Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
FEBS Open Bio ; 11(8): 2364-2382, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34235895

ABSTRACT

We previously found that DNA methyltransferase 3a (DNMT3a) plays an important role in regulating embryonic cardiomyocyte gene expression, morphology, and function. In this study, we investigated the role of the most abundant DNMT in mammalian cells, DNMT1, in these processes. It is known that DNMT1 is essential for embryonic development, during which it is involved in regulating cardiomyocyte DNA methylation and gene expression. We used siRNA to knock down DNMT1 expression in primary cultures of mouse embryonic cardiomyocytes. Immunofluorescence staining and multielectrode array were, respectively, utilized to evaluate cardiomyocyte growth and electrophysiology. RNA sequencing (RNA-Seq) and multiplex bisulfite sequencing were, respectively, performed to examine gene expression and promoter methylation. At 72 h post-transfection, reduction of DNMT1 expression decreased the number and increased the size of embryonic cardiomyocytes. Beat frequency and the amplitude of field action potentials were decreased by DNMT1 siRNA. RNA-Seq analysis identified 801 up-regulated genes and 494 down-regulated genes in the DNMT1 knockdown cells when compared to controls. Pathway analysis of the differentially expressed genes revealed pathways that were associated with cell death and survival, cell morphology, cardiac function, and cardiac disease. Alternative splicing analysis identified 929 differentially expressed exons, including 583 up-regulated exons and 308 down-regulated exons. Moreover, decreased methylation levels were found in the promoters of cardiac genes Myh6, Myh7, Myh7b, Tnnc1, Tnni3, Tnnt2, Nppa, Nppb, mef2c, mef2d, Camta2, Cdkn1A, and Cdkn1C. Of these 13 genes, 6 (Myh6, Tnnc1, Tnni3, Tnnt2, Nppa, Nppb) and 1 (Cdkn1C) had increased or decreased gene expression, respectively. Altogether, these data show that DNMT1 is important in embryonic cardiomyocytes by regulating DNA methylation, gene expression, gene splicing, and cell function.

2.
Mol Aspects Med ; 55: 110-117, 2017 06.
Article in English | MEDLINE | ID: mdl-28202385

ABSTRACT

There is growing evidence that disruption in the prenatal environment can have long-lasting effects on an individual's health in adulthood. Research on the fetal programming of adult diseases, including cardiovascular disease, focuses on epi-mutations, which alter the normal pattern of epigenetic factors such as DNA methylation, miRNA expression, or chromatin modification, rather than traditional genetic alteration. Thus, understanding how in utero chemical exposures alter epigenetics and lead to adult disease is of considerable public health concern. Few signaling molecules have the potential to influence the developing mammal as the nucleoside adenosine. Adenosine levels increase rapidly with tissue hypoxia and inflammation. Adenosine antagonists including the methlyxanthines caffeine and theophylline are widely consumed during pregnancy. The receptors that transduce adenosine action are the A1, A2a, A2b, and A3 adenosine receptors (ARs). We examined the long-term effects of in utero disruption of adenosine signaling on cardiac gene expression, morphology, and function in adult offspring. One substance that fetuses are frequently exposed to is caffeine, which is a non-selective adenosine receptor antagonist. Over the past several years, we examined the role of adenosine signaling during embryogenesis and cardiac development. We discovered that in utero alteration in adenosine action leads to adverse effects on embryonic and adult murine hearts. We find that cardiac A1ARs protect the embryo from in utero hypoxic stress, a condition that causes an increase in adenosine levels. After birth in mice, we observed that in utero caffeine exposure leads to abnormal cardiac function and morphology in adults, including an impaired response to ß-adrenergic stimulation. Recently, we observed that in utero caffeine exposure induces transgenerational effects on cardiac morphology, function, and gene expression. Our findings indicate that the effects of altered adenosine signaling are dependent on signaling through the A1ARs and timing of disruption. In addition, the long-term effects of altered adenosine signaling appear to be mediated by alterations in DNA methylation, an epigenetic process critical for normal development.


Subject(s)
Adenosine/genetics , Embryonic Development/genetics , Epigenesis, Genetic/genetics , Receptors, Purinergic P1/genetics , Adenosine/metabolism , Animals , Caffeine/toxicity , DNA Methylation/genetics , Embryonic Development/drug effects , Humans , Mice , Receptors, Purinergic P1/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
3.
Cereb Cortex ; 27(5): 3015-3027, 2017 05 01.
Article in English | MEDLINE | ID: mdl-27235101

ABSTRACT

Insufficient or excessive thyroid hormone (TH) levels during fetal development can cause long-term neurological and cognitive problems. Studies in animal models of perinatal hypo- and hyperthyroidism suggest that these problems may be a consequence of the formation of maladaptive circuitry in the cerebral cortex, which can persist into adulthood. Here we used mouse models of maternal hypo- and hyperthyroidism to investigate the long-term effects of altering thyroxine (T4) levels during pregnancy (corresponding to embryonic days 6.5-18.5) on thalamocortical (TC) axon dynamics in adult offspring. Because perinatal hypothyroidism has been linked to visual processing deficits in humans, we performed chronic two-photon imaging of TC axons and boutons in primary visual cortex (V1). We found that a decrease or increase in maternal serum T4 levels was associated with atypical steady-state dynamics of TC axons and boutons in V1 of adult offspring. Hypothyroid offspring exhibited axonal branch and bouton dynamics indicative of an abnormal increase in TC connectivity, whereas changes in hyperthyroid offspring were indicative of an abnormal decrease in TC connectivity. Collectively, our data suggest that alterations to prenatal T4 levels can cause long-term synaptic instability in TC circuits, which could impair early stages of visual processing.


Subject(s)
Hyperthyroidism/pathology , Hypothyroidism/pathology , Prenatal Exposure Delayed Effects/physiopathology , Synapses/physiology , Thalamus/pathology , Visual Cortex/pathology , Adult , Animals , Animals, Newborn , Antithyroid Agents/toxicity , Brain Mapping , Disease Models, Animal , Female , Gestational Age , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Hyperthyroidism/diagnostic imaging , Hypothyroidism/diagnostic imaging , Image Processing, Computer-Assisted , Male , Methimazole/toxicity , Mice , Mice, Inbred C57BL , Neuroimaging , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/diagnostic imaging , Synapsins/genetics , Synapsins/metabolism , Thalamus/diagnostic imaging , Thyroxine/toxicity , Time Factors , Transduction, Genetic , Visual Cortex/diagnostic imaging
4.
Sci Rep ; 6: 34106, 2016 Sep 28.
Article in English | MEDLINE | ID: mdl-27677355

ABSTRACT

Each year millions of pregnant woman are exposed to caffeine, which acts to antagonize adenosine action. The long-term consequences of this exposure on the developing fetus are largely unknown, although in animal models we have found adverse effects on cardiac function. To assess if these effects are transmitted transgenerationally, we exposed pregnant mice to caffeine equivalent to 2-4 cups of coffee at two embryonic stages. Embryos (F1 generation) exposed to caffeine early from embryonic (E) day 6.5-9.5 developed a phenotype similar to dilated cardiomyopathy by 1 year of age. Embryos exposed to caffeine later (E10.5-13.5) were not affected. We next examined the F2 generation and F3 generation of mice exposed to caffeine from E10.5-13.5, as this coincides with germ cell development. These F2 generation adult mice developed a cardiac phenotype similar to hypertrophic cardiomyopathy. The F3 generation exhibited morphological changes in adult hearts, including increased mass. This report shows that in utero caffeine exposure has long-term effects into adulthood and that prenatal caffeine exposure can exert adverse transgenerational effects on adult cardiac function.

5.
FASEB J ; 30(9): 3238-55, 2016 09.
Article in English | MEDLINE | ID: mdl-27306334

ABSTRACT

We previously found that in utero caffeine exposure causes down-regulation of DNA methyltransferases (DNMTs) in embryonic heart and results in impaired cardiac function in adulthood. To assess the role of DNMTs in these events, we investigated the effects of reduced DNMT expression on embryonic cardiomyocytes. siRNAs were used to knock down individual DNMT expression in primary cultures of mouse embryonic cardiomyocytes. Immunofluorescence staining was conducted to evaluate cell morphology. A video-based imaging assay and multielectrode array were used to assess cardiomyocyte contractility and electrophysiology, respectively. RNA-Seq and multiplex bisulfite sequencing were performed to examine gene expression and promoter methylation, respectively. At 72 h after transfection, reduced DNMT3a expression, but not DNMT1 or -3b, disrupted sarcomere assembly and decreased beating frequency, contractile movement, amplitude of field action potential, and cytosolic calcium signaling of cardiomyocytes. RNA-Seq analysis revealed that the DNMT3a-deficient cells had deactivated gene networks involved in calcium, endothelin-1, renin-angiotensin, and cardiac ß-adrenergic receptor signaling, which were not inhibited by DNMT3b siRNA. Moreover, decreased methylation levels were found in the promoters of Myh7, Myh7b, Tnni3, and Tnnt2, consistent with the up-regulation of these genes by DNMT3a siRNA. These data show that DNMT3a plays an important role in regulating embryonic cardiomyocyte gene expression, morphology and function.-Fang, X., Poulsen, R. R., Wang-Hu, J., Shi, O., Calvo, N. S., Simmons, C. S., Rivkees, S. A., Wendler, C. C. Knockdown of DNA methyltransferase 3a alters gene expression and inhibits function of embryonic cardiomyocytes.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Embryo, Mammalian/physiology , Gene Expression Regulation, Developmental/physiology , Myocytes, Cardiac/enzymology , Action Potentials/physiology , Animals , Apoptosis , Calcium Signaling/physiology , Cell Survival , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Down-Regulation , Gene Knockdown Techniques , Mice , Sarcomeres , DNA Methyltransferase 3B
6.
Am J Physiol Cell Physiol ; 309(6): C425-36, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26224577

ABSTRACT

cAMP is a highly regulated secondary messenger involved in many biological processes. Chronic activation of the cAMP pathway by catecholamines results in cardiac hypertrophy and fibrosis; however, the mechanism by which elevated cAMP leads to cardiomyopathy is not fully understood. To address this issue, we increased intracellular cAMP levels in HL-1 cardiomyocytes, a cell line derived from adult mouse atrium, using either the stable cAMP analog N(6),2'-O-dibutyryladenosine 3',5'-cyclic monophosphate (DBcAMP) or phosphodiesterase (PDE) inhibitors caffeine and theophylline. Elevated cAMP levels increased cell size and altered expression levels of cardiac genes and micro-RNAs associated with hypertrophic cardiomyopathy (HCM), including Myh6, Myh7, Myh7b, Tnni3, Anp, Bnp, Gata4, Mef2c, Mef2d, Nfatc1, miR208a, and miR208b. In addition, DBcAMP altered the expression of DNA methyltransferases (Dnmts) and Tet methylcytosine dioxygenases (Tets), enzymes that regulate genomic DNA methylation levels. Changes in expression of DNA methylation genes induced by elevated cAMP led to increased global DNA methylation in HL-1 cells. In contrast, inhibition of DNMT activity with 5-azacytidine treatment decreased global DNA methylation levels and blocked the increased expression of several HCM genes (Myh7, Gata4, Mef2c, Nfatc1, Myh7b, Tnni3, and Bnp) observed with DBcAMP treatment. These results demonstrate that cAMP induces cardiomyocyte hypertrophy and altered HCM gene expression in vitro and that DNA methylation patterns mediate the upregulation of HCM genes induced by cAMP. These data identify a previously unknown mechanism by which elevated levels of cAMP lead to increased expression of genes associated with cardiomyocyte hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomyopathies/genetics , Cyclic AMP/metabolism , DNA Methylation/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Animals , Cardiomegaly/genetics , Cardiomyopathies/pathology , Cell Line , DNA Methylation/physiology , Gene Expression/genetics , Heart/physiopathology , Mice , MicroRNAs/genetics , Up-Regulation/genetics
7.
Am J Physiol Regul Integr Comp Physiol ; 307(12): R1471-87, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25354728

ABSTRACT

Previous studies demonstrated that in utero caffeine treatment at embryonic day (E) 8.5 alters DNA methylation patterns, gene expression, and cardiac function in adult mice. To provide insight into the mechanisms, we examined cardiac gene and microRNA (miRNA) expression in cardiomyocytes shortly after exposure to physiologically relevant doses of caffeine. In HL-1 and primary embryonic cardiomyocytes, caffeine treatment for 48 h significantly altered the expression of cardiac structural genes (Myh6, Myh7, Myh7b, Tnni3), hormonal genes (Anp and BnP), cardiac transcription factors (Gata4, Mef2c, Mef2d, Nfatc1), and microRNAs (miRNAs; miR208a, miR208b, miR499). In addition, expressions of these genes were significantly altered in embryonic hearts exposed to in utero caffeine. For in utero experiments, pregnant CD-1 dams were treated with 20-60 mg/kg of caffeine, which resulted in maternal circulation levels of 37.3-65.3 µM 2 h after treatment. RNA sequencing was performed on embryonic ventricles treated with vehicle or 20 mg/kg of caffeine daily from E6.5-9.5. Differential expression (DE) analysis revealed that 124 genes and 849 transcripts were significantly altered, and differential exon usage (DEU) analysis identified 597 exons that were changed in response to prenatal caffeine exposure. Among the DE genes identified by RNA sequencing were several cardiac structural genes and genes that control DNA methylation and histone modification. Pathway analysis revealed that pathways related to cardiovascular development and diseases were significantly affected by caffeine. In addition, global cardiac DNA methylation was reduced in caffeine-treated cardiomyocytes. Collectively, these data demonstrate that caffeine exposure alters gene expression and DNA methylation in embryonic cardiomyocytes.


Subject(s)
Caffeine/pharmacology , Gene Expression Regulation, Developmental/drug effects , Heart/drug effects , Myocytes, Cardiac/drug effects , Animals , Cell Shape/drug effects , Cell Survival/drug effects , Cells, Cultured , DNA Methylation/drug effects , Dose-Response Relationship, Drug , Female , Gene Expression Profiling , Gestational Age , Heart/embryology , Maternal Exposure , Mice , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Pregnancy , RNA, Messenger/metabolism , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Brain Res ; 1586: 64-72, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25157906

ABSTRACT

Periventricular white matter injury (PWMI) is the most common cause of brain injury in preterm infants. It is believed that loss of late oligodendrocyte progenitor cells (OPCs) and disrupted maturation of oligodendrocytes contributes to defective myelination in PWMI. At present, no clinically approved drugs are available for treating PWMI. Previously, we found that diazoxide promotes myelination and attenuates brain injury in the chronic sublethal hypoxia model of PWMI. In this study, we investigated the mechanisms by which diazoxide promotes myelination. We observed that diazoxide increases the ratio of differentiated oligodendrocytes in the cerebral white matter, promotes the expression of differentiation-associated transcriptional factors Nkx2.2 and Sox10, and increases the expression of myelin genes CNP and MBP. These results show that diazoxide promotes oligodendrocyte differentiation in the developing brain.


Subject(s)
Brain/cytology , Cell Differentiation/drug effects , Cell Hypoxia/physiology , Diazoxide/pharmacology , Oligodendroglia/drug effects , Vasodilator Agents/pharmacology , Adenomatous Polyposis Coli Protein/metabolism , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bromodeoxyuridine/metabolism , Gene Expression Regulation, Developmental/drug effects , Homeobox Protein Nkx-2.2 , Hypoxia/drug therapy , Hypoxia/pathology , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2 , RNA, Messenger/metabolism , Stem Cells/drug effects
9.
Birth Defects Res B Dev Reprod Toxicol ; 101(4): 300-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24980470

ABSTRACT

BACKGROUND: Propylthiouracil (PTU) and methimazole (MMI) are antithyroid drugs used to treat hyperthyroidism. Despite the widespread use of PTU and MMI during pregnancy, modest clinical data and less animal data are available on the teratogenic potential of these drugs. METHODS: We evaluated the teratogenicity of in utero exposure to PTU or MMI in mice and rats. First, pregnant C57Bl/6 mice were treated daily with PTU (10 or 100 mg/kg), MMI (2 or 20 mg/kg), or vehicle from gestation day (GD) 6 to 16. GD 18 fetuses were evaluated for gross and histopathological abnormalities. Next, pregnant Sprague-Dawley rats were treated daily with PTU (50 or 100 mg/kg), MMI (10 or 20 mg/kg), or vehicle from GD 6 to 19, followed by evaluation for gross and histopathological abnormalities at GD 20. RESULTS: In mice treated with PTU or MMI, no significant histopathological abnormalities or external gross malformations, and no adverse effects on placental weight, litter size, resorption rates, or fetal weight were observed at GD 18. In rats, no adverse effects on litter size, placental weights, or maternal body weights were observed with either PTU or MMI treatment. PTU treatment (50 and 100 mg/kg) and MMI (10 mg/kg) treatment resulted in a decrease in crown-rump length in rat fetuses but no external gross malformations or histopathological abnormalities were observed. CONCLUSION: We did not observe either gross external malformations or histopathological malformations in mice or rats treated long-term with high doses of PTU or MMI during pregnancy.


Subject(s)
Antithyroid Agents/toxicity , Hyperthyroidism/drug therapy , Methimazole/toxicity , Pregnancy Complications/chemically induced , Propylthiouracil/toxicity , Animals , Antithyroid Agents/pharmacology , Female , Methimazole/pharmacology , Mice , Mice, Inbred C57BL , Pregnancy , Propylthiouracil/pharmacology , Rats , Rats, Sprague-Dawley , Teratogens/toxicity
10.
PLoS One ; 9(1): e87547, 2014.
Article in English | MEDLINE | ID: mdl-24475304

ABSTRACT

Evidence indicates that disruption of normal prenatal development influences an individual's risk of developing obesity and cardiovascular disease as an adult. Thus, understanding how in utero exposure to chemical agents leads to increased susceptibility to adult diseases is a critical health related issue. Our aim was to determine whether adenosine A1 receptors (A1ARs) mediate the long-term effects of in utero caffeine exposure on cardiac function and whether these long-term effects are the result of changes in DNA methylation patterns in adult hearts. Pregnant A1AR knockout mice were treated with caffeine (20 mg/kg) or vehicle (0.09% NaCl) i.p. at embryonic day 8.5. This caffeine treatment results in serum levels equivalent to the consumption of 2-4 cups of coffee in humans. After dams gave birth, offspring were examined at 8-10 weeks of age. A1AR+/+ offspring treated in utero with caffeine were 10% heavier than vehicle controls. Using echocardiography, we observed altered cardiac function and morphology in adult mice exposed to caffeine in utero. Caffeine treatment decreased cardiac output by 11% and increased left ventricular wall thickness by 29% during diastole. Using DNA methylation arrays, we identified altered DNA methylation patterns in A1AR+/+ caffeine treated hearts, including 7719 differentially methylated regions (DMRs) within the genome and an overall decrease in DNA methylation of 26%. Analysis of genes associated with DMRs revealed that many are associated with cardiac hypertrophy. These data demonstrate that A1ARs mediate in utero caffeine effects on cardiac function and growth and that caffeine exposure leads to changes in DNA methylation.


Subject(s)
Caffeine/toxicity , DNA Methylation/drug effects , Heart/drug effects , Maternal-Fetal Exchange , Prenatal Exposure Delayed Effects/physiopathology , Receptor, Adenosine A1/metabolism , Analysis of Variance , Animals , Caffeine/metabolism , DNA Primers/genetics , Echocardiography , Female , Heart Function Tests , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Knockout , Pregnancy , Real-Time Polymerase Chain Reaction , Receptor, Adenosine A1/genetics
11.
Online J Biol Sci ; 14(1): 49-56, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-25587244

ABSTRACT

Diazoxide is an ATP-sensitive potassium channel (KATP) agonist that has been shown to neuroprotective effects. These observations raise the possibility that diazoxide may have potential as a therapeutic agent for other applications. This study investigated (1) the long term effects of chronic neonatal administration of diazoxide and (2) the role of KATP on murin behavior and neurohistology. C57B/6J pups were injected daily with diazoxide (10, 20 or 50 mg kg-1) or vehicle from Postnatal days 2 (P2) through P12. Pups were allow to mature and underwent behavioral testing at 5-7 months of age. After behavioral testing, animals were euthanized and morphology of the brains was assessed. No long term adverse effects of neonatal diazoxide therapy on physical characteristics, visual acuity, sensori-motor reflexes, spontaneous locomotor activity, motor coordination/balance or motor learning and memory were observed. In addition, no morphological changes were observed on brains. However, we did observe that diazoxide therapy causes depressive-like phenotypes in female murine mice. Chronic neonatal diazoxide therapy does not cause deficits or enhancements in mice behavior. Diazoxide does not cause abnormal morphological changes in brain anatomy. However, diazoxide does cause gender specific depressive-like phenotype in mice.

12.
FASEB J ; 27(2): 684-91, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23150524

ABSTRACT

Antithyroid medications are the preferred therapy for the treatment of Graves' disease during pregnancy. Propylthiouracil (PTU) is favored over methimazole (MMI) due to potential teratogenic concerns with MMI. This study was to determine the teratogenic potential of MMI and PTU using a validated Xenopus tropicalis embryo model. Embryos were exposed to 1 mM PTU (EC(50)=0.88 mM), 1 mM MMI, or vehicle control (water) from stages 2 to 45. Treated embryos were examined for gross morphological defects, ciliary function, and gene expression by in situ hybridization. Exposure to PTU, but not MMI, led to cardiac and gut looping defects and shortening along the anterior-posterior axis. PTU exposure during gastrulation (stage 8-12.5) was identified as the critical period of exposure leading to left-right (LR) patterning defects. Abnormal cilia polarization, abnormal cilia-driven leftward flow at the gastrocoel roof plate (GRP), and aberrant expression of both Coco and Pitx2c were associated with abnormal LR symmetry observed following PTU exposure. PTU is teratogenic during late blastula, gastrulation, and neurulation; whereas MMI is not. PTU alters ciliary-driven flow and disrupts the normal genetic program involved in LR axis determination. These studies have important implications for women taking PTU during early pregnancy.


Subject(s)
Antithyroid Agents/toxicity , Body Patterning/drug effects , Propylthiouracil/toxicity , Teratogens/toxicity , Xenopus/embryology , Animals , Antithyroid Agents/administration & dosage , Body Patterning/genetics , Cilia/drug effects , Digestive System Abnormalities/chemically induced , Digestive System Abnormalities/embryology , Female , Gene Expression Regulation, Developmental/drug effects , Graves Disease/complications , Graves Disease/drug therapy , Heart Defects, Congenital/chemically induced , Heart Defects, Congenital/embryology , Humans , Methimazole/administration & dosage , Methimazole/toxicity , Models, Animal , Pregnancy , Pregnancy Complications/drug therapy , Propylthiouracil/administration & dosage , Time Factors , Triiodothyronine/pharmacology , Xenopus/genetics
13.
PLoS One ; 7(4): e35213, 2012.
Article in English | MEDLINE | ID: mdl-22529993

ABSTRACT

BACKGROUND: Hyperthyroidism during pregnancy is treated with the antithyroid drugs (ATD) propylthiouracil (PTU) and methimazole (MMI). PTU currently is recommended as the drug of choice during early pregnancy. Yet, despite widespread ATD use in pregnancy, formal studies of ATD teratogenic effects have not been performed. METHODS: We examined the teratogenic effects of PTU and MMI during embryogenesis in mice. To span different periods of embryogenesis, dams were treated with compounds or vehicle daily from embryonic day (E) 7.5 to 9.5 or from E3.5 to E7.5. Embryos were examined for gross malformations at E10.5 or E18.5 followed by histological and micro-CT analysis. Influences of PTU on gene expression levels were examined by RNA microarray analysis. RESULTS: When dams were treated from E7.5 to E9.5 with PTU, neural tube and cardiac abnormalities were observed at E10.5. Cranial neural tube defects were significantly more common among the PTU-exposed embryos than those exposed to MMI or vehicle. Blood in the pericardial sac, which is a feature indicative of abnormal cardiac function and/or abnormal vasculature, was observed more frequently in PTU-treated than MMI-treated or vehicle-treated embryos. Following PTU treatment, a total of 134 differentially expressed genes were identified. Disrupted genetic pathways were those associated with cytoskeleton remodeling and keratin filaments. At E 18.5, no gross malformations were evident in either ATD group, but the number of viable PTU embryos per dam at E18.5 was significantly lower from those at E10.5, indicating loss of malformed embryos. These data show that PTU exposure during embryogenesis is associated with delayed neural tube closure and cardiac abnormalities. In contrast, we did not observe structural or cardiac defects associated with MMI exposure except at the higher dose. We find that PTU exposure during embryogenesis is associated with fetal loss. These observations suggest that PTU has teratogenic potential.


Subject(s)
Antithyroid Agents/toxicity , Embryonic Development/drug effects , Propylthiouracil/toxicity , Teratogens/toxicity , Animals , Antithyroid Agents/administration & dosage , Embryo, Mammalian/abnormalities , Embryo, Mammalian/drug effects , Embryo, Mammalian/pathology , Embryonic Development/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Heart/drug effects , Heart/embryology , Male , Methimazole/pharmacology , Mice , Mice, Inbred C57BL , Pregnancy , Propylthiouracil/administration & dosage , Signal Transduction/drug effects
14.
Arterioscler Thromb Vasc Biol ; 32(4): 851-5, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22423036

ABSTRACT

Few signaling molecules have as much potential to influence the developing mammal as the nucleoside adenosine. Adenosine levels increase rapidly with tissue hypoxia and inflammation. Adenosine antagonists include the methylxanthines caffeine and theophylline. The receptors that transduce adenosine action are the A1, A2a, A2b, and A3 adenosine receptors (A1AR, A2aAR, A2bAR, and A3AR). We examined how adenosine acts via A1ARs to influence embryo development. Transgenic mice were studied along with embryo cultures. Embryos lacking A1ARs were markedly growth retarded following intrauterine hypoxia exposure. Studies of mice selectively lacking A1AR in the heart identify the heart as a key site of adenosine's embryo-protective effects. Studies of isolated embryos showed that adenosine plays a key role in modulating embryo cardiac function, especially in the setting of hypoxia. When pregnant mice were treated during embryogenesis with the adenosine antagonist caffeine, adult mice had abnormal heart function. Adenosine acts via A1ARs to play an essential role in protecting the embryo against intrauterine stress, and adenosine antagonists, including caffeine, may be an unwelcome exposure for the embryo.


Subject(s)
Adenosine/metabolism , Heart/embryology , Myocardium/metabolism , Receptor, Adenosine A1/metabolism , Signal Transduction , Abnormalities, Drug-Induced/etiology , Abnormalities, Drug-Induced/metabolism , Adenosine A1 Receptor Antagonists/pharmacology , Adenosine A1 Receptor Antagonists/toxicity , Animals , Embryo Culture Techniques , Heart/drug effects , Heart Defects, Congenital/chemically induced , Heart Defects, Congenital/metabolism , Mice , Mice, Transgenic , Receptor, Adenosine A1/drug effects , Receptor, Adenosine A1/genetics , Signal Transduction/drug effects
15.
PLoS One ; 6(12): e28296, 2011.
Article in English | MEDLINE | ID: mdl-22164264

ABSTRACT

BACKGROUND: Evidence suggests that adenosine acts via cardiac A1 adenosine receptors (A1ARs) to protect embryos against hypoxia. During embryogenesis, A1ARs are the dominant regulator of heart rate, and A1AR activation reduces heart rate. Adenosine action is inhibited by caffeine, which is widely consumed during pregnancy. In this study, we tested the hypothesis that caffeine influences developing embryos by altering cardiac function. METHODOLOGY/PRINCIPAL FINDINGS: Effects of caffeine and adenosine receptor-selective antagonists on heart rate were studied in vitro using whole murine embryos at E9.5 and isolated hearts at E12.5. Embryos were examined in room air (21% O(2)) or hypoxic (2% O(2)) conditions. Hypoxia decreased heart rates of E9.5 embryos by 15.8% and in E12.5 isolated hearts by 27.1%. In room air, caffeine (200 µM) had no effect on E9.5 heart rates; however, caffeine increased heart rates at E12.5 by 37.7%. Caffeine abolished hypoxia-mediated bradycardia at E9.5 and blunted hypoxia-mediated bradycardia at E12.5. Real-time PCR analysis of RNA from isolated E9.5 and E12.5 hearts showed that A1AR and A2aAR genes were expressed at both ages. Treatment with adenosine receptor-selective antagonists revealed that SCH-58261 (A2aAR-specific antagonist) had no affects on heart function, whereas DPCPX (A1AR-specific antagonist) had effects similar to caffeine treatment at E9.5 and E12.5. At E12.5, embryonic hearts lacking A1AR expression (A1AR-/-) had elevated heart rates compared to A1AR+/- littermates, A1AR-/- heart rates failed to decrease to levels comparable to those of controls. Caffeine did not significantly affect heart rates of A1AR-/- embryos. CONCLUSIONS/SIGNIFICANCE: These data show that caffeine alters embryonic cardiac function and disrupts the normal cardiac response to hypoxia through blockade of A1AR action. Our results raise concern for caffeine exposure during embryogenesis, particularly in pregnancies with increased risk of embryonic hypoxia.


Subject(s)
Caffeine/pharmacology , Gene Expression Regulation, Developmental , Heart/embryology , Receptor, Adenosine A1/metabolism , Air , Animals , Heart Rate/drug effects , Hypoxia , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxygen/metabolism , Real-Time Polymerase Chain Reaction/methods , Receptor, Adenosine A1/genetics , Time Factors
16.
BMC Dev Biol ; 11: 37, 2011 Jun 13.
Article in English | MEDLINE | ID: mdl-21668976

ABSTRACT

BACKGROUND: Sphingosine-1-phosophate (S1P) is a biologically active sphingolipid metabolite that influences cellular events including differentiation, proliferation, and migration. S1P acts through five distinct cell surface receptors designated S1P1-5R, with S1P1R having the highest expression level in the developing heart. S1P1R is critical for vascular maturation, with its loss leading to embryonic death by E14.5; however, its function during early cardiac development is not well known. Our previous studies demonstrated that altered S1P levels adversely affects atrioventricular (AV) canal development in vitro, with reduced levels leading to cell death and elevated levels inhibiting cell migration and endothelial to mesenchymal cell transformation (EMT). RESULTS: We determined, by real-time PCR analysis, that S1P1R was expressed at least 10-fold higher than other S1P receptors in the developing heart. Immunohistochemical analysis revealed S1P1R protein expression in both endothelial and myocardial cells in the developing atrium and ventricle. Using AV canal cultures, we observed that treatment with either FTY720 (an S1P1,3,4,5R agonist) or KRP203 (an S1P1R-specific agonist) caused similar effects on AV canal cultures as S1P treatment, including induction of cell rounding, inhibition of cell migration, and inhibition of EMT. In vivo, morphological analysis of embryonic hearts at E10.5 revealed that S1P1R-/- hearts were malformed with reduced myocardial tissue. In addition to reduced myocardial tissue, E12.5 S1P1R-/- hearts had disrupted morphology of the heart wall and trabeculae, with thickened and disorganized outer compact layer and reduced fibronectin (FN) deposition compared to S1P1R+/+ littermates. The reduced myocardium was accompanied by a decrease in cell proliferation but not an increase in apoptosis. CONCLUSIONS: These data indicate that S1P1R is the primary mediator of S1P action in AV canal cultures and that loss of S1P1R expression in vivo leads to malformed embryonic hearts, in part due to reduced fibronectin expression and reduced cell proliferation.


Subject(s)
Heart/embryology , Lysophospholipids/metabolism , Myocardium/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Animals , Cell Proliferation , Embryo, Mammalian/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fibronectins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Sphingosine/metabolism
17.
Pediatr Res ; 69(4): 271-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21228731

ABSTRACT

Few signaling molecules have the potential to influence the developing mammal as the nucleoside adenosine. Adenosine levels increase rapidly with tissue hypoxia and inflammation. Adenosine antagonists include the methylxanthines caffeine and theophylline. The receptors that transduce adenosine action are the A1, A2a, A2b, and A3 adenosine receptors (ARs). In the postnatal period, A1AR activation may contribute to white matter injury in the preterm infant by altering oligodendrocyte (OL) development. In models of perinatal brain injury, caffeine is neuroprotective against periventricular white matter injury (PWMI) and hypoxic-ischemic encephalopathy (HIE). Supporting the notion that blockade of adenosine action is of benefit in the premature infant, caffeine reduces the incidence of bronchopulmonary dysplasia and CP in clinical studies. In comparison with the adverse effects on the postnatal brain, adenosine acts via A1ARs to play an essential role in protecting the embryo from hypoxia. Embryo protective effects are blocked by caffeine, and caffeine intake during early pregnancy increases the risk of miscarriage and fetal growth retardation. Adenosine and adenosine antagonists play important modulatory roles during mammalian development. The protective and deleterious effects of adenosine depend on the time of exposure and target sites of action.


Subject(s)
Adenosine/pharmacology , Embryo, Mammalian/drug effects , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Myelinated/pathology , Neuroprotective Agents/pharmacology , Adenosine/antagonists & inhibitors , Adenosine/therapeutic use , Animals , Caffeine/pharmacology , Embryo, Mammalian/physiology , Female , Humans , Hypoxia-Ischemia, Brain/drug therapy , Infant, Newborn , Infant, Premature , Leukomalacia, Periventricular/drug therapy , Neuroprotective Agents/therapeutic use , Oligodendroglia/drug effects , Oligodendroglia/pathology , Pregnancy , Protein Isoforms/genetics , Protein Isoforms/metabolism , Purinergic P1 Receptor Antagonists/pharmacology , Receptors, Purinergic P1/genetics , Receptors, Purinergic P1/metabolism , Respiration/drug effects
18.
BMC Dev Biol ; 10: 57, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20509906

ABSTRACT

BACKGROUND: Our understanding of the mechanisms that protect the developing embryo from intrauterine stress is limited. Recently, adenosine has been demonstrated to play a critical role in protecting the embryo against hypoxia via adenosine A1 receptors (A1ARs), which are expressed in the heart, nervous system, and other sites during development. However, the sites of A1AR action that mediate embryo protection are not known. To determine if the heart is a key site of adenosine-mediated embryo protection, A1ARs were selectively deleted in the embryonic heart using a Cre-LoxP system in which the alpha-myosin heavy chain promoter drives Cre-recombinase expression and excision of the A1AR gene from cardiomyocytes. RESULTS: With increasing exposure of maternal hypoxia (10% O2) from 48-96 hours beginning at embryonic day (E) 8.5, embryo viability decreased in the cardiac-A1AR deleted embryos. 48 hours of hypoxia reduced embryonic viability by 49% in embryos exposed from E10.5-12.5 but no effect on viability was observed in younger embryos exposed to hypoxia from E8.5-10.5. After 72 hours of hypoxia, 57.8% of the cardiac-A1AR deleted embryos were either dead or re-absorbed compared to 13.7% of control littermates and after 96 hours 81.6% of cardiac-A1AR deleted embryos were dead or re-absorbed. After 72 hours of hypoxia, cardiac size was reduced significantly more in the cardiac-A1AR deleted hearts compared to controls. Gene expression analysis revealed clusters of genes that are regulated by both hypoxia and A1AR expression. CONCLUSIONS: These data identify the embryonic heart as the critical site where adenosine acts to protect the embryo against hypoxia. As such these studies identify a previously unrecognized mechanism of embryo protection.


Subject(s)
Adenosine/metabolism , Fetal Hypoxia/metabolism , Heart/embryology , Animals , Crosses, Genetic , Female , Gene Expression Regulation , Male , Mice , Receptor, Adenosine A1/genetics , Receptor, Adenosine A1/metabolism
19.
FASEB J ; 23(4): 1272-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19088180

ABSTRACT

The purpose of this study was to determine both the short-term effects on cardiac development and embryo growth and the long-term effects on cardiac function and body composition of in utero caffeine exposure. Pregnant mice (C57BL/6) were exposed to hypoxia (10% O(2)) or room air from embryonic days (E) 8.5-10.5, and treated with caffeine (20 mg/kg, i.p.) or vehicle (normal saline, 0.9% NaCl). This caffeine dose results in a circulating level that is equivalent to 2 cups of coffee in humans. Hypoxic exposure acutely reduced embryonic growth by 30%. Exposure to a single dose of caffeine inhibited cardiac ventricular development by 53% in hypoxia and 37% in room air. Caffeine exposure resulted in inhibition of hypoxia-induced HIF1alpha protein expression in embryos by 40%. When offspring from dams treated with a single dose of caffeine were studied in adulthood, we observed that caffeine treatment alone resulted in a decrease in cardiac function of 38%, as assessed by echocardiography. We also observed a 20% increase in body fat with male mice exposed to caffeine. Caffeine was dissolved in normal saline, so it was used as a control. Room air controls were used to compare to the hypoxic mice. Exposure to a single dose of caffeine during embryogenesis results in both short-term effects on cardiac development and long-term effects on cardiac function.


Subject(s)
Caffeine/toxicity , Growth/drug effects , Maternal Exposure , Prenatal Exposure Delayed Effects/metabolism , Time , Animals , Caffeine/metabolism , Embryo, Mammalian/metabolism , Female , Hypoxia/metabolism , Maternal-Fetal Exchange/drug effects , Mice , Mice, Inbred C57BL , Pregnancy
20.
Birth Defects Res A Clin Mol Teratol ; 82(3): 121-30, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18186126

ABSTRACT

BACKGROUND: The current understanding of the effects of hypoxia on early embryogenesis is limited. Potential mediators of hypoxic effects include adenosine, which increases dramatically during hypoxic conditions and activates A(1) adenosine receptors (A(1)ARs). METHODS: To examine the influences of hypoxia and adenosine signaling on cardiac development, chicken embryos were studied. Real time RT-PCR assay was used to examine the A(1)AR gene expression during embryogenesis and after siRNA- mediated knock down. Cell proliferation was determined by counting cell nuclei and PhosphoHistone H3 positive cells. Apoptosis was determined by TUNEL assay. RESULTS: A(1)ARs were found to be expressed in chicken embryos during early embryogenesis. Treatment of Hamburger and Hamilton stage 4 embryos with the A(1)AR agonist N(6)-cyclopentyladenosine caused cardiac bifida and looping defects in 55% of embryos. Hamburger and Hamilton stage 4 embryos exposed to 10% oxygen for 6, 12, 18, and 24 h followed by recovery in room air until stage 11, exhibited cardia bifida and looping defects in 34, 45, 60, and 86% of embryos respectively. Hypoxia-induced abnormalities were reduced when A(1)AR signaling was inhibited by the A(1)AR antagonist 1,3 dipropyl-8-cyclopentylxanthine or by siRNA-targeting A(1)ARs. Hypoxia treatment did not increase apoptosis, but decreased embryonic cell proliferation. CONCLUSIONS: These data indicate that hypoxia adversely influences cardiac malformations during development, in part by A(1)AR signaling.


Subject(s)
Heart Defects, Congenital/etiology , Hypoxia/physiopathology , Receptor, Adenosine A1/physiology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A1 Receptor Agonists , Animals , Base Sequence , Chick Embryo , In Situ Nick-End Labeling , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...