Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
J Appl Toxicol ; 44(7): 965-977, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38419361

ABSTRACT

Triphenyl phosphate (TPhP) is an organophosphate flame retardant and plasticizer that is added to a wide variety of consumer and industrial products. It is also a ubiquitous environmental pollutant. Exposure to TPhP has been shown to alter gene expression in metabolic and estrogenic signaling pathways in in vitro and in vivo models of a variety of species, and as such, is considered to be an endocrine disrupting chemical. Exposure to endocrine disrupting chemicals is increasingly being associated with changes to the epigenome, especially during embryonic development. The aim of this study was to evaluate whether TPhP exposure in aquatic ecosystems has the ability to alter the epigenome in two immortal cell lines derived from trout (Oncorhynchus mykiss). This study assessed whether 24 h exposure to TPhP resulted in changes to histone modification and DNA methylation profiles in steelhead trout embryonic cells and rainbow trout gill epithelial cells. Results show that several epigenetic modifications on histone H3 and DNA methylation are altered in the embryonic cells following TPhP exposure, but not in the gill epithelial cells. Specifically, histone H3 acetylation, histone H3 mono-methylation and global DNA methylation were found to be reduced. The alterations of these epigenetic modification profiles in the embryonic cells suggest that exposure to TPhP during fetal development may alter gene expression in the developing embryo, likely in metabolic and estrogenic pathways. The impacts to the epigenome determined in this study may even carry multigenerational detrimental effects on human and ecosystem health, which requires further investigation.


Subject(s)
DNA Methylation , Flame Retardants , Oncorhynchus mykiss , Organophosphates , Water Pollutants, Chemical , Animals , Flame Retardants/toxicity , Oncorhynchus mykiss/embryology , Oncorhynchus mykiss/genetics , DNA Methylation/drug effects , Organophosphates/toxicity , Water Pollutants, Chemical/toxicity , Epigenome/drug effects , Cell Line , Endocrine Disruptors/toxicity , Gills/drug effects , Gills/metabolism , Epigenesis, Genetic/drug effects , Embryo, Nonmammalian/drug effects , Histones/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism
2.
J Appl Toxicol ; 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39295171

ABSTRACT

The reliability of relative quantification RT-qPCR depends upon the gene of interest being normalized to one or more reference genes, with the assumption that the chosen reference genes do not experience altered expression with experimental conditions. The correct choice of stable reference genes is critical when investigating alterations to gene transcript levels following exposure to endocrine and metabolic disrupting chemicals, such as the flame retardant triphenyl phosphate (TPhP). This study assessed the stability of eight reference genes following TPhP exposure in embryonic cells derived from rainbow trout (Oncorhynchus mykiss). The genes ß-actin (actb) and 18s rRNA (18s) were stable, while glyceraldehyde-3-phosphate dehydrogenase (gapdh) relative expression was found to be increased. gapdh is a popular reference gene and has been previously used in the literature for investigating TPhP exposure in teleost fish models. We discuss the implications of gapdh upregulation in the context of TPhP as a metabolic disrupting chemical. Furthermore, we quantified the expression of the tumor suppressor gene p53 following TPhP exposure in relation to different reference genes to use as an example to report on how discrepancies in findings might arise depending on the stability of the chosen reference gene.

3.
Environ Res ; 205: 112435, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34843719

ABSTRACT

Pregnancy is a complex process requiring tremendous physiological changes in the mother in order to fulfill the needs of the growing fetus, and to give birth, expel the placenta and nurse the newborn. These physiological modifications are accompanied with psychological changes, as well as with variations in habits and behaviors. As a result, this period of life is considered as a sensitive window as impaired functional and physiological changes in the mother can have short- and long-term impacts on her health. In addition, dysregulation of the placenta and of mechanisms governing placentation have been linked to chronic diseases later-on in life for the fetus, in a concept known as the Developmental Origin of Health and Diseases (DOHaD). This concept stipulates that any change in the environment during the pre-conception and perinatal (in utero life and neonatal) period to puberty, can be "imprinted" in the organism, thereby impacting the health and risk of chronic diseases later in life. Pregnancy is a succession of events that is regulated, in large part, by hormones and growth factors. Therefore, small changes in hormonal balance can have important effects on both the mother and the developing fetus. An increasing number of studies demonstrate that exposure to endocrine disrupting compounds (EDCs) affect both the mother and the fetus giving rise to growing concerns surrounding these exposures. This review will give an overview of changes that happen during pregnancy with respect to the mother, the placenta, and the fetus, and of the current literature regarding the effects of EDCs during this specific sensitive window of exposure.


Subject(s)
Mothers , Sexual Maturation , Female , Fetus , Humans , Infant, Newborn , Placenta/metabolism , Placentation , Pregnancy
4.
J Appl Toxicol ; 40(8): 1018-1032, 2020 08.
Article in English | MEDLINE | ID: mdl-32112456

ABSTRACT

Benzene is an environmental toxicant found in many consumer products. It is an established human carcinogen and is known to cause acute myeloid leukemia in adults. Epidemiological evidence has since shown that benzene can cross the placenta and affect the fetal liver. Animal studies have shown that in utero exposure to benzene can increase tumor incidence in offspring. Although there have been risk factors established for acute myeloid leukemia, they still do not account for many of the cases. Clearly then, current efforts to elucidate the mechanism by which benzene exerts its carcinogenic properties have been superficial. Owing to the critical role of cell signaling pathways in the development of an organism and its various organ systems, it seems plausible to suspect that these pathways may have a role in leukemogenesis. This review article assesses current evidence of the effects of benzene on critical hematopoietic signaling pathways. Pathways discussed included Hedgehog, Notch/Delta, Wingless/Integrated, nuclear factor-kappaB and others. Following a review of the literature, it seems that current evidence about the effects of benzene on these critical signaling pathways remains limited. Given the important role of these pathways in hematopoiesis, more attention should be given to them.


Subject(s)
Benzene/toxicity , Carcinogens/toxicity , Cell Differentiation/drug effects , Environmental Exposure/adverse effects , Hematopoiesis/drug effects , Leukemia/chemically induced , Signal Transduction/drug effects , Humans , Leukemia/physiopathology
5.
Biol Reprod ; 97(1): 143-152, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28859286

ABSTRACT

INTRODUCTION: Pre-eclampsia is associated with increased risk of subsequent cardiovascular and metabolic disease in the affected mothers. While aberrant inflammation contributes to the pathophysiology of pre-eclampsia, it is unclear whether maternal inflammation contributes to the increased risk of disease. Here, we determined the effect of aberrant inflammation in pregnancy on cardiovascular and metabolic disease risk factors. METHODS: Wistar rats were administered low doses of lipopolysaccharide (LPS) on gestational days (GD) 13.5-16.5 to induce inflammation. Controls included pregnant rats treated with saline and nonpregnant rats treated with LPS or saline. We previously showed that LPS-treated pregnant rats exhibit key features of pre-eclampsia. Echocardiographic parameters, heart weight, blood pressure, blood lipids, pulse-wave velocity, and glucose tolerance, were assessed at 16 weeks postpartum. Messenger RNA levels of transcription factors associated with cardiac growth were measured in left ventricular tissue; histone modifications and global DNA methylation were determined in hearts and livers at GD 17.5 and at 16 weeks postpartum. RESULTS: Compared with saline-treated pregnant rats and nonpregnant rats treated with LPS or saline, LPS-treated pregnant rats exhibited left ventricular hypertrophy and increased blood cholesterol and low-density lipoprotein levels at 16 weeks postdelivery. LPS-treated rats had increased left ventricular mRNA levels of hypertrophy-associated transcription factors at GD 17.5 and increased levels of modified histones in hearts and livers at GD 17.5 and 16 weeks postpartum. Other parameters remained unchanged. CONCLUSION: Aberrant inflammation during pregnancy results in persistent alterations in maternal physiological parameters and epigenetic modifications that could contribute to the pathophysiology of cardiovascular disease.


Subject(s)
Inflammation/chemically induced , Lipopolysaccharides/toxicity , Pregnancy Complications, Cardiovascular , Animals , Blood Pressure , DNA/genetics , DNA/metabolism , Echocardiography , Female , Gene Expression Regulation , Heart , Histones/genetics , Histones/metabolism , Inflammation/complications , Pregnancy , Rats , Rats, Wistar , Risk Factors
6.
Toxicol Appl Pharmacol ; 306: 69-78, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27381264

ABSTRACT

The teratogenicity of the antiepileptic drug valproic acid (VPA) is well established and its inhibition of histone deacetylases (HDAC) is proposed as an initiating factor. Recently, VPA-mediated HDAC inhibition was demonstrated to involve transcriptional downregulation of histone acetyltransferases (HATs), which was proposed to compensate for the increased acetylation resulting from HDAC inhibition. Cbp and p300 are HATs required for embryonic development and deficiencies in either are associated with congenital malformations and embryolethality. The objective of the present study was to characterize Cbp/p300 following VPA exposure in P19 cells. Consistent with previous studies, exposure to 5mM VPA over 24h induced a moderate decrease in Cbp/p300 mRNA, which preceded a strong decrease in total cellular protein mediated by ubiquitin-proteasome degradation. Nuclear Cbp/p300 protein was also decreased following VPA exposure, although to a lesser extent. Total cellular and nuclear p300 HAT activity was reduced proportionately to p300 protein levels, however while total cellular HAT activity also decreased, nuclear HAT activity was unaffected. Using the Cbp/p300 HAT inhibitor C646, we demonstrated that HAT inhibition similarly affected many of the same endpoints as VPA, including increased reactive oxygen species and caspase-3 cleavage, the latter of which could be attenuated by pre-treatment with the antioxidant catalase. C646 exposure also decreased NF-κB/p65 protein, which was not due to reduced mRNA and was not attenuated with catalase pre-treatment. This study provides support for an adaptive HAT response following VPA exposure and suggests that reduced Cbp/p300 HAT activity could contribute to VPA-mediated alterations.


Subject(s)
Anticonvulsants/pharmacology , Histone Acetyltransferases/metabolism , Valproic Acid/pharmacology , p300-CBP Transcription Factors/metabolism , Animals , Apoptosis/drug effects , Benzoates/pharmacology , Caspase 3/metabolism , Cell Line , Down-Regulation , Mice , Nitrobenzenes , Pyrazoles/pharmacology , Pyrazolones , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , p300-CBP Transcription Factors/antagonists & inhibitors , p300-CBP Transcription Factors/genetics
7.
J Appl Toxicol ; 36(8): 1015-24, 2016 08.
Article in English | MEDLINE | ID: mdl-26456483

ABSTRACT

Benzene is an environmental pollutant known to cause leukemia in adults, and may be associated with childhood leukemia. While the mechanisms of benzene-mediated carcinogenicity have not been fully elucidated, increased reactive oxygen species (ROS) and DNA damage are implicated. Sulforaphane (SFN) induces nuclear factor erythroid 2-related factor 2 (Nrf2), which contributes to SFN-mediated protection against carcinogenesis. We exposed cultured CD-1 mouse fetal liver cells to the benzene metabolite, benzoquinone, to determine its potential to cause DNA damage and alter DNA repair. Cells were also exposed to SFN to determine potential protective effects. Initially, cells were exposed to benzoquinone to confirm increased ROS and SFN to confirm Nrf2 induction. Subsequently, cells were treated with benzoquinone (with or without SFN) and levels of ROS, 8-hydroxy-2-deoxyguanosine (8-OHdG; marker of oxidative DNA damage), gamma histone 2A variant X (γH2AX; marker of DNA double-stranded breaks; DSBs) and transcript levels of genes involved in DNA repair were measured. Benzoquinone exposure led to a significant increase in ROS, which was not prevented by pretreatment with SFN or the antioxidative enzyme, catalase. DNA damage was increased after benzoquinone exposure, which was not prevented by SFN. Benzoquinone exposure significantly decreased the transcript levels of the critical base excision repair gene, 8-oxoguanine glycosylase (Ogg1), which was not prevented by SFN. The findings of this study demonstrate for the first time that DNA damage and altered DNA repair are a consequence of benzoquinone exposure in CD-1 mouse fetal liver cells and that SFN conferred little protection in this model. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Benzoquinones/toxicity , Hepatocytes/drug effects , Isothiocyanates/pharmacology , Liver/drug effects , 8-Hydroxy-2'-Deoxyguanosine , Animals , Antioxidants/pharmacology , Catalase/genetics , Catalase/metabolism , Cell Line , DNA Damage/drug effects , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA Repair , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Female , Histones/genetics , Histones/metabolism , Leukemia/blood , Leukemia/chemically induced , Liver/cytology , Male , Mice , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Sulfoxides
8.
Toxicol Appl Pharmacol ; 289(1): 12-9, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26341289

ABSTRACT

Exposure to the ubiquitous environmental pollutant benzene is positively correlated with leukemia in adults and may be associated with childhood leukemia following in utero exposure. While numerous studies implicate oxidative stress and DNA damage as playing a role in benzene-mediated carcinogenicity, emerging evidence suggests that alterations in epigenetic regulations may be involved. The present study aimed to determine whether DNA methylation and/or various histone modifications were altered following in utero benzene exposure in CD-1 mice. Global DNA methylation and promoter-specific methylation of the tumor suppressor gene, p15, were assessed. Additionally, levels of acetylated histones H3, H4, and H3K56, as well as methylated histones H3K9 and H3K27 were assessed by Western blotting. A significant decrease in global DNA methylation of maternal bone marrow was observed following benzene exposure; however no effect on global DNA methylation was detected in fetal livers. Additionally, no effect of benzene exposure was observed on p15 promoter methylation or any measured histone modifications in both maternal bone marrow and fetal livers. These results suggest that the methodology used in the present study did not reveal alterations in DNA methylation and histone modifications following in utero exposure to benzene; however further experimentation investigating these modifications at the whole genome/epigenome level, as well as at later stages of benzene-induced carcinogenesis, are warranted.


Subject(s)
Benzene/toxicity , DNA Methylation , Epigenesis, Genetic , Maternal Exposure/adverse effects , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cyclin-Dependent Kinase Inhibitor p15/metabolism , DNA Damage , Female , Fetus/drug effects , Fetus/metabolism , Histones/metabolism , Male , Mice , Pregnancy , Promoter Regions, Genetic
9.
Article in English | MEDLINE | ID: mdl-24412381

ABSTRACT

Benzo[a]pyrene (BaP) is a polycyclic aromatic hydrocarbon and carcinogen that is released into the environment through natural and anthropogenic sources. BaP toxicity is dependent on its metabolism by cytochrome P450s to the reactive metabolite benzo[a]pyrene diol epoxide (BPDE), which is strongly associated with increased mutation frequency. BaP can also be metabolized to benzo[a]pyrene quinones that can undergo redox cycling and induce oxidative stress. The purpose of this study was to examine if BaP exposure induces DNA double strand breaks (DSBs) and subsequently activate DNA DSB repair pathways in the CHO 3-6 cell line and pKZ1 mouse model. In vitro assessment of homologous recombination (HR) showed significantly increased HR frequency following exposure to 10µM of BaP. In vivo evaluations of BaP-induced DNA DSB repair demonstrated positive staining for intrachromosomal recombination events, which are associated with non-homologous end joining (NHEJ), in the lung and thymus of exposed animals that were statistically significant in the thymus when quantified by Western blotting. Gene expression analyses from mouse tissues showed significantly decreased expression of ATM and Xrcc6 in BaP-treated liver and lung. In addition, BaP exposure significantly reduced the expression of Xrcc5, p53, and DNA-PKcs in lung. Taken together, our results demonstrate that BaP increases DNA DSB repair in vitro and in vivo, and induces expression changes in DNA repair pathway genes. As repair of DNA DSBs is not error-free, aberrant DNA repair may be contributing to the mechanism of BaP-induced toxicity.


Subject(s)
Benzo(a)pyrene/toxicity , DNA Breaks, Double-Stranded/drug effects , DNA Repair , Signal Transduction/drug effects , Animals , Antigens, Nuclear/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Blotting, Western , CHO Cells , Cricetinae , Cricetulus , DNA-Binding Proteins/genetics , Gene Expression/drug effects , Homologous Recombination/drug effects , Kidney/drug effects , Kidney/metabolism , Ku Autoantigen , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Male , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Spleen/drug effects , Spleen/metabolism , Thymus Gland/drug effects , Thymus Gland/metabolism , Tumor Suppressor Protein p53/genetics , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
10.
Reprod Toxicol ; 124: 108551, 2024 03.
Article in English | MEDLINE | ID: mdl-38280688

ABSTRACT

Gestational exposure to the anticonvulsant drug valproic acid (VPA) is associated with congenital malformations and neurodevelopmental disorders through its action as a histone deacetylase inhibitor. VPA can elicit placental toxicity and affect placental growth and development. The objective of this study was to evaluate the impact of maternal exposure to VPA on the mouse placenta following exposure on gestational day (GD) 13 since previous studies have shown that mice exposed at this time during gestation give birth to offspring with an autism spectrum disorder-like phenotype. We exposed CD-1 dams to a teratogenic dose (600 mg/kg) of VPA or saline on GD13 and assessed fetoplacental growth and development on GD18. We evaluated epigenetic modifications, including acetylated histone H4 (H4ac), methylated H3K4 (H3K4me2) using immunohistochemistry, and global DNA methylation in the placenta at 1, 3, and 24 h following maternal exposure on GD13. In utero exposure to VPA on GD13 significantly decreased placental weight and increased fetal resorptions. Moreover, VPA significantly increased the staining intensity of histone H4 acetylation and H3K4 di-methylation across the placenta at 1 and 3 h post maternal dose. Our results also demonstrate that VPA significantly decreased global DNA methylation levels in placental tissue. These results show that gestational exposure to VPA interferes with placental growth and elicits epigenetic modifications, which may play a vital role in VPA-induced developmental toxicity.


Subject(s)
Autism Spectrum Disorder , Valproic Acid , Pregnancy , Female , Mice , Animals , Valproic Acid/toxicity , Histones/metabolism , Placenta/metabolism , Epigenesis, Genetic
11.
Birth Defects Res ; 115(3): 338-347, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36369707

ABSTRACT

INTRODUCTION: Triphenyl phosphate (TPHP) is a chemical flame retardant and plasticizer which is added to consumer and industrial products. The developmental origins of health and disease hypothesis postulate that in utero exposures can have later-in-life effects on the developing fetus and can alter fetal gene expression. This study aimed to determine whether epigenetic modifications occurred following in utero TPHP exposure in mice and whether these changes were dose and/or sex-dependent. METHODS: Pregnant C57Bl/6 mice were treated with 0, 5, 25, or 50 mg/kg of TPHP on gestational days (GD) 8, 10, 12, and 14 via intraperitoneal injection and fetal livers were collected on GD 19. Changes in the levels of acetylation of H3 and H4, as well as methylation of H3K9 and global DNA methylation were assessed in the fetal livers by western blot. RESULTS: Results showed that there was a significant decrease in fetal DNA methylation following in utero exposure to 50 mg/kg TPHP compared to the control (0 mg/kg) independent of the sex of the fetus. While there were no significant alterations compared to controls in any histone modifications at any dose or sex following in utero TPHP exposure, we did note a decrease (t test, p = .025) in the levels of acetylated H3 in males versus females following a maternal dose of 25 mg/kg. The monomethylated H3K9 levels were also increased in females versus males following exposure to TPHP at 5 mg/kg (p = .018) and 25 mg/kg (p = .027) when analyzed via unpaired t tests, although not significantly different from controls. DISCUSSION: The results suggest that gestational TPHP exposure can induce epigenetic modifications in murine fetal tissue. Specifically, global DNA methylation levels were downregulated in response to TPHP. Additionally, males appear to be more sensitive to TPHP-induced histone modifications than females. These data support the need for further studies investigating the impacts of gestational TPHP exposure on the developing fetus.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Pregnancy , Male , Female , Animals , Mice , Organophosphates/toxicity , Mice, Inbred C57BL , Liver
12.
Toxicol Lett ; 368: 47-55, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-35963423

ABSTRACT

Benzene is an environmental toxicant and known human carcinogen. Recent epidemiological studies show a relationship between exposure to benzene in pregnant women and increased incidence of childhood leukemias. Studies in murine models demonstrate a relationship between carcinogenicity and in utero benzene exposure which was sex dependent, thus the cellular mechanisms of benzene toxicity by sex require further studies. A hypothesized mechanism of benzene-induced in utero carcinogenicity is through increased DNA damage and reduced fetal DNA repair capacity. This includes the potential inhibition of topoisomerase IIα (topo IIα), in part, to generate double stranded DNA (dsDNA) breaks and induction of error-prone DNA repair. Using a mouse model of transplacental benzene carcinogenicity, gestational day (GD) 14 fetal livers were harvested 2, 6, and 24 h following maternal exposure to 200 mg/kg benzene and used to assess DNA damage, DNA repair gene expression and topo IIα activity. DNA damage, measured by levels of modified histone H2AX (γH2AX), is significantly increased in benzene exposed pups, with sex-dependent significance seen only in female pups. Comet assay results confirmed that benzene exposure in utero induces dsDNA damage in the GD14 fetal liver. Genes involved in DNA repair were assessed, and DNA repair gene expression changes were observed after 24 h in genes related to nucleotide excision repair, homologous recombination, and non-homologous end-joining. There were no significant differences in topo IIα activity in GD14 fetal livers at any timepoint, or between sexes. Overall, this study shows that 200 mg/kg benzene exposure induces dsDNA damage and alters fetal DNA repair gene expression in utero, without perturbing fetal topo IIα in CD-1 mice.


Subject(s)
Antigens, Neoplasm , Benzene , DNA Damage , DNA Repair , DNA Topoisomerases, Type II , Animals , Antigens, Neoplasm/genetics , Benzene/toxicity , DNA , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/genetics , Female , Gene Expression , Maternal Exposure , Mice , Pregnancy
13.
J Dev Orig Health Dis ; 13(6): 706-718, 2022 12.
Article in English | MEDLINE | ID: mdl-35593438

ABSTRACT

Children of women with pre-eclampsia have increased risk of cardiovascular (CV) and metabolic disease in adult life. Furthermore, the risk of pregnancy complications is higher in daughters born to women affected by pre-eclampsia than in daughters born after uncomplicated pregnancies. While aberrant inflammation contributes to the pathophysiology of pregnancy complications, including pre-eclampsia, the contribution of maternal inflammation to subsequent risk of CV and metabolic disease as well as pregnancy complications in the offspring remains unclear. Here, we demonstrate that 24-week-old female rats (F1) born to dams (F0) exposed to lipopolysaccharide (LPS) during pregnancy (to induce inflammation) exhibited mild systolic dysfunction, increased cardiac growth-related gene expression, altered glucose tolerance, and coagulopathy; whereas male F1 offspring exhibited altered glucose tolerance and increased visceral fat accumulation compared with F1 sex-matched offspring born to saline-treated dams. Both male and female F1 offspring born to LPS-treated dams had evidence of anemia. Fetuses (F2) from F1 females born to LPS-treated dams were growth restricted, and this reduction in fetal growth was associated with increased CD68 positivity (indicative of macrophage presence) and decreased expression of glucose transporter-1 in their utero-placental units. These results indicate that abnormal maternal inflammation can contribute to increased risk of CV and metabolic disease in the offspring, and that the effects of inflammation may cross generations. Our findings provide evidence in support of early screening for CV and metabolic disease, as well as pregnancy complications in offspring affected by pre-eclampsia or other pregnancy complications associated with aberrant inflammation.


Subject(s)
Cardiovascular Diseases , Pre-Eclampsia , Prenatal Exposure Delayed Effects , Humans , Rats , Female , Pregnancy , Male , Animals , Fetal Growth Retardation , Pre-Eclampsia/etiology , Placenta/metabolism , Lipopolysaccharides/metabolism , Inflammation/metabolism , Cardiovascular Diseases/metabolism , Glucose/metabolism
14.
Mol Pharmacol ; 80(6): 979-87, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21868484

ABSTRACT

Exposure to the anticonvulsant valproic acid (VPA) during the first trimester of pregnancy is associated with an increased risk of congenital malformations including heart defects, craniofacial abnormalities, skeletal and limb defects, and, most frequently, neural tube defects (NTDs). The mechanisms by which VPA induces teratogenic effects are not fully understood, although previous studies support a role for oxidative stress. To investigate the effects of VPA on early development, a whole-embryo culture model was used to evaluate the protective effects of antioxidants, measure intracellular reactive oxygen species (ROS) levels, and assess markers of oxidative damage and apoptosis. Furthermore, in vivo teratological evaluations of antioxidant protection were also completed. VPA (0.60 mM in embryo culture, 400 mg/kg in vivo) induced significant decreases in embryonic growth and increases in NTDs. Of the antioxidants tested, catalase provided partial protection against VPA-mediated reductions in morphological and developmental growth parameters in both whole-embryo culture and in vivo systems. VPA exposure resulted in an increase in ROS staining in the head region, as assessed by whole-mount staining with 5-(and-6)-chloromethyl-2',7'-dichlorodihydrofluorescein diacetate. Markers of embryonic oxidative damage including 8-hydroxyguanosine, 4-hydroxynonenal adducts, and 3-nitrotyrosine were not affected by VPA treatment. Increased ROS levels were correlated with increased staining for apoptotic markers, as assessed by Western blotting and immunohistochemistry. Addition of catalase to the medium attenuated VPA-induced increases in ROS formation and apoptosis. These studies identify regions of the embryo susceptible to ROS and apoptosis induced by VPA, thus establishing a possible molecular pathway by which VPA exerts teratogenicity.


Subject(s)
Apoptosis/physiology , Embryonic Development/physiology , Neural Tube Defects/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Valproic Acid/toxicity , Animals , Apoptosis/drug effects , Dose-Response Relationship, Drug , Embryonic Development/drug effects , Female , Mice , Neural Tube Defects/chemically induced , Organ Culture Techniques , Oxidative Stress/drug effects , Pregnancy
15.
Toxicol Appl Pharmacol ; 257(3): 429-36, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22005274

ABSTRACT

In the last two decades, nanoparticles (NPs) have found applications in a wide variety of consumer goods. Titanium dioxide (TiO(2)) and silver (Ag) NPs are both found in cosmetics and foods, but their increasing use is of concern due to their ability to be taken up by biological systems. While there are some reports of TiO(2) and Ag NPs affecting complex organisms, their effects on reproduction and development have been largely understudied. Here, the effects of orally administered TiO(2) or Ag NPs on reproduction and development in two different model organisms were investigated. TiO(2) NPs reduced the developmental success of CD-1 mice after a single oral dose of 100 or 1000 mg/kg to dams, resulting in a statistically significant increase in fetal deformities and mortality. Similarly, TiO(2) NP addition to food led to a significant progeny loss in the fruit fly, Drosophila, as shown by a decline in female fecundity. Ag NP administration resulted in an increase in the mortality of fetal mice. Similarly in Drosophila, Ag NP feeding led to a significant decrease in developmental success, but unlike TiO(2) NP treatment, there was no decline in fecundity. The distinct response associated with each type of NP likely reflects differences in NP administration as well as the biology of the particular model. Taken together, however, this study warns that these common NPs could be detrimental to the reproductive and developmental health of both invertebrates and vertebrates.


Subject(s)
Metal Nanoparticles , Reproduction/drug effects , Silver/toxicity , Titanium/toxicity , Administration, Oral , Animals , Dose-Response Relationship, Drug , Drosophila melanogaster , Female , Male , Mice , Pregnancy , Pregnancy Outcome , Silver/administration & dosage , Species Specificity , Titanium/administration & dosage
16.
Placenta ; 112: 141-152, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34352490

ABSTRACT

INTRODUCTION: Valproic acid (VPA) is an effective anti-epileptic drug clinically used to treat seizures, bipolar disorders and neuropathic pain in women of reproductive age. Current approval of VPA for psychiatric conditions and migraine has increased the number of VPA exposed pregnancies. VPA crosses the placental barrier and induces birth defects in about 10% of exposed pregnancies. In addition, VPA exposure results in neurodevelopmental disorders in children without any overt birth defects. The current study was designed to investigate the effects of in utero VPA exposure on fetoplacental growth in a mouse model. METHODS: Pregnant CD-1 dams were exposed to a single teratogenic dose of 400 mg/kg VPA or saline via subcutaneous injection on gestational day (GD) 9 and fetuses were harvested on GD 13, 15, 17 and 19, respectively. Resorptions, gross malformations, fetal weight, fetal head weight, fetal crown-rump length, fetal head transverse and anteroposterior diameters, placental weight and placental diameter were noted. RESULTS: VPA exposure led to multiple external deformities including exencephaly, open eye defect, subcutaneous hemorrhage and underdevelopment of tail. All fetoplacental growth parameters fetal weight, fetal head weight, fetal crown-rump length, placental weight and placental diameter were significantly reduced in VPA-exposed fetuses with and without congenital malformations such as exencephaly, compared to control fetuses. DISCUSSION: In conclusion, the effects of in utero VPA exposure on fetal and placental growth persisted throughout pregnancy and our results suggest that the effects of VPA on placental growth may play a role in VPA-induced toxicity.


Subject(s)
Abnormalities, Drug-Induced/pathology , Anticonvulsants/adverse effects , Fetal Development/drug effects , Fetus/pathology , Valproic Acid/adverse effects , Abnormalities, Drug-Induced/etiology , Animals , Female , Fetus/drug effects , Male , Maternal Exposure , Mice , Pregnancy
17.
Placenta ; 107: 31-40, 2021 04.
Article in English | MEDLINE | ID: mdl-33735658

ABSTRACT

INTRODUCTION: Valproic acid (VPA), a widely prescribed antiepileptic drug and an effective treatment for bipolar disorder and neuropathic pain, results in multiple developmental defects following in utero exposure. Uterine decidua provides nutritional and physical support during implantation and early embryonic development. Perturbations in the molecular mechanisms within decidual tissue during early pregnancy might affect early embryonic growth, result in early pregnancy loss or cause complications in the later gestational stage. VPA is a known histone deacetylase inhibitor and epigenetic changes such as histone hyperacetylation and methylation have been proposed as a mechanism of VPA-induced teratogenesis. METHODS: This study investigated the effects of in utero VPA exposure on histone modifications in murine decidual tissue. Pregnant CD-1 mice were exposed to 400 mg/kg VPA or saline on GD9 via subcutaneous injection. Decidual tissue from each gestational sac was harvested at 1, 3 and 6 h following exposure. Levels of acetylated histones H3, H4 and H3K56, as well as methylated histones H3K9 and H3K27 were acid extracted and assessed by western blotting followed by acid histone extraction. RESULTS: VPA exposure induced a significant increase (p < 0.05) in the levels of acetylated H3 at 1, 3 h; acetylated H4 at 1, 3 and 6 h and trimethylated H3K9 at 6 h. In contrast, no significant perturbations were noted in the levels of monomethylated H3K9, trimethylated H3K27 and acetylated H3K56. DISCUSSION: The results from this study suggest that VPA-induced decidual histone modifications might play an important role as a mechanism of VPA-induced teratogenesis during early embryonic growth.


Subject(s)
Decidua/drug effects , Epigenesis, Genetic/drug effects , Valproic Acid/pharmacology , Animals , Decidua/metabolism , Female , Histones/metabolism , Mice , Pregnancy
18.
Neurotoxicol Teratol ; 83: 106941, 2021.
Article in English | MEDLINE | ID: mdl-33212164

ABSTRACT

Nuclear factor kappa B (NF-κB) is a heterodimer of protein subunits p65 and p50, that regulates the expression of a large number of genes related to cell growth and proliferation. The p65 subunit is activated after phosphorylation by Pim-1, while the p50 subunit is the cleaved product of its precursor molecule p105. Valproic acid (VPA), an antiepileptic drug, is a known teratogen and its exposure during pregnancy is associated with 1-2% of neural tube defects in the offspring. The current study aimed at investigating the effects of in utero VPA exposure on the key components of the NF-κB signaling pathway including p65, p50, and Pim-1 in CD-1 mouse embryos during the critical period of neural tube closure. Here we report that p65, Pim-1 and p105/p50 mRNA were significantly (p < 0.05) downregulated at 1 and 3 h following in utero exposure to a teratogenic dose (400 mg/kg) of VPA in gestational day (GD)9 exposed embryos. At GD13 heads of control, non-exencephalic and exencephalic embryos were used for analysis and we found significant upregulation of p65 protein expression in non-exencephalic GD13 heads while p50 protein levels were significantly downregulated in both non-exencephalic and exencephalic groups. On the other hand, p65 and p50 protein levels remained unchanged in the nuclear extracts of the VPA-exposed non-exencephalic and exencephalic GD13 embryo heads. The reported results suggest that VPA exposure perturbates p65, p105/p50, Pim-1 transcript and p65/p50 protein levels in mouse embryos.


Subject(s)
NF-kappa B/metabolism , Neural Tube/drug effects , Neural Tube/embryology , Valproic Acid/toxicity , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/toxicity , Disease Models, Animal , Female , Gene Expression Regulation, Developmental/drug effects , Male , Maternal-Fetal Exchange , Mice , NF-kappa B/genetics , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , Neural Tube/metabolism , Neural Tube Defects/chemically induced , Neural Tube Defects/embryology , Neural Tube Defects/metabolism , Neurotoxins/administration & dosage , Neurotoxins/toxicity , Neurulation/drug effects , Neurulation/physiology , Pregnancy , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Valproic Acid/administration & dosage
19.
Carcinogenesis ; 31(6): 1142-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20400480

ABSTRACT

Childhood cancer is the leading cause of disease-related death in children aged 1-14 years in Canada and the USA and it has been hypothesized that transplacental exposure to environmental carcinogens such as benzene may contribute to the etiology of these cancers. Our objectives were to determine if transplacental benzene exposure increased tumor incidence in mouse offspring and assess fetal benzene metabolism capability. Pregnant CD-1 and C57Bl/6N mice were given intraperitoneal injections of corn oil, 200 mg/kg, or 400 mg/kg benzene on gestational days 8, 10, 12 and 14. A significant increase in tumor incidence was observed in CD-1, but not C57BL/6N, 1-year-old offspring exposed transplacentally to 200 mg/kg benzene. Hepatic and hematopoietic tumors were predominantly observed in male and female CD-1 offspring, respectively. Female CD-1 offspring exposed transplacentally to 200 mg/kg benzene had significantly suppressed bone marrow CD11b(+) cells 1 year after birth, correlating with reduced colony-forming unit granulocyte/macrophage numbers in 2-day-old pups. CD-1 and C57Bl/6N maternal blood benzene levels and fetal liver benzene, t, t-muconic acid, hydroquinone and catechol levels were analyzed by gas chromatography/mass spectrometry. Significant strain-, gender- and dose-related differences were observed. Male CD-1 fetuses had high hydroquinone levels, whereas females had high catechol levels after maternal exposure to 200 mg/kg benzene. This is the first demonstration that transplacental benzene exposure can induce hepatic and hematopoietic tumors in mice, which may be dependent on fetal benzene metabolism capability.


Subject(s)
Benzene/toxicity , Maternal-Fetal Exchange , Neoplasms, Experimental/chemically induced , Animals , Base Sequence , Benzene/pharmacokinetics , DNA Primers , Female , Flow Cytometry , Gas Chromatography-Mass Spectrometry , Male , Mice , Mice, Inbred C57BL , Pregnancy
20.
Toxicol Appl Pharmacol ; 244(3): 273-9, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20083130

ABSTRACT

Benzene is a ubiquitous occupational and environmental toxicant. Exposures to benzene both prenatally and during adulthood are associated with the development of disorders such as aplastic anemia and leukemia. Mechanisms of benzene toxicity are unknown; however, generation of reactive oxygen species (ROS) by benzene metabolites may play a role. Little is known regarding the effects of benzene metabolites on erythropoiesis. Therefore, to determine the effects of in utero exposure to benzene on the growth and differentiation of fetal erythroid progenitor cells (CFU-E), pregnant CD-1 mice were exposed to benzene and CFU-E numbers were assessed in fetal liver (hematopoietic) tissue. In addition, to determine the effect of benzene metabolite-induced ROS generation on erythropoiesis, HD3 chicken erythroblast cells were exposed to benzene, phenol, or hydroquinone followed by stimulation of erythrocyte differentiation. Our results show that in utero exposure to benzene caused significant alterations in female offspring CFU-E numbers. In addition, exposure to hydroquinone, but not benzene or phenol, significantly reduced the percentage of differentiated HD3 cells, which was associated with an increase in ROS. Pretreatment of HD3 cells with polyethylene glycol-conjugated superoxide dismutase (PEG-SOD) prevented hydroquinone-induced inhibition of erythropoiesis, supporting the hypothesis that ROS generation is involved in the development of benzene erythrotoxicity. In conclusion, this study provided evidence that ROS generated as a result of benzene metabolism may significantly alter erythroid differentiation, potentially leading to the development of Blood Disorders.


Subject(s)
Benzene/toxicity , Cell Differentiation/drug effects , Environmental Pollutants/toxicity , Erythroid Cells/drug effects , Maternal Exposure , Reactive Oxygen Species/metabolism , Animals , Benzene/metabolism , Environmental Pollutants/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/drug effects , Erythropoiesis/drug effects , Female , Growth Inhibitors/toxicity , Hydroquinones/toxicity , Liver/cytology , Liver/drug effects , Liver/metabolism , Metabolic Networks and Pathways , Mice , Models, Animal , Phenol/toxicity , Pregnancy , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL