Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Nature ; 616(7955): 143-151, 2023 04.
Article in English | MEDLINE | ID: mdl-36991123

ABSTRACT

The relationship between the human placenta-the extraembryonic organ made by the fetus, and the decidua-the mucosal layer of the uterus, is essential to nurture and protect the fetus during pregnancy. Extravillous trophoblast cells (EVTs) derived from placental villi infiltrate the decidua, transforming the maternal arteries into high-conductance vessels1. Defects in trophoblast invasion and arterial transformation established during early pregnancy underlie common pregnancy disorders such as pre-eclampsia2. Here we have generated a spatially resolved multiomics single-cell atlas of the entire human maternal-fetal interface including the myometrium, which enables us to resolve the full trajectory of trophoblast differentiation. We have used this cellular map to infer the possible transcription factors mediating EVT invasion and show that they are preserved in in vitro models of EVT differentiation from primary trophoblast organoids3,4 and trophoblast stem cells5. We define the transcriptomes of the final cell states of trophoblast invasion: placental bed giant cells (fused multinucleated EVTs) and endovascular EVTs (which form plugs inside the maternal arteries). We predict the cell-cell communication events contributing to trophoblast invasion and placental bed giant cell formation, and model the dual role of interstitial EVTs and endovascular EVTs in mediating arterial transformation during early pregnancy. Together, our data provide a comprehensive analysis of postimplantation trophoblast differentiation that can be used to inform the design of experimental models of the human placenta in early pregnancy.


Subject(s)
Multiomics , Pregnancy Trimester, First , Trophoblasts , Female , Humans , Pregnancy , Cell Movement , Placenta/blood supply , Placenta/cytology , Placenta/physiology , Pregnancy Trimester, First/physiology , Trophoblasts/cytology , Trophoblasts/metabolism , Trophoblasts/physiology , Decidua/blood supply , Decidua/cytology , Maternal-Fetal Relations/physiology , Single-Cell Analysis , Myometrium/cytology , Myometrium/physiology , Cell Differentiation , Organoids/cytology , Organoids/physiology , Stem Cells/cytology , Transcriptome , Transcription Factors/metabolism , Cell Communication
3.
Placenta ; 125: 54-60, 2022 07.
Article in English | MEDLINE | ID: mdl-34952691

ABSTRACT

Correct establishment of the placenta is critical to the success of a pregnancy, but many of the key events take place during or shortly after implantation and are inaccessible for study. This inaccessibility, coupled with the lack of a suitable preclinical animal model, means that knowledge of human early placental development and function is extremely limited. Hence, the first trimester is often referred to as the 'black box' of pregnancy. However, recent advances in the derivation of trophoblast stem cells and organoid cultures of the trophoblast and endometrium are opening new opportunities for basic and translational research, providing for the first time cells that faithfully replicate their tissue of origin and proliferate and differentiate in culture in a stable and reproducible manner. These cells are valuable new tools for investigating cell-lineage differentiation and maternal-fetal interactions, but become even more powerful when combined with advances in bioengineering, microfabrication and microfluidic technologies. Assembloids of the endometrium comprising various cell types as model systems to investigate events at implantation, and placentas-on-a-chip for the study of nutrient transfer or drug screening are just two examples. This is a rapidly advancing field that may usher in more personalised approaches to infertility and pregnancy complications. Many of the developments are still at the proof-of-principle phase, but with continued refinement they are likely to shed important light on events that are fundamental to our reproduction as individuals and as a species, yet for ethical reasons are hidden from view.


Subject(s)
Awards and Prizes , Placenta , Animals , Embryo Implantation , Female , Humans , Placenta/metabolism , Placentation , Pregnancy , Trophoblasts/metabolism
4.
Nat Genet ; 53(12): 1698-1711, 2021 12.
Article in English | MEDLINE | ID: mdl-34857954

ABSTRACT

The endometrium, the mucosal lining of the uterus, undergoes dynamic changes throughout the menstrual cycle in response to ovarian hormones. We have generated dense single-cell and spatial reference maps of the human uterus and three-dimensional endometrial organoid cultures. We dissect the signaling pathways that determine cell fate of the epithelial lineages in the lumenal and glandular microenvironments. Our benchmark of the endometrial organoids reveals the pathways and cell states regulating differentiation of the secretory and ciliated lineages both in vivo and in vitro. In vitro downregulation of WNT or NOTCH pathways increases the differentiation efficiency along the secretory and ciliated lineages, respectively. We utilize our cellular maps to deconvolute bulk data from endometrial cancers and endometriotic lesions, illuminating the cell types dominating in each of these disorders. These mechanistic insights provide a platform for future development of treatments for common conditions including endometriosis and endometrial carcinoma.


Subject(s)
Endometrium/physiology , Menstrual Cycle , Cell Differentiation , Cell Lineage , Cellular Microenvironment , Endometrial Neoplasms/pathology , Endometrium/embryology , Endometrium/pathology , Female , Gonadal Steroid Hormones/metabolism , Humans , In Vitro Techniques , Organoids , Receptors, Notch/metabolism , Signal Transduction , Spatio-Temporal Analysis , Tissue Culture Techniques , Transcriptome , Uterus/pathology , Wnt Proteins/metabolism
5.
Development ; 148(21)2021 11 01.
Article in English | MEDLINE | ID: mdl-34651188

ABSTRACT

Two recently developed models, trophoblast organoids and trophoblast stem cells (TSCs), are useful tools to further the understanding of human placental development. Both differentiate from villous cytotrophoblast (VCT) to either extravillous trophoblast (EVT) or syncytiotrophoblast (SCT). Here, we compare the transcriptomes and miRNA profiles of these models to identify which trophoblast they resemble in vivo. Our findings indicate that TSCs do not readily undergo SCT differentiation and closely resemble cells at the base of the cell columns from where EVT derives. In contrast, organoids are similar to VCT and undergo spontaneous SCT differentiation. A defining feature of human trophoblast is that VCT and SCT are human leukocyte antigen (HLA) null, whereas EVT expresses HLA-C, -G and -E molecules. We find that trophoblast organoids retain these in vivo characteristics. In contrast, TSCs express classical HLA-A and HLA-B molecules, and maintain their expression after EVT differentiation, with upregulation of HLA-G. Furthermore, HLA expression in TSCs differs when grown in 3D rather than in 2D, suggesting that mechanical cues are important. Our results can be used to select the most suitable model for the study of trophoblast development, function and pathology.


Subject(s)
Models, Biological , Trophoblasts/cytology , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Female , HLA Antigens/genetics , HLA Antigens/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Organoids/cytology , Organoids/growth & development , Organoids/metabolism , Placentation , Pregnancy , Stem Cells/cytology , Stem Cells/metabolism , Transcriptome , Trophoblasts/metabolism
6.
Commun Biol ; 4(1): 651, 2021 06 17.
Article in English | MEDLINE | ID: mdl-34140633

ABSTRACT

Assessment of the endometrium often necessitates a biopsy, which currently involves an invasive, transcervical procedure. Here, we present an alternative technique based on deriving organoids from menstrual flow. We demonstrate that organoids can be derived from gland fragments recovered from menstrual flow. To confirm they faithfully reflect the in vivo state we compared organoids derived from paired scratch biopsies and ensuing menstrual flow from patients undergoing in vitro fertilisation (IVF). We demonstrate that the two sets of organoids share the same transcriptome signature, derivation efficiency and proliferation rate. Furthermore, they respond similarly to sex steroids and early-pregnancy hormones, with changes in morphology, receptor expression, and production of 'uterine milk' proteins that mimic those during the late-secretory phase and early pregnancy. This technique has wide-ranging impact for non-invasive investigation and personalised approaches to treatment of common gynaecological conditions, such as endometriosis, and reproductive disorders, including failed implantation after IVF and recurrent miscarriage.


Subject(s)
Endometrium/cytology , Menstruation , Organoids/cytology , Adult , Cells, Cultured , Endometrium/growth & development , Endometrium/metabolism , Female , Fertilization in Vitro , Humans , Organoids/growth & development , Organoids/metabolism , Pilot Projects
7.
Elife ; 102021 06 25.
Article in English | MEDLINE | ID: mdl-34170818

ABSTRACT

Normal function of the placenta depends on the earliest developmental stages when trophoblast cells differentiate and invade into the endometrium to establish the definitive maternal-fetal interface. Previously, we identified the ubiquitously expressed tumour suppressor BRCA1-associated protein 1 (BAP1) as a central factor of a novel molecular node controlling early mouse placentation. However, functional insights into how BAP1 regulates trophoblast biology are still missing. Using CRISPR/Cas9 knockout and overexpression technology in mouse trophoblast stem cells, here we demonstrate that the downregulation of BAP1 protein is essential to trigger epithelial-mesenchymal transition (EMT) during trophoblast differentiation associated with a gain of invasiveness. Moreover, we show that the function of BAP1 in suppressing EMT progression is dependent on the binding of BAP1 to additional sex comb-like (ASXL1/2) proteins to form the polycomb repressive deubiquitinase (PR-DUB) complex. Finally, both endogenous expression patterns and BAP1 overexpression experiments in human trophoblast stem cells suggest that the molecular function of BAP1 in regulating trophoblast differentiation and EMT progression is conserved in mice and humans. Our results reveal that the physiological modulation of BAP1 determines the invasive properties of the trophoblast, delineating a new role of the BAP1 PR-DUB complex in regulating early placentation.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Animals , Cell Differentiation/genetics , Gene Expression Regulation , Humans , Mice , Repressor Proteins/metabolism , Trophoblasts/physiology , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism
8.
Reproduction ; 161(5): R113-R127, 2021 05.
Article in English | MEDLINE | ID: mdl-33621191

ABSTRACT

Infertility is a common problem in modern societies with significant socio-psychological implications for women. Therapeutic interventions are often needed which, depending on the cause, can either be medical treatment, surgical procedures or assisted reproductive technology (ART). However, the treatment of infertility is not always successful due to our limited understanding of the preparation of the lining of the uterus, the endometrium, for pregnancy. The endometrium is of central importance for successful reproduction as it is the site of placental implantation providing the interface between the mother and her baby. Due to the dynamic, structural and functional changes the endometrium undergoes throughout the menstrual cycle, it is challenging to study. A major advancement is the establishment of 3D organoid models of the human endometrium to study this dynamic tissue in health and disease. In this review, we describe the changes that the human endometrium undergoes through the different phases of the menstrual cycle in preparation for pregnancy. We discuss defects in the processes of endometrial repair, decidualization and acquisition of receptivity that are associated with infertility. Organoids could be utilized to investigate the underlying cellular and molecular mechanisms occurring in non-pregnant endometrium and early pregnancy. These studies may lead to therapeutic applications that could transform the treatment of reproductive failure.


Subject(s)
Endometrium/cytology , Infertility, Female/pathology , Infertility, Female/therapy , Organoids/cytology , Organoids/physiology , Reproduction , Female , Humans
9.
J Mol Med (Berl) ; 99(4): 531-553, 2021 04.
Article in English | MEDLINE | ID: mdl-33580825

ABSTRACT

Healthy functioning of the female reproductive tract (FRT) depends on balanced and dynamic regulation by hormones during the menstrual cycle, pregnancy and childbirth. The mucosal epithelial lining of different regions of the FRT-ovaries, fallopian tubes, uterus, cervix and vagina-facilitates the selective transport of gametes and successful transfer of the zygote to the uterus where it implants and pregnancy takes place. It also prevents pathogen entry. Recent developments in three-dimensional (3D) organoid systems from the FRT now provide crucial experimental models that recapitulate the cellular heterogeneity and physiological, anatomical and functional properties of the organ in vitro. In this review, we summarise the state of the art on organoids generated from different regions of the FRT. We discuss the potential applications of these powerful in vitro models to study normal physiology, fertility, infections, diseases, drug discovery and personalised medicine.


Subject(s)
Genitalia, Female/cytology , Organoids , Animals , CRISPR-Cas Systems , Drug Discovery/methods , Estrous Cycle/physiology , Extracellular Matrix/physiology , Female , Fertility , Gene Editing/methods , Gene Editing/trends , Genital Diseases, Female/pathology , Genital Diseases, Female/therapy , Genitalia, Female/anatomy & histology , Genitalia, Female/physiology , Gestational Age , Gonadal Steroid Hormones/physiology , Humans , Maternal-Fetal Exchange , Mice , Organoids/cytology , Pituitary Hormones, Anterior/physiology , Placenta/cytology , Precision Medicine/methods , Precision Medicine/trends , Pregnancy , Stem Cells/cytology , Tissue Engineering/methods , Tissue Engineering/trends
10.
Cell Death Differ ; 28(1): 35-51, 2021 01.
Article in English | MEDLINE | ID: mdl-32494027

ABSTRACT

Both the proper functioning of the female reproductive tract (FRT) and normal placental development are essential for women's health, wellbeing, and pregnancy outcome. The study of the FRT in humans has been challenging due to limitations in the in vitro and in vivo tools available. Recent developments in 3D organoid technology that model the different regions of the FRT include organoids of the ovaries, fallopian tubes, endometrium and cervix, as well as placental trophoblast. These models are opening up new avenues to investigate the normal biology and pathology of the FRT. In this review, we discuss the advances, potential, and limitations of organoid cultures of the human FRT.


Subject(s)
Biomedical Research , Genital Diseases, Female , Genital Neoplasms, Female , Organoids , Reproductive Medicine , Animals , Cell Culture Techniques, Three Dimensional , Drug Evaluation, Preclinical , Endometrium , Fallopian Tubes , Female , Humans , Ovary , Pregnancy , Trophoblasts
11.
Placenta ; 102: 21-26, 2020 12.
Article in English | MEDLINE | ID: mdl-33218574

ABSTRACT

Development of the placenta must always be in advance of that of the embryo. Evidence from domestic species demonstrates that the placenta is capable of stimulating its own development through a signalling dialogue with the endometrial glands. Placental lactogens produced by the trophoblast lead to increased expression and release of uterine secretions and mitogenic growth factors, including epidermal growth factor, that have a close temporal and spatial relationship with trophoblast proliferation. Here, we review evidence that an equivalent mechanism operates in the human. The same repertoire of receptors is present on the endometrial gland cells, and the epithelial cells have long been known to adopt a hypersecretory phenotype following an implantation. Furthermore, early pregnancy hormones stimulate the secretion of glycodelin-A and osteopontin, two 'uterine milk proteins' that have multiple potential effects at the maternal-placental interface, from organoid cultures derived from endometrial glands. Prolactin appears to be an important stimulant, but unlike in domestic species the human trophoblast does not secrete this hormone. Instead, it is a major product of decidual cells. Hence, complications of pregnancy that have their pathophysiological roots in deficient trophoblast proliferation may be due primarily to problems of decidualisation. Ensuring the endometrium is in an optimal state pre-conceptionally should therefore be a priority for women's health. Trophoblast stemness and proliferative capacity show a sharp decline at the switch from histotrophic to haemotrophic nutrition. This may reflect the increase in oxygen concentration or loss of growth factor support. Either way, there are implications for adaptive growth of the organ.


Subject(s)
Endometrium/physiology , Placentation , Animals , Bodily Secretions/physiology , Embryonic Development , Female , Humans , Maternal-Fetal Exchange , Pregnancy
12.
Reproduction ; 160(6): 819-831, 2020 12.
Article in English | MEDLINE | ID: mdl-33112764

ABSTRACT

The endometrium, the inner uterine lining, is composed of cell layers that come in direct contact with an embryo during early pregnancy and later with the fetal placenta. The endometrium is responsible for signals associated with normal reproductive cyclicity as well as maintenance of pregnancy. In the mare, functionally competent in vitro models of the endometrium have not been successful. Furthermore, the ability to study various reproductive processes in vitro may allow critical evaluation of signaling pathways involved in the reproductive diseases of animals that cannot be handled frequently, such as various wildlife species. Here we report the establishment of organoids, 3D structures, derived from fresh and frozen-thawed equine endometrium (Equus ferus caballus and E. f. przewalskii). Although organoids from domestic mares responded to exogenous hormonal stimuli, organoids from Przewalski's horse failed to respond to exogenous hormones. The present study represents a 'first' for any large animal model or endangered species. These physiologically functional organoids may facilitate improved understanding of normal reproductive mechanisms, uterine pathologies, and signaling mechanisms between the conceptus and endometrium and may lead to the development of novel bioassays for drug discovery.


Subject(s)
Endometrium/drug effects , Gene Expression Regulation/drug effects , Hormones/pharmacology , Organoids/drug effects , Animals , Animals, Wild , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Endometrium/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Horses , Organoids/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism
13.
Nat Protoc ; 15(10): 3441-3463, 2020 10.
Article in English | MEDLINE | ID: mdl-32908314

ABSTRACT

The human placenta is essential for successful reproduction. There is great variation in the anatomy and development of the placenta in different species, meaning that animal models provide limited information about human placental development and function. Until recently, it has been impossible to isolate trophoblast cells from the human placenta that proliferate in vitro. This has limited our ability to understand pregnancy disorders. Generating an in vitro model that recapitulates the unique features of the human placenta has been challenging. The first in vitro model system of human trophoblast that could be cultured long term and differentiated to syncytiotrophoblast (SCT) and extravillous trophoblast (EVT) was a two-dimensional (2D) culture system of human trophoblast stem cells. Here, we describe a protocol to isolate trophoblast from first-trimester human placentas that can be grown long term in a three-dimensional (3D) organoid culture system. Trophoblast organoids can be established within 2-3 weeks, passaged every 7-10 d, and cultured for over a year. The structural organization of these human trophoblast organoids closely resembles the villous placenta with a layer of cytotrophoblast (VCT) that differentiates into superimposed SCT. Altering the composition of the medium leads to differentiation of the trophoblast organoids into HLA-G+ EVT cells which rapidly migrate and invade through the Matrigel droplet in which they are cultured. Our previous research confirmed that there is similarity between the trophoblast organoids and in vivo placentas in their transcriptomes and ability to produce placental hormones. This organoid culture system provides an experimental model to investigate human placental development and function as well as interactions of trophoblast cells with the local and systemic maternal environment.


Subject(s)
Cell Culture Techniques/methods , Placenta/cytology , Trophoblasts/cytology , Cell Differentiation , Female , Humans , Organoids/cytology , Organoids/metabolism , Placenta/metabolism , Pregnancy , Stem Cells , Trophoblasts/metabolism , Trophoblasts/physiology
14.
Dev Cell ; 54(3): 295-296, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32781021

ABSTRACT

A fully functional placenta is critical for a successful pregnancy. In this issue of Developmental Cell, Singh et al. reveal that excessive placental DNA damage in murine models for Cornelia de Lange syndrome results in an inefficient and senescent placenta that impairs embryonic development.


Subject(s)
De Lange Syndrome , Animals , DNA Damage , Female , Mice , Placenta , Pregnancy
15.
Hum Reprod Update ; 26(4): 501-513, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32441309

ABSTRACT

BACKGROUND: In humans, inadequate trophoblast invasion into the decidua is associated with the 'great obstetrical syndromes' which include pre-eclampsia, foetal growth restriction (FGR) and stillbirth. The mechanisms regulating invasion remain poorly understood, although interactions with the uterine environment are clearly of central importance. Extravillous trophoblast (EVT) cells invade the uterus and transform the spiral arteries. Progress in understanding how they invade has been limited due to the lack of good in vitro models. Firstly, there are no non-malignant cell lines that have an EVT phenotype. Secondly, the invasion assays used are of limited use for the small numbers of primary EVT available from first-trimester placentas. We discuss recent progress in this field with the generation of new EVT lines and invasion assays using microfluidic technology. OBJECTIVE AND RATIONALE: Our aim is to describe the established models used to study human trophoblast invasion in vivo and in vitro. The difficulties of obtaining primary cells and cell lines that recapitulate the phenotype of EVT are discussed together with the advantages and pitfalls of the different invasion assays. We compare these traditional end point assays to microfluidic assays where the dynamics of migration can be measured. SEARCH METHODS: Relevant studies were identified by PubMed search, last updated on February 2020. A search was conducted to determine the number of journal articles published using the cell lines JEG-3, BeWo, JAR, HTR-8/Svneo, Swan-71 and primary human extravillous trophoblast in the last 5 years. OUTCOMES: Deep trophoblast invasion into the maternal decidua is a particular feature of human pregnancy. This invasion needs to be finely regulated to allocate resources between mother and baby. A reliable source of EVT is needed to study in vitro how the uterine environment regulates this process. First, we critically discuss the issues with the trophoblast cell lines currently used; for example, most of them lack expression of the defining marker of EVT, HLA-G. Recently, advances in human stem cell and organoid technology have been applied to extraembryonic tissues to develop trophoblast cell lines that can grow in two (2D) and three dimensions (3D) and differentiate to EVT. This means that the 'trophoblast' cell lines currently in use should rapidly become obsolete. Second, we critically discuss the problems with assays to study trophoblast invasion. These lack physiological relevance and have simplified migration dynamics. Microfluidic assays are a powerful tool to study cell invasion because they require only a few cells, which are embedded in 3D in an extracellular matrix. Their major advantage is real-time monitoring of cell movement, enabling detailed analysis of the dynamics of trophoblast migration. WIDER IMPLICATIONS: Trophoblast invasion in the first trimester of pregnancy remains poorly understood despite the importance of this process in the pathogenesis of pre-eclampsia, FGR, stillbirth and recurrent miscarriage. The new technologies described here will allow investigation into this critical process.


Subject(s)
Cell Culture Techniques/methods , Cell Movement/physiology , Trophoblasts/physiology , Abortion, Habitual/metabolism , Abortion, Habitual/pathology , Cell Line , Embryo Implantation/physiology , Female , Humans , Models, Biological , Placenta/cytology , Placenta/metabolism , Placenta/pathology , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Pregnancy , Pregnancy Trimester, First , Trophoblasts/cytology
16.
Interface Focus ; 10(2): 20190079, 2020 Apr 06.
Article in English | MEDLINE | ID: mdl-32194932

ABSTRACT

The endometrium is the secretory lining of the uterus that undergoes dynamic changes throughout the menstrual cycle in preparation for implantation and a pregnancy. Recently, endometrial organoids (EO) were established to study the glandular epithelium. We have built upon this advance and developed a multi-cellular model containing both endometrial stromal and epithelial cells. We use porous collagen scaffolds produced with controlled lyophilization to direct cellular organization, integrating organoids with primary isolates of stromal cells. The internal pore structure of the scaffold was optimized for stromal cell culture in a systematic study, finding an optimal average pore size of 101 µm. EO seeded organize to form a luminal-like epithelial layer, on the surface of the scaffold. The cells polarize with their apical surface carrying microvilli and cilia that face the pore cavities and their basal surface attaching to the scaffold with the formation of extracellular matrix proteins. Both cell types are hormone responsive on the scaffold, with hormone stimulation resulting in epithelial differentiation and stromal decidualization.

17.
Development ; 146(22)2019 11 27.
Article in English | MEDLINE | ID: mdl-31776138

ABSTRACT

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.


Subject(s)
Placenta/physiology , Placentation , Trophoblasts/physiology , Animals , Bioengineering , Cell Culture Techniques , Cell Differentiation , Cell Lineage , Decidua/physiology , Endometrium/pathology , Female , Fetal Growth Retardation , Humans , Immune System , Leukocytes/cytology , Mice , Organoids , Placenta/cytology , Pre-Eclampsia/physiopathology , Pregnancy , Pregnancy Complications , Sequence Analysis, RNA , Single-Cell Analysis , Trophoblasts/cytology , Uterus/pathology
18.
Nature ; 564(7735): 263-267, 2018 12.
Article in English | MEDLINE | ID: mdl-30487605

ABSTRACT

The placenta is the extraembryonic organ that supports the fetus during intrauterine life. Although placental dysfunction results in major disorders of pregnancy with immediate and lifelong consequences for the mother and child, our knowledge of the human placenta is limited owing to a lack of functional experimental models1. After implantation, the trophectoderm of the blastocyst rapidly proliferates and generates the trophoblast, the unique cell type of the placenta. In vivo, proliferative villous cytotrophoblast cells differentiate into two main sub-populations: syncytiotrophoblast, the multinucleated epithelium of the villi responsible for nutrient exchange and hormone production, and extravillous trophoblast cells, which anchor the placenta to the maternal decidua and transform the maternal spiral arteries2. Here we describe the generation of long-term, genetically stable organoid cultures of trophoblast that can differentiate into both syncytiotrophoblast and extravillous trophoblast. We used human leukocyte antigen (HLA) typing to confirm that the organoids were derived from the fetus, and verified their identities against four trophoblast-specific criteria3. The cultures organize into villous-like structures, and we detected the secretion of placental-specific peptides and hormones, including human chorionic gonadotropin (hCG), growth differentiation factor 15 (GDF15) and pregnancy-specific glycoprotein (PSG) by mass spectrometry. The organoids also differentiate into HLA-G+ extravillous trophoblast cells, which vigorously invade in three-dimensional cultures. Analysis of the methylome reveals that the organoids closely resemble normal first trimester placentas. This organoid model will be transformative for studying human placental development and for investigating trophoblast interactions with the local and systemic maternal environment.


Subject(s)
Maternal-Fetal Relations , Models, Biological , Organoids/cytology , Organoids/physiology , Placentation , Tissue Culture Techniques , Trophoblasts/cytology , Trophoblasts/physiology , Cell Differentiation , Cell Movement , Chorionic Gonadotropin/metabolism , DNA Methylation , Decidua/cytology , Female , Growth Differentiation Factor 15/metabolism , HLA Antigens/metabolism , Humans , Organoids/metabolism , Pregnancy , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Transcriptome/genetics , Trophoblasts/metabolism
19.
Nature ; 563(7731): 347-353, 2018 11.
Article in English | MEDLINE | ID: mdl-30429548

ABSTRACT

During early human pregnancy the uterine mucosa transforms into the decidua, into which the fetal placenta implants and where placental trophoblast cells intermingle and communicate with maternal cells. Trophoblast-decidual interactions underlie common diseases of pregnancy, including pre-eclampsia and stillbirth. Here we profile the transcriptomes of about 70,000 single cells from first-trimester placentas with matched maternal blood and decidual cells. The cellular composition of human decidua reveals subsets of perivascular and stromal cells that are located in distinct decidual layers. There are three major subsets of decidual natural killer cells that have distinctive immunomodulatory and chemokine profiles. We develop a repository of ligand-receptor complexes and a statistical tool to predict the cell-type specificity of cell-cell communication via these molecular interactions. Our data identify many regulatory interactions that prevent harmful innate or adaptive immune responses in this environment. Our single-cell atlas of the maternal-fetal interface reveals the cellular organization of the decidua and placenta, and the interactions that are critical for placentation and reproductive success.


Subject(s)
Cell Communication , Fetus/cytology , Histocompatibility, Maternal-Fetal/immunology , Placenta/cytology , Placenta/metabolism , Pregnancy/immunology , Single-Cell Analysis , Cell Communication/immunology , Cell Differentiation/genetics , Decidua/cytology , Decidua/immunology , Decidua/metabolism , Female , Fetus/immunology , Fetus/metabolism , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Ligands , Placenta/immunology , RNA, Small Cytoplasmic/genetics , Sequence Analysis, RNA , Stromal Cells/cytology , Stromal Cells/metabolism , Transcriptome , Trophoblasts/cytology , Trophoblasts/immunology , Trophoblasts/metabolism
20.
Development ; 145(16)2018 04 16.
Article in English | MEDLINE | ID: mdl-29540503

ABSTRACT

During pregnancy the trophoblast cells of the placenta are the only fetal cells in direct contact with maternal blood and decidua. Their functions include transport of nutrients and oxygen, secretion of pregnancy hormones, remodelling of the uterine arteries, and communicating with maternal cells. Despite the importance of trophoblast cells in placental development and successful pregnancy, little is known about the identity, location and differentiation of human trophoblast progenitors. We identify a proliferative trophoblast niche at the base of the cytotrophoblast cell columns in first trimester placentas that is characterised by integrin α2 (ITGA2) expression. Pulse-chase experiments with 5-iodo-2'-deoxyuridine indicate that these cells might contribute to both villous (VCT) and extravillous (EVT) lineages. These proliferating trophoblast cells can be isolated by flow cytometry using ITGA2 as a marker and express genes from both VCT and EVT. Microarray expression analysis shows that ITAG2+ cells display a unique transcriptional signature, including genes involved in NOTCH signalling, and exhibit a combination of epithelial and mesenchymal characteristics. ITGA2 thus marks a niche allowing the study of pure populations of trophoblast progenitor cells.


Subject(s)
Integrin alpha2/metabolism , Placenta/cytology , Placentation/physiology , Receptor, Notch1/metabolism , Stem Cells/cytology , Trophoblasts/cytology , Biomarkers/metabolism , Cell Proliferation , Female , Humans , Placenta/metabolism , Pregnancy , Pregnancy Trimester, First , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...