Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Acta Biol Hung ; 69(4): 395-410, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30587022

ABSTRACT

Cancer drug resistance and poor selectivity towards cancer cells demand the constant search for new therapeutics. PI3K-Akt-mTOR and RAS-MAPK-ERK signaling pathways are key mechanisms involved in cell survival, proliferation, differentiation, and metabolism and their deregulation in cancer can promote development of therapy resistance. We investigated the effects of targeted inhibitors (wortmannin, GSK690693, AZD2014 and tipifarnib) towards these two pathways on early zebrafish and sea urchin development to assess their toxicity in normal, fast proliferating cells. PI3K inhibitor wortmannin and RAS inhibitor tipifarnib displayed highest toxicity while GSK690693, a pan-Akt kinase inhibitor, exhibited a less significant impact on embryo survival and development. Moreover, inhibition of the upstream part of the PI3K-Akt-mTOR pathway (wortmannin/GSK690693 co-treatment) produced a synergistic effect and impacted zebrafish embryo survival and development at much lower concentrations. Dual mTORC1/mTORC2 inhibitor AZD2014 showed no considerable effects on embryonic cells of zebrafish in concentrations substantially toxic in cancer cells. AZD2014 also caused the least prominent effects on sea urchin embryo development compared to other inhibitors. Significant toxicity of AZD2014 in human cancer cells, its capacity to sensitize resistant cancers, lower antiproliferative activity against human normal cell lines and fast proliferating embryonic cells could make this agent a promising candidate for anticancer therapy.


Subject(s)
Antineoplastic Agents/toxicity , Cell Proliferation/drug effects , Enzyme Inhibitors/toxicity , Molecular Targeted Therapy/adverse effects , Signal Transduction/drug effects , Abnormalities, Drug-Induced/enzymology , Abnormalities, Drug-Induced/etiology , Abnormalities, Drug-Induced/pathology , Animals , Arbacia/embryology , Benzamides , Dose-Response Relationship, Drug , Embryonic Development/drug effects , Morpholines/toxicity , Oxadiazoles/toxicity , Pyrimidines , Quinolones/toxicity , Wortmannin/toxicity , Zebrafish/embryology
2.
Bull Exp Biol Med ; 163(5): 605-607, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28948551

ABSTRACT

In 60-day-old Wistar rats with fetal valproate syndrome, the brain to body weight ratio was higher by 9.4% and activity of dipeptidyl peptidase IV in the serum and cerebrospinal fluid was higher by 18.4 and 40.6%, respectively, than in healthy controls. Activity of prolylendopeptidase in the serum and cerebrospinal fluid in rats with the fetal valproate syndrome did not differ from the control.


Subject(s)
Abnormalities, Drug-Induced/enzymology , Serine Endopeptidases/metabolism , Valproic Acid/adverse effects , Abnormalities, Drug-Induced/blood , Abnormalities, Drug-Induced/cerebrospinal fluid , Animals , Dipeptidyl Peptidase 4/blood , Dipeptidyl Peptidase 4/cerebrospinal fluid , Male , Prolyl Oligopeptidases , Rats , Rats, Wistar , Serine Endopeptidases/blood , Serine Endopeptidases/cerebrospinal fluid , Valproic Acid/blood , Valproic Acid/cerebrospinal fluid
3.
Birth Defects Res A Clin Mol Teratol ; 106(10): 814-830, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27488927

ABSTRACT

BACKGROUND: Orofacial clefts (OFCs) are common birth defects, which include a range of disorders with a complex etiology affecting formation of craniofacial structures. Some forms of syndromic OFCs are produced by defects in the cholesterol pathway. The principal enzyme of the cholesterol pathway is the 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR). Our aim is to study whether defects of HMGCR function would produce orofacial malformation similar to those found in disorders of cholesterol synthesis. METHODS: We used zebrafish hmgcrb mutants and HMGCR inhibition assay using atorvastatin during early and late stages of orofacial morphogenesis in zebrafish. To describe craniofacial phenotypes, we stained cartilage and bone and performed in situ hybridization using known craniofacial markers. Also, we visualized neural crest cell migration in a transgenic fish. RESULTS: Our results showed that mutants displayed loss of cartilage and diminished orofacial outgrowth, and in some cases palatal cleft. Late treatments with statin show a similar phenotype. Affected-siblings displayed a moderate phenotype, whereas early-treated embryos had a minor cleft. We found reduced expression of the downstream component of Sonic Hedgehog-signaling gli1 in ventral brain, oral ectoderm, and pharyngeal endoderm in mutants and in late atorvastatin-treated embryos. CONCLUSION: Our results suggest that HMGCR loss-of-function primarily affects postmigratory cranial neural crest cells through abnormal Sonic Hedgehog signaling, probably induced by reduction in metabolites of the cholesterol pathway. Malformation severity correlates with the grade of HMGCR inhibition, developmental stage of its disruption, and probably with availability of maternal lipids. Together, our results might help to understand the spectrum of orofacial phenotypes found in cholesterol synthesis disorders. Birth Defects Research (Part A) 106:814-830, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Abnormalities, Drug-Induced , Atorvastatin/adverse effects , Cleft Lip , Cleft Palate , Hydroxymethylglutaryl CoA Reductases , Mutation , Zebrafish Proteins , Zebrafish , Abnormalities, Drug-Induced/enzymology , Abnormalities, Drug-Induced/genetics , Animals , Atorvastatin/pharmacology , Cleft Lip/chemically induced , Cleft Lip/enzymology , Cleft Lip/genetics , Cleft Lip/pathology , Cleft Palate/chemically induced , Cleft Palate/enzymology , Cleft Palate/genetics , Cleft Palate/pathology , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
4.
Pharmacogenomics J ; 16(5): 411-29, 2016 10.
Article in English | MEDLINE | ID: mdl-27401223

ABSTRACT

Mendelian diseases contain important biological information regarding developmental effects of gene mutations that can guide drug discovery and toxicity efforts. In this review, we focus on Smith-Lemli-Opitz syndrome (SLOS), a rare Mendelian disease characterized by compound heterozygous mutations in 7-dehydrocholesterol reductase (DHCR7) resulting in severe fetal deformities. We present a compilation of SLOS-inducing DHCR7 mutations and the geographic distribution of those mutations in healthy and diseased populations. We observed that several mutations thought to be disease causing occur in healthy populations, indicating an incomplete understanding of the condition and highlighting new research opportunities. We describe the functional environment around DHCR7, including pharmacological DHCR7 inhibitors and cholesterol and vitamin D synthesis. Using PubMed, we investigated the fetal outcomes following prenatal exposure to DHCR7 modulators. First-trimester exposure to DHCR7 inhibitors resulted in outcomes similar to those of known teratogens (50 vs 48% born-healthy). DHCR7 activity should be considered during drug development and prenatal toxicity assessment.


Subject(s)
Abnormalities, Drug-Induced/genetics , Enzyme Inhibitors/adverse effects , Maternal Exposure/adverse effects , Mutation , Oxidoreductases Acting on CH-CH Group Donors/genetics , Pharmacogenetics , Smith-Lemli-Opitz Syndrome/genetics , Abnormalities, Drug-Induced/enzymology , Abnormalities, Drug-Induced/epidemiology , Animals , Cholesterol/metabolism , Evolution, Molecular , Female , Gene Frequency , Genetic Drift , Genetic Predisposition to Disease , Heredity , Humans , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Phenotype , Pregnancy , Risk Assessment , Risk Factors , Smith-Lemli-Opitz Syndrome/drug therapy , Smith-Lemli-Opitz Syndrome/enzymology , Smith-Lemli-Opitz Syndrome/epidemiology , Vitamin D/metabolism
5.
Article in English | MEDLINE | ID: mdl-26183885

ABSTRACT

We aimed to investigate the impact of the epigenome in inducting fetal alcohol spectrum disorder (FASD) phenotypes in Japanese rice fish embryogenesis. One of the significant events in epigenome is DNA methylation which is catalyzed by DNA methyltransferase (DNMT) enzymes. We analyzed DNMT enzyme mRNA expressions in Japanese rice fish development starting from fertilized eggs to hatching and also in embryos exposed for first 48h of development either to ethanol (300mM) or to 5-azacytidine (5-azaC; 2mM), an inhibitor of DNMT enzyme activity. As observed in FASD phenotypes, 5-azaC exposure was able to induce microcephaly and craniofacial cartilage deformities in Japanese rice fish. Moreover, we have observed that expression of DNMTs (dnmt1, dnmt3aa, and dnmt3bb.1) are developmentally regulated; high mRNA copies were found in early stages (1-2day-post-fertilization, dpf), followed by gradual reduction until hatched. In ethanol-treated embryos, compared to controls, dnmt1 mRNA is in reduced level in 2dpf and in enhanced level in 6dpf embryos. While dnmt3aa and 3bb.1 remained unaltered. In contrast, embryos exposed to 5-azaC have an enhanced level of dnmt1 and dnmt3bb.1 mRNAs both in 2 and 6dpf embryos while dnmt3aa is enhanced only in 6dpf embryos. Moreover, endocannabinoid receptor 1a (cnr1a) mRNA which was found to be reduced by ethanol remained unaltered and cnr1b and cnr2 mRNAs, which were remained unaltered by ethanol, were increased significantly by 5-azaC in 6dpf embryos. This study indicates that the craniofacial defects observed in FASD phenotypes are the results of dysregulations in DNMT expressions.


Subject(s)
Abnormalities, Drug-Induced/etiology , Azacitidine/toxicity , DNA (Cytosine-5-)-Methyltransferases/metabolism , Epigenesis, Genetic/drug effects , Ethanol/toxicity , Fetal Alcohol Spectrum Disorders/etiology , Fish Proteins/metabolism , Oryzias/metabolism , Abnormalities, Drug-Induced/enzymology , Abnormalities, Drug-Induced/genetics , Animals , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , DNA Methyltransferase 3A , Disease Models, Animal , Fetal Alcohol Spectrum Disorders/enzymology , Fetal Alcohol Spectrum Disorders/genetics , Fish Proteins/genetics , Gene Expression Regulation, Enzymologic , Oryzias/embryology , Oryzias/genetics , RNA, Messenger/metabolism , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/drug effects , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism , Time Factors , DNA Methyltransferase 3B
6.
Toxicol Appl Pharmacol ; 287(3): 232-9, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26074427

ABSTRACT

Reactive oxygen species (ROS) have been implicated in the mechanism of ethanol (EtOH) teratogenicity, but the protective role of the embryonic antioxidative enzyme catalase is unclear, as embryonic activity is only about 5% of maternal levels. We addressed this question in a whole embryo culture model. C57BL/6 mouse embryos expressing human catalase (hCat) or their wild-type (C57BL/6 WT) controls, and C3Ga.Cg-Cat(b)/J catalase-deficient, acatalasemic (aCat) mouse embryos or their wild-type C3HeB/FeJ (C3H WT) controls, were explanted on gestational day (GD) 9 (plug=GD 1), exposed for 24h to 2 or 4mg/mL EtOH or vehicle, and evaluated for functional and morphological changes. hCat and C57BL/6 WT vehicle-exposed embryos developed normally, while EtOH was embryopathic in C57BL/6 WT embryos, evidenced by decreases in anterior neuropore closure, somites developed, turning and head length, whereas hCat embryos were protected (p<0.001). Maternal pretreatment of C57BL/6 WT dams with 50kU/kg PEG-catalase (PEG-cat) 8h prior to embryo culture, which increases embryonic catalase activity, blocked all EtOH embryopathies (p<0.001). Vehicle-exposed aCat mouse embryos had lower yolk sac diameters compared to WT controls, suggesting that endogenous ROS are embryopathic. EtOH was more embryopathic in aCat embryos than WT controls, evidenced by reduced head length and somite development (p<0.01), and trends for reduced anterior neuropore closure, turning and crown-rump length. Maternal pretreatment of aCat dams with PEG-Cat blocked all EtOH embryopathies (p<0.05). These data suggest that embryonic catalase is a determinant of risk for EtOH embryopathies.


Subject(s)
Abnormalities, Drug-Induced/prevention & control , Acatalasia/enzymology , Catalase/metabolism , Embryo, Mammalian/drug effects , Ethanol/toxicity , Abnormalities, Drug-Induced/embryology , Abnormalities, Drug-Induced/enzymology , Abnormalities, Drug-Induced/genetics , Acatalasia/embryology , Acatalasia/genetics , Animals , Catalase/genetics , Catalase/pharmacology , DNA Damage , Disease Models, Animal , Dose-Response Relationship, Drug , Embryo Culture Techniques , Embryo, Mammalian/enzymology , Embryo, Mammalian/pathology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Gestational Age , Humans , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oxidative Stress , Polyethylene Glycols/pharmacology , Reactive Oxygen Species/metabolism
7.
Toxicology ; 328: 142-51, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25527867

ABSTRACT

Exposure to environmental toxic chemicals in utero during the neural tube development period can cause developmental disorders. To evaluate the disruption of neural tube development programming, the murine neural tube defects (NTDs) model was induced by interrupting folate metabolism using methotrexate in our previous study. The present study aimed to examine the effects of dNTP deficiency induced by hydroxyurea (HU), a specific ribonucleotide reductase (RNR) inhibitor, during murine neural tube development. Pregnant C57BL/6J mice were intraperitoneally injected with various doses of HU on gestation day (GD) 7.5, and the embryos were checked on GD 11.5. RNR activity and deoxynucleoside triphosphate (dNTP) levels were measured in the optimal dose. Additionally, DNA damage was examined by comet analysis and terminal deoxynucleotidyl transferase mediated dUTP nick end-labeling (TUNEL) assay. Cellular behaviors in NTDs embryos were evaluated with phosphorylation of histone H3 (PH-3) and caspase-3 using immunohistochemistry and western blot analysis. The results showed that NTDs were observed mostly with HU treatment at an optimal dose of 225 mg/kg b/w. RNR activity was inhibited and dNTP levels were decreased in HU-treated embryos with NTDs. Additionally, increased DNA damage, decreased proliferation, and increased caspase-3 were significant in NTDs embryos compared to the controls. Results indicated that HU induced murine NTDs model by disturbing dNTP metabolism and further led to the abnormal cell balance between proliferation and apoptosis.


Subject(s)
Abnormalities, Drug-Induced/etiology , Deoxyribonucleotides/metabolism , Enzyme Inhibitors/toxicity , Hydroxyurea/toxicity , Maternal Exposure/adverse effects , Neural Tube Defects/chemically induced , Neural Tube/drug effects , Ribonucleotide Reductases/antagonists & inhibitors , Abnormalities, Drug-Induced/embryology , Abnormalities, Drug-Induced/enzymology , Animals , Apoptosis/drug effects , Brain/abnormalities , Brain/drug effects , Brain/enzymology , Caspase 3/metabolism , Cell Proliferation/drug effects , DNA Damage , Down-Regulation , Female , Gestational Age , Histones/metabolism , Mice, Inbred C57BL , Neural Tube/abnormalities , Neural Tube/enzymology , Neural Tube Defects/embryology , Neural Tube Defects/enzymology , Phosphorylation , Pregnancy , Ribonucleotide Reductases/metabolism , Time Factors
8.
Environ Sci Technol ; 47(18): 10556-66, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-24003986

ABSTRACT

Atlantic killifish (Fundulus heteroclitus) inhabiting the Atlantic Wood Industries Superfund Site (Elizabeth River, Portsmouth, VA, USA) are resistant to the acute toxicity and cardiac teratogenesis caused by high levels of polycyclic aromatic hydrocarbons (PAHs) from creosote. The resistance is linked to down regulation of the aryl hydrocarbon receptor (AHR) pathway. We investigated the association between CYP1 activity, as a marker of potential AHR pathway suppression, and contaminant resistance in killifish subpopulations from sites throughout the estuary that varied significantly in PAH contamination level. Adult killifish and sediments were collected from seven sites across approximately 13.7 km in river length within the estuary and from a nearby reference site. Sediment PAH levels were determined using gas chromatography mass spectrometry. Embryos obtained via manual spawning were exposed to individual AHR agonists and PAH mixtures 24 h post fertilization (hpf); CYP1 activity was determined by in ovo ethoxyresorufin-o-deethylase (EROD) at 96 hpf, and cardiac deformity severity was scored at 144 hpf. The total PAH levels measured among the sites varied from approximately 200 to 125,000 ng/g dry sediment. Overall, the resistance to teratogenesis was strongest in the subpopulations from sites in or closest to the major PAH contamination sites, but even embryos from less-contaminated sites within the Elizabeth River demonstrated at least partial resistance to many challenges. Surprisingly, all of the subpopulations tested were highly resistant to PCB-126 (3,3',4,4',5-pentachlorobiphenyl). However, the degree of CYP1 activity response varied significantly among subpopulations and did not always correlate strongly with resistance to teratogenesis; some subpopulations resisted the cardiac teratogenesis caused by the challenges at doses that still elicited strong EROD induction. Our results suggest that there is variation in the adaptive phenotype exhibited by laboratory-spawned embryos from killifish subpopulations throughout the estuary. Furthermore, the results show that contaminants have affected killifish subpopulations throughout the estuary, even in sites with lower levels of PAHs.


Subject(s)
Cytochrome P-450 CYP1A1/metabolism , Fundulidae/physiology , Polychlorinated Biphenyls/toxicity , Polycyclic Aromatic Hydrocarbons/toxicity , Teratogens/toxicity , Water Pollutants, Chemical/toxicity , Abnormalities, Drug-Induced/enzymology , Adaptation, Physiological , Animals , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/enzymology , Embryonic Development/drug effects , Fundulidae/abnormalities , Geologic Sediments/analysis , Heart Defects, Congenital/chemically induced , Heart Defects, Congenital/enzymology , Polycyclic Aromatic Hydrocarbons/analysis , Receptors, Aryl Hydrocarbon/metabolism , Teratogens/analysis , Virginia , Water Pollutants, Chemical/analysis
9.
J Appl Toxicol ; 33(1): 32-40, 2013 Jan.
Article in English | MEDLINE | ID: mdl-21751222

ABSTRACT

SP600125 (anthrapyrazolone) is a synthetic polyaromatic chemical that inhibits c-Jun N-terminal kinase (JNK) signaling by interfering with phosphorylation of c-Jun. To determine the pharmacological impact of SP600125 on zebrafish development, we incubated embryos in various concentrations of SP600125 from 18 h postfertilization (hpf) to 48 hpf. Embryos treated with 1.25 µm appeared with occasional pericardium edema. Treatment with 12.5 µm resulted in complete mortality by 120 hpf, preventing an assessment of physiological defects. Embryos treated with 5 µm exhibited slowed overall growth, a delay in hatching and numerous morphological defects such as pericardium edema, yolk sac edema, swim bladder deflation, bent vertebrae and eye and jaw malformations. Whole-mount immunohistochemical studies using an anti-acetylated ß-tubulin antibody confirmed developmental defects in the nervous system. Within the retina, fish treated with 1.25 µm showed a mild reduction of immunoreactivity. Immunoreactivity in the retina was further reduced in fish treated with 5 µm of SP600125. In these fish, eyes and olfactory organs were half the size compared with other groups. Multiple lenses were observed in 67% of these fish. A second experiment with a shorter exposure period of SP600125 (6 h) presented significantly fewer morphological defects. The treatment led to a delay in hatching, and increased incidences of swim bladder deflation and pericardium edema with increasing concentrations. In summary, SP600125 caused developmental abnormalities during zebrafish organogenesis starting at 1.25 µm and the defects were exacerbated with increasing concentrations. Our study suggests that SP600125 at 1.25 µm and beyond has devastating consequences for zebrafish development.


Subject(s)
Abnormalities, Drug-Induced/etiology , Anthracenes/toxicity , Embryo, Nonmammalian/drug effects , Enzyme Inhibitors/toxicity , Nervous System Malformations/chemically induced , Abnormalities, Drug-Induced/enzymology , Abnormalities, Drug-Induced/pathology , Air Sacs/abnormalities , Air Sacs/drug effects , Air Sacs/enzymology , Animals , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/enzymology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Longevity/drug effects , Nervous System Malformations/enzymology , Nervous System Malformations/pathology , Olfactory Bulb/abnormalities , Olfactory Bulb/drug effects , Olfactory Bulb/enzymology , Pericardial Effusion/chemically induced , Pericardial Effusion/enzymology , Pericardial Effusion/pathology , Retina/abnormalities , Retina/drug effects , Retina/enzymology , Time Factors , Zebrafish
10.
Reprod Toxicol ; 36: 33-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23207165

ABSTRACT

The role of catalase in methanol (MeOH) teratogenesis is unclear. In rodents it both detoxifies reactive oxygen species (ROS) and metabolizes MeOH and its formic acid (FA) metabolite. We treated pregnant mice expressing either high (hCat) or low catalase activity (aCat), or their wild-type (WT) controls, with either MeOH (4g/kg ip) or saline. hCat mice and WTs were similarly susceptible to MeOH-initiated ophthalmic abnormalities and cleft palates. aCat and WT mice appeared resistant, precluding assessment of the developmental impact of catalase deficiency. Catalase activity was respectively increased at least 1.5-fold, and decreased by at least 35%, in hCat and aCat embryos and maternal livers. MeOH and FA pharmacokinetic profiles were similar among hCat, aCat and WT strains. Although the hCat results imply no ROS involvement, embryo culture studies suggest this may be confounded by maternal factors and/or a requirement for higher catalase activity in the hCat mice.


Subject(s)
Abnormalities, Multiple/chemically induced , Catalase/metabolism , Embryo, Mammalian/drug effects , Maternal Exposure/adverse effects , Methanol/toxicity , Solvents/toxicity , Teratogens/toxicity , Abnormalities, Drug-Induced/embryology , Abnormalities, Drug-Induced/enzymology , Abnormalities, Drug-Induced/metabolism , Abnormalities, Multiple/embryology , Abnormalities, Multiple/enzymology , Abnormalities, Multiple/metabolism , Acatalasia/enzymology , Acatalasia/metabolism , Animals , Catalase/genetics , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Female , Humans , Liver/drug effects , Liver/enzymology , Liver/metabolism , Male , Methanol/blood , Methanol/pharmacokinetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Pregnancy , Solvents/analysis , Solvents/pharmacokinetics , Teratogens/analysis , Teratogens/pharmacokinetics
11.
FASEB J ; 25(7): 2188-200, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21478259

ABSTRACT

Oxidative stress and reactive oxygen species (ROS) such as hydrogen peroxide (H(2)O(2)), which is detoxified by catalase, are implicated in fetal death and birth defects. However, embryonic levels of catalase are only ∼ 5% of adult activity, and its protective role is not understood completely. Herein, we used mutant catalase-deficient mice [acatalasemic (aCat)] and transgenic mice expressing human catalase (hCat), which, respectively, exhibited 40-50% reductions and 2-fold elevations in the activities of embryonic and fetal brain catalase, to show that embryonic catalase protects the embryo from both physiological oxidative stress and the ROS-initiating antiepileptic drug phenytoin. Compared to wild-type (WT) catalase-normal controls, both untreated and phenytoin-exposed aCat mice exhibited a 30% increase in embryonic DNA oxidation and a >2-fold increase in embryopathies, both of which were completely blocked by protein therapy with exogenous catalase. Conversely, compared to WT controls, untreated and, to a lesser extent, phenytoin-exposed hCat mice were protected, with untreated hCat embryos exhibiting a 40% decrease in embryonic DNA oxidation and up to a 67% decrease in embryopathies. Embryonic catalase accordingly plays an important protective role, and both physiological and phenytoin-enhanced oxidative stress can be embryopathic.


Subject(s)
Catalase/metabolism , DNA/metabolism , Embryo, Mammalian/enzymology , Phenytoin/toxicity , Abnormalities, Drug-Induced/enzymology , Abnormalities, Drug-Induced/genetics , Abnormalities, Drug-Induced/mortality , Acatalasia/enzymology , Acatalasia/genetics , Acatalasia/mortality , Animals , Anticonvulsants/metabolism , Anticonvulsants/toxicity , Brain/abnormalities , Brain/drug effects , Brain/enzymology , Catalase/genetics , Dose-Response Relationship, Drug , Embryo, Mammalian/abnormalities , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oxidation-Reduction/drug effects , Phenytoin/metabolism , Pregnancy , Survival Rate
12.
J Biol Chem ; 286(23): 20970-6, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21518755

ABSTRACT

Membrane PTK7 pseudo-kinase plays an essential role in planar cell polarity and the non-canonical Wnt pathway in vertebrates. Recently, a new N-ethyl-N-nitrosourea-induced mutant named chuzhoi (chz) was isolated in mice. chz embryos have severe birth defects, including a defective neural tube, defective heart and lung development, and a shortened anterior-posterior body axis. The chz mutation was mapped to the Ala-Asn-Pro tripeptide insertion into the junction region between the fifth and the sixth Ig-like domains of PTK7. Unexpectedly, chz reduced membrane localization of the PTK7 protein. We hypothesized and then proved that the chz mutation caused an insertion of an additional membrane type 1 matrix metalloproteinase cleavage site in PTK7 and that the resulting aberrant proteolysis of chz affected the migratory parameters of the cells. It is likely that aberrations in the membrane type 1 matrix metalloproteinase/PTK7 axis are detrimental to cell movements that shape the body plan and that chz represents a novel model system for increasing our understanding of the role of proteolysis in developmental pathologies, including congenital defects.


Subject(s)
Abnormalities, Drug-Induced/enzymology , Cell Adhesion Molecules/metabolism , Matrix Metalloproteinase 14/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Abnormalities, Drug-Induced/genetics , Alkylating Agents/adverse effects , Alkylating Agents/pharmacology , Animals , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Shape/drug effects , Cell Shape/genetics , Ethylnitrosourea/adverse effects , Ethylnitrosourea/pharmacology , Humans , Matrix Metalloproteinase 14/genetics , Mice , Mutation , Protein Structure, Tertiary , Receptor Protein-Tyrosine Kinases/genetics
13.
Toxicol Sci ; 120(2): 428-38, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21252394

ABSTRACT

Reactive oxygen species (ROS) are implicated in spontaneous and xenobiotic-enhanced embryopathies, and protein therapy with exogenous catalase suggests an embryoprotective role, although embryonic catalase activity is only about 5% of adult activity. Using mutant catalase-deficient (acatalasemic, aCat) mice and transgenic mice expressing human catalase (hCat, enhanced catalase activity) compared with a confirmed outbred CD-1 mouse model, we investigated the protective importance of constitutive embryonic catalase against endogenous ROS and the ROS-initiating teratogen phenytoin in embryo culture. Vehicle-exposed aCat and hCat embryos, respectively, exhibited reduced and enhanced catalase activity compared with wild-type (WT) controls, with conversely enhanced and reduced spontaneous embryopathies. Phenytoin was embryopathic in all strains without altering catalase activity but less so in the WT embryos for the aCat and hCat strains, which exhibited about half the catalase activity of CD-1 embryos. Phenytoin, respectively, enhanced and reduced embryopathies in aCat and hCat embryos. Among aCat embryos exposed to phenytoin, embryopathies increased with decreasing catalase activity and were completely blocked by addition of exogenous catalase, which increased embryonic catalase activity to WT levels. These results provide the first direct evidence that (1) the low level of constitutive embryonic catalase protects the conceptus from developmental and xenobiotic-enhanced oxidative stress and (2) embryonic variations in activity of this enzyme affect development.


Subject(s)
Catalase/physiology , Embryo, Mammalian/drug effects , Embryonic Development/drug effects , Oxidative Stress/drug effects , Phenytoin/toxicity , Abnormalities, Drug-Induced/enzymology , Abnormalities, Drug-Induced/metabolism , Animals , Catalase/genetics , Catalase/metabolism , Embryo Culture Techniques , Embryo, Mammalian/enzymology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pregnancy , Reactive Oxygen Species/metabolism
14.
Toxicol Sci ; 118(2): 686-95, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20889679

ABSTRACT

Exposure during the organogenesis stage of the mouse embryo to the model teratogen, hydroxyurea (HU), induces curly tail and limb malformations. Oxidative stress contributes to the developmental toxicity of HU. Reactive oxygen species (ROS) interact with polyunsaturated bilipid membranes to form α,ß-unsaturated reactive aldehydes; 4-hydroxy-2-nonenal (4-HNE), one of the most cytotoxic of these aldehydes, covalently adducts with proteins, lipids, and nucleic acids. The goal of the current study is to determine if HU exposure of CD1 mice on gestation day 9 generates region-specific 4-HNE-protein adducts in the embryo and to identify the proteins targeted. The formation of 4-HNE-protein adducts was elevated in the caudal region of control embryos; HU exposure further increased 4-HNE-protein adduct formation in this area. Interestingly, three of the 4-HNE-modified proteins, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), glutamate oxaloacetate transaminase 2, and aldolase 1, A isoform, are involved in energy metabolism. The formation of 4-HNE-GAPDH protein adducts reduced GAPDH enzymatic activity by 20% and attenuated lactate production by 40%. Furthermore, HU exposure induced the nuclear translocation of GAPDH in the caudal region of exposed embryos; this nuclear translocation may be associated with the reactivation of oxidized proteins involved in DNA repair, such as apurinic/apyrimidinic endonuclease-1, and the stimulation of E1A-associated P300 protein/creb-binding protein (p300/CBP) activity, initiating cell death in a p53-dependent pathway. We propose that GAPDH is a redox-sensitive target in the embryo and may play a role in a stress response during development.


Subject(s)
Embryo, Mammalian/drug effects , Embryonic Development/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Hydroxyurea/toxicity , Organogenesis/drug effects , Oxidative Stress/drug effects , Teratogens/toxicity , Abnormalities, Drug-Induced/enzymology , Abnormalities, Drug-Induced/etiology , Aldehydes/metabolism , Animals , Aspartate Aminotransferase, Mitochondrial/metabolism , Cell Nucleus/drug effects , Cell Nucleus/genetics , Embryo Culture Techniques , Embryo, Mammalian/embryology , Embryo, Mammalian/enzymology , Fructose-Bisphosphate Aldolase/metabolism , Mice , Organogenesis/physiology , Translocation, Genetic
15.
Toxicol Appl Pharmacol ; 244(2): 156-61, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20045017

ABSTRACT

Dithiocarbamates (DTCs) are a class of compounds that are extensively used in agriculture as pesticides. As such, humans and wildlife are undoubtedly exposed to these chemicals. Although DTCs are thought to be relatively safe due to their short half lives, it is well established that they are teratogenic to vertebrates, especially to fish. In zebrafish, these teratogenic effects are characterized by distorted notochord development and shortened anterior to posterior axis. DTCs are known copper (Cu) chelators but this does not fully explain the observed teratogenic effects. We show here that DTCs cause malformations in zebrafish that highly resemble teratogenic effects observed by direct inhibition of a group of cuproenzymes termed lysyl oxidases (LOX). Additionally, we demonstrate that partial knockdown of three LOX genes, lox, loxl1 and loxl5b, sensitizes the developing embryo to DTC exposure. Finally, we show that DTCs directly inhibit zebrafish LOX activity in an ex vivo amine oxidase assay. Taken together, these results provide the first evidence that DTC induced teratogenic effects are, at least in part, caused by direct inhibition of LOX activity.


Subject(s)
Abnormalities, Drug-Induced/enzymology , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Protein-Lysine 6-Oxidase/metabolism , Teratogens/toxicity , Thiocarbamates/toxicity , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/metabolism , Abnormalities, Drug-Induced/embryology , Animals , Enzyme Inhibitors/toxicity , Gene Knockdown Techniques/methods , Protein-Lysine 6-Oxidase/genetics , Zebrafish , Zebrafish Proteins/genetics
16.
Birth Defects Res A Clin Mol Teratol ; 85(9): 800-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19492401

ABSTRACT

BACKGROUND: Oxidative stress is hypothesized to mediate embryotoxicity during organogenesis, yet the reactive oxygen species involved are not defined. The superoxide oxygen radical is converted to hydrogen peroxide, a less reactive species, by superoxide dismutases (SODs). If superoxide is important in mediating embryotoxicity, increased SOD expression should protect embryos against insult. Exposure to hydroxyurea during organogenesis causes brain defects, cleft palate, tail anomalies, and limb defects; administration of D-mannitol, a free radical scavenger, ameliorates hydroxyurea embryotoxicity, suggesting that oxidative stress is important. To elucidate the role of superoxide in mediating hydroxyurea embryotoxicity, we assessed the impact of human SOD1 expression in a murine model. METHODS: hSOD1 hemizygous male mice, carrying the human SOD1 gene, were mated to wild-type or hSOD1 hemizygous females. Dams were treated on gestation day (GD) 9 with saline (control) or 400 (low) or 600 (high) mg/kg hydroxyurea (n = 8-13/group). Mice were euthanized on GD 18 and developmental toxicity was assessed. RESULTS: Exposure to hydroxyurea caused a dose-dependent increase in fetal deaths that was not affected by hSOD1 expression; hydroxyurea decreased fetal weights in litters from wild-type but not hemizygous dams. Hydroxyurea increased the incidence of external and skeletal malformations; fetuses from hemizygous dams treated with high-dose hydroxyurea had fewer malformations compared to wild-type dams. There was no correlation between embryonic phenotype and genotype or SOD activity. CONCLUSION: Maternal hSOD1 expression protected fetuses against malformations induced by hydroxyurea, providing evidence that superoxide plays a role in mediating the response of organogenesis stage embryos to this teratogen.


Subject(s)
Abnormalities, Drug-Induced/etiology , Enzyme Inhibitors/toxicity , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Hydroxyurea/toxicity , Superoxide Dismutase/genetics , Abnormalities, Drug-Induced/enzymology , Abnormalities, Drug-Induced/pathology , Animals , Bone and Bones/abnormalities , Bone and Bones/drug effects , Disease Models, Animal , Female , Fetal Death/chemically induced , Fetal Weight/drug effects , Humans , Male , Mice , Mice, Transgenic , Oxidative Stress/drug effects , Pregnancy , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
17.
Article in English | MEDLINE | ID: mdl-19194987

ABSTRACT

Maternal diabetes causes neural tube defects in embryos, which are associated with increased apoptosis in the neuroepithelium. Many factors, including effector caspases, have been shown to be involved in the events. However, the key regulators have not been identified and the underlying mechanisms remain to be addressed. Caspase-8, an initiator caspase, has been shown to be altered in diabetic embryopathy, suggesting a role as an upstream apoptotic regulator. Using mouse embryos as a model system, this study demonstrates that caspase-8 is required for the production of hyperglycemia-associated embryonic malformations. Caspase-8 was shown to be expressed in the developing neural tube. Its activity, as evidenced by enhanced cleavage, was increased by hyperglycemia. These changes were associated with increased formation of the active cleavage of Bid. Inhibition of caspase-8 activity in high glucose-challenged embryos reduced the rate of embryonic malformation and this was associated with decreased apoptosis in the neuroepithelium of the neural tube. Inhibition of caspase-8 activity also reduced hyperglycemia-induced Bid activation and caspase-9 cleavage. These data suggest that caspase-8 may control diabetic embryopathy-associated apoptosis via regulation of the Bid-stimulated mitochondrion/caspase-9 pathway.


Subject(s)
Abnormalities, Drug-Induced/enzymology , Caspase 8/metabolism , Diabetes Mellitus, Experimental/enzymology , Neural Tube Defects/enzymology , Neural Tube/enzymology , Pregnancy in Diabetics/enzymology , Abnormalities, Drug-Induced/etiology , Animals , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Fluorescent Antibody Technique, Indirect , Glucose/pharmacology , Image Processing, Computer-Assisted , Mice , Neural Tube/abnormalities , Neural Tube Defects/etiology , Pregnancy , Pregnancy in Diabetics/etiology
18.
J Pharmacol Exp Ther ; 326(3): 764-72, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18577701

ABSTRACT

Mitogen-activated protein kinase (MAPK) signaling plays an important role during embryo development. We hypothesize that MAPK activation is a determinant of the fate of organogenesis-stage embryos exposed to insult. To test this hypothesis, CD1 mice were exposed to a model teratogen, hydroxyurea, on gestational day 9. Hydroxyurea exposure triggered a dramatic, transient increase in the activation of p38 MAPKs and c-Jun N-terminal kinases (JNKs) in embryos, without activating extracellular signal-regulated kinases 1 and 2. Selectively blocking p38 MAPKs with 4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imidazole (SB203580) enhanced hydroxyurea-induced fetal mortality without affecting growth retardation or the incidence of deformities among surviving fetuses. In contrast, selectively blocking JNKs with JNK peptide inhibitor 1, L-stereoisomer did not affect hydroxyurea-induced fetal death but increased the incidence of the hindlimb defects observed. Thus, p38 MAPKs and JNKs play distinct roles in protecting the conceptus against insult. Pharmacological inhibition of teratogen exposure induced MAPK activation has adverse consequences on the embryo.


Subject(s)
Abnormalities, Drug-Induced/embryology , Abnormalities, Drug-Induced/enzymology , JNK Mitogen-Activated Protein Kinases/physiology , MAP Kinase Signaling System/physiology , Organogenesis/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Embryo, Mammalian/drug effects , Embryo, Mammalian/embryology , Embryo, Mammalian/enzymology , Female , Imidazoles/pharmacology , Imidazoles/therapeutic use , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/physiology , Organogenesis/drug effects , Pregnancy , Pyridines/pharmacology , Pyridines/therapeutic use , Teratogens , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
19.
Toxicol Sci ; 104(2): 397-404, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18483001

ABSTRACT

In spite of the large use of salicylates, introduced into clinical practice more than 100 years ago, their anti-inflammatory and cancer preventive mechanisms are still under study. Teratogenic effects of salicylates have been reported in experimental animals since 1959 but the pathogenic pathways and the mechanisms of action were never described until now. The aim of this work is to verify if the inhibition of embryonic histone deacetylase (HDAC) enzymes and the consequent tissue hyperacetylation could be the mechanism responsible for axial skeletal defects described after the exposure of pregnant rodents to sodium salicylate (SAL). E8 pregnant CD-1 mice were intraperitoneally treated with SAL 0-150-300-450 mg/kg and sacrificed at 1, 3, 5 h after treatment or at term of gestation (E18). E8 embryos were processed for Western blotting and immunostaining analyses, while skeletons of E18 fetuses were double stained for bone and cartilage. A group of control E8 embryos were used to prepare embryonic nuclear extract for the HDAC enzyme assay. A significant SAL dose-related HDAC inhibition activity, compatible with a mixed-type partial inhibition mechanism, was detected. A clear dose-related hyperacetylation of histones was observed in embryos exposed in utero to SAL, with a peak at 3 h after treatment of dams. The most hyperacetylated organs were somites and the heart. Histone hyperacetylation is suggested to be the mechanism accounting for SAL-related axial skeletal and cardiovascular defects and is proposed as the mechanism responsible for other biological effects of salicylates.


Subject(s)
Abnormalities, Drug-Induced/enzymology , Bone and Bones/drug effects , Cyclooxygenase Inhibitors/toxicity , Histone Acetyltransferases/antagonists & inhibitors , Sodium Salicylate/toxicity , Teratogens/toxicity , Abnormalities, Drug-Induced/etiology , Abnormalities, Drug-Induced/pathology , Acetylation , Animals , Bone and Bones/abnormalities , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Fluorescent Antibody Technique, Indirect , Histones/chemistry , Histones/metabolism , Injections, Intraperitoneal , Mice , Mice, Inbred Strains , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...