Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Commun ; 12(1): 3447, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34103494

RESUMEN

Congenital heart disease (CHD) is the most common class of human birth defects, with a prevalence of 0.9% of births. However, two-thirds of cases have an unknown cause, and many of these are thought to be caused by in utero exposure to environmental teratogens. Here we identify a potential teratogen causing CHD in mice: maternal iron deficiency (ID). We show that maternal ID in mice causes severe cardiovascular defects in the offspring. These defects likely arise from increased retinoic acid signalling in ID embryos. The defects can be prevented by iron administration in early pregnancy. It has also been proposed that teratogen exposure may potentiate the effects of genetic predisposition to CHD through gene-environment interaction. Here we show that maternal ID increases the severity of heart and craniofacial defects in a mouse model of Down syndrome. It will be important to understand if the effects of maternal ID seen here in mice may have clinical implications for women.


Asunto(s)
Sistema Cardiovascular/embriología , Embrión de Mamíferos/patología , Deficiencias de Hierro , Animales , Aorta Torácica/anomalías , Biomarcadores/metabolismo , Diferenciación Celular , Vasos Coronarios/embriología , Vasos Coronarios/patología , Suplementos Dietéticos , Edema/patología , Embrión de Mamíferos/anomalías , Desarrollo Embrionario , Femenino , Perfilación de la Expresión Génica , Interacción Gen-Ambiente , Proteínas Fluorescentes Verdes/metabolismo , Hierro/metabolismo , Vasos Linfáticos/embriología , Vasos Linfáticos/patología , Ratones Endogámicos C57BL , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Penetrancia , Fenotipo , Embarazo , Transducción de Señal , Células Madre/patología , Transgenes , Tretinoina/metabolismo
2.
J Ethnopharmacol ; 239: 111895, 2019 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-31014960

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Chuanxiong (Chuanxiong Rhizoma, CR), the dried rhizome of Ligusticum chuanxiong Hort, has been used during pregnancy for more than 2000 years. However, the embryotoxicity of CR was not evaluated so far. The purpose of this study was to examine the safety and rational use of CR during pregnancy on mice and mouse embryonic stem cell (ES), and to explore the mechanism of embryotoxicity. AIM OF THE STUDY: This study was carried out to evaluate embryotoxicity of CR decoction in vivo and in vitro, and to explore the mechanism of embryotoxicity from the perspective of bone metabolism. MATERIALS AND METHODS: In animal experiments, pregnant mice were randomly assigned into 5 groups, i.e. mice were orally treated with CR decoction at dosages of 0 (distilled water, as negative controls), 2, 8, 32 g/kg/d (low, medium and high-dose group), and vitamin A (as positive controls), respectively. Maternal and embryo-fetal parameters were registered after cesarean section. The fetal skeletal development was further assessed with the alizarin red S and Hematoxylin-Eosin staining (H&E staining) and fluorescent imaging. Meanwhile, the mouse embryonic stem cell test model (EST model) was established to objectively evaluate the toxicity of CR on the embryo development. The median inhibitory proliferation values (IC50) for both the mouse embryonic stem cell D3 (ES) and mouse embryonic fibroblast 3T3 (3T3) were detected with MTT assays. After removal of inhibiting factor (LIF), mouse embryonic stem cells spontaneously differentiated into cardiomyocytes, the expression of specific myosin heavy chain gene (ß-MHC) contained in cardiomyocytes were detected by q-PCR quantitative analysis, and median inhibitory differentiation concentration (ID50) of ES was obtained. The development toxicity calculation formula was used to determine the embryotoxicity grade of CR decoction. finally, based on the successful induction of osteoblasts, the molecular mechanism of CR embryotoxicity was preliminarily studied based on BMP-Smads signal pathway. RESULTS: Compared with the negative control group, high, medium, and low doses of CR decoction had no significant effect on the maternal body weight and uterine weight (P > 0.05), as well as on the maternal liver, heart, and kidneys. The observation results showed that high dose of CR decoction significantly increase the number of absorbed fetuses (P < 0.05). The EST model was successfully established, the IC50 3T3, IC50 ES and ID50 ES of CR were 9.39 mg/mL, 18.78 mg/mL, and 10.20 mg/mL, respectively. CR was classified as weak embryonic development toxicity by the EST linear discriminant formula. Meanwhile, osteoblasts were successfully induced in vitro, the relative expression levels of BMP2, BMPR2, Smad1, and Smad5 were down-regulated in varying degrees after 3, 6, and 9 days of treatment with different concentration gradients of CR decoction. CONCLUSIONS: Combining in vivo and in vitro experiments, CR showed a potential embryotoxicity. The mechanism of embryotoxicity may be related to inhibiting the expression of key genes in the BMP-SMADs signaling pathway. In the clinical application, the normal dosage of CR is safe to a certain extent. However, when the dosage is too high (160 g/60 kg/d), there may be a risk of embryotoxicity.


Asunto(s)
Embrión de Mamíferos/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Ligusticum , Osteoblastos/efectos de los fármacos , Extractos Vegetales/toxicidad , Animales , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Células Cultivadas , Embrión de Mamíferos/anomalías , Femenino , Reabsorción del Feto/inducido químicamente , Ratones , Osteoblastos/metabolismo , Embarazo , Rizoma , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo , Esternón/anomalías , Esternón/efectos de los fármacos
3.
Hum Reprod ; 34(5): 851-862, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30989206

RESUMEN

STUDY QUESTION: Could clinically-relevant moderate and/or high dose maternal folic acid supplementation prevent aberrant developmental and epigenetic outcomes associated with assisted reproductive technologies (ART)? SUMMARY ANSWER: Our results demonstrate dose-dependent and sex-specific effects of folic acid supplementation in ART and provide evidence that moderate dose supplements may be optimal for both sexes. WHAT IS KNOWN ALREADY: Children conceived using ART are at an increased risk for growth and genomic imprinting disorders, often associated with DNA methylation defects. Folic acid supplementation is recommended during pregnancy to prevent adverse offspring outcomes; however, the effects of folic acid supplementation in ART remain unclear. STUDY DESIGN, SIZE, DURATION: Outbred female mice were fed three folic acid-supplemented diets, control (rodent daily recommended intake or DRI; CD), moderate (4-fold DRI; 4FASD) or high (10-fold DRI; 10FASD) dose, for six weeks prior to ART and throughout gestation. Mouse ART involved a combination of superovulation, in vitro fertilisation, embryo culture and embryo transfer. PARTICIPANTS/MATERIALS, SETTING, METHODS: Midgestation embryos and placentas (n = 74-99/group) were collected; embryos were assessed for developmental delay and gross morphological abnormalities and embryos and placentas were examined for epigenetic defects. We assessed methylation at four imprinted genes (Snrpn, Kcnq1ot1, Peg1 and H19) in matched midgestation embryos and placentas (n = 31-32/group) using bisulfite pyrosequencing. In addition, we examined genome-wide DNA methylation patterns in placentas (n = 6 normal placentas per sex/group) and embryos (n = 6 normal female embryos/group; n = 3 delayed female embryos/group) using reduced representation bisulfite sequencing (RRBS). MAIN RESULTS AND THE ROLE OF CHANCE: Moderate, but not high dose supplementation, was associated with a decrease in the proportion of developmentally delayed embryos. Although moderate dose folic acid supplementation reduced DNA methylation variance at certain imprinted genes in embryonic and placental tissues, high dose supplementation exacerbated the negative effects of ART at imprinted loci. Furthermore, folic acid supplements resolved female-biased aberrant imprinted gene methylation. Supplementation was more effective at correcting ART-induced genome-wide methylation defects in male versus female placentas; however, folic acid supplementation also led to additional methylation perturbations which were more pronounced in males. LARGE-SCALE DATA: The RRBS data from this study have been submitted to the NCBI Gene Expression Omnibus under the accession number GSE123143. LIMITATIONS REASONS FOR CAUTION: Although the combination of mouse ART utilised in this study consisted of techniques commonly used in human fertility clinics, there may be species differences. Therefore, human studies, designed to determine the optimal levels of folic acid supplementation for ART pregnancies, and taking into account foetal sex, are warranted. WIDER IMPLICATIONS OF THE FINDINGS: Taken together, our findings support moderation in the dose of folic acid supplements taken during ART. STUDY FUNDING/COMPETING INTEREST(S): This work was funded by the Canadian Institutes of Health Research (FDN-148425). The authors declare no conflict of interest.


Asunto(s)
Anomalías Congénitas/prevención & control , Suplementos Dietéticos , Ácido Fólico/administración & dosificación , Impresión Genómica/efectos de los fármacos , Técnicas Reproductivas Asistidas/efectos adversos , Administración Oral , Animales , Anomalías Congénitas/genética , Metilación de ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/efectos de los fármacos , Femenino , Sitios Genéticos/efectos de los fármacos , Humanos , Masculino , Ratones , Embarazo
4.
Development ; 142(17): 2981-95, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26329601

RESUMEN

Yin Yang 1 (YY1) is a multifunctional zinc-finger-containing transcription factor that plays crucial roles in numerous biological processes by selectively activating or repressing transcription, depending upon promoter contextual differences and specific protein interactions. In mice, Yy1 null mutants die early in gestation whereas Yy1 hypomorphs die at birth from lung defects. We studied how the epithelial-specific inactivation of Yy1 impacts on lung development. The Yy1 mutation in lung epithelium resulted in neonatal death due to respiratory failure. It impaired tracheal cartilage formation, altered cell differentiation, abrogated lung branching and caused airway dilation similar to that seen in human congenital cystic lung diseases. The cystic lung phenotype in Yy1 mutants can be partly explained by the reduced expression of Shh, a transcriptional target of YY1, in lung endoderm, and the subsequent derepression of mesenchymal Fgf10 expression. Accordingly, SHH supplementation partially rescued the lung phenotype in vitro. Analysis of human lung tissues revealed decreased YY1 expression in children with pleuropulmonary blastoma (PPB), a rare pediatric lung tumor arising during fetal development and associated with DICER1 mutations. No evidence for a potential genetic interplay between murine Dicer and Yy1 genes during lung morphogenesis was observed. However, the cystic lung phenotype resulting from the epithelial inactivation of Dicer function mimics the Yy1 lung malformations with similar changes in Shh and Fgf10 expression. Together, our data demonstrate the crucial requirement for YY1 in lung morphogenesis and identify Yy1 mutant mice as a potential model for studying the genetic basis of PPB.


