
P-value
Estimate (Std. Error)
P-value
(Intercept)
−4721.71 * (693.49)
<0.001
−3782.55 (1161.8)
0.002
−4546.38 (1075.15)
<0.001
−4251.48 (869.47)
<0.001
Moderate Cd
−125.97 (160.05)
0.432
−264.59 (264.87)
0.321
−70.98 (247.9)
0.775
−111.12 (172.28)
0.520
High Cd
−382.74 (210.06)
0.070
−812.25 (346.1)
0.021
−201.44 (356.57)
0.573
−290.2 (264.06)
0.273
Moderate Pb
−68.42 (160.65)
0.671
−225.21 (359.43)
0.533
35.51 (199.73)
0.859
−22.5 (173.71)
0.897
High Pb
−120.42 (200.95)
0.550
11.01 (395.46)
0.978
−58.88 (262.59)
0.823
−191.24 (223.77)
0.394
Moderate As
−366.5 (175.2)
0.038
−870.69 (372.09)
0.022
−180.59 (218.97)
0.412
−464.29 (202.74)
0.023
High As
72.82 (139.83)
0.603
−310.42 (242.22)
0.204
199.52 (195.24)
0.309
163.61 (161.11)
0.311
Moderate Cd:oderate Pb
68.5 (164.92)
0.678
3.27 (290.12)
0.991
23.69 (254.14)
0.926
95.5 (176.05)
0.588
High Cd:Moderate Pb
129 (200.79)
0.521
477.08 (428.43)
0.269
−63.06 (312.3)
0.84
−68.99 (277.85)
0.804
Moderate Cd:High Pb
403.84 (226.22)
0.076
65.54 (368.9)
0.859
785.05 (363.23)
0.033
322.81 (251.94)
0.202
High Cd:High Pb
113.2 (220.94)
0.609
47.79 (413.45)
0.908
345.83 (357.83)
0.336
−68.44 (301.77)
0.821
Moderate NAN
69.17 (171.44)
0.687
−272.91 (274.51)
0.323
309.38 (332.21)
0.354
−24.77 (187.92)
0.895
High NAN
133.53 (179.46)
0.458
62.14 (319.67)
0.846
48.16 (248.6)
0.847
104.1 (192.18)
0.589
Moderate PAN
−106.41 (173.19)
0.540
−527.03 (279.37)
0.063
154.08 (286.47)
0.592
−16.12 (188.07)
0.932
High PAN
−306.32 (196.91)
0.121
−736.08 (353.06)
0.04
−171.63 (276.41)
0.536
−271.49 (213.82)
0.206
Moderate Cd:Moderate NAN
−15.56 (187.42)
0.934
−83.86 (330.37)
0.8
−52.55 (276.51)
0.85
−19.69 (205.13)
0.924
High Cd: Moderate NAN
103.44 (190.11)
0.587
433.38 (344.66)
0.212
−114.8 (278.45)
0.681
220.18 (219.16)
0.316
Moderate Cd:High NAN
99.37 (213.17)
0.642
−197.68 (361.34)
0.586
317.79 (318.09)
0.320
42.21 (230.92)
0.855
High Cd:High NAN
170.97 (226.96)
0.452
316.61 (462.66)
0.496
77.43 (309.97)
0.803
190.84 (278.84)
0.495
Moderate Cd:Moderate PAN
55.91 (194.67)
0.774
307.21 (334.27)
0.361
12.31 (270.51)
0.964
63.2 (212.11)
0.766
High Cd:Moderate PAN
361.59 (188.45)
0.056
368.06 (366.16)
0.318
458.2 (281.48)
0.107
456.64 (233.05)
0.052
Moderate Cd:High PAN
202.64 (230.04)
0.379
894.08 (404.05)
0.030
−155.77 (322.56)
0.630
263.34 (250.81)
0.295
High Cd:High PAN
477.25 (229.66)
0.039
711.43 (445.54)
0.114
481.47 (334.96)
0.154
449.77 (286.85)
0.119
Moderate Pb:Moderate NAN
−187.78 (187.41)
0.317
170.59 (340.91)
0.618
−351.75 (296.46)
0.238
−224.7 (210.09)
0.286
High Pb:Moderate NAN
−153.04 (222.77)
0.493
−131.75 (396.94)
0.741
−542.56 (339.57)
0.113
−145.29 (255.15)
0.570
Moderate Pb:High NAN
−126.16 (204.89)
0.539
118.2 (388.7)
0.762
−92.68 (292.05)
0.752
−125.5 (231.1)
0.588
High Pb:High NAN
6.28 (235.71)
0.979
−194.48 (469.11)
0.68
−142.03 (353.1)
0.688
3.89 (282.4)
0.989
Moderate Pb:Moderate PAN
30.45 (179.31)
0.865
43.46 (340.04)
0.899
−124.46 (281.42)
0.659
−43.8 (206)
0.832
High Pb:Moderate PAN
−109.95 (226.29)
0.628
21.11 (404.4)
0.958
−210.73 (364.93)
0.565
−62.16 (267.16)
0.816
Moderate Pb:High PAN
210.19 (199.96)
0.294
200.26 (365.37)
0.585
266.68 (278.08)
0.340
210.73 (223.78)
0.348
High Pb:High PAN
−236.39 (263.92)
0.371
11.09 (464.4)
0.981
−416.52 (406.03)
0.308
−68.75 (321.64)
0.831
Moderate As:Moderate NAN
131.74 (201.75)
0.514
284.7 (390.7)
0.468
34.85 (275.87)
0.900
313.29 (229.55)
0.174
High As:Moderate NAN
−272.79 (213.48)
0.203
−274.26 (368.65)
0.459
−199.63 (316.75)
0.530
−243.52 (263.43)
0.356
Median As:High NAN
42.14 (236.38)
0.859
215.27 (442.65)
0.628
5.1 (326.28)
0.988
202.7 (273.88)
0.46
High As:High NAN
−430.35 (210.5)
0.042
−548.28 (352.69)
0.124
−163.32 (326.11)
0.618
−464.49 (236.44)
0.051
Median As:Median PAN
177.57 (180.43)
0.326
683.69 (326.62)
0.039
24.95 (251.2)
0.921
162.32 (201.93)
0.423
High As:Median PAN
81.74 (202.68)
0.687
710.58 (315.78)
0.027
−394.93 (305.35)
0.199
7.11 (244.53)
0.977
Median As:High PAN
195.33 (218.9)
0.373
294.86 (394.46)
0.457
206.5 (318.7)
0.519
119.09 (244.02)
0.626
High As:High PAN
158.09 (231.82)
0.496
435.1 (411.65)
0.294
−23.33 (326.51)
0.943
63.95 (256.37)
0.803
*: Coefficients that are statistically significant are shown in bold. Negative-association nutrient (NAN), obtained by the sum of the standardized values of Fe, Se, Ca and total folate. Positive-association nutrient (PAN), obtained by the sum of the standardized values of Zn, Cu, Mg and Mn. Models adjusted for maternal age, ethnicity, cigarette smoking, educational attainment, gestational age at delivery and blood draw and sex
There were also significant metal and nutrient interacitons observed in all-subject analysis, including between Cd and PAN (where higher PAN levels reduced the magnitude of birthweight loss) as well as between As and NAN (for which, though high As and high NAN individually are positively associated with birthweight, the existence of both leads to birthweight loss).
We did not find significant co-exposure effects between Pb and Cd except in the female-only analysis (Table 2C), where we observed that female infants with moderate Cd and high Pb exposure tend to have higher birthweight compared to the baseline group of low Cd and low Pb.

Discussion

We evaluated associations between Cd, Pb and As, and eight nutrients in pregnant women, and determined if exposure to these metals were associated with lower birthweight. We found that higher levels of a combination comprising Fe, Ca, Se and folate was robustly related to lower Pb and Cd, regardless of concentration of these toxic metals, whereas the negative relationships with single nutrients were within very narrow ranges of exposure. Surprisingly, higher levels of Cu, Zn, Mg and Mn were associated with higher levels of Pb and Cd. With the exception of recent data supporting positive associations between Mn and Cd [61], these findings are despite accumulated animal data and human single nutrient data to the contrary. Finally, exposure to Cd and As were negatively associated with birthweight as would be expected; but these associations were modified by select nutrient combinations. These findings are consistent with previous studies demonstrating that dietary repletion with Fe, Ca, Se and folate can mitigate exposure to Cd and Pb. However, our findings suggesting that Cu, Zn, Mg and Mn are positively associated with Cd and Pb levels contrast with previous studies that form the basis for existing intervention guidelines as they suggest these minerals may exacerbate exposure.
Our data showing negative, but weak and inconsistent associations between Pb, As or Cd and single nutrients are consistent with these divalent cations Fe2+ or Ca2+ competitively displacing Cd2+ in transmetallation reactions. In animals, Fe reduces intestinal absorption of Cd [35]. These animal data are corroborated by human studies showing that Fe-deficiency is consistently associated with higher blood and urine Cd levels, independent of smoking, poverty, age, race, obesity and parity [35]. Our data however contrast with in vitro evidence that show >50% lower Cd levels with Zn and Mn treatment, and negative relationships between Zn, Cu and Mg with Cd [62]. These data provide early evidence that some essential metals, including Mn [63] may not mitigate exposure and effects of these metals. Specific mineral combinations that mitigate exposure may depend on the underlying nutritional status and may therefore vary by population exposed to Pb or Cd.
One-carbon cycle nutrients are critical in generating S-adenosyl methionine, key in the clearance of As [64]. In mice [65] and humans [6669], higher circulating one-carbon nutrients are associated with a lower body burden of inorganic As. Depletion of one-carbon cycle nutrients reduces As excretion and methylation [49, 70], whereas their repletion lowers blood As levels [51]. In 6-year old As-exposed children, one-carbon cycle supplementation was associated with re-methylation of interspersed repeat elements [50] and lower As levels [51]. Although fewer studies have been conducted on Pb or Cd, negative associations with folate or vitamin B12 have been reported [71]. Our data provides evidence for negative associations between folate in combination with Fe, Ca and Se, and Cd or Pb levels.
Consistent with previous studies, we also found that prenatal exposure to Cd or As is associated with lower birthweight [1620]. This adverse birth outcome increases the risk of rapid adiposity gain in young children; a consistent risk factor for cardiometabolic impairment in adulthood [7278]. In adult cross-sectional studies, elevated Cd, Pb and As have been associated with cardiometabolic risk markers [7985]. While metabolic syndrome is not clinically discernible in young children [86], individual cardiometabolic risk markers that include central adiposity, elevated systolic blood pressure and elevated levels of fasting insulin and/or glucose, triglycerides, cholesterol, accelerated adiposity, [8791, 7678, 9295] sometimes without overt obesity [96], predict metabolic syndrome, atherosclerosis, diabetes and hypertension in adulthood [97104]. While not confirmed by others [47], increased adiposity and higher insulin have been associated with deficiency of Cu [36], Fe [3740], Mn [41], Mg [4244] and Zn [45, 46]. Thus, the low birthweight that is associated with early exposure to these toxic metals supports the developmental origins of these cardiometabolic diseases, and may portend, and/or contribute to the increase in incidence of these diseases.
While these data suggest that some nutrients may mitigate toxic metal exposure and effects, mechanisms are poorly understood. In animal and in vitro models, ferritin reduces intestinal absorption of Cd [35] and Pb. Mn also shares transporters with Cd and Pb in vivo and in vitro. Cd induces oxidative stress via decreasing cellular antioxidant capacity, increases lipid peroxidation, and depletes glutathione and protein-bound sulfhydryl groups [105107]. Because the target organs for these metals such as the kidney and liver also play a critical role in the maintenance of blood glucose levels [108111], associations between these toxic metals and low birthweight that is often followed by accelerated adiposity gains and insulin resistance, are to be expected.
Our findings should be interpreted in the context of the study limitations. The relatively small sample size limited our ability to assess subgroup effects, and particularly higher order interactions (e.g. by combined maternal smoking, nutrient combinations and race/ethnicity), which may inform public health interventions. In addition, while we anticipate that some nutrient combinations may mitigate toxic metal exposure, cause-and-effect cannot be inferred in this cross-sectional study. Furthemore, we assessed maternal nutrient concentrations at a single time-point, yet these nutritional markers may vary throughout gestation, as the physiologic changes that occur during pregnancy may impact levels of toxic metals in maternal blood. For example, the increase in erythrocytes and plasma with advancing gestation may lower levels, presumably due to hemodilution, whereas essential elements may increase, in part due to increased supplementation. In support, a Canadian study reported that while Cd levels did not change, Pb levels decreased over the course of pregnancy, while Mn levels increased during the same period [63]. Thus, given our specimens were collected during a short gestational window (median 12 weeks, IQR 8–14 weeks), the full effect of toxic metal and nutrients on birthweight may only be partially realized. However, sensitivity analyses showed that restricting analysis to women with gestational age > 14 weeks at blood draw did not alter our findings. Despite these limitations, our study has several strengths including the multiethnic composition of the cohort and our ability to examine multiple nutrients to mitigate exposure and effects very early in gestation, when many metabolic set points are established.

Conclusions

In summary, we provide early data suggesting that a combination of Fe, Ca, Se and folate is negatively associated with Cd and As exposure, while Cu, Zn, Mg and Mn may exacerbate exposure to these toxic compounds. We also confirmed negative associations between birthweight and toxic compounds, Cd and As. Larger studies are required to identify other nutrients, which in combination, may mitigate exposure to these ubiquitous toxic metals in exposed populations.

Acknowledgments

We thank participants of the NEST study project. We also acknowledge Stacy Murray, Kennetra Irby, Siobhan Greene and Anna Tsent for their recruiting efforts and Carole Grenier, Erin Erginer for specimen handling.

Funding

This work was supported in part by the generous donation from Howard and Julia Clark, and research grants from the National Institute of Health (Grant no. R01-ES016772, P30 ES025128) and National Cancer Institute (R25CA057726). The funders had no role in the study design; in the collection, analysis and interpretation of data; in the writing of the report; or in the decision to submit the article for publication.

Availability of data and materials

Data are not publically available but may be requested through the corresponding author.

Authors’ contributions

CH, JYT, YL, LM developed the research question. TD, AV, RM, SM, CH provided resources and data to support these analyses. YL, JYT, AM performed the statistical analyses. LM, YL, CSH, JYT, MM, CH were responsible for data intrpretation. LM, YL drafted the manuscript. All authors read and approved the final manuscript.

Competing interests

The authors declare they have no competing interest.
Not applicable.
The study protocol was approved by the Institutional Review Boards of Duke University, Durham Regional Hospital and North Carolina State University. All participants were informed of the study aims, benefits and potential limitations and provided written consent prior to participation.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat King KE, Darrah TH, Money E, et al. Geographic clustering of elevated blood heavy metal levels in pregnant women. BMC Public Health. 2015;15:1035.PubMedPubMedCentralCrossRef King KE, Darrah TH, Money E, et al. Geographic clustering of elevated blood heavy metal levels in pregnant women. BMC Public Health. 2015;15:1035.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Satarug S, Moore MR. Adverse health effects of chronic exposure to low-level cadmium in foodstuffs and cigarette smoke. Environ Health Perspect. 2004;112:1099–103.PubMedPubMedCentralCrossRef Satarug S, Moore MR. Adverse health effects of chronic exposure to low-level cadmium in foodstuffs and cigarette smoke. Environ Health Perspect. 2004;112:1099–103.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Satarug S, Moore MR. Emerging roles of cadmium and heme oxygenase in type-2 diabetes and cancer susceptibility. Tohoku J Exp Med. 2012;228:267–88.PubMedCrossRef Satarug S, Moore MR. Emerging roles of cadmium and heme oxygenase in type-2 diabetes and cancer susceptibility. Tohoku J Exp Med. 2012;228:267–88.PubMedCrossRef
4.
Zurück zum Zitat Agarwal S, Zaman T, Tuzcu EM, Kapadia SR. Heavy metals and cardiovascular disease: results from the National Health and nutrition examination survey (NHANES) 1999–2006. Angiology. 2011;62:422–9.PubMedCrossRef Agarwal S, Zaman T, Tuzcu EM, Kapadia SR. Heavy metals and cardiovascular disease: results from the National Health and nutrition examination survey (NHANES) 1999–2006. Angiology. 2011;62:422–9.PubMedCrossRef
5.
Zurück zum Zitat Tellez-Plaza M, Guallar E, Howard BV, Navas-Acien A. Cadmium and cardiovascular risk. Epidemiology. 2013;24:784–5.PubMedCrossRef Tellez-Plaza M, Guallar E, Howard BV, Navas-Acien A. Cadmium and cardiovascular risk. Epidemiology. 2013;24:784–5.PubMedCrossRef
6.
Zurück zum Zitat Chen L, Lei L, Jin T, Nordberg M, Nordberg GF. Plasma metallothionein antibody, urinary cadmium, and renal dysfunction in a Chinese type 2 diabetic population. Diabetes Care. 2006;29:2682–7.PubMedCrossRef Chen L, Lei L, Jin T, Nordberg M, Nordberg GF. Plasma metallothionein antibody, urinary cadmium, and renal dysfunction in a Chinese type 2 diabetic population. Diabetes Care. 2006;29:2682–7.PubMedCrossRef
7.
Zurück zum Zitat Huang M, Choi SJ, Kim DW, et al. Evaluation of factors associated with cadmium exposure and kidney function in the general population. Environ Toxicol. 2013;28:563–70.PubMedCrossRef Huang M, Choi SJ, Kim DW, et al. Evaluation of factors associated with cadmium exposure and kidney function in the general population. Environ Toxicol. 2013;28:563–70.PubMedCrossRef
8.
Zurück zum Zitat Liang Y, Lei L, Nilsson J, et al. Renal function after reduction in cadmium exposure: an 8-year follow-up of residents in cadmium-polluted areas. Environ Health Perspect. 2012;120:223–8.PubMedCrossRef Liang Y, Lei L, Nilsson J, et al. Renal function after reduction in cadmium exposure: an 8-year follow-up of residents in cadmium-polluted areas. Environ Health Perspect. 2012;120:223–8.PubMedCrossRef
10.
Zurück zum Zitat Christensen BC, Marsit CJ, Houseman EA, et al. Differentiation of lung adenocarcinoma, pleural mesothelioma, and nonmalignant pulmonary tissues using DNA methylation profiles. Cancer Res. 2009;69:6315–21.PubMedPubMedCentralCrossRef Christensen BC, Marsit CJ, Houseman EA, et al. Differentiation of lung adenocarcinoma, pleural mesothelioma, and nonmalignant pulmonary tissues using DNA methylation profiles. Cancer Res. 2009;69:6315–21.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Gallagher CM, Chen JJ, Kovach JS. Environmental cadmium and breast cancer risk. Aging (Albany NY). 2010;2:804–14.CrossRef Gallagher CM, Chen JJ, Kovach JS. Environmental cadmium and breast cancer risk. Aging (Albany NY). 2010;2:804–14.CrossRef
12.
Zurück zum Zitat Benbrahim-Tallaa L, Tokar EJ, Diwan BA, Dill AL, Coppin JF, Waalkes MP. Cadmium malignantly transforms normal human breast epithelial cells into a basal-like phenotype. Environ Health Perspect. 2009;117:1847–52.PubMedPubMedCentralCrossRef Benbrahim-Tallaa L, Tokar EJ, Diwan BA, Dill AL, Coppin JF, Waalkes MP. Cadmium malignantly transforms normal human breast epithelial cells into a basal-like phenotype. Environ Health Perspect. 2009;117:1847–52.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Tokar EJ, Benbrahim-Tallaa L, Waalkes MP. Metal ions in human cancer development. Met Ions Life Sci. 2011;8:375–401.PubMed Tokar EJ, Benbrahim-Tallaa L, Waalkes MP. Metal ions in human cancer development. Met Ions Life Sci. 2011;8:375–401.PubMed
14.
Zurück zum Zitat Ferraro PM, Sturniolo A, Naticchia A, D'Alonzo S, Gambaro G. Temporal trend of cadmium exposure in the United States population suggests gender specificities. Intern Med J. 2012;42:691–7.PubMedCrossRef Ferraro PM, Sturniolo A, Naticchia A, D'Alonzo S, Gambaro G. Temporal trend of cadmium exposure in the United States population suggests gender specificities. Intern Med J. 2012;42:691–7.PubMedCrossRef
15.
Zurück zum Zitat Scinicariello F, Abadin HG, Murray HE. Association of low-level blood lead and blood pressure in NHANES 1999–2006. Environ Res. 2011;111:1249–57.PubMedCrossRef Scinicariello F, Abadin HG, Murray HE. Association of low-level blood lead and blood pressure in NHANES 1999–2006. Environ Res. 2011;111:1249–57.PubMedCrossRef
16.
Zurück zum Zitat Nishijo M, Nakagawa H, Honda R, et al. Effects of maternal exposure to cadmium on pregnancy outcome and breast milk. Occup Environ Med. 2002;59:394–6. discussion 7PubMedPubMedCentralCrossRef Nishijo M, Nakagawa H, Honda R, et al. Effects of maternal exposure to cadmium on pregnancy outcome and breast milk. Occup Environ Med. 2002;59:394–6. discussion 7PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Nishijo M, Tawara K, Honda R, Nakagawa H, Tanebe K, Saito S. Relationship between newborn size and mother's blood cadmium levels, Toyama, Japan. Arch Environ Health. 2004;59:22–5.PubMedCrossRef Nishijo M, Tawara K, Honda R, Nakagawa H, Tanebe K, Saito S. Relationship between newborn size and mother's blood cadmium levels, Toyama, Japan. Arch Environ Health. 2004;59:22–5.PubMedCrossRef
18.
Zurück zum Zitat Salpietro CD, Gangemi S, Minciullo PL, et al. Cadmium concentration in maternal and cord blood and infant birth weight: a study on healthy non-smoking women. J Perinat Med. 2002;30:395–9.PubMedCrossRef Salpietro CD, Gangemi S, Minciullo PL, et al. Cadmium concentration in maternal and cord blood and infant birth weight: a study on healthy non-smoking women. J Perinat Med. 2002;30:395–9.PubMedCrossRef
19.
Zurück zum Zitat Ronco AM, Arguello G, Munoz L, Gras N, Llanos M. Metals content in placentas from moderate cigarette consumers: correlation with newborn birth weight. Biometals. 2005;18:233–41.PubMedCrossRef Ronco AM, Arguello G, Munoz L, Gras N, Llanos M. Metals content in placentas from moderate cigarette consumers: correlation with newborn birth weight. Biometals. 2005;18:233–41.PubMedCrossRef
20.
Zurück zum Zitat Fei DL, Koestler DC, Li Z, et al. Association between in Utero arsenic exposure, placental gene expression, and infant birth weight: a US birth cohort study. Environmental health: a global access science source. 2013;12:58.CrossRef Fei DL, Koestler DC, Li Z, et al. Association between in Utero arsenic exposure, placental gene expression, and infant birth weight: a US birth cohort study. Environmental health: a global access science source. 2013;12:58.CrossRef
21.
Zurück zum Zitat Rodosthenous RS, Burris HH, Svensson K, et al. Prenatal lead exposure and fetal growth: smaller infants have heightened susceptibility. Environ Int. 2017;99:228–33.PubMedCrossRef Rodosthenous RS, Burris HH, Svensson K, et al. Prenatal lead exposure and fetal growth: smaller infants have heightened susceptibility. Environ Int. 2017;99:228–33.PubMedCrossRef
22.
Zurück zum Zitat Jedrychowski W, Perera F, Jankowski J, et al. Gender specific differences in neurodevelopmental effects of prenatal exposure to very low-lead levels: the prospective cohort study in three-year olds. Early Hum Dev. 2009;85:503–10.PubMedPubMedCentralCrossRef Jedrychowski W, Perera F, Jankowski J, et al. Gender specific differences in neurodevelopmental effects of prenatal exposure to very low-lead levels: the prospective cohort study in three-year olds. Early Hum Dev. 2009;85:503–10.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Kaji M, Nishi Y. Lead and growth. Clinical pediatric endocrinology : case reports and clinical investigations: official journal of the Japanese Society for Pediatric Endocrinology. 2006;15:123–8. Kaji M, Nishi Y. Lead and growth. Clinical pediatric endocrinology : case reports and clinical investigations: official journal of the Japanese Society for Pediatric Endocrinology. 2006;15:123–8.
24.
Zurück zum Zitat Al-Saleh I, Shinwari N, Mashhour A, Rabah A. Birth outcome measures and maternal exposure to heavy metals (lead, cadmium and mercury) in Saudi Arabian population. Int J Hyg Environ Health. 2014;217:205–18.PubMedCrossRef Al-Saleh I, Shinwari N, Mashhour A, Rabah A. Birth outcome measures and maternal exposure to heavy metals (lead, cadmium and mercury) in Saudi Arabian population. Int J Hyg Environ Health. 2014;217:205–18.PubMedCrossRef
25.
Zurück zum Zitat Gundacker C, Frohlich S, Graf-Rohrmeister K, et al. Perinatal lead and mercury exposure in Austria. Sci Total Environ. 2010;408:5744–9.PubMedCrossRef Gundacker C, Frohlich S, Graf-Rohrmeister K, et al. Perinatal lead and mercury exposure in Austria. Sci Total Environ. 2010;408:5744–9.PubMedCrossRef
26.
Zurück zum Zitat Jones L, Parker JD, Mendola P. Blood lead and mercury levels in pregnant women in the United States, 2003–2008. NCHS data brief. 2010;52:1–8. Jones L, Parker JD, Mendola P. Blood lead and mercury levels in pregnant women in the United States, 2003–2008. NCHS data brief. 2010;52:1–8.
27.
Zurück zum Zitat Koppen G, Den Hond E, Nelen V, et al. Organochlorine and heavy metals in newborns: results from the Flemish environment and health survey (FLEHS 2002–2006). Environ Int. 2009;35:1015–22.PubMedCrossRef Koppen G, Den Hond E, Nelen V, et al. Organochlorine and heavy metals in newborns: results from the Flemish environment and health survey (FLEHS 2002–2006). Environ Int. 2009;35:1015–22.PubMedCrossRef
28.
Zurück zum Zitat Piasek M, Blanusa M, Kostial K, Laskey JW. Placental cadmium and progesterone concentrations in cigarette smokers. Reproductive toxicology (Elmsford, NY). 2001;15:673–81.CrossRef Piasek M, Blanusa M, Kostial K, Laskey JW. Placental cadmium and progesterone concentrations in cigarette smokers. Reproductive toxicology (Elmsford, NY). 2001;15:673–81.CrossRef
29.
Zurück zum Zitat Shirai S, Suzuki Y, Yoshinaga J, Mizumoto Y. Maternal exposure to low-level heavy metals during pregnancy and birth size. J Environ Sci Health A Tox Hazard Subst Environ Eng. 2010;45:1468–74.PubMedCrossRef Shirai S, Suzuki Y, Yoshinaga J, Mizumoto Y. Maternal exposure to low-level heavy metals during pregnancy and birth size. J Environ Sci Health A Tox Hazard Subst Environ Eng. 2010;45:1468–74.PubMedCrossRef
30.
Zurück zum Zitat Faulk C, Barks A, Sanchez BN, et al. Perinatal lead (Pb) exposure results in sex-specific effects on food intake, fat, weight, and insulin response across the Murine life-course. PLoS One. 2014;9:e104273.PubMedPubMedCentralCrossRef Faulk C, Barks A, Sanchez BN, et al. Perinatal lead (Pb) exposure results in sex-specific effects on food intake, fat, weight, and insulin response across the Murine life-course. PLoS One. 2014;9:e104273.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Ogden CL, Carroll MD, Flegal KM. High body mass index for age among US children and adolescents, 2003–2006. JAMA. 2008;299:2401–5.PubMedCrossRef Ogden CL, Carroll MD, Flegal KM. High body mass index for age among US children and adolescents, 2003–2006. JAMA. 2008;299:2401–5.PubMedCrossRef
32.
Zurück zum Zitat Ong KK. Catch-up growth in small for gestational age babies: good or bad? Curr Opin Endocrinol Diabetes Obes. 2007;14:30–4.PubMedCrossRef Ong KK. Catch-up growth in small for gestational age babies: good or bad? Curr Opin Endocrinol Diabetes Obes. 2007;14:30–4.PubMedCrossRef
33.
Zurück zum Zitat Ong KK, Ahmed ML, Emmett PM, Preece MA, Dunger DB. Association between postnatal catch-up growth and obesity in childhood: prospective cohort study. BMJ. 2000;320:967–71.PubMedPubMedCentralCrossRef Ong KK, Ahmed ML, Emmett PM, Preece MA, Dunger DB. Association between postnatal catch-up growth and obesity in childhood: prospective cohort study. BMJ. 2000;320:967–71.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Hu H, Kotha S, Brennan T. The role of nutrition in mitigating environmental insults: policy and ethical issues. Environ Health Perspect. 1995;103(Suppl 6):185–90.PubMedPubMedCentralCrossRef Hu H, Kotha S, Brennan T. The role of nutrition in mitigating environmental insults: policy and ethical issues. Environ Health Perspect. 1995;103(Suppl 6):185–90.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Gallagher CM, Chen JJ, Kovach JS. The relationship between body iron stores and blood and urine cadmium concentrations in US never-smoking, non-pregnant women aged 20–49 years. Environ Res. 2011;111:702–7.PubMedCrossRef Gallagher CM, Chen JJ, Kovach JS. The relationship between body iron stores and blood and urine cadmium concentrations in US never-smoking, non-pregnant women aged 20–49 years. Environ Res. 2011;111:702–7.PubMedCrossRef
36.
Zurück zum Zitat Wildman RE, Mao S. Tissue-specific alterations in lipoprotein lipase activity in copper-deficient rats. Biol Trace Elem Res. 2001;80:221–9.PubMedCrossRef Wildman RE, Mao S. Tissue-specific alterations in lipoprotein lipase activity in copper-deficient rats. Biol Trace Elem Res. 2001;80:221–9.PubMedCrossRef
37.
Zurück zum Zitat Bourque SL, Komolova M, Nakatsu K, Adams MA. Long-term circulatory consequences of perinatal iron deficiency in male Wistar rats. Hypertension. 2008;51:154–9.PubMedCrossRef Bourque SL, Komolova M, Nakatsu K, Adams MA. Long-term circulatory consequences of perinatal iron deficiency in male Wistar rats. Hypertension. 2008;51:154–9.PubMedCrossRef
38.
Zurück zum Zitat Komolova M, Bourque SL, Nakatsu K, Adams MA. Sedentariness and increased visceral adiposity in adult perinatally iron-deficient rats. International journal of obesity (2005). 2008;32:1441–4.CrossRef Komolova M, Bourque SL, Nakatsu K, Adams MA. Sedentariness and increased visceral adiposity in adult perinatally iron-deficient rats. International journal of obesity (2005). 2008;32:1441–4.CrossRef
39.
Zurück zum Zitat Menzie CM, Yanoff LB, Denkinger BI, et al. Obesity-related hypoferremia is not explained by differences in reported intake of heme and nonheme iron or intake of dietary factors that can affect iron absorption. J Am Diet Assoc. 2008;108:145–8.PubMedPubMedCentralCrossRef Menzie CM, Yanoff LB, Denkinger BI, et al. Obesity-related hypoferremia is not explained by differences in reported intake of heme and nonheme iron or intake of dietary factors that can affect iron absorption. J Am Diet Assoc. 2008;108:145–8.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Yanoff LB, Menzie CM, Denkinger B, et al. Inflammation and iron deficiency in the hypoferremia of obesity. International journal of obesity (2005). 2007;31:1412–9.CrossRef Yanoff LB, Menzie CM, Denkinger B, et al. Inflammation and iron deficiency in the hypoferremia of obesity. International journal of obesity (2005). 2007;31:1412–9.CrossRef
41.
Zurück zum Zitat Ganeshan M, Sainath PB, Padmavathi IJ, et al. Maternal manganese restriction increases susceptibility to high-fat diet-induced dyslipidemia and altered adipose function in WNIN male rat offspring. Exp Diabetes Res. 2011;2011:486316.PubMedPubMedCentralCrossRef Ganeshan M, Sainath PB, Padmavathi IJ, et al. Maternal manganese restriction increases susceptibility to high-fat diet-induced dyslipidemia and altered adipose function in WNIN male rat offspring. Exp Diabetes Res. 2011;2011:486316.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Guerrero-Romero F, Bermudez-Pena C, Rodriguez-Moran M. Severe hypomagnesemia and low-grade inflammation in metabolic syndrome. Magnes Res. 2011;24:45–53.PubMed Guerrero-Romero F, Bermudez-Pena C, Rodriguez-Moran M. Severe hypomagnesemia and low-grade inflammation in metabolic syndrome. Magnes Res. 2011;24:45–53.PubMed
43.
Zurück zum Zitat Guerrero-Romero F, Rodriguez-Moran M. Low serum magnesium levels and metabolic syndrome. Acta Diabetol. 2002;39:209–13.PubMedCrossRef Guerrero-Romero F, Rodriguez-Moran M. Low serum magnesium levels and metabolic syndrome. Acta Diabetol. 2002;39:209–13.PubMedCrossRef
44.
Zurück zum Zitat Guerrero-Romero F, Rodriguez-Moran M. Hypomagnesemia, oxidative stress, inflammation, and metabolic syndrome. Diabetes Metab Res Rev. 2006;22:471–6.PubMedCrossRef Guerrero-Romero F, Rodriguez-Moran M. Hypomagnesemia, oxidative stress, inflammation, and metabolic syndrome. Diabetes Metab Res Rev. 2006;22:471–6.PubMedCrossRef
45.
Zurück zum Zitat Jain CK, Gupta H, Chakrapani GJ. Enrichment and fractionation of heavy metals in bed sediments of river Narmada, India. Environ Monit Assess. 2008;141:35–47.PubMedCrossRef Jain CK, Gupta H, Chakrapani GJ. Enrichment and fractionation of heavy metals in bed sediments of river Narmada, India. Environ Monit Assess. 2008;141:35–47.PubMedCrossRef
46.
Zurück zum Zitat Jain CK, Rao VV, Prakash BA, Kumar KM, Yoshida M. Metal fractionation study on bed sediments of Hussainsagar Lake, Hyderabad, India. Environ Monit Assess. 2010;166:57–67.PubMedCrossRef Jain CK, Rao VV, Prakash BA, Kumar KM, Yoshida M. Metal fractionation study on bed sediments of Hussainsagar Lake, Hyderabad, India. Environ Monit Assess. 2010;166:57–67.PubMedCrossRef
47.
Zurück zum Zitat Cheng HL, Griffin HJ, Bryant CE, Rooney KB, Steinbeck KS, O'Connor HT. Impact of diet and weight loss on iron and zinc status in overweight and obese young women. Asia Pac J Clin Nutr. 2013;22:574–82.PubMed Cheng HL, Griffin HJ, Bryant CE, Rooney KB, Steinbeck KS, O'Connor HT. Impact of diet and weight loss on iron and zinc status in overweight and obese young women. Asia Pac J Clin Nutr. 2013;22:574–82.PubMed
48.
Zurück zum Zitat Mistry HD, Kurlak LO, Young SD, et al. Maternal selenium, copper and zinc concentrations in pregnancy associated with small-for-gestational-age infants. Matern Child Nutr. 2014;10:327–34.PubMedCrossRef Mistry HD, Kurlak LO, Young SD, et al. Maternal selenium, copper and zinc concentrations in pregnancy associated with small-for-gestational-age infants. Matern Child Nutr. 2014;10:327–34.PubMedCrossRef
49.
Zurück zum Zitat Rossman TG, Klein CB. Genetic and epigenetic effects of environmental arsenicals. Metallomics. 2011;3:1135–41.PubMedCrossRef Rossman TG, Klein CB. Genetic and epigenetic effects of environmental arsenicals. Metallomics. 2011;3:1135–41.PubMedCrossRef
50.
Zurück zum Zitat Hall MN, Liu X, Slavkovich V, et al. Folate, Cobalamin, Cysteine, Homocysteine, and arsenic metabolism among children in Bangladesh. Environ Health Perspect. 2009;117:825–31.PubMedPubMedCentralCrossRef Hall MN, Liu X, Slavkovich V, et al. Folate, Cobalamin, Cysteine, Homocysteine, and arsenic metabolism among children in Bangladesh. Environ Health Perspect. 2009;117:825–31.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Gamble MV, Liu X, Slavkovich V, et al. Folic acid supplementation lowers blood arsenic. Am J Clin Nutr. 2007;86:1202–9.PubMedPubMedCentral Gamble MV, Liu X, Slavkovich V, et al. Folic acid supplementation lowers blood arsenic. Am J Clin Nutr. 2007;86:1202–9.PubMedPubMedCentral
52.
Zurück zum Zitat Heindel JJ, Vandenberg LN. Developmental origins of health and disease: a paradigm for understanding disease cause and prevention. Curr Opin Pediatr. 2015;27:248–53.PubMedPubMedCentralCrossRef Heindel JJ, Vandenberg LN. Developmental origins of health and disease: a paradigm for understanding disease cause and prevention. Curr Opin Pediatr. 2015;27:248–53.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Liu Y, Murphy SK, Murtha AP, et al. Depression in pregnancy, infant birth weight and DNA methylation of imprint regulatory elements. Epigenetics. 2012;7:735–46.PubMedPubMedCentralCrossRef Liu Y, Murphy SK, Murtha AP, et al. Depression in pregnancy, infant birth weight and DNA methylation of imprint regulatory elements. Epigenetics. 2012;7:735–46.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Horne DW, Patterson D. Lactobacillus casei microbiological assay of folic acid derivatives in 96-well microtiter plates. Clin Chem. 1988;34:2357–9.PubMed Horne DW, Patterson D. Lactobacillus casei microbiological assay of folic acid derivatives in 96-well microtiter plates. Clin Chem. 1988;34:2357–9.PubMed
55.
Zurück zum Zitat Darrah TH, Prutsman-Pfeiffer JJ, Poreda RJ, Ellen Campbell M, Hauschka PV, Hannigan RE. Incorporation of excess gadolinium into human bone from medical contrast agents. Metallomics. 2009;1:479–88.PubMedCrossRef Darrah TH, Prutsman-Pfeiffer JJ, Poreda RJ, Ellen Campbell M, Hauschka PV, Hannigan RE. Incorporation of excess gadolinium into human bone from medical contrast agents. Metallomics. 2009;1:479–88.PubMedCrossRef
56.
Zurück zum Zitat DeLoid G, Cohen JM, Darrah T, et al. Estimating the effective density of engineered nanomaterials for in vitro dosimetry. Nat Commun. 2014;5:3514.PubMedPubMedCentralCrossRef DeLoid G, Cohen JM, Darrah T, et al. Estimating the effective density of engineered nanomaterials for in vitro dosimetry. Nat Commun. 2014;5:3514.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat McLaughlin MP, Darrah TH, Holland PL. Palladium (II) and platinum (II) bind strongly to an engineered blue copper protein. Inorg Chem. 2011;50:11294–6.PubMedPubMedCentralCrossRef McLaughlin MP, Darrah TH, Holland PL. Palladium (II) and platinum (II) bind strongly to an engineered blue copper protein. Inorg Chem. 2011;50:11294–6.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Sprauten M, Darrah TH, Peterson DR, et al. Impact of long-term serum platinum concentrations on neuro- and ototoxicity in Cisplatin-treated survivors of testicular cancer. J Clin Oncol. 2012;30:300–7.PubMedCrossRef Sprauten M, Darrah TH, Peterson DR, et al. Impact of long-term serum platinum concentrations on neuro- and ototoxicity in Cisplatin-treated survivors of testicular cancer. J Clin Oncol. 2012;30:300–7.PubMedCrossRef
59.
Zurück zum Zitat Koenker R, Bassett G. Regression quantiles. Econometrica: journal of the Econometric Society. 1978;46:33–50.CrossRef Koenker R, Bassett G. Regression quantiles. Econometrica: journal of the Econometric Society. 1978;46:33–50.CrossRef
60.
Zurück zum Zitat Faulk C, Barks A, Liu K, Goodrich JM, Dolinoy DC. Early-life lead exposure results in dose- and sex-specific effects on weight and epigenetic gene regulation in weanling mice. Epigenomics. 2013;5:487–500.PubMedCrossRef Faulk C, Barks A, Liu K, Goodrich JM, Dolinoy DC. Early-life lead exposure results in dose- and sex-specific effects on weight and epigenetic gene regulation in weanling mice. Epigenomics. 2013;5:487–500.PubMedCrossRef
61.
Zurück zum Zitat Govarts E, Remy S, Bruckers L, et al. Combined effects of prenatal exposures to environmental chemicals on birth weight. Int J Environ Res Public Health. 2016;13 Govarts E, Remy S, Bruckers L, et al. Combined effects of prenatal exposures to environmental chemicals on birth weight. Int J Environ Res Public Health. 2016;13
62.
Zurück zum Zitat Djukic-Cosic D, Ninkovic M, Malicevic Z, Plamenac-Bulat Z, Matovic V. Effect of supplemental magnesium on the kidney levels of cadmium, zinc, and copper of mice exposed to toxic levels of cadmium. Biol Trace Elem Res. 2006;114:281–91.PubMedCrossRef Djukic-Cosic D, Ninkovic M, Malicevic Z, Plamenac-Bulat Z, Matovic V. Effect of supplemental magnesium on the kidney levels of cadmium, zinc, and copper of mice exposed to toxic levels of cadmium. Biol Trace Elem Res. 2006;114:281–91.PubMedCrossRef
63.
Zurück zum Zitat Arbuckle TE, Liang CL, Morisset AS, et al. Maternal and fetal exposure to cadmium, lead, manganese and mercury: the MIREC study. Chemosphere. 2016;163:270–82.PubMedCrossRef Arbuckle TE, Liang CL, Morisset AS, et al. Maternal and fetal exposure to cadmium, lead, manganese and mercury: the MIREC study. Chemosphere. 2016;163:270–82.PubMedCrossRef
64.
Zurück zum Zitat McKay JA, Mathers JC. Diet induced epigenetic changes and their implications for health. Acta Physiol. 2011;202:103–18.CrossRef McKay JA, Mathers JC. Diet induced epigenetic changes and their implications for health. Acta Physiol. 2011;202:103–18.CrossRef
65.
Zurück zum Zitat Tsang V, Fry RC, Niculescu MD, et al. The epigenetic effects of a high prenatal folate intake in male mouse fetuses exposed in utero to arsenic. Toxicol Appl Pharmacol. 2012;264:439–50.PubMedPubMedCentralCrossRef Tsang V, Fry RC, Niculescu MD, et al. The epigenetic effects of a high prenatal folate intake in male mouse fetuses exposed in utero to arsenic. Toxicol Appl Pharmacol. 2012;264:439–50.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Gruber JF, Karagas MR, Gilbert-Diamond D, et al. Associations between toenail arsenic concentration and dietary factors in a New Hampshire population. Nutr J. 2012;11:45.PubMedPubMedCentralCrossRef Gruber JF, Karagas MR, Gilbert-Diamond D, et al. Associations between toenail arsenic concentration and dietary factors in a New Hampshire population. Nutr J. 2012;11:45.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Deb D, Biswas A, Ghose A, Das A, Majumdar KK, Guha Mazumder DN. Nutritional deficiency and arsenical manifestations: a perspective study in an arsenic-endemic region of West Bengal, India. Public Health Nutr. 2013;16:1644–55.PubMedCrossRef Deb D, Biswas A, Ghose A, Das A, Majumdar KK, Guha Mazumder DN. Nutritional deficiency and arsenical manifestations: a perspective study in an arsenic-endemic region of West Bengal, India. Public Health Nutr. 2013;16:1644–55.PubMedCrossRef
69.
Zurück zum Zitat Heck JE, Gamble MV, Chen Y, et al. Consumption of folate-related nutrients and metabolism of arsenic in Bangladesh. Am J Clin Nutr. 2007;85:1367–74.PubMed Heck JE, Gamble MV, Chen Y, et al. Consumption of folate-related nutrients and metabolism of arsenic in Bangladesh. Am J Clin Nutr. 2007;85:1367–74.PubMed
70.
Zurück zum Zitat Vahter M, Marafante E. Effects of low dietary intake of methionine, choline or proteins on the biotransformation of arsenite in the rabbit. Toxicol Lett. 1987;37:41–6.PubMedCrossRef Vahter M, Marafante E. Effects of low dietary intake of methionine, choline or proteins on the biotransformation of arsenite in the rabbit. Toxicol Lett. 1987;37:41–6.PubMedCrossRef
71.
Zurück zum Zitat Suarez-Ortegon MF, Mosquera M, Caicedo DM, De Plata CA, Mendez F. Nutrients intake as determinants of blood lead and cadmium levels in Colombian pregnant women. Am J Hum Biol. 2013;25:344–50.PubMedCrossRef Suarez-Ortegon MF, Mosquera M, Caicedo DM, De Plata CA, Mendez F. Nutrients intake as determinants of blood lead and cadmium levels in Colombian pregnant women. Am J Hum Biol. 2013;25:344–50.PubMedCrossRef
73.
Zurück zum Zitat Whincup PH, Kaye SJ, Owen CG, et al. Birth weight and risk of type 2 diabetes: a systematic review. JAMA. 2008;300:2886–97.PubMedCrossRef Whincup PH, Kaye SJ, Owen CG, et al. Birth weight and risk of type 2 diabetes: a systematic review. JAMA. 2008;300:2886–97.PubMedCrossRef
74.
Zurück zum Zitat Ezzahir N, Alberti C, Deghmoun S, et al. Time course of catch-up in adiposity influences adult anthropometry in individuals who were born small for gestational age. Pediatr Res. 2005;58:243–7.PubMedCrossRef Ezzahir N, Alberti C, Deghmoun S, et al. Time course of catch-up in adiposity influences adult anthropometry in individuals who were born small for gestational age. Pediatr Res. 2005;58:243–7.PubMedCrossRef
75.
Zurück zum Zitat Meas T, Deghmoun S, Armoogum P, Alberti C, Levy-Marchal C. Consequences of being born small for gestational age on body composition: an 8-year follow-up study. J Clin Endocrinol Metab. 2008;93:3804–9.PubMedPubMedCentralCrossRef Meas T, Deghmoun S, Armoogum P, Alberti C, Levy-Marchal C. Consequences of being born small for gestational age on body composition: an 8-year follow-up study. J Clin Endocrinol Metab. 2008;93:3804–9.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Howe LD, Tilling K, Benfield L, et al. Changes in ponderal index and body mass index across childhood and their associations with fat mass and cardiovascular risk factors at age 15. PLoS One. 2010;5:e15186.PubMedPubMedCentralCrossRef Howe LD, Tilling K, Benfield L, et al. Changes in ponderal index and body mass index across childhood and their associations with fat mass and cardiovascular risk factors at age 15. PLoS One. 2010;5:e15186.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Anderson EL, Howe LD, Fraser A, et al. Weight trajectories through infancy and childhood and risk of non-alcoholic fatty liver disease in adolescence: the ALSPAC study. J Hepatol. 2014;61:626–32.PubMedPubMedCentralCrossRef Anderson EL, Howe LD, Fraser A, et al. Weight trajectories through infancy and childhood and risk of non-alcoholic fatty liver disease in adolescence: the ALSPAC study. J Hepatol. 2014;61:626–32.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat de Kroon ML, Renders CM, van Wouwe JP, van Buuren S, Hirasing RA. The Terneuzen birth cohort: BMI change between 2 and 6 years is most predictive of adult cardiometabolic risk. PLoS One. 2010;5:e13966.PubMedPubMedCentralCrossRef de Kroon ML, Renders CM, van Wouwe JP, van Buuren S, Hirasing RA. The Terneuzen birth cohort: BMI change between 2 and 6 years is most predictive of adult cardiometabolic risk. PLoS One. 2010;5:e13966.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Gallagher CM, Meliker JR. Blood and urine cadmium, blood pressure, and hypertension: a systematic review and meta-analysis. Environ Health Perspect. 2010;118:1676–84.PubMedPubMedCentralCrossRef Gallagher CM, Meliker JR. Blood and urine cadmium, blood pressure, and hypertension: a systematic review and meta-analysis. Environ Health Perspect. 2010;118:1676–84.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Wallia A, Allen NB, Badon S, El Muayed M. Association between urinary cadmium levels and prediabetes in the NHANES 2005–2010 population. Int J Hyg Environ Health. 2014;217:854–60.PubMedPubMedCentralCrossRef Wallia A, Allen NB, Badon S, El Muayed M. Association between urinary cadmium levels and prediabetes in the NHANES 2005–2010 population. Int J Hyg Environ Health. 2014;217:854–60.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Tellez-Plaza M, Jones MR, Dominguez-Lucas A, Guallar E, Navas-Acien A. Cadmium exposure and clinical cardiovascular disease: a systematic review. Current atherosclerosis reports. 2013;15:356.PubMedCrossRef Tellez-Plaza M, Jones MR, Dominguez-Lucas A, Guallar E, Navas-Acien A. Cadmium exposure and clinical cardiovascular disease: a systematic review. Current atherosclerosis reports. 2013;15:356.PubMedCrossRef
82.
Zurück zum Zitat Karim MR, Rahman M, Islam K, et al. Increases in oxidized low-density lipoprotein and other inflammatory and adhesion molecules with a concomitant decrease in high-density lipoprotein in the individuals exposed to arsenic in Bangladesh. Toxicological sciences: an official journal of the Society of Toxicology. 2013;135:17–25.CrossRef Karim MR, Rahman M, Islam K, et al. Increases in oxidized low-density lipoprotein and other inflammatory and adhesion molecules with a concomitant decrease in high-density lipoprotein in the individuals exposed to arsenic in Bangladesh. Toxicological sciences: an official journal of the Society of Toxicology. 2013;135:17–25.CrossRef
83.
Zurück zum Zitat Mendez MA, González-Horta C, Sánchez-Ramírez B, Ballinas-Casarrubias L, Hernández Cerón R, Viniegra Morales D, Baeza Terrazas FA, Ishida MC, Gutiérrez-Torres DS, Saunders RJ, Drobná Z, Fry RC, Buse JB, Loomis D, García-Vargas GG, Del Razo LM, Stýblo M. Chronic exposure to arsenic and markers of cardiometabolic risk: a cross-sectional study in Chihuahua, Mexico. Environ Health Perspect 2016. Mendez MA, González-Horta C, Sánchez-Ramírez B, Ballinas-Casarrubias L, Hernández Cerón R, Viniegra Morales D, Baeza Terrazas FA, Ishida MC, Gutiérrez-Torres DS, Saunders RJ, Drobná Z, Fry RC, Buse JB, Loomis D, García-Vargas GG, Del Razo LM, Stýblo M. Chronic exposure to arsenic and markers of cardiometabolic risk: a cross-sectional study in Chihuahua, Mexico. Environ Health Perspect 2016.
84.
Zurück zum Zitat Kuo CC, Moon K, Thayer KA, Navas-Acien A. Environmental chemicals and type 2 diabetes: an updated systematic review of the epidemiologic evidence. Current diabetes reports. 2013;13:831–49.PubMedPubMedCentralCrossRef Kuo CC, Moon K, Thayer KA, Navas-Acien A. Environmental chemicals and type 2 diabetes: an updated systematic review of the epidemiologic evidence. Current diabetes reports. 2013;13:831–49.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Schober SE, Mirel LB, Graubard BI, Brody DJ, Flegal KM. Blood lead levels and death from all causes, cardiovascular disease, and cancer: results from the NHANES III mortality study. Environ Health Perspect. 2006;114:1538–41.PubMedPubMedCentral Schober SE, Mirel LB, Graubard BI, Brody DJ, Flegal KM. Blood lead levels and death from all causes, cardiovascular disease, and cancer: results from the NHANES III mortality study. Environ Health Perspect. 2006;114:1538–41.PubMedPubMedCentral
86.
Zurück zum Zitat Daniels SR, Greer FR. Lipid screening and cardiovascular health in childhood. Pediatrics. 2008;122:198–208.PubMedCrossRef Daniels SR, Greer FR. Lipid screening and cardiovascular health in childhood. Pediatrics. 2008;122:198–208.PubMedCrossRef
87.
Zurück zum Zitat Chinapaw MJ, Altenburg TM, van Eijsden M, Gemke RJ, Vrijkotte TG. Screen time and cardiometabolic function in Dutch 5–6 year olds: cross-sectional analysis of the ABCD-study. BMC Public Health. 2014;14:933.PubMedPubMedCentralCrossRef Chinapaw MJ, Altenburg TM, van Eijsden M, Gemke RJ, Vrijkotte TG. Screen time and cardiometabolic function in Dutch 5–6 year olds: cross-sectional analysis of the ABCD-study. BMC Public Health. 2014;14:933.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Falaschetti E, Hingorani AD, Jones A, et al. Adiposity and cardiovascular risk factors in a large contemporary population of pre-pubertal children. Eur Heart J. 2010;31:3063–72.PubMedPubMedCentralCrossRef Falaschetti E, Hingorani AD, Jones A, et al. Adiposity and cardiovascular risk factors in a large contemporary population of pre-pubertal children. Eur Heart J. 2010;31:3063–72.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Toschke AM, Reinehr T. Different anthropometric index changes in relation to cardiovascular risk profile change. Clin Nutr. 2008;27:457–63.PubMedCrossRef Toschke AM, Reinehr T. Different anthropometric index changes in relation to cardiovascular risk profile change. Clin Nutr. 2008;27:457–63.PubMedCrossRef
90.
Zurück zum Zitat Calarge CA, Xie D, Fiedorowicz JG, Burns TL, Haynes WG. Rate of weight gain and cardiometabolic abnormalities in children and adolescents. J Pediatr. 2012;161:1010–5.PubMedPubMedCentralCrossRef Calarge CA, Xie D, Fiedorowicz JG, Burns TL, Haynes WG. Rate of weight gain and cardiometabolic abnormalities in children and adolescents. J Pediatr. 2012;161:1010–5.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Messiah SE, Arheart KL, Natale RA, Hlaing WM, Lipshultz SE, Miller TL. BMI, waist circumference, and selected cardiovascular disease risk factors among preschool-age children. Obesity. 2012;20:1942–9.PubMedCrossRef Messiah SE, Arheart KL, Natale RA, Hlaing WM, Lipshultz SE, Miller TL. BMI, waist circumference, and selected cardiovascular disease risk factors among preschool-age children. Obesity. 2012;20:1942–9.PubMedCrossRef
92.
Zurück zum Zitat Eriksson JG, Kajantie E, Lampl M, Osmond C. Trajectories of body mass index among children who develop type 2 diabetes as adults. J Intern Med. 2015; Eriksson JG, Kajantie E, Lampl M, Osmond C. Trajectories of body mass index among children who develop type 2 diabetes as adults. J Intern Med. 2015;
93.
Zurück zum Zitat Huang RC, de Klerk NH, Smith A, et al. Lifecourse childhood adiposity trajectories associated with adolescent insulin resistance. Diabetes Care. 2011;34:1019–25.PubMedPubMedCentralCrossRef Huang RC, de Klerk NH, Smith A, et al. Lifecourse childhood adiposity trajectories associated with adolescent insulin resistance. Diabetes Care. 2011;34:1019–25.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Lawlor DA, Benfield L, Logue J, et al. Association between general and central adiposity in childhood, and change in these, with cardiovascular risk factors in adolescence: prospective cohort study. BMJ. 2010;341:c 6224.CrossRef Lawlor DA, Benfield L, Logue J, et al. Association between general and central adiposity in childhood, and change in these, with cardiovascular risk factors in adolescence: prospective cohort study. BMJ. 2010;341:c 6224.CrossRef
95.
Zurück zum Zitat Puder JJ, Schindler C, Zahner L, Kriemler S. Adiposity, fitness and metabolic risk in children: a cross-sectional and longitudinal study. International journal of pediatric obesity : IJPO: an official journal of the International Association for the Study of Obesity. 2011;6:e297–306.CrossRef Puder JJ, Schindler C, Zahner L, Kriemler S. Adiposity, fitness and metabolic risk in children: a cross-sectional and longitudinal study. International journal of pediatric obesity : IJPO: an official journal of the International Association for the Study of Obesity. 2011;6:e297–306.CrossRef
96.
Zurück zum Zitat Ong KK, Dunger DB. Birth weight, infant growth and insulin resistance. Eur J Endocrinol. 2004;151(Suppl 3):U131–9.PubMedCrossRef Ong KK, Dunger DB. Birth weight, infant growth and insulin resistance. Eur J Endocrinol. 2004;151(Suppl 3):U131–9.PubMedCrossRef
97.
Zurück zum Zitat Juonala M, Viikari JS, Raitakari OT. Main findings from the prospective cardiovascular risk in young Finns study. Curr Opin Lipidol. 2013;24:57–64.PubMedCrossRef Juonala M, Viikari JS, Raitakari OT. Main findings from the prospective cardiovascular risk in young Finns study. Curr Opin Lipidol. 2013;24:57–64.PubMedCrossRef
98.
Zurück zum Zitat Laitinen TT, Pahkala K, Magnussen CG, et al. Ideal cardiovascular health in childhood and cardiometabolic outcomes in adulthood: the cardiovascular risk in young Finns study. Circulation. 2012;125:1971–8.PubMedCrossRef Laitinen TT, Pahkala K, Magnussen CG, et al. Ideal cardiovascular health in childhood and cardiometabolic outcomes in adulthood: the cardiovascular risk in young Finns study. Circulation. 2012;125:1971–8.PubMedCrossRef
99.
Zurück zum Zitat Berenson GS, Srnivasan SR. Cardiovascular risk factors in youth with implications for aging: the Bogalusa Heart study. Neurobiol Aging. 2005;26:303–7.PubMedCrossRef Berenson GS, Srnivasan SR. Cardiovascular risk factors in youth with implications for aging: the Bogalusa Heart study. Neurobiol Aging. 2005;26:303–7.PubMedCrossRef
100.
Zurück zum Zitat Chen W, Srinivasan SR, Li S, Xu J, Berenson GS. Metabolic syndrome variables at low levels in childhood are beneficially associated with adulthood cardiovascular risk: the Bogalusa Heart study. Diabetes Care. 2005;28:126–31.PubMedCrossRef Chen W, Srinivasan SR, Li S, Xu J, Berenson GS. Metabolic syndrome variables at low levels in childhood are beneficially associated with adulthood cardiovascular risk: the Bogalusa Heart study. Diabetes Care. 2005;28:126–31.PubMedCrossRef
101.
Zurück zum Zitat Morrison JA, Glueck CJ, Wang P. Childhood risk factors predict cardiovascular disease, impaired fasting glucose plus type 2 diabetes mellitus, and high blood pressure 26 years later at a mean age of 38 years: the Princeton-lipid research clinics follow-up study. Metab Clin Exp. 2012;61:531–41.PubMedCrossRef Morrison JA, Glueck CJ, Wang P. Childhood risk factors predict cardiovascular disease, impaired fasting glucose plus type 2 diabetes mellitus, and high blood pressure 26 years later at a mean age of 38 years: the Princeton-lipid research clinics follow-up study. Metab Clin Exp. 2012;61:531–41.PubMedCrossRef
102.
Zurück zum Zitat Sun SS, Grave GD, Siervogel RM, Pickoff AA, Arslanian SS, Daniels SR. Systolic blood pressure in childhood predicts hypertension and metabolic syndrome later in life. Pediatrics. 2007;119:237–46.PubMedCrossRef Sun SS, Grave GD, Siervogel RM, Pickoff AA, Arslanian SS, Daniels SR. Systolic blood pressure in childhood predicts hypertension and metabolic syndrome later in life. Pediatrics. 2007;119:237–46.PubMedCrossRef
103.
Zurück zum Zitat Nguyen QM, Srinivasan SR, Xu JH, Chen W, Kieltyka L, Berenson GS. Utility of childhood glucose homeostasis variables in predicting adult diabetes and related cardiometabolic risk factors: the Bogalusa Heart study. Diabetes Care. 2010;33:670–5.PubMedCrossRef Nguyen QM, Srinivasan SR, Xu JH, Chen W, Kieltyka L, Berenson GS. Utility of childhood glucose homeostasis variables in predicting adult diabetes and related cardiometabolic risk factors: the Bogalusa Heart study. Diabetes Care. 2010;33:670–5.PubMedCrossRef
104.
Zurück zum Zitat Nguyen QM, Srinivasan SR, Xu JH, Chen W, Berenson GS. Changes in risk variables of metabolic syndrome since childhood in pre-diabetic and type 2 diabetic subjects: the Bogalusa Heart study. Diabetes Care. 2008;31:2044–9.PubMedPubMedCentralCrossRef Nguyen QM, Srinivasan SR, Xu JH, Chen W, Berenson GS. Changes in risk variables of metabolic syndrome since childhood in pre-diabetic and type 2 diabetic subjects: the Bogalusa Heart study. Diabetes Care. 2008;31:2044–9.PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Nigam D, Shukla GS, Agarwal AK. Glutathione depletion and oxidative damage in mitochondria following exposure to cadmium in rat liver and kidney. Toxicol Lett. 1999;106:151–7.PubMedCrossRef Nigam D, Shukla GS, Agarwal AK. Glutathione depletion and oxidative damage in mitochondria following exposure to cadmium in rat liver and kidney. Toxicol Lett. 1999;106:151–7.PubMedCrossRef
106.
Zurück zum Zitat Jurczuk M, Brzoska MM, Moniuszko-Jakoniuk J, Galazyn-Sidorczuk M, Kulikowska-Karpinska E. Antioxidant enzymes activity and lipid peroxidation in liver and kidney of rats exposed to cadmium and ethanol. Food and chemical toxicology: an international journal published for the British Industrial Biological Research Association. 2004;42:429–38.CrossRef Jurczuk M, Brzoska MM, Moniuszko-Jakoniuk J, Galazyn-Sidorczuk M, Kulikowska-Karpinska E. Antioxidant enzymes activity and lipid peroxidation in liver and kidney of rats exposed to cadmium and ethanol. Food and chemical toxicology: an international journal published for the British Industrial Biological Research Association. 2004;42:429–38.CrossRef
107.
Zurück zum Zitat Li R, Yuan C, Dong C, Shuang S, Choi MM. In vivo antioxidative effect of isoquercitrin on cadmium-induced oxidative damage to mouse liver and kidney. Naunyn Schmiedeberg's Arch Pharmacol. 2011;383:437–45.CrossRef Li R, Yuan C, Dong C, Shuang S, Choi MM. In vivo antioxidative effect of isoquercitrin on cadmium-induced oxidative damage to mouse liver and kidney. Naunyn Schmiedeberg's Arch Pharmacol. 2011;383:437–45.CrossRef
108.
Zurück zum Zitat Gerich JE. Role of the kidney in normal glucose homeostasis and in the hyperglycaemia of diabetes mellitus: therapeutic implications. Diabetic medicine: a journal of the British Diabetic Association. 2010;27:136–42.CrossRef Gerich JE. Role of the kidney in normal glucose homeostasis and in the hyperglycaemia of diabetes mellitus: therapeutic implications. Diabetic medicine: a journal of the British Diabetic Association. 2010;27:136–42.CrossRef
109.
Zurück zum Zitat Meyer C, Dostou JM, Gerich JE. Role of the human kidney in glucose counterregulation. Diabetes. 1999;48:943–8.PubMedCrossRef Meyer C, Dostou JM, Gerich JE. Role of the human kidney in glucose counterregulation. Diabetes. 1999;48:943–8.PubMedCrossRef
110.
Zurück zum Zitat Meyer C, Dostou JM, Welle SL, Gerich JE. Role of human liver, kidney, and skeletal muscle in postprandial glucose homeostasis. Am J Physiol Endocrinol Metab. 2002;282:E419–27.PubMedCrossRef Meyer C, Dostou JM, Welle SL, Gerich JE. Role of human liver, kidney, and skeletal muscle in postprandial glucose homeostasis. Am J Physiol Endocrinol Metab. 2002;282:E419–27.PubMedCrossRef
111.
Zurück zum Zitat DeFronzo RA, Davidson JA, Del Prato S. The role of the kidneys in glucose homeostasis: a new path towards normalizing glycaemia. Diabetes Obes Metab. 2012;14:5–14.PubMedCrossRef DeFronzo RA, Davidson JA, Del Prato S. The role of the kidneys in glucose homeostasis: a new path towards normalizing glycaemia. Diabetes Obes Metab. 2012;14:5–14.PubMedCrossRef
Metadaten
Titel
Maternal blood cadmium, lead and arsenic levels, nutrient combinations, and offspring birthweight
verfasst von
Yiwen Luo
Lauren E. McCullough
Jung-Ying Tzeng
Thomas Darrah
Avner Vengosh
Rachel L. Maguire
Arnab Maity
Carmen Samuel-Hodge
Susan K. Murphy
Michelle A. Mendez
Cathrine Hoyo
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
BMC Public Health / Ausgabe 1/2017
Elektronische ISSN: 1471-2458
DOI
https://doi.org/10.1186/s12889-017-4225-8

Weitere Artikel der Ausgabe 1/2017

BMC Public Health 1/2017 Zur Ausgabe