Your browser doesn't support javascript.
loading
Mostrar: 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.
J Ethnopharmacol ; 267: 113538, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33144170

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Clerodendrum cyrtophyllum Turcz has been used in traditional medicine for the treatment of various diseases. In spite of its therapeutic applications, research on its toxicity and teratogenicity is still limited. AIM OF THE STUDY: The study aimed to investigate the developmental toxicity of the ethanol extract of C. cyrtophyllum (EE) in zebrafish embryo model. MATERIAL AND METHODS: Major compounds from crude ethanol extract of Clerodendron cyrtophyllum Turcz leaves were determined using HPLC-DAD-Orbitrap-MS analysis. The developmental toxicity of EE were investigated using zebrafish embryo model. Zebrafish embryos at 6 h post-fertilization (hpf) were treated with EE at different concentrations. Egg coagulation, mortality, hatching, yolk sac edema, pericardial edema and teratogenicity were recorded each day for during a 5-day exposure. At time point 120 hpf, body length, pericardial area, heartbeat and yolk sac area were assessed. In order to elucidate molecular mechanisms for the developmental toxicity of EE, we further evaluated the effects of the EE on the expression of genes involved on signaling pathways affecting fish embryo's development such as heart development (gata5, myl7, myh6, has2, hand2, nkx 2.5), oxidative stress (cat, sod1, gpx4, gstp2), wnt pathway (ß-catenin, wnt3a, wnt5, wnt8a, wnt11), or cell apoptosis (p53, bax, bcl2, casp3, casp8, casp9, apaf-1, gadd45bb) using qRT-PCR analysis. RESULTS: Our results demonstrated that three major components including acteoside, cirsilineol and cirsilineol-4'-O-ß-D-glucopyranoside were identified from EE. EE exposure during 6-96 h post-fertilization (hpf) at doses ranging from 80 to 200 µg/mL increased embryo mortality and reduced hatching rate. EE exposure at 20 and 40 µg/mL until 72-120 hpf induced a series of malformations, including yolk sac edema, pericardial edema, spine deformation, shorter body length. Based on two prediction models using a teratogenic index (TI), a 25% lethality concentration (LD25) and the no observed-adverse-effect level (NOAEL), EE is considered as teratogenic for zebrafish embryos with TI (LC50/EC50) and LD25/NOAEC values at 96 hpf reaching 3.87 and 15.73 respectively. The mRNA expression levels of p53, casp8, bax/bcl2, gstp2, nkx2.5, wnt3a, wnt11, gadd45bb and gata5 were significantly upregulated by EE exposure at 20 and 40 µg/mL while the expression of wnt5, hand2 and bcl2 were downregulated. CONCLUSIONS: These results provide evidence for toxicity effects of EE to embryo stages and provide an insight into the potential toxicity mechanisms on embryonic development.


Asunto(s)
Anomalías Inducidas por Medicamentos/etiología , Clerodendrum/toxicidad , Embrión no Mamífero/efectos de los fármacos , Extractos Vegetales/toxicidad , Pez Cebra/embriología , Anomalías Inducidas por Medicamentos/genética , Anomalías Inducidas por Medicamentos/metabolismo , Anomalías Inducidas por Medicamentos/patología , Animales , Apoptosis/efectos de los fármacos , Clerodendrum/química , Relación Dosis-Respuesta a Droga , Embrión no Mamífero/metabolismo , Embrión no Mamífero/patología , Etanol/química , Regulación del Desarrollo de la Expresión Génica , Nivel sin Efectos Adversos Observados , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/aislamiento & purificación , Solventes/química , Vía de Señalización Wnt/efectos de los fármacos , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
4.
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
5.
Reprod Toxicol ; 95: 86-94, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32445665

RESUMEN

Valproic acid (VPA), an antiepileptic and mood-stabilizing drug, is prescribed to women of reproductive age. VPA is associated with a 1-2% increase in neural tube defects in offspring following gestational exposure and results in epigenetic modifications induced by perturbations in transcription cofactors. Cbp and p300, two transcription cofactors, play key roles in embryonic neural development. p300 is a downstream target of Akt, a protein kinase B associated with cell survival and anti-apoptotic mechanisms, as part of the Akt-p300 axis. We examined the effects of in utero VPA exposure on Cbp, p300, and Akt in gestational day (GD)9, GD10 and GD13 CD-1 mouse embryos following a teratogenic maternal dose of 400 mg/kg. Embryos were collected at 0, 1, 3 and 6 h post-dosing on GD9, 24 h post-dosing on GD10 and on GD13. GD10 embryos were grouped according to the status of neural tube closure in control, closed and open groups. GD13 heads were grouped as control, exposed but non-exencephalic and exencephalic. Our data indicate that Cbp, p300 and Akt mRNA levels were downregulated at 1 and 3 h post-exposure in GD9 embryos while Cbp and p300 protein levels remained stable. Akt protein levels were significantly increased 1 h post-exposure. No significant changes were observed in either mRNA or protein expression in embryos with closed or open neural tubes compared to the control group at GD10. Downregulated expression of Cbp, p300, and Akt may play a key role in VPA-induced neural tube defects considering their vitally important role in embryonic development.


Asunto(s)
Anomalías Inducidas por Medicamentos/metabolismo , Proteína de Unión a CREB , Proteína p300 Asociada a E1A , Defectos del Tubo Neural/metabolismo , Proteínas Proto-Oncogénicas c-akt , Ácido Valproico , Anomalías Inducidas por Medicamentos/genética , Animales , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Regulación hacia Abajo , Proteína p300 Asociada a E1A/genética , Proteína p300 Asociada a E1A/metabolismo , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Ratones , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo
6.
Cold Spring Harb Protoc ; 2020(11)2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32404313

RESUMEN

Failure to predict drug-induced toxicity reactions is a major problem contributing to a high attrition rate and tremendous cost in drug development. Drug screening in X. laevis embryos is high-throughput relative to screening in rodents, potentially making them ideal for this use. Xenopus embryos have been used as a toxicity model in the frog embryo teratogenesis assay on Xenopus (FETAX) for the early stages of drug safety evaluation. We previously developed compound-screening methods using Xenopus embryos and believe they could be used for in vitro drug-induced toxicity safety assessment before expensive preclinical trials in mammals. Specifically, Xenopus embryos could help predict drug-induced hepatotoxicity and consequently aid lead candidate prioritization. Here we present methods, which we have modified for use on Xenopus embryos, to help measure the potential for a drug to induce liver toxicity. One such method examines the release of the liver-specific microRNA (miRNA) miR-122 from the liver into the vasculature as a result of hepatocellular damage, which could be due to drug-induced acute liver injury. Paracetamol, a known hepatotoxin at high doses, can be used as a positive control. We previously showed that some of the phenotypes of mammalian paracetamol overdose are reflected in Xenopus embryos. Consequently, we have also included here a method that measures the concentration of free glutathione (GSH), which is an indicator of paracetamol-induced liver injury. These methods can be used as part of a panel of protocols to help predict the hepatoxicity of a drug at an early stage in drug development.


Asunto(s)
Anomalías Inducidas por Medicamentos/diagnóstico , Bioensayo/métodos , Enfermedad Hepática Inducida por Sustancias y Drogas/diagnóstico , Embrión no Mamífero/efectos de los fármacos , Xenopus laevis/embriología , Anomalías Inducidas por Medicamentos/genética , Anomalías Inducidas por Medicamentos/metabolismo , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Embrión no Mamífero/metabolismo , Hígado/efectos de los fármacos , Hígado/embriología , Hígado/metabolismo , MicroARNs/genética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Pruebas de Toxicidad/métodos , Xenopus laevis/genética , Xenopus laevis/metabolismo
7.
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
8.
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
9.
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
10.
Skelet Muscle ; 7(1): 8, 2017 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-28526071

RESUMEN

BACKGROUND: We previously reported that Wnt5a/CaMKIIδ (calcium/calmodulin-dependent protein kinase II delta) pathway was involved in the embryonic tongue deformity induced by excess retinoic acid (RA). Our latest study found that the expression of miR-31-5p, which was predicted to target the 3'UTR of CamkIIδ, was raised in the RA-treated embryonic tongue. Thus, we hypothesized that the excess RA regulated Wnt5a/CaMKIIδ pathway through miR-31-5p in embryonic tongue. METHODS: C2C12 myoblast line was employed as an in vitro model to examine the suppression of miR-31-5p on CamkIIδ expression, through which RA impaired the myoblast proliferation and differentiation in embryonic tongue. RESULTS: RA stimulated the expression of miR-31-5p in both embryonic tongue and C2C12 myoblasts. Luciferase reporter assay confirmed that the 3'UTR of CamkIIδ was a target of miR-31-5p. MiR-31-5p mimics disrupted CamkIIδ expression, C2C12 proliferation and differentiation as excess RA did, while miR-31-5p inhibitor partially rescued these defects in the presence of RA. CONCLUSIONS: Excess RA can stimulate miR-31-5p expression to suppress CamkIIδ, which represses the proliferation and differentiation of tongue myoblasts.


Asunto(s)
Anomalías Inducidas por Medicamentos/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , MicroARNs/genética , Desarrollo de Músculos , Lengua/anomalías , Tretinoina/farmacología , Regiones no Traducidas 3' , Anomalías Inducidas por Medicamentos/genética , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Diferenciación Celular , Proliferación Celular , Células HEK293 , Humanos , Ratones , MicroARNs/metabolismo , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Mioblastos/fisiología , Lengua/embriología , Tretinoina/toxicidad
11.
Curr Mol Med ; 17(2): 108-117, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28429672

RESUMEN

Thalidomide is a teratogen that affects many organs but primarily induces limb truncations like phocomelia. Rodents are thalidomide resistant. In the 1950s, this has led to misinterpretations of animal tests and to the fatal assumption that the drug was safe for pregnant women to use against morning sickness. The result was one of the biggest scandals in medical history: 10.000 and more infants with birth defects in Europe. Nonetheless, thalidomide still has its place in modern medicine as it has strong therapeutic potential: it has been approved by the FDA for multiple myeloma and erythema nodosum leprosum, and its anti-inflammatory, immunomodulatory and antiangiogenic activities are considered in many other refractory diseases. The aim is to develop derivatives that are not teratogenic but maintain the therapeutic potential. This requires detailed knowledge about the underlying molecular mechanisms. Much progress has been made in deciphering the teratogenic mechanisms in the last decade. Here, we summarize these mechanisms, explain thalidomide resistance of rodents, and discuss possible mechanisms that could explain why the drug primarily targets the developing limb in the embryo. We also summarize the most important therapeutic mechanisms. Finally, we discuss which therapeutic and teratogenic mechanisms do and do not overlap, and if there is a chance for the development of non-teratogenic thalidomide derivatives with therapeutic potential.


Asunto(s)
Anomalías Inducidas por Medicamentos/etiología , Anomalías Inducidas por Medicamentos/metabolismo , Medicina , Teratógenos/toxicidad , Talidomida/efectos adversos , Anomalías Inducidas por Medicamentos/diagnóstico , Animales , Biomarcadores , Femenino , Regulación de la Expresión Génica , Humanos , Deformidades Congénitas de las Extremidades , Medicina/tendencias , Neovascularización Patológica/etiología , Neovascularización Patológica/metabolismo , Estrés Oxidativo , Fenotipo , Embarazo , Transducción de Señal
12.
Birth Defects Res ; 109(6): 452-459, 2017 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-28398707

RESUMEN

BACKGROUND: The objective was to study pregnancy outcomes between groups of Danish women, with pregnancy ending between 1998 and 2009, according to their exposure to montelukast. METHODS: Cross-sectional observational study in Danish women, selecting live births and stillbirths (Birth Registry) and spontaneous abortions and induced terminations (Patient Registry). Montelukast exposure was obtained from the Prescription Registry (ATC code R03DC03). Exposure period was from 3 months before the last menstrual period until the end of the first trimester. Four groups were studied: (1) women with prescription for montelukast, (2) women with prescription for montelukast and other anti-asthmatic medications, (3) women with prescription for other anti-asthmatic medications, (4) women without prescription for any anti-asthmatic medications. RESULTS: A total of 754,300 singleton pregnancies (> 12 weeks) were identified: 401 pregnancies in group 1, 426 pregnancies in group 2, 24878 in group 3 and 728,595 in group 4. Risk of preterm birth, maternal preeclampsia and gestational diabetes was increased for pregnancies exposed to montelukast. No significant differences were found for the risk of major congenital anomalies (CA). Adjusted odds ratio for CA was 1.4 (95% CI 0.9-2.3) for the group 1 and 1.0 (95% CI 0.6-1.8) for group 2. CONCLUSION: Pregnant women with prescriptions for montelukast had a higher risk of preterm birth and maternal complications. These risks are known to be associated with maternal asthma, no increased risk of CA was found. Further analysis including more exposed pregnancies will be needed to determine the risk of specific CA. Birth Defects Research 109:452-459, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Acetatos/efectos adversos , Quinolinas/efectos adversos , Anomalías Inducidas por Medicamentos/metabolismo , Antiasmáticos/uso terapéutico , Asma/tratamiento farmacológico , Anomalías Congénitas/etiología , Estudios Transversales , Ciclopropanos , Dinamarca/epidemiología , Femenino , Humanos , Oportunidad Relativa , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico , Resultado del Embarazo/epidemiología , Primer Trimestre del Embarazo , Efectos Tardíos de la Exposición Prenatal/epidemiología , Sistema de Registros , Sulfuros
13.
Seizure ; 46: 7-12, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28212902

RESUMEN

PURPOSE: Valproate is one of the most commonly used anticonvulsive drugs. Despite its significant benefits, the teratogenicity of valproate is a relevant problem in the treatment of women of childbearing age. In addition to major congenital malformations, such as neural tube defects, reduced intelligence and attention after intrauterine valproate exposure are reported. Until now the mechanisms of teratogenicity of VPA are poorly understood and concepts how to reduce valproate teratogenicity are lacking. METHODS: In a rat model of valproate teratogenicity we examined hippocampal cell structure in 4 week old animals with a stereological approach. As potential mechanisms of VPA teratogenicity we examined histone acetylation by western blotting and metabolites of the folate metabolism as well as global DNA methylation by tandem mass spectrometry in the brain and liver tissue of newborn pups (p0). RESULTS: We found an increase in the number of neurons in the hippocampal areas CA1/2 (p=0.018) and CA3 (p=0.022), as well as a decreased number of astrocytes in CA1/2 (p=0.004) and CA3 (p=0.003) after intrauterine VPA exposure, as a possible indication of altered cell differentiation during intrauterine VPA exposure. Valproate exposure was also associated with an increase in 5-methyl-tetrahydrofolate (THF) (p=0.002) and a decrease in 5-10-methenyl-THF in the brain of newborn pups, as well as a reduced homocysteine plasma level (p<0.001). The described changes in hippocampal cell numbers and folate metabolism were only significant after high-dose intrauterine VPA exposure indicating a dose-dependent effect. VPA exposure was not associated with changes in histone acetylation or global DNA methylation in brain tissue in newborn pups. CONCLUSION: This study shows that intrauterine VPA exposure is associated with changes in hippocampal cell numbers in the CA1/2 and CA3 region and in folate metabolism.


Asunto(s)
Anomalías Inducidas por Medicamentos/metabolismo , Anomalías Inducidas por Medicamentos/patología , Ácido Fólico/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/patología , Ácido Valproico/toxicidad , Animales , Anticonvulsivantes/toxicidad , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Recuento de Células , Metilación de ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Hipocampo/metabolismo , Homocisteína/sangre , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Neurogénesis/efectos de los fármacos , Neurogénesis/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Tamaño de los Órganos , Ratas Wistar
14.
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
15.
Drug Res (Stuttg) ; 66(5): 246-50, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26676852

RESUMEN

Prenatal opioid exposure such as oxycodone is linked to significant adverse effects on the developing brain. Endothelin (ET) and its receptors are involved in normal development of the central nervous system. Opioid tolerance and withdrawal are mediated through ET receptors. It is possible that adverse effect of oxycodone on the developing brain is mediated through ET receptors. We evaluated brain ETA and ETB receptor expression during postnatal development in rats with prenatal oxycodone exposure. Timed pregnant Sprague-Dawley rats received either oxycodone or placebo throughout gestation. After birth, male rat pups were sacrificed on postnatal day (PND) 1, 7, 14 or 28. Brain ETA and ETB receptor expression was determined by Western blot analysis. Oxycodone pups compared to placebo demonstrated congenital malformations of the face, mouth, and vertebrae at the time of birth [4/69 (5.7%) vs. 0/60 (0%); respectively] and intrauterine growth retardation [10/69 (15%) vs. 2/60 (3.3%); respectively]. On PND 28, oxycodone pups compared to placebo had lower body and kidney weight. ETA receptor expression in the oxycodone group was significantly higher compared to placebo on PND 1 (p=0.035), but was similar on PND 7, 14, or 28. ETB receptor expression decreased in oxycodone compared to placebo on PND 1 and 7 (p=0.001); and increased on PND 28 (p=0.002), but was similar on PND 14. Oxycodone-exposed rat pups had lower birth weight and postnatal weight gain and greater congenital malformations. ETB receptor expression is altered in the brain of oxycodone-treated rat pups indicating a possible delay in CNS development.


Asunto(s)
Anomalías Inducidas por Medicamentos/metabolismo , Analgésicos Opioides/efectos adversos , Encéfalo/efectos de los fármacos , Oxicodona/efectos adversos , Receptores de Endotelina/metabolismo , Analgésicos Opioides/administración & dosificación , Animales , Animales Recién Nacidos , Western Blotting , Peso Corporal/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Femenino , Masculino , Oxicodona/administración & dosificación , Embarazo , Ratas , Ratas Sprague-Dawley , Síndrome de Abstinencia a Sustancias/metabolismo
16.
Drug Saf ; 38(7): 651-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26017034

RESUMEN

INTRODUCTION: Drug use in pregnancy is very common but may cause harm to the fetus. The teratogenic effect of a drug is partly dependent on the drug level in the fetal circulation, which is associated with the transport across the placenta. Many drugs are substrates of P-glycoprotein (P-gp), an efflux transporter that acts as a protective barrier for the fetus. We aim to identify whether drug interactions associated with P-gp promote any changes in fetal drug exposure, as measured by the risk of having children with congenital anomalies. METHODS: In this study, cases (N = 4634) were mothers of children with congenital anomalies registered in the EUROCAT Northern Netherlands registry, and the reference population were mothers of children (N = 25,126) from a drug prescription database (IADB.nl). RESULTS: Drugs that are associated with P-gp transport were commonly used in pregnancy in cases (10 %) and population (12 %). Several drug classes, which are substrates for P-gp, were shown to have a higher user rate in mothers of cases with specific anomalies. The use of this subset of drugs in combination with other P-gp substrates increased the risk for specific anomalies (odds ratio [OR] 4.17, 95 % CI 1.75-9.91), and the addition of inhibitors further increased the risk (OR 13.03, 95 % CI 3.37-50.42). The same pattern of risk increment was observed when the drugs were analyzed separately according to substrate specificity. CONCLUSIONS: The use of drugs associated with P-gp transport was common during pregnancy. For several drug classes associated with specific anomalies, P-gp-mediated drug interactions are associated with an increased risk for those specific anomalies.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Anomalías Inducidas por Medicamentos/etiología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/etiología , Exposición Materna/efectos adversos , Placenta/metabolismo , Medicamentos bajo Prescripción/efectos adversos , Anomalías Inducidas por Medicamentos/diagnóstico , Anomalías Inducidas por Medicamentos/metabolismo , Transporte Biológico , Estudios de Casos y Controles , Distribución de Chi-Cuadrado , Interacciones Farmacológicas , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/diagnóstico , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/metabolismo , Femenino , Humanos , Masculino , Países Bajos , Oportunidad Relativa , Embarazo , Medicamentos bajo Prescripción/clasificación , Sistema de Registros , Medición de Riesgo , Factores de Riesgo
17.
Circ Arrhythm Electrophysiol ; 8(2): 420-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25648353

RESUMEN

BACKGROUND: N629D KCNH2 is a human missense long-QT2 mutation. Previously, we reported that the N629D/N629D mutation embryos disrupted cardiac looping, right ventricle development, and ablated IKr activity at E9.5. The present study evaluates the role of KCNH2 in vasculogenesis. METHODS AND RESULTS: N629D/N629D yolk sac vessels and aorta consist of sinusoids without normal arborization. Isolated E9.5 +/+ first branchial arches showed normal outgrowth of mouse ERG-positive/α-smooth muscle actin coimmunolocalized cells; however, outgrowth was grossly reduced in N629D/N629D. N629D/N629D aortas showed fewer α-smooth muscle actin positive cells that were not coimmunolocalized with mouse ERG cells. Transforming growth factor-ß treatment of isolated N629D/N629D embryoid bodies partially rescued this phenotype. Cultured N629D/N629D embryos recapitulate the same cardiovascular phenotypes as seen in vivo. Transforming growth factor-ß treatment significantly rescued these embryonic phenotypes. Both in vivo and in vitro, dofetilide treatment, over a narrow window of time, entirely recapitulated the N629D/N629D fetal phenotypes. Exogenous transforming growth factor-ß treatment also rescued the dofetilide-induced phenotype toward normal. CONCLUSIONS: Loss of function of KCNH2 mutations results in defects in cardiogenesis and vasculogenesis. Because many medications inadvertently block the KCNH2 potassium current, these novel findings seem to have clinical relevance.


Asunto(s)
Anomalías Inducidas por Medicamentos/prevención & control , Células Madre Embrionarias/efectos de los fármacos , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/genética , Muerte Fetal , Cardiopatías Congénitas/prevención & control , Mutación Missense , Neovascularización Fisiológica/efectos de los fármacos , Fenetilaminas/toxicidad , Bloqueadores de los Canales de Potasio/toxicidad , Sulfonamidas/toxicidad , Factor de Crecimiento Transformador beta/farmacología , Malformaciones Vasculares/prevención & control , Anomalías Inducidas por Medicamentos/embriología , Anomalías Inducidas por Medicamentos/genética , Anomalías Inducidas por Medicamentos/metabolismo , Animales , Células Cultivadas , Canal de Potasio ERG1 , Técnicas de Cultivo de Embriones , Células Madre Embrionarias/metabolismo , Canales de Potasio Éter-A-Go-Go/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genotipo , Cardiopatías Congénitas/inducido químicamente , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Humanos , Ratones de la Cepa 129 , Ratones Transgénicos , Morfogénesis/efectos de los fármacos , Fenotipo , Transducción de Señal , Malformaciones Vasculares/inducido químicamente , Malformaciones Vasculares/embriología , Malformaciones Vasculares/genética , Malformaciones Vasculares/metabolismo
18.
Toxicol Sci ; 144(1): 90-104, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25490952

RESUMEN

Although Cadmium (Cd) is a well-known heavy metal pollutant and teratogen, the mechanism behind Cd-mediated teratogenicity remains unknown. Previously, we have reported of the protective role of Nitric oxide (NO), a key signaling molecule in the embryonic developmental process, against Thalidomide-induced teratogenicity. The objective of this study was to obtain a mechanistic in-sight of the antiteratogenic potential of NO against Cd-mediated teratogenicity. To achieve this goal, we first studied the effect of Cd on the vasculature of developing embryos and then we investigated whether Cd mediated its effects by interfering with the redox regulation of NO signaling in the early development milieu. We used a chick embryonic model to determine the time and dose-dependent effects of Cd and NO recovery against Cd assault. The effects of Cd and NO recovery were assessed using various angiogenic assays. Redox and NO levels were also measured. Results demonstrated that exposure to Cd at early stage of development caused multiple birth defects in the chick embryos. Exposure to Cd suppressed endogenous NO levels and cGMP signaling, inhibiting angioblast activation and subsequently impairing yolk sac vascular development. Furthermore, Cd-induced superoxide and lipid peroxidation mediated activation of proapoptotic markers p21 and p53 in the developing embryo. Cd also caused the down-regulation of FOXO1, and up-regulation of FOXO3a and Caspase 3-mediated apoptosis. Addition of exogenous NO through a NO donor was able to blunt Cd-mediated effects and restore normal vascular and embryonic development. In conclusion, Cd-mediated teratogenicity occurs as a result of impaired NO-cGMP signaling, increased oxidative stress, and the activation of apoptotic pathways. Subsequent addition of exogenous NO through NO donor negated Cd-mediated effects and protected the developing embryo.


Asunto(s)
Anomalías Inducidas por Medicamentos/prevención & control , Cloruro de Cadmio/toxicidad , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico/metabolismo , Teratógenos/toxicidad , Anomalías Inducidas por Medicamentos/etiología , Anomalías Inducidas por Medicamentos/genética , Anomalías Inducidas por Medicamentos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Células Cultivadas , Embrión de Pollo , GMP Cíclico/metabolismo , Citoprotección , Relación Dosis-Respuesta a Droga , Regulación del Desarrollo de la Expresión Génica , Peroxidación de Lípido/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
19.
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
20.
PLoS One ; 9(5): e96010, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24816763

RESUMEN

AIM: To characterize the abnormal metabolic profile of all-trans-retinoic acid (ATRA)-induced craniofacial development in mouse embryos using proton magnetic resonance spectroscopy (1H-MRS). METHODS: Timed-pregnant mice were treated by oral gavage on the morning of embryonic gestation day 11 (E11) with all-trans-retinoic acid (ATRA). Dosing solutions were adjusted by maternal body weight to provide 30, 70, or 100 mg/kg RA. The control group was given an equivalent volume of the carrier alone. Using an Agilent 7.0 T MR system and a combination of surface coil coils, a 3 mm×3 mm×3 mm 1H-MRS voxel was selected along the embryonic craniofacial tissue. 1H-MRS was performed with a single-voxel method using PRESS sequence and analyzed using LCModel software. Hematoxylin and eosin was used to detect and confirm cleft palate. RESULT: 1H-MRS revealed elevated choline levels in embryonic craniofacial tissue in the RA70 and RA100 groups compared to controls (P<0.05). Increased choline levels were also found in the RA70 and RA100 groups compared with the RA30 group (P<0.01). High intra-myocellular lipids at 1.30 ppm (IMCL13) in the RA100 group compared to the RA30 group were found (P<0.01). There were no significant changes in taurine, intra-myocellular lipids at 2.10 ppm (IMCL21), and extra-myocellular lipids at 2.30 ppm (EMCL23). Cleft palate formation was observed in all fetuses carried by mice administered 70 and 100 mg/kg RA. CONCLUSIONS: This novel study suggests that the elevated choline and lipid levels found by 1H-MRS may represent early biomarkers of craniofacial defects. Further studies will determine performance of this test and pathogenetic mechanisms of craniofacial malformation.


Asunto(s)
Anomalías Inducidas por Medicamentos/metabolismo , Anomalías Craneofaciales/metabolismo , Embrión de Mamíferos/metabolismo , Espectroscopía de Protones por Resonancia Magnética/métodos , Tretinoina/toxicidad , Anomalías Inducidas por Medicamentos/embriología , Anomalías Inducidas por Medicamentos/etiología , Animales , Antineoplásicos/toxicidad , Colina/metabolismo , Fisura del Paladar/inducido químicamente , Fisura del Paladar/embriología , Fisura del Paladar/metabolismo , Anomalías Craneofaciales/inducido químicamente , Anomalías Craneofaciales/embriología , Creatina/metabolismo , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/efectos de los fármacos , Femenino , Lípidos/análisis , Masculino , Ratones , Embarazo , Taurina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...