Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 257
Filtrar
1.
Alcohol Clin Exp Res ; 45(10): 1965-1979, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34581462

RESUMEN

BACKGROUND: Alcohol exposure during the gastrulation stage of development causes the craniofacial and brain malformations that define fetal alcohol syndrome. These malformations, such as a deficient philtrum, are exemplified by a loss of midline tissue and correspond, at least in part, to regionally selective cell death in the embryo. The tumor suppressor protein Tp53 is an important mechanism for cell death, but the role of Tp53 in the consequences of alcohol exposure during the gastrulation stage has yet to be examined. The current studies used mice and zebrafish to test whether genetic loss of Tp53 is a conserved mechanism to protect against the effects of early developmental stage alcohol exposure. METHODS: Female mice, heterozygous for a mutation in the Tp53 gene, were mated with Tp53 heterozygous males, and the resulting embryos were exposed during gastrulation on gestational day 7 (GD 7) to alcohol (two maternal injections of 2.9 g/kg, i.p., 4 h apart) or a vehicle control. Zebrafish mutants or heterozygotes for the tp53zdf1  M214K mutation and their wild-type controls were exposed to alcohol (1.5% or 2%) beginning 6 h postfertilization (hpf), the onset of gastrulation. RESULTS: Examination of GD 17 mice revealed that eye defects were the most common phenotype among alcohol-exposed fetuses, occurring in nearly 75% of the alcohol-exposed wild-type fetuses. Tp53 gene deletion reduced the incidence of eye defects in both the heterozygous and mutant fetuses (to about 35% and 20% of fetuses, respectively) and completely protected against alcohol-induced facial malformations. Zebrafish (4 days postfertilization) also demonstrated alcohol-induced reductions of eye size and trabeculae length that were less common and less severe in tp53 mutants, indicating a protective effect of tp53 deletion. CONCLUSIONS: These results identify an evolutionarily conserved role of Tp53 as a pathogenic mechanism for alcohol-induced teratogenesis.


Asunto(s)
Anomalías Inducidas por Medicamentos/etiología , Anomalías Craneofaciales/etiología , Etanol/efectos adversos , Trastornos del Espectro Alcohólico Fetal/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Anomalías Inducidas por Medicamentos/metabolismo , Animales , Anomalías Craneofaciales/metabolismo , Femenino , Masculino , Ratones , Embarazo , Teratogénesis , Pez Cebra
2.
Pediatr Dev Pathol ; 24(5): 438-444, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34082612

RESUMEN

INTRODUCTION: Angiotensin-Converting Enzyme Inhibitors and Angiotensin II Receptor Blockers (AAs) are used for several indications, with cessation recommended in pregnancy due to toxic effects. AA fetopathy phenotype is similar to renal tubular dysgenesis including reduced proximal convoluted tubules (PCTs). Our study aimed to quantify the reduction of PCTs in fetuses and infants with prenatal exposure to AAs. MATERIALS AND METHODS: We identified 5 fetal AA exposure cases that underwent autopsy at our institution between 2011 and 2018 and compared with 5 gestational age-matched controls. Immunohistochemistry with CD10 and epithelial membrane antigen (EMA) was utilized. RESULTS: CD10 and EMA identified a median PCT density of 19.0% ± 12.3% in AA fetopathy patients, significantly less than controls (52.8% ± 4.4%; p < 0.0001). One case with in utero cessation had a PCT density of 34.2% ± 0.2%. Among other AA fetopathy findings, 1 case demonstrated unilateral renal vein thrombosis and 4 had hypocalvaria. CONCLUSIONS: We have quantified the reduction in AA fetopathy PCT density, and demonstrated in utero cessation may recover PCT differentiation. Future studies may benefit from calculating PCT percentage as a potential biomarker to correlate with post-natal renal function and maternal factors including medication type, dosage, duration, and time from medication cessation.


Asunto(s)
Anomalías Inducidas por Medicamentos/etiología , Antagonistas de Receptores de Angiotensina/efectos adversos , Inhibidores de la Enzima Convertidora de Angiotensina/efectos adversos , Enfermedades Fetales/inducido químicamente , Enfermedades Renales/inducido químicamente , Túbulos Renales Proximales/anomalías , Anomalías Inducidas por Medicamentos/diagnóstico , Anomalías Inducidas por Medicamentos/metabolismo , Anomalías Inducidas por Medicamentos/patología , Biomarcadores/metabolismo , Estudios de Casos y Controles , Femenino , Muerte Fetal/etiología , Enfermedades Fetales/diagnóstico , Enfermedades Fetales/metabolismo , Enfermedades Fetales/patología , Humanos , Inmunohistoquímica , Recién Nacido , Enfermedades Renales/congénito , Enfermedades Renales/diagnóstico , Enfermedades Renales/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Masculino , Mucina-1/metabolismo , Neprilisina/metabolismo , Estudios Retrospectivos
3.
PLoS Med ; 17(9): e1003322, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32870921

RESUMEN

BACKGROUND: Despite the widespread use, only sparse information is available on the safety of gabapentin during pregnancy. We sought to evaluate the association between gabapentin exposure during pregnancy and risk of adverse neonatal and maternal outcomes. METHODS AND FINDINGS: Using the United States Medicaid Analytic eXtract (MAX) dataset, we conducted a population-based study of 1,753,865 Medicaid-eligible pregnancies between January 2000 and December 2013. We examined the risk of major congenital malformations and cardiac defects associated with gabapentin exposure during the first trimester (T1), and the risk of preeclampsia (PE), preterm birth (PTB), small for gestational age (SGA), and neonatal intensive care unit admission (NICUa) associated with gabapentin exposure early, late, or both early and late in pregnancy. Gabapentin-unexposed pregnancies served as the reference. We estimated relative risks (RRs) and 95% confidence intervals (CIs) using fine stratification on the propensity score (PS) to control for over 70 confounders (e.g., maternal age, race/ethnicity, indications for gabapentin, other pain conditions, hypertension, diabetes, use of opioids, and specific morphine equivalents). We identified 4,642 pregnancies exposed in T1 (mean age = 28 years; 69% white), 3,745 exposed in early pregnancy only (28 years; 67% white), 556 exposed in late pregnancy only (27 years; 60% white), and 1,275 exposed in both early and late pregnancy (29 years; 75% white). The reference group consisted of 1,744,447 unexposed pregnancies (24 years; 40% white). The adjusted RR for major malformations was 1.07 (95% CI 0.94-1.21, p = 0.33) and for cardiac defects 1.12 (0.89-1.40, p = 0.35). Requiring ≥2 gabapentin dispensings moved the RR to 1.40 (1.03-1.90, p = 0.03) for cardiac defects. There was a higher risk of preterm birth among women exposed to gabapentin either late (RR, 1.28 [1.08-1.52], p < 0.01) or both early and late in pregnancy (RR, 1.22 [1.09-1.36], p < 0.001), SGA among women exposed to gabapentin early (1.17 [1.02-1.33], p = 0.02), late (1.39 [1.01-1.91], p = 0.05), or both early and late in pregnancy (RR, 1.32 [1.08-1.60], p < 0.01), and NICU admission among women exposed to gabapentin both early and late in pregnancy (RR, 1.35 [1.20-1.52], p < 0.001). There was no higher risk of preeclampsia among women exposed to gabapentin after adjustment. Study limitations include the potential for residual confounding and exposure misclassification. CONCLUSIONS: In this large population-based study, we did not find evidence for an association between gabapentin exposure during early pregnancy and major malformations overall, although there was some evidence of a higher risk of cardiac malformations. Maternal use of gabapentin, particularly late in pregnancy, was associated with a higher risk of PTB, SGA, and NICUa.


Asunto(s)
Gabapentina/efectos adversos , Efectos Tardíos de la Exposición Prenatal/metabolismo , Anomalías Inducidas por Medicamentos/metabolismo , Adulto , Estudios de Cohortes , Femenino , Gabapentina/uso terapéutico , Humanos , Recién Nacido Pequeño para la Edad Gestacional , Preeclampsia/fisiopatología , Embarazo , Complicaciones del Embarazo , Primer Trimestre del Embarazo , Nacimiento Prematuro , Puntaje de Propensión , Riesgo , Estados Unidos
4.
Pak J Pharm Sci ; 33(6(Supplementary)): 2721-2728, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33879430

RESUMEN

The study is aimed to investigate the protective role of Moringa oleifera extracts against sodium arsenate induced embryo toxicity in albino mice. Forty four pregnant mice were divided into 11groups (A-K). Group A was control while B and C were sodium arsenate treated groups with dose, A (0.00), B (6.00, 0.00), C (12.00, 0.00). Group D to G were of sodium arsenate+Moringa oleifera flower extract treated groups with doses D (6.00, 150.00), E (6.00, 300.00), F (12.00, 150.00), G (12.00, 300.00) and groups H to K were sodium arsenate+Moringa oleifera leaf extract treated groups H (6.00, 150.00), I (6.00, 300.00), J (12.00, 150.00) and K (12.00, 300.00) mg/kg B.W. Moringa oleifera leaf extract treated groups showed significant (p<0.05) amelioration against sodium arsenate induced histopathological changes as malformed heart, spina bifida, enlarged ventricles, poorly developed kidneys, anopthalmia and cavitation in brain. Significant (p<0.05) increased in malondialdehyde 36±0.81 and decreased glutathione 8.25±0.95 values in sodium arsenate treated groups were observed as compared to control 22.5±0.57 and 19±0.81.Whereas Moringa oleifera leaf extract at dose of 300mg/kg B.W normalizesd the malondialdehyde 23±0.81 and glutathione 17.75±3.20 values. So concluded that Moringa oleifera leaf extract has ameliorative effects against sodium arsenate induced embryotoxicity.


Asunto(s)
Anomalías Inducidas por Medicamentos/prevención & control , Arseniatos/toxicidad , Moringa oleifera , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacología , Anomalías Inducidas por Medicamentos/metabolismo , Anomalías Inducidas por Medicamentos/patología , Animales , Peso Corporal/efectos de los fármacos , Femenino , Masculino , Ratones , Moringa oleifera/química , Hojas de la Planta , Embarazo
5.
Ecotoxicol Environ Saf ; 181: 559-571, 2019 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-31238190

RESUMEN

Warfarin is the most worldwide used anticoagulant drug and rodenticide. Since it crosses placental barrier it can induce warfarin embryopathy (WE), a fetal mortality in neonates characterized by skeletal deformities in addition to brain hemorrhages. Although the effects of warfarin exposure in aquatic off target species were already described, the particular molecular toxicological mechanisms during early development are still unclear. Here, we used zebrafish (Danio rerio) to describe and compare the developmental effects of warfarin exposure (0, 15.13, 75.68 and 378.43 mM) on two distinct early developmental phases (embryos and eleuthero-embryos). Although exposure to both developmental phases induced fish mortality, only embryos exposed to the highest warfarin level exhibited features mimicking mammalian WE, e.g. high mortality, higher incidence of hemorrhages and altered skeletal development, among other effects. To gain insights into the toxic mechanisms underlying warfarin exposure, the transcriptome of embryos exposed to warfarin was explored through RNA-Seq and compared to that of control embryos. 766 differentially expressed (564 up- and 202 down-regulated) genes were identified. Gene Ontology analysis revealed particular cellular components (cytoplasm, extracellular matrix, lysosome and vacuole), biological processes (mainly amino acid and lipid metabolism and response to stimulus) and pathways (oxidative stress response and apoptosis signaling pathways) being significantly overrepresented in zebrafish embryos upon warfarin exposure. Protein-protein interaction further evidenced an altered redox system, blood coagulation and vasculogenesis, visual phototransduction and collagen formation upon warfarin exposure. The present study not only describes for the first time the WE in zebrafish, it provides new insights for a better risk assessment, and highlights the need for programming the rat eradication actions outside the fish spawning season to avoid an impact on off target fish community. The urge for the development of more species-specific anticoagulants for rodent pest control is also highlighted.


Asunto(s)
Anomalías Inducidas por Medicamentos/metabolismo , Anticoagulantes/toxicidad , Hueso Nasal/anomalías , Rodenticidas/toxicidad , Warfarina/efectos adversos , Warfarina/toxicidad , Contaminantes Químicos del Agua/toxicidad , Anomalías Inducidas por Medicamentos/genética , Animales , Modelos Animales de Enfermedad , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Humanos , Hueso Nasal/metabolismo , Estrés Oxidativo/efectos de los fármacos , Transcriptoma , Warfarina/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo
6.
Ecotoxicol Environ Saf ; 156: 34-40, 2018 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-29525683

RESUMEN

Selenium (Se) is an essential element and its biological activity is related to its speciation. It is also well-known that in excess it can cause teratogenesis in fish and birds. In this study we compared dietary toxicity of elemental selenium nanoparticles (SeNPs) with selenite and selenomethionine (Se-Met). Japanese medaka (Oryzias latipes) was used as a laboratory model to determine Se effects on adults and their offspring. Adult females were individually exposed using a dry diet fortified with 0, 10 or 20 µg/g of the three Se species for 7 days and then allowed to breed for 3 days. Fertilization rate and the proportion of malformed offspring were examined. The three Se diets led to significant increase in maternal tissue Se concentration in the order of Se-Met >>selenite > SeNP. However, in terms of proportion of malformed offspring, the effect of Se-Met = selenite > SeNP. The malformations included pericardial edema and craniofacial changes, which were typical for Se toxicity. The mismatch of maternal ovary Se concentration and proportion of malformed offspring suggested total Se concentration is a poor predictor of toxicity and teratogenesis. Comparing expression of four genes related to oxidative stress in maternal tissue also showed that there were significant differences in expression patterns between three Se diets in the order of selenite = SeNP > Se-Met. Our results showed that SeNPs cause similar toxicity as other Se species but require further study to elucidate the underlying mechanism.


Asunto(s)
Anomalías Inducidas por Medicamentos , Exposición Dietética , Exposición Materna , Nanopartículas , Selenio/toxicidad , Anomalías Inducidas por Medicamentos/genética , Anomalías Inducidas por Medicamentos/metabolismo , Animales , Femenino , Oryzias/genética , Oryzias/metabolismo , Estrés Oxidativo , Ácido Selenioso/toxicidad , Selenometionina/toxicidad
7.
Birth Defects Res A Clin Mol Teratol ; 106(10): 803-813, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27435288

RESUMEN

BACKGROUND: Craniosynostosis, the premature fusion of one or more of the cranial sutures, is estimated to occur in 1:1800 to 2500 births. Genetic murine models of craniosynostosis exist, but often imperfectly model human patients. Case, cohort, and surveillance studies have identified excess thyroid hormone as an agent that can either cause or exacerbate human cases of craniosynostosis. METHODS: Here we investigate the influence of in utero and in vitro exogenous thyroid hormone exposure on a murine model of craniosynostosis, Twist 1 +/-. RESULTS: By 15 days post-natal, there was evidence of coronal suture fusion in the Twist 1 +/- model, regardless of exposure. With the exception of craniofacial width, there were no significant effects of exposure; however, the Twist 1 +/- phenotype was significantly different from the wild-type control. Twist 1 +/- cranial suture cells did not respond to thyroxine treatment as measured by proliferation, osteogenic differentiation, and gene expression of osteogenic markers. However, treatment of these cells did result in modulation of thyroid associated gene expression. CONCLUSION: Our findings suggest the phenotypic effects of the genetic mutation largely outweighed the effects of thyroxine exposure in the Twist 1 +/- model. These results highlight difficultly in experimentally modeling gene-environment interactions for craniosynostotic phenotypes. Birth Defects Research (Part A) 106:803-813, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Anomalías Inducidas por Medicamentos , Craneosinostosis , Interacción Gen-Ambiente , Proteínas Nucleares/genética , Fenotipo , Tiroxina/efectos adversos , Proteína 1 Relacionada con Twist/genética , Anomalías Inducidas por Medicamentos/genética , Anomalías Inducidas por Medicamentos/metabolismo , Anomalías Inducidas por Medicamentos/patología , Animales , Craneosinostosis/inducido químicamente , Craneosinostosis/genética , Craneosinostosis/metabolismo , Craneosinostosis/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Ratones , Ratones Mutantes , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Tiroxina/farmacología
8.
Differentiation ; 88(2-3): 70-83, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25449353

RESUMEN

The effect of neonatal exposure to diethylstilbestrol (DES), a potent synthetic estrogen, was examined to evaluate whether the CD-1 (estrogen insensitive, outbred) and C57 (estrogen sensitive, inbred) mouse strains differ in their response to estrogen disruption of male ExG differentiation. CD-1 and C57BL/6 litters were injected with sesame oil or DES (200 ng/g/5 µl in sesame oil vehicle) every other day from birth to day 10. Animals were sacrificed at the following time points: birth, 5, 10 and 60 days postnatal. Neonatally DES-treated mice from both strains had many ExG abnormalities that included the following: (a) severe truncation of the prepuce and glans penis, (b) an abnormal urethral meatus, (c) ventral tethering of the penis, (d) reduced os penis length and glans width, (e) impaired differentiation of cartilage, (f) absence of urethral flaps, and (g) impaired differentiation of erectile bodies. Adverse effects of DES correlated with the expression of estrogen receptors within the affected tissues. While the effects of DES were similar in the more estrogen-sensitive C57BL/6 mice versus the less estrogen-sensitive CD-1 mice, the severity of DES effects was consistently greater in C57BL/6 mice. We suggest that many of the effects of DES, including the induction of hypospadias, are due to impaired growth and tissue fusion events during development.


Asunto(s)
Anomalías Inducidas por Medicamentos/patología , Dietilestilbestrol/toxicidad , Estrógenos/metabolismo , Genitales Masculinos/anomalías , Receptores de Estrógenos/genética , Anomalías Inducidas por Medicamentos/etiología , Anomalías Inducidas por Medicamentos/metabolismo , Animales , Femenino , Genitales Masculinos/efectos de los fármacos , Genitales Masculinos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Receptores de Estrógenos/metabolismo , Especificidad de la Especie
9.
Arch Toxicol ; 88(9): 1749-63, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24623309

RESUMEN

The tumor suppressor protein p53 is a powerful regulator of the embryo's susceptibility to diverse teratogenic stimuli, functioning both as a teratogenesis inducer and suppressor. However, the targets that p53 engages to fulfill its functions remain largely undefined. We asked whether the microRNA (miRNA) miR-34 family, identified as one of the main targets of p53, mediates its function as a teratogenesis inducer. For this, pregnant ICR-, p53- and miR-34a-deficient mice, as well as rats, were exposed to 5-aza-2'-deoxycytidine (5-aza), a teratogen inducing limb reduction anomalies (LRA) of the hindlimbs in mice and either the hindlimbs or forelimbs in rats. Using hind- and forelimb buds of 5-aza-exposed embryos, we identified that the miR-34 family members are the most upregulated miRNAs in mouse and rat limb buds, with their increase level being significantly higher in limb buds destined for LRA. We showed that p53 mediates the 5-aza-induced miR-34 transcription followed by met proto-oncogene and growth-arrest-specific 1 target suppression in embryonic limb buds. We demonstrated that p53 regulates the teratogenic response to 5-aza acting as a teratogenesis inducer albeit miR-34a deletion does not affect the susceptibility of mice to 5-aza. Overall, our study thoroughly characterizes the expression and regulation of miR-34 family in teratogen-resistant and teratogen-sensitive embryonic structures and discusses the involvement of epigenetic miRNA-mediated pathway(s) in induced teratogenesis.


Asunto(s)
Anomalías Inducidas por Medicamentos/metabolismo , Esbozos de los Miembros/efectos de los fármacos , Deformidades Congénitas de las Extremidades/inducido químicamente , MicroARNs/metabolismo , Teratógenos/toxicidad , Proteína p53 Supresora de Tumor/metabolismo , Animales , Azacitidina/administración & dosificación , Azacitidina/análogos & derivados , Azacitidina/toxicidad , Decitabina , Relación Dosis-Respuesta a Droga , Resistencia a Medicamentos , Femenino , Desarrollo Fetal/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Heterocigoto , Esbozos de los Miembros/anomalías , Esbozos de los Miembros/metabolismo , Deformidades Congénitas de las Extremidades/metabolismo , Masculino , Ratones Endogámicos ICR , Ratones Noqueados , Embarazo , Ratas Sprague-Dawley , Proteína p53 Supresora de Tumor/genética , Regulación hacia Arriba/efectos de los fármacos
10.
Indian J Exp Biol ; 52(2): 159-67, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24597149

RESUMEN

Citrinin is the one of the well-known mycotoxins, which is possibly spread all over the world. The graded doses of citrinin (1, 3 and 5 ppm CIT in feed) in female Wistar rats 10 weeks prior to mating, during mating and during organogenesis resulted in resorptions and post implantation losses, decreased fetal body weights and crown-rump lengths in fetuses of all groups. Various developmental anomalies recorded in fetuses of treated rats included gross (wrist drop, curled tail, stretched forelimb, subcutaneous haematoma), skeletal (incomplete ossification of skull bones, incomplete fusion of vertebral bodies, complete and partial agenesis of sternaebrae, metacarpals, metatarsals and phalanges, fused ribs and swing out ribs) and visceral (internal and external hydrocephalus, cerebellar hypoplasia, microphthalmia, roundening of heart, contracted kidneys, dilated renal pelvis and cryptorchid testes). The results suggest that CIT has adverse effects on fetal development which may be due to the longer bioavailability of citrinin in the animals.


Asunto(s)
Anomalías Inducidas por Medicamentos/patología , Citrinina/efectos adversos , Pérdida del Embrión/patología , Desarrollo Fetal/efectos de los fármacos , Anomalías Inducidas por Medicamentos/clasificación , Anomalías Inducidas por Medicamentos/metabolismo , Animales , Citrinina/administración & dosificación , Pérdida del Embrión/inducido químicamente , Desarrollo Embrionario/efectos de los fármacos , Femenino , Masculino , Micotoxinas/toxicidad , Ratas , Ratas Wistar , Reproducción/efectos de los fármacos , Teratología
11.
Mol Pharmacol ; 84(3): 353-60, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23775563

RESUMEN

Activation of the transcription factor aryl hydrocarbon receptor by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) prevents the formation of the epicardium and leads to severe heart malformations in developing zebrafish (Danio rerio). The downstream genes that cause heart malformation are not known. Because TCDD causes craniofacial malformations in zebrafish by downregulating the sox9b gene, we hypothesized that cardiotoxicity might also result from sox9b downregulation. We found that sox9b is expressed in the developing zebrafish heart ventricle and that TCDD exposure markedly reduces this expression. Furthermore, we found that manipulation of sox9b expression could phenocopy many but not all of the effects of TCDD at the heart. Loss of sox9b prevented the formation of epicardium progenitors comprising the proepicardium on the pericardial wall, and prevented the formation and migration of the epicardial layer around the heart. Zebrafish lacking sox9b showed pericardial edema, an elongated heart, and reduced blood circulation. Fish lacking sox9b failed to form valve cushions and leaflets. Sox9b is one of two mammalian Sox9 homologs, sox9b and sox9a. Knock down of sox9a expression did not cause cardiac malformations, or defects in epicardium development. We conclude that the decrease in sox9b expression in the heart caused by TCDD plays a role in many of the observed signs of cardiotoxicity. We find that while sox9b is expressed in myocardial cells, it is not normally expressed in the affected epicardial cells or progenitors. We therefore speculate that sox9b is involved in signals between the cardiomyocytes and the nascent epicardial cells.


Asunto(s)
Anomalías Inducidas por Medicamentos/metabolismo , Cardiopatías Congénitas/inducido químicamente , Pericardio/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Factor de Transcripción SOX9/metabolismo , Proteínas de Pez Cebra/metabolismo , Anomalías Inducidas por Medicamentos/fisiopatología , Animales , Circulación Coronaria , Regulación hacia Abajo , Edema/inducido químicamente , Edema/metabolismo , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/fisiopatología , Válvulas Cardíacas/anomalías , Válvulas Cardíacas/efectos de los fármacos , Válvulas Cardíacas/embriología , Válvulas Cardíacas/crecimiento & desarrollo , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/embriología , Ventrículos Cardíacos/crecimiento & desarrollo , Ventrículos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Pericardio/embriología , Pericardio/crecimiento & desarrollo , Pericardio/metabolismo , Pez Cebra
12.
Arterioscler Thromb Vasc Biol ; 32(4): 851-5, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22423036

RESUMEN

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.


Asunto(s)
Adenosina/metabolismo , Corazón/embriología , Miocardio/metabolismo , Receptor de Adenosina A1/metabolismo , Transducción de Señal , Anomalías Inducidas por Medicamentos/etiología , Anomalías Inducidas por Medicamentos/metabolismo , Antagonistas del Receptor de Adenosina A1/farmacología , Antagonistas del Receptor de Adenosina A1/toxicidad , Animales , Técnicas de Cultivo de Embriones , Corazón/efectos de los fármacos , Cardiopatías Congénitas/inducido químicamente , Cardiopatías Congénitas/metabolismo , Ratones , Ratones Transgénicos , Receptor de Adenosina A1/efectos de los fármacos , Receptor de Adenosina A1/genética , Transducción de Señal/efectos de los fármacos
13.
Cell Mol Life Sci ; 68(9): 1569-79, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21207098

RESUMEN

Fifty years ago, prescription of the sedative thalidomide caused a worldwide epidemic of multiple birth defects. The drug is now used in the treatment of leprosy and multiple myeloma. However, its use is limited due to its potent teratogenic activity. The mechanism by which thalidomide causes limb malformations and other developmental defects is a long-standing question. Multiple hypotheses exist to explain the molecular mechanism of thalidomide action. Among them, theories involving oxidative stress and anti-angiogenesis have been widely supported. Nevertheless, until recently, the direct target of thalidomide remained elusive. We identified a thalidomide-binding protein, cereblon (CRBN), as a primary target for thalidomide teratogenicity. Our data suggest that thalidomide initiates its teratogenic effects by binding to CRBN and inhibiting its ubiquitin ligase activity. In this review, we summarize the biology of thalidomide, focusing on the molecular mechanisms of its teratogenic effects. In addition, we discuss the questions still to be addressed.


Asunto(s)
Anomalías Inducidas por Medicamentos/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Estrés Oxidativo , Péptido Hidrolasas/metabolismo , Teratógenos/toxicidad , Talidomida/toxicidad , Proteínas Adaptadoras Transductoras de Señales , Animales , Embrión de Pollo , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Humanos , Estrés Oxidativo/efectos de los fármacos , Conejos , Especificidad de la Especie , Teratógenos/química , Teratógenos/farmacocinética , Talidomida/química , Talidomida/farmacocinética , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Pez Cebra
14.
Arch Environ Contam Toxicol ; 63(3): 445-52, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22878731

RESUMEN

Deoxynivalenol (DON [vomitoxin]), one of trichothecene mycotoxins produced by the fungus Fusarium, is commonly detected in cereal foods across the world. DON induces diverse toxic effects in humans and animals, including emesis and diarrhea, anorexia, and immunotoxicity, and impaired maternal reproduction and fetal development. Recently, the teratogenic potential of DON has been shown and has received much attention. DON can cause various skeletal deformities in fetuses, but the underlying mechanisms have not yet been fully examined. In this study, fetal skeletal malformations in DON-treated maternal mice were thoroughly investigated using microarray assay. The results showed that DON administration caused various skeletal defects in fetuses, including misaligned or fused sternebrae and vertebrae, divided or fused ribs and polydactyly, hemivertebrae, short toes, and tail anomalies. Microarray analysis showed that 282 genes, including 148 downregulated and 134 upregulated genes, were abnormally expressed in fetal vertebral bones after maternal DON exposure. These identified genes can be classified into several categories: skeletal development, carcinogenesis, nervous disorders, sperm development and embryogenesis, and inflammation. Of these, 6 genes, mostly related to bone development, were intentionally selected for further validation using real-time reverse transcription-Polymerase Chain Reaction (RT-PCR). It was confirmed that the mRNA expression of 4 genes, i.e., fibrillin-1, Col9A2, 3'-phosphoadenosine 5'-phosphosulfate synthase 2, and Pax1, was upregulated significantly by DON administration, whereas that of 2 other genes, Runx2 and parathyroid hormone-like hormone, was downregulated significantly. Taken together, the results of our study suggest that altered expression of these 6 genes plays a critical role in fetal skeletal deformities induced by DON and thus they are worthy of further investigation.


Asunto(s)
Anomalías Inducidas por Medicamentos/patología , Desarrollo Fetal/efectos de los fármacos , Teratógenos/toxicidad , Tricotecenos/toxicidad , Anomalías Inducidas por Medicamentos/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices
15.
J Environ Monit ; 14(8): 2254-60, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22777571

RESUMEN

The Eastern coast of Tunisia hosts a mosaic of populations of the Mediterranean killifish A. fasciatus, an excellent monitoring species for the analysis of environmental impact in coastal Mediterranean areas. Among these populations, fish from the Sfax area, in the Gulf of Gabès, are known for presenting a high prevalence of different physiological alterations, including spinal deformation. Compared to a reference clean area, Luza, Sfax water and sediments are polluted with heavy metals (20 to 40 times more Cd, for example) and other compounds like PAHs (10 times higher levels). Histological analyses of deformed fish from this area showed an abnormal abundance of collagen fibers infiltrated in the spinal area, either as a physiological mechanism to alleviate the consequences of the deformation of the bones or as a secondary effect of the same pathological/toxicity mechanism inducing bone deformation. Deformed fish from Sfax showed elevated (4-5 fold) levels of transcripts from the collagen COL1A2 gene in bone tissue, compared to either non-deformed animals from the same location or normal animals from the reference Luza site. These results are consistent with an overexpression of collagen in the bone tissue, in agreement with the histological results. We propose that the deformation observed in the Sfax Aphanius population is a degenerative process occurring in adult animals and probably terminal, as it results in severe limitations on the fish capacity to swim, to catch food, and ultimately to survive.


Asunto(s)
Anomalías Inducidas por Medicamentos/veterinaria , Monitoreo del Ambiente , Enfermedades de los Peces/epidemiología , Peces Killi/anomalías , Contaminantes Químicos del Agua/toxicidad , Anomalías Inducidas por Medicamentos/epidemiología , Anomalías Inducidas por Medicamentos/metabolismo , Anomalías Inducidas por Medicamentos/patología , Animales , Colágeno Tipo I/metabolismo , Monitoreo Epidemiológico , Femenino , Enfermedades de los Peces/metabolismo , Enfermedades de los Peces/patología , Peces Killi/metabolismo , Masculino , Metales Pesados/análisis , Metales Pesados/toxicidad , Hidrocarburos Policíclicos Aromáticos/análisis , Hidrocarburos Policíclicos Aromáticos/toxicidad , Columna Vertebral/anomalías , Columna Vertebral/patología , Túnez , Contaminantes Químicos del Agua/análisis
16.
Alcohol Clin Exp Res ; 35(9): 1644-61, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21554333

RESUMEN

BACKGROUND: Microphthalmia (reduced eye size), generally accompanied by vision defects, is a hallmark of fetal alcohol spectrum disorder (FASD) in humans. In zebrafish, embryonic ethanol exposure over the time of retinal neurogenesis also results in microphthalmia. This microphthalmia is in part the consequence of reduced retinal cell differentiation, including photoreceptors. Here we pursue 2 signaling pathways implicated in other aspects of FASD pathogenesis: retinoic acid (RA) and Sonic hedgehog (Shh). METHODS: We evaluated markers for RA and Shh signaling within the eyes of embryos treated with ethanol during the period of retinal neurogenesis. We also performed rescue experiments using administration of exogenous RA and microinjection of cholesterol, which augments Shh signaling. RESULTS: Using sequential or co-treatments, RA did not rescue ethanol-induced microphthalmia at any concentration tested. In addition, RA itself caused microphthalmia, although the underlying mechanisms were distinct from those of ethanol. Interestingly, RA treatment appeared to recover photoreceptor differentiation in a concentration-dependent manner. This may be an independent effect of exogenous RA, as ethanol treatment alone did not alter RA signaling in the eye. Cholesterol injection also did not rescue ethanol-induced microphthalmia at any concentration tested, and ethanol treatments did not alter expression of shh, or of ptc-2, which is normally regulated by Shh signaling. CONCLUSIONS: Together these findings indicate that, during the time of retinal neurogenesis, effects of ethanol on eye development are likely independent of the RA and Shh signaling pathways. These studies suggest that FASD intervention strategies based upon augmentation of RA or Shh signaling may not prevent ethanol-induced microphthalmia.


Asunto(s)
Depresores del Sistema Nervioso Central/toxicidad , Etanol/toxicidad , Proteínas Hedgehog/metabolismo , Microftalmía/inducido químicamente , Neurogénesis , Retina/embriología , Tretinoina/metabolismo , Anomalías Inducidas por Medicamentos/metabolismo , Anomalías Inducidas por Medicamentos/patología , Naranja de Acridina/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Femenino , Microftalmía/metabolismo , Microftalmía/patología , Retina/metabolismo , Retina/patología , Transducción de Señal/efectos de los fármacos , Transgenes , Tretinoina/farmacología , Tretinoina/toxicidad , Vitaminas/metabolismo , Vitaminas/farmacología , Vitaminas/toxicidad , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo
17.
J Appl Toxicol ; 31(3): 210-22, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20878907

RESUMEN

Perfluorooctane sulfonate (PFOS), a kind of widely distributed environmentally organic compound, has been found to cause developmental toxicity. Although microRNAs (miRNAs) play an important role in many metabolic tasks, whether and how they are involved in the process of PFOS-induced toxicity is largely unknown. To address this problem, PFOS-induced changes in miRNAs and target gene expression in zebrafish embryos, and the potential mechanism of PFOS-induced toxic action were studied in this research. Zebrafish embryos were exposed to 1 µg ml(-1) PFOS or DMSO control from 6 h post-fertilization (hpf) to 24 or 120 hpf. Subsequently, RNA was isolated from the embryo pool and the expression profiles of 219 known zebrafish miRNAs were analyzed using microarray. Finally, quantitative real-time polymerase chain reaction was used to validate several miRNAs expression of microarray data. The analysis revealed that PFOS exposure induced significant changes in miRNA expression profiles. A total of 39 and 81 miRNAs showed significantly altered expression patterns after PFOS exposure 24 and 120 hpf. Of the changed miRNAs, 20 were significantly up-regulated and 19 were significantly down-regulated (p < 0.01) at 24 hpf, whereas 41 were significantly up-regulated and 40 were significantly down-regulated (p < 0.01) at 120 hpf. These miRNAs were involved in development, apoptosis and cell signal pathway, cell cycle progression and proliferation, oncogenesis, adipose metabolism and hormone secretion, whereas there is still little functional information available for 32 miRNAs. Our results demonstrate that PFOS exposure alters the expression of a suite of miRNAs and may induce developmental toxicity.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Embrión no Mamífero/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Fluorocarburos/toxicidad , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , MicroARNs/metabolismo , Pez Cebra/embriología , Anomalías Inducidas por Medicamentos/etiología , Anomalías Inducidas por Medicamentos/genética , Anomalías Inducidas por Medicamentos/metabolismo , Animales , Embrión no Mamífero/anomalías , Desarrollo Embrionario/genética , Longevidad/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero , Factores de Tiempo , Pez Cebra/genética , Pez Cebra/fisiología
18.
Environ Toxicol ; 26(1): 103-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19746408

RESUMEN

We identified a novel zebrafish mutant that has wavy-notochord phenotypes, such as severely twisted notochord and posterior malformations, but has normal melanocytes. Histological evidences showed that proliferating vacuolar cells extended their growth to the muscle region, and consequently caused the wavy-notochord phenotypes. Interestingly, those malformations can be greatly reversed by exposure with copper, suggesting that copper plays an important role on wavy-notochord phenotypes. In addition, after long-term copper exposure, the surviving larvae derived from wavy-notochord mutants displayed bone malformations, such as twisted axial skeleton and osteophyte. These phenotypic changes and molecular evidences of wavy-notochord mutants are highly similar to those embryos whose lysyl oxidases activities have been inactivated. Taken together, we propose that (i) the putative mutated genes of this wavy-notochord mutant might be highly associated with the lysyl oxidase genes in zebrafish; and (ii) this fish model is an effective tool for monitoring copper pollution of water from natural resources.


Asunto(s)
Anomalías Inducidas por Medicamentos/embriología , Cobre/toxicidad , Mutación , Notocorda/anomalías , Contaminantes Químicos del Agua/toxicidad , Pez Cebra/anomalías , Anomalías Inducidas por Medicamentos/metabolismo , Animales , Embrión no Mamífero/anomalías , Embrión no Mamífero/efectos de los fármacos , Monitoreo del Ambiente/métodos , Modelos Biológicos , Notocorda/metabolismo , Notocorda/patología , Proteína-Lisina 6-Oxidasa/genética , Proteína-Lisina 6-Oxidasa/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo
19.
Indian J Exp Biol ; 49(3): 183-90, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21452597

RESUMEN

Heterocyclic compounds such as quinazolinones have variety of biological and pharmacological properties (anticancer, antiinflammatory, antimicrobial, antimalaria, etc.). Effects of two new quinazolinones viz., 4(3H)-quinazolinone-2-propyl-2-phenylethyl (QPPE) and 4(3H)quinazolinone-2-ethyl-2-phenylethyl (QEPE) were investigated on Balb/C mice embryos livers-the major organ of metabolism and detoxification of drugs and toxins. Histological and pathological studies demonstrated QPPE and QEPE as producers of toxic metabolites after biotransformation, creating necrosis, fatty changes, increase in the number of band cells, hepatocytes' diameters and alkaline phosphatase, in addition to sinusoid dilation, hemorrhages and hyperemia. Transmission electron micrographs showed lipid droplets in hepatocytes' cytoplasm, necrosis, vacuolization, cytoplasm disintegration, disfigured and swollen mitochondria, irregular and abnormal nuclei, nuclei with heterochromatin, condensed chromatins, myelin figures and autophages in injured hepatocytes. In conclusion, QPPE and QEPE make toxic components after biotransformation injuring membranes and creating inflammatory reactions. They also disturb metabolism of lipids pathways and cause the appearances of lipid droplets in hepatocytes.


Asunto(s)
Hígado/anomalías , Hígado/efectos de los fármacos , Quinazolinonas/toxicidad , Anomalías Inducidas por Medicamentos/embriología , Anomalías Inducidas por Medicamentos/metabolismo , Anomalías Inducidas por Medicamentos/patología , Animales , Biotransformación , Femenino , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/embriología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Embarazo , Quinazolinonas/farmacocinética , Teratógenos/farmacocinética , Teratógenos/toxicidad
20.
Bull Environ Contam Toxicol ; 86(4): 389-93, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21404046

RESUMEN

The toxic effects of water-soluble fraction (WSF) of crude oil (API27, Petrobras Campos Basin, Brazil) were evaluated during the early life stages of zebrafish, as well as its biotransformation in juvenile fish. Embryonic development was studied during 96 h. Reduced heartbeat rate, weak pigmentation, tail defects, and embryo mortality were observed for all of the tested concentrations of the WSF. Activities of the biotransformation enzymes were induced at the highest concentrations, showing that these enzymes played a role in its elimination. As shown in this study the crude oil WSF altered the normal embryonic development of fish.


Asunto(s)
Embrión no Mamífero/efectos de los fármacos , Petróleo/toxicidad , Contaminantes Químicos del Agua/toxicidad , Pez Cebra/metabolismo , Anomalías Inducidas por Medicamentos/embriología , Anomalías Inducidas por Medicamentos/metabolismo , Animales , Biotransformación , Citocromo P-450 CYP1A1/metabolismo , Relación Dosis-Respuesta a Droga , Embrión no Mamífero/metabolismo , Glutatión Transferasa/metabolismo , Pez Cebra/embriología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda