Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Assist Reprod Genet ; 39(11): 2625-2633, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36264444

RESUMEN

PURPOSE: To report outcome of planned oocyte cryopreservation (POC) in the first 8 years of this treatment in our center. METHODS: A retrospective study in a university-affiliated medical center. RESULTS: A total of 446 women underwent POC during 2011-2018. Fifty-seven (13%) women presented to use these oocytes during the study period (until June 2021). POC was performed at a mean age of 37.9 ± 2.0 (range 33-41). Age at thawing was 43.3 ± 2.1 (range 38-49). A total of 34 (60%) women transferred their oocytes for thawing at other units. Oocyte survival after thawing was significantly higher at our center than following shipping to ancillary sites (78 vs. 63%, p = 0.047). Forty-nine women completed their treatment, either depleting their cryopreserved oocytes without conceiving (36) or attaining a live birth (13)-27% live birth rate per woman. Only one of eleven women who cryopreserved oocytes aged 40 and older had a live birth using thawed oocytes. CONCLUSION: Women should be advised to complete planned oocyte cryopreservation before age 40, given low success rates in women who underwent cryopreservation at advanced reproductive age. In this study, oocyte shipping was associated with lower survival rate. These findings may be relevant for women considering POC and utilization of cryopreserved oocytes.


Asunto(s)
Criopreservación , Transferencia de Embrión , Embarazo , Femenino , Humanos , Masculino , Índice de Embarazo , Estudios Retrospectivos , Oocitos
2.
Int J Mol Sci ; 21(12)2020 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-32604954

RESUMEN

Each follicle represents the basic functional unit of the ovary. From its very initial stage of development, the follicle consists of an oocyte surrounded by somatic cells. The oocyte grows and matures to become fertilizable and the somatic cells proliferate and differentiate into the major suppliers of steroid sex hormones as well as generators of other local regulators. The process by which a follicle forms, proceeds through several growing stages, develops to eventually release the mature oocyte, and turns into a corpus luteum (CL) is known as "folliculogenesis". The task of this review is to define the different stages of folliculogenesis culminating at ovulation and CL formation, and to summarize the most recent information regarding the newly identified factors that regulate the specific stages of this highly intricated process. This information comprises of either novel regulators involved in ovarian biology, such as Ube2i, Phoenixin/GPR73, C1QTNF, and α-SNAP, or recently identified members of signaling pathways previously reported in this context, namely PKB/Akt, HIPPO, and Notch.


Asunto(s)
Hormonas Esteroides Gonadales/metabolismo , Oogénesis , Folículo Ovárico/citología , Ovulación , Transducción de Señal , Animales , Femenino , Humanos , Folículo Ovárico/metabolismo
3.
FASEB J ; 32(4): 2124-2136, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29259033

RESUMEN

Members of the TGF-ß superfamily take part in the control of folliculogenesis. Vasorin (Vasn) is a newly identified negative regulator of TGF-ß signaling whose possible involvement in ovarian physiology has never been studied. Here, we demonstrate that Vasn is expressed in the ovary by somatic cells of follicles, and that its expression is up-regulated by LH. We established a conditional knockout (cKO) mouse model in which Vasn is deleted specifically in granulosa cells of growing follicles from the secondary stage onwards. Using this model, we show that, upon hormonal stimulation, follicle ovulation size is almost 2-fold higher. This enhanced ovulatory response is associated with overactivation of the TGF-ß signaling pathway and a lower number of atretic antral follicles. Of importance, we demonstrate that the number of primordial follicles is reduced in prepubertal cKO mouse ovaries, which suggests that the production of VASN by growing follicles protects the ovarian reserve. Finally, analysis of systemic KO mice revealed that the ovarian reserve is almost 2.5-fold higher, which implies that Vasn may also play a role in primordial follicle formation. Overall, our findings reveal that Vasn is a new regulator that exerts an effect on several key ovarian functions, including folliculogenesis, maintenance of the ovarian reserve, and ovulation.-Rimon-Dahari, N., Heinemann-Yerushalmi, L., Hadas, R., Kalich-Philosoph, L., Ketter, D., Nevo, N., Galiani, D., Dekel, N. Vasorin: a newly identified regulator of ovarian folliculogenesis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de la Membrana/metabolismo , Folículo Ovárico/crecimiento & desarrollo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Células Cultivadas , Femenino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Folículo Ovárico/metabolismo , Reserva Ovárica , Ovulación
4.
J Assist Reprod Genet ; 36(1): 159-164, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30402730

RESUMEN

PURPOSE: To study the outcome of repeated biopsy for pre-implantation genetic testing in case of failed genetic diagnosis in the first biopsy. METHODS: The study group included 81 cycles where embryos underwent re-biopsy because there were no transferable embryos after the first biopsy: in 55 cycles, the first procedure was polar body biopsy (PBs) and the second cleavage-stage (BB); in 26 cycles, the first was BB and the second trophectoderm (BLAST) biopsy. The control group included 77 cycles where embryos underwent successful genetic diagnosis following the first biopsy, matched by maternal age, egg number, genetic inheritance type, and embryonic stage at the first biopsy. We measured genetic diagnosis rate, clinical pregnancy rates (PRs), live-birth rates (LBRs), gestational age, and birth weight. RESULTS: For repeated biopsy, genetic diagnosis was received in 67/81 cycles (82.7%); at a higher rate in PB + BB than in BB + BLAST (49/55, 89.1% and 18/26, 69.2% respectively, p = 0.055). Transferable embryos were found in 47 and 68 cycles in the study and the control groups. PRs/ET were 20/47 (42.6%) and 36/68 (52.9%) (p = 0.27), 16/36 (44.4%) following PB + BB, and 4/11 (36.4%) following BB + BLAST (p = 0.74). LBRs/ET were 13/47 (27.7%) in study group, and 28/68 (41.2%) in the controls (p = 0.14), 10/36 (27.8%) following PB + BB group, and 3/11 (27.3%) following BB + BLAST (p > 0.99). Gestational age and birth weight were similar in all groups. CONCLUSIONS: Re-biopsy of embryos when no genetic diagnosis could be reached following the first biopsy, achieved high rates of genetic diagnosis, pregnancies, and live births.


Asunto(s)
Aneuploidia , Tasa de Natalidad , Implantación del Embrión , Fertilización In Vitro , Enfermedades Genéticas Congénitas/diagnóstico , Pruebas Genéticas/métodos , Diagnóstico Preimplantación/métodos , Adulto , Biopsia , Transferencia de Embrión , Femenino , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/prevención & control , Humanos , Nacimiento Vivo , Embarazo , Índice de Embarazo , Resultado del Tratamiento
5.
BMC Genomics ; 19(1): 28, 2018 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-29310578

RESUMEN

BACKGROUND: Adenosine-to-inosine (A-to-I) RNA editing is an epigenetic modification catalyzed by adenosine deaminases acting on RNA (ADARs), and is especially prevalent in the brain. We used the highly accurate microfluidics-based multiplex PCR sequencing (mmPCR-seq) technique to assess the effects of development and environmental stress on A-to-I editing at 146 pre-selected, conserved sites in the rat prefrontal cortex and amygdala. Furthermore, we asked whether changes in editing can be observed in offspring of stress-exposed rats. In parallel, we assessed changes in ADARs expression levels. RESULTS: In agreement with previous studies, we found editing to be generally higher in adult compared to neonatal rat brain. At birth, editing was generally lower in prefrontal cortex than in amygdala. Stress affected editing at the serotonin receptor 2c (Htr2c), and editing at this site was significantly altered in offspring of rats exposed to prereproductive stress across two generations. Stress-induced changes in Htr2c editing measured with mmPCR-seq were comparable to changes measured with Sanger and Illumina sequencing. Developmental and stress-induced changes in Adar and Adarb1 mRNA expression were observed but did not correlate with editing changes. CONCLUSIONS: Our findings indicate that mmPCR-seq can accurately detect A-to-I RNA editing in rat brain samples, and confirm previous accounts of a developmental increase in RNA editing rates. Our findings also point to stress in adolescence as an environmental factor that alters RNA editing patterns several generations forward, joining a growing body of literature describing the transgenerational effects of stress.


Asunto(s)
Adenosina/metabolismo , Encéfalo/metabolismo , Ambiente , Interacción Gen-Ambiente , Inosina/metabolismo , Edición de ARN , ARN/genética , ARN/metabolismo , Estrés Fisiológico/genética , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Factores de Edad , Animales , Epigénesis Genética , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Especificidad de Órganos/genética , Ratas , Receptor de Serotonina 5-HT2C/genética , Receptor de Serotonina 5-HT2C/metabolismo
6.
Proc Natl Acad Sci U S A ; 111(46): E4972-80, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25368174

RESUMEN

The HDL receptor scavenger receptor, class B type I (SR-BI) controls the structure and fate of plasma HDL. Female SR-BI KO mice are infertile, apparently because of their abnormal cholesterol-enriched HDL particles. We examined the growth and meiotic progression of SR-BI KO oocytes and found that they underwent normal germinal vesicle breakdown; however, SR-BI KO eggs, which had accumulated excess cholesterol in vivo, spontaneously activated, and they escaped metaphase II (MII) arrest and progressed to pronuclear, MIII, and anaphase/telophase III stages. Eggs from fertile WT mice were activated when loaded in vitro with excess cholesterol by a cholesterol/methyl-ß-cyclodextrin complex, phenocopying SR-BI KO oocytes. In vitro cholesterol loading of eggs induced reduction in maturation promoting factor and MAPK activities, elevation of intracellular calcium, extrusion of a second polar body, and progression to meiotic stages beyond MII. These results suggest that the infertility of SR-BI KO females is caused, at least in part, by excess cholesterol in eggs inducing premature activation and that cholesterol can activate WT mouse eggs to escape from MII arrest. Analysis of SR-BI KO female infertility raises the possibility that abnormalities in cholesterol metabolism might underlie some cases of human female infertility of unknown etiology.


Asunto(s)
HDL-Colesterol/metabolismo , Colesterol/toxicidad , Infertilidad Femenina/etiología , Meiosis/efectos de los fármacos , Oocitos/efectos de los fármacos , Receptores Depuradores de Clase B/deficiencia , Animales , Supervivencia Celular , Ácido Egtácico/farmacología , Femenino , Sistema de Señalización de MAP Quinasas , Meiosis/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oocitos/citología , Cuerpos Polares , Receptores Depuradores de Clase B/fisiología , Estroncio/farmacología , beta-Ciclodextrinas/farmacología
7.
FASEB J ; 29(11): 4670-81, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26207029

RESUMEN

Timely degradation of protein regulators of the cell cycle is essential for the completion of cell division. This degradation is promoted by the E3 anaphase-promoting complex/cyclosome (APC/C) and mediated by the E2 ubiquitin-conjugating enzymes (Ube2s). Unlike the ample information gathered regarding the meiotic E3 APC/C, the E2s participating in this cell division have never been studied. We identified Ube2C, -S, and -D3 as the E2 enzymes that regulate APC/C activity during meiosis of mouse oocytes. Their depletion reduces the levels of the first meiotic cytokinesis by 50%, and their overexpression doubles and accelerates its completion (50% as compared with 4% at 11 h). We also demonstrated that these E2s take part in ensuring appropriate spindle formation. It is noteworthy that high levels of Ube2C bring about the resumption of the first meiotic division, regardless of the formation of the spindle, overriding the spindle assembly checkpoint. Thus, alongside their canonical function in protein degradation, Ube2C and -S also control the extrusion of the first polar body. Overall, our study characterizes new regulators and unveils the novel roles they play during the meiotic division. These findings shed light on faithful chromosome segregation in oocytes and may contribute to better understanding of aneuploidy and its consequent genetic malformations.


Asunto(s)
Segregación Cromosómica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Meiosis/fisiología , Cuerpos Polares/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ciclosoma-Complejo Promotor de la Anafase/genética , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Animales , Femenino , Ratones , Cuerpos Polares/citología , Proteolisis , Enzimas Ubiquitina-Conjugadoras/genética
8.
PLoS Genet ; 8(2): e1002477, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22383887

RESUMEN

Fundamental aspects of embryonic and post-natal development, including maintenance of the mammalian female germline, are largely unknown. Here we employ a retrospective, phylogenetic-based method for reconstructing cell lineage trees utilizing somatic mutations accumulated in microsatellites, to study female germline dynamics in mice. Reconstructed cell lineage trees can be used to estimate lineage relationships between different cell types, as well as cell depth (number of cell divisions since the zygote). We show that, in the reconstructed mouse cell lineage trees, oocytes form clusters that are separate from hematopoietic and mesenchymal stem cells, both in young and old mice, indicating that these populations belong to distinct lineages. Furthermore, while cumulus cells sampled from different ovarian follicles are distinctly clustered on the reconstructed trees, oocytes from the left and right ovaries are not, suggesting a mixing of their progenitor pools. We also observed an increase in oocyte depth with mouse age, which can be explained either by depth-guided selection of oocytes for ovulation or by post-natal renewal. Overall, our study sheds light on substantial novel aspects of female germline preservation and development.


Asunto(s)
Envejecimiento , Linaje de la Célula/genética , Células Germinativas , Envejecimiento/genética , Animales , Femenino , Células Germinativas/citología , Células Germinativas/metabolismo , Mutación de Línea Germinal , Células Madre Mesenquimatosas/citología , Ratones , Oogénesis/genética , Especificidad de Órganos , Ovario/citología , Ovario/fisiología , Ovulación
9.
Reproduction ; 148(5): 507-17, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25118304

RESUMEN

Similar expression to FGF (Sef or IL17-RD), is a tumor suppressor and an inhibitor of growth factors as well as of pro-inflammatory cytokine signaling. In this study, we examined the regulation of Sef expression by gonadotropins during ovarian folliculogenesis. In sexually immature mice, in situ hybridization (ISH) localized Sef gene expression to early developing oocytes and granulosa cells (GC) but not to theca cells. Sef was also expressed in mouse ovarian endothelial cells, in the fallopian tube epithelium as well as in adipose tissue venules. SEF protein expression, determined by immunohistochemistry (IHC), correlated well with Sef mRNA expression in GC, while differential expression was noticed in oocytes. High Sef mRNA but undetectable SEF protein levels were observed in the oocytes of primary/secondary follicles, while an inverse correlation was found in the oocytes of preantral and small antral follicles. Sef mRNA expression dropped after pregnant mare's serum gonadotropin (PMSG) administration, peaked at 6-8 h after human chorionic gonadotropin (hCG) treatment, and declined by 12 h after this treatment. ISH and IHC localized the changes to oocytes and mural GC following PMSG treatment, whereas Sef expression increased in mural GC and declined in granulosa-lutein cells upon hCG treatment. The ovarian expression of SEF was confirmed using human samples. ISH localized SEF transcripts to human GC of antral follicles but not to corpora lutea. Furthermore, SEF mRNA was detected in human GC recovered from preovulatory follicles. These results are the first to demonstrate SEF expression in a healthy ovary during folliculogenesis. Hormonal regulation of its expression suggests that SEF may be an important factor involved in intra-ovarian control mechanisms.


Asunto(s)
Células de la Granulosa/metabolismo , Proteínas de la Membrana/metabolismo , Oocitos/metabolismo , Receptores de Interleucina/metabolismo , Animales , Gonadotropina Coriónica/farmacología , Femenino , Fármacos para la Fertilidad/farmacología , Regulación del Desarrollo de la Expresión Génica , Gonadotropinas Equinas/farmacología , Células de la Granulosa/efectos de los fármacos , Humanos , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Oocitos/efectos de los fármacos , ARN Mensajero/metabolismo , Receptores de Interleucina/genética
10.
Reproduction ; 148(1): 87-98, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24700326

RESUMEN

Oocyte quality is a well-established determinant of embryonic fate. However, the molecular participants and biological markers that affect and may predict adequate embryonic development are largely elusive. Our aim was to identify the components of the oocyte molecular machinery that part take in the production of a healthy embryo. For this purpose, we used an animal model, generated by us previously, the oocytes of which do not express Cx43 (Cx43(del/del)). In these mice, oogenesis appears normal, fertilisation does occur, early embryonic development is successful but implantation fails. We used magnetic resonance imaging analysis combined with histological examination to characterise the embryonic developmental incompetence. Reciprocal embryo transfer confirmed that the blastocyst evolved from the Cx43(del/del) oocyte is responsible for the implantation disorder. In order to unveil the genes, the impaired expression of which brings about the development of defective embryos, we carried out a genomic screening of both the oocytes and the resulting blastocysts. This microarray analysis revealed a low expression of Egr1, Rpl21 and Eif4a1 in Cx43(del/del) oocytes and downregulation of Rpl15 and Eif4g2 in the resulting blastocysts. We propose that global deficiencies in genes related to the expression of ribosomal proteins and translation initiation factors in apparently normal oocytes bring about accumulation of defects, which significantly compromise their developmental capacity. The blastocysts resulting from such oocytes, which grow within a confined space until implantation, may be unable to generate enough biological mass to allow their expansion. This information could be implicated to diagnosis and treatment of infertility, particularly to IVF.


Asunto(s)
Blastocisto/metabolismo , Implantación Tardía del Embrión/genética , Regulación del Desarrollo de la Expresión Génica , Oocitos/metabolismo , Biosíntesis de Proteínas/genética , Animales , Conexina 43/deficiencia , Conexina 43/genética , Transferencia de Embrión , Factores Eucarióticos de Iniciación/deficiencia , Factores Eucarióticos de Iniciación/genética , Femenino , Genotipo , Imagen por Resonancia Magnética , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Embarazo , Proteínas Ribosómicas/deficiencia , Proteínas Ribosómicas/genética
11.
Proc Natl Acad Sci U S A ; 108(4): 1462-7, 2011 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-21220312

RESUMEN

Ovulation is stimulated by the preovulatory surge of the pituitary luteinizing hormone (LH). Because the ovulatory response is commonly identified with inflammation, we explored the involvement of reactive oxygen species (ROS) in this process. Our experiments show that administration of broad-range scavengers of oxidative species into the ovarian bursa of mice, hormonally induced to ovulate, significantly reduced the rate of ovulation. LH-induced cumulus mucification/expansion, a necessary requirement for ovulation, was prevented by antioxidants both in vivo and in an ex vivo system of isolated intact ovarian follicles. Along this line, H(2)O(2) fully mimicked the effect of LH, bringing about an extensive mucification/expansion of the follicle-enclosed cumulus-oocyte complexes. Impaired progesterone production was observed in isolated follicles incubated with LH in the presence of the antioxidant agents. Furthermore, LH-stimulated up-regulation of genes, the expression of which is crucial for ovulation, was substantially attenuated upon ROS ablation. This system was also used for demonstrating the role of ROS in phosphorylation and activation of the EGF receptor as well as its downstream effector, p42/44 MAPK. Together, our results provide evidence that ovarian production of ROS is an essential preovulatory signaling event, most probably transiently triggered by LH.


Asunto(s)
Antioxidantes/farmacología , Ovulación/fisiología , Especies Reactivas de Oxígeno/metabolismo , Acetilcisteína/farmacología , Animales , Western Blotting , Hidroxianisol Butilado/farmacología , Femenino , Expresión Génica/efectos de los fármacos , Gonadotropinas/farmacología , Humanos , Peróxido de Hidrógeno/farmacología , Hormona Luteinizante/farmacología , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/metabolismo , Ovulación/efectos de los fármacos , Ovulación/genética , Oxidantes/farmacología , Fosforilación/efectos de los fármacos , Progesterona/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Técnicas de Cultivo de Tejidos
12.
PLoS Genet ; 7(7): e1002192, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21829376

RESUMEN

Stem cell dynamics in vivo are often being studied by lineage tracing methods. Our laboratory has previously developed a retrospective method for reconstructing cell lineage trees from somatic mutations accumulated in microsatellites. This method was applied here to explore different aspects of stem cell dynamics in the mouse colon without the use of stem cell markers. We first demonstrated the reliability of our method for the study of stem cells by confirming previously established facts, and then we addressed open questions. Our findings confirmed that colon crypts are monoclonal and that, throughout adulthood, the process of monoclonal conversion plays a major role in the maintenance of crypts. The absence of immortal strand mechanism in crypts stem cells was validated by the age-dependent accumulation of microsatellite mutations. In addition, we confirmed the positive correlation between physical and lineage proximity of crypts, by showing that the colon is separated into small domains that share a common ancestor. We gained new data demonstrating that colon epithelium is clustered separately from hematopoietic and other cell types, indicating that the colon is constituted of few progenitors and ruling out significant renewal of colonic epithelium from hematopoietic cells during adulthood. Overall, our study demonstrates the reliability of cell lineage reconstruction for the study of stem cell dynamics, and it further addresses open questions in colon stem cells. In addition, this method can be applied to study stem cell dynamics in other systems.


Asunto(s)
Linaje de la Célula , Colon/citología , Células Madre/citología , Animales , Linfocitos B/metabolismo , Linaje de la Célula/genética , Colon/metabolismo , Epitelio/metabolismo , Estudio de Asociación del Genoma Completo , Células Madre Hematopoyéticas , Células Secretoras de Insulina/metabolismo , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Páncreas/citología , Páncreas/metabolismo , Células Madre/metabolismo
13.
JBRA Assist Reprod ; 28(1): 2-8, 2024 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-37850846

RESUMEN

OBJECTIVE: Pre-treatment (PT) therapies in IVF are known to be used as pre-stimulation modality to improve cycle outcomes. This study aims to assess whether PT in GnRH antagonist cycles triggered with GnRH-agonist impact oocyte maturation response. METHODS: Data were retrospectively collected for patients who underwent GnRH antagonist cycle with agonist triggering with and without PT. The patients were allocated to groups according to their PT status. The primary outcome evaluated was suboptimal maturation response. Suboptimal maturation to trigger was defined as no oocyte upon retrieval when adequate response was expected. RESULTS: The study population included 196 patients who underwent GnRH antagonist cycle with agonist triggering. The study group included 69 patients who received PT. The control group included 127 patients with no PT. In univariate analysis, the PT group significantly displayed suboptimal response compared to the controls (p = 0.008). All the patients in the study group with suboptimal response (with or without hCG re-triggering) were treated with GnRH-agonist as PT. Basal and pre-trigger LH values were significantly lower in the study group compared to controls (p < 0.001). Multivariate regression analysis revealed that PT with GnRH agonist was a significant predictor for suboptimal response. CONCLUSIONS: Pre-treatment, and particularly the use of GnRH-agonist as PT in antagonist cycles triggered with agonist, increases the risk of suboptimal response to GnRH-agonist trigger. This might be explained by prolonged pituitary suppression, which lasts beyond the PT cessation.


Asunto(s)
Fertilización In Vitro , Hormona Liberadora de Gonadotropina , Humanos , Estudios Retrospectivos , Inducción de la Ovulación , Oogénesis , Oocitos , Gonadotropina Coriónica
14.
Elife ; 132024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38314803

RESUMEN

Background: Fetal growth restriction (FGR) is a pregnancy complication in which a newborn fails to achieve its growth potential, increasing the risk of perinatal morbidity and mortality. Chronic maternal gestational hypoxia, as well as placental insufficiency are associated with increased FGR incidence; however, the molecular mechanisms underlying FGR remain unknown. Methods: Pregnant mice were subjected to acute or chronic hypoxia (12.5% O2) resulting in reduced fetal weight. Placenta oxygen transport was assessed by blood oxygenation level dependent (BOLD) contrast magnetic resonance imaging (MRI). The placentae were analyzed via immunohistochemistry and in situ hybridization. Human placentae were selected from FGR and matched controls and analyzed by immunohistochemistry (IHC). Maternal and cord sera were analyzed by mass spectrometry. Results: We show that murine acute and chronic gestational hypoxia recapitulates FGR phenotype and affects placental structure and morphology. Gestational hypoxia decreased labyrinth area, increased the incidence of red blood cells (RBCs) in the labyrinth while expanding the placental spiral arteries (SpA) diameter. Hypoxic placentae exhibited higher hemoglobin-oxygen affinity compared to the control. Placental abundance of Bisphosphoglycerate mutase (BPGM) was upregulated in the syncytiotrophoblast and spiral artery trophoblast cells (SpA TGCs) in the murine gestational hypoxia groups compared to the control. Hif1α levels were higher in the acute hypoxia group compared to the control. In contrast, human FGR placentae exhibited reduced BPGM levels in the syncytiotrophoblast layer compared to placentae from healthy uncomplicated pregnancies. Levels of 2,3 BPG, the product of BPGM, were lower in cord serum of human FGR placentae compared to control. Polar expression of BPGM was found in both human and mouse placentae syncytiotrophoblast, with higher expression facing the maternal circulation. Moreover, in the murine SpA TGCs expression of BPGM was concentrated exclusively in the apical cell side, in direct proximity to the maternal circulation. Conclusions: This study suggests a possible involvement of placental BPGM in maternal-fetal oxygen transfer, and in the pathophysiology of FGR. Funding: This work was supported by the Weizmann Krenter Foundation and the Weizmann - Ichilov (Tel Aviv Sourasky Medical Center) Collaborative Grant in Biomedical Research, by the Minerva Foundation, by the ISF KillCorona grant 3777/19.


Asunto(s)
Retardo del Crecimiento Fetal , Placenta , Humanos , Embarazo , Femenino , Ratones , Animales , Placenta/metabolismo , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/metabolismo , Bisfosfoglicerato Mutasa/genética , Bisfosfoglicerato Mutasa/metabolismo , Trofoblastos/metabolismo , Hipoxia/metabolismo , Oxígeno/metabolismo
15.
FASEB J ; 26(11): 4495-505, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22859367

RESUMEN

Completion of the first meiotic division, manifested by extrusion of the first polar body (PBI), depends on proteasomal degradation of cyclin B1 and securin and the subsequent respective CDK1 inactivation and chromosome segregation. We aimed at identifying the polyubiquitin signal that mediates proteasomal action and at a better characterization of the role of CDK1 inactivation at this stage of meiosis. Microinjections of mutated ubiquitin proteins into mouse oocytes revealed that interference with lysine-11 polyubiquitin chains abrogated chromosome segregation and reduced PBI extrusion by 63% as compared to WT ubiquitin-injected controls. Inactivation of CDK1 in oocytes arrested at first metaphase by a proteasome inhibitor fully rescued PBI extrusion. However, removal of CDK1 inhibition failed to allow progression to the second metaphase, rather, inducing PBI reengulfment in 62% of the oocytes. Inhibition of either PLK1 or MEK1/2 during the first anaphase changed spindle dimensions. The PLK1 inhibitor also blocked PBI emission and prevented RhoA translocation. Our results identified lysine-11 rather than the canonic lysine-48 ubiquitin chains as the degradation signal in oocytes resuming meiosis, further disclosing that CDK1 inactivation is necessary and sufficient for PBI emission. This information significantly contributes to our understanding of faulty chromosome segregation that may lead to aneuploidy.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Oocitos/citología , Oocitos/metabolismo , Cuerpos Polares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Animales , Proteína Quinasa CDC2/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Segregación Cromosómica , Citocinesis , Femenino , Regulación Enzimológica de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Meiosis/fisiología , Ratones , Cuerpos Polares/citología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Securina , Transducción de Señal , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo , Quinasa Tipo Polo 1
16.
Reprod Fertil Dev ; 25(3): 484-94, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23206331

RESUMEN

Meiosis in oocytes consists of two consecutive asymmetric cell divisions, each completed by the extrusion of one set of chromosomes into a small polar body. First polar body (PBI) extrusion is triggered by the inactivation of cyclin-dependent kinase 1 (CDK1), following the degradation of its regulatory subunit cyclin B1 by the ubiquitin proteasome pathway. The present review covers the sequence of events leading to PBI extrusion, and compares them to the corresponding events in mitotic cell division. The latest findings regarding the contribution of ubiquitin chain topology, separase, securin, cyclin B1, CDK1, Polo-like kinase 1 and mitogen-activated protein kinase kinase 1/2 to the regulation of meiosis are discussed.


Asunto(s)
Meiosis , Oocitos/metabolismo , Animales , Proteína Quinasa CDC2/metabolismo , Femenino , Humanos , Mamíferos , Oocitos/citología , Oocitos/crecimiento & desarrollo , Oogénesis , Cuerpos Polares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal
17.
Biomedicines ; 11(10)2023 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-37892988

RESUMEN

This multi-center study evaluated a novel microscope system capable of quantitative phase microscopy (QPM) for label-free sperm-cell selection for intracytoplasmic sperm injection (ICSI). Seventy-three patients were enrolled in four in vitro fertilization (IVF) units, where senior embryologists were asked to select 11 apparently normal and 11 overtly abnormal sperm cells, in accordance with current clinical practice, using a micromanipulator and 60× bright field microscopy. Following sperm selection and imaging via QPM, the individual sperm cell was chemically stained per World Health Organization (WHO) 2021 protocols and imaged via bright field microscopy for subsequent manual measurements by embryologists who were blinded to the QPM measurements. A comparison of the two modalities resulted in mean differences of 0.18 µm (CI -0.442-0.808 µm, 95%, STD-0.32 µm) for head length, -0.26 µm (CI -0.86-0.33 µm, 95%, STD-0.29 µm) for head width, 0.17 (CI -0.12-0.478, 95%, STD-0.15) for length-width ratio and 5.7 for acrosome-head area ratio (CI -12.81-24.33, 95%, STD-9.6). The repeatability of the measurements was significantly higher in the QPM modality. Surprisingly, only 19% of the subjectively pre-selected normal cells were found to be normal according to the WHO2021 criteria. The measurements of cells imaged stain-free through QPM were found to be in good agreement with the measurements performed on the reference method of stained cells imaged through bright field microscopy. QPM is non-toxic and non-invasive and can improve the clinical effectiveness of ICSI by choosing sperm cells that meet the strict criteria of the WHO2021.

18.
Cell Cycle ; 21(8): 792-804, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35104175

RESUMEN

Fertilization triggers physiological degradation of maternal-mRNAs, which are then replaced by embryonic transcripts. Ample evidence suggests that Argonaut 2 (AGO2) is a possible post-fertilization regulator of maternal-mRNAs degradation; but its role in degradation of maternal-mRNAs during oocyte maturation remains obscure. Fyn, a member of the Src family kinases (SFKs), and an essential factor in oocyte maturation, was reported to inhibit AGO2 activity in oligodendrocytes. Our aim was to examine the role of Fyn and AGO2 in degradation of maternal-mRNAs during oocyte maturation by either suppressing their activity with SU6656 - an SFKs inhibitor; or by microinjecting DN-Fyn RNA for suppression of Fyn and BCl-137 for suppression of AGO2. Batches of fifteen mouse oocytes or embryos were analyzed by qPCR to measure the expression level of nine maternal-mRNAs that were selected for their known role in oocyte growth, maturation and early embryogenesis. We found that Fyn/SFKs are involved in maintaining the stability of at least four pre-transcribed mRNAs in oocytes at the germinal vesicle (GV) stage, whereas AGO2 had no role at this stage. During in-vivo oocyte maturation, eight maternal-mRNAs were significantly degraded. Inhibition of AGO2 prevented the degreadation of at least five maternal-mRNAs, whereas inhibition of Fyn/SFK prevented degradation of at least five Fyn maternal-mRNAs and two SFKs maternal-mRNAs; pointing at their role in promoting the physiological degradation which occurs during in-vivo oocyte maturation. Our findings imply the involvement of Fyn/SFKs in stabilization of maternal-mRNA at the GV stage and the involvement of Fyn, SFKs and AGO2 in degradation of maternal mRNAs during oocyte maturation.


Asunto(s)
Oogénesis , ARN Mensajero Almacenado , Animales , Ratones , Oocitos/metabolismo , Estabilidad del ARN/genética , ARN Mensajero Almacenado/metabolismo , Familia-src Quinasas/metabolismo
19.
J Clin Invest ; 118(12): 3954-65, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19033665

RESUMEN

Implantation is a key stage during pregnancy, as the fate of the embryo is often decided upon its first contact with the maternal endometrium. Around this time, DCs accumulate in the uterus; however, their role in pregnancy and, more specifically, implantation, remains unknown. We investigated the function of uterine DCs (uDCs) during implantation using a transgenic mouse model that allows conditional ablation of uDCs in a spatially and temporally regulated manner. Depletion of uDCs resulted in a severe impairment of the implantation process, leading to embryo resorption. Depletion of uDCs also caused embryo resorption in syngeneic and T cell-deficient pregnancies, which argues against a failure to establish immunological tolerance during implantation. Moreover, even in the absence of embryos, experimentally induced deciduae failed to adequately form. Implantation failure was associated with impaired decidual proliferation and differentiation. Dynamic contrast-enhanced MRI revealed perturbed angiogenesis characterized by reduced vascular expansion and attenuated maturation. We suggest therefore that uDCs directly fine-tune decidual angiogenesis by providing two critical factors, sFlt1 and TGF-beta1, that promote coordinated blood vessel maturation. Collectively, uDCs appear to govern uterine receptivity, independent of their predicted role in immunological tolerance, by regulating tissue remodeling and angiogenesis. Importantly, our results may aid in understanding the limited implantation success of embryos transferred following in vitro fertilization.


Asunto(s)
Células Dendríticas/inmunología , Implantación del Embrión/inmunología , Embrión de Mamíferos/inmunología , Endometrio/inmunología , Tolerancia Inmunológica/fisiología , Embarazo/inmunología , Animales , Diferenciación Celular/inmunología , Proliferación Celular , Células Dendríticas/citología , Implantación del Embrión/genética , Pérdida del Embrión/genética , Pérdida del Embrión/inmunología , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Endometrio/irrigación sanguínea , Endometrio/citología , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/inmunología , Embarazo/genética , Linfocitos T/citología , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/inmunología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/inmunología
20.
Biol Reprod ; 84(3): 537-45, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20980686

RESUMEN

The importance of placental circulation is exemplified by the correlation of placental size and blood flow with fetal weight and survival during normal and compromised human pregnancies in such conditions as preeclampsia and intrauterine growth restriction (IUGR). Using noninvasive magnetic resonance imaging, we evaluated the role of PKBalpha/AKT1, a major mediator of angiogenesis, on placental vascular function. PKBalpha/AKT1 deficiency reduced maternal blood volume fraction without affecting the integrity of the fetomaternal blood barrier. In addition to angiogenesis, PKBalpha/AKT1 regulates additional processes related to survival and growth. In accordance with reports in adult mice, we demonstrated a role for PKBalpha/AKT1 in regulating chondrocyte organization in fetal long bones. Using tetraploid complementation experiments with PKBalpha/AKT1-expressing placentas, we found that although placental PKBalpha/AKT1 restored fetal survival, fetal PKBalpha/AKT1 regulated fetal size, because tetraploid complementation did not prevent intrauterine growth retardation. Histological examination of rescued fetuses showed reduced liver blood vessel and renal glomeruli capillary density in PKBalpha/Akt1 null fetuses, both of which were restored by tetraploid complementation. However, bone development was still impaired in tetraploid-rescued PKBalpha/Akt1 null fetuses. Although PKBalpha/AKT1-expressing placentas restored chondrocyte cell number in the hypertrophic layer of humeri, fetal PKBalpha/AKT1 was found to be necessary for chondrocyte columnar organization. Remarkably, a dose-dependent phenotype was exhibited for PKBalpha/AKT1 when examining PKBalpha/Akt1 heterozygous fetuses as well as those complemented by tetraploid placentas. The differential role of PKBalpha/AKT1 on mouse fetal survival and growth may shed light on its roles in human IUGR.


Asunto(s)
Tamaño Corporal/genética , Viabilidad Fetal/genética , Feto/fisiología , Placenta/metabolismo , Proteínas Proto-Oncogénicas c-akt/fisiología , Animales , Embrión de Mamíferos , Femenino , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/metabolismo , Retardo del Crecimiento Fetal/patología , Viabilidad Fetal/fisiología , Feto/metabolismo , Edad Gestacional , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Embarazo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA