Skip to main content
Biology of Reproduction logoLink to Biology of Reproduction
. 2018 Feb 22;99(1):196–211. doi: 10.1093/biolre/ioy049

Hemochorial placentation: development, function, and adaptations

Michael J Soares 1,2,, Kaela M Varberg 1, Khursheed Iqbal 1
PMCID: PMC6044390  PMID: 29481584

Abstract

Placentation is a reproductive adaptation that permits fetal growth and development within the protected confines of the female reproductive tract. Through this important role, the placenta also determines postnatal health and susceptibility to disease. The hemochorial placenta is a prominent feature in primate and rodent development. This manuscript provides an overview of the basics of hemochorial placental development and function, provides perspectives on major discoveries that have shaped placental research, and thoughts on strategies for future investigation.

Keywords: trophoblast, hemochorial placentation, pregnancy


The review presents basic concepts of hemochorial placentation, discusses significant contributions to the field, and highlights experimental approaches for future investigation.

Introduction

Everyone had a placenta, but few of us know much about its contributions to the most critical time of our existence, and at a broader level appreciate its importance in the survival of our species. The placenta is much more than reproduction. It sets the stage for all aspects of embryonic and fetal development and largely determines postnatal health. The placenta has a rich history of scientific discovery, some designed and some through serendipity. Investigation of the placenta is somewhat unique and has had its challenges. Evolutionary divergence and ethical considerations have presented experimental obstacles for placental research. Additionally, nomenclature differences across species have also proven to be a hindrance. In this review, we provide a perspective on the placenta and placental research, as well as offer some thoughts for future investigation. The focus will be on hemochorial placentation, which reflects the authors’ familiarity. We approach the task from a trophoblast-centric perspective that is biased by our past research efforts. Initially, we will provide a general framework for understanding placenta development and function and then proceed to highlight discoveries that have shaped placenta research and our understanding of hemochorial placentation. The reader is directed to a series of excellent recent reviews, which expertly discuss a wide range of topics related to placental development and function [1–7].

What is a placenta?

A placenta is a reproductive adaptation that enabled viviparity to become a successful reproductive strategy [8]. The placenta is comprised of specialized epithelial cell types, collectively referred to as trophoblast cells, situated among mesenchymal cells and vasculature, at the maternal-fetal interface. Trophoblast cells are specialized cell types capable of accessing and modifying maternal structures and controlling the bidirectional flow of nutrients and wastes [9]. Effectively, the placenta and its cellular constituents regulate the milieu in which the fetus develops. Among species, different types of placentas exist and exhibit a range of shapes, sizes, cellular constituents, and levels of integration within the female reproductive tract [6]. Humans, the rat and mouse, and many nonhuman primates possess a hemochorial placenta, characterized by a limited cellular barrier for maternal-fetal delivery of nutrients and, in some instances, extensive intrauterine trophoblast cell invasion and uterine arterial remodeling.

Trophoblast cell lineage

Embryonic development is initiated at fertilization by the blending of sperm and egg haploid genomes. The initial cell differentiation event during embryonic development is the establishment of the trophoblast cell lineage [10–12]. As an embryo increases in cell number, cell–cell interactions, and microenvironments change. Cells on the surface of the embryo are directed to differentiate into the trophoblast lineage. These early trophoblast cells, referred to as trophectoderm, encase a cluster of cells called the inner cell mass (ICM). Trophectoderm cells further differentiate into parenchymal cell lineages comprising the placenta, whereas cells from the ICM have the capacity to develop into the embryo and other extraembryonic tissues (allantois, amnion, yolk sac). ICM–trophoblast cell signaling is vital to the growth and development of the trophoblast lineage [13, 14]. Development of specialized trophoblast cell lineages is engineered through interactions with maternal and fetal structures.

Specialized trophoblast cell lineages

Not all trophoblast cells are alike. As stated above, trophoblast stem (TS) and progenitor cell populations give rise to specialized trophoblast cell lineages (Table 1). In the mouse and the rat, TS and progenitor cells differentiate into trophoblast giant cells, spongiotrophoblast cells, glycogen trophoblast cells, invasive trophoblast cells, and syncytiotrophoblast [15–17]. Trophoblast giant cells and syncytiotrophoblast arise via endoreduplication and cell fusion, respectively, which are processes that overtly alter nucleocytoplasmic organization. The human trophoblast lineage also includes TS and progenitor cell populations, which are generically classified within the moniker of cytotrophoblast [2, 18]. This classification is broad and includes both mitotic and postmitotic cell types. Invasive trophoblast cell lineages (extravillous trophoblast) and syncytiotrophoblast also exist in the human placentation site [19, 20]. Collectively, these specialized trophoblast cell types can be defined by their epithelial nature, their location within the placentation site, and their functionality. The precise hierarchy of trophoblast cell lineage development has not been determined in rodents or the human.

Table 1.

Trophoblast cell types.

Cell type Speciesa Placentation site location Principal function(s)
Trophectoderm Mouse, rat, human Outer cellular layer of blastocyst Embryo implantation; inner cell mass/epiblast protection; source of trophoblast stem and progenitor cells
Trophoblast stem progenitor cells Mouse, rat Prominently located in extraembryonic ectoderm; ectoplacental cone Seed differentiated trophoblast lineages
Cytotrophoblast Human Beneath syncytiotrophoblast layer (villous compartment); Base of extravillous trophoblast column (extravillous compartment) Trophoblast stem and progenitor cell population; seed differentiated trophoblast lineages; also, postmitotic component of villous compartment
Primary syncytium Human Multinucleated trophoblast at the embryo implantation site of early pregnancy Invasion/facilitating embryo interstitial implantation into the uterine decidua
Trophoblast giant cell Mouse, rat Junctional zone: uterine-placental interface (primary), labyrinth zone (secondary) Endocrine activity: steroid and peptide hormonogenesis
Glycogen trophoblast cell Mouse, rat Junctional zone (transient) Energy reserve
Spongiotrophoblast cell Mouse, rat Junctional zone Endocrine activity: peptide hormonogenesis
Endovascular invasive trophoblast cell Mouse, rat, human Intrauterine: intravascular; extravillous compartment (human) Uterine spiral artery restructuring
Interstitial invasive trophoblast cell Mouse, rat, human Intrauterine: extravascular; extravillous compartment (human) Placental anchor; uterine stroma remodeling
Syncytiotrophoblast Mouse, rat, human Labyrinth zone (mouse, rat); villous compartment (human) Barrier/transport (mouse, rat, human); endocrine activity: steroid and peptide hormonogenesis (human)

aTrophoblast cell types found in mouse, rat, and human.

How are trophoblast cells organized into a placentation site?

Embryo implantation is different across species, which affects organization of trophoblast cells and placentation [12]. In the mouse and rat, folds develop within the uterine lumen to create a chamber where mural (abembryonic) trophectoderm attaches to the antimesometrial uterine epithelium. Trophectoderm attachment initiates differentiation of uterine stromal cells into decidual cells, followed by mesometrial uterine epithelial cell death and expansion of the primordial placentation site into the mesometrial decidua. Alternatively, interstitial embryo implantation occurs in the human when polar trophectoderm cells attach to uterine epithelium facilitating complete penetration of the embryo into the underlying decidualized uterine stroma. In the human, this embedding process is facilitated by differentiation of trophectoderm into a primary syncytium [12, 21]. Formation of a primary syncytium associated with embryo implantation is also observed in the guinea pig but not the mouse or rat [22, 23]. As hemochorial placentation progresses, two organizing centers emerge and include the (i) maternal interface and (ii) fetal interface (Figure 1). These organizing centers are determined by the depth extraembryonic mesenchyme and associated vasculature penetrate into the trophoblast compartment. The region encompassing trophoblast and extraembryonic mesenchyme interaction delineates the labyrinth zone of the mouse and rat placenta and the villous compartment of the human placenta [16, 21, 24]. Labyrinth and villous compartments are further demarcated by the presence of syncytiotrophoblast layers. The villous compartment of the human placenta is subdivided into functional units referred to as cotyledons, which are each bathed by maternal blood. Trophoblast cells extending beyond the trophoblast-extraembryonic mesenchyme admixture are located at the maternal boundary. These trophoblast cells are arranged into defined structures in both rodents and the human, and are referred to as the junctional zone and extravillous trophoblast column, respectively [15, 16, 18, 25]. As gestation advances, invasive trophoblast cells arise from the junctional zone in the mouse and rat, and from the extravillous trophoblast column in the human, and migrate into the uterine parenchyma [25–28]. Two types of invasive trophoblast cells have been identified and include (i) interstitial and (ii) endovascular. Interstitial invasive trophoblast cells coalesce between the uterine vasculature, whereas endovascular invasive trophoblast infiltrate uterine blood vessels, especially arteries/arterioles, and supplant the vascular endothelium. During invasive trophoblast cell differentiation, shifts occur in integrin expression altering interactions with surrounding extracellular matrices [29]. As endovascular invasive trophoblast cells differentiate, they acquire an endothelial cell-like phenotype [30–32]. The rat and human exhibit deep intrauterine trophoblast cell invasion, whereas dissemination of trophoblast cells into the mouse uterine parenchyma is nominal [16, 33, 34]. Cellular constituents of the maternal uterine interface, including decidual cells, endometrial glands, and immune/inflammatory cell populations, modulate trophoblast cell behavior (see below).

Figure 1.

Figure 1.

Schematic representations of hemochorial placentation sites showing homologous structures within human, rat, and mouse. Note human and rat exhibit deep intrauterine trophoblast invasion, whereas trophoblast invasion in the mouse is shallow. (adapted from Ref. 241)

Placental function

Trophoblast cells support development through facilitating delivery of maternal resources to the fetus and protecting the fetus from adverse exposures emanating from the mother. Acquisition of maternal nutrients is facilitated through several trophoblast-guided processes, including (i) direct modification of arterial vasculature; (ii) production of hormones, which affect a range of maternal targets that act to maintain pregnancy and mobilize nutrient reserves for fetal development; and (iii) expression of nutrient transporters. Invasive trophoblast cell populations are principal directors of uterine arterial vascular remodeling [27, 28]. Several trophoblast cell populations contribute to the endocrine activities of the placenta; however, chief among them are trophoblast giant cells and spongiotrophoblast in the mouse and rat, and syncytiotrophoblast in the human placenta [35–37]. Syncytiotrophoblast is paramount in facilitating nutrient delivery to the fetus [38–40]. The protective function of the placenta is multifaceted and includes the ability to abate harmful actions of the maternal immune system [41] and through the exclusion and biomodification of xenobiotics [4, 42, 43]. Although all trophoblast cell populations interact at some level with the maternal immune system, those embedded within the uterine parenchyma experience the greatest immune challenge. Syncytiotrophoblast express a robust assortment of efflux transporters and xenobiotic metabolizing enzymes, which act to limit delivery of injurious compounds to the fetus [42, 43]. The barrier function of the placenta is also of critical importance in regulating the vertical transmission of pathogens [44].

Experimental tools for placental investigation

In the preceding paragraphs, we have described several trophoblast cell lineages, morphogenetic processes, and placental functions. Each is critical to the success of pregnancy. Mechanistic insights about placentation have been achieved through key experimental advancements (Figure 2).

Figure 2.

Figure 2.

Timeline of selected technical advancements in placental research. In vitro and in vivo approaches that have positively impacted placental research over the past 50 years are presented.

Learning from placental pathology and pregnancy diseases

Historically, the tools of anatomy and cell biology have had a major impact on placental research, especially studies of human placenta organization as well as inferences about placenta function. Attributes of specific trophoblast cell lineages have been defined and can be monitored within tissues using techniques such as immunocytochemistry, in situ hybridization, or laser capture microdissection. The arrangement of specific trophoblast cell lineages within the placentation site is precisely determined. Aberrations in the organization and behaviors of specific trophoblast cell populations are correlated with pregnancy disease states [27, 28]. This basic premise of linking placental structure and/or function with disease has been examined at many levels with a variety of approaches, including transcriptomics, epigenomics, proteomics, metabolomics, etc. Genome-wide association studies have also been implemented to identify candidate genes regulating placentation in disease states [45].

Assessment of analyses based on placental pathology and pregnancy diseases

The effectiveness of this approach on human placental investigation is based on the availability of a diversity of tissue specimens. Placental samples spanning gestation from both normal and pregnancy disease states are useful resources. However, access to tissue specimens can be dependent upon geographic location and the socio-political climate. The establishment of placental tissue biobanks (e.g. Research Centre for Women's and Infant's Health BioBank, Sinai Health System, Toronto, Ontario, Canada; National Centers for Translational Research in Reproduction and Infertility Human Placental Tissue Bank, University of California, San Francisco, CA, etc.) has helped facilitate placental research. The strength of a human tissue specimen-based research strategy is in establishing correlations that can be further tested with experimental model systems. However, it is important to appreciate that analyses of human placental specimens do not permit the separation of cause versus effect. A 9-month-old (term) placenta can be carefully interrogated, but it represents a compounding history of successful and failed adaptations. Early gestation samples can be informative; however, they are generally devoid of clinical outcomes. Chorionic villus specimens are an intriguing exception [46], with the caveat that their acquisition presaged a potential disease state. Noninvasive retrieval of sloughed trophoblast cells accumulating at the cervix is a relatively new sampling method that provides sufficient material for molecular analyses, including prenatal diagnostics, and opportunities for establishing correlations with pregnancy outcomes [47, 48]. The utility of the trophoblast cells residing in the cervix as harbingers of in situ placenta function is yet to be determined.

Pregnancy disease can arise from multiple etiologies and can be difficult to compartmentalize into homogenous classifications. Biochemical and molecular analyses of the placentation site are advancing beyond assessment of heterogeneous tissue specimens, with current approaches enabling interrogation of specific cell populations and single cells [49–51]. Tissue architecture impacts cell behavior. Thus, in situ validations will be critical in evaluating isolated cell populations and single cell data.

In vitro approaches

Placental research expanded exponentially as regulatory processes governing trophoblast cell development and function were investigated in a culture dish. Several landmark developments that spurred these efforts are highlighted in the following paragraphs.

Transformed/immortalized trophoblast cell lines

First and foremost, the establishment of cell lines from trophoblast cancers, referred to as choriocarcinomas, represented a pivotal development in placental research. These efforts, first reported approximately 50 years ago, yielded choriocarcinoma cell lines including BeWo (ATCC CCL98), JEG-3 (ATCC HTB-36), and JAR (ATCC HTB-144) [52–55]. Importantly, these cell lines continue to be used in placental research. As methodologies for immortalizing and adapting cells to culture emerged, additional cell lines with trophoblast properties were established, e.g., HTR-8/SVneo (ATCC CRL-3271) [56], SGHPL-4 [57], SWAN-71 (Sw.71, Applied Biological Materials Inc., Richmond, BC, Canada) [58], and others [59–61]. Among these various cell models, BeWo cells are unique in that they can be induced to differentiate into syncytiotrophoblast via elevation of intracellular cyclic adenosine monophosphate [62]. BeWo cells have proven particularly useful for elucidating molecular mechanisms controlling syncytialization [63, 64]. Much has been learned with these cell models; however, several challenges remain due to the nature of the procedures used to establish the cell lines. First, it is a challenge to ascribe a specific trophoblast lineage identity to a cell line, and it is difficult to understand the impact of the transformation/immortalization process on the elaboration of an exact trophoblast cell phenotype. The latter is an especially problematic issue when features of trophoblast cell and transformed/immortalized cell behaviors overlap, e.g., proliferation, invasive properties, immune cell evasion, etc.

Primary trophoblast cell and explant cultures

An assortment of primary cultures derived from isolated trophoblast cells and placental explants has provided a means for investigating trophoblast cell biology [65–70]. Strauss and colleagues established an efficient and reproducible technique for isolating human cytotrophoblast [65]. The procedure, or modified versions of the procedure, has been effectively used by many laboratories to investigate fundamental properties of human trophoblast cells. Primary trophoblast culture systems resolve issues of cellular identity, but present challenges due to biological variation associated with their origin, and their limited life span in culture, and their potential heterogeneity. In addition to primary culture systems, in vitro perfusion of isolated human placental cotyledons has proven to be an effective method for examining bidirectional transport and barrier function of the human placenta [71].

Rodent TS cells

In 1983, Teshima and colleagues published a report describing the generation of a transplantable rat cell population with trophoblast properties, including the presence of large cells that resemble trophoblast giant cells [72]. The cells originated from the mid-gestation rat placenta following fetus removal and peritoneal exposure. The resulting tissue mass, was referred to as a choriocarcinoma, and was used to establish cell lines referred to as RCHO [73] and Rcho-1 [74]. The cells proved remarkable in that they could be maintained in a stem/proliferative state or induced to differentiate by mitogen removal [74]. Differentiation was signified by the development of trophoblast giant cells that exhibited functional attributes comparable to trophoblast giant cells of the placenta [74, 75]. Although Rcho-1 cells have been utilized to effectively investigate the regulation of trophoblast cell differentiation [76–80], they also exhibit some limitations which include aneuploidy (pseudo-tetraploid) and autonomy from developmental growth regulators [81].

Authentic TS cells were first derived from mouse blastocysts and extraembryonic ectoderm [82] and subsequently from single blastomeres [83]. Ex vivo proliferation of TS cells is dependent on fibroblast growth factor (FGF) and transforming growth factor beta (TGFB) family regulatory cues, the same signals that control TS, and progenitor cell expansion in the early embryo [82, 84–86]. Mouse TS cells are diploid and can be indefinitely propagated in vitro [82]. When mitogenic factors are removed, TS cells undergo differentiation and when TS cells are reintroduced into an early embryo, they can contribute to all differentiated trophoblast lineages [82]. Two groups independently established defined culture conditions for maintaining mouse TS cells [86, 87]. The essential components defined by Kubaczka and coworkers include FGF4, heparin, and TGFB, with TS cells plated on an extracellular Matrigel matrix [86]. Alternatively, Ohinata and Tsukiyama identified FGF2, activin A, and inhibitors of Rho-associated protein kinase and WNT signaling as key components for TS cell culture [87]. TS cells have also been derived from rat blastocysts [88] and from blastocysts of mouse and rat mutant models [89–91]. Collectively, mouse and rat TS cells have proven to be extraordinary in vitro model systems for investigating regulatory pathways controlling trophoblast cell stem and differentiation states.

Human embryonic stem cell-derived trophoblast

Unlike the rat and mouse, direct expansion of TS cells from human embryos has proven more difficult. Human embryonic stem (ES) cells are readily propagated from human embryos [92], and have become a model system for investigating development of the human trophoblast cell lineage [93]. A serendipitous observation, first reported in 2002 by Thomson and colleagues, showed that exposure of human ES cells to bone morphogenetic protein-4 (BMP4) resulted in the development of the trophoblast cell lineage [94]. Trophoblast derivation from human ES cells has been optimized by the addition of FGF and activin/nodal signaling inhibitors. These human ES-derived trophoblast cells are proposed to represent a window into early events of human trophoblast lineage development [95]. The physiological relevance of BMP-directed differentiation of human ES cells to trophoblast has been debated [61, 93, 96]. Capture of human TS cells from BMP4-treated human ES cell cultures has been elusive.

Human TS cells

At the end of 2017, a report appeared that described a stem cell population established from first trimester human trophoblast tissue or human blastocysts with all the known and expected features of human TS cells [97]. To identify potential signaling pathways necessary to support a stem and proliferative state, Okae and coworkers performed transcriptome profiling of first trimester cytotrophoblast. Their investigation implicated epidermal growth factor (EGF) and WNT signals as candidate regulators of human TS cells. EGF and WNT pathway activation combined with inhibition of epithelial cell differentiation pathways (TGFB, histone deacetylase, and Rho-associated protein kinase inhibitors) resulted in culture conditions conducive to human TS cell expansion. The stem cell population could be induced to differentiate into both syncytiotrophoblast and extravillous trophoblast through modulation of culture conditions. Similarities and differences were noted between rodent and human TS cells. Importantly, a core group of transcription factors associated with the TS cell stem state, including GATA binding protein 3 (GATA3), TEA domain transcription factor 4 (TEAD4), and transcription factor AP-2 gamma (TFAP2C), are shared between mouse and human [61, 97].

“Extended/expanded” potential stem cell populations

Two recent reports describe exciting new tools for investigating trophoblast development in both the human and mouse [98, 99]. Each report used a different approach to generate pluripotent stem cell populations with “extended/expanded” properties. In the first approach, a chemical library screen was used to generate key components of a cocktail (dimethindene maleate, and minocycline hydrochloride) that could be added to leukemia inhibitory factor (LIF) and a glycogen synthase kinase-3 inhibitor to establish human ES cells with an ICM-like, “naïve” state, from human ES cells with an epiblast-like, “primed” state. Subsequently, an equivalent cell population was also established from the mouse [98]. The second approach used a combination of small molecule signaling pathway inhibitors (mitogen-activated protein kinases, SRC proto-oncogene, non-receptor tyrosine kinase, WNT/HIPPO/poly-ADP-ribosylation), known to interfere with blastomere differentiation and LIF to capture a stem cell population equivalent to a blastomere from a 4- or 8-cell mouse embryo [99]. When reintroduced into early embryos, both “extended/expanded” stem cell populations formed chimeras and contributed to both ICM-derived and extraembryonic cell lineages. These results differ dramatically from chimeras generated using mouse ES cells or TS cells, which contribute only to epiblast or trophoblast lineages, respectively [14]. Thus, “extended/expanded” pluripotent stem cells exhibit properties expected for a multipotent blastomere of an early embryo prior to trophoblast lineage determination.

An earlier report, from Fisher and colleagues described the ex vivo expansion of stem cells from 8-cell human embryos [100]. These cells have the capacity for self-renewal, development of ICM-derived germ layers, and differentiation towards both villous and extravillous human trophoblast cell lineages. Interestingly, the human trophoblast progenitor cells prominently express caudal type homeobox 2 (CDX2) [100], a key regulator of mouse trophoblast lineage development [5] that is not a feature characteristic of human TS cells [97]. Collectively, based on these initial reports, it appears that “extended/expanded” potential stem cells may represent new tools for investigation of early decision-making within the trophoblast lineage.

Induced mouse TS cells

Mouse TS cells have also been engineered through introduction of a core group of transcription factors (Eomesodermin, EOMES; GATA3, TFAP2C) into fibroblasts [101, 102]. This significant accomplishment built on earlier discoveries, in which genes controlling trophoblast lineage development were identified using mouse mutagenesis and mouse TS cells [5]. Induced-TS cells behave like authentic TS cells isolated from mouse embryos. These two cell populations possess similar transcriptomic and epigenomic profiles. Furthermore, they can differentiate into specialized trophoblast lineages and when introduced into blastocysts can contribute to the formation of the placenta. Capturing mouse-induced TS cells was facilitated by well-established culture conditions for mouse TS cell propagation.

Assembly of embryo-like structures with mouse ES and TS cells

Mouse ES cells and TS cells can be assembled into three-dimensional extracellular matrix scaffolds to recapitulate several significant embryonic and extraembryonic developmental events [103]. Such an in vitro model system is remarkable, and can be exploited to investigate molecular mechanisms controlling embryonic-extraembryonic cell cross-talk during embryogenesis.

Overview and assessment of in vitro approaches

In vitro experimentation is a powerful tool to investigate molecular mechanisms controlling trophoblast cell biology. An extensive evaluation of the merits and limitations of various human trophoblast cell models is provided by Moffett and coworkers [61]. The culture and manipulation of rodent and, very recently, human TS cells represent major methodological advancements that permit the discovery of regulatory events that control the TS cell stem state as well as trophoblast differentiation. Differences in rodent and human TS cells exist and delayed progress in the ex vivo expansion of human TS cells. Differentiation along the trophoblast giant cell lineage has been the focus of much effort with rodent TS cells. Effective strategies for directing TS cells to other differentiated trophoblast cell lineages are emerging [104]. Several approaches have surfaced for investigating human trophoblast cell differentiation, including the recent establishment of human TS cells [97]. The discovery of culture conditions for sustaining human TS cells will facilitate the identification of core conserved signaling pathways defining the trophoblast lineage. Human TS cells may supplant other in vitro culture systems and become the in vitro model of choice for investigating human trophoblast cell development. Culture conditions have also been established for monkey, vole, rabbit, cow, and pig TS or TS-like cells [105-109]. Finally, it is important to recognize that in vitro analyses illuminate the “potential” of a cell in the context of the culture environment, which may or may not reflect physiology. Hypotheses generated using in vitro models are best tested in vivo. Knowledge of comparative placentation is a prerequisite for identifying the appropriate animal model to match the regulatory process under investigation.

In vivo approaches

Conservation of genes regulating mammalian placentation is evident [2, 110-113]. Approximately, 80% of genes regulating placentation are conserved in their expression and biological activities between mouse and human [112]. Thus, the mouse has been a valuable animal model for studying many aspects of placentation [2, 114]. Organization of rat and human placentation sites shows conservation regarding deep trophoblast invasion and trophoblast-directed remodeling of uterine spiral arteries [16, 33, 34] (Figure 1). Similarities between guinea pig and human placentation are evident, making the guinea pig another intriguing model for investigating the hemochorial placenta [115, 116]. However, the requisite experimental tools (TS cell culture models, antibodies, in vivo genetic manipulation, etc.) for implementing the guinea pig as a model system for placentation research are not yet available. Nonhuman primate models are advantageous in secondary evaluation of regulatory pathways established in other models but have practical limitations (costs, compliance issues, etc.) in most primary analyses. However, in some instances nonhuman primates may be the only relevant alternative for in vivo analysis of primate-specific aspects of placentation.

Targeted mutagenesis in the mouse

The advent of gene targeting in mouse ES cells and subsequent production of mutant mouse models provided useful discovery tools for identifying and dissecting pathways controlling trophoblast lineage development and placentation [114]. Identification of some genes critical to placental development was based on a foundation of earlier experimentation, while identification of others was a consequence of serendipity. Global in vivo gene disruption experiments identified numerous key regulators of the trophoblast lineage and placental development including the following selected genes: Achaete-scute family bHLH transcription factor 2 (Ascl2), Cdx2, Estrogen related receptor, beta (Esrrb), Gata3, Heart and neural crest derivatives expressed 1 (Hand1), ETS proto-oncogene 2 (Ets2), Peroxisome proliferator activated receptor gamma (Pparg), Glial cells missing homolog 1 (Gcm1), Eomes, Tfap2c, E74 like ETS transcription factor 5 (Elf5), and Tead4 [117-132]. These phenotypic outcomes in the mouse directly led to evaluations of functional conservation in human placentation [61, 91, 133-145]. Refinements to mouse mutagenesis techniques resulted in trophoblast lineage-specific targeting using Cre/Lox strategies. Regulatory sequences associated with the human Cytochrome P450 family 19 subfamily A member 1 (Cyp19a1) gene, several mouse genes (Trophoblast specific protein alpha, Tpbpa; Prolactin family 3, subfamily d, member 1, Prl3d1; Prolactin family 2, subfamily c, member 2, Prl2c2; PR/SET domain 1, Prdm1; Keratin 5, Krt5; Prolactin family 3, subfamily b, member 1, Prl3b1; Gcm1; Cathepsin Q, Ctsq), and a chimeric Tpbpa/Adenosine deaminase (Ada) enhancer have all been effectively utilized to produce gene disruptions in specific lineages of trophoblast cells [146-155] (Table 2). Strengths and limitations of Cre mouse lines for placenta-specific gene disruptions have been discussed [156].

Table 2.

Regulatory sequences demonstrated to drive Cre expression in mouse trophoblast cell lineages.

Regulatory sequencea Cellular target(s) References
Cyp19a1 All trophoblast lineages first detectable in extraembryonic ectoderm and ectoplacental cone structures 146
Chimeric Tpbpa/Ada enhancer All trophoblast lineages 151
Krt5 All trophoblast lineages first detectable in the ectoplacental cone; visceral yolk sac 152
Tpbpa Trophoblast progenitors in the ectoplacental cone that give rise to subsets of trophoblast giant cells, spongiotrophoblast, glycogen trophoblast cells 148
Prdm1 Trophoblast progenitors in the junctional zone, spongiotrophoblast, subsets of trophoblast giant cells, and glycogen trophoblast cells 149
Prl3d1 Trophoblast giant cells 150
Prl2c2 Trophoblast giant cells 150
Prl3b1 Placenta (specific cell types were not reported; would expect trophoblast giant cell expression) 153
Ctsq Trophoblast giant cells of the labyrinth zone and lining channels in the junctional zone 155
Gcm1 Syncytiotrophoblast layer-II 147, 154

aAll regulatory sequences are from the mouse except for human Cyp19a1.

Targeted mutagenesis in other species

Over the past few years, an assortment of genome editing strategies (zinc finger nucleases, transcription activator-like effector nucleases, Crispr/Cas9) has been implemented in additional species to generate global gene disruptions [157, 158]. These technical breakthroughs have expanded the utility of other species that may be more appropriate for investigating specific events in the hemochorial placentation process. Genome editing has led to a renaissance in using the rat as an experimental model for investigating hemochorial placental development [16, 91], and will continue to create additional opportunities for in vivo mechanistic approaches in a wider range of species.

Lentiviral-mediated trophectoderm gene delivery

A clever strategy to genetically manipulate trophectoderm on the surface of the mouse blastocyst was independently reported by three groups [159-161]. The approach takes advantage of the tight surface epithelium of the blastocyst for selective and efficient gene delivery to trophectoderm without ICM access as well as the capacity of trophectoderm to develop into all trophoblast cell lineages. In brief, the zona pellucida encasing the embryo is removed, the denuded blastocyst is incubated with lentiviral constructs and transferred into appropriately timed pseudopregnant mice, followed by subsequent analysis of placentation sites at various stages of gestation (Figure 3). Importantly, both gain-of-function and loss-of-function gene manipulations can be performed [162]. This technique has been adapted for manipulating trophectoderm of embryos from other species, including the rat, rabbit, pig, and sheep [163-166]. The merit of the lentiviral trophectoderm delivery approach is the ability to selectively modify the trophoblast lineage.

Figure 3.

Figure 3.

Lentiviral-mediated trophectoderm gene delivery. (a) Procedure for lentiviral-mediated gene delivery. (b–d) Ubiquitin C promoter driven enhanced green fluorescent protein (EGFP) trophectoderm lentiviral delivery, transfer to pseudopregnant rats, and assessment at gestation day 13.5. Fetal–placental tissues were examined under bright field (b), fluorescence microscopy (c), or a merge of panels b and c (d). Note the placental-specific expression of EGFP. Scale bar = 2 mm (adapted from Ref. 163).

Overview and assessment of in vivo approaches

Understanding molecular mechanisms in a physiological context is essential. However, the efficacy of the approach is not simply in using an animal model but in using a relevant animal model. Furthermore, the usefulness of the approach depends on what you are trying to model. Modeling specific physiological processes, or events such as trophoblast cell intrauterine invasion, uterine spiral artery remodeling, or transplacental movement of solutes, can be achieved using an appropriate species. In contrast, modeling the full clinical spectrum of a human pregnancy/placentation disease state or syndrome is inherently more problematic. There are fundamental differences in litter-bearing species versus singleton or twin pregnancies. A negatively impacted pregnancy in a litter-bearing species is associated with activation of mechanisms that cull progeny during pregnancy. The mistake is trying to use one species to model all aspects of placentation and related diseases. Knowledge of hemochorial placentation in a range of species is critical for matching the appropriate animal model to the biology that is being investigated.

Future discovery of conserved regulatory processes controlling hemochorial placental development

Placental research is at an exciting stage. In vitro and in vivo model systems have been established that permit careful dissection of regulatory processes controlling placental development. An effective strategy for future placental research will be the use of a range of TS cell culture systems, appropriately validated animal models that exhibit the relevant trait and that can be genetically manipulated, and the continued interrogation of archived normal and diseased human placental specimens. Combining these experimental tools to address specific biological questions will spur scientific advancement.

Placental biology: selected discoveries

In this section, we highlight discoveries that have impacted our understanding of hemochorial placentation.

Placental structure and pregnancy disease

Relationships between placentation and diseases of pregnancy have been established. This is best illustrated through a landmark discovery of the connection between extravillous trophoblast cells and uterine spiral arteries with preeclampsia [166]. Shallow trophoblast invasion and failures in the modification of the uterine vasculature were identified as pathological features of preeclampsia and subsequently other pregnancy diseases including early pregnancy loss, intrauterine growth restriction, pre-term birth, and placenta abruption [27, 28, 167–169]. Aberrations in trophoblast cell differentiation are apparent in diseases of placentation. Specifically, in preeclampsia, trophoblast cells at the uterine interface fail to acquire an appropriate integrin expression pattern and endothelial cell-like phenotype [170, 171]. Molecular mechanisms underlying the development of invasive trophoblast cell lineages and uterine spiral artery remodeling are poorly understood, as is their efficient removal at the end of pregnancy [172].

Uterine environment—directing placentation

In cancer biology, reference is made to “seed and soil” when describing the ability of tumor cells (seed) to successfully colonize target organs (soil) [173]. Trophoblast cell development within the uterus can be viewed in an analogous manner (trophoblast cell: seed; uterus: soil). During the establishment of pregnancy, the uterus undergoes pivotal changes including the differentiation of uterine stromal cells to multifunctional decidual cells, modulation of endometrial gland function, alterations of the uterine vasculature, and a pronounced redistribution of immune cells within the uterine endometrium [41, 174-177]. Disruptions in uterine development compromise placentation [178-182]. Impairments in uterine stromal cell decidualization may contribute to diseases of placentation, including preeclampsia [183-185] and aging-associated placental insufficiency and pregnancy failure [186].

Identifying the function of various immune cells (natural killer cells, macrophages, dendritic cells, and T-regulatory cells, etc.) within the uterus has been the focus of substantial investigation [41, 176]. As an example, NK cells accumulate in significant numbers at the placentation site and are viewed as both maternal protectors and collaborators in promoting placentation [175, 187, 188]. NK cells carry out several functions including the ability to modify uterine arterial vasculature to facilitate the delivery of nutrients to the placenta [175, 189-191], promote placental development [192], and restrain trophoblast invasion. NK cells effectively regulate the gestational timing of deep placentation [33, 191, 193]. The role of uterine NK cells in restraining trophoblast invasion is not fully accepted. Instead, it has been proffered from in vitro experimentation that uterine NK cells promote trophoblast cell invasiveness [194]; however, this in vitro observation contradicts in vivo findings described above [33, 191, 193]. In human pregnancies, extravillous trophoblast-NK cell signaling is influenced by histocompatibility antigen-killer immunoglobulin-like receptor polymorphic profiles, which influence the depth and extent of intrauterine trophoblast invasion and uterine spiral artery remodeling [195]. Failed pregnancies trigger NK cell killing activity, which contributes to ridding the uterus of compromised extraembryonic and embryonic tissues [188, 196, 197]. Although the NK cell actions listed above are compelling, it is also evident that NK cells are not required for progression of pregnancy, at least when NK cell-deficient mice or rats are maintained in the controlled conditions of a modern research vivarium [191, 193]. It remains possible that resident uterine NK cells, in addition to other immune cells, possess an expanded spectrum of biological roles and an intrinsic purpose in pregnancies that occur in feral environments [198].

Placental endocrine signaling—fodder for evolutionary thought

The placenta is a rich source of hormones, growth factors, and cytokines. Some of the most innovative experimentation prior to the genome era is represented in the identification and characterization of hormones produced by trophoblast cells. These hormones are placental-specific mimics of hormones produced by a wide range of endocrine organs and notably include chorionic gonadotropin (CG), placental lactogen (PL), interferon (IFN), as well as the enzymatic machinery for the biosynthesis of steroid hormones [36, 37, 199-201]. These discoveries were consequential, and in the case of CG became the central component of treatments for infertility and early pregnancy tests [202]. As individual hormone signaling systems were characterized and genomes were sequenced, it became evident that the endocrinology of the placenta exhibits considerable species diversity. This is best demonstrated with a few examples.

CG is a hormone characteristic of primate and equine placentas, but is not present in placentas of other species, including the mouse and rat [203]. PL, which is also referred to as chorionic somatomammotropin, represents an expansion of the growth hormone family in the human [204]. In the rat and mouse, PL was derived from duplications of an ancestral prolactin gene [36]. Furthermore, the expanded prolactin family of the mouse and rat consists of approximately two dozen related proteins representing a diverse array of functions, most of which are not yet fully understood [205-207]. Placentas of some species do not produce hormones with prolactin or growth hormone-related actions [36, 205]. Trophoblast IFNs were discovered through efforts to identify factors controlling the establishment of pregnancy in sheep, a species not exhibiting a hemochorial placenta but instead the less invasive synepitheliochorial placenta [201]. Specifically, these studies identified a protein secreted by trophoblast cells of elongating embryos capable of facilitating the maintenance of corpus luteum function during early pregnancy. This protein was determined to be a new member of the Type I IFN family and was termed IFN tau [208-212]. Other IFNs have been identified in placentas of nonruminant species, but not IFN tau [200]. Finally, the enzymes responsible for the biosynthesis of androgens and estrogens, key hormones regulating reproductive function, exhibit species-specific placental expression profiles. Specifically, 17 alpha hydroxylase (CYP17A1), an enzyme essential for androgen production, is prominently expressed in mouse and rat placentas, but less so in the human placenta [35, 213-216]. Conversely, aromatase (CYP19A1), the rate-limiting enzyme in estrogen biosynthesis, is prominently expressed in human placenta [217], but not in placentas of the mouse and rat [218]. In addition to the hormones listed here, several other examples could be presented to underscore the lack of placenta endocrine system conservation.

Even though most of the tasks required to establish and maintain pregnancy are shared among species, the hormones regulating the physiological adaptations to pregnancy are not necessarily conserved across species. Historically, the extensive divergence in placental endocrinology has been one of the best arguments for not using animal models to investigate the human placenta. Origins of placental hormone diversity arose from fundamental adaptive drivers of evolution such as (i) gene amplification; (ii) selection-induced subfunctionalization and neofunctionalization; (iii) birth of new genes and regulatory sequences. Gene amplification increases protein production and is a known adaptive response to environmental stressors [219]. Once the stressor is removed, genes within the amplified locus can mutate and expand functionality. New genes and regulatory sequences can also arise from co-optation of transposable elements.

Co-opting transposable elements and placental development

As genomes were sequenced, it became apparent that the contribution of DNA designated for coding proteins was a small portion of the total genome, and that the accumulation of repetitive transposable elements in the genome was substantial [220]. Transposable elements arose from ancient viral infections and have served as substrates for the evolution of species, providing contributions to the birth of new genes and regulatory elements controlling gene expression [221-224]. The placenta has provided several examples for co-opting transposable elements. The syncytin genes are one of the best examples of the endogenization of viral genes. Heidmann and colleagues showed that retroviral genes encoding envelope proteins have independently been used in multiple mammalian species to facilitate the process of cell fusion in the development of syncytiotrophoblast [225, 226]. Syncytin gene disruption interferes with the formation of syncytial trophoblast layers in the mouse placenta and compromises pregnancy [227]. In sheep, endogenous Jaagsiekte retroviral genes regulate synepitheliochorial placental development [228]. Furthermore, gene regulatory elements associated with the trophoblast lineage have been generated from transposable elements [229]. The increased involvement of transposable elements in placental biology may be linked to a global hypomethylation of DNA in the placenta [230-234].

Placental plasticity

Ten years ago, we published a report that shifted our thinking regarding the process of placentation [235]. Until that time, we viewed placentation as a programmed outcome of gene regulatory pathways controlling the differentiation of trophoblast cell lineages within the uterine milieu. We did not appreciate how placental development could be shaped by the environment. Prior to the report, we had rediscovered deep placentation in the rat [33] and were motivated to investigate its regulation. Reports suggested that low oxygen hinders the in vitro invasive properties of trophoblast cells [236, 237]. We seized on these findings and sought to use oxygen tension as an in vivo experimental tool with the intent to disrupt intrauterine trophoblast invasion and compromise pregnancy. To our surprise, the low oxygen tensions selected (10%–11% oxygen) evoked a remarkable adaptation. Hypoxia (e.g., low oxygen) exposure led to changes at the placentation site, including expansion of the uterine mesometrial vasculature, alterations to placental compartment sizes, activation of invasive endovascular trophoblast cells, uterine spiral artery remodeling, and, amazingly, maintenance of pregnancy [235] (Figure 4). A critical 24-h period of sensitivity to oxygen tension could be mapped between gestation days 8.5 and 9.5. This adaptive response is highly reproducible in the rat [90, 190] and elements of the adaptive response are also observed in the mouse [238]. Much of what was learned can be summarized in three main lessons: (i) use caution when extrapolating in vitro findings to in vivo animal models; (ii) the intrinsic value for in vivo experimentation is the ability to test mechanisms controlling placentation; and (iii) although hypoxia can be a consequence of a failed placenta and compromised pregnancy, it is also an important signal guiding placental development. Our observations are illustrative of an intrinsic placental/placentation plasticity. Placentas are engineered to adapt to the maternal environment, which has led us to conclude that a signature feature of a healthy placenta is its plasticity [25].

Figure 4.

Figure 4.

Placentation site plasticity: responses to hypoxia exposure during rat pregnancy. (a) Chicken beta actin promoter-enhanced green fluorescent protein (chβA-EGFP) transgenic model for monitoring intrauterine trophoblast cell invasion. (b) Gestation day 13.5 placentation site from a rat exposed to ambient oxygen. (c) Gestation day 13.5 placentation site from a rat exposed to hypoxia (∼11% oxygen) from gestation day 6.5–13.5. Maternal hypoxia activated endovascular trophoblast invasion. Scale bar = 0.5 mm (adapted from Ref. 235).

We have not been the only laboratory to observe an effect of an environmental challenge on the placenta. The organization of placentation sites is affected by an assortment of environmental exposures, including maternal diet, chemical exposures, disease states, infectious agents, stressors, etc. [16, 239, 240]. Although hypoxia-inducible factor has been identified as a regulatory mechanism mediating placental adaptations to low oxygen tensions [90, 190, 241-243], regulatory pathways controlling placental adaptations to other environmental challenges are yet to be identified. We infer that disease results when adaptive pathways impacting placentation are overwhelmed or dysregulated.

Sexual dimorphisms exist in placental responses to environmental challenges [244-247]. In general, early stages of female placenta development show heightened sensitivity to disruption, while male placentas are more vulnerable as gestation advances [247]. Sexual dimorphisms may be due to differences in sex chromosome composition, sex-specific modulation of the epigenome, or the ability to relax allele-specific gene repression when exposed to stressors [240, 248, 249]. Genes exhibiting allele-specific expression (e.g., imprinted genes) are fundamentally involved in the regulation of placentation [250]. Epigenetic modifications dictate allele-specific expression and are mediators of environmental impacts on the genome [240, 251].

It appears that the plasticity of the placenta may be enhanced in comparison to other tissues. Mechanisms that impact the epigenome and genome structure are likely at the core of cellular plasticity. The global DNA hypomethylation characteristic of trophoblast cells is not common, and among other cell types has only been observed in cancer cells [230-234]. Epigenetic modifications can relax DNA repression, thereby enabling structural changes, including copy number variation, activation of transposable elements, access to typically unavailable regions of the genome, and increased responses to environmental stressors [233, 245]. Importantly, these structural changes are the same adaptive mechanisms utilized in the evolution of species. Unlike other cell types, trophoblast cells can tolerate profound changes in the organization of their genomes and continue to function normally [252-254]. Interestingly, DNA structural variations are less abundant in diseased versus normal placentas [255]. If plasticity is a positive attribute of the placenta, and is enabled, at least in part, by the epigenetic landscape, then understanding dynamics of epigenome modification during trophoblast cell development and following environmental exposures should be fruitful research endeavors.

Final Thoughts

In the preceding paragraphs, we described tactics for investigating placentation and highlighted selected observations defining the placentation process. We will close by commenting on some potentially provocative directions of placental research.

We have attempted to convince the reader that meaningful tests of hypotheses directed at understanding hemochorial placentation require in vivo animal experimentation. However, different from other organs, the placenta exhibits considerable species diversity, which has led some to a Homo sapian-centric approach to placenta research [256]. No single species represents the perfect animal model for investigating the human placenta, but species exist that have elements of placentation shared with humans. Consequently, rather than be restrictive and relegated to in vitro human trophoblast cell models and establishing correlations of putative regulators impacted by pathology, we need to embrace species diversity. Experimental tools are in hand that permit expanding the effective repertoire of animal models for placenta research.

The placenta is charged with a monumental task: “provide a controlled environment for fetal development that is optimal for postnatal growth, maturation, and survival.” The origins of adult health and disease can be traced to the placenta [4]. Moreover, postnatal success can be predicted by simple measurements of the size and shape of the placenta at birth [257, 258]. The implications are profound. How do disruptions in placental form and function affect the trajectory of postnatal development? Can we identify sensitive events in placentation affecting postnatal development and in a preventative mode introduce placental modifications via stem cell or pharmaceutic treatments with the goal of optimizing adult health? Such an approach will have merits but circumventing an injurious maternal environment may be a limiting factor in the prospects of a successful pregnancy.

Preterm birth is a serious health problem and financial burden [259]. The impact of a shortened gestation that obviates the protective and nutritive functions of the placenta in conjunction with premature fetal exposure to extrauterine life can have devastating life-long consequences. However, prolonging exposure to an unhealthy in utero environment also has costs. Flake and colleagues recently provided a strategy for extrauterine life maintained by an artificial placenta [260]. Fetal sheep were removed during the last month of gestation by caesarian delivery and placed into a plastic bag instrumented to provide a secure and supportive environment for the promotion of growth and maturation. At term, newborn sheep extricated from the artificial womb were of appropriate size and in good health. Optimization of conditions for extrauterine fetal development is required and will benefit from knowledge of placentation and placental function. Extension of the technology to other species, including the human, will present challenges, including ethical issues, which are commonplace for placenta research. To the lay person, extrauterine fetal development may seem to be science fiction, but to the placental researcher it represents a new opportunity.

Footnotes

Grant Support: The authors would like to thank the research contributions of past and current members of our laboratory and support from the National Institutes of Health (HD020676, HD079363, HD082535) and the Sosland Foundation.

References

  • 1. Gundling WE Jr, Wildman DE. A review of inter- and intraspecific variation in the eutherian placenta. Philos Trans R Soc Lond B Biol Sci 2015; 370:20140072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Maltepe E, Fisher SJ. Placenta: the forgotten organ. Annu Rev Cell Dev Biol 2015; 31:523–552. [DOI] [PubMed] [Google Scholar]
  • 3. Soncin F, Natale D, Parast MM. Signaling pathways in mouse and human trophoblast differentiation: a comparative review. Cell Mol Life Sci 2015; 72:1291–1302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Burton GJ, Fowden AL, Thornburg KL. Placental origins of chronic disease. Physiol Rev 2016; 96:1509–1565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Roberts RM, Green JA, Schulz LC. The evolution of the placenta. Reproduction 2016; 152:R179–R189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Latos PA, Hemberger M. From the stem of the placental tree: trophoblast stem cells and their progeny. Development 2016; 143:3650–3660. [DOI] [PubMed] [Google Scholar]
  • 7. Sibley CP. Treating the dysfunctional placenta. J Endocrinol 2017; 234:R81–R97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Amoroso EC. The evolution of viviparity. Proc R Soc Med 1968; 61:1188–1200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Soares MJ, Varberg KM. Trophoblast. In: Spencer, T, Flaws, J (eds.), Encyclopedia of Reproduction, 2 New York: Elsevier, 2018. [Google Scholar]
  • 10. Biggers JD, Bell JE, Benos DJ. Mammalian blastocyst: transport functions in a developing epithelium. Am J Physiol 1988; 255:C419–C432. [DOI] [PubMed] [Google Scholar]
  • 11. Cockburn K, Rossant J. Making the blastocyst: lessons from the mouse. J Clin Invest 2010; 120:995–1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Aplin JD, Ruane PT. Embryo–epithelium interactions during implantation at a glance. J Cell Sci 2017; 130:15–22. [DOI] [PubMed] [Google Scholar]
  • 13. Gardner RL, Beddington RS. Multi-lineage 'stem' cells in the mammalian embryo. J Cell Sci 1988; 10(Supplement 10):11–27. [DOI] [PubMed] [Google Scholar]
  • 14. Rossant J. Stem cells from the mammalian blastocyst. Stem Cells 2001; 19:477–482. [DOI] [PubMed] [Google Scholar]
  • 15. Simmons DG, Cross JC. Determinants of trophoblast lineage and cell subtype specification in the mouse placenta. Dev Biol 2005; 284:12–24. [DOI] [PubMed] [Google Scholar]
  • 16. Soares MJ, Chakraborty D, Karim Rumi MA, Konno T, Renaud SJ. Rat placentation: an experimental model for investigating the hemochorial maternal-fetal interface. Placenta 2012; 33:233–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Walentin K, Hinze C, Schmidt-Ott KM. The basal chorionic trophoblast cell layer: an emerging coordinator of placenta development. Bioessays 2016; 38:254–265. [DOI] [PubMed] [Google Scholar]
  • 18. Red-Horse K, Zhou Y, Genbacev O, Prakobphol A, Foulk R, McMaster M, Fisher SJ. Trophoblast differentiation during embryo implantation and formation of the maternal-fetal interface. J Clin Invest 2004; 114:744–754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Huppertz B. The anatomy of the normal placenta. J Clin Pathol 2008; 61:1296–1302. [DOI] [PubMed] [Google Scholar]
  • 20. Velicky P, Knöfler M, Pollheimer J. Function and control of human invasive trophoblast subtypes: intrinsic vs. maternal control. Cell Adh Migr 2016; 10:154–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. James JL, Carter AM, Chamley LW. Human placentation from nidation to 5 weeks of gestation. Part I: What do we know about formative placental development following implantation? Placenta 2012; 33:327–334. [DOI] [PubMed] [Google Scholar]
  • 22. Enders AC, Schlafke S. Cytological aspects of trophoblast-uterine interaction in early implantation. Am J Anat 1969; 125:1–29. [DOI] [PubMed] [Google Scholar]
  • 23. Schlafke S, Enders AC. Cellular basis of interaction between trophoblast and uterus at implantation. Biol Reprod 1975; 12:41–65. [DOI] [PubMed] [Google Scholar]
  • 24. Georgiades P, Ferguson-Smith AC, Burton GJ. Comparative developmental anatomy of the murine and human definitive placentae. Placenta 2002; 23:3–19. [DOI] [PubMed] [Google Scholar]
  • 25. Soares MJ, Chakraborty D, Kubota K, Renaud SJ, Rumi MA. Adaptive mechanisms controlling uterine spiral artery remodeling during the establishment of pregnancy. Int J Dev Biol 2014; 58:247–259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Pijnenborg R, Robertson WB, Brosens I, Dixon G. Review article: Trophoblast invasion and the establishment of haemochorial placentation in man and laboratory animals. Placenta 1981; 2:71–91. [DOI] [PubMed] [Google Scholar]
  • 27. Pijnenborg R, Vercruysse L, Hanssens M. The uterine spiral arteries in human pregnancy: facts and controversies. Placenta 2006; 27:939–958. [DOI] [PubMed] [Google Scholar]
  • 28. Kaufmann P, Black S, Huppertz B. Endovascular trophoblast invasion: implications for the pathogenesis of intrauterine growth retardation and preeclampsia. Biol Reprod 2003; 69:1–7. [DOI] [PubMed] [Google Scholar]
  • 29. Damsky CH, Librach C, Lim KH, Fitzgerald ML, McMaster MT, Janatpour M, Zhou Y, Logan SK, Fisher SJ. Integrin switching regulates normal trophoblast invasion. Development 1994; 120:3657–3666. [DOI] [PubMed] [Google Scholar]
  • 30. Zhou Y, Fisher SJ, Janatpour M, Genbacev O, Dejana E, Wheelock M, Damsky CH. Human cytotrophoblasts adopt a vascular phenotype as they differentiate. A strategy for successful endovascular invasion? J Clin Invest 1997; 99:2139–2151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Damsky CH, Fisher SJ. Trophoblast pseudo-vasculogenesis: faking it with endothelial adhesion receptors. Curr Opin Cell Biol 1998; 10:660–666. [DOI] [PubMed] [Google Scholar]
  • 32. Harris LK. Review: Trophoblast-vascular cell interactions in early pregnancy: how to remodel a vessel. Placenta 2010; 31(Suppl):S93–S98. [DOI] [PubMed] [Google Scholar]
  • 33. Ain R, Canham LN, Soares MJ. Gestation stage-dependent intrauterine trophoblast cell invasion in the rat and mouse: novel endocrine phenotype and regulation. Dev Biol 2003; 260:176–190. [DOI] [PubMed] [Google Scholar]
  • 34. Pijnenborg R, Vercruysse L. Animal models of deep trophoblast invasion. In: Pijnenborg, R, Brosens, I, Romero, R (eds.), Placental Bed Disorders: Basic Science and its Translation to Obstetrics. Cambridge: Cambridge University Press; 2010:127–139. [Google Scholar]
  • 35. Soares MJ, Chapman BM, Rasmussen CA, Dai G, Kamei T, Orwig KE. Differentiation of trophoblast endocrine cells. Placenta 1996; 17:277–289. [DOI] [PubMed] [Google Scholar]
  • 36. Soares MJ. The prolactin and growth hormone families: pregnancy-specific hormones/cytokines at the maternal-fetal interface. Reprod Biol Endocrinol 2004; 2:51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Costa MA. The endocrine function of human placenta: an overview. Reprod Biomed Online 2016; 32:14–43. [DOI] [PubMed] [Google Scholar]
  • 38. Carter AM. Evolution of placental function in mammals: the molecular basis of gas and nutrient transfer, hormone secretion, and immune responses. Physiol Rev 2012; 92:1543–1576. [DOI] [PubMed] [Google Scholar]
  • 39. Dilworth MR, Sibley CP. Review: Transport across the placenta of mice and women. Placenta 2013; 34(Suppl):S34–S39. [DOI] [PubMed] [Google Scholar]
  • 40. Díaz P, Powell TL, Jansson T. The role of placental nutrient sensing in maternal-fetal resource allocation. Biol Reprod 2014; 91:82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Erlebacher A. Immunology of the maternal-fetal interface. Annu Rev Immunol 2013; 31:387–411. [DOI] [PubMed] [Google Scholar]
  • 42. Hakkola J, Pelkonen O, Pasanen M, Raunio H. Xenobiotic-metabolizing cytochrome P450 enzymes in the human feto-placental unit: role in intrauterine toxicity. Crit Rev Toxicol 1998; 28:35–72. [DOI] [PubMed] [Google Scholar]
  • 43. Joshi AA, Vaidya SS, St-Pierre MV, Mikheev AM, Desino KE, Nyandege AN, Audus KL, Unadkat JD, Gerk PM. Placental ABC transporters: biological impact and pharmaceutical significance. Pharm Res 2016; 33:2847–2878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Arora N, Sadovsky Y, Dermody TS, Coyne CB. Microbial vertical transmission during human pregnancy. Cell Host Microbe 2017; 21:561–567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. van Dijk M, Oudejans C (Epi)genetics of pregnancy-associated diseases. Front Genet 2013; 4:180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Pisarska MD, Akhlaghpour M, Lee B, Barlow GM, Xu N, Wang ET, Mackey AJ, Farber CR, Rich SS, Rotter JI, Chen YI, Goodarzi MO et al. Optimization of techniques for multiple platform testing in small, precious samples such as human chorionic villus sampling. Prenat Diagn 2016; 36:1061–1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Imudia AN, Suzuki Y, Kilburn BA, Yelian FD, Diamond MP, Romero R, Armant DR. Retrieval of trophoblast cells from the cervical canal for prediction of abnormal pregnancy: a pilot study. Hum Reprod 2009; 24:2086–2092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Drewlo S, Armant DR. Quo vadis, trophoblast? Exploring the new ways of an old cell lineage. Placenta 2017; 60(Suppl 1):S27–S31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Nelson AC, Mould AW, Bikoff EK, Robertson EJ. Single-cell RNA-seq reveals cell type-specific transcriptional signatures at the maternal–foetal interface during pregnancy. Nat Comms 2016; 7:11414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Gormley M, Ona K, Kapidzic M, Garrido-Gomez T, Zdravkovic T, Fisher SJ. Preeclampsia: novel insights from global RNA profiling of trophoblast subpopulations. Am J Obstet Gynecol 2017; 217:200.e1–200.e17. [DOI] [PubMed] [Google Scholar]
  • 51. Pavličev M, Wagner GP, Chavan AR, Owens K, Maziarz J, Dunn-Fletcher C, Kallapur SG, Muglia L, Jones H. Single-cell transcriptomics of the human placenta: inferring the cell communication network of the maternal-fetal interface. Genome Res 2017; 27:349–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Pattillo RA, Gey GO. The establishment of a cell line of human hormone-synthesizing trophoblastic cells in vitro. Cancer Res 1968; 28:1231–1236. [PubMed] [Google Scholar]
  • 53. Kohler PO, Bridson WE. Isolation of hormone-producing clonal lines of human choriocarcinoma. J Clin Endocrinol Metab 1971; 32:683–687. [DOI] [PubMed] [Google Scholar]
  • 54. Pattillo RA, Ruckert A, Hussa R, Bernstein R, Delfs E. The Jar cell line – continuous human multihormone production and controls. In Vitro 1971; 6:398. [Google Scholar]
  • 55. Ringler GE. In vitro systems for the study of human placental endocrine function. Endocr Rev 1990; 11:105–123. [DOI] [PubMed] [Google Scholar]
  • 56. Graham CH, Hawley TS, Hawley RG, MacDougall JR, Kerbel RS, Khoo N, Lala PK. Establishment and characterization of first trimester human trophoblast cells with extended lifespan. Exp Cell Res 1993; 206:204–211. [DOI] [PubMed] [Google Scholar]
  • 57. Choy MY, Manyonda IT. The phagocytic activity of human first trimester extravillous trophoblast. Hum Reprod 1998; 13:2941–2949. [DOI] [PubMed] [Google Scholar]
  • 58. Straszewski-Chavez SL, Abrahams VM, Alvero AB, Aldo PB, Ma Y, Guller S, Romero R, Mor G. The isolation and characterization of a novel telomerase immortalized first trimester trophoblast cell line, Swan 7. Placenta 2009; 30:939–948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Sullivan MH. Endocrine cell lines from the placenta. Mol Cell Endocrinol 2004; 22:103–119. [DOI] [PubMed] [Google Scholar]
  • 60. Whitley GS. Production of human trophoblast cell lines. Methods Mol Med 2006; 121:219–228. [DOI] [PubMed] [Google Scholar]
  • 61. Lee CQ, Gardner L, Turco M, Zhao N, Murray MJ, Coleman N, Rossant J, Hemberger M, Moffett A. What is trophoblast? A combination of criteria define human first-trimester trophoblast. Stem Cell Rep 2016; 6:257–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Wice B, Menton D, Geuze H, Schwartz AL. Modulators of cyclic AMP metabolism induce syncytiotrophoblast formation in vitro. Exp Cell Res 1990; 186:306–316. [DOI] [PubMed] [Google Scholar]
  • 63. Matsuura K, Jigami T, Taniue K, Morishita Y, Adachi S, Senda T, Nonaka A, Aburatani H, Nakamura T, Akiyama T. Identification of a link between Wnt/β-catenin signalling and the cell fusion pathway. Nat Commun 2011; 2:548. [DOI] [PubMed] [Google Scholar]
  • 64. Lu J, Zhang S, Nakano H, Simmons DG, Wang S, Kong S, Wang Q, Shen L, Tu Z, Wang W, Wang B, Wang H et al. A positive feedback loop involving Gcm1 and Fzd5 directs chorionic branching morphogenesis in the placenta. PLoS Biol 2013; 11:e1001536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Kliman HJ, Nestler JE, Sermasi E, Sanger JM, Purification, characterization, and in vitro differentiation of cytotrophoblasts from human term placentae. Endocrinology 1986; 118:1567–1582. [DOI] [PubMed] [Google Scholar]
  • 66. Thordarson G, Folger P, Talamantes F. Development of a placental cell culture system for studying the control of mouse placental lactogen II secretion. Placenta 1987; 8:573–585. [DOI] [PubMed] [Google Scholar]
  • 67. Lu XJ, Deb S, Soares MJ. Spontaneous differentiation of trophoblast cells along the spongiotrophoblast cell pathway: expression of members of the placental prolactin gene family and modulation by retinoic acid. Dev Biol 1994; 163:86–97. [DOI] [PubMed] [Google Scholar]
  • 68. Aplin JD. In vitro analysis of trophoblast invasion. Methods Mol Med 2006; 122:45–57. [DOI] [PubMed] [Google Scholar]
  • 69. Petroff MG, Phillips TA, Ka H, Pace JL, Hunt JS. Isolation and culture of term human trophoblast cells. Methods Mol Med 2006; 121:203–217. [DOI] [PubMed] [Google Scholar]
  • 70. Orendi K, Kivity V, Sammar M, Grimpel Y, Gonen R, Meiri H, Lubzens E, Huppertz B. Placental and trophoblastic in vitro models to study preventive and therapeutic agents for preeclampsia. Placenta 2011; 32:S49–S54. [DOI] [PubMed] [Google Scholar]
  • 71. Schneider H. The role of the placenta in nutrition of the human fetus. Am J Obstet Gynecol 1991; 164:967–973. [DOI] [PubMed] [Google Scholar]
  • 72. Teshima S, Shimosato Y, Koide T, Kuroki M, Kikuchi Y, Aizawa M. Transplantable choriocarcinoma of rats induced by fetectomy and its biological activities. Gan 1983; 74:205–212. [PubMed] [Google Scholar]
  • 73. Verstuyf A, Sobis H, Goebels J, Fonteyn E, Cassiman JJ, Vandeputte M. Establishment and characterization of a continuousin vitro line from a rat choriocarcinoma. Int J Cancer 1990; 45:752–756. [DOI] [PubMed] [Google Scholar]
  • 74. Faria TN, Soares MJ. Trophoblast cell differentiation: establishment, characterization, and modulation of a rat trophoblast cell line expressing members of the placental prolactin family. Endocrinology 1991; 129:2895–2906. [DOI] [PubMed] [Google Scholar]
  • 75. Hamlin GP, Lu XJ, Roby KF, Soares MJ. Recapitulation of the pathway for trophoblast giant cell differentiation in vitro: stage-specific expression of members of the prolactin gene family. Endocrinology 1994; 134:2390–2396. [DOI] [PubMed] [Google Scholar]
  • 76. Kamei T, Hamlin GP, Chapman BM, Burkhardt AL, Bolen JB, Soares MJ. Signaling pathways controlling trophoblast cell differentiation: src family protein tyrosine kinases in the rat. Biol Reprod 1997; 57:1302–1311. [DOI] [PubMed] [Google Scholar]
  • 77. Kamei T, Jones SR, Chapman BM, MCGonigle KL, Dai G, Soares MJ. The phosphatidylinositol 3-kinase/Akt signaling pathway modulates the endocrine differentiation of trophoblast cells. Mol Endocrinol 2002; 16:1469–1481. [DOI] [PubMed] [Google Scholar]
  • 78. Kent LN, Konno T, Soares MJ. Phosphatidylinositol 3 kinase modulation of trophoblast cell differentiation. BMC Dev Biol 2010; 10:97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Kent LN, Rumi MA, Kubota K, Lee DS, Soares MJ. FOSL1 is integral to establishing the maternal-fetal interface. Mol Cell Biol 2011; 31:4801–4813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Kubota K, Kent LN, Rumi MA, Roby KF, Soares MJ. Dynamic regulation of AP-1 transcriptional complexes directs trophoblast differentiation. Mol Cell Biol 2015; 35:3163–3177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Sahgal N, Canham LN, Canham B, Soares MJ. Rcho-1 trophoblast stem cells: a model system for studying trophoblast cell differentiation. Methods Mol Med 2006; 121:159–178. [PubMed] [Google Scholar]
  • 82. Tanaka S, Kunath T, Hadjantonakis AK, Nagy A, Rossant J. Promotion of trophoblast stem cell proliferation by FGF4. Science 1998; 282:2072–2075. [DOI] [PubMed] [Google Scholar]
  • 83. Chung Y, Klimanskaya I, Becker S, Marh J, Lu SJ, Johnson J, Meisner L, Lanza R. Embryonic and extraembryonic stem cell lines derived from single mouse blastomeres. Nature 2006; 439:216–219. [DOI] [PubMed] [Google Scholar]
  • 84. Erlebacher A, Price KA, Glimcher LH. Maintenance of mouse trophoblast stem cell proliferation by TGF-beta/activin. Dev Biol 2004; 275:158–169. [DOI] [PubMed] [Google Scholar]
  • 85. Guzman-Ayala M, Ben-Haim N, Beck S, Constam DB. Nodal protein processing and fibroblast growth factor 4 synergize to maintain a trophoblast stem cell microenvironment. Proc Natl Acad Sci USA 2004; 101:15656–15660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Kubaczka C, Senner C, Araúzo-Bravo MJ, Sharma N, Kuckenberg P, Becker A, Zimmer A, Brüstle O, Peitz M, Hemberger M, Schorle H. Derivation and maintenance of murine trophoblast stem cells under defined conditions. Stem Cell Rep 2014; 2:232–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Ohinata Y, Tsukiyama T. Establishment of trophoblast stem cells under defined culture conditions in mice. PLoS One 2014; 9:e107308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Asanoma K, Rumi MA, Kent LN, Chakraborty D, Renaud SJ, Wake N, Lee DS, Kubota K, Soares MJ. FGF4-dependent stem cells derived from rat blastocysts differentiate along the trophoblast lineage. Dev Biol 2011; 351:110–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Kibschull M, Winterhager E. Connexins and trophoblast cell lineage development. Methods Mol Med 2006; 121:149–158. [DOI] [PubMed] [Google Scholar]
  • 90. Konno T, Rempel LA, Rumi MA, Graham AR, Asanoma K, Renaud SJ, Soares MJ. Chromosome-substituted rat strains provide insights into the genetics of placentation. Physiol Genomics 2011; 43:930–941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Chakraborty D, Cui W, Rosario GX, Scott RL, Dhakal P, Renaud SJ, Tachibana M, Rumi MA, Mason CW, Krieg AJ, Soares MJ. HIF-KDM3A-MMP12 regulatory circuit ensures trophoblast plasticity and placental adaptations to hypoxia. Proc Natl Acad Sci USA 2016; 113:E7212–E7221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. Embryonic stem cell lines derived from human blastocysts. Science 1998; 282:1145–1147. [DOI] [PubMed] [Google Scholar]
  • 93. Ezashi T, Telugu BP, Roberts RM. Model systems for studying trophoblast differentiation from human pluripotent stem cells. Cell Tissue Res 2012; 349:809–824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Xu RH, Chen X, Li DS, Li R, Addicks GC, Glennon C, Zwaka TP, Thomson JA. BMP4 initiates human embryonic stem cell differentiation to trophoblast. Nat Biotechnol 2002; 20:1261–1264. [DOI] [PubMed] [Google Scholar]
  • 95. Yabe S, Alexenko AP, Amita M, Yang Y, Schust DJ, Sadovsky Y, Ezashi T, Roberts RM. Comparison of syncytiotrophoblast generated from human embryonic stem cells and from term placentas. Proc Natl Acad Sci USA 2016; 113:E2598–E2607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Roberts RM, Loh KM, Amita M, Bernardo AS, Adachi K, Alexenko AP, Schust DJ, Schulz LC, Telugu BP, Ezashi T, Pedersen RA. Differentiation of trophoblast cells from human embryonic stem cells: to be or not to be? Reproduction 2014; 147:D1–D12. [DOI] [PubMed] [Google Scholar]
  • 97. Okae H, Toh H, Sato T, Hiura H, Takahashi S, Shirane K, Kabayama Y, Suyama M, Sasaki H, Arima T. Derivation of human trophoblast stem cells. Cell Stem Cell 2018; 22:50.e6–63.e6. [DOI] [PubMed] [Google Scholar]
  • 98. Yang Y, Liu B, Xu J, Wang J, Wu J, Shi C, Xu Y, Dong J, Wang C, Lai W, Zhu J, Xiong L et al. Derivation of Pluripotent Stem Cells with In Vivo Embryonic and Extraembryonic Potency. Cell 2017; 169:243–257.e25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Yang J, Ryan DJ, Wang W, Tsang JC, Lan G, Masaki H, Gao X, Antunes L, Yu Y, Zhu Z, Wang J, Kolodziejczyk AA et al. Establishment of mouse expanded potential stem cells. Nature 2017; 550:393–397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Zdravkovic T, Nazor KL, Larocque N, Gormley M, Donne M, Hunkapillar N, Giritharan G, Bernstein HS, Wei G, Hebrok M, Zeng X, Genbacev O et al. Human stem cells from single blastomeres reveal pathways of embryonic or trophoblast fate specification. Development 2015; 142:4010–4025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Benchetrit H, Herman S, van Wietmarschen N, Wu T, Makedonski K, Maoz N, Yom Tov N, Stave D, Lasry R, Zayat V, Xiao A, Lansdorp PM et al. Extensive nuclear reprogramming underlies lineage conversion into functional trophoblast stem-like cells. Cell Stem Cell 2015; 17:543–556. [DOI] [PubMed] [Google Scholar]
  • 102. Kubaczka C, Senner CE, Cierlitza M, Araúzo-Bravo MJ, Kuckenberg P, Peitz M, Hemberger M, Schorle H. Direct induction of trophoblast stem cells from murine fibroblasts. Cell Stem Cell 2015; 17:557–568. [DOI] [PubMed] [Google Scholar]
  • 103. Harrison SE, Sozen B, Christodoulou N, Kyprianou C, Zernicka-Goetz M. Assembly of embryonic and extraembryonic stem cells to mimic embryogenesis in vitro. Science 2017; 356:6334. [DOI] [PubMed] [Google Scholar]
  • 104. Zhu D, Gong X, Miao L, Fang J, Zhang J. Efficient induction of syncytiotrophoblast layer II cells from trophoblast stem cells by canonical Wnt signaling activation. Stem Cell Rep 2017; 9:2034–2049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. VandeVoort CA, Thirkill TL, Douglas GC. Blastocyst-derived trophoblast stem cells from the rhesus monkey. Stem Cells Dev 2007; 16:779–788. [DOI] [PubMed] [Google Scholar]
  • 106. Grigor’eva EV, Shevchenko AI, Mazurok NA, Elisaphenko EA, Zhelezova AI, Shilov AG, Dyban PA, Dyban AP, Noniashvili EM, Slobodyanyuk SY, Nesterova TB, Brockdorff N et al. FGF4 independent derivation of trophoblast stem cells from the common vole. PLoS One 2009; 4:e7161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Tan T, Tang X, Zhang J, Niu Y, Chen H, Li B, Wei Q, Ji W. Generation of trophoblast stem cells from rabbit embryonic stem cells with BMP4. PLoS One 2011; 6:e17124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Huang X, Han X, Uyunbilig B, Zhang M, Duo S, Zuo Y, Zhao Y, Yun T, Tai D, Wang C, Li J, Li X et al. Establishment of bovine trophoblast stem-like cells from in vitro - produced blastocyst-stage embryos using two inhibitors. Stem Cells Dev 2014; 23:1501–1514. [DOI] [PubMed] [Google Scholar]
  • 109. Hou D, Su M, Li X, Li Z, Yun T, Zhao Y, Zhang M, Zhao L, Li R, Yu H, Li X. The efficient derivation of trophoblast cells from porcine in vitro fertilized and parthenogenetic blastocysts and culture with ROCK inhibitor Y-27632. PLoS One 2015; 10:e0142442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Hou Z, Romero R, Uddin M, Than NG, Wildman DE. Adaptive history of single copy genes highly expressed in the term human placenta. Genomics 2009; 93:33–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Wildman DE. Review: Toward an integrated evolutionary understanding of the mammalian placenta. Placenta 2011; 32(Suppl 2):S142–S145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Cox B, Kotlyar M, Evangelou AI, Ignatchenko V, Ignatchenko A, Whiteley K, Jurisica I, Adamson SL, Rossant J, Kislinger T. Comparative systems biology of human and mouse as a tool to guide the modeling of human placental pathology. Mol Syst Biol 2009; 5:279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Armstrong DL, McGowen MR, Weckle A, Pantham P, Caravas J, Agnew D, Benirschke K, Savage-Rumbaugh S, Nevo E, Kim CJ, Wagner GP, Romero R et al. The core transcriptome of mammalian placentas and the divergence of expression with placental shape. Placenta 2017; 57:71–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Rossant J, Cross JC. Placental development: lessons from mouse mutants. Nat Rev Genet 2001; 2:538–548. [DOI] [PubMed] [Google Scholar]
  • 115. Carter AM. Animal models of human placentation – a review. Placenta 2007; 28(Suppl A):S41–S47. [DOI] [PubMed] [Google Scholar]
  • 116. Mess A, Zaki N, Kadyrov M, Korr H, Kaufmann P. Caviomorph placentation as a model for trophoblast invasion. Placenta 2007; 28:1234–1238. [DOI] [PubMed] [Google Scholar]
  • 117. Guillemot F, Nagy A, Auerbach A, Rossant J, Joyner AL. Essential role of Mash-2 in extraembryonic development. Nature 1994; 371:333–336. [DOI] [PubMed] [Google Scholar]
  • 118. Chawengsaksophak K, de Graaff W, Rossant J, Deschamps J, Beck F. Cdx2 is essential for axial elongation in mouse development. Proc Natl Acad Sci USA 2004; 101:7641–7645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Chawengsaksophak K, James R, Hammond VE, Köntgen F, Beck F. Homeosis and intestinal tumours in Cdx2 mutant mice. Nature 1997; 386:84–87. [DOI] [PubMed] [Google Scholar]
  • 120. Luo J, Sladek R, Bader JA, Matthyssen A, Rossant J, Giguère V. Placental abnormalities in mouse embryos lacking the orphan nuclear receptor ERR-?. Nature 1997; 388:778–782. [DOI] [PubMed] [Google Scholar]
  • 121. Ma GT, Roth ME, Groskopf JC, Tsai FY, Orkin SH, Grosveld F, Engel JD, Linzer DI. GATA-2 and GATA-3 regulate trophoblast-specific gene expression in vivo. Development 1997; 124:907–914. [DOI] [PubMed] [Google Scholar]
  • 122. Riley P, Anson-Cartwright L, Cross JC. The Hand1 bHLH transcription factor is essential for placentation and cardiac morphogenesis. Nat Genet 1998; 18:271–275. [DOI] [PubMed] [Google Scholar]
  • 123. Yamamoto H, Flannery ML, Kupriyanov S, Pearce J, McKercher SR, Henkel GW, Maki RA, Werb Z, Oshima RG. Defective trophoblast function in mice with a targeted mutation of Ets2. Genes Dev 1998; 12:1315–1326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Barak Y, Nelson MC, Ong ES, Jones YZ, Ruiz-Lozano P, Chien KR, Koder A, Evans RM. PPAR? Is required for placental, cardiac, and adipose tissue development. Mol Cell 1999; 4:585–595. [DOI] [PubMed] [Google Scholar]
  • 125. Morasso MI, Grinberg A, Robinson G, Sargent TD, Mahon KA. Placental failure in mice lacking the homeobox gene Dlx3. Proc Natl Acad Sci USA 1999; 96:162–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Anson-Cartwright L, Dawson K, Holmyard D, Fisher SJ, Lazzarini RA, Cross JC. The glial cells missing-1 protein is essential for branching morphogenesis in the chorioallantoic placenta. Nat Genet 2000; 25:311–314. [DOI] [PubMed] [Google Scholar]
  • 127. Russ AP, Wattler S, Colledge WH, Aparicio SA, Carlton MB, Pearce JJ, Barton SC, Surani MA, Ryan K, Nehls MC, Wilson V, Evans MJ. Eomesodermin is required for mouse trophoblast development and mesoderm formation. Nature 2000; 404:95–99. [DOI] [PubMed] [Google Scholar]
  • 128. Auman HJ, Nottoli T, Lakiza O, Winger Q, Donaldson S, Williams T. Transcription factor AP-2 gamma is essential in the extra-embryonic lineages for early postimplantation development. Development 2002; 129:2733–2747. [DOI] [PubMed] [Google Scholar]
  • 129. Werling U, Schorle H. Transcription factor gene AP-2 gamma essential for early murine development. Mol Cell Biol 2002; 22:3149–3156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Donnison M, Beaton A, Davey HW, Broadhurst R, L’Huillier P, Pfeffer PL. Loss of the extraembryonic ectoderm in Elf5 mutants leads to defects in embryonic patterning. Development 2005; 132:2299–2308. [DOI] [PubMed] [Google Scholar]
  • 131. Yagi R, Kohn MJ, Karavanova I, Kaneko KJ, Vullhorst D, DePamphilis ML, Buonanno A. Transcription factor TEAD4 specifies the trophectoderm lineage at the beginning of mammalian development. Development 2007; 134:3827–3836. [DOI] [PubMed] [Google Scholar]
  • 132. Nishioka N, Yamamoto S, Kiyonari H, Sato H, Sawada A, Ota M, Nakao K, Sasaki H. Tead4 is required for specification of trophectoderm in pre-implantation mouse embryos. Mech Dev 2008; 125:270–283. [DOI] [PubMed] [Google Scholar]
  • 133. Alders M, Hodges M, Hadjantonakis AK, Postmus J, van Wijk I, Bliek J, de Meulemeester M, Westerveld A, Guillemot F, Oudejans C, Little P, Mannens M. The Human Achaete-Scute Homologue 2 (ASCL2, HASH2) maps to chromosome 11p15.5, close to IGF2 and is expressed in extravillus trophoblasts. Hum Mol Genet 1997; 6:859–867. [DOI] [PubMed] [Google Scholar]
  • 134. Schaiff WT, Carlson MG, Smith SD, Levy R, Nelson DM, Sadovsky Y. Peroxisome proliferator-activated receptor-gamma modulates differentiation of human trophoblast in a ligand-specific manner. J Clin Endocrinol Metab 2000; 85:3874–3881. [DOI] [PubMed] [Google Scholar]
  • 135. Loregger T, Pollheimer J, Knöfler M. Regulatory transcription factors controlling function and differentiation of human trophoblast—a review. Placenta 2003; 24:S104–S110. [DOI] [PubMed] [Google Scholar]
  • 136. Adjaye J, Huntriss J, Herwig R, BenKahla A, Brink TC, Wierling C, Hultschig C, Groth D, Yaspo ML, Picton HM, Gosden RG, Lehrach H. Primary differentiation in the human blastocyst: comparative molecular portraits of inner cell mass and trophectoderm cells. Stem Cells 2005; 23:1514–1525. [DOI] [PubMed] [Google Scholar]
  • 137. Baczyk D, Drewlo S, Proctor L, Dunk C, Lye S, Kingdom J. Glial cell missing-1 transcription factor is required for the differentiation of the human trophoblast. Cell Death Differ 2009; 16:719–727. [DOI] [PubMed] [Google Scholar]
  • 138. Staun-Ram E, Goldman S, Shalev E. Ets-2 and p53 mediate cAMP-induced MMP-2 expression, activity and trophoblast invasion. Reprod Biol Endocrinol 2009; 7:135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Hemberger M, Udayashankar R, Tesar P, Moore H, Burton GJ. ELF5-enforced transcriptional networks define an epigenetically regulated trophoblast stem cell compartment in the human placenta. Hum Mol Genet 2010; 19:2456–2467. [DOI] [PubMed] [Google Scholar]
  • 140. Chui A, Evseenko DA, Brennecke SP, Keelan JA, Kalionis B, Murthi P. Homeobox gene Distal-less 3 (DLX3) is a regulator of villous cytotrophoblast differentiation. Placenta 2011; 32:745–751. [DOI] [PubMed] [Google Scholar]
  • 141. Kumar P, Mendelson CR. Estrogen-related receptor gamma (ERRgamma) mediates oxygen-dependent induction of aromatase (CYP19) gene expression during human trophoblast differentiation. Mol Endocrinol 2011; 25:1513–1526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Niakan KK, Eggan K. Analysis of human embryos from zygote to blastocyst reveals distinct gene expression patterns relative to the mouse. Dev Biol 2013; 375:54–64. [DOI] [PubMed] [Google Scholar]
  • 143. Haider S, Meinhardt G, Saleh L, Fiala C, Pollheimer J, Knöfler M. Notch1 controls development of the extravillous trophoblast lineage in the human placenta. Proc Natl Acad Sci USA 2016; 113:E7710–E7719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Lee B, Kroener LL, Xu N, Wang ET, Banks A, Williams J 3rd, Goodarzi MO, Chen YI, Tang J, Wang Y, Gangalapudi V, Pisarska MD. Function and hormonal regulation of GATA3 in human first trimester placentation. Biol Reprod 2016; 95:113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Krendl C, Shaposhnikov D, Rishko V, Ori C, Ziegenhain C, Sass S, Simon L, Müller NS, Straub T, Brooks KE, Chavez SL, Enard W et al. GATA2/3-TFAP2A/C transcription factor network couples human pluripotent stem cell differentiation to trophectoderm with repression of pluripotency. Proc Natl Acad Sci USA 2017; 114:E9579–E9588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Wenzel PL, Leone G. Expression of Cre recombinase in early diploid trophoblast cells of the mouse placenta. Genesis 2007; 45:129–134. [DOI] [PubMed] [Google Scholar]
  • 147. Nadeau V, Guillemette S, Bélanger LF, Jacob O, Roy S, Charron J. Map2k1 and Map2k2 genes contribute to the normal development of syncytiotrophoblasts during placentation. Development 2009; 136:1363–1374. [DOI] [PubMed] [Google Scholar]
  • 148. Hu D, Cross JC. Ablation of Tpbpa-positive trophoblast precursors leads to defects in maternal spiral artery remodeling in the mouse placenta. Dev Biol 2011; 358:231–239. [DOI] [PubMed] [Google Scholar]
  • 149. Mould A, Morgan MA, Li L, Bikoff EK, Robertson EJ. Blimp1/Prdm1 governs terminal differentiation of endovascular trophoblast giant cells and defines multipotent progenitors in the developing placenta. Genes Dev 2012; 26:2063–2074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Ouseph MM, Li J, Chen HZ, Pécot T, Wenzel P, Thompson JC, Comstock G, Chokshi V, Byrne M, Forde B, Chong JL, Huang K et al. Atypical E2F repressors and activators coordinate placental development. Dev Cell 2012; 22:849–862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Zhou CC, Chang J, Mi T, Abbasi S, Gu D, Huang L, Zhang W, Kellems RE, Schwartz RJ, Xia Y. Targeted expression of Cre recombinase provokes placental-specific DNA recombination in transgenic mice. PLoS One 2012; 7:e29236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Crish J, Conti MA, Sakai T, Adelstein RS, Egelhoff TT. Keratin 5-Cre-driven excision of nonmuscle myosin IIA in early embryo trophectoderm leads to placenta defects and embryonic lethality. Dev Biol 2013; 382:136–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Al-Soudy AS, Nakanishi T, Mizuno S, Hasegawa Y, Shawki HH, Katoh MC, Basha WA, Ibrahim AE, El-Shemy HA, Iseki H, Yoshiki A, Hiromori Y et al. Germline recombination in a novel Cre transgenic line, Prl3b1 -Cre mouse. Genesis 2016; 54:389–397. [DOI] [PubMed] [Google Scholar]
  • 154. Nadeau V, Charron J. Essential role of the ERK/MAPK pathway in blood-placental barrier formation. Development 2014; 141:2825–2837. [DOI] [PubMed] [Google Scholar]
  • 155. Outhwaite JE, McGuire V, Simmons DG. Genetic ablation of placental sinusoidal trophoblast giant cells causes fetal growth restriction and embryonic lethality. Placenta 2015; 36:951–955. [DOI] [PubMed] [Google Scholar]
  • 156. Moreau JL, Artap ST, Shi H, Chapman G, Leone G, Sparrow DB, Dunwoodie SL. Cited2 is required in trophoblasts for correct placental capillary patterning. Dev Biol 2014; 392:62–79. [DOI] [PubMed] [Google Scholar]
  • 157. Dow LE, Lowe SW. Life in the fast lane: mammalian disease models in the genomics era. Cell 2012; 148:1099–1109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Dow LE. Modeling disease in vivo with CRISPR/Cas9. Trends Mol Med 2015; 21:609–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Georgiades P, Cox B, Gertsenstein M, Chawengsaksophak K, Rossant J. Trophoblast-specific gene manipulation using lentivirus-based vectors. Biotech 2007; 42:317–325. [DOI] [PubMed] [Google Scholar]
  • 160. Malashicheva A, Kanzler B, Tolkunova E, Trono D, Tomilin A. Lentivirus as a tool for lineage-specific gene manipulations. Genesis 2007; 45:456–459. [DOI] [PubMed] [Google Scholar]
  • 161. Okada Y, Ueshin Y, Isotani A, Saito-Fujita T, Nakashima H, Kimura K, Mizoguchi A, Oh-Hora M, Mori Y, Ogata M, Oshima RG, Okabe M et al. Complementation of placental defects and embryonic lethality by trophoblast-specific lentiviral gene transfer. Nat Biotechnol 2007; 25:233–237. [DOI] [PubMed] [Google Scholar]
  • 162. Tobita T, Kiyozumi D, Ikawa M. Placenta-specific gene manipulation using lentiviral vector and its application. Placenta 2017; 59(Suppl 1):S37–S43. [DOI] [PubMed] [Google Scholar]
  • 163. Lee DS, Rumi MA, Konno T, Soares MJ. In vivo genetic manipulation of the rat trophoblast cell lineage using lentiviral vector delivery. Genesis 2009; 47:433–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Purcell SH, Cantlon JD, Wright CD, Henkes LE, Seidel GE Jr, Anthony RV. The involvement of proline-rich 15 in early conceptus development in sheep. Biol Reprod 2009; 81:1112–1121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Bou G, Liu S, Sun M, Zhu J, Xue B, Guo J, Zhao Y, Qu B, Weng X, Wei Y, Lei L, Liu Z. CDX2 is essential for cell proliferation and polarity in porcine blastocysts. Development 2017; 144:1296–1306. [DOI] [PubMed] [Google Scholar]
  • 166. Skoda G, Hoffmann OI, Gócza E, Bodrogi L, Kerekes A, Bösze Z, Hiripi L. Placenta-specific gene manipulation in rabbits. J Biotechnol 2017; 259:86–90. [DOI] [PubMed] [Google Scholar]
  • 167. Brosens IA, Robertson WB, Dixon HG. The role of the spiral arteries in the pathogenesis of preeclampsia. Obstet Gynecol Annu 1972; 1:177–191. [PubMed] [Google Scholar]
  • 168. Fisher SJ. The placental problem: linking abnormal cytotrophoblast differentiation to the maternal symptoms of preeclampsia. Reprod Biol Endocrinol 2004; 2:53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Brosens I, Pijnenborg R, Vercruysse L, Romero R. The “Great Obstetrical Syndromes” are associated with disorders of deep placentation. Am J Obstet Gynecol 2011; 204:193–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Zhou Y, Damsky CH, Chiu K, Roberts JM, Fisher SJ. Preeclampsia is associated with abnormal expression of adhesion molecules by invasive cytotrophoblasts. J Clin Invest 1993; 91:950–960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Zhou Y, Damsky CH, Fisher SJ. Preeclampsia is associated with failure of human cytotrophoblasts to mimic a vascular adhesion phenotype. One cause of defective endovascular invasion in this syndrome? J Clin Invest 1997; 99:2152–2164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Rosario GX, Ain R, Konno T, Soares MJ. Intrauterine fate of invasive trophoblast cells? Placenta 2009; 30:457–463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Fidler IJ. Timeline: The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis revisited. Nat Rev Cancer 2003; 3:453–458. [DOI] [PubMed] [Google Scholar]
  • 174. Cha J, Sun X, Dey SK. Mechanisms of implantation: strategies for successful pregnancy. Nat Med 2012; 18:1754–1767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Zhang J, Chen Z, Smith GN, Croy BA. Natural killer cell-triggered vascular transformation: maternal care before birth? Cell Mol Immunol 2011; 8:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Zhang J, Dunk C, Croy AB, Lye SJ. To serve and to protect: the role of decidual innate immune cells on human pregnancy. Cell Tissue Res 2016; 363:249–265. [DOI] [PubMed] [Google Scholar]
  • 177. Nancy P, Tagliani E, Tay CS, Asp P, Levy DE, Erlebacher A. Chemokine gene silencing in decidual stromal cells limits T cell access to the maternal-fetal interface. Science 2012; 336:1317–1321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Konno T, Graham AR, Rempel LA, Ho-Chen JK, Alam SM, Bu P, Rumi MA, Soares MJ. Subfertility linked to combined luteal insufficiency and uterine progesterone resistance. Endocrinology 2010; 151:4537–4550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Cooke PS, Spencer TE, Bartol FF, Hayashi K. Uterine glands: development, function and experimental model systems. Mol Hum Reprod 2013; 19:547–558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Filant J, Spencer TE. Endometrial glands are essential for blastocyst implantation and decidualization in the mouse uterus. Biol Reprod 2013; 88:93. [DOI] [PubMed] [Google Scholar]
  • 181. Davila J, Laws MJ, Kannan A, Li Q, Taylor RN, Bagchi MK, Bagchi IC. Rac1 regulates endometrial secretory function to control placental development. PLoS Genet 2015; 11:e1005458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Dhakal P, Rumi MA, Kubota K, Chakraborty D, Chien J, Roby KF, Soares MJ. Neonatal progesterone programs adult uterine responses to progesterone and susceptibility to uterine dysfunction. Endocrinology 2015; 156:3791–3803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Rabaglino MB, Post Uiterweer ED, Jeyabalan A, Hogge WA, Conrad KP. Bioinformatics approach reveals evidence for impaired endometrial maturation before and during early pregnancy in women who developed preeclampsia. Hypertension. 2015; 65:421–429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Conrad KP, Rabaglino MB, Post Uiterweer ED. Emerging role for dysregulated decidualization in the genesis of preeclampsia. Placenta 2017; 60:119–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Garrido-Gomez T, Dominguez F, Quiñonero A, Diaz-Gimeno P, Kapidzic M, Gormley M, Ona K, Padilla-Iserte P, McMaster M, Genbacev O, Perales A, Fisher SJ et al. Defective decidualization during and after severe preeclampsia reveals a possible maternal contribution to the etiology. Proc Natl Acad Sci USA 2017; 114:E8468–E8477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Woods L, Perez-Garcia V, Kieckbusch J, Wang X, DeMayo F, Colucci F, Hemberger M. Decidualisation and placentation defects are a major cause of age-related reproductive decline. Nat Commun 2017; 8:352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Hanna J, Mandelboim O. When killers become helpers. Trends Immunol 2007; 28:201–206. [DOI] [PubMed] [Google Scholar]
  • 188. Sharma S. Natural killer cells and regulatory T cells in early pregnancy loss. Int J Dev Biol 2014; 58:219–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Guimond MJ, Luross JA, Wang B, Terhorst C, Danial S, Croy BA. Absence of natural killer cells during murine pregnancy is associated with reproductive compromise in TgE26 Mice. Biol Reprod 1997; 56:169–179. [DOI] [PubMed] [Google Scholar]
  • 190. Barber EM, Pollard JW. The uterine NK cell population requires IL-15 but these cells are not required for pregnancy nor the resolution of a Listeria monocytogenes infection. J Immunol 2003; 171:37–46. [DOI] [PubMed] [Google Scholar]
  • 191. Chakraborty D, Rumi MA, Konno T, Soares MJ. Natural killer cells direct hemochorial placentation by regulating hypoxia-inducible factor dependent trophoblast lineage decisions. Proc Natl Acad Sci USA 2011; 108:16295–16300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Fu B, Zhou Y, Ni X, Tong X, Xu X, Dong Z, Sun R, Tian Z, Wei H. Natural killer cells promote fetal development through the secretion of growth-promoting factors. Immunity 2017; 47:1100.e6–1113.e6. [DOI] [PubMed] [Google Scholar]
  • 193. Renaud SJ, Scott RL, Chakraborty D, Rumi MA, Soares MJ. Natural killer-cell deficiency alters placental development in rats. Biol Reprod 2017; 96:145–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Hanna J, Goldman-Wohl D, Hamani Y, Avraham I, Greenfield C, Natanson-Yaron S, Prus D, Cohen-Daniel L, Arnon TI, Manaster I, Gazit R, Yutkin V et al. Decidual NK cells regulate key developmental processes at the human fetal-maternal interface. Nat Med 2006; 12:1065–1074. [DOI] [PubMed] [Google Scholar]
  • 195. Moffett A, Chazara O, Colucci F. Maternal allo-recognition of the fetus. Fertil Steril 2017; 107:1269–1272. [DOI] [PubMed] [Google Scholar]
  • 196. Murphy SP, Fast LD, Hanna NN, Sharma S. Uterine NK cells mediate inflammation-induced fetal demise in IL-10-null mice. J Immunol 2005; 175:4084–4090. [DOI] [PubMed] [Google Scholar]
  • 197. Yougbaré I, Tai WS, Zdravic D, Oswald BE, Lang S, Zhu G, Leong-Poi H, Qu D, Yu L, Dunk C, Zhang J, Sled JG et al. Activated NK cells cause placental dysfunction and miscarriages in fetal alloimmune thrombocytopenia. Nat Commun 2017; 8:224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Beura LK, Hamilton SE, Bi K, Schenkel JM, Odumade OA, Casey KA, Thompson EA, Fraser KA, Rosato PC, Filali-Mouhim A, Sekaly RP, Jenkins MK et al. Normalizing the environment recapitulates adult human immune traits in laboratory mice. Nature 2016; 532:512–516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Simpson ER, Zhao Y, Agarwal VR, Michael MD, Bulun SE, Hinshelwood MM, Graham-Lorence S, Sun T, Fisher CR, Qin K, Mendelson CR. Aromatase expression in health and disease. Recent Prog Horm Res 1997; 52:185–213. [PubMed] [Google Scholar]
  • 200. Roberts RM, Chen Y, Ezashi T, Walker AM. Interferons and the maternal–conceptus dialog in mammals. Semin Cell Dev Biol 2008; 19:170–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Bazer FW, Thatcher WW. Chronicling the discovery of interferon tau. Reproduction 2017; 154:F11–F20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Ross GT. Clinical relevance of research on the structure of human chorionic gonadotropin. Am J Obstet Gynecol 1977; 129:795–808. [DOI] [PubMed] [Google Scholar]
  • 203. Henke A, Gromoll J. New insights into the evolution of chorionic gonadotrophin. Mol Cell Endocrinol 2008; 291:11–19. [DOI] [PubMed] [Google Scholar]
  • 204. Cooke NE, Liebhaber SA. Molecular biology of the growth hormone-prolactin gene system. Vitam Horm 1995; 50:385–459. [DOI] [PubMed] [Google Scholar]
  • 205. Soares MJ, Konno T, Alam SM. The prolactin family: effectors of pregnancy-dependent adaptations. Trends Endocrinol Metab 2007; 18:114–121. [DOI] [PubMed] [Google Scholar]
  • 206. Wiemers DO, Shao LJ, Ain R, Dai G, Soares MJ. The mouse prolactin gene family locus. Endocrinology 2003; 144:313–325. [DOI] [PubMed] [Google Scholar]
  • 207. Alam SM, Ain R, Konno T, Ho-Chen JK, Soares MJ. The rat prolactin gene family locus: species-specific gene family expansion. Mamm Genome 2006; 17:858–877. [DOI] [PubMed] [Google Scholar]
  • 208. Martal J, Lacroix MC, Loudes C, Saunier M, Wintenberger-Torrès S. Trophoblastin, an antiluteolytic protein present in early pregnancy in sheep. Reproduction 1979; 56:63–73. [DOI] [PubMed] [Google Scholar]
  • 209. Godkin JD, Bazer FW, Moffatt J, Sessions F, Roberts RM. Purification and properties of a major, low molecular weight protein released by the trophoblast of sheep blastocysts at Day 13-21. Reproduction 1982; 65:141–150. [DOI] [PubMed] [Google Scholar]
  • 210. Godkin JD, Bazer FW, Thatcher WW, Roberts RM. Proteins released by cultured Day 15-16 conceptuses prolong luteal maintenance when introduced into the uterine lumen of cyclic ewes. Reproduction 1984; 71:57–64. [DOI] [PubMed] [Google Scholar]
  • 211. Imakawa K, Anthony RV, Kazemi M, Marotti KR, Polites HG, Roberts RM. Interferon-like sequence of ovine trophoblast protein secreted by embryonic trophectoderm. Nature 1987; 330:377–379. [DOI] [PubMed] [Google Scholar]
  • 212. Stewart HJ, McCann SH, Barker PJ, Lee KE, Lamming GE, Flint AP. Interferon sequence homology and receptor binding activity of ovine trophoblast antiluteolytic protein. J Endocrinol 1987; 115:R13–R15. [DOI] [PubMed] [Google Scholar]
  • 213. Gibori G, Khan I, Warshaw ML, McLean MP, Puryear TK, Nelson S, Durkee TJ, Azhar S, Steinschneider A, Rao MC. Placental-derived regulators and the complex control of luteal cell function. Recent Prog Horm Res 1988; 44:377–429. [DOI] [PubMed] [Google Scholar]
  • 214. Yamamoto T, Chapman BM, Johnson DC, Givens CR, Mellon SH, Soares MJ. Cytochrome P450 17 alpha-hydroxylase gene expression in differentiating rat trophoblast cells. J Endocrinol 1996; 150:161–168. [DOI] [PubMed] [Google Scholar]
  • 215. Waterman MR, Simpson ER. Regulation of steroid hydroxylase gene expression is multifactorial in nature. Recent Prog Horm Res 1989; 45:533–563. [DOI] [PubMed] [Google Scholar]
  • 216. Escobar JC, Patel SS, Beshay VE, Suzuki T, Carr BR. The Human Placenta Expresses CYP17 and Generates Androgens De Novo. J Clin Endocrinol Metab 2011; 96:1385–1392. [DOI] [PubMed] [Google Scholar]
  • 217. Simpson ER, Zhao Y, Agarwal VR, Michael MD, Bulun SE, Hinshelwood MM, Graham-Lorence S, Sun T, Fisher CR, Qin K, Mendelson CR. Aromatase expression in health and disease. Recent Prog Horm Res 1997; 52:185–213. [PubMed] [Google Scholar]
  • 218. Kamat A, Hinshelwood MM, Murry BA, Mendelson CR. Mechanisms in tissue-specific regulation of estrogen biosynthesis in humans. Trends Endocrinol Metab 2002; 13:122–128. [DOI] [PubMed] [Google Scholar]
  • 219. Francino MP. An adaptive radiation model for the origin of new gene functions. Nat Genet 2005; 37:573–578. [DOI] [PubMed] [Google Scholar]
  • 220. Kidwell MG, Lisch DR. Perspective: transposable elements, parasitic DNA, and genome evolution. Evolution 2001; 55:1–24. [DOI] [PubMed] [Google Scholar]
  • 221. Feschotte C, Gilbert C. Endogenous viruses: insights into viral evolution and impact on host biology. Nat Rev Genet 2012; 13:283–296. [DOI] [PubMed] [Google Scholar]
  • 222. Rebollo R, Romanish MT, Mager DL. Transposable elements: an abundant and natural source of regulatory sequences for host genes. Annu Rev Genet 2012; 46:21–42. [DOI] [PubMed] [Google Scholar]
  • 223. Chuong EB, Elde NC, Feschotte C. Regulatory activities of transposable elements: from conflicts to benefits. Nat Rev Genet 2017; 18:71–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224. Frank JA, Feschotte C. Co-option of endogenous viral sequences for host cell function. Curr Opin Virol 2017; 25:81–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225. Dupressoir A, Lavialle C, Heidmann T. From ancestral infectious retroviruses to bona fide cellular genes: role of the captured syncytins in placentation. Placenta 2012;33:663–671. [DOI] [PubMed] [Google Scholar]
  • 226. Lavialle C, Cornelis G, Dupressoir A, Esnault C, Heidmann O, Vernochet C, Heidmann T. Paleovirology of 'syncytins', retroviral env genes exapted for a role in placentation. Philos Trans R Soc Lond B Biol Sci 2013; 368:20120507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227. Dupressoir A, Vernochet C, Harper F, Guégan J, Dessen P, Pierron G, Heidmann T. A pair of co-opted retroviral envelope syncytin genes is required for formation of the two-layered murine placental syncytiotrophoblast. Proc Natl Acad Sci USA 2011; 108:E1164–E1173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228. Dunlap KA, Palmarini M, Varela M, Burghardt RC, Hayashi K, Farmer JL, Spencer TE. Endogenous retroviruses regulate periimplantation placental growth and differentiation. Proc Natl Acad Sci USA 2006; 103:14390–14395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229. Chuong EB, Rumi MA, Soares MJ, Baker JC. Endogenous retroviruses function as species-specific enhancer elements in the placenta. Nat Genet 2013; 45:325–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230. Chapman V, Forrester L, Sanford J, Hastie N, Rossant J. Cell lineage-specific undermethylation of mouse repetitive DNA. Nature 1984; 307:284–286. [DOI] [PubMed] [Google Scholar]
  • 231. Rossant J, Sanford JP, Chapman VM, Andrews GK. Undermethylation of structural gene sequences in extraembryonic lineages of the mouse. Dev Biol 1986; 117:567–573. [DOI] [PubMed] [Google Scholar]
  • 232. Novakovic B, Saffery R. Placental pseudo-malignancy from a DNA methylation perspective: unanswered questions and future directions. Front Genet 2013; 4:285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233. Macaulay EC, Chatterjee A, Cheng X, Baguley BC, Eccles MR, Morison IM. The genes of life and death: a potential role for placental-specific genes in cancer. Bioessays. 2017; 39:11. [DOI] [PubMed] [Google Scholar]
  • 234. Wilson SL, Robinson WP. Utility of DNA methylation to assess placental health. Placenta. 2017. DOI: 10.1016/j.placenta.2017.12.013. [DOI] [PubMed]
  • 235. Rosario GX, Konno T, Soares MJ. Maternal hypoxia activates endovascular trophoblast cell invasion. Dev Biol 2008; 314:362–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236. Genbacev O, Joslin R, Damsky CH, Polliotti BM, Fisher SJ. Hypoxia alters early gestation human cytotrophoblast differentiation/invasion in vitro and models the placental defects that occur in preeclampsia. J Clin Invest 1996; 97:540–550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237. Genbacev O, Zhou Y, Ludlow JW, Fisher SJ. Regulation of human placental development by oxygen tension. Science 1997; 277:1669–1672. [DOI] [PubMed] [Google Scholar]
  • 238. Bu P, Alam SM, Dhakal P, Vivian JL, Soares MJ. A prolactin family paralog regulates placental adaptations to a physiological stressor. Biol Reprod 2016; 94:107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239. Bronson SL, Bale TL. The placenta as a mediator of stress effects on neurodevelopmental reprogramming. Neuropsychopharmacol 2016;41:207–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. Vrooman LA, Xin F, Bartolomei MS. Morphologic and molecular changes in the placenta: what we can learn from environmental exposures. Fertil Steril 2016; 106:930–940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241. Soares MJ, Iqbal K, Kozai K. Hypoxia and placental development. Birth Defects Res 2017; 109:1309–1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242. Wakeland AK, Soncin F, Moretto-Zita M, Chang CW, Horii M, Pizzo D, Nelson KK, Laurent LC, Parast MM. Hypoxia directs human extravillous trophoblast differentiation in a hypoxia-inducible factor–dependent manner. Am J Pathol 2017; 187:767–780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243. Chang CW, Wakeland AK, Parast MM. Trophoblast lineage specification, differentiation and their regulation by oxygen tension. J Endocrinol 2018; 236:R43–R56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244. Clifton VL. Review: sex and the human placenta: mediating differential strategies of fetal growth and survival. Placenta 2010; 31(Suppl):S33–S39. [DOI] [PubMed] [Google Scholar]
  • 245. Nugent BM, Bale TL. The omniscient placenta: metabolic and epigenetic regulation of fetal programming. Front Neuroendocrinol 2015; 39:28–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246. Rosenfeld CS. Sex-specific placental responses in fetal development. Endocrinology 2015; 156:3422–3434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247. Kalisch-Smith JI, Simmons DG, Dickinson H, Moritz KM. Review: sexual dimorphism in the formation, function and adaptation of the placenta. Placenta 2017; 54:10–16. [DOI] [PubMed] [Google Scholar]
  • 248. Gallou-Kabani C, Gabory A, Tost J, Karimi M, Mayeur S, Lesage J, Boudadi E, Gross MS, Taurelle J, Vigé A, Breton C, Reusens B et al. Sex- and diet-specific changes of imprinted gene expression and DNA methylation in mouse placenta under a high-fat diet. PLoS One 2010; 5:e14398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249. Gabory A, Ferry L, Fajardy I, Jouneau L, Gothié JD, Vigé A, Fleur C, Mayeur S, Gallou-Kabani C, Gross MS, Attig L, Vambergue A et al. Maternal diets trigger sex-specific divergent trajectories of gene expression and epigenetic systems in mouse placenta. PLoS One 2012; 7:e47986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250. Cleaton MA, Edwards CA, Ferguson-Smith AC. Phenotypic outcomes of imprinted gene models in mice: elucidation of pre- and postnatal functions of imprinted genes. Annu Rev Genom Hum Genet 2014; 15:93–126. [DOI] [PubMed] [Google Scholar]
  • 251. Marsit CJ. Placental epigenetics in children's environmental health. Semin Reprod Med 2016; 34:36–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252. Eakin GS, Behringer RR. Tetraploid development in the mouse. Dev Dyn 2003; 228:751–766. [DOI] [PubMed] [Google Scholar]
  • 253. Weier JF, Weier HU, Jung CJ, Gormley M, Zhou Y, Chu LW, Genbacev O, Wright AA, Fisher SJ. Human cytotrophoblasts acquire aneuploidies as they differentiate to an invasive phenotype. Dev Biol 2005; 279:420–432. [DOI] [PubMed] [Google Scholar]
  • 254. Yuen RK, Robinson WP. Review: a high capacity of the human placenta for genetic and epigenetic variation: implications for assessing pregnancy outcome. Placenta 2011; 32(Suppl 2):S136–S141. [DOI] [PubMed] [Google Scholar]
  • 255. Kasak L, Rull K, Sõber S, Laan M. Copy number variation profile in the placental and parental genomes of recurrent pregnancy loss families. Sci Rep 2017; 7:45327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256. Guttmacher AE, Maddox YT, Spong CY. The human placenta project: placental structure, development, and function in real time. Placenta 2014; 35:303–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257. Barker DJ, Bull AR, Osmond C, Simmonds SJ. Fetal and placental size and risk of hypertension in adult life. BMJ 1990; 301:259–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258. Barker DJ, Thornburg KL. Placental programming of chronic diseases, cancer and lifespan: a review. Placenta 2013; 34:841–845. [DOI] [PubMed] [Google Scholar]
  • 259. Rubens CE, Sadovsky Y, Muglia L, Gravett MG, Lackritz E, Gravett C. Prevention of preterm birth: harnessing science to address the global epidemic. Sci Transl Med 2014; 6:262sr5–262sr5. [DOI] [PubMed] [Google Scholar]
  • 260. Partridge EA, Davey MG, Hornick MA, McGovern PE, Mejaddam AY, Vrecenak JD, Mesas-Burgos C, Olive A, Caskey RC, Weiland TR, Han J, Schupper AJ et al. An extra-uterine system to physiologically support the extreme premature lamb. Nat Commun 2017; 8:15112. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Biology of Reproduction are provided here courtesy of Oxford University Press

RESOURCES