Abstract
The goal of this investigation was to evaluate the effects of gestational administrations of arsenic trioxide (ATO; As2O3) on fetal neuroendocrine development (the thyroid-cerebrum axis). Pregnant Wistar rats were orally administered ATO (5 or 10 mg/kg) from gestation day (GD) 1 to 20. Both doses of ATO diminished free thyroxine and free triiodothyronine levels and augmented thyrotropin level in both dams and fetuses at GD 20. Also, the maternofetal hypothyroidism in both groups caused a dose-dependent reduction in the fetal serum growth hormone, insulin growth factor-I (IGF-I), and IGF-II levels at embryonic day (ED) 20. These disorders perturbed the maternofetal body weight, fetal brain weight, and survival of pregnant and their fetuses. In addition, destructive degeneration, vacuolation, hyperplasia, and edema were observed in the fetal thyroid and cerebrum of both ATO groups at ED 20. These disruptions appear to depend on intensification in the values of lipid peroxidation, nitric oxide, and H2O2, suppression of messenger RNA (mRNA) expression of nuclear factor erythroid 2-related factor 2 and peroxisome proliferator-activated receptor gamma, and activation of mRNA expression of caspase-3, nuclear factor kappa-light-chain-enhancer of activated B cells, cyclooxygenase-2, Bcl-2–associated X protein, and inducible nitric oxide synthase in the fetal cerebrum. These data suggest that gestational ATO may disturb thyroid-cerebrum axis generating fetal neurodevelopmental toxicity.
Keywords: arsenic trioxide, thyroid, cerebrum, fetus, pregnant rats
Introduction
Arsenic, a natural metalloid, is a known ubiquitous environmental contaminant1–4 found in human foods (meat and fish), drinking water, soil, and air.5–8 In particular, the effective harmful source of arsenic is in drinking water.9 In 8 states of America, the levels of arsenic in drinking water are 50 to 100 ppb above the approved maximum contaminant level, 10 ppb.10 In addition, arsenic trioxide (ATO; As2O3) is a potent toxic agent used in agriculture as herbicide and pesticide, and in the manufacturing of many industrial materials such as glass, wood, dyes, and pigments.11–13 Arsenic is a harmful toxin to human6,14,15 and animal health4,11 causing a wide range of diseases, including skin cancer and cognitive dysfunction,16 diabetes mellitus and cardiovascular diseases,17 and abdominal problems.18 These abnormalities are amplified during the fetal developmental period.11,19
Thyroid hormones (THs) are susceptible to disruption by arsenic,20 particularly during the fetal development. Exposure of guinea pigs to 50 ppm arsenic in the basal diet decreased the levels of THs.13 However, rats exposed to 100 mg arsenic/L for 4 weeks exhibited an increase in the level of serum T3.21 Epidemiological and animal studies have proposed that arsenic penetrates the placenta22 causing miscarriage, death, and several developmental toxicities.23,24 In human, low values of arsenic (0.8-2 ppb) can increase the risk of spontaneous abortion25 and fetal mortality.26 In utero arsenic exposure caused morbidity in rural Bangladesh.27 In pregnant rats, Holson et al28 reported that a reduction in the food intake for the dams and body weight of both dams and their fetuses was progressed from doses 1 to 10 mg ATO. In addition, gestational arsenic can accumulate in the fetal brain of rats29 causing several brain disorders.30 Administration of rats to 50 mg/L inorganic arsenic (arsenite) as a chronic dose induced apoptosis in the brain.31 Exposure of mice to 0.5 and 5 ppm ATO via the drinking water for 6 weeks induced apoptosis in the neuronal cells of the cerebral cortex.32 Additional investigations have shown that exposure to arsenic causes neuronal apoptosis30,33 and neural dysfunction.34 In clinical trials and animal models, chronic exposure to arsenic causes neurotoxicity35,36 and cognitive impairments.37 These disturbances could be attributed to the ability of arsenic to deplete sulfhydryl groups, increase reactive oxygen species (ROS),4,38 and disrupt the activity of peroxisome proliferator-activated receptor gamma (PPAR-γ),39 Nrf-2 expression,40 and phosphoinositide 3-kinase (PI3K)/AKT phosphorylation pathways.41 These variations can disrupt the neural gene expression and brain activities.42,43 However, the mechanisms by which gestational ATO induces developmental neuroendocrine disruption are still unclear.
Thyroid hormones,44 growth factors,45 and gene expression factors46 display dynamic roles during brain development. As the sensitivity of the developing neuroendocrine axis to any stress is found to be obviously critical,47,48 the purpose of the current investigation was to follow the novel action of gestational administrations of ATO (5 or 10 mg/kg) from gestation day (GD) 1 to 20 on the fetal thyroid-cerebrum axis.
Materials and Methods
Chemicals
Arsenic trioxide (As2O3; 99.99% purity) was procured from Sigma-Aldrich (St. Louis, MO, USA). The T4, T3, thyrotropin (TSH), growth hormone (GH), insulin growth factor-I (IGF-I), and IGF-II kits were purchased from Millipore (St. Charles, MO, USA) ELISA Kit. All chemicals were of the optimum analytic grades.
Ethics Statement
All animal experimentations were approved with the overall rules of Egyptian Animal Care and Animal Ethics Committee in Beni-Suef University, Faculty of Science, Zoology Department (BSU/FS/ 2016/9). All efforts were made to ensure the proper care and use of animals; http://www.bsu.edu.eg/Content.aspx?section_id=874&cat_id=43&.
Animals and Experimental Design
The experiment was conducted on 24 mature virgin female Wistar rats (Rattus norvegicus) weighing 155 to 165 g. Also, 12 adult males were used for the coupling only and were otherwise not part of the study. Rats were obtained from the animal house of VACSERA (Holding Company for Biological Products & Vaccine) in Helwan, Egypt. Female and male rats were given tap water and food daily ad libitum, and kept in stainless steel cages at 25°C ± 2°C temperature, 50% ± 5% relative humidity, and light/dark cycle of 12 hours each (lights on at 7:00 hours)49–53 for 2 weeks to get rid of any intercurrent infection and familiarization. Then, pregnancy was carried out in a separate cage by coupling 1 male with 2 proestrous females overnight for 1 or 2 days54,55 and confirmed by the presence of a copulatory plug or sperms in the vaginal smears (GD 0). Pregnant rats were then weighed and housed separately in cages. Notably, rats are used in this experiment because their erythrocytes are extremely prone to retain arsenic.56
Arsenic trioxide was dissolved in deionized water. Then, 1 M NaOH was added to form a clear solution. HCl of 1 M was added to adjust pH between 7.0 and 8.0. Then, a suitable volume of deionized water was added to each preparation to achieve the desired concentration. Aqueous solutions of ATO were freshly prepared and administered to pregnant rats by gavage (0, 5, or 10 mg ATO/kg) from GD 1 to GD 20. These dosages were designated according to Holson et al28 The control pregnant was orally administrated deionized water only as a vehicle. The maternal mortality and abortion were observed throughout the experimental period, and the maternal weight gain, fetal weight, fetal death, and fetal brain weight were recorded at GD 20.
Anesthetized dams and fetuses by mild diethyl ether were killed at GD 20. The quantity of the mild diethyl ether was very low with very short exposure-time to avoid any irritation to the eyes, nose, and respiratory airways according to the local ethics committee and several papers.48,49,51–53,55,57–62 Maternal blood samples were obtained from the jugular vein whereas blood samples of fetuses were obtained directly from the umbilical cord.57,59,60,63 Samples of coagulated blood were centrifuged at 3000 rpm (1006.2 g) for 20 minutes, and the clear supernatants were retained at −20°C for the following biochemical examinations. On the other hand, thyroid and cerebrum of dissected fetuses at embryonic day (ED) 20 were rapidly excised and fixed in 10% neutral buffered formalin for the histopathological examination. The remaining cerebrum tissue was perfused in ice-cold phosphate-buffered saline (PBS). Homogenates of frozen cerebrum samples in chilled PBS (10% wt/vol) were centrifuged at 3000 rpm (1006.2 g) for 20 minutes. The homogenates were retained at −70°C for subsequent quantitative reverse transcription polymerase chain reaction (qRT-PCR) assay. Furthermore, homogenates of other-remaining cerebrum samples in 0.9% NaCl (10% wt/vol) were prepared for further biochemical assay.
Enzyme-Linked Immunosorbent Assay Examination
The concentrations of maternal and fetal serum T3, T4, and TSH, and fetal GH, IGF-I, and IGF-II were assessed by ELISA (Spectra Max 190-Molecular Devices, USA) in Egyptian Cairo University, Faculty of Medicine, Department of Biochemistry, using a spectrophotometer at 450 nm. The standard plots were assembled using standards and used to calculate the concentrations of unknown samples.
Histological Examination
After fixation of the thyroid and cerebral tissues in 10% neutral buffered formalin for 1 day at room temperature, the samples were transferred to the Histopathological Department, Veterinary Faculty, Beni-Suef University, Egypt for additional processing, paraffin blocking, sectioning at 6 µm, mounting on glass slides, deparaffinized, and staining with hematoxylin and eosin (H&E) stain.64 The stained slides were evaluated under the light microscope.
RNA Isolation and qRT-PCR Examination
The homogenate of frozen cerebrum samples was transferred to the Department of Biochemistry, Faculty of Medicine, Cairo University, Egypt for additional processing. A Fermentas RNA isolation kit was used to isolate the total RNA. The concentrations were measured at 260 nm and the selection of RNA samples was A260/A280 ratios ≥1.7. A reverse transcription Fermentas kit was used in reverse transcription of RNA to complementary DNA (cDNA) with 1 µg RNA. A SYBR Green master mix was used to augmentation the produced cDNA with a total volume of 20 µL. Table 1 shows the primer set of examined PPARγ, nuclear factor erythroid 2-related factor 2 (Nrf2), cysteine-aspartic acid protease-3 (caspase-3); cyclooxygenase-2 (Cox2), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), Bcl-2–associated X protein (BAX), and inducible nitric oxide synthase (iNOS). A 96-well plate was used for the sample seeding reactions. The PCR thermal cycles involved initial denaturation at 95°C for 10 minutes and 35 cycles of denaturation at 95°C for 30 seconds, annealing at Tm-5 for 60 seconds and extension at 72°C for 30 seconds. Nonspecific amplifications were detected by nontemplate and water controls. Each standard and experimental sample was examined in duplicate. The acquired amplification results were assessed by the 2−ΔΔCt method.65 Notably, β-actin and glyceraldehyde-3-phosphate dehydrogenase, 2 reference genes, were utilized for normalization of all values.
Table 1.
Primer Sequences for the Quantitative RT-PCR Analyses.
Gene | Sequence 5’-3’ |
---|---|
PPARγ | F: GGACGCTGAAGAAGAGACCTG |
R: CCGGGTCCTGTCTGAGTATG | |
Nrf2 | F: TTGTAGATGACCATGAGTCGC |
R: TGTCCTGCTGTATGCTGCTT | |
iNOS | F: ATTCCCAGCCCAACAACACA |
R: GCAGCTTGTCCAGGGATTCT | |
NF-κB | F: TCTCAGCTGCGACCCCG |
R: TGGGCTGCTCAATGATCTCC | |
BAX | F: TGAGCGAGGCGGTGAGG |
R: AAGTTCAACGGCACAGTCAAGG | |
Cox2 | F: GTGGGATGACGAGCGACTG |
R: CCGTGTTCAAGGAGGATGG | |
Caspase-3 | F: ACTCTTGTGGGCAAACCCAA |
R: CTCTCCATGAGCAGTAGCCG | |
β-Actin | F: TACAACCTTCTTGCAGCTCCT |
R: CCTTCTGACCCATACCCACC | |
GAPDH | F: AGGTCGGAGTCA ACGGATTTGGT |
R: CATGTGGGCCATGAGGTCCACCAC |
Abbreviations: BAX, Bcl-2–associated X protein; Cox2, cyclooxygenase-2; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; iNOS, inducible nitric oxide synthase; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; Nrf2, nuclear factor erythroid 2-related factor 2; PPARγ, peroxisome proliferator-activated receptor gamma; RT-PCR, reverse transcription polymerase chain reaction.
Biochemical Assay
The homogenates of frozen cerebrum samples were centrifuged at 3000 rpm (1006.2 g) for 20 minutes. The levels of ROS were assayed in the Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt. Lipid peroxidation (LPO) level was estimated by the formation of the malondialdehyde (MDA) as per the methodology of Preuss et al.66 In addition, nitric oxide (NO) and H2O2 levels were determined as per the procedures of Dutta et al,67 and Sergiev et al,68 respectively.
Statistical Analysis
The data were stated as a mean ± standard error. Difference between the experimental groups was analyzed by PC-STAT program software (version 1A(C) PC-STAT, University of Georgia).69 The groups were compared using analysis of one-way of variance and the least significant degree (LSD). The general variations between the experimental groups were recognized by F-probability. The number of considered samples/marker/group was 6. The variations between the experimental groups were P <.01 and P < .001.
Results
Gestational ATO-Perturbed the Maternofetal Thyroid Functions and Development
In 5 and 10 mg maternal ATO-treated groups, the concentrations of serum T3 and T4 were considerably (LSD; P < .01) decreased while the concentration of serum TSH was profoundly (LSD; P < .01) increased at GD 20 in both dams and fetuses (Figure 1A and B). These disorders indicate a maternofetal hypothyroid state. Fetal serum GH, IGF-I, and IGF-II levels were decreased by 3.56, 4.64, and 2.1-fold, respectively in the maternal ATO-low treated group and by 6.09, 9.26, and 5.47-fold, respectively in the maternal ATO-high treated group, respectively in relative to the control group (Figure 1C). In both maternal treated groups, there was a substantial drop in the maternal body weight gain (−32.22% in the As2O3-low treated group and −54.35% in the As2O3-high treated group), fetal body weight (−38.98% in the As2O3-low treated group and −54.47% in the As2O3-high treated group), and fetal brain weight (−11.65% in the As2O3-low treated group and −21.69% in the As2O3-high treated group; Table 2). Otherwise, both administrations of ATO caused a death in only 1 pregnant rat. However, abortion was only observed in 1 pregnant rat in the ATO-high treated group only. The fetal death was folded in the maternal ATO-high treated group compared to the maternal ATO-low treated group at ED 20 (Table 2).
Figure 1.
Administrations of As2O3 caused (A) maternal hypothyroidism at GD 20, (B) fetal hypothyroidism at ED 20, and (C) Fetal growth retardation at ED 20. Data are expressed as mean ± SE. Number of animals in each group is 6. Where * is a highly significant (P < .01) change between the control and treated group. ED indicates embryonic day; GD, gestation day; SE, standard error.
Table 2.
Gestational Administrations of As2O3 Changed the Maternal and Fetal Markers at GD 20.a
Day | As2O3 (mg/kg) | Maternal Body Weight Gain (g) | No. of Dead Dams/Pregnant Rats | No. of Aborted Dams | No. of Dead Fetuses/Total Fetuses | Fetal Body Weight (g) | Fetal Brain Weight (mg) |
---|---|---|---|---|---|---|---|
GD 20 | 0 | 45.81 ± 0.57 | 0/8 | 0 | 0/80 | 4.13 ± 0.090 | 152.46±0.95 |
5 | 31.05 ± 0.52b | 1/8 | 0 | 2/65 | 2.52 ± 0.049b | 134.69±0.68 b | |
−32.22% | −38.98% | −11.65% | |||||
10 | 20.91 ± 0.37 b | 1/8 | 1 | 4/62 | 1.88 ± 0.055 b | 119.39±0.77 b | |
−54.35% | −54.47% | −21.69% | |||||
ANOVA | P < .001 | P < .001 | |||||
LSD 5% | 1.5035 | 0.2034 | 3.6285 | ||||
LSD 1% | 2.0792 | 0.2813 | 5.0179 |
Abbreviations: ANOVA, analysis of one way of variance; GD, gestation day; LSD, least significant degree; SE, standard error.
aData are expressed as mean ± SE. Number of animals in each group is 6.
bIndicates a highly significant (P < .01) change between the control and treated group and P < .001 is very highly significant change for whole experiment.
Gestational ATO-Induced a Fetal Thyroid and Cerebrum Dysgenesis
In the control group, the follicles of the fetal thyroid gland appeared normally in their distribution at ED 20 (Figure 2A). Fetal thyroid sections of maternal ATO-low treated group showed a flattened cell lining epithelium, hyperplasia, edema, atrophy, and destructive degeneration (Figure 2B). In addition to the previous lesions, colloid vacuoles were observed in the fetal thyroid sections of maternal ATO-high treated group (Figure 2C). On the other hand, the fetal cerebrum sections showed a normal histological architecture at ED 20 (Figure 3A). Vacuoles (Figure 3B 1), congested blood vessels and degenerative changes (Figure 3B 2), and pericellular edema (Figure 3B 1,2) were noticed in the fetal cerebrum of maternal ATO-low treated group. Moreover, Figure 3C 1 showed a vacuolar degenerative and hyperplastic proliferation, and Figure 3C 2 appeared severe congested blood vessels and several degenerative changes in the fetal cerebrum of maternal ATO-high treated group.
Figure 2.
Sagittal sections in the thyroid gland of fetal rats at ED 20 in control (A), 5 mg As2O3 (B), and 10 mg As2O3 (C). Hematoxylin and eosin stain. Where, A indicates atrophy; C, colloid; CCLE, cuboidal cell lining epithelium; CV, colloid vacuoles; DD, destructive degeneration; ED embryonic day; FCLE, flattened cell lining epithelium; HYP, hyperplasia; O, edema; PFC, parafollicular cell.
Figure 3.
Sagittal sections in the cerebrum of fetal rats at ED 20 in control (A), 5 mg As2O3 (B1,2), and 10 mg As2O3 (C1,2). Hematoxylin and eosin stain. Where, CBV indicates congested blood vessel; DC, degenerative changes; ED, embryonic day; HP, hyperplastic proliferation; PCO, pericellular edema; PYC, pyramidal cell; V, vacuoles, and the arrows refer to the apical dendrites.
Gestational ATO-Disturbed the Gene Expression and Increased ROS in the Fetal Cerebrum
In both maternal ATO-treated groups, qPCR analysis exhibited a considerable (LSD; P < .01) downregulation of messenger RNA (mRNA) expression of Nrf2 and PPARγ in the fetal cerebrum at ED 20 (Table 3). On the contrary, both maternal administrations of ATO showed significant (LSD; P < .01) upregulation of mRNA expression of caspase-3, NF-κB, Cox2, BAX, and iNOS in the fetal cerebrum, where their means in the ATO-high treated group were 5.16, 2.91, 2.65, 2.80, and 3.25, respectively compared to their means in the ATO-low treated group (4.50, 2.45, 1.96, 1.54, and 2.47, respectively) or in the control group (3.68, 1.89, 1.14, 0.98, and 1.97, respectively; Table 3). On the other hand, LPO (MDA) level of both maternal ATO-treated groups was considerably (LSD; P < .01) elevated in the fetal cerebrum at ED 20 in comparison with the control group (Figure 4). Nitric oxide and H2O2 levels displayed a parallel pattern where their levels were significantly increased in the fetal cerebrum at ED 20, as described in Figure 4.
Table 3.
Gestational Administrations of As2O3 Disrupted the Relative mRNA Expression of PPARγ, Nrf2, iNOS, NF-κB, BAX, Cox2, and Caspase-3 in the Fetal Cerebrum at ED 20.a
Day | As2O3 (mg/kg) | PPARγ | Nrf2 | iNOS | NF-κB | BAX | Cox2 | Caspase-3 |
---|---|---|---|---|---|---|---|---|
Relative mRNA expression | ||||||||
ED 20 | 0 | 1.04 ± 0.001 | 1.50 ± 0.003 | 1.97 ± 0.024 | 1.89 ± 0.003 | 0.98 ± 0.04 | 1.14 ± 0.003 | 3.68 ± 0.08 |
5 | 0.57 ± 0.002b | 1.20 ± 0.001b | 2.47 ± 0.049b | 2.45 ± 0.049b | 1.54 ± 0.05b | 1.96 ± 0.055b | 4.50 ± 0.05b | |
−45.19% | −20.00% | +25.38% | +29.62% | +57.14% | +71.92% | +22.28% | ||
10 | 0.26 ± 0.003b | 0.95 ± 0.004b | 3.25 ± 0.002b | 2.91 ± 0.078b | 2.80 ± 0.08b | 2.65 ± 0.029b | 5.16 ± 0.04b | |
−75.00% | −36.66% | +64.97% | +53.96% | +185.71% | +132.45% | +40.21% | ||
ANOVA | P < .001 | |||||||
LSD 5% | 0.0081 | 0.0090 | 0.1082 | 0.1724 | 0.1949 | 0.1224 | 0.1946 | |
LSD 1% | 0.1120 | 0.1249 | 0.1496 | 0.2385 | 0.2695 | 0.1692 | 0.2692 |
Abbreviations: ANOVA, analysis of one-way of variance; BAX, Bcl-2–associated X protein; Cox2, cyclooxygenase-2; ED, embryonic day; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; LSD, least significant degree; iNOS, inducible nitric oxide synthase; mRNA, messenger RNA; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; Nrf2, nuclear factor erythroid 2-related factor 2; PPARγ, peroxisome proliferator-activated receptor gamma; SE, standard error.
aExpression levels were normalized using β-Actin and GAPDH as the reference genes. For each gene, levels are expressed relative to the average level in the control group. Data are expressed as mean ± SE. Number of animals in each group is 6.
bIndicates a highly significant (P < .01) change between the control and treated group and P < .001 is very highly significant change for whole experiment.
Figure 4.
Gestational administrations of As2O3 increased LPO, NO, and H2O2 levels in the fetal cerebrum at ED 20. Data are expressed as mean ± SE. Number of animals in each group is 6. Where * is a highly significant (P < .01) change between the control and treated group. ED indicates embryonic day; LPO, lipid peroxidation; NO, nitric oxide; SE, standard error.
Discussion
The gestational administrations of ATO (5 or 10 mg/kg) from GD 1 to 20 induced a hypothyroidism/thyrotoxicity in both dams and fetuses at GD 20. This state was evident by elevated level of serum TSH, along with the declined levels of serum T4 and T3. Also, some histological alterations such as a flattened cell lining epithelium, hyperplasia, edema, atrophy, colloid vacuoles, and destructive degeneration were observed in the fetal thyroid gland at ED 20. Previously, ATO accumulates in the thyroid follicles70 and binds the protein sulfhydryl group of thyroid peroxidase (THs synthesis)20,71 decreasing THs levels,13,72 altering THs-metabolism,73 perturbing thyroid receptors (TRs),72,74 and thus initiating goiter.75 Another possible mechanism is that exposure to arsenic can decrease the levels of selenium (necessary for conversion of T4 to T3) in blood and tissue causing a negative action on thyroid functions.76,77 These results suggest that gestational ATO may cause maternofetal thyroid dyshormonogenesis, fetal thyroid dysgenesis, and fetal pituitary-thyroid axis dysregulation.
The current study demonstrated that administrations of ATO during pregnancy induced a marked reduction in the weight of both dams and fetuses at GD 20. This disturbance may reduce the uteroplacental transfer, interrupt the pregnancy, and delay the fetal development. Maternal and fetal death was observed in both ATO-treated groups while abortion was only recorded in the ATO-high treated group. Alternatively, the maternofetal hypothyroidism in both ATO-experimental groups resulted in a substantial reduction in the values of fetal serum GH, IGF-I, and IGF-II at ED 20. Interestingly, gestational ATO-high dose group (10 mg/kg) effects were more severe than those in the gestational ATO-low dose group (5 mg/kg). These findings may reflect the toxicity and teratogenic outcomes of gestational ATO. In parallel, rats exposed to 1, 2.5, 5, or 10 mg/kg ATO displayed a diminution in the maternal food intake and in the body weight of both dams and their fetuses.28 Epidemiological and animal studies have proposed that arsenic penetrates the placenta22 causing a spontaneous abortion, low maternal food consumption, stillbirth, low maternofetal body weight, preterm delivery, and developmental retardation in human,24,27 in rats,19,78,79 and in mice.11,79 These complications could be attributed to the following causes: (1) accumulation of arsenic in the placenta might reduce the placental blood flow and disturb the placental vasculogenesis causing growth retardation11,19,23; and (2) growth retardation due to exposure to arsenic might be mediated, at least partly, by inhibiting mRNA expression of IGF-I and its activity.80 Also, the maternofetal hypothyroidism described here may be associated with a reduction of GH and IGF-I/IGF-II, and intensification in the ROS levels that may retard the fetal growth. On the basis of these data, it can be inferred that (1) the gestational administrations of ATO may induce imbalance between the hormonal system (hypothalamic–pituitary–thyroid axis) and growth axis, and (2) The dysregulation due to the gestational ATO may disrupt the dam appetite, increase the risk of mortality, and cause several developmental disorders.
The histopathological findings showed an ATO dose-dependent vacuolar degeneration, hyperplastic proliferation, congested blood vessels, pericellular edema, and destructive changes at ED 20 in the fetal cerebrum sections of maternal ATO-treated groups. The observed cortical deformation and neurodegeneration may be attributed to (1) elevation in the levels of LPO, NO, and H2O2; (2) attenuation of mRNA expression of Nrf2 and PPARγ; (3) upregulation of mRNA expression of caspase-3, NF-κB, Cox2, BAX, and iNOS; and (4) maternofetal hypothyroidism/thyrotoxicity and fetal growth dysfunction. In parallel, the fetal brain weight was decreased in both maternal ATO-treated groups compared to the equivalent averages of the corresponding control group. Concomitantly, arsenic accumulates in the brain81 initiating the excess formation of ROS82 such as ·OH, H2O2 and MDA,83 damaging the cell membrane and mitochondrial function,33 diminishing DNA and protein synthesis,19 and disrupting the DNA methylation.43 These abnormalities could induce apoptosis in a caspase-3,33,84 BAX,31,85 Cox2,86,87 NF-κB,88,89 and mitogen-activated protein kinase independent manners,32,90 and inhibit PPAR-γ activity,39,91,92 Nrf-2 expression,4,39,40 and PI3K)/AKT phosphorylation pathways.4,41 Additionally, Hu et al3 reported that the activation of NF-κB signaling by ATO induces the inflammatory damage pathways. Moreover, the elevation in NO level in the present study may be the result of the upregulation of mRNA expression of iNOS and NF-κB, and a downregulation of mRNA expression of Nrf2 and PPARγ.14 This alteration may result in the degree of degeneration in the fetal cerebrum at ED 20. On the other hand, oxidative DNA damage can induce several pathological changes in the brain of mice.93 Another interesting interpretation is that hypothyroidism can retard the developing brain via distorting the peripheral THs levels and TRs expression.46,94,95 These mechanisms could be explained by the neuronal degeneration in the cerebrum82 and hippocampus of mice,85 neuronal destruction in guinea pig,96 axons demyelination,97 neurotoxicity43 and neuropsychological deficits in rats,98 and mental retardation in human infancy.99 In clinical trials and animal models, chronic exposure to arsenic can impair the neurocognitive behaviors37 and cause neurotoxicity.36 This disruption may directly or indirectly delay the morphogenesis and functional development of thyroid-cerebrum axis, and the differentiation of the fetal neuroendocrine system. From the aforementioned interpretations and the current view, it can be concluded that the gestational ATO may cause fetal toxicity and neuroendocrine dysfunction.
Conclusion and Future Direction
Gestational exposure of ATO initiated a hypothyroidism/thyrotoxicity in both dams and fetuses indicating a stress-responsive aspect for a fetal thyroid-cerebrum axis and in general prenatal development. Gestational ATO accumulated and concentrated in the fetal thyroid and cerebrum causing several histopathological lesions in these tissues. These disruptions appear to rely on augmentation of ROS (LPO, NO, and H2O2), suppression of mRNA expression of Nrf2 and PPARγ, and upregulation of mRNA expression of caspase-3, NF-κB, Cox2, BAX, and iNOS in the fetal cerebrum. A novel interpretation of this study is that the gestational ATO may disrupt thyroid-cerebrum axis generating fetal neurodevelopmental toxicity (Figure 5). The variations were dose-dependent for all estimated markers except for the dead of pregnant dams, where the alterations were similar in both treated groups. However, these disorders may be dependent on the experimental design, developmental period, concentration, absorption, and metabolism of ATO. Additional studies are warranted to discovering the detailed molecular mechanisms of the gestational ATO-induced apoptosis to avoid any disturbance in the fetal and neonatal development.
Figure 5.
Diagram about the gestational As2O3 and fetal neuroendocrine disruptions.
Acknowledgments
The authors acknowledge all the staff in their departments for general assistance. The authors also pleasantly thank Dr Manushree Bharadwaj (National Institute of Environmental Health Sciences, Durham, North Carolina) and Prof Dr Douglas B. Learn (Director of Photobiology and Cellular Therapeutic Safety at Charles River Laboratories, Pennsylvania) for their assistance.
Footnotes
Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.
References
- 1. Madhyastha H, Madhyastha R, Nakajima Y, Maruyama M. Deciphering the molecular events during arsenic induced transcription signal cascade activation in cellular milieu. Biometals. 2018;31(1):7–15. [DOI] [PubMed] [Google Scholar]
- 2. Mohammadi Kian M, Mohammadi S, Tavallaei M, et al. Inhibitory effects of arsenic trioxide and thalidomide on angiogenesis and vascular endothelial growth factor expression in leukemia cells. Asian Pac J Cancer Prev. 2018;19(4):1127–1134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Hu Y, Wei M, Niu Q, et al. Grape seed proanthocyanidin extract alleviates arsenic-induced lung damage through NF-κB signaling. Exp Biol Med. 2019;244(3):213–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Thangapandiyan S, Ramesh M, Hema T, et al. Sulforaphane potentially ameliorates arsenic induced hepatotoxicity in albino Wistar rats: implication of PI3K/Akt/Nrf2 signaling pathway. Cell Physiol Biochem. 2019;52(5):1203–1222. [DOI] [PubMed] [Google Scholar]
- 5. Jeong CH, Seok JS, Petriello MC, Han SG. Arsenic downregulates tight junction claudin proteins through p38 and NF-κB in intestinal epithelial cell line, HT-29. Toxicology. 2017;379:31–39. [DOI] [PubMed] [Google Scholar]
- 6. Jia C, Wei Y, Land Y, et al. Comprehensive analysis of the metabolomic characteristics on the health lesions induced by chronic arsenic exposure: a metabolomics study. Int J Hyg Environ Health. 2019;222(3):434–445. [DOI] [PubMed] [Google Scholar]
- 7. Szymkowicz DB, Sims KC, Schwendinger KL, et al. Exposure to arsenic during embryogenesis impairs olfactory sensory neuron differentiation and function into adulthood. Toxicology. 2019;420:73–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Twaddle NC, Vanlandingham M, Beland FA, Fisher JW, Doerge DR. Metabolism and disposition of arsenic species from oral dosing with sodium arsenite in neonatal CD-1 mice IV toxicokinetics following gavage administration and lactational transfer. Food Chem Toxicol. 2019;123:28–41. [DOI] [PubMed] [Google Scholar]
- 9. National Research Council. Arsenic in Drinking Water 2001 Update. Washington, DC: National Academy Press; 2001. [Google Scholar]
- 10. Brwown KG, Ross GL. Arsenic, drinking water, and health: a position paper of the American Council on Sciences and Health. Regul Toxicol Pharmacol. 2002;36(2):162–174. [DOI] [PubMed] [Google Scholar]
- 11. He W, Greenwell RJ, Brooks DM, Calderón-Garcidueñas L, Beall HD, Coffin JD. Arsenic exposure in pregnant mice disrupts placental vasculogenesis and causes spontaneous abortion. Toxicol Sci. 2007;99(1):244–253. [DOI] [PubMed] [Google Scholar]
- 12. Watanabe T, Hirano S. Metabolism of arsenic and its toxicological relevance. Arch Toxicol. 2013;87(6):969–979. [DOI] [PubMed] [Google Scholar]
- 13. Mohanta RK, Garg AK, Dass RS, Behera SK. Blood biochemistry, thyroid hormones, and oxidant/antioxidant status of guinea pigs challenged with sodium arsenite or arsenic trioxide. Biol Trace Elem Res. 2014;160(2):238–244. [DOI] [PubMed] [Google Scholar]
- 14. Li S, Wang Y, Zhao H, et al. NF-κB-mediated inflammation correlates with calcium overload under arsenic trioxide-induced myocardial damage in Gallus gallus . Chemosphere. 2017;185:618–627. [DOI] [PubMed] [Google Scholar]
- 15. Moore CL, Flanigan TJ, Law CD, et al. Developmental neurotoxicity of inorganic arsenic exposure in Sprague Dawley rats. Neurotoxicol Teratol. 2019;72:49–57. [DOI] [PubMed] [Google Scholar]
- 16. Castro-Coronel Y, Del Razo LM, Huerta M, Hernandez-Lopez A, Ortega A, López-Bayghen E. Arsenite exposure downregulates EAAT1/GLAST transporter expression in glial cells. Toxicol Sci. 2011;122(2):539–550. [DOI] [PubMed] [Google Scholar]
- 17. Argos M, Ahsan H, Graziano JH. Arsenic and human health: epidemiologic progress and public health implications. Rev Environ Health. 2012;27(4):191–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. ATSDR. Toxicological profile for arsenic Agency for Toxic Substances and Disease Registry, US Department of Health and Human Services. Atlanta, GA, 2007. http://wwwatsdrcdcgov/ToxProfiles/tp2pdf. Accessed July 27, 2011. [PubMed] [Google Scholar]
- 19. Wang A, Holladay SD, Wolf DC, Ahmed SA, Robertson JL. Reproductive and developmental toxicity of arsenic in rodents: a review. Int J of Toxicol. 2006;25(5):319–331. [DOI] [PubMed] [Google Scholar]
- 20. Palazzolo DL, Ely EA. Arsenic trioxide and reduced glutathione act synergistically to augment inhibition of thyroid peroxidase activity in vitro. Biol Trace Elem Res. 2015;165(1):110–117. [DOI] [PubMed] [Google Scholar]
- 21. Glatter E, Mravcova A, Lencer J, Vobecky M, Egertova E, Mysliveckova M. Study of the distribution and interaction of arsenic and selenium in the thyroid. Biol Trace Elem Res. 1995;49(2-3):177–186. [DOI] [PubMed] [Google Scholar]
- 22. Jin Y, Xi S, Li X, et al. Arsenic speciation transported through the placenta from mother mice to their newborn pups. Environ Res. 2006;101(3):349–355. [DOI] [PubMed] [Google Scholar]
- 23. Hopenhayn C, Ferreccio C, Browning SR, et al. Arsenic exposure from drinking water and birth weight. Epidemiology. 2003;14(5):593–602. [DOI] [PubMed] [Google Scholar]
- 24. von Ehrenstein OS, Guha Mazumder DN, Hira-Smith M, et al. Pregnancy outcomes, infant mortality, and arsenic in drinking water in West Bengal, India. Am J Epidemiol. 2006;163(7):662–669. [DOI] [PubMed] [Google Scholar]
- 25. Aschengrau A, Zierler S, Cohen A. Quality of community drinking water and the occurrence of spontaneous abortion. Arch Environ Health. 1989;44(5):283–290. [DOI] [PubMed] [Google Scholar]
- 26. Hopenhayn-Rich C, Browning SR, Hertz-Picciotto I, Ferreccio C, Peralta C, Gibb H. Chronic arsenic exposure and risk of infant mortality in two areas of Chile. Environ Health Perspect. 2000;108(7):667–673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Raqib R, Ahmed S, Sultana R, et al. Effects of in utero arsenic exposure on child immunity and morbidity in rural Bangladesh. Toxicol Lett. 2009;185(3):197–202. [DOI] [PubMed] [Google Scholar]
- 28. Holson JF, Stump DG, Clevidence KJ, Knapp JF, Farr CH. Evaluation of the prenatal developmental toxicity of orally administered arsenic trioxide in rats. Food Chem Toxicol. 2000;38(5):459–466. [DOI] [PubMed] [Google Scholar]
- 29. Rodríguez VM, Carrizales L, Mendoza MS, Fajardo OR, Giordano M. Effects of sodium arsenite exposure on development and behavior in the rat. Neurotoxicol Teratol. 2002;24(6):743–750. [DOI] [PubMed] [Google Scholar]
- 30. Flora SJ, Bhatt K, Mehta A. Arsenic moiety in gallium arsenide is responsible for neuronal apoptosis and behavioral alterations in rats. Toxicol Appl Pharmacol. 2009;240(2):236–244. [DOI] [PubMed] [Google Scholar]
- 31. Zhang W, Feng H, Gao Y, et al. Role of pigment epithelium-derived factor (PEDF) in arsenic-induced cell apoptosis of liver and brain in a rat model. Biol Trace Elem Res. 2013;151(2):269–276. [DOI] [PubMed] [Google Scholar]
- 32. Yen CC, Ho TJ, Wu CC, et al. Inorganic arsenic causes cell apoptosis in mouse cerebrum through an oxidative stress-regulated signaling pathway. Arch Toxicol. 2011;85(6):565–575. [DOI] [PubMed] [Google Scholar]
- 33. Atashrazm F, Lowenthal RM, Dickinson JL, Holloway AF, Woods GM. Fucoidan enhances the therapeutic potential of arsenic trioxide and all-trans retinoic acid in acute promyelocytic leukemia, in vitro and in vivo. Oncotarget. 2016;7(29):46028–46041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Zhang J, Liu X, Zhao L, Hu S, Li S, Piao F. Subchronic exposure to arsenic disturbed the biogenic amine neurotransmitter level and the mRNA expression of the synthetase in mice brains. Neuroscience. 2013;41:2–8. [DOI] [PubMed] [Google Scholar]
- 35. Rodríguez VM, Carrizales L, Jiménez-Capdeville ME, Dufour L, Giordano M. The effects of sodium arsenite exposure on behavioral parameters in the rat. Brain Res Bull. 2001;55(2):301–308. [DOI] [PubMed] [Google Scholar]
- 36. Chattopadhyay S, Bhaumik S, Nag Chaudhury A, Das Gupta S. Arsenic induced changes in growth development and apoptosis in neonatal and adult brain cells in vivo and in tissue culture. Toxicol Lett. 2002;128(1-3):73–84. [DOI] [PubMed] [Google Scholar]
- 37. Tsai SY, Chou HY, The HW, Chen CM, Chen CJ. The effects of chronic arsenic exposure from drinking water on the neurobehavioral development in adolescence. Neurotoxicol. 2003;24(4-5):747–753. [DOI] [PubMed] [Google Scholar]
- 38. Sabir S, Akash MSH, Fiayyaz F, Saleem U, Mehmood MH, Rehman K. Role of cadmium and arsenic as endocrine disruptors in the metabolism of carbohydrates: inserting the association into perspectives. Biomed Pharmacother. 2019;114:108802. [DOI] [PubMed] [Google Scholar]
- 39. Bai J, Yao X, Jiang L, et al. Taurine protects against As2O3-induced autophagy in livers of rat offspring through PPARγ pathway. Sci Rep. 2016;6:27733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Mukherjee AA, Kandhare AD, Bodhankar SL. Elucidation of protective efficacy of pentahydroxy flavone isolated from Madhuca indica against arsenite-induced cardiomyopathy: role of Nrf-2, PPAR-γ, c-fos and c-jun. Environ Toxicol Pharmacol. 2017;56:172–185. [DOI] [PubMed] [Google Scholar]
- 41. Mann KK, Colombo M, Miller WH., Jr Arsenic trioxide decreases AKT protein in a caspase-dependent manner. Mol Cancer Ther. 2008;7(6):1680–1687. [DOI] [PubMed] [Google Scholar]
- 42. Herrera A, Pineda J, Antonio MT. Toxic effects of perinatal arsenic exposure on the brain of developing rats and the beneficial role of natural antioxidants. Environ Toxicol Pharmacol. 2013;36(1):73–79. [DOI] [PubMed] [Google Scholar]
- 43. Du X, Tian M, Wang X, et al. Cortex and hippocampus DNA epigenetic response to a long-term arsenic exposure via drinking water. Environ Pollut. 2018;234:590–600. [DOI] [PubMed] [Google Scholar]
- 44. Morte B, Gil-Ibáñez P, Bernal J. Regulation of gene expression by thyroid hormone in primary astrocytes: factors influencing the genomic response. Endocrinology. 2018;159(5):2083–2092. [DOI] [PubMed] [Google Scholar]
- 45. Candelotti E, De Vito P, Ahmed RG, et al. Thyroid hormones crosstalk with growth factors: old facts and new hypotheses. Immunol Endocr Metab Agents Med Chem. 2015;15(1):71–85. [Google Scholar]
- 46. Van Herck SLJ, Geysens S, Bald E, et al. Maternal transfer of methimazole and effects on thyroid hormone availability in embryonic tissues. J Endocrinol. 2013;218(1):105–115. [DOI] [PubMed] [Google Scholar]
- 47. Ahmed OM, Ahmed RG, El-Gareib AW, El-Bakry AM, Abd El-Tawab SM. . Effects of experimentally induced maternal hypothyroidism and hyperthyroidism on the development of rat offspring: II-The developmental pattern of neurons in relation to oxidative stress and antioxidant defense system. Int J Dev Neurosci. 2012;30(6):517–537. [DOI] [PubMed] [Google Scholar]
- 48. Ahmed RG, Incerpi S. Gestational doxorubicin alters fetal thyroid-brain axis. Int J Dev Neurosci. 2013;31(2):96–104. [DOI] [PubMed] [Google Scholar]
- 49. El-bakry AM, El-Ghareeb AW, Ahmed RG. Comparative study of the effects of experimentally-induced hypothyroidism and hyperthyroidism in some brain regions in albino rats. Int J Dev Neurosci. 2010;28(5):371–389. [DOI] [PubMed] [Google Scholar]
- 50. Ahmed RG, El-Gareib AW. Maternal carbamazepine alters fetal neuroendocrine-cytokines axis. Toxicology. 2017;382:59–66. [DOI] [PubMed] [Google Scholar]
- 51. Ahmed RG, El-Gareib AW, Shaker HM. Gestational 3,3′,4,4′,5-pentachlorobiphenyl (PCB 126) exposure disrupts fetoplacental unit: fetal thyroid-cytokines dysfunction. Life Sci. 2018;192:213–220. [DOI] [PubMed] [Google Scholar]
- 52. Ahmed RG, Walaa GH, Asmaa FS. Suppressive effects of neonatal bisphenol A on the neuroendocrine system. Toxicol Ind Health. 2018;34(6):397–407. [DOI] [PubMed] [Google Scholar]
- 53. Ahmed RG. Overdoses of acetaminophen disrupt the thyroid-liver axis in neonatal rats. Endocr Metab Immune Disord Drug Targets. 2019;19(5):705–714. [DOI] [PubMed] [Google Scholar]
- 54. Marcondes FK, Bianchi FJ, Tanno AP. Determination of the estrous cycle phases of rats: some helpful considerations. Braz J Biol. 2002;62(4A):609–614. [DOI] [PubMed] [Google Scholar]
- 55. Ahmed RG. Gestational caffeine exposure acts as a fetal thyroid-cytokine disruptor by activating caspase-3/BAX/Bcl-2/Cox2/NF-κB at ED 20. Toxicol Res. 2019;8(2):196–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Lu M, Wang H, Li XF, et al. Evidence of hemoglobin binding to arsenic as a basis for the accumulation of arsenic in rat blood. Chem Res Toxicol. 2004;17(12):1733–1742. [DOI] [PubMed] [Google Scholar]
- 57. Ahmed RG. Perinatal TCDD exposure alters developmental neuroendocrine system. Food Chem Toxicol. 2011;49(6):1276–1284. [DOI] [PubMed] [Google Scholar]
- 58. Ahmed RG. Early weaning PCB 95 exposure alters the neonatal endocrine system: thyroid adipokine dysfunction. J Endocrinol. 2013;219(3):205–215. [DOI] [PubMed] [Google Scholar]
- 59. Ahmed RG. Maternal bisphenol A alters fetal endocrine system: thyroid adipokine dysfunction. Food Chem Toxicol. 2016;95:168–174. [DOI] [PubMed] [Google Scholar]
- 60. Ahmed RG. Gestational dexamethasone alters fetal neuroendocrine axis. Toxicol Lett. 2016;258:46–54. [DOI] [PubMed] [Google Scholar]
- 61. Ahmed RG, Abdel-Latif M, Ahmed F. Protective effects of GM-CSF in experimental neonatal hypothyroidism. Int Immunopharmacol. 2015;29(2):538–543. [DOI] [PubMed] [Google Scholar]
- 62. Ahmed OM, Abd El-Tawab SM, Ahmed RG. Effects of experimentally induced maternal hypothyroidism and hyperthyroidism on the development of rat offspring: I. The development of the thyroid hormones-neurotransmitters and adenosinergic system interactions. Int J Dev Neurosci. 2010;28(6):437–454. [DOI] [PubMed] [Google Scholar]
- 63. Ahmed RG, Abdel-Latif M, Emad M, Khalid El-N. Immune stimulation improves endocrine and neural fetal outcomes in a model of maternofetal thyrotoxicosis. Int Immunopharmacol. 2015;29(2):714–721. [DOI] [PubMed] [Google Scholar]
- 64. Bancroft JD, Gamble M. Theory and Practice of Histological Techniques 6th ed Philadelphia, PA: Churchill Livingstone/Elsevier; 2008. [Google Scholar]
- 65. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-delta delta C(T)) method. Methods. 2001;25(4):402–408. [DOI] [PubMed] [Google Scholar]
- 66. Preuss HG, Jarrel ST, Seheckenbach R, Lieberman S, Anderson RA. Comparative effects of chromium, vanadium and gymnema sylvestre on sugar-induced blood pressure elevations in SHR. J Am Coll Nutr. 1998;17(2):116–123. [DOI] [PubMed] [Google Scholar]
- 67. Dutta A, Sarkar D, Gurib-Fakim A, Mandal C, Chatterjee M. In vitro and in vivo activity of aloe vera leaf exudate in experimental visceral leishmaniasis. Parasitol Res. 2008;102(6):1235–1242. [DOI] [PubMed] [Google Scholar]
- 68. Sergiev I, Alexieva V, Karanov E. Effect of spermine, atrazine and combination between them on some endogenous protective systems and stress markers in plants. Proceedings of the Bulgarian Acad of Sci. 1997;51:121–124. [Google Scholar]
- 69. Roa M, Blane K, Zonneberg M. One-Way Analysis, version 1A(C) PC-STAT. Athens, Georgia: University of Georgia; 1985. [Google Scholar]
- 70. Kotyzova D, Eybl V, Mihaljevic M, Glattre E. Effect of long-term administration of arsenic (III) and bromine with and without selenium and iodine supplementation on the element level in the thyroid of rat. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 2005;149(2):329–333. [DOI] [PubMed] [Google Scholar]
- 71. Palazzolo DL, Jansen KP. The minimal arsenic concentration required to inhibit the activity of thyroid peroxidase in vitro. Biol Trace Elem Res. 2008;126(1-3):49–55. [DOI] [PubMed] [Google Scholar]
- 72. Sun HJ, Xiang P, Tang MH, Sun L, Ma LQ. Arsenic impacted the development, thyroid hormone and gene transcription of thyroid hormone receptors in bighead carp larvae (Hypophthalmichthys nobilis). J Hazard Mater. 2016;303:76–82. [DOI] [PubMed] [Google Scholar]
- 73. Meltzer HM, Maage A, Ydersbond TA, Haug E, Glattre E, Holm H. Fish arsenic may influence human blood arsenic, selenium, and T4: T3 ratio. Biol Trace Elem Res. 2002;90(1-3):83–98. [DOI] [PubMed] [Google Scholar]
- 74. Takagi S, Hummel BC, Walfish PG. Thioamides and arsenite inhibit specific T3 binding to the hepatic nuclear receptor. Biochem Cell Biol. 1990;68(3):616–621. [DOI] [PubMed] [Google Scholar]
- 75. Chang TC, Hong MC, Chen CJ. Higher prevalence of goiter in endemic area of blackfoot disease of Taiwan. J Formos Med Assoc. 1991;90(10):941–946. [PubMed] [Google Scholar]
- 76. Gailer J. Chronic toxicity of AsIII in mammals: the role of (GS)(2)AsSe(-). Biochimie. 2009;91(10):1268–1272. [DOI] [PubMed] [Google Scholar]
- 77. George CM, Gamble M, Slavkovich V, et al. A cross-sectional study of the impact of blood selenium on blood and urinary arsenic concentrations in Bangladesh. Environ Health. 2013;12(1):52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Holson JF, Desesso JM, Jacobson CF, Farr CH. Appropriate use of animal models in the assessment of risk during prenatal development: an illustration using inorganic arsenic. Teratology. 2000;62(1):51–71. [DOI] [PubMed] [Google Scholar]
- 79. DeSesso JM. Teratogen update: inorganic arsenic. Teratology. 2001;64(3):170–173. [DOI] [PubMed] [Google Scholar]
- 80. Ahmed S, Rekha RS, Ahsan KB, et al. Arsenic exposure affects plasma insulin-like growth factor 1 (IGF-1) in children in rural Bangladesh. PLoS One. 2013;8(11):e81530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Xi S, Guo L, Qi R, Sun W, Jin Y, Sun G. Prenatal and early life arsenic exposure induced oxidative damage and altered activities and mRNA expressions of neurotransmitter metabolic enzymes in offspring rat brain. J Biochem Mol Toxicol. 2010;24(6):368–378. [DOI] [PubMed] [Google Scholar]
- 82. Piao F, Ma N, Hiraku Y, et al. Oxidative DNA damage in relation to neurotoxicity in the brain of mice exposed to arsenic at environmentally relevant levels. J Occup Health. 2005;47(5):445–449. [DOI] [PubMed] [Google Scholar]
- 83. Jomova K, Jenisova Z, Feszterova M, et al. Arsenic: toxicity, oxidative stress and human disease. J Appl Toxicol. 2011;31(2):95–107. [DOI] [PubMed] [Google Scholar]
- 84. Kang YH, Yi MJ, Kim MJ, et al. Caspase-independent cell death by arsenic trioxide in human cervical cancer cells: reactive oxygen species-mediated poly(ADP-ribose) polymerase-1 activation signals apoptosis-inducing factor release from mitochondria. Cancer Res. 2004;64(24):8960–8967. [DOI] [PubMed] [Google Scholar]
- 85. Wang Y, Bai C, Guan H, et al. Subchronic exposure to arsenic induces apoptosis in the hippocampus of the mouse brains through the Bcl-2/Bax pathway. J Occup Health. 2015;57(3):212–221. [DOI] [PubMed] [Google Scholar]
- 86. Tsai SH, Liang YC, Chen L, Ho FM, Hsieh MS, Lin JK. Arsenite stimulates cyclooxygenase-2 expression through activating IkappaB kinase and nuclear factor kappaB in primary and ECV304 endothelial cells. J Cell Biochem. 2002;84(4):750–758. [DOI] [PubMed] [Google Scholar]
- 87. Trouba KJ, Germolec DR. Micromolar concentrations of sodium arsenite induce cyclooxygenase-2 expression and stimulate p42/44 mitogen-activated protein kinase phosphorylation in normal human epidermal keratinocytes. Toxicol Sci. 2004;79(2):248–257. [DOI] [PubMed] [Google Scholar]
- 88. Wijeweera JB, Gandolfi AJ, Parrish A, Lantz RC. Sodium arsenite enhances AP-1 and NFκB DNA binding and induces stress protein expression in precision-cut rat lung slices. Toxicol Sci. 2001;61(2):283–294. [DOI] [PubMed] [Google Scholar]
- 89. Zhang K, Zhao P, Guo G, et al. Arsenic trioxide attenuates NF-κB and cytokine mRNA levels in the livers of cocks. Biol Trace Elem Res. 2016;170(2):432–437. [DOI] [PubMed] [Google Scholar]
- 90. Wang X, Meng D, Chang Q, et al. Arsenic inhibits neurite outgrowth by inhibiting the LKB1-AMPK signaling pathway. Environ Health Perspect. 2010;118(5):627–634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Wauson EM, Langan AS, Vorce RL. Sodium arsenite inhibits and reverses expression of adipogenic and fat cell-specific genes during in vitro adipogenesis. Toxicol Sci. 2002;65(2):211–219. [DOI] [PubMed] [Google Scholar]
- 92. Yadav S, Anbalagan M, Shi Y, Wang F, Wang H. Arsenic inhibits the adipogenic differentiation of mesenchymal stem cells by down-regulating peroxisome proliferator-activated receptor gamma and CCAAT enhancer-binding proteins. Toxicol In Vitro. 2013;27(1):211–219. [DOI] [PubMed] [Google Scholar]
- 93. Ding X, Su Q, Jiang M, Xie H, Cong J, Wang L. Arsenic affects on cerebellar development of mice. Toxicol Mech Methods. 2013;23(9):672–677. [DOI] [PubMed] [Google Scholar]
- 94. Ahmed OM, El-Gareib AW, El-Bakry AM, Abd El-Tawab SM, Ahmed RG. Thyroid hormones states and brain development interactions. Int J Dev Neurosci. 2008;26(2):147–209. [DOI] [PubMed] [Google Scholar]
- 95. Ahmed RG. Hypothyroidism and brain developmental players. Thyroid Res. 2015;8(2):1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Pachauri V, Mehta A, Mishra D, Flora SJ. Arsenic induced neuronal apoptosis in guinea pigs is Ca2+ dependent and abrogated by chelation therapy: role of voltage gated calcium channels. Neurotoxicology. 2013;35:137–145. [DOI] [PubMed] [Google Scholar]
- 97. García-Chávez E, Segura B, Merchant H, Jiménez I, Del Razo LM. Functional and morphological effects of repeated sodium arsenite exposure on rat peripheral sensory nerves. J Neurol Sci. 2007;258(1-2):104–110. [DOI] [PubMed] [Google Scholar]
- 98. Gumilar F, Lencinas I, Bras C, Giannuzzi L, Minetti A. Locomotor activity and sensory–motor developmental alterations in rat offspring exposed to arsenic prenatally and via lactation. Neurotoxicol Teratol. 2015;49:1–9. [DOI] [PubMed] [Google Scholar]
- 99. Dakeishi M, Murata K, Grandjean P. Long-term consequences of arsenic poisoning during infancy due to contaminated milk powder. Environ Health. 2006;5:31. [DOI] [PMC free article] [PubMed] [Google Scholar]