Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 739
Filtrar
1.
Reprod Fertil Dev ; 362024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38976640

RESUMEN

Context There is mounting evidence implicating kisspeptin signalling in placental development and function. Aims This study aimed to elucidate kisspeptin's role in trophoblast invasion and migration using three experimental models. Methods First, we examined the mouse fetus and placenta in a kisspeptin receptor (Kiss1r) knockout (KO) model. Fetal/placental weights and gene expression (quantitative polymerase chain reaction) were assessed. Second, we determined kisspeptin effects on a human trophoblast (BeWo) cell line in vitro . Third, we examined KISS1 and KISS1R gene expression in human placenta from term and pre-term pregnancies. Key results No difference was found in fetal or placental weight between Kiss1r KO and wildtype mice. However, expression of the trophoblast invasion marker, Mmp2 mRNA, was greater in the placental labyrinth zone of Kiss1r KO mice. BeWo cell models of villus cytotrophoblast and syncytiotrophoblast cells exhibited kisspeptin protein expression, with greater expression in syncytiotrophoblast, consistent with KISS1 mRNA. Kisspeptin treatment inhibited the migratory potential of cytotrophoblast-like cells. Finally, while no difference was seen in KISS1 and KISS1R mRNA between term and pre-term placentas, we saw a difference in the relative expression of each gene pre-term. We also observed a positive correlation between KISS1 expression and maternal body mass index. Conclusions Our results indicate that kisspeptin may inhibit trophoblast invasion. Implications Further investigation is required to clarify specific regulatory mechanisms.


Asunto(s)
Movimiento Celular , Kisspeptinas , Ratones Noqueados , Placenta , Receptores de Kisspeptina-1 , Trofoblastos , Kisspeptinas/metabolismo , Kisspeptinas/genética , Femenino , Trofoblastos/metabolismo , Receptores de Kisspeptina-1/metabolismo , Receptores de Kisspeptina-1/genética , Animales , Embarazo , Placenta/metabolismo , Movimiento Celular/fisiología , Humanos , Ratones , Línea Celular , Placentación/fisiología
2.
Ceska Gynekol ; 89(2): 151-155, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38704229

RESUMEN

The human placenta serves as a vital barrier between the mother and the developing fetus during pregnancy. A defect in the early development of the placenta is associated with severe pregnancy disorders. Despite its complex development, various molecular processes control placental development, and the specialization of trophoblast cells is still not fully understood. One primary obstacle is the lack of suitable cell model systems. Traditional two-dimensional (2D) cell cultures fail to mimic in vivo conditions and do not capture the intricate intercellular interactions vital for studying placental development. However, three-dimensional (3D) organoid models derived from stem cells that replicate natural cell organization and architecture have greatly improved our understanding of trophoblast behavior and its medicinal applications. Organoids with relevant phenotypes provide a valuable platform to model both placental physiology and pathology, including the modeling of placental disorders. They hold great promise for personalized medicine, improved diagnostics, and the evaluation of pharmaceutical drug efficacy and safety. This article provides a concise overview of trophoblast stem cells, trophoblast invasion, and the evolving role of organoids in gynecology.


Asunto(s)
Organoides , Células Madre , Trofoblastos , Humanos , Trofoblastos/fisiología , Organoides/fisiología , Femenino , Embarazo , Células Madre/fisiología , Placenta/citología , Placenta/fisiología , Placenta/patología , Placentación/fisiología
3.
Placenta ; 152: 53-64, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38805949

RESUMEN

INTRODUCTION: The placenta differs greatly among species, and deep extra-villous trophoblast (EVT) invasion is a unique feature of placentation of higher primates including humans. We reported serine protease HtrA4 being found predominantly in human placentas with aberrant expression linked to preeclampsia. However, it remains unclear where HtrA4 is produced in the placenta, how it is expressed in other species, and whether it is essential for human placentation. METHODS: We first compared HtrA4 protein sequences of over 100 species, then scrutinized the key characteristics of HtrA4 in the human, rhesus macaque and mouse, and determined cellular localization in the placenta. We next investigated functional significance of HtrA4 in EVT differentiation using human trophoblast stem cells (TSCs). RESULTS: Across broader species HtrA4 is well conserved only in higher primates. In humans, only the placenta expressed HtrA4, localising to trophoblasts of villous as well as extra-villous lineages. Rhesus macaques produced HtrA4 but again only in placentas, whereas mice showed no abundant HtrA4 expression anywhere including the placenta, yet it was an active protease if produced. The functional importance of HtrA4 in human EVT was demonstrated using TSCs, which expressed low levels of HtrA4 but significantly up-regulated it during EVT differentiation, and knockdown of HtrA4 severely inhibited the differentiation process. DISCUSSION: HtrA4 is expressed in placentas of humans and macaques but not mice; it is critical for human EVT differentiation. Together with previous reports showing HtrA4 is also indispensable for syncytialization, this study further revealed HtrA4 as a functionally important protease for human placentation.


Asunto(s)
Diferenciación Celular , Macaca mulatta , Serina Endopeptidasas , Trofoblastos , Animales , Trofoblastos/metabolismo , Humanos , Femenino , Embarazo , Diferenciación Celular/fisiología , Ratones , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/genética , Placenta/metabolismo , Placentación/fisiología , Serina Proteasas
4.
Placenta ; 151: 37-47, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38703713

RESUMEN

Caspases, a family of cysteine proteases, are pivotal regulators of apoptosis, the tightly controlled cell death process crucial for eliminating excessive or unnecessary cells during development, including placental development. Collecting research has unveiled the multifaceted roles of caspases in the placenta, extending beyond apoptosis. Apart from their involvement in placental tissue remodeling via apoptosis, caspases actively participate in essential regulatory processes, such as trophoblast fusion and differentiation, significantly influencing placental growth and functionality. In addition, growing evidence indicates an elevation in caspase activity under pathological conditions like pre-eclampsia (PE) and intrauterine growth restriction (IUGR), leading to excessive cell death as well as inflammation. Drawing from advancements in caspase research and placental development under both normal and abnormal conditions, we examine the significance of caspases in both cell death (apoptosis) and non-cell death-related processes within the placenta. We also discuss potential therapeutics targeting caspase-related pathways for placenta disorders.


Asunto(s)
Apoptosis , Caspasas , Placenta , Animales , Femenino , Humanos , Embarazo , Caspasas/metabolismo , Placenta/patología , Placenta/metabolismo , Enfermedades Placentarias/patología , Enfermedades Placentarias/metabolismo , Placentación/fisiología , Preeclampsia/patología , Preeclampsia/metabolismo , Trofoblastos/fisiología , Trofoblastos/patología
5.
Biol Reprod ; 111(2): 414-426, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38647664

RESUMEN

OBJECTIVE: The purpose of this study is to investigate the role of high mobility group protein B1 (HMGB1) in placental development and fetal growth. METHODS: We employed the Cre-loxP recombination system to establish a placenta-specific HMGB1 knockout mouse model. Breeding HMGB1flox/flox mice with Elf5-Cre mice facilitated the knockout, leveraging Elf5 expression in extra-embryonic ectoderm, ectoplacental cone, and trophoblast giant cells at 12.5 days of embryonic development. The primary goal of this model was to elucidate the molecular mechanism of HMGB1 in placental development, assessing parameters such as placental weight, fetal weight, and bone development. Additionally, we utilized lentiviral interference and overexpression of HMGB1 in human trophoblast cells to further investigate HMGB1's functional role. RESULTS: Our findings indicate that the HMGB1flox/floxElf5cre/+ mouse displays fetal growth restriction, characterized by decreased placental and fetal weight and impaired bone development. The absence of HMGB1 inhibits autophagosome formation, impairs lysosomal degradation, and disrupts autophagic flux. Depletion of HMGB1 in human trophoblast cells also suppresses cell viability, proliferation, migration, and invasion by inhibiting the ERK signaling pathway. Overexpression of HMGB1 observed the opposite phenotypes. CONCLUSIONS: HMGB1 participates in the regulation of autophagy through the ERK signaling pathway and affects placental development.


Asunto(s)
Autofagia , Proteína HMGB1 , Sistema de Señalización de MAP Quinasas , Placenta , Trofoblastos , Animales , Femenino , Humanos , Ratones , Embarazo , Autofagia/fisiología , Proteína HMGB1/metabolismo , Proteína HMGB1/genética , Sistema de Señalización de MAP Quinasas/fisiología , Ratones Noqueados , Placenta/metabolismo , Placentación/fisiología , Trofoblastos/metabolismo , Trofoblastos/fisiología , Masculino
6.
Hum Reprod Update ; 30(4): 442-471, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38519450

RESUMEN

BACKGROUND: The placenta is a unique and pivotal organ in reproduction, controlling crucial growth and cell differentiation processes that ensure a successful pregnancy. Placental development is a tightly regulated and dynamic process, in which the transforming growth factor beta (TGFß) superfamily plays a central role. This family of pleiotropic growth factors is heavily involved in regulating various aspects of reproductive biology, particularly in trophoblast differentiation during the first trimester of pregnancy. TGFß signalling precisely regulates trophoblast invasion and the cell transition from cytotrophoblasts to extravillous trophoblasts, which is an epithelial-to-mesenchymal transition-like process. Later in pregnancy, TGFß signalling ensures proper vascularization and angiogenesis in placental endothelial cells. Beyond its role in trophoblasts and endothelial cells, TGFß signalling contributes to the polarization and function of placental and decidual macrophages by promoting maternal tolerance of the semi-allogeneic foetus. Disturbances in early placental development have been associated with several pregnancy complications, including preeclampsia (PE) which is one of the severe complications. Emerging evidence suggests that TGFß is involved in the pathogenesis of PE, thereby offering a potential target for intervention in the human placenta. OBJECTIVE AND RATIONALE: This comprehensive review aims to explore and elucidate the roles of the major members of the TGFß superfamily, including TGFßs, bone morphogenetic proteins (BMPs), activins, inhibins, nodals, and growth differentiation factors (GDFs), in the context of placental development and function. The review focusses on their interactions within the major cell types of the placenta, namely trophoblasts, endothelial cells, and immune cells, in both normal pregnancies and pregnancies complicated by PE throughout pregnancy. SEARCH METHODS: A literature search was carried out using PubMed and Google Scholar, searching terms: 'TGF signalling preeclampsia', 'pregnancy TGF signalling', 'preeclampsia tgfß', 'preeclampsia bmp', 'preeclampsia gdf', 'preeclampsia activin', 'endoglin preeclampsia', 'endoglin pregnancy', 'tgfß signalling pregnancy', 'bmp signalling pregnancy', 'gdf signalling pregnancy', 'activin signalling pregnancy', 'Hofbauer cell tgfß signalling', 'placental macrophages tgfß', 'endothelial cells tgfß', 'endothelium tgfß signalling', 'trophoblast invasion tgfß signalling', 'trophoblast invasion Smad', 'trophoblast invasion bmp', 'trophoblast invasion tgfß', 'tgfß preeclampsia', 'tgfß placental development', 'TGFß placental function', 'endothelial dysfunction preeclampsia tgfß signalling', 'vascular remodelling placenta TGFß', 'inflammation pregnancy tgfß', 'immune response pregnancy tgfß', 'immune tolerance pregnancy tgfß', 'TGFß pregnancy NK cells', 'bmp pregnancy NK cells', 'bmp pregnancy tregs', 'tgfß pregnancy tregs', 'TGFß placenta NK cells', 'TGFß placenta tregs', 'NK cells preeclampsia', 'Tregs preeclampsia'. Only articles published in English until 2023 were used. OUTCOMES: A comprehensive understanding of TGFß signalling and its role in regulating interconnected cell functions of the main placental cell types provides valuable insights into the processes essential for successful placental development and growth of the foetus during pregnancy. By orchestrating trophoblast invasion, vascularization, immune tolerance, and tissue remodelling, TGFß ligands contribute to the proper functioning of a healthy maternal-foetal interface. However, dysregulation of TGFß signalling has been implicated in the pathogenesis of PE, where the shallow trophoblast invasion, defective vascular remodelling, decreased uteroplacental perfusion, and endothelial cell and immune dysfunction observed in PE, are all affected by an altered TGFß signalling. WIDER IMPLICATIONS: The dysregulation of TGFß signalling in PE has important implications for research and clinical practice. Further investigation is required to understand the underlying mechanisms, including the role of different ligands and their regulation under pathophysiological conditions, in order to discover new therapeutic targets. Distinguishing between clinically manifested subtypes of PE and studying TGFß signalling in different placental cell types holistically is an important first step. To put this knowledge into practice, pre-clinical animal models combined with new technologies are needed. This may also lead to improved human research models and identify potential therapeutic targets, ultimately improving outcomes for affected pregnancies and reducing the burden of PE.


Asunto(s)
Inflamación , Placenta , Preeclampsia , Transducción de Señal , Factor de Crecimiento Transformador beta , Humanos , Embarazo , Femenino , Preeclampsia/metabolismo , Preeclampsia/fisiopatología , Factor de Crecimiento Transformador beta/metabolismo , Placenta/metabolismo , Inflamación/metabolismo , Trofoblastos/metabolismo , Trofoblastos/fisiología , Placentación/fisiología
7.
Methods Mol Biol ; 2781: 93-103, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38502446

RESUMEN

The placenta is a vital organ that regulates nutrient supply to the developing embryo during gestation. In mice, the placenta is composed of trophoblast lineage and mesodermal derivatives, which merge through the chorioallantoic fusion process in a critical event for the progression of placenta development. The trophoblast lineage is derived from self-renewing, multipotent cells known as mouse trophoblast stem cells (mTSCs). These cells are a valuable tool that allows scientists to comprehend the signals regulating major placental cell types' self-renewal and differentiation capacity. Recent advances in CRISPR-Cas9 genome editing applied in mTSCs have provided novel insights into the molecular networks involved in placentation. Here, we present a comprehensive CRISPR activation (CRISPRa) protocol based on the CRISPR/gRNA-directed synergistic activation mediator (SAM) method to overexpress specific target genes in mTSCs.


Asunto(s)
Placenta , ARN Guía de Sistemas CRISPR-Cas , Embarazo , Femenino , Animales , Ratones , Trofoblastos , Placentación/fisiología , Diferenciación Celular/genética , Células Madre
8.
J Assist Reprod Genet ; 41(3): 591-608, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38315418

RESUMEN

Precise extravillous trophoblast (EVT) invasion is crucial for successful placentation and pregnancy. This review focuses on elucidating the mechanisms that promote heightened EVT invasion. We comprehensively summarize the pivotal roles of hormones, angiogenesis, hypoxia, stress, the extracellular matrix microenvironment, epithelial-to-mesenchymal transition (EMT), immunity, inflammation, programmed cell death, epigenetic modifications, and microbiota in facilitating EVT invasion. The molecular mechanisms underlying enhanced EVT invasion may provide valuable insights into potential pathogenic mechanisms associated with diseases characterized by excessive invasion, such as the placenta accreta spectrum (PAS), thereby offering novel perspectives for managing pregnancy complications related to deficient EVT invasion.


Asunto(s)
Trofoblastos Extravellosos , Trofoblastos , Embarazo , Femenino , Humanos , Trofoblastos/metabolismo , Placentación/fisiología , Células Epiteliales , Placenta/metabolismo
9.
Nat Commun ; 15(1): 1487, 2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38374152

RESUMEN

Proper placental development in early pregnancy ensures a positive outcome later on. The developmental relationship between the placenta and embryonic organs, such as the heart, is crucial for a normal pregnancy. However, the mechanism through which the placenta influences the development of embryonic organs remains unclear. Trophoblasts fuse to form multinucleated syncytiotrophoblasts (SynT), which primarily make up the placental materno-fetal interface. We discovered that endogenous progesterone immunomodulatory binding factor 1 (PIBF1) is vital for trophoblast differentiation and fusion into SynT in humans and mice. PIBF1 facilitates communication between SynT and adjacent vascular cells, promoting vascular network development in the primary placenta. This process affected the early development of the embryonic cardiovascular system in mice. Moreover, in vitro experiments showed that PIBF1 promotes the development of cardiovascular characteristics in heart organoids. Our findings show how SynTs organize the barrier and imply their possible roles in supporting embryogenesis, including cardiovascular development. SynT-derived factors and SynT within the placenta may play critical roles in ensuring proper organogenesis of other organs in the embryo.


Asunto(s)
Sistema Cardiovascular , Placenta , Proteínas Gestacionales , Animales , Femenino , Humanos , Ratones , Embarazo , Diferenciación Celular , Desarrollo Embrionario , Placenta/metabolismo , Placentación/fisiología , Proteínas Gestacionales/genética , Proteínas Gestacionales/metabolismo , Factores Supresores Inmunológicos/metabolismo , Trofoblastos/metabolismo , Sistema Cardiovascular/embriología
10.
J Anat ; 244(6): 1054-1066, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38288680

RESUMEN

The mammalian placenta's interface with the parent is a richly vascularized tissue whose development relies upon communication between many different cell types within the uterine microenvironment. The uterine blood vessels of the interface are reshaped during pregnancy into wide-bore, flaccid vessels that convey parental blood to the exchange region of the placenta. Invasive trophoblast as well as parental uterine macrophages and Natural Killer cells are involved in the stepwise remodeling of these vessels and their respective contributions to this crucial process are still being delineated. However, the earliest steps in arteriole remodeling are understudied as they are difficult to study in humans, and other species lack the deep trophoblast invasion that is so prominent a feature of placentation in humans. Here, we further characterize the rat, with deep hemochorial placentation akin to humans, as a model system in which to tease apart the earliest, relatively understudied events in spiral arteriole remodeling. We show that the rat uterine-placental interface increases in size and vascularity rapidly, before trophoblast invasion. The remodeling stages in the arterioles of the rat uterine-placental interface follow a sequence of anatomical changes similar to those in humans, and there are changes to the arterioles' muscular tunica media prior to the marked influx of immune cells. The rat is a tractable model in which to better understand the cell/cell interactions occurring in vivo in an intact tissue microenvironment over time.


Asunto(s)
Placenta , Útero , Remodelación Vascular , Animales , Femenino , Embarazo , Arteriolas , Ratas , Útero/irrigación sanguínea , Placenta/irrigación sanguínea , Remodelación Vascular/fisiología , Placentación/fisiología , Modelos Animales , Ratas Sprague-Dawley
11.
Cell Stem Cell ; 31(2): 181-195.e9, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-38237587

RESUMEN

In humans, balanced invasion of trophoblast cells into the uterine mucosa, the decidua, is critical for successful pregnancy. Evidence suggests that this process is regulated by uterine natural killer (uNK) cells, but how they influence reproductive outcomes is unclear. Here, we used our trophoblast organoids and primary tissue samples to determine how uNK cells affect placentation. By locating potential interaction axes between trophoblast and uNK cells using single-cell transcriptomics and in vitro modeling of these interactions in organoids, we identify a uNK cell-derived cytokine signal that promotes trophoblast differentiation at the late stage of the invasive pathway. Moreover, it affects transcriptional programs involved in regulating blood flow, nutrients, and inflammatory and adaptive immune responses, as well as gene signatures associated with disorders of pregnancy such as pre-eclampsia. Our findings suggest mechanisms on how optimal immunological interactions between uNK cells and trophoblast enhance reproductive success.


Asunto(s)
Trofoblastos Extravellosos , Útero , Embarazo , Femenino , Humanos , Útero/metabolismo , Placentación/fisiología , Trofoblastos , Células Asesinas Naturales
12.
Endocrinology ; 165(3)2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38195194

RESUMEN

BACKGROUND: Repeated implantation failure (RIF) leads to a waste of high-quality embryos and remains a challenge in assisted reproductive technology. During early human placentation, the invasion of trophoblast cells into the decidua is an essential step for the establishment of maternal-fetal interactions and subsequent successful pregnancy. Bone morphogenetic protein 2 (BMP2) has been reported to regulate endometrial receptivity and promote trophoblast invasion. However, whether there is dysregulation of endometrial BMP2 expression in patients with RIF remains unknown. Additionally, the molecular mechanisms underlying the effects of BMP2 on human trophoblast invasion and early placentation remain to be further elucidated. METHODS: Midluteal phase endometrial samples were biopsied from patients with RIF and from routine control in vitro fertilization followed by quantitative polymerase chain reaction and immunoblotting analyses. Human trophoblast organoids, primary human trophoblast cells, and an immortalized trophoblast cell line (HTR8/SVneo) were used as study models. RESULTS: We found that BMP2 was aberrantly low in midluteal phase endometrial tissues from patients with RIF. Recombinant human BMP2 treatment upregulated integrin ß3 (ITGB3) in a SMAD2/3-SMAD4 signaling-dependent manner in both HTR8/SVneo cells and primary trophoblast cells. siRNA-mediated integrin ß3 downregulation reduced both basal and BMP2-upregulated trophoblast invasion and vascular mimicry in HTR8/SVneo cells. Importantly, shRNA-mediated ITGB3 knockdown significantly decreased the formation ability of human trophoblast organoids. CONCLUSION: Our results demonstrate endometrial BMP2 deficiency in patients with RIF. ITGB3 mediates both basal and BMP2-promoted human trophoblast invasion and is essential for early placentation. These findings broaden our knowledge regarding the regulation of early placentation and provide candidate diagnostic and therapeutic targets for RIF clinical management.


Asunto(s)
Proteína Morfogenética Ósea 2 , Integrina beta3 , Embarazo , Humanos , Femenino , Integrina beta3/genética , Integrina beta3/metabolismo , Proteína Morfogenética Ósea 2/metabolismo , Trofoblastos/metabolismo , Línea Celular , Placentación/fisiología , ARN Interferente Pequeño/metabolismo , Movimiento Celular
13.
Methods Mol Biol ; 2767: 175-188, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-36773273

RESUMEN

Human pluripotent stem cells (hPSCs) form an ideal system to study the formation of placental cells, from an undifferentiated human embryonic stem cell state. The conventional human in vitro model systems to study the human placenta cannot be employed for understanding placental dysfunctions or the development of specialized placental cell types. Hence, human PSCs make an ideal model system to study human placental development and disorders. Here, we describe an efficient and validated protocol to reproducibly study the formation of human cytotrophoblasts (CTBs) and syncytiotrophoblast (STBs) from undifferentiated hPSCs. CTBs are the trophoblast stem cells that can differentiate into specialized placental cell types such as STBs. The multinucleated STB plays vital role in the exchange of nutrients and gases across the placenta and secretes several hormones during pregnancy, such as human chorionic gonadotropin ß (hCGß). Here we describe two methods of seeding the hPSCs: chemical (clumps method) and enzymatic methods (single cells) to differentiate them to CTB and STB, activating BMP (B) signaling and inhibiting ACTIVIN/NODAL and FGF signaling pathways (2i), thus naming our protocol as "B2i" (Sudheer et al., Stem Cells Dev 21:2987-3000, 2012). This protocol forms the perfect model system for understanding in vitro placentation, modeling diseases arising from abnormal placentation that cause complications such as miscarriage, preeclampsia or intrauterine growth restriction (IUGR), and drug discovery for placental disorders.


Asunto(s)
Enfermedades Placentarias , Células Madre Pluripotentes , Humanos , Embarazo , Femenino , Placenta , Trofoblastos , Placentación/fisiología , Diferenciación Celular/fisiología , Enfermedades Placentarias/metabolismo
14.
Biol Reprod ; 109(5): 759-771, 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37665239

RESUMEN

Proper extravillous trophoblast invasion is essential for normal placentation and pregnancy. However, the molecular mechanisms by which cytotrophoblasts differentiate into extravillous trophoblast are unclear. We discovered that in the first-trimester placenta, progesterone receptor membrane component 2 was highly expressed in syncytiotrophoblast but significantly lower in extravillous trophoblast and cytotrophoblasts, indicating a divergent role for progesterone receptor membrane component 2 in trophoblast functions. We aim to examine the role of progesterone receptor membrane component 2 in extravillous trophoblasts invasion mediated by both intracellular and extracellular signals. Progesterone receptor membrane component 2 knockdown and overexpression cells were established in HTR8/SVneo cells, a first-trimester extravillous trophoblast-derived cell model, by transfection with small-interfering RNA or progesterone receptor membrane component 2 plasmids, respectively. Progesterone receptor membrane component 2 knockdown led to cellular morphological changes , enhanced trophoblast proliferation,invasion, and promoted tube formation. These effects were mediated by the activation of hypoxia-inducible factor 1alpha and an increased expression of vascular endothelial growth factor A. The culture supernatant collected from progesterone receptor membrane component 2 knockdown cells did not significantly affect extravillous trophoblast invasion compared to the controls, indicating that extracellular signaling did not robustly regulate extravillous trophoblast invasion in this study. In conclusion, attenuation of progesterone receptor membrane component 2 plays a role in placentation by promoting cell proliferation, invasion, and angiogenesis in extravillous trophoblasts via activation of hypoxia-inducible factor 1 alpha signaling. We thus identified a new function of progesterone receptor membrane component 2 and provide insights on understanding the mechanisms of trophoblast invasion.


Asunto(s)
Placenta , Factor A de Crecimiento Endotelial Vascular , Femenino , Humanos , Embarazo , Línea Celular , Movimiento Celular , Trofoblastos Extravellosos , Placenta/metabolismo , Placentación/fisiología , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Trofoblastos/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Proc Biol Sci ; 290(2001): 20230940, 2023 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-37357861

RESUMEN

Reproduction is a central activity for all living organisms but is also associated with a diversity of costs that are detrimental for survival. Until recently, the cost of cancer as a selective force has been poorly considered. Considering 191 mammal species, we found cancer mortality was more likely to be detected in species having large, rather than low, litter sizes and long lactation lengths regardless of the placentation types. However, increasing litter size and gestation length are not per se associated with an enhanced cancer mortality risk. Contrary to basic theoretical expectations, the species with the highest cancer mortality were not those with the most invasive (i.e. haemochorial) placentation, but those with a moderately invasive (i.e. endotheliochorial) one. Overall, these results suggest that (i) high reproductive efforts favour oncogenic processes' dynamics, presumably because of trade-offs between allocation in reproduction effort and anti-cancer defences, (ii) cancer defence mechanisms in animals are most often adjusted to align reproductive lifespan, and (iii) malignant cells co-opt existing molecular and physiological pathways for placentation, but species with the most invasive placentation have also selected for potent barriers against lethal cancers. This work suggests that the logic of Peto's paradox seems to be applicable to other traits that promote tumorigenesis.


Asunto(s)
Neoplasias , Placentación , Embarazo , Animales , Femenino , Placentación/fisiología , Tamaño de la Camada , Lactancia/fisiología , Reproducción/fisiología , Mamíferos , Neoplasias/etiología
16.
Cells ; 12(12)2023 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-37371117

RESUMEN

Human chorionic gonadotropin (hCG) is produced by the placenta and its roles have been studied for over a century, being the first known pregnancy-related protein. Although its main role is to stimulate the production of progesterone by corpus luteal cells, hCG does not represent just one biologically active molecule, but a group of at least five variants, produced by different cells and each with different functions. The hyperglycosylated variant of hCG (H-hCG) plays a key role in trophoblast invasion, placental development and fetal growth. During trophoblast invasion, H-hCG promotes extravillous cytotrophoblast cells to infiltrate the decidua, and also to colonize and remodel the spiral arteries in to low resistance, larger-diameter vessels. As fetal growth is heavily reliant on nutrient availability, impaired trophoblast invasion and remodeling of the uterine arteries, leads to a defective perfusion of the placenta and fetal growth restriction. Understanding the function of H-hCG in the evolution of the placenta might unveil new ways to manage and treat fetal growth restriction.


Asunto(s)
Gonadotropina Coriónica , Retardo del Crecimiento Fetal , Placenta , Trofoblastos , Femenino , Humanos , Embarazo , Gonadotropina Coriónica/metabolismo , Retardo del Crecimiento Fetal/metabolismo , Placenta/metabolismo , Placentación/fisiología , Trofoblastos/metabolismo
17.
Reproduction ; 166(1): 1-11, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37078791

RESUMEN

In brief: Healthy development of the placenta is dependent on trophoblast cell migration and reduced oxidative stress presence. This article describes how a phytoestrogen found in spinach and soy causes impaired placental development during pregnancy. Abstract: Although vegetarianism has grown in popularity, especially among pregnant women, the effects of phytoestrogens in placentation lack understanding. Factors such as cellular oxidative stress and hypoxia and external factors including cigarette smoke, phytoestrogens, and dietary supplements can regulate placental development. The isoflavone phytoestrogen coumestrol was identified in spinach and soy and was found to not cross the fetal-placental barrier. Since coumestrol could be a valuable supplement or potent toxin during pregnancy, we sought to examine its role in trophoblast cell function and placentation in murine pregnancy. After treating trophoblast cells (HTR8/SVneo) with coumestrol and performing an RNA microarray, we determined 3079 genes were significantly changed with the top differentially changed pathways related to the oxidative stress response, cell cycle regulation, cell migration, and angiogenesis. Upon treatment with coumestrol, trophoblast cells exhibited reduced migration and proliferation. Additionally, we observed increased reactive oxygen species accumulation with coumestrol administration. We then examined the role of coumestrol within an in vivo pregnancy by treating wildtype pregnant mice with coumestrol or vehicle from day 0 to 12.5 of gestation. Upon euthanasia, fetal and placental weights were significantly decreased in coumestrol-treated animals with the placenta exhibiting a proportional decrease with no obvious changes in morphology. Therefore, we conclude that coumestrol impairs trophoblast cell migration and proliferation, causes accumulation of reactive oxygen species, and reduces fetal and placental weights in murine pregnancy.


Asunto(s)
Cumestrol , Placenta , Embarazo , Femenino , Ratones , Humanos , Animales , Placenta/metabolismo , Cumestrol/farmacología , Cumestrol/metabolismo , Fitoestrógenos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Línea Celular , Placentación/fisiología , Trofoblastos/metabolismo , Estrés Oxidativo
18.
Biochem Soc Trans ; 51(2): 639-653, 2023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-36929183

RESUMEN

Protein N-linked glycosylation is a structurally diverse post-translational modification that stores biological information in a larger order of magnitude than other post-translational modifications such as phosphorylation, ubiquitination and acetylation. This gives N-glycosylated proteins a diverse range of properties and allows glyco-codes (glycan-related information) to be deciphered by glycan-binding proteins (GBPs). The intervillous space of the placenta is richly populated with membrane-bound and secreted glycoproteins. Evidence exists to suggest that altering the structural nature of their N-glycans can impact several trophoblast functions, which include those related to interactions with decidual cells. This review summarizes trophoblast-related activities influenced by N-glycan-GBP recognition, exploring how different subtypes of trophoblasts actively adapt to characteristics of the decidualized endometrium through cell-specific expression of N-glycosylated proteins, and how these cells receive decidua-derived signals via N-glycan-GBP interactions. We highlight work on how changes in N-glycosylation relates to the success of trophoblast infiltration, interactions of immunomodulators, and uterine angiogenesis. We also discuss studies that suggest aberrant N-glycosylation of trophoblasts may contribute to the pathogenesis of pregnancy complications (e.g. pre-eclampsia, early spontaneous miscarriages and hydatidiform mole). We propose that a more in-depth understanding of how N-glycosylation shapes trophoblast phenotype during early pregnancy has the potential to improve our approach to predicting, diagnosing and alleviating poor maternal/fetal outcomes associated with placental dysfunction.


Asunto(s)
Placentación , Trofoblastos , Embarazo , Femenino , Humanos , Placentación/fisiología , Trofoblastos/metabolismo , Placenta/metabolismo , Glicosilación , Proteínas Portadoras/metabolismo , Proteínas/metabolismo , Inmunomodulación
19.
Hum Cell ; 36(3): 1190-1198, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36995581

RESUMEN

Argininosuccinate synthase (ASS1) is involved in nitric oxide production, which has a key role in placental development improving pregnancy outcomes. Syncytiotrophoblast and extravillous trophoblast differentiations are milestones of placental development and their impairment can cause pathologies, such as preeclampsia (PE) and fetal growth restriction (FGR). Immunohistochemistry and Western blotting were used to localize and quantify ASS1 in first trimester (8.2 ± 1.8 weeks), third trimester (38.6 ± 1.1 weeks), and PE (36.3 ± 1.5 weeks) placentas. In addition, cell cultures were used to evaluate ASS1 expression under hypoxic conditions and the syncytialization process. Our data showed that ASS1 is localized in the villous cytotrophoblast of first trimester, third trimester, and PE placentas, while the villous cytotrophoblast adjacent to the extravillous trophoblast of cell columns as well as the extravillous trophoblast were negative for ASS1 in first trimester placentas. In addition, ASS1 was decreased in third trimester compared to the first trimester placentas (p = 0.003) and no differences were detected between third trimester and PE placentas. Moreover, ASS1 expression was decreased in hypoxic conditions and syncytialized cells compared to those not syncytialized. In conclusion, we suggest that the expression of ASS1 in villous cytotrophoblast is related to maintaining proliferative phenotype, while ASS1 absence may be involved in promoting the differentiation of villous cytotrophoblast in extravillous cytotrophoblast of cell columns in first trimester placentas.


Asunto(s)
Placentación , Preeclampsia , Humanos , Embarazo , Femenino , Placentación/fisiología , Placenta , Argininosuccinato Sintasa/metabolismo , Regulación hacia Abajo , Trofoblastos/patología , Preeclampsia/patología , Hipoxia/patología
20.
Am J Obstet Gynecol ; 228(6): 613-621, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36539026

RESUMEN

The importance of uterine microvascular adaptations during placentation in pregnancy has been well established for decades. Inadequate dilatation of spiral arteries is associated with gestational complications, such as preeclampsia and/or intrauterine growth restriction. More recently, it has become clear that trophoblast cells invade and adapt decidual veins and lymphatic vessels 1 month before spiral arteries become patent and before intervillous space perfusion starts. Normal intervillous space hemodynamics is characterized by high volume flow at low velocity and pressure in the interseptal compartments surrounding the chorionic villi, hereby facilitating efficient maternal-fetal exchange. In case of shallow decidual vein dilatation, intervillous arterial supply exceeds venous drainage. This will cause congestion in the interseptal compartments with subsequently reduced perfusion and increased pressure. An efficient mechanism to counteract venous congestion and safeguard the viability of the conceptus is by reducing arterial inflow via shallow dilatation of the spiral arteries. This review made the case for intervillous space congestion as an unexplored trigger for inadequate spiral artery dilatation during the placentation process, eventually leading to abnormal systemic circulatory dysfunctions. An abnormal maternal venous function can result from an abnormal maternal immune response to paternal antigens with an imbalanced release of vasoactive mediators or can exist before conception. To get the full picture of abnormal placentation, maternal veins must not be forgotten.


Asunto(s)
Placentación , Preeclampsia , Embarazo , Femenino , Humanos , Placentación/fisiología , Placenta/irrigación sanguínea , Trofoblastos/fisiología , Intercambio Materno-Fetal , Arterias
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA