Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 577
Filtrar
1.
Development ; 146(18)2019 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-31444215

RESUMEN

Developmental defects affecting the heart and aortic arch arteries are a significant phenotype observed in individuals with 22q11 deletion syndrome and are caused by a microdeletion on chromosome 22q11. TBX1, one of the deleted genes, is expressed throughout the pharyngeal arches and is considered a key gene, when mutated, for the arch artery defects. Pax9 is expressed in the pharyngeal endoderm and is downregulated in Tbx1 mutant mice. We show here that Pax9-deficient mice are born with complex cardiovascular malformations that affect the outflow tract and aortic arch arteries with failure of the 3rd and 4th pharyngeal arch arteries to form correctly. Transcriptome analysis indicated that Pax9 and Tbx1 may function together, and mice double heterozygous for Tbx1/Pax9 presented with a significantly increased incidence of interrupted aortic arch when compared with Tbx1 heterozygous mice. Using a novel Pax9Cre allele, we demonstrated that the site of this Tbx1-Pax9 genetic interaction is the pharyngeal endoderm, therefore revealing that a Tbx1-Pax9-controlled signalling mechanism emanating from the pharyngeal endoderm is required for crucial tissue interactions during normal morphogenesis of the pharyngeal arch artery system.


Asunto(s)
Arterias/embriología , Región Branquial/irrigación sanguínea , Sistema Cardiovascular/embriología , Endodermo/embriología , Morfogénesis , Factor de Transcripción PAX9/metabolismo , Faringe/embriología , Proteínas de Dominio T Box/metabolismo , Animales , Sistema Cardiovascular/metabolismo , Diferenciación Celular/genética , Embrión de Mamíferos/anomalías , Eliminación de Gen , Redes Reguladoras de Genes , Heterocigoto , Ratones Endogámicos C57BL , Modelos Biológicos , Mutación/genética , Cresta Neural/patología , Factor de Transcripción PAX9/deficiencia , Unión Proteica , Transducción de Señal
2.
Development ; 145(19)2018 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-30228104

RESUMEN

Midface dysgenesis is a feature of more than 200 genetic conditions in which upper airway anomalies frequently cause respiratory distress, but its etiology is poorly understood. Mouse models of Apert and Crouzon craniosynostosis syndromes exhibit midface dysgenesis similar to the human conditions. They carry activating mutations of Fgfr2, which is expressed in multiple craniofacial tissues during development. Magnetic resonance microscopy of three mouse models of Apert and Crouzon syndromes revealed decreased nasal passage volume in all models at birth. Histological analysis suggested overgrowth of the nasal cartilage in the two Apert syndrome mouse models. We used tissue-specific gene expression and transcriptome analysis to further dissect the structural, cellular and molecular alterations underlying midface and upper airway dysgenesis in Apert Fgfr2+/S252W mutants. Cartilage thickened progressively during embryogenesis because of increased chondrocyte proliferation in the presence of Fgf2 Oral epithelium expression of mutant Fgfr2, which resulted in a distinctive nasal septal fusion defect, and premature facial suture fusion contributed to the overall dysmorphology. Midface dysgenesis in Fgfr2-related craniosynostosis is a complex phenotype arising from the combined effects of aberrant signaling in multiple craniofacial tissues.


Asunto(s)
Ciclo Celular , Craneosinostosis/embriología , Cara/anomalías , Especificidad de Órganos , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Anomalías del Sistema Respiratorio/embriología , Anomalías del Sistema Respiratorio/patología , Acrocefalosindactilia/patología , Animales , Cartílago/patología , Proliferación Celular , Condrocitos/patología , Suturas Craneales/patología , Disostosis Craneofacial/embriología , Disostosis Craneofacial/patología , Craneosinostosis/patología , Modelos Animales de Enfermedad , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/patología , Cara/embriología , Cara/patología , Regulación del Desarrollo de la Expresión Génica , Ratones Endogámicos C57BL , Ratones Mutantes , Nariz/anomalías , Nariz/embriología , Nariz/patología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética
3.
Reprod Biomed Online ; 42(5): 892-900, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33810985

RESUMEN

RESEARCH QUESTION: To assess incidence of abnormal cleavage among biopsied blastocysts; to compare euploidy rates of the blastocysts with abnormal and normal cleavage; and to compare single euploid blastocyst transfer (SEBT) outcome derived from embryos with normal or abnormal cleavage. DESIGN: Retrospective analysis of prospectively collected data in a private IVF clinic. Consecutive 554 patients (749 cycles) undergoing preimplantation genetic testing for aneuploidy (n = 497; 671 cycles) or monogenic diseases (n = 57; 78 cycles) were included. All assessments for abnormal cleavage were carried out retrospectively; presence of abnormal cleavage was not a factor in deciding which euploid embryo to transfer. A total of 1015 blastocysts were biopsied and 295 SEBT procedures were carried out. Main outcome measure was live birth rate (LBR). RESULTS: Incidence of reverse cleavage, direct cleavage, and reverse plus direct cleavage, were 7.7%, 6.4% and 2.3%, respectively. Of the 1015 biopsied blastocysts, 35.0% were euploid. Blastocysts with abnormal cleavage, in total, had a significantly higher euploidy rate compared with blastocysts with normal cleavage (44.6% [74/166] versus 33.1% [281/849]; P = 0.017). The LBR after SEBT with normal, reverse and direct cleavage, and direct cleavage plus reverse cleavage, was 133/238 (55.9%), 6/26 (23.1%), 8/24 (33.3%) and 0/3 (0.0%) (P < 0.001). Generalized estimating equation analysis showed that the presence of abnormal cleavage pattern was the only independent predictor of LBR (OR 0.316; 95% CI 0.115 to 0.867; P = 0.013). CONCLUSIONS: Blastocysts with direct or reverse cleavage should be biopsied in preimplantation genetic testing cycles if they are morphologically eligible. Euploid blastocysts with abnormal cleavage, however, have approximately half the LBR of those euploid blastocyst with normal cleavage, hence, blastocysts with abnormal cleavage should have lower priority for transfer.


Asunto(s)
Aneuploidia , Blastocisto/patología , Transferencia de Embrión/estadística & datos numéricos , Desarrollo Embrionario , Adulto , Embrión de Mamíferos/anomalías , Femenino , Humanos , Nacimiento Vivo , Embarazo , Estudios Retrospectivos
4.
Reprod Biomed Online ; 43(4): 627-636, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34474978

RESUMEN

RESEARCH QUESTION: What is the prevalence of embryo abnormal early cleavage (ACL) identified by time lapse and factors related to patients and treatment that explain ACL occurrence? DESIGN: A single-centre, retrospective cohort study. Data were collected on all IVF cycles for which embryos were observed in the EmbryoScope® between December 2015 and August 2017. Only diploid zygotes cleaved on day 2 were included. The study included 318 cycles (250 couples and 1343 embryos). Embryo videos were retrospectively analysed for ACL. The prevalence of each type of ACL was recorded. The influence of clinical factors (whether they were intrinsic to patients or specific to IVF treatment) on ACL occurrence was analysed in multivariate multilevel mixed-effect logistic regression analysis. RESULTS: A high prevalence of ACL was observed: 37.6% (505/1343) of embryos presented at least one ACL, 22.8% (306/1343) a trichotomous mitosis, 25.8% (347/1343) a rapid cleavage, 6.7% (90/1343) a cell fusion and two or more ACL (16.1%). Part of the variation (12-25%) in ACL occurrence could be explained by embryo origin. Trichotomous mitosis and two or more ACL phenotypes were less likely to occur in women with endometriosis or tubal pathology and tubal pathology alone, respectively. No factor related to IVF cycles was found to be statistically associated with ACL occurrence. CONCLUSIONS: Our findings emphasize the importance of considering embryo origin when interpreting studies focusing on embryo characteristics and factors that could affect their quality. The present study is limited by a small sample size of known embryo implantations and monocentric criterion.


Asunto(s)
Embrión de Mamíferos/anomalías , Desarrollo Embrionario , Técnicas Reproductivas Asistidas , Imagen de Lapso de Tiempo , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Adulto Joven
5.
Reprod Biomed Online ; 43(4): 645-654, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34446374

RESUMEN

RESEARCH QUESTION: Does maternal preconceptional body mass index (BMI) associate with mean blastocyst euploidy rate (m-ER) per patient and live birth rate (LBR) after vitrified-warmed euploid single embryo transfer (SET)? DESIGN: Observational study conducted between April 2013 and March 2020 at a private IVF clinic, involving 1811 Caucasian women undergoing trophectoderm biopsy and comprehensive chromosome testing. The outcomes of 1125 first vitrified-warmed euploid SET were also analysed. Patients were clustered as normal weight (BMI 18.5-25; n = 1392 performing 859 SET), underweight (BMI <18.5; n = 160 performing 112 SET) and overweight (BMI >25; n = 259 performing 154 SET). m-ER per patient was the primary outcome. The secondary outcomes were all clinical outcomes per euploid SET. All data were adjusted for confounders through regression analyses. RESULTS: The m-ER per patient decreases as maternal BMI increases from 17 up to 22-23 before reaching a plateau. A linear regression adjusted for maternal age confirmed this moderate association (unstandardized coefficient B: -0.6%, 95% confidence interval [CI]: -1.1 to -0.1%, P = 0.02). All clinical outcomes were similar between normal weight and underweight women. Overweight women, instead, showed higher miscarriage rate per clinical pregnancy (n = 20/75, 26.7% versus n = 67/461, 14.5%; odds ratio [OR] adjusted for blastocyst quality and day of full blastulation: 2.0, 95% CI: 1.1-3.6, P = 0.01) and lower LBR per SET (n = 55/154, 35.7% versus n = 388/859, 45.2%; OR adjusted for blastocyst quality and day of full blastulation: 0.67, 95% CI: 0.46-0.96, P = 0.03). CONCLUSION: These data indicate a need for future research on more sensitive metrics to assess body fat mass and distribution, as well as on the mechanisms leading to lipotoxicity, thereby impairing embryo competence and/or endometrial receptivity. Overweight women should be informed of their higher risk for miscarriage and, whenever possible, encouraged to lose weight, especially before transfer.


Asunto(s)
Aborto Espontáneo/etiología , Tasa de Natalidad , Índice de Masa Corporal , Embrión de Mamíferos/anomalías , Sobrepeso/complicaciones , Adulto , Femenino , Humanos , Persona de Mediana Edad , Embarazo , Estudios Retrospectivos , Adulto Joven
6.
Gynecol Endocrinol ; 37(2): 137-140, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32342711

RESUMEN

OBJECTIVES: We aimed to explore the effect of three pro-nuclei (3PN) incidence on laboratory and clinical outcomes after early rescue intracytoplasmic sperm injection (rescue-ICSI). METHODS: This study included 509 early rescue-ICSI cycles from February 2014 to February 2019. The patients were divided into 3PN = 0% (394 cycles) and 3PN > 0% (115 cycles) group. Main outcomes compared were good quality embryo, available embryo, implantation (IR), clinical pregnancy (CPR), abortion (AR) and live birth rates (LBR). RESULTS: There were no significant differences in the basal characteristics between two groups (p > .05). We observed that 3PN = 0% and 3PN > 0% groups had similar good quality embryo (47.02 versus 46.80%; p = .917) and available embryo (83.28 versus 81.37%; p = .247) rates. Our results showed that the IR (47.26 versus 51.05%; p = .357), CPR (61.17 versus 66.08%; p = .338) and LBR (52.80 versus 50.43%; p = .656) were comparable between 3PN = 0% and 3PN > 0% groups. The 3PN = 0% group showed significantly lower AR than that in the 3PN > 0% group (11.20 versus 21.05%; p = .029; OR 2.114; 95% CI 1.069-4.178). CONCLUSIONS: 3PN incidence made negative effects on the clinical outcomes after early rescue-ICSI.


Asunto(s)
Embrión de Mamíferos/anomalías , Inyecciones de Esperma Intracitoplasmáticas/estadística & datos numéricos , Adulto , Femenino , Humanos , Masculino , Embarazo , Estudios Retrospectivos
7.
FASEB J ; 33(12): 14204-14220, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31665922

RESUMEN

Polymorphic variants of the gene encoding for metabotropic glutamate receptor 3 (mGlu3) are linked to schizophrenia. Because abnormalities of cortical GABAergic interneurons lie at the core of the pathophysiology of schizophrenia, we examined whether mGlu3 receptors influence the developmental trajectory of cortical GABAergic transmission in the postnatal life. mGlu3-/- mice showed robust changes in the expression of interneuron-related genes in the prefrontal cortex (PFC), including large reductions in the expression of parvalbumin (PV) and the GluN1 subunit of NMDA receptors. The number of cortical cells enwrapped by perineuronal nets was increased in mGlu3-/- mice, suggesting that mGlu3 receptors shape the temporal window of plasticity of PV+ interneurons. Electrophysiological measurements of GABAA receptor-mediated responses revealed a more depolarized reversal potential of GABA currents in the somata of PFC pyramidal neurons in mGlu3-/- mice at postnatal d 9 associated with a reduced expression of the K+/Cl- symporter. Finally, adult mGlu3-/- mice showed lower power in electroencephalographic rhythms at 1-45 Hz in quiet wakefulness as compared with their wild-type counterparts. These findings suggest that mGlu3 receptors have a strong impact on the development of cortical GABAergic transmission and cortical neural synchronization mechanisms corroborating the concept that genetic variants of mGlu3 receptors may predispose to psychiatric disorders.-Imbriglio, T., Verhaeghe, R., Martinello, K., Pascarelli, M. T., Chece, G., Bucci, D., Notartomaso, S., Quattromani, M., Mascio, G., Scalabrì, F., Simeone, A., Maccari, S., Del Percio, C., Wieloch, T., Fucile, S., Babiloni, C., Battaglia, G., Limatola, C., Nicoletti, F., Cannella, M. Developmental abnormalities in cortical GABAergic system in mice lacking mGlu3 metabotropic glutamate receptors.


Asunto(s)
Corteza Cerebral/anomalías , Embrión de Mamíferos/anomalías , Neuronas GABAérgicas/fisiología , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Biomarcadores , Corteza Cerebral/metabolismo , Femenino , Regulación de la Expresión Génica , Genes Homeobox , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , ARN Mensajero , Receptores de Glutamato Metabotrópico/genética
8.
Pediatr Nephrol ; 35(5): 725-731, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-30923969

RESUMEN

Aberrant hedgehog (Hh) signaling during embryogenesis results in various severe congenital abnormalities, including renal malformations. The molecular mechanisms that underlie congenital renal malformations remain poorly understood. Here, we review the current understanding of the lineage-specific roles of Hh signaling during renal morphogenesis and how aberrant Hh signaling during embryonic kidney development contributes to renal malformation.


Asunto(s)
Desarrollo Embrionario/genética , Proteínas Hedgehog/metabolismo , Riñón/embriología , Transducción de Señal/genética , Proteína con Dedos de Zinc GLI1/metabolismo , Animales , Linaje de la Célula/genética , Embrión de Mamíferos/anomalías , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Humanos , Riñón/anomalías , Riñón/citología , Ratones , Modelos Animales , Mutación , Proteína con Dedos de Zinc GLI1/genética
9.
Nature ; 514(7521): 228-32, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25119037

RESUMEN

CHARGE syndrome is a multiple anomaly disorder in which patients present with a variety of phenotypes, including ocular coloboma, heart defects, choanal atresia, retarded growth and development, genitourinary hypoplasia and ear abnormalities. Despite 70-90% of CHARGE syndrome cases resulting from mutations in the gene CHD7, which encodes an ATP-dependent chromatin remodeller, the pathways underlying the diverse phenotypes remain poorly understood. Surprisingly, our studies of a knock-in mutant mouse strain that expresses a stabilized and transcriptionally dead variant of the tumour-suppressor protein p53 (p53(25,26,53,54)), along with a wild-type allele of p53 (also known as Trp53), revealed late-gestational embryonic lethality associated with a host of phenotypes that are characteristic of CHARGE syndrome, including coloboma, inner and outer ear malformations, heart outflow tract defects and craniofacial defects. We found that the p53(25,26,53,54) mutant protein stabilized and hyperactivated wild-type p53, which then inappropriately induced its target genes and triggered cell-cycle arrest or apoptosis during development. Importantly, these phenotypes were only observed with a wild-type p53 allele, as p53(25,26,53,54)(/-) embryos were fully viable. Furthermore, we found that CHD7 can bind to the p53 promoter, thereby negatively regulating p53 expression, and that CHD7 loss in mouse neural crest cells or samples from patients with CHARGE syndrome results in p53 activation. Strikingly, we found that p53 heterozygosity partially rescued the phenotypes in Chd7-null mouse embryos, demonstrating that p53 contributes to the phenotypes that result from CHD7 loss. Thus, inappropriate p53 activation during development can promote CHARGE phenotypes, supporting the idea that p53 has a critical role in developmental syndromes and providing important insight into the mechanisms underlying CHARGE syndrome.


Asunto(s)
Anomalías Múltiples/metabolismo , Síndrome CHARGE/genética , Síndrome CHARGE/metabolismo , Fenotipo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Anomalías Múltiples/genética , Alelos , Animales , Apoptosis/genética , Puntos de Control del Ciclo Celular/genética , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Oído/anomalías , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/metabolismo , Femenino , Fibroblastos , Eliminación de Gen , Heterocigoto , Humanos , Masculino , Ratones , Proteínas Mutantes/metabolismo , Regiones Promotoras Genéticas/genética
10.
RNA ; 23(1): 23-31, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27780844

RESUMEN

The exon junction complex (EJC) is a multiprotein complex integral to mRNA metabolism. Biochemistry and genetic studies have concluded that the EJC is composed of four core proteins, MAGOH, EIF4A3, RBM8A, and CASC3. Yet recent studies in Drosophila indicate divergent physiological functions for Barentsz, the mammalian Casc3 ortholog, raising the question as to whether CASC3 is a constitutive component of the EJC. This issue remains poorly understood, particularly in an in vivo mammalian context. We previously found that haploinsufficiency for Magoh, Eif4a3, or Rbm8a disrupts neuronal viability and neural progenitor proliferation, resulting in severe microcephaly. Here, we use two new Casc3 mouse alleles to demonstrate developmental phenotypes that sharply contrast those of other core EJC components. Homozygosity for either null or hypomorphic Casc3 alleles led to embryonic and perinatal lethality, respectively. Compound embryos lacking Casc3 expression were smaller with proportionately reduced brain size. Mutant brains contained fewer neurons and progenitors, but no apoptosis, all phenotypes explained by developmental delay. This finding, which contrasts with severe neural phenotypes evident in other EJC mutants, indicates Casc3 is largely dispensable for brain development. In the developing brain, CASC3 protein expression is substoichiometric relative to MAGOH, EIF4A3, and RBM8A. Taken together, this argues that CASC3 is not an essential EJC component in brain development and suggests it could function in a tissue-specific manner.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Factor 4A Eucariótico de Iniciación/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas/genética , Animales , Encéfalo/anomalías , Encéfalo/metabolismo , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Letales , Ratones , Modelos Animales , Proteínas de Neoplasias , Especificidad de Órganos
11.
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
12.
Cells Tissues Organs ; 207(1): 1-14, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31189166

RESUMEN

The number of human embryology collections is very limited worldwide. Some of these comprise the Carnegie Collection, Kyoto Collection, and the Blechschmidt Collection. One further embryonic collection is the Hinrichsen Collection of the Ruhr University Bochum, Germany, which also contains very well-preserved embryos/fetuses, along with approximately 16,000 histological sections. The digitization of this collection is indispensable to enable conservation of the collection for the future and to provide a large group of embryologists, researchers, and physicians access to these histological slides. A small selection of these scans is available at the website of the Digital Embryology Consortium [https://-human-embryology.org/wiki/Main_Page].


Asunto(s)
Embrión de Mamíferos/patología , Feto/diagnóstico por imagen , Imagenología Tridimensional , Imagen por Resonancia Magnética , Embrión de Mamíferos/anomalías , Femenino , Feto/anomalías , Humanos , Recién Nacido , Masculino
13.
Reprod Biomed Online ; 39(6): 981-989, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31606300

RESUMEN

RESEARCH QUESTION: Does the quality of transferred embryos have an impact on the rate of congenital malformations in IVF/intracytoplasmic sperm injection (ICSI)-conceived babies? DESIGN: Retrospective cohort study involving 6637 pregnancies of ≥20 weeks' gestation from women undergoing embryo transfer with a single Day 5 embryo at a private multisite IVF clinic between 2005 and 2015. Embryos were classified as good quality (n = 5537) or poor quality (n = 1100) based on an internal grading system of morphological parameters; malformation rates were compared. RESULTS: In pregnancies proceeding to delivery (≥20 weeks' gestation), poor quality embryos were associated with increased odds of at least one anomaly (adjusted odds ratio [OR] 1.33, 95% confidence interval [CI] 1.03-1.71), major anomalies (adjusted OR 1.42, 95% CI 1.05-1.91), musculoskeletal anomalies (adjusted OR 2.09, 95% CI 1.35-3.22), particularly talipes (adjusted OR 2.88, 95% CI 1.33-6.25), and the International Classification of Diseases (ICD) classification 'Other congenital malformations' (adjusted OR 2.34, 95% CI 1.13-4.34). Furthermore, for pregnancies ≥9 weeks' gestation, poor embryos had more than double the odds of chromosomal anomalies than good embryos (adjusted OR 2.33, 95% CI 1.30-4.18, P = 0.005). CONCLUSIONS: This is the first study to compare the rates of individual congenital malformations for good and poor quality embryos. It provides insight into potential risks of transferring poor quality embryos. In pregnancies ≥20 weeks' gestation, poor quality Day 5 embryos are associated with major malformations, at least one anomaly, musculoskeletal anomalies, talipes and the ICD classification 'Other congenital malformations'. In pregnancies ≥9 weeks' gestation, poor quality Day 5 embryos are associated with chromosomal anomalies.


Asunto(s)
Anomalías Congénitas/embriología , Embrión de Mamíferos/anomalías , Adulto , Femenino , Humanos , Embarazo , Estudios Retrospectivos , Inyecciones de Esperma Intracitoplasmáticas
14.
Int J Mol Sci ; 20(6)2019 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-30884872

RESUMEN

An increasing number of publications indicate that babies born after IVF (in vitro fertilization) procedures have higher rates of anomalies related to imprinting/epigenetic changes, which may be attributed to suboptimal culture conditions. Appropriate maintenance of DNA methylation during the first few days of an in vitro culture requires a supply of methyl donors, which are lacking in current in vitro culture systems. The absence of protection against oxidative stress in the culture increases the risks for errors in methylation. A decrease in the methylation processes is sometimes observed immediately post fertilization, due to delays that occur during the maternal⁻zygotic transition period. Care should be exercised in ART (assisted reproductive technology) procedures in order to avoid the risk of generating errors in methylation during the in vitro culture period immediately post fertilization, which has an impact on imprinting/epigenetics. Formulation of IVF culture media needs to be re-assessed in the perspective of current knowledge regarding embryo physiology.


Asunto(s)
Metilación de ADN , Embrión de Mamíferos/metabolismo , Epigénesis Genética , Fertilización In Vitro , Regulación del Desarrollo de la Expresión Génica , Técnicas de Cultivo de Célula/métodos , Embrión de Mamíferos/anomalías , Fertilización In Vitro/efectos adversos , Fertilización In Vitro/métodos , Impresión Genómica , Humanos , Estrés Oxidativo , Cigoto/citología , Cigoto/metabolismo
15.
J Biol Chem ; 292(44): 18091-18097, 2017 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-28912269

RESUMEN

Glycoprotein A repetitions predominant (GARP) (encoded by the Lrrc32 gene) plays important roles in cell-surface docking and activation of TGFß. However, GARP's role in organ development in mammalian systems is unclear. To determine the function of GARP in vivo, we generated a GARP KO mouse model. Unexpectedly, the GARP KO mice died within 24 h after birth and exhibited defective palatogenesis without apparent abnormalities in other major organs. Furthermore, we observed decreased apoptosis and SMAD2 phosphorylation in the medial edge epithelial cells of the palatal shelf of GARP KO embryos at embryonic day 14.5 (E14.5), indicating a defect in the TGFß signaling pathway in the GARP-null developing palates. Of note, the failure to develop the secondary palate and concurrent reduction of SMAD phosphorylation without other defects in GARP KO mice phenocopied TGFß3 KO mice, although GARP has not been suggested previously to interact with TGFß3. We found that GARP and TGFß3 co-localize in medial edge epithelial cells at E14.5. In vitro studies confirmed that GARP and TGFß3 directly interact and that GARP is indispensable for the surface expression of membrane-associated latent TGFß3. Our findings indicate that GARP is essential for normal morphogenesis of the palate and demonstrate that GARP plays a crucial role in regulating TGFß3 signaling during embryogenesis. In conclusion, we have uncovered a novel function of GARP in positively regulating TGFß3 activation and function.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas de la Membrana/metabolismo , Organogénesis , Hueso Paladar/metabolismo , Procesamiento Proteico-Postraduccional , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta3/agonistas , Animales , Animales Recién Nacidos , Apoptosis , Fisura del Paladar/embriología , Fisura del Paladar/metabolismo , Fisura del Paladar/patología , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Femenino , Técnicas de Sustitución del Gen , Células HEK293 , Heterocigoto , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones Noqueados , Hueso Paladar/anomalías , Hueso Paladar/embriología , Hueso Paladar/patología , Fosforilación , Embarazo , Multimerización de Proteína , Transporte de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta3/química , Factor de Crecimiento Transformador beta3/genética , Factor de Crecimiento Transformador beta3/metabolismo
16.
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
17.
Nucleic Acids Res ; 44(D1): D855-61, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26519470

RESUMEN

The Deciphering the Mechanisms of Developmental Disorders (DMDD) consortium is a research programme set up to identify genes in the mouse, which if mutated (or knocked-out) result in embryonic lethality when homozygous, and initiate the study of why disruption of their function has such profound effects on embryo development and survival. The project uses a combination of comprehensive high resolution 3D imaging and tissue histology to identify abnormalities in embryo and placental structures of embryonic lethal lines. The image data we have collected and the phenotypes scored are freely available through the project website (http://dmdd.org.uk). In this article we describe the web interface to the images that allows the embryo data to be viewed at full resolution in different planes, discuss how to search the database for a phenotype, and our approach to organising the data for an embryo and a mutant line so it is easy to comprehend and intuitive to navigate.


Asunto(s)
Bases de Datos Factuales , Embrión de Mamíferos/anomalías , Genes Letales , Animales , Anomalías Congénitas/genética , Embrión de Mamíferos/anatomía & histología , Desarrollo Embrionario/genética , Imagenología Tridimensional , Internet , Ratones Mutantes , Fenotipo
18.
Dev Biol ; 409(1): 95-105, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26481065

RESUMEN

Mutations in endoglin, a TGFß/BMP coreceptor, are causal for hereditary hemorrhagic telangiectasia (HHT). Endoglin-null (Eng-/-) mouse embryos die at embryonic day (E)10.5-11.5 due to defects in angiogenesis. In part, this is due to an absence of vascular smooth muscle cell differentiation and vessel investment. Prior studies from our lab and others have shown the importance of endoglin expression in embryonic development in both endothelial cells and neural crest stem cells. These studies support the hypothesis that endoglin may play cell-autonomous roles in endothelial and vascular smooth muscle cell precursors. However, the requirement for endoglin in vascular cell precursors remains poorly defined. Our objective was to specifically delete endoglin in neural crest- and somite-derived Pax3-positive vascular precursors to understand the impact on somite progenitor cell contribution to embryonic vascular development. Pax3Cre mice were crossed with Eng+/- mice to obtain compound mutant Pax3(Cre/+);Eng+/- mice. These mice were then crossed with homozygous endoglin LoxP-mutated (Eng(LoxP/LoxP)) mice to conditionally delete the endoglin gene in specific lineages that contribute to endothelial and smooth muscle constituents of developing embryonic vessels. Pax3(Cre/+);Eng(LoxP/)(-) mice showed a variety of vascular defects at E10.5, and none of these mice survived past E12.5. Embryos analyzed at E10.5 showed malformations suggestive of misdirection of the intersomitic vessels. The dorsal aorta showed significant dilation with associated vascular smooth muscle cells exhibiting disorganization and enhanced expression of smooth muscle differentiation proteins, including smooth muscle actin. These results demonstrate a requirement for endoglin in descendants of Pax3-expressing vascular cell precursors, and thus provides new insight into the cellular basis underlying adult vascular diseases such as HHT.


Asunto(s)
Vasos Sanguíneos/embriología , Vasos Sanguíneos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neovascularización Fisiológica , Factores de Transcripción Paired Box/metabolismo , Actinas/metabolismo , Alelos , Animales , Aorta/embriología , Aorta/patología , Pérdida del Embrión/metabolismo , Pérdida del Embrión/patología , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Endoglina , Células Endoteliales/metabolismo , Eliminación de Gen , Integrasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Ratones , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Factor de Transcripción PAX3 , Fenotipo , Recombinación Genética/genética , Somitos/irrigación sanguínea , Coloración y Etiquetado
19.
Dev Biol ; 420(1): 148-165, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27671872

RESUMEN

Growth and maturation of the cerebrovasculature is a vital event in neocortical development however mechanisms that control cerebrovascular development remain poorly understood. Mutations in or deletions that include the FOXC1 gene are associated with congenital cerebrovascular anomalies and increased stroke risk in patients. Foxc1 mutant mice display severe cerebrovascular hemorrhage at late gestational ages. While these data demonstrate Foxc1 is required for cerebrovascular development, its broad expression in the brain vasculature combined with Foxc1 mutant's complex developmental defects have made it difficult to pinpoint its function(s). Using global and conditional Foxc1 mutants, we find 1) significant cerebrovascular growth defects precede cerebral hemorrhage and 2) expression of Foxc1 in neural crest-derived meninges and brain pericytes, though not endothelial cells, is required for normal cerebrovascular development. We provide evidence that reduced levels of meninges-derived retinoic acid (RA), caused by defects in meninges formation in Foxc1 mutants, is a major contributing factor to the cerebrovascular growth defects in Foxc1 mutants. We provide data that suggests that meninges-derived RA ensures adequate growth of the neocortical vasculature via regulating expression of WNT pathway proteins and neural progenitor derived-VEGF-A. Our findings offer the first evidence for a role of the meninges in brain vascular development and provide new insight into potential causes of cerebrovascular defects in patients with FOXC1 mutations.


Asunto(s)
Encéfalo/anomalías , Factores de Transcripción Forkhead/genética , Meninges/metabolismo , Mutación/genética , Transducción de Señal , Tretinoina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Wnt/metabolismo , Animales , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/patología , Encéfalo/irrigación sanguínea , Encéfalo/patología , Células Cultivadas , Hemorragia Cerebral/patología , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/patología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Factores de Transcripción Forkhead/metabolismo , Inmunohistoquímica , Integrasas/metabolismo , Meninges/efectos de los fármacos , Ratones , Neocórtex/irrigación sanguínea , Neocórtex/embriología , Neocórtex/patología , Pericitos/efectos de los fármacos , Pericitos/metabolismo , Transducción de Señal/efectos de los fármacos , Tretinoina/farmacología , beta-Galactosidasa/metabolismo
20.
J Biol Chem ; 291(2): 681-90, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26582204

RESUMEN

A-kinase anchoring proteins (AKAPs) represent a family of structurally diverse proteins, all of which bind PKA. A member of this family is glycogen synthase kinase 3ß (GSK3ß) interaction protein (GSKIP). GSKIP interacts with PKA and also directly interacts with GSK3ß. The physiological function of the GSKIP protein in vivo is unknown. We developed and characterized a conditional knock-out mouse model and found that GSKIP deficiency caused lethality at birth. Embryos obtained through Caesarean section at embryonic day 18.5 were cyanotic, suffered from respiratory distress, and failed to initiate breathing properly. Additionally, all GSKIP-deficient embryos showed an incomplete closure of the palatal shelves accompanied by a delay in ossification along the fusion area of secondary palatal bones. On the molecular level, GSKIP deficiency resulted in decreased phosphorylation of GSK3ß at Ser-9 starting early in development (embryonic day 10.5), leading to enhanced GSK3ß activity. At embryonic day 18.5, GSK3ß activity decreased to levels close to that of wild type. Our findings reveal a novel, crucial role for GSKIP in the coordination of GSK3ß signaling in palatal shelf fusion.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Hueso Paladar/embriología , Hueso Paladar/metabolismo , Proteínas Represoras/metabolismo , Alelos , Animales , Fisura del Paladar/embriología , Fisura del Paladar/enzimología , Fisura del Paladar/patología , Pérdida del Embrión/metabolismo , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/embriología , Embrión de Mamíferos/enzimología , Femenino , Regulación del Desarrollo de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta , Hemicigoto , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Hueso Paladar/anomalías , Hueso Paladar/enzimología , Fenotipo , Fosforilación , Fosfoserina/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/deficiencia , Respiración
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda