Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Toxicol Appl Pharmacol ; 459: 116344, 2023 01 15.
Article in English | MEDLINE | ID: mdl-36526072

ABSTRACT

P-glycoprotein (P-gp, encoded by the ABCB1 gene) and breast cancer resistance protein (BCRP/ABCG2) are efflux multidrug resistance (MDR) transporters localized at the syncytiotrophoblast barrier of the placenta and protect the conceptus from drug and toxin exposure throughout pregnancy. Infection is an important modulator of MDR expression and function. This review comprehensively examines the effect of infection on the MDR transporters, P-gp and BCRP in the placenta. Infection PAMPs such as bacterial lipopolysaccharide (LPS) and viral polyinosinic-polycytidylic acid (poly I:C) and single-stranded (ss)RNA, as well as infection with Zika virus (ZIKV), Plasmodium berghei ANKA (modeling malaria in pregnancy - MiP) and polymicrobial infection of intrauterine tissues (chorioamnionitis) all modulate placental P-gp and BCRP at the levels of mRNA, protein and or function; with specific responses varying according to gestational age, trophoblast type and species (human vs. mice). Furthermore, we describe the expression and localization profile of Toll-like receptor (TLR) proteins of the innate immune system at the maternal-fetal interface, aiming to better understand how infective agents modulate placental MDR. We also highlight important gaps in the field and propose future research directions. We conclude that alterations in placental MDR expression and function induced by infective agents may not only alter the intrauterine biodistribution of important MDR substrates such as drugs, toxins, hormones, cytokines, chemokines and waste metabolites, but also impact normal placentation and adversely affect pregnancy outcome and maternal/neonatal health.


Subject(s)
Zika Virus Infection , Zika Virus , Pregnancy , Female , Humans , Mice , Animals , Placenta/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Tissue Distribution , Neoplasm Proteins/genetics , Drug Resistance, Multiple , Membrane Transport Proteins/metabolism
2.
Sci Rep ; 12(1): 10262, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35715474

ABSTRACT

Limited information is available about the effect of mid-pregnancy viral infections on the placental expression of efflux transporters and offspring behavior. We hypothesized that maternal exposure to polyinosinic-polycytidylic acid [poly(I:C)], a synthetic double-stranded RNA viral mimic, would impair placental cell turnover, the expression of selected ABC transporters and adult offspring behavior. C57BL/6 mice were administered poly(I:C) (10 mg/Kg;ip) or vehicle at gestational day (GD) 13.5 (mid-pregnancy). Dams were euthanized for blood collection 4 h after injection, fetal and placental collection at GD18.5 or allowed to deliver spontaneously at term. At GD 13.5, poly(I:C) induced an acute pro-inflammatory response characterized by an increase in maternal plasma levels of IL-6, CXCL-1 and CCL-2/MCP-1. At GD 18.5, poly(I:C) decreased cell proliferation/death in the labyrinthine and increased cell death in the junctional zones, characterizing a disruption of placental cell turnover. Abca1 and Abcg1 immunolabelling was decreased in the labyrinthine zone, whereas Abca1, Abcg1 and breast cancer resistance transporter (Bcrp) expression increased in the junctional zone. Moreover, adult offspring showed motor and cognitive impairments in the Rotarod and T-water maze tests. These results indicate that viral infection during mid-pregnancy may disrupt relevant placental efflux transporters, as well as placental cell turnover and offspring behavior in adult life.


Subject(s)
ATP-Binding Cassette Transporters , Cognitive Dysfunction , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Cognitive Dysfunction/metabolism , Female , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Proteins/metabolism , Placenta/metabolism , Poly I-C/pharmacology , Pregnancy
3.
Reprod Toxicol ; 98: 82-91, 2020 12.
Article in English | MEDLINE | ID: mdl-32916274

ABSTRACT

Bacterial infection alters placental ABC transporters expression. These transporters provide fetal protection against circulating xenobiotics and environmental toxins present in maternal blood. We hypothesized that lipopolysaccharide (LPS-bacterial mimic) alters the yolk sac morphology and expression of key ABC transporters in a gestational-age dependent manner. Yolk sac samples from C57BL/6 mice were obtained at gestational ages (GD) 15.5 and GD18.5, 4 or 24 h after LPS exposure (150ug/kg; n = 8/group). Samples underwent morphometrical, qPCR and immunohistochemistry analysis. The volumetric proportions of the histological components of the yolk sac did not change in response to LPS. LPS increased Abcg2 expression at GD15.5, after 4 h of treatment (p < 0.05). No changes in Abca1, Abcb1a/b, Abcg1, Glut1, Snat1, Il-1ß, Ccl2 and Mif were observed. Il-6 and Cxcl1 were undetectable in the yolk sac throughout pregnancy. Abca1, breast cancer resistance protein (Bcrp, encoded by Abcg2) and P-glycoprotein (P-gp/ Abcb1a/b) were localized in the endodermal (uterine-facing) epithelium and to a lesser extent in the mesothelium (amnion-facing), whereas Abca1 was also localized to the endothelium of the yolk sac blood vessels. LPS increased the labeling area and intensity of Bcrp in the yolk sac's mesothelial cells at GD15.5 (4 h), whereas at GD18.5, the area of Bcrp labeling in the mesothelium (4 and 24 h) was decreased (p < 0.05). Bacterial infection has the potential to change yolk sac barrier function by affecting Bcrp and Abcg2 expression in a gestational-age dependent-manner. These changes may alter fetal exposure to xenobiotics and toxic substances present in the maternal circulation and in the uterine cavity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Lipopolysaccharides/pharmacology , Yolk Sac/drug effects , Animals , Female , Gestational Age , Mice, Inbred C57BL , Pregnancy , Yolk Sac/metabolism
4.
Neurosci Biobehav Rev ; 117: 198-210, 2020 10.
Article in English | MEDLINE | ID: mdl-28528960

ABSTRACT

In utero exposure to environmental stress in both animals and humans could result in long-term epigenome alterations which further lead to consequences for adaptation and development in the offspring. Epigenetics, especially DNA methylation, is considered one of the most widely studied and well-characterized mechanisms involved in the long-lasting effects of in utero stress exposure. In this review, we outlined evidence from animal and human prenatal research supporting the view that prenatal stress could lead to lasting, broad and functionally organized signatures in DNA methylation which, in turn, could mediate exposure-phenotype associations. We also emphasized the advantage of using stressor from quasi-randomly assigned experiments. Furthermore, we discuss challenges that still need to be addressed in this field in the future.


Subject(s)
Prenatal Exposure Delayed Effects , Animals , DNA Methylation , Epigenesis, Genetic , Epigenomics , Female , Humans , Phenotype , Pregnancy , Prenatal Exposure Delayed Effects/genetics
5.
Sci Rep ; 9(1): 11488, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31391498

ABSTRACT

Malaria in Pregnancy (MiP) is characterized by placental accumulation of Plasmodium-infected erythrocytes, intrauterine growth restriction (IUGR) and preterm delivery (PTD). Placental ATP-binding cassette (ABC) transporters mediate the efflux of nutrients, cytokines and xenobiotics. The expression and activity of these transporters are highly responsive to infection. We hypothesized that MiP would perturb the expression of placental ABC transporters, promoting PTD. Peripheral blood, spleens, livers and placentas of pregnant mice, infected with Plasmodium berghei ANKA on gestational day (GD) 13.5, were collected and analyzed on GD18.5. The primary consequences of human MiP, including IUGR, PTD (20%) and placental inflammation, were recapitulated in our mouse model. Electron microscopy revealed attenuated presence of labyrinthine microvilli and dilated spongiotrophoblasts -granular endoplasmic reticulum cisternae. Additionally, a decrease in placental Abca1 (ABCA1), Abcb1b (P-glycoprotein), Abcb9 and Abcg2 (BCRP) expression was observed in MiP mice. In conclusion, MiP associated with PTD impairs placental ABC transporters' expression, potentially modulating placental nutrient, environmental toxin and xenobiotic biodistribution within the fetal compartment, and may, at some degree, be involved with pregnancy outcome in MiP.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Malaria/complications , Obstetric Labor, Premature/immunology , Placenta/pathology , Plasmodium berghei/immunology , Animals , Disease Models, Animal , Female , Humans , Malaria/immunology , Malaria/parasitology , Maternal-Fetal Exchange/immunology , Mice , Nutrients/metabolism , Obstetric Labor, Premature/parasitology , Obstetric Labor, Premature/pathology , Placenta/metabolism , Pregnancy , Xenobiotics/metabolism
6.
J Neuroendocrinol ; 28(3): 12360, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26718627

ABSTRACT

P-glycoprotein (P-gp) encoded by Abcb1 provides protection to the developing brain from xenobiotics. P-gp in brain endothelial cells (BECs) derived from the developing brain microvasculature is up-regulated by glucocorticoids and inhibited by pro-inflammatory cytokines in vitro. However, little is known about how prenatal maternal glucocorticoid treatment can affect Abcb1/P-gp function and subsequent cytokine regulation in foetal BECs. We hypothesised that glucocorticoid exposure increases Abcb1/P-gp in the foetal brain microvasculature and enhances the sensitivity of Abcb1/P-gp in BECs to the inhibitory effects of cytokines. BECs isolated from dexamethasone- or vehicle-exposed foetal guinea pigs were cultured and treated with interleukin-1ß, interleukin-6 or tumour necrosis factor-α, and Abcb1/P-gp expression and function were assessed. Prenatal dexamethasone exposure significantly increased Abcb1/P-gp expression/activity and cytokine receptor levels in BECs of the foetal brain microvasculature. Foetal dexamethasone exposure in vivo also increased the subsequent responsiveness of BECs to pro-inflammatory cytokines in vitro. In conclusion, maternal treatment with synthetic glucocorticoids appears to prematurely mature P-gp mediated drug resistance at the foetal BBB in vivo and profoundly impact the subsequent responsiveness of P-gp to pro-inflammatory cytokines in the foetal BEC. The significance of these findings to foetal brain protection against xenobiotics and other P-gp substrates in vivo requires further elaboration. However, the results of the present study may have implications for human pregnancy and foetal brain protection, particularly in cases of preterm birth combined with infection.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Blood-Brain Barrier/embryology , Blood-Brain Barrier/metabolism , Cytokines/pharmacology , Glucocorticoids/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Blood-Brain Barrier/drug effects , Cells, Cultured , Dexamethasone/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Guinea Pigs , Inflammation Mediators/pharmacology , Male , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/metabolism
7.
Hum Reprod Update ; 22(2): 164-81, 2016.
Article in English | MEDLINE | ID: mdl-26545808

ABSTRACT

BACKGROUND: The transmembrane ATP-binding cassette (ABC) transporters actively efflux an array of clinically relevant compounds across biological barriers, and modulate biodistribution of many physiological and pharmacological factors. To date, over 48 ABC transporters have been identified and shown to be directly and indirectly involved in peri-implantation events and fetal/placental development. They efflux cholesterol, steroid hormones, vitamins, cytokines, chemokines, prostaglandins, diverse xenobiotics and environmental toxins, playing a critical role in regulating drug disposition, immunological responses and lipid trafficking, as well as preventing fetal accumulation of drugs and environmental toxins. METHODS: This review examines ABC transporters as important mediators of placental barrier functions and key reproductive processes. Expression, localization and function of all identified ABC transporters were systematically reviewed using PubMed and Google Scholar websites to identify relevant studies examining ABC transporters in reproductive tissues in physiological and pathophysiological states. Only reports written in English were incorporated with no restriction on year of publication. While a major focus has been placed on the human, extensive evidence from animal studies is utilized to describe current understanding of the regulation and function of ABC transporters relevant to human reproduction. RESULTS: ABC transporters are modulators of steroidogenesis, fertilization, implantation, nutrient transport and immunological responses, and function as 'gatekeepers' at various barrier sites (i.e. blood-testes barrier and placenta) against potentially harmful xenobiotic factors, including drugs and environmental toxins. These roles appear to be species dependent and change as a function of gestation and development. The best-described ABC transporters in reproductive tissues (primarily in the placenta) are the multidrug transporters p-glycoprotein and breast cancer-related protein, the multidrug resistance proteins 1 through 5 and the cholesterol transporters ABCA1 and ABCG1. CONCLUSIONS: The ABC transporters have various roles across multiple reproductive tissues. Knowledge of efflux direction, tissue distribution, substrate specificity and regulation of the ABC transporters in the placenta and other reproductive tissues is rapidly expanding. This will allow better understanding of the disposition of specific substrates within reproductive tissues, and facilitate development of novel treatments for reproductive disorders as well as improved approaches to protecting the developing fetus.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Reproduction/physiology , ATP-Binding Cassette Transporters/genetics , Animals , Blastocyst/metabolism , Embryonic Development/genetics , Female , Humans , Placenta/metabolism , Pregnancy , Reproduction/genetics , Tissue Distribution
8.
Placenta ; 35(5): 324-30, 2014 May.
Article in English | MEDLINE | ID: mdl-24685282

ABSTRACT

INTRODUCTION: The placenta contains efflux transporters, including P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP), that limit the passage of xenobiotics, certain hormones and nutrients from the maternal to the fetal circulation. The expression of these transporters changes with gestational age, yet the mechanisms involved remain unknown. However, the changes in P-gp and BCRP transporter expression coincide with those of oxygen tension in the placenta, and oxygen tension has been shown to modulate P-gp and BCRP expression in other tissues. The objective of this study was to investigate the effects of oxygen tension on P-gp and BCRP expression in the term human placenta. METHODS: Following equilibration in culture (96 h), term placental explants (n = 7) were cultured in 3% or 20% oxygen for 24 and 48 h. Culture medium was collected every 24 h to measure lactate dehydrogenase (LDH; explant viability) and human chorionic gonadotropin (hCG; syncytiotrophoblast function). P-gp (encoded by ABCB1) and BCRP (encoded by ABCG2) protein and mRNA, as well as VEGFA mRNA were measured using western blot and qRT-PCR. P-gp localization was determined using immunofluorescence. RESULTS: Oxygen tension had a significant effect on P-gp expression, with ABCB1/P-gp mRNA and protein levels increased in the hypoxic condition (3% O2) after 48 h (p < 0.05). VEGFA mRNA was elevated by hypoxia at both 24 and 48 h (p < 0.05). In contrast, placental ABCG2/BCRP mRNA and protein expression were stable with changes in oxygen tension. We identified profound differences in the glycosylation of P-gp between cultured and non-cultured placental tissue, with cultured explants expressing deglycosylated P-gp. CONCLUSIONS: These findings demonstrate that, at term, the expression of placental P-gp, is regulated by oxygen tension. This suggests that changes in oxygenation of the placenta in the third trimester may alter levels of placental P-gp, and in doing so alter fetal exposure to P-gp substrates, including xenobiotics and certain hormones.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Drug Resistance, Multiple/drug effects , Neoplasm Proteins/metabolism , Oxygen/administration & dosage , Placenta/drug effects , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Chorionic Gonadotropin/metabolism , Female , Gestational Age , Humans , L-Lactate Dehydrogenase/metabolism , Placenta/metabolism , Pregnancy , Pregnancy Trimester, Third , Trophoblasts/metabolism
9.
Placenta ; 34(9): 817-23, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23790363

ABSTRACT

INTRODUCTION: The multidrug resistance proteins, P-glycoprotein (P-gp, encoded by the ABCB1 gene) and breast cancer resistance protein (BCRP, encoded by ABCG2) are highly expressed in the first trimester placenta. These transporters protect the fetus from exposure to maternally derived toxins and xenobiotics. Since oxygen is a regulator of multidrug resistance in various tissues, we hypothesized that changes in oxygen tension alter placental ABCB1/P-gp and ABCG2/BCRP expression in the first trimester. METHODS: Placental specimens were collected from first (n = 7), second (n = 5) and term pregnancies (n = 5). First trimester placental villous explants were incubated (24 or 48 h) in different oxygen tension (3-20%). ABCB1, ABCG2 and VEGFA mRNA expression levels were assessed by RT-PCR and protein was localized by IHC. RESULTS: ABCB1 is expressed most highly in the first trimester placenta (p < 0.05), whereas ABCG2 expression does not change significantly over pregnancy. P-gp and BCRP staining is present in the syncytiotrophoblast and in cytotrophoblasts. ABCG2 mRNA is increased in hyperoxic (20%) conditions after 48 h (p < 0.05). In contrast, hypoxia (3%) did not change ABCB1 mRNA expression but significantly increased VEGFA mRNA (p < 0.05). Hypoxia resulted in increased BCRP staining in cytotrophoblasts and in the microvillous membrane of the syncytium. Whereas, hypoxia resulted in increased P-gp staining in proliferating cytotrophoblasts. CONCLUSION: We conclude that placental multidrug resistance expression, specifically ABCG2, is regulated by oxygen tension in the first trimester. It is possible that changes in placental oxygen supply are capable of altering fetal drug exposure especially during early pregnancy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Drug Resistance, Multiple , Gene Expression Regulation, Developmental , Neoplasm Proteins/metabolism , Oxygen/metabolism , Placenta/metabolism , Vascular Endothelial Growth Factor A/metabolism , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Cell Hypoxia , Female , Giant Cells/cytology , Giant Cells/drug effects , Giant Cells/metabolism , Humans , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Placenta/cytology , Placenta/drug effects , Placentation , Pregnancy , Pregnancy Trimester, First , Pregnancy Trimester, Second , Pregnancy Trimester, Third , RNA, Messenger/metabolism , Tissue Banks , Tissue Culture Techniques , Trophoblasts/cytology , Trophoblasts/drug effects , Trophoblasts/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
10.
J Dev Orig Health Dis ; 4(1): 77-89, 2013 Feb.
Article in English | MEDLINE | ID: mdl-25080184

ABSTRACT

In this study, we determined the gene and/or protein expression of hypothalamic-pituitary-adrenal (HPA) axis regulatory molecules following synthetic glucocorticoid exposures. Pregnant sheep received intramuscular saline or betamethasone (BET) injections at 104 (BET-1), 104 and 111(BET-2) or 104, 111 and 118 (BET-3) days of gestation (dG). Samples were collected at numerous time-points between 75 dG and 12 weeks postnatal age. In the BET-3 treatment group, fetal plasma cortisol levels were lower at 145 dG than controls and gestational length was lengthened significantly. The cortisol:adrenocorticotropic hormone (ACTH) ratio in fetal plasma of control and BET-3 fetuses rose significantly between132 and 145 dG, and remained elevated in lambs at 6 and 12 weeks of age; this rise was truncated at day 145 in fetuses of BET-3 treated mothers. After BET treatment, fetal and postnatal pituitary proopiomelanocortin mRNA levels were reduced from 109 dG to 12 weeks postnatal age; pituitary prohormone convertase 1 and 2 mRNA levels were reduced at 145 dG and postnatally; hypothalamic arginine vasopressin mRNA levels were lowered at all time-points, but corticotrophin-releasing hormone mRNA levels were reduced only in postnatal lambs. Maternal BET increased late fetal and/or postnatal adrenal mRNA levels of ACTH receptor and 3ß hydroxysteroid dehydrogenase but decreased steroidogenic acute regulatory protein and P450 17-α hydroxylase. The altered mRNA levels of key HPA axis regulatory proteins after maternal BET injections suggests processes that may subserve long-term changes in HPA activity in later life after prenatal exposure to synthetic glucocorticoids.


Subject(s)
Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Pituitary-Adrenal System/drug effects , Prenatal Exposure Delayed Effects/metabolism , 3-Hydroxysteroid Dehydrogenases/metabolism , Age Factors , Animals , Animals, Newborn , Betamethasone/administration & dosage , Betamethasone/pharmacology , Dose-Response Relationship, Drug , Female , Fetus/drug effects , Gene Expression Regulation/genetics , Gestational Age , Glucocorticoids/administration & dosage , Hydrocortisone/blood , Phosphoproteins/metabolism , Pregnancy , Receptors, Corticotropin/metabolism , Sheep
11.
Genes Brain Behav ; 11(6): 684-94, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22574669

ABSTRACT

The dopamine pathway and especially the dopamine receptors 1 and 2 (DRD1 and DRD2) are implicated in the regulation of mothering in rats. Evidence for this in humans is lacking. Here, we show that genetic variation in both DRD1 and DRD2 genes in a sample of 187 Caucasian mothers predicts variation in distinct maternal behaviors during a 30-min mother-infant interaction at 6 months postpartum. Two DRD1 single-nucleotide polymorphisms (SNPs rs265981 and rs686) significantly associated with maternal orienting away from the infant (P = 0.002 and P = 0.003, respectively), as did DRD1 haplotypes (P = 0.03). Two DRD2 SNPs (rs1799732 and rs6277) significantly associated with maternal infant-directed vocalizing (P = 0.001 and P = 0.04, respectively), as did DRD2 haplotypes (P = 0.01). We present evidence for heterosis in DRD1 where heterozygote mothers orient away from their infants significantly less than either homozygote group. Our findings provide important evidence that genetic variation in receptors critical for mothering in non-human species also affect human maternal behaviors. The findings also highlight the importance of exploring multiple dimensions of the complex human mothering phenotype.


Subject(s)
Maternal Behavior/physiology , Receptors, Dopamine D1/physiology , Receptors, Dopamine D2/physiology , Adult , Female , Haplotypes , Humans , Hybrid Vigor/genetics , Hybrid Vigor/physiology , Infant , Maternal Behavior/psychology , Mothers/psychology , Polymorphism, Single Nucleotide/genetics , Pregnancy , Receptors, Dopamine D1/genetics , Receptors, Dopamine D2/genetics , Young Adult
12.
Placenta ; 33(3): 137-42, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22265855

ABSTRACT

The human placenta has a number of protective mechanisms that help to prevent potentially harmful compounds from entering the fetal compartment. Two important transporter proteins are phospho-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) which are mainly expressed in the syncytiotrophoblast where they actively extrude a wide range of xenobiotics. The expression profile of these transporters varies with advancing gestation. P-gp has been shown to decline near term, leaving the fetus susceptible to potentially teratogenic drugs commonly administered to pregnant women (i.e., synthetic glucocorticoids, selective serotonin reuptake inhibitors, glyburide, antiretrovirals, etc.). Drug transporter expression is regulated by a number of transcription factors, and steroid hormones present during pregnancy, such as progesterone, estrogen and corticosteroids. Drug transporter levels have also been found to be altered in pathological pregnancies (preterm, pre-eclampsia, growth restriction and infection). Genetic variation in the genes that encode these drug transporters can significantly alter transporter function and may play a significant role in determining the susceptibility of a fetus to maternally-administered therapeutic drugs. Understanding the regulation of placental drug transporters in normal and pathological pregnancies is critical to further our knowledge of fetal development, and may lead to the development of more selectively-targeted maternal and fetal drug treatments.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Fetal Diseases/chemically induced , Fetal Diseases/prevention & control , Membrane Transport Proteins/physiology , Pharmaceutical Preparations/metabolism , Placenta/metabolism , Animals , Biological Transport/genetics , Biological Transport/physiology , Female , Fetal Diseases/genetics , Fetus/drug effects , Fetus/metabolism , Humans , Maternal-Fetal Exchange/drug effects , Maternal-Fetal Exchange/genetics , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Placenta/physiology , Pregnancy
13.
Genes Brain Behav ; 10(3): 325-33, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21232011

ABSTRACT

Maternal behavior in the new mother is a multidimensional set of responses to infant cues that are influenced by the mother's early life experiences. In this study, we wanted to test if mothers' early life experiences and mothers' genotype have interactive effects on maternal behaviors and attitudes, something which has not been previously explored. In a sample of 204 mothers, we assessed maternal genotype at the serotonin transporter-linked polymorphic region (5-HTTLPR) and an adjacent upstream polymorphism (rs25531), together giving rise to three alleles: short (S), L(G) and L(A). Controlling for maternal age and parity, we showed that this genotype can predict differences in maternal sensitivity at 6 months postpartum: mothers with an S (or the functionally similar L(G)) allele were more sensitive than mothers who lacked the allele during a 30-min recorded mother-infant interaction (F (4,140) = 3.43; P = 0.01). Furthermore, we found highly significant gene-environment interactions in association with maternal behavior, such that mothers with no S or L(G) alleles oriented away more frequently from their babies if they also reported more negative early care quality (F (5,138) = 3.28; P = 0.008). Finally, we found significant gene-environment associations with maternal attitudes; mothers with the S allele and with greater early care quality scored higher on ratings of their perceived attachment to their baby (F (5,125) = 3.27; P = 0.008). The regression results show significant interactions between the reported quality of care mothers received from their own parents and genotype on both their frequency of orienting away from the infant during the interaction (F(5, 138) = 3.28; P = 0.008, Fig. 1a) and their perceived attachment feelings to the infant (F(5, 125) = 3.27; P = 0.008, Fig. 1b); however the direction of the effects for these two outcome measures were different from one another. With increasing care quality, mothers with the L(A)L(A) genotype (no S or L(G) allele) oriented away less frequently, while S or L(G) allele carriers showed no significant change. In contrast, with increasing early care quality. L(A)L(A) (no S or L(G) allele) mothers scored lower on perceived attachment to their infants, whereas S or L(G) allele carrying mothers scored higher. [corrected].


Subject(s)
Brain Chemistry/genetics , Gene Frequency/genetics , Genetic Variation/genetics , Maternal Behavior/physiology , Mother-Child Relations , Serotonin Plasma Membrane Transport Proteins/genetics , Child, Preschool , Cohort Studies , Female , Genotype , Humans , Infant , Infant, Newborn , Longitudinal Studies , Polymorphism, Genetic/genetics , Predictive Value of Tests , Pregnancy , Serotonin/metabolism
14.
Placenta ; 31(9): 803-10, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20655586

ABSTRACT

Previously, we and others have shown that placental Abcb1 mRNA and phosphoglycoprotein (P-gp; encoded by Abcb1 mRNA) decreases over the second half of gestation, resulting in increased accumulation of P-gp substrates in the fetal compartment. Very little is known pertaining to the regulation of placental Abcb1 mRNA and P-gp. In non-placental adult murine tissues, synthetic glucocorticoids have been shown to regulate Abcb1 (Abcb1a and Abcb1b) mRNA in an isoform and tissue-specific manner. Furthermore, given that maternal and fetal endogenous glucocorticoid levels increase dramatically in late gestation, we hypothesized that synthetic glucocorticoids down-regulate placental Abcb1 and P-gp expression, consequently decreasing placental P-gp mediated fetal protection. Pregnant FVB mice were treated with dexamethasone (0.1 mg/kg or 1 mg/kg; s.c.) or vehicle (saline) from either embryonic day (E)9.5-15.5 or E12.5-E18.5 and then injected with [3H]digoxin (i.v.) to assess placental P-gp function. Dexamethasone treatment from E12.5-E18.5 significantly up-regulated Abcb1a mRNA (1 mg/kg) and P-gp (0.1 mg/kg and 1 mg/kg) expression on E18.5; however, this did not correlate to changes in drug accumulation in the fetal compartment. Similarly, dexamethasone (1 mg/kg) treatment during mid-gestation (E9.5-E15.5) significantly increased placental Abcb1a mRNA. In conclusion, glucocorticoids exhibit complex regulation of the P-gp transport system at the level of gene transcription and translation. Dexamethasone exposure up-regulates Abcb1a mRNA and P-gp protein, particularly in late gestation. However, these changes do not appear to be reflected by changes in P-gp mediated drug transfer. While the latter is somewhat reassuring with respect to antenatal use of glucocorticoids for management of preterm labour, further studies are required to understand regulation of these important drug transporters in the placenta.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP-Binding Cassette Transporters/biosynthesis , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Placenta/drug effects , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Digoxin/metabolism , Down-Regulation , Female , Gene Expression Regulation, Developmental/drug effects , Mice , Placenta/metabolism , Pregnancy , Up-Regulation
15.
Placenta ; 31(5): 351-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20347140

ABSTRACT

Recent studies have illustrated the importance of placental drug transport proteins, such as P-glycoprotein (Pgp) and breast cancer resistance protein (BCRP) in limiting fetal exposure to drugs and toxins. Moreover, increasing evidence supports a role for Pgp and BCRP in the normal development and physiological function of the placenta. Several single nucleotide polymorphisms (SNPs) in the genes encoding Pgp and BCRP have been described and are associated with altered protein expression, transporter activity, and clinical outcome in studies focusing on tissues other than the placenta. This review aims to summarize current research regarding the association between these polymorphisms and expression and function in the placenta. The influence of these genotypes on fetal drug exposure and altered placental physiology or development is also presented. To date, evidence suggests that SNPs in both ABCB1 and ABCG1 can alter expression of their respective protein; however, the functional significance of these polymorphisms is less clear. An understanding of this genotype-phenotype relationship will allow for prediction of susceptible or favorable genotypes in order to personalize medication choices to minimize fetal exposure to teratogens, or to maximize pharmacological therapy to the fetus.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP-Binding Cassette Transporters/genetics , Drug Resistance, Multiple/genetics , Fetus/metabolism , Maternal-Fetal Exchange/genetics , Neoplasm Proteins/genetics , Placenta/metabolism , Polymorphism, Single Nucleotide , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Adult , Biological Transport , Female , Fetus/drug effects , Genotype , Humans , Maternal-Fetal Exchange/drug effects , Neoplasm Proteins/metabolism , Placenta/drug effects , Pregnancy , Xenobiotics/pharmacokinetics
16.
Placenta ; 31(2): 97-105, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20045184

ABSTRACT

Synthetic glucocorticoids (GCs) are given to women with threatened preterm labour but their administration has been linked to reduced infant birthweight. The underlying mechanisms are unknown, but impaired placental development and/or function has been implicated. The activity of the system A amino acid transporter is decreased in placentas from pregnancies complicated by fetal growth restriction. Whether GCs adversely affect placental amino acid transport is unknown. The objective of this study was to determine the regulatory effects of GCs on system A transport using a human in vitro placental explant model. Term explants (n=7) were treated with dexamethasone (DEX 10(-8)M or 10(-6)M) or vehicle for 48 h. System A activity was measured by the uptake of (14)C-N-methylated aminoisobutyric acid by explants. Explants were also processed for electron microscopy (EM), immunohistochemistry, and qRT-PCR. Lactate dehydrogenase (LDH), human chorionic gonadotropin (hCG) and human placental lactogen (hPL) release into the culture medium was measured. DEX (10(-6)M) stimulated system A activity compared to vehicle (p<0.05). System A transporter proteins were localized to the newly regenerating syncytiotrophoblast layer, but mRNA levels were unchanged with DEX treatment. DEX did not adversely affect explant viability as determined by reduced LDH release (p<0.05). DEX treatment was associated with morphological (accelerated apical microvilli formation, nuclear maturation, and increased cell organelle number) and functional (elevated hCG secretion, increased 11beta-HSD2 mRNA expression and reduced cytotrophoblast proliferation (p<0.05 for all)) markers of syncytiotrophoblast differentiation. These findings suggest that DEX stimulates system A activity and promotes syncytiotrophoblast differentiation and maturation.


Subject(s)
Amino Acid Transport System A/metabolism , Cell Differentiation/drug effects , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Placenta/drug effects , Trophoblasts/drug effects , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Cell Proliferation/drug effects , Cell Shape/drug effects , Chorionic Gonadotropin/metabolism , Dose-Response Relationship, Drug , Female , Gene Expression Regulation/drug effects , Gene Products, env/genetics , Gene Products, env/metabolism , Humans , In Vitro Techniques , Ki-67 Antigen/metabolism , Placenta/cytology , Placenta/enzymology , Placenta/physiology , Pregnancy , Pregnancy Proteins/genetics , Pregnancy Proteins/metabolism , Protein Isoforms/metabolism , Regeneration/drug effects , Term Birth , Time Factors , Trophoblasts/cytology , Trophoblasts/metabolism
17.
Reprod Fertil Dev ; 20(2): 328-34, 2008.
Article in English | MEDLINE | ID: mdl-18255023

ABSTRACT

Breast cancer resistance protein (BCRP) is a multidrug resistant ABC transport protein (ABCG-2). It extrudes a wide range of substrates, including many chemotherapy drugs, steroids and folate. It is present in many cancers, as well as normal tissues, in particular barrier tissues such as the blood-brain barrier, the intestine, blood vessels and the human placenta. Human fetal membranes (amnion and chorion laeve) provide the barrier between the maternal uterine environment and the fetus. In the present study, we defined the expression and localisation of BCRP mRNA and protein in human fetal membranes (amnion and chorion) and attached decidua obtained before and following labour at term. BCRP protein and mRNA was expressed in all tissues examined and the levels of expression were not altered by labour. BCRP was localised to the amnion epithelial cells, chorion trophoblast cells and decidua stromal cells, as well as the endothelial cells of maternal blood vessels in the decidua, but was absent from mesenchymal cells. In the amnion epithelium, BCRP protein was localised to the apical surface, cytoplasm and membrane between cells. In the chorion trophoblast and decidua stromal cells, BCRP protein was localised to the plasma membrane. However, in the chorion trophoblast, BCRP protein was also highly expressed in the nucleus. The level of BCRP protein in the membranes was comparable to that in the placenta. These high levels raise the possibility that this transporter plays an important role in the physiological function of the tissues.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Decidua/metabolism , Extraembryonic Membranes/metabolism , Gene Expression Regulation, Developmental/physiology , Neoplasm Proteins/metabolism , RNA, Messenger/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Analysis of Variance , Blotting, Western , DNA Primers/genetics , Female , Humans , Immunohistochemistry , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction
18.
Placenta ; 28(10): 1073-81, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17524480

ABSTRACT

Breast Cancer Resistance Protein (BCRP), a recently-discovered transporter belonging to ABC superfamily, is highly expressed within the labyrinth of the placenta, the primary site of exchange between the maternal and fetal circulation. It has been proposed to function as an efflux pump protecting the fetus from a wide range of xenobiotics. It has also been recently shown that the yolk sac, in addition to the placenta, may be involved in transport of certain substances to and from the fetus. We hypothesised that there are changes in placental Bcrp1 (the mouse orthologue of human BCRP) expression during pregnancy and that these correlate with changes in progesterone production that occur in late gestation. We also hypothesised that Bcrp1 is expressed in the yolk sac, and that levels change with advancing gestation. Either whole concepti, or placenta and yolk sac, were collected from pregnant mice and analysed at embryonic (E) day 9.5, 12.5, 15.5 and 18.5 (term approximately E19.5). Peak expression of Bcrp1 mRNA was detected using in situ hybridisation within the placenta at E9.5 and the yolk sac at E12.5. There was a significant decrease thereafter in both tissues (p<0.001). In contrast, expression of Bcrp1 protein as assessed by immunohistochemistry and Western immunoblots did not change significantly during gestation either in the placenta nor the yolk sac, and no sex difference in Bcrp1 protein expression in either tissue was observed at E12.5. Daily progesterone treatment starting at E14.5 and continuing until E18.5 significantly increased maternal progesterone levels, but did not elicit any changes in the Bcrp1 mRNA or Bcrp1 protein expression either in the placenta or the yolk sac. Significant expression of Bcrp1 protein in fetal tissue was evident at the end of gestation, while expression in the fetal brain endothelium was evident as early as E12.5. We suggest that the placenta and the yolk sac, both of which express Bcrp1, may limit fetal exposure to the potentially adverse effects of xenobiotics including therapeutic drugs which the mother may be exposed to during pregnancy. The significant decrease in Bcrp1 mRNA expression in both the yolk sac and the placenta from mid to late gestation may be counter-balanced by an increase in Bcrp1 expression in fetal organs involved in absorption, excretion and protection.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Placenta/metabolism , Progesterone/pharmacology , Yolk Sac/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Animals , Female , Mice , Placenta/drug effects , Pregnancy , Progesterone/blood , RNA, Messenger/metabolism , Yolk Sac/drug effects
19.
J Neuroendocrinol ; 19(3): 172-80, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17280590

ABSTRACT

The neurodevelopmental consequences of prenatal glucocorticoid exposure are not well-understood, particularly in species that give birth to neuroanatomically mature offspring. In the present study, we hypothesised that repeated prenatal glucocorticoid administration would alter hypothalamo-pituitary-adrenal (HPA) function in juvenile guinea pig offspring. Pregnant guinea pigs were injected with betamethasone (1 mg/kg) or vehicle on gestational days 40, 41, 50, 51, 60 and 61 (six doses). Prenatal glucocorticoid exposure abolished the pituitary-adrenal response to maternal separation in juvenile males, but had no effect in female offspring. Indeed, female offspring (vehicle and betamethasone) did not mount a significant HPA response to separation at 10 days of age. Although there were no effects of prenatal glucocorticoid exposure on hippocampal or hypothalamic corticosteroid receptor expression or corticotrophin-releasing factor (CRF) mRNA, there were significant effects in the pituitary and adrenal; again males were more affected than females. Prenatal glucocorticoid exposure increased pituitary pro-opiomelanocortin and CRF receptor mRNA, and markedly decreased adrenocortical CYP17 mRNA. In conclusion, repeated prenatal glucocorticoid exposure has profound influences on HPA function and regulation in the juvenile guinea pig, and this involves altered regulation at the level of the pituitary and adrenal cortex. Furthermore, juvenile males appear to be more vulnerable to the effects of prenatal glucocorticoid exposure than females.


Subject(s)
Betamethasone/pharmacology , Glucocorticoids/pharmacology , Pituitary-Adrenal System/drug effects , Prenatal Exposure Delayed Effects , Stress, Psychological/physiopathology , Adrenocorticotropic Hormone/blood , Analysis of Variance , Animals , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Drug Administration Schedule , Exploratory Behavior/physiology , Female , Guinea Pigs , Hippocampus/metabolism , Hydrocortisone/blood , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiopathology , Hypothalamus/metabolism , Male , Maternal Deprivation , Pituitary-Adrenal System/physiopathology , Pregnancy , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , RNA, Messenger/analysis , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Sex Factors , Social Isolation , Statistics, Nonparametric
20.
J Neuroendocrinol ; 18(11): 826-34, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17026532

ABSTRACT

Exposure to high cortisol concentration can injure the developing brain, possibly via an excitotoxic mechanism involving glutamate (Glu). The present study tested the hypothesis that chronic prenatal ethanol exposure (CPEE) activates the foetal hypothalamic-pituitary-adrenal axis to produce high cortisol exposure in the foetal compartment and alters sensitivity to glucocorticoid-induced Glu release in the foetal hippocampus. Pregnant guinea pigs received daily oral administration of ethanol (4 g/kg maternal body weight/day) or isocaloric-sucrose/pair-feeding from gestational day (GD) 2 until GD 63 (term, approximately GD 68) at which time they were euthanised, 1 h after their final treatment. Adrenocorticotrophic hormone (ACTH) and cortisol concentrations were determined in foetal plasma. Basal and electrically stimulated Glu and gamma-aminobutyric acid (GABA) efflux in the presence or absence of dexamethasone (DEX), a selective glucocorticoid-receptor agonist, were determined ex vivo in foetal hippocampal slices. Glucocorticoid receptor (GR), mineralocorticoid receptor (MR) and N-methyl-D-aspartate (NMDA) receptor NR1 subunit mRNA expression were determined in situ in the hippocampus and dentate gyrus. In the near-term foetus, CPEE increased foetal plasma ACTH and cortisol concentrations. Electrically stimulated glutamate, but not GABA, release was increased in CPEE foetal hippocampal slices. Low DEX concentration (0.3 microM) decreased stimulated glutamate, but not GABA, release in both CPEE and control foetal hippocampal slices. High DEX concentration (3.0 microM) increased basal release of Glu, but not GABA, in CPEE foetal hippocampal slices. GR, but not MR, mRNA expression was elevated in the hippocampus and dentate gyrus, whereas NR1 mRNA expression was increased in the CA1 and CA3 fields of the foetal hippocampus. These data demonstrate that CPEE increases high glucocorticoid concentration-induced Glu release in the foetal hippocampus, presumably as a consequence of increased GR expression. These effects of CPEE, coupled with increased glutamate release and increased NMDA receptor expression, may predispose the near-term foetal hippocampus to GR and Glu-NMDA receptor-mediated neurodevelopmental toxicity.


Subject(s)
Ethanol/toxicity , Fetus/drug effects , Glucocorticoids/metabolism , Glutamic Acid/drug effects , Hippocampus/drug effects , Neurotoxins/toxicity , Adrenocorticotropic Hormone/blood , Animals , Central Nervous System Depressants/toxicity , Electric Stimulation , Female , Fetus/metabolism , Glutamic Acid/metabolism , Guinea Pigs , Hippocampus/metabolism , Hydrocortisone/blood , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Maternal-Fetal Exchange , Organ Culture Techniques , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Pregnancy , RNA, Messenger/analysis , Random Allocation , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/drug effects , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Statistics, Nonparametric , Toxicity Tests, Chronic
SELECTION OF CITATIONS
SEARCH DETAIL
...