Asunto(s)
Epitelio/embriología , Epitelio/metabolismo , Pulmón/embriología , Pulmón/metabolismo , Morfogénesis , Factor de Transcripción YY1/metabolismo , Animales , Apoptosis , Tipificación del Cuerpo , Cartílago/anomalías , Cartílago/embriología , Cartílago/patología , Diferenciación Celular , Proliferación Celular , ARN Helicasas DEAD-box/metabolismo , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/patología , Endodermo/embriología , Endodermo/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Enfermedades Pulmonares/congénito , Enfermedades Pulmonares/patología , Ratones , Ratones Transgénicos , Modelos Biológicos , Miocitos del Músculo Liso/metabolismo , Miofibroblastos/patología , Fenotipo , Blastoma Pulmonar/metabolismo , Blastoma Pulmonar/patología , Ribonucleasa III/metabolismo , Tráquea/anomalías , Tráquea/embriología , Tráquea/patología
5.
Int J Med Mushrooms ; 16(6): 519-28, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25404217

RESUMEN

In this study, we investigated the reproductive capacity of pregnant rats exposed to daily orally administered powder-dehydrated reconstituted of Agaricus brasiliensis (=Agaricus blazei sensu Murrill), the fetal organogenesis, and the development of the pups. Pregnant rats were exposed for the entire gestational period to water (control) and A. brasiliensis at 300 or 600 mg/kg/day. Fertility and body weight of dams were monitored. Pups were monitored for body weight, offspring vitality, morphology, and physical and neurobehavioral development. An increase in sternebrae agenesis was observed at the 600 mg/kg/day dose of A. brasiliensis, while incomplete ossification of sternebrae was seen even at a 300 mg/kg/day dose. In conclusion, this study is the first to demonstrate the impact of maternal exposure to A. brasiliensis on the fetal organogenesis and development of offspring in a rat model. The 600 mg/kg/day dose showed some negative effects, and low toxicity was observed at the 300 mg/kg/day dose.


Asunto(s)
Basidiomycota , Embrión de Mamíferos/anomalías , Animales , Femenino , Inocuidad de los Alimentos , Masculino , Embarazo , Ratas , Ratas Wistar , Esternón/anomalías
6.
J Ethnopharmacol ; 151(1): 114-22, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24184264

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Dipsaci Radix (DR), the dried root of Dipsacus asper Wall. ex Henry, has been used to treat pregnant disorders for thousands of years, and currently has been ranked as the first selective herb for prevention of miscarriage clinically; however, there is no sufficient evidence so far to assess its safety. The purpose of this study was to examine the impacts of DR aqueous extracts on embryonic development with mice and embryonic stem cells (ESCs). MATERIAL AND METHOD: In a segment II study, pregnant ICR mice were randomly assigned into 5 groups, i.e. mice were orally treated with DR aqueous extracts at dosages of 0 (distilled water, as negative controls (G1 group)), 2, 8, 32 g/kg/d (G2, G3, G4 group), and vitamin A (as positive controls (G5 group)) respectively. Maternal and embryo-fetal parameters were evaluated after cesarean section. The fetal skeletal development was further assessed with the alizarin red S and H&E staining and fluorescent imaging. Meanwhile, IC50 values for both ESCs and 3T3 cells were detected with MTT assays. RESULT: Compared to G1 group, the maternal body-weight in G3 and G4 groups was significantly lower (P<0.05-0.001), and the fetal malformation rate increased in G2, G3 and G4 groups as a dose-dependent manner, although a statistical significance was only reached in G4 group (P<0.001). The morphologic and histochemistry abnormalities of fetal skeletal development such as delayed osteogenesis and mineralization in the cartilaginous tissue were found after DR treatments (32 g/kg/d). There was no significant difference between IC50 ESC (6.826 ± 0.311 mg/ml) and IC50 3T3 (5.132 ± 0.142 mg/ml, P>0.05). CONCLUSION: DR aqueous extracts at the dosage of 8 or 32 g/kg/d (4.3 or 17.2 folds of recommended daily-dosage for adult human respectively) might cause adverse impacts in maternal healthy and embryo-fetal development. It suggests that high-dose and long-term administration of DR preparations should be unsafe to pregnant women.


Asunto(s)
Dipsacaceae/química , Desarrollo Embrionario/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Desarrollo Fetal/efectos de los fármacos , Extractos Vegetales/toxicidad , Raíces de Plantas/química , Animales , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/efectos de los fármacos , Femenino , Feto/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Extractos Vegetales/química , Embarazo , Distribución Aleatoria
7.
Reprod Toxicol ; 41: 57-66, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23811354

RESUMEN

Transcriptomic evaluations may improve toxicity prediction of in vitro-based developmental models. In this study, transcriptomics was used to identify VPA-induced gene expression changes in rat whole embryo culture (WEC). Furthermore, VPA-induced responses were compared across in vitro-based developmental models, such as the cardiac and neural embryonic stem cells (ESTc and ESTn, respectively) and the zebrafish embryotoxicity model. VPA-induced gene regulation in WEC corresponded with observed morphological effects and previously suggested mechanisms of toxicity. Gene Ontology term-directed analysis showed conservation of VPA-induced gene expression changes across in vitro-based developmental models, with ESTc and ESTn exhibiting complementary responses. Furthermore, comparison of in vitro-based developmental and non-developmental models revealed that more generalized VPA-induced effects can be detected using non-developmental models whereas developmental models provide added value when assessing developmental-specific effects. These analyses can be used to optimize test batteries for the detection of developmental toxicants in vitro.


Asunto(s)
Embrión de Mamíferos/efectos de los fármacos , Embrión no Mamífero/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Teratógenos/toxicidad , Ácido Valproico/toxicidad , Animales , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/metabolismo , Embrión no Mamífero/metabolismo , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Ratas Wistar , Pez Cebra
8.
Mech Dev ; 129(1-4): 1-12, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22522080

RESUMEN

Haploinsufficiency for the HMG-box transcription factor SOX2 results in abnormalities of the human ventral forebrain and its derivative structures. These defects include anophthalmia (absence of eye), microphthalmia (small eye) and hypothalamic hamartoma (HH), an overgrowth of the ventral hypothalamus. To determine how Sox2 deficiency affects the morphogenesis of the ventral diencephalon and eye, we generated a Sox2 allelic series (Sox2(IR), Sox2(LP), and Sox2(EGFP)), allowing for the generation of mice that express germline hypomorphic levels (<40%) of SOX2 protein and that faithfully recapitulate SOX2 haploinsufficient human phenotypes. We find that Sox2 hypomorphism significantly disrupts the development of the posterior hypothalamus, resulting in an ectopic protuberance of the prechordal floor, an upregulation of Shh signaling, and abnormal hypothalamic patterning. In the anterior diencephalon, both the optic stalks and optic cups (OC) of Sox2 hypomorphic (Sox2(HYP)) embryos are malformed. Furthermore, Sox2(HYP) eyes exhibit a loss of neural potential and coloboma, a common phenotype in SOX2 haploinsufficient humans that has not been described in a mouse model of SOX2 deficiency. These results establish for the first time that germline Sox2 hypomorphism disrupts the morphogenesis and patterning of the hypothalamus, optic stalk, and the early OC, establishing a model of the development of the abnormalities that are observed in SOX2 haploinsufficient humans.


Asunto(s)
Anomalías del Ojo/genética , Hipotálamo/anomalías , Factores de Transcripción SOXB1/genética , Animales , Diencéfalo/anomalías , Diencéfalo/patología , Modelos Animales de Enfermedad , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/patología , Haploinsuficiencia , Humanos , Hipotálamo/patología , Ratones , Ratones Transgénicos , Microscopía Electrónica de Rastreo , Especificidad de Órganos , Factores de Transcripción SOXB1/deficiencia
9.
BMC Genomics ; 11: 497, 2010 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-20846364

RESUMEN

BACKGROUND: Congenital heart defect (CHD) account for 25% of all human congenital abnormalities. However, very few CHD-causing genes have been identified so far. A promising approach for the identification of essential cardiac regulators whose mutations may be linked to human CHD, is the molecular and genetic analysis of heart development. With the use of a triple retinoic acid competitive antagonist (BMS189453) we previously developed a mouse model of congenital heart defects (81%), thymic abnormalities (98%) and neural tube defects (20%). D-TGA (D-transposition of great arteries) was the most prevalent cardiac defect observed (61%). Recently we were able to partially rescue this abnormal phenotype (CHD were reduced to 64.8%, p = 0.05), by oral administration of folic acid (FA). Now we have performed a microarray analysis in our mouse models to discover genes/transcripts potentially implicated in the pathogenesis of this CHD. RESULTS: We analysed mouse embryos (8.5 dpc) treated with BMS189453 alone and with BMS189453 plus folic acid (FA) by microarray and qRT-PCR. By selecting a fold change (FC) ≥ ± 1.5, we detected 447 genes that were differentially expressed in BMS-treated embryos vs. untreated control embryos, while 239 genes were differentially expressed in BMS-treated embryos whose mothers had also received FA supplementation vs. BMS-treated embryos. On the basis of microarray and qRT-PCR results, we further analysed the Hif1α gene. In fact Hif1α is down-regulated in BMS-treated embryos vs. untreated controls (FCmicro = -1.79; FCqRT-PCR = -1.76; p = 0.005) and its expression level is increased in BMS+FA-treated embryos compared to BMS-treated embryos (FCmicro = +1.17; FCqRT-PCR = +1.28: p = 0.005). Immunofluorescence experiments confirmed the under-expression of Hif1α protein in BMS-treated embryos compared to untreated and BMS+FA-treated embryos and, moreover, we demonstrated that at 8.5 dpc, Hif1α is mainly expressed in the embryo heart region. CONCLUSIONS: We propose that Hif1α down-regulation in response to blocking retinoic acid binding may contribute to the development of cardiac defects in mouse newborns. In line with our hypothesis, when Hif1α expression level is restored (by supplementation of folic acid), a decrement of CHD is found. To the best of our knowledge, this is the first report that links retinoic acid metabolism to Hif1α regulation and the development of D-TGA.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Retinoides/farmacología , Transposición de los Grandes Vasos/genética , Tretinoina/antagonistas & inhibidores , Secuencia de Bases , Suplementos Dietéticos , Regulación hacia Abajo/genética , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Embrionario/genética , Técnica del Anticuerpo Fluorescente , Ácido Fólico/farmacología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Elementos de Respuesta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alineación de Secuencia , Transactivadores/genética , Transactivadores/metabolismo
10.
Birth Defects Res B Dev Reprod Toxicol ; 89(4): 313-25, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20803691

RESUMEN

A large body of evidence supports the concept that human pregnancy outcome is significantly influenced by the nutritional status of the mother. The consumption of "poor diets" has been associated with an increased risk for pregnancy complications, including gross structural birth defects, prematurity, low birth weight, and an increased risk for neurobehavioral and immunological abnormalities after birth. Forty-four years ago, zinc deficiency in mammals was shown to be teratogenic. Maternal zinc deficiency produces effects ranging from infertility and embryo/fetal death, to intrauterine growth retardation and teratogenesis. Postnatal complications of maternal zinc deficiency can also occur, and include behavioral abnormalities, impaired immunocompetence, and an elevated risk for high blood pressure in the offspring. It has been suggested that developmental zinc deficiency in humans can present a significant challenge to the conceptus, increasing the risk for numerous defects. Developmental zinc deficiency can occur through multiple pathways, and the concept that acute phase response-induced changes in maternal zinc metabolism may be a common cause of embryonic and fetal zinc deficiency is presented. Potential mechanisms underlying the teratogenic effects of zinc deficiency are reviewed. The potential value of maternal zinc supplementation in high risk pregnancies is discussed.


Asunto(s)
Anomalías Congénitas/embriología , Embrión de Mamíferos/embriología , Fenómenos Fisiologicos de la Nutrición Prenatal , Zinc/deficiencia , Reacción de Fase Aguda/metabolismo , Embrión de Mamíferos/anomalías , Femenino , Retardo del Crecimiento Fetal , Humanos , Recién Nacido , Embarazo , Complicaciones del Embarazo/etiología , Resultado del Embarazo
11.
Dis Model Mech ; 2(9-10): 467-78, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19638421

RESUMEN

Elevated plasma homocysteine (HCy), which results from folate (folic acid, FA) deficiency, and the mood-stabilizing drug lithium (Li) are both linked to the induction of human congenital heart and neural tube defects. We demonstrated previously that acute administration of Li to pregnant mice on embryonic day (E)6.75 induced cardiac valve defects by potentiating Wnt-beta-catenin signaling. We hypothesized that HCy may similarly induce cardiac defects during gastrulation by targeting the Wnt-beta-catenin pathway. Because dietary FA supplementation protects from neural tube defects, we sought to determine whether FA also protects the embryonic heart from Li- or HCy-induced birth defects and whether the protection occurs by impacting Wnt signaling. Maternal elevation of HCy or Li on E6.75 induced defective heart and placental function on E15.5, as identified non-invasively using echocardiography. This functional analysis of HCy-exposed mouse hearts revealed defects in tricuspid and semilunar valves, together with altered myocardial thickness. A smaller embryo and placental size was observed in the treated groups. FA supplementation ameliorates the observed developmental errors in the Li- or HCy-exposed mouse embryos and normalized heart function. Molecular analysis of gene expression within the avian cardiogenic crescent determined that Li, HCy or Wnt3A suppress Wnt-modulated Hex (also known as Hhex) and Islet-1 (also known as Isl1) expression, and that FA protects from the gene misexpression that is induced by all three factors. Furthermore, myoinositol with FA synergistically enhances the protective effect. Although the specific molecular epigenetic control mechanisms remain to be defined, it appears that Li or HCy induction and FA protection of cardiac defects involve intimate control of the canonical Wnt pathway at a crucial time preceding, and during, early heart organogenesis.


Asunto(s)
Ácido Fólico/farmacología , Cardiopatías Congénitas/prevención & control , Proteínas Wnt/metabolismo , Animales , Proteínas Aviares/genética , Proteínas Aviares/metabolismo , Pollos , Suplementos Dietéticos , Modelos Animales de Enfermedad , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/diagnóstico por imagen , Embrión de Mamíferos/efectos de los fármacos , Embrión no Mamífero/anomalías , Embrión no Mamífero/efectos de los fármacos , Gastrulación/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Cardiopatías Congénitas/inducido químicamente , Cardiopatías Congénitas/diagnóstico por imagen , Cardiopatías Congénitas/fisiopatología , Pruebas de Función Cardíaca/efectos de los fármacos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Homocisteína , Inositol/farmacología , Proteínas con Homeodominio LIM , Litio , Ratones , Miocardio/metabolismo , Miocardio/patología , Factores de Transcripción , Ultrasonografía , Proteína Wnt3 , Proteína Wnt3A
12.
Artículo en Inglés | MEDLINE | ID: mdl-19170234

RESUMEN

BACKGROUND: Pregnancy in mammals with diabetes mellitus results in low birth weight, malformations, and intrauterine death. Parenteral application of natural polyamines or their precursor, L-arginine, to diabetic pregnant rats partially prevents the alterations of development caused by diabetes mellitus. This experiment has been designed to understand if this preventive action also occurs in rat whole embryo in culture. MATERIALS AND METHODS: Rat embryos of gestational day 10 were cultured for 24 h in normal medium, high glucose medium, or high glucose medium supplemented with polyamines or L-arginine, and furthermore embryo growth and development were evaluated. RESULTS: L-arginine and putrescine partially prevents the dysmorphogenic effects of high glucose, whereas spermidine and spermine prevent these effects almost completely. CONCLUSIONS: Polyamines directly protect the embryo from the toxic effect of high glucose concentration on growth and development, although the mechanism remains to be elucidated.


Asunto(s)
Anomalías Inducidas por Medicamentos/prevención & control , Diabetes Mellitus Experimental/metabolismo , Embrión de Mamíferos/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Glucosa/toxicidad , Espermina/farmacología , Anomalías Inducidas por Medicamentos/etiología , Animales , Arginina/farmacología , Combinación de Medicamentos , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/metabolismo , Femenino , Técnicas de Cultivo de Órganos , Ratas , Ratas Wistar
13.
Dev Biol ; 325(1): 94-105, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18955041

RESUMEN

Mammalian eye development requires vitamin A (retinol, ROL). The role of vitamin A at specific times during eye development was studied in rat fetuses made vitamin A deficient (VAD) after embryonic day (E) 10.5 (late VAD). The optic fissure does not close in late VAD embryos, and severe folding and collapse of the retina is observed at E18.5. Pitx2, a gene required for normal optic fissure closure, is dramatically downregulated in the periocular mesenchyme in late VAD embryos, and dissolution of the basal lamina does not occur at the optic fissure margin. The addition of ROL to late VAD embryos by E12.5 restores Pitx2 expression, supports dissolution of the basal lamina, and prevents coloboma, whereas supplementation at E13.5 does not. Surprisingly, ROL given as late as E13.5 completely prevents folding of the retina despite the presence of an open fetal fissure, showing that coloboma and retinal folding represent distinct VAD-dependent defects. Retinal folding due to VAD is preceded by an overall reduction in the percentage of cyclin D1 positive cells in the developing retina, (initially resulting in retinal thinning), as well as a dramatic reduction in the cell adhesion-related molecules, N-cadherin and beta-catenin. Reduction of retinal cell number combined with a loss of the normal cell-cell adhesion proteins may contribute to the collapse and folding of the retina that occurs in late VAD fetuses.


Asunto(s)
Retina/citología , Retina/embriología , Vitamina A/metabolismo , Animales , Membrana Basal/efectos de los fármacos , Membrana Basal/patología , Cadherinas/metabolismo , Adhesión Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Coloboma/complicaciones , Coloboma/embriología , Coloboma/genética , Ciclina D1/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/efectos de los fármacos , Feto/anomalías , Feto/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Retina/anomalías , Retina/efectos de los fármacos , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/efectos de los fármacos , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vitamina A/farmacología , Deficiencia de Vitamina A/embriología , Deficiencia de Vitamina A/genética , beta Catenina/metabolismo , Proteína del Homeodomínio PITX2
14.
Reprod Toxicol ; 23(1): 92-7, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17034988

RESUMEN

The effect of glycine in preventing cadmium (Cd) teratogenicity in mice was studied. Cadmium chloride (CdCl2) was administered subcutaneously at 1, 2 or 4 mg/kg doses on gestation days (GD) 7, 8 and 9. Glycine was given ad libitum (in the drinking water) from GD0 through GD18 (the day when animals were killed), as a 1% and 2% drinking water solution. Cd and nucleic acid concentrations in embryos were determined. The most common finding seen after CdCl2 4 mg/kg exposure was exencephaly. The incidence of this malformation was significantly reduced in mice receiving 2% glycine while fetal Cd significantly decreased as compared to cadmium-treated positive control animals. Increased nucleic acid levels were seen in the same embryos. In glycine non-supplemented mice given CdCl2 4 mg/kg, embryonic lipid peroxidation proved to be increased. In conclusion, lipid peroxidation was associated with cadmium-induced teratogenicity, and glycine inhibited the cadmium-induced effect by inhibiting placental transport of cadmium. However, further detailed studies are needed to establish the mechanism(s) of action.


Asunto(s)
Anomalías Inducidas por Medicamentos/prevención & control , Cloruro de Cadmio/toxicidad , Embrión de Mamíferos/efectos de los fármacos , Glicina/farmacología , Teratógenos/toxicidad , Anomalías Inducidas por Medicamentos/patología , Animales , ADN/análisis , Relación Dosis-Respuesta a Droga , Antagonismo de Drogas , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/metabolismo , Femenino , Peroxidación de Lípido/efectos de los fármacos , Malondialdehído/metabolismo , Ratones , Embarazo
15.
Toxicol Appl Pharmacol ; 213(1): 55-63, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16226775

RESUMEN

Splotch (Sp/Sp) mice homozygous for a mutation in the Pax3 gene inevitably present with neural tube defects (NTDs), along with other associated congenital anomalies. The affected mutant embryos usually die by gestation days (E) 12-13. In the present study, the effect of modifier genes from a new genetic background (CXL-Sp) and periconceptional supplementation with selected micronutrients (folic acid, 5-formyltetrahydrofolate, 5-methyltetrahydrofolate, methionine, myoinositol, thiamine, thymidine, and alpha-tocopherol) was determined with respect to the incidence of NTDs. In order to explore how different exposure parameters (time, dose, and route of compound administration) modulate the beneficial effects of micronutrient supplementation, female mice received either short- or long-term nutrient supplements via enteral or parenteral routes. Embryos were collected on E12.5 and examined for the presence of anterior or posterior NTDs. Additionally, whole mount in situ hybridization studies were conducted in order to reveal/confirm normal expression patterns of the Pax3 gene during neurulation in the wild-type and Sp/Sp homozygous mutant mouse embryos utilized in this study. A strong Pax3 signal was demonstrated in CXL-Sp embryos during neural tube closure (E9.5 to E10.5). The intensity and spatial pattern of expression were similar to other Splotch mutant mice. Of all the micronutrients tested, only supplementation with folic acid or 5-methyltetrahydrofolate rescued the normal phenotype in Sp/Sp embryos. When the folate supplementation dose was increased to 200 mg/kg in the diet, the incidence of rescued splotch homozygotes reached 30%; however, this was accompanied by six-fold increased resorption rate.


Asunto(s)
Micronutrientes/farmacología , Defectos del Tubo Neural/prevención & control , Factores de Transcripción Paired Box/genética , Animales , Embrión de Mamíferos/anomalías , Femenino , Hibridación in Situ , Ratones , Ratones Mutantes , Mutación , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/genética , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/biosíntesis , Embarazo
16.
Development ; 131(22): 5551-60, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15496447

RESUMEN

We have generated a series of Erbb2 cDNA knock-in animals to explore the role of signaling pathways coupled to Erbb2 during development. Although this knock-in allele was hypomorphic, expressing tenfold less Erbb2 protein than wild type, the knock-in animals were healthy. However, a further twofold reduction in Erbb2 levels in hemizygous knock-in animals resulted in perinatal lethality with defects in the innervation of the diaphragm. Genetic rescue of this hypomorph was accomplished by expression of the Erbb2-Y1028F mutant in a comparable knock-in allele. Interestingly, hemizygous Y1028F animals were viable with normal innervation of the diaphragm. Molecular analyses revealed that the Y1028F allele expressed higher levels of Erbb2 and that Y1028 promoted the turnover of the receptor. In addition, ablation of the Shc-binding site in Erbb2 (Y1227) resulted in subtle defects in the sensory nerves not observed in the other mutant erbb2 strains. Thus, we have established how Erbb2 levels may be modulated through development and that a minimum threshold level of Erbb2 is required.


Asunto(s)
Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Receptor ErbB-2/metabolismo , Transducción de Señal , Animales , Animales Recién Nacidos , Línea Celular , ADN Complementario/genética , Regulación hacia Abajo/genética , Embrión de Mamíferos/anomalías , Regulación del Desarrollo de la Expresión Génica , Genes Letales/genética , Genotipo , Heterocigoto , Ratones , Ratones Transgénicos , Mutación/genética , Neuronas/citología , Neuronas/metabolismo , Fosfotirosina/genética , Ratas , Receptor ErbB-2/química , Receptor ErbB-2/genética , Tasa de Supervivencia , Tirosina/genética , Tirosina/metabolismo
17.
Dev Dyn ; 231(1): 221-31, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15305303

RESUMEN

Inactivation of folate binding protein-1 (Folbp1) adversely impacts murine embryonic development, as nullizygous embryos (Folbp1(-/-)) die in utero. Administration of folinic acid (N5-formyl-tetrahydrofolate) to Folbp1-deficient dams before and throughout gestation rescues the majority of embryos from premature death; however, a portion of surviving embryos develop structural malformations, including neural tube defects. We examined whether maternal supplementation with L-N5-methyl-tetrahydrofolate (L-5M-THF) has superior protective effects on embryonic development of Folbp1(-/-) fetuses compared with L-N5-formyl-tetrahydrofolate (L-5F-THF). We also examined the critical period during gestation when folate supplementation is most beneficial to the developing Folbp1(-/-) embryos. Folbp1(-/-) pups presented with a range of malformations involving the neural tube, craniofacies, eyes, and abdominal wall. The frequencies of these malformations decreased with increasing folate dose, regardless of the form used. There was no additional benefit provided by L-5M-THF compared with L-5F-THF. Despite rescuing the phenotype in Folbp1(-/-) embryos, no significant elevation of Folbp1(-/-) maternal folate levels was observed with supplementation.


Asunto(s)
Proteínas Portadoras/genética , Embrión de Mamíferos/anomalías , Leucovorina/farmacología , Fenotipo , Receptores de Superficie Celular/genética , Animales , Proteínas Portadoras/metabolismo , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Femenino , Receptores de Folato Anclados a GPI , Leucovorina/farmacocinética , Ratones , Ratones Noqueados , Embarazo , Receptores de Superficie Celular/metabolismo , Tasa de Supervivencia
18.
Ann Nutr Metab ; 47(1): 6-10, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12624481

RESUMEN

BACKGROUND/AIMS: Previously we have shown that administration of 150 mg of vitamin E (alpha-tocopherol) per day to rats having diabetes decreases the rate of embryo malformations and increases their maturation and size. The present study was addressed to determine the effects of different doses of vitamin E upon these parameters. METHODS: Female rats were made diabetic (D) with streptozotocin, and from day 0 of gestation they were treated daily with 25 (D+25), 50 (D+50), 100 (D+100), 150 (D+150), and 500 (D+500) mg of vitamin E administered orally and were compared with control (C) animals. RESULTS: On day 11.5 of gestation, crown-rump length, somite number, and protein and DNA levels were lower in D than in C embryos. Crown-rump length and somite number increased with 100 mg or higher doses of vitamin E, although the values observed in C embryos were not reached. The proportions of reabsorption and malformations were 24.7 and 50%, respectively, in D rats, and in the rats supplemented with vitamin E they decreased to 22.7 and 19% in D+25, 16.4 and 21.3% in D+50, 16.2 and 12% in D+100, 12.9 and 13.9% in D+150, and to 43.9 and 10.8% in D+500 rats, whereas the values were 6.8 and 4.9% in C animals. CONCLUSIONS: Administration of vitamin E to D rats decreases the rate of embryo malformations, dependent on the dose administered. However, high doses have a negative effect in the conceptus, as shown by the increased rate of reabsorptions in the D+500 group.


Asunto(s)
Antioxidantes/farmacología , Diabetes Mellitus Experimental/embriología , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/efectos de los fármacos , Embarazo en Diabéticas/embriología , Vitamina E/farmacología , Animales , Largo Cráneo-Cadera , ADN/efectos de los fármacos , Diabetes Mellitus Experimental/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Embarazo , Embarazo en Diabéticas/metabolismo , Proteínas/efectos de los fármacos , Ratas , Ratas Wistar , Somitos/efectos de los fármacos , Estreptozocina
19.
J Cell Biol ; 158(4): 801-15, 2002 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-12186857

RESUMEN

Glycoprotein fucosylation enables fringe-dependent modulation of signal transduction by Notch transmembrane receptors, contributes to selectin-dependent leukocyte trafficking, and is faulty in leukocyte adhesion deficiency (LAD) type II, also known as congenital disorder of glycosylation (CDG)-IIc, a rare human disorder characterized by psychomotor defects, developmental abnormalities, and leukocyte adhesion defects. We report here that mice with an induced null mutation in the FX locus, which encodes an enzyme in the de novo pathway for GDP-fucose synthesis, exhibit a virtually complete deficiency of cellular fucosylation, and variable frequency of intrauterine demise determined by parental FX genotype. Live-born FX(-/-) mice exhibit postnatal failure to thrive that is suppressed with a fucose-supplemented diet. FX(-/-) adults suffer from an extreme neutrophilia, myeloproliferation, and absence of leukocyte selectin ligand expression reminiscent of LAD-II/CDG-IIc. Contingent restoration of leukocyte and endothelial selectin ligand expression, general cellular fucosylation, and normal postnatal physiology is achieved by modulating dietary fucose to supply a salvage pathway for GDP-fucose synthesis. Conditional control of fucosylation in FX(-/-) mice identifies cellular fucosylation events as essential concomitants to fertility, early growth and development, and leukocyte adhesion.


Asunto(s)
Carbohidrato Epimerasas/metabolismo , Proteínas de Escherichia coli/metabolismo , Fucosa/metabolismo , Integrinas/metabolismo , Cetona Oxidorreductasas/metabolismo , Leucocitosis/genética , Complejos Multienzimáticos/metabolismo , Selectinas/metabolismo , Animales , Animales Modificados Genéticamente , Carbohidrato Epimerasas/genética , Suplementos Dietéticos , Embrión de Mamíferos/anomalías , Proteínas de Escherichia coli/genética , Femenino , Viabilidad Fetal , Genotipo , Cetona Oxidorreductasas/genética , Síndrome de Deficiencia de Adhesión del Leucocito/genética , Síndrome de Deficiencia de Adhesión del Leucocito/metabolismo , Leucocitosis/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multienzimáticos/genética , Mutación , Fenotipo , Polisacáridos/metabolismo
20.
Diabetes ; 47(4): 677-84, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9568703

RESUMEN

Previous studies have suggested that the metabolism of arachidonic acid and radical oxygen species (ROS) are altered in diabetes and that these disturbances may induce severe embryonic dysmorphogenesis in diabetic pregnancy. We tested this hypothesis by studying whether an inhibition of the rate-limiting enzyme of prostaglandin biosynthesis, cyclooxygenase (COX), caused developmental disturbances analogous to those seen in embryos exposed to high glucose concentration. Whether antioxidants could prevent such developmental alterations was also investigated. Whole embryo culture was used in which day-9 embryos were exposed to high concentrations of glucose, arachidonic acid, prostaglandin (PG)E2, COX inhibitors, and antioxidants for 48 h. Increased glucose concentration (from 10 to 30 mmol/l) caused embryonic dysmorphogenesis, and addition of either 60 pmol/l arachidonic acid or 280 nmol/l PGE2 largely protected the embryo from this maldevelopment. Furthermore, exposure to the COX inhibitors indomethacin (200 micromol/l) or acetylsalicylic acid (700 micromol/l) in 10 mmol/l glucose concentration yielded embryonic dysmorphogenesis similar to that caused by 30 mmol/l glucose. Supplementation of either arachidonic acid or PGE2 to the culture medium with COX inhibitors in low glucose rectified the embryonic development, and PGE2 supplementation also normalized the development of embryos cultured with COX inhibitors in high glucose concentration. Interestingly, the antioxidants superoxide dismutase (SOD) and N-acetylcysteine (NAC) were each able to diminish the dysmorphogenesis induced by the COX inhibitors, at doses previously shown to diminish glucose-induced embryonic damage in the same in vitro culture system. In conclusion, the present study shows that a high glucose concentration disturbs embryonic development and that this disturbance may be partly mediated via altered metabolism of arachidonic acid and ROS in the embryo.


Asunto(s)
Antioxidantes/farmacología , Inhibidores de la Ciclooxigenasa/farmacología , Embrión de Mamíferos , Desarrollo Embrionario y Fetal/efectos de los fármacos , Glucosa/toxicidad , Prostaglandina-Endoperóxido Sintasas/metabolismo , Acetilcisteína/farmacología , Animales , Ácido Araquidónico/metabolismo , Ácido Araquidónico/farmacología , Aspirina/farmacología , Técnicas de Cultivo , Dinoprostona/metabolismo , Dinoprostona/farmacología , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Indometacina/farmacología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA