Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.560
Filtrar
1.
FASEB J ; 38(9): e23632, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38686936

RESUMEN

The upper Müllerian duct (MD) is patterned and specified into two morphologically and functionally distinct organs, the oviduct and uterus. It is known that this regionalization process is instructed by inductive signals from the adjacent mesenchyme. However, the interaction landscape between epithelium and mesenchyme during upper MD development remains largely unknown. Here, we performed single-cell transcriptomic profiling of mouse neonatal oviducts and uteri at the initiation of MD epithelial differentiation (postnatal day 3). We identified major cell types including epithelium, mesenchyme, pericytes, mesothelium, endothelium, and immune cells in both organs with established markers. Moreover, we uncovered region-specific epithelial and mesenchymal subpopulations and then deduced region-specific ligand-receptor pairs mediating mesenchymal-epithelial interactions along the craniocaudal axis. Unexpectedly, we discovered a mesenchymal subpopulation marked by neurofilaments with specific localizations at the mesometrial pole of both the neonatal oviduct and uterus. Lastly, we analyzed and revealed organ-specific signature genes of pericytes and mesothelial cells. Taken together, our study enriches our knowledge of upper MD development, and provides a manageable list of potential genes, pathways, and region-specific cell subtypes for future functional studies.


Asunto(s)
Conductos Paramesonéfricos , Oviductos , Análisis de la Célula Individual , Transcriptoma , Útero , Animales , Femenino , Ratones , Útero/metabolismo , Útero/citología , Conductos Paramesonéfricos/metabolismo , Oviductos/metabolismo , Oviductos/citología , Perfilación de la Expresión Génica , Animales Recién Nacidos , Diferenciación Celular , Mesodermo/metabolismo , Mesodermo/citología , Células Epiteliales/metabolismo , Ratones Endogámicos C57BL , Regulación del Desarrollo de la Expresión Génica
2.
Cell ; 187(4): 981-998.e25, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38325365

RESUMEN

The female reproductive tract (FRT) undergoes extensive remodeling during reproductive cycling. This recurrent remodeling and how it shapes organ-specific aging remains poorly explored. Using single-cell and spatial transcriptomics, we systematically characterized morphological and gene expression changes occurring in ovary, oviduct, uterus, cervix, and vagina at each phase of the mouse estrous cycle, during decidualization, and into aging. These analyses reveal that fibroblasts play central-and highly organ-specific-roles in FRT remodeling by orchestrating extracellular matrix (ECM) reorganization and inflammation. Our results suggest a model wherein recurrent FRT remodeling over reproductive lifespan drives the gradual, age-related development of fibrosis and chronic inflammation. This hypothesis was directly tested using chemical ablation of cycling, which reduced fibrotic accumulation during aging. Our atlas provides extensive detail into how estrus, pregnancy, and aging shape the organs of the female reproductive tract and reveals the unexpected cost of the recurrent remodeling required for reproduction.


Asunto(s)
Envejecimiento , Genitales Femeninos , Animales , Femenino , Ratones , Embarazo , Genitales Femeninos/citología , Genitales Femeninos/metabolismo , Inflamación/metabolismo , Útero/citología , Vagina/citología , Análisis de la Célula Individual
3.
Development ; 150(15)2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37417811

RESUMEN

The invasive trophoblast cell lineages in rat and human share crucial responsibilities in establishing the uterine-placental interface of the hemochorial placenta. These observations have led to the rat becoming an especially useful animal model for studying hemochorial placentation. However, our understanding of similarities or differences between regulatory mechanisms governing rat and human invasive trophoblast cell populations is limited. In this study, we generated single-nucleus ATAC-seq data from gestation day 15.5 and 19.5 rat uterine-placental interface tissues, and integrated the data with single-cell RNA-seq data generated at the same stages. We determined the chromatin accessibility profiles of invasive trophoblast, natural killer, macrophage, endothelial and smooth muscle cells, and compared invasive trophoblast chromatin accessibility with extravillous trophoblast cell accessibility. In comparing chromatin accessibility profiles between species, we found similarities in patterns of gene regulation and groups of motifs enriched in accessible regions. Finally, we identified a conserved gene regulatory network in invasive trophoblast cells. Our data, findings and analysis will facilitate future studies investigating regulatory mechanisms essential for the invasive trophoblast cell lineage.


Asunto(s)
Redes Reguladoras de Genes , Trofoblastos , Animales , Embarazo , Ratas , Núcleo Celular , Cromatina , Placenta/citología , Análisis de Expresión Génica de una Sola Célula , Factores de Transcripción/metabolismo , Trofoblastos/citología , Trofoblastos/metabolismo , Útero/citología , Femenino
4.
Nature ; 619(7970): 595-605, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37468587

RESUMEN

Beginning in the first trimester, fetally derived extravillous trophoblasts (EVTs) invade the uterus and remodel its spiral arteries, transforming them into large, dilated blood vessels. Several mechanisms have been proposed to explain how EVTs coordinate with the maternal decidua to promote a tissue microenvironment conducive to spiral artery remodelling (SAR)1-3. However, it remains a matter of debate regarding which immune and stromal cells participate in these interactions and how this evolves with respect to gestational age. Here we used a multiomics approach, combining the strengths of spatial proteomics and transcriptomics, to construct a spatiotemporal atlas of the human maternal-fetal interface in the first half of pregnancy. We used multiplexed ion beam imaging by time-of-flight and a 37-plex antibody panel to analyse around 500,000 cells and 588 arteries within intact decidua from 66 individuals between 6 and 20 weeks of gestation, integrating this dataset with co-registered transcriptomics profiles. Gestational age substantially influenced the frequency of maternal immune and stromal cells, with tolerogenic subsets expressing CD206, CD163, TIM-3, galectin-9 and IDO-1 becoming increasingly enriched and colocalized at later time points. By contrast, SAR progression preferentially correlated with EVT invasion and was transcriptionally defined by 78 gene ontology pathways exhibiting distinct monotonic and biphasic trends. Last, we developed an integrated model of SAR whereby invasion is accompanied by the upregulation of pro-angiogenic, immunoregulatory EVT programmes that promote interactions with the vascular endothelium while avoiding the activation of maternal immune cells.


Asunto(s)
Intercambio Materno-Fetal , Trofoblastos , Útero , Femenino , Humanos , Embarazo , Arterias/fisiología , Decidua/irrigación sanguínea , Decidua/citología , Decidua/inmunología , Decidua/fisiología , Primer Trimestre del Embarazo/genética , Primer Trimestre del Embarazo/metabolismo , Primer Trimestre del Embarazo/fisiología , Trofoblastos/citología , Trofoblastos/inmunología , Trofoblastos/fisiología , Útero/irrigación sanguínea , Útero/citología , Útero/inmunología , Útero/fisiología , Intercambio Materno-Fetal/genética , Intercambio Materno-Fetal/inmunología , Intercambio Materno-Fetal/fisiología , Factores de Tiempo , Proteómica , Perfilación de la Expresión Génica , Conjuntos de Datos como Asunto , Edad Gestacional
5.
PLoS Genet ; 18(1): e1010018, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35025868

RESUMEN

Approximately 75% of failed pregnancies are considered to be due to embryo implantation failure or defects. Nevertheless, the explicit signaling mechanisms governing this process have not yet been elucidated. Here, we found that conditional deletion of the Shp2 gene in mouse uterine stromal cells deferred embryo implantation and inhibited the decidualization of stromal cells, which led to embryonic developmental delay and to the death of numerous embryos mid-gestation, ultimately reducing female fertility. The absence of Shp2 in stromal cells increased the proliferation of endometrial epithelial cells, thereby disturbing endometrial epithelial remodeling. However, Shp2 deletion impaired the proliferation and polyploidization of stromal cells, which are distinct characteristics of decidualization. In human endometrial stromal cells (hESCs), Shp2 expression gradually increased during the decidualization process. Knockout of Shp2 blocked the decidual differentiation of hESCs, while Shp2 overexpression had the opposite effect. Shp2 knockout inhibited the proliferation of hESCs during decidualization. Whole gene expression profiling analysis of hESCs during the decidualization process showed that Shp2 deficiency disrupted many signaling transduction pathways and gene expression. Analyses of hESCs and mouse uterine tissues confirmed that the signaling pathways extracellular regulated protein kinases (ERK), protein kinase B (AKT), signal transducer and activator of transcription 3 (STAT3) and their downstream transcription factors CCAAT/enhancer binding protein ß (C/EBPß) and Forkhead box transcription factor O1 (FOXO-1) were involved in the Shp2 regulation of decidualization. In summary, these results demonstrate that Shp2 plays a crucial role in stromal decidualization by mediating and coordinating multiple signaling pathways in uterine stromal cells. Our discovery possibly provides a novel key regulator of embryo implantation and novel therapeutic target for pregnancy failure.


Asunto(s)
Decidua/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Útero/citología , Animales , Línea Celular , Proliferación Celular , Implantación del Embrión , Femenino , Eliminación de Gen , Perfilación de la Expresión Génica , Humanos , Ratones , Embarazo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Transducción de Señal , Células del Estroma/citología , Células del Estroma/metabolismo , Útero/metabolismo
6.
Physiol Genomics ; 54(2): 71-85, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34890509

RESUMEN

In cattle, starting 4-5 days after estrus, preimplantation embryonic development occurs in the confinement of the uterine lumen. Cells in the endometrial epithelial layer control the molecular traffic to and from the lumen and, thereby determine luminal composition. Starting early postestrus, endometrial function is regulated by sex steroids, but the effects of progesterone on luminal cells transcription have not been measured in vivo. The first objective was to determine the extent to which progesterone controls transcription in luminal epithelial cells 4 days (D4) after estrus. The second objective was to discover luminal transcripts that predict pregnancy outcomes when the effect of progesterone is controlled. Endometrial luminal epithelial cells were collected from embryo transfer recipients on D4 using a cytological brush and their transcriptome was determined by RNASeq. Pregnancy by embryo transfer was measured on D30 (25 pregnant and 18 nonpregnant). Progesterone concentration on D4 was associated positively (n = 182) and negatively (n = 58) with gene expression. Progesterone-modulated transcription indicated an increase in oxidative phosphorylation, biosynthetic activity, and proliferation of epithelial cells. When these effects of progesterone were controlled, different genes affected positively (n = 22) and negatively (n = 292) odds of pregnancy. These set of genes indicated that a receptive uterine environment was characterized by the inhibition of phosphoinositide signaling and innate immune system responses. A panel of 25 genes predicted the pregnancy outcome with sensitivity and specificity ranging from 64%-96% and 44%-83%, respectively. In conclusion, in the early diestrus, both progesterone-dependent and progesterone-independent mechanisms regulate luminal epithelial transcription associated with pregnancy outcomes in cattle.


Asunto(s)
Endometrio/metabolismo , Células Epiteliales/metabolismo , Progesterona/metabolismo , Transcriptoma/genética , Útero/metabolismo , Animales , Bovinos , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Análisis por Conglomerados , Transferencia de Embrión , Desarrollo Embrionario , Endometrio/citología , Estro/genética , Femenino , Perfilación de la Expresión Génica/métodos , Embarazo , Progesterona/farmacología , RNA-Seq/métodos , Transducción de Señal/genética , Transcriptoma/efectos de los fármacos , Útero/citología
7.
Reprod Toxicol ; 107: 81-89, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34864119

RESUMEN

Zearalenone (ZEN)-contaminated diets induce detrimental effects on the bovine reproduction. Recently, we reported that active sperm induce pro-inflammatory responses in bovine endometrial epithelial cells (BEECs) in vitro. This study aimed to investigate the impact of presence of ZEN on the sperm-uterine crosstalk in vitro. BEECs monolayers were stimulated by ZEN (10, 100, and 1000 ng/mL) for 0, 3, 6, 12, or 24 h and gene expressions were analyzed by real-time PCR. Moreover, BEECs were pre-exposed to ZEN (10, 100, and 1000 ng/mL) for 24 h then, co-incubated with sperm for 6 h. Conditioned media (CM) from a sperm-BEECs co-culture, after pre-exposure to ZEN, were harvested and exploited to challenge either polymorphonuclear cells (PMNs) or sperm. Both PMNs phagocytic activity toward sperm and sperm motility parameters were then assessed. Results showed that ZEN alone induced pro-inflammatory responses in BEECs through the induction of mRNA expressions of pro-inflammatory cytokines (TNFA and IL1B) and PGES1 at different time points. Pre-exposure of BEECs to ZEN, amplified the sperm-triggered upregulation of pro-inflammatory cytokines (TNFA and IL1B) and chemokine IL8 mRNA abundance in BEECs. Sperm-BEECs conditioned media, primed by ZEN, stimulated the PMNs phagocytosis for sperm whereas suppressed sperm motility parameters. Taken together, these findings indicate that the presence of ZEN augments the pro-inflammatory cascade triggered by sperm in BEECs, provokes PMNs phagocytosis for sperm, and reduces sperm motility parameters. Such immunological reactions may create a hostile environment for sperm competence and survival in the bovine uterus, thus impair fertility.


Asunto(s)
Estrógenos no Esteroides/toxicidad , Inflamación , Motilidad Espermática/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Útero , Zearalenona/toxicidad , Animales , Bovinos , Células Cultivadas , Técnicas de Cocultivo , Citocinas/genética , Células Epiteliales/efectos de los fármacos , Femenino , Inflamación/genética , Masculino , Neutrófilos/fisiología , Fagocitosis , Espermatozoides/fisiología , Útero/citología
8.
J Neuroinflammation ; 18(1): 294, 2021 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-34920745

RESUMEN

BACKGROUND: The emerging role of microglia in neurological disorders requires a novel method for obtaining massive amounts of adult microglia. We aim to develop a new method for obtaining bankable and expandable adult-like microglia in mice. METHODS: The head neuroepithelial layer (NEL) that composed of microglial progenitor and neuroepithelial cells at mouse E13.5 was dissected and then cultured or banked. Microglia (MG) isolated from the cultured NEL by magnetic-activated cell sorting system were obtained and named NEL-MG. RESULTS: The NEL included microglia progenitors that proliferate and ramify over time with neuroepithelial cells as feeder. In functional analysis, NEL-MG exhibited microglial functions, such as phagocytosis (microbeads, amyloid ß, synaptosome), migration, and inflammatory response following lipopolysaccharide (LPS) stimulation. NEL was passage cultured and the NEL-MG exhibited a higher expression of microglia signature genes than the neonatal microglia, a widely used in vitro surrogate. Banking or long-term passage culture of NEL did not affect NEL-MG characteristics. Transcriptome analysis revealed that NEL-MG exhibited better conservation of microglia signature genes with a closer fidelity to freshly isolated adult microglia than neonatal microglia. NEL-MG could be re-expandable when they were plated again on neuroepithelial cells. CONCLUSIONS: This new method effectively contributes to obtaining sufficient matured form of microglia (adult-like microglia), even when only a small number of experimental animals are available, leading to a broad application in the field of neuroscience.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Corteza Cerebral/fisiología , Células Epiteliales/fisiología , Perfilación de la Expresión Génica/métodos , Microglía/fisiología , Útero/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Línea Celular , Corteza Cerebral/citología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Útero/citología
9.
Biochem Biophys Res Commun ; 584: 66-72, 2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34768084

RESUMEN

In the adult uterus of mice, rats and humans, the initially closely packed muscle bundles of the inner myometrium (muscular tissue that encircles the endometrium where the conceptus implants) undergo a pregnancy-induced dispersal that is clinically significant and hypothesized to regulate important pregnancy events. However, where, when and how this dispersal occurs, what its functions are, as well as its spatial relationship to the mouse metrial gland/mesometrial lymphoid aggregate of pregnancy (MG/MLAp), are unknown. The MG/MLAp, is a pregnancy-induced uterine structure required for successful rodent pregnancy located mesometrial to (above) the decidua basalis (pregnancy-modified mesometrial endometrium) and defined by its accumulation of maternal lymphocytes known as uterine Natural Killer (uNK) cells. To begin to understand how mouse inner myometrium dispersal (IMD) occurs, we spatiotemporally described it by observing the distribution of its muscle bundles and measuring their volume fraction (VF), as well as the VF of uNKs and stromal cells of inner myometrium. We discovered that (a) IMD (defined as reduction in VF of inner myometrium muscle bundles) is restricted to the mesometrial half of the uterus, is first evident at Embryonic day (E) 5.5 (early postimplantation) but not at E3.5 (preimplantation), further increases between E6.5 and E7.5 and remains unchanged from E7.5 to E10.5, (b) IMD initiation (observed between E3.5 and E5.5) occurs in the absence of uNKs and is associated with VF increases of pre-existing inner myometrium stromal cells and (c) the IMD observed between E6.5 and E7.5 is not associated with VF increases of uNKs or stromal cells. To get functional clues about IMD, we examined whether stromal cells between the dispersed muscle bundles undergo decidualization (important for correct fetomaternal interactions) and provide evidence that they do by E10.5, based on their production of Desmin (decidualization marker). Lastly, we examined whether mouse MG/MLAp only comprises the dispersed inner myometrium or additionally includes the mesometrial triangle (a triangular-like area mesometrial to the inner myometrium at the mesometrium-uterus attachment site), as is the case in rats. Our data supports that the dispersed inner myometrium is the only tissue that makes up the mouse MG/MLAp. In conclusion, we provide novel cellular and spatiotemporal insights about IMD that will contribute to understanding its mechanism and function and allow more informed inter-species comparisons about this process.


Asunto(s)
Decidua/metabolismo , Glándula Metrial/metabolismo , Miometrio/metabolismo , Útero/metabolismo , Animales , Biomarcadores/metabolismo , Proteínas de Unión al Calcio/metabolismo , Decidua/citología , Desmina/metabolismo , Femenino , Inmunohistoquímica , Células Asesinas Naturales/metabolismo , Lectinas/metabolismo , Glándula Metrial/citología , Ratones Endogámicos ICR , Proteínas de Microfilamentos/metabolismo , Miometrio/citología , Embarazo , Células del Estroma/metabolismo , Factores de Tiempo , Útero/citología , Calponinas
10.
Front Immunol ; 12: 712614, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34335628

RESUMEN

The gut microbiota is influenced by environmental factors such as food. Maternal diet during pregnancy modifies the gut microbiota composition and function, leading to the production of specific compounds that are transferred to the fetus and enhance the ontogeny and maturation of the immune system. Prebiotics are fermented by gut bacteria, leading to the release of short-chain fatty acids that can specifically interact with the immune system, inducing a switch toward tolerogenic populations and therefore conferring health benefits. In this study, pregnant BALB/cJRj mice were fed either a control diet or a diet enriched in prebiotics (Galacto-oligosaccharides/Inulin). We hypothesized that galacto-oligosaccharides/inulin supplementation during gestation could modify the maternal microbiota, favoring healthy immune imprinting in the fetus. Galacto-oligosaccharides/inulin supplementation during gestation increases the abundance of Bacteroidetes and decreases that of Firmicutes in the gut microbiota, leading to increased production of fecal acetate, which was found for the first time in amniotic fluid. Prebiotic supplementation increased the abundance of regulatory B and T cells in gestational tissues and in the fetus. Interestingly, these regulatory cells remained later in life. In conclusion, prebiotic supplementation during pregnancy leads to the transmission of specific microbial and immune factors from mother to child, allowing the establishment of tolerogenic immune imprinting in the fetus that may be beneficial for infant health outcomes.


Asunto(s)
Líquido Amniótico/metabolismo , Suplementos Dietéticos , Microbioma Gastrointestinal , Tolerancia Inmunológica , Prebióticos , Preñez , Acetatos/metabolismo , Animales , Subgrupos de Linfocitos B/inmunología , Butiratos/metabolismo , Células Dendríticas/inmunología , Heces/química , Heces/microbiología , Femenino , Feto/inmunología , Humanos , Inulina/administración & dosificación , Inulina/farmacología , Intercambio Materno-Fetal , Ratones , Ratones Endogámicos BALB C , Oligosacáridos/administración & dosificación , Oligosacáridos/farmacología , Placenta/citología , Placenta/inmunología , Embarazo , Resultado del Embarazo , Preñez/inmunología , Preñez/metabolismo , Efectos Tardíos de la Exposición Prenatal , Propionatos/metabolismo , Ribotipificación , Subgrupos de Linfocitos T/inmunología , Útero/citología , Útero/inmunología
11.
Endocrinology ; 162(11)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34402888

RESUMEN

Recent studies have demonstrated that the formation of an implantation chamber composed of a uterine crypt, an implantation-competent blastocyst, and uterine glands is a critical step in blastocyst implantation in mice. Leukemia inhibitory factor (LIF) activates signal transducer and activator of transcription 3 (STAT3) precursors via uterine LIF receptors (LIFRs), allowing successful blastocyst implantation. Our recent study revealed that the role of epithelial STAT3 is different from that of stromal STAT3. However, both are essential for blastocyst attachment, suggesting the different roles of epithelial and stromal LIFR in blastocyst implantation. However, how epithelial and stromal LIFR regulate the blastocyst implantation process remains unclear. To investigate the roles of LIFR in the uterine epithelium and stroma, we generated Lifr-floxed/lactoferrin (Ltf)-iCre (Lifr eKO) and Lifr-floxed/antimüllerian hormone receptor type 2 (Amhr2)-Cre (Lifr sKO) mice with deleted epithelial and stromal LIFR, respectively. Surprisingly, fertility and blastocyst implantation in the Lifr sKO mice were normal despite stromal STAT3 inactivation. In contrast, blastocyst attachment failed, and no implantation chambers were formed in the Lifr eKO mice with epithelial inactivation of STAT3. In addition, normal responsiveness to ovarian hormones was observed in the peri-implantation uteri of the Lifr eKO mice. These results indicate that the epithelial LIFR-STAT3 pathway initiates the formation of implantation chambers, leading to complete blastocyst attachment, and that stromal STAT3 regulates blastocyst attachment without stromal LIFR control. Thus, uterine epithelial LIFR is critical to implantation chamber formation and blastocyst attachment.


Asunto(s)
Implantación del Embrión/genética , Epitelio/metabolismo , Receptores OSM-LIF/fisiología , Útero/metabolismo , Animales , Blastocisto/fisiología , Decidua/fisiología , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Receptores OSM-LIF/genética , Receptores OSM-LIF/metabolismo , Útero/citología
12.
FASEB J ; 35(9): e21812, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34411354

RESUMEN

Blastocysts hatch from the zona pellucida (ZP) to enable implantation into the uterine endometrial epithelium, but little is known regarding the effect of hatching sites on pregnancy outcomes. Murine hatching embryos were categorized into five groups based on initial trophectoderm projection (TEP)/ZP position corresponding to the inner cell mass center. In blastocysts (3.5 dpc) post-12 hours in vitro culture, TEP rates of A-site (44.4%) and B-site (38.6%) embryos were higher than those of C-site (12.5%) and D-site (3.1%) embryos, while the O-site (1.4%) was the lowest (P < .05). Post-ET A-site (55.6%) and B-site (65.6%) birth rates were higher than those of C-site embryos (21.3%) and controls (P < .05). Furthermore, live birth rate of B-site embryos remained higher than C-site embryos (68.8% vs 31.3%; P < .05) when both were transferred into the same recipients. Different TEP site blastocysts exhibited different implantation competences: the implantation rate of C-site embryos was lower than that of both A- and B-site groups (67.7% vs 84.3% and 83.2%, respectively; P < .05) at 2 days post-ET. C-site embryos also had a distinctly higher ratio of developmental defects (47.5%) than A- and B-site embryos (22.5% and 14.6%, respectively), with implantation failure mainly associated with poor birth rate, a finding corroborated by differential gene expression analysis such as LIF, LIFR, and S100a9. Surprisingly, acidified Tyrode's solution (AAH)-treated B-site blastocysts had a significantly increased birth rate (77.1%) than C-site (55.3%) and controls (43.4%). Site specificity and differential gene expression during embryo hatching can be applied in ART screening. More importantly, assisted hatching by AAH is effective and feasible for improving pregnancy and term development, particularly at the B-site, for humans and in animal husbandry.


Asunto(s)
Tasa de Natalidad , Blastocisto/citología , Implantación del Embrión , Trofoblastos/citología , Zona Pelúcida/metabolismo , Animales , Transferencia de Embrión , Femenino , Fertilización In Vitro , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Embarazo , Resultado del Embarazo , Útero/citología
13.
Anim Sci J ; 92(1): e13621, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34448516

RESUMEN

To determine the effects of Follicle Stimulating Hormone (FSH) treatment and subsequent withdrawal on uterine proliferation and estrogen receptor (ESR), Brahman crossbred heifers (n = 12) were twice daily injected with FSH (4, 3, and 2 mg/injection) on Days 17-19 of the estrous cycle (FSH 3 days) and (4 and 3 mg/injection) on Days 17-18 (FSH 2 days) and withdrawal with saline on Day 19 and (4 mg/injection) on Day 17 (FSH 1 day) and withdrawal with saline on Days 18-19. Uterine tissue was subjectively collected on Day 20 and microscopically classified to four regions: endometrial stroma (ES), surface endometrial gland (EG), deep endometrial gland (DG), and myometrium (Myo). The cell proliferation marker, Ki-67, was quantified as labeling index (LI) in uterine regions, and tissues were immunostained to detect ESR2 followed by image analysis. The LI of ES, EG, and DG was greater (P = 0.0018, P = 0.0005, and P = 0.0103; respectively) in heifers received FSH for 3 days. The expression of ESR2 protein on ES and EG was greatest (P < 0.0001 and P = 0.0036, respectively) in FSH 3 days-treated group. Thus, FSH administration during proestrus stimulates uterine cell proliferation, and ESR2 expressions are affected by FSH during proestrus and differentially distributed in the uterine regions.


Asunto(s)
Bovinos/genética , Bovinos/metabolismo , Proliferación Celular/efectos de los fármacos , Ciclo Estral/metabolismo , Ciclo Estral/fisiología , Hormona Folículo Estimulante/administración & dosificación , Hormona Folículo Estimulante/farmacología , Expresión Génica/efectos de los fármacos , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Útero/citología , Útero/metabolismo , Animales , Ciclo Estral/genética , Femenino
14.
Front Immunol ; 12: 607669, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34234770

RESUMEN

Innate lymphoid cells (ILCs) are the most abundant immune cells in the uterine mucosa both before and during pregnancy. Circumstantial evidence suggests they play important roles in regulating placental development but exactly how they contribute to the successful outcome of pregnancy is still unclear. Uterine ILCs (uILCs) include subsets of tissue-resident natural killer (NK) cells and ILCs, and until recently the phenotype and functions of uILCs were poorly defined. Determining the specific roles of each subset is intrinsically challenging because of the rapidly changing nature of the tissue both during the menstrual cycle and pregnancy. Single-cell RNA sequencing (scRNAseq) and high dimensional flow and mass cytometry approaches have recently been used to analyse uILC populations in the uterus in both humans and mice. This detailed characterisation has significantly changed our understanding of the heterogeneity within the uILC compartment. It will also enable key clinical questions to be addressed including whether specific uILC subsets are altered in infertility, miscarriage and pregnancy disorders such as foetal growth restriction and pre-eclampsia. Here, we summarise recent advances in our understanding of the phenotypic and functional diversity of uILCs in non-pregnant endometrium and first trimester decidua, and review how these cells may contribute to successful placental development.


Asunto(s)
Inmunidad Innata , Células Asesinas Naturales/inmunología , Linfocitos/inmunología , Resultado del Embarazo , Útero/citología , Útero/inmunología , Animales , Recuento de Células , Citocinas/inmunología , Endometrio/citología , Endometrio/inmunología , Femenino , Humanos , Células Asesinas Naturales/clasificación , Células Asesinas Naturales/fisiología , Ratones , Fenotipo , Embarazo
15.
JCI Insight ; 6(17)2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34292881

RESUMEN

Retinoic acid (RA) signaling has long been speculated to regulate embryo implantation, because many enzymes and proteins responsible for maintaining RA homeostasis and transducing RA signals are tightly regulated in the endometrium during this critical period. However, due to a lack of genetic data, it was unclear whether RA signaling is truly required for implantation and which specific RA signaling cascades are at play. Herein we utilize a genetic murine model that expresses a dominant-negative form of RA receptor (RAR) specifically in female reproductive organs to show that functional RA signaling is fundamental to female fertility, particularly implantation and decidualization. Reduction in RA signaling activity severely affects the ability of the uterus to achieve receptive status and decidualize, partially through dampening follistatin expression and downstream activin B/bone morphogenetic protein 2 signaling. To confirm translational relevance of these findings to humans, human endometrial stromal cells (hESCs) were treated with a pan-RAR antagonist to show that in vitro decidualization is impaired. RNA interference perturbation of individual RAR transcripts in hESCs revealed that RARα in particular was essential for proper decidualization. These data provide direct functional evidence that uterine RAR-mediated RA signaling was crucial for mammalian embryo implantation, and its disruption led to failure of uterine receptivity and decidualization, resulting in severely compromised fertility.


Asunto(s)
ADN/genética , Endometrio/metabolismo , Mutación , Receptores de Ácido Retinoico/genética , Útero/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Análisis Mutacional de ADN , Endometrio/citología , Femenino , Masculino , Ratones , Ratones Mutantes , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal , Útero/citología
16.
Int J Mol Sci ; 22(14)2021 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-34299317

RESUMEN

Decidualization is a crucial step for human reproduction, which is a prerequisite for embryo implantation, placentation and pregnancy maintenance. Despite rapid advances over recent years, the molecular mechanism underlying decidualization remains poorly understood. Here, we used the mouse as an animal model and generated a single-cell transcriptomic atlas of a mouse uterus during decidualization. By analyzing the undecidualized inter-implantation site of the uterus as a control, we were able to identify global gene expression changes associated with decidualization in each cell type. Additionally, we identified intercellular crosstalk between decidual cells and niche cells, including immune cells, endothelial cells and trophoblast cells. Our data provide a valuable resource for deciphering the molecular mechanism underlying decidualization.


Asunto(s)
Decidua/citología , Decidua/metabolismo , Útero/citología , Útero/metabolismo , Animales , Comunicación Celular/genética , Comunicación Celular/inmunología , Decidua/inmunología , Implantación del Embrión/genética , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Humanos , Ratones , Modelos Animales , Placentación/genética , Embarazo , Mantenimiento del Embarazo/genética , RNA-Seq , Análisis de la Célula Individual , Células del Estroma/citología , Células del Estroma/metabolismo , Transcriptoma , Trofoblastos/citología , Trofoblastos/metabolismo , Útero/inmunología
17.
Cells ; 10(5)2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-34068340

RESUMEN

Here we report the use of a microfluidic system to assess the differential metabolomics of murine embryos cultured with endometrial cells-conditioned media (CM). Groups of 10, 1-cell murine B6C3F1 × B6D2F1 embryos were cultured in the microfluidic device. To produce CM, mouse uterine epithelial cells were cultured in potassium simplex optimized medium (KSOM) for 24 h. Media samples were collected from devices after 5 days of culture with KSOM (control) and CM, analyzed by reverse phase liquid chromatography and untargeted positive ion mode mass spectrometry analysis. Blastocyst rates were significantly higher (p < 0.05) in CM (71.8%) compared to control media (54.6%). We observed significant upregulation of 341 compounds and downregulation of 214 compounds in spent media from CM devices when compared to control. Out of these, 353 compounds were identified showing a significant increased abundance of metabolites involved in key metabolic pathways (e.g., arginine, proline and pyrimidine metabolism) in the CM group, suggesting a beneficial effect of CM on embryo development. The metabolomic study carried out in a microfluidic environment confirms our hypothesis on the potential of uterine epithelial cells to enhance blastocyst development. Further investigations are required to highlight specific pathways involved in embryo development and implantation.


Asunto(s)
Blastocisto/metabolismo , Técnicas de Cultivo de Embriones/instrumentación , Células Epiteliales/metabolismo , Dispositivos Laboratorio en un Chip , Metaboloma , Metabolómica , Técnicas Analíticas Microfluídicas/instrumentación , Comunicación Paracrina , Útero/metabolismo , Animales , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Desarrollo Embrionario , Femenino , Espectrometría de Masas , Ratones , Transducción de Señal , Útero/citología
18.
Int J Mol Sci ; 22(10)2021 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-34068395

RESUMEN

As a crucial step for human reproduction, embryo implantation is a low-efficiency process. Despite rapid advances in recent years, the molecular mechanism underlying embryo implantation remains poorly understood. Here, we used the mouse as an animal model and generated a single-cell transcriptomic atlas of embryo implantation sites. By analyzing inter-implantation sites of the uterus as control, we were able to identify global gene expression changes associated with embryo implantation in each cell type. Additionally, we predicted signaling interactions between uterine luminal epithelial cells and mural trophectoderm of blastocysts, which represent the key mechanism of embryo implantation. We also predicted signaling interactions between uterine epithelial-stromal crosstalk at implantation sites, which are crucial for post-implantation development. Our data provide a valuable resource for deciphering the molecular mechanism underlying embryo implantation.


Asunto(s)
Blastocisto/fisiología , Comunicación Celular , Implantación del Embrión , Desarrollo Embrionario , Análisis de la Célula Individual/métodos , Transcriptoma , Útero/fisiología , Animales , Blastocisto/citología , Femenino , Masculino , Ratones , Transducción de Señal , Útero/citología
19.
Commun Biol ; 4(1): 749, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-34140619

RESUMEN

The uterus is the organ for embryo implantation and fetal development. Most current models of the uterus are centred around capturing its function during later stages of pregnancy to increase the survival in pre-term births. However, in vitro models focusing on the uterine tissue itself would allow modelling of pathologies including endometriosis and uterine cancers, and open new avenues to investigate embryo implantation and human development. Motivated by these key questions, we discuss how stem cell-based uteri may be engineered from constituent cell parts, either as advanced self-organising cultures, or by controlled assembly through microfluidic and print-based technologies.


Asunto(s)
Células Madre/fisiología , Ingeniería de Tejidos/métodos , Útero/citología , Útero/fisiología , Animales , Implantación del Embrión/fisiología , Femenino , Desarrollo Fetal/fisiología , Humanos , Modelos Biológicos , Placenta/fisiología , Embarazo , Primates , Andamios del Tejido
20.
Biomed Mater ; 16(4)2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-33946053

RESUMEN

Data on how the immune system reacts to decellularized scaffolds after implantation is scarce and difficult to interpret due to many heterogeneous parameters such as tissue-type match, decellularization method and treatment application. The engraftment of these scaffolds must prove safe and that they remain inert to the recipient's immune system to enable successful translational approaches and potential future clinical evaluation. Herein, we investigated the immune response after the engraftment of three decellularized scaffold types that previously showed potential to repair a uterine injury in the rat. Protocol (P) 1 and P2 were based on Triton-X100 and generated scaffolds containing 820 ng mg-1and 33 ng mg-1donor DNA per scaffold weight, respectively. Scaffolds obtained with a sodium deoxycholate-based protocol (P3) contained 160 ng donor DNA per mg tissue. The total number of infiltrating cells, and the population of CD45+leukocytes, CD4+T-cells, CD8a+cytotoxic T-cells, CD22+B-cells, NCR1+NK-cells, CD68+and CD163+macrophages were quantified on days 5, 15 and 30 after a subcutaneous allogenic (Lewis to Sprague Dawley) transplantation. Gene expression for the pro-inflammatory cytokines INF-γ, IL-1ß, IL-2, IL-6 and TNF were also examined. P1 scaffolds triggered an early immune response that may had been negative for tissue regeneration but it was stabilized after 30 d. Conversely, P3 initiated a delayed immune response that appeared negative for scaffold survival. P2 scaffolds were the least immunogenic and remained similar to autologous tissue implants. Hence, an effective decellularization protocol based on a mild detergent was advantageous from an immunological perspective and appears the most promising for futurein vivouterus bioengineering applications.


Asunto(s)
Matriz Extracelular Descelularizada , Andamios del Tejido , Trasplante Homólogo , Útero , Animales , Bioingeniería , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Matriz Extracelular Descelularizada/efectos adversos , Matriz Extracelular Descelularizada/química , Femenino , Inflamación/inducido químicamente , Inflamación/metabolismo , Ratas , Ratas Endogámicas Lew , Ratas Sprague-Dawley , Andamios del Tejido/efectos adversos , Andamios del Tejido/química , Trasplante Homólogo/efectos adversos , Trasplante Homólogo/métodos , Útero/citología , Útero/trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...