Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 793
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Proc Natl Acad Sci U S A ; 119(38): e2200252119, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36095212

RESUMEN

In humans, the uterus undergoes a dramatic transformation to form an endometrial stroma-derived secretory tissue, termed decidua, during early pregnancy. The decidua secretes various factors that act in an autocrine/paracrine manner to promote stromal differentiation, facilitate maternal angiogenesis, and influence trophoblast differentiation and development, which are critical for the formation of a functional placenta. Here, we investigated the mechanisms by which decidual cells communicate with each other and with other cell types within the uterine milieu. We discovered that primary human endometrial stromal cells (HESCs) secrete extracellular vesicles (EVs) during decidualization and that this process is controlled by a conserved HIF2α-RAB27B pathway. Mass spectrometry revealed that the decidual EVs harbor a variety of protein cargo, including cell signaling molecules, growth modulators, metabolic regulators, and factors controlling endothelial cell expansion and remodeling. We tested the hypothesis that EVs secreted by the decidual cells mediate functional communications between various cell types within the uterus. We demonstrated that the internalization of EVs, specifically those carrying the glucose transporter 1 (GLUT1), promotes glucose uptake in recipient HESCs, supporting and advancing the decidualization program. Additionally, delivery of HESC-derived EVs into human endothelial cells stimulated their proliferation and led to enhanced vascular network formation. Strikingly, stromal EVs also promoted the differentiation of trophoblast stem cells into the extravillous trophoblast lineage. Collectively, these findings provide a deeper understanding of the pleiotropic roles played by EVs secreted by the decidual cells to ensure coordination of endometrial differentiation and angiogenesis with trophoblast function during the progressive phases of decidualization and placentation.


Asunto(s)
Decidua , Vesículas Extracelulares , Trofoblastos , Diferenciación Celular , Decidua/citología , Decidua/fisiología , Células Endoteliales/citología , Células Endoteliales/fisiología , Vesículas Extracelulares/fisiología , Femenino , Humanos , Neovascularización Fisiológica , Embarazo , Células del Estroma/citología , Células del Estroma/fisiología , Trofoblastos/citología , Trofoblastos/fisiología
3.
Development ; 147(6)2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32184271

RESUMEN

Reproductive decline in older female mice can be attributed to a failure of the uterus to decidualise in response to steroid hormones. Here, we show that normal decidualisation is associated with significant epigenetic changes. Notably, we identify a cohort of differentially methylated regions (DMRs), most of which gain DNA methylation between the early and late stages of decidualisation. These DMRs are enriched at progesterone-responsive gene loci that are essential for reproductive function. In female mice nearing the end of their reproductive lifespan, DNA methylation fidelity is lost at a number of CpG islands (CGIs) resulting in CGI hypermethylation at key decidualisation genes. Importantly, this hypermethylated state correlates with the failure of the corresponding genes to become transcriptionally upregulated during the implantation window. Thus, age-associated DNA methylation changes may underlie the decidualisation defects that are a common occurrence in older females. Alterations to the epigenome of uterine cells may therefore contribute significantly to the reproductive decline associated with advanced maternal age.


Asunto(s)
Envejecimiento/genética , Implantación del Embrión/genética , Epigénesis Genética/fisiología , Reproducción/fisiología , Animales , Células Cultivadas , Islas de CpG/genética , Metilación de ADN/fisiología , Decidua/fisiología , Embrión de Mamíferos , Femenino , Masculino , Edad Materna , Ratones , Ratones Endogámicos C57BL , Embarazo , Reproducción/genética
4.
Cell Biol Toxicol ; 39(3): 1077-1098, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-34773530

RESUMEN

Uterine deficiency of Dnmt3b impairs decidualization and consequent embryo implantation defects. Recent advances in molecular technologies have allowed the unprecedented mapping of epigenetic modifications during embryo implantation. DNA methyltransferase 3a (DNMT3A) and DNMT3B are responsible for establishing DNA methylation patterns produced through their de novo-type DNA methylation activity in implantation stage embryos and during germ cell differentiation. It was reported that conditional knockout of Dnmt3a in the uterus does not markedly affect endometrial function during embryo implantation, but the tissue-specific functions of Dnmt3b in the endometrium during embryo implantation remain poorly understood to investigate the role of Dnmt3b during peri-implantation period. Here, we generated Dnmt3b conditional knockout (Dnmt3bd/d) female mice using progesterone receptor-Cre mice and examined the role of Dnmt3b during embryo implantation. Dnmt3bd/d female mice exhibited compromised fertility, which was associated with defective decidualization, but not endometrial receptivity. Furthermore, results showed loss of Dnmt3b did not lead to altered genomic methylation patterns of the decidual endometrium during early pregnancy. Transcriptome sequencing analysis of uteri from day 6 pregnant mice identified phosphoglycerate kinase 1 (Pgk1) as one of the most variable genes in Dnmt3bd/d decidual endometrium. Potential roles of PGK1 in the decidualization process during early pregnancy were confirmed. Lastly, the compromised decidualization upon the downregulation of Dnmt3b could be reversed by overexpression of Pgk1. Collectively, our findings indicate that uterine deficiency of Dnmt3b impairs decidualization and consequent embryo implantation defects.


Asunto(s)
Decidua , Útero , Animales , Femenino , Ratones , Embarazo , Decidua/fisiología , Metilación de ADN/genética , Implantación del Embrión/fisiología , Endometrio/metabolismo , ADN Metiltransferasa 3B
5.
Development ; 146(22)2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31776138

RESUMEN

The placenta is essential for normal in utero development in mammals. In humans, defective placental formation underpins common pregnancy disorders such as pre-eclampsia and fetal growth restriction. The great variation in placental types across mammals means that animal models have been of limited use in understanding human placental development. However, new tools for studying human placental development, including 3D organoids, stem cell culture systems and single cell RNA sequencing, have brought new insights into this field. Here, we review the morphological, molecular and functional aspects of human placental formation, with a focus on the defining cell of the placenta - the trophoblast.


Asunto(s)
Placenta/fisiología , Placentación , Trofoblastos/fisiología , Animales , Bioingeniería , Técnicas de Cultivo de Célula , Diferenciación Celular , Linaje de la Célula , Decidua/fisiología , Endometrio/patología , Femenino , Retardo del Crecimiento Fetal , Humanos , Sistema Inmunológico , Leucocitos/citología , Ratones , Organoides , Placenta/citología , Preeclampsia/fisiopatología , Embarazo , Complicaciones del Embarazo , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Trofoblastos/citología , Útero/patología
6.
Biol Reprod ; 107(5): 1166-1176, 2022 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-35947987

RESUMEN

Human endometrial and decidual stromal cells are the same cells in different environments (nonpregnancy and pregnancy, respectively). Although some authors consider decidual stromal cells to arise solely from the differentiation of endometrial stromal cells, this is a debatable issue given that decidualization processes do not end with the formation of the decidua, as shown by the presence of stromal cells from both the endometrium and decidua in both undifferentiated (nondecidualized) and decidualized states. Furthermore, recent functional and transcriptomic results have shown that there are differences in the decidualization process of endometrial and decidual stromal cells, with the latter having a greater decidualization capacity than the former. These differences suggest that in the terminology and study of their characteristics, endometrial and decidual stromal cells should be clearly distinguished, as should their undifferentiated or decidualized status. There is, however, considerable confusion in the designation and identification of uterine stromal cells. This confusion may impede a judicious understanding of the functional processes in normal and pathological situations. In this article, we analyze the different terms used in the literature for different types of uterine stromal cells, and propose that a combination of differentiation status (undifferentiated, decidualized) and localization (endometrium, decidua) criteria should be used to arrive at a set of accurate, unambiguous terms. The cell identity of uterine stromal cells is also a debatable issue: phenotypic, functional, and transcriptomic studies in recent decades have related these cells to different established cells. We discuss the relevance of these associations in normal and pathological situations.


Asunto(s)
Decidua , Endometrio , Embarazo , Femenino , Humanos , Decidua/fisiología , Células del Estroma , Diferenciación Celular , Células Cultivadas
7.
Biol Reprod ; 107(2): 529-545, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35357464

RESUMEN

Uterine dysfunctions lead to fertility disorders and pregnancy complications. Normal uterine functions at pregnancy depend on crosstalk among multiple cell types in uterine microenvironments. Here, we performed the spatial transcriptomics and single-cell RNA-seq assays to determine local gene expression profiles at the embryo implantation site of the mouse uterus on pregnancy day 7.5 (D7.5). The spatial transcriptomic annotation identified 11 domains of distinct gene signatures, including a mesometrial myometrium, an anti-mesometrial myometrium, a mesometrial decidua enriched with natural killer cells, a vascular sinus zone for maternal vessel remodeling, a fetal-maternal interface, a primary decidual zone, a transition decidual zone, a secondary decidual zone, undifferentiated stroma, uterine glands, and the embryo. The scRNA-Seq identified 12 types of cells in the D7.5 uterus including three types of stromal fibroblasts with differentiated and undifferentiated markers, one cluster of epithelium including luminal and glandular epithelium, mesothelium, endothelia, pericytes, myelomonocytic cell, natural killer cells, and lymphocyte B. These single-cell RNA signatures were then utilized to deconvolute the cell-type compositions of each individual uterine microenvironment. Functional annotation assays on spatial transcriptomic data revealed uterine microenvironments with distinguished metabolic preferences, immune responses, and various cellular behaviors that are regulated by region-specific endocrine and paracrine signals. Global interactome among regions is also projected based on the spatial transcriptomic data. This study provides high-resolution transcriptome profiles with locality information at the embryo implantation site to facilitate further investigations on molecular mechanisms for normal pregnancy progression.


Asunto(s)
Transcriptoma , Útero , Animales , Decidua/fisiología , Implantación del Embrión/genética , Epitelio , Femenino , Células Asesinas Naturales , Ratones , Miometrio , Embarazo , Útero/metabolismo
8.
Biol Reprod ; 106(1): 155-172, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-34591094

RESUMEN

The decidua is a hallmark of reproduction in many placental mammals. Differentiation of decidual stromal cells is known to be induced by progesterone and the cyclic AMP/protein kinase A (cAMP/PKA) pathway. Several candidates have been identified as the physiological stimulus for adenylyl cyclase activation, but their relative importance remains unclear. To bypass this uncertainty, the standard approach for in vitro experiments uses membrane-permeable cAMP and progestin. We phylogenetically infer that prostaglandin E2 (PGE2) likely was the signal that ancestrally induced decidualization in conjunction with progesterone. This suggests that PGE2 and progestin should be able to activate the core gene regulatory network of decidual cells. To test this prediction, we performed a genome-wide study of gene expression in human endometrial fibroblasts decidualized with PGE2 and progestin. Comparison to a cAMP-based protocol revealed shared activation of core decidual genes and decreased induction of senescence-associated genes. Single-cell transcriptomics of PGE2-mediated decidualization revealed a distinct, early-activated state transitioning to a differentiated decidual state. PGE2-mediated decidualization was found to depend upon progestin-dependent induction of PGE2 receptor 2 (PTGER2) which in turn leads to PKA activation upon PGE2 stimulation. Progesterone-dependent induction of PTGER2 is absent in opossum, an outgroup taxon of placental mammals which is incapable of decidualization. Together, these findings suggest that the origin of decidualization involved the evolution of progesterone-dependent activation of the PGE2/PTGER2/PKA axis, facilitating entry into a PKA-dominant rather than AKT-dominant cellular state. We propose the use of PGE2 for in vitro decidualization as an alternative to 8-Br-cAMP.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Decidua/citología , Dinoprostona/farmacología , Línea Celular Transformada , Células Cultivadas , AMP Cíclico/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Decidua/fisiología , Endometrio/citología , Endometrio/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Acetato de Medroxiprogesterona/farmacología , Embarazo , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual
9.
Mol Reprod Dev ; 89(5-6): 256-268, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35474595

RESUMEN

Decidualization is an essential process for embryo implantation and maintenance of pregnancy, and abnormal decidualization contributed to several pregnancy disorders like a miscarriage. The objective of this study was to explore the regulation and function of CD55 in human decidualization. By immunohistochemical staining, it was found that CD55 expression was higher in first-trimester decidua than in the endometrium. In both primary endometrial stromal cells and immortalized cell line T-hESCs, CD55 was upregulated by induction of in vitro decidualization with medroxyprogesterone acetate (MPA) and 8-Br-cAMP. During decidualization in vitro, CD55 was stimulated by 8-Br-cAMP in a time- and concentration-dependent manner, which was reversed by a PKA inhibitor H89 and partially by an AKT activator SC79. Knocking down CD55 expression diminished the expression of decidualization markers prolactin (PRL) and insulin-like growth factor-binding protein 1 (IGFBP1), accompanied by inhibition of Src, aberrant activation of ERK and decreased expression of several decidualization-related genes, including FOXO1, EGFR, and STAT3. Furthermore, the decidua of unexplained miscarriage women and the endometrium of unexplained infertile women both exhibited decreased CD55 expression. Collectively, these findings revealed that 8-Br-cAMP promotes CD55 expression via PKA activation and AKT dephosphorylation, and decreased CD55 impairs decidualization by inactivation of Src, aberrant activation of ERK pathway, and compromised expression of decidualization-related genes, indicating that CD55 deficiency may contribute to the pathogenesis of spontaneous miscarriage and infertility.


Asunto(s)
Aborto Espontáneo , Antígenos CD55 , Decidua , Infertilidad Femenina , Aborto Espontáneo/metabolismo , Antígenos CD55/metabolismo , Células Cultivadas , Decidua/fisiología , Endometrio/fisiología , Femenino , Humanos , Infertilidad Femenina/metabolismo , Sistema de Señalización de MAP Quinasas , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células del Estroma/metabolismo
10.
Proc Natl Acad Sci U S A ; 116(39): 19541-19551, 2019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31501330

RESUMEN

Failure of embryo implantation accounts for a significant percentage of female infertility. Exquisitely coordinated molecular programs govern the interaction between the competent blastocyst and the receptive uterus. Decidualization, the rapid proliferation and differentiation of endometrial stromal cells into decidual cells, is required for implantation. Decidualization defects can cause poor placentation, intrauterine growth restriction, and early parturition leading to preterm birth. Decidualization has not yet been systematically studied at the genetic level due to the lack of a suitable high-throughput screening tool. Herein we describe the generation of an immortalized human endometrial stromal cell line that uses yellow fluorescent protein under the control of the prolactin promoter as a quantifiable visual readout of the decidualization response (hESC-PRLY cells). Using this cell line, we performed a genome-wide siRNA library screen, as well as a screen of 910 small molecules, to identify more than 4,000 previously unrecognized genetic and chemical modulators of decidualization. Ontology analysis revealed several groups of decidualization modulators, including many previously unappreciated transcription factors, sensory receptors, growth factors, and kinases. Expression studies of hits revealed that the majority of decidualization modulators are acutely sensitive to ovarian hormone exposure. Gradient treatment of exogenous factors was used to identify EC50 values of small-molecule hits, as well as verify several growth factor hits identified by the siRNA screen. The high-throughput decidualization reporter cell line and the findings described herein will aid in the development of patient-specific treatments for decidualization-based recurrent pregnancy loss, subfertility, and infertility.


Asunto(s)
Decidua/metabolismo , Endometrio/metabolismo , Infertilidad Femenina/metabolismo , Blastocisto/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Decidua/fisiología , Implantación del Embrión/fisiología , Endometrio/patología , Femenino , Fertilidad , Estudio de Asociación del Genoma Completo , Humanos , Infertilidad Femenina/genética , Embarazo , Prolactina/metabolismo , ARN Interferente Pequeño/genética , Receptores de Progesterona/genética , Células del Estroma/metabolismo , Útero/metabolismo , Útero/patología
11.
Biol Reprod ; 104(3): 539-547, 2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33284968

RESUMEN

The endometrium undergoes a pregnancy-delivery-repair cycle multiple times during the reproductive lifespan in females. Decidualization is one of the critical events for the success of this essential process. We have previously reported that Notch1 is essential for artificial decidualization in mice. However, in a natural pregnancy, the deletion of Notch1 (PgrCre/+Notch1f/f, or Notch1d/d) only affects female fertility in the first 30 days of a 6-month fertility test, but not the later stages. In the present study, we undertook a closer evaluation at the first pregnancy of these mice to attempt to understand this puzzling phenomenon. We observed a large number of pregnancy losses in Notch1d/d mice in their first pregnancy, which led to the subfertility observed in the first 30 days of the fertility test. We then demonstrated that the initial pregnancy loss is a consequence of impaired decidualization. Furthermore, we identified a group of genes that contribute to Notch1 regulated decidualization in a natural pregnancy. Gene ontogeny analysis showed that these differentially expressed genes in the natural pregnancy are involved in cell-cell and cell-matrix interactions, different from genes that have been previously identified from the artificial decidualization model, which contribute to cell proliferation and apoptosis. In summary, we determined that Notch1 is essential for normal decidualization in the mouse uterus only in the first pregnancy but not in subsequent ones.


Asunto(s)
Decidua/fisiología , Regulación de la Expresión Génica/fisiología , Preñez , Receptor Notch1/metabolismo , Aborto Veterinario/genética , Animales , Proliferación Celular , Implantación del Embrión/genética , Femenino , Ratones , Ratones Noqueados , Embarazo , Preñez/genética , Preñez/metabolismo , Receptor Notch1/genética , Transducción de Señal , Transcriptoma
12.
Reproduction ; 162(6): 461-472, 2021 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-34591784

RESUMEN

As a multifunctional transcription factor, YY1 regulates the expression of many genes essential for early embryonic development. RTCB is an RNA ligase that plays a role in tRNA maturation and Xbp1 mRNA splicing. YY1 can bind in vitro to the response element in the proximal promoter of Rtcb and regulate Rtcb promoter activity. However, the in vivo regulation and whether these two genes are involved in the mother-fetal dialogue during early pregnancy remain unclear. In this study, we validated that YY1 bound in vivo to the proximal promoter of Rtcb in mouse uterus of early pregnancy. Moreover, via building a variety of animal models, our study suggested that both YY1 and RTCB might play a role in mouse uterus decidualization and embryo implantation during early pregnancy.


Asunto(s)
Aminoacil-ARNt Sintetasas/metabolismo , Implantación del Embrión , Factores de Transcripción , Factor de Transcripción YY1/metabolismo , Animales , Decidua/fisiología , Implantación del Embrión/fisiología , Femenino , Ratones , Embarazo , Empalme del ARN , Factores de Transcripción/genética , Útero
13.
Reproduction ; 161(4): 477-487, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33606664

RESUMEN

Recurrent pregnancy loss (RPL) is one of the most common complications of early pregnancy associated in most cases with local or systemic immune abnormalities such as the diminished proportion of regulatory T cells (Tregs). Mesenchymal stem cells (MSCs) have been shown to modulate the immune responses by de novo induction and expansion of Tregs. In this study, we analyzed the molecular and cellular mechanisms involved in Treg-associated pregnancy protection following MSCs administration in an abortion-prone mouse mating. In a case-control study, syngeneic abdominal fat-derived MSCs were administered intraperitoneally (i.p) to the DBA/2-mated CBA/J female mice on day 4.5 of pregnancy. Abortion rate, Tregs proportion in spleen and inguinal lymph nodes, Ho1, Foxp3, Pd1 and Ctla4 genes expression at the feto-maternal interface were then measured on day 13.5 of pregnancy using flow cytometry and quantitative RT-PCR, respectively. The abortion rate in MSCs-treated mice reduced significantly and normalized to the level observed in normal pregnant animals. We demonstrated a significant induction of Tregs in inguinal lymph nodes but not in the spleen following MSCs administration. Administration of MSCs remarkably upregulated the expression of Ho1, Foxp3, Pd1 and Ctla4 genes in both placenta and decidua. Here, we show that MSCs therapy could protect the fetus in the abortion-prone mice through Tregs expansion and upregulation of Treg-related genes. These events could establish an immune-privileged microenvironment, which participates in the regulation of detrimental maternal immune responses against the semi-allogeneic fetus.


Asunto(s)
Aborto Espontáneo/patología , Decidua/fisiología , Intercambio Materno-Fetal , Células Madre Mesenquimatosas/fisiología , Linfocitos T Reguladores/inmunología , Aborto Inducido , Aborto Espontáneo/inmunología , Aborto Espontáneo/metabolismo , Animales , Citocinas/metabolismo , Decidua/citología , Femenino , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos CBA , Ratones Endogámicos DBA , Embarazo , Linfocitos T Reguladores/citología
14.
Reprod Biol Endocrinol ; 19(1): 96, 2021 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-34176510

RESUMEN

BACKGROUND: Successful human embryo implantation requires the differentiation of endometrial stromal cells (ESCs) into decidual cells during a process called decidualization. ESCs express specific markers of decidualization, including prolactin, insulin-like growth factor-binding protein-1 (IGFBP-1), and connexin-43. Decidual cells also control of trophoblast invasion by secreting various factors, such as matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases. Preimplantation factor (PIF) is a recently identified, embryo-derived peptide with activities at the fetal-maternal interface. It creates a favorable pro-inflammatory environment in human endometrium and directly controls placental development by increasing the human trophoblastic cells' ability to invade the endometrium. We hypothesized that PIF's effects on the endometrium counteract its pro-invasive effects. METHODS: We tested sPIF effect on the expression of three decidualization markers by RT-qPCR and/or immunochemiluminescence assay. We examined sPIF effect on human ESC migration by performing an in vitro wound healing assay. We analyzed sPIF effect on endometrial control of human trophoblast invasion by performing a zymography and an invasion assay. RESULTS: Firstly, we found that a synthetic analog of PIF (sPIF) significantly upregulates the mRNA expression of IGFBP-1 and connexin-43, and prolactin secretion in ESCs - suggesting a pro-differentiation effect. Secondly, we showed that the HTR-8/SVneo trophoblastic cell line's invasive ability was low in the presence of conditioned media from ESCs cultured with sPIF. Thirdly, this PIF's anti-invasive action was associated with a specifically decrease in MMP-9 activity. CONCLUSION: Taken as a whole, our results suggest that PIF accentuates the decidualization process and the production of endometrial factors that limit trophoblast invasion. By controlling both trophoblast and endometrial cells, PIF therefore appears to be a pivotal player in the human embryo implantation process.


Asunto(s)
Decidua/citología , Decidua/efectos de los fármacos , Endometrio/citología , Endometrio/efectos de los fármacos , Proteínas Gestacionales/administración & dosificación , Trofoblastos/efectos de los fármacos , Adulto , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Decidua/fisiología , Endometrio/fisiología , Femenino , Humanos , Células del Estroma/efectos de los fármacos , Células del Estroma/fisiología , Trofoblastos/fisiología
15.
PLoS Biol ; 16(8): e2005594, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30142145

RESUMEN

Among animal species, cell types vary greatly in terms of number and kind. The number of cell types found within an organism differs considerably between species, and cell type diversity is a significant contributor to differences in organismal structure and function. These observations suggest that cell type origination is a significant source of evolutionary novelty. The molecular mechanisms that result in the evolution of novel cell types, however, are poorly understood. Here, we show that a novel cell type of eutherians mammals, the decidual stromal cell (DSC), evolved by rewiring an ancestral cellular stress response. We isolated the precursor cell type of DSCs, endometrial stromal fibroblasts (ESFs), from the opossum Monodelphis domestica. We show that, in opossum ESFs, the majority of decidual core regulatory genes respond to decidualizing signals but do not regulate decidual effector genes. Rather, in opossum ESFs, decidual transcription factors function in apoptotic and oxidative stress response. We propose that rewiring of cellular stress responses was an important mechanism for the evolution of the eutherian decidual cell type.


Asunto(s)
Decidua/fisiología , Estrés Fisiológico/fisiología , Animales , Evolución Biológica , Endometrio/fisiología , Evolución Molecular , Femenino , Fibroblastos , Mamíferos , Monodelphis/fisiología , Estrés Fisiológico/genética , Células del Estroma/metabolismo , Células del Estroma/fisiología , Factores de Transcripción/metabolismo
16.
Cell Mol Life Sci ; 77(11): 2091-2101, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31813015

RESUMEN

Decidualization is a critical event for the blastocyst implantation, placental development and fetal growth and the normal term. In mice, the embryo implantation to the uterine epithelial would trigger the endometrial stromal cells to differentiate into decidual stromal cells. However, decidualization in women takes place from the secretory phase of each menstrual cycle and continues to early pregnancy if there is conceptus. Deficient decidualization is often associated with pregnancy specific complications and reproductive disorders. Dramatic changes occur in the gene expression profiles during decidualization, which is coordinately regulated by steroid hormones, growth factors, and molecular and epigenetic mechanisms. Recently, emerging evidences showed that epigenetic modifications, mainly including DNA methylation, histone modification, and non-coding RNAs, play an important role in the decidualization process via affecting the target genes' expression. In this review, we will focus on the epigenetic modifications in decidualization and open novel avenues to predict and treat the pregnancy complications caused by abnormal decidualization.


Asunto(s)
Decidua/fisiología , Endometrio/fisiología , Epigénesis Genética , Animales , Metilación de ADN , Decidua/citología , Endometrio/citología , Femenino , Código de Histonas , Humanos , Embarazo , Células del Estroma/citología , Células del Estroma/metabolismo
17.
PLoS Genet ; 14(11): e1007787, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30452456

RESUMEN

Successful embryo implantation requires a receptive endometrium. Poor uterine receptivity can account for implantation failure in women who experience recurrent pregnancy loss or multiple rounds of unsuccessful in vitro fertilization cycles. Here, we demonstrate that the transcription factor Forkhead Box O1 (FOXO1) is a critical regulator of endometrial receptivity in vivo. Uterine ablation of Foxo1 using the progesterone receptor Cre (PgrCre) mouse model resulted in infertility due to altered epithelial cell polarity and apoptosis, preventing the embryo from penetrating the luminal epithelium. Analysis of the uterine transcriptome after Foxo1 ablation identified alterations in gene expression for transcripts involved in the activation of cell invasion, molecular transport, apoptosis, ß-catenin (CTNNB1) signaling pathway, and an increase in PGR signaling. The increase of PGR signaling was due to PGR expression being retained in the uterine epithelium during the window of receptivity. Constitutive expression of epithelial PGR during this receptive period inhibited expression of FOXO1 in the nucleus of the uterine epithelium. The reciprocal expression of PGR and FOXO1 was conserved in human endometrial samples during the proliferative and secretory phase. This demonstrates that expression of FOXO1 and the loss of PGR during the window of receptivity are interrelated and critical for embryo implantation.


Asunto(s)
Implantación del Embrión/genética , Implantación del Embrión/fisiología , Endometrio/metabolismo , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Animales , Núcleo Celular/metabolismo , Polaridad Celular/genética , Polaridad Celular/fisiología , Decidua/fisiología , Endometrio/citología , Femenino , Proteína Forkhead Box O1/deficiencia , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Embarazo , Receptores de Progesterona/deficiencia , Transducción de Señal
18.
Biol Reprod ; 103(4): 695-710, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32591788

RESUMEN

Despite sharing many common features, the relationship between ageing and parturition remains poorly understood. The decidua is a specialized lining of endometrial tissue, which develops in preparation for pregnancy. The structure and location of the decidua support its role as the physical scaffold for the growing embryo and placenta, and thus, it is vital to sustain pregnancy. Approaching term, the physical support properties of the decidua are naturally weakened to permit parturition. In this review, we hypothesize that the natural weakening of decidual tissue at parturition is promoted by the ageing process. Studies of the ageing-related functional and molecular changes in the decidua at parturition are reviewed and classified using hallmarks of ageing as the framework. The potential roles of decidual mesenchymal stem/stromal cell (DMSC) ageing in labor are also discussed because, although stem cell exhaustion is also a hallmark of ageing, its role in labor is not completely understood. In addition, the potential roles of extracellular vesicles secreted by DMSCs in labor, and their parturition-related miRNAs, are reviewed to gain further insight into this research area. In summary, the literature supports the notion that the decidua ages as the pregnancy progresses, and this may facilitate parturition, suggesting that ageing is the probable impetus of the gestational clocks in the decidua. This conceptual framework was developed to provide a better understanding of the natural ageing process of the decidua during parturition as well as to encourage future studies of the importance of healthy ageing for optimal pregnancy outcomes.


Asunto(s)
Decidua/fisiología , Parto/fisiología , Placenta/fisiología , Femenino , Humanos , Embarazo
19.
Mol Hum Reprod ; 26(11): 850-865, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32898274

RESUMEN

A critical component of early human placental development includes migration of extravillous trophoblasts (EVTs) into the decidua. EVTs migrate toward and displace vascular smooth muscle cells (SMCs) surrounding several uterine structures, including spiral arteries. Shallow trophoblast invasion features in several pregnancy complications including preeclampsia. Maternal obesity is a risk factor for placental dysfunction, suggesting that factors within an obese environment may impair early placental development. Herein, we tested the hypothesis that palmitic acid, a saturated fatty acid circulating at high levels in obese women, induces an inflammatory response in EVTs that hinders their capacity to migrate toward SMCs. We found that SMCs and SMC-conditioned media stimulated migration and invasion of an EVT-like cell line, HTR8/SVneo. Palmitic acid impaired EVT migration and invasion toward SMCs, and induced expression of several vasoactive and inflammatory mediators in EVTs, including endothelin, interleukin (IL)-6, IL-8 and PAI1. PAI1 was increased in plasma of women with early-onset preeclampsia, and PAI1-deficient EVTs were protected from the anti-migratory effects of palmitic acid. Using first trimester placental explants, palmitic acid exposure decreased EVT invasion through Matrigel. Our findings reveal that palmitic acid induces an inflammatory response in EVTs and attenuates their migration through a mechanism involving PAI1. High levels of palmitic acid in pathophysiological situations like obesity may impair early placental development and predispose to placental dysfunction.


Asunto(s)
Movimiento Celular , Inflamación , Ácido Palmítico/farmacología , Inhibidor 1 de Activador Plasminogénico/fisiología , Trofoblastos/fisiología , Adulto , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Células Cultivadas , Decidua/efectos de los fármacos , Decidua/fisiología , Femenino , Células HEK293 , Humanos , Inflamación/sangre , Inflamación/inducido químicamente , Inflamación/patología , Inflamación/fisiopatología , Mediadores de Inflamación/sangre , Mediadores de Inflamación/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/fisiología , Obesidad/sangre , Obesidad/complicaciones , Obesidad/fisiopatología , Ácido Palmítico/sangre , Placenta/citología , Placenta/efectos de los fármacos , Placentación/efectos de los fármacos , Placentación/fisiología , Inhibidor 1 de Activador Plasminogénico/sangre , Embarazo , Complicaciones del Embarazo/sangre , Complicaciones del Embarazo/fisiopatología , Trofoblastos/citología , Trofoblastos/efectos de los fármacos , Adulto Joven
20.
Hum Reprod ; 35(3): 617-640, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32219408

RESUMEN

STUDY QUESTION: Do seminal plasma (SP) and its constituents affect the decidualization capacity and transcriptome of human primary endometrial stromal fibroblasts (eSFs)? SUMMARY ANSWER: SP promotes decidualization of eSFs from women with and without inflammatory disorders (polycystic ovary syndrome (PCOS), endometriosis) in a manner that is not mediated through semen amyloids and that is associated with a potent transcriptional response, including the induction of interleukin (IL)-11, a cytokine important for SP-induced decidualization. WHAT IS KNOWN ALREADY: Clinical studies have suggested that SP can promote implantation, and studies in vitro have demonstrated that SP can promote decidualization, a steroid hormone-driven program of eSF differentiation that is essential for embryo implantation and that is compromised in women with the inflammatory disorders PCOS and endometriosis. STUDY DESIGN, SIZE, DURATION: This is a cross-sectional study involving samples treated with vehicle alone versus treatment with SP or SP constituents. SP was tested for the ability to promote decidualization in vitro in eSFs from women with or without PCOS or endometriosis (n = 9). The role of semen amyloids and fractionated SP in mediating this effect and in eliciting transcriptional changes in eSFs was then studied. Finally, the role of IL-11, a cytokine with a key role in implantation and decidualization, was assessed as a mediator of the SP-facilitated decidualization. PARTICIPANTS/MATERIALS, SETTING, METHODS: eSFs and endometrial epithelial cells (eECs) were isolated from endometrial biopsies from women of reproductive age undergoing benign gynecologic procedures and maintained in vitro. Assays were conducted to assess whether the treatment of eSFs with SP or SP constituents affects the rate and extent of decidualization in women with and without inflammatory disorders. To characterize the response of the endometrium to SP and SP constituents, RNA was isolated from treated eSFs or eECs and analyzed by RNA sequencing (RNAseq). Secreted factors in conditioned media from treated cells were analyzed by Luminex and ELISA. The role of IL-11 in SP-induced decidualization was assessed through Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas-9-mediated knockout experiments in primary eSFs. MAIN RESULTS AND THE ROLE OF CHANCE: SP promoted decidualization both in the absence and presence of steroid hormones (P < 0.05 versus vehicle) in a manner that required seminal proteins. Semen amyloids did not promote decidualization and induced weak transcriptomic and secretomic responses in eSFs. In contrast, fractionated SP enriched for seminal microvesicles (MVs) promoted decidualization. IL-11 was one of the most potently SP-induced genes in eSFs and was important for SP-facilitated decidualization. LARGE SCALE DATA: RNAseq data were deposited in the Gene Expression Omnibus repository under series accession number GSE135640. LIMITATIONS, REASONS FOR CAUTION: This study is limited to in vitro analyses. WIDER IMPLICATIONS OF THE FINDINGS: Our results support the notion that SP promotes decidualization, including within eSFs from women with inflammatory disorders. Despite the general ability of amyloids to induce cytokines known to be important for implantation, semen amyloids poorly signaled to eSFs and did not promote their decidualization. In contrast, fractionated SP enriched for MVs promoted decidualization and induced a transcriptional response in eSFs that overlapped with that of SP. Our results suggest that SP constituents, possibly those associated with MVs, can promote decidualization of eSFs in an IL-11-dependent manner in preparation for implantation. STUDY FUNDING/COMPETING INTEREST(S): This project was supported by NIH (R21AI116252, R21AI122821 and R01AI127219) to N.R.R. and (P50HD055764) to L.C.G. The authors declare no conflict of interest.


Asunto(s)
Decidua , Fibroblastos/citología , Interleucina-11/fisiología , Semen , Estudios Transversales , Decidua/fisiología , Endometriosis , Endometrio/citología , Femenino , Humanos , Interleucina-11/genética , Síndrome del Ovario Poliquístico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